Identification and functional characterization of G6PC2 coding variants influencing glycemic traits define an effector transcript at the G6PC2-ABCB11 locus

Anubha Mahajan, Xueling Sim, Hui Jin Ng, Alisa Manning, Manuel A Rivas, Heather M Highland, Adam E Locke, Niels Grarup, Hae Kyung Im, Pablo Cingolani, Jason Flannick, Pierre Fontanillas, Christian Fuchsberger, Kyle J Gaulton, Tanya M Teslovich, N William Rayner, Neil R Robertson, Nicola L Beer, Jana K Rundle, Jette Bork-Jensen, Claes Ladenvall, Christine Blancher, David Buck, Gemma Buck, Noël P Burtt, Stacey Gabriel, Anette P Gjesing, Christopher J Groves, Mette Hollensted, Jeroen R Huyghe, Anne U Jackson, Goo Jun, Johanne Marie Justesen, Massimo Mangino, Jacquelyn Murphy, Matt Neville, Robert Onofrio, Kerrin S Small, Heather M Stringham, Ann-Christine Syvänen, Joseph Trakalo, Goncalo Abecasis, Graeme I Bell, John Blangero, Nancy J Cox, Ravindranath Duggirala, Craig L Hanis, Mark Seielstad, James G Wilson, Cramer Christensen, Ivan Brandslund, Rainer Rauramaa, Gabriela L Surdulescu, Alex S F Doney, Lars Lannfelt, Allan Linneberg, Bo Isomaa, Tiinamaija Tuomi, Marit E Jørgensen, Torben Jørgensen, Johanna Kuusisto, Matti Uusitupa, Veikko Salomaa, Timothy D Spector, Andrew D Morris, Colin N A Palmer, Francis S Collins, Karen L Mohlke, Richard N Bergman, Erik Ingelsson, Lars Lind, Jaakko Tuomilehto, Torben Hansen, Richard M Watanabe, Inga Prokopenko, Josee Dupuis, Fredrik Karpe, Leif Groop, Markku Laakso, Oluf Pedersen, Jose C Florez, Andrew P Morris, David Altshuler, James B Meigs, Michael Boehnke, Mark I McCarthy, Cecilia M Lindgren, Anna L Gloyn, T2D-GENES consortium and GoT2D consortium, Anubha Mahajan, Xueling Sim, Hui Jin Ng, Alisa Manning, Manuel A Rivas, Heather M Highland, Adam E Locke, Niels Grarup, Hae Kyung Im, Pablo Cingolani, Jason Flannick, Pierre Fontanillas, Christian Fuchsberger, Kyle J Gaulton, Tanya M Teslovich, N William Rayner, Neil R Robertson, Nicola L Beer, Jana K Rundle, Jette Bork-Jensen, Claes Ladenvall, Christine Blancher, David Buck, Gemma Buck, Noël P Burtt, Stacey Gabriel, Anette P Gjesing, Christopher J Groves, Mette Hollensted, Jeroen R Huyghe, Anne U Jackson, Goo Jun, Johanne Marie Justesen, Massimo Mangino, Jacquelyn Murphy, Matt Neville, Robert Onofrio, Kerrin S Small, Heather M Stringham, Ann-Christine Syvänen, Joseph Trakalo, Goncalo Abecasis, Graeme I Bell, John Blangero, Nancy J Cox, Ravindranath Duggirala, Craig L Hanis, Mark Seielstad, James G Wilson, Cramer Christensen, Ivan Brandslund, Rainer Rauramaa, Gabriela L Surdulescu, Alex S F Doney, Lars Lannfelt, Allan Linneberg, Bo Isomaa, Tiinamaija Tuomi, Marit E Jørgensen, Torben Jørgensen, Johanna Kuusisto, Matti Uusitupa, Veikko Salomaa, Timothy D Spector, Andrew D Morris, Colin N A Palmer, Francis S Collins, Karen L Mohlke, Richard N Bergman, Erik Ingelsson, Lars Lind, Jaakko Tuomilehto, Torben Hansen, Richard M Watanabe, Inga Prokopenko, Josee Dupuis, Fredrik Karpe, Leif Groop, Markku Laakso, Oluf Pedersen, Jose C Florez, Andrew P Morris, David Altshuler, James B Meigs, Michael Boehnke, Mark I McCarthy, Cecilia M Lindgren, Anna L Gloyn, T2D-GENES consortium and GoT2D consortium

Abstract

Genome wide association studies (GWAS) for fasting glucose (FG) and insulin (FI) have identified common variant signals which explain 4.8% and 1.2% of trait variance, respectively. It is hypothesized that low-frequency and rare variants could contribute substantially to unexplained genetic variance. To test this, we analyzed exome-array data from up to 33,231 non-diabetic individuals of European ancestry. We found exome-wide significant (P<5×10-7) evidence for two loci not previously highlighted by common variant GWAS: GLP1R (p.Ala316Thr, minor allele frequency (MAF)=1.5%) influencing FG levels, and URB2 (p.Glu594Val, MAF = 0.1%) influencing FI levels. Coding variant associations can highlight potential effector genes at (non-coding) GWAS signals. At the G6PC2/ABCB11 locus, we identified multiple coding variants in G6PC2 (p.Val219Leu, p.His177Tyr, and p.Tyr207Ser) influencing FG levels, conditionally independent of each other and the non-coding GWAS signal. In vitro assays demonstrate that these associated coding alleles result in reduced protein abundance via proteasomal degradation, establishing G6PC2 as an effector gene at this locus. Reconciliation of single-variant associations and functional effects was only possible when haplotype phase was considered. In contrast to earlier reports suggesting that, paradoxically, glucose-raising alleles at this locus are protective against type 2 diabetes (T2D), the p.Val219Leu G6PC2 variant displayed a modest but directionally consistent association with T2D risk. Coding variant associations for glycemic traits in GWAS signals highlight PCSK1, RREB1, and ZHX3 as likely effector transcripts. These coding variant association signals do not have a major impact on the trait variance explained, but they do provide valuable biological insights.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Figure 1. Haplotypes of the lead non-coding…
Figure 1. Haplotypes of the lead non-coding GWAS SNP rs560887 and the three coding variants.
rs138726309 (p.His177Tyr), rs2232323 (p.Tyr207Ser), and rs492594 (p.Val219Leu), obtained from 4,442 unrelated individuals from the Oxford Biobank. (A) Percentage minor allele frequency (MAF) and effect size estimates (β^) of the four variants reported for the minor allele in mmol/L of FG after adjustment for age, sex, and BMI. (B) Haplotypes of the four associated variants in G6PC2 revealed that the glucose-lowering Leu219 allele was carried exclusively in cis with the glucose-raising allele at the GWAS SNP. Wild-type, glucose-raising alleles are circled in blue and the mutant, glucose-lowering alleles are circled in red. Diameter of the circle is proportional to the effect size estimates. Haplotype association was performed with FG derived residuals (after adjustment for age, sex, and BMI) using the most frequent haplotype as baseline.
Figure 2. Functional characterization of wild type…
Figure 2. Functional characterization of wild type and variant G6PC2 proteins.
(A) Expression levels in HEK293 and (B) INS-1E cells were determined by western blot and densitometry analysis. The multiple bands on the western blot are likely to represent glycosylated G6PC2 protein products. Data are presented as mean ± standard error of the mean for at least three independent experiments. Significant differences are indicated as ** P<0.01; *** P<0.001; **** P<0.0001. EV, empty vector; WT, wild type. (C) Expression levels in HEK293 and INS-1E cells in the presence of proteasomal inhibitor MG-132 or lysosomal inhibitor chloroquine were determined by western blot. (D) Cellular localization in HEK293 cells was assessed by immunofluorescence microscopy. Cells were double immunostained for FLAG tag (green) and calnexin (red), and merged images with a DNA stain (blue) are shown. Images were taken with laser settings that were optimized separately for each sample. Scale bar, 10µm.

References

    1. Prokopenko I, Langenberg C, Florez JC, Saxena R, Soranzo N, et al. (2009) Variants in MTNR1B influence fasting glucose levels. Nat Genet 41: 77–81. 10.1038/ng.290
    1. Dupuis J, Langenberg C, Prokopenko I, Saxena R, Soranzo N, et al. (2010) New genetic loci implicated in fasting glucose homeostasis and their impact on type 2 diabetes risk. Nat Genet 42: 105–116. 10.1038/ng.520
    1. Manning AK, Hivert M-F, Scott RA, Grimsby JL, Bouatia-Naji N, et al. (2012) A genome-wide approach accounting for body mass index identifies genetic variants influencing fasting glycemic traits and insulin resistance. Nat Genet 44: 659–669. 10.1038/ng.2274
    1. Scott RA, Lagou V, Welch RP, Wheeler E, Montasser ME, et al. (2012) Large-scale association analyses identify new loci influencing glycemic traits and provide insight into the underlying biological pathways. Nat Genet 44: 991–991005. 10.1038/ng.2385
    1. Kang HM, Sul JH, Service SK, Zaitlen NA, Kong S-Y, et al. (2010) Variance component model to account for sample structure in genome-wide association studies. Nat Genet 42: 348–354. 10.1038/ng.548
    1. Liu DJ, Peloso GM, Zhan X, Holmen OL, Zawistowski M, et al. (2014) Meta-analysis of gene-level tests for rare variant association. Nat Genet 46: 200–204. 10.1038/ng.2852
    1. Feng S, Liu D, Zhan X, Wing MK, Abecasis GR (2014) RAREMETAL: fast and powerful meta-analysis for rare variants. Bioinformatics. 10.1093/bioinformatics/btu367
    1. Wu MC, Lee S, Cai T, Li Y, Boehnke M, et al. (2011) Rare-variant association testing for sequencing data with the sequence kernel association test. Am J Hum Genet 89: 82–93. 10.1016/j.ajhg.2011.05.029
    1. Magi R, Kumar A, Morris AP (2011) Assessing the impact of missing genotype data in rare variant association analysis. BMC Proc 5 Suppl 9 10.1186/1753-6561-5-S9-S107
    1. Purcell SM, Moran JL, Fromer M, Ruderfer D, Solovieff N, et al. (2014) A polygenic burden of rare disruptive mutations in schizophrenia. Nature 506: 185–190. 10.1038/nature12975
    1. Vaxillaire M, Cavalcanti-Proenca C, Dechaume A, Tichet J, Marre M, et al. (2008) The common P446L polymorphism in GCKR inversely modulates fasting glucose and triglyceride levels and reduces type 2 diabetes risk in the DESIR prospective general French population. Diabetes 57: 2253–2257. 10.2337/db07-1807
    1. Beer NL, Tribble ND, McCulloch LJ, Roos C, Johnson PRV, et al. (2009) The P446L variant in GCKR associated with fasting plasma glucose and triglyceride levels exerts its effect through increased glucokinase activity in liver. Hum Mol Genet 18: 4081–4088. 10.1093/hmg/ddp357
    1. Rees MG, Wincovitch S, Schultz J, Waterstradt R, Beer NL, et al. (2012) Cellular characterisation of the GCKR P446L variant associated with type 2 diabetes risk. Diabetologia 55: 114–122. 10.1007/s00125-011-2348-5
    1. Drucker DJ, Nauck MA (2006) The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet 368: 1696–1705. 10.1016/S0140-6736(06)69705-5
    1. Service SK, Teslovich TM, Fuchsberger C, Ramensky V, Yajnik P, et al. (2014) Re-sequencing expands our understanding of the phenotypic impact of variants at GWAS loci. PLoS genetics 10: e1004147 10.1371/journal.pgen.1004147
    1. Khajavi M, Inoue K, Lupski JR. (2006) Nonsense-mediated mRNA decay modulates clinical outcome of genetic disease. Eur J Hum Genet 14: 1074–81. 10.1038/sj.ejhg.5201649
    1. Chen W-M, Erdos MR, Jackson AU, Saxena R, Sanna S, et al. (2008) Variations in the G6PC2/ABCB11 genomic region are associated with fasting glucose levels. J Clin Invest 118: 2620–2628. 10.1172/JCI34566
    1. Bouatia-Naji N, Rocheleau G, Van Lommel L, Lemaire K, Schuit F, et al. (2008) A polymorphism within the G6PC2 gene is associated with fasting plasma glucose levels. Science 320: 1085–1088. 10.1126/science.1156849
    1. Baerenwald DA, Bonnefond A, Bouatia-Naji N, Flemming BP, Umunakwe OC, et al. (2013) Multiple functional polymorphisms in the G6PC2 gene contribute to the association with higher fasting plasma glucose levels. Diabetologia 56: 1306–1316. 10.1007/s00125-013-2875-3
    1. Pound LD, Oeser JK, O’Brien TP, Wang Y, Faulman CJ, et al. (2013) G6PC2: a negative regulator of basal glucose-stimulated insulin secretion. Diabetes 62: 1547–1556. 10.2337/db12-1067
    1. Morris AP, Voight BF, Teslovich TM, Ferreira T, Segre AV, et al. (2012) Large-scale association analysis provides insights into the genetic architecture and pathophysiology of type 2 diabetes. Nat Genet 44: 981–990. 10.1038/ng.2383
    1. Wang H, Liu L, Zhao J, Cui G, Chen C, et al. (2013) Large scale meta-analyses of fasting plasma glucose raising variants in GCK, GCKR, MTNR1B and G6PC2 and their impacts on type 2 diabetes mellitus risk. PLoS One 8: e67665 10.1371/journal.pone.0067665
    1. Hu C, Zhang R, Wang C, Ma X, Fang Q, et al. (2009) A genetic variant of G6PC2 is associated with type 2 diabetes and fasting plasma glucose level in the Chinese population. Diabetologia 52: 451–456. 10.1007/s00125-008-1241-3
    1. Pickett LA, Yourshaw M, Albornoz V, Chen Z, Solorzano-Vargas RS, et al. (2013) Functional consequences of a novel variant of PCSK1. PLoS One 8: e55065 10.1371/journal.pone.0055065
    1. Liu G, Clement LC, Kanwar YS, Avila-Casado C, Chugh SS (2006) ZHX proteins regulate podocyte gene expression during the development of nephrotic syndrome. J Biol Chem 281: 39681–39692. 10.1074/jbc.M606664200
    1. Gargalovic PS, Erbilgin A, Kohannim O, Pagnon J, Wang X, et al. (2010) Quantitative trait locus mapping and identification of Zhx2 as a novel regulator of plasma lipid metabolism. Circ Cardiovasc Genet 3: 60–67. 10.1161/CIRCGENETICS.109.902320
    1. Mahajan A (2014) Genome-wide trans-ancestry meta-analysis provides insight into the genetic architecture of type 2 diabetes susceptibility. Nat Genet 46: 234–244. 10.1038/ng.2897
    1. Vlcek S, Foisner R, Wilson KL (2004) Lco1 is a novel widely expressed lamin-binding protein in the nuclear interior. Exp Cell Res 298: 499–511. 10.1016/j.yexcr.2004.04.028
    1. Hegele RA, Anderson CM, Wang J, Jones DC, Cao H (2000) Association between nuclear lamin A/C R482Q mutation and partial lipodystrophy with hyperinsulinemia, dyslipidemia, hypertension, and diabetes. Genome Res 10: 652–658. 10.1101/gr.10.5.652
    1. Mesa JL, Loos RJF, Franks PW, Ong KK, Luan Ja, et al. (2007) Lamin A/C polymorphisms, type 2 diabetes, and the metabolic syndrome: case-control and quantitative trait studies. Diabetes 56: 884–889. 10.2337/db06-1055
    1. Lappalainen T, Montgomery SB, Nica AC, Dermitzakis ET (2011) Epistatic selection between coding and regulatory variation in human evolution and disease. Am J Hum Genet 89: 459–463. 10.1016/j.ajhg.2011.08.004
    1. Rees MG, Ng D, Ruppert S, Turner C, Beer NL, et al. (2012) Correlation of rare coding variants in the gene encoding human glucokinase regulatory protein with phenotypic, cellular, and kinetic outcomes. J Clin Invest 122: 205–217. 10.1172/JCI46425
    1. Bonnefond A, Clement N, Fawcett K, Yengo L, Vaillant E, et al. (2012) Rare MTNR1B variants impairing melatonin receptor 1B function contribute to type 2 diabetes. Nat Genet 44: 297–301. 10.1038/ng.1053
    1. Zuk O, Schaffner SF, Samocha K, Do R, Hechter E, et al. (2014) Searching for missing heritability: designing rare variant association studies. Proc Natl Acad Sci U S A 111: 455–464. 10.1073/pnas.1322563111
    1. D’Orazio P, Burnett RW, Fogh-Andersen N, Jacobs E, Kuwa K, et al. (2005) Approved IFCC recommendation on reporting results for blood glucose (abbreviated). Clin Chem 51: 1573–1576. 10.1373/clinchem.2005.051979
    1. Willer CJ, Li Y, Abecasis GR (2010) METAL: fast and efficient meta-analysis of genomewide association scans. Bioinformatics 26: 2190–2191. 10.1093/bioinformatics/btq340
    1. McLaren W, Pritchard B, Rios D, Chen Y, Flicek P, et al. (2010) Deriving the consequences of genomic variants with the Ensembl API and SNP Effect Predictor. Bioinformatics 26: 2069–2070. 10.1093/bioinformatics/btq330
    1. Cingolani P, Platts A, Wang LL, Coon M, Nguyen T, et al. (2012) A program for annotating and predicting the effects of single nucleotide polymorphisms, SnpEff: SNPs in the genome of Drosophila melanogaster strain w1118; iso-2; iso-3. Fly (Austin) 6: 80–92. 10.4161/fly.19695
    1. Kumar P, Henikoff S, Ng PC (2009) Predicting the effects of coding non-synonymous variants on protein function using the SIFT algorithm. Nat Protoc 4: 1073–1081. 10.1038/nprot.2009.86
    1. Adzhubei IA, Schmidt S, Peshkin L, Ramensky VE, Gerasimova A, et al. (2010) A method and server for predicting damaging missense mutations. Nat Methods 7: 248–249. 10.1038/nmeth0410-248
    1. Gonzalez-Perez A, Lopez-Bigas N (2011) Improving the assessment of the outcome of nonsynonymous SNVs with a consensus deleteriousness score, Condel. Am J Hum Genet 88: 440–449. 10.1016/j.ajhg.2011.03.004
    1. Cooper GM, Stone EA, Asimenos G, Green ED, Batzoglou S, et al. (2005) Distribution and intensity of constraint in mammalian genomic sequence. Genome Res 15: 901–913. 10.1101/gr.3577405

Source: PubMed

3
Subskrybuj