Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Gut ; 72(4): 612-623, 2023 04.
Article in English | MEDLINE | ID: mdl-35882562

ABSTRACT

OBJECTIVE: Oesophageal cancer (EC) is the sixth leading cause of cancer-related deaths. Oesophageal adenocarcinoma (EA), with Barrett's oesophagus (BE) as a precursor lesion, is the most prevalent EC subtype in the Western world. This study aims to contribute to better understand the genetic causes of BE/EA by leveraging genome wide association studies (GWAS), genetic correlation analyses and polygenic risk modelling. DESIGN: We combined data from previous GWAS with new cohorts, increasing the sample size to 16 790 BE/EA cases and 32 476 controls. We also carried out a transcriptome wide association study (TWAS) using expression data from disease-relevant tissues to identify BE/EA candidate genes. To investigate the relationship with reported BE/EA risk factors, a linkage disequilibrium score regression (LDSR) analysis was performed. BE/EA risk models were developed combining clinical/lifestyle risk factors with polygenic risk scores (PRS) derived from the GWAS meta-analysis. RESULTS: The GWAS meta-analysis identified 27 BE and/or EA risk loci, 11 of which were novel. The TWAS identified promising BE/EA candidate genes at seven GWAS loci and at five additional risk loci. The LDSR analysis led to the identification of novel genetic correlations and pointed to differences in BE and EA aetiology. Gastro-oesophageal reflux disease appeared to contribute stronger to the metaplastic BE transformation than to EA development. Finally, combining PRS with BE/EA risk factors improved the performance of the risk models. CONCLUSION: Our findings provide further insights into BE/EA aetiology and its relationship to risk factors. The results lay the foundation for future follow-up studies to identify underlying disease mechanisms and improving risk prediction.


Subject(s)
Adenocarcinoma , Barrett Esophagus , Esophageal Neoplasms , Humans , Barrett Esophagus/pathology , Genome-Wide Association Study , Esophageal Neoplasms/pathology , Adenocarcinoma/pathology
2.
Cancer Epidemiol Biomarkers Prev ; 31(9): 1735-1745, 2022 09 02.
Article in English | MEDLINE | ID: mdl-35709760

ABSTRACT

BACKGROUND: Over 20 susceptibility single-nucleotide polymorphisms (SNP) have been identified for esophageal adenocarcinoma (EAC) and its precursor, Barrett esophagus (BE), explaining a small portion of heritability. METHODS: Using genetic data from 4,323 BE and 4,116 EAC patients aggregated by international consortia including the Barrett's and Esophageal Adenocarcinoma Consortium (BEACON), we conducted a comprehensive transcriptome-wide association study (TWAS) for BE/EAC, leveraging Genotype Tissue Expression (GTEx) gene-expression data from six tissue types of plausible relevance to EAC etiology: mucosa and muscularis from the esophagus, gastroesophageal (GE) junction, stomach, whole blood, and visceral adipose. Two analytical approaches were taken: standard TWAS using the predicted gene expression from local expression quantitative trait loci (eQTL), and set-based SKAT association using selected eQTLs that predict the gene expression. RESULTS: Although the standard approach did not identify significant signals, the eQTL set-based approach identified eight novel associations, three of which were validated in independent external data (eQTL SNP sets for EXOC3, ZNF641, and HSP90AA1). CONCLUSIONS: This study identified novel genetic susceptibility loci for EAC and BE using an eQTL set-based genetic association approach. IMPACT: This study expanded the pool of genetic susceptibility loci for EAC and BE, suggesting the potential of the eQTL set-based genetic association approach as an alternative method for TWAS analysis.


Subject(s)
Adenocarcinoma , Barrett Esophagus , Esophageal Neoplasms , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Barrett Esophagus/genetics , Barrett Esophagus/pathology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Genetic Predisposition to Disease , Humans , Quantitative Trait Loci
3.
Gut ; 71(6): 1053-1061, 2022 06.
Article in English | MEDLINE | ID: mdl-34187846

ABSTRACT

OBJECTIVE: Gastro-oesophageal reflux disease (GERD) has heterogeneous aetiology primarily attributable to its symptom-based definitions. GERD genome-wide association studies (GWASs) have shown strong genetic overlaps with established risk factors such as obesity and depression. We hypothesised that the shared genetic architecture between GERD and these risk factors can be leveraged to (1) identify new GERD and Barrett's oesophagus (BE) risk loci and (2) explore potentially heterogeneous pathways leading to GERD and oesophageal complications. DESIGN: We applied multitrait GWAS models combining GERD (78 707 cases; 288 734 controls) and genetically correlated traits including education attainment, depression and body mass index. We also used multitrait analysis to identify BE risk loci. Top hits were replicated in 23andMe (462 753 GERD cases, 24 099 BE cases, 1 484 025 controls). We additionally dissected the GERD loci into obesity-driven and depression-driven subgroups. These subgroups were investigated to determine how they relate to tissue-specific gene expression and to risk of serious oesophageal disease (BE and/or oesophageal adenocarcinoma, EA). RESULTS: We identified 88 loci associated with GERD, with 59 replicating in 23andMe after multiple testing corrections. Our BE analysis identified seven novel loci. Additionally we showed that only the obesity-driven GERD loci (but not the depression-driven loci) were associated with genes enriched in oesophageal tissues and successfully predicted BE/EA. CONCLUSION: Our multitrait model identified many novel risk loci for GERD and BE. We present strong evidence for a genetic underpinning of disease heterogeneity in GERD and show that GERD loci associated with depressive symptoms are not strong predictors of BE/EA relative to obesity-driven GERD loci.


Subject(s)
Barrett Esophagus , Esophageal Neoplasms , Esophagitis, Peptic , Gastroesophageal Reflux , Barrett Esophagus/complications , Barrett Esophagus/diagnosis , Barrett Esophagus/genetics , Esophageal Neoplasms/diagnosis , Esophageal Neoplasms/genetics , Gastroesophageal Reflux/complications , Gastroesophageal Reflux/diagnosis , Gastroesophageal Reflux/genetics , Genome-Wide Association Study , Humans , Obesity/complications , Obesity/genetics
4.
Article in German | MEDLINE | ID: mdl-34519834

ABSTRACT

Since May 2020, manufacturers have been able to submit an application to the Federal Institute for Drugs and Medical Devices (BfArM) for the inclusion of a digital health application (DiGA) in the directory pursuant to section 139e of the German Social Code Book V. This new application procedure is based on specific requirements and assessment parameters for which a large number of procedural and scientific questions arise on the side of the manufacturers.In order to meet this need, the BfArM has specifically expanded its established information and advisory services for these new questions. This article describes in detail what these are in relation to DiGA, where the corresponding information and documents can be found and what distinguishes them in each case. Furthermore, with a view to the counselling interviews conducted between May 2020 and April 2021, their influence on the outcome of the respective application procedure for inclusion in the DiGA directory is analysed. This confirms that the early discussion of parameters relevant to the evaluation and the consideration of the advisory recommendations correlate with a positive evaluation result. For the DiGA listed in the directory, 80% of the manufacturers had previously received advice from the BfArM. The rate of withdrawn or rejected applications, on the other hand, was significantly higher if manufacturers had not previously sought advice from the BfArM in advance compared to manufacturers who discussed key aspects (63% vs. 35%). Overall, all sides benefit from the early, intensive exchange - in the end especially the patients through introduction of DiGA into standard healthcare due to higher application quality without delay.


Subject(s)
Academies and Institutes , Referral and Consultation , Germany , Humans
5.
Nat Commun ; 12(1): 246, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33431812

ABSTRACT

Previous Mendelian randomization (MR) studies on 25-hydroxyvitamin D (25(OH)D) and cancer have typically adopted a handful of variants and found no relationship between 25(OH)D and cancer; however, issues of horizontal pleiotropy cannot be reliably addressed. Using a larger set of variants associated with 25(OH)D (74 SNPs, up from 6 previously), we perform a unified MR analysis to re-evaluate the relationship between 25(OH)D and ten cancers. Our findings are broadly consistent with previous MR studies indicating no relationship, apart from ovarian cancers (OR 0.89; 95% C.I: 0.82 to 0.96 per 1 SD change in 25(OH)D concentration) and basal cell carcinoma (OR 1.16; 95% C.I.: 1.04 to 1.28). However, after adjustment for pigmentation related variables in a multivariable MR framework, the BCC findings were attenuated. Here we report that lower 25(OH)D is unlikely to be a causal risk factor for most cancers, with our study providing more precise confidence intervals than previously possible.


Subject(s)
Genetic Predisposition to Disease , Mendelian Randomization Analysis , Neoplasms/genetics , Vitamin D/metabolism , Case-Control Studies , Child , Humans , Multivariate Analysis , Pigmentation/genetics , Risk Factors , Sunburn/genetics
6.
Drug Discov Today ; 26(2): 283-288, 2021 02.
Article in English | MEDLINE | ID: mdl-33127567

ABSTRACT

Truly disruptive medicine innovation and new treatment paradigms tend to start in non-commercial research institutions. However, the lack of mutual understanding between medicine developers and regulators when it comes to medicine development significantly delays or even prevents the access of patients to these innovations. Here, we outline what regulatory-related barriers hamper the translational development of novel products or new treatment paradigms initiated in academia, and propose key steps towards improved regulatory dialogue among academia, funding bodies and regulatory authorities. Moreover, we briefly describe how the STARS (Strengthening Training of Academia in Regulatory Science) project aims to reach out to medicine innovators in academia to bridge the regulatory knowledge gap and enhance this dialogue to facilitate the implementation of academic research findings in clinical practice.


Subject(s)
Diffusion of Innovation , Translational Research, Biomedical/organization & administration , Disruptive Technology/legislation & jurisprudence , European Union , Humans , Translational Research, Biomedical/legislation & jurisprudence
7.
Carcinogenesis ; 42(3): 369-377, 2021 04 17.
Article in English | MEDLINE | ID: mdl-33300568

ABSTRACT

Genome-wide association studies (GWAS) of esophageal adenocarcinoma (EAC) and its precursor, Barrett's esophagus (BE), have uncovered significant genetic components of risk, but most heritability remains unexplained. Targeted assessment of genetic variation in biologically relevant pathways using novel analytical approaches may identify missed susceptibility signals. Central obesity, a key BE/EAC risk factor, is linked to systemic inflammation, altered hormonal signaling and insulin-like growth factor (IGF) axis dysfunction. Here, we assessed IGF-related genetic variation and risk of BE and EAC. Principal component analysis was employed to evaluate pathway-level and gene-level associations with BE/EAC, using genotypes for 270 single-nucleotide polymorphisms (SNPs) in or near 12 IGF-related genes, ascertained from 3295 BE cases, 2515 EAC cases and 3207 controls in the Barrett's and Esophageal Adenocarcinoma Consortium (BEACON) GWAS. Gene-level signals were assessed using Multi-marker Analysis of GenoMic Annotation (MAGMA) and SNP summary statistics from BEACON and an expanded GWAS meta-analysis (6167 BE cases, 4112 EAC cases, 17 159 controls). Global variation in the IGF pathway was associated with risk of BE (P = 0.0015). Gene-level associations with BE were observed for GHR (growth hormone receptor; P = 0.00046, false discovery rate q = 0.0056) and IGF1R (IGF1 receptor; P = 0.0090, q = 0.0542). These gene-level signals remained significant at q < 0.1 when assessed using data from the largest available BE/EAC GWAS meta-analysis. No significant associations were observed for EAC. This study represents the most comprehensive evaluation to date of inherited genetic variation in the IGF pathway and BE/EAC risk, providing novel evidence that variation in two genes encoding cell-surface receptors, GHR and IGF1R, may influence risk of BE.


Subject(s)
Adenocarcinoma/genetics , Barrett Esophagus/genetics , Biomarkers, Tumor/genetics , Esophageal Neoplasms/genetics , Somatomedins/metabolism , Adenocarcinoma/pathology , Aged , Barrett Esophagus/pathology , Biomarkers, Tumor/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Esophageal Neoplasms/pathology , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , Germ-Line Mutation , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Risk Factors , Signal Transduction/genetics
8.
Gastroenterology ; 159(6): 2065-2076.e1, 2020 12.
Article in English | MEDLINE | ID: mdl-32918910

ABSTRACT

BACKGROUND & AIMS: Esophageal adenocarcinoma (EA) and its premalignant lesion, Barrett's esophagus (BE), are characterized by a strong and yet unexplained male predominance (with a male-to-female ratio in EA incidence of up to 6:1). Genome-wide association studies (GWAS) have identified more than 20 susceptibility loci for these conditions. However, potential sex differences in genetic associations with BE/EA remain largely unexplored. METHODS: Given strong genetic overlap, BE and EA cases were combined into a single case group for analysis. These were compared with population-based controls. We performed sex-specific GWAS of BE/EA in 3 separate studies and then used fixed-effects meta-analysis to provide summary estimates for >9 million variants for male and female individuals. A series of downstream analyses were conducted separately in male and female individuals to identify genes associated with BE/EA and the genetic correlations between BE/EA and other traits. RESULTS: We included 6758 male BE/EA cases, 7489 male controls, 1670 female BE/EA cases, and 6174 female controls. After Bonferroni correction, our meta-analysis of sex-specific GWAS identified 1 variant at chromosome 6q11.1 (rs112894788, KHDRBS2-MTRNR2L9, PBONF = .039) that was statistically significantly associated with BE/EA risk in male individuals only, and 1 variant at chromosome 8p23.1 (rs13259457, PRSS55-RP1L1, PBONF = 0.057) associated, at borderline significance, with BE/EA risk in female individuals only. We also observed strong genetic correlations of BE/EA with gastroesophageal reflux disease in male individuals and obesity in female individuals. CONCLUSIONS: The identified novel sex-specific variants associated with BE/EA could improve the understanding of the genetic architecture of the disease and the reasons for the male predominance.


Subject(s)
Adenocarcinoma/genetics , Barrett Esophagus/genetics , Biomarkers, Tumor/genetics , Esophageal Neoplasms/genetics , Genetic Predisposition to Disease , Adenocarcinoma/epidemiology , Barrett Esophagus/epidemiology , Case-Control Studies , Esophageal Neoplasms/epidemiology , Eye Proteins/genetics , Female , Gastroesophageal Reflux/epidemiology , Gastroesophageal Reflux/genetics , Genetic Loci , Genome-Wide Association Study , Humans , Male , Obesity/epidemiology , Obesity/genetics , Polymorphism, Single Nucleotide , RNA-Binding Proteins/genetics , Risk Assessment , Risk Factors , Serine Endopeptidases/genetics , Sex Factors
9.
Cancer Epidemiol Biomarkers Prev ; 29(2): 427-433, 2020 02.
Article in English | MEDLINE | ID: mdl-31748258

ABSTRACT

BACKGROUND: Obesity is a major risk factor for esophageal adenocarcinoma (EA) and its precursor Barrett's esophagus (BE). Research suggests that individuals with high genetic risk to obesity have a higher BE/EA risk. To facilitate understanding of biological factors that lead to progression from BE to EA, the present study investigated the shared genetic background of BE/EA and obesity-related traits. METHODS: Cross-trait linkage disequilibrium score regression was applied to summary statistics from genome-wide association meta-analyses on BE/EA and on obesity traits. Body mass index (BMI) was used as a proxy for general obesity, and waist-to-hip ratio (WHR) for abdominal obesity. For single marker analyses, all genome-wide significant risk alleles for BMI and WHR were compared with summary statistics of the BE/EA meta-analyses. RESULTS: Sex-combined analyses revealed a significant genetic correlation between BMI and BE/EA (rg = 0.13, P = 2 × 10-04) and a rg of 0.12 between WHR and BE/EA (P = 1 × 10-02). Sex-specific analyses revealed a pronounced genetic correlation between BMI and EA in females (rg = 0.17, P = 1.2 × 10-03), and WHR and EA in males (rg = 0.18, P = 1.51 × 10-02). On the single marker level, significant enrichment of concordant effects was observed for BMI and BE/EA risk variants (P = 8.45 × 10-03) and WHR and BE/EA risk variants (P = 2 × 10-02). CONCLUSIONS: Our study provides evidence for sex-specific genetic correlations that might reflect specific biological mecha-nisms. The data demonstrate that shared genetic factors are particularly relevant in progression from BE to EA. IMPACT: Our study quantifies the genetic correlation between BE/EA and obesity. Further research is now warranted to elucidate these effects and to understand the shared pathophysiology.


Subject(s)
Adenocarcinoma/genetics , Barrett Esophagus/genetics , Esophageal Neoplasms/genetics , Obesity/genetics , Quantitative Trait Loci , Adenocarcinoma/pathology , Barrett Esophagus/pathology , Body Mass Index , Disease Progression , Esophageal Neoplasms/pathology , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Linkage Disequilibrium , Male , Meta-Analysis as Topic , Polymorphism, Single Nucleotide , Risk Assessment , Risk Factors , Sex Factors , Waist-Hip Ratio
11.
Nat Commun ; 10(1): 4219, 2019 09 16.
Article in English | MEDLINE | ID: mdl-31527586

ABSTRACT

Gastroesophageal reflux disease (GERD) is caused by gastric acid entering the esophagus. GERD has high prevalence and is the major risk factor for Barrett's esophagus (BE) and esophageal adenocarcinoma (EA). We conduct a large GERD GWAS meta-analysis (80,265 cases, 305,011 controls), identifying 25 independent genome-wide significant loci for GERD. Several of the implicated genes are existing or putative drug targets. Loci discovery is greatest with a broad GERD definition (including cases defined by self-report or medication data). Further, 91% of the GERD risk-increasing alleles also increase BE and/or EA risk, greatly expanding gene discovery for these traits. Our results map genes for GERD and related traits and uncover potential new drug targets for these conditions.


Subject(s)
Esophageal Diseases/genetics , Gastroesophageal Reflux/genetics , Genome-Wide Association Study , Female , Genetic Loci , Genetic Predisposition to Disease , Humans , Male , Polymorphism, Single Nucleotide
12.
Clin Gastroenterol Hepatol ; 17(11): 2227-2235.e1, 2019 10.
Article in English | MEDLINE | ID: mdl-30716477

ABSTRACT

BACKGROUND & AIMS: Epidemiology studies of circulating concentrations of 25 hydroxy vitamin D (25(OH)D) and risk of esophageal adenocarcinoma (EAC) have produced conflicting results. We conducted a Mendelian randomization study to determine the associations between circulating concentrations of 25(OH)D and risks of EAC and its precursor, Barrett's esophagus (BE). METHODS: We conducted a Mendelian randomization study using a 2-sample (summary data) approach. Six single-nucleotide polymorphisms (SNPs; rs3755967, rs10741657, rs12785878, rs10745742, rs8018720, and rs17216707) associated with circulating concentrations of 25(OH)D were used as instrumental variables. We collected data from 6167 patients with BE, 4112 patients with EAC, and 17,159 individuals without BE or EAC (controls) participating in the Barrett's and Esophageal Adenocarcinoma Consortium, as well as studies from Bonn, Germany, and Cambridge and Oxford, United Kingdom. Analyses were performed separately for BE and EAC. RESULTS: Overall, we found no evidence for an association between genetically estimated 25(OH)D concentration and risk of BE or EAC. The odds ratio per 20 nmol/L increase in genetically estimated 25(OH)D concentration for BE risk estimated by combining the individual SNP association using inverse variance weighting was 1.21 (95% CI, 0.77-1.92; P = .41). The odds ratio for EAC risk, estimated by combining the individual SNP association using inverse variance weighting, was 0.68 (95% CI, 0.39-1.19; P = .18). CONCLUSIONS: In a Mendelian randomization study, we found that low genetically estimated 25(OH)D concentrations were not associated with risk of BE or EAC.


Subject(s)
Adenocarcinoma/genetics , Barrett Esophagus/genetics , Esophageal Neoplasms/genetics , Mendelian Randomization Analysis/methods , Polymorphism, Single Nucleotide , Risk Assessment , Vitamin D/blood , Adenocarcinoma/blood , Adenocarcinoma/epidemiology , Barrett Esophagus/blood , Barrett Esophagus/epidemiology , Biomarkers, Tumor/blood , DNA, Neoplasm/genetics , Esophageal Neoplasms/blood , Esophageal Neoplasms/epidemiology , Europe/epidemiology , Female , Humans , Male , Morbidity , North America/epidemiology , Risk Factors
13.
PLoS One ; 14(12): e0227072, 2019.
Article in English | MEDLINE | ID: mdl-31891614

ABSTRACT

Esophageal adenocarcinoma (EA) and its precancerous condition Barrett's esophagus (BE) are multifactorial diseases with rising prevalence rates in Western populations. A recent meta-analysis of genome-wide association studies (GWAS) data identified 14 BE/EA risk loci located in non-coding genomic regions. Knowledge about the impact of non-coding variation on disease pathology is incomplete and needs further investigation. The aim of the present study was (i) to identify candidate genes of functional relevance to BE/EA at known risk loci and (ii) to find novel risk loci among the suggestively associated variants through the integration of expression quantitative trait loci (eQTL) and genetic association data. eQTL data from two BE/EA-relevant tissues (esophageal mucosa and gastroesophageal junction) generated within the context of the GTEx project were cross-referenced with the GWAS meta-analysis data. Variants representing an eQTL in at least one of the two tissues were categorized into genome-wide significant loci (P < 5×10-8) and novel candidate loci (5×10-8 ≤ P ≤ 5×10-5). To follow up these novel candidate loci, a genetic association study was performed in a replication cohort comprising 1,993 cases and 967 controls followed by a combined analysis with the GWAS meta-analysis data. The cross-referencing of eQTL and genetic data yielded 2,180 variants that represented 25 loci. Among the previously reported genome-wide significant loci, 22 eQTLs were identified in esophageal mucosa and/or gastroesophageal junction tissue. The regulated genes, most of which have not been linked to BE/EA etiology so far, included C2orf43/LDAH, ZFP57, and SLC9A3. Among the novel candidate loci, replication was achieved for two variants (rs7754014, Pcombined = 3.16×10-7 and rs1540, Pcombined = 4.16×10-6) which represent eQTLs for CFDP1 and SLC22A3, respectively. In summary, the present approach identified candidate genes whose expression was regulated by risk variants in disease-relevant tissues. These findings may facilitate the elucidation of BE/EA pathophysiology.


Subject(s)
Adenocarcinoma/genetics , Barrett Esophagus/genetics , Esophageal Mucosa/pathology , Esophageal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Quantitative Trait Loci , Adenocarcinoma/pathology , Barrett Esophagus/pathology , Esophageal Neoplasms/pathology , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Polymorphism, Single Nucleotide , Proteins/genetics , Repressor Proteins/genetics , Sodium-Hydrogen Exchanger 3/genetics
14.
Cancer Med ; 7(10): 5057-5065, 2018 10.
Article in English | MEDLINE | ID: mdl-30191681

ABSTRACT

Genetic associations between variants on chromosome 5p13 and 8q24 and gastric cancer (GC) have been previously reported in the Asian population. We aimed to replicate these findings and to characterize the associations at the genome and transcriptome level. We performed a fine-mapping association study in 1926 GC patients and 2012 controls of European descent using high dense SNP marker sets on both chromosomal regions. Next, we performed expression quantitative trait locus (eQTL) analyses using gastric transcriptome data from 143 individuals focusing on the GC associated variants. On chromosome 5p13 the strongest association was observed at rs6872282 (P = 2.53 × 10-04 ) and on chromosome 8q24 at rs2585176 (P = 1.09 × 10-09 ). On chromosome 5p13 we found cis-eQTL effects with an upregulation of PTGER4 expression in GC risk allele carrier (P = 9.27 × 10-11 ). On chromosome 8q24 we observed cis-eQTL effects with an upregulation of PSCA expression in GC risk allele carrier (P = 2.17 × 10-47 ). In addition, we found trans-eQTL effects for the same variants on 8q24 with a downregulation of MBOAT7 expression in GC risk allele carrier (P = 3.11 × 10-09 ). In summary, we confirmed and refined the previously reported GC associations at both chromosomal regions. Our data point to shared etiological factors between Asians and Europeans. Furthermore, our data imply an upregulated expression of PTGER4 and PSCA as well as a downregulated expression of MBOAT7 in gastric tissue as risk-conferring GC pathomechanisms.


Subject(s)
Acyltransferases/genetics , Antigens, Neoplasm/genetics , Gene Expression Profiling/methods , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Receptors, Prostaglandin E, EP4 Subtype/genetics , Stomach Neoplasms/genetics , Case-Control Studies , Chromosome Mapping/methods , Chromosomes, Human, Pair 5/genetics , Chromosomes, Human, Pair 8/genetics , Female , GPI-Linked Proteins/genetics , Gene Expression Regulation, Neoplastic , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , Humans , Male , Polymorphism, Single Nucleotide , Quantitative Trait Loci
15.
Birth Defects Res ; 110(10): 871-882, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29498243

ABSTRACT

BACKGROUND: Nonsyndromic cleft palate only (nsCPO) is a common and multifactorial form of orofacial clefting. In contrast to successes achieved for the other common form of orofacial clefting, that is, nonsyndromic cleft lip with/without cleft palate (nsCL/P), genome wide association studies (GWAS) of nsCPO have identified only one genome wide significant locus. Aim of the present study was to investigate whether common variants contribute to nsCPO and, if so, to identify novel risk loci. METHODS: We genotyped 33 SNPs at 27 candidate loci from 2 previously published nsCPO GWAS in an independent multiethnic sample. It included: (i) a family-based sample of European ancestry (n = 212); and (ii) two case/control samples of Central European (n = 94/339) and Arabian ancestry (n = 38/231), respectively. A separate association analysis was performed for each genotyped dataset, and meta-analyses were performed. RESULTS: After association analysis and meta-analyses, none of the 33 SNPs showed genome-wide significance. Two variants showed nominally significant association in the imputed GWAS dataset and exhibited a further decrease in p-value in a European and an overall meta-analysis including imputed GWAS data, respectively (rs395572: PMetaEU = 3.16 × 10-4 ; rs6809420: PMetaAll = 2.80 × 10-4 ). CONCLUSION: Our findings suggest that there is a limited contribution of common variants to nsCPO. However, the individual effect sizes might be too small for detection of further associations in the present sample sizes. Rare variants may play a more substantial role in nsCPO than in nsCL/P, for which GWAS of smaller sample sizes have identified genome-wide significant loci. Whole-exome/genome sequencing studies of nsCPO are now warranted.


Subject(s)
Cleft Palate/genetics , Arabs/genetics , Exome/genetics , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study/methods , Genotype , Humans , Polymorphism, Single Nucleotide/genetics , White People/genetics
16.
Transl Psychiatry ; 8(1): 31, 2018 01 31.
Article in English | MEDLINE | ID: mdl-29382824

ABSTRACT

The large biological distance between genetic risk loci and their mechanistic consequences in the tissue of interest limits the ability to establish functionality of susceptibility variants for genetically complex traits. Such a biological gap may be reduced through the systematic study of molecular mediators of genomic action, such as epigenetic modification. Here, we report the identification of robust genetic estimators of whole-blood CpG methylation, which can serve as intermediate molecular traits amenable to association testing with other genetically complex traits. We describe the relationship between these estimators and gene expression, demonstrate their genome-wide applicability to association testing even in the absence of individual genotypic data, and show that these estimators powerfully identify methylation-related genomic loci associated with polygenic traits and common diseases, such as schizophrenia. The use of genetic estimators for blood DNA methylation, which are made publically available, can serve as a valuable tool for the identification of epigenetic underpinnings of complex traits.


Subject(s)
CpG Islands/genetics , DNA Methylation/genetics , Gene Expression/genetics , Genetic Loci/genetics , Genome-Wide Association Study/methods , Multifactorial Inheritance/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Female , Genotype , Humans , Male , Schizophrenia/genetics , Young Adult
17.
Birth Defects Res ; 110(4): 336-341, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29134786

ABSTRACT

Nonsyndromic cleft lip with or without cleft palate (nsCL/P) is one of the most common congenital malformation worldwide, and its etiology involves both genetic and environmental factors. Recent genome-wide and targeted genetic studies of nsCL/P have identified numerous genetic risk loci, under the hypothesis of a multiplicative mode of inheritance. The present study investigated whether novel nsCL/P risk loci could be identified by analyzing dominant/recessive genetic effects in single nucleotide polymorphism (SNP) data from genome-wide association studies. For this purpose, a genome-wide investigation of dominant/recessive common SNP effects was performed in our previously published meta-analysis data set. Twenty-four loci were identified as candidate regions. In a subsequent association analysis in an independent study cohort of 224 nsCL/P patients and 986 controls of European descent, none of the loci could be replicated. Therefore, our strategy of identifying novel loci by applying different genetic models did not yield any novel findings, suggesting that recessive/dominant common variation only make a limited contribution to nsCL/P in Europeans. However, we cannot rule out that such effects are present at some of the loci that have previously been identified, or are present in different populations.


Subject(s)
Cleft Lip/genetics , Cleft Palate/genetics , Genes, Dominant , Genes, Recessive , Genome-Wide Association Study , Models, Genetic , Female , Humans , Male
18.
Psychiatr Genet ; 27(3): 96-102, 2017 06.
Article in English | MEDLINE | ID: mdl-28272115

ABSTRACT

OBJECTIVES: Social anxiety disorder (SAD) is a common and heritable psychiatric disorder. However, genetic studies in SAD are rare and only a few candidate genes have been implicated so far. In the present study, we investigated whether single-nucleotide polymorphisms (SNPs) associated with other psychiatric disorders also contribute toward the development of SAD and followed up variants associated with SAD on the phenotypic level. PATIENTS AND METHODS: We genotyped a total of 24 SNPs in a German sample of 321 SAD patients and 804 controls. We carried out single-marker analyses as well as quantitative association analyses of SAD severity and harm avoidance. RESULTS: None of the variants investigated showed an association with SAD in our case-control sample after Bonferroni correction. Two SNPs reached nominal significance (rs818702, P=0.032; rs140701, P=0.048). Of these, only rs140701 showed an association in the same allelic direction as reported previously. This SNP is located within the serotonin transporter gene SLC6A4, which is the primary target of selective-serotonin reuptake inhibitors used for the treatment of depressive and anxiety disorders. The quantitative association analysis of all cases with available data on symptom severity showed four SNPs with a nominal significant association. Among these SNPs, rs10994359 showed the strongest association (P=0.001) and was located near the ANK3 gene. In addition, rs10994359 was nominally associated with harm avoidance scores (P=0.001). CONCLUSION: Our results provide further evidence for an involvement of the serotonin transporter gene SLC6A4 in the etiology of anxiety-related traits. Furthermore, our study implicates that genetic variation at the genome-wide associated bipolar disorder locus ANK3 might influence anxiety-related personality traits.


Subject(s)
Anxiety Disorders/genetics , Serotonin Plasma Membrane Transport Proteins/genetics , Adult , Anxiety/genetics , Anxiety/metabolism , Case-Control Studies , Female , Gene Frequency , Genetic Predisposition to Disease/genetics , Genetic Variation , Genotype , Germany , Humans , Linkage Disequilibrium , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Risk Factors , Serotonin Plasma Membrane Transport Proteins/metabolism
19.
Hum Mol Genet ; 26(4): 829-842, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28087736

ABSTRACT

Nonsyndromic cleft lip with or without cleft palate (nsCL/P) is among the most common human birth defects with multifactorial etiology. Here, we present results from a genome-wide imputation study of nsCL/P in which, after adding replication cohort data, four novel risk loci for nsCL/P are identified (at chromosomal regions 2p21, 14q22, 15q24 and 19p13). On a systematic level, we show that the association signals within this high-density dataset are enriched in functionally-relevant genomic regions that are active in both human neural crest cells (hNCC) and mouse embryonic craniofacial tissue. This enrichment is also detectable in hNCC regions primed for later activity. Using GCTA analyses, we suggest that 30% of the estimated variance in risk for nsCL/P in the European population can be attributed to common variants, with 25.5% contributed to by the 24 risk loci known to date. For each of these, we identify credible SNPs using a Bayesian refinement approach, with two loci harbouring only one probable causal variant. Finally, we demonstrate that there is no polygenic component of nsCL/P detectable that is shared with nonsyndromic cleft palate only (nsCPO). Our data suggest that, while common variants are strongly contributing to risk for nsCL/P, they do not seem to be involved in nsCPO which might be more often caused by rare deleterious variants. Our study generates novel insights into both nsCL/P and nsCPO etiology and provides a systematic framework for research into craniofacial development and malformation.


Subject(s)
Chromosomes, Human/genetics , Cleft Lip/genetics , Cleft Palate/genetics , Databases, Genetic , Genetic Loci , Genome-Wide Association Study , Polymorphism, Single Nucleotide , Animals , Cleft Lip/metabolism , Cleft Lip/pathology , Cleft Palate/metabolism , Cleft Palate/pathology , Female , Humans , Male , Mice
20.
Genom Data ; 10: 22-9, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27630819

ABSTRACT

Previous research suggests a genetic overlap between nonsyndromic cleft lip with or without cleft palate (NSCL/P) and cancer. The aim of the present study was to identify common genetic risk loci for NSCL/P and cancer entities that have been reported to co-occur with orofacial clefting. This was achieved through the investigation of large genome-wide association study datasets. Investigations of 12 NSCL/P single nucleotide polymorphisms (SNPs) in 32 cancer datasets, and 204 cancer SNPs in two NSCL/P datasets, were performed. The SNPs rs13041247 (20q12) and rs6457327 (6p21.33) showed suggestive evidence for an association with both NSCL/P and a specific cancer entity. These loci harbor genes of biological relevance to oncogenesis (MAFB and OCT4, respectively). This study is the first to characterize possible pleiotropic risk loci for NSCL/P and cancer in a systematic manner. The data represent a starting point for future research by identifying a genetic link between NSCL/P and cancer.

SELECTION OF CITATIONS
SEARCH DETAIL