Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Mol Cancer ; 23(1): 101, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38745269

RESUMEN

BACKGROUND: Long noncoding RNAs (lncRNAs) have surpassed the number of protein-coding genes, yet the majority have no known function. We previously discovered 844 lncRNAs that were genetically linked to breast cancer through genome-wide association studies (GWAS). Here, we show that a subset of these lncRNAs alter breast cancer risk by modulating cell proliferation, and provide evidence that a reduced expression on one lncRNA increases breast cancer risk through aberrant DNA replication and repair. METHODS: We performed pooled CRISPR-Cas13d-based knockdown screens in breast cells to identify which of the 844 breast cancer-associated lncRNAs alter cell proliferation. We selected one of the lncRNAs that increased cell proliferation, KILR, for follow-up functional studies. KILR pull-down followed by mass spectrometry was used to identify binding proteins. Knockdown and overexpression studies were performed to assess the mechanism by which KILR regulates proliferation. RESULTS: We show that KILR functions as a tumor suppressor, safeguarding breast cells against uncontrolled proliferation. The half-life of KILR is significantly reduced by the risk haplotype, revealing an alternative mechanism by which variants alter cancer risk. Mechanistically, KILR sequesters RPA1, a subunit of the RPA complex required for DNA replication and repair. Reduced KILR expression promotes breast cancer cell proliferation by increasing the available pool of RPA1 and speed of DNA replication. Conversely, KILR overexpression promotes apoptosis in breast cancer cells, but not normal breast cells. CONCLUSIONS: Our results confirm lncRNAs as mediators of breast cancer risk, emphasize the need to annotate noncoding transcripts in relevant cell types when investigating GWAS variants and provide a scalable platform for mapping phenotypes associated with lncRNAs.


Asunto(s)
Neoplasias de la Mama , Sistemas CRISPR-Cas , Proliferación Celular , Reparación del ADN , Replicación del ADN , ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Femenino , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo
2.
Genome Biol ; 24(1): 59, 2023 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-36991492

RESUMEN

BACKGROUND: Genome-wide association studies (GWAS) have identified > 200 loci associated with breast cancer risk. The majority of candidate causal variants are in non-coding regions and likely modulate cancer risk by regulating gene expression. However, pinpointing the exact target of the association, and identifying the phenotype it mediates, is a major challenge in the interpretation and translation of GWAS. RESULTS: Here, we show that pooled CRISPR screens are highly effective at identifying GWAS target genes and defining the cancer phenotypes they mediate. Following CRISPR mediated gene activation or suppression, we measure proliferation in 2D, 3D, and in immune-deficient mice, as well as the effect on DNA repair. We perform 60 CRISPR screens and identify 20 genes predicted with high confidence to be GWAS targets that promote cancer by driving proliferation or modulating the DNA damage response in breast cells. We validate the regulation of a subset of these genes by breast cancer risk variants. CONCLUSIONS: We demonstrate that phenotypic CRISPR screens can accurately pinpoint the gene target of a risk locus. In addition to defining gene targets of risk loci associated with increased breast cancer risk, we provide a platform for identifying gene targets and phenotypes mediated by risk variants.


Asunto(s)
Estudio de Asociación del Genoma Completo , Neoplasias , Animales , Ratones , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Predisposición Genética a la Enfermedad , Fenotipo , Polimorfismo de Nucleótido Simple
3.
Am J Hum Genet ; 107(4): 778-787, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32871102

RESUMEN

Breast cancer genome-wide association studies (GWASs) have identified 150 genomic risk regions containing more than 13,000 credible causal variants (CCVs). The CCVs are predominantly noncoding and enriched in regulatory elements. However, the genes underlying breast cancer risk associations are largely unknown. Here, we used genetic colocalization analysis to identify loci at which gene expression could potentially explain breast cancer risk phenotypes. Using data from the Breast Cancer Association Consortium (BCAC) and quantitative trait loci (QTL) from the Genotype-Tissue Expression (GTEx) project and The Cancer Genome Project (TCGA), we identify shared genetic relationships and reveal novel associations between cancer phenotypes and effector genes. Seventeen genes, including NTN4, were identified as potential mediators of breast cancer risk. For NTN4, we showed the rs61938093 CCV at this region was located within an enhancer element that physically interacts with the NTN4 promoter, and the risk allele reduced NTN4 promoter activity. Furthermore, knockdown of NTN4 in breast cells increased cell proliferation in vitro and tumor growth in vivo. These data provide evidence linking risk-associated variation to genes that may contribute to breast cancer predisposition.


Asunto(s)
Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Proteínas de Neoplasias/genética , Netrinas/genética , Alelos , Animales , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Elementos de Facilitación Genéticos , Femenino , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Genómica/métodos , Xenoinjertos , Humanos , Células MCF-7 , Ratones , Ratones Desnudos , Proteínas de Neoplasias/metabolismo , Netrinas/metabolismo , Fenotipo , Sitios de Carácter Cuantitativo , Riesgo
4.
Genome Biol ; 21(1): 8, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31910858

RESUMEN

BACKGROUND: Genome-wide association studies have identified 196 high confidence independent signals associated with breast cancer susceptibility. Variants within these signals frequently fall in distal regulatory DNA elements that control gene expression. RESULTS: We designed a Capture Hi-C array to enrich for chromatin interactions between the credible causal variants and target genes in six human mammary epithelial and breast cancer cell lines. We show that interacting regions are enriched for open chromatin, histone marks for active enhancers, and transcription factors relevant to breast biology. We exploit this comprehensive resource to identify candidate target genes at 139 independent breast cancer risk signals and explore the functional mechanism underlying altered risk at the 12q24 risk region. CONCLUSIONS: Our results demonstrate the power of combining genetics, computational genomics, and molecular studies to rationalize the identification of key variants and candidate target genes at breast cancer GWAS signals.


Asunto(s)
Neoplasias de la Mama/genética , Cromatina/metabolismo , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Genoma Humano , Estudio de Asociación del Genoma Completo , Humanos
5.
Genome Biol ; 21(1): 7, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31910864

RESUMEN

BACKGROUND: Genetic variants identified through genome-wide association studies (GWAS) are predominantly non-coding and typically attributed to altered regulatory elements such as enhancers and promoters. However, the contribution of non-coding RNAs to complex traits is not clear. RESULTS: Using targeted RNA sequencing, we systematically annotated multi-exonic non-coding RNA (mencRNA) genes transcribed from 1.5-Mb intervals surrounding 139 breast cancer GWAS signals and assessed their contribution to breast cancer risk. We identify more than 4000 mencRNA genes and show their expression distinguishes normal breast tissue from tumors and different breast cancer subtypes. Importantly, breast cancer risk variants, identified through genetic fine-mapping, are significantly enriched in mencRNA exons, but not the promoters or introns. eQTL analyses identify mencRNAs whose expression is associated with risk variants. Furthermore, chromatin interaction data identify hundreds of mencRNA promoters that loop to regions that contain breast cancer risk variants. CONCLUSIONS: We have compiled the largest catalog of breast cancer-associated mencRNAs to date and provide evidence that modulation of mencRNAs by GWAS variants may provide an alternative mechanism underlying complex traits.


Asunto(s)
Neoplasias de la Mama/genética , ARN no Traducido/genética , Estudio de Asociación del Genoma Completo , Humanos , Análisis de Secuencia de ARN
7.
Am J Hum Genet ; 101(2): 255-266, 2017 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-28777932

RESUMEN

Breast cancer risk is strongly associated with an intergenic region on 11q13. We have previously shown that the strongest risk-associated SNPs fall within a distal enhancer that regulates CCND1. Here, we report that, in addition to regulating CCND1, this enhancer regulates two estrogen-regulated long noncoding RNAs, CUPID1 and CUPID2. We provide evidence that the risk-associated SNPs are associated with reduced chromatin looping between the enhancer and the CUPID1 and CUPID2 bidirectional promoter. We further show that CUPID1 and CUPID2 are predominantly expressed in hormone-receptor-positive breast tumors and play a role in modulating pathway choice for the repair of double-strand breaks. These data reveal a mechanism for the involvement of this region in breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Cromosomas Humanos Par 11/genética , Ciclina D1/genética , Reparación del ADN/genética , ARN Largo no Codificante/genética , Línea Celular Tumoral , Cromatina/metabolismo , Roturas del ADN de Doble Cadena , Daño del ADN/genética , Elementos de Facilitación Genéticos/genética , Estrógenos/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad/genética , Humanos , Células MCF-7 , Polimorfismo de Nucleótido Simple/genética , Regiones Promotoras Genéticas/genética , Interferencia de ARN , ARN Guía de Kinetoplastida/genética , ARN Interferente Pequeño/genética
8.
Am J Hum Genet ; 99(4): 903-911, 2016 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-27640304

RESUMEN

Genome-wide association studies (GWASs) have revealed increased breast cancer risk associated with multiple genetic variants at 5p12. Here, we report the fine mapping of this locus using data from 104,660 subjects from 50 case-control studies in the Breast Cancer Association Consortium (BCAC). With data for 3,365 genotyped and imputed SNPs across a 1 Mb region (positions 44,394,495-45,364,167; NCBI build 37), we found evidence for at least three independent signals: the strongest signal, consisting of a single SNP rs10941679, was associated with risk of estrogen-receptor-positive (ER+) breast cancer (per-g allele OR ER+ = 1.15; 95% CI 1.13-1.18; p = 8.35 × 10-30). After adjustment for rs10941679, we detected signal 2, consisting of 38 SNPs more strongly associated with ER-negative (ER-) breast cancer (lead SNP rs6864776: per-a allele OR ER- = 1.10; 95% CI 1.05-1.14; p conditional = 1.44 × 10-12), and a single signal 3 SNP (rs200229088: per-t allele OR ER+ = 1.12; 95% CI 1.09-1.15; p conditional = 1.12 × 10-05). Expression quantitative trait locus analysis in normal breast tissues and breast tumors showed that the g (risk) allele of rs10941679 was associated with increased expression of FGF10 and MRPS30. Functional assays demonstrated that SNP rs10941679 maps to an enhancer element that physically interacts with the FGF10 and MRPS30 promoter regions in breast cancer cell lines. FGF10 is an oncogene that binds to FGFR2 and is overexpressed in ∼10% of human breast cancers, whereas MRPS30 plays a key role in apoptosis. These data suggest that the strongest signal of association at 5p12 is mediated through coordinated activation of FGF10 and MRPS30, two candidate genes for breast cancer pathogenesis.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Cromosomas Humanos Par 5/genética , Factor 10 de Crecimiento de Fibroblastos/genética , Predisposición Genética a la Enfermedad/genética , Polimorfismo de Nucleótido Simple/genética , Receptores de Estrógenos/metabolismo , Alelos , Estudios de Casos y Controles , Línea Celular Tumoral , Elementos de Facilitación Genéticos/genética , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Haplotipos/genética , Humanos , Regiones Promotoras Genéticas/genética , Sitios de Carácter Cuantitativo/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo
9.
Am J Hum Genet ; 98(6): 1159-1169, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-27259051

RESUMEN

A recent meta-analysis of multiple genome-wide association and follow-up endometrial cancer case-control datasets identified a novel genetic risk locus for this disease at chromosome 14q32.33. To prioritize the functional SNP(s) and target gene(s) at this locus, we employed an in silico fine-mapping approach using genotyped and imputed SNP data for 6,608 endometrial cancer cases and 37,925 controls of European ancestry. Association and functional analyses provide evidence that the best candidate causal SNP is rs2494737. Multiple experimental analyses show that SNP rs2494737 maps to a silencer element located within AKT1, a member of the PI3K/AKT/MTOR intracellular signaling pathway activated in endometrial tumors. The rs2494737 risk A allele creates a YY1 transcription factor-binding site and abrogates the silencer activity in luciferase assays, an effect mimicked by transfection of YY1 siRNA. Our findings suggest YY1 is a positive regulator of AKT1, mediating the stimulatory effects of rs2494737 increasing endometrial cancer risk. Identification of an endometrial cancer risk allele within a member of the PI3K/AKT signaling pathway, more commonly activated in tumors by somatic alterations, raises the possibility that well tolerated inhibitors targeting this pathway could be candidates for evaluation as chemopreventive agents in individuals at high risk of developing endometrial cancer.


Asunto(s)
Cromosomas Humanos Par 14/genética , Neoplasias Endometriales/genética , Fosfatidilinositol 3-Quinasas/genética , Polimorfismo de Nucleótido Simple/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias Uterinas/genética , Factor de Transcripción YY1/metabolismo , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/patología , Femenino , Sitios Genéticos , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/genética , Factores de Riesgo , Transducción de Señal , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patología , Factor de Transcripción YY1/genética
10.
Nat Genet ; 48(4): 374-86, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26928228

RESUMEN

We analyzed 3,872 common genetic variants across the ESR1 locus (encoding estrogen receptor α) in 118,816 subjects from three international consortia. We found evidence for at least five independent causal variants, each associated with different phenotype sets, including estrogen receptor (ER(+) or ER(-)) and human ERBB2 (HER2(+) or HER2(-)) tumor subtypes, mammographic density and tumor grade. The best candidate causal variants for ER(-) tumors lie in four separate enhancer elements, and their risk alleles reduce expression of ESR1, RMND1 and CCDC170, whereas the risk alleles of the strongest candidates for the remaining independent causal variant disrupt a silencer element and putatively increase ESR1 and RMND1 expression.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas Portadoras/genética , Proteínas de Ciclo Celular/genética , Cromosomas Humanos Par 6/genética , Receptor alfa de Estrógeno/genética , Secuencia de Bases , Neoplasias de la Mama/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Receptor alfa de Estrógeno/metabolismo , Femenino , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Fenotipo , Polimorfismo de Nucleótido Simple , Unión Proteica , Factores de Riesgo
11.
Oncotarget ; 7(6): 6353-68, 2016 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-26840454

RESUMEN

Women with epithelial ovarian cancer (EOC) are usually treated with platinum/taxane therapy after cytoreductive surgery but there is considerable inter-individual variation in response. To identify germline single-nucleotide polymorphisms (SNPs) that contribute to variations in individual responses to chemotherapy, we carried out a multi-phase genome-wide association study (GWAS) in 1,244 women diagnosed with serous EOC who were treated with the same first-line chemotherapy, carboplatin and paclitaxel. We identified two SNPs (rs7874043 and rs72700653) in TTC39B (best P=7x10-5, HR=1.90, for rs7874043) associated with progression-free survival (PFS). Functional analyses show that both SNPs lie in a putative regulatory element (PRE) that physically interacts with the promoters of PSIP1, CCDC171 and an alternative promoter of TTC39B. The C allele of rs7874043 is associated with poor PFS and showed increased binding of the Sp1 transcription factor, which is critical for chromatin interactions with PSIP1. Silencing of PSIP1 significantly impaired DNA damage-induced Rad51 nuclear foci and reduced cell viability in ovarian cancer lines. PSIP1 (PC4 and SFRS1 Interacting Protein 1) is known to protect cells from stress-induced apoptosis, and high expression is associated with poor PFS in EOC patients. We therefore suggest that the minor allele of rs7874043 confers poor PFS by increasing PSIP1 expression.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Elementos de Facilitación Genéticos/genética , Neoplasias de las Trompas Uterinas/mortalidad , Mutación de Línea Germinal/genética , Neoplasias Ováricas/mortalidad , Neoplasias Peritoneales/mortalidad , Polimorfismo de Nucleótido Simple/genética , Factores de Transcripción/genética , Apoptosis , Biomarcadores de Tumor/genética , Proliferación Celular , Inmunoprecipitación de Cromatina , Estudios de Cohortes , Cistadenocarcinoma Seroso/tratamiento farmacológico , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/mortalidad , Cistadenocarcinoma Seroso/patología , Ensayo de Cambio de Movilidad Electroforética , Neoplasias de las Trompas Uterinas/tratamiento farmacológico , Neoplasias de las Trompas Uterinas/genética , Neoplasias de las Trompas Uterinas/patología , Femenino , Estudios de Seguimiento , Humanos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Neoplasias Peritoneales/tratamiento farmacológico , Neoplasias Peritoneales/genética , Neoplasias Peritoneales/patología , Pronóstico , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tasa de Supervivencia , Células Tumorales Cultivadas
12.
Hum Mol Genet ; 25(22): 5046-5058, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28171565

RESUMEN

Genome-wide association studies (GWAS) have identified markers within the WNT4 region on chromosome 1p36.12 showing consistent and strong association with increasing endometriosis risk. Fine mapping using sequence and imputed genotype data has revealed strong candidates for the causal SNPs within these critical regions; however, the molecular pathogenesis of these SNPs is currently unknown. We used gene expression data collected from whole blood from 862 individuals and endometrial tissue from 136 individuals from independent populations of European descent to examine the mechanism underlying endometriosis susceptibility. Association mapping results from 7,090 individuals (2,594 cases and 4,496 controls) supported rs3820282 as the SNP with the strongest association for endometriosis risk (P = 1.84 × 10−5, OR = 1.244 (1.126-1.375)). SNP rs3820282 is a significant eQTL in whole blood decreasing expression of LINC00339 (also known as HSPC157) and increasing expression of CDC42 (P = 2.0 ×10−54 and 4.5x10−4 respectively). The largest effects were for two LINC00339 probes (P = 2.0 ×10−54; 1.0 × 10−34). The eQTL for LINC00339 was also observed in endometrial tissue (P = 2.4 ×10−8) with the same direction of effect for both whole blood and endometrial tissue. There was no evidence for eQTL effects for WNT4. Chromatin conformation capture provides evidence for risk SNPs interacting with the promoters of both LINC00339 and CDC4 and luciferase reporter assays suggest the risk SNP rs12038474 is located in a transcriptional silencer for CDC42 and the risk allele increases expression of CDC42. However, no effect of rs3820282 was observed in the LINC00339 expression in Ishikawa cells. Taken together, our results suggest that SNPs increasing endometriosis risk in this region act through CDC42, but further functional studies are required to rule out inverse regulation of both LINC00339 and CDC42.


Asunto(s)
Endometriosis/genética , ARN Largo no Codificante/genética , Proteína de Unión al GTP cdc42/genética , Estudios de Casos y Controles , Cromosomas Humanos Par 1 , Endometriosis/sangre , Femenino , Expresión Génica , Regulación de la Expresión Génica , Frecuencia de los Genes , Predisposición Genética a la Enfermedad/genética , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Sitios de Carácter Cuantitativo , ARN Largo no Codificante/metabolismo , Factores de Riesgo , Población Blanca/genética , Proteína Wnt4/genética , Proteína Wnt4/metabolismo , Proteína de Unión al GTP cdc42/metabolismo
13.
Am J Hum Genet ; 97(2): 329-36, 2015 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-26211970

RESUMEN

The gene(s) whose expression is regulated by allergy risk variants is unknown for many loci identified through genome-wide association studies. Addressing this knowledge gap might point to new therapeutic targets for allergic disease. The aim of this study was to identify the target gene(s) and the functional variant(s) underlying the association between rs7009110 on chromosome 8q21 and allergies. Eight genes are located within 1 Mb of rs7009110. Multivariate association analysis of publicly available exon expression levels from lymphoblastoid cell lines (LCLs) identified a significant association between rs7009110 and the expression of a single gene, PAG1 (p = 0.0017), 732 kb away. Analysis of histone modifications and DNase I hypersensitive sites in LCLs identified four putative regulatory elements (PREs) in the region. Chromosome conformation capture confirmed that two PREs interacted with the PAG1 promoter, one in allele-specific fashion. To determine whether these PREs were functional, LCLs were transfected with PAG1 promoter-driven luciferase reporter constructs. PRE3 acted as a transcriptional enhancer for PAG1 exclusively when it carried the rs2370615:C allergy predisposing allele, a variant in complete linkage disequilibrium with rs7009110. As such, rs2370615, which overlaps RelA transcription factor (TF) binding in LCLs and was found to disrupt Foxo3a binding to PRE3, represents the putative functional variant in this locus. Our studies suggest that the risk-associated allele of rs2370615 predisposes to allergic disease by increasing PAG1 expression, which might promote B cell activation and have a pro-inflammatory effect. Inhibition of PAG1 expression or function might have therapeutic potential for allergic diseases.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Cromosomas Humanos Par 8/genética , Regulación de la Expresión Génica/genética , Hipersensibilidad/genética , Proteínas de la Membrana/genética , Linfocitos B/inmunología , Estudios de Asociación Genética , Humanos , Hipersensibilidad/inmunología , Desequilibrio de Ligamiento , Luciferasas , Activación de Linfocitos/genética , Análisis Multivariante , Conformación de Ácido Nucleico , Polimorfismo de Nucleótido Simple/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética
14.
Hum Mol Genet ; 24(5): 1478-92, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25378557

RESUMEN

Common variants in the hepatocyte nuclear factor 1 homeobox B (HNF1B) gene are associated with the risk of Type II diabetes and multiple cancers. Evidence to date indicates that cancer risk may be mediated via genetic or epigenetic effects on HNF1B gene expression. We previously found single-nucleotide polymorphisms (SNPs) at the HNF1B locus to be associated with endometrial cancer, and now report extensive fine-mapping and in silico and laboratory analyses of this locus. Analysis of 1184 genotyped and imputed SNPs in 6608 Caucasian cases and 37 925 controls, and 895 Asian cases and 1968 controls, revealed the best signal of association for SNP rs11263763 (P = 8.4 × 10(-14), odds ratio = 0.86, 95% confidence interval = 0.82-0.89), located within HNF1B intron 1. Haplotype analysis and conditional analyses provide no evidence of further independent endometrial cancer risk variants at this locus. SNP rs11263763 genotype was associated with HNF1B mRNA expression but not with HNF1B methylation in endometrial tumor samples from The Cancer Genome Atlas. Genetic analyses prioritized rs11263763 and four other SNPs in high-to-moderate linkage disequilibrium as the most likely causal SNPs. Three of these SNPs map to the extended HNF1B promoter based on chromatin marks extending from the minimal promoter region. Reporter assays demonstrated that this extended region reduces activity in combination with the minimal HNF1B promoter, and that the minor alleles of rs11263763 or rs8064454 are associated with decreased HNF1B promoter activity. Our findings provide evidence for a single signal associated with endometrial cancer risk at the HNF1B locus, and that risk is likely mediated via altered HNF1B gene expression.


Asunto(s)
Mapeo Cromosómico , Neoplasias Endometriales/genética , Sitios Genéticos , Factor Nuclear 1-beta del Hepatocito/genética , Alelos , Estudios de Casos y Controles , Línea Celular Tumoral , Biología Computacional , Bases de Datos Genéticas , Epigénesis Genética , Femenino , Variación Genética , Estudio de Asociación del Genoma Completo , Genotipo , Haplotipos , Factor Nuclear 1-beta del Hepatocito/metabolismo , Humanos , Desequilibrio de Ligamiento , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Riesgo , Población Blanca/genética
15.
Am J Hum Genet ; 96(1): 5-20, 2015 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-25529635

RESUMEN

Genome-wide association studies (GWASs) have revealed SNP rs889312 on 5q11.2 to be associated with breast cancer risk in women of European ancestry. In an attempt to identify the biologically relevant variants, we analyzed 909 genetic variants across 5q11.2 in 103,991 breast cancer individuals and control individuals from 52 studies in the Breast Cancer Association Consortium. Multiple logistic regression analyses identified three independent risk signals: the strongest associations were with 15 correlated variants (iCHAV1), where the minor allele of the best candidate, rs62355902, associated with significantly increased risks of both estrogen-receptor-positive (ER(+): odds ratio [OR] = 1.24, 95% confidence interval [CI] = 1.21-1.27, ptrend = 5.7 × 10(-44)) and estrogen-receptor-negative (ER(-): OR = 1.10, 95% CI = 1.05-1.15, ptrend = 3.0 × 10(-4)) tumors. After adjustment for rs62355902, we found evidence of association of a further 173 variants (iCHAV2) containing three subsets with a range of effects (the strongest was rs113317823 [pcond = 1.61 × 10(-5)]) and five variants composing iCHAV3 (lead rs11949391; ER(+): OR = 0.90, 95% CI = 0.87-0.93, pcond = 1.4 × 10(-4)). Twenty-six percent of the prioritized candidate variants coincided with four putative regulatory elements that interact with the MAP3K1 promoter through chromatin looping and affect MAP3K1 promoter activity. Functional analysis indicated that the cancer risk alleles of four candidates (rs74345699 and rs62355900 [iCHAV1], rs16886397 [iCHAV2a], and rs17432750 [iCHAV3]) increased MAP3K1 transcriptional activity. Chromatin immunoprecipitation analysis revealed diminished GATA3 binding to the minor (cancer-protective) allele of rs17432750, indicating a mechanism for its action. We propose that the cancer risk alleles act to increase MAP3K1 expression in vivo and might promote breast cancer cell survival.


Asunto(s)
Neoplasias de la Mama/genética , Mapeo Cromosómico , Cromosomas Humanos Par 5/genética , Quinasa 1 de Quinasa de Quinasa MAP/genética , Sitios de Carácter Cuantitativo , Alelos , Estudios de Casos y Controles , Línea Celular Tumoral , Femenino , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Técnicas de Genotipaje , Humanos , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Células MCF-7 , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Grupos Raciales/genética , Factores de Riesgo
16.
Nat Commun ; 4: 4999, 2014 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-25248036

RESUMEN

GWAS have identified a breast cancer susceptibility locus on 2q35. Here we report the fine mapping of this locus using data from 101,943 subjects from 50 case-control studies. We genotype 276 SNPs using the 'iCOGS' genotyping array and impute genotypes for a further 1,284 using 1000 Genomes Project data. All but two, strongly correlated SNPs (rs4442975 G/T and rs6721996 G/A) are excluded as candidate causal variants at odds against >100:1. The best functional candidate, rs4442975, is associated with oestrogen receptor positive (ER+) disease with an odds ratio (OR) in Europeans of 0.85 (95% confidence interval=0.84-0.87; P=1.7 × 10(-43)) per t-allele. This SNP flanks a transcriptional enhancer that physically interacts with the promoter of IGFBP5 (encoding insulin-like growth factor-binding protein 5) and displays allele-specific gene expression, FOXA1 binding and chromatin looping. Evidence suggests that the g-allele confers increased breast cancer susceptibility through relative downregulation of IGFBP5, a gene with known roles in breast cell biology.


Asunto(s)
Neoplasias de la Mama/genética , Cromosomas Humanos Par 2/genética , Proteína 5 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Regiones Promotoras Genéticas/genética , ARN Mensajero/metabolismo , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Cromatina/metabolismo , Cromosomas Humanos Par 2/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Humanos , Proteína 5 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Células MCF-7 , Polimorfismo de Nucleótido Simple
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...