Untargeted Metabolome- and Transcriptome-Wide Association Study Suggests Causal Genes Modulating Metabolite Concentrations in UrineClick to copy article linkArticle link copied!
- Reyhan Sönmez Flitman*Reyhan Sönmez Flitman*Email: [email protected]Department of Computational Biology, University of Lausanne, Lausanne 1015, SwitzerlandSwiss Institute of Bioinformatics, Lausanne 1015, SwitzerlandMore by Reyhan Sönmez Flitman
- Bita KhaliliBita KhaliliDepartment of Computational Biology, University of Lausanne, Lausanne 1015, SwitzerlandSwiss Institute of Bioinformatics, Lausanne 1015, SwitzerlandMore by Bita Khalili
- Zoltan KutalikZoltan KutalikDepartment of Computational Biology, University of Lausanne, Lausanne 1015, SwitzerlandUniversity Center for Primary Care and Public Health, University of Lausanne, Lausanne 1010, SwitzerlandSwiss Institute of Bioinformatics, Lausanne 1015, SwitzerlandMore by Zoltan Kutalik
- Rico RueediRico RueediDepartment of Computational Biology, University of Lausanne, Lausanne 1015, SwitzerlandSwiss Institute of Bioinformatics, Lausanne 1015, SwitzerlandMore by Rico Rueedi
- Anneke BrümmerAnneke BrümmerDepartment of Computational Biology, University of Lausanne, Lausanne 1015, SwitzerlandSwiss Institute of Bioinformatics, Lausanne 1015, SwitzerlandMore by Anneke Brümmer
- Sven Bergmann*Sven Bergmann*Email: [email protected]Department of Computational Biology, University of Lausanne, Lausanne 1015, SwitzerlandSwiss Institute of Bioinformatics, Lausanne 1015, SwitzerlandDepartment of Integrative Biomedical Sciences, University of Cape Town, Cape Town 7700, South AfricaMore by Sven Bergmann
Abstract
Gene products can affect the concentrations of small molecules (aka “metabolites”), and conversely, some metabolites can modulate the concentrations of gene transcripts. While many specific instances of this interplay have been revealed, a global approach to systematically uncover human gene-metabolite interactions is still lacking. We performed a metabolome- and transcriptome-wide association study to identify genes influencing the human metabolome using untargeted metabolome features, extracted from 1H nuclear magnetic resonance spectroscopy (NMR) of urine samples, and gene expression levels, quantified from RNA-Seq of lymphoblastoid cell lines (LCL) from 555 healthy individuals. We identified 20 study-wide significant associations corresponding to 15 genes, of which 5 associations (with 2 genes) were confirmed with follow-up NMR data. Using metabomatching, we identified the metabolites corresponding to metabolome features associated with the genes, namely, N-acetylated compounds with ALMS1 and trimethylamine (TMA) with HPS1. Finally, Mendelian randomization analysis supported a potential causal link between the expression of genes in both the ALMS1- and HPS1-loci and their associated metabolite concentrations. In the case of HPS1, we additionally observed that TMA concentration likely exhibits a reverse causal effect on HPS1 expression levels, indicating a negative feedback loop. Our study highlights how the integration of metabolomics, gene expression, and genetic data can pinpoint causal genes modulating metabolite concentrations.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Introduction
Materials and Methods
Study Samples
Metabolomics Data
Gene Expression Data
Genotypic Data
Association Analysis
Metabomatching
Mendelian Randomization
Results
Association Analysis Identifies 20 Significant Metabolome- and Transcriptome-Wide Associations
Figure 1
Figure 1. QQ-plot of −log10 (p values) of metabolome- and transcriptome-wide association analysis. The highly significant associations (FDR < 0.05) with ALMS1 expression are ranked 1st and 2nd and with HPS1 expression 3rd and 4th.
genes | metabolite | association | published as mGWAS | |||
---|---|---|---|---|---|---|
ensembl gene ID | Chr | gene symbol | feature(s) | effect size | p value | body fluid |
ENSG00000116127 | 2 | ALMS1 | 2.0375, 2.0325, 2.0275, 2.0425 | 0.72, 0.69, 0.45, 0.41 | 1.1 × 10–20, 2.0 × 10–19, 7.8 × 10–09, 1.2 × 10–07 | serum, (9,33) urine (27,34) |
ENSG00000107521 | 10 | HPS1 | 2.8575, 2.8725 | –0.38, −0.37 | 1.1 × 10–10, 5.6 × 10–10 | serum, (9,33) urine (27,34) |
ENSG00000149089 | 11 | APIP | 2.7925 | –0.33 | 4.7 × 10–09 | serum, (9,33) urine (27) |
ENSG00000256029 | 1 | RP11-190A12.7 | 3.0925 | 0.26 | 9.7 × 10–09 | serum (9) |
ENSG00000100603 | 14 | SNW1 | 8.1275 | –0.38 | 1.5 × 10–08 | serum (33) |
ENSG00000163016 | 2 | ALMS1P | 2.0325, 2.0375 | 0.27, 0.27 | 2.5 × 10–08, 2.7 × 10–08 | serum, (9,33) urine (27) |
ENSG00000219257 | 6 | RP11-14I4.2 | 2.3275 | –0.25 | 5.7 × 10–08 | |
ENSG00000259357 | 1 | RP11-316M1.12 | 7.7875 | 0.33 | 6.1 × 10–08 | |
ENSG00000163520 | 3 | FBLN2 | 5.4375 | 0.29 | 9.5 × 10–08 | serum (9,33) |
ENSG00000226430 | 8 | USP17L7 | 2.7075 | –0.24 | 1.8 × 10–07 | |
ENSG00000219355 | 12 | RPL31P52 | 2.8675 | –0.23 | 2.1 × 10–07 | |
ENSG00000266795 | 17 | RP11-744K17.9 | 7.2725 | 0.26 | 2.2 × 10–07 | serum (9) |
ENSG00000150593 | 10 | PDCD4 | 5.4075 | 0.42 | 2.2 × 10–07 | serum, (9,33) urine (27) |
ENSG00000266805 | 18 | RP11-61L19.1 | 5.3525 | 0.24 | 2.7 × 10–07 | |
ENSG00000254396 | 9 | RP11-56F10.3 | 3.0925 | 0.23 | 3.6 × 10–07 |
20 study-wide significant associations involving 15 unique genes and 17 unique features. Associations are grouped by genes and sorted by the lowest association p value for each gene.
Identification of Metabolites Corresponding to Metabolome Features Associated with Gene Expression
Figure 2
Figure 2. Metabomatching (22) results for pseudospectra derived from gene expression - metabolome feature associations for ALMS1 (A) and HPS1 (B). Upper panels show the features in each pseudospectrum, color-coded according to the direction of the effect (positive in blue and negative in orange). Lower panels show the highest ranking candidate metabolites with their reference NMR spectra (color coded to indicate their relative peak intensities). Leading features allowing metabolite identification are in (A) at 2.04 ppm, which matches well with the highest intensity peak of the NAA spectrum and in (B) at 2.87 ppm, which matches well with the TMA singlet.
Validation of Significant Metabolome–Transcriptome Associations Using Follow-up Urine NMR Data
mGWAS for NAC and TMA Indicates Numerous Significant SNPs in the ALMS1 and HPS1 Gene Loci
Figure 3
Figure 3. SNP - metabolome feature and SNP - gene expression associations in ALMS1/NAT8 locus. (A) LocusZoom plot for ALMS1/NAT8 locus, where the SNPs are associated with metabolome feature at 2.0375 ppm, LD colored with respect to lead mQTL. (B) Bar plot shows −log10 transformed p values from associating expression values of nine genes in the locus with the five NAA features.
Figure 4
Figure 4. SNP - metabolome feature and SNP - gene expression associations in HPS1/PYROXD2 locus. (A) LocusZoom plot for HPS1/PYROXD2 locus, showing the association significance of SNP with the metabolome feature at 2.8725 ppm. Colors indicate the correlation (LD) to the lead QTL. (B) Bar plot shows −log10 transformed p values from associating expression values of seven genes in the locus with the same feature.
reference | platform | biofluid | locus | metabolite |
---|---|---|---|---|
Nicholson et al. 2011 (34) | MS + NMR | urine + plasma | ALMS1, NAT8 | N-acetylated compounds |
Montoliu et al. 2013 (44) | NMR | urine | ALMS1 | N-acetylated compounds |
Rueedi et al. 2014 (18) | NMR | urine | ALMS1 | 2.0375 (suggested as N-acetylated compounds) |
Raffler et al. 2015 (27) | NMR | urine | NAT8 | 2.031 (suggested as N-acetyl-l-aspartate) |
Suhre et al. 2011 (38) | MS | serum | NAT8 | N-acetylornithine |
Yu et al. 2014 (39) | MS | serum | NAT8 | N-acetylornithine |
Shin et al. 2014 (33) | MS | serum | NAT8 | N-acetyllysine, unknown compounds |
Nicholson et al. 2011 (34) | MS + NMR | urine + plasma | HPS1, PYROXD2 | trimethylamine (urine), dimethylamine (plasma), |
Rueedi et al. 2014 (18) | NMR | urine | PYROXD2 | trimethylamine, unknown compound, 2.8575, 1.8025 |
Raffler et al. 2015 (27) | NMR | urine | PYROXD2 | 2.854 (suggested as trimethylamine) |
Raffler et al. 2013 (40) | NMR | plasma | PYROXD2 | 2.757 |
Rhee et al. 2013 (41) | MS | plasma | HPS1 | asymmetric dimethylarginine |
Krumsiek et al. 2012 (42) | MS | serum | HPS1, PYROXD2 | multiple compounds, unknown compounds |
Hong et al. 2013 (43) | MS | serum | HPS1 | caprolactam |
Shin et al. 2014 (33) | MS | serum | PYROXD2 | unknown compounds |
MS: mass spectrometry; numbers in metabolite section refer to NMR spectral shift positions in ppm. Reported genes are mostly based on proximity to the mQTL or based on gene function.
Figure 5
Figure 5. Scatter plot of the mQTL effect of SNP (rs7566315) on NAC and its eQTL effect on ALMS1 gene expression. Each point represents a study sample. NAC concentration is approximated by the feature at 2.0375 ppm that is log10 transformed after feature- and sample-wise z-scoring (y axis). ALMS1 expression is z-scored after log2 transforming RPKM+1 values (x axis). Color code represents the genotype of rs7566315 (legend) that is an eQTL of ALMS1 and mQTL of NAA.
Mendelian Randomization Analysis Suggests ALMS1 Expression in Blood and Confirms NAT8 Expression in Other Tissues to Causally Effect NAC Concentration
method | causal effect size estimate | std. error | 95% CI | p value | Cochran’s Q-statistic p value | ||
---|---|---|---|---|---|---|---|
A | ALMS1 → NAC | inverse variance weighted | 0.967 | 0.061 | 0.847–1.087 | <2 × 10–16 | 0.2323 |
weighted median | 1.111 | 0.075 | 0.965–1.257 | <2 × 10–16 | NA | ||
MR - Egger | 0.994 | 0.092 | 0.812–1.175 | <2 × 10–16 | 0.1776 | ||
maximum-likelihood | 0.999 | 0.065 | 0.872–1.126 | <2 × 10–16 | 0.249 | ||
B | NAC → ALMS1 | inverse variance weighted | –0.015 | 0.264 | –0.532–0.502 | 0.955 | 0.7443 |
weighted median | 0.122 | 0.321 | –0.507–0.751 | 0.704 | NA | ||
MR - Egger | 1.495 | 1.976 | –2.377–5.368 | 0.449 | 0.7256 | ||
maximum-likelihood | –0.015 | 0.266 | –0.535–0.505 | 0.955 | 0.7443 | ||
C | ALMS1 → NAC | inverse variance weighted | 0.796 | 0.183 | 0.437–1.155 | <2 × 10–16 | 0.1902 |
(NAT8 related SNPs removed) | weighted median | 0.668 | 0.242 | 0.193–1.142 | 0.006 | NA | |
MR - Egger | 1.912 | 0.704 | 0.532–3.291 | 0.007 | 0.4144 | ||
maximum-likelihood | 0.805 | 0.185 | 0.444–1.167 | < 2 × 10–16 | 0.2199 |
MR results for testing (A) causal effect of ALMS1 gene expression levels on N-acetylated compounds (ALMS1 → NAC), (B) causal effect of N-acetylated compounds on ALMS1 gene expression levels (NAC → ALMS1), (C) causal effect of ALMS1 gene expression levels on N-acetylated compounds (ALMS1 → NAC) when NAT8-related SNPs were removed from the instrument set.
Mendelian Randomization Analysis Suggests HPS1 as Causal Gene Modulating TMA Concentration and Indicates a Reverse Causal Effect between Both
method | causal effect size estimate | std. error | 95% CI | p value | Cochran’s Q-statistic p value | ||
---|---|---|---|---|---|---|---|
A | HPS1 → TMA | inverse variance weighted | 0.266 | 0.094 | 0.082–0.450 | 0.005 | 0.0803 |
weighted median | 0.311 | 0.072 | 0.170–0.453 | <2 × 10–16 | NA | ||
MR - Egger | 0.37 | 0.126 | 0.123–0.617 | 0.003 | 0.0852 | ||
maximum-likelihood | 0.267 | 0.094 | 0.083–0.452 | 0.004 | 0.0829 | ||
B | TMA → HPS1 | inverse variance weighted | –0.089 | 0.012 | –0.113 to –0.065 | <2 × 10–16 | 0.0958 |
weighted median | –0.09 | 0.011 | –0.111 to –0.068 | <2 × 10–16 | NA | ||
MR - Egger | –0.086 | 0.013 | –0.111 to –0.061 | <2 × 10–16 | 0.0758 | ||
maximum-likelihood | –0.09 | 0.012 | –0.114 to –0.066 | <2 × 10–16 | 0.1258 | ||
C | HPS1 → TMA | inverse variance weighted | |||||
(PYROXD2 related SNPs removed) | weighted median | 1.079 | 0.121 | 0.842–1.315 | <2 × 10–16 | NA | |
MR - Egger | 1.705 | 0.305 | 1.107–2.303 | <2 × 10–16 | 0.5575 | ||
maximum-likelihood |
MR results for testing (A) causal effect of HPS1 gene expression levels on TMA (HPS1 → TMA), (B) causal effect of TMA on HPS1 gene expression levels (TMA → HPS1), (C) causal effect of HPS1 gene expression levels on TMA (HPS1 → TMA) when PYROXD2-related SNPs were removed from the instrument set.
Discussion
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jproteome.1c00585.
Figure S1: Principal component analysis of baseline and follow-up metabolomics data; Figure S2: Overview of RNA-Seq read count and quantifiable genes in 555 individuals; Figure S3: Scatter plots of removed associations; Figure S4: Scatter plots of 20 study-wide significant metabolome feature - gene expression associations; Figure S5: Metabomatching figure showing the pseudospectrum derived from ALMS1 gene expression - metabolome features associations; Figure S6: Metabomatching figure showing the pseudospectrum derived from ALMS1P gene expression - metabolome features associations; Figure S7: NMR profiles of 3 different spike-in experiments; Figure S8: Metabomatching figure showing the pseudospectrum derived from APIP gene expression - metabolome feature associations; Figure S9: Metabomatching figure showing the pseudospectrum derived from SNW1 gene expression - metabolome feature associations; Table S1: Validation of all associations discovered in CoLaus baseline; Table S2: MR results of testing causal effect of ALMS1 gene expression levels on N-acetylated compounds, using R-squared threshold of 0.05; Table S3: MR results of testing causal effect of HPS1 gene expression levels on trimethylamine, using R-squared threshold of 0.05 (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
This work was supported by the Swiss National Science Foundation (grant FN 310030_152724/1) and the NIH (grant R03 CA211815).
References
This article references 53 other publications.
- 1Buniello, A.; MacArthur, J. A. L.; Cerezo, M.; Harris, L. W.; Hayhurst, J.; Malangone, C. The NHGRI-EBI GWAS Catalog of published genome-wide association studies, targeted arrays and summary statistics 2019. Nucleic Acids Res. 2019, 47, D1005– D1012, DOI: 10.1093/nar/gky1120Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhs1GqtrvO&md5=08a31cc6157f3ffe9be66587fdeed121The NHGRI-EBI GWAS catalog of published genome-wide association studies, targeted arrays and summary statistics 2019Buniello, Annalisa; MacArthu, Jacqueline A. L.; Cerez, Maria; Harris, Laura W.; Hayhurst, James; Malangone, Cinzia; McMahon, Aoife; Morales, Joannella; Mountjoy, Edward; Sollis, Elliot; Suveges, Daniel; Vrousgou, Olga; Whetzel, Patricia L.; Amode, Ridwan; Guillen, Jose A.; Riat, Harpreet S.; Trevanion, Stephen J.; Hall, Peggy; Junkins, Heather; Flicek, Paul; Burdett, Tony; Hindorff, Lucia A.; Cunningham, Fiona; Parkinson, HelenNucleic Acids Research (2019), 47 (D1), D1005-D1012CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)The GWAS Catalog delivers a high-quality curated collection of all published genome-wide assocn. studies enabling investigations to identify causal variants, understand disease mechanisms, and establish targets for novel therapies. The scope of the Catalog has also expanded to targeted and exome arrays with 1000 new assocns. added for these technologies. As of Sept. 2018, the Catalog contains 5687 GWAS comprising 71673 variant-trait assocns. from 3567 publications. New content includes 284 full P-value summary statistics datasets for genome-wide and new targeted array studies, representing 6 × 109 individual variant-trait statistics. In the last 12 mo, the Catalog's user interface was accessed by ∼90000 unique users who viewed >1 million pages. We have improved data access with the release of a new RESTful API to support high-throughput programmatic access, an improved web interface and a new summary statistics database. Summary statistics provision is supported by a new format proposed as a community std. for summary statistics data representation. This format was derived from our experience in standardizing heterogeneous submissions, mapping formats and in harmonizing content.
- 2GTEx Consortium The GTEx Consortium atlas of genetic regulatory effects across human tissues. Science 2020, 369, 1318– 1330, DOI: 10.1126/science.aaz1776Google ScholarThere is no corresponding record for this reference.
- 3Maurano, M. T.; Humbert, R.; Rynes, E.; Thurman, R. E.; Haugen, E.; Wang, H. Systematic localization of common disease-associated variation in regulatory DNA. Science 2012, 337, 1190– 1195, DOI: 10.1126/science.1222794Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xht1ylsLfL&md5=6b4de239d584cc09c6e513580e6caa3cSystematic localization of common disease-associated variation in regulatory DNAMaurano, Matthew T.; Humbert, Richard; Rynes, Eric; Thurman, Robert E.; Haugen, Eric; Wang, Hao; Reynolds, Alex P.; Sandstrom, Richard; Qu, Hongzhu; Brody, Jennifer; Shafer, Anthony; Neri, Fidencio; Lee, Kristen; Kutyavin, Tanya; Stehling-Sun, Sandra; Johnson, Audra K.; Canfield, Theresa K.; Giste, Erika; Diegel, Morgan; Bates, Daniel; Hansen, R. Scott; Neph, Shane; Sabo, Peter J.; Heimfeld, Shelly; Raubitschek, Antony; Ziegler, Steven; Cotsapas, Chris; Sotoodehnia, Nona; Glass, Ian; Sunyaev, Shamil R.; Kaul, Rajinder; Stamatoyannopoulos, John A.Science (Washington, DC, United States) (2012), 337 (6099), 1190-1195CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Genome-wide assocn. studies have identified many noncoding variants assocd. with common diseases and traits. We show that these variants are concd. in regulatory DNA marked by DNase I hypersensitive sites (DHSs). Eighty-eight percent of such DHSs are active during fetal development and are enriched in variants assocd. with gestational exposure-related phenotypes. We identified distant gene targets for hundreds of variant-contg. DHSs that may explain phenotype assocns. Disease-assocd. variants systematically perturb transcription factor recognition sequences, frequently alter allelic chromatin states, and form regulatory networks. We also demonstrated tissue-selective enrichment of more weakly disease-assocd. variants within DHSs and the de novo identification of pathogenic cell types for Crohn's disease, multiple sclerosis, and an ECG trait, without prior knowledge of physiol. mechanisms. Our results suggest pervasive involvement of regulatory DNA variation in common human disease and provide pathogenic insights into diverse disorders.
- 4Nicolae, D. L.; Gamazon, E.; Zhang, W.; Duan, S.; Dolan, M. E.; Cox, N. J. Trait-associated SNPs are more likely to be eQTLs: annotation to enhance discovery from GWAS. PLoS Genet. 2010, 6, e1000888, DOI: 10.1371/journal.pgen.1000888Google ScholarThere is no corresponding record for this reference.
- 5Ward, L. D.; Kellis, M. Interpreting non-coding variation in complex disease genetics. Nat. Biotechnol. 2012, 30, 1095, DOI: 10.1038/nbt.2422Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xhs1elsr%252FL&md5=bc1dd6f524db613547c87ecac384675cInterpreting noncoding genetic variation in complex traits and human diseaseWard, Lucas D.; Kellis, ManolisNature Biotechnology (2012), 30 (11), 1095-1106CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)A review. Assocn. studies provide genome-wide information about the genetic basis of complex disease, but medical research has focused primarily on protein-coding variants, owing to the difficulty of interpreting noncoding mutations. This picture has changed with advances in the systematic annotation of functional noncoding elements. Evolutionary conservation, functional genomics, chromatin state, sequence motifs and mol. quant. trait loci all provide complementary information about the function of noncoding sequences. These functional maps can help with prioritizing variants on risk haplotypes, filtering mutations encountered in the clinic and performing systems-level analyses to reveal processes underlying disease assocns. Advances in predictive modeling can enable data-set integration to reveal pathways shared across loci and alleles, and richer regulatory models can guide the search for epistatic interactions. Lastly, new massively parallel reporter expts. can systematically validate regulatory predictions. Ultimately, advances in regulatory and systems genomics can help unleash the value of whole-genome sequencing for personalized genomic risk assessment, diagnosis and treatment.
- 6Lloyd-Jones, L. R.; Holloway, A.; McRae, A.; Yang, J.; Small, K.; Zhao, J. The genetic architecture of gene expression in peripheral blood. Am. J. Hum. Genet. 2017, 100, 228– 237, DOI: 10.1016/j.ajhg.2016.12.008Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXlt12qsQ%253D%253D&md5=667b0bb41842675e780eaee9cb6879e2The Genetic Architecture of Gene Expression in Peripheral BloodLloyd-Jones, Luke R.; Holloway, Alexander; McRae, Allan; Yang, Jian; Small, Kerrin; Zeng, Biao; Bakshi, Andrew; Metspalu, Andres; Dermitzakis, Manolis; Gibson, Greg; Spector, Tim; Montgomery, Grant; Esko, Tonu; Visscher, Peter M.; Powell, Joseph E.American Journal of Human Genetics (2017), 100 (2), 228-237CODEN: AJHGAG; ISSN:0002-9297. (Cell Press)We analyzed the mRNA levels for 36,778 transcript expression traits (probes) from 2,765 individuals to comprehensively investigate the genetic architecture and degree of missing heritability for gene expression in peripheral blood. We identified 11,204 cis and 3,791 trans independent expression quant. trait loci (eQTL) by using linear mixed models to perform genome-wide assocn. analyses. Furthermore, using information on both closely and distantly related individuals, heritability was estd. for all expression traits. Of the set of expressed probes (15,966), 10,580 (66%) had an estd. narrow-sense heritability (h2) greater than zero with a mean (median) value of 0.192 (0.142). Across these probes, on av. the proportion of genetic variance explained by all eQTL (h2COJO) was 31% (0.060/0.192), meaning that 69% is missing, with the sentinel SNP of the largest eQTL explaining 87% (0.052/0.060) of the variance attributed to all identified cis- and trans-eQTL. For the same set of probes, the genetic variance attributed to genome-wide common (MAF > 0.01) HapMap 3 SNPs (h2g) accounted for on av. 48% (0.093/0.192) of h2. Taken together, the evidence suggests that approx. half the genetic variance for gene expression is not tagged by common SNPs, and of the variance that is tagged by common SNPs, a large proportion can be attributed to identifiable eQTL of large effect, typically in cis. Finally, we present evidence that, compared with a meta-anal., using individual-level data results in an increase of approx. 50% in power to detect eQTL.
- 7Montgomery, S. B.; Sammeth, M.; Gutierrez-Arcelus, M.; Lach, R. P.; Ingle, C.; Nisbett, J. Transcriptome genetics using second generation sequencing in a Caucasian population. Nature 2010, 464, 773, DOI: 10.1038/nature08903Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXivFKnsbc%253D&md5=5bb32c958242d7bbd5c43c4aeaa3fc53Transcriptome genetics using second generation sequencing in a Caucasian populationMontgomery, Stephen B.; Sammeth, Micha; Gutierrez-Arcelus, Maria; Lach, Radoslaw P.; Ingle, Catherine; Nisbett, James; Guigo, Roderic; Dermitzakis, Emmanouil T.Nature (London, United Kingdom) (2010), 464 (7289), 773-777CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Gene expression is an important phenotype that informs about genetic and environmental effects on cellular state. Many studies have previously identified genetic variants for gene expression phenotypes using custom and com. available microarrays. Second generation sequencing technologies are now providing unprecedented access to the fine structure of the transcriptome. We have sequenced the mRNA fraction of the transcriptome in 60 extended HapMap individuals of European descent and have combined these data with genetic variants from the HapMap3 project. We have quantified exon abundance based on read depth and have also developed methods to quantify whole transcript abundance. We have found that approx. 10 million reads of sequencing can provide access to the same dynamic range as arrays with better quantification of alternative and highly abundant transcripts. Correlation with SNPs (small nucleotide polymorphisms) leads to a larger discovery of eQTLs (expression quant. trait loci) than with arrays. We also detect a substantial no. of variants that influence the structure of mature transcripts indicating variants responsible for alternative splicing. Finally, measures of allele-specific expression allowed the identification of rare eQTLs and allelic differences in transcript structure. This anal. shows that high throughput sequencing technologies reveal new properties of genetic effects on the transcriptome and allow the exploration of genetic effects in cellular processes.
- 8Wright, F. A.; Sullivan, P. F.; Brooks, A. I.; Zou, F.; Sun, W.; Xia, K. Heritability and genomics of gene expression in peripheral blood. Nat. Genet. 2014, 46, 430, DOI: 10.1038/ng.2951Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXmtlWrtb8%253D&md5=e687d946efdf04987a94b474ba930e27Heritability and genomics of gene expression in peripheral bloodWright, Fred A.; Sullivan, Patrick F.; Brooks, Andrew I.; Zou, Fei; Sun, Wei; Xia, Kai; Madar, Vered; Jansen, Rick; Chung, Wonil; Zhou, Yi-Hui; Abdellaoui, Abdel; Batista, Sandra; Butler, Casey; Chen, Guanhua; Chen, Ting-Huei; D'Ambrosio, David; Gallins, Paul; Ha, Min Jin; Hottenga, Jouke Jan; Huang, Shunping; Kattenberg, Mathijs; Kochar, Jaspreet; Middeldorp, Christel M.; Qu, Ani; Shabalin, Andrey; Tischfield, Jay; Todd, Laura; Tzeng, Jung-Ying; van Grootheest, Gerard; Vink, Jacqueline M.; Wang, Qi; Wang, Wei; Wang, Weibo; Willemsen, Gonneke; Smit, Johannes H.; de Geus, Eco J.; Yin, Zhaoyu; Penninx, Brenda W. J. H.; Boomsma, Dorret I.Nature Genetics (2014), 46 (5), 430-437CODEN: NGENEC; ISSN:1061-4036. (Nature Publishing Group)We assessed gene expression profiles in 2752 twins, using a classic twin design to quantify expression heritability and quant. trait loci (eQTLs) in peripheral blood. The most highly heritable genes (∼777) were grouped into distinct expression clusters, enriched in gene-poor regions, assocd. with specific gene function or ontol. classes, and strongly assocd. with disease designation. The design enabled a comparison of twin-based heritability to ests. based on dizygotic identity-by-descent sharing and distant genetic relatedness. Consideration of sampling variation suggests that previous heritability ests. have been upwardly biased. Genotyping of 2494 twins enabled powerful identification of eQTLs, which we further examd. in a replication set of 1895 unrelated subjects. A large no. of non-redundant local eQTLs (6756) met replication criteria, whereas a relatively small no. of distant eQTLs (165) met quality control and replication stds. Our results provide a new resource toward understanding the genetic control of transcription.
- 9Suhre, K.; Wallaschofski, H.; Raffler, J.; Friedrich, N.; Haring, R.; Michael, K. A genome-wide association study of metabolic traits in human urine. Nat. Genet. 2011, 43, 565– 569, DOI: 10.1038/ng.837Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXmtV2hs7g%253D&md5=6a695aed032888c41f97b16552775b2aA genome-wide association study of metabolic traits in human urineSuhre, Karsten; Wallaschofski, Henri; Raffler, Johannes; Friedrich, Nele; Haring, Robin; Michael, Kathrin; Wasner, Christina; Krebs, Alexander; Kronenberg, Florian; Chang, David; Meisinger, Christa; Wichmann, H.-Erich; Hoffmann, Wolfgang; Voelzke, Henry; Voelker, Uwe; Teumer, Alexander; Biffar, Reiner; Kocher, Thomas; Felix, Stephan B.; Illig, Thomas; Kroemer, Heyo K.; Gieger, Christian; Roemisch-Margl, Werner; Nauck, MatthiasNature Genetics (2011), 43 (6), 565-569CODEN: NGENEC; ISSN:1061-4036. (Nature Publishing Group)We present a genome-wide assocn. study of metabolic traits in human urine, designed to investigate the detoxification capacity of the human body. Using NMR spectroscopy, we tested for assocns. between 59 metabolites in urine from 862 male participants in the population-based SHIP study. We replicated the results using 1,039 addnl. samples of the same study, including a 5-yr follow-up, and 992 samples from the independent KORA study. We report five loci with joint P values of assocn. from 3.2 × 10-19 to 2.1 × 10-182. Variants at three of these loci have previously been linked with important clin. outcomes: SLC7A9 is a risk locus for chronic kidney disease, NAT2 for coronary artery disease and genotype-dependent response to drug toxicity, and SLC6A20 for iminoglycinuria. Moreover, we identify rs37369 in AGXT2 as the genetic basis of hyper-β-aminoisobutyric aciduria.
- 10Bartel, J.; Krumsiek, J.; Schramm, K.; Adamski, J.; Gieger, C.; Herder, C. The Human Blood Metabolome-Transcriptome Interface. PLoS Genet. 2015, 11, e1005274, DOI: 10.1371/journal.pgen.1005274Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXjvVKisLs%253D&md5=b9e5564d646b90c0e91499383bbbd54bThe human blood metabolome-transcriptome interfaceBartel, Joerg; Krumsiek, Jan; Schramm, Katharina; Adamski, Jerzy; Gieger, Christian; Herder, Christian; Carstensen, Maren; Peters, Annette; Rathmann, Wolfgang; Roden, Michael; Strauch, Konstantin; Suhre, Karsten; Kastenmueller, Gabi; Prokisch, Holger; Theis, Fabian J.PLoS Genetics (2015), 11 (6), e1005274/1-e1005274/32CODEN: PGLEB5; ISSN:1553-7404. (Public Library of Science)Biol. systems consist of multiple organizational levels all densely interacting with each other to ensure function and flexibility of the system. Simultaneous anal. of cross-sectional multi-omics data from large population studies is a powerful tool to comprehensively characterize the underlying mol. mechanisms on a physiol. scale. In this study, we systematically analyzed the relationship between fasting serum metabolomics and whole blood transcriptomics data from 712 individuals of the German KORA F4 cohort. Correlation-based anal. identified 1,109 significant assocns. between 522 transcripts and 114 metabolites summarized in an integrated network, the 'human blood metabolometranscriptome interface' (BMTI). Bidirectional causality anal. using Mendelian randomization did not yield any statistically significant causal assocns. between transcripts and metabolites. A knowledge-based interpretation and integration with a genome-scale human metabolic reconstruction revealed systematic signatures of signaling, transport and metabolic processes, i.e. metabolic reactions mainly belonging to lipid, energy and amino acid metab. Moreover, the construction of a network based on functional categories illustrated the cross-talk between the biol. layers at a pathway level. Using a transcription factor binding site enrichment anal., this pathway cross-talk was further confirmed at a regulatory level. Finally, we demonstrated how the constructed networks can be used to gain novel insights into mol. mechanisms assocd. to intermediate clin. traits. Overall, our results demonstrate the utility of a multi-omics integrative approach to understand the mol. mechanisms underlying both normal physiol. and disease.
- 11Burkhardt, R.; Kirsten, H.; Beutner, F.; Holdt, L. M.; Gross, A.; Teren, A. Integration of Genome-Wide SNP Data and Gene-Expression Profiles Reveals Six Novel Loci and Regulatory Mechanisms for Amino Acids and Acylcarnitines in Whole Blood. PLoS Genet. 2015, 11, e1005510, DOI: 10.1371/journal.pgen.1005510Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XmtFGltrg%253D&md5=9f36470c050b23fa8b6425d998e68d91Integration of genome-wide SNP data and gene-expression profiles reveals six novel loci and regulatory mechanisms for amino acids and acylcarnitines in whole bloodBurkhardt, Ralph; Kirsten, Holger; Beutner, Frank; Holdt, Lesca M.; Gross, Arnd; Teren, Andrej; Toenjes, Anke; Becker, Susen; Krohn, Knut; Kovacs, Peter; Stumvoll, Michael; Teupser, Daniel; Thiery, Joachim; Ceglarek, Uta; Scholz, MarkusPLoS Genetics (2015), 11 (9), e1005510/1-e1005510/25CODEN: PGLEB5; ISSN:1553-7404. (Public Library of Science)Profiling amino acids and acylcarnitines in whole blood spots is a powerful tool in the lab. diagnosis of several inborn errors of metab. Emerging data suggests that altered blood levels of amino acids and acylcarnitines are also assocd. with common metabolic diseases in adults. Thus, the identification of common genetic determinants for blood metabolites might shed light on pathways contributing to human physiol. and common diseases. We applied a targeted mass-spectrometry-based method to analyze whole blood concns. of 96 amino acids, acylcarnitines and pathway assocd. metabolite ratios in a Central European cohort of 2,107 adults and performed genome-wide assocn. (GWA) to identify genetic modifiers of metabolite concns. We discovered and replicated six novel loci assocd. with blood levels of total acylcarnitine, arginine (both on chromosome 6; rs12210538, rs17657775), propionylcarnitine (chromosome 10; rs12779637), 2-hydroxyisovalerylcarnitine (chromosome 21; rs1571700), stearoylcarnitine (chromosome 1; rs3811444), and aspartic acid traits (chromosome 8; rs750472). Based on an integrative anal. of expression quant. trait loci in blood mononuclear cells and correlations between gene expressions and metabolite levels, we provide evidence for putative causative genes: SLC22A16 for total acylcarnitines, ARG1 for arginine, HLCS for 2-hydroxyisovalerylcarnitine, JAM3 for stearoylcarnitine via a trans-effect at chromosome 1, and PPP1R16A for aspartic acid traits. Further, we report replication and provide addnl. functional evidence for ten loci that have previously been published for metabolites measured in plasma, serum or urine. In conclusion, our integrative anal. of SNP, gene-expression and metabolite data points to novel genetic factors that may be involved in the regulation of human metab. At several loci, we provide evidence for metabolite regulation via gene-expression and obsd. overlaps with GWAS loci for common diseases. These results form a strong rationale for subsequent functional and disease-related studies.
- 12Inouye, M.; Kettunen, J.; Soininen, P.; Silander, K.; Ripatti, S.; Kumpula, L. S.; Hämäläinen, E.; Jousilahti, P.; Kangas, A. J.; Männistö, S.; Savolainen, M. J.; Jula, A.; Leiviskä, J.; Palotie, A.; Salomaa, V.; Perola, M.; Ala-Korpela, M.; Peltonen, L. Metabonomic, transcriptomic, and genomic variation of a population cohort. Mol. Syst. Biol. 2010, 6, 441, DOI: 10.1038/msb.2010.93Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3M%252FmtFygug%253D%253D&md5=b4f99d89873a563ebf32c668fd9c9066Metabonomic, transcriptomic, and genomic variation of a population cohortInouye Michael; Kettunen Johannes; Soininen Pasi; Silander Kaisa; Ripatti Samuli; Kumpula Linda S; Hamalainen Eija; Jousilahti Pekka; Kangas Antti J; Mannisto Satu; Savolainen Markku J; Jula Antti; Leiviska Jaana; Palotie Aarno; Salomaa Veikko; Perola Markus; Ala-Korpela Mika; Peltonen LeenaMolecular systems biology (2010), 6 (), 441 ISSN:.Comprehensive characterization of human tissues promises novel insights into the biological architecture of human diseases and traits. We assessed metabonomic, transcriptomic, and genomic variation for a large population-based cohort from the capital region of Finland. Network analyses identified a set of highly correlated genes, the lipid-leukocyte (LL) module, as having a prominent role in over 80 serum metabolites (of 134 measures quantified), including lipoprotein subclasses, lipids, and amino acids. Concurrent association with immune response markers suggested the LL module as a possible link between inflammation, metabolism, and adiposity. Further, genomic variation was used to generate a directed network and infer LL module's largely reactive nature to metabolites. Finally, gene co-expression in circulating leukocytes was shown to be dependent on serum metabolite concentrations, providing evidence for the hypothesis that the coherence of molecular networks themselves is conditional on environmental factors. These findings show the importance and opportunity of systematic molecular investigation of human population samples. To facilitate and encourage this investigation, the metabonomic, transcriptomic, and genomic data used in this study have been made available as a resource for the research community.
- 13Bullaughey, K.; Chavarria, C. I.; Coop, G.; Gilad, Y. Expression quantitative trait loci detected in cell lines are often present in primary tissues. Hum. Mol. Genet. 2009, 18, 4296– 4303, DOI: 10.1093/hmg/ddp382Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtlWhu7fN&md5=00faca2e530fbbbd42b01e61c4408802Expression quantitative trait loci detected in cell lines are often present in primary tissuesBullaughey, Kevin; Chavarria, Claudia I.; Coop, Graham; Gilad, YoavHuman Molecular Genetics (2009), 18 (22), 4296-4303CODEN: HMGEE5; ISSN:0964-6906. (Oxford University Press)Expression quant. trait loci (eQTL) mapping is a powerful tool for identifying genetic regulatory variation. However, at present, most eQTLs in humans were identified using gene expression data from cell lines, and it remains unknown whether these eQTLs also have a regulatory function in other expression contexts, such as human primary tissues. Here we investigate this question using a targeted strategy. Specifically, we selected a subset of large-effect eQTLs identified in the HapMap lymphoblastoid cell lines, and examd. the assocn. of these eQTLs with gene expression levels across individuals in five human primary tissues (heart, kidney, liver, lung and testes). We show that genotypes at the eQTLs we selected are often predictive of variation in gene expression levels in one or more of the five primary tissues. The genotype effects in the primary tissues are consistently in the same direction as the effects inferred in the cell lines. Addnl., a no. of the eQTLs we tested are found in more than one of the tissues. Our results indicate that functional studies in cell lines may uncover a substantial amt. of genetic variation that affects gene expression levels in human primary tissues.
- 14Çalışkan, M.; Cusanovich, D. A.; Ober, C.; Gilad, Y. The effects of EBV transformation on gene expression levels and methylation profiles. Hum. Mol. Genet. 2011, 20, 1643– 1652, DOI: 10.1093/hmg/ddr041Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXjvVKrur4%253D&md5=b0ef2fc5995f0b08e47c759ef1725bc2The effects of EBV transformation on gene expression levels and methylation profilesCaliskan, Minal; Cusanovich, Darren A.; Ober, Carole; Gilad, YoavHuman Molecular Genetics (2011), 20 (8), 1643-1652CODEN: HMGEE5; ISSN:0964-6906. (Oxford University Press)Epstein-Barr virus (EBV) transformed lymphoblastoid cell lines (LCLs) provide a conveniently accessible and renewable resource for functional genomic studies in humans. The ability to accumulate multidimensional data pertaining to the same individual cell lines, from complete genomic sequences to detailed gene regulatory profiles, further enhances the utility of LCLs as a model system. A lingering concern, however, is that the changes assocd. with EBV transformation of B cells reduce the usefulness of LCLs as a surrogate model for primary tissues. To evaluate the validity of this concern, the authors compared global gene expression and methylation profiles between CD20+ primary B cells sampled from six individuals and six independent replicates of transformed LCLs derived from each sample. These data allowed the authors to obtain a detailed catalog of the genes and pathways whose regulation is affected by EBV transformation. The expression levels and promoter methylation profiles of more than half of the studied genes were affected by the EBV transformation, including enrichments of genes involved in transcription regulation, cell cycle and immune response. However, most of the differences in gene expression levels between LCLs and B cells are of small magnitude, and that LCLs can often recapitulate the naturally occurring gene expression variation in primary B cells. Thus, these observations suggest that inference of the genetic architecture that underlies regulatory variation in LCLs can typically be generalized to primary B cells. In contrast, inference based on functional studies in LCLs may be more limited to the cell lines.
- 15Dimas, A. S.; Deutsch, S.; Stranger, B. E.; Montgomery, S. B.; Borel, C.; Attar-Cohen, H. Common regulatory variation impacts gene expression in a cell type–dependent manner. Science 2009, 325, 1246– 1250, DOI: 10.1126/science.1174148Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtVOmsrjM&md5=c1c8c0127f5791bbf889eb29caaf5a82Common Regulatory Variation Impacts Gene Expression in a Cell Type-Dependent MannerDimas, Antigone S.; Deutsch, Samuel; Stranger, Barbara E.; Montgomery, Stephen B.; Borel, Christelle; Attar-Cohen, Homa; Ingle, Catherine; Beazley, Claude; Gutierrez Arcelus, Maria; Sekowska, Magdalena; Gagnebin, Marilyne; Nisbett, James; Deloukas, Panos; Dermitzakis, Emmanouil T.; Antonarakis, Stylianos E.Science (Washington, DC, United States) (2009), 325 (5945), 1246-1250CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Studies correlating genetic variation to gene expression facilitate the interpretation of common human phenotypes and disease. As functional variants may be operating in a tissue-dependent manner, we performed gene expression profiling and assocn. with genetic variants (single-nucleotide polymorphisms) on three cell types of 75 individuals. We detected cell type-specific genetic effects, with 69 to 80% of regulatory variants operating in a cell type-specific manner, and identified multiple expressive quant. trait loci (eQTLs) per gene, unique or shared among cell types and pos. correlated with the no. of transcripts per gene. Cell type-specific eQTLs were found at larger distances from genes and at lower effect size, similar to known enhancers. These data suggest that the complete regulatory variant repertoire can only be uncovered in the context of cell-type specificity.
- 16Ding, J.; Gudjonsson, J. E.; Liang, L.; Stuart, P. E.; Li, Y.; Chen, W. Gene expression in skin and lymphoblastoid cells: Refined statistical method reveals extensive overlap in cis-eQTL signals. Am. J. Hum. Genet. 2010, 87, 779– 789, DOI: 10.1016/j.ajhg.2010.10.024Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsFags7bM&md5=763f424eb72a032e6393b2bb36b1116eGene expression in skin and lymphoblastoid cells: refined statistical method reveals extensive overlap in cis-eQTL signalsDing, Jun; Gudjonsson, Johann E.; Liang, Liming; Stuart, Philip E.; Li, Yun; Chen, Wei; Weichenthal, Michael; Ellinghaus, Eva; Franke, Andre; Cookson, William; Nair, Rajan P.; Elder, James T.; Abecasis, Goncalo R.American Journal of Human Genetics (2010), 87 (6), 779-789CODEN: AJHGAG; ISSN:0002-9297. (Cell Press)Psoriasis, an immune-mediated, inflammatory disease of the skin and joints, provides an ideal system for expression quant. trait locus (eQTL) anal., because it has a strong genetic basis and disease-relevant tissue (skin) is readily accessible. To better understand the role of genetic variants regulating cutaneous gene expression, we identified 841 cis-acting eQTLs using RNA extd. from skin biopsies of 53 psoriatic individuals and 57 healthy controls. We found substantial overlap between cis-eQTLs of normal control, uninvolved psoriatic, and lesional psoriatic skin. Consistent with recent studies and with the idea that control of gene expression can mediate relationships between genetic variants and disease risk, we found that eQTL SNPs are more likely to be assocd. with psoriasis than are randomly selected SNPs. To explore the tissue specificity of these eQTLs and hence to quantify the benefits of studying eQTLs in different tissues, we developed a refined statistical method for estg. eQTL overlap and used it to compare skin eQTLs to a published panel of lymphoblastoid cell line (LCL) eQTLs. Our method accounts for the fact that most eQTL studies are likely to miss some true eQTLs as a result of power limitations and shows that ∼70% of cis-eQTLs in LCLs are shared with skin, as compared with the naive est. of <50% sharing. Our results provide a useful method for estg. the overlap between various eQTL studies and provide a catalog of cis-eQTLs in skin that can facilitate efforts to understand the functional impact of identified susceptibility variants on psoriasis and other skin traits.
- 17Firmann, M.; Mayor, V.; Vidal, P. M.; Bochud, M.; Pécoud, A.; Hayoz, D. The CoLaus study: a population-based study to investigate the epidemiology and genetic determinants of cardiovascular risk factors and metabolic syndrome. BMC Cardiovasc. Disord. 2008, 8, 6, DOI: 10.1186/1471-2261-8-6Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1c3ltF2luw%253D%253D&md5=6ffcf0d9742f2c91a6d24583188b9380The CoLaus study: a population-based study to investigate the epidemiology and genetic determinants of cardiovascular risk factors and metabolic syndromeFirmann Mathieu; Mayor Vladimir; Vidal Pedro Marques; Bochud Murielle; Pecoud Alain; Hayoz Daniel; Paccaud Fred; Preisig Martin; Song Kijoung S; Yuan Xin; Danoff Theodore M; Stirnadel Heide A; Waterworth Dawn; Mooser Vincent; Waeber Gerard; Vollenweider PeterBMC cardiovascular disorders (2008), 8 (), 6 ISSN:.BACKGROUND: Cardiovascular diseases and their associated risk factors remain the main cause of mortality in western societies. In order to assess the prevalence of cardiovascular risk factors (CVRFs) in the Caucasian population of Lausanne, Switzerland, we conducted a population-based study (Colaus Study). A secondary aim of the CoLaus study will be to determine new genetic determinants associated with CVRFs. METHODS: Single-center, cross-sectional study including a random sample of 6,188 extensively phenotyped Caucasian subjects (3,251 women and 2,937 men) aged 35 to 75 years living in Lausanne, and genotyped using the 500 K Affymetrix chip technology. RESULTS: Obesity (body mass index > or = 30 kg/m2), smoking, hypertension (blood pressure > or = 140/90 mmHg and/or treatment), dyslipidemia (high LDL-cholesterol and/or low HDL-cholesterol and/or high triglyceride levels) and diabetes (fasting plasma glucose > or = 7 mmol/l and/or treatment) were present in 947 (15.7%), 1673 (27.0%), 2268 (36.7%), 2113 (34.2%) and 407 (6.6%) of the participants, respectively, and the prevalence was higher in men than in women. In both genders, the prevalence of obesity, hypertension and diabetes increased with age. CONCLUSION: The prevalence of major CVRFs is high in the Lausanne population in particular in men. We anticipate that given its size, the depth of the phenotypic analysis and the availability of dense genome-wide genetic data, the CoLaus Study will be a unique resource to investigate not only the epidemiology of isolated, or aggregated CVRFs like the metabolic syndrome, but can also serve as a discovery set, as well as replication set, to identify novel genes associated with these conditions.
- 18Rueedi, R.; Ledda, M.; Nicholls, A. W.; Salek, R. M.; Marques-Vidal, P.; Morya, E. Genome-wide association study of metabolic traits reveals novel gene-metabolite-disease links. PLoS Genet. 2014, 10, e1004132, DOI: 10.1371/journal.pgen.1004132Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXkvVOnsbg%253D&md5=69bc097be50d0028080e1391f85db825Genome-wide association study of metabolic traits reveals novel gene-metabolite-disease linksRueedi, Rico; Ledda, Mirko; Nicholls, Andrew W.; Salek, Reza M.; Marques-Vidal, Pedro; Morya, Edgard; Sameshima, Koichi; Montoliu, Ivan; Da Silva, Laeticia; Collino, Sebastiano; Martin, Francois-Pierre; Rezzi, Serge; Steinbeck, Christoph; Waterworth, Dawn M.; Waeber, Gerard; Vollenweider, Peter; Beckmann, Jacques S.; Le Coutre, Johannes; Mooser, Vincent; Bergmann, Sven; Genick, Ulrich K.; Kutalik, ZoltanPLoS Genetics (2014), 10 (2), e1004132/1-e1004132/10, 10 pp.CODEN: PGLEB5; ISSN:1553-7404. (Public Library of Science)Metabolic traits are mol. phenotypes that can drive clin. phenotypes and may predict disease progression. Here, we report results from a metabolome- and genome-wide assocn. study on 1H-NMR urine metabolic profiles. The study was conducted within an untargeted approach, employing a novel method for compd. identification. From our discovery cohort of 835 Caucasian individuals who participated in the CoLaus study, we identified 139 suggestively significant (P<5 × 10-8) and independent assocns. between single nucleotide polymorphisms (SNP) and metabolome features. Fifty-six of these assocns. replicated in the TasteSensomics cohort, comprising 601 individuals from S~ao Paulo of vastly diverse ethnic background. They correspond to eleven gene-metabolite assocns., six of which had been previously identified in the urine metabolome and three in the serum metabolome. Our key novel findings are the assocns. of two SNPs with NMR spectral signatures pointing to fucose (rs492602, P = 6.9 × 10-44) and lysine (rs8101881, P = 1.2 × 10-33), resp. Fine-mapping of the first locus pinpointed the FUT2 gene, which encodes a fucosyltransferase enzyme and has previously been assocd. with Crohn's disease. This implicates fucose as a potential prognostic disease marker, for which there is already published evidence from a mouse model. The second SNP lies within the SLC7A9 gene, rare mutations of which have been linked to severe kidney damage. The replication of previous assocns. and our new discoveries demonstrate the potential of untargeted metabolomics GWAS to robustly identify mol. disease markers.
- 19Marco-Sola, S.; Sammeth, M.; Guigó, R.; Ribeca, P. The GEM mapper: fast, accurate and versatile alignment by filtration. Nat. Methods 2012, 9, 1185, DOI: 10.1038/nmeth.2221Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsFOmt73O&md5=559ff724a6a064d7280d553793f21d4fThe GEM mapper: fast, accurate and versatile alignment by filtrationMarco-Sola, Santiago; Sammeth, Michael; Guigo, Roderic; Ribeca, PaoloNature Methods (2012), 9 (12), 1185-1188CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)Because of ever-increasing throughput requirements of sequencing data, most existing short-read aligners have been designed to focus on speed at the expense of accuracy. The Genome Multitool (GEM) mapper can leverage string matching by filtration to search the alignment space more efficiently, simultaneously delivering precision (performing fully tunable exhaustive searches that return all existing matches, including gapped ones) and speed (being several times faster than comparable state-of-the-art tools).
- 20MATLAB 8.5.0.197613 (R2015a); The MathWorks Inc.: Natick, Massachusetts, 2015.Google ScholarThere is no corresponding record for this reference.
- 21Gao, X.; Starmer, J.; Martin, E. R. A multiple testing correction method for genetic association studies using correlated single nucleotide polymorphisms. Genet. Epidemiol. 2008, 32, 361– 369, DOI: 10.1002/gepi.20310Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1c3msl2gug%253D%253D&md5=ea3ae76ef9371956e694e05c166ebbceA multiple testing correction method for genetic association studies using correlated single nucleotide polymorphismsGao Xiaoyi; Starmer Joshua; Martin Eden RGenetic epidemiology (2008), 32 (4), 361-9 ISSN:0741-0395.Multiple testing is a challenging issue in genetic association studies using large numbers of single nucleotide polymorphism (SNP) markers, many of which exhibit linkage disequilibrium (LD). Failure to adjust for multiple testing appropriately may produce excessive false positives or overlook true positive signals. The Bonferroni method of adjusting for multiple comparisons is easy to compute, but is well known to be conservative in the presence of LD. On the other hand, permutation-based corrections can correctly account for LD among SNPs, but are computationally intensive. In this work, we propose a new multiple testing correction method for association studies using SNP markers. We show that it is simple, fast and more accurate than the recently developed methods and is comparable to permutation-based corrections using both simulated and real data. We also demonstrate how it might be used in whole-genome association studies to control type I error. The efficiency and accuracy of the proposed method make it an attractive choice for multiple testing adjustment when there is high intermarker LD in the SNP data set.
- 22Rueedi, R.; Mallol, R.; Raffler, J.; Lamparter, D.; Friedrich, N.; Vollenweider, P. Metabomatching: Using genetic association to identify metabolites in proton NMR spectroscopy. PLoS Comput. Biol. 2017, 13, e1005839, DOI: 10.1371/journal.pcbi.1005839Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsFags7vK&md5=842831a300f5401d3db64e43c372ce62Metabomatching: using genetic association to identify metabolites in proton NMR spectroscopyRueedi, Rico; Mallol, Roger; Raffler, Johannes; Lamparter, David; Friedrich, Nele; Vollenweider, Peter; Waeber, Gerard; Kastenmuller, Gabi; Kutalik, Zoltan; Bergmann, SvenPLoS Computational Biology (2017), 13 (12), e1005839/1-e1005839/17CODEN: PCBLBG; ISSN:1553-7358. (Public Library of Science)A metabolome-wide genome-wide assocn. study aims to discover the effects of genetic variants on metabolome phenotypes. Most mGWASes use as phenotypes concns. of limited sets of metabolites that can be identified and quantified from spectral information. In contrast, in an untargeted mGWAS both identification and quantification are forgone and, instead, all measured metabolome features are tested for assocn. with genetic variants. While the untargeted approach does not discard data that may have eluded identification, the interpretation of assocd. features remains a challenge. To address this issue, we developed metabomatching to identify the metabolites underlying significant assocns. obsd. in untargeted mGWASes on proton NMR metabolome data. Metabomatching capitalizes on genetic spiking, the concept that because metabolome features assocd. with a genetic variant tend to correspond to the peaks of the NMR spectrum of the underlying metabolite, genetic assocn. can allow for identification. Applied to the untargeted mGWASes in the SHIP and CoLaus cohorts and using 180 ref. NMR spectra of the urine metabolome database, metabomatching successfully identified the underlying metabolite in 14 of 19, and 8 of 9 assocns., resp. The accuracy and efficiency of our method make it a strong contender for facilitating or complementing metabolomics analyses in large cohorts, where the availability of genetic, or other data, enables our approach, but targeted quantification is limited.
- 23Wishart, D. S.; Feunang, Y. D.; Marcu, A.; Guo, A. C.; Liang, K.; Vázquez-Fresno, R. HMDB 4.0: the human metabolome database for 2018. Nucleic Acids Res. 2018, 46, D608– D617, DOI: 10.1093/nar/gkx1089Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitlGisb%252FF&md5=29725f192d00a0e31a35401058a32082HMDB 4.0: the human metabolome database for 2018Wishart, David S.; Feunang, Yannick Djoumbou; Marcu, Ana; Guo, An Chi; Liang, Kevin; Vazquez-Fresno, Rosa; Sajed, Tanvir; Johnson, Daniel; Li, Carin; Karu, Naama; Sayeeda, Zinat; Lo, Elvis; Assempour, Nazanin; Berjanskii, Mark; Singhal, Sandeep; Arndt, David; Liang, Yonjie; Badran, Hasan; Grant, Jason; Serra-Cayuela, Arnau; Liu, Yifeng; Mandal, Rupa; Neveu, Vanessa; Pon, Allison; Knox, Craig; Wilson, Michael; Manach, Claudine; Scalbert, AugustinNucleic Acids Research (2018), 46 (D1), D608-D617CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)A review. The Human Metabolome Database or HMDB (www. hmdb.ca) is a web-enabled metabolomic database contg. comprehensive information about human metabolites along with their biol. roles, physiol. concns., disease assocns., chem. reactions, metabolic pathways, and ref. spectra. First described in 2007, the HMDB is now considered the std. metabolomic resource for human metabolic studies. Over the past decade the HMDB has continued to grow and evolve in response to emerging needs for metabolomics researchers and continuing changes in web stds. This year's update, HMDB 4.0, represents the most significant upgrade to the database in its history. For instance, the no. of fully annotated metabolites has increased by nearly threefold, the no. of exptl. spectra has grown by almost fourfold and the no. of illustrated metabolic pathways has grown by a factor of almost 60. Significant improvements have also been made to the HMDB's chem. taxonomy, chem. ontol., spectral viewing, and spectral/text searching tools. A great deal of brand new data has also been added to HMDB 4.0. This includes large quantities of predicted MS/MS and GC-MS ref. spectral data as well as predicted (physiol. feasible) metabolite structures to facilitate novel metabolite identification. Addnl. information on metabolite-SNP interactions and the influence of drugs on metabolite levels (pharmaco-metabolomics) has also been added. Many other important improvements in the content, the interface, and the performance of the HMDB website have been made and these should greatly enhance its ease of use and its potential applications in nutrition, biochem., clin. chem., clin. genetics, medicine, and metabolomics science.
- 24Khalili, B.; Tomasoni, M.; Mattei, M.; Parera, R. M.; Sonmez, R.; Krefl, D. Automated analysis of large-scale NMR data generates metabolomic signatures and links them to candidate metabolites. J. Proteome Res. 2019, 613935, DOI: 10.1021/acs.jproteome.9b00295Google ScholarThere is no corresponding record for this reference.
- 25Burgess, S.; Small, D. S.; Thompson, S. G. A review of instrumental variable estimators for Mendelian randomization. Stat. Methods Med. Res. 2017, 26, 2333– 2355, DOI: 10.1177/0962280215597579Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC287jsl2quw%253D%253D&md5=6c22b011ffcad08533fbaa060bf97b9bA review of instrumental variable estimators for Mendelian randomizationBurgess Stephen; Thompson Simon G; Small Dylan SStatistical methods in medical research (2017), 26 (5), 2333-2355 ISSN:.Instrumental variable analysis is an approach for obtaining causal inferences on the effect of an exposure (risk factor) on an outcome from observational data. It has gained in popularity over the past decade with the use of genetic variants as instrumental variables, known as Mendelian randomization. An instrumental variable is associated with the exposure, but not associated with any confounder of the exposure-outcome association, nor is there any causal pathway from the instrumental variable to the outcome other than via the exposure. Under the assumption that a single instrumental variable or a set of instrumental variables for the exposure is available, the causal effect of the exposure on the outcome can be estimated. There are several methods available for instrumental variable estimation; we consider the ratio method, two-stage methods, likelihood-based methods, and semi-parametric methods. Techniques for obtaining statistical inferences and confidence intervals are presented. The statistical properties of estimates from these methods are compared, and practical advice is given about choosing a suitable analysis method. In particular, bias and coverage properties of estimators are considered, especially with weak instruments. Settings particularly relevant to Mendelian randomization are prioritized in the paper, notably the scenario of a continuous exposure and a continuous or binary outcome.
- 26Davey Smith, G.; Ebrahim, S. ‘Mendelian randomization’: can genetic epidemiology contribute to understanding environmental determinants of disease?. Int. J. Epidemiol. 2003, 32, 1– 22, DOI: 10.1093/ije/dyg070Google ScholarThere is no corresponding record for this reference.
- 27Raffler, J.; Friedrich, N.; Arnold, M.; Kacprowski, T.; Rueedi, R.; Altmaier, E. Genome-wide association study with targeted and non-targeted NMR metabolomics identifies 15 novel loci of urinary human metabolic individuality. PLoS Genet. 2015, 11, e1005487, DOI: 10.1371/journal.pgen.1005487Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xls1ygu7g%253D&md5=46d58208d9b118aad16308ff59f3b0daGenome-wide association study with targeted and non-targeted nmr metabolomics identifies 15 novel loci of urinary human metabolic individualityRaffler, Johannes; Friedrich, Nele; Arnold, Matthias; Kacprowski, Tim; Rueedi, Rico; Altmaier, Elisabeth; Bergmann, Sven; Budde, Kathrin; Gieger, Christian; Homuth, Georg; Pietzner, Maik; Roemisch-Margl, Werner; Strauch, Konstantin; Voelzke, Henry; Waldenberger, Melanie; Wallaschofski, Henri; Nauck, Matthias; Voelker, Uwe; Kastenmueller, Gabi; Suhre, KarstenPLoS Genetics (2015), 11 (9), e1005487/1-e1005487/28CODEN: PGLEB5; ISSN:1553-7404. (Public Library of Science)Genome-wide assocn. studies with metabolic traits (mGWAS) uncovered many genetic variants that influence human metab. These genetically influenced metabotypes (GIMs) contribute to our metabolic individuality, our capacity to respond to environmental challenges, and our susceptibility to specific diseases. While metabolic homeostasis in blood is a well investigated topic in large mGWAS with over 150 known loci, metabolic detoxification through urinary excretion has only been addressed by few small mGWAS with only 11 assocd. loci so far. Here we report the largest mGWAS to date, combining targeted and non-targeted 1H NMR anal. of urine samples from 3,861 participants of the SHIP-0 cohort and 1,691 subjects of the KORA F4 cohort. We identified and replicated 22 loci with significant assocns. with urinary traits, 15 of which are new (HIBCH, CPS1, AGXT, XYLB, TKT, ETNPPL, SLC6A19, DMGDH, SLC36A2, GLDC, SLC6A13, ACSM3, SLC5A11, PNMT, SLC13A3). Two-thirds of the urinary loci also have a metabolite assocn. in blood. For all but one of the 6 loci where significant assocns. target the same metabolite in blood and urine, the genetic effects have the same direction in both fluids. In contrast, for the SLC5A11 locus, we found increased levels of myo-inositol in urine whereas mGWAS in blood reported decreased levels for the same genetic variant. This might indicate less effective re-absorption of myo-inositol in the kidneys of carriers. In summary, our study more than doubles the no. of known loci that influence urinary phenotypes. It thus allows novel insights into the relationship between blood homeostasis and its regulation through excretion. The newly discovered loci also include variants previously linked to chronic kidney disease (CPS1, SLC6A13), pulmonary hypertension (CPS1), and ischemic stroke (XYLB). By establishing connections from gene to disease via metabolic traits our results provide novel hypotheses about mol. mechanisms involved in the etiol. of diseases.
- 28Võsa, U.; Claringbould, A.; Westra, H.-J.; Bonder, M. J.; Deelen, P.; Zeng, B.; Kirsten, H.; Saha, A.; Kreuzhuber, R.; Brugge, H. Large-scale cis-and trans-eQTL analyses identify thousands of genetic loci and polygenic scores that regulate blood gene expression. Nat. Genet. 2021, 53, 1300– 1310, DOI: 10.1038/s41588-021-00913-zGoogle Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvFGmtbfE&md5=d78fcc21a5127342cbdddadc10a596fdLarge-scale cis- and trans-eQTL analyses identify thousands of genetic loci and polygenic scores that regulate blood gene expressionVosa, Urmo; Claringbould, Annique; Westra, Harm-Jan; Bonder, Marc Jan; Deelen, Patrick; Zeng, Biao; Kirsten, Holger; Saha, Ashis; Kreuzhuber, Roman; Yazar, Seyhan; Brugge, Harm; Oelen, Roy; de Vries, Dylan H.; van der Wijst, Monique G. P.; Kasela, Silva; Pervjakova, Natalia; Alves, Isabel; Fave, Marie-Julie; Agbessi, Mawusse; Christiansen, Mark W.; Jansen, Rick; Seppala, Ilkka; Tong, Lin; Teumer, Alexander; Schramm, Katharina; Hemani, Gibran; Verlouw, Joost; Yaghootkar, Hanieh; Sonmez Flitman, Reyhan; Brown, Andrew; Kukushkina, Viktorija; Kalnapenkis, Anette; Rueger, Sina; Porcu, Eleonora; Kronberg, Jaanika; Kettunen, Johannes; Lee, Bernett; Zhang, Futao; Qi, Ting; Hernandez, Jose Alquicira; Arindrarto, Wibowo; Beutner, Frank; Dmitrieva, Julia; Elansary, Mahmoud; Fairfax, Benjamin P.; Georges, Michel; Heijmans, Bastiaan T.; Hewitt, Alex W.; Kahonen, Mika; Kim, Yungil; Knight, Julian C.; Kovacs, Peter; Krohn, Knut; Li, Shuang; Loeffler, Markus; Marigorta, Urko M.; Mei, Hailang; Momozawa, Yukihide; Muller-Nurasyid, Martina; Nauck, Matthias; Nivard, Michel G.; Penninx, Brenda W. J. H.; Pritchard, Jonathan K.; Raitakari, Olli T.; Rotzschke, Olaf; Slagboom, Eline P.; Stehouwer, Coen D. A.; Stumvoll, Michael; Sullivan, Patrick; 't Hoen, Peter A. C.; Thiery, Joachim; Tonjes, Anke; van Dongen, Jenny; van Iterson, Maarten; Veldink, Jan H.; Volker, Uwe; Warmerdam, Robert; Wijmenga, Cisca; Swertz, Morris; Andiappan, Anand; Montgomery, Grant W.; Ripatti, Samuli; Perola, Markus; Kutalik, Zoltan; Dermitzakis, Emmanouil; Bergmann, Sven; Frayling, Timothy; van Meurs, Joyce; Prokisch, Holger; Ahsan, Habibul; Pierce, Brandon L.; Lehtimaki, Terho; Boomsma, Dorret I.; Psaty, Bruce M.; Gharib, Sina A.; Awadalla, Philip; Milani, Lili; Ouwehand, Willem H.; Downes, Kate; Stegle, Oliver; Battle, Alexis; Visscher, Peter M.; Yang, Jian; Scholz, Markus; Powell, Joseph; Gibson, Greg; Esko, Tonu; Franke, LudeNature Genetics (2021), 53 (9), 1300-1310CODEN: NGENEC; ISSN:1061-4036. (Nature Portfolio)Trait-assocd. genetic variants affect complex phenotypes primarily via regulatory mechanisms on the transcriptome. To investigate the genetics of gene expression, we performed cis- and trans-expression quant. trait locus (eQTL) analyses using blood-derived expression from 31,684 individuals through the eQTLGen Consortium. We detected cis-eQTL for 88% of genes, and these were replicable in numerous tissues. Distal trans-eQTL (detected for 37% of 10,317 trait-assocd. variants tested) showed lower replication rates, partially due to low replication power and confounding by cell type compn. However, replication analyses in single-cell RNA-seq data prioritized intracellular trans-eQTL. Trans-eQTL exerted their effects via several mechanisms, primarily through regulation by transcription factors. Expression of 13% of the genes correlated with polygenic scores for 1,263 phenotypes, pinpointing potential drivers for those traits. In summary, this work represents a large eQTL resource, and its results serve as a starting point for in-depth interpretation of complex phenotypes.
- 29Wald, A. The fitting of straight lines if both variables are subject to error. Ann. Math. Stat. 1940, 11, 284– 300, DOI: 10.1214/aoms/1177731868Google ScholarThere is no corresponding record for this reference.
- 30Hartung, J.; Knapp, G.; Sinha, B. K.; Sinha, B. K. Statistical meta-analysis with applications; Wiley: New York, 2008.Google ScholarThere is no corresponding record for this reference.
- 31Greco, M. F. D.; Minelli, C.; Sheehan, N. A.; Thompson, J. R. Detecting pleiotropy in Mendelian randomisation studies with summary data and a continuous outcome. Stat. Med. 2015, 34, 2926– 2940, DOI: 10.1002/sim.6522Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2MfhtlCjtg%253D%253D&md5=dab1f5ee1ce3851064ebee6727d9bf16Detecting pleiotropy in Mendelian randomisation studies with summary data and a continuous outcomeGreco M Fabiola Del; Minelli Cosetta; Sheehan Nuala A; Thompson John RStatistics in medicine (2015), 34 (21), 2926-40 ISSN:.Mendelian randomisation (MR) estimates causal effects of modifiable phenotypes on an outcome by using genetic variants as instrumental variables, but its validity relies on the assumption of no pleiotropy, that is, genes influence the outcome only through the given phenotype. Excluding pleiotropy is difficult, but the use of multiple instruments can indirectly address the issue: if all genes represent valid instruments, their MR estimates should vary only by chance. The Sargan test detects pleiotropy when individual phenotype, outcome and genotype data are measured in the same subjects. We propose an alternative approach to be used when only summary genetic data are available or data on gene-phenotype and gene-outcome come from different subjects. The presence of pleiotropy is investigated using the between-instrument heterogeneity Q test (together with the I(2) index) in a meta-analysis of MR Wald estimates, derived separately from each instrument. For a continuous outcome, we evaluate the approach through simulations and illustrate it using published data. For the scenario where all data come from the same subjects, we compare it with the Sargan test. The Q test tends to be conservative in small samples. Its power increases with the degree of pleiotropy and the sample size, as does the precision of the I(2) index, in which case results are similar to those of the Sargan test. In MR studies with large sample sizes based on summary data, the between-instrument Q test represents a useful tool to explore the presence of heterogeneity due to pleiotropy or other causes.
- 32Yavorska, O. O.; Burgess, S. MendelianRandomization: an R package for performing Mendelian randomization analyses using summarized data. Int. J. Epidemiol. 2017, 46, 1734– 1739, DOI: 10.1093/ije/dyx034Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1cvmslehsA%253D%253D&md5=d8d13430779261f1a45cf0cbdde85f8eMendelianRandomization: an R package for performing Mendelian randomization analyses using summarized dataYavorska Olena O; Burgess StephenInternational journal of epidemiology (2017), 46 (6), 1734-1739 ISSN:.MendelianRandomization is a software package for the R open-source software environment that performs Mendelian randomization analyses using summarized data. The core functionality is to implement the inverse-variance weighted, MR-Egger and weighted median methods for multiple genetic variants. Several options are available to the user, such as the use of robust regression, fixed- or random-effects models and the penalization of weights for genetic variants with heterogeneous causal estimates. Extensions to these methods, such as allowing for variants to be correlated, can be chosen if appropriate. Graphical commands allow summarized data to be displayed in an interactive graph, or the plotting of causal estimates from multiple methods, for comparison. Although the main method of data entry is directly by the user, there is also an option for allowing summarized data to be incorporated from the PhenoScanner database of genotype-phenotype associations. We hope to develop this feature in future versions of the package. The R software environment is available for download from [https://www.r-project.org/]. The MendelianRandomization package can be downloaded from the Comprehensive R Archive Network (CRAN) within R, or directly from [https://cran.r-project.org/web/packages/MendelianRandomization/]. Both R and the MendelianRandomization package are released under GNU General Public Licenses (GPL-2 GPL-3).
- 33Shin, S.-Y.; Fauman, E. B.; Petersen, A.-K.; Krumsiek, J.; Santos, R.; Huang, J. An atlas of genetic influences on human blood metabolites. Nat. Genet. 2014, 46, 543, DOI: 10.1038/ng.2982Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXnslGktro%253D&md5=ba0b0c68c921ef6ea01105fce89818d4An atlas of genetic influences on human blood metabolitesShin, So-Youn; Fauman, Eric B.; Petersen, Ann-Kristin; Krumsiek, Jan; Santos, Rita; Huang, Jie; Arnold, Matthias; Erte, Idil; Forgetta, Vincenzo; Yang, Tsun-Po; Walter, Klaudia; Menni, Cristina; Chen, Lu; Vasquez, Louella; Valdes, Ana M.; Hyde, Craig L.; Wang, Vicky; Ziemek, Daniel; Roberts, Phoebe; Xi, Li; Grundberg, Elin; Waldenberger, Melanie; Richards, J. Brent; Mohney, Robert P.; Milburn, Michael V.; John, Sally L.; Trimmer, Jeff; Theis, Fabian J.; Overington, John P.; Suhre, Karsten; Brosnan, M. Julia; Gieger, Christian; Kastenmuller, Gabi; Spector, Tim D.; Soranzo, NicoleNature Genetics (2014), 46 (6), 543-550CODEN: NGENEC; ISSN:1061-4036. (Nature Publishing Group)Genome-wide assocn. scans with high-throughput metabolic profiling provide unprecedented insights into how genetic variation influences metab. and complex disease. Here the authors report the most comprehensive exploration of genetic loci influencing human metab. thus far, comprising 7,824 adult individuals from 2 European population studies. The authors report genome-wide significant assocns. at 145 metabolic loci and their biochem. connectivity with more than 400 metabolites in human blood. The authors extensively characterize the resulting in vivo blueprint of metab. in human blood by integrating it with information on gene expression, heritability and overlap with known loci for complex disorders, inborn errors of metab. and pharmacol. targets. The authors further developed a database and web-based resources for data mining and results visualization. The authors' findings provide new insights into the role of inherited variation in blood metabolic diversity and identify potential new opportunities for drug development and for understanding disease.
- 34Nicholson, G.; Rantalainen, M.; Li, J. V.; Maher, A. D.; Malmodin, D.; Ahmadi, K. R. A genome-wide metabolic QTL analysis in Europeans implicates two loci shaped by recent positive selection. PLoS Genet. 2011, 7, e1002270, DOI: 10.1371/journal.pgen.1002270Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXht1ChsbvE&md5=3115de8a4d2238d5ce02ef0ed273d58fA genome-wide metabolic QTL analysis in Europeans implicates two loci shaped by recent positive selectionNicholson, George; Rantalainen, Mattias; Li, Jia V.; Maher, Anthony D.; Malmodin, Daniel; Ahmadi, Kourosh R.; Faber, Johan H.; Barrett, Amy; Min, Josine L.; Rayner, N. William; Toft, Henrik; Krestyaninova, Maria; Viksna, Juris; Neogi, Sudeshna Guha; Dumas, Marc-Emmanuel; Sarkans, Ugis; Donnelly, Peter; Illig, Thomas; Adamski, Jerzy; Suhre, Karsten; Allen, Maxine; Zondervan, Krina T.; Spector, Tim D.; Nicholson, Jeremy K.; Lindon, John C.; Baunsgaard, Dorrit; Holmes, Elaine; McCarthy, Mark I.; Holmes, Chris C.PLoS Genetics (2011), 7 (9), e1002270CODEN: PGLEB5; ISSN:1553-7404. (Public Library of Science)We have performed a metabolite quant. trait locus (mQTL) study of the 1H NMR spectroscopy metabolome in humans, building on recent targeted knowledge of genetic drivers of metabolic regulation. Urine and plasma samples were collected from two cohorts of individuals of European descent, with one cohort comprised of female twins donating samples longitudinally. Sample metabolite concns. were quantified by 1H NMR and tested for assocn. with genome-wide single-nucleotide polymorphisms (SNPs). 4 Metabolites' concns. exhibited significant, replicable assocn. with SNP variation (8.6 × 10-11 < p < 2.8 × 10-23). 3 Of these, trimethylamine, 3-amino-isobutyrate, and an N-acetylated compd., were measured in urine. The other, dimethylamine, was measured in plasma. Trimethylamine and dimethylamine mapped to a single genetic region (hence we report a total of three implicated genomic regions). 2 Of the 3 hit regions lie within haplotype blocks (at 2p13.1 and 10q24.2) that carry the genetic signature of strong, recent, pos. selection in European populations. Genes NAT8 and PYROXD2, both with relatively uncharacterized functional roles, are good candidates for mediating the corresponding mQTL assocns. The studies longitudinal twin design allowed detailed variance-components anal. of the sources of population variation in metabolite levels. The mQTLs explained 40%-64% of biol. population variation in the corresponding metabolites' concns. These effect sizes are stronger than those reported in a recent, targeted mQTL study of metabolites in serum using the targeted-metabolomics Biocrates platform. By re-analyzing our plasma samples using the Biocrates platform, we replicated the mQTL findings of the previous study and discovered a previously uncharacterized yet substantial familial component of variation in metabolite levels in addn. to the heritability contribution from the corresponding mQTL effects.
- 35Engelke, U. F.; Liebrand-van Sambeek, M. L.; De Jong, J. G.; Leroy, J. G.; Morava, E.; Smeitink, J. A. N-acetylated metabolites in urine: proton nuclear magnetic resonance spectroscopic study on patients with inborn errors of metabolism. Clin. Chem. 2004, 50, 58– 66, DOI: 10.1373/clinchem.2003.020214Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXovFWhtw%253D%253D&md5=80e2e8bb8453ccce361195b7b8e695faN-acetylated metabolites in urine: Proton nuclear magnetic resonance spectroscopic study on patients with inborn errors of metabolismEngelke, Udo F. H.; Liebrand-van Sambeek, Maria L. F.; de Jong, Jan G. N.; Leroy, Jules G.; Morava, Eva; Smeitink, Jan A. M.; Wevers, Ron A.Clinical Chemistry (Washington, DC, United States) (2004), 50 (1), 58-66CODEN: CLCHAU; ISSN:0009-9147. (American Association for Clinical Chemistry)There is no comprehensive anal. technique to analyze N-acetylated metabolites in urine. Many of these compds. are involved in inborn errors of metab. In the present study, the authors examd. the potential of proton NMR (1H-NMR) spectroscopy as a tool to identify and quantify N-acetylated metabolites in urine of patients with various inborn errors of metab. The authors performed 1H-NMR spectroscopy on a 500 MHz spectrometer. Using a combination of 1- and 2-dimensional correlation spectroscopy (COSY) 1H-NMR spectra, the authors were able to assign and quantify resonances of characteristic N-acetylated compds. products in urine of patients with 13 inborn errors of metab. The disease-specific N-acetylated metabolites were excreted at concns. >100 μmol/mmol of creatinine in the patients' urine. In control urine samples, the concn. of individual N-acetyl-contg. compds. was <40 μmol/mmol of creatinine. The combination of one- and two-dimensional COSY NMR spectroscopy led to the correct diagnosis of nine different inborn errors of metab. No abnormalities were obsd. in the spectra of urine from patients with GM1- or GM2-gangliosidosis. The authors also detd. the 1H-NMR characteristics of N-acetylated metabolites that may be relevant to human metab. 1H-NMR spectroscopy may be used to identify and quantify N-acetylated metabolites of diagnostic importance for the field of inborn errors of metab.
- 36Benjamini, Y.; Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. Royal Stat. Soc.: B 1995, 57, 289– 300, DOI: 10.1111/j.2517-6161.1995.tb02031.xGoogle ScholarThere is no corresponding record for this reference.
- 37Kastenmüller, G.; Raffler, J.; Gieger, C.; Suhre, K. Genetics of human metabolism: an update. Hum. Mol. Genet. 2015, 24, R93– R101, DOI: 10.1093/hmg/ddv263Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXitVCqu7nP&md5=a421ead21475ebed82390dbead5d6d97Genetics of human metabolism: an updateKastenmueller, Gabi; Raffler, Johannes; Gieger, Christian; Suhre, KarstenHuman Molecular Genetics (2015), 24 (Rev. Issue 1), R93-R101CODEN: HMGEE5; ISSN:0964-6906. (Oxford University Press)A review. Genome-wide assocn. studies with metabolomics (mGWAS) identify genetically influenced metabotypes (GIMs), their ensemble defining the heritable part of every human's metabolic individuality. Knowledge of genetic variation in metab. has many applications of biomedical and pharmaceutical interests, including the functional understanding of genetic assocns. with clin. end points, design of strategies to correct dysregulations in metabolic disorders and the identification of genetic effect modifiers of metabolic disease biomarkers. Furthermore, it has been shown that GIMs provide testable hypotheses for functional genomics and metabolomics and for the identification of novel gene functions and metabolite identities. mGWAS with growing sample sizes and increasingly complex metabolic trait panels are being conducted, allowing for more comprehensive and systems-based downstream analyses. The generated large datasets of genetic assocns. can now be mined by the biomedical research community and provide valuable resources for hypothesis-driven studies. In this review, we provide a brief summary of the key aspects of mGWAS, followed by an update of recently published mGWAS. We then discuss new approaches of integrating and exploring mGWAS results and finish by presenting selected applications of GIMs in recent studies.
- 38Suhre, K.; Shin, S. Y.; Petersen, A. K.; Mohney, R. P.; Meredith, D.; Wägele, B. Human metabolic individuality in biomedical and pharmaceutical research. Nature 2011, 477, 54– 60, DOI: 10.1038/nature10354Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtFWku7nN&md5=da346121e93b1d123d72dc4c0b634833Human metabolic individuality in biomedical and pharmaceutical researchSuhre, Karsten; Shin, So-Youn; Petersen, Ann-Kristin; Mohney, Robert P.; Meredith, David; Waegele, Brigitte; Altmaier, Elisabeth; Deloukas, Panos; Erdmann, Jeanette; Grundberg, Elin; Hammond, Christopher J.; Hrabe de Angelis, Martin; Kastenmueller, Gabi; Koettgen, Anna; Kronenberg, Florian; Mangino, Massimo; Meisinger, Christa; Meitinger, Thomas; Mewes, Hans-Werner; Milburn, Michael V.; Prehn, Cornelia; Raffler, Johannes; Ried, Janina S.; Roemisch-Margl, Werner; Samani, Nilesh J.; Small, Kerrin S.; Wichmann, H.-Erich; Zhai, Guang-Ju; Illig, Thomas; Spector, Tim D.; Adamski, Jerzy; Soranzo, Nicole; Gieger, ChristianNature (London, United Kingdom) (2011), 477 (7362), 54-60CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)A review based on author's and other researchers studies. Genome-wide assocn. studies (GWAS) have identified many risk loci for complex diseases, but effect sizes are typically small and information on the underlying biol. processes is often lacking. Assocns. with metabolic traits as functional intermediates can overcome these problems and potentially inform individualized therapy. Here we report a comprehensive anal. of genotype-dependent metabolic phenotypes using a GWAS with non-targeted metabolomics. We identified 37 genetic loci assocd. with blood metabolite concns., of which 25 show effect sizes that are unusually high for GWAS and account for 10-60% differences in metabolite levels per allele copy. Our assocns. provide new functional insights for many disease-related assocns. that have been reported in previous studies, including those for cardiovascular and kidney disorders, type 2 diabetes, cancer, gout, venous thromboembolism and Crohn's disease. The study advances our knowledge of the genetic basis of metabolic individuality in humans and generates many new hypotheses for biomedical and pharmaceutical research.
- 39Yu, B.; Zheng, Y.; Alexander, D.; Morrison, A. C.; Coresh, J.; Boerwinkle, E. Genetic determinants influencing human serum metabolome among African Americans. PLoS Genet. 2014, 10, e1004212 DOI: 10.1371/journal.pgen.1004212Google Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhsVGlsLnI&md5=8dc1533231f3e76be1739d1a8b1fc491Genetic determinants influencing human serum metabolome among African AmericansYu, Bing; Zheng, Yan; Alexander, Danny; Morrison, Alanna C.; Coresh, Josef; Boerwinkle, EricPLoS Genetics (2014), 10 (3), e1004212/1-e1004212/8, 8 pp.CODEN: PGLEB5; ISSN:1553-7404. (Public Library of Science)Phenotypes proximal to gene action generally reflect larger genetic effect sizes than those that are distant. The human metabolome, a result of multiple cellular and biol. processes, are functional intermediate phenotypes proximal to gene action. Here, we present a genome-wide assocn. study of 308 untargeted metabolite levels among African Americans from the Atherosclerosis Risk in Communities (ARIC) Study. Nineteen significant common variant-metabolite assocns. were identified, including 13 novel loci (p<1.6 × 1010). These loci were assocd. with 7-50 % of the difference in metabolite levels per allele, and the variance explained ranged from 4% to 20%. Fourteen genes were identified within the nineteen loci, and four of them contained non-synonymous substitutions in four enzyme-encoding genes (KLKB1, SIAE, CPS1, and NAT8); the other significant loci consist of eight other enzyme-encoding genes (ACE, GATM, ACY3, ACSM2B, THEM4, ADH4, UGT1A, TREH), a transporter gene (SLC6A13) and a polycystin protein gene (PKD2L1). In addn., four potential disease-assocd. paths were identified, including two direct longitudinal predictive relationships: NAT8 with N-acetylornithine, N-acetyl-1-methylhistidine and incident chronic kidney disease, and TREH with trehalose and incident diabetes. These results highlight the value of using endophenotypes proximal to gene function to discover new insights into biol. and disease pathol.
- 40Raffler, J.; Römisch-Margl, W.; Petersen, A. K.; Pagel, P.; Blöchl, F.; Hengstenberg, C. Identification and MS-assisted interpretation of genetically influenced NMR signals in human plasma. Genome Med. 2013, 5, 13, DOI: 10.1186/gm417Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtVGqtbnE&md5=cf4b62ba8345c74d16110c4f7ab47396Identification and MS-assisted interpretation of genetically influenced NMR signals in human plasmaRaffler, Johannes; Roemisch-Margl, Werner; Petersen, Ann-Kristin; Pagel, Philipp; Bloechl, Florian; Hengstenberg, Christian; Illig, Thomas; Meisinger, Christa; Stark, Klaus; Wichmann, H.-Erich; Adamski, Jerzy; Gieger, Christian; Kastenmueller, Gabi; Suhre, KarstenGenome Medicine (2013), 5 (2), 13CODEN: GMEECG; ISSN:1756-994X. (BioMed Central Ltd.)NMR spectroscopy provides robust readouts of many metabolic parameters in one expt. However, identification of clin. relevant markers in 1H NMR spectra is a major challenge. Assocn. of NMR-derived quantities with genetic variants can uncover biol. relevant metabolic traits. Using NMR data of plasma samples from 1,757 individuals from the KORA study together with 655,658 genetic variants, we show that ratios between NMR intensities at two chem. shift positions can provide informative and robust biomarkers. We report seven loci of genetic assocn. with NMR-derived traits (APOA1, CETP, CPS1, GCKR, FADS1, LIPC, PYROXD2) and characterize these traits biochem. using mass spectrometry. These ratios may now be used in clin. studies.
- 41Rhee, E. P.; Ho, J. E.; Chen, M. H.; Shen, D.; Cheng, S.; Larson, M. G. A genome-wide association study of the human metabolome in a community-based cohort. Cell Metab. 2013, 18, 130– 143, DOI: 10.1016/j.cmet.2013.06.013Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtVGrsr7P&md5=dcaac8e4c86e2360ad15c820c2839998A Genome-wide Association Study of the Human Metabolome in a Community-Based CohortRhee, Eugene P.; Ho, Jennifer E.; Chen, Ming-Huei; Shen, Dongxiao; Cheng, Susan; Larson, Martin G.; Ghorbani, Anahita; Shi, Xu; Helenius, Iiro T.; O'Donnell, Christopher J.; Souza, Amanda L.; Deik, Amy; Pierce, Kerry A.; Bullock, Kevin; Walford, Geoffrey A.; Vasan, Ramachandran S.; Florez, Jose C.; Clish, Clary; Yeh, J.-R. Joanna; Wang, Thomas J.; Gerszten, Robert E.Cell Metabolism (2013), 18 (1), 130-143CODEN: CMEEB5; ISSN:1550-4131. (Elsevier Inc.)Because metabolites are hypothesized to play key roles as markers and effectors of cardiometabolic diseases, recent studies have sought to annotate the genetic determinants of circulating metabolite levels. We report a genome-wide assocn. study (GWAS) of 217 plasma metabolites, including >100 not measured in prior GWAS, in 2076 participants of the Framingham Heart Study (FHS). For the majority of analytes, we find that estd. heritability explains >20% of interindividual variation, and that variation attributable to heritable factors is greater than that attributable to clin. factors. Further, we identify 31 genetic loci assocd. with plasma metabolites, including 23 that have not previously been reported. Importantly, we include GWAS results for all surveyed metabolites and demonstrate how this information highlights a role for AGXT2 in cholesterol ester and triacylglycerol metab. Thus, our study outlines the relative contributions of inherited and clin. factors on the plasma metabolome and provides a resource for metab. research.
- 42Krumsiek, J.; Suhre, K.; Evans, A. M.; Mitchell, M. W.; Mohney, R. P.; Milburn, M. V. Mining the unknown: a systems approach to metabolite identification combining genetic and metabolic information. PLoS Genet. 2012, 8, e1003005, DOI: 10.1371/journal.pgen.1003005Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xhs1antr7N&md5=cb9c81f3de0729332ee6b22a7c935c96Mining the unknown: a systems approach to metabolite identification combining genetic and metabolic informationKrumsiek, Jan; Suhre, Karsten; Evans, Anne M.; Mitchell, Matthew W.; Mohney, Robert P.; Milburn, Michael V.; Waegele, Brigitte; Roemisch-Margl, Werner; Illig, Thomas; Adamski, Jerzy; Gieger, Christian; Theis, Fabian J.; Kastenmueller, GabiPLoS Genetics (2012), 8 (10), e1003005CODEN: PGLEB5; ISSN:1553-7404. (Public Library of Science)Recent genome-wide assocn. studies (GWAS) with metabolomics data linked genetic variation in the human genome to differences in individual metabolite levels. A strong relevance of this metabolic individuality for biomedical and pharmaceutical research has been reported. However, a considerable amt. of the mols. currently quantified by modern metabolomics techniques are chem. unidentified. The identification of these "unknown metabolites" is still a demanding and intricate task, limiting their usability as functional markers of metabolic processes. As a consequence, previous GWAS largely ignored unknown metabolites as metabolic traits for the anal. Here we present a systems-level approach that combines genome-wide assocn. anal. and Gaussian graphical modeling with metabolomics to predict the identity of the unknown metabolites. We apply our method to original data of 517 metabolic traits, of which 225 are unknowns, and genotyping information on 655,658 genetic variants, measured in 1,768 human blood samples. We report previously undescribed genotype-metabotype assocns. for six distinct gene loci (SLC22A2, COMT, CYP3A5, CYP2C18, GBA3, UGT3A1) and one locus not related to any known gene (rs12413935). Overlaying the inferred genetic assocns., metabolic networks, and knowledge-based pathway information, we derive testable hypotheses on the biochem. identities of 106 unknown metabolites. As a proof of principle, we exptl. confirm nine concrete predictions. We demonstrate the benefit of our method for the functional interpretation of previous metabolomics biomarker studies on liver detoxification, hypertension, and insulin resistance. Our approach is generic in nature and can be directly transferred to metabolomics data from different exptl. platforms.
- 43Hong, M. G.; Karlsson, R.; Magnusson, P. K.; Lewis, M. R.; Isaacs, W.; Zheng, L. S. A Genome-Wide Assessment of Variability in Human Serum Metabolism. Hum. Mutat. 2013, 34, 515– 524, DOI: 10.1002/humu.22267Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXivFKktL0%253D&md5=79fda7875ae6787b539bc283cadc3221A Genome-Wide Assessment of Variability in Human Serum MetabolismHong, Mun-Gwan; Karlsson, Robert; Magnusson, Patrik K. E.; Lewis, Matthew R.; Isaacs, William; Zheng, Lilly S.; Xu, Jianfeng; Groenberg, Henrik; Ingelsson, Erik; Pawitan, Yudi; Broeckling, Corey; Prenni, Jessica E.; Wiklund, Fredrik; Prince, Jonathan A.Human Mutation (2013), 34 (3), 515-524CODEN: HUMUE3; ISSN:1059-7794. (Wiley-Liss, Inc.)The study of the genetic regulation of metab. in human serum samples can contribute to a better understanding of the intermediate biol. steps that lead from polymorphism to disease. Here, we conducted a genome-wide assocn. study (GWAS) to discover metabolic quant. trait loci (mQTLs) utilizing samples from a study of prostate cancer in Swedish men, consisting of 402 individuals (214 cases and 188 controls) in a discovery set and 489 case-only samples in a replication set. A global nontargeted metabolite profiling approach was utilized resulting in the detection of 6,138 mol. features followed by targeted identification of assocd. metabolites. Seven replicating loci were identified (PYROXD2, FADS1, PON1, CYP4F2, UGT1A8, ACADL, and LIPC) with assocd. sequence variants contributing significantly to trait variance for one or more metabolites (P = 10-13-10-91). Regional mQTL enrichment analyses implicated two loci that included FADS1 and a novel locus near PDGFC. Biol. pathway anal. implicated ACADM, ACADS, ACAD8, ACAD10, ACAD11, and ACOXL, reflecting significant enrichment of genes with acyl-CoA dehydrogenase activity. mQTL SNPs and mQTL-harboring genes were over-represented across GWASs conducted to date, suggesting that these data may have utility in tracing the mol. basis of some complex disease assocns.
- 44Montoliu, I.; Genick, U.; Ledda, M.; Collino, S.; Martin, F.-P.; le Coutre, J. Current status on genome–metabolome-wide associations: an opportunity in nutrition research. Gene Nutr. 2013, 8, 19, DOI: 10.1007/s12263-012-0313-7Google ScholarThere is no corresponding record for this reference.
- 45Chambers, J. C.; Zhang, W.; Lord, G. M.; Van Der Harst, P.; Lawlor, D. A.; Sehmi, J. S. Genetic loci influencing kidney function and chronic kidney disease. Nat. Genet. 2010, 42, 373– 375, DOI: 10.1038/ng.566Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXksFakurk%253D&md5=0c2bb874fec4125b00fc801bc8359464Genetic loci influencing kidney function and chronic kidney diseaseChambers, John C.; Zhang, Weihua; Lord, Graham M.; van der Harst, Pim; Lawlor, Debbie A.; Sehmi, Joban S.; Gale, Daniel P.; Wass, Mark N.; Ahmadi, Kourosh R.; Bakker, Stephan J. L.; Beckmann, Jacqui; Bilo, Henk J. G.; Bochud, Murielle; Brown, Morris J.; Caulfield, Mark J.; Connell, John M. C.; Cook, H. Terence; Cotlarciuc, Ioana; Smith, George Davey; de Silva, Ranil; Deng, Guohong; Devuyst, Olivier; Dikkeschei, Lambert D.; Dimkovic, Nada; Dockrell, Mark; Dominiczak, Anna; Ebrahim, Shah; Eggermann, Thomas; Farrall, Martin; Ferrucci, Luigi; Floege, Jurgen; Forouhi, Nita G.; Gansevoort, Ron T.; Han, Xijin; Hedblad, Bo; van der Heide, Jaap J. Homan; Hepkema, Bouke G.; Hernandez-Fuentes, Maria; Hypponen, Elina; Johnson, Toby; de Jong, Paul E.; Kleefstra, Nanne; Lagou, Vasiliki; Lapsley, Marta; Li, Yun; Loos, Ruth J. F.; Luan, Jian'an; Luttropp, Karin; Marechal, Celine; Melander, Olle; Munroe, Patricia B.; Nordfors, Louise; Parsa, Afshin; Peltonen, Leena; Penninx, Brenda W.; Perucha, Esperanza; Pouta, Anneli; Prokopenko, Inga; Roderick, Paul J.; Ruokonen, Aimo; Samani, Nilesh J.; Sanna, Serena; Schalling, Martin; Schlessinger, David; Schlieper, Georg; Seelen, Marc A. J.; Shuldiner, Alan R.; Sjoegren, Marketa; Smit, Johannes H.; Snieder, Harold; Soranzo, Nicole; Spector, Timothy D.; Stenvinkel, Peter; Sternberg, Michael J. E.; Swaminathan, Ramasamyiyer; Tanaka, Toshiko; Ubink-Veltmaat, Lielith J.; Uda, Manuela; Vollenweider, Peter; Wallace, Chris; Waterworth, Dawn; Zerres, Klaus; Waeber, Gerard; Wareham, Nicholas J.; Maxwell, Patrick H.; McCarthy, Mark I.; Jarvelin, Marjo-Riitta; Mooser, Vincent; Abecasis, Goncalo R.; Lightstone, Liz; Scott, James; Navis, Gerjan; Elliott, Paul; Kooner, Jaspal S.Nature Genetics (2010), 42 (5), 373-375CODEN: NGENEC; ISSN:1061-4036. (Nature Publishing Group)Using genome-wide assocn., we identify common variants at 2p12-p13, 6q26, 17q23 and 19q13 assocd. with serum creatinine, a marker of kidney function (P = 10-10 to 10-15). Of these, rs10206899 (near NAT8, 2p12-p13) and rs4805834 (near SLC7A9, 19q13) were also assocd. with chronic kidney disease (P = 5.0 × 10-5 and P = 3.6 × 10-4, resp.). Our findings provide insight into metabolic, solute and drug-transport pathways underlying susceptibility to chronic kidney disease.
- 46Simmons, M. L.; Frondoza, C. G.; Coyle, J. T. Immunocytochemical localization of N-acetyl-aspartate with monoclonal antibodies. Neuroscience 1991, 45, 37– 45, DOI: 10.1016/0306-4522(91)90101-SGoogle Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3MXmslKgtb8%253D&md5=361a6c94e99c27148b37e32321f739f3Immunocytochemical localization of N-acetyl-aspartate with monoclonal antibodiesSimmons, M. L.; Frondoza, C. G.; Coyle, J. T.Neuroscience (Oxford, United Kingdom) (1991), 45 (1), 37-45CODEN: NRSCDN; ISSN:0306-4522.N-Acetyl-aspartate is found in high concns. in all areas of the brain, but is undetectable in non-neuronal tissue. In order to characterize the cellular localization of N-acetyl-aspartate in brain, highly specific monoclonal antibodies against N-acetyl-aspartate were produced by fusing spleen lymphocytes obtained from mice immunized with N-acetyl-aspartate conjugated to thyroglobulin by carbodiimide with P3/x63-Ag8.653 mouse myeloma cells. Clones were selected which secrete IgG2a(k) antibodies highly specific for conjugated N-acetyl-aspartate. Only 3-6% cross-reactivity with conjugated N-acetyl-aspartate-glutamate was obsd. at high antibody concns., whereas no cross-reactivity (<1%) was obsd. with conjugated N-acetyl-glutamate or aspartate. Preincubation of the antibodies with 0.5 mg/mL conjugated N-acetyl-aspartate blocked immunoreactivity more than 90%, while preincubation with conjugated N-acetyl-aspartate-glutamate and free N-acetyl-aspartate had no effect. Immunocytochem. staining has shown that N-acetyl-aspartate-like immunoreactivity is localized in neurons, which are widely distributed throughout the brain. The immunoreactive neurons exhibited intense staining of the perikarya, proximal dendrites and axons. No consistent pattern of distribution of immunoreactivity was obsd. with regard to primary neurotransmitter characteristics of stained neurons although neurons with long projections or extensive arbors, such as pyramidal cells in cortex, locus ceruleus, motor neurons and Purkinje cells, stained much more intensively than local circuit neurons.
- 47Masaharu, M.; Hideo, M.; Mutsuhiko, M.; Yasuo, K. N-acetyl-l-aspartic acid, N-acetyl-α-l-aspartyl-l-glutamic acid and β-citryl-l-glutamic acid in human urine. Clin. Chim. Acta 1982, 120, 119– 126, DOI: 10.1016/0009-8981(82)90082-1Google ScholarThere is no corresponding record for this reference.
- 48Barker, P. B. N-acetyl aspartate─a neuronal marker?. Ann. Neurol. 2001, 49, 423– 424, DOI: 10.1002/ana.90Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD3M3hvVyltg%253D%253D&md5=b94bddfa307a732255479dd70f6d7ae4N-acetyl aspartate--a neuronal marker?Barker P BAnnals of neurology (2001), 49 (4), 423-4 ISSN:0364-5134.There is no expanded citation for this reference.
- 49Jung, R. E.; Brooks, W. M.; Yeo, R. A.; Chiulli, S. J.; Weers, D. C.; Sibbitt, W. L., Jr. Biochemical markers of intelligence: a proton MR spectroscopy study of normal human brain. Proc. R. Soc. London, Ser. B 1999, 266, 1375– 1379, DOI: 10.1098/rspb.1999.0790Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaK1MznsVeiug%253D%253D&md5=28668501ddca01938cdcecec067146adBiochemical markers of intelligence: a proton MR spectroscopy study of normal human brainJung R E; Brooks W M; Yeo R A; Chiulli S J; Weers D C; Sibbitt W L JrProceedings. Biological sciences / The Royal Society (1999), 266 (1426), 1375-9 ISSN:0962-8452.Proton magnetic resonance spectroscopy (1H-MRS) offers a unique non-invasive approach to measurement of N-acetylaspartate (NAA) and choline (Cho), putative markers of neuronal and glial integrity. Previous studies revealed that these neurochemicals predict cognitive impairment in diseased subjects, although little is known about their relationship to cognitive functioning in healthy people. We measured the concentrations of NAA and Cho in the left occipitoparietal white matter of 26 healthy adults and compared them with intellectual performance assessed by the Wechsler Adult Intelligence Scale-3. We found that NAA (b = 0.6, p < 0.01) and Cho (b = -0.42, p < 0.01) were independently associated with the Full-Scale Intelligence Quotient. Together, these metabolites accounted for a large proportion of the variance in intelligence (r2 = 0.45). Possible mechanisms underlying these correlations, such as mitochondrial function and myelin turnover, are discussed. 1H-MRS is a sensitive new tool to assess the neuronal underpinnings of cognitive function non-invasively.
- 50Patel, T.; Blyth, J. C.; Griffiths, G.; Kelly, D.; Talcott, J. B. Moderate relationships between NAA and cognitive ability in healthy adults: implications for cognitive spectroscopy. Front. Hum. Neurosci. 2014, 8, 39, DOI: 10.3389/fnhum.2014.00039Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhs1CrtL3O&md5=d73d396cdd39f3cfc6dda110ddd55e63Moderate relationships between NAA and cognitive ability in healthy adults: implications for cognitive spectroscopyPatel, Tulpesh; Blyth, Jacqueline C.; Griffiths, Gareth; Kelly, Deirdre; Talcott, Joel B.Frontiers in Human Neuroscience (2014), 8 (), 39/1-39/10, 10CODEN: FHNRAI; ISSN:1662-5161. (Frontiers Media S.A.)Background: Proton Magnetic Resonance Spectroscopy (1H-MRS)is a non-invasive imaging technique that enables quantification of neurochem. in vivo and thereby facilitates investigation of the biochem. underpinnings of human cognitive variability. Studies in the field of cognitive spectroscopy have commonly focused on relationships between measures of N-acetyl aspartate (NAA), a surrogate marker of neuronal health and function, and broad measures of cognitive performance, such as IQ. Methodol./Principal Findings: In this study, we used 1H-MRS to interrogate single-voxels in occipitoparietal and frontal cortex, in parallel with assessments of psychometric intelligence, in a sample of 40 healthy adult participants. We found correlations between NAA and IQ that were within the range reported in previous studies. However, the magnitude of these effects was significantly modulated by the stringency of data screening and the extent to which outlying values contributed to statistical analyses. Conclusions/Significance:1H-MRS offers a sensitive tool for assessing neurochem. non-invasively, yet the relationships between brain metabolites and broad aspects of human behavior such as IQ are subtle. We highlight the need to develop an increasingly rigorous anal. and interpretive framework for collecting and reporting data obtained from cognitive spectroscopy studies of this kind.
- 51Davies, G.; Lam, M.; Harris, S. E.; Trampush, J. W.; Luciano, M.; Hill, W. D. Study of 300,486 individuals identifies 148 independent genetic loci influencing general cognitive function. Nat. Commun. 2018, 9, 2098, DOI: 10.1038/s41467-018-04362-xGoogle Scholar52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1MbhsFCksg%253D%253D&md5=524b8e4875a84c1948df7aa439e49e4fStudy of 300,486 individuals identifies 148 independent genetic loci influencing general cognitive functionDavies Gail; Harris Sarah E; Luciano Michelle; Hill W David; Hagenaars Saskia P; Ritchie Stuart J; Marioni Riccardo E; Fawns-Ritchie Chloe; Liewald David C M; Okely Judith A; Corley Janie; Cox Simon R; Porteous David J; Starr John M; McIntosh Andrew M; Gale Catharine R; Deary Ian J; Lam Max; DeRosse Pamela; Harris Sarah E; Marioni Riccardo E; Porteous David J; Trampush Joey W; Trampush Joey W; Hagenaars Saskia P; Ahola-Olli Ari V; Rovio Suvi P; Raitakari Olli T; Ahola-Olli Ari V; Barnes Catriona L K; Campbell Harry; Joshi Peter K; Rudan Igor; Wilson James F; Bertram Lars; Liu Tian; Bis Joshua C; Psaty Bruce M; Burdick Katherine E; Roussos Panos; Burdick Katherine E; Burdick Katherine E; Christoforou Andrea; Djurovic Srdjan; Giddaluru Sudheer; Melle Ingrid; Le Hellard Stephanie; Steen Vidar M; Christoforou Andrea; Giddaluru Sudheer; Le Hellard Stephanie; Steen Vidar M; DeRosse Pamela; Yu Jin; Malhotra Anil; Lencz Todd; Djurovic Srdjan; Espeseth Thomas; Melle Ingrid; Andreassen Ole A; Sundet Kjetil; Espeseth Thomas; Sundet Kjetil; Reinvang Ivar; Giakoumaki Stella; Gustavson Daniel E; Dale Anders M; Kremen William S; Gustavson Daniel E; Kremen William S; Hayward Caroline; Vitart Veronique; Wilson James F; Hayward Caroline; Murray Alison D; Porteous David J; Hofer Edith; Petrovic Katja; Koini Marisa; Schmidt Reinhold; Hofer Edith; Ikram M Arfan; Terzikhan Natalie; van der Lee Sven J; Amin Najaf; Uitterlinden Andre; Wen K Hoyan; Adams Hieab H H; Ikram M Kamran; Van Duijn Cornelia M; Ikram M Arfan; Ikram M Arfan; Ikram M Kamran; Karlsson Robert; Karlsson Ida; Hagg Sara; Pedersen Nancy L; Knowles Emma; Glahn David C; Lahti Jari; Raikkonen Katri; Lahti Jari; Leber Markus; Ramirez Alfredo; Li Shuo; Yang Qiong; Mather Karen A; Thalamuthu Anbupalam; Assareh Amelia A; Brodaty Henry; Kochan Nicole A; Lee Teresa; Reppermund Simone; Sachdev Perminder S; Trollor Julian N; Morris Derek; Donohoe Gary; Oldmeadow Christopher; Palviainen Teemu; Loukola Anu; Kaprio Jaakko; Widen Elisabeth; Vuoksimaa Eero; Palotie Aarno; Payton Antony; Pazoki Raha; Dehghan Abbas; Evangelou Evangelos; Gao He; Tzoulaki Ioanna; Elliott Paul; Reynolds Chandra A; Sargurupremraj Muralidharan; Soumare Aicha; Tzourio Christophe; Debette Stephanie; Scholz Markus; Scholz Markus; Luck Tobias; Smith Jennifer A; Kardia Sharon L R; Zhao Wei; Smith Jennifer A; Ware Erin B; Faul Jessica D; Weir David R; Smith Albert V; Gudnason Vilmundur; Smith Albert V; Gudnason Vilmundur; Terzikhan Natalie; Trompet Stella; Noordam Raymond; Windham B Gwen; Mosley Thomas H Jr; Wright Margaret J; Hansell Narelle K; Wright Margaret J; Yang Jingyun; Bennett David A; Boyle Patricia A; Yang Jingyun; Bennett David A; Ames David; Ames David; Amouyel Philippe; Andreassen Ole A; Armstrong Nicola J; Attia John R; Holliday Elizabeth G; Scott Rodney J; Attix Deborah; Chiba-Falek Ornit; Attix Deborah; Avramopoulos Dimitrios; Avramopoulos Dimitrios; Arking Dan E; Bohmer Anne C; Ramirez Alfredo; Bohmer Anne C; Boyle Patricia A; Brodaty Henry; Cannon Tyrone D; Cirulli Elizabeth T; Congdon Eliza; Freimer Nelson A; London Edythe; Bilder Robert M; Conley Emily Drabant; Dale Anders M; Dale Anders M; Dale Anders M; Dehghan Abbas; Tzoulaki Ioanna; Elliott Paul; Dick Danielle; Dickinson Dwight; Eriksson Johan G; Evangelou Evangelos; Kaprio Jaakko; Eriksson Johan G; Eriksson Johan G; Eriksson Johan G; Ford Ian; Giegling Ina; Hartmann Annette M; Konte Bettina; Dan Rujescu; Gillespie Nathan A; Gordon Scott D; Martin Nicholas G; Montgomery Grant W; Gottesman Rebecca F; Gottesman Rebecca F; Griswold Michael E; Simino Jeannette; Harris Tamara B; Launer Lenore; Hatzimanolis Alex; Smyrnis Nikolaos; Stefanis Nikos C; Hatzimanolis Alex; Smyrnis Nikolaos; Stefanis Nikos C; Hatzimanolis Alex; Stefanis Nikos C; Heiss Gerardo; Kahonen Mika; Kahonen Mika; Kahonen Mika; Kleineidam Luca; Kleineidam Luca; Wagner Holger; Ramirez Alfredo; Kleineidam Luca; Wagner Michael; Kleineidam Luca; Wagner Michael; Knopman David S; Kochan Nicole A; Lee Teresa; Sachdev Perminder S; Kwok John B; Kwok John B; Lehtimaki Terho; Lyytikainen Leo-Pekka; Lehtimaki Terho; Lyytikainen Leo-Pekka; Li Shu-Chen; Liu Tian; Papenberg Goran; Lindenberger Ulman; Li Shu-Chen; Lill Christina M; Longstreth Will T Jr; Longstreth Will T Jr; Fitzpatrick Annette L; Lopez Oscar L; Luck Tobias; Riedel-Heller Steffi G; Lundervold Astri J; Lundervold Astri J; Lundquist Anders; Nyberg Lars; Lundquist Anders; Montgomery Grant W; Murray Alison D; Need Anna C; Nyberg Lars; Nyberg Lars; Ollier William; Papenberg Goran; Pattie Alison; Taylor Adele M; Polasek Ozren; Polasek Ozren; Poldrack Russell A; Psaty Bruce M; Psaty Bruce M; Reppermund Simone; Trollor Julian N; Rose Richard J; Rotter Jerome I; Taylor Kent D; Rotter Jerome I; Taylor Kent D; Roussos Panos; Roussos Panos; Saba Yasaman; Schmidt Helena; Sabb Fred W; Seshadri Sudha; Satizabal Claudia L; Seshadri Sudha; Satizabal Claudia L; Schmid Matthias; Weinhold Leonie; Scult Matthew A; Hariri Ahmad R; Slagboom P Eline; Stott David J; Straub Richard E; Weinberger Daniel R; Tzoulaki Ioanna; Tzourio Christophe; Uitterlinden Andre; Voineskos Aristotle N; Kaprio Jaakko; Vuoksimaa Eero; Adams Hieab H H; Bitsios Panos; Boerwinkle Eric; Bressler Jan; Boerwinkle Eric; Corvin Aiden; Gill Michael; De Jager Philip L; De Jager Philip L; Debette Stephanie; Fitzpatrick Annette L; Jukema J Wouter; Keller Matthew C; Palotie Aarno; Palotie Aarno; Pendleton Neil; Raitakari Olli T; Schofield Peter W; Schofield Peter R; Schofield Peter R; Starr John M; Turner Steven T; Villringer Arno; Villringer Arno; Malhotra Anil; Malhotra Anil; McIntosh Andrew M; Lencz Todd; Gale Catharine R; Seshadri SudhaNature communications (2018), 9 (1), 2098 ISSN:.General cognitive function is a prominent and relatively stable human trait that is associated with many important life outcomes. We combine cognitive and genetic data from the CHARGE and COGENT consortia, and UK Biobank (total N = 300,486; age 16-102) and find 148 genome-wide significant independent loci (P < 5 × 10(-8)) associated with general cognitive function. Within the novel genetic loci are variants associated with neurodegenerative and neurodevelopmental disorders, physical and psychiatric illnesses, and brain structure. Gene-based analyses find 709 genes associated with general cognitive function. Expression levels across the cortex are associated with general cognitive function. Using polygenic scores, up to 4.3% of variance in general cognitive function is predicted in independent samples. We detect significant genetic overlap between general cognitive function, reaction time, and many health variables including eyesight, hypertension, and longevity. In conclusion we identify novel genetic loci and pathways contributing to the heritability of general cognitive function.
- 52Lee, J. J.; Wedow, R.; Okbay, A.; Kong, E.; Maghzian, O.; Zacher, M. Gene discovery and polygenic prediction from a genome-wide association study of educational attainment in 1.1 million individuals. Nat. Genet. 2018, 50, 1112– 1121, DOI: 10.1038/s41588-018-0147-3Google Scholar53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtlOis7zI&md5=5259e727f3e6ec901d14a7f088dafedeGene discovery and polygenic prediction from a genome-wide association study of educational attainment in 1.1 million individualsLee, James J.; Wedow, Robbee; Okbay, Aysu; Kong, Edward; Maghzian, Omeed; Zacher, Meghan; Nguyen-Viet, Tuan Anh; Bowers, Peter; Sidorenko, Julia; Karlsson Linner, Richard; Fontana, Mark Alan; Kundu, Tushar; Lee, Chanwook; Li, Hui; Li, Ruoxi; Royer, Rebecca; Timshel, Pascal N.; Walters, Raymond K.; Willoughby, Emily A.; Yengo, Loic; 23andMe Research Team; Cogent; Social Science Genetic Association Consortium; Alver, Maris; Bao, Yanchun; Clark, David W.; Day, Felix R.; Furlotte, Nicholas A.; Joshi, Peter K.; Kemper, Kathryn E.; Kleinman, Aaron; Langenberg, Claudia; Magi, Reedik; Trampush, Joey W.; Verma, Shefali Setia; Wu, Yang; Lam, Max; Zhao, Jing Hua; Zheng, Zhili; Boardman, Jason D.; Campbell, Harry; Freese, Jeremy; Harris, Kathleen Mullan; Hayward, Caroline; Herd, Pamela; Kumari, Meena; Lencz, Todd; Luan, Jian'an; Malhotra, Anil K.; Metspalu, Andres; Milani, Lili; Ong, Ken K.; Perry, John R. B.; Porteous, David J.; Ritchie, Marylyn D.; Smart, Melissa C.; Smith, Blair H.; Tung, Joyce Y.; Wareham, Nicholas J.; Wilson, James F.; Beauchamp, Jonathan P.; Conley, Dalton C.; Esko, Tonu; Lehrer, Steven F.; Magnusson, Patrik K. E.; Oskarsson, Sven; Pers, Tune H.; Robinson, Matthew R.; Thom, Kevin; Watson, Chelsea; Chabris, Christopher F.; Meyer, Michelle N.; Laibson, David I.; Yang, Jian; Johannesson, Magnus; Koellinger, Philipp D.; Turley, Patrick; Visscher, Peter M.; Benjamin, Daniel J.; Cesarini, DavidNature Genetics (2018), 50 (8), 1112-1121CODEN: NGENEC; ISSN:1061-4036. (Nature Research)Here we conducted a large-scale genetic assocn. anal. of educational attainment in a sample of approx. 1.1 million individuals and identify 1,271 independent genome-wide-significant SNPs. For the SNPs taken together, we found evidence of heterogeneous effects across environments. The SNPs implicate genes involved in brain-development processes and neuron-to-neuron communication. In a sep. anal. of the X chromosome, we identify 10 independent genome-wide-significant SNPs and est. a SNP heritability of around 0.3% in both men and women, consistent with partial dosage compensation. A joint (multi-phenotype) anal. of educational attainment and three related cognitive phenotypes generates polygenic scores that explain 11-13% of the variance in educational attainment and 7-10% of the variance in cognitive performance. This prediction accuracy substantially increases the utility of polygenic scores as tools in research.
- 53Hearn, T. ALMS1 and Alström syndrome: a recessive form of metabolic, neurosensory and cardiac deficits. J. Mol Med. 2019, 97, 1– 7, DOI: 10.1007/s00109-018-1714-xGoogle Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitlSrtbzL&md5=ef0b17a82be038f1f685abc63d0a87eaALMS1 and Alstrom syndrome: a recessive form of metabolic, neurosensory and cardiac deficitsHearn, TomJournal of Molecular Medicine (Heidelberg, Germany) (2019), 97 (1), 1-17CODEN: JMLME8; ISSN:0946-2716. (Springer)A review. Alstrom syndrome (AS) is characterised by metabolic deficits, retinal dystrophy, sensorineural hearing loss, dilated cardiomyopathy and multi-organ fibrosis. Elucidating the function of the mutated gene, ALMS1, is crit. for the development of specific treatments and may uncover pathways relevant to a range of other disorders including common forms of obesity and type 2 diabetes. Interest in ALMS1 is heightened by the recent discovery of its involvement in neonatal cardiomyocyte cell cycle arrest, a process with potential relevance to regenerative medicine. ALMS1 encodes a ∼ 0.5 megadalton protein that localises to the base of centrioles. Some studies have suggested a role for this protein in maintaining centriole-nucleated sensory organelles termed primary cilia, and AS is now considered to belong to the growing class of human genetic disorders linked to ciliary dysfunction (ciliopathies). However, mechanistic details are lacking, and recent studies have implicated ALMS1 in several processes including endosomal trafficking, actin organization, maintenance of centrosome cohesion and transcription. In line with a more complex picture, multiple isoforms of the protein likely exist and non-centrosomal sites of localisation have been reported. This review outlines the evidence for both ciliary and extra-ciliary functions of ALMS1.
Cited By
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by ACS Publications if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
This article is cited by 7 publications.
- Zhipeng Li, Li You, Xiping Du, Haoyi Yang, Liang Yang, Yanbing Zhu, Lijun Li, Zedong Jiang, Qingbiao Li, Ning He, Rui Lin, Zhen Chen, Hui Ni. New strategies to study in depth the metabolic mechanism of astaxanthin biosynthesis in
Phaffia rhodozyma. Critical Reviews in Biotechnology 2025, 45
(2)
, 454-472. https://doi.org/10.1080/07388551.2024.2344578
- Anneke Brümmer, Sven Bergmann. Disentangling genetic effects on transcriptional and post-transcriptional gene regulation through integrating exon and intron expression QTLs. Nature Communications 2024, 15
(1)
https://doi.org/10.1038/s41467-024-48244-x
- Krista Freimann, Anneke Brümmer, Robert Warmerdam, Tarran S. Rupall, Ana Laura Hernández-Ledesma, Joshua Chiou, Emily R. Holzinger, Joseph C. Maranville, Nikolina Nakic, Halit Ongen, Luca Stefanucci, Michael C. Turchin, , Lude Franke, Urmo Võsa, Carla P. Jones, Alejandra Medina-Rivera, Gosia Trynka, Kai Kisand, Sven Bergmann, Kaur Alasoo. Trans
-eQTL mapping prioritises
USP18
as a negative regulator of interferon response at a lupus risk locus. 2024https://doi.org/10.1101/2024.07.15.24310442
- Kaylia M. Reynolds, Andrea R. V. R. Horimoto, Bridget M. Lin, Ying Zhang, Nuzulul Kurniansyah, Bing Yu, Eric Boerwinkle, Qibin Qi, Robert Kaplan, Martha Daviglus, Lifang Hou, Laura Y. Zhou, Jianwen Cai, Saame Raza Shaikh, Tamar Sofer, Sharon R. Browning, Nora Franceschini. Ancestry-driven metabolite variation provides insights into disease states in admixed populations. Genome Medicine 2023, 15
(1)
https://doi.org/10.1186/s13073-023-01209-z
- Anneke Brümmer, Sven Bergmann. Disentangling genetic effects on transcriptional and post-transcriptional gene regulation through integrating exon and intron expression QTLs. 2023https://doi.org/10.1101/2023.04.27.538308
- Keenan S. Fine, John T. Wilkins, Konrad Teodor Sawicki. Clinical Metabolomic Landscape of Cardiovascular Physiology and Disease. Journal of the American Heart Association 2023, 12
(6)
https://doi.org/10.1161/JAHA.122.027725
- E. Molinero, R.N. Pena, J. Estany, R. Ros-Freixedes. 260. Prioritisation of variants from whole-genome sequence data for validation of their association with traits of interest. 2022, 1098-1101. https://doi.org/10.3920/978-90-8686-940-4_260
- Marie C. Sadler, Chiara Auwerx, Kaido Lepik, Eleonora Porcu, Zoltán Kutalik. Quantifying the role of transcript levels in mediating DNA methylation effects on complex traits and diseases. Nature Communications 2022, 13
(1)
https://doi.org/10.1038/s41467-022-35196-3
- Xianyong Yin, Debraj Bose, Annie Kwon, Sarah C. Hanks, Anne U. Jackson, Heather M. Stringham, Ryan Welch, Anniina Oravilahti, Lilian Fernandes Silva, Adam E. Locke, Christian Fuchsberger, Susan K. Service, Michael R. Erdos, Lori L. Bonnycastle, Johanna Kuusisto, Nathan O. Stitziel, Ira M. Hall, Jean Morrison, Samuli Ripatti, Aarno Palotie, Nelson B. Freimer, Francis S. Collins, Karen L. Mohlke, Laura J. Scott, Eric B. Fauman, Charles Burant, Michael Boehnke, Markku Laakso, Xiaoquan Wen. Integrating transcriptomics, metabolomics, and GWAS helps reveal molecular mechanisms for metabolite levels and disease risk. The American Journal of Human Genetics 2022, 109
(10)
, 1727-1741. https://doi.org/10.1016/j.ajhg.2022.08.007
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
Abstract
Figure 1
Figure 1. QQ-plot of −log10 (p values) of metabolome- and transcriptome-wide association analysis. The highly significant associations (FDR < 0.05) with ALMS1 expression are ranked 1st and 2nd and with HPS1 expression 3rd and 4th.
Figure 2
Figure 2. Metabomatching (22) results for pseudospectra derived from gene expression - metabolome feature associations for ALMS1 (A) and HPS1 (B). Upper panels show the features in each pseudospectrum, color-coded according to the direction of the effect (positive in blue and negative in orange). Lower panels show the highest ranking candidate metabolites with their reference NMR spectra (color coded to indicate their relative peak intensities). Leading features allowing metabolite identification are in (A) at 2.04 ppm, which matches well with the highest intensity peak of the NAA spectrum and in (B) at 2.87 ppm, which matches well with the TMA singlet.
Figure 3
Figure 3. SNP - metabolome feature and SNP - gene expression associations in ALMS1/NAT8 locus. (A) LocusZoom plot for ALMS1/NAT8 locus, where the SNPs are associated with metabolome feature at 2.0375 ppm, LD colored with respect to lead mQTL. (B) Bar plot shows −log10 transformed p values from associating expression values of nine genes in the locus with the five NAA features.
Figure 4
Figure 4. SNP - metabolome feature and SNP - gene expression associations in HPS1/PYROXD2 locus. (A) LocusZoom plot for HPS1/PYROXD2 locus, showing the association significance of SNP with the metabolome feature at 2.8725 ppm. Colors indicate the correlation (LD) to the lead QTL. (B) Bar plot shows −log10 transformed p values from associating expression values of seven genes in the locus with the same feature.
Figure 5
Figure 5. Scatter plot of the mQTL effect of SNP (rs7566315) on NAC and its eQTL effect on ALMS1 gene expression. Each point represents a study sample. NAC concentration is approximated by the feature at 2.0375 ppm that is log10 transformed after feature- and sample-wise z-scoring (y axis). ALMS1 expression is z-scored after log2 transforming RPKM+1 values (x axis). Color code represents the genotype of rs7566315 (legend) that is an eQTL of ALMS1 and mQTL of NAA.
References
This article references 53 other publications.
- 1Buniello, A.; MacArthur, J. A. L.; Cerezo, M.; Harris, L. W.; Hayhurst, J.; Malangone, C. The NHGRI-EBI GWAS Catalog of published genome-wide association studies, targeted arrays and summary statistics 2019. Nucleic Acids Res. 2019, 47, D1005– D1012, DOI: 10.1093/nar/gky11201https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhs1GqtrvO&md5=08a31cc6157f3ffe9be66587fdeed121The NHGRI-EBI GWAS catalog of published genome-wide association studies, targeted arrays and summary statistics 2019Buniello, Annalisa; MacArthu, Jacqueline A. L.; Cerez, Maria; Harris, Laura W.; Hayhurst, James; Malangone, Cinzia; McMahon, Aoife; Morales, Joannella; Mountjoy, Edward; Sollis, Elliot; Suveges, Daniel; Vrousgou, Olga; Whetzel, Patricia L.; Amode, Ridwan; Guillen, Jose A.; Riat, Harpreet S.; Trevanion, Stephen J.; Hall, Peggy; Junkins, Heather; Flicek, Paul; Burdett, Tony; Hindorff, Lucia A.; Cunningham, Fiona; Parkinson, HelenNucleic Acids Research (2019), 47 (D1), D1005-D1012CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)The GWAS Catalog delivers a high-quality curated collection of all published genome-wide assocn. studies enabling investigations to identify causal variants, understand disease mechanisms, and establish targets for novel therapies. The scope of the Catalog has also expanded to targeted and exome arrays with 1000 new assocns. added for these technologies. As of Sept. 2018, the Catalog contains 5687 GWAS comprising 71673 variant-trait assocns. from 3567 publications. New content includes 284 full P-value summary statistics datasets for genome-wide and new targeted array studies, representing 6 × 109 individual variant-trait statistics. In the last 12 mo, the Catalog's user interface was accessed by ∼90000 unique users who viewed >1 million pages. We have improved data access with the release of a new RESTful API to support high-throughput programmatic access, an improved web interface and a new summary statistics database. Summary statistics provision is supported by a new format proposed as a community std. for summary statistics data representation. This format was derived from our experience in standardizing heterogeneous submissions, mapping formats and in harmonizing content.
- 2GTEx Consortium The GTEx Consortium atlas of genetic regulatory effects across human tissues. Science 2020, 369, 1318– 1330, DOI: 10.1126/science.aaz1776There is no corresponding record for this reference.
- 3Maurano, M. T.; Humbert, R.; Rynes, E.; Thurman, R. E.; Haugen, E.; Wang, H. Systematic localization of common disease-associated variation in regulatory DNA. Science 2012, 337, 1190– 1195, DOI: 10.1126/science.12227943https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xht1ylsLfL&md5=6b4de239d584cc09c6e513580e6caa3cSystematic localization of common disease-associated variation in regulatory DNAMaurano, Matthew T.; Humbert, Richard; Rynes, Eric; Thurman, Robert E.; Haugen, Eric; Wang, Hao; Reynolds, Alex P.; Sandstrom, Richard; Qu, Hongzhu; Brody, Jennifer; Shafer, Anthony; Neri, Fidencio; Lee, Kristen; Kutyavin, Tanya; Stehling-Sun, Sandra; Johnson, Audra K.; Canfield, Theresa K.; Giste, Erika; Diegel, Morgan; Bates, Daniel; Hansen, R. Scott; Neph, Shane; Sabo, Peter J.; Heimfeld, Shelly; Raubitschek, Antony; Ziegler, Steven; Cotsapas, Chris; Sotoodehnia, Nona; Glass, Ian; Sunyaev, Shamil R.; Kaul, Rajinder; Stamatoyannopoulos, John A.Science (Washington, DC, United States) (2012), 337 (6099), 1190-1195CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Genome-wide assocn. studies have identified many noncoding variants assocd. with common diseases and traits. We show that these variants are concd. in regulatory DNA marked by DNase I hypersensitive sites (DHSs). Eighty-eight percent of such DHSs are active during fetal development and are enriched in variants assocd. with gestational exposure-related phenotypes. We identified distant gene targets for hundreds of variant-contg. DHSs that may explain phenotype assocns. Disease-assocd. variants systematically perturb transcription factor recognition sequences, frequently alter allelic chromatin states, and form regulatory networks. We also demonstrated tissue-selective enrichment of more weakly disease-assocd. variants within DHSs and the de novo identification of pathogenic cell types for Crohn's disease, multiple sclerosis, and an ECG trait, without prior knowledge of physiol. mechanisms. Our results suggest pervasive involvement of regulatory DNA variation in common human disease and provide pathogenic insights into diverse disorders.
- 4Nicolae, D. L.; Gamazon, E.; Zhang, W.; Duan, S.; Dolan, M. E.; Cox, N. J. Trait-associated SNPs are more likely to be eQTLs: annotation to enhance discovery from GWAS. PLoS Genet. 2010, 6, e1000888, DOI: 10.1371/journal.pgen.1000888There is no corresponding record for this reference.
- 5Ward, L. D.; Kellis, M. Interpreting non-coding variation in complex disease genetics. Nat. Biotechnol. 2012, 30, 1095, DOI: 10.1038/nbt.24225https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xhs1elsr%252FL&md5=bc1dd6f524db613547c87ecac384675cInterpreting noncoding genetic variation in complex traits and human diseaseWard, Lucas D.; Kellis, ManolisNature Biotechnology (2012), 30 (11), 1095-1106CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)A review. Assocn. studies provide genome-wide information about the genetic basis of complex disease, but medical research has focused primarily on protein-coding variants, owing to the difficulty of interpreting noncoding mutations. This picture has changed with advances in the systematic annotation of functional noncoding elements. Evolutionary conservation, functional genomics, chromatin state, sequence motifs and mol. quant. trait loci all provide complementary information about the function of noncoding sequences. These functional maps can help with prioritizing variants on risk haplotypes, filtering mutations encountered in the clinic and performing systems-level analyses to reveal processes underlying disease assocns. Advances in predictive modeling can enable data-set integration to reveal pathways shared across loci and alleles, and richer regulatory models can guide the search for epistatic interactions. Lastly, new massively parallel reporter expts. can systematically validate regulatory predictions. Ultimately, advances in regulatory and systems genomics can help unleash the value of whole-genome sequencing for personalized genomic risk assessment, diagnosis and treatment.
- 6Lloyd-Jones, L. R.; Holloway, A.; McRae, A.; Yang, J.; Small, K.; Zhao, J. The genetic architecture of gene expression in peripheral blood. Am. J. Hum. Genet. 2017, 100, 228– 237, DOI: 10.1016/j.ajhg.2016.12.0086https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXlt12qsQ%253D%253D&md5=667b0bb41842675e780eaee9cb6879e2The Genetic Architecture of Gene Expression in Peripheral BloodLloyd-Jones, Luke R.; Holloway, Alexander; McRae, Allan; Yang, Jian; Small, Kerrin; Zeng, Biao; Bakshi, Andrew; Metspalu, Andres; Dermitzakis, Manolis; Gibson, Greg; Spector, Tim; Montgomery, Grant; Esko, Tonu; Visscher, Peter M.; Powell, Joseph E.American Journal of Human Genetics (2017), 100 (2), 228-237CODEN: AJHGAG; ISSN:0002-9297. (Cell Press)We analyzed the mRNA levels for 36,778 transcript expression traits (probes) from 2,765 individuals to comprehensively investigate the genetic architecture and degree of missing heritability for gene expression in peripheral blood. We identified 11,204 cis and 3,791 trans independent expression quant. trait loci (eQTL) by using linear mixed models to perform genome-wide assocn. analyses. Furthermore, using information on both closely and distantly related individuals, heritability was estd. for all expression traits. Of the set of expressed probes (15,966), 10,580 (66%) had an estd. narrow-sense heritability (h2) greater than zero with a mean (median) value of 0.192 (0.142). Across these probes, on av. the proportion of genetic variance explained by all eQTL (h2COJO) was 31% (0.060/0.192), meaning that 69% is missing, with the sentinel SNP of the largest eQTL explaining 87% (0.052/0.060) of the variance attributed to all identified cis- and trans-eQTL. For the same set of probes, the genetic variance attributed to genome-wide common (MAF > 0.01) HapMap 3 SNPs (h2g) accounted for on av. 48% (0.093/0.192) of h2. Taken together, the evidence suggests that approx. half the genetic variance for gene expression is not tagged by common SNPs, and of the variance that is tagged by common SNPs, a large proportion can be attributed to identifiable eQTL of large effect, typically in cis. Finally, we present evidence that, compared with a meta-anal., using individual-level data results in an increase of approx. 50% in power to detect eQTL.
- 7Montgomery, S. B.; Sammeth, M.; Gutierrez-Arcelus, M.; Lach, R. P.; Ingle, C.; Nisbett, J. Transcriptome genetics using second generation sequencing in a Caucasian population. Nature 2010, 464, 773, DOI: 10.1038/nature089037https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXivFKnsbc%253D&md5=5bb32c958242d7bbd5c43c4aeaa3fc53Transcriptome genetics using second generation sequencing in a Caucasian populationMontgomery, Stephen B.; Sammeth, Micha; Gutierrez-Arcelus, Maria; Lach, Radoslaw P.; Ingle, Catherine; Nisbett, James; Guigo, Roderic; Dermitzakis, Emmanouil T.Nature (London, United Kingdom) (2010), 464 (7289), 773-777CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Gene expression is an important phenotype that informs about genetic and environmental effects on cellular state. Many studies have previously identified genetic variants for gene expression phenotypes using custom and com. available microarrays. Second generation sequencing technologies are now providing unprecedented access to the fine structure of the transcriptome. We have sequenced the mRNA fraction of the transcriptome in 60 extended HapMap individuals of European descent and have combined these data with genetic variants from the HapMap3 project. We have quantified exon abundance based on read depth and have also developed methods to quantify whole transcript abundance. We have found that approx. 10 million reads of sequencing can provide access to the same dynamic range as arrays with better quantification of alternative and highly abundant transcripts. Correlation with SNPs (small nucleotide polymorphisms) leads to a larger discovery of eQTLs (expression quant. trait loci) than with arrays. We also detect a substantial no. of variants that influence the structure of mature transcripts indicating variants responsible for alternative splicing. Finally, measures of allele-specific expression allowed the identification of rare eQTLs and allelic differences in transcript structure. This anal. shows that high throughput sequencing technologies reveal new properties of genetic effects on the transcriptome and allow the exploration of genetic effects in cellular processes.
- 8Wright, F. A.; Sullivan, P. F.; Brooks, A. I.; Zou, F.; Sun, W.; Xia, K. Heritability and genomics of gene expression in peripheral blood. Nat. Genet. 2014, 46, 430, DOI: 10.1038/ng.29518https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXmtlWrtb8%253D&md5=e687d946efdf04987a94b474ba930e27Heritability and genomics of gene expression in peripheral bloodWright, Fred A.; Sullivan, Patrick F.; Brooks, Andrew I.; Zou, Fei; Sun, Wei; Xia, Kai; Madar, Vered; Jansen, Rick; Chung, Wonil; Zhou, Yi-Hui; Abdellaoui, Abdel; Batista, Sandra; Butler, Casey; Chen, Guanhua; Chen, Ting-Huei; D'Ambrosio, David; Gallins, Paul; Ha, Min Jin; Hottenga, Jouke Jan; Huang, Shunping; Kattenberg, Mathijs; Kochar, Jaspreet; Middeldorp, Christel M.; Qu, Ani; Shabalin, Andrey; Tischfield, Jay; Todd, Laura; Tzeng, Jung-Ying; van Grootheest, Gerard; Vink, Jacqueline M.; Wang, Qi; Wang, Wei; Wang, Weibo; Willemsen, Gonneke; Smit, Johannes H.; de Geus, Eco J.; Yin, Zhaoyu; Penninx, Brenda W. J. H.; Boomsma, Dorret I.Nature Genetics (2014), 46 (5), 430-437CODEN: NGENEC; ISSN:1061-4036. (Nature Publishing Group)We assessed gene expression profiles in 2752 twins, using a classic twin design to quantify expression heritability and quant. trait loci (eQTLs) in peripheral blood. The most highly heritable genes (∼777) were grouped into distinct expression clusters, enriched in gene-poor regions, assocd. with specific gene function or ontol. classes, and strongly assocd. with disease designation. The design enabled a comparison of twin-based heritability to ests. based on dizygotic identity-by-descent sharing and distant genetic relatedness. Consideration of sampling variation suggests that previous heritability ests. have been upwardly biased. Genotyping of 2494 twins enabled powerful identification of eQTLs, which we further examd. in a replication set of 1895 unrelated subjects. A large no. of non-redundant local eQTLs (6756) met replication criteria, whereas a relatively small no. of distant eQTLs (165) met quality control and replication stds. Our results provide a new resource toward understanding the genetic control of transcription.
- 9Suhre, K.; Wallaschofski, H.; Raffler, J.; Friedrich, N.; Haring, R.; Michael, K. A genome-wide association study of metabolic traits in human urine. Nat. Genet. 2011, 43, 565– 569, DOI: 10.1038/ng.8379https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXmtV2hs7g%253D&md5=6a695aed032888c41f97b16552775b2aA genome-wide association study of metabolic traits in human urineSuhre, Karsten; Wallaschofski, Henri; Raffler, Johannes; Friedrich, Nele; Haring, Robin; Michael, Kathrin; Wasner, Christina; Krebs, Alexander; Kronenberg, Florian; Chang, David; Meisinger, Christa; Wichmann, H.-Erich; Hoffmann, Wolfgang; Voelzke, Henry; Voelker, Uwe; Teumer, Alexander; Biffar, Reiner; Kocher, Thomas; Felix, Stephan B.; Illig, Thomas; Kroemer, Heyo K.; Gieger, Christian; Roemisch-Margl, Werner; Nauck, MatthiasNature Genetics (2011), 43 (6), 565-569CODEN: NGENEC; ISSN:1061-4036. (Nature Publishing Group)We present a genome-wide assocn. study of metabolic traits in human urine, designed to investigate the detoxification capacity of the human body. Using NMR spectroscopy, we tested for assocns. between 59 metabolites in urine from 862 male participants in the population-based SHIP study. We replicated the results using 1,039 addnl. samples of the same study, including a 5-yr follow-up, and 992 samples from the independent KORA study. We report five loci with joint P values of assocn. from 3.2 × 10-19 to 2.1 × 10-182. Variants at three of these loci have previously been linked with important clin. outcomes: SLC7A9 is a risk locus for chronic kidney disease, NAT2 for coronary artery disease and genotype-dependent response to drug toxicity, and SLC6A20 for iminoglycinuria. Moreover, we identify rs37369 in AGXT2 as the genetic basis of hyper-β-aminoisobutyric aciduria.
- 10Bartel, J.; Krumsiek, J.; Schramm, K.; Adamski, J.; Gieger, C.; Herder, C. The Human Blood Metabolome-Transcriptome Interface. PLoS Genet. 2015, 11, e1005274, DOI: 10.1371/journal.pgen.100527410https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXjvVKisLs%253D&md5=b9e5564d646b90c0e91499383bbbd54bThe human blood metabolome-transcriptome interfaceBartel, Joerg; Krumsiek, Jan; Schramm, Katharina; Adamski, Jerzy; Gieger, Christian; Herder, Christian; Carstensen, Maren; Peters, Annette; Rathmann, Wolfgang; Roden, Michael; Strauch, Konstantin; Suhre, Karsten; Kastenmueller, Gabi; Prokisch, Holger; Theis, Fabian J.PLoS Genetics (2015), 11 (6), e1005274/1-e1005274/32CODEN: PGLEB5; ISSN:1553-7404. (Public Library of Science)Biol. systems consist of multiple organizational levels all densely interacting with each other to ensure function and flexibility of the system. Simultaneous anal. of cross-sectional multi-omics data from large population studies is a powerful tool to comprehensively characterize the underlying mol. mechanisms on a physiol. scale. In this study, we systematically analyzed the relationship between fasting serum metabolomics and whole blood transcriptomics data from 712 individuals of the German KORA F4 cohort. Correlation-based anal. identified 1,109 significant assocns. between 522 transcripts and 114 metabolites summarized in an integrated network, the 'human blood metabolometranscriptome interface' (BMTI). Bidirectional causality anal. using Mendelian randomization did not yield any statistically significant causal assocns. between transcripts and metabolites. A knowledge-based interpretation and integration with a genome-scale human metabolic reconstruction revealed systematic signatures of signaling, transport and metabolic processes, i.e. metabolic reactions mainly belonging to lipid, energy and amino acid metab. Moreover, the construction of a network based on functional categories illustrated the cross-talk between the biol. layers at a pathway level. Using a transcription factor binding site enrichment anal., this pathway cross-talk was further confirmed at a regulatory level. Finally, we demonstrated how the constructed networks can be used to gain novel insights into mol. mechanisms assocd. to intermediate clin. traits. Overall, our results demonstrate the utility of a multi-omics integrative approach to understand the mol. mechanisms underlying both normal physiol. and disease.
- 11Burkhardt, R.; Kirsten, H.; Beutner, F.; Holdt, L. M.; Gross, A.; Teren, A. Integration of Genome-Wide SNP Data and Gene-Expression Profiles Reveals Six Novel Loci and Regulatory Mechanisms for Amino Acids and Acylcarnitines in Whole Blood. PLoS Genet. 2015, 11, e1005510, DOI: 10.1371/journal.pgen.100551011https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XmtFGltrg%253D&md5=9f36470c050b23fa8b6425d998e68d91Integration of genome-wide SNP data and gene-expression profiles reveals six novel loci and regulatory mechanisms for amino acids and acylcarnitines in whole bloodBurkhardt, Ralph; Kirsten, Holger; Beutner, Frank; Holdt, Lesca M.; Gross, Arnd; Teren, Andrej; Toenjes, Anke; Becker, Susen; Krohn, Knut; Kovacs, Peter; Stumvoll, Michael; Teupser, Daniel; Thiery, Joachim; Ceglarek, Uta; Scholz, MarkusPLoS Genetics (2015), 11 (9), e1005510/1-e1005510/25CODEN: PGLEB5; ISSN:1553-7404. (Public Library of Science)Profiling amino acids and acylcarnitines in whole blood spots is a powerful tool in the lab. diagnosis of several inborn errors of metab. Emerging data suggests that altered blood levels of amino acids and acylcarnitines are also assocd. with common metabolic diseases in adults. Thus, the identification of common genetic determinants for blood metabolites might shed light on pathways contributing to human physiol. and common diseases. We applied a targeted mass-spectrometry-based method to analyze whole blood concns. of 96 amino acids, acylcarnitines and pathway assocd. metabolite ratios in a Central European cohort of 2,107 adults and performed genome-wide assocn. (GWA) to identify genetic modifiers of metabolite concns. We discovered and replicated six novel loci assocd. with blood levels of total acylcarnitine, arginine (both on chromosome 6; rs12210538, rs17657775), propionylcarnitine (chromosome 10; rs12779637), 2-hydroxyisovalerylcarnitine (chromosome 21; rs1571700), stearoylcarnitine (chromosome 1; rs3811444), and aspartic acid traits (chromosome 8; rs750472). Based on an integrative anal. of expression quant. trait loci in blood mononuclear cells and correlations between gene expressions and metabolite levels, we provide evidence for putative causative genes: SLC22A16 for total acylcarnitines, ARG1 for arginine, HLCS for 2-hydroxyisovalerylcarnitine, JAM3 for stearoylcarnitine via a trans-effect at chromosome 1, and PPP1R16A for aspartic acid traits. Further, we report replication and provide addnl. functional evidence for ten loci that have previously been published for metabolites measured in plasma, serum or urine. In conclusion, our integrative anal. of SNP, gene-expression and metabolite data points to novel genetic factors that may be involved in the regulation of human metab. At several loci, we provide evidence for metabolite regulation via gene-expression and obsd. overlaps with GWAS loci for common diseases. These results form a strong rationale for subsequent functional and disease-related studies.
- 12Inouye, M.; Kettunen, J.; Soininen, P.; Silander, K.; Ripatti, S.; Kumpula, L. S.; Hämäläinen, E.; Jousilahti, P.; Kangas, A. J.; Männistö, S.; Savolainen, M. J.; Jula, A.; Leiviskä, J.; Palotie, A.; Salomaa, V.; Perola, M.; Ala-Korpela, M.; Peltonen, L. Metabonomic, transcriptomic, and genomic variation of a population cohort. Mol. Syst. Biol. 2010, 6, 441, DOI: 10.1038/msb.2010.9312https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3M%252FmtFygug%253D%253D&md5=b4f99d89873a563ebf32c668fd9c9066Metabonomic, transcriptomic, and genomic variation of a population cohortInouye Michael; Kettunen Johannes; Soininen Pasi; Silander Kaisa; Ripatti Samuli; Kumpula Linda S; Hamalainen Eija; Jousilahti Pekka; Kangas Antti J; Mannisto Satu; Savolainen Markku J; Jula Antti; Leiviska Jaana; Palotie Aarno; Salomaa Veikko; Perola Markus; Ala-Korpela Mika; Peltonen LeenaMolecular systems biology (2010), 6 (), 441 ISSN:.Comprehensive characterization of human tissues promises novel insights into the biological architecture of human diseases and traits. We assessed metabonomic, transcriptomic, and genomic variation for a large population-based cohort from the capital region of Finland. Network analyses identified a set of highly correlated genes, the lipid-leukocyte (LL) module, as having a prominent role in over 80 serum metabolites (of 134 measures quantified), including lipoprotein subclasses, lipids, and amino acids. Concurrent association with immune response markers suggested the LL module as a possible link between inflammation, metabolism, and adiposity. Further, genomic variation was used to generate a directed network and infer LL module's largely reactive nature to metabolites. Finally, gene co-expression in circulating leukocytes was shown to be dependent on serum metabolite concentrations, providing evidence for the hypothesis that the coherence of molecular networks themselves is conditional on environmental factors. These findings show the importance and opportunity of systematic molecular investigation of human population samples. To facilitate and encourage this investigation, the metabonomic, transcriptomic, and genomic data used in this study have been made available as a resource for the research community.
- 13Bullaughey, K.; Chavarria, C. I.; Coop, G.; Gilad, Y. Expression quantitative trait loci detected in cell lines are often present in primary tissues. Hum. Mol. Genet. 2009, 18, 4296– 4303, DOI: 10.1093/hmg/ddp38213https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtlWhu7fN&md5=00faca2e530fbbbd42b01e61c4408802Expression quantitative trait loci detected in cell lines are often present in primary tissuesBullaughey, Kevin; Chavarria, Claudia I.; Coop, Graham; Gilad, YoavHuman Molecular Genetics (2009), 18 (22), 4296-4303CODEN: HMGEE5; ISSN:0964-6906. (Oxford University Press)Expression quant. trait loci (eQTL) mapping is a powerful tool for identifying genetic regulatory variation. However, at present, most eQTLs in humans were identified using gene expression data from cell lines, and it remains unknown whether these eQTLs also have a regulatory function in other expression contexts, such as human primary tissues. Here we investigate this question using a targeted strategy. Specifically, we selected a subset of large-effect eQTLs identified in the HapMap lymphoblastoid cell lines, and examd. the assocn. of these eQTLs with gene expression levels across individuals in five human primary tissues (heart, kidney, liver, lung and testes). We show that genotypes at the eQTLs we selected are often predictive of variation in gene expression levels in one or more of the five primary tissues. The genotype effects in the primary tissues are consistently in the same direction as the effects inferred in the cell lines. Addnl., a no. of the eQTLs we tested are found in more than one of the tissues. Our results indicate that functional studies in cell lines may uncover a substantial amt. of genetic variation that affects gene expression levels in human primary tissues.
- 14Çalışkan, M.; Cusanovich, D. A.; Ober, C.; Gilad, Y. The effects of EBV transformation on gene expression levels and methylation profiles. Hum. Mol. Genet. 2011, 20, 1643– 1652, DOI: 10.1093/hmg/ddr04114https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXjvVKrur4%253D&md5=b0ef2fc5995f0b08e47c759ef1725bc2The effects of EBV transformation on gene expression levels and methylation profilesCaliskan, Minal; Cusanovich, Darren A.; Ober, Carole; Gilad, YoavHuman Molecular Genetics (2011), 20 (8), 1643-1652CODEN: HMGEE5; ISSN:0964-6906. (Oxford University Press)Epstein-Barr virus (EBV) transformed lymphoblastoid cell lines (LCLs) provide a conveniently accessible and renewable resource for functional genomic studies in humans. The ability to accumulate multidimensional data pertaining to the same individual cell lines, from complete genomic sequences to detailed gene regulatory profiles, further enhances the utility of LCLs as a model system. A lingering concern, however, is that the changes assocd. with EBV transformation of B cells reduce the usefulness of LCLs as a surrogate model for primary tissues. To evaluate the validity of this concern, the authors compared global gene expression and methylation profiles between CD20+ primary B cells sampled from six individuals and six independent replicates of transformed LCLs derived from each sample. These data allowed the authors to obtain a detailed catalog of the genes and pathways whose regulation is affected by EBV transformation. The expression levels and promoter methylation profiles of more than half of the studied genes were affected by the EBV transformation, including enrichments of genes involved in transcription regulation, cell cycle and immune response. However, most of the differences in gene expression levels between LCLs and B cells are of small magnitude, and that LCLs can often recapitulate the naturally occurring gene expression variation in primary B cells. Thus, these observations suggest that inference of the genetic architecture that underlies regulatory variation in LCLs can typically be generalized to primary B cells. In contrast, inference based on functional studies in LCLs may be more limited to the cell lines.
- 15Dimas, A. S.; Deutsch, S.; Stranger, B. E.; Montgomery, S. B.; Borel, C.; Attar-Cohen, H. Common regulatory variation impacts gene expression in a cell type–dependent manner. Science 2009, 325, 1246– 1250, DOI: 10.1126/science.117414815https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtVOmsrjM&md5=c1c8c0127f5791bbf889eb29caaf5a82Common Regulatory Variation Impacts Gene Expression in a Cell Type-Dependent MannerDimas, Antigone S.; Deutsch, Samuel; Stranger, Barbara E.; Montgomery, Stephen B.; Borel, Christelle; Attar-Cohen, Homa; Ingle, Catherine; Beazley, Claude; Gutierrez Arcelus, Maria; Sekowska, Magdalena; Gagnebin, Marilyne; Nisbett, James; Deloukas, Panos; Dermitzakis, Emmanouil T.; Antonarakis, Stylianos E.Science (Washington, DC, United States) (2009), 325 (5945), 1246-1250CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Studies correlating genetic variation to gene expression facilitate the interpretation of common human phenotypes and disease. As functional variants may be operating in a tissue-dependent manner, we performed gene expression profiling and assocn. with genetic variants (single-nucleotide polymorphisms) on three cell types of 75 individuals. We detected cell type-specific genetic effects, with 69 to 80% of regulatory variants operating in a cell type-specific manner, and identified multiple expressive quant. trait loci (eQTLs) per gene, unique or shared among cell types and pos. correlated with the no. of transcripts per gene. Cell type-specific eQTLs were found at larger distances from genes and at lower effect size, similar to known enhancers. These data suggest that the complete regulatory variant repertoire can only be uncovered in the context of cell-type specificity.
- 16Ding, J.; Gudjonsson, J. E.; Liang, L.; Stuart, P. E.; Li, Y.; Chen, W. Gene expression in skin and lymphoblastoid cells: Refined statistical method reveals extensive overlap in cis-eQTL signals. Am. J. Hum. Genet. 2010, 87, 779– 789, DOI: 10.1016/j.ajhg.2010.10.02416https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsFags7bM&md5=763f424eb72a032e6393b2bb36b1116eGene expression in skin and lymphoblastoid cells: refined statistical method reveals extensive overlap in cis-eQTL signalsDing, Jun; Gudjonsson, Johann E.; Liang, Liming; Stuart, Philip E.; Li, Yun; Chen, Wei; Weichenthal, Michael; Ellinghaus, Eva; Franke, Andre; Cookson, William; Nair, Rajan P.; Elder, James T.; Abecasis, Goncalo R.American Journal of Human Genetics (2010), 87 (6), 779-789CODEN: AJHGAG; ISSN:0002-9297. (Cell Press)Psoriasis, an immune-mediated, inflammatory disease of the skin and joints, provides an ideal system for expression quant. trait locus (eQTL) anal., because it has a strong genetic basis and disease-relevant tissue (skin) is readily accessible. To better understand the role of genetic variants regulating cutaneous gene expression, we identified 841 cis-acting eQTLs using RNA extd. from skin biopsies of 53 psoriatic individuals and 57 healthy controls. We found substantial overlap between cis-eQTLs of normal control, uninvolved psoriatic, and lesional psoriatic skin. Consistent with recent studies and with the idea that control of gene expression can mediate relationships between genetic variants and disease risk, we found that eQTL SNPs are more likely to be assocd. with psoriasis than are randomly selected SNPs. To explore the tissue specificity of these eQTLs and hence to quantify the benefits of studying eQTLs in different tissues, we developed a refined statistical method for estg. eQTL overlap and used it to compare skin eQTLs to a published panel of lymphoblastoid cell line (LCL) eQTLs. Our method accounts for the fact that most eQTL studies are likely to miss some true eQTLs as a result of power limitations and shows that ∼70% of cis-eQTLs in LCLs are shared with skin, as compared with the naive est. of <50% sharing. Our results provide a useful method for estg. the overlap between various eQTL studies and provide a catalog of cis-eQTLs in skin that can facilitate efforts to understand the functional impact of identified susceptibility variants on psoriasis and other skin traits.
- 17Firmann, M.; Mayor, V.; Vidal, P. M.; Bochud, M.; Pécoud, A.; Hayoz, D. The CoLaus study: a population-based study to investigate the epidemiology and genetic determinants of cardiovascular risk factors and metabolic syndrome. BMC Cardiovasc. Disord. 2008, 8, 6, DOI: 10.1186/1471-2261-8-617https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1c3ltF2luw%253D%253D&md5=6ffcf0d9742f2c91a6d24583188b9380The CoLaus study: a population-based study to investigate the epidemiology and genetic determinants of cardiovascular risk factors and metabolic syndromeFirmann Mathieu; Mayor Vladimir; Vidal Pedro Marques; Bochud Murielle; Pecoud Alain; Hayoz Daniel; Paccaud Fred; Preisig Martin; Song Kijoung S; Yuan Xin; Danoff Theodore M; Stirnadel Heide A; Waterworth Dawn; Mooser Vincent; Waeber Gerard; Vollenweider PeterBMC cardiovascular disorders (2008), 8 (), 6 ISSN:.BACKGROUND: Cardiovascular diseases and their associated risk factors remain the main cause of mortality in western societies. In order to assess the prevalence of cardiovascular risk factors (CVRFs) in the Caucasian population of Lausanne, Switzerland, we conducted a population-based study (Colaus Study). A secondary aim of the CoLaus study will be to determine new genetic determinants associated with CVRFs. METHODS: Single-center, cross-sectional study including a random sample of 6,188 extensively phenotyped Caucasian subjects (3,251 women and 2,937 men) aged 35 to 75 years living in Lausanne, and genotyped using the 500 K Affymetrix chip technology. RESULTS: Obesity (body mass index > or = 30 kg/m2), smoking, hypertension (blood pressure > or = 140/90 mmHg and/or treatment), dyslipidemia (high LDL-cholesterol and/or low HDL-cholesterol and/or high triglyceride levels) and diabetes (fasting plasma glucose > or = 7 mmol/l and/or treatment) were present in 947 (15.7%), 1673 (27.0%), 2268 (36.7%), 2113 (34.2%) and 407 (6.6%) of the participants, respectively, and the prevalence was higher in men than in women. In both genders, the prevalence of obesity, hypertension and diabetes increased with age. CONCLUSION: The prevalence of major CVRFs is high in the Lausanne population in particular in men. We anticipate that given its size, the depth of the phenotypic analysis and the availability of dense genome-wide genetic data, the CoLaus Study will be a unique resource to investigate not only the epidemiology of isolated, or aggregated CVRFs like the metabolic syndrome, but can also serve as a discovery set, as well as replication set, to identify novel genes associated with these conditions.
- 18Rueedi, R.; Ledda, M.; Nicholls, A. W.; Salek, R. M.; Marques-Vidal, P.; Morya, E. Genome-wide association study of metabolic traits reveals novel gene-metabolite-disease links. PLoS Genet. 2014, 10, e1004132, DOI: 10.1371/journal.pgen.100413218https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXkvVOnsbg%253D&md5=69bc097be50d0028080e1391f85db825Genome-wide association study of metabolic traits reveals novel gene-metabolite-disease linksRueedi, Rico; Ledda, Mirko; Nicholls, Andrew W.; Salek, Reza M.; Marques-Vidal, Pedro; Morya, Edgard; Sameshima, Koichi; Montoliu, Ivan; Da Silva, Laeticia; Collino, Sebastiano; Martin, Francois-Pierre; Rezzi, Serge; Steinbeck, Christoph; Waterworth, Dawn M.; Waeber, Gerard; Vollenweider, Peter; Beckmann, Jacques S.; Le Coutre, Johannes; Mooser, Vincent; Bergmann, Sven; Genick, Ulrich K.; Kutalik, ZoltanPLoS Genetics (2014), 10 (2), e1004132/1-e1004132/10, 10 pp.CODEN: PGLEB5; ISSN:1553-7404. (Public Library of Science)Metabolic traits are mol. phenotypes that can drive clin. phenotypes and may predict disease progression. Here, we report results from a metabolome- and genome-wide assocn. study on 1H-NMR urine metabolic profiles. The study was conducted within an untargeted approach, employing a novel method for compd. identification. From our discovery cohort of 835 Caucasian individuals who participated in the CoLaus study, we identified 139 suggestively significant (P<5 × 10-8) and independent assocns. between single nucleotide polymorphisms (SNP) and metabolome features. Fifty-six of these assocns. replicated in the TasteSensomics cohort, comprising 601 individuals from S~ao Paulo of vastly diverse ethnic background. They correspond to eleven gene-metabolite assocns., six of which had been previously identified in the urine metabolome and three in the serum metabolome. Our key novel findings are the assocns. of two SNPs with NMR spectral signatures pointing to fucose (rs492602, P = 6.9 × 10-44) and lysine (rs8101881, P = 1.2 × 10-33), resp. Fine-mapping of the first locus pinpointed the FUT2 gene, which encodes a fucosyltransferase enzyme and has previously been assocd. with Crohn's disease. This implicates fucose as a potential prognostic disease marker, for which there is already published evidence from a mouse model. The second SNP lies within the SLC7A9 gene, rare mutations of which have been linked to severe kidney damage. The replication of previous assocns. and our new discoveries demonstrate the potential of untargeted metabolomics GWAS to robustly identify mol. disease markers.
- 19Marco-Sola, S.; Sammeth, M.; Guigó, R.; Ribeca, P. The GEM mapper: fast, accurate and versatile alignment by filtration. Nat. Methods 2012, 9, 1185, DOI: 10.1038/nmeth.222119https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsFOmt73O&md5=559ff724a6a064d7280d553793f21d4fThe GEM mapper: fast, accurate and versatile alignment by filtrationMarco-Sola, Santiago; Sammeth, Michael; Guigo, Roderic; Ribeca, PaoloNature Methods (2012), 9 (12), 1185-1188CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)Because of ever-increasing throughput requirements of sequencing data, most existing short-read aligners have been designed to focus on speed at the expense of accuracy. The Genome Multitool (GEM) mapper can leverage string matching by filtration to search the alignment space more efficiently, simultaneously delivering precision (performing fully tunable exhaustive searches that return all existing matches, including gapped ones) and speed (being several times faster than comparable state-of-the-art tools).
- 20MATLAB 8.5.0.197613 (R2015a); The MathWorks Inc.: Natick, Massachusetts, 2015.There is no corresponding record for this reference.
- 21Gao, X.; Starmer, J.; Martin, E. R. A multiple testing correction method for genetic association studies using correlated single nucleotide polymorphisms. Genet. Epidemiol. 2008, 32, 361– 369, DOI: 10.1002/gepi.2031021https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1c3msl2gug%253D%253D&md5=ea3ae76ef9371956e694e05c166ebbceA multiple testing correction method for genetic association studies using correlated single nucleotide polymorphismsGao Xiaoyi; Starmer Joshua; Martin Eden RGenetic epidemiology (2008), 32 (4), 361-9 ISSN:0741-0395.Multiple testing is a challenging issue in genetic association studies using large numbers of single nucleotide polymorphism (SNP) markers, many of which exhibit linkage disequilibrium (LD). Failure to adjust for multiple testing appropriately may produce excessive false positives or overlook true positive signals. The Bonferroni method of adjusting for multiple comparisons is easy to compute, but is well known to be conservative in the presence of LD. On the other hand, permutation-based corrections can correctly account for LD among SNPs, but are computationally intensive. In this work, we propose a new multiple testing correction method for association studies using SNP markers. We show that it is simple, fast and more accurate than the recently developed methods and is comparable to permutation-based corrections using both simulated and real data. We also demonstrate how it might be used in whole-genome association studies to control type I error. The efficiency and accuracy of the proposed method make it an attractive choice for multiple testing adjustment when there is high intermarker LD in the SNP data set.
- 22Rueedi, R.; Mallol, R.; Raffler, J.; Lamparter, D.; Friedrich, N.; Vollenweider, P. Metabomatching: Using genetic association to identify metabolites in proton NMR spectroscopy. PLoS Comput. Biol. 2017, 13, e1005839, DOI: 10.1371/journal.pcbi.100583922https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsFags7vK&md5=842831a300f5401d3db64e43c372ce62Metabomatching: using genetic association to identify metabolites in proton NMR spectroscopyRueedi, Rico; Mallol, Roger; Raffler, Johannes; Lamparter, David; Friedrich, Nele; Vollenweider, Peter; Waeber, Gerard; Kastenmuller, Gabi; Kutalik, Zoltan; Bergmann, SvenPLoS Computational Biology (2017), 13 (12), e1005839/1-e1005839/17CODEN: PCBLBG; ISSN:1553-7358. (Public Library of Science)A metabolome-wide genome-wide assocn. study aims to discover the effects of genetic variants on metabolome phenotypes. Most mGWASes use as phenotypes concns. of limited sets of metabolites that can be identified and quantified from spectral information. In contrast, in an untargeted mGWAS both identification and quantification are forgone and, instead, all measured metabolome features are tested for assocn. with genetic variants. While the untargeted approach does not discard data that may have eluded identification, the interpretation of assocd. features remains a challenge. To address this issue, we developed metabomatching to identify the metabolites underlying significant assocns. obsd. in untargeted mGWASes on proton NMR metabolome data. Metabomatching capitalizes on genetic spiking, the concept that because metabolome features assocd. with a genetic variant tend to correspond to the peaks of the NMR spectrum of the underlying metabolite, genetic assocn. can allow for identification. Applied to the untargeted mGWASes in the SHIP and CoLaus cohorts and using 180 ref. NMR spectra of the urine metabolome database, metabomatching successfully identified the underlying metabolite in 14 of 19, and 8 of 9 assocns., resp. The accuracy and efficiency of our method make it a strong contender for facilitating or complementing metabolomics analyses in large cohorts, where the availability of genetic, or other data, enables our approach, but targeted quantification is limited.
- 23Wishart, D. S.; Feunang, Y. D.; Marcu, A.; Guo, A. C.; Liang, K.; Vázquez-Fresno, R. HMDB 4.0: the human metabolome database for 2018. Nucleic Acids Res. 2018, 46, D608– D617, DOI: 10.1093/nar/gkx108923https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitlGisb%252FF&md5=29725f192d00a0e31a35401058a32082HMDB 4.0: the human metabolome database for 2018Wishart, David S.; Feunang, Yannick Djoumbou; Marcu, Ana; Guo, An Chi; Liang, Kevin; Vazquez-Fresno, Rosa; Sajed, Tanvir; Johnson, Daniel; Li, Carin; Karu, Naama; Sayeeda, Zinat; Lo, Elvis; Assempour, Nazanin; Berjanskii, Mark; Singhal, Sandeep; Arndt, David; Liang, Yonjie; Badran, Hasan; Grant, Jason; Serra-Cayuela, Arnau; Liu, Yifeng; Mandal, Rupa; Neveu, Vanessa; Pon, Allison; Knox, Craig; Wilson, Michael; Manach, Claudine; Scalbert, AugustinNucleic Acids Research (2018), 46 (D1), D608-D617CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)A review. The Human Metabolome Database or HMDB (www. hmdb.ca) is a web-enabled metabolomic database contg. comprehensive information about human metabolites along with their biol. roles, physiol. concns., disease assocns., chem. reactions, metabolic pathways, and ref. spectra. First described in 2007, the HMDB is now considered the std. metabolomic resource for human metabolic studies. Over the past decade the HMDB has continued to grow and evolve in response to emerging needs for metabolomics researchers and continuing changes in web stds. This year's update, HMDB 4.0, represents the most significant upgrade to the database in its history. For instance, the no. of fully annotated metabolites has increased by nearly threefold, the no. of exptl. spectra has grown by almost fourfold and the no. of illustrated metabolic pathways has grown by a factor of almost 60. Significant improvements have also been made to the HMDB's chem. taxonomy, chem. ontol., spectral viewing, and spectral/text searching tools. A great deal of brand new data has also been added to HMDB 4.0. This includes large quantities of predicted MS/MS and GC-MS ref. spectral data as well as predicted (physiol. feasible) metabolite structures to facilitate novel metabolite identification. Addnl. information on metabolite-SNP interactions and the influence of drugs on metabolite levels (pharmaco-metabolomics) has also been added. Many other important improvements in the content, the interface, and the performance of the HMDB website have been made and these should greatly enhance its ease of use and its potential applications in nutrition, biochem., clin. chem., clin. genetics, medicine, and metabolomics science.
- 24Khalili, B.; Tomasoni, M.; Mattei, M.; Parera, R. M.; Sonmez, R.; Krefl, D. Automated analysis of large-scale NMR data generates metabolomic signatures and links them to candidate metabolites. J. Proteome Res. 2019, 613935, DOI: 10.1021/acs.jproteome.9b00295There is no corresponding record for this reference.
- 25Burgess, S.; Small, D. S.; Thompson, S. G. A review of instrumental variable estimators for Mendelian randomization. Stat. Methods Med. Res. 2017, 26, 2333– 2355, DOI: 10.1177/096228021559757925https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC287jsl2quw%253D%253D&md5=6c22b011ffcad08533fbaa060bf97b9bA review of instrumental variable estimators for Mendelian randomizationBurgess Stephen; Thompson Simon G; Small Dylan SStatistical methods in medical research (2017), 26 (5), 2333-2355 ISSN:.Instrumental variable analysis is an approach for obtaining causal inferences on the effect of an exposure (risk factor) on an outcome from observational data. It has gained in popularity over the past decade with the use of genetic variants as instrumental variables, known as Mendelian randomization. An instrumental variable is associated with the exposure, but not associated with any confounder of the exposure-outcome association, nor is there any causal pathway from the instrumental variable to the outcome other than via the exposure. Under the assumption that a single instrumental variable or a set of instrumental variables for the exposure is available, the causal effect of the exposure on the outcome can be estimated. There are several methods available for instrumental variable estimation; we consider the ratio method, two-stage methods, likelihood-based methods, and semi-parametric methods. Techniques for obtaining statistical inferences and confidence intervals are presented. The statistical properties of estimates from these methods are compared, and practical advice is given about choosing a suitable analysis method. In particular, bias and coverage properties of estimators are considered, especially with weak instruments. Settings particularly relevant to Mendelian randomization are prioritized in the paper, notably the scenario of a continuous exposure and a continuous or binary outcome.
- 26Davey Smith, G.; Ebrahim, S. ‘Mendelian randomization’: can genetic epidemiology contribute to understanding environmental determinants of disease?. Int. J. Epidemiol. 2003, 32, 1– 22, DOI: 10.1093/ije/dyg070There is no corresponding record for this reference.
- 27Raffler, J.; Friedrich, N.; Arnold, M.; Kacprowski, T.; Rueedi, R.; Altmaier, E. Genome-wide association study with targeted and non-targeted NMR metabolomics identifies 15 novel loci of urinary human metabolic individuality. PLoS Genet. 2015, 11, e1005487, DOI: 10.1371/journal.pgen.100548727https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xls1ygu7g%253D&md5=46d58208d9b118aad16308ff59f3b0daGenome-wide association study with targeted and non-targeted nmr metabolomics identifies 15 novel loci of urinary human metabolic individualityRaffler, Johannes; Friedrich, Nele; Arnold, Matthias; Kacprowski, Tim; Rueedi, Rico; Altmaier, Elisabeth; Bergmann, Sven; Budde, Kathrin; Gieger, Christian; Homuth, Georg; Pietzner, Maik; Roemisch-Margl, Werner; Strauch, Konstantin; Voelzke, Henry; Waldenberger, Melanie; Wallaschofski, Henri; Nauck, Matthias; Voelker, Uwe; Kastenmueller, Gabi; Suhre, KarstenPLoS Genetics (2015), 11 (9), e1005487/1-e1005487/28CODEN: PGLEB5; ISSN:1553-7404. (Public Library of Science)Genome-wide assocn. studies with metabolic traits (mGWAS) uncovered many genetic variants that influence human metab. These genetically influenced metabotypes (GIMs) contribute to our metabolic individuality, our capacity to respond to environmental challenges, and our susceptibility to specific diseases. While metabolic homeostasis in blood is a well investigated topic in large mGWAS with over 150 known loci, metabolic detoxification through urinary excretion has only been addressed by few small mGWAS with only 11 assocd. loci so far. Here we report the largest mGWAS to date, combining targeted and non-targeted 1H NMR anal. of urine samples from 3,861 participants of the SHIP-0 cohort and 1,691 subjects of the KORA F4 cohort. We identified and replicated 22 loci with significant assocns. with urinary traits, 15 of which are new (HIBCH, CPS1, AGXT, XYLB, TKT, ETNPPL, SLC6A19, DMGDH, SLC36A2, GLDC, SLC6A13, ACSM3, SLC5A11, PNMT, SLC13A3). Two-thirds of the urinary loci also have a metabolite assocn. in blood. For all but one of the 6 loci where significant assocns. target the same metabolite in blood and urine, the genetic effects have the same direction in both fluids. In contrast, for the SLC5A11 locus, we found increased levels of myo-inositol in urine whereas mGWAS in blood reported decreased levels for the same genetic variant. This might indicate less effective re-absorption of myo-inositol in the kidneys of carriers. In summary, our study more than doubles the no. of known loci that influence urinary phenotypes. It thus allows novel insights into the relationship between blood homeostasis and its regulation through excretion. The newly discovered loci also include variants previously linked to chronic kidney disease (CPS1, SLC6A13), pulmonary hypertension (CPS1), and ischemic stroke (XYLB). By establishing connections from gene to disease via metabolic traits our results provide novel hypotheses about mol. mechanisms involved in the etiol. of diseases.
- 28Võsa, U.; Claringbould, A.; Westra, H.-J.; Bonder, M. J.; Deelen, P.; Zeng, B.; Kirsten, H.; Saha, A.; Kreuzhuber, R.; Brugge, H. Large-scale cis-and trans-eQTL analyses identify thousands of genetic loci and polygenic scores that regulate blood gene expression. Nat. Genet. 2021, 53, 1300– 1310, DOI: 10.1038/s41588-021-00913-z28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvFGmtbfE&md5=d78fcc21a5127342cbdddadc10a596fdLarge-scale cis- and trans-eQTL analyses identify thousands of genetic loci and polygenic scores that regulate blood gene expressionVosa, Urmo; Claringbould, Annique; Westra, Harm-Jan; Bonder, Marc Jan; Deelen, Patrick; Zeng, Biao; Kirsten, Holger; Saha, Ashis; Kreuzhuber, Roman; Yazar, Seyhan; Brugge, Harm; Oelen, Roy; de Vries, Dylan H.; van der Wijst, Monique G. P.; Kasela, Silva; Pervjakova, Natalia; Alves, Isabel; Fave, Marie-Julie; Agbessi, Mawusse; Christiansen, Mark W.; Jansen, Rick; Seppala, Ilkka; Tong, Lin; Teumer, Alexander; Schramm, Katharina; Hemani, Gibran; Verlouw, Joost; Yaghootkar, Hanieh; Sonmez Flitman, Reyhan; Brown, Andrew; Kukushkina, Viktorija; Kalnapenkis, Anette; Rueger, Sina; Porcu, Eleonora; Kronberg, Jaanika; Kettunen, Johannes; Lee, Bernett; Zhang, Futao; Qi, Ting; Hernandez, Jose Alquicira; Arindrarto, Wibowo; Beutner, Frank; Dmitrieva, Julia; Elansary, Mahmoud; Fairfax, Benjamin P.; Georges, Michel; Heijmans, Bastiaan T.; Hewitt, Alex W.; Kahonen, Mika; Kim, Yungil; Knight, Julian C.; Kovacs, Peter; Krohn, Knut; Li, Shuang; Loeffler, Markus; Marigorta, Urko M.; Mei, Hailang; Momozawa, Yukihide; Muller-Nurasyid, Martina; Nauck, Matthias; Nivard, Michel G.; Penninx, Brenda W. J. H.; Pritchard, Jonathan K.; Raitakari, Olli T.; Rotzschke, Olaf; Slagboom, Eline P.; Stehouwer, Coen D. A.; Stumvoll, Michael; Sullivan, Patrick; 't Hoen, Peter A. C.; Thiery, Joachim; Tonjes, Anke; van Dongen, Jenny; van Iterson, Maarten; Veldink, Jan H.; Volker, Uwe; Warmerdam, Robert; Wijmenga, Cisca; Swertz, Morris; Andiappan, Anand; Montgomery, Grant W.; Ripatti, Samuli; Perola, Markus; Kutalik, Zoltan; Dermitzakis, Emmanouil; Bergmann, Sven; Frayling, Timothy; van Meurs, Joyce; Prokisch, Holger; Ahsan, Habibul; Pierce, Brandon L.; Lehtimaki, Terho; Boomsma, Dorret I.; Psaty, Bruce M.; Gharib, Sina A.; Awadalla, Philip; Milani, Lili; Ouwehand, Willem H.; Downes, Kate; Stegle, Oliver; Battle, Alexis; Visscher, Peter M.; Yang, Jian; Scholz, Markus; Powell, Joseph; Gibson, Greg; Esko, Tonu; Franke, LudeNature Genetics (2021), 53 (9), 1300-1310CODEN: NGENEC; ISSN:1061-4036. (Nature Portfolio)Trait-assocd. genetic variants affect complex phenotypes primarily via regulatory mechanisms on the transcriptome. To investigate the genetics of gene expression, we performed cis- and trans-expression quant. trait locus (eQTL) analyses using blood-derived expression from 31,684 individuals through the eQTLGen Consortium. We detected cis-eQTL for 88% of genes, and these were replicable in numerous tissues. Distal trans-eQTL (detected for 37% of 10,317 trait-assocd. variants tested) showed lower replication rates, partially due to low replication power and confounding by cell type compn. However, replication analyses in single-cell RNA-seq data prioritized intracellular trans-eQTL. Trans-eQTL exerted their effects via several mechanisms, primarily through regulation by transcription factors. Expression of 13% of the genes correlated with polygenic scores for 1,263 phenotypes, pinpointing potential drivers for those traits. In summary, this work represents a large eQTL resource, and its results serve as a starting point for in-depth interpretation of complex phenotypes.
- 29Wald, A. The fitting of straight lines if both variables are subject to error. Ann. Math. Stat. 1940, 11, 284– 300, DOI: 10.1214/aoms/1177731868There is no corresponding record for this reference.
- 30Hartung, J.; Knapp, G.; Sinha, B. K.; Sinha, B. K. Statistical meta-analysis with applications; Wiley: New York, 2008.There is no corresponding record for this reference.
- 31Greco, M. F. D.; Minelli, C.; Sheehan, N. A.; Thompson, J. R. Detecting pleiotropy in Mendelian randomisation studies with summary data and a continuous outcome. Stat. Med. 2015, 34, 2926– 2940, DOI: 10.1002/sim.652231https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2MfhtlCjtg%253D%253D&md5=dab1f5ee1ce3851064ebee6727d9bf16Detecting pleiotropy in Mendelian randomisation studies with summary data and a continuous outcomeGreco M Fabiola Del; Minelli Cosetta; Sheehan Nuala A; Thompson John RStatistics in medicine (2015), 34 (21), 2926-40 ISSN:.Mendelian randomisation (MR) estimates causal effects of modifiable phenotypes on an outcome by using genetic variants as instrumental variables, but its validity relies on the assumption of no pleiotropy, that is, genes influence the outcome only through the given phenotype. Excluding pleiotropy is difficult, but the use of multiple instruments can indirectly address the issue: if all genes represent valid instruments, their MR estimates should vary only by chance. The Sargan test detects pleiotropy when individual phenotype, outcome and genotype data are measured in the same subjects. We propose an alternative approach to be used when only summary genetic data are available or data on gene-phenotype and gene-outcome come from different subjects. The presence of pleiotropy is investigated using the between-instrument heterogeneity Q test (together with the I(2) index) in a meta-analysis of MR Wald estimates, derived separately from each instrument. For a continuous outcome, we evaluate the approach through simulations and illustrate it using published data. For the scenario where all data come from the same subjects, we compare it with the Sargan test. The Q test tends to be conservative in small samples. Its power increases with the degree of pleiotropy and the sample size, as does the precision of the I(2) index, in which case results are similar to those of the Sargan test. In MR studies with large sample sizes based on summary data, the between-instrument Q test represents a useful tool to explore the presence of heterogeneity due to pleiotropy or other causes.
- 32Yavorska, O. O.; Burgess, S. MendelianRandomization: an R package for performing Mendelian randomization analyses using summarized data. Int. J. Epidemiol. 2017, 46, 1734– 1739, DOI: 10.1093/ije/dyx03432https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1cvmslehsA%253D%253D&md5=d8d13430779261f1a45cf0cbdde85f8eMendelianRandomization: an R package for performing Mendelian randomization analyses using summarized dataYavorska Olena O; Burgess StephenInternational journal of epidemiology (2017), 46 (6), 1734-1739 ISSN:.MendelianRandomization is a software package for the R open-source software environment that performs Mendelian randomization analyses using summarized data. The core functionality is to implement the inverse-variance weighted, MR-Egger and weighted median methods for multiple genetic variants. Several options are available to the user, such as the use of robust regression, fixed- or random-effects models and the penalization of weights for genetic variants with heterogeneous causal estimates. Extensions to these methods, such as allowing for variants to be correlated, can be chosen if appropriate. Graphical commands allow summarized data to be displayed in an interactive graph, or the plotting of causal estimates from multiple methods, for comparison. Although the main method of data entry is directly by the user, there is also an option for allowing summarized data to be incorporated from the PhenoScanner database of genotype-phenotype associations. We hope to develop this feature in future versions of the package. The R software environment is available for download from [https://www.r-project.org/]. The MendelianRandomization package can be downloaded from the Comprehensive R Archive Network (CRAN) within R, or directly from [https://cran.r-project.org/web/packages/MendelianRandomization/]. Both R and the MendelianRandomization package are released under GNU General Public Licenses (GPL-2 GPL-3).
- 33Shin, S.-Y.; Fauman, E. B.; Petersen, A.-K.; Krumsiek, J.; Santos, R.; Huang, J. An atlas of genetic influences on human blood metabolites. Nat. Genet. 2014, 46, 543, DOI: 10.1038/ng.298233https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXnslGktro%253D&md5=ba0b0c68c921ef6ea01105fce89818d4An atlas of genetic influences on human blood metabolitesShin, So-Youn; Fauman, Eric B.; Petersen, Ann-Kristin; Krumsiek, Jan; Santos, Rita; Huang, Jie; Arnold, Matthias; Erte, Idil; Forgetta, Vincenzo; Yang, Tsun-Po; Walter, Klaudia; Menni, Cristina; Chen, Lu; Vasquez, Louella; Valdes, Ana M.; Hyde, Craig L.; Wang, Vicky; Ziemek, Daniel; Roberts, Phoebe; Xi, Li; Grundberg, Elin; Waldenberger, Melanie; Richards, J. Brent; Mohney, Robert P.; Milburn, Michael V.; John, Sally L.; Trimmer, Jeff; Theis, Fabian J.; Overington, John P.; Suhre, Karsten; Brosnan, M. Julia; Gieger, Christian; Kastenmuller, Gabi; Spector, Tim D.; Soranzo, NicoleNature Genetics (2014), 46 (6), 543-550CODEN: NGENEC; ISSN:1061-4036. (Nature Publishing Group)Genome-wide assocn. scans with high-throughput metabolic profiling provide unprecedented insights into how genetic variation influences metab. and complex disease. Here the authors report the most comprehensive exploration of genetic loci influencing human metab. thus far, comprising 7,824 adult individuals from 2 European population studies. The authors report genome-wide significant assocns. at 145 metabolic loci and their biochem. connectivity with more than 400 metabolites in human blood. The authors extensively characterize the resulting in vivo blueprint of metab. in human blood by integrating it with information on gene expression, heritability and overlap with known loci for complex disorders, inborn errors of metab. and pharmacol. targets. The authors further developed a database and web-based resources for data mining and results visualization. The authors' findings provide new insights into the role of inherited variation in blood metabolic diversity and identify potential new opportunities for drug development and for understanding disease.
- 34Nicholson, G.; Rantalainen, M.; Li, J. V.; Maher, A. D.; Malmodin, D.; Ahmadi, K. R. A genome-wide metabolic QTL analysis in Europeans implicates two loci shaped by recent positive selection. PLoS Genet. 2011, 7, e1002270, DOI: 10.1371/journal.pgen.100227034https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXht1ChsbvE&md5=3115de8a4d2238d5ce02ef0ed273d58fA genome-wide metabolic QTL analysis in Europeans implicates two loci shaped by recent positive selectionNicholson, George; Rantalainen, Mattias; Li, Jia V.; Maher, Anthony D.; Malmodin, Daniel; Ahmadi, Kourosh R.; Faber, Johan H.; Barrett, Amy; Min, Josine L.; Rayner, N. William; Toft, Henrik; Krestyaninova, Maria; Viksna, Juris; Neogi, Sudeshna Guha; Dumas, Marc-Emmanuel; Sarkans, Ugis; Donnelly, Peter; Illig, Thomas; Adamski, Jerzy; Suhre, Karsten; Allen, Maxine; Zondervan, Krina T.; Spector, Tim D.; Nicholson, Jeremy K.; Lindon, John C.; Baunsgaard, Dorrit; Holmes, Elaine; McCarthy, Mark I.; Holmes, Chris C.PLoS Genetics (2011), 7 (9), e1002270CODEN: PGLEB5; ISSN:1553-7404. (Public Library of Science)We have performed a metabolite quant. trait locus (mQTL) study of the 1H NMR spectroscopy metabolome in humans, building on recent targeted knowledge of genetic drivers of metabolic regulation. Urine and plasma samples were collected from two cohorts of individuals of European descent, with one cohort comprised of female twins donating samples longitudinally. Sample metabolite concns. were quantified by 1H NMR and tested for assocn. with genome-wide single-nucleotide polymorphisms (SNPs). 4 Metabolites' concns. exhibited significant, replicable assocn. with SNP variation (8.6 × 10-11 < p < 2.8 × 10-23). 3 Of these, trimethylamine, 3-amino-isobutyrate, and an N-acetylated compd., were measured in urine. The other, dimethylamine, was measured in plasma. Trimethylamine and dimethylamine mapped to a single genetic region (hence we report a total of three implicated genomic regions). 2 Of the 3 hit regions lie within haplotype blocks (at 2p13.1 and 10q24.2) that carry the genetic signature of strong, recent, pos. selection in European populations. Genes NAT8 and PYROXD2, both with relatively uncharacterized functional roles, are good candidates for mediating the corresponding mQTL assocns. The studies longitudinal twin design allowed detailed variance-components anal. of the sources of population variation in metabolite levels. The mQTLs explained 40%-64% of biol. population variation in the corresponding metabolites' concns. These effect sizes are stronger than those reported in a recent, targeted mQTL study of metabolites in serum using the targeted-metabolomics Biocrates platform. By re-analyzing our plasma samples using the Biocrates platform, we replicated the mQTL findings of the previous study and discovered a previously uncharacterized yet substantial familial component of variation in metabolite levels in addn. to the heritability contribution from the corresponding mQTL effects.
- 35Engelke, U. F.; Liebrand-van Sambeek, M. L.; De Jong, J. G.; Leroy, J. G.; Morava, E.; Smeitink, J. A. N-acetylated metabolites in urine: proton nuclear magnetic resonance spectroscopic study on patients with inborn errors of metabolism. Clin. Chem. 2004, 50, 58– 66, DOI: 10.1373/clinchem.2003.02021435https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXovFWhtw%253D%253D&md5=80e2e8bb8453ccce361195b7b8e695faN-acetylated metabolites in urine: Proton nuclear magnetic resonance spectroscopic study on patients with inborn errors of metabolismEngelke, Udo F. H.; Liebrand-van Sambeek, Maria L. F.; de Jong, Jan G. N.; Leroy, Jules G.; Morava, Eva; Smeitink, Jan A. M.; Wevers, Ron A.Clinical Chemistry (Washington, DC, United States) (2004), 50 (1), 58-66CODEN: CLCHAU; ISSN:0009-9147. (American Association for Clinical Chemistry)There is no comprehensive anal. technique to analyze N-acetylated metabolites in urine. Many of these compds. are involved in inborn errors of metab. In the present study, the authors examd. the potential of proton NMR (1H-NMR) spectroscopy as a tool to identify and quantify N-acetylated metabolites in urine of patients with various inborn errors of metab. The authors performed 1H-NMR spectroscopy on a 500 MHz spectrometer. Using a combination of 1- and 2-dimensional correlation spectroscopy (COSY) 1H-NMR spectra, the authors were able to assign and quantify resonances of characteristic N-acetylated compds. products in urine of patients with 13 inborn errors of metab. The disease-specific N-acetylated metabolites were excreted at concns. >100 μmol/mmol of creatinine in the patients' urine. In control urine samples, the concn. of individual N-acetyl-contg. compds. was <40 μmol/mmol of creatinine. The combination of one- and two-dimensional COSY NMR spectroscopy led to the correct diagnosis of nine different inborn errors of metab. No abnormalities were obsd. in the spectra of urine from patients with GM1- or GM2-gangliosidosis. The authors also detd. the 1H-NMR characteristics of N-acetylated metabolites that may be relevant to human metab. 1H-NMR spectroscopy may be used to identify and quantify N-acetylated metabolites of diagnostic importance for the field of inborn errors of metab.
- 36Benjamini, Y.; Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. Royal Stat. Soc.: B 1995, 57, 289– 300, DOI: 10.1111/j.2517-6161.1995.tb02031.xThere is no corresponding record for this reference.
- 37Kastenmüller, G.; Raffler, J.; Gieger, C.; Suhre, K. Genetics of human metabolism: an update. Hum. Mol. Genet. 2015, 24, R93– R101, DOI: 10.1093/hmg/ddv26337https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXitVCqu7nP&md5=a421ead21475ebed82390dbead5d6d97Genetics of human metabolism: an updateKastenmueller, Gabi; Raffler, Johannes; Gieger, Christian; Suhre, KarstenHuman Molecular Genetics (2015), 24 (Rev. Issue 1), R93-R101CODEN: HMGEE5; ISSN:0964-6906. (Oxford University Press)A review. Genome-wide assocn. studies with metabolomics (mGWAS) identify genetically influenced metabotypes (GIMs), their ensemble defining the heritable part of every human's metabolic individuality. Knowledge of genetic variation in metab. has many applications of biomedical and pharmaceutical interests, including the functional understanding of genetic assocns. with clin. end points, design of strategies to correct dysregulations in metabolic disorders and the identification of genetic effect modifiers of metabolic disease biomarkers. Furthermore, it has been shown that GIMs provide testable hypotheses for functional genomics and metabolomics and for the identification of novel gene functions and metabolite identities. mGWAS with growing sample sizes and increasingly complex metabolic trait panels are being conducted, allowing for more comprehensive and systems-based downstream analyses. The generated large datasets of genetic assocns. can now be mined by the biomedical research community and provide valuable resources for hypothesis-driven studies. In this review, we provide a brief summary of the key aspects of mGWAS, followed by an update of recently published mGWAS. We then discuss new approaches of integrating and exploring mGWAS results and finish by presenting selected applications of GIMs in recent studies.
- 38Suhre, K.; Shin, S. Y.; Petersen, A. K.; Mohney, R. P.; Meredith, D.; Wägele, B. Human metabolic individuality in biomedical and pharmaceutical research. Nature 2011, 477, 54– 60, DOI: 10.1038/nature1035438https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtFWku7nN&md5=da346121e93b1d123d72dc4c0b634833Human metabolic individuality in biomedical and pharmaceutical researchSuhre, Karsten; Shin, So-Youn; Petersen, Ann-Kristin; Mohney, Robert P.; Meredith, David; Waegele, Brigitte; Altmaier, Elisabeth; Deloukas, Panos; Erdmann, Jeanette; Grundberg, Elin; Hammond, Christopher J.; Hrabe de Angelis, Martin; Kastenmueller, Gabi; Koettgen, Anna; Kronenberg, Florian; Mangino, Massimo; Meisinger, Christa; Meitinger, Thomas; Mewes, Hans-Werner; Milburn, Michael V.; Prehn, Cornelia; Raffler, Johannes; Ried, Janina S.; Roemisch-Margl, Werner; Samani, Nilesh J.; Small, Kerrin S.; Wichmann, H.-Erich; Zhai, Guang-Ju; Illig, Thomas; Spector, Tim D.; Adamski, Jerzy; Soranzo, Nicole; Gieger, ChristianNature (London, United Kingdom) (2011), 477 (7362), 54-60CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)A review based on author's and other researchers studies. Genome-wide assocn. studies (GWAS) have identified many risk loci for complex diseases, but effect sizes are typically small and information on the underlying biol. processes is often lacking. Assocns. with metabolic traits as functional intermediates can overcome these problems and potentially inform individualized therapy. Here we report a comprehensive anal. of genotype-dependent metabolic phenotypes using a GWAS with non-targeted metabolomics. We identified 37 genetic loci assocd. with blood metabolite concns., of which 25 show effect sizes that are unusually high for GWAS and account for 10-60% differences in metabolite levels per allele copy. Our assocns. provide new functional insights for many disease-related assocns. that have been reported in previous studies, including those for cardiovascular and kidney disorders, type 2 diabetes, cancer, gout, venous thromboembolism and Crohn's disease. The study advances our knowledge of the genetic basis of metabolic individuality in humans and generates many new hypotheses for biomedical and pharmaceutical research.
- 39Yu, B.; Zheng, Y.; Alexander, D.; Morrison, A. C.; Coresh, J.; Boerwinkle, E. Genetic determinants influencing human serum metabolome among African Americans. PLoS Genet. 2014, 10, e1004212 DOI: 10.1371/journal.pgen.100421239https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhsVGlsLnI&md5=8dc1533231f3e76be1739d1a8b1fc491Genetic determinants influencing human serum metabolome among African AmericansYu, Bing; Zheng, Yan; Alexander, Danny; Morrison, Alanna C.; Coresh, Josef; Boerwinkle, EricPLoS Genetics (2014), 10 (3), e1004212/1-e1004212/8, 8 pp.CODEN: PGLEB5; ISSN:1553-7404. (Public Library of Science)Phenotypes proximal to gene action generally reflect larger genetic effect sizes than those that are distant. The human metabolome, a result of multiple cellular and biol. processes, are functional intermediate phenotypes proximal to gene action. Here, we present a genome-wide assocn. study of 308 untargeted metabolite levels among African Americans from the Atherosclerosis Risk in Communities (ARIC) Study. Nineteen significant common variant-metabolite assocns. were identified, including 13 novel loci (p<1.6 × 1010). These loci were assocd. with 7-50 % of the difference in metabolite levels per allele, and the variance explained ranged from 4% to 20%. Fourteen genes were identified within the nineteen loci, and four of them contained non-synonymous substitutions in four enzyme-encoding genes (KLKB1, SIAE, CPS1, and NAT8); the other significant loci consist of eight other enzyme-encoding genes (ACE, GATM, ACY3, ACSM2B, THEM4, ADH4, UGT1A, TREH), a transporter gene (SLC6A13) and a polycystin protein gene (PKD2L1). In addn., four potential disease-assocd. paths were identified, including two direct longitudinal predictive relationships: NAT8 with N-acetylornithine, N-acetyl-1-methylhistidine and incident chronic kidney disease, and TREH with trehalose and incident diabetes. These results highlight the value of using endophenotypes proximal to gene function to discover new insights into biol. and disease pathol.
- 40Raffler, J.; Römisch-Margl, W.; Petersen, A. K.; Pagel, P.; Blöchl, F.; Hengstenberg, C. Identification and MS-assisted interpretation of genetically influenced NMR signals in human plasma. Genome Med. 2013, 5, 13, DOI: 10.1186/gm41740https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtVGqtbnE&md5=cf4b62ba8345c74d16110c4f7ab47396Identification and MS-assisted interpretation of genetically influenced NMR signals in human plasmaRaffler, Johannes; Roemisch-Margl, Werner; Petersen, Ann-Kristin; Pagel, Philipp; Bloechl, Florian; Hengstenberg, Christian; Illig, Thomas; Meisinger, Christa; Stark, Klaus; Wichmann, H.-Erich; Adamski, Jerzy; Gieger, Christian; Kastenmueller, Gabi; Suhre, KarstenGenome Medicine (2013), 5 (2), 13CODEN: GMEECG; ISSN:1756-994X. (BioMed Central Ltd.)NMR spectroscopy provides robust readouts of many metabolic parameters in one expt. However, identification of clin. relevant markers in 1H NMR spectra is a major challenge. Assocn. of NMR-derived quantities with genetic variants can uncover biol. relevant metabolic traits. Using NMR data of plasma samples from 1,757 individuals from the KORA study together with 655,658 genetic variants, we show that ratios between NMR intensities at two chem. shift positions can provide informative and robust biomarkers. We report seven loci of genetic assocn. with NMR-derived traits (APOA1, CETP, CPS1, GCKR, FADS1, LIPC, PYROXD2) and characterize these traits biochem. using mass spectrometry. These ratios may now be used in clin. studies.
- 41Rhee, E. P.; Ho, J. E.; Chen, M. H.; Shen, D.; Cheng, S.; Larson, M. G. A genome-wide association study of the human metabolome in a community-based cohort. Cell Metab. 2013, 18, 130– 143, DOI: 10.1016/j.cmet.2013.06.01341https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtVGrsr7P&md5=dcaac8e4c86e2360ad15c820c2839998A Genome-wide Association Study of the Human Metabolome in a Community-Based CohortRhee, Eugene P.; Ho, Jennifer E.; Chen, Ming-Huei; Shen, Dongxiao; Cheng, Susan; Larson, Martin G.; Ghorbani, Anahita; Shi, Xu; Helenius, Iiro T.; O'Donnell, Christopher J.; Souza, Amanda L.; Deik, Amy; Pierce, Kerry A.; Bullock, Kevin; Walford, Geoffrey A.; Vasan, Ramachandran S.; Florez, Jose C.; Clish, Clary; Yeh, J.-R. Joanna; Wang, Thomas J.; Gerszten, Robert E.Cell Metabolism (2013), 18 (1), 130-143CODEN: CMEEB5; ISSN:1550-4131. (Elsevier Inc.)Because metabolites are hypothesized to play key roles as markers and effectors of cardiometabolic diseases, recent studies have sought to annotate the genetic determinants of circulating metabolite levels. We report a genome-wide assocn. study (GWAS) of 217 plasma metabolites, including >100 not measured in prior GWAS, in 2076 participants of the Framingham Heart Study (FHS). For the majority of analytes, we find that estd. heritability explains >20% of interindividual variation, and that variation attributable to heritable factors is greater than that attributable to clin. factors. Further, we identify 31 genetic loci assocd. with plasma metabolites, including 23 that have not previously been reported. Importantly, we include GWAS results for all surveyed metabolites and demonstrate how this information highlights a role for AGXT2 in cholesterol ester and triacylglycerol metab. Thus, our study outlines the relative contributions of inherited and clin. factors on the plasma metabolome and provides a resource for metab. research.
- 42Krumsiek, J.; Suhre, K.; Evans, A. M.; Mitchell, M. W.; Mohney, R. P.; Milburn, M. V. Mining the unknown: a systems approach to metabolite identification combining genetic and metabolic information. PLoS Genet. 2012, 8, e1003005, DOI: 10.1371/journal.pgen.100300542https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xhs1antr7N&md5=cb9c81f3de0729332ee6b22a7c935c96Mining the unknown: a systems approach to metabolite identification combining genetic and metabolic informationKrumsiek, Jan; Suhre, Karsten; Evans, Anne M.; Mitchell, Matthew W.; Mohney, Robert P.; Milburn, Michael V.; Waegele, Brigitte; Roemisch-Margl, Werner; Illig, Thomas; Adamski, Jerzy; Gieger, Christian; Theis, Fabian J.; Kastenmueller, GabiPLoS Genetics (2012), 8 (10), e1003005CODEN: PGLEB5; ISSN:1553-7404. (Public Library of Science)Recent genome-wide assocn. studies (GWAS) with metabolomics data linked genetic variation in the human genome to differences in individual metabolite levels. A strong relevance of this metabolic individuality for biomedical and pharmaceutical research has been reported. However, a considerable amt. of the mols. currently quantified by modern metabolomics techniques are chem. unidentified. The identification of these "unknown metabolites" is still a demanding and intricate task, limiting their usability as functional markers of metabolic processes. As a consequence, previous GWAS largely ignored unknown metabolites as metabolic traits for the anal. Here we present a systems-level approach that combines genome-wide assocn. anal. and Gaussian graphical modeling with metabolomics to predict the identity of the unknown metabolites. We apply our method to original data of 517 metabolic traits, of which 225 are unknowns, and genotyping information on 655,658 genetic variants, measured in 1,768 human blood samples. We report previously undescribed genotype-metabotype assocns. for six distinct gene loci (SLC22A2, COMT, CYP3A5, CYP2C18, GBA3, UGT3A1) and one locus not related to any known gene (rs12413935). Overlaying the inferred genetic assocns., metabolic networks, and knowledge-based pathway information, we derive testable hypotheses on the biochem. identities of 106 unknown metabolites. As a proof of principle, we exptl. confirm nine concrete predictions. We demonstrate the benefit of our method for the functional interpretation of previous metabolomics biomarker studies on liver detoxification, hypertension, and insulin resistance. Our approach is generic in nature and can be directly transferred to metabolomics data from different exptl. platforms.
- 43Hong, M. G.; Karlsson, R.; Magnusson, P. K.; Lewis, M. R.; Isaacs, W.; Zheng, L. S. A Genome-Wide Assessment of Variability in Human Serum Metabolism. Hum. Mutat. 2013, 34, 515– 524, DOI: 10.1002/humu.2226743https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXivFKktL0%253D&md5=79fda7875ae6787b539bc283cadc3221A Genome-Wide Assessment of Variability in Human Serum MetabolismHong, Mun-Gwan; Karlsson, Robert; Magnusson, Patrik K. E.; Lewis, Matthew R.; Isaacs, William; Zheng, Lilly S.; Xu, Jianfeng; Groenberg, Henrik; Ingelsson, Erik; Pawitan, Yudi; Broeckling, Corey; Prenni, Jessica E.; Wiklund, Fredrik; Prince, Jonathan A.Human Mutation (2013), 34 (3), 515-524CODEN: HUMUE3; ISSN:1059-7794. (Wiley-Liss, Inc.)The study of the genetic regulation of metab. in human serum samples can contribute to a better understanding of the intermediate biol. steps that lead from polymorphism to disease. Here, we conducted a genome-wide assocn. study (GWAS) to discover metabolic quant. trait loci (mQTLs) utilizing samples from a study of prostate cancer in Swedish men, consisting of 402 individuals (214 cases and 188 controls) in a discovery set and 489 case-only samples in a replication set. A global nontargeted metabolite profiling approach was utilized resulting in the detection of 6,138 mol. features followed by targeted identification of assocd. metabolites. Seven replicating loci were identified (PYROXD2, FADS1, PON1, CYP4F2, UGT1A8, ACADL, and LIPC) with assocd. sequence variants contributing significantly to trait variance for one or more metabolites (P = 10-13-10-91). Regional mQTL enrichment analyses implicated two loci that included FADS1 and a novel locus near PDGFC. Biol. pathway anal. implicated ACADM, ACADS, ACAD8, ACAD10, ACAD11, and ACOXL, reflecting significant enrichment of genes with acyl-CoA dehydrogenase activity. mQTL SNPs and mQTL-harboring genes were over-represented across GWASs conducted to date, suggesting that these data may have utility in tracing the mol. basis of some complex disease assocns.
- 44Montoliu, I.; Genick, U.; Ledda, M.; Collino, S.; Martin, F.-P.; le Coutre, J. Current status on genome–metabolome-wide associations: an opportunity in nutrition research. Gene Nutr. 2013, 8, 19, DOI: 10.1007/s12263-012-0313-7There is no corresponding record for this reference.
- 45Chambers, J. C.; Zhang, W.; Lord, G. M.; Van Der Harst, P.; Lawlor, D. A.; Sehmi, J. S. Genetic loci influencing kidney function and chronic kidney disease. Nat. Genet. 2010, 42, 373– 375, DOI: 10.1038/ng.56646https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXksFakurk%253D&md5=0c2bb874fec4125b00fc801bc8359464Genetic loci influencing kidney function and chronic kidney diseaseChambers, John C.; Zhang, Weihua; Lord, Graham M.; van der Harst, Pim; Lawlor, Debbie A.; Sehmi, Joban S.; Gale, Daniel P.; Wass, Mark N.; Ahmadi, Kourosh R.; Bakker, Stephan J. L.; Beckmann, Jacqui; Bilo, Henk J. G.; Bochud, Murielle; Brown, Morris J.; Caulfield, Mark J.; Connell, John M. C.; Cook, H. Terence; Cotlarciuc, Ioana; Smith, George Davey; de Silva, Ranil; Deng, Guohong; Devuyst, Olivier; Dikkeschei, Lambert D.; Dimkovic, Nada; Dockrell, Mark; Dominiczak, Anna; Ebrahim, Shah; Eggermann, Thomas; Farrall, Martin; Ferrucci, Luigi; Floege, Jurgen; Forouhi, Nita G.; Gansevoort, Ron T.; Han, Xijin; Hedblad, Bo; van der Heide, Jaap J. Homan; Hepkema, Bouke G.; Hernandez-Fuentes, Maria; Hypponen, Elina; Johnson, Toby; de Jong, Paul E.; Kleefstra, Nanne; Lagou, Vasiliki; Lapsley, Marta; Li, Yun; Loos, Ruth J. F.; Luan, Jian'an; Luttropp, Karin; Marechal, Celine; Melander, Olle; Munroe, Patricia B.; Nordfors, Louise; Parsa, Afshin; Peltonen, Leena; Penninx, Brenda W.; Perucha, Esperanza; Pouta, Anneli; Prokopenko, Inga; Roderick, Paul J.; Ruokonen, Aimo; Samani, Nilesh J.; Sanna, Serena; Schalling, Martin; Schlessinger, David; Schlieper, Georg; Seelen, Marc A. J.; Shuldiner, Alan R.; Sjoegren, Marketa; Smit, Johannes H.; Snieder, Harold; Soranzo, Nicole; Spector, Timothy D.; Stenvinkel, Peter; Sternberg, Michael J. E.; Swaminathan, Ramasamyiyer; Tanaka, Toshiko; Ubink-Veltmaat, Lielith J.; Uda, Manuela; Vollenweider, Peter; Wallace, Chris; Waterworth, Dawn; Zerres, Klaus; Waeber, Gerard; Wareham, Nicholas J.; Maxwell, Patrick H.; McCarthy, Mark I.; Jarvelin, Marjo-Riitta; Mooser, Vincent; Abecasis, Goncalo R.; Lightstone, Liz; Scott, James; Navis, Gerjan; Elliott, Paul; Kooner, Jaspal S.Nature Genetics (2010), 42 (5), 373-375CODEN: NGENEC; ISSN:1061-4036. (Nature Publishing Group)Using genome-wide assocn., we identify common variants at 2p12-p13, 6q26, 17q23 and 19q13 assocd. with serum creatinine, a marker of kidney function (P = 10-10 to 10-15). Of these, rs10206899 (near NAT8, 2p12-p13) and rs4805834 (near SLC7A9, 19q13) were also assocd. with chronic kidney disease (P = 5.0 × 10-5 and P = 3.6 × 10-4, resp.). Our findings provide insight into metabolic, solute and drug-transport pathways underlying susceptibility to chronic kidney disease.
- 46Simmons, M. L.; Frondoza, C. G.; Coyle, J. T. Immunocytochemical localization of N-acetyl-aspartate with monoclonal antibodies. Neuroscience 1991, 45, 37– 45, DOI: 10.1016/0306-4522(91)90101-S47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3MXmslKgtb8%253D&md5=361a6c94e99c27148b37e32321f739f3Immunocytochemical localization of N-acetyl-aspartate with monoclonal antibodiesSimmons, M. L.; Frondoza, C. G.; Coyle, J. T.Neuroscience (Oxford, United Kingdom) (1991), 45 (1), 37-45CODEN: NRSCDN; ISSN:0306-4522.N-Acetyl-aspartate is found in high concns. in all areas of the brain, but is undetectable in non-neuronal tissue. In order to characterize the cellular localization of N-acetyl-aspartate in brain, highly specific monoclonal antibodies against N-acetyl-aspartate were produced by fusing spleen lymphocytes obtained from mice immunized with N-acetyl-aspartate conjugated to thyroglobulin by carbodiimide with P3/x63-Ag8.653 mouse myeloma cells. Clones were selected which secrete IgG2a(k) antibodies highly specific for conjugated N-acetyl-aspartate. Only 3-6% cross-reactivity with conjugated N-acetyl-aspartate-glutamate was obsd. at high antibody concns., whereas no cross-reactivity (<1%) was obsd. with conjugated N-acetyl-glutamate or aspartate. Preincubation of the antibodies with 0.5 mg/mL conjugated N-acetyl-aspartate blocked immunoreactivity more than 90%, while preincubation with conjugated N-acetyl-aspartate-glutamate and free N-acetyl-aspartate had no effect. Immunocytochem. staining has shown that N-acetyl-aspartate-like immunoreactivity is localized in neurons, which are widely distributed throughout the brain. The immunoreactive neurons exhibited intense staining of the perikarya, proximal dendrites and axons. No consistent pattern of distribution of immunoreactivity was obsd. with regard to primary neurotransmitter characteristics of stained neurons although neurons with long projections or extensive arbors, such as pyramidal cells in cortex, locus ceruleus, motor neurons and Purkinje cells, stained much more intensively than local circuit neurons.
- 47Masaharu, M.; Hideo, M.; Mutsuhiko, M.; Yasuo, K. N-acetyl-l-aspartic acid, N-acetyl-α-l-aspartyl-l-glutamic acid and β-citryl-l-glutamic acid in human urine. Clin. Chim. Acta 1982, 120, 119– 126, DOI: 10.1016/0009-8981(82)90082-1There is no corresponding record for this reference.
- 48Barker, P. B. N-acetyl aspartate─a neuronal marker?. Ann. Neurol. 2001, 49, 423– 424, DOI: 10.1002/ana.9049https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD3M3hvVyltg%253D%253D&md5=b94bddfa307a732255479dd70f6d7ae4N-acetyl aspartate--a neuronal marker?Barker P BAnnals of neurology (2001), 49 (4), 423-4 ISSN:0364-5134.There is no expanded citation for this reference.
- 49Jung, R. E.; Brooks, W. M.; Yeo, R. A.; Chiulli, S. J.; Weers, D. C.; Sibbitt, W. L., Jr. Biochemical markers of intelligence: a proton MR spectroscopy study of normal human brain. Proc. R. Soc. London, Ser. B 1999, 266, 1375– 1379, DOI: 10.1098/rspb.1999.079050https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaK1MznsVeiug%253D%253D&md5=28668501ddca01938cdcecec067146adBiochemical markers of intelligence: a proton MR spectroscopy study of normal human brainJung R E; Brooks W M; Yeo R A; Chiulli S J; Weers D C; Sibbitt W L JrProceedings. Biological sciences / The Royal Society (1999), 266 (1426), 1375-9 ISSN:0962-8452.Proton magnetic resonance spectroscopy (1H-MRS) offers a unique non-invasive approach to measurement of N-acetylaspartate (NAA) and choline (Cho), putative markers of neuronal and glial integrity. Previous studies revealed that these neurochemicals predict cognitive impairment in diseased subjects, although little is known about their relationship to cognitive functioning in healthy people. We measured the concentrations of NAA and Cho in the left occipitoparietal white matter of 26 healthy adults and compared them with intellectual performance assessed by the Wechsler Adult Intelligence Scale-3. We found that NAA (b = 0.6, p < 0.01) and Cho (b = -0.42, p < 0.01) were independently associated with the Full-Scale Intelligence Quotient. Together, these metabolites accounted for a large proportion of the variance in intelligence (r2 = 0.45). Possible mechanisms underlying these correlations, such as mitochondrial function and myelin turnover, are discussed. 1H-MRS is a sensitive new tool to assess the neuronal underpinnings of cognitive function non-invasively.
- 50Patel, T.; Blyth, J. C.; Griffiths, G.; Kelly, D.; Talcott, J. B. Moderate relationships between NAA and cognitive ability in healthy adults: implications for cognitive spectroscopy. Front. Hum. Neurosci. 2014, 8, 39, DOI: 10.3389/fnhum.2014.0003951https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhs1CrtL3O&md5=d73d396cdd39f3cfc6dda110ddd55e63Moderate relationships between NAA and cognitive ability in healthy adults: implications for cognitive spectroscopyPatel, Tulpesh; Blyth, Jacqueline C.; Griffiths, Gareth; Kelly, Deirdre; Talcott, Joel B.Frontiers in Human Neuroscience (2014), 8 (), 39/1-39/10, 10CODEN: FHNRAI; ISSN:1662-5161. (Frontiers Media S.A.)Background: Proton Magnetic Resonance Spectroscopy (1H-MRS)is a non-invasive imaging technique that enables quantification of neurochem. in vivo and thereby facilitates investigation of the biochem. underpinnings of human cognitive variability. Studies in the field of cognitive spectroscopy have commonly focused on relationships between measures of N-acetyl aspartate (NAA), a surrogate marker of neuronal health and function, and broad measures of cognitive performance, such as IQ. Methodol./Principal Findings: In this study, we used 1H-MRS to interrogate single-voxels in occipitoparietal and frontal cortex, in parallel with assessments of psychometric intelligence, in a sample of 40 healthy adult participants. We found correlations between NAA and IQ that were within the range reported in previous studies. However, the magnitude of these effects was significantly modulated by the stringency of data screening and the extent to which outlying values contributed to statistical analyses. Conclusions/Significance:1H-MRS offers a sensitive tool for assessing neurochem. non-invasively, yet the relationships between brain metabolites and broad aspects of human behavior such as IQ are subtle. We highlight the need to develop an increasingly rigorous anal. and interpretive framework for collecting and reporting data obtained from cognitive spectroscopy studies of this kind.
- 51Davies, G.; Lam, M.; Harris, S. E.; Trampush, J. W.; Luciano, M.; Hill, W. D. Study of 300,486 individuals identifies 148 independent genetic loci influencing general cognitive function. Nat. Commun. 2018, 9, 2098, DOI: 10.1038/s41467-018-04362-x52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1MbhsFCksg%253D%253D&md5=524b8e4875a84c1948df7aa439e49e4fStudy of 300,486 individuals identifies 148 independent genetic loci influencing general cognitive functionDavies Gail; Harris Sarah E; Luciano Michelle; Hill W David; Hagenaars Saskia P; Ritchie Stuart J; Marioni Riccardo E; Fawns-Ritchie Chloe; Liewald David C M; Okely Judith A; Corley Janie; Cox Simon R; Porteous David J; Starr John M; McIntosh Andrew M; Gale Catharine R; Deary Ian J; Lam Max; DeRosse Pamela; Harris Sarah E; Marioni Riccardo E; Porteous David J; Trampush Joey W; Trampush Joey W; Hagenaars Saskia P; Ahola-Olli Ari V; Rovio Suvi P; Raitakari Olli T; Ahola-Olli Ari V; Barnes Catriona L K; Campbell Harry; Joshi Peter K; Rudan Igor; Wilson James F; Bertram Lars; Liu Tian; Bis Joshua C; Psaty Bruce M; Burdick Katherine E; Roussos Panos; Burdick Katherine E; Burdick Katherine E; Christoforou Andrea; Djurovic Srdjan; Giddaluru Sudheer; Melle Ingrid; Le Hellard Stephanie; Steen Vidar M; Christoforou Andrea; Giddaluru Sudheer; Le Hellard Stephanie; Steen Vidar M; DeRosse Pamela; Yu Jin; Malhotra Anil; Lencz Todd; Djurovic Srdjan; Espeseth Thomas; Melle Ingrid; Andreassen Ole A; Sundet Kjetil; Espeseth Thomas; Sundet Kjetil; Reinvang Ivar; Giakoumaki Stella; Gustavson Daniel E; Dale Anders M; Kremen William S; Gustavson Daniel E; Kremen William S; Hayward Caroline; Vitart Veronique; Wilson James F; Hayward Caroline; Murray Alison D; Porteous David J; Hofer Edith; Petrovic Katja; Koini Marisa; Schmidt Reinhold; Hofer Edith; Ikram M Arfan; Terzikhan Natalie; van der Lee Sven J; Amin Najaf; Uitterlinden Andre; Wen K Hoyan; Adams Hieab H H; Ikram M Kamran; Van Duijn Cornelia M; Ikram M Arfan; Ikram M Arfan; Ikram M Kamran; Karlsson Robert; Karlsson Ida; Hagg Sara; Pedersen Nancy L; Knowles Emma; Glahn David C; Lahti Jari; Raikkonen Katri; Lahti Jari; Leber Markus; Ramirez Alfredo; Li Shuo; Yang Qiong; Mather Karen A; Thalamuthu Anbupalam; Assareh Amelia A; Brodaty Henry; Kochan Nicole A; Lee Teresa; Reppermund Simone; Sachdev Perminder S; Trollor Julian N; Morris Derek; Donohoe Gary; Oldmeadow Christopher; Palviainen Teemu; Loukola Anu; Kaprio Jaakko; Widen Elisabeth; Vuoksimaa Eero; Palotie Aarno; Payton Antony; Pazoki Raha; Dehghan Abbas; Evangelou Evangelos; Gao He; Tzoulaki Ioanna; Elliott Paul; Reynolds Chandra A; Sargurupremraj Muralidharan; Soumare Aicha; Tzourio Christophe; Debette Stephanie; Scholz Markus; Scholz Markus; Luck Tobias; Smith Jennifer A; Kardia Sharon L R; Zhao Wei; Smith Jennifer A; Ware Erin B; Faul Jessica D; Weir David R; Smith Albert V; Gudnason Vilmundur; Smith Albert V; Gudnason Vilmundur; Terzikhan Natalie; Trompet Stella; Noordam Raymond; Windham B Gwen; Mosley Thomas H Jr; Wright Margaret J; Hansell Narelle K; Wright Margaret J; Yang Jingyun; Bennett David A; Boyle Patricia A; Yang Jingyun; Bennett David A; Ames David; Ames David; Amouyel Philippe; Andreassen Ole A; Armstrong Nicola J; Attia John R; Holliday Elizabeth G; Scott Rodney J; Attix Deborah; Chiba-Falek Ornit; Attix Deborah; Avramopoulos Dimitrios; Avramopoulos Dimitrios; Arking Dan E; Bohmer Anne C; Ramirez Alfredo; Bohmer Anne C; Boyle Patricia A; Brodaty Henry; Cannon Tyrone D; Cirulli Elizabeth T; Congdon Eliza; Freimer Nelson A; London Edythe; Bilder Robert M; Conley Emily Drabant; Dale Anders M; Dale Anders M; Dale Anders M; Dehghan Abbas; Tzoulaki Ioanna; Elliott Paul; Dick Danielle; Dickinson Dwight; Eriksson Johan G; Evangelou Evangelos; Kaprio Jaakko; Eriksson Johan G; Eriksson Johan G; Eriksson Johan G; Ford Ian; Giegling Ina; Hartmann Annette M; Konte Bettina; Dan Rujescu; Gillespie Nathan A; Gordon Scott D; Martin Nicholas G; Montgomery Grant W; Gottesman Rebecca F; Gottesman Rebecca F; Griswold Michael E; Simino Jeannette; Harris Tamara B; Launer Lenore; Hatzimanolis Alex; Smyrnis Nikolaos; Stefanis Nikos C; Hatzimanolis Alex; Smyrnis Nikolaos; Stefanis Nikos C; Hatzimanolis Alex; Stefanis Nikos C; Heiss Gerardo; Kahonen Mika; Kahonen Mika; Kahonen Mika; Kleineidam Luca; Kleineidam Luca; Wagner Holger; Ramirez Alfredo; Kleineidam Luca; Wagner Michael; Kleineidam Luca; Wagner Michael; Knopman David S; Kochan Nicole A; Lee Teresa; Sachdev Perminder S; Kwok John B; Kwok John B; Lehtimaki Terho; Lyytikainen Leo-Pekka; Lehtimaki Terho; Lyytikainen Leo-Pekka; Li Shu-Chen; Liu Tian; Papenberg Goran; Lindenberger Ulman; Li Shu-Chen; Lill Christina M; Longstreth Will T Jr; Longstreth Will T Jr; Fitzpatrick Annette L; Lopez Oscar L; Luck Tobias; Riedel-Heller Steffi G; Lundervold Astri J; Lundervold Astri J; Lundquist Anders; Nyberg Lars; Lundquist Anders; Montgomery Grant W; Murray Alison D; Need Anna C; Nyberg Lars; Nyberg Lars; Ollier William; Papenberg Goran; Pattie Alison; Taylor Adele M; Polasek Ozren; Polasek Ozren; Poldrack Russell A; Psaty Bruce M; Psaty Bruce M; Reppermund Simone; Trollor Julian N; Rose Richard J; Rotter Jerome I; Taylor Kent D; Rotter Jerome I; Taylor Kent D; Roussos Panos; Roussos Panos; Saba Yasaman; Schmidt Helena; Sabb Fred W; Seshadri Sudha; Satizabal Claudia L; Seshadri Sudha; Satizabal Claudia L; Schmid Matthias; Weinhold Leonie; Scult Matthew A; Hariri Ahmad R; Slagboom P Eline; Stott David J; Straub Richard E; Weinberger Daniel R; Tzoulaki Ioanna; Tzourio Christophe; Uitterlinden Andre; Voineskos Aristotle N; Kaprio Jaakko; Vuoksimaa Eero; Adams Hieab H H; Bitsios Panos; Boerwinkle Eric; Bressler Jan; Boerwinkle Eric; Corvin Aiden; Gill Michael; De Jager Philip L; De Jager Philip L; Debette Stephanie; Fitzpatrick Annette L; Jukema J Wouter; Keller Matthew C; Palotie Aarno; Palotie Aarno; Pendleton Neil; Raitakari Olli T; Schofield Peter W; Schofield Peter R; Schofield Peter R; Starr John M; Turner Steven T; Villringer Arno; Villringer Arno; Malhotra Anil; Malhotra Anil; McIntosh Andrew M; Lencz Todd; Gale Catharine R; Seshadri SudhaNature communications (2018), 9 (1), 2098 ISSN:.General cognitive function is a prominent and relatively stable human trait that is associated with many important life outcomes. We combine cognitive and genetic data from the CHARGE and COGENT consortia, and UK Biobank (total N = 300,486; age 16-102) and find 148 genome-wide significant independent loci (P < 5 × 10(-8)) associated with general cognitive function. Within the novel genetic loci are variants associated with neurodegenerative and neurodevelopmental disorders, physical and psychiatric illnesses, and brain structure. Gene-based analyses find 709 genes associated with general cognitive function. Expression levels across the cortex are associated with general cognitive function. Using polygenic scores, up to 4.3% of variance in general cognitive function is predicted in independent samples. We detect significant genetic overlap between general cognitive function, reaction time, and many health variables including eyesight, hypertension, and longevity. In conclusion we identify novel genetic loci and pathways contributing to the heritability of general cognitive function.
- 52Lee, J. J.; Wedow, R.; Okbay, A.; Kong, E.; Maghzian, O.; Zacher, M. Gene discovery and polygenic prediction from a genome-wide association study of educational attainment in 1.1 million individuals. Nat. Genet. 2018, 50, 1112– 1121, DOI: 10.1038/s41588-018-0147-353https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtlOis7zI&md5=5259e727f3e6ec901d14a7f088dafedeGene discovery and polygenic prediction from a genome-wide association study of educational attainment in 1.1 million individualsLee, James J.; Wedow, Robbee; Okbay, Aysu; Kong, Edward; Maghzian, Omeed; Zacher, Meghan; Nguyen-Viet, Tuan Anh; Bowers, Peter; Sidorenko, Julia; Karlsson Linner, Richard; Fontana, Mark Alan; Kundu, Tushar; Lee, Chanwook; Li, Hui; Li, Ruoxi; Royer, Rebecca; Timshel, Pascal N.; Walters, Raymond K.; Willoughby, Emily A.; Yengo, Loic; 23andMe Research Team; Cogent; Social Science Genetic Association Consortium; Alver, Maris; Bao, Yanchun; Clark, David W.; Day, Felix R.; Furlotte, Nicholas A.; Joshi, Peter K.; Kemper, Kathryn E.; Kleinman, Aaron; Langenberg, Claudia; Magi, Reedik; Trampush, Joey W.; Verma, Shefali Setia; Wu, Yang; Lam, Max; Zhao, Jing Hua; Zheng, Zhili; Boardman, Jason D.; Campbell, Harry; Freese, Jeremy; Harris, Kathleen Mullan; Hayward, Caroline; Herd, Pamela; Kumari, Meena; Lencz, Todd; Luan, Jian'an; Malhotra, Anil K.; Metspalu, Andres; Milani, Lili; Ong, Ken K.; Perry, John R. B.; Porteous, David J.; Ritchie, Marylyn D.; Smart, Melissa C.; Smith, Blair H.; Tung, Joyce Y.; Wareham, Nicholas J.; Wilson, James F.; Beauchamp, Jonathan P.; Conley, Dalton C.; Esko, Tonu; Lehrer, Steven F.; Magnusson, Patrik K. E.; Oskarsson, Sven; Pers, Tune H.; Robinson, Matthew R.; Thom, Kevin; Watson, Chelsea; Chabris, Christopher F.; Meyer, Michelle N.; Laibson, David I.; Yang, Jian; Johannesson, Magnus; Koellinger, Philipp D.; Turley, Patrick; Visscher, Peter M.; Benjamin, Daniel J.; Cesarini, DavidNature Genetics (2018), 50 (8), 1112-1121CODEN: NGENEC; ISSN:1061-4036. (Nature Research)Here we conducted a large-scale genetic assocn. anal. of educational attainment in a sample of approx. 1.1 million individuals and identify 1,271 independent genome-wide-significant SNPs. For the SNPs taken together, we found evidence of heterogeneous effects across environments. The SNPs implicate genes involved in brain-development processes and neuron-to-neuron communication. In a sep. anal. of the X chromosome, we identify 10 independent genome-wide-significant SNPs and est. a SNP heritability of around 0.3% in both men and women, consistent with partial dosage compensation. A joint (multi-phenotype) anal. of educational attainment and three related cognitive phenotypes generates polygenic scores that explain 11-13% of the variance in educational attainment and 7-10% of the variance in cognitive performance. This prediction accuracy substantially increases the utility of polygenic scores as tools in research.
- 53Hearn, T. ALMS1 and Alström syndrome: a recessive form of metabolic, neurosensory and cardiac deficits. J. Mol Med. 2019, 97, 1– 7, DOI: 10.1007/s00109-018-1714-x54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitlSrtbzL&md5=ef0b17a82be038f1f685abc63d0a87eaALMS1 and Alstrom syndrome: a recessive form of metabolic, neurosensory and cardiac deficitsHearn, TomJournal of Molecular Medicine (Heidelberg, Germany) (2019), 97 (1), 1-17CODEN: JMLME8; ISSN:0946-2716. (Springer)A review. Alstrom syndrome (AS) is characterised by metabolic deficits, retinal dystrophy, sensorineural hearing loss, dilated cardiomyopathy and multi-organ fibrosis. Elucidating the function of the mutated gene, ALMS1, is crit. for the development of specific treatments and may uncover pathways relevant to a range of other disorders including common forms of obesity and type 2 diabetes. Interest in ALMS1 is heightened by the recent discovery of its involvement in neonatal cardiomyocyte cell cycle arrest, a process with potential relevance to regenerative medicine. ALMS1 encodes a ∼ 0.5 megadalton protein that localises to the base of centrioles. Some studies have suggested a role for this protein in maintaining centriole-nucleated sensory organelles termed primary cilia, and AS is now considered to belong to the growing class of human genetic disorders linked to ciliary dysfunction (ciliopathies). However, mechanistic details are lacking, and recent studies have implicated ALMS1 in several processes including endosomal trafficking, actin organization, maintenance of centrosome cohesion and transcription. In line with a more complex picture, multiple isoforms of the protein likely exist and non-centrosomal sites of localisation have been reported. This review outlines the evidence for both ciliary and extra-ciliary functions of ALMS1.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jproteome.1c00585.
Figure S1: Principal component analysis of baseline and follow-up metabolomics data; Figure S2: Overview of RNA-Seq read count and quantifiable genes in 555 individuals; Figure S3: Scatter plots of removed associations; Figure S4: Scatter plots of 20 study-wide significant metabolome feature - gene expression associations; Figure S5: Metabomatching figure showing the pseudospectrum derived from ALMS1 gene expression - metabolome features associations; Figure S6: Metabomatching figure showing the pseudospectrum derived from ALMS1P gene expression - metabolome features associations; Figure S7: NMR profiles of 3 different spike-in experiments; Figure S8: Metabomatching figure showing the pseudospectrum derived from APIP gene expression - metabolome feature associations; Figure S9: Metabomatching figure showing the pseudospectrum derived from SNW1 gene expression - metabolome feature associations; Table S1: Validation of all associations discovered in CoLaus baseline; Table S2: MR results of testing causal effect of ALMS1 gene expression levels on N-acetylated compounds, using R-squared threshold of 0.05; Table S3: MR results of testing causal effect of HPS1 gene expression levels on trimethylamine, using R-squared threshold of 0.05 (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.