ArticlePDF Available

Functional Polymorphisms in the TERT Promoter Are Associated with Risk of Serous Epithelial Ovarian and Breast Cancers

PLOS
PLOS One
Authors:

Abstract and Figures

Genetic variation at the TERT-CLPTM1L locus at 5p15.33 is associated with susceptibility to several cancers, including epithelial ovarian cancer (EOC). We have carried out fine-mapping of this region in EOC which implicates an association with a single nucleotide polymorphism (SNP) within the TERT promoter. We demonstrate that the minor alleles at rs2736109, and at an additional TERT promoter SNP, rs2736108, are associated with decreased breast cancer risk, and that the combination of both SNPs substantially reduces TERT promoter activity.
Content may be subject to copyright.
Functional Polymorphisms in the
TERT
Promoter Are
Associated with Risk of Serous Epithelial Ovarian and
Breast Cancers
Jonathan Beesley
1
*
.
, Hilda A. Pickett
2,3.
, Sharon E. Johnatty
1
, Alison M. Dunning
4
, Xiaoqing Chen
1
, Jun
Li
1
, Kyriaki Michailidou
4
,YiLu
1
, David N. Rider
5
, Rachel T. Palmieri
6
, Michael D. Stutz
2,3
, Diether
Lambrechts
7,8
, Evelyn Despierre
9
, Sandrina Lambrechts
9
, Ignace Vergote
9
, Jenny Chang-Claude
10
,
Stefan Nickels
10
, Alina Vrieling
10
, Dieter Flesch-Janys
11
, Shan Wang-Gohrke
12
, Ursula Eilber
10
, Natalia
Bogdanova
13,14,15
, Natalia Antonenkova
14
, Ingo B. Runnebaum
16
, Thilo Do
¨rk
13
, Marc T. Goodman
17
,
Galina Lurie
17
, Lynne R. Wilkens
17
, Rayna K. Matsuno
17
, Lambertus A. Kiemeney
18,19,20
, Katja K. H.
Aben
18,20
, Tamara Marees
18
, Leon F. A. G. Massuger
21
, Brooke L. Fridley
5
, Robert A. Vierkant
5
, Elisa V.
Bandera
22
, Sara H. Olson
23
, Irene Orlow
23
, Lorna Rodriguez-Rodriguez
22
, Linda S. Cook
24
, Nhu D. Le
24
,
Angela Brooks-Wilson
24
, Linda E. Kelemen
24
, Ian Campbell
25
, Simon A. Gayther
26
, Susan J. Ramus
26
,
Aleksandra Gentry-Maharaj
27
, Usha Menon
27
, Shahana Ahmed
4
, Caroline Baynes
4
, Paul D. Pharoah
4
,
kConFab Investigators
25
, Australian Ovarian Cancer Study Group
1,25,28
, ABCTB Investigators, Kenneth
Muir
30
, Artitaya Lophatananon
30
, Arkom Chaiwerawattana
31
, Surapon Wiangnon
32
, Stuart Macgregor
1
,
Douglas F. Easton
4
, Roger R. Reddel
2,3
, Ellen L. Goode
5
, Georgia Chenevix-Trench
1
on behalf of the
Ovarian Cancer Association Consortium
1Division of Genetics and Population Health, Queensland Institute of Medical Research, Brisbane, Queensland, Australia, 2Cancer Research Unit, Children’s Medical
Research Institute, Westmead, New South Wales, Australia, 3Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia, 4Department of Oncology,
University of Cambridge, Strangeways Research Laboratory, Cambridge, United Kingdom, 5Department of Health Sciences Research, Mayo Clinic College of Medicine,
Rochester, Minnesota, United States of America, 6Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina, United
States of America, 7Vesalius Research Center, VIB, Leuven, Belgium, 8Vesalius Research Center, University of Leuven, Leuven, Belgium, 9Division of Gynaecologic
Oncology, Department of Obstetrics and Gynaecology, University Hospitals Leuven, University of Leuven, Leuven, Belgium, 10 Division of Cancer Epidemiology, German
Cancer Research Center (DKFZ), Heidelberg, Germany, 11 Institute for Medical Biometrics and Epidemiology, University Clinic Hamburg-Eppendorf, Hamburg, Germany,
12 Department of Obstetrics and Gynecology, University of Ulm, Ulm, Germany, 13 Clinics of Obstetrics and Gynaecology, Hannover Medical School, Hannover, Germany,
14 Byelorussian Institute for Oncology and Medical Radiology Aleksandrov N.N., Minsk, Belarus, 15 Clinics of Radiation Oncology, Hannover Medical School, Hannover,
Germany, 16 Clinics of Obstetrics and Gynaecology, Friedrich Schiller University, Jena, Germany, 17 Cancer Epidemiology Program, University of Hawaii Cancer Center,
Honolulu, Hawaii, United States of America, 18 Department of Epidemiology, Biostatistics and HTA, Radboud University Nijmegen Medical Centre, Nijmegen, The
Netherlands, 19 Department of Urology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands, 20 Comprehensive Cancer Center, The Netherlands,
Nijmegen, The Netherlands, 21 Department of Gynaecology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands, 22 The Cancer Institute of New
Jersey/Robert Wood Johnson Medical School, New Brunswick, New Jersey, United States of America, 23 Department of Epidemiology & Biostatistics, Memorial Sloan
Kettering Cancer Center, New York, New York, United States of America, 24 Department of Population Health Research, Alberta Health Services-Cancer Care, Calgary,
Alberta, Canada, 25 Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia, 26 Department of Preventive Medicine, Keck School of Medicine, University of
Southern California, Los Angeles, California, United States of America, 27 Department of Gynaecological Oncology, University College London, EGA Institute for Women’s
Health, London, United Kingdom, 28 Department of Gynaecological Oncology and Westmead Institute for Cancer Research, University of Sydney at the Westmead
Millennium Institute, Westmead Hospital, Sydney, New South Wales, Australia, 29 Westmead Millennium Institute, Sydney Medical School, Westmead, The University of
Sydney, Sydney, New South Wales, Australia, 30 Health Sciences Research Institute, Warwick Medical School, University of Warwick, Coventry, United Kingdom,
31 Department of Academic Support, The National Cancer Institute of Thailand, Ministry of Public Health, Nonthaburi, Thailand, 32 Department of Pediatrics, Medical
School, Khon Kaen University, Khon Kaen, Thailand
Abstract
Genetic variation at the TERT-CLPTM1L locus at 5p15.33 is associated with susceptibility to several cancers, including
epithelial ovarian cancer (EOC). We have carried out fine-mapping of this region in EOC which implicates an association with
a single nucleotide polymorphism (SNP) within the TERT promoter. We demonstrate that the minor alleles at rs2736109, and
at an additional TERT promoter SNP, rs2736108, are associated with decreased breast cancer risk, and that the combination
of both SNPs substantially reduces TERT promoter activity.
PLoS ONE | www.plosone.org 1 September 2011 | Volume 6 | Issue 9 | e24987
Citation: Beesley J, Pickett HA, Johnatty SE, Dunning AM, Chen X, et al. (2011) Functional Polymorphisms in the TERT Promoter Are Associated with Risk of Serous
Epithelial Ovarian and Breast Cancers. PLoS ONE 6(9): e24987. doi:10.1371/journal.pone.0024987
Editor: Lin Zhang, University of Pennsylvania School of Medicine, United States of America
Received August 12, 2011; Accepted August 21, 2011; Published September 15, 2011
This is an open-access article distributed under the terms of the Creative Commons Public Domain declaration which stipulates that, once placed in the public
domain, this work may be freely reproduced, distributed, transmitted, modified, built upon, or otherwise used by anyone for any lawful purpose.
Funding: The Ovarian Cancer Association Consortium is supported by a grant from the Ovarian Cancer Research Fund. kConFaB is supported by the National
Breast Cancer Foundation, the National Health and Medical Research Council of Australia, and the Cancer Councils of Queensland, New South Wales, Western
Australia, South Australia, and Victoria. The Clinical Follow-up Study of kConFaB is funded by the National Health and Medical Research Council (NHMRC) of
Australia (145684 and 288704). AOCS was supported by the U.S. Army Medical Research and Materiel Command (DAMD17-01-1-0729), the National Health and
Medical Research Council of Australia (199600), and the Cancer Council Tasmania and Cancer Foundation of Western Australia. Tissues and samples were received
from the Australian Breast Cancer Tissue Bank which is supported by the National Health and Medical Research Council of Australia, The Cancer Institute NSW and
the National Breast Cancer Foundation. The tissues and samples are made available to researchers on a non-exclusive basis. The BEL study was supported by the
Nationaal Kankerplan – actie 29. The HAW study is supported by the U.S. National Institutes of Health (R01 CA58598, N01-CN-55424, N01-PC-67001). SEARCH was
supported by Cancer Research-UK grants [C1287/A10118, C490/A11021, C8197/A10123]. The MAYO study was supported by R01 CA122443 and P50 CA136939.
The MARIE study was supported by the Deutsche Krebshilfe e.V., grant number 70-2892-BR I, the Hamburg Cancer Society, the German Cancer Research Center
(DKFZ) and the DNA extraction and genotype work in part by the Federal Ministry of Education and Research (BMBF) Germany grant 01KH0402. The GESBC
epidemiological study was supported by the Deutsche Krebshilfe e. V. [70492] and GESBC genotyping in part by the state of Baden-Wu
¨rttemberg through the
Medical Faculty of the University of Ulm [P.685]. The OVAL-BC Study is supported by the British Columbia Workers Compensation Board and the Canadian
Institutes for Health Research. The NJO study was funded by NIH-K07 CA095666, R01CA83918, and The Cancer Institute of New Jersey. The UKO study is
supported by funding from Cancer Research UK, the Eve Appeal, and the OAK Foundation; some of this work was undertaken at UCLH/UCL which received some
funding from the Department of Health’s NIHR Biomedical Research Centre funding scheme. ACP study is funded by The Breast Cancer Research Trust, UK. JB,
HAP, SEJ, MDS and RRR are supported by NHMRC project grant 1012023. DFE is a Principal Research Fellow of Cancer Research UK. SM is supported by an NHMRC
career development award. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
Competing Interests: The authors have declared that no competing interests exist.
* E-mail: jonathan.beesley@qimr.edu.au
.These authors contributed equally to this work.
Introduction
Genome-wide association studies (GWAS) have identified more
than 140 cancer susceptibility loci for 17 different cancers (www.
genome.gov/gwastudies), including a locus at 5p15.33 which has
been implicated in susceptibility to melanoma [1,2], glioma [3],
lung [4,5], pancreas [6], prostate [2], testicular [7], and bladder
cancers [8]. This locus harbours TERT, encoding the reverse
transcriptase component of telomerase, and cleft lip and palate
transmembrane protein 1-like (CLPTM1L). GWAS of ovarian and
breast cancer have not detected an association with this locus to
date, which may be due to poor tagging of this region on the chips
employed, or the lack of statistical power to detect associations.
Using a candidate gene approach we previously reported evidence
of an association between an intronic SNP in TERT (rs7726159)
and EOC risk, particularly of the serous histological subtype, using
a per-allele model [9]. In this study we carried out fine-mapping,
followed by functional analyses of associated SNPs identified
within the promoter region of TERT. We directly demonstrate
that the presence of a common haplotype, which is associated with
decreased cancer risk, substantially reduces TERT promoter
activity. The vast majority of cancers depend on expression of
telomerase, which requires substantial upregulation of TERT
expression, for their continued proliferation, strongly implicating
TERT as the predominant gene involved in this association.
Results and Discussion
To further clarify the association previously reported with
TERT, we employed a fine-mapping strategy in nine case-control
studies from the Ovarian Cancer Association Consortium (OCAC)
(Table S1). SNPs in a region spanning 6250 kb across the TERT,
CLPT1ML,SLC6A18 and SLC6A19 genes that were correlated
(0.2#r
2
#0.99) with rs7726159, rs11133719, rs2735940 and
rs2736100 [9] were selected from the 1000 Genomes low coverage
pilot release of April 2009. The SNPs implicated by our previous
study of EOC [9] (rs11133719, rs7726159, rs2736100 and
rs2735940) or by cancer GWAS (rs2736100) were also included
in the panel. We genotyped 36 SNPs by iPLEX (Sequenom Inc.)
in 2,130 invasive EOC cases and 3,975 controls, all of Caucasian
ancestry. After excluding monomorphic loci (n = 6) and two SNPs
that failed OCAC’s quality control criteria [10], 28 SNPs were
analysed for association with risk of EOC. We used single marker
and stepwise logistic regression models, adjusted for study and age
(at interview for controls and at diagnosis for cases) with a
threshold of P#0.05 for addition (forward stepwise) or removal
(backward stepwise) of SNPs (Table S2). We found that
rs2736109 showed the strongest association with serous EOC
(adjusted OR
per-allele
0.86 (0.77–0.96), P= 0.005) (Table S3), but
not with invasive EOC risk overall (adjusted OR
per-allele
0.96
(0.89–1.05), P= 0.38) (data not shown). Likelihood ratio tests
comparing logistic regression models with and without a genotype-
by-study interaction term revealed no significant study heteroge-
neity (P= 0.4). rs2736109 is in a region of low linkage
disequilibrium that encompasses the 59end of the TERT gene
and the TERT promoter (Figure 1). This region also contains the
SNPs, rs2736108 and rs2853669, which have pairwise correlations
(r
2
) with each other, and with rs2736109 of greater than 0.6. It has
previously been reported that rs2853669 is associated with breast
cancer risk [11].
Since other loci confer susceptibility to both EOC and breast
cancer [12,13] we investigated associations of TERT SNPs with
breast cancer risk in various data sets. First, we genotyped
rs2736109 by iPLEX in 4,277 invasive breast cancer cases and
7,000 controls from Australian, German and Thai studies from the
Breast Cancer Association Consortium (BCAC) (Table S4). In the
combined analysis of the Australian and German studies, there
was no association with invasive breast cancer risk overall
(adjusted OR
per-allele
0.95 (0.90–1.01), P= 0.10), but there was
an association in cases $50 years at diagnosis that approached
significance (adjusted OR
per-allele
0.94 (0.88–1.00), P= 0.049). We
also found a stronger association for ER-negative tumours
(adjusted OR
per-allele
0.88 (0.80–0.98), P= 0.022; for ER-positive
tumours adjusted OR
per-allele
0.98 (0.92–1.05), P= 0.562) (Tables
S5, S6, and S7). We found the strongest evidence of association
among ER-negative cases over the age of 50 (n = 636) (adjusted
OR
per-allele
0.84 (0.75–0.95), P= 0.005). We did not find any
TERT SNPs and Ovarian and Breast Cancer Risk
PLoS ONE | www.plosone.org 2 September 2011 | Volume 6 | Issue 9 | e24987
association with breast cancer risk in a Thai study (n = 327 cases)
genotyped for rs2736109.
Next, we analysed genotypes imputed using MaCH for
rs2736109 (r
2
= 0.45) from a breast cancer GWAS of 3,931 cases
and 3,622 controls from the United Kingdom, from which neither
age nor ER status were available [14]. We observed a significant
association between rs2736109 and overall breast cancer risk in
this population (OR
per-allele
0.91 (0.83–0.99), P= 0.037), and from
a weighted meta-analysis of imputed and genotyped data from all
studies (OR
per-allele
0.94 (0.89–0.98), P= 0.011).
In an additional replication sample set (SEARCH) we
genotyped a correlated SNP, rs2736108 (r
2
= 0.96, 1000 Genomes
Project, Dec 2009, r
2
= 0.63 based on 345 Australian controls) in
6,788 cases and 6,426 controls because rs2736109 was not
amenable to genotyping by TaqMan (the genotyping platform
used by this group). We observed a significant association with
breast cancer risk overall (adjusted OR
per-allele
0.92 (0.87–0.97),
P= 0.003), which was restricted to the subset of cases diagnosed at
age 50 or older (adjusted OR
per-allele
0.91 (0.85–0.97), P= 0.002)
(Table S6). Estimates according to ER status showed a significant
association between the rs2736108 genotypes and ER-positive
tumours (adjusted OR
per-allele
0.93 (0.88–0.99), P= 0.031); there
was no significant association in ER-negative tumours but the
estimated OR was similar (OR
per-allele
0.95 (0.85–1.06)) and the
sample size much smaller. Comparison of models for both
rs2736109 and rs2736108 with and without genotype by age
group interaction terms showed no evidence of a statistical
interaction on the multiplicative scale (P
interaction
$0.25).
The SNPs of interest (rs2736108, rs2736109 and rs2853669) lie
within the upstream promoter region of TERT. To determine the
functional significance of these sites, we generated combinations of
these variants in a luciferase reporter construct containing 3.9 kb
of the TERT promoter [15]. Relative promoter activity was
determined in an EOC cell line (27/87), a breast adenocarcinoma
cell line (MDA-MB-468), and in post-selection normal breast
epithelial cells (Bre16) (Figure 2). Introduction of a mutation into
the human estrogen-responsive element in the TERT promoter
(TERT-ERE) [16] was used as a positive control and confirmed
diminished reporter activity. In all three cell types, luciferase
activity was substantially reduced for the construct carrying both
minor (A) alleles at rs2736108 and rs2736109, but remained
unaltered for those with the individual minor allele at either SNP.
We observed no change in expression for the minor allele at
rs2853669.
Our analysis of Australian controls estimated a frequency of 32%
for the A-A haplotype at rs2736108 and rs2736109, suggesting that
this relatively common promoter haplotype may lower the risk of
ovarian and breast cancer through decreasing TERT expression. This
finding provides no support for the hypothesis that decreased
telomerase activity predisposes to genomic instability and consequent
oncogenic progression but instead our data imply the opposite,
namely, that decreased TERT expression confers decreased cancer risk.
Figure 1.
TERT-CLPTM1L
locus SNPs genotyped in epithelial ovarian cancer cases and controls. Panel Adepicts the pattern of linkage
disequilibrium using data from the HapMap phase II+III CEU population (2010), where white represents r
2
= 0, and black r
2
= 1. The plot in B
represents 2log
10
pagainst the chromosomal position for genotyped SNPs in the ovarian cancer fine mapping panel. The purple diamond represents
the SNP (rs2736109) with the strongest observed association. The point colours represent the strength of LD according to 1000 Genomes Data.
doi:10.1371/journal.pone.0024987.g001
TERT SNPs and Ovarian and Breast Cancer Risk
PLoS ONE | www.plosone.org 3 September 2011 | Volume 6 | Issue 9 | e24987
We also examined tumour expression and germline variants at
the TERT-CLPTM1L locus using data from The Cancer Genome
Atlas (TCGA) ovarian serous cystadenocarcinoma set. mRNA
expression profiling was available for 574 tumour samples using
Affymetrix U133A platform This assay has one probe for TERT,
but none for CLPTM1L.TERT was expressed at low levels, as
expected, but this does not necessarily reflect low levels of
telomerase protein expression or activity [17]. 508 normal DNA
samples were genotyped using Illumina 1 M array, and the SNP
rs2736108 was typed on this array. We also imputed the TCGA
samples with reference to the 1000 Genome data (June 2010
release); rs2853669 was successfully imputed, but rs2736109 failed
quality control. There was no evidence suggesting that the
genotyped SNP, rs2736108, or the imputed SNP, rs2853669,
are associated with expression of TERT. However one SNP,
rs2735845, in partial linkage disequilibrium with the typed SNP,
rs2736108 (r
2
= 0.306), was associated with significantly altered
transcript abundance of TERT (P=4610
24
and P= 0.0029 after
correction for multiple testing). Gene copy number aberration was
found in TERT; it was amplified in ,20% of tumour samples.
This SNP showed robust association after adjusting for the copy
number variation (P= 1.8610
23
and P= 0.015 after correction for
multiple testing), and it explained 1.85% of variance of TERT
transcription in ovarian cancer tumours.
A recently published meta-analysis identified a significantly
decreased risk of breast cancer with a TERT SNP, rs2853669 (OR
0?76 (0?64–0?91), P=0?002) [11]. This SNP lies in the same
linkage disequilibrium block as rs2736108 and rs2736109 (r
2
.0.6
with rs2736108); therefore, this meta-analysis provides additional
support for an association between TERT promoter SNPs and
breast cancer risk. However, our functional analyses, which were
carried out in normal breast epithelial cells and a breast
adenocarcinoma cell line did not indicate any change to TERT
expression with this individual SNP.
We computed a gene-based test [18] of association at TERT
and CLPTM1L which yielded evidence in GWAS data available
from dbGAP (http://www.ncbi.nlm.nih.gov/gap) for association
with risk of lung, prostate and pancreatic cancer, but not breast
cancer overall. Combining data for all cancers in a cross-cancer
meta-analysis, revealed a genome-wide significant gene-based
P= 4.1610
27
for CLPTM1L (P= 0.008 after correction for 19,000
genes). A similar result was obtained for TERT (all cancer
P= 7.7610
25
). This gene-based test includes all SNPs within
50 kb of the start/stop site of each gene. Most of the associated
SNPs lie in the interval between TERT and CLPTM1L but with
slightly more evidence for SNPs near to CLPTM1L, leading to a
slightly higher gene-based Pvalue for CLPTM1L. These results are
based on marker data from Illumina GWAS arrays and hence the
exact location of the maximum association test statistic is
dependent to some degree upon the arbitrary set of SNPs that
are on the arrays. Clearly, additional analysis of the entire TERT-
Figure 2.
TERT
promoter activity. Luciferase reporter assays
following transient transfection with pGL2-control (SV40 promoter
and enhancer), pGL2-basic (lacks promoter and enhancer), transfection
control (blank), and the TERT reporter vectors TERT WT (3.9 kb of TERT
promoter), the positive control TERT-ERE (containing a mutation in the
estrogen-responsive element), and the minor alleles of rs2736108 (A
allele), rs2736109 (A allele), rs2736108/9 (A alleles at both sites),
rs2853669 (C allele) in (A) an EOC cell line (27/87), (B) a breast
adenocarcinoma cell line (MDA-MB-468) and (C) a normal breast
epithelial cell strain (Bre16). Error bars represent standard deviation
between three separate experiments. * represents statistical signifi-
cance using one-way ANOVA with post hoc Dunnett’s tests.
doi:10.1371/journal.pone.0024987.g002
TERT SNPs and Ovarian and Breast Cancer Risk
PLoS ONE | www.plosone.org 4 September 2011 | Volume 6 | Issue 9 | e24987
CLPM1L locus is warranted in breast, and other, cancers to
validate the associations we have identified and fine map putative
causal variants if our findings are confirmed.
The failure to date to identify an association between the
TERT-CLPTM1L locus and risk of EOC or breast cancer by
GWAS may be explained by the pattern of linkage disequilibrium
of the relevant SNPs: rs2736100, the tagSNP most commonly
identified by GWAS of other cancers is poorly correlated with
rs2736108 and rs2736109 (r
2
= 0.141 and 0.105, respectively), and
neither of these SNPs are on the Illumina 300 K, 610 K or 650 K
arrays used for most cancer GWAS.
In summary, we have demonstrated a direct association
between functional SNPs in the TERT promoter, which confer
decreased risk of ovarian and breast cancer, and reduced TERT
promoter activity. Decreased levels of TERT result in progressive
telomere shortening and the onset of cellular senescence, which
ultimately acts to suppress tumorigenesis. The association of
hypomorphic sequence variants in the TERT promoter with
decreased risk of cancer implicates downregulation of telomerase
and telomere shortening as an intrinsic tumour suppressive
mechanism. It is also possible that TERT variants associated with
elevated cancer risk may alter the stringency with which TERT is
regulated, potentially facilitating TERT activation and conse-
quently providing a tumorigenic advantage. Potential non-
canonical roles of TERT in cell signalling pathways may also
underlie cancer risk [19]. Our results add functional insight into
the increasingly important role of TERT as a cancer risk factor and
demonstrate the need for further mechanistic analysis of this multi-
cancer susceptibility locus.
Materials and Methods
Ethics Statement
Approval for this study was obtained from The Queensland
Institute of Medical Research Human Research Ethics Commit-
tee. All studies were approved by the review boards and ethics
committees of their respective institutions, and all participants
provided written informed consent.
Genotyping
iPLEX genotyping was carried out using MALDI-TOF
spectroscopy utilising Sequenom’s MassARRAY platform and
iPLEX GOLD chemistry. 10 ng of genomic DNA was used as
template, to which a PCR mix containing Qiagen Hot-StarTaq
was added. Shrimp alkaline phosphatase and primer extension
steps were carried out using Sequenom’s protocol and reagents.
Primers were obtained from Integrated DNA Technologies (Ohio
USA). Assays were designed with MassARRAY Assay Design
version 3.1(Sequenom). Raw genotype data were visualised and
processed with MassARRAY Typer software version 3.4. TaqMan
genotyping (SEARCH) was carried out as previously described
[20]. Strict quality control criteria were adhered to as part of the
Ovarian Cancer Association Consortium’s guidelines [10].
Statistical methods
We used single marker and stepwise logistic regression models to
screen 28 SNPs in non-Hispanic white ovarian cancer cases
(n = 2,130) and controls (n = 3,975) from nine OCAC studies
(Supplementary Table 1). Genotype data for all the previously
reported TERT SNPs [9] has been excluded from the current
analysis. Stepwise models were adjusted for study and age (at
interview for controls and at diagnosis for cases), with a threshold
of P#0.05 for addition (forward stepwise) or removal (backward
stepwise) of SNPs. All single marker risk estimates were obtained
from unconditional logistic regression models adjusted for age
(where available) and additionally for study where data was pooled
across multiple studies. Assuming a log additive model of
inheritance, the per-allele odds ratios (ORs) and their 95%
confidence intervals (CIs) associated for selected SNPs were
estimated by fitting the number of rare alleles carried as a
continuous covariate. Ovarian cancer risk associated with SNP
genotypes were obtained for all invasive cases as well as a subset of
serous cases. Breast cancer risk estimates were obtained for
invasive cases and by estrogen receptor (ER) status. Separate
comparisons were made for cases diagnosed before 50 vs. $50
years of age to explore effect modification by advancing age of
diagnosis. Summary estimates from pooled analyses using
genotyped and imputed SNP data were obtained from weighted
meta-analysis of study-specific parameter estimates (bcoefficients
and Standard Error). The minor allele frequency (MAF) for each
SNP was estimated from the control population for each study.
Study heterogeneity and risk differences associated with age
groups (,50 vs. $50) were assessed using the likelihood ratio test
to compare logistic regression models with and without a
multiplicative interaction term. All tests for association were two-
tailed, statistical significance was assessed at P#0.05, and were
performed in STATA SE v.11 (StataCorp, USA), and SAS v. 9.1.
Tests for study heterogeneity and age group interaction tests were
implemented in the R project for Statistical Computing (http://
www.r-project.org/).
VEGAS applied to dbGAP data
To evaluate evidence for association at TERT and CLPTM1L
with various cancers, we applied the gene-based test implemented
in VEGAS (all SNPs in gene test) [18] to data from dbGAP. In
brief, we selected cancer cases and controls from dbGAP that were
genotyped on ,550,000 SNPs (Illumina 610 quad or Illumina
HumanHap550 arrays). Studies were CGEMS breast (1145 cases,
1142 controls), CGEMS pancreatic cancer (2328 cases, 2351
controls), GENEVA lung cancer (EAGLE and PLCO combined
2748 cases, 2840 controls) and CGEMS prostate (1145 cases, 1054
controls). For full details see dbGAP website (http://www.ncbi.
nlm.nih.gov/gap). In each case, following standard quality control,
the genomic control lambda was as would be expected if cases and
controls were well matched; breast, pancreatic, lung, prostate
lambda values 1.01, 1.01, 1.03, 1.03, respectively. The VEGAS
gene based Pvalues from each of the four dbGAP studies were
combined in a meta-analysis using Fisher’s method for combining
Pvalues.
Luciferase assays
Variants were introduced into pGL3-hTERT-3915 [15] by
site-directed mutagenesis (Agilent Technologies). The A-A vector
was generated by introducing the variants sequentially. Cells were
transfected using siPORT NeoFX Transfection Agent (Ambion),
according to the manufacturer’s instructions, and harvested after
48 h. Cells were washed with phosphate buffered saline (PBS)
and lysed with 200 mL/well lysis buffer (25 mM tris pH 7.8,
2 mM EDTA, 10% glycerol, 1% Triton X-100, 0.2 mM DTT).
Luciferase activity was assayed in triplicate for each transfection
using 20 mL lysate and 50 mL reconstituted luciferase assay
reagent (Promega). Luminescence was measured immediately for
4 s in each well with a Wallac Victor
3
1420 multilabel counter.
The experiment was repeated three times and the results
averaged. Data were analysed by one-way ANOVA with post
hoc Dunnett’s tests in GraphPad Prism version 5.03 (GraphPad
Software).
TERT SNPs and Ovarian and Breast Cancer Risk
PLoS ONE | www.plosone.org 5 September 2011 | Volume 6 | Issue 9 | e24987
Supporting Information
Table S1 Participating EOC case-control studies.
(DOC)
Table S2 Per allele OR for all SNPs in EOC.
(DOC)
Table S3 The association of rs2736109 with EOC, by study.
(DOC)
Table S4 Participating invasive breast case-control studies.
(DOC)
Table S5 The association of rs2736109/rs2736108 with risk of
invasive breast cancer, by study.
(DOC)
Table S6 Breast cancer risk by age.
(DOC)
Table S7 Breast cancer risk by ER status.
(DOC)
Acknowledgments
We are grateful to the family and friends of Kathryn Sladek Smith for their
generous support of Ovarian Cancer Association Consortium through their
donations to the Ovarian Cancer Research Fund. MARIE thanks Tracy
Slanger and Elke Mutschelknauss for their valuable contributions, and S.
Behrens, R. Birr, W. Busch, U. Eilber, B. Kaspereit, N. Knese, K. Smit, for
their excellent technical assistance. GESBC thank Ursula Eilber and Tanya
Koehler for competent technical assistance. HJO and HMO acknowledge
the support of Matthias Du¨rst, Peter Schu¨ rmann and Peter Hillemanns.
UKO thanks Ian Jacobs, Eva Wozniak, Andy Ryan, Jeremy Ford and
Nayala Balogun for their contribution to the study.
Author Contributions
Conceived and designed the experiments: GC-T RRR ELG DNR.
Performed the experiments: JB HAP XC MDS SA CB. Analyzed the data:
JB HAP SEJ JL KM SM YL. Contributed reagents/materials/analysis
tools: RTP DL ED SL IV JC-C SN AV DF-J MTG GL LRW RKM NB
NA IBR TD BLF RAV ELG EVB SHO IO LR-R LEK NDL LSC AB-W
IC SAG SJR AG-M UM PDP AMD SW-G UE KM AL AC SW DFE
RRR GC-T LAK KKHA TM LFAGM. Wrote the paper: JB HAP GC-T.
References
1. Baird DM (2010) Variation at the TERT locus and predisposition for cancer.
Expert Rev Mol Med 12: e16.
2. Rafnar T, Sulem P, Stacey SN, Geller F, Gudmundsson J, et al. (2009) Sequence
variants at the TERT-CLPTM1L locus associate with many cancer types. Nat
Genet 41: 221–227.
3. Shete S, Hosking FJ, Robertson LB, Dobbins SE, Sanson M, et al. (2009)
Genome-wide association study identifies five susceptibility loci for glioma. Nat
Genet 41: 899–904.
4. Landi MT, Chatterjee N, Yu K, Goldin LR, Golds tein AM, et al. (2009) A
genome-wide association study of lung cancer identifies a region of chromosome
5p15 associated with risk for adenocarcinoma. Am J Hum Genet 85: 679–691.
5. McKay JD, Hung RJ, Gaborieau V, Boffetta P, Chabrier A, et al. (2008) Lung
cancer susceptibility locus at 5p15.33. Nat Genet 40: 1404–1406.
6. Petersen GM, Amundado ttir L, Fuchs CS, Kraft P, Stolzenberg-Solomon RZ,
et al. (2010) A genome-wide association study identifies pancreatic cancer
susceptibility loci on chromosomes 13q22.1, 1q32.1 and 5p15.33. Nat Genet 42:
224–228.
7. Turnbull C, Rapley EA, Seal S, Pernet D, Renwick A, et al. (2010) Variants near
DMRT1, TERT and ATF7IP are associated with testicular germ cell cancer.
Nat Genet 42: 604–607.
8. Rothman N, Garcia-Closas M, Chatterjee N, Malats N, Wu X, et al. (2010) A
multi-stage genome-wide association study of bladder cancer identifies multiple
susceptibility loci. Nat Genet 42: 978–984.
9. Johnatty SE, Beesley J, Chen X, Macgregor S, Duffy DL, et al. (2010)
Evaluation of candidate stromal epithelial cross-talk genes identifies association
between risk of serous ovarian cancer and TERT, a cancer susceptibility ‘‘hot-
spot’’. PLoS Genet 6: e1001016.
10. Ramus SJ, Vierkant RA, Johnatty SE, Pike MC, Van Den Berg DJ, et al. (2008)
Consortium analysis of 7 candidate SNPs for ovarian cancer. Int J Cancer 123:
380–388.
11. Zhang B, Beeghly-Fadiel A, Long J, Zheng W (2011) Genetic variants associated
with breast-cancer risk: comprehensive research synopsis, meta-analysis, and
epidemiological evidence. Lancet Oncol 12: 477–488.
12. Antoniou AC, Wang X, Frederick sen ZS, McGuffog L, Tarrell R, et al. (2010) A
locus on 19p13 modifies risk of breast cancer in BRCA1 mutation carriers and is
associated with hormone receptor-negative breast cancer in the general
population. Nat Genet 42: 885–892.
13. Bolton KL, Tyrer J, Song H, Ramus SJ, Notaridou M, et al. (2010) Common
variants at 19p13 are associated with susceptibility to ovarian cancer. Nat Genet
42: 880–884.
14. Turnbull C, Ahmed S, Morrison J, Pernet D, Renwick A, et al. (2010) Genome-
wide association study identifies five new breast cancer susceptibility loci. Nat
Genet 42: 504–507.
15. Chen YJ, Campbell HG, Wiles AK, Eccles MR, Reddel RR, et al. (2008) PAX8
regulates telomerase reverse transcriptase and telomerase RNA component in
glioma. Cancer Res 68: 5724–5732.
16. Kyo S, Takakura M, Kanaya T, Zhuo W, Fujimoto K, et al. (1999) Estrogen
activates telomerase. Cancer Res 59: 5917–5921.
17. Yi X, Shay JW, Wright WE (2001) Quantitation of telomerase components and
hTERT mRNA splicing patterns in immortal human cells. Nucleic Acids Res
29: 4818–4825.
18. Liu JZ, McRae AF, Nyholt DR, Medland SE, Wray NR, et al. (2010) A versatile
gene-based test for genome-wide association studies. Am J Hum Genet 87:
139–145.
19. Choi J, Southworth LK, Sarin KY, Venteicher AS, Ma W, et al. (2008) TERT
promotes epithelial proliferation through transcriptional control of a Myc- and
Wnt-related developmental program. PLoS Genet 4: e10.
20. Dunning AM, Healey CS, Baynes C, Maia AT, Scollen S, et al. (2009)
Association of ESR1 gene tagging SNPs with breast cancer risk. Hum Mol
Genet 18: 1131–1139.
TERT SNPs and Ovarian and Breast Cancer Risk
PLoS ONE | www.plosone.org 6 September 2011 | Volume 6 | Issue 9 | e24987
... Consequently, two distinct haplotypes can be observed (see Table 1). Functional genomics work observed that the allele in common haplotype, i.e., C of rs2736107, rs2736108 and rs2736109 and A of rs2853669, can increase TERT promoter activity [15][16][17]. Further analysis verified that rs2736108 is significantly associated with telomere length [17][18][19][20][21]. ...
... The result indicated that the common allele of rs2736098, C, is with the ability to enhance TERT expression. Interestingly, the common allele at other four positions (see Table 1) can also increase TERT promoter activity [15][16][17], thus constituting a haplotype with high expression and leading to a significant signal at rs2736108 in GWAS [10][11][12][13][14]. ...
... Our and previous functional genomics work indicated that the allele in common haplotype, i.e., C of rs2736098, rs2736107, rs2736108 and rs2736109 and A of rs2853669, is the high expression one for TERT [15][16][17]. Therefore, the common haplotype should induce a high TERT expression. ...
Article
Full-text available
rs2736098 is a synonymous polymorphism in TERT (telomerase reverse transcriptase), an enzyme involved in tumor onset of multiple tissues, and should play no roles in carcinogenesis. However, a search in cancer somatic mutation database indicated that the mutation frequency at rs2736098 is much higher than the average one for TERT. Moreover, there are significant H3K4me1 and H3K27Ac signals, two universal histone modifications for active enhancers, surrounding rs2736098. Therefore, we hypothesized that rs2736098 might be within an enhancer region, regulate TERT expression and influence cancer risk. Through luciferase assay, it was verified that the enhancer activity of rs2736098C allele is significantly higher than that of T in multiple tissues. Transfection of plasmids containing TERT coding region with two different alleles indicated that rs2736098C allele can induce a significantly higher TERT expression than T. By chromatin immunoprecipitation, it was observed that the fragment spanning rs2736098 can interact with USF1 (upstream transcription factor 1). The two alleles of rs2736098 present evidently different binding affinity with nuclear proteins. Database and literature search indicated that rs2736098 is significantly associated with carcinogenesis in multiple tissues and count of multiple cell types. All these facts indicated that rs2736098 is also an oncogenic polymorphism and plays important role in cell proliferation.
... [6][7][8][9][10][11][12][13][14][15][16] The 5p15.33 region has been associated with more than ten types of cancer, with multiple independent risk alleles identified. [17][18][19][20][21][22][23][24] Various biological mechanisms, including inflammation, epigenetics, gene expression, and telomere structure, have been proposed to explain these identified pleiotropic associations. For example, the 5p15.33 region harbors the TERT gene, which encodes the catalytic subunit of telomerase, 25 as well as the CLPTM1L gene, which encodes the cleft lip and palate-associated transmembrane-1 like protein. ...
... 12,59,60 The 5p15.33 region, containing the TERT and CLPTM1L genes, has also been associated with multiple cancers. [17][18][19][20][21][22][23] Other significant genomic regions identified in our study, including 1q32.1 (ER-negative breast and prostate), 4q24 (colorectal and prostate), 5q11.2 (overall breast and colorectal), 10q26.13 (ER-positive breast and prostate), 17q12 (endometrial and prostate), and 19p13.11 ...
Article
Full-text available
Genome-wide association studies (GWAS) have identified thousands of cancer risk loci revealing many risk regions shared across multiple cancers. Characterizing the cross-cancer shared genetic basis can increase our understanding of global mechanisms of cancer development. In this study, we collected GWAS summary statistics based on up to 375,468 cancer cases and 530,521 controls for fourteen types of cancer, including breast (overall, estrogen receptor (ER)-positive and ER-negative), colorectal, endometrial, esophageal, glioma, head/neck, lung, melanoma, ovarian, pancreatic, prostate, and renal cancer, to characterize the shared genetic basis of cancer risk. We identified thirteen pairs of cancers with statistically significant local genetic correlations across eight distinct genomic regions. Specifically, the 5p15.33 region, harboring the TERT and CLPTM1L genes, showed statistically significant local genetic correlations for multiple cancer pairs. We conducted a cross-cancer fine-mapping of the 5p15.33 region based on eight cancers that showed genome-wide significant associations in this region (ER-negative breast, colorectal, glioma, lung, melanoma, ovarian, pancreatic and prostate cancer). We used an iterative analysis pipeline implementing a subset-based meta-analysis approach based on cancer-specific conditional analyses and identified ten independent cross-cancer associations within this region. For each signal, we conducted cross-cancer fine-mapping to prioritize the most plausible causal variants. Our findings provide a more in-depth understanding of the shared inherited basis across human cancers and expand our knowledge of the 5p15.33 region in carcinogenesis.
... Some 50 kbp apart lies TERT, the gene encoding human telomerase reverse transcriptase. The CLPTM1L-TERT locus is relevant for several gynaecological cancers [121,[145][146][147][148], however, the functional significance of the identified variant and its contribution to cervical cancer pathogenesis via the CLPTM1L-TERT locus remain to be elucidated. ...
Article
Full-text available
Cervical cancer is the fourth common cancer amongst women worldwide. Infection by high-risk human papilloma virus is necessary in most cases, but not sufficient to develop invasive cervical cancer. Despite a predicted genetic heritability in the range of other gynaecological cancers, only few genomic susceptibility loci have been identified thus far. Various case-control association studies have found corroborative evidence for several independent risk variants at the 6p21.3 locus (HLA), while many reports of associations with variants outside the HLA region remain to be validated in other cohorts. Here, we review cervical cancer susceptibility variants arising from recent genome-wide association studies and meta-analysis in large cohorts and propose 2q14 (PAX8), 17q12 (GSDMB), and 5p15.33 (CLPTM1L) as consistently replicated non-HLA cervical cancer susceptibility loci. We further discuss the available evidence for these loci, knowledge gaps, future perspectives, and the potential impact of these findings on precision medicine strategies to combat cervical cancer.
... It is noteworthy here that germline variation near or within the h-TERT gene is equated with telomere length in peripheral blood leukocytes und risk of h-TERT promoter mutation mutants [21,22]. Moreover, the h-TERT promoter polymorphism viz rs 2,853,669 modulates the prognostic value in various tumor types as well as showed reduction in overall survival and increased tumor recurrence rate in Bladder cancer [23][24][25]. Thus, the prognostic power of h-TERT promoter mutation highlights their potential use as a clinical biomarker. ...
Article
Full-text available
Buccal mucosa cancer has an aggressive nature as it rapidly grows and penetrates with high recurrence rate. Strikingly, carcinoma of buccal mucosa is the most common cancer of oral cavity in India. Recently, telomerase and telomere biology have been implicated in the pathogenesis and progression in various cancers via regulation of telomere maintenance by telomerase expression which is controlled by telomerase reverse transcriptase (TERT) promoter. Strikingly, h-TERT promoter mutations have been incriminated in regulation of telomerase gene expression. Here, we present a 35 years old male with intense coughing, short breathlessness and fever since 15 days, was admitted to the pulmonary unit. He was a chronic smoker and gutka user. The cytopathological analysis of gastric aspirate revealed buccal mucosa carcinoma of IV stage. We identified h-TERT promoter mutations in isolated genomic DNA from whole blood using DNA sequencer. Genetic analysis disclosed that h-TERT promoter region was highly mutated in this patient. Identified mutations include C.-248 del G, C.-272 del G, C.-279 del G, C.-331 del G, C.-349 del G, C.-351 del C, C.-360 G > A, C.-362 T > A, C.-371 del T and C.-372 del T. Further, all identified mutations were subjected to predict the pathologic functional consequences using bioinformatics tools viz TFsitescan and CiiiDER which showed either loss or gain of transcription factors binding sites in h-TERT promoter. This is a unique case in which total 9 mutations were observed in h-TERT promoter in a single case. In conclusion, all together these mutations in h-TERT promoter may alter the epigenetics and subsequently the tenacity of binding transcription factors which are of functional significance.
Article
Programmed cell death 6 (PDCD6) is known to be involved in apoptosis and tumorigenesis. Given the reported association with urinary cancer susceptibility through SNP analysis, we further analyzed the entire genomic structure of PDCD6. Three VNTR regions (MS1-MS3) were identified through the analysis of the genomic structure of PDCD6. To investigate the association between these VNTR regions and urinary cancer susceptibility, genomic DNA was extracted from 413 cancer-free male controls, 267 bladder cancer patients, and 331 prostate cancer patients. Polymerase chain reaction (PCR) was performed to analyze the PDCD6-MS regions. Statistical analysis was performed to determine the association between specific genotypes and cancer risk. In addition, the effect of specific VNTRs on PDCD6 expression was also confirmed using a reporter vector. Among the three VNTR regions, MS1 and MS2 exhibited monomorphism, while the MS3 region represented polymorphism, with its transmission to subsequent generations through meiosis substantiating its utility as a DNA typing marker. In a case–control study, the presence of rare alleles within PDCD6-MS3 exhibited significant associations with both bladder cancer (OR = 2.37, 95% CI: 1.33–4.95, P = 0.019) and prostate cancer (OR = 2.11, 95% CI: 1.03–4.36, P = 0.038). Furthermore, through luciferase assays, we validated the impact of the MS3 region on modulating PDCD6 expression. This study suggests that the PDCD6-MS3 region could serve as a prognostic marker for urinary cancers, specifically bladder cancer and prostate cancer. Moreover, the subdued influence exerted by PDCD6-MS3 on the expression of PDCD6 offers another insight concerning the progression of urinary cancer.
Article
Full-text available
Telomerase reverse transcriptase (TERT/hTERT) serves as the pivotal catalytic subunit of telomerase, a crucial enzyme responsible for telomere maintenance and human genome stability. The high activation of hTERT, observed in over 90% of tumors, plays a significant role in tumor initiation and progression. An in-depth exploration of hTERT activation mechanisms in cancer holds promise for advancing our understanding of the disease and developing more effective treatment strategies. In breast cancer, the expression of hTERT is regulated by epigenetic, transcriptional, post-translational modification mechanisms and DNA variation. Besides its canonical function in telomere maintenance, hTERT exerts non-canonical roles that contribute to disease progression through telomerase-independent mechanisms. This comprehensive review summarizes the regulatory mechanisms governing hTERT in breast cancer and elucidates the functional implications of its activation. Given the overexpression of hTERT in most breast cancer cells, the detection of hTERT and its associated molecules are potential for enhancing early screening and prognostic evaluation of breast cancer. Although still in its early stages, therapeutic approaches targeting hTERT and its regulatory molecules show promise as viable strategies for breast cancer treatment. These methods are also discussed in this paper.
Article
Lung cancer is a severe and the leading cause of cancer related deaths worldwide. The recurrent h-TERT promoter mutations have been implicated in various cancer types. Thus, the present study is extended to analyze h-TERT promoter mutations from the North Indian lung carcinoma patients. Total 20 histopathologically and clinically confirmed cases of lung cancer were enrolled in this study. The genomic DNA was extracted from venous blood and subjected to amplification using appropriate h-TERT promoter primers. Amplified PCR products were subjected for DNA Sanger sequencing for the identification of novel h-TERT mutations. Further, these identified h-TERT promoter mutations were analysed for the prediction of pathophysiological consequences using bioinformatics tools such as Tfsitescan and CIIDER. The average age of patients was 45 ± 8 years which was categorized in early onset of lung cancer with predominance of male patients by 5.6 fold. Interestingly, h-TERT promoter mutations were observed highly frequent in lung cancer. Identified mutations include c. G272A, c. T122A, c. C150A, c. 123 del C, c. C123T, c. G105A, c. 107 Ins A, c. 276 del C corresponding to −168 G>A, −18 T>A, −46 C>A, −19 del C, −19 C>T, −1 G>A, −3 Ins A, −172 del C respectively from the translation start site in the promoter of the telomerase reverse transcriptase gene which are the first time reported in germline genome from lung cancer. Strikingly, c. −18 T>A [C.T122A] was found the most prevalent variant with 75% frequency. Notwithstanding, other mutations viz c. -G168A [c. G272A] and c. −1 G>A [c. G105A] were found to be at 35% and 15% frequency respectively whilst the rest of the mutations were present at 10% and 5% frequency. Additionally, bioinformatics analysis revealed that these mutations can lead to either loss or gain of various transcription factor binding sites in the h-TERT promoter region. Henceforth, these mutations may play a pivotal role in h-TERT gene expression. Taken together, these identified novel promoter mutations may alter the epigenetics and subsequently various transcription factor binding sites which are of great functional significance. Thereby, it is plausible that these germline mutations may involve either as predisposing factor or direct participation in the pathophysiology of lung cancer through entangled molecular mechanisms.
Article
Full-text available
The role of telomerase reverse transcriptase (TERT) induction and telomere maintenance in carcinogenesis including cervical cancer (CC) pathogenesis has been well established. However, it remains unclear whether they affect infection of high-risk human papillomavirus (hrHPV), an initiating event for CC development. Similarly, genetic variants at the TERT locus are shown to be associated with susceptibility to CC, but it is unclear whether these SNPs modify the risk for cervical HPV infection. Here we show that in CC-derived HeLa cells, TERT overexpression inhibits, while its depletion upregulates expression of Syndecan-1 (SDC-1), a key component for HPV entry receptors. The TCGA cohort of CC analyses reveals an inverse correlation between TERT and SDC-1 expression (R = −0.23, P = 0.001). We further recruited 1330 females (520 non-HPV and 810 hrHPV-infected) without CC or high-grade cervical intraepithelial neoplasia to analyze telomeres in cervical epithelial cells and SNPs at rs2736098, rs2736100 and rs2736108, previously identified TERT SNPs for CC risk. Non-infected females exhibited age-related telomere shortening in cervical epithelial cells and their telomeres were significantly longer than those in hrHPV-infected group (1.31 ± 0.62 vs 1.19 ± 0.48, P < 0.001). There were no differences in rs2736098 and rs2736100 genotypes, but non-infected individuals had significantly a higher C-allele frequency (associated with higher TERT expression) while lower T-allele levels at rs2736108 compared with those in the hrHPV group (P = 0.020). Collectively, appropriate telomere maintenance and TERT expression in normal cervical cells may prevent CC by modulating hrHPV infection predisposition, although they are required for CC development and progression.
Article
Full-text available
Telomerase serves a critical role in stem cell function and tissue homeostasis. This role depends on its ability to synthesize telomere repeats in a manner dependent on the reverse transcriptase (RT) function of its protein component telomerase RT (TERT), as well as on a novel pathway whose mechanism is poorly understood. Here, we use a TERT mutant lacking RT function (TERTci) to study the mechanism of TERT action in mammalian skin, an ideal tissue for studying progenitor cell biology. We show that TERTci retains the full activities of wild-type TERT in enhancing keratinocyte proliferation in skin and in activating resting hair follicle stem cells, which triggers initiation of a new hair follicle growth phase and promotes hair synthesis. To understand the nature of this RT-independent function for TERT, we studied the genome-wide transcriptional response to acute changes in TERT levels in mouse skin. We find that TERT facilitates activation of progenitor cells in the skin and hair follicle by triggering a rapid change in gene expression that significantly overlaps the program controlling natural hair follicle cycling in wild-type mice. Statistical comparisons to other microarray gene sets using pattern-matching algorithms revealed that the TERT transcriptional response strongly resembles those mediated by Myc and Wnt, two proteins intimately associated with stem cell function and cancer. These data show that TERT controls tissue progenitor cells via transcriptional regulation of a developmental program converging on the Myc and Wnt pathways.
Article
Full-text available
Telomerase serves a critical role in stem cell function and tissue homeostasis. This role depends on its ability to synthesize telomere repeats in a manner dependent on the reverse transcriptase (RT) function of its protein component telomerase RT (TERT), as well as on a novel pathway whose mechanism is poorly understood. Here, we use a TERT mutant lacking RT function (TERTci) to study the mechanism of TERT action in mammalian skin, an ideal tissue for studying progenitor cell biology. We show that TERTci retains the full activities of wild-type TERT in enhancing keratinocyte proliferation in skin and in activating resting hair follicle stem cells, which triggers initiation of a new hair follicle growth phase and promotes hair synthesis. To understand the nature of this RT-independent function for TERT, we studied the genome-wide transcriptional response to acute changes in TERT levels in mouse skin. We find that TERT facilitates activation of progenitor cells in the skin and hair follicle by triggering a rapid change in gene expression that significantly overlaps the program controlling natural hair follicle cycling in wild-type mice. Statistical comparisons to other microarray gene sets using pattern-matching algorithms revealed that the TERT transcriptional response strongly resembles those mediated by Myc and Wnt, two proteins intimately associated with stem cell function and cancer. These data show that TERT controls tissue progenitor cells via transcriptional regulation of a developmental program converging on the Myc and Wnt pathways.
Article
Full-text available
We conducted a genome-wide association study for testicular germ cell tumor, genotyping 298,782 SNPs in 979 affected individuals and 4,947 controls from the UK and replicating associations in a further 664 cases and 3,456 controls. We identified three new susceptibility loci, two of which include genes that are involved in telomere regulation. We identified two independent signals within the TERT-CLPTM1L locus on chromosome 5, which has previously been associated with multiple other cancers (rs4635969, OR=1.54, P=1.14x10(-23); rs2736100, OR=1.33, P=7.55x10(-15)). We also identified a locus on chromosome 12 (rs2900333, OR=1.27, P=6.16x10(-10)) that contains ATF7IP, a regulator of TERT expression. Finally, we identified a locus on chromosome 9 (rs755383, OR=1.37, P=1.12x10(-23)), containing the sex determination gene DMRT1, which has been linked to teratoma susceptibility in mice
Article
Full-text available
Paired box (PAX) developmental genes are frequently expressed in cancers and confer survival advantages on cancer cells. We have previously found that PAX genes are deregulated in glioma. We have now investigated the expression of PAX genes in glioma and their role in telomere maintenance. The mRNA level of PAX8 showed a positive correlation with telomerase activity in glioma biopsies (r(2) = 0.75, P < 0.001) and in established glioma cell lines (r(2) = 0.97, P = 0.0025). We found that PAX8 is able to coordinately transactivate the promoter for both the telomerase catalytic subunit (hTERT) and the telomerase RNA component (hTR) genes. By electrophoretic mobility shift assay, quantitative PCR, and a telomerase activity assay, we show that PAX8 binds directly to the hTERT and hTR promoters, up-regulating hTERT and hTR mRNA, as well as telomerase activity. Additionally, PAX8 small interfering RNA down-regulated hTERT and hTR. Collectively, these results show that PAX8 may have a role in telomerase regulation.
Article
Full-text available
We conducted a multi-stage, genome-wide association study of bladder cancer with a primary scan of 591,637 SNPs in 3,532 affected individuals (cases) and 5,120 controls of European descent from five studies followed by a replication strategy, which included 8,382 cases and 48,275 controls from 16 studies. In a combined analysis, we identified three new regions associated with bladder cancer on chromosomes 22q13.1, 19q12 and 2q37.1: rs1014971, (P = 8 × 10⁻¹²) maps to a non-genic region of chromosome 22q13.1, rs8102137 (P = 2 × 10⁻¹¹) on 19q12 maps to CCNE1 and rs11892031 (P = 1 × 10⁻⁷) maps to the UGT1A cluster on 2q37.1. We confirmed four previously identified genome-wide associations on chromosomes 3q28, 4p16.3, 8q24.21 and 8q24.3, validated previous candidate associations for the GSTM1 deletion (P = 4 × 10⁻¹¹) and a tag SNP for NAT2 acetylation status (P = 4 × 10⁻¹¹), and found interactions with smoking in both regions. Our findings on common variants associated with bladder cancer risk should provide new insights into the mechanisms of carcinogenesis.
Article
Telomerase is a reverse transcriptase that adds telomeric repeats to chromosomal ends. In most normal human somatic cells, telomerase is repressed and telomeres progressively shorten, leading to limited proliferative life-span. Telomerase reactivation is associated with cellular immortalization and is a frequent event during tumorigenesis. The telomerase ribonucleoprotein complex consists of two essential components, a catalytic protein subunit [human telomerase reverse transcriptase (hTERT)] and a template RNA (hTR). hTR is constitutively expressed, while hTERT is almost universally absent in telomerase-negative cells. Although repression of telomerase is transcriptional in telomerase-negative cells, post-transcriptional and assembly processes are likely to play important roles in regulating telomerase activity in those that are telomerase-positive. The telomerase transcript can also be alternatively spliced into a variety of non-functional forms. To establish the quantitative relationships between telomerase activity and its various components, we determined the numbers of molecules of hTR and hTERT mRNA, and the levels of alternatively spliced hTERT mRNA variants in normal, in vitro immortalized and cancer cell lines. We report here that there is surprisingly little variation in the proportion of alternatively spliced forms of hTERT in different cell lines. The only variation observed occurred when a change in splicing to non-functional forms appeared in response to conditions that repress telomerase activity in IDH4 cells. We also found that most telomerase-positive cell lines only contain a few molecules of potentially functional hTERT mRNA, and there is a correlation between telomerase activity and the levels of both hTR and hTERT +α+β mRNA.
Article
More than 1000 reports have been published in the past two decades on associations between variants in candidate genes and risk of breast cancer. Results have been generally inconsistent. We did a literature search and meta-analyses to provide a synopsis of the current understanding of the genetic architecture of breast-cancer risk. A systematic literature search for candidate-gene association studies of breast-cancer risk was done in two stages, using PubMed on or before Feb 28, 2010. A total of 24,500 publications were identified, of which 1059 were deemed eligible for inclusion. Meta-analyses were done for 279 genetic variants in 128 candidate genes or chromosomal loci that had at least three data sources. Variants with significant associations by meta-analysis were assessed using the Venice criteria and scored as having strong, moderate, or weak cumulative evidence for an association with breast-cancer risk. 51 variants in 40 genes showed significant associations with breast-cancer risk. Cumulative epidemiological evidence of an association was graded as strong for ten variants in six genes (ATM, CASP8, CHEK2, CTLA4, NBN, and TP53), moderate for four variants in four genes (ATM, CYP19A1, TERT, and XRCC3), and weak for 37 variants. Additionally, in meta-analyses that included a minimum of 10,000 cases and 10,000 controls, convincing evidence of no association with breast-cancer risk was identified for 45 variants in 37 genes. Whereas most genetic variants assessed in previous candidate-gene studies showed no association with breast-cancer risk in meta-analyses, 14 variants in nine genes had moderate to strong evidence for an association. Further evaluation of these variants is warranted. US National Cancer Institute.