Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters

Publication year range
1.
Nat Genet ; 39(8): 984-8, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17618284

ABSTRACT

Much of the variation in inherited risk of colorectal cancer (CRC) is probably due to combinations of common low risk variants. We conducted a genome-wide association study of 550,000 tag SNPs in 930 familial colorectal tumor cases and 960 controls. The most strongly associated SNP (P = 1.72 x 10(-7), allelic test) was rs6983267 at 8q24.21. To validate this finding, we genotyped rs6983267 in three additional CRC case-control series (4,361 affected individuals and 3,752 controls; 1,901 affected individuals and 1,079 controls; 1,072 affected individuals and 415 controls) and replicated the association, providing P = 1.27 x 10(-14) (allelic test) overall, with odds ratios (ORs) of 1.27 (95% confidence interval (c.i.): 1.16-1.39) and 1.47 (95% c.i.: 1.34-1.62) for heterozygotes and rare homozygotes, respectively. Analyses based on 1,477 individuals with colorectal adenoma and 2,136 controls suggest that susceptibility to CRC is mediated through development of adenomas (OR = 1.21, 95% c.i.: 1.10-1.34; P = 6.89 x 10(-5)). These data show that common, low-penetrance susceptibility alleles predispose to colorectal neoplasia.


Subject(s)
Colorectal Neoplasms/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Aged , Chromosomes, Human, Pair 8 , Female , Genotype , Humans , Male , Middle Aged
2.
Genet Res (Camb) ; 97: e11, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25992589

ABSTRACT

BACKGROUND: A 40 kb ancestral germline duplication upstream of the GREM1 gene was reported in Ashkenazi families with hereditary mixed polyposis syndrome (HMPS). OBJECTIVE: Assess the contribution of the GREM1 mutation to familial colorectal cancer (CRC) in Ashkenazim. METHODS: Jewish Ashkenazi individuals (n = 472 155 males, 317 females) were genotyped for the GREM1 duplication, 194 with CRC, 131 had other cancer types (endometrial, pancreatic and ovarian) that show a syndromic association with CRC, and 147 were cancer-free with a suggestive family history of CRC. RESULTS: One mutation carrier was found who fulfills the Amsterdam criteria for Lynch Syndrome (LS). The prevalence of this mutation amongst LS Ashkenazim is 0·7%. CONCLUSION: If validated in additional studies it seems rational to recommend to look for the GREM1 founder mutation in Ashkenazi individuals with multiple colorectal polyps and/or fulfill the criteria for LS.


Subject(s)
Colorectal Neoplasms/genetics , Genetic Predisposition to Disease/genetics , Germ-Line Mutation , Intercellular Signaling Peptides and Proteins/genetics , Adult , Aged , Aged, 80 and over , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA Mutational Analysis , Family Health , Female , Genetic Testing , Humans , Jews/genetics , Male , Middle Aged , Young Adult
3.
PLoS Genet ; 7(6): e1002105, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21655089

ABSTRACT

Genome-wide association studies (GWAS) have identified 14 tagging single nucleotide polymorphisms (tagSNPs) that are associated with the risk of colorectal cancer (CRC), and several of these tagSNPs are near bone morphogenetic protein (BMP) pathway loci. The penalty of multiple testing implicit in GWAS increases the attraction of complementary approaches for disease gene discovery, including candidate gene- or pathway-based analyses. The strongest candidate loci for additional predisposition SNPs are arguably those already known both to have functional relevance and to be involved in disease risk. To investigate this proposition, we searched for novel CRC susceptibility variants close to the BMP pathway genes GREM1 (15q13.3), BMP4 (14q22.2), and BMP2 (20p12.3) using sample sets totalling 24,910 CRC cases and 26,275 controls. We identified new, independent CRC predisposition SNPs close to BMP4 (rs1957636, P = 3.93×10(-10)) and BMP2 (rs4813802, P = 4.65×10(-11)). Near GREM1, we found using fine-mapping that the previously-identified association between tagSNP rs4779584 and CRC actually resulted from two independent signals represented by rs16969681 (P = 5.33×10(-8)) and rs11632715 (P = 2.30×10(-10)). As low-penetrance predisposition variants become harder to identify-owing to small effect sizes and/or low risk allele frequencies-approaches based on informed candidate gene selection may become increasingly attractive. Our data emphasise that genetic fine-mapping studies can deconvolute associations that have arisen owing to independent correlation of a tagSNP with more than one functional SNP, thus explaining some of the apparently missing heritability of common diseases.


Subject(s)
Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 4/genetics , Colorectal Neoplasms/genetics , Genetic Predisposition to Disease , Intercellular Signaling Peptides and Proteins/genetics , Aged , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 4/metabolism , Case-Control Studies , Colorectal Neoplasms/metabolism , Gene Frequency , Genetic Variation , Genome-Wide Association Study , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Quantitative Trait, Heritable , Signal Transduction
4.
Article in English | MEDLINE | ID: mdl-38815859

ABSTRACT

BACKGROUND: Research has demonstrated an association between elevated systemic inflammation and changes in brain function. Affective areas of the brain involved in processing threat (e.g., amygdala) and reward (e.g., nucleus accumbens [NAcc]) appear to be sensitive to inflammation. Early life stress (ELS), such as experiencing low socioeconomic status (SES), may also potentiate this association, but relevant evidence has come primarily from cross-sectional studies of brain function. It is unclear whether similar associations are present between ELS, inflammation, and brain structure, particularly in typically developing populations. METHODS: We recruited and assessed 50 adolescents (31F/19M) from the community (M±SD age=15.5±1.1; range=13.1-17.5 years ) and in exploratory analyses examined whether changes in C-reactive protein (ΔCRP) from blood spots predict changes in gray matter volumes (ΔGMV) in the bilateral amygdala and NAcc over a two-year period. We also investigated whether experiencing ELS, operationalized using a comprehensive composite score of SES disadvantage at the family and neighborhood levels, significantly moderated the association between ΔCRP and ΔGMV. RESULTS: We found that ΔCRP was negatively associated with ΔAmygdala GMV (i.e. increasing CRP levels were associated with decreasing amygdala volume; ß=-0.84; p=0.012). This effect was stronger in youth who experienced greater SES disadvantage (ß=-0.56; p=0.025). CONCLUSIONS: These findings suggest that increases in systemic inflammation are associated with reductions in amygdala GMV in adolescents, potentially signaling accelerated maturation, and that these neuroimmune processes are compounded in adolescents who experienced greater SES disadvantage. Our findings are consistent with theoretical frameworks of neuroimmune associations and suggest they may influence adolescent neurodevelopment.

5.
Nat Genet ; 30(4): 406-10, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11865300

ABSTRACT

Uterine leiomyomata (fibroids) are common and clinically important tumors, but little is known about their etiology and pathogenesis. We previously mapped a gene that predisposes to multiple fibroids, cutaneous leiomyomata and renal cell carcinoma to chromosome 1q42.3-q43 (refs 4-6). Here we show, through a combination of mapping critical recombinants, identifying individuals with germline mutations and screening known and predicted transcripts, that this gene encodes fumarate hydratase, an enzyme of the tricarboxylic acid cycle. Leiomyomatosis-associated mutations are predicted to result in absent or truncated protein, or substitutions or deletions of highly conserved amino acids. Activity of fumarate hydratase is reduced in lymphoblastoid cells from individuals with leiomyomatosis. This enzyme acts as a tumor suppressor in familial leiomyomata, and its measured activity is very low or absent in tumors from individuals with leiomyomatosis. Mutations in FH also occur in the recessive condition fumarate hydratase deficiency, and some parents of people with this condition are susceptible to leiomyomata. Thus, heterozygous and homozygous or compound heterozygous mutants have very different clinical phenotypes. Our results provide clues to the pathogenesis of fibroids and emphasize the importance of mutations of housekeeping and mitochondrial proteins in the pathogenesis of common types of tumor.


Subject(s)
Carcinoma, Papillary/genetics , Carcinoma, Renal Cell/genetics , Fumarate Hydratase/genetics , Germ-Line Mutation , Kidney Neoplasms/genetics , Leiomyoma, Epithelioid/genetics , Leiomyoma/genetics , Uterine Neoplasms/genetics , Alleles , Chromosomes, Human, Pair 1 , Exons , Female , Fumarate Hydratase/metabolism , Gene Deletion , Genes, Dominant , Genetic Markers , Humans , In Situ Hybridization, Fluorescence , Male , Mutation , Pedigree , Recombination, Genetic , Sequence Analysis, DNA
6.
MicroPubl Biol ; 20232023.
Article in English | MEDLINE | ID: mdl-36824381

ABSTRACT

Tadpoles display preferences for different environments but the sensory modalities that govern these choices are not well understood. Here, we examined light preferences and associated sensory mechanisms of albino and wild-type Xenopus laevis tadpoles. We found that albino tadpoles spent more time in darker environments compared to the wild type, although they showed no differences in overall activity. This preference persisted when the tadpoles had their optic nerve severed or pineal glands removed, suggesting these sensory systems alone are not necessary for phototaxis. These experiments were conducted by an undergraduate laboratory course, highlighting how X. laevis tadpole behavior assays in a classroom setting can reveal new insights into animal behavior.

7.
MicroPubl Biol ; 20232023.
Article in English | MEDLINE | ID: mdl-37008729

ABSTRACT

Many ant species are equipped with chemical defenses, although how these compounds impact nervous system function is unclear. Here, we examined the utility of Caenorhabditis elegans chemotaxis assays for investigating how ant chemical defense compounds are detected by heterospecific nervous systems. We found that C. elegans respond to extracts from the invasive Argentine Ant ( Linepithema humile ) and the osm-9 ion channel is required for this response. Divergent strains varied in their response to L. humile extracts, suggesting genetic variation underlying chemotactic responses. These experiments were conducted by an undergraduate laboratory course, highlighting how C. elegans chemotaxis assays in a classroom setting can provide genuine research experiences and reveal new insights into interspecies interactions.

8.
Hum Mol Genet ; 17(23): 3720-7, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-18753146

ABSTRACT

The common single-nucleotide polymorphism (SNP) rs3802842 at 11q23.1 has recently been reported to be associated with risk of colorectal cancer (CRC). To examine this association in detail we genotyped rs3802842 in eight independent case-control series comprising a total of 10 638 cases and 10 457 healthy individuals. A significant association between the C allele of rs3802842 and CRC risk was found (per allele OR = 1.17; 95% confidence interval [CI]: 1.12-1.22; P = 1.08 x 10(-12)) with the risk allele more frequent in rectal than colonic disease (P = 0.02). In combination with 8q21, 8q24, 10p14, 11q, 15q13.3 and 18q21 variants, the risk of CRC increases with an increasing numbers of variant alleles for the six loci (OR(per allele) = 1.19; 95% CI: 1.15-1.23; P(trend) = 7.4 x 10(-24)). Using the data from our genome-wide association study of CRC, LD mapping and imputation, we were able to refine the location of the causal locus to a 60 kb region and screened for coding changes. The absence of exonic mutations in any of the transcripts (FLJ45803, LOC120376, C11orf53 and POU2AF1) mapping to this region makes the association likely to be a consequence of non-coding effects on gene expression.


Subject(s)
Chromosomes, Human, Pair 11/genetics , Colorectal Neoplasms/genetics , Genetic Predisposition to Disease , Genetic Variation , Adult , Aged , Case-Control Studies , Female , Genotype , Humans , Male , Middle Aged , Mutation , Polymorphism, Single Nucleotide , Risk Factors
9.
J Natl Cancer Inst ; 111(8): 828-836, 2019 08 01.
Article in English | MEDLINE | ID: mdl-30649440

ABSTRACT

BACKGROUND: Constitutional loss of function (LOF) single nucleotide polymorphisms (SNPs) in pattern recognition receptors FPR1, TLR3, and TLR4 have previously been reported to predict oxaliplatin benefit in colorectal cancer. Confirmation of this association could substantially improve patient stratification. METHODS: We performed a retrospective biomarker analysis of the Short Course in Oncology Therapy (SCOT) and COIN/COIN-B trials. Participant status for LOF variants in FPR1 (rs867228), TLR3 (rs3775291), and TLR4 (rs4986790/rs4986791) was determined by genotyping array or genotype imputation. Associations between LOF variants and disease-free survival (DFS) and overall survival (OS) were analyzed by Cox regression, adjusted for confounders, using additive, dominant, and recessive genetic models. All statistical tests were two-sided. RESULTS: Our validation study populations included 2929 and 1948 patients in the SCOT and COIN/COIN-B cohorts, respectively, of whom 2728 and 1672 patients had functional status of all three SNPs determined. We found no evidence of an association between any SNP and DFS in the SCOT cohort, or with OS in either cohort, irrespective of the type of model used. This included models for which an association was previously reported for rs867228 (recessive model, multivariable-adjusted hazard ratio [HR] for DFS in SCOT = 1.19, 95% confidence interval [CI] = 0.99 to 1.45, P = .07; HR for OS in COIN/COIN-B = 0.92, 95% CI = 0.63 to 1.34, P = .66), and rs4986790 (dominant model, multivariable-adjusted HR for DFS in SCOT = 0.86, 95% CI = 0.65 to 1.13, P = .27; HR for OS in COIN/COIN-B = 1.08, 95% CI = 0.90 to 1.31, P = .40). CONCLUSION: In this prespecified analysis of two large clinical trials, we found no evidence that constitutional LOF SNPs in FPR1, TLR3, or TLR4 are associated with differential benefit from oxaliplatin. Our results suggest these SNPs are unlikely to be clinically useful biomarkers.


Subject(s)
Colorectal Neoplasms/drug therapy , Receptors, Formyl Peptide/genetics , Toll-Like Receptor 3/genetics , Toll-Like Receptor 4/genetics , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease-Free Survival , Female , Genotype , Humans , Male , Middle Aged , Oxaliplatin/adverse effects , Oxaliplatin/therapeutic use , Polymorphism, Genetic , Polymorphism, Single Nucleotide/genetics , Receptors, Pattern Recognition/genetics
10.
Nat Commun ; 10(1): 2154, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31089142

ABSTRACT

Colorectal cancer (CRC) is a leading cause of cancer-related death worldwide, and has a strong heritable basis. We report a genome-wide association analysis of 34,627 CRC cases and 71,379 controls of European ancestry that identifies SNPs at 31 new CRC risk loci. We also identify eight independent risk SNPs at the new and previously reported European CRC loci, and a further nine CRC SNPs at loci previously only identified in Asian populations. We use in situ promoter capture Hi-C (CHi-C), gene expression, and in silico annotation methods to identify likely target genes of CRC SNPs. Whilst these new SNP associations implicate target genes that are enriched for known CRC pathways such as Wnt and BMP, they also highlight novel pathways with no prior links to colorectal tumourigenesis. These findings provide further insight into CRC susceptibility and enhance the prospects of applying genetic risk scores to personalised screening and prevention.


Subject(s)
Colorectal Neoplasms/genetics , Genetic Loci , Genetic Predisposition to Disease , Asian People/genetics , Case-Control Studies , Colorectal Neoplasms/pathology , Datasets as Topic , Female , Genome-Wide Association Study , Humans , Inheritance Patterns , Male , Middle Aged , Polymorphism, Single Nucleotide , Risk Factors , White People/genetics
11.
Eur J Hum Genet ; 16(12): 1477-86, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18628789

ABSTRACT

Previously we have localized to chromosome 3q21-q24, a predisposition locus for colorectal cancer (CRC), through a genome-wide linkage screen (GWLS) of 69 families without familial adenomatous polyposis or hereditary non-polyposis CRC. To further investigate Mendelian susceptibility to CRC, we extended our screen to include a further GWLS of an additional 34 CRC families. We also searched for a disease gene at 3q21-q24 by linkage disequilibrium mapping in 620 familial CRC cases and 960 controls by genotyping 1676 tagging SNPs and sequencing 30 candidate genes from the region. Linkage analysis was conducted using the Affymetrix 10K SNP array. Data from both GWLSs were pooled and multipoint linkage statistics computed. The maximum NPL score (3.01; P=0.0013) across all families was at 3q22, maximal evidence for linkage coming from families segregating rectal CRC. The same genomic position also yielded the highest multipoint heterogeneity LOD (HLOD) score under a dominant model (HLOD=2.79; P=0.00034), with an estimated 43% of families linked. In the case-control analysis, the strongest association was obtained at rs698675 (P=0.0029), but this was not significant after adjusting for multiple testing. Analysis of candidate gene mapping to the region of maximal linkage on 3q22 failed to identify a causal mutation. There was no evidence for linkage to the previously reported 9q CRC locus (NPL=0.95, P=0.23; HLOD(dominant)=0.40, HLOD(recessive)=0.20). Our findings are consistent with the hypothesis that variation at 3q22 contributes to the risk of CRC, but this is unlikely to be mediated through a restricted set of alleles.


Subject(s)
Adenoma/genetics , Colorectal Neoplasms/genetics , Family , Genetic Linkage , Case-Control Studies , Chromosome Mapping , Chromosomes, Human, Pair 3 , DNA Mutational Analysis , Family Health , Gene Dosage , Genome-Wide Association Study , Humans , Lod Score , Polymorphism, Single Nucleotide
12.
Cancer Res ; 66(10): 5003-6, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16707420

ABSTRACT

About 30% of all colorectal cancers are thought to have a genetic basis and the known predisposing genes can only account for a small fraction of cases. A previous report suggested that a colorectal cancer candidate gene, explaining at least 20% of colorectal cancer cases with family history, was located within a 25 cM region on chromosome 9q22.2-q31.3. We typed 16 polymorphic markers encompassing the region of putative linkage in 57 colorectal tumor families from the United Kingdom. Known Mendelian syndromes had been excluded. We found suggestive evidence of linkage, as positive parametric (HLOD = 1.23) and nonparametric (NPL = 1.21, P = 0.11) LOD scores were obtained by analysis of the whole family set. Enrichment for cases with a priori genetic etiology by analyzing families with at least one person affected at <45 years of age (n = 39 families) gave a maximum multipoint NPL score of 2.65 (P = 0.007). In this group, significant NPL scores >1.67 (P < 0.05) were found in a 6.5 cM region between D9S1851 and D9S277. With a more stringent threshold (NPL>2.4, P < 0.01), the linked region was 1.7 cM between D9S971 and D9S272/D9S173. Exclusion from the analysis of kindreds with a phenotype of multiple polyposis also found evidence of linkage in the same region (NPL = 2.47 at close to D9S277, P = 0.009). The type I transforming growth factor-beta receptor, a prime candidate gene, was excluded as a cause of disease. The results presented here further support the existence of a colorectal cancer susceptibility gene on chromosome 9q and refine its likely location.


Subject(s)
Chromosomes, Human, Pair 9/genetics , Colorectal Neoplasms/genetics , Genetic Linkage , Adult , Genetic Predisposition to Disease , Humans , Middle Aged , United Kingdom
13.
J Clin Endocrinol Metab ; 10(3): 1098-1103, 2016 03 01.
Article in English | MEDLINE | ID: mdl-26691890

ABSTRACT

CONTEXT: A recent study reported the non-synonymous G534E (rs7080536, allele A) variant in the HABP2 gene as causal in familial non-medullary thyroid cancer (NMTC). OBJECTIVE: The objective of this study was to evaluate the causality of HABP2 G534E in the TCUKIN study, a multi-center population based study of NMTC cases from the British Isles. DESIGN AND SETTING: A case-control analysis of rs7080536 genotypes was performed using 2,105 TCUKIN cases and 5,172 UK controls. PARTICIPANTS: Cases comprised 2,105 NMTC cases. Patients sub-groups with papillary (N=1,056), follicular (N=691) and Hurthle cell (N=86) TC cases were studied separately. Controls comprised 5,172 individuals from the 1958 Birth Cohort (58C) and the National Blood Donor Service (NBS) study. The controls had previously been genotyped using genome-wide SNP arrays by the Wellcome Trust Case Control Consortium study. OUTCOME: Measures: Association between HABP2 G534E (rs7080536A) and NMTC risk was evaluated using logistic regression. RESULTS: The frequency of HABP2 G534E was 4.2% in cases and 4.6% in controls. We did not detect an association between this variant and NMTC risk (OR=0.896, 95% CI: 0.746-1.071, P=0.233). We also failed to detect an association between HABP2 G534E and cases with papillary (1056 cases, G534E frequency= 3.5%, OR=0.74, P=0.017), follicular (691 cases, G534E frequency= 4.7%, OR=1.00, P=1.000) or Hurthle cell (86 cases, G534E frequency= 6.3%, OR=1.40, P=0.279) histology. CONCLUSIONS: We found that HABP2 G534E is a low-to-moderate frequency variant in the British Isles and failed to detect an association with NMTC risk, independent of histological type. Hence, our study does not implicate HABP2 G534E or a correlated polymorphism in familial NMTC and additional data are required before using this variant in NMTC risk assessment.

14.
Nat Med ; 21(1): 62-70, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25419707

ABSTRACT

Hereditary mixed polyposis syndrome (HMPS) is characterized by the development of mixed-morphology colorectal tumors and is caused by a 40-kb genetic duplication that results in aberrant epithelial expression of the gene encoding mesenchymal bone morphogenetic protein antagonist, GREM1. Here we use HMPS tissue and a mouse model of the disease to show that epithelial GREM1 disrupts homeostatic intestinal morphogen gradients, altering cell fate that is normally determined by position along the vertical epithelial axis. This promotes the persistence and/or reacquisition of stem cell properties in Lgr5-negative progenitor cells that have exited the stem cell niche. These cells form ectopic crypts, proliferate, accumulate somatic mutations and can initiate intestinal neoplasia, indicating that the crypt base stem cell is not the sole cell of origin of colorectal cancer. Furthermore, we show that epithelial expression of GREM1 also occurs in traditional serrated adenomas, sporadic premalignant lesions with a hitherto unknown pathogenesis, and these lesions can be considered the sporadic equivalents of HMPS polyps.


Subject(s)
Carcinogenesis/genetics , Colorectal Neoplasms/genetics , Intercellular Signaling Peptides and Proteins/biosynthesis , Stem Cell Niche/genetics , Animals , Cell Proliferation/genetics , Colorectal Neoplasms/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation, Neoplastic , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mutation , Receptors, G-Protein-Coupled/genetics
15.
Nat Commun ; 6: 6336, 2015 Mar 19.
Article in English | MEDLINE | ID: mdl-25790038

ABSTRACT

Papillary renal cell carcinoma (pRCC) is an important subtype of kidney cancer with a problematic pathological classification and highly variable clinical behaviour. Here we sequence the genomes or exomes of 31 pRCCs, and in four tumours, multi-region sequencing is undertaken. We identify BAP1, SETD2, ARID2 and Nrf2 pathway genes (KEAP1, NHE2L2 and CUL3) as probable drivers, together with at least eight other possible drivers. However, only ~10% of tumours harbour detectable pathogenic changes in any one driver gene, and where present, the mutations are often predicted to be present within cancer sub-clones. We specifically detect parallel evolution of multiple SETD2 mutations within different sub-regions of the same tumour. By contrast, large copy number gains of chromosomes 7, 12, 16 and 17 are usually early, monoclonal changes in pRCC evolution. The predominance of large copy number variants as the major drivers for pRCC highlights an unusual mode of tumorigenesis that may challenge precision medicine approaches.


Subject(s)
Carcinoma, Renal Cell/genetics , Chromosomes/ultrastructure , Kidney Neoplasms/genetics , Mutation , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/chemistry , Chromosome Mapping , DNA Copy Number Variations , Exome , Exons , Female , Gene Expression Regulation, Neoplastic , Histone-Lysine N-Methyltransferase/genetics , Humans , Loss of Heterozygosity , Male , Middle Aged , Phylogeny , Polymorphism, Single Nucleotide , Sequence Analysis, DNA
16.
Cell Rep ; 8(4): 983-90, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-25131200

ABSTRACT

A rare germline duplication upstream of the bone morphogenetic protein antagonist GREM1 causes a Mendelian-dominant predisposition to colorectal cancer (CRC). The underlying disease mechanism is strong, ectopic GREM1 overexpression in the intestinal epithelium. Here, we confirm that a common GREM1 polymorphism, rs16969681, is also associated with CRC susceptibility, conferring ∼20% differential risk in the general population. We hypothesized the underlying cause to be moderate differences in GREM1 expression. We showed that rs16969681 lies in a region of active chromatin with allele- and tissue-specific enhancer activity. The CRC high-risk allele was associated with stronger gene expression, and higher Grem1 mRNA levels increased the intestinal tumor burden in Apc(Min) mice. The intestine-specific transcription factor CDX2 and Wnt effector TCF7L2 bound near rs16969681, with significantly higher affinity for the risk allele, and CDX2 overexpression in CDX2/GREM1-negative cells caused re-expression of GREM1. rs16969681 influences CRC risk through effects on Wnt-driven GREM1 expression in colorectal tumors.


Subject(s)
Colonic Neoplasms/genetics , Homeodomain Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Transcription Factor 7-Like 2 Protein/metabolism , Animals , Base Sequence , CDX2 Transcription Factor , Cell Line, Tumor , Enhancer Elements, Genetic , Gene Expression Regulation, Neoplastic , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Mice, Transgenic , Organ Specificity , Polymorphism, Single Nucleotide , Risk
17.
Nat Genet ; 44(6): 699-703, 2012 May 06.
Article in English | MEDLINE | ID: mdl-22561515

ABSTRACT

Hereditary mixed polyposis syndrome (HMPS) is characterized by apparent autosomal dominant inheritance of multiple types of colorectal polyp, with colorectal carcinoma occurring in a high proportion of affected individuals. Here, we use genetic mapping, copy-number analysis, exclusion of mutations by high-throughput sequencing, gene expression analysis and functional assays to show that HMPS is caused by a duplication spanning the 3' end of the SCG5 gene and a region upstream of the GREM1 locus. This unusual mutation is associated with increased allele-specific GREM1 expression. Whereas GREM1 is expressed in intestinal subepithelial myofibroblasts in controls, GREM1 is predominantly expressed in the epithelium of the large bowel in individuals with HMPS. The HMPS duplication contains predicted enhancer elements; some of these interact with the GREM1 promoter and can drive gene expression in vitro. Increased GREM1 expression is predicted to cause reduced bone morphogenetic protein (BMP) pathway activity, a mechanism that also underlies tumorigenesis in juvenile polyposis of the large bowel.


Subject(s)
Colonic Polyps/genetics , Gene Duplication , Intercellular Signaling Peptides and Proteins/genetics , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Morphogenetic Proteins/genetics , Chromosome Mapping , Chromosomes, Human, Pair 15/genetics , Colorectal Neoplasms/genetics , DNA Copy Number Variations , Fetal Proteins , Formins , Gene Expression , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Microfilament Proteins , Neuroendocrine Secretory Protein 7B2 , Nuclear Proteins , Receptors, Immunologic , Signaling Lymphocytic Activation Molecule Family , Syndrome
18.
Nat Genet ; 42(11): 973-7, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20972440

ABSTRACT

Genome-wide association studies (GWAS) have identified ten loci harboring common variants that influence risk of developing colorectal cancer (CRC). To enhance the power to identify additional CRC risk loci, we conducted a meta-analysis of three GWAS from the UK which included a total of 3,334 affected individuals (cases) and 4,628 controls followed by multiple validation analyses including a total of 18,095 cases and 20,197 controls. We identified associations at four new CRC risk loci: 1q41 (rs6691170, odds ratio (OR) = 1.06, P = 9.55 × 10⁻¹° and rs6687758, OR = 1.09, P = 2.27 × 10⁻9, 3q26.2 (rs10936599, OR = 0.93, P = 3.39 × 10⁻8), 12q13.13 (rs11169552, OR = 0.92, P = 1.89 × 10⁻¹° and rs7136702, OR = 1.06, P = 4.02 × 10⁻8) and 20q13.33 (rs4925386, OR = 0.93, P = 1.89 × 10⁻¹°). In addition to identifying new CRC risk loci, this analysis provides evidence that additional CRC-associated variants of similar effect size remain to be discovered.


Subject(s)
Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 13/genetics , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 3/genetics , Colorectal Neoplasms/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Chromosome Mapping/methods , Female , Genome-Wide Association Study/methods , Genotype , Humans , Male , Meta-Analysis as Topic , Odds Ratio , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Risk Assessment
20.
Nat Genet ; 40(1): 26-8, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18084292

ABSTRACT

We mapped a high-penetrance gene (CRAC1; also known as HMPS) associated with colorectal cancer (CRC) in the Ashkenazi population to a 0.6-Mb region on chromosome 15 containing SCG5 (also known as SGNE1), GREM1 and FMN1. We hypothesized that the CRAC1 locus harbored low-penetrance variants that increased CRC risk in the general population. In a large series of colorectal cancer cases and controls, SNPs near GREM1 and SCG5 were strongly associated with increased CRC risk (for rs4779584, P = 4.44 x 10(-14)).


Subject(s)
Colorectal Neoplasms/genetics , Genetic Predisposition to Disease , Adenoma/genetics , Cell Line, Tumor , Cells, Cultured , Chromosomes, Human, Pair 15 , Humans , Jews/genetics , Polymorphism, Single Nucleotide
SELECTION OF CITATIONS
SEARCH DETAIL