Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
PLoS One ; 17(10): e0274867, 2022.
Article in English | MEDLINE | ID: mdl-36227936

ABSTRACT

Lipoedema is a chronic adipose tissue disorder mainly affecting women, causing excess subcutaneous fat deposition on the lower limbs with pain and tenderness. There is often a family history of lipoedema, suggesting a genetic origin, but the contribution of genetics is currently unclear. A tightly phenotyped cohort of 200 lipoedema patients was recruited from two UK specialist clinics. Objective clinical characteristics and measures of quality of life data were obtained. In an attempt to understand the genetic architecture of the disease better, genome-wide single nucleotide polymorphism (SNP) genotype data were obtained, and a genome wide association study (GWAS) was performed on 130 of the recruits. The analysis revealed genetic loci suggestively associated with the lipoedema phenotype, with further support provided by an independent cohort taken from the 100,000 Genomes Project. The top SNP rs1409440 (ORmeta ≈ 2.01, Pmeta ≈ 4 x 10-6) is located upstream of LHFPL6, which is thought to be involved with lipoma formation. Exactly how this relates to lipoedema is not yet understood. This first GWAS of a UK lipoedema cohort has identified genetic regions of suggestive association with the disease. Further replication of these findings in different populations is warranted.


Subject(s)
Genome-Wide Association Study , Lipedema , Female , Genotype , Humans , Polymorphism, Single Nucleotide , Quality of Life , United Kingdom
2.
Blood Cancer J ; 9(8): 60, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31387987

ABSTRACT

To gain insight into multiple myeloma (MM) tumorigenesis, we analyzed the mutational signatures in 874 whole-exome and 850 whole-genome data from the CoMMpass Study. We identified that coding and non-coding regions are differentially dominated by distinct single-nucleotide variant (SNV) mutational signatures, as well as five de novo structural rearrangement signatures. Mutational signatures reflective of different principle mutational processes-aging, defective DNA repair, and apolipoprotein B editing complex (APOBEC)/activation-induced deaminase activity-characterize MM. These mutational signatures show evidence of subgroup specificity-APOBEC-attributed signatures associated with MAF translocation t(14;16) and t(14;20) MM; potentially DNA repair deficiency with t(11;14) and t(4;14); and aging with hyperdiploidy. Mutational signatures beyond that associated with APOBEC are independent of established prognostic markers and appear to have relevance to predicting high-risk MM.


Subject(s)
Multiple Myeloma/genetics , Mutation , APOBEC Deaminases/genetics , DNA Mutational Analysis , Humans , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Prognosis , Proto-Oncogene Proteins c-maf/genetics , Survival Rate , Transcriptome , Translocation, Genetic , Exome Sequencing/methods , Whole Genome Sequencing/methods
3.
Blood Adv ; 3(1): 21-32, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30606723

ABSTRACT

The identification of driver mutations is fundamental to understanding oncogenesis. Although genes frequently mutated in B-cell lymphoma have been identified, the search for driver mutations has largely focused on the coding genome. Here we report an analysis of the noncoding genome using whole-genome sequencing data from 117 patients with B-cell lymphoma. Using promoter capture Hi-C data in naive B cells, we define cis-regulatory elements, which represent an enriched subset of the noncoding genome in which to search for driver mutations. Regulatory regions were identified whose mutation significantly alters gene expression, including copy number variation at cis-regulatory elements targeting CD69, IGLL5, and MMP14, and single nucleotide variants in a cis-regulatory element for TPRG1 We also show the commonality of pathways targeted by coding and noncoding mutations, exemplified by MMP14, which regulates Notch signaling, a pathway important in lymphomagenesis and whose expression is associated with patient survival. This study provides an enhanced understanding of lymphomagenesis and describes the advantages of using chromosome conformation capture to decipher noncoding mutations relevant to cancer biology.


Subject(s)
Genetic Association Studies , Genetic Predisposition to Disease , Lymphoma, B-Cell/genetics , Mutation , RNA, Untranslated/genetics , Computational Biology/methods , DNA Copy Number Variations , Databases, Genetic , Gene Amplification , Gene Expression Regulation, Neoplastic , Genetic Association Studies/methods , Humans , Lymphoma, B-Cell/mortality , Lymphoma, B-Cell/pathology , Open Reading Frames , Promoter Regions, Genetic , Whole Genome Sequencing
4.
Nat Genet ; 50(10): 1375-1380, 2018 10.
Article in English | MEDLINE | ID: mdl-30224643

ABSTRACT

Efforts are being directed to systematically analyze the non-coding regions of the genome for cancer-driving mutations1-6. cis-regulatory elements (CREs) represent a highly enriched subset of the non-coding regions of the genome in which to search for such mutations. Here we use high-throughput chromosome conformation capture techniques (Hi-C) for 19,023 promoter fragments to catalog the regulatory landscape of colorectal cancer in cell lines, mapping CREs and integrating these with whole-genome sequence and expression data from The Cancer Genome Atlas7,8. We identify a recurrently mutated CRE interacting with the ETV1 promoter affecting gene expression. ETV1 expression influences cell viability and is associated with patient survival. We further refine our understanding of the regulatory effects of copy-number variations, showing that RASL11A is targeted by a previously identified enhancer amplification1. This study reveals new insights into the complex genetic alterations driving tumor development, providing a paradigm for employing chromosome conformation capture to decipher non-coding CREs relevant to cancer biology.


Subject(s)
Cell Transformation, Neoplastic/genetics , Chromosomes, Human/chemistry , Codon, Nonsense , Colorectal Neoplasms/genetics , Computational Biology/methods , High-Throughput Nucleotide Sequencing/methods , Caco-2 Cells , Colorectal Neoplasms/epidemiology , DNA, Neoplasm/chemistry , Databases, Genetic , Gene Frequency , HT29 Cells , HeLa Cells , Hep G2 Cells , Humans , K562 Cells , MCF-7 Cells , Nucleic Acid Conformation , Promoter Regions, Genetic/genetics , Regulatory Sequences, Nucleic Acid/genetics , Tumor Cells, Cultured
5.
Neuro Oncol ; 20(11): 1485-1493, 2018 10 09.
Article in English | MEDLINE | ID: mdl-29762745

ABSTRACT

Background: Meningiomas are adult brain tumors originating in the meningeal coverings of the brain and spinal cord, with significant heritable basis. Genome-wide association studies (GWAS) have previously identified only a single risk locus for meningioma, at 10p12.31. Methods: To identify a susceptibility locus for meningioma, we conducted a meta-analysis of 2 GWAS, imputed using a merged reference panel from the 1000 Genomes Project and UK10K data, with validation in 2 independent sample series totaling 2138 cases and 12081 controls. Results: We identified a new susceptibility locus for meningioma at 11p15.5 (rs2686876, odds ratio = 1.44, P = 9.86 × 10-9). A number of genes localize to the region of linkage disequilibrium encompassing rs2686876, including RIC8A, which plays a central role in the development of neural crest-derived structures, such as the meninges. Conclusions: This finding advances our understanding of the genetic basis of meningioma development and provides additional support for a polygenic model of meningioma.


Subject(s)
Biomarkers, Tumor/genetics , Chromosomes, Human, Pair 11/genetics , Genetic Loci , Genome-Wide Association Study , Meningeal Neoplasms/genetics , Meningioma/genetics , Polymorphism, Single Nucleotide , Adult , Aged , Case-Control Studies , Female , Follow-Up Studies , Genetic Predisposition to Disease , Genotype , Humans , Linkage Disequilibrium , Male , Meningeal Neoplasms/pathology , Meningioma/pathology , Middle Aged , Prognosis , Risk Factors , Young Adult
6.
Leukemia ; 32(11): 2459-2470, 2018 11.
Article in English | MEDLINE | ID: mdl-29654271

ABSTRACT

Multiple myeloma (MM) is a biologically heterogeneous malignancy, however, the mechanisms underlying this complexity are incompletely understood. We report an analysis of the whole-genome sequencing of 765 MM patients from CoMMpass. By employing promoter capture Hi-C in naïve B-cells, we identify cis-regulatory elements (CREs) that represent a highly enriched subset of the non-coding genome in which to search for driver mutations. We identify regulatory regions whose mutation significantly alters the expression of genes as candidate non-coding drivers, including copy number variation (CNV) at CREs of MYC and single-nucleotide variants (SNVs) in a PAX5 enhancer. To better inform the interplay between non-coding driver mutations with other driver mechanisms, and their respective roles in oncogenic pathways, we extended our analysis identifying coding drivers in 40 genes, including 11 novel candidates. We demonstrate the same pathways can be targeted by coding and non-coding mutations; exemplified by IRF4 and PRDM1, along with BCL6 and PAX5, genes that are central to plasma cell differentiation. This study reveals new insights into the complex genetic alterations driving MM development and an enhanced understanding of oncogenic pathways.


Subject(s)
Multiple Myeloma/genetics , Oncogenes/genetics , Adult , Aged , Aged, 80 and over , Carcinogenesis/genetics , Cell Differentiation/genetics , DNA Copy Number Variations/genetics , Female , Genome, Human/genetics , High-Throughput Nucleotide Sequencing/methods , Humans , Male , Middle Aged , Mutation/genetics , Polymorphism, Single Nucleotide/genetics , Promoter Regions, Genetic/genetics , Regulatory Sequences, Nucleic Acid/genetics , Whole Genome Sequencing/methods
7.
Cell Rep ; 20(11): 2556-2564, 2017 Sep 12.
Article in English | MEDLINE | ID: mdl-28903037

ABSTRACT

Multiple myeloma (MM) is a malignancy of plasma cells. Genome-wide association studies have shown that variation at 5q15 influences MM risk. Here, we have sought to decipher the causal variant at 5q15 and the mechanism by which it influences tumorigenesis. We show that rs6877329 G > C resides in a predicted enhancer element that physically interacts with the transcription start site of ELL2. The rs6877329-C risk allele is associated with reduced enhancer activity and lowered ELL2 expression. Since ELL2 is critical to the B cell differentiation process, reduced ELL2 expression is consistent with inherited genetic variation contributing to arrest of plasma cell development, facilitating MM clonal expansion. These data provide evidence for a biological mechanism underlying a hereditary risk of MM at 5q15.


Subject(s)
Chromosomes, Human, Pair 5/genetics , Enhancer Elements, Genetic , Genetic Predisposition to Disease , Multiple Myeloma/genetics , Polymorphism, Single Nucleotide/genetics , Transcriptional Elongation Factors/genetics , Alleles , Diploidy , Epigenesis, Genetic , Epigenomics , Genetic Loci , Humans , Nuclear Proteins/metabolism , Physical Chromosome Mapping , Prognosis , Protein Binding , Risk Factors , Transcription Elongation, Genetic , Unfolded Protein Response/genetics
8.
Oncotarget ; 8(22): 36203-36210, 2017 May 30.
Article in English | MEDLINE | ID: mdl-28404951

ABSTRACT

The genetic basis underlying the inherited risk of developing multiple myeloma (MM) is largely unknown. To examine the impact of rare protein altering variants on the risk of developing MM we analyzed high-coverage exome sequencing data on 513 MM cases and 1,569 healthy controls, performing both single variant and gene burden tests. We did not identify any recurrent coding low-frequency alleles (1-5%) with moderate effect that were statistically associated with MM. In a gene burden analysis we did however identify a promising relationship between variation in the marrow kinetochore microtubule stromal gene KIF18A, which plays a role in control mitotic chromosome positioning dynamics, and risk of MM (P =3.6x10-6). Further analysis showed KIF18A displays a distinct pattern of expression across molecular subgroups of MM as well as being associated with patient survival. Our results inform future study design and provide a resource for contextualizing the impact of candidate MM susceptibility genes.


Subject(s)
Gene Dosage , Kinesins/genetics , Multiple Myeloma/genetics , Aged , DNA Mutational Analysis , Exome , Female , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , High-Throughput Nucleotide Sequencing , Humans , Male , Mitosis , Multiple Myeloma/mortality , Polymorphism, Single Nucleotide , Risk , Transcriptome
9.
Gastroenterology ; 152(1): 75-77.e4, 2017 01.
Article in English | MEDLINE | ID: mdl-27713038

ABSTRACT

High-throughput sequencing analysis has accelerated searches for genes associated with risk for colorectal cancer (CRC); germline mutations in NTHL1, RPS20, FANCM, FAN1, TP53, BUB1, BUB3, LRP6, and PTPN12 have been recently proposed to increase CRC risk. We attempted to validate the association between variants in these genes and development of CRC in a systematic review of 11 publications, using sequence data from 863 familial CRC cases and 1604 individuals without CRC (controls). All cases were diagnosed at an age of 55 years or younger and did not carry mutations in an established CRC predisposition gene. We found sufficient evidence for NTHL1 to be considered a CRC predisposition gene-members of 3 unrelated Dutch families were homozygous for inactivating p.Gln90Ter mutations; a Canadian woman with polyposis, CRC, and multiple tumors was reported to be heterozygous for the inactivating NTHL1 p.Gln90Ter/c.709+1G>A mutations; and a man with polyposis was reported to carry p.Gln90Ter/p.Gln287Ter; whereas no inactivating homozygous or compound heterozygous mutations were detected in controls. Variants that disrupted RPS20 were detected in a Finnish family with early-onset CRC (p.Val50SerfsTer23), a 39-year old individual with metachronous CRC (p.Leu61GlufsTer11 mutation), and a 41-year-old individual with CRC (missense p.Val54Leu), but not in controls. We therefore found published evidence to support the association between variants in NTHL1 and RPS20 with CRC, but not of other recently reported CRC susceptibility variants. We urge the research community to adopt rigorous statistical and biological approaches coupled with independent replication before making claims of pathogenicity.


Subject(s)
Adenomatous Polyposis Coli/genetics , Colorectal Neoplasms/genetics , Deoxyribonuclease (Pyrimidine Dimer)/genetics , Genetic Predisposition to Disease/genetics , Ribosomal Proteins/genetics , Germ-Line Mutation , Humans
10.
Fam Cancer ; 15(4): 593-9, 2016 10.
Article in English | MEDLINE | ID: mdl-27356891

ABSTRACT

Although family history is a major risk factor for colorectal cancer (CRC) a genetic diagnosis cannot be obtained in over 50 % of familial cases when screened for known CRC cancer susceptibility genes. The genetics of undefined-familial CRC is complex and recent studies have implied additional clinically actionable mutations for CRC in susceptibility genes for other cancers. To clarify the contribution of non-CRC susceptibility genes to undefined-familial CRC we conducted a mutational screen of 114 cancer susceptibility genes in 847 patients with early-onset undefined-familial CRC and 1609 controls by analysing high-coverage exome sequencing data. We implemented American College of Medical Genetics and Genomics standards and guidelines for assigning pathogenicity to variants. Globally across all 114 cancer susceptibility genes no statistically significant enrichment of likely pathogenic variants was shown (6.7 % cases 57/847, 5.3 % controls 85/1609; P = 0.15). Moreover there was no significant enrichment of mutations in genes such as TP53 or BRCA2 which have been proposed for clinical testing in CRC. In conclusion, while we identified genes that may be considered interesting candidates as determinants of CRC risk warranting further research, there is currently scant evidence to support a role for genes other than those responsible for established CRC syndromes in the clinical management of familial CRC.


Subject(s)
Colorectal Neoplasms/genetics , Genetic Pleiotropy , Genetic Predisposition to Disease , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Case-Control Studies , Colorectal Neoplasms/etiology , Heterozygote , Humans , Mutation , Proto-Oncogene Proteins/genetics , RecQ Helicases/genetics , Tumor Suppressor Proteins/genetics
11.
Nat Commun ; 7: 11883, 2016 06 22.
Article in English | MEDLINE | ID: mdl-27329137

ABSTRACT

Colorectal cancer (CRC) displays a complex pattern of inheritance. It is postulated that much of the missing heritability of CRC is enshrined in high-impact rare alleles, which are mechanistically and clinically important. In this study, we assay the impact of rare germline mutations on CRC, analysing high-coverage exome sequencing data on 1,006 early-onset familial CRC cases and 1,609 healthy controls, with additional sequencing and array data on up to 5,552 cases and 6,792 controls. We identify highly penetrant rare mutations in 16% of familial CRC. Although the majority of these reside in known genes, we identify POT1, POLE2 and MRE11 as candidate CRC genes. We did not identify any coding low-frequency alleles (1-5%) with moderate effect. Our study clarifies the genetic architecture of CRC and probably discounts the existence of further major high-penetrance susceptibility genes, which individually account for >1% of the familial risk. Our results inform future study design and provide a resource for contextualizing the impact of new CRC genes.


Subject(s)
Colorectal Neoplasms/genetics , Genetic Predisposition to Disease , Germ-Line Mutation , Adult , Age Factors , Age of Onset , Alleles , Case-Control Studies , Exome , Female , Genetic Variation , Humans , Male , Middle Aged , Pedigree , Phenotype , Risk Factors , United Kingdom
13.
F1000Res ; 5: 2813, 2016.
Article in English | MEDLINE | ID: mdl-28105316

ABSTRACT

The advent of high-throughput sequencing has accelerated our ability to discover genes predisposing to disease and is transforming clinical genomic sequencing. In both contexts knowledge of the spectrum and frequency of genetic variation in the general population and in disease cohorts is vital to the interpretation of sequencing data. While population level data is becoming increasingly available from publicly accessible sources, as exemplified by The Exome Aggregation Consortium (ExAC), the availability of large-scale disease-specific frequency information is limited. These data are of particular importance to contextualise findings from clinical mutation screens and small gene discovery projects. This is especially true for cancer, which is typified by a number of hereditary predisposition syndromes.  Although mutation frequencies in tumours are available from resources such as Cosmic and The Cancer Genome Atlas, a similar facility for germline variation is lacking. Here we present the Cancer Variation Resource (CanVar) an online database which has been developed using the ExAC framework to provide open access to germline variant frequency data from the sequenced exomes of cancer patients. In its first release, CanVar catalogues the exomes of 1,006 familial early-onset colorectal cancer (CRC) patients sequenced at The Institute of Cancer Research. It is anticipated that CanVar will host data for additional cancers, providing a resource for others studying cancer predisposition and an example of how the research community can utilise the ExAC framework to share sequencing data.

15.
Nat Commun ; 6: 7207, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-26068201

ABSTRACT

Anaplastic oligodendroglioma (AO) are rare primary brain tumours that are generally incurable, with heterogeneous prognosis and few treatment targets identified. Most oligodendrogliomas have chromosomes 1p/19q co-deletion and an IDH mutation. Here we analysed 51 AO by whole-exome sequencing, identifying previously reported frequent somatic mutations in CIC and FUBP1. We also identified recurrent mutations in TCF12 and in an additional series of 83 AO. Overall, 7.5% of AO are mutated for TCF12, which encodes an oligodendrocyte-related transcription factor. Eighty percent of TCF12 mutations identified were in either the bHLH domain, which is important for TCF12 function as a transcription factor, or were frameshift mutations leading to TCF12 truncated for this domain. We show that these mutations compromise TCF12 transcriptional activity and are associated with a more aggressive tumour type. Our analysis provides further insights into the unique and shared pathways driving AO.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Brain Neoplasms/genetics , Mutation , Oligodendroglioma/genetics , Down-Regulation , Humans , Transcriptional Activation/genetics
16.
Sci Rep ; 5: 10442, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25990418

ABSTRACT

Genome-wide association studies (GWAS) of colorectal cancer (CRC) have identified 23 susceptibility loci thus far. Analyses of previously conducted GWAS indicate additional risk loci are yet to be discovered. To identify novel CRC susceptibility loci, we conducted a new GWAS and performed a meta-analysis with five published GWAS (totalling 7,577 cases and 9,979 controls of European ancestry), imputing genotypes utilising the 1000 Genomes Project. The combined analysis identified new, significant associations with CRC at 1p36.2 marked by rs72647484 (minor allele frequency [MAF] = 0.09) near CDC42 and WNT4 (P = 1.21 × 10(-8), odds ratio [OR] = 1.21 ) and at 16q24.1 marked by rs16941835 (MAF = 0.21, P = 5.06 × 10(-8); OR = 1.15) within the long non-coding RNA (lncRNA) RP11-58A18.1 and ~500 kb from the nearest coding gene FOXL1. Additionally we identified a promising association at 10p13 with rs10904849 intronic to CUBN (MAF = 0.32, P = 7.01 × 10(-8); OR = 1.14). These findings provide further insights into the genetic and biological basis of inherited genetic susceptibility to CRC. Additionally, our analysis further demonstrates that imputation can be used to exploit GWAS data to identify novel disease-causing variants.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Genetic Predisposition to Disease , Chromosome Mapping , Female , Forkhead Transcription Factors/genetics , Gene Frequency/genetics , Genome-Wide Association Study , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Receptors, Cell Surface/genetics
17.
J Clin Oncol ; 33(5): 426-32, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25559809

ABSTRACT

PURPOSE: Knowledge of the contribution of high-penetrance susceptibility to familial colorectal cancer (CRC) is relevant to the counseling, treatment, and surveillance of CRC patients and families. PATIENTS AND METHODS: To quantify the impact of germline mutation to familial CRC, we sequenced the mismatch repair genes (MMR) APC, MUTYH, and SMAD4/BMPR1A in 626 early-onset familial CRC cases ascertained through a population-based United Kingdom national registry. In addition, we evaluated the contribution of mutations in the exonuclease domain (exodom) of POLE and POLD1 genes that have recently been reported to confer CRC risk. RESULTS: Overall mutations (pathogenic, likely pathogenic) in MMR genes make the highest contribution to familial CRC (10.9%). Mutations in the other established CRC genes account for 3.3% of cases. POLE/POLD1 exodom mutations were identified in three patients with family histories consistent with dominant transmission of CRC. Collectively, mutations in the known genes account for 14.2% of familial CRC (89 of 626 cases; 95% CI = 11.5, 17.2). CONCLUSION: A genetic diagnosis is feasible in a high proportion of familial CRC. Mainstreaming such analysis in clinical practice should enable the medical management of patients and their families to be optimized. Findings suggest CRC screening of POLE and POLD1 mutation carriers should be comparable to that afforded to those at risk of HNPCC. Although the risk of CRC associated with unexplained familial CRC is in general moderate, in some families the risk is substantive and likely to be the consequence of unidentified genes, as exemplified by POLE and POLD1. Our findings have utility in the design of genetic analyses to identify such novel CRC risk genes.


Subject(s)
Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , DNA Mismatch Repair , Exome , Genetic Testing/methods , Germ-Line Mutation , Adenomatous Polyposis Coli Protein/genetics , Adult , Aged , Bone Morphogenetic Protein Receptors, Type I/genetics , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA Glycosylases/genetics , DNA Mismatch Repair/genetics , DNA Polymerase II/genetics , DNA Polymerase III/genetics , Feasibility Studies , Female , Genetic Predisposition to Disease , Germ-Line Mutation/genetics , Humans , Male , Middle Aged , Pedigree , Poly-ADP-Ribose Binding Proteins , Predictive Value of Tests , Registries , Risk Assessment , Risk Factors , Sequence Analysis, DNA , Smad4 Protein/genetics , United Kingdom/epidemiology
18.
Hum Mol Genet ; 23(17): 4729-37, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24737748

ABSTRACT

To identify common variants influencing colorectal cancer (CRC) risk, we performed a meta-analysis of five genome-wide association studies, comprising 5626 cases and 7817 controls of European descent. We conducted replication of top ranked single nucleotide polymorphisms (SNPs) in additional series totalling 14 037 cases and 15 937 controls, identifying a new CRC risk locus at 10q24.2 [rs1035209; odds ratio (OR) = 1.13, P = 4.54 × 10(-11)]. We also performed meta-analysis of our studies, with previously published data, of several recently purported CRC risk loci. We failed to find convincing evidence for a previously reported genome-wide association at rs11903757 (2q32.3). Of the three additional loci for which evidence of an association in Europeans has been previously described we failed to show an association between rs59336 (12q24.21) and CRC risk. However, for the other two SNPs, our analyses demonstrated new, formally significant associations with CRC. These are rs3217810 intronic in CCND2 (12p13.32; OR = 1.19, P = 2.16 × 10(-10)) and rs10911251 near LAMC1 (1q25.3; OR = 1.09, P = 1.75 × 10(-8)). Additionally, we found some evidence to support a relationship between, rs647161, rs2423297 and rs10774214 and CRC risk originally identified in East Asians in our European datasets. Our findings provide further insights into the genetic and biological basis of inherited genetic susceptibility to CRC.


Subject(s)
Colorectal Neoplasms/genetics , Genetic Loci/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Asian People/genetics , Female , Humans , Male , Middle Aged , Odds Ratio , Polymorphism, Single Nucleotide/genetics , Quantitative Trait Loci/genetics , White People/genetics
19.
Hum Mol Genet ; 22(24): 5075-82, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-23904454

ABSTRACT

Recent genome-wide association studies (GWASs) have identified common variants at 16 autosomal regions influencing the risk of developing colorectal cancer (CRC). To decipher the genetic basis of the association signals at these loci, we performed a meta-analysis of data from five GWASs, totalling 5626 cases and 7817 controls, using imputation to recover un-typed genotypes. To enhance our ability to discover low-frequency risk variants, in addition to using 1000 Genomes Project data as a reference panel, we made use of high-coverage sequencing data on 253 individuals, 199 with early-onset familial CRC. For 13 of the regions, it was possible to refine the association signal identifying a smaller region of interest likely to harbour the functional variant. Our analysis did not provide evidence that any of the associations at the 16 loci being a consequence of synthetic associations rather than linkage disequilibrium with a common risk variant.


Subject(s)
Colorectal Neoplasms/genetics , Genetic Predisposition to Disease , Penetrance , Adult , Aged , Alleles , Case-Control Studies , Chromosome Mapping , Gene Frequency , Genetic Loci , Genome-Wide Association Study , Genotype , Humans , Middle Aged , Polymorphism, Single Nucleotide
20.
Nat Genet ; 45(10): 1221-1225, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23955597

ABSTRACT

To identify variants for multiple myeloma risk, we conducted a genome-wide association study with validation in additional series totaling 4,692 individuals with multiple myeloma (cases) and 10,990 controls. We identified four risk loci at 3q26.2 (rs10936599, P = 8.70 × 10(-14)), 6p21.33 (rs2285803, PSORS1C2, P = 9.67 × 10(-11)), 17p11.2 (rs4273077, TNFRSF13B, P = 7.67 × 10(-9)) and 22q13.1 (rs877529, CBX7, P = 7.63 × 10(-16)). These data provide further evidence for genetic susceptibility to this B-cell hematological malignancy, as well as insight into the biological basis of predisposition.


Subject(s)
Chromosome Aberrations , Chromosomes, Human , Genetic Predisposition to Disease , Multiple Myeloma/genetics , Case-Control Studies , Humans
SELECTION OF CITATIONS
SEARCH DETAIL