• Non ci sono risultati.

Genome-wide association study for colorectal cancer identifies risk polymorphisms in German familial cases and implicates MAPK signalling pathways in disease susceptibility

N/A
N/A
Protected

Academic year: 2021

Condividi "Genome-wide association study for colorectal cancer identifies risk polymorphisms in German familial cases and implicates MAPK signalling pathways in disease susceptibility"

Copied!
8
0
0

Testo completo

(1)

Advance Access publication July 7, 2010

Genome-wide association study for colorectal cancer identifies risk polymorphisms in

German familial cases and implicates MAPK signalling pathways in disease

susceptibility

Jesu

´ s Lascorz

1,

, Asta Fo¨rsti

1,2

, Bowang Chen

1

,

Stephan Buch

3,4

, Verena Steinke

5

, Nils Rahner

5

,

Elke Holinski-Feder

6

, Monika Morak

6

,

Hans K.Schackert

7

, Heike Go¨rgens

7

, Karsten Schulmann

8

,

Timm Goecke

9

, Matthias Kloor

10

, Cristoph Engel

11

,

Reinhard Bu¨ttner

12

, Nelli Kunkel

1

, Marianne Weires

1

,

Michael Hoffmeister

13

, Barbara Pardini

14

,

Alessio Naccarati

14

, Ludmila Vodickova

15

, Jan Novotny

16

,

Stefan Schreiber

4,17

, Michael Krawczak

4,18

,

Clemens D.Bro¨ring

19

, Henry Vo¨lzke

20

,

Clemens Schafmayer

4,19

, Pavel Vodicka

14

, Jenny

Chang-Claude

21

, Hermann Brenner

13

, Barbara Burwinkel

22,23

,

Peter Propping

5

, Jochen Hampe

3

and Kari Hemminki

1,2 1

Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany,2Center for Primary Health Care Research, Clinical Research Center, Lund University, Malmo¨ 20502, Sweden, 3Department of General Internal Medicine, Christian-Albrechts-University, Kiel 24105, Germany,4PopGen Biobank project, Christian-Albrechts-University, Kiel 24105, Germany,5Institute of Human Genetics, University of Bonn, Bonn 53127, Germany,6Department of Internal Medicine, Campus Innenstadt, University Hospital of Ludwig-Maximilians-University, Munich 81377, Germany,7Department of Surgical Research, Technische Universita¨t Dresden, Dresden 01069, Germany,8Medical Department,

Knappschaftskrankenhaus, Ruhr University, Bochum 44892, Germany, 9

Institute of Human Genetics, University of Du¨sseldorf, Du¨sseldorf 40225, Germany,10Department of Applied Tumour Biology, Institute of Pathology, University of Heidelberg, Heidelberg 69120, Germany,11Institute of Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig 04107 Germany,12Institute of Pathology, University of Bonn, Bonn 53127, Germany,13Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg 69115, Germany,14Department of Molecular Biology of Cancer, Institute of Experimental Medicine, Academy of Science of Czech Republic, Prague 14220, Czech Republic, 15

Center of Occupational Health, National Institute of Public Health, Prague 10042, Czech Republic,16Department of Oncology, General Teaching Hospital, Prague 12808, Czech Republic,17Institute of Clinical Molecular Biology, Christian-Albrechts-University, Kiel 24105, Germany,18Institute of Medical Informatics and Statistics, Christian-Albrechts-University, Kiel 24105, Germany,19Department of General and Thoracic Surgery, Christian-Albrechts-University, Kiel 24105, Germany,20Institute for Community Medicine, University Hospital Greifswald, Greifswald 17489, Germany, 21

Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany,22Helmholtz Group Molecular Epidemiology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany and23Molecular Oncology, Department of Gynecology and Obstetrics, University of Heidelberg, Heidelberg 69115, Germany To whom correspondence should be addressed. Tel:þ49 6221 421803; Fax:þ49 6221 421810;

Email: j.lascorz@dkfz.de

Genetic susceptibility accounts for

35% of all colorectal

can-cer (CRC). Ten common low-risk variants contributing to CRC

risk have been identified through genome-wide association

studies (GWASs). In our GWAS, 610 664 genotyped

single-nucleotide polymorphisms (SNPs) passed the quality control

filtering in 371 German familial CRC patients and 1263

con-trols, and replication studies were conducted in four additional

case–control sets (4915 cases and 5607 controls). Known risk

loci at 8q24.21 and 11q23 were confirmed, and a previously

un-reported association, rs12701937, located between the genes

GLI3 (GLI family zinc finger 3) and INHBA (inhibin, beta A)

[P 5 1.1 3 10

23

, odds ratio (OR) 1.14, 95% confidence interval

(CI) 1.05–1.23, dominant model in the combined cohort], was

identified. The association was stronger in familial cases

com-pared with unselected cases (P 5 2.0 3 10

24

, OR 1.36, 95% CI

1.16–1.60, dominant model). Two other unreported SNPs,

rs6038071, 40 kb upstream of CSNK2A1 (casein kinase 2, alpha

1 polypeptide) and an intronic marker in MYO3A (myosin IIIA),

rs11014993, associated with CRC only in the familial CRC cases

(P 5 2.5 3 10

23

, recessive model, and P 5 2.7 3 10

24

,

domi-nant model). Three software tools successfully pointed to the

overrepresentation of genes related to the mitogen-activated

protein

kinase

(MAPK)

signalling

pathways

among

the

1340 most strongly associated markers from the GWAS

(allelic P value

< 10

23

). The risk of CRC increased significantly

with an increasing number of risk alleles in seven genes

involved in MAPK signalling events (P

trend

5 2.2 3 10

216

,

OR

per allele

5 1.34, 95% CI 1.11–1.61).

Introduction

Colorectal cancer (CRC) is the third most common cancer and the

fourth-leading cause of cancer death worldwide, with a lifetime

risk in Western European and North American populations

5%.

The majority of CRC cases (up to 80%) are sporadic (1), indicating

that both genetic and environmental factors contribute to the

disease aetiology, with inherited susceptibility being responsible

for

35% of all CRC (2). However, hereditary high-risk germ

line mutations in the APC gene, the DNA mismatch repair genes,

MUTYH, and more rarely in the SMAD4, BMPR1A and STK11/

LKB1 genes account for 3.6% of all cases, according to a Finish

study (3).

To date, three genome-wide association studies (GWASs) (4–6),

followed by a series of fast-tracked publications (7–9), have been

carried out for CRC, leading to the identification of six susceptibility

loci (8q23.3, 8q24.21, 10p14, 11q23, 15q13.3 and 18q21). A

subse-quent meta-analysis (10) of two of the three GWASs yielded four

novel risk loci (14q22.2, 16q22.1, 19p13.1 and 20p12.3), including

confirmation of these associations in additional large sample sets.

Several follow-up studies have replicated these associations through

genotyping of thousands of individuals (11–19). Despite this

enor-mous effort, these 10 common low-penetrance variants exert only

subtle effects on cancer risk, with odds ratios (ORs) ranging

1.1

to 1.2 (20), whereas the joint population attributable fraction for the

first six risk loci described (without including the meta-analysis) was

52% (21). Thus, most of the estimated inherited susceptibility for

CRC remains unknown.

We performed a GWAS on 384 German familial cases and 1285

ethnically matched healthy controls on the Affymetrix Genome

Wide Human SNP Array 6.0 [906 600 single-nucleotide

polymor-phisms (SNPs)]. Familial cases were selected because they might

be genetically enriched for susceptibility alleles, thus providing

enhanced power to detect an association (22,23). Selected

associ-ated SNPs were included in up to four replication studies. There is

an increasing focus on the idea of a complex interplay of molecules

in a common pathway contributing to disease aetiology (24).

Abbreviations:CI, confidence interval; CRC, colorectal cancer; EGFR, epi-dermal growth factor receptor; GO, Gene Ontology; GWAS, genome-wide association study; LD, linkage disequilibrium; MAPK, mitogen-activated pro-tein kinase; MDR, multifactor-dimensionality reduction; OR, odds ratio; SNP, single-nucleotide polymorphism.

at Ustavy AV - Krc on September 1, 2010

http://carcin.oxfordjournals.org

(2)

Accordingly, we performed overrepresentation analyses among

the most strongly associated SNPs from our GWAS (allelic

P value , 10

3

) using three software tools, in order to search

for enrichment of pathways or Gene Ontology (GO) categories.

Materials and methods

Study populations

A total of 384 unrelated familial CRC cases were included in the GWAS, 315 of them were identified from the regional cancer registry of Schleswig-Holstein (Germany) (25) and another 69 were included from the German HNPCC Consortium (Table I). Samples from the HNPCC Consortium were tested to be microsatellite stable (none of the five markers tested showed in-stability) and thus negative for germ line mutations in MSH2 and MLH1 (26). All patients had at least one first-degree relative with CRC (71% one relative, 16% two relatives, 7% three or more relatives and 6% with unknown number). The 1285 healthy controls, free of CRC at the time of recruitment, were randomly selected from the population-based control pool of the PopGen pro-ject in Schleswig-Holstein (Germany) (27).

Replication study 1 included 575 additional, independent CRC cases from the German HNPCC Consortium (26), also tested to be microsatellite stable. The 760 healthy controls were healthy voluntary blood donors recruited in Mannheim (Germany) (28). Replication 2 comprised 2173 or 1908 additional, independent cases identified from the cancer registry of Schleswig-Holstein, as well as 2552 or 1752 controls (Table II): 322/293 additional controls from the PopGen project and 2230/1459 randomly selected population-derived controls from the SHIP (Study of Health In Pomerania) study, free of CRC at the time of recruitment, and also from Northern Germany (29). Replication 3 included 1373 CRC cases and 1480 controls from the population-based DACHS (DArmkrebs: CHancen der Verhuetung durch Screening—CRC: chances of prevention through screening) study in the Rhine-Neckar region in the south-west of Germany (30). DACHS cases were recruited during first hospitalization due to cancer treatment or shortly afterwards at their homes, whereas controls were randomly selected from lists of residents supplied by population regis-tries. Finally, replication 4 consisted of 794 CRC cases and 815 hospital-based colonoscopy negative controls from the Czech Republic (31). Controls were undergoing colonoscopy for various gastrointestinal complaints and in the frame of preventive examination. Only subjects whose colonoscopy results were negative for malignancy, colorectal adenomas, benign polyps or idio-pathic bowel disease were chosen as controls, and this guarantees a very low risk of CRC for the next20 years (30). For a further description of all patient and control groups see Table I.

The study protocols were approved by the ethics committee of all partici-pating clinical centres. Written informed consent was obtained from all study participants.

GWAS

DNA was extracted from peripheral venous blood. All samples initially in-cluded in the GWAS (384 cases, 1285 controls) were genotyped for 906 600 SNPs using the Affymetrix 6.0 array by Affymetrix (Santa Clara, CA) accord-ing to the Affymetrix’s protocol, together with 14 Affymetrix controls.

Thirteen CRC cases and 22 controls failed genotyping due to standard quality control of genotype data by Affymetrix (call rate , 86% or preliminary Birdseed call rate , 95%), leaving a total of 371 cases and 1263 controls that were included in the association study.

Primary data analysis was performed with the Affymetrix Genotyping Con-sole software, and the genotype calling was done using the Birdseed 2 algorithm in Affymetrix Power Tool (http://www.affymetrix.com/partners_ programs/programs/developer/tools/powertools.affx). Before analysis, we ex-cluded markers with one or more of the following criteria: ,90% genotype call rate (61 152 SNPs), minor allele frequency , 5% in cases or controls (224 200 SNPs) or Hardy–Weinberg equilibrium exact P value , 105 in cases or controls (78 289 SNPs). Additional 352 SNPs were rejected after visual quality inspection of the clustering pattern (32) using Affymetrix Genotyping Console (v3.0.2). Finally, 610 664 SNPs passed all quality control filters, with an average call rate of 97.5%.

To evaluate the evidence for association between each genotyped SNP and CRC, we first calculated the association between SNPs and CRC for allelic, genotype, dominant and recessive models based on logistic regression, using the whole-genome analysis package PLINK with the ‘model’ option (33), as we do not know whether variants are probably to add additively, dominantly or recessively. A total of 523 SNPs showed at least one of the four calculated P values for association (allelic, genotype, dominant or recessive) , 104and reliable clustering. The Manhattan plot, which plots log10 of the allelic P value of each SNP along chromosomes and physical positions, was created by the genetic analysis package in CRAN R (http://cran.r-project.org/web /packages/gap/index.html). The impact of population stratification was evalu-ated by calculating the genomic control inflation factor lambda (k) using the ‘qqunif()’ function in CRAN R. The genomic control k value of 1.08 indicated a minimal overall inflation due to population stratification. This value of k is identical with the recent results comparing Northern and Southern German populations (34). Given that the impact factor was found to be minimal, all the statistical results are reported without genomic control correction.

Since Bonferroni correction for multiple testing is agreed to be overly con-servative in GWASs (35), we followed the Wellcome Trust Case Control Con-sortium thresholds of P , 1 105and P , 5 107as moderate and strong evidence for association, respectively (36).

Forward SNP selection procedure

Among the 523 SNPs with one of the four calculated P values for association ,104and reliable clustering, selection for replication studies was based not only on the most extreme P values, but also on the following criteria, in an attempt to select the best candidate markers for CRC (37):

Table I. Details of individual case–controls series used in the genome-wide and the replication studies

Origin Sample size Average age at

recruitment (range) Male:female ratio Average age of CRC diagnosis (range) GWAS

Cases Germany (Kielþ HNPCC

Consortium)

384 68.2 (49–86)a 0.99 64.4 (47–83)a

No data (HNPCC) 46.5 (15–76)b

Controls Germany (PopGen) 1285 56.0 (19–77) 1.25 —

Replication 1

Cases Germany (HNPCC

Consortium)

575 No data 0.98 43.4 (9–82)

Controls Germany (Mannheim) 760 45.9 (26–68) 1 —

Replication 2

Cases Germany (Kiel) 2173 66.9 (17–94) 1.17 63.0 (10–90)

Controls Germany (PopGen) 322 73.6 (50–81) 0.86 —

Germany (SHIP) 2230 60.9 (21–81) 0.92 —

Replication 3

Cases Germany (DACHS) 1373 68.6 (33–95) 1.35 68.1 (33–94)

Controls Germany (DACHS) 1480 68.0 (34–98) 0.94 —

Replication 4

Cases Czech Republic 794 62.3 (27–90) 1.33 61.1 (27–90)

Controls Czech Republic 815 54.4 (28–91) 1.38 —

SHIP, Study of Health In Pomerania; DACHS, Darmkrebs: CHancen der Verhuetung durch Screening (CRC: chances of prevention through screening). a

Only for the 170 cases from Kiel with full data. b

HNPCC Consortium samples.

at Ustavy AV - Krc on September 1, 2010

http://carcin.oxfordjournals.org

(3)

(i) Two or more SNPs located within or close to a gene or intergenic but not further than 150 kb from each other (so-called ‘SNP-clusters’) (37 SNPs); (ii) SNPs located within known risk loci for CRC, according to previous GWASs [8q24.21 (5,6), 18q21 (7), 8q23.3 (9), 10p14 (9), 15q13.3 (8), 11q23 (4), the not confirmed 9p24 (6) and the four loci from a meta-analysis 14q22.2, 16q22.1, 19p13.1 and 20p12.3 (10)] (2 SNPs); (iii) SNPs located in 451 candidate genes for CRC, based on high-throughput mutation analy-ses in CRC tumours (38,39), their possible relevance for CRC biology (6), or a transposon-based genetic screen in mice (40) (15 SNPs); (iv) coding SNPs (5 SNPs) and/or (v) comparison with the results of an unpublished GWAS for CRC (Jochen Hampe, personal communication) (18 SNPs). This resulted in selection of 53 SNPs for replication, 51 of them were successfully genotyped in replication 1 (see supplementary Table 1, available at Carcinogenesis Online). The 470 SNPs not included in replication studies are listed in supplementary Table 2, available at Carcinogenesis Online.

Replication studies

All DNAs used in the replication studies were extracted from peripheral ve-nous blood and subjected to genome-wide amplification using the illustra GenomiPhi V2 DNA Amplification Kit (GE Healthcare, Freiburg, Germany) according to manufacturer’s instructions. Genotyping was performed at the German Cancer Research Center in Heidelberg and at the University of Kiel (Germany), using 5 ng of genomic amplified DNA in 384-well plate format, using KASPar assays on demand (50 SNPs) (KBiosciences, Hoddesdon, UK) or TaqMan (one SNP) (Applied Biosystems, Darmstadt, Germany) technolo-gies and following the manufacturers’ protocols. Average genotyping call rate was 98%. Calculation of linkage disequilibrium (LD) and reconstruction of

haplotypes between rs12701937 and rs10441105 at 7p14.1 were performed using Haploview (41). All joint analyses of replication studies were adjusted for study centre.

Overrepresentation analysis and gene–gene interactions

Three different tools were used to search for enrichment of pathways or Gene Ontology (GO) categories among the associated polymorphisms. The recently published approach ALIGATOR (42) considers the biological pathways as a unit of analysis, and tests for overrepresentation of categories of genes de-fined by GO terms on a list of significant SNPs, which are selected according to an arbitrary threshold of significance in the GWAS. The 610 664 SNPs ana-lysed and their corresponding allelic P values for association were used as input, with default settings for calculations (5000 replicate gene lists and 1000 replicate studies). Three different thresholds for allelic P values of SNPs in the GWAS were tested in order to define a significantly associated SNP set for the enrichment analysis; 104(153 SNPs), 103(1340 SNPs) and 5 103 (5130 SNPs). The highest level of significance for overrepresentation of GO categories was obtained using, as input list, the 1340 SNPs with allelic P value , 103, located within 350 different genes (Table IV). For all three cut-offs, ALIGATOR analysis was restricted to those SNPs from the input list located within genes, and only GO categories containing two or more genes were counted.

ConsensusPathDB (43) integrates more than 12 different interaction data-bases and 1738 pathways and carries out overrepresentation analysis within an uploaded gene list (pathway-based set enrichment tool). Input list of genes consisted of the 350 genes with SNPs associated in the GWAS with allelic P value , 103, also used by ALIGATOR for overrepresentation analysis. Table II. Association for the six SNPs genotyped in replication 1 (575 familial cases and 760 controls) and results for the three SNPs selected for further replications

Cases Controls MAF

cases

MAF controls

ORhet.(95% CI) ORhom. (95% CI)

Best model P value OR (95% CI)

rs12701937 (7p14.1) [T] GWAS 371 1263 0.47 0.39 1.09 (0.83–1.42) 2.22 (1.59–3.11) Replication 1 575 760 0.48 0.40 1.58 (1.23–2.03) 1.76 (1.28–2.41) Replication 2a 2173 2552 0.46 0.44 1.12 (0.98–1.28) 1.06 (0.90–1.26) Replication 3 1373 1480 0.42 0.42 1.00 (0.85–1.19) 1.02 (0.82–1.26) Replication 4 794 815 0.45 0.46 1.00 (0.77–1.29) 0.88 (0.64–1.21)

All replications 4915 5607 1.12 (1.02–1.22) 1.10 (0.98–1.23) Dominant 1.3 102 1.11 (1.02–1.21)

GWASþ replications 5286 6870 1.11 (1.02–1.21) 1.21 (1.09–1.35) Dominant 1.1 103 1.14 (1.05–1.23) rs6038071 (20p13) [T] GWAS 371 1263 0.11 0.07 1.66 (1.22–2.24) 4.61 (1.23–17.3) Replication 1 575 760 0.12 0.09 1.19 (0.90–1.57) 2.54 (1.01–6.42) Replication 2a 1908 1752 0.09 0.09 0.94 (0.79–1.12) 0.73 (0.33–1.60) Replication 3 1373 1480 0.09 0.09 0.89 (0.72–1.08) 2.28 (1.03–5.05)

All replications 3856 3992 0.96 (0.85–1.08) 1.50 (0.94–2.39) Recessive 8.4 102 1.50 (0.94–2.40)

GWASþ replications 4227 5225 1.05 (0.94–1.17) 1.80 (1.15–2.80) Recessive 9.4 103 1.78 (1.15–2.77) rs11014993 (10p12.1) [C] GWAS 371 1263 0.24 0.21 0.86 (0.67–1.10) 3.05 (1.78–5.22) Replication 1 575 760 0.25 0.20 1.49 (1.18–1.88) 1.18 (0.73–1.91) Replication 2a 1908 1752 0.21 0.22 0.98 (0.85–1.14) 0.78 (0.56–1.08) Replication 3 1373 1480 0.20 0.21 0.96 (0.82–1.13) 0.81 (0.56–1.18)

All replications 3856 3992 1.05 (0.95–1.16) 0.85 (0.69–1.06) Recessive 1.0 101 0.84 (0.67–1.03)

GWASþ replications 4227 5225 0.99 (0.91–1.08) 1.05 (0.86–1.29) Recessive 7.2 101 1.04 (0.85–1.27) rs12296076 (11q23 locus) [G] GWAS 371 1263 0.38 0.30 1.99 (1.51–2.62) 1.49 (0.92–2.40) Dominant 1.8 106 1.90 (1.46–2.49) Replication 1 575 760 0.33 0.28 1.30 (1.03–1.64) 1.53 (1.04–2.24) Dominant 8.8 103 1.34 (1.08–1.67) rs10956369 (8q24.21 locus) [T] GWAS 371 1263 0.48 0.40 1.25 (0.96–1.64) 2.03 (1.46–2.82) Recessive 6.4 105 1.77 (1.33–2.35) Replication 1 575 760 0.44 0.37 1.35 (1.06–1.72) 1.75 (1.27–2.41) Recessive 7.1 103 1.48 (1.11–1.97) rs10441105 (7p14.1) [C] GWAS 371 1263 0.45 0.53 0.55 (0.41–0.72) 0.55 (0.40–0.78) Dominant 2.2 105 0.55 (0.42–0.72) Replication 1 575 760 0.42 0.48 0.83 (0.64–1.07) 0.61 (0.44–0.83) Recessive 6.1 103 0.68 (0.52–0.90) Minor allele for each SNP is indicated in square brackets; MAF, minor allele frequency; OR (95% CI) for heterozygous (ORhet.) and homozygous (ORhom.) genotypes are shown; P value and OR (95% CI) for the best model. All joint analyses were adjusted for study centre.

a

Due to DNA availability, different numbers of samples were genotyped for the three SNPs in replication 2 (see Materials and Methods).

at Ustavy AV - Krc on September 1, 2010

http://carcin.oxfordjournals.org

(4)

Background consisted of all genes with SNPs among the 610 664 analysed in the GWAS.

Finally, ToppGene Suite (44) was applied to rank a list of genes (test set) based on functional similarity (ToppGene tool) or topological feature in protein–protein interaction network (ToppNet tool) to a training gene list (training set). In both cases (ToppGene and ToppNet tool), the test gene list (test set) consisted of the same 350 genes from associated SNPs of the GWAS, also used for ALIGATOR and ConsensusPathDB. The training gene list (train-ing set) consisted of 459 candidate genes for CRC, which included the 451 genes used for selection of SNPs for replication analysis (criterion c, see Materials and Methods/forward SNP selection procedure) and the eight genes located in or close to the known risk loci for CRC from previous GWASs (SMAD7, EIF3H, SCG5, GREM1, FMN1, BMP4, CDH1 and RHPN2) (20). For each gene in the test gene list, the ToppGene tool derives a similarity score to the training gene list for 14 different features and combines all similarity scores into an overall score using statistical meta-analysis.

All three software tools pointed out to an overrepresentation of genes related to mitogen-activated protein kinase (MAPK) signalling events, with seven genes directly implicated in the MAPK signalling pathways in CRC (45). Therefore, the effect of an increasing number of risk alleles (one risk allele per each of the seven genes) on CRC risk was calculated from the GWAS data by counting two for a homozygote and one for a heterozygote genotype.

Epistasis between associated polymorphisms of the seven selected MAPK-related genes was tested using both logistic regression and multifactor-dimensionality reduction (MDR) methods for interaction. Logistic regression and MDR are two commonly used methods to estimate interaction between polymorphisms in association studies for complex diseases (46). Pair-wise interactions between the seven SNPs from MAPK-related genes were studied by logistic regression using the option ‘epistasis’ in the package PLINK (33), which assumes an allelic by allelic epistasis model for the interactions. For each SNP, all possible pair-wise combinations were tested. MDR is considered a non-parametric alternative to logistic regression for detecting interaction between polymorphisms. This model-free, non-parametric data reduction method classifies multi-locus genotypes into high-risk and low-risk groups (47). The MDR version 2.0 and the MDRpt version 1.0 module for permutation testing are open-source and freely available software (http://www.epistasis. org/). The software estimates the importance of the signals by using both cross-validation and permutation testing, which generates an empirical P value for the result. Additionally, a dendrogram shows the type of interaction between each pair of SNPs (epistasis, independence or redundancy).

Results

Supplementary Figure 1, available at Carcinogenesis Online,

de-scribes the individual steps from GWAS to replication studies and

overrepresentation analysis.

GWAS

Four SNPs reached genome-wide strong evidence for association at

allelic level (P , 5

 10

7

) (Figure 1): rs2209907 (P 5 3.4

 10

8

),

located 0.8 Mb from TLE4 (transducin-like enhancer of split 4);

rs16823149 (P 5 5.5

 10

8

), intronic in the open reading frame

C1orf21; rs4140904 (P 5 1.4

 10

7

), located 0.5 Mb from NCAPG

(non-SMC condensing I complex, subunit G) and rs4574118

(P 5 1.8

 10

7

), located 250 kb from PLGLA (plasminogen-like

A, non-coding RNA). The polymorphism rs2209907 was selected for

replication because it fulfilled two of the selection criteria used. For

the other three markers, none of the linked SNPs showed association,

and they were considered association artefacts.

No clear SNP peaks were identified in the Manhattan plot at the

ge-nome-wide significance level (P , 5

 10

7

), whereas a cluster of three

SNPs at chromosome 3 on the gene FAM19A1 [family with sequence

similarity 19 (chemokine C-C motif-like), member A1] reached

moder-ate evidence for association (P , 1

 10

5

) (Figure 1).

Sixteen additional SNPs reached moderate evidence for association

(allelic P , 1

 10

5

) (Figure 1). From the 10 CRC risk loci

iden-tified in previous GWASs (20), our study ideniden-tified rs10956369

(al-lelic P 5 4.2

 10

5

) at locus 8q24.21 and rs12296076 (allelic

P 5 2.4

 10

4

) at locus 11q23 among the 523 SNPs with a P value ,

10

4

. At chromosome 8q24.21, three additional polymorphisms

re-ported in the previous GWASs were also associated, but at a lower

level (rs10505477, P 5 5.9

 10

4

; rs6983267, P 5 7

 10

4

and

rs7014346, P 5 2.5

 10

4

). At the other eight risk loci from

pre-vious GWASs, our study did not show association at P , 10

2

for any

of the reported SNPs or for any marker in LD with them. At the

20p12.3 region, the two previously reported SNPs rs961253 and

rs355527 (10) were not present in our array, and the linked

polymor-phism rs2423154 showed a P value of 2.1

 10

3

for a dominant

model.

Using additional functional and positional criteria as described in

Materials and Methods, we selected 53 SNPs for replication studies

from the SNPs with a P value ,10

4

(supplementary Tables 1 and 2

are available at Carcinogenesis Online).

Replication 1

From the 53 SNPs selected for replication, 51 were successfully

gen-otyped in 575 familial CRC cases (collected by the German HNPCC

Consortium) and 760 controls (supplementary Table 1 is available at

Carcinogenesis Online). A total of six SNPs showed nominal

associ-ation at allelic level (P , 5

 10

2

): the two SNPs located in the

known CRC risk loci, rs10956369 at 8q24.21 (P 5 2.5

 10

4

) and

rs12296076 at 11q23 (P 5 5.9

 10

3

); one intronic marker in

MYO3A (myosin IIIA), rs11014993 (P 5 7.2

 10

3

); rs6038071

(P 5 3.9

 10

2

), located 40 kb upstream of CSNK2A1 (casein

ki-nase 2, alpha 1 polypeptide); two SNPs in high LD (Haploview LD

plot: D# 5 1.0, r

2

5 0.62 in the 760 controls) located between the

genes GLI3 (GLI family zinc finger 3) and INHBA (inhibin,

beta A), rs12701937 (P 5 1.1

 10

4

) and rs10441105 (P 5 2.1



10

3

). For these last two SNPs, the haplotype T-G, containing the

risk alleles of the two SNPs, was also associated with CRC

(P 5 8.0

 10

4

in the GWAS, P 5 1.0

 10

4

in replication 1).

Additional replications

From the six SNPs associated in replication 1, we selected three for

further replication analysis (Table II): rs12701937 between GLI3 and

INHBA, which is in high LD with rs10441105; rs6038071, close to

CSKN2A1, and rs11014993, the MYO3A intronic polymorphism. The

two SNPs located in the risk loci 8q24.21 (rs10956369) and 11q23

(rs12296076) were not included in the further replications.

For rs12701937, none of the replications 2–4 showed association of

this SNP with CRC. Joint analysis of all replication studies reached

a borderline significance (P 5 1.3

 10

2

for dominant model,

OR 1.11, 95% CI 1.02–1.21) (Table II).

For rs11014993 and the rare rs6038071, neither replications 2–3

nor joint analysis of all of them showed association for any of the two

SNPs with the risk of CRC (Table II). Only for rs6038071, a recessive

inheritance model did show an association (P 5 9.4

 10

3

, OR 1.78,

95% CI 1.15–2.77, for the joint analysis of GWAS and the

replica-tions, Table II). However, this result should be taken with caution due

to the low number of samples homozygous for the risk allele (48 cases

and 34 controls).

When considering only familial cases of CRC (all cases from

rep-lication 1, 6% from reprep-lication 2 and 13% from reprep-lication 3), the

associations were stronger than for all patients together, which were

mostly sporadic cases (Table III). For a total of 880 familial cases and

4790 controls from replications 1–3, the P value for rs12701937 was

2.0

 10

4

and OR 1.36 (95% CI 1.16–1.60) for a dominant model. For

rs6038071, the P value was 2.5

 10

3

for a recessive model (OR 2.49,

95% CI 1.35–4.59), and for rs11014993, it was 2.7

 10

4

for a

dom-inant model (OR 1.32, 95% CI 1.14–1.54).

Overrepresentation analysis and gene–gene interactions

All three software tools used to detect enrichment of particular GO or

pathway categories among associated SNPs from the GWAS pointed

out to an overrepresentation of genes related to MAPK signalling

events. First, the ALIGATOR software identified a significant

enrich-ment of 73 GO categories among the 1340 most significantly

associ-ated SNPs (in 350 genes) from the GWAS with allelic P value , 10

3

(Table IV). Nine of these enriched categories were related to the

MAPK activities and four of them ranked in the first eight most

at Ustavy AV - Krc on September 1, 2010

http://carcin.oxfordjournals.org

(5)

significant positions (supplementary Table 3A is available at

Carci-nogenesis Online). Genes with SNPs among the nine MAPK

catego-ries were MAP4K1, MAP4K5, epidermal growth factor receptor

(EGFR) (ERBB1), AXIN1, GRM4, RAPGEF2 and TNIK. Second,

and using the same 350 input genes from the GWAS,

Consensus-PathDB identified ErbB4 signalling events as the category with the

most prominent overlap (six overlapping genes) with the input gene

list (P 5 8.8

 10

4

). This overlap included the genes ERBB4, EGFR

(ERBB1), FYN, GRIN2B, PTPRZ1 and WWOX (supplementary Table

3B is available at Carcinogenesis Online). Finally, and also with the

same 350 input genes, ToppGene ranked three genes related to MAPK

signalling pathways (EGFR, AXIN1 and PRKCA) among the four

best candidate genes from the GWAS (P , 2

 10

5

), whereas

Top-pNet tool selected five genes from the same categories (EGRF, FYN,

AXIN1, PRKCA and GRIN2B) among the 10 best candidates (P , 3



10

4

) (supplementary Tables 3C and D are available at

Carcinogen-esis Online). Combining the three approaches, a total of 15 genes

related to the overrepresented MAPK signalling pathways were

de-tected among the 350 genes from the 1340 most significantly

associ-ated SNPs from the GWAS.

Based on their biological function in the Entrez Gene database and

their direct implication in the MAPK signalling pathways for CRC

(45), only 7 of the 15 genes were selected as being directly involved in

MAPK signalling events. These genes were EGFR (ERBB1), ERBB4,

Table III. Association for the three SNPs in familial CRC cases of replications 1–3 Familial

cases

Controls ORhet.(95% CI) ORhom.(95% CI) Best model P value OR (95% CI)

rs12701937 (7p14.1) [T]

All replications 880 4790 1.35 (1.13–1.60) 1.39 (1.13–1.71) Dominant 2.0 104 1.36 (1.16–1.60)

GWASþ replications 1251 6053 1.27 (1.09–1.46) 1.51 (1.27–1.80) Dominant 3.5 105 1.33 (1.16–1.52) rs6038071 (20p13) [T]

All replications 861 3992 1.09 (0.90–1.32) 2.53 (1.37–4.67) Recessive 2.5 103 2.49 (1.35–4.59)

GWASþ replications 1232 5225 1.21 (1.03–1.42) 2.73 (1.58–4.73) Recessive 3.0 104 2.64 (1.52–4.57) rs11014993 (10p12.1) [C]

All replications 861 3992 1.36 (1.17–1.60) 1.06 (0.75–1.50) Dominant 2.7 104 1.32 (1.14–1.54)

GWASþ replications 1232 5225 1.20 (1.05–1.37) 1.38 (1.04–1.82) Dominant 2.0 103 1.22 (1.08–1.39) Minor allele for each SNP is indicated in square brackets; OR (95% CI) for heterozygous (ORhet.) and homozygous (ORhom.) genotypes are shown; P value and OR (95% CI) for the best model. All results were adjusted for study centre.

Table IV. ALIGATOR results P value SNPs (GWAS) Number top SNPs Number genes P , 0.05 P , 0.01 P , 0.001

Number categories P value Number categories P value Number categories P value

104 153 46 16 0.740 5 0.607 1 0.445

103 1340 350 213 0 73 0.001 7 0.072

5 103 5130 1057 156 0.155 27 0.342 3 0.400

Number of GO categories reaching various levels of significance for overrepresentation (P , 0.05, P , 0.01 or P , 0.001) on the list of significant SNPs and their corresponding genes, together with P values indicating whether this number is significantly greater than that expected by chance. Most significant categories were obtained using the 1340 SNPs from the GWAS with allelic P value , 103, located within 350 different genes.

Fig. 1. Genome-wide association results from the initial GWAS analysis. Thelog10 of the allelic P values from 610 664 polymorphic SNPs comparing 371 familial CRC cases and 1263 controls of German origin are represented. The chromosomal distribution is shown (Manhattan plot). The horizontal line represents a threshold of 105for moderate evidence for association.

at Ustavy AV - Krc on September 1, 2010

http://carcin.oxfordjournals.org

(6)

MAP4K1, MAP4K5, PRKCA, AXIN1 and RAPGEF2. Combining

gen-otypes of the seven most significantly associated SNPs (one from each

gene) for the 371 cases and 1263 controls from the GWAS, we

cal-culated ORs corresponding to an increasing number of risk alleles.

The risk of CRC increased significantly, with a per allele OR of 1.34,

95% CI 1.11–1.61 (P

trend

5 2.2

 10

16

). For carriers of more than

four risk alleles, the risk of disease was increased

3-fold (OR 2.88,

95% CI 2.26–3.67), compared with carriers of less than or equal to

four risk alleles (Figure 2; supplementary Table 4 is available at

Carcinogenesis Online).

Pair-wise interactions using logistic regression did not yield evidence

for epistasis between any pair of risk alleles, with only borderline

evidences of interaction for rs6593210–rs7251456 (EGFR–MAP4K1,

P 5 0.095)

and

rs11157745–rs7251456

(MAP4K5–MAP4K1,

P 5 0.064). Using MDR, the pair rs6593210–rs7251456 (EGFR–

MAP4K1) showed nominal evidence of an interaction, with a

cross-validation consistency of 10/10, testing balanced accuracy of 55.5%

and permutation P value of 0.015 (based on 1000 permutations).

How-ever, visualization of the corresponding interaction dendrogram

re-vealed redundant interaction between any pair of SNPs, without

evidence of epistatic relationship.

Discussion

In our GWAS, 371 familial CRC cases and 1263 healthy controls were

successfully genotyped. After stringent quality control, a total of 523

polymorphic SNPs showed a P value for association ,10

4

for either

allelic, genotype, dominant or recessive model.

Application of several functional and positional criteria led to the

selection of 51 SNPs for replication in a cohort of 575 familial CRC

cases and 760 controls (replication 1). Six of them showed nominal

association (P , 5

 10

2

) with the same allele as in the GWAS.

Three SNPs were selected for further replications (replications 2–4).

Polymorphism rs12701937, located between the genes GLI3 and

INHBA remained associated in the combined cohort (P 5 1.1



10

3

, OR 1.14, 95% CI 1.05–1.23 for a dominant model). Both

rs12701937 and its neighbouring marker rs10441105 (D# 5 1.0,

r

2

5 0.62 in the HapMap CEU population) are located in a 37 kb

LD block at chromosome 7 (41.588–41.625 Mb), 50 kb upstream

from INHBA and 180 kb downstream from GLI3, not in LD with

any of the genes. The role of the sonic hedgehog (Shh) signalling,

which includes GLI3 as a mediator, in CRC remains controversial.

Alterations in the signalling have been shown in several cancer types

(48), although they do not seem to be common in CRC (49). INHBA

has been proposed as a potential marker for gastric (50) and head and

neck squamous cell carcinomas (51).

Already published GWASs for CRC and a meta-analysis have so far

identified 10 risk loci, with ORs ranging between 1.10 and 1.26 (20).

For loci on chromosome regions 8q24.21 and 11q23, our GWAS

identified SNPs (rs10956369 and rs12296076, respectively) with

P values ,10

4

, which were confirmed in replication 1. The CRC

susceptibility locus at 8q24.21 (rs6983267) (5,6), also associated with

prostate, breast, ovarian and other cancers (52), was represented

with several SNPs in our GWAS. Apart from rs10956369, the most

significantly associated SNP in the region (allelic P 5 4.2

 10

5

,

OR 1.41, 95% CI 1.20–1.66), the three polymorphisms reported in the

previous GWASs showed also association (rs10505477, P 5 5.9



10

4

; rs6983267, P 5 7

 10

4

and rs7014346, P 5 2.5

 10

4

).

Recently, rs6983267 has been functionally identified as the plausible

causal variant in the region (53,54). Since all these markers belong to

the same LD block, our association with rs10956369 and its adjacent

SNPs reflects the same signal as rs6983267. At chromosome 11q23,

rs12296076 (allelic P 5 2.4

 10

4

, OR 1.42, 95% CI 1.18–1.71) is

located 5 kb upstream of rs3802842, the most significantly associated

SNP in the original GWAS (4). This SNP is not present in the

Affy-metrix 6.0 array used by us, but it belongs to the same LD block

(D# 5 0.95, r

2

5 0.79 in the HapMap CEU population).

Interest-ingly, rs12296076 has been identified in silico as a

polymorphic-binding site for miRNAs (4). No mutations have been identified in

any of the transcripts in the region (16), thus the exact nature of the

association remains unknown.

In an attempt to enhance the case group with ‘genetically enriched’

cases (55), all 371 CRC patients in our GWAS had at least one

first-degree relative with CRC. This strategy of selecting only familial

cases increases the efficiency and power of the case–control

associa-tion study (23) and reduces the sample size required to detect common

disease susceptibility alleles (22). Association results for familial

cases, which were genotyped for three SNPs in replications 1–3, are

consistent with this theory because the association signal for familial

cases was stronger than for all patients, which were mostly sporadic.

These results might indicate that markers selected from the GWAS for

replication are more specific for familial than for sporadic CRC cases.

On the other hand, the limited number of patients investigated in the

GWAS (371 patients) compromised the power to detect weak signals

expected in a common disease like CRC. For example, the known risk

locus at 8q24.21 would not have passed our SNP selection criteria, if it

had not been implicated in the previous GWASs.

Only a small fraction of the estimated heritability for CRC (6% of

the full-sibling relative risk) can be explained by the so far discovered

10 low-risk loci (20). The missing heritability might be accounted by

many additional common genetic variants with even smaller effect

sizes (56), by rare variants with moderate to high penetrance which

are not gauged by the GWA platforms (21), by copy number variants

(57) or by more complex gene–gene (46) and gene–environment

inter-actions (58). There is an increasing interest in searching for networks of

genes, instead of single genes, contributing to disease (24).

Accord-ingly, we searched for enrichment of particular pathways or GO

cate-gories among the 1340 most significantly associated markers from the

GWAS (allelic P value , 10

3

), located within 350 genes. Using this

input information, three different software tools pointed to the

over-representation of genes related to the MAPK signalling pathways. We

consider these results of special interest because enrichment tools are

being an object of discussion due to their heterogeneity in the type of

statistical analysis performed and lack of reproducibility of results

among tools (59). The direct involvement of the MAPK signalling

pathways in CRC is well known (45), and several recently developed

drugs for CRC treatment are antagonist of EGFR, activation of which is

the main trigger of the MAPK signalling cascade (60). Thus, these

results demonstrate the validity of the enrichment approaches used

and they provide a proof-of-principle demonstration (42). Using the

genotypes from the GWAS, we observed a significant increase in ORs

with the number of risk alleles from seven SNPs selected from the

Fig. 2. Sample distribution according to the number of risk alleles in seven of the most significantly associated SNPs from the MAPK signalling pathway genes. CRC cases (n 5 371, grey columns), controls (n 5 1263, black columns).

at Ustavy AV - Krc on September 1, 2010

http://carcin.oxfordjournals.org

(7)

overrepresented

MAPK

categories

(P

trend

5 2.2



10

16

,

OR

per allele

5 1.34, 95% CI 1.11–1.61), with an OR 2.88 (95% CI

2.26–3.67) for carriers of more than four risk alleles. Pair-wise

combinations and MDR analysis provided no evidence of

interactive effects between any of the seven risk alleles from

MAPK-related genes, suggesting that each locus independently

car-ries CRC risk.

In conclusion, our GWAS replicated two known risk loci for CRC

at chromosomes 8q24.21 and 11q23 and identified a possible new risk

polymorphism, rs12701937, which should now be tested in larger

studies. The overrepresentation analysis successfully identified an

enrichment of the MAPK signalling pathways among the most

sig-nificantly associated markers, and accumulation of risk alleles in an

individual increased CRC risk. This and other similar approaches are

clearly needed in order to reveal the large proportion of missing

heritability of complex diseases such as CRC.

Supplementary material

Supplementary Tables 1–4 and Figure 1 can be found at http://carcin

.oxfordjournals.org/

Funding

German

National

Genome

Research

Network

(NGFN-Plus)

(01GS08181); Deutsche Krebshilfe (German Cancer Aid) (107318);

European Union (HEALTH-F4-2007-200767); Czech Science

Foun-dation (GACR) (310/07/1430).

Acknowledgements

We would like to thank all participants who joined the study and the NGFN-Plus CCN Group.

Conflict of Interest Statement: None declared.

References

1. Cheah,P.Y. (2009) Recent advances in colorectal cancer genetics and diag-nostics. Crit. Rev. Oncol. Hematol., 69, 45–55.

2. Lichtenstein,P. et al. (2000) Environmental and heritable factors in the causation of cancer–analyses of cohorts of twins from Sweden, Denmark, and Finland. N. Engl. J. Med., 343, 78–85.

3. Aaltonen,L. et al. (2007) Explaining the familial colorectal cancer risk associated with mismatch repair (MMR)-deficient and MMR-stable tumors. Clin. Cancer Res., 13, 356–361.

4. Tenesa,A. et al. (2008) Genome-wide association scan identifies a colorec-tal cancer susceptibility locus on 11q23 and replicates risk loci at 8q24 and 18q21. Nat. Genet., 40, 631–637.

5. Tomlinson,I. et al. (2007) A genome-wide association scan of tag SNPs identifies a susceptibility variant for colorectal cancer at 8q24.21. Nat. Genet., 39, 984–988.

6. Zanke,B.W. et al. (2007) Genome-wide association scan identifies a colo-rectal cancer susceptibility locus on chromosome 8q24. Nat. Genet., 39, 989–994.

7. Broderick,P. et al. (2007) A genome-wide association study shows that common alleles of SMAD7 influence colorectal cancer risk. Nat. Genet., 39, 1315–1317.

8. Jaeger,E. et al. (2008) Common genetic variants at the CRAC1 (HMPS) locus on chromosome 15q13.3 influence colorectal cancer risk. Nat. Genet., 40, 26–28.

9. Tomlinson,I.P. et al. (2008) A genome-wide association study identifies colorectal cancer susceptibility loci on chromosomes 10p14 and 8q23.3. Nat. Genet., 40, 623–630.

10. Houlston,R.S. et al. (2008) Meta-analysis of genome-wide association data identifies four new susceptibility loci for colorectal cancer. Nat. Genet., 40, 1426–1435.

11. Berndt,S.I. et al. (2008) Pooled analysis of genetic variation at chromosome 8q24 and colorectal neoplasia risk. Hum. Mol. Genet., 17, 2665–2672.

12. Curtin,K. et al. (2009) Meta association of colorectal cancer confirms risk alleles at 8q24 and 18q21. Cancer Epidemiol. Biomarkers Prev., 18, 616– 621.

13. Li,L. et al. (2008) A common 8q24 variant and the risk of colon cancer: a population-based case-control study. Cancer Epidemiol. Biomarkers Prev., 17, 339–342.

14. Matsuo,K. et al. (2009) Association between an 8q24 locus and the risk of colorectal cancer in Japanese. BMC Cancer, 9, 379.

15. Middeldorp,A. et al. (2009) Enrichment of low penetrance susceptibility loci in a Dutch familial colorectal cancer cohort. Cancer Epidemiol. Bio-markers Prev., 18, 3062–3067.

16. Pittman,A.M. et al. (2008) Refinement of the basis and impact of common 11q23.1 variation to the risk of developing colorectal cancer. Hum. Mol. Genet., 17, 3720–3727.

17. Poynter,J.N. et al. (2007) Variants on 9p24 and 8q24 are associated with risk of colorectal cancer: results from the Colon Cancer Family Registry. Cancer Res., 67, 11128–11132.

18. Schafmayer,C. et al. (2009) Investigation of the colorectal cancer suscep-tibility region on chromosome 8q24.21 in a large German case-control sample. Int. J. Cancer, 124, 75–80.

19. Slattery,M.L. et al. (2010) Increased risk of colon cancer associated with a genetic polymorphism of SMAD7. Cancer Res., 70, 1479–1485. 20. Tenesa,A. et al. (2009) New insights into the aetiology of colorectal cancer

from genome-wide association studies. Nat. Rev. Genet., 10, 353–358. 21. Hemminki,K. et al. (2009) Surveying the genomic landscape of colorectal

cancer. Am. J. Gastroenterol., 104, 789–790.

22. Antoniou,A.C. et al. (2003) Polygenic inheritance of breast cancer: impli-cations for design of association studies. Genet. Epidemiol., 25, 190–202. 23. Lange,E.M. et al. (2008) Family-based samples can play an important role in genetic association studies. Cancer Epidemiol. Biomarkers Prev., 17, 2208–2214.

24. Hardy,J. et al. (2009) Genomewide association studies and human disease. N. Engl. J. Med., 360, 1759–1768.

25. Schafmayer,C. et al. (2007) Genetic investigation of DNA-repair pathway genes PMS2, MLH1, MSH2, MSH6, MUTYH, OGG1 and MTH1 in spo-radic colon cancer. Int. J. Cancer, 121, 555–558.

26. Mangold,E. et al. (2005) Spectrum and frequencies of mutations in MSH2 and MLH1 identified in 1,721 German families suspected of hereditary nonpolyposis colorectal cancer. Int. J. Cancer, 116, 692–702.

27. Krawczak,M. et al. (2006) PopGen: population-based recruitment of pa-tients and controls for the analysis of complex genotype-phenotype rela-tionships. Community Genet., 9, 55–61.

28. Frank,B. et al. (2008) Ten recently identified associations between nsSNPs and colorectal cancer could not be replicated in German families. Cancer Lett., 271, 153–157.

29. John,U. et al. (2001) Study of Health In Pomerania (SHIP): a health ex-amination survey in an east German region: objectives and design. Soz. Praventivmed., 46, 186–194.

30. Brenner,H. et al. (2006) Does a negative screening colonoscopy ever need to be repeated? Gut, 55, 1145–1150.

31. Pechlivanis,S. et al. (2009) Genetic variation in adipokine genes and risk of colorectal cancer. Eur. J. Endocrinol., 160, 933–940.

32. McCarthy,M.I. et al. (2008) Genome-wide association studies for com-plex traits: consensus, uncertainty and challenges. Nat. Rev. Genet., 9, 356–369.

33. Purcell,S. et al. (2007) PLINK: a tool set for whole-genome association and population-based linkage analyses. Am. J. Hum. Genet., 81, 559–575. 34. Nelis,M. et al. (2009) Genetic structure of Europeans: a view from the

North-East. PLoS One, 4, e5472.

35. McKay,J.D. et al. (2008) Lung cancer susceptibility locus at 5p15.33. Nat. Genet., 40, 1404–1406.

36. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature, 447, 661–678.

37. Williams,S.M. et al. (2007) Problems with genome-wide association stud-ies. Science, 316, 1840–1842.

38. Sjoblom,T. et al. (2006) The consensus coding sequences of human breast and colorectal cancers. Science, 314, 268–274.

39. Wood,L.D. et al. (2007) The genomic landscapes of human breast and colorectal cancers. Science, 318, 1108–1113.

40. Starr,T.K. et al. (2009) A transposon-based genetic screen in mice identifies genes altered in colorectal cancer. Science, 323, 1747–1750.

41. Barrett,J.C. et al. (2005) Haploview: analysis and visualization of LD and haplotype maps. Bioinformatics, 21, 263–265.

42. Holmans,P. et al. (2009) Gene ontology analysis of GWA study data sets provides insights into the biology of bipolar disorder. Am. J. Hum. Genet., 85, 13–24.

at Ustavy AV - Krc on September 1, 2010

http://carcin.oxfordjournals.org

(8)

43. Kamburov,A. et al. (2009) ConsensusPathDB—a database for integrating human functional interaction networks. Nucleic Acids Res., 37, D623– D628.

44. Chen,J. et al. (2009) ToppGene Suite for gene list enrichment analysis and candidate gene prioritization. Nucleic Acids Res., 37, W305–W311. 45. Fang,J.Y. et al. (2005) The MAPK signalling pathways and colorectal

cancer. Lancet Oncol., 6, 322–327.

46. Cordell,H.J. (2009) Detecting gene-gene interactions that underlie human diseases. Nat. Rev. Genet., 10, 392–404.

47. Ritchie,M.D. et al. (2001) Multifactor-dimensionality reduction reveals high-order interactions among estrogen-metabolism genes in sporadic breast cancer. Am. J. Hum. Genet., 69, 138–147.

48. Kim,J.E. et al. (2009) Sonic hedgehog signaling proteins and ATP-binding cassette G2 are aberrantly expressed in diffuse large B-cell lymphoma. Mod. Pathol., 22, 1312–1320.

49. Chatel,G. et al. (2007) Hedgehog signaling pathway is inactive in colorec-tal cancer cell lines. Int. J. Cancer, 121, 2622–2627.

50. Takeno,A. et al. (2008) Integrative approach for differentially overexpressed genes in gastric cancer by combining large-scale gene expression profiling and network analysis. Br. J. Cancer, 99, 1307– 1315.

51. Shimizu,S. et al. (2007) Identification of molecular targets in head and neck squamous cell carcinomas based on genome-wide gene expression profil-ing. Oncol. Rep., 18, 1489–1497.

52. Ghoussaini,M. et al. (2008) Multiple loci with different cancer specificities within the 8q24 gene desert. J. Natl Cancer Inst., 100, 962–966.

53. Pomerantz,M.M. et al. (2009) The 8q24 cancer risk variant rs6983267 shows long-range interaction with MYC in colorectal cancer. Nat. Genet., 41, 882–884.

54. Tuupanen,S. et al. (2009) The common colorectal cancer predisposition SNP rs6983267 at chromosome 8q24 confers potential to enhanced Wnt signaling. Nat. Genet., 41, 885–890.

55. Thomas,D.C. et al. (2005) Recent developments in genomewide association scans: a workshop summary and review. Am. J. Hum. Genet., 77, 337–345. 56. Frazer,K.A. et al. (2009) Human genetic variation and its contribution to

complex traits. Nat. Rev. Genet., 10, 241–251.

57. Manolio,T.A. et al. (2009) Finding the missing heritability of complex diseases. Nature, 461, 747–753.

58. Maher,B. (2008) Personal genomes: the case of the missing heritability. Nature, 456, 18–21.

59. Huang da,W. et al. (2009) Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res., 37, 1–13.

60. Ciardiello,F. et al. (2008) EGFR antagonists in cancer treatment. N. Engl. J. Med., 358, 1160–1174.

Received March 29, 2010; revised July 1, 2010; accepted July 3, 2010

at Ustavy AV - Krc on September 1, 2010

http://carcin.oxfordjournals.org

Riferimenti

Documenti correlati

that cortisol and dehydroepiandrosterone (DHEA) can influence alternative splicing of the GR, controlling the level of exon inclusion/exclusion in the mRNA transcript [17,18],

La città e il suo porto, infatti, nella maggior parte dei casi non costituiscono la destinazione finale di quanto vi è movimentato, ma sempre più come un luogo di transito e

Keywords: Masonry vault; laser-scanning; thrust network analysis; point cloud; geometric configuration; structural analysis; equilibrium of nodes Citation: Maria Grazia D'Urso,

Hanno parlato di aspettative e motivazioni che sono mutate nel corso dello svolgimento del Progetto; un'evoluzione data in relazione alle sfide giornaliere che ognuno si pone.

Let’s say that in general there’s a communication activity that we haven’t centralised or obliged if not within the terms of community obligations, in the sense that you have to

Les indications que l’on peut envisager pour l’amélioration du travail et pour la santé et la sécurité des opérateurs, en ce qui concerne le processus de travail dans son

Proper storage, at appropriate temperature, humidity, packaging, concentration of oxygen contributes during the aging to the wine quality thanks to the chemical reaction