Publications

Rafaella E. Sigala, Vasiliki Lagou, Aleksey Shmeliov, Sara Atito, Samaneh Kouchaki, Muhammad Awais, Inga Prokopenko, Adam Mahdi, Ayse Demirkan (2023)Machine Learning to Advance Human Genome-Wide Association Studies, In: Genes15(1)

Machine learning, including deep learning, reinforcement learning, and generative artificial intelligence are revolutionising every area of our lives when data are made available. With the help of these methods, we can decipher information from larger datasets while addressing the complex nature of biological systems in a more efficient way. Although machine learning methods have been introduced to human genetic epidemiological research as early as 2004, those were never used to their full capacity. In this review, we outline some of the main applications of machine learning to assigning human genetic loci to health outcomes. We summarise widely used methods and discuss their advantages and challenges. We also identify several tools, such as Combi, GenNet, and GMSTool, specifically designed to integrate these methods for hypothesis-free analysis of genetic variation data. We elaborate on the additional value and limitations of these tools from a geneticist’s perspective. Finally, we discuss the fast-moving field of foundation models and large multi-modal omics biobank initiatives.

Inga Prokopenko, Ayşe Demirkan, Marika Kaakinen (2023)51st European Mathematical Genetics Meeting (EMGM) 2023, In: Human heredity88(Suppl 1)pp. 1-72

NA

N. Amin, O. Jovanova, H.H.H. Adams, A. Dehghan, M. Kavousi, M.W. Vernooij, R.P. Peeters, F.M.S. de Vrij, S.J. van der Lee, J.G.J. van Rooij, E.M. van Leeuwen, L. Chaker, A. Demirkan, A. Hofman, R.W.W. Brouwer, R. Kraaij, K. Willems Van Dijk, T. Hankemeier, W.F.J. van Ijcken, A.G. Uitterlinden, W.J. Niessen, O.H. Franco, S.A. Kushner, M.A. Ikram, H. Tiemeier, C.M. van Duijn (2017)Exome-sequencing in a large population-based study reveals a rare Asn396Ser variant in the LIPG gene associated with depressive symptoms22(4)pp. 537-543
J. Yang, R.J.F. Loos, J.E. Powell, S.E. Medland, E.K. Speliotes, D.I. Chasman, L.M. Rose, G. Thorleifsson, V. Steinthorsdottir, R. Maegi, L. Waite, A.V. Smith, L.M. Yerges-Armstrong, K.L. Monda, D. Hadley, A. Mahajan, G. Li, K. Kapur, V. Vitart, J.E. Huffman, S.R. Wang, C. Palmer, T. Esko, K. Fischer, J.H. Zhao, A. Demirkan, A. Isaacs, M.F. Feitosa, J. Luan, N.L. Heard-Costa, C. White, A.U. Jackson, M. Preuss, A. Ziegler, J. Eriksson, Z. Kutalik, F. Frau, I.M. Nolte, J.V. van Vliet-Ostaptchouk, J.J. Hottenga, K.B. Jacobs, N. Verweij, A. Goel, C. Medina-Gomez, K. Estrada, J.L. Bragg-Gresham, S. Sanna, C. Sidore, J. Tyrer, A. Teumer, I. Prokopenko, M. Mangino, C.M. Lindgren, T.L. Assimes, A.R. Shuldiner, J. Hui, J.P. Beilby, W.L. McArdle, P. Hall, T. Haritunians, L. Zgaga, I. Kolcic, O. Polasek, T. Zemunik, B.A. Oostra, M.J. Junttila, H. Groenberg, S. Schreiber, A. Peters, A.A. Hicks, J. Stephens, N.S. Foad, J. Laitinen, A. Pouta, M. Kaakinen, G. Willemsen, J.M. Vink, S.H. Wild, G. Navis, F.W. Asselbergs, G. Homuth, U. John, C. Iribarren, T. Harris, L. Launer, V. Gudnason, J.R. O'Connell, E. Boerwinkle, G. Cadby, L.J. Palmer, A.L. James, A.W. Musk, E. Ingelsson, B.M. Psaty, J.S. Beckmann, G. Waeber, P. Vollenweider, C. Hayward, A.F. Wright, I. Rudan, L.C. Groop, A. Metspalu, K.T. Khaw, C.M. van Duijn, I.B. Borecki, M.A. Province, N.J. Wareham, J.C. Tardif, H.V. Huikuri, L.A. Cupples, L.D. Atwood, C.S. Fox, M. Boehnke, F.S. Collins, K.L. Mohlke, J. Erdmann, H. Schunkert, C. Hengstenberg, K. Stark, M. Lorentzon, C. Ohlsson, D. Cusi, J.A. Staessen, M.M. van der Klauw, P.P. Pramstaller, S. Kathiresan, J.D. Jolley, S. Ripatti, M.R. Jarvelin, E.J.C. de Geus, D.I. Boomsma, B. Penninx, J.F. Wilson, H. Campbell, S.J. Chanock, P. van der Harst, A. Hamsten, H. Watkins, A. Hofman, J.C. Witteman, M.C. Zillikens, A.G. Uitterlinden, F. Rivadeneira, L.A. Kiemeney, S.H. Vermeulen, G.R. Abecasis, D. Schlessinger, S. Schipf, M. Stumvoll, A. Toenjes, T.D. Spector, K.E. North, G. Lettre, M.I. McCarthy, S.I. Berndt, A.C. Heath, P.A.F. Madden, D.R. Nyholt, G.W. Montgomery, N.G. Martin, B. McKnight, D.P. Strachan, W.G. Hill, H. Snieder, P.M. Ridker, U. Thorsteinsdottir, K. Stefansson, T.M. Frayling, J.N. Hirschhorn, M.E. Goddard, P.M. Visscher (2012)FTO genotype is associated with phenotypic variability of body mass index, In: Nature490(7419)pp. 267-+
Ebru Nur Vanli-Yavuz, Ozkan Ozdemir, Ayse Demirkan, Suzin Catal, Nerses Bebek, Ugur Ozbek, Betul Baykan (2015)Investigation of the possible association of NEDD4-2 (NEDD4L) gene with idiopathic photosensitive epilepsy, In: Acta neurologica Belgica115(3)241pp. 241-245 Springer Nature

NEDD4-2 alias NEDD4L (neural precursor cell expressed, developmentally downregulated) gene was reported as a candidate gene for epileptic photo-sensitivity. We aimed to investigate this possible association of NEDD4-2 variants with idiopathic photosensitive epilepsy. Consecutive patients who had been followed up at our epilepsy center and diagnosed with idiopathic epilepsy according to ILAE criteria and clear-cut photoparoxysmal responses in their electroencephalograms and 100 ethnically matched healthy subjects were included in the study. The regions around previously reported three variants, namely, S233L, E271A and H515P were tracked with DHPLC and the samples showing variations were sequenced. 81 patients (63 females) aged between 12-63 years (45 had juvenile myoclonic epilepsy, 11 childhood absence epilepsy, 14 juvenile absence epilepsy, 7 late onset idiopathic generalized epilepsy, 1 unclassified idiopathic generalized epilepsy, and 3 patients with idiopathic photosensitive occipital lobe epilepsy) were included in this study. We found only one heterozygous S233L variant in a 23-year-old man who has photosensitive form of juvenile absence epilepsy and pattern sensitivity to striped carpets. Other two variants were not found in any of the other patients and controls. Our results suggest that three screened NEDD4-2 variants do not play a leading role in the pathogenesis of photosensitive epilepsy in the Turkish population.

Johanna M. Colijn, Anneke den Hollander, Ayse Demirkan, Audrey Cougnard-Gregoire, Timo Verzijden, Eveline Kersten, Magda A. Meester-Smoor, Benedicte M. J. Merle, Grigorios Papageorgiou, Shahzad Ahmad, Monique T. Mulder, Miguel Angelo Costa, Pascale Benlian, Geir Bertelsen, Alain M. Bron, Birte Claes, Catherine Creuzot-Garcher, Maja Gran Erke, Sascha Fauser, Paul J. Foster, Christopher J. Hammond, Hans-Werner Hense, Carel B. Hoyng, Anthony P. Khawaja, Jean-Francois Korobelnik, Stefano Piermarocchi, Tatiana Segato, Rufino Silva, Eric H. Souied, Katie M. Williams, Cornelia M. van Duijn, Cecile Delcourt, Caroline C. W. Klaver, Niyazi Acar, Lebriz Altay, Eleftherios Anastosopoulos, Augusto Azuara-Blanco, Tos Berendschot, Arthur Bergen, Christine Binquet, Alan Bird, Martin Bobak, Morten Bogelund Larsen, Camiel Boon, Rupert Bourne, Lionel Bretillon, Rebecca Broe, Alain Bron, Gabrielle Buitendijk, Maria Luz Cachulo, Vittorio Capuano, Isabelle Carriere, Usha Chakravarthy, Michelle Chan, Petrus Chang, Johanna Colijn, Angela Cree, Phillippa Cumberland, Jose Cunha-Vaz, Vincent Daien, Eiko De Jong, Gabor Deak, Marie-Noelle Delyfer, Anneke den Hollander, Martha Dietzel, Pedro Faria, Claudia Farinha, Robert Finger, Astrid Fletcher, Paul Foster, Panayiota Founti, Theo Gorgels, Jakob Grauslund, Franz Grus, Christopher Hammond, Thomas Heesterbeek, Manuel Hermann, Rene Hoehn, Ruth Hogg, Frank Holz, Carel Hoyng, Nomdo Jansonius, Sarah Janssen, Eiko de Jonga, Anthony Khawaja, Caroline Klaver, Julia Lamparter, Melanie Le Goff, Terho Lehtimaki, Irene Leung, Andrew Lotery, Matthias Mauschitz, Magda Meester, Benedicte Merle, Verena Meyer Zu Westrup, Edoardo Midena, Stefania Miotto, Alireza Mirshahi, Sadek Mohan-Said, Michael Mueller (2019)Increased High-Density Lipoprotein Levels Associated with Age-Related Macular Degeneration Evidence from the EYE-RISK and European Eye Epidemiology Consortia, In: Ophthalmology (Rochester, Minn.)126(3)pp. 393-406 Elsevier

Purpose: Genetic and epidemiologic studies have shown that lipid genes and high-density lipoproteins (HDLs) are implicated in age-related macular degeneration (AMD). We studied circulating lipid levels in relationship to AMD in a large European dataset. Design: Pooled analysis of cross-sectional data. Participants: Individuals (N = 30 953) aged 50 years or older participating in the European Eye Epidemiology (E3) consortium and 1530 individuals from the Rotterdam Study with lipid subfraction data. Methods: AMD features were graded on fundus photographs using the Rotterdam classification. Routine blood lipid measurements, genetics, medication, and potential confounders were extracted from the E3 database. In a subgroup of the Rotterdam Study, lipid subfractions were identified by the Nightingale biomarker platform. Random-intercepts mixed-effects models incorporating confounders and study site as a random effect were used to estimate associations. Main Outcome Measures: AMD features and stage; lipid measurements. Results: HDL was associated with an increased risk of AMD (odds ratio [OR], 1.21 per 1-mmol/l increase; 95% confidence interval [CI], 1.14-1.29), whereas triglycerides were associated with a decreased risk (OR, 0.94 per 1-mmol/l increase; 95% CI, 0.91-0.97). Both were associated with drusen size. Higher HDL raised the odds of larger drusen, whereas higher triglycerides decreases the odds. LDL cholesterol reached statistical significance only in the association with early AMD (P = 0.045). Regarding lipid subfractions, the concentration of extra-large HDL particles showed the most prominent association with AMD (OR, 1.24; 95% CI, 1.10-1.40). The cholesteryl ester transfer protein risk variant (rs17231506) for AMD was in line with increased HDL levels (P = 7.7 x 10(-7)), but lipase C risk variants (rs2043085, rs2070895) were associated in an opposite way (P = 1.0 x 10(-6) and P = 1.6 x 10(-4)). Conclusions: Our study suggested that HDL cholesterol is associated with increased risk of AMD and that triglycerides are negatively associated. Both show the strongest association with early AMD and drusen. Extra-large HDL subfractions seem to be drivers in the relationship with AMD, and variants in lipid genes play a more ambiguous role in this association. Whether systemic lipids directly influence AMD or represent lipid metabolism in the retina remains to be answered. (C) 2018 by the American Academy of Ophthalmology

Ron Do, Cristen J. Willer, Ellen M. Schmidt, Sebanti Sengupta, Chi Gao, Gina M. Peloso, Stefan Gustafsson, Stavroula Kanoni, Andrea Ganna, Jin Chen, Martin L. Buchkovich, Samia Mora, Jacques S. Beckmann, Jennifer L. Bragg-Gresham, Hsing-Yi Chang, Ayse Demirkan, Heleen M. Den Hertog, Louise A. Donnelly, Georg B. Ehret, Tonu Esko, Mary F. Feitosa, Teresa Ferreira, Krista Fischer, Pierre Fontanillas, Ross M. Fraser, Daniel F. Freitag, Deepti Gurdasani, Kauko Heikkila, Elina Hyppoenen, Aaron Isaacs, Anne U. Jackson, Asa Johansson, Toby Johnson, Marika Kaakinen, Johannes Kettunen, Marcus E. Kleber, Xiaohui Li, Jian'an Luan, Leo-Pekka Lyytikainen, Patrik K. E. Magnusson, Massimo Mangino, Evelin Mihailov, May E. Montasser, Martina Mueller-Nurasyid, Ilja M. Nolte, Jeffrey R. O'Connell, Cameron D. Palmer, Markus Perola, Ann-Kristin Petersen, Serena Sanna, Richa Saxena, Susan K. Service, Sonia Shah, Dmitry Shungin, Carlo Sidore, Ci Song, Rona J. Strawbridge, Ida Surakka, Toshiko Tanaka, Tanya M. Teslovich, Gudmar Thorleifsson, Evita G. Van den Herik, Benjamin F. Voight, Kelly A. Volcik, Lindsay L. Waite, Andrew Wong, Ying Wu, Weihua Zhang, Devin Absher, Gershim Asiki, Ines Barroso, Latonya F. Been, Jennifer L. Bolton, Lori L. Bonnycastle, Paolo Brambilla, Mary S. Burnett, Giancarlo Cesana, Maria Dimitriou, Alex S. F. Doney, Angela Doering, Paul Elliott, Stephen E. Epstein, Gudmundur Ingi Eyjolfsson, Bruna Gigante, Mark O. Goodarzi, Harald Grallert, Martha L. Gravito, Christopher J. Groves, Goran Hallmans, Anna-Liisa Hartikainen, Caroline Hayward, Dena Hernandez, Andrew A. Hicks, Hilma Holm, Yi-Jen Hung, Thomas Illig, Michelle R. Jones, Pontiano Kaleebu, John J. P. Kastelein, Kay-Tee Khaw (2013)Common variants associated with plasma triglycerides and risk for coronary artery disease, In: Nature genetics45(11)1345pp. 1345-1352 Springer Nature

Triglycerides are transported in plasma by specific triglyceride-rich lipoproteins; in epidemiological studies, increased triglyceride levels correlate with higher risk for coronary artery disease (CAD). However, it is unclear whether this association reflects causal processes. We used 185 common variants recently mapped for plasma lipids (P < 5 x 10(-8) for each) to examine the role of triglycerides in risk for CAD. First, we highlight loci associated with both low-density lipoprotein cholesterol (LDL-C) and triglyceride levels, and we show that the direction and magnitude of the associations with both traits are factors in determining CAD risk. Second, we consider loci with only a strong association with triglycerides and show that these loci are also associated with CAD. Finally, in a model accounting for effects on LDL-C and/or high-density lipoprotein cholesterol (HDL-C) levels, the strength of a polymorphism's effect on triglyceride levels is correlated with the magnitude of its effect on CAD risk. These results suggest that triglyceride-rich lipoproteins causally influence risk for CAD.

Johannes Kettunen, Ayse Demirkan, Peter Wurtz, Harmen H. M. Draisma, Toomas Haller, Rajesh Rawal, Anika Vaarhorst, Antti J. Kangas, Leo-Pekka Lyytikaeinen, Matti Pirinen, Rene Pool, Antti-Pekka Sarin, Pasi Soininen, Taru Tukiainen, Qin Wang, Mika Tiainen, Tuulia Tynkkynen, Najaf Amin, Tanja Zeller, Marian Beekman, Joris Deelen, Ko Willems van Dijk, Tonu Esko, Jouke-Jan Hottenga, Elisabeth M. van Leeuwen, Terho Lehtimaki, Evelin Mihailov, Richard J. Rose, Anton J. M. de Craen, Christian Gieger, Mika Kahonen, Markus Perola, Stefan Blankenberg, Markku J. Savolainen, Aswin Verhoeven, Jorma Viikari, Gonneke Willemsen, Dorret I. Boomsma, Cornelia M. van Duijn, Johan Eriksson, Antti Jula, Marjo-Riitta Jarvelin, Jaakko Kaprio, Andres Metspalu, Olli Raitakari, Veikko Salomaa, P. Eline Slagboom, Melanie Waldenberger, Samuli Ripatti, Mika Ala-Korpela (2016)Genome-wide study for circulating metabolites identifies 62 loci and reveals novel systemic effects of LPA, In: Nature communications7(1)11122pp. 11122-11122 Springer Nature

Genome-wide association studies have identified numerous loci linked with complex diseases, for which the molecular mechanisms remain largely unclear. Comprehensive molecular profiling of circulating metabolites captures highly heritable traits, which can help to uncover metabolic pathophysiology underlying established disease variants. We conduct an extended genome-wide association study of genetic influences on 123 circulating metabolic traits quantified by nuclear magnetic resonance metabolomics from up to 24,925 individuals and identify eight novel loci for amino acids, pyruvate and fatty acids. The LPA locus link with cardiovascular risk exemplifies how detailed metabolic profiling may inform underlying aetiology via extensive associations with very-low-density lipoprotein and triglyceride metabolism. Genetic fine mapping and Mendelian randomization uncover wide-spread causal effects of lipoprotein(a) on overall lipoprotein metabolism and we assess potential pleiotropic consequences of genetically elevated lipoprotein(a) on diverse morbidities via electronic health-care records. Our findings strengthen the argument for safe LPA-targeted intervention to reduce cardiovascular risk.

Martin A. Kohli, Susanne Lucae, Philipp G. Saemann, Mathias V. Schmidt, Ayse Demirkan, Karin Hek, Darina Czamara, Michael Alexander, Daria Salyakina, Stephan Ripke, David Hoehn, Michael Specht, Andreas Menke, Johannes Hennings, Angela Heck, Christiane Wolf, Marcus Ising, Stefan Schreiber, Michael Czisch, Marianne B. Mueller, Manfred Uhr, Thomas Bettecken, Albert Becker, Johannes Schramm, Marcella Rietschel, Wolfgang Maier, Bekh Bradley, Kerry J. Ressler, Markus M. Noethen, Sven Cichon, Ian W. Craig, Gerome Breen, Cathryn M. Lewis, Albert Hofman, Henning Tiemeier, Cornelia M. van Duijn, Florian Holsboer, Bertram Mueller-Myhsok, Elisabeth B. Binder (2011)The Neuronal Transporter Gene SLC6A15 Confers Risk to Major Depression, In: Neuron (Cambridge, Mass.)70(2)pp. 252-265 Elsevier

Major depression (MD) is one of the most prevalent psychiatric disorders and a leading cause of loss in work productivity. A combination of genetic and environmental risk factors probably contributes to MD. We present data from a genome-wide association study revealing a neuron-specific neutral amino acid transporter (SLC6A15) as a susceptibility gene for MD. Risk allele carrier status in humans and chronic stress in mice were associated with a down-regulation of the expression of this gene in the hippocampus, a brain region implicated in the pathophysiology of MD. The same polymorphisms also showed associations with alterations in hippocampal volume and neuronal integrity. Thus, decreased SLC6A15 expression, due to genetic or environmental factors, might alter neuronal circuits related to the susceptibility for MD. Our convergent data from human genetics, expression studies, brain imaging, and animal models suggest a pathophysiological mechanism for MD that may be accessible to drug targeting.

Yukinori Okada, Xueling Sim, Min Jin Go, Jer-Yuarn Wu, Dongfeng Gu, Fumihiko Takeuchi, Atsushi Takahashi, Shiro Maeda, Tatsuhiko Tsunoda, Peng Chen, Su-Chi Lim, Tien-Yin Wong, Jianjun Liu, Terri L. Young, Tin Aung, Mark Seielstad, Yik-Ying Teo, Young Jin Kim, Jong-Young Lee, Bok-Ghee Han, Daehee Kang, Chien-Hsiun Chen, Fuu-Jen Tsai, Li-Ching Chang, S-J Cathy Fann, Hao Mei, Dabeeru C. Rao, James E. Hixson, Shufeng Chen, Tomohiro Katsuya, Masato Isono, Toshio Ogihara, John C. Chambers, Weihua Zhang, Jaspal S. Kooner, Eva Albrecht, Kazuhiko Yamamoto, Michiaki Kubo, Yusuke Nakamura, Naoyuki Kamatani, Norihiro Kato, Jiang He, Yuan-Tsong Chen, Yoon Shin Cho, E-Shyong Tai, Toshihiro Tanaka, Ayse Demirkan (2012)Meta-analysis identifies multiple loci associated with kidney function-related traits in east Asian populations, In: Nature genetics44(8)pp. 904-909 Springer Nature

Chronic kidney disease (CKD), impairment of kidney function, is a serious public health problem, and the assessment of genetic factors influencing kidney function has substantial clinical relevance. Here, we report a meta-analysis of genomewide association studies for kidney function-related traits, including 71,149 east Asian individuals from 18 studies in 11 population-, hospital- or family-based cohorts, conducted as part of the Asian Genetic Epidemiology Network (AGEN). Our meta-analysis identified 17 loci newly associated with kidney function-related traits, including the concentrations of blood urea nitrogen, uric acid and serum creatinine and estimated glomerular filtration rate based on serum creatinine levels (eGFRcrea) (P < 5.0 x 10(-8)). We further examined these loci with in silico replication in individuals of European ancestry from the KidneyGen, CKDGen and GUGC consortia, including a combined total of similar to 110,347 individuals. We identify pleiotropic associations among these loci with kidney function-related traits and risk of CKD. These findings provide new insights into the genetics of kidney function.

Beben Benyamin, Tonu Esko, Janina S Ried, Aparna Radhakrishnan, Sita H Vermeulen, Michela Traglia, Martin Gögele, Denise Anderson, Linda Broer, Clara Podmore, Jian'an Luan, Zoltan Kutalik, Serena Sanna, Peter van der Meer, Toshiko Tanaka, Fudi Wang, Harm-Jan Westra, Lude Franke, Evelin Mihailov, Lili Milani, Jonas Hälldin, Jonas Häldin, Juliane Winkelmann, Thomas Meitinger, Joachim Thiery, Annette Peters, Melanie Waldenberger, Augusto Rendon, Jennifer Jolley, Jennifer Sambrook, Lambertus A Kiemeney, Fred C Sweep, Cinzia F Sala, Christine Schwienbacher, Irene Pichler, Jennie Hui, Ayse Demirkan, Aaron Isaacs, Najaf Amin, Maristella Steri, Gérard Waeber, Niek Verweij, Joseph E Powell, Dale R Nyholt, Andrew C Heath, Pamela A F Madden, Peter M Visscher, Margaret J Wright, Grant W Montgomery, Nicholas G Martin, Dena Hernandez, Stefania Bandinelli, Pim van der Harst, Manuela Uda, Peter Vollenweider, Robert A Scott, Claudia Langenberg, Nicholas J Wareham, Cornelia van Duijn, John Beilby, Peter P Pramstaller, Andrew A Hicks, Willem H Ouwehand, Konrad Oexle, Christian Gieger, Andres Metspalu, Clara Camaschella, Daniela Toniolo, Dorine W Swinkels, John B Whitfield (2014)Novel loci affecting iron homeostasis and their effects in individuals at risk for hemochromatosis, In: Nature communications5(1)4926pp. 4926-4926

Variation in body iron is associated with or causes diseases, including anaemia and iron overload. Here, we analyse genetic association data on biochemical markers of iron status from 11 European-population studies, with replication in eight additional cohorts (total up to 48,972 subjects). We find 11 genome-wide-significant (P

Ayse Demirkan, Brenda W.J.H. Penninx, Karin Hek, Naomi Wray, Najaf Amin, Yurii Aulchenko, Richard van Dyck, Eco de Geus, Albert Hofman, André Uitterlinden, Jouke-Jan Hottenga, Willem Nolen, Ben Oostra, Patrick Sullivan, G. Willemsen, Frans Zitman, Henning Tiemeier, Cecile Janssens, Dorret Boomsma, Cornelia van Duijn, Christel Middeldorp (2011)Genetic risk profiles for depression and anxiety in adult and elderly cohorts, In: Molecular psychiatry16(7)773pp. 773-783 Nature Publishing Group

The first generation of genome-wide association studies (GWA studies) for psychiatric disorders has led to new insights regarding the genetic architecture of these disorders. We now start to realize that a larger number of genes, each with a small contribution, are likely to explain the heritability of psychiatric diseases. The contribution of a large number of genes to complex traits can be investigated with genome-wide profiling. In a discovery sample a genetic risk profile for depression was defined based on a GWA study of 1738 adult cases and 1802 controls. The genetic risk scores were tested in two population based samples of elderly participants. The genetic risk profiles were evaluated for depression and anxiety in the Rotterdam Study cohort and the Erasmus Rucphen Family (ERF) study. The genetic risk scores were significantly associated to different measures of depression and explained up to ~0.7% of the variance in depression in Rotterdam Study and up to ~1 % in ERF study. The genetic score for depression was also significantly associated to anxiety explaining up to 2.1% in Rotterdam study. These findings suggest the presence of many genetic loci of small effect that influence both depression and anxiety. Remarkably, the predictive value of these profiles was as large in the sample of elderly participants as in the middle-aged samples.

Ayse Demirkan, Peter Henneman, Aswin Verhoeven, Harish Dharuri, Najaf Amin, Jan Bert van Klinken, Lennart C. Karssen, Boukje de Vries, Axel Meissner, Sibel Goraler, Arn M. J. M. van den Maagdenberg, Andre M. Deelder, Peter A. C. 't Hoen, Cornelia M. van Duijn, Ko Willems van Dijk (2015)Insight in Genome-Wide Association of Metabolite Quantitative Traits by Exome Sequence Analyses, In: PLoS genetics11(1)1004835pp. e1004835-e1004835 Public Library Science

Metabolite quantitative traits carry great promise for epidemiological studies, and their genetic background has been addressed using Genome-Wide Association Studies (GWAS). Thus far, the role of less common variants has not been exhaustively studied. Here, we set out a GWAS for metabolite quantitative traits in serum, followed by exome sequence analysis to zoom in on putative causal variants in the associated genes. H-1 Nuclear Magnetic Resonance (H-1-NMR) spectroscopy experiments yielded successful quantification of 42 unique metabolites in 2,482 individuals from The Erasmus Rucphen Family (ERF) study. Heritability of metabolites were estimated by SOLAR. GWAS was performed by linear mixed models, using HapMap imputations. Based on physical vicinity and pathway analyses, candidate genes were screened for coding region variation using exome sequence data. Heritability estimates for metabolites ranged between 10% and 52%. GWAS replicated three known loci in the metabolome wide significance: CPS1 with glycine (P-value = 1.27x10(-32)), PRODH with proline (P-value = 1.11x10(-19)), SLC16A9 with carnitine level (P-value = 4.81x10(-14)) and uncovered a novel association between DMGDH and dimethyl-glycine (P-value = 1.65x10(-19)) level. In addition, we found three novel, suggestively significant loci: TNP1 with pyruvate (P-value = 1.26x10(-8)), KCNJ16 with 3-hydroxybutyrate (P-value = 1.65x10(-8)) and 2p12 locus with valine (P-value = 3.49x10(-8)). Exome sequence analysis identified potentially causal coding and regulatory variants located in the genes CPS1, KCNJ2 and PRODH, and revealed allelic heterogeneity for CPS1 and PRODH. Combined GWAS and exome analyses of metabolites detected by high-resolution H-1-NMR is a robust approach to uncover metabolite quantitative trait loci (mQTL), and the likely causative variants in these loci. It is anticipated that insight in the genetics of intermediate phenotypes will provide additional insight into the genetics of complex traits.

Natasha Ng, Sara Willems, Anna Gloyn, Inês Barroso, Meta-Analyses Of Glucose And Insulin-Related Traits Consortium, Ayse Demirkan Tissue-Specific Alteration of Metabolic Pathways Influences Glycemic Regulation, In: bioRxiv Cold Spring Harbor Laboratory Press

Metabolic dysregulation in multiple tissues alters glucose homeostasis and influences risk for type 2 diabetes (T2D). To identify pathways and tissues influencing T2D-relevant glycemic traits (fasting glucose [FG], fasting insulin [FI], two-hour glucose [2hGlu] and glycated hemoglobin [HbA1c]), we investigated associations of exome-array variants in up to 144,060 individuals without diabetes of multiple ancestries. Single-variant analyses identified novel associations at 21 coding variants in 18 novel loci, whilst gene-based tests revealed signals at two genes, TF (HbA1c) and G6PC (FG, FI). Pathway and tissue enrichment analyses of trait-associated transcripts confirmed the importance of liver and kidney for FI and pancreatic islets for FG regulation, implicated adipose tissue in FI and the gut in 2hGlu, and suggested a role for the non-endocrine pancreas in glucose homeostasis. Functional studies demonstrated that a novel FG/FI association at the liver-enriched G6PC transcript was driven by multiple rare loss-of-function variants. The FG/HbA1c-associated, islet-specific G6PC2 transcript also contained multiple rare functional variants, including two alleles within the same codon with divergent effects on glucose levels. Our findings highlight the value of integrating genomic and functional data to maximize biological inference.

Ayse Demirkan, Najaf Amin, Aaron Isaacs, Marjo-Riitta Jarvelin, John B. Whitfield, Heinz-Erich Wichmann, Kirsten Ohm Kyvik, Igor Rudan, Christian Gieger, Andrew A. Hicks, Asa Johansson, Jouke-Jan Hottenga, Johannes J. Smith, Sarah H. Wild, Nancy L. Pedersen, Gonneke Willemsen, Massimo Mangino, Caroline Hayward, Andre G. Uitterlinden, Albert Hofman, Jacqueline Witteman, Grant W. Montgomery, Kirsi H. Pietilainen, Taina Rantanen, Jaakko Kaprio, Angela Doering, Peter P. Pramstaller, Ulf Gyllensten, Eco J. C. de Geus, Brenda W. Penninx, James F. Wilson, Fernando Rivadeneria, Patrik K. E. Magnusson, Dorret I. Boomsma, Tim Spector, Harry Campbell, Birgit Hoehne, Nicholas G. Martin, Ben A. Oostra, Mark McCarthy, Leena Peltonen-Palotie, Yurii Aulchenko, Peter M. Visscher, Samuli Ripatti, A. Cecile J. W. Janssens, Cornelia M. van Duijn (2011)Genetic architecture of circulating lipid levels, In: European journal of human genetics : EJHG19(7)pp. 813-819 Springer Nature

Serum concentrations of low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), triglycerides (TGs) and total cholesterol (TC) are important heritable risk factors for cardiovascular disease. Although genome-wide association studies (GWASs) of circulating lipid levels have identified numerous loci, a substantial portion of the heritability of these traits remains unexplained. Evidence of unexplained genetic variance can be detected by combining multiple independent markers into additive genetic risk scores. Such polygenic scores, constructed using results from the ENGAGE Consortium GWAS on serum lipids, were applied to predict lipid levels in an independent population-based study, the Rotterdam Study-II (RS-II). We additionally tested for evidence of a shared genetic basis for different lipid phenotypes. Finally, the polygenic score approach was used to identify an alternative genome-wide significance threshold before pathway analysis and those results were compared with those based on the classical genome-wide significance threshold. Our study provides evidence suggesting that many loci influencing circulating lipid levels remain undiscovered. Cross-prediction models suggested a small overlap between the polygenic backgrounds involved in determining LDL-C, HDL-C and TG levels. Pathway analysis utilizing the best polygenic score for TC uncovered extra information compared with using only genome-wide significant loci. These results suggest that the genetic architecture of circulating lipids involves a number of undiscovered variants with very small effects, and that increasing GWAS sample sizes will enable the identification of novel variants that regulate lipid levels. European Journal of Human Genetics (2011) 19, 813-819; doi:10.1038/ejhg.2011.21; published online 30 March 2011

Anna Koettgen, Eva Albrecht, Alexander Teumer, Veronique Vitart, Jan Krumsiek, Claudia Hundertmark, Giorgio Pistis, Daniela Ruggiero, Conall M. O'Seaghdha, Toomas Haller, Qiong Yang, Toshiko Tanaka, Andrew D. Johnson, Zoltan Kutalik, Albert V. Smith, Julia Shi, Maksim Struchalin, Rita P. S. Middelberg, Morris J. Brown, Angelo L. Gaffo, Nicola Pirastu, Guo Li, Caroline Hayward, Tatijana Zemunik, Jennifer Huffman, Loic Yengo, Jing Hua Zhao, Ayse Demirkan, Mary F. Feitosa, Xuan Liu, Giovanni Malerba, Lorna M. Lopez, Pim van der Harst, Xinzhong Li, Marcus E. Kleber, Andrew A. Hicks, Ilja M. Nolte, Asa Johansson, Federico Murgia, Sarah H. Wild, Stephan J. L. Bakker, John F. Peden, Abbas Dehghan, Maristella Steri, Albert Tenesa, Vasiliki Lagou, Perttu Salo, Massimo Mangino, Lynda M. Rose, Terho Lehtimaki, Owen M. Woodward, Yukinori Okada, Adrienne Tin, Christian Mueller, Christopher Oldmeadow, Margus Putku, Darina Czamara, Peter Kraft, Laura Frogheri, Gian Andri Thun, Anne Grotevendt, Gauti Kjartan Gislason, Tamara B. Harris, Lenore J. Launer, Patrick McArdle, Alan R. Shuldiner, Eric Boerwinkle, Josef Coresh, Helena Schmidt, Michael Schallert, Nicholas G. Martin, Grant W. Montgomery, Michiaki Kubo, Yusuke Nakamura, Toshihiro Tanaka, Patricia B. Munroe, Nilesh J. Samani, David R. Jacobs, Kiang Liu, Pio D'Adamo, Sheila Ulivi, Jerome I. Rotter, Bruce M. Psaty, Peter Vollenweider, Gerard Waeber, Susan Campbell, Olivier Devuyst, Pau Navarro, Ivana Kolcic, Nicholas Hastie, Beverley Balkau, Philippe Froguel, Tonu Esko, Andres Salumets, Kay Tee Khaw, Claudia Langenberg, Nicholas J. Wareham, Aaron Isaacs, Aldi Kraja, Qunyuan Zhang, Inga Prokopenko (2013)Genome-wide association analyses identify 18 new loci associated with serum urate concentrations, In: Nature genetics45(2)145pp. 145-154 Springer Nature

Elevated serum urate concentrations can cause gout, a prevalent and painful inflammatory arthritis. By combining data from >140,000 individuals of European ancestry within the Global Urate Genetics Consortium (GUGC), we identified and replicated 28 genome-wide significant loci in association with serum urate concentrations (18 new regions in or near TRIM46, INHBB, SEMBT1, TMEM171, VEGFA, BAZ1B, PRKAG2, STC1, HNF4G, A1CF, ATXN2, UBE2Q2, IGF1R, NFAT5, MAF, HLF, ACVR1B-ACVRL1 and B3GNT4). Associations for many of the loci were of similar magnitude in individuals of non-European ancestry. We further characterized these loci for associations with gout, transcript expression and the fractional excretion of urate. Network analyses implicate the inhibins-activins signaling pathways and glucose metabolism in systemic urate control. New candidate genes for serum urate concentration highlight the importance of metabolic control of urate production and excretion, which may have implications for the treatment and prevention of gout.

Fabiola M. Del Greco, Luisa Foco, Alexander Teumer, Niek Verweij, Giuseppe Paglia, Vivian Meraviglia, Roberto Melotti, Vladimir Vukovic, Werner Rauhe, Peter K. Joshi, Ayse Demirkan, Stephan B. Felix, Maik Pietzner, Abdullah Said, Yordi J. van de Vegte, Pim van der Harst, Alan F. Wright, Andrew A. Hicks, Harry Campbell, Marcus Doerr, Harold Snieder, James F. Wilson, Peter P. Pramstaller, Alessandra Rossini, Cristian Pattaro (2019)Lipidomics, Atrial Conduction, and Body Mass Index Evidence From Association, Mediation, and Mendelian Randomization Models, In: Circulation. Genomic and precision medicine12(7)002384pp. 315-325 Lippincott Williams & Wilkins

Background: Lipids are increasingly involved in cardiovascular risk prediction as potential proarrhythmic influencers. However, knowledge is limited about the specific mechanisms connecting lipid alterations with atrial conduction. Methods: To shed light on this issue, we conducted a broad assessment of 151 sphingo- and phospholipids, measured using mass spectrometry, for association with atrial conduction, measured by P wave duration (PWD) from standard electrocardiograms, in the MICROS study (Microisolates in South Tyrol) (n=839). Causal pathways involving lipidomics, body mass index (BMI), and PWD were assessed using 2-sample Mendelian randomization analyses based on published genome-wide association studies of lipidomics (n=4034) and BMI (n=734 481), and genetic association analysis of PWD in 5 population-based studies (n=24 236). Results: We identified an association with relative phosphatidylcholine 38:3 (%PC 38:3) concentration, which was replicated in the ORCADES (Orkney Complex Disease Study; n=951), with a pooled association across studies of 2.59 (95% CI, 1.3-3.9; P=1.1x10(-4)) ms PWD per mol% increase. While being independent of cholesterol, triglycerides, and glucose levels, the %PC 38:3-PWD association was mediated by BMI. Results supported a causal effect of BMI on both PWD (P=8.3x10(-5)) and %PC 38:3 (P=0.014). Conclusions: Increased %PC 38:3 levels are consistently associated with longer PWD, partly because of the confounding effect of BMI. The causal effect of BMI on PWD reinforces evidence of BMI's involvement into atrial electrical activity.

Tuomas O Kilpeläinen, Amy R Bentley, Raymond Noordam, Yun Ju Sung, Karen Schwander, Thomas W Winkler, Hermina Jakupović, Daniel I Chasman, Alisa Manning, Ioanna Ntalla, Hugues Aschard, Michael R Brown, Lisa de las Fuentes, Nora Franceschini, Xiuqing Guo, Dina Vojinovic, Stella Aslibekyan, Mary F Feitosa, Minjung Kho, Solomon K Musani, Melissa Richard, Heming Wang, Zhe Wang, Traci M Bartz, Lawrence F Bielak, Archie Campbell, Rajkumar Dorajoo, Virginia Fisher, Fernando P Hartwig, Andrea R V R Horimoto, Changwei Li, Kurt K Lohman, Jonathan Marten, Xueling Sim, Albert V Smith, Salman M Tajuddin, Maris Alver, Marzyeh Amini, Mathilde Boissel, Jin Fang Chai, Xu Chen, Jasmin Divers, Evangelos Evangelou, Chuan Gao, Mariaelisa Graff, Sarah E Harris, Meian He, Fang-Chi Hsu, Anne U Jackson, Jing Hua Zhao, Aldi T Kraja, Brigitte Kühnel, Federica Laguzzi, Leo-Pekka Lyytikäinen, Ilja M Nolte, Rainer Rauramaa, Muhammad Riaz, Antonietta Robino, Rico Rueedi, Heather M Stringham, Fumihiko Takeuchi, Peter J van der Most, Tibor V Varga, Niek Verweij, Erin B Ware, Wanqing Wen, Xiaoyin Li, Lisa R Yanek, Najaf Amin, Donna K Arnett, Eric Boerwinkle, Marco Brumat, Brian Cade, Mickaël Canouil, Yii-Der Ida Chen, Maria Pina Concas, John Connell, Renée de Mutsert, H Janaka de Silva, Paul S de Vries, Ayşe Demirkan, Jingzhong Ding, Charles B Eaton, Jessica D Faul, Yechiel Friedlander, Kelley P Gabriel, Mohsen Ghanbari, Franco Giulianini, Chi Charles Gu, Dongfeng Gu, Tamara B Harris, Jiang He, Sami Heikkinen, Chew-Kiat Heng, Steven C Hunt, M Arfan Ikram, Jost B Jonas, Woon-Puay Koh, Pirjo Komulainen, Jose E Krieger (2019)Multi-ancestry study of blood lipid levels identifies four loci interacting with physical activity, In: Nature communications10(1)376pp. 376-11

Many genetic loci affect circulating lipid levels, but it remains unknown whether lifestyle factors, such as physical activity, modify these genetic effects. To identify lipid loci interacting with physical activity, we performed genome-wide analyses of circulating HDL cholesterol, LDL cholesterol, and triglyceride levels in up to 120,979 individuals of European, African, Asian, Hispanic, and Brazilian ancestry, with follow-up of suggestive associations in an additional 131,012 individuals. We find four loci, in/near CLASP1, LHX1, SNTA1, and CNTNAP2, that are associated with circulating lipid levels through interaction with physical activity; higher levels of physical activity enhance the HDL cholesterol-increasing effects of the CLASP1, LHX1, and SNTA1 loci and attenuate the LDL cholesterol-increasing effect of the CNTNAP2 locus. The CLASP1, LHX1, and SNTA1 regions harbor genes linked to muscle function and lipid metabolism. Our results elucidate the role of physical activity interactions in the genetic contribution to blood lipid levels.

Selcuk Gormez, Ayse Demirkan, Fatmahan Atalar, Baris Caynak, Refik Erdim, Volkan Sozer, Demet Gunay, Bellhan Akpinar, Ugur Ozbek, Ahmet Sevim Buyukdevrim (2011)Adipose Tissue Gene Expression of Adiponectin, Tumor Necrosis Factor-alpha and Leptin in Metabolic Syndrome Patients with Coronary Artery Disease, In: Internal medicine (Tokyo, 1992)50(8)pp. 805-810 Japan Soc Internal Medicine

Objective Metabolic syndrome (MS) is associated with an increased risk of coronary artery disease (CAD) and type 2 diabetes mellitus (DM). In MS, adipose tissue has been shown to function as a paracrine and an endocrine organ secreting various adipocytokines. In the current study, adiponectin, tumor necrosis factor-alpha (TNF-alpha) and leptin gene expressions in the epicardial adipose tissue (EAT), paracardial adipose tissue (PAT) and subcutaneous adipose tissue (SAT) were investigated in MS patients with CAD and in non-MS patients without CAD. Methods and Results Thirty-seven patients with MS undergoing coronary artery bypass grafting due to CAD (MS group) and twenty-three non-MS patients without CAD undergoing heart valve surgery (control group) were recruited prospectively to the study. Relative gene expressions of adiponectin, TNF-alpha and leptin in EAT, PAT and SAT were compared between two groups of patients. Adiponectin gene expression in EAT and PAT were significantly lower in MS group compared to the control group (p < 0.0001, p=0.04, respectively) while SAT adiponectin gene expression did not differ significantly (p=0.64). TNF-alpha and leptin gene expressions were found to be statistically significantly higher in EAT, PAT and SAT of the MS group (p < 0.0001, for all). Conclusion Our results demonstrate that TNF-alpha and leptin gene expressions increase prominently in the EAT, PAT and SAT while adiponectin gene expression decreases significantly in EAT and PAT in MS patients with CAD. These findings suggest that disturbances in expression of adiponectin, TNF-alpha and leptin in EAT, PAT and SAT might play an important role in MS patients with CAD.

Tuomas O Kilpeläinen, Jayne F Martin Carli, Alicja A Skowronski, Qi Sun, Jennifer Kriebel, Mary F Feitosa, Åsa K Hedman, Alexander W Drong, James E Hayes, Jinghua Zhao, Tune H Pers, Ursula Schick, Niels Grarup, Zoltán Kutalik, Stella Trompet, Massimo Mangino, Kati Kristiansson, Marian Beekman, Leo-Pekka Lyytikäinen, Joel Eriksson, Peter Henneman, Jari Lahti, Toshiko Tanaka, Jian'an Luan, Fabiola Del Greco M, Dorota Pasko, Frida Renström, Sara M Willems, Anubha Mahajan, Lynda M Rose, Xiuqing Guo, Yongmei Liu, Marcus E Kleber, Louis Pérusse, Tom Gaunt, Tarunveer S Ahluwalia, Yun Ju Sung, Yolande F Ramos, Najaf Amin, Antoinette Amuzu, Inês Barroso, Claire Bellis, John Blangero, Brendan M Buckley, Stefan Böhringer, Yii-Der I Chen, Anton J N de Craen, David R Crosslin, Caroline E Dale, Zari Dastani, Felix R Day, Joris Deelen, Graciela E Delgado, Ayse Demirkan, Francis M Finucane, Ian Ford, Melissa E Garcia, Christian Gieger, Stefan Gustafsson, Göran Hallmans, Susan E Hankinson, Aki S Havulinna, Christian Herder, Dena Hernandez, Andrew A Hicks, David J Hunter, Thomas Illig, Erik Ingelsson, Andreea Ioan-Facsinay, John-Olov Jansson, Nancy S Jenny, Marit E Jørgensen, Torben Jørgensen, Magnus Karlsson, Wolfgang Koenig, Peter Kraft, Joanneke Kwekkeboom, Tiina Laatikainen, Karl-Heinz Ladwig, Charles A LeDuc, Gordon Lowe, Yingchang Lu, Pedro Marques-Vidal, Christa Meisinger, Cristina Menni, Andrew P Morris, Richard H Myers, Satu Männistö, Mike A Nalls, Lavinia Paternoster, Annette Peters, Aruna D Pradhan, Tuomo Rankinen, Laura J Rasmussen-Torvik, Wolfgang Rathmann, Treva K Rice, J Brent Richards, Paul M Ridker, Naveed Sattar, David B Savage (2016)Genome-wide meta-analysis uncovers novel loci influencing circulating leptin levels, In: Nature communications7(1)10494pp. 10494-14

Leptin is an adipocyte-secreted hormone, the circulating levels of which correlate closely with overall adiposity. Although rare mutations in the leptin (LEP) gene are well known to cause leptin deficiency and severe obesity, no common loci regulating circulating leptin levels have been uncovered. Therefore, we performed a genome-wide association study (GWAS) of circulating leptin levels from 32,161 individuals and followed up loci reaching P

BACKGROUND: The datafiles deposited here are products of the research project "Linking the gut microbiome to host DNA methylation by a discovery and replication epigenome-wide association study" Authors: Ayşe Demirkan1,2, Jenny van Dongen3,4, Casey T. Finnicum5, Harm-Jan Westra1, Soesma Jankipersadsing1, Gonneke Willemsen3,4, Richard G. Ijzerman6, Dorret I. Boomsma3,4, Erik A. Ehli5, Marc Jan Bonder1, Jingyuan Fu,1,7 Lude Franke1, Cisca Wijmenga1, Eco J.C. de Geus3,4, Alexander Kurilshikov1, Alexandra Zhernakova1 1 Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands 2 Section of Statistical Multi-omics, Department of Clinical and Experimental Medicine, School of Biosciences and Medicine & People-Centered AI institute University of Surrey, Guildford, United Kingdom 3 Biological Psychology, Vrije Universiteit, Amsterdam, the Netherlands 4 Amsterdam Public Health Research Institute, Amsterdam, the Netherlands 5 Avera Institute of Human Genetics, Avera McKennan Hospital & University Health Center, Sioux Falls, SD, USA 6 Department of Endocrinology, Amsterdam University Medical Center, location VUMC, Amsterdam, the Netherlands 7 Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands Corresponding Authors: Alexandra Zhernakova; Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands Ayse Demirkan; Section of Statistical Multi-omics, Department of Clinical and Experimental Medicine, School of Biosciences and Medicine & People-Centered AI institute University of Surrey, Guildford, United Kingdom. FILES:  1-merged_lld16s.Rata: Epigenome-wide association of 16s microbial abundances in LifeLines-Deep (LLD, n = 616, 450k methylation array)  2-lld_mgs.Rdata: Epigenome-wide association ofshotgun metagenomic sequencing derived taxa relative abundances (n = 683, 450k methylation array)) 3-lld_mgs_pathways. Rdata: Epigenome-wide association ofshotgun metagenomic sequencing derived bacterial pathway relative abundances (n = 683, 450k methylation array) FUNDING: The Lifelines initiative has been made possible by subsidy from the Dutch Ministry of Health, Welfare and Sport, the Dutch Ministry of Economic Affairs, the University Medical Center Groningen (UMCG), Groningen University and the Provinces in the North of the Netherlands (Drenthe, Friesland, Groningen). The Netherlands Twin Register acknowledges funding from the Netherlands Organization for Scientific Research (NWO): (NWO 911–09–032; NWO 480-04-004; 480-15-001/674, NWO 916-130-82), Biobanking and Biomolecular Research Infrastructure (184.033.111),  and the BBRMI-NL-financed BIOS Consortium (NWO 184.021.007), NWO Large Scale infrastructures X-Omics (184.034.019), Genotype/phenotype database for behaviour genetic and genetic epidemiological studies (ZonMw Middelgroot 911-09-032); Netherlands Twin Registry Repository: researching the interplay between genome and environment (NWO-Groot 480-15-001/674); the Avera Institute, Sioux Falls (USA), the European Research Council (Genetics of Mental Illness 230374), the European Research Council (Genetics of Mental Illness 230374), and INRA-Pfizer. Pfizer provided support for data collection, but did not have any additional role in the study design, data analysis, decision to publish, or preparation of the manuscript.  

Beben Benyamin, Tonu Esko, Janina S. Ried, Aparna Radhakrishnan, Sita H. Vermeulen, Michela Traglia, Martin Goegele, Denise Anderson, Linda Broer, Clara Podmore, Jianan Luan, Zoltan Kutalik, Serena Sanna, Peter van der Meer, Toshiko Tanaka, Fudi Wang, Harm-Jan Westra, Lude Franke, Evelin Mihailov, Lili Milani, Jonas Haelldin, Juliane Winkelmann, Thomas Meitinger, Joachim Thiery, Annette Peters, Melanie Waldenberger, Augusto Rendon, Jennifer Jolley, Jennifer Sambrook, Lambertus A. Kiemeney, Fred C. Sweep, Cinzia F. Sala, Christine Schwienbacher, Irene Pichler, Jennie Hui, Ayse Demirkan, Aaron Isaacs, Najaf Amin, Maristella Steri, Gerard Waeber, Niek Verweij, Joseph E. Powell, Dale R. Nyholt, Andrew C. Heath, Pamela A. F. Madden, Peter M. Visscher, Margaret J. Wright, Grant W. Montgomery, Nicholas G. Martin, Dena Hernandez, Stefania Bandinelli, Pim van der Harst, Manuela Uda, Peter Vollenweider, Robert A. Scott, Claudia Langenberg, Nicholas J. Wareham, Cornelia van Duijn, John Beilby, Peter P. Pramstaller, Andrew A. Hicks, Willem H. Ouwehand, Konrad Oexle, Christian Gieger, Andres Metspalu, Clara Camaschella, Daniela Toniolo, Dorine W. Swinkels, John B. Whitfield (2015)Novel loci affecting iron homeostasis and their effects in individuals at risk for hemochromatosis (vol 5, 4926, 2014), In: Nature communications66542 Springer Nature
Adam E. Locke, Bratati Kahali, Sonja I. Berndt, Anne E. Justice, Tune H. Pers, Felix R. Day, Corey Powell, Sailaja Vedantam, Martin L. Buchkovich, Jian Yang, Damien C. Croteau-Chonka, Tonu Esko, Tove Fall, Teresa Ferreira, Stefan Gustafsson, Zoltan Kutalik, Jian'an Luan, Reedik Maegi, Joshua C. Randall, Thomas W. Winkler, Andrew R. Wood, Tsegaselassie Workalemahu, Jessica D. Faul, Jennifer A. Smith, Jing Hua Zhao, Wei Zhao, Jin Chen, Rudolf Fehrmann, Asa K. Hedman, Juha Karjalainen, Ellen M. Schmidt, Devin Absher, Najaf Amin, Denise Anderson, Marian Beekman, Jennifer L. Bolton, L. Bragg-Gresham, Steven Buyske, Ayse Demirkan, Guohong Deng, Georg B. Ehret, Bjarke Feenstra, Mary F. Feitosa, Krista Fischer, Anuj Goel, Jian Gong, Anne U. Jackson, Stavroula Kanoni, Marcus E. Kleber, Kati Kristiansson, Unhee Lim, Vaneet Lotay, Massimo Mangino, Irene Mateo Leach, Carolina Medina-Gomez, Sarah E. Medland, Michael A. Nalls, Cameron D. Palmer, Dorota Pasko, Sonali Pechlivanis, Marjolein J. Peters, Inga Prokopenko, Dmitry Shungin, Alena Stancakova, Rona J. Strawbridge, Yun Ju Sung, Toshiko Tanaka, Alexander Teumer, Stella Trompet, Sander W. van der Laan, Jessica van Settee, Jana V. Van Vliet-Ostaptchouk, Zhaoming Wang, Loic Yengo, Weihua Zhang, Aaron Isaacs, Eva Albrecht, Johan Arnlov, Gillian M. Arscott, Antony P. Attwood, Stefania Bandinelli, Amy Barrett, Isabelita N. Bas, Claire Bellis, Amanda J. Bennett, Christian Berne, Roza Blagieva, Matthias Blueher, Stefan Bohringer, Lori L. Bonnycastle, Yvonne Boettcher, Heather A. Boyd, Marcel Bruinenberg, Ida H. Caspersen, Yii-Der Ida Chen, Robert Clarke, E. Warwick Daw, Anton J. M. de Craen, Graciela Delgado, Maria Dimitriou (2015)Genetic studies of body mass index yield new insights for obesity biology, In: Nature (London)518(7538)197pp. 197-206 NATURE PORTFOLIO

Obesity is heritable and predisposes to many diseases. To understand the genetic basis of obesity better, here we conduct a genome-wide association study and Metabochip meta-analysis of body mass index (BMI), a measure commonly used to define obesity and assess adiposity, in upto 339,224 individuals. This analysis identifies 97 BMI-associated loci (P < 5 x 10(-8)), 56 of which are novel. Five loci demonstrate clear evidence of several independent association signals, and many loci have significant effects on other metabolic phenotypes. The 97 loci account for similar to 2.7% of BMI variation, and genome-wide estimates suggest that common variation accounts for >20% of BMI variation. Pathway analyses provide strong support for a role of the central nervous systemin obesity susceptibility and implicate new genes and pathways, including those related to synaptic function, glutamate signalling, insulin secretion/action, energy metabolism, lipid biology and adipogenesis.

Harish Dharuri, Aye Demirkan, Jan Bert van Klinken, Dennis Owen Mook-Kanamori, Cornelia M. van Duijn, Peter A. C. 't Hoen, Ko Willems van Dijk (2014)Genetics of the human metabolome, what is next?, In: Biochimica et biophysica acta. Molecular basis of disease1842(10)pp. 1923-1931 Elsevier

Increases in throughput and decreases in costs have facilitated large scale metabolomics studies, the simultaneous measurement of large numbers of biochemical components in biological samples. Initial large scale studies focused on biomarker discovery for disease or disease progression and helped to understand biochemical pathways underlying disease. The first population-based studies that combined metabolomics and genome wide association studies (mGWAS) have increased our understanding of the (genetic) regulation of biochemical conversions. Measurements of metabolites as intermediate phenotypes are a potentially very powerful approach to uncover how genetic variation affects disease susceptibility and progression. However, we still face many hurdles in the interpretation of mGWAS data. Due to the composite nature of many metabolites, single enzymes may affect the levels of multiple metabolites and, conversely, levels of single metabolites may be affected by multiple enzymes. Here, we will provide a global review of the current status of mGWAS. We will specifically discuss the application of prior biological knowledge present in databases to the interpretation of mGWAS results and discuss the potential of mathematical models. As the technology continuously improves to detect metabolites and to measure genetic variation, it is clear that comprehensive systems biology based approaches are required to further our insight in the association between genes, metabolites and disease. This article is part of a Special Issue entitled: From Genome to Function. (C) 2014 Elsevier B.V. All rights reserved.

Karin Hek, Ayse Demirkan, Jari Lahti, Antonio Terracciano, Alexander Teumer, Marilyn C. Cornelis, Najaf Amin, Erin Bakshis, Jens Baumert, Jingzhong Ding, Yongmei Liu, Kristin Marciante, Osorio Meirelles, Michael A. Nalls, Yan V. Sun, Nicole Vogelzangs, Lei Yu, Stefania Bandinelli, Emelia J. Benjamin, David A. Bennett, Dorret Boomsma, Alessandra Cannas, Laura H. Coker, Eco de Geus, Philip L. De Jager, Ana V. Diez-Roux, Shaun Purcell, Frank B. Hu, Eric B. Rimm, David J. Hunter, Majken K. Jensen, Gary Curhan, Kenneth Rice, Alan D. Penman, Jerome I. Rotter, Nona Sotoodehnia, Rebecca Emeny, Johan G. Eriksson, Denis A. Evans, Luigi Ferrucci, Myriam Fornage, Vilmundur Gudnason, Albert Hofman, Thomas Illig, Sharon Kardia, Margaret Kelly-Hayes, Karestan Koenen, Peter Kraft, Maris Kuningas, Joseph M. Massaro, David Melzer, Antonella Mulas, Cornelis L. Mulder, Anna Murray, Ben A. Oostra, Aarno Palotie, Brenda Penninx, Astrid Petersmann, Luke C. Pilling, Bruce Psaty, Rajesh Rawal, Eric M. Reiman, Andrea Schulz, Joshua M. Shulman, Andrew B. Singleton, Albert V. Smith, Angelina R. Sutin, André G. Uitterlinden, Henry Völzke, Elisabeth Widen, Kristine Yaffe, Alan B. Zonderman, Francesco Cucca, Tamara Harris, Karl-Heinz Ladwig, David J. Llewellyn, Katri Räikkönen, Toshiko Tanaka, Cornelia M. van Duijn, Hans J. Grabe, Lenore J. Launer, Kathryn L. Lunetta, Thomas H. Mosley, Anne B. Newman, Henning Tiemeier, Joanne Murabito (2013)A Genome-Wide Association Study of Depressive Symptoms, In: Biological psychiatry (1969)73(7)667pp. 667-678 Elsevier Inc

Depression is a heritable trait that exists on a continuum of varying severity and duration. Yet, the search for genetic variants associated with depression has had few successes. We exploit the entire continuum of depression to find common variants for depressive symptoms. In this genome-wide association study, we combined the results of 17 population-based studies assessing depressive symptoms with the Center for Epidemiological Studies Depression Scale. Replication of the independent top hits (p

Nese Direk, Stephanie Williams, Jennifer A. Smith, Stephan Ripke, Tracy Air, Azmeraw T. Amare, Najaf Amin, Bernhard T. Baune, David A. Bennett, Douglas H.R. Blackwood, Dorret Boomsma, Gerome Breen, Henriette N. Buttenschøn, Enda M. Byrne, Anders D. Børglum, Enrique Castelao, Sven Cichon, Toni-Kim Clarke, Marilyn C. Cornelis, Udo Dannlowski, Philip L. De Jager, Ayse Demirkan, Enrico Domenici, Cornelia M. van Duijn, Erin C. Dunn, Johan G. Eriksson, Tonu Esko, Jessica D. Faul, Luigi Ferrucci, Myriam Fornage, Eco de Geus, Michael Gill, Scott D. Gordon, Hans Jörgen Grabe, Gerard van Grootheest, Steven P. Hamilton, Catharina A. Hartman, Andrew C. Heath, Karin Hek, Albert Hofman, Georg Homuth, Carsten Horn, Jouke Jan Hottenga, Sharon L.R. Kardia, Stefan Kloiber, Karestan Koenen, Zoltán Kutalik, Karl-Heinz Ladwig, Jari Lahti, Douglas F. Levinson, Cathryn M. Lewis, Glyn Lewis, Qingqin S. Li, David J. Llewellyn, Susanne Lucae, Kathryn L. Lunetta, Donald J. MacIntyre, Pamela Madden, Nicholas G. Martin, Andrew M. McIntosh, Andres Metspalu, Yuri Milaneschi, Grant W. Montgomery, Ole Mors, Thomas H. Mosley, Joanne M. Murabito, Bertram Müller-Myhsok, Markus M. Nöthen, Dale R. Nyholt, Michael C. O’Donovan, Brenda W. Penninx, Michele L. Pergadia, Roy Perlis, James B. Potash, Martin Preisig, Shaun M. Purcell, Jorge A. Quiroz, Katri Räikkönen, John P. Rice, Marcella Rietschel, Margarita Rivera, Thomas G. Schulze, Jianxin Shi, Stanley Shyn, Grant C. Sinnamon, Johannes H. Smit, Jordan W. Smoller, Harold Snieder, Toshiko Tanaka, Katherine E. Tansey, Alexander Teumer, Rudolf Uher, Daniel Umbricht, Sandra Van der Auwera, Erin B. Ware, David R. Weir, Myrna M. Weissman, Gonneke Willemsen, Jingyun Yang, Wei Zhao, Henning Tiemeier, Patrick F. Sullivan (2017)An Analysis of Two Genome-wide Association Meta-analyses Identifies a New Locus for Broad Depression Phenotype, In: Biological psychiatry (1969)82(5)322pp. 322-329 Elsevier Inc

The genetics of depression has been explored in genome-wide association studies that focused on either major depressive disorder or depressive symptoms with mostly negative findings. A broad depression phenotype including both phenotypes has not been tested previously using a genome-wide association approach. We aimed to identify genetic polymorphisms significantly associated with a broad phenotype from depressive symptoms to major depressive disorder. We analyzed two prior studies of 70,017 participants of European ancestry from general and clinical populations in the discovery stage. We performed a replication meta-analysis of 28,328 participants. Single nucleotide polymorphism (SNP)-based heritability and genetic correlations were calculated using linkage disequilibrium score regression. Discovery and replication analyses were performed using a p-value-based meta-analysis. Lifetime major depressive disorder and depressive symptom scores were used as the outcome measures. The SNP-based heritability of major depressive disorder was 0.21 (SE = 0.02), the SNP-based heritability of depressive symptoms was 0.04 (SE = 0.01), and their genetic correlation was 1.001 (SE = 0.2). We found one genome-wide significant locus related to the broad depression phenotype (rs9825823, chromosome 3: 61,082,153, p = 8.2 × 10–9) located in an intron of the FHIT gene. We replicated this SNP in independent samples (p = .02) and the overall meta-analysis of the discovery and replication cohorts (1.0 × 10–9). This large study identified a new locus for depression. Our results support a continuum between depressive symptoms and major depressive disorder. A phenotypically more inclusive approach may help to achieve the large sample sizes needed to detect susceptibility loci for depression.

Tao Xu, Stefan Brandmaier, Ana C. Messias, Christian Herder, Harmen H. M. Draisma, Ayse Demirkan, Zhonghao Yu, Janina S. Ried, Toomas Haller, Margit Heier, Monica Campillos, Gisela Fobo, Renee Stark, Christina Holzapfel, Jonathan Adam, Shen Chi, Markus Rotter, Tommaso Panni, Anne S. Quante, Ying He, Cornelia Prehn, Werner Roemisch-Margl, Gabi Kastenmueller, Gonneke Willemsen, Rene Pool, Katarina Kasa, Ko Willems van Dijk, Thomas Hankemeier, Christa Meisinger, Barbara Thorand, Andreas Ruepp, Martin Hrabe de Angelis, Yixue Li, H. -Erich Wichmann, Bernd Stratmann, Konstantin Strauch, Andres Metspalu, Christian Gieger, Karsten Suhre, Jerzy Adamski, Thomas Illig, Wolfgang Rathmann, Michael Roden, Annette Peters, Cornelia M. van Duijn, Dorret I. Boomsma, Thomas Meitinger, Rui Wang-Sattler (2015)Effects of Metformin on Metabolite Profiles and LDL Cholesterol in Patients With Type 2 Diabetes, In: Diabetes care38(10)pp. 1858-1867 Amer Diabetes Assoc

OBJECTIVEMetformin is used as a first-line oral treatment for type 2 diabetes (T2D). However, the underlying mechanism is not fully understood. Here, we aimed to comprehensively investigate the pleiotropic effects of metformin.RESEARCH DESIGN AND METHODSWe analyzed both metabolomic and genomic data of the population-based KORA cohort. To evaluate the effect of metformin treatment on metabolite concentrations, we quantified 131 metabolites in fasting serum samples and used multivariable linear regression models in three independent cross-sectional studies (n = 151 patients with T2D treated with metformin [mt-T2D]). Additionally, we used linear mixed-effect models to study the longitudinal KORA samples (n = 912) and performed mediation analyses to investigate the effects of metformin intake on blood lipid profiles. We combined genotyping data with the identified metformin-associated metabolites in KORA individuals (n = 1,809) and explored the underlying pathways.RESULTSWe found significantly lower (P < 5.0E-06) concentrations of three metabolites (acyl-alkyl phosphatidylcholines [PCs]) when comparing mt-T2D with four control groups who were not using glucose-lowering oral medication. These findings were controlled for conventional risk factors of T2D and replicated in two independent studies. Furthermore, we observed that the levels of these metabolites decreased significantly in patients after they started metformin treatment during 7 years' follow-up. The reduction of these metabolites was also associated with a lowered blood level of LDL cholesterol (LDL-C). Variations of these three metabolites were significantly associated with 17 genes (including FADS1 and FADS2) and controlled by AMPK, a metformin target.CONCLUSIONSOur results indicate that metformin intake activates AMPK and consequently suppresses FADS, which leads to reduced levels of the three acyl-alkyl PCs and LDL-C. Our findings suggest potential beneficial effects of metformin in the prevention of cardiovascular disease.

B Vural, A Demirkan, E Ugurel, Z KALAYLIOGLU-WHEELER, B. A Esen, A. O Gure, A Gül, U Ozbek (2009)Seroreactivity against PTEN-induced putative kinase 1 (PINK1) in Turkish patients with Behçet's disease, In: Clinical and experimental rheumatology (Testo stampato)27(2)pp. S67-S72 Clinical and Experimental Rheumatology
Paolo Zanoni, Sumeet A. Khetarpal, Daniel B. Larach, William F. Hancock-Cerutti, John S. Millar, Marina Cuchel, Stephanie DerOhannessian, Anatol Kontush, Praveen Surendran, Danish Saleheen, Stella Trompet, J. Wouter Jukema, Anton De Craen, Panos Deloukas, Naveed Sattar, Ian Ford, Chris Packard, Abdullah al Shafi Majumder, Dewan S. Alam, Emanuele Di Angelantonio, Goncalo Abecasis, Rajiv Chowdhury, Jeanette Erdmann, Borge G. Nordestgaard, Sune F. Nielsen, Anne Tybjaerg-Hansen, Ruth Frikke Schmidt, Kari Kuulasmaa, Dajiang J. Liu, Markus Perola, Stefan Blankenberg, Veikko Salomaa, Satu Mannisto, Philippe Amouyel, Dominique Arveiler, Jean Ferrieres, Martina Muller-Nurasyid, Marco Ferrario, Frank Kee, Cristen J. Willer, Nilesh Samani, Heribert Schunkert, Adam S. Butterworth, Joanna M. M. Howson, Gina M. Peloso, Nathan O. Stitziel, John Danesh, Sekar Kathiresan, Daniel J. Rader, Ayse Demirkan (2016)Rare variant in scavenger receptor BI raises HDL cholesterol and increases risk of coronary heart disease, In: Science (American Association for the Advancement of Science)351(6278)1166pp. 1166-1171 Amer Assoc Advancement Science

Scavenger receptor BI (SR-BI) is the major receptor for high-density lipoprotein (HDL) cholesterol (HDL-C). In humans, high amounts of HDL-C in plasma are associated with a lower risk of coronary heart disease (CHD). Mice that have depleted Scarb1 (SR-BI knockout mice) have markedly elevated HDL-C levels but, paradoxically, increased atherosclerosis. The impact of SR-BI on HDL metabolism and CHD risk in humans remains unclear. Through targeted sequencing of coding regions of lipid-modifying genes in 328 individuals with extremely high plasma HDL-C levels, we identified a homozygote for a loss-of-function variant, in which leucine replaces proline 376 (P376L), in SCARB1, the gene encoding SR-BI. The P376L variant impairs posttranslational processing of SR-BI and abrogates selective HDL cholesterol uptake in transfected cells, in hepatocyte-like cells derived from induced pluripotent stem cells from the homozygous subject, and in mice. Large population-based studies revealed that subjects who are heterozygous carriers of the P376L variant have significantly increased levels of plasma HDL-C. P376L carriers have a profound HDL-related phenotype and an increased risk of CHD (odds ratio = 1.79, which is statistically significant).

Sven J. van der Lee, Charlotte E. Teunissen, Rene Pool, Martin J. Shipley, Alexander Teumer, Vincent Chouraki, Debora Melo van Lent, Juho Tynkkynen, Krista Fischer, Jussi Hernesniemi, Toomas Haller, Archana Singh-Manoux, Aswin Verhoeven, Gonneke Willemsen, Francisca A. de Leeuw, Holger Wagner, Jenny van Dongen, Johannes Hertel, Kathrin Budde, Ko Willems van Dijk, Leonie Weinhold, M. Arfan Ikram, Maik Pietzner, Markus Perola, Michael Wagner, Nele Friedrich, P. Eline Slagboom, Philip Scheltens, Qiong Yang, Robert E. Gertzen, Sarah Egert, Shuo Li, Thomas Hankemeier, Catharina E. M. van Beijsterveldt, Ramachandran S. Vasan, Wolfgang Maier, Carel F. W. Peeters, Hans Joergen Grabe, Alfredo Ramirez, Sudha Seshadri, Andres Metspalu, Mika Kivimaki, Veikko Salomaa, Ayse Demirkan, Dorret I. Boomsma, Wiesje M. van der Flier, Najaf Amin, Cornelia M. van Duijn (2018)Circulating metabolites and general cognitive ability and dementia: Evidence from 11 cohort studies, In: Alzheimer's & dementia14(6)pp. 707-722 Wiley

Introduction: Identifying circulating metabolites that are associated with cognition and dementia may improve our understanding of the pathogenesis of dementia and provide crucial readouts for preventive and therapeutic interventions. Methods: We studied 299 metabolites in relation to cognition (general cognitive ability) in two discovery cohorts (N total = 5658). Metabolites significantly associated with cognition after adjusting for multiple testing were replicated in four independent cohorts (N total = 6652), and the associations with dementia and Alzheimer's disease (N = 25,872) and lifestyle factors (N = 5168) were examined. Results: We discovered and replicated 15 metabolites associated with cognition including subfractions of high-density lipoprotein, docosahexaenoic acid, ornithine, glutamine, and glycoprotein acetyls. These associations were independent of classical risk factors including high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, triglycerides, glucose, and apolipoprotein E (APOE) genotypes. Six of the cognition-associated metabolites were related to the risk of dementia and lifestyle factors. Discussion: Circulating metabolites were consistently associated with cognition, dementia, and lifestyle factors, opening new avenues for prevention of cognitive decline and dementia. (C) 2018 The Authors. Published by Elsevier Inc. on behalf of the Alzheimer's Association.

Adrienne Tin, Jonathan Marten, Victoria L Halperin Kuhns, Yong Li, Matthias Wuttke, Holger Kirsten, Karsten B Sieber, Chengxiang Qiu, Mathias Gorski, Zhi Yu, Ayush Giri, Gardar Sveinbjornsson, Man Li, Audrey Y Chu, Anselm Hoppmann, Luke J O'Connor, Bram Prins, Teresa Nutile, Damia Noce, Masato Akiyama, Massimiliano Cocca, Sahar Ghasemi, Peter J van der Most, Katrin Horn, Yizhe Xu, Christian Fuchsberger, Sanaz Sedaghat, Saima Afaq, Najaf Amin, Johan Ärnlöv, Stephan J L Bakker, Nisha Bansal, Daniela Baptista, Sven Bergmann, Mary L Biggs, Ginevra Biino, Eric Boerwinkle, Erwin P Bottinger, Thibaud S Boutin, Marco Brumat, Ralph Burkhardt, Eric Campana, Archie Campbell, Harry Campbell, Robert J Carroll, Eulalia Catamo, John C Chambers, Marina Ciullo, Maria Pina Concas, Josef Coresh, Tanguy Corre, Daniele Cusi, Sala Cinzia Felicita, Martin H de Borst, Alessandro De Grandi, Renée de Mutsert, Aiko P J de Vries, Graciela Delgado, Ayşe Demirkan, Olivier Devuyst, Katalin Dittrich, Kai-Uwe Eckardt, Georg Ehret, Karlhans Endlich, Michele K Evans, Ron T Gansevoort, Paolo Gasparini, Vilmantas Giedraitis, Christian Gieger, Giorgia Girotto, Martin Gögele, Scott D Gordon, Daniel F Gudbjartsson, Vilmundur Gudnason, Toomas Haller, Pavel Hamet, Tamara B Harris, Caroline Hayward, Andrew A Hicks, Edith Hofer, Hilma Holm, Wei Huang, Nina Hutri-Kähönen, Shih-Jen Hwang, M Arfan Ikram, Raychel M Lewis, Erik Ingelsson, Johanna Jakobsdottir, Ingileif Jonsdottir, Helgi Jonsson, Peter K Joshi, Navya Shilpa Josyula, Bettina Jung, Mika Kähönen, Yoichiro Kamatani, Masahiro Kanai, Shona M Kerr, Wieland Kiess, Marcus E Kleber, Wolfgang Koenig (2019)Target genes, variants, tissues and transcriptional pathways influencing human serum urate levels, In: Nature genetics51(10)1459pp. 1459-1474

Elevated serum urate levels cause gout and correlate with cardiometabolic diseases via poorly understood mechanisms. We performed a trans-ancestry genome-wide association study of serum urate in 457,690 individuals, identifying 183 loci (147 previously unknown) that improve the prediction of gout in an independent cohort of 334,880 individuals. Serum urate showed significant genetic correlations with many cardiometabolic traits, with genetic causality analyses supporting a substantial role for pleiotropy. Enrichment analysis, fine-mapping of urate-associated loci and colocalization with gene expression in 47 tissues implicated the kidney and liver as the main target organs and prioritized potentially causal genes and variants, including the transcriptional master regulators in the liver and kidney, HNF1A and HNF4A. Experimental validation showed that HNF4A transactivated the promoter of ABCG2, encoding a major urate transporter, in kidney cells, and that HNF4A p.Thr139Ile is a functional variant. Transcriptional coregulation within and across organs may be a general mechanism underlying the observed pleiotropy between urate and cardiometabolic traits.

F. Atalar, B. Vural, C. Ciftci, A. Demirkan, G. Akan, B. Susleyici-Duman, D. Gunay, B. Akpinar, E. Sagbas, U. Ozbek, A. S. Buyukdevrim (2012)11 beta-hydroxysteroid dehydrogenase type 1 gene expression is increased in ascending aorta tissue of metabolic syndrome patients with coronary artery disease, In: Genetics and molecular research11(3)pp. 3122-3132 Funpec-Editora

11 beta-hydroxysteroid dehydrogenase type 1 (11 beta-HSD-1) activity and mRNA levels are increased in visceral and subcutaneous adipose tissues of metabolic syndrome subjects. We analyzed 11 beta-HSD-1 expression in human epicardial adipose (EA) and ascending aorta (AA) tissues of metabolic syndrome patients and examined their contribution to the development of coronary atherosclerosis. The 11 beta-HSD-1 expression was evaluated by qRT-PCR in EA and AA tissues of 20 metabolic syndrome patients with coronary artery disease (metabolic syndrome group) and 10 non-metabolic syndrome patients without coronary artery disease (controls). 11 beta-HSD-1 expression was increased in EA and AA tissues of the metabolic syndrome group (4.1-and 5.5-fold, respectively). A significant positive correlation was found between 11 beta-HSD-1 expression in EA tissue and waist hip ratio and 11 beta-HSD-1 expression in AA tissue and body mass index, while a negative correlation was found between 11 beta-HSD-1 expression in EA tissue and HDL. Expression of CD68, a macrophage marker, was significantly increased in both tissues of the metabolic syndrome group; it was 2-fold higher in AA tissue compared to EA tissue in the metabolic syndrome group. Our findings of increased expression of 11 beta-HSD-1 and CD68 in AA tissue of the metabolic syndrome group lead us to suggest that they contribute to coronary atherosclerosis in metabolic syndrome. This positive correlation between obesity markers and 11 beta-HSD-1 in AA and EA tissues strengthens the evidence that 11 beta-HSD-1 has a role in metabolic syndrome. To the best of our knowledge, this is the first report showing 11 beta-HSD-1 and CD68 expression in AA tissue of metabolic syndrome patients. We suggest that there is tissue-specific expression of 11 beta-HSD-1 in metabolic syndrome and associated cardiovascular disorders.

Thomas W. Winkler, Anne E. Justice, Mariaelisa Graff, Llilda Barata, Mary F. Feitosa, Su Chu, Jacek Czajkowski, Tonu Esko, Tove Fall, Tuomas O. Kilpelainen, Yingchang Lu, Reedik Magi, Evelin Mihailov, Tune H. Pers, Sina Rueger, Alexander Teumer, Georg B. Ehret, Teresa Ferreira, Nancy L. Heard-Costa, Juha Karjalainen, Vasiliki Lagou, Anubha Mahajan, Michael D. Neinast, Inga Prokopenko, Jeannette Simino, Tanya M. Teslovich, Rick Jansen, Harm-Jan Westra, Charles C. White, Devin Absher, Tarunveer S. Ahluwalia, Shafqat Ahmad, Eva Albrecht, Alexessander Couto Alves, Jennifer L. Bragg-Gresham, Anton J. M. de Craen, Joshua C. Bis, Amelie Bonnefond, Gabrielle Boucher, Gemma Cadby, Yu-Ching Cheng, Charleston W. K. Chiang, Graciela Delgado, Ayse Demirkan, Nicole Dueker, Niina Eklund, Gudny Eiriksdottir, Joel Eriksson, Bjarke Feenstra, Krista Fischer, Francesca Frau, Tessel E. Galesloot, Frank Geller, Anuj Goel, Mathias Gorski, Tanja B. Grammer, Stefan Gustafsson, Saskia Haitjema, Jouke-Jan Hottenga, Jennifer E. Huffman, Anne U. Jackson, Kevin B. Jacobs, Asa Johansson, Marika Kaakinen, Marcus E. Kleber, Jari Lahti, Irene Mateo Leach, Benjamin Lehne, Youfang Liu, Ken Sin Lo, Mattias Lorentzon, Jian'an Luan, Pamela A. F. Madden, Massimo Mangino, Barbara McKnight, Carolina Medina-Gomez, Keri L. Monda, May E. Montasser, Gabriele Muller, Martina Muller-Nurasyid, Ilja M. Nolte, Kalliope Panoutsopoulou, Laura Pascoe, Lavinia Paternoster, Nigel W. Rayner, Frida Renstrom, Federica Rizzi, Lynda M. Rose, Kathy A. Ryan, Perttu Salo, Serena Sanna, Hubert Scharnagl, Jianxin Shi, Albert Vernon Smith, Lorraine Southam, Alena Stancakova, Valgerdur Steinthorsdottir, Rona J. Strawbridge, Yun Ju Sung, Ioanna Tachmazidou (2016)The Influence of Age and Sex on Genetic Associations with Adult Body Size and Shape: A Large-Scale Genome-Wide Interaction Study (vol 11, e1005378, 2015), In: PLoS genetics12(6)1006166 Public Library Science
Matthias Wuttke, Yong Li, Man Li, Karsten B Sieber, Mary F Feitosa, Mathias Gorski, Adrienne Tin, Lihua Wang, Audrey Y Chu, Anselm Hoppmann, Holger Kirsten, Ayush Giri, Jin-Fang Chai, Gardar Sveinbjornsson, Bamidele O Tayo, Teresa Nutile, Christian Fuchsberger, Jonathan Marten, Massimiliano Cocca, Sahar Ghasemi, Yizhe Xu, Katrin Horn, Damia Noce, Peter J van der Most, Sanaz Sedaghat, Zhi Yu, Masato Akiyama, Saima Afaq, Tarunveer S Ahluwalia, Peter Almgren, Najaf Amin, Johan Ärnlöv, Stephan J L Bakker, Nisha Bansal, Daniela Baptista, Sven Bergmann, Mary L Biggs, Ginevra Biino, Michael Boehnke, Eric Boerwinkle, Mathilde Boissel, Erwin P Bottinger, Thibaud S Boutin, Hermann Brenner, Marco Brumat, Ralph Burkhardt, Adam S Butterworth, Eric Campana, Archie Campbell, Harry Campbell, Mickaël Canouil, Robert J Carroll, Eulalia Catamo, John C Chambers, Miao-Ling Chee, Miao-Li Chee, Xu Chen, Ching-Yu Cheng, Yurong Cheng, Kaare Christensen, Renata Cifkova, Marina Ciullo, Maria Pina Concas, James P Cook, Josef Coresh, Tanguy Corre, Cinzia Felicita Sala, Daniele Cusi, John Danesh, E Warwick Daw, Martin H de Borst, Alessandro De Grandi, Renée de Mutsert, Aiko P J de Vries, Frauke Degenhardt, Graciela Delgado, Ayse Demirkan, Emanuele Di Angelantonio, Katalin Dittrich, Jasmin Divers, Rajkumar Dorajoo, Kai-Uwe Eckardt, Georg Ehret, Paul Elliott, Karlhans Endlich, Michele K Evans, Janine F Felix, Valencia Hui Xian Foo, Oscar H Franco, Andre Franke, Barry I Freedman, Sandra Freitag-Wolf, Yechiel Friedlander, Philippe Froguel, Ron T Gansevoort, He Gao, Paolo Gasparini, J Michael Gaziano, Vilmantas Giedraitis, Christian Gieger (2019)A catalog of genetic loci associated with kidney function from analyses of a million individuals, In: Nature genetics51(6)957pp. 957-972

Chronic kidney disease (CKD) is responsible for a public health burden with multi-systemic complications. Through trans-ancestry meta-analysis of genome-wide association studies of estimated glomerular filtration rate (eGFR) and independent replication (n = 1,046,070), we identified 264 associated loci (166 new). Of these, 147 were likely to be relevant for kidney function on the basis of associations with the alternative kidney function marker blood urea nitrogen (n = 416,178). Pathway and enrichment analyses, including mouse models with renal phenotypes, support the kidney as the main target organ. A genetic risk score for lower eGFR was associated with clinically diagnosed CKD in 452,264 independent individuals. Colocalization analyses of associations with eGFR among 783,978 European-ancestry individuals and gene expression across 46 human tissues, including tubulo-interstitial and glomerular kidney compartments, identified 17 genes differentially expressed in kidney. Fine-mapping highlighted missense driver variants in 11 genes and kidney-specific regulatory variants. These results provide a comprehensive priority list of molecular targets for translational research.

Saqib Hassan, Marika Kaakinen, Harmen Draisma, Mohd Ashraf Ganie, Zhanna Balkhiyarova, Paris Vogazianos, Christos Shammas, Joseph Selvin, Athos Antoniades, Ayse Demirkan, Inga Prokopenko Bifidobacterium is enriched in gut microbiome of Kashmiri women with polycystic ovary syndrome, In: bioRxiv Cold Spring Harbor Laboratory Press

Polycystic ovary syndrome (PCOS) is a common endocrine condition in women of reproductive age understudied in non-European populations. In India, PCOS affects the life of up to 19.4 million women of age 14-25 years. Gut microbiome composition might contribute to PCOS susceptibility. We profiled the microbiome in DNA isolated from faecal samples by 16S rRNA sequencing in 19/20 women with/without PCOS from Kashmir, India. We assigned genera to sequenced species with an average 121k reads depth and included bacteria detected in at least 1/3 of the subjects or with average relative abundance ≥0.1%. We compared the relative abundances of 40/58 operational taxonomic units in family/genus level between cases and controls, and in relation to 33 hormonal and metabolic factors, by multivariate analyses adjusted for confounders, and corrected for multiple testing. Seven genera were significantly enriched in PCOS cases: Sarcina, Alkalibacterium and Megasphaera, and previously reported for PCOS Bifidobacterium, Collinsella, Paraprevotella and Lactobacillus. We identified significantly increased relative abundance of Bifidobacteriaceae (median 6.07% vs. 2.77%) and Aerococcaceae (0.03% vs. 0.004%), whereas we detected lower relative abundance Peptococcaceae (0.16% vs. 0.25%) in PCOS cases. For the first time, we identified a significant direct association between butyrate producing Eubacterium and follicle-stimulating hormone levels. We observed increased relative abundance of Collinsella and Paraprevotella with higher fasting blood glucose levels, and Paraprevotella and Alkalibacterium with larger hip and waist circumference, and weight. We show a relationship between gut microbiome composition and PCOS linking it to specific reproductive health metabolic and hormonal predictors in Indian women.

Mariaelisa Graff, Robert A. Scott, Anne E. Justice, Kristin L. Young, Mary F. Feitosa, Llilda Barata, Thomas W. Winkler, Audrey Y. Chu, Anubha Mahajan, David Hadley, Luting Xue, Tsegaselassie Workalemahu, Nancy L. Heard-Costa, Marcel den Hoed, Tarunveer S. Ahluwalia, Qibin Qi, Julius S. Ngwa, Frida Renstrom, Lydia Quaye, John D. Eicher, James E. Hayes, Marilyn Cornelis, Zoltan Kutalik, Elise Lim, Jian'an Luan, Jennifer E. Huffman, Weihua Zhang, Wei Zhao, Paula J. Griffin, Toomas Haller, Shafqat Ahmad, Pedro M. Marques-Vidal, Stephanie Bien, Loic Yengo, Alexander Teumer, Albert Vernon Smith, Meena Kumari, Marie Neergaard Harder, Johanne Marie Justesen, Marcus E. Kleber, Mette Hollensted, Kurt Lohman, Natalia V. Rivera, John B. Whitfield, Jing Hua Zhao, Heather M. Stringham, Leo-Pekka Lyytikainen, Charlotte Huppertz, Gonneke Willemsen, Wouter J. Peyrot, Ying Wu, Kati Kristiansson, Ayse Demirkan, Myriam Fornage, Maija Hassinen, Lawrence F. Bielak, Gemma Cadby, Toshiko Tanaka, Reedlk Magl, Peter J. Van der Most, Anne U. Jackson, Jennifer L. Bragg-Gresham, Veronique Vitart, Jonathan Marten, Pau Navarro, Claire Bellis, Dorota Pasko, Asa Johansson, Soren Snitker, Yu-Ching Cheng, Joel Eriksson, Unhee Lim, Mette Aadahl, Linda S. Adair, Najaf Amin, Beverley Balkau, Juha Auvinen, John Beilby, Richard N. Bergman, Sven Bergmann, Alain G. Bertoni, John Blangero, Amelle Bonnefond, Lori L. Bonnycastle, Judith B. Borja, Soren Brage, Fabio Busonero, Steve Buyske, Harry Campbell, Peter S. Chines, Francis S. Collins, Tanguy Corre, George Davey Smith, Graciela E. Delgado, Nicole Dueker, Marcus Doerr, Tapani Ebeling, Gudny Eiriksdottir, Tonu Esko, Jessica D. Faul, Inga Prokopenko (2017)Genome-wide physical activity interactions in adiposity. A meta-analysis of 200,452 adults, In: PLoS genetics13(4)1006528pp. e1006528-e1006528 Public Library Science

Physical activity (PA) may modify the genetic effects that give rise to increased risk of obesity. To identify adiposity loci whose effects are modified by PA, we performed genome-wide interaction meta-analyses of BMI and BMI-adjusted waist circumference and waist-hip ratio from up to 200,452 adults of European (n = 180,423) or other ancestry (n = 20,029). We standardized PA by categorizing it into a dichotomous variable where, on average, 23% of participants were categorized as inactive and 77% as physically active. While we replicate the interaction with PA for the strongest known obesity-risk locus in the FTO gene, of which the effect is attenuated by similar to 30% in physically active individuals compared to inactive individuals, we do not identify additional loci that are sensitive to PA. In additional genome-wide meta-analyses adjusting for PA and interaction with PA, we identify 11 novel adiposity loci, suggesting that accounting for PA or other environmental factors that contribute to variation in adiposity may facilitate gene discovery.

A. Demirkan, J. Lahti, N. Direk, A. Viktorin, K. L. Lunetta, A. Terracciano, M. A. Nalls, T. Tanaka, K. Hek, M. Fornage, J. Wellmann, M. C. Cornelis, H. M. Ollila, L. Yu, J. A. Smith, L. C. Pilling, A. Isaacs, A. Palotie, W. V. Zhuang, A. Zonderman, J. D. Faul, A. Sutin, O. Meirelles, A. Mulas, A. Hofman, A. Uitterlinden, F. Rivadeneira, M. Perola, W. Zhao, V. Salomaa, K. Yaffe, A. I. Luik, Y. Liu, J. Ding, P. Lichtenstein, M. Landen, E. Widen, D. R. Weir, D. J. Llewellyn, A. Murray, S. L. R. Kardia, J. G. Eriksson, K. Koenen, P. K. E. Magnusson, L. Ferrucci, T. H. Mosley, F. Cucca, B. A. Oostra, D. A. Bennett, T. Paunio, K. Berger, T. B. Harris, N. L. Pedersen, J. M. Murabito, H. Tiemeier, C. M. van Duijn, K. Raeikkoenen (2016)Somatic, positive and negative domains of the Center for Epidemiological Studies Depression (CES-D) scale: a meta-analysis of genome-wide association studies, In: Psychological medicine46(8)1613pp. 1613-1623 Cambridge Univ Press

Background. Major depressive disorder (MDD) is moderately heritable, however genome-wide association studies (GWAS) for MDD, as well as for related continuous outcomes, have not shown consistent results. Attempts to elucidate the genetic basis of MDD may be hindered by heterogeneity in diagnosis. The Center for Epidemiological Studies Depression (CES-D) scale provides a widely used tool for measuring depressive symptoms clustered in four different domains which can be combined together into a total score but also can be analysed as separate symptom domains. Method. We performed a meta-analysis of GWAS of the CES-D symptom clusters. We recruited 12 cohorts with the 20-or 10-item CES-D scale (32 528 persons). Results. One single nucleotide polymorphism (SNP), rs713224, located near the brain-expressed melatonin receptor (MTNR1A) gene, was associated with the somatic complaints domain of depression symptoms, with borderline genome-wide significance (p(discovery) = 3.82 x 10(-8)). The SNP was analysed in an additional five cohorts comprising the replication sample (6813 persons). However, the association was not consistent among the replication sample (p(discovery+replication) = 1.10 x 10(-6)) with evidence of heterogeneity. Conclusions. Despite the effort to harmonize the phenotypes across cohorts and participants, our study is still underpowered to detect consistent association for depression, even by means of symptom classification. On the contrary, the SNP-based heritability and co-heritability estimation results suggest that a very minor part of the variation could be captured by GWAS, explaining the reason of sparse findings.

Francisca A. de Leeuw, Carel F.W. Peeters, Maartje I. Kester, Amy C. Harms, Eduard A. Struys, Thomas Hankemeier, Herman W.T. van Vlijmen, Sven J. van der Lee, Cornelia M. van Duijn, Philip Scheltens, Ayşe Demirkan, Mark A. van de Wiel, Wiesje M. van der Flier, Charlotte E. Teunissen (2017)Blood-based metabolic signatures in Alzheimer's disease, In: Alzheimer's & dementia : diagnosis, assessment & disease monitoring8(1)196pp. 196-207 Elsevier Inc

Identification of blood-based metabolic changes might provide early and easy-to-obtain biomarkers. We included 127 Alzheimer's disease (AD) patients and 121 control subjects with cerebrospinal fluid biomarker-confirmed diagnosis (cutoff tau/amyloid β peptide 42: 0.52). Mass spectrometry platforms determined the concentrations of 53 amine compounds, 22 organic acid compounds, 120 lipid compounds, and 40 oxidative stress compounds. Multiple signatures were assessed: differential expression (nested linear models), classification (logistic regression), and regulatory (network extraction). Twenty-six metabolites were differentially expressed. Metabolites improved the classification performance of clinical variables from 74% to 79%. Network models identified five hubs of metabolic dysregulation: tyrosine, glycylglycine, glutamine, lysophosphatic acid C18:2, and platelet-activating factor C16:0. The metabolite network for apolipoprotein E (APOE) ε4 negative AD patients was less cohesive compared with the network for APOE ε4 positive AD patients. Multiple signatures point to various promising peripheral markers for further validation. The network differences in AD patients according to APOE genotype may reflect different pathways to AD. •Multiple metabolic signatures point to peripheral AD markers for future validation.•AD may be described by changes in the metabolism of amines and oxidative stressors.•APOE ε4-driven AD and non- APOE ε4-driven AD represent different biochemical pathways.•Network analyses of metabolomics data enable the study of metabolic changes in AD.

Yingchang Lu, Felix R Day, Stefan Gustafsson, Martin L Buchkovich, Jianbo Na, Veronique Bataille, Diana L Cousminer, Zari Dastani, Alexander W Drong, Tõnu Esko, David M Evans, Mario Falchi, Mary F Feitosa, Teresa Ferreira, Åsa K Hedman, Robin Haring, Pirro G Hysi, Mark M Iles, Anne E Justice, Stavroula Kanoni, Vasiliki Lagou, Rui Li, Xin Li, Adam Locke, Chen Lu, Reedik Mägi, John R B Perry, Tune H Pers, Qibin Qi, Marianna Sanna, Ellen M Schmidt, William R Scott, Dmitry Shungin, Alexander Teumer, Anna A E Vinkhuyzen, Ryan W Walker, Harm-Jan Westra, Mingfeng Zhang, Weihua Zhang, Jing Hua Zhao, Zhihong Zhu, Uzma Afzal, Tarunveer Singh Ahluwalia, Stephan J L Bakker, Claire Bellis, Amélie Bonnefond, Katja Borodulin, Aron S Buchman, Tommy Cederholm, Audrey C Choh, Hyung Jin Choi, Joanne E Curran, Lisette C P G M de Groot, Philip L De Jager, Rosalie A M Dhonukshe-Rutten, Anke W Enneman, Elodie Eury, Daniel S Evans, Tom Forsen, Nele Friedrich, Frédéric Fumeron, Melissa E Garcia, Simone Gärtner, Bok-Ghee Han, Aki S Havulinna, Caroline Hayward, Dena Hernandez, Hans Hillege, Till Ittermann, Jack W Kent, Ivana Kolcic, Tiina Laatikainen, Jari Lahti, Irene Mateo Leach, Christine G Lee, Jong-Young Lee, Tian Liu, Youfang Liu, Stéphane Lobbens, Marie Loh, Leo-Pekka Lyytikäinen, Carolina Medina-Gomez, Karl Michaëlsson, Mike A Nalls, Carrie M Nielson, Laticia Oozageer, Laura Pascoe, Lavinia Paternoster, Ozren Polašek, Samuli Ripatti, Mark A Sarzynski, Chan Soo Shin, Nina Smolej Narančić, Dominik Spira, Priya Srikanth, Elisabeth Steinhagen-Thiessen, Yun Ju Sung, Karin M A Swart, Leena Taittonen, Toshiko Tanaka, Ayse Demirkan (2016)New loci for body fat percentage reveal link between adiposity and cardiometabolic disease risk, In: Nature communications7(1)10495pp. 10495-10495

To increase our understanding of the genetic basis of adiposity and its links to cardiometabolic disease risk, we conducted a genome-wide association meta-analysis of body fat percentage (BF%) in up to 100,716 individuals. Twelve loci reached genome-wide significance (P

Adrienne Tin, Yong Li, Jennifer A. Brody, Teresa Nutile, Audrey Y. Chu, Jennifer E. Huffman, Qiong Yang, Ming-Huei Chen, Cassianne Robinson-Cohen, Aurélien Macé, Jun Liu, Ayşe Demirkan, Rossella Sorice, Sanaz Sedaghat, Melody Swen, Bing Yu, Sahar Ghasemi, Alexanda Teumer, Peter Vollenweider, Marina Ciullo, Meng Li, André G. Uitterlinden, Robert Kraaij, Najaf Amin, Jeroen van Rooij, Zoltán Kutalik, Abbas Dehghan, Barbara McKnight, Cornelia M. van Duijn, Alanna Morrison, Bruce M. Psaty, Eric Boerwinkle, Caroline S. Fox, Owen M. Woodward, Anna Köttgen (2018)Large-scale whole-exome sequencing association studies identify rare functional variants influencing serum urate levels, In: Nature communications9(1)4228pp. 4228-11 Nature Publishing Group UK

Elevated serum urate levels can cause gout, an excruciating disease with suboptimal treatment. Previous GWAS identified common variants with modest effects on serum urate. Here we report large-scale whole-exome sequencing association studies of serum urate and kidney function among ≤19,517 European ancestry and African-American individuals. We identify aggregate associations of low-frequency damaging variants in the urate transporters SLC22A12 (URAT1; p  = 1.3 × 10 −56 ) and SLC2A9 ( p  = 4.5 × 10 −7 ). Gout risk in rare SLC22A12 variant carriers is halved (OR = 0.5, p  = 4.9 × 10 −3 ). Selected rare variants in SLC22A12 are validated in transport studies, confirming three as loss-of-function (R325W, R405C, and T467M) and illustrating the therapeutic potential of the new URAT1-blocker lesinurad. In SLC2A9 , mapping of rare variants of large effects onto the predicted protein structure reveals new residues that may affect urate binding. These findings provide new insights into the genetic architecture of serum urate, and highlight molecular targets in SLC22A12 and SLC2A9 for lowering serum urate and preventing gout. Elevated serum urate levels are a risk factor for gout. Here, Tin et al. perform whole-exome sequencing in 19,517 individuals and detect low-frequency genetic variants in urate transporter genes, SLC22A12 and SLC2A9 , associated with serum urate levels and confirm their damaging nature in vitro and in silico.

Pradeep Natarajan, Joshua C. Bis, Lawrence F. Bielak, Amanda J. Cox, Marcus Dorr, Mary F. Feitosa, Nora Franceschini, Xiuqing Guo, Shih-Jen Hwang, Aaron Isaacs, Min A. Jhun, Maryam Kavousi, Ruifang Li-Gao, Leo-Pekka Lyytikainen, Riccardo E. Marioni, Ulf Schminke, Nathan O. Stitziel, Hayato Tada, Jessica van Setten, Albert V. Smith, Dina Vojinovic, Lisa R. Yanek, Jie Yao, Laura M. Yerges-Armstrong, Najaf Amin, Usman Baber, Ingrid B. Borecki, J. Jeffrey Carr, Yii-Der Ida Chen, L. Adrienne Cupples, Pim A. de Jong, Harry de Koning, Bob D. de Vos, Ayse Demirkan, Valentin Fuster, Oscar H. Franco, Mark O. Goodarzi, Tamara B. Harris, Susan R. Heckbert, Gerardo Heiss, Udo Hoffmann, Albert Hofman, Ivana Isgum, J. Wouter Jukema, Mika Kahonen, Sharon L. R. Kardia, Brian G. Kral, Lenore J. Launer, Joe Massaro, Roxana Mehran, Braxton D. Mitchell, Thomas H. Mosley, Renee de Mutsert, Anne B. Newman, Khanh-dung Nguyen, Kari E. North, Jeffrey R. O'Connell, Matthijs Oudkerk, James S. Pankow, Gina M. Peloso, Wendy Post, Michael A. Province, Laura M. Raffield, Olli T. Raitakari, Dermot F. Reilly, Fernando Rivadeneira, Frits Rosendaal, Samantha Sartori, Kent D. Taylor, Alexander Teumer, Stella Trompet, Stephen T. Turner, Andre G. Uitterlinden, Dhananjay Vaidya, Aad van der Lugt, Uwe Volker, Joanna M. Wardlaw, Christina L. Wassel, Stefan Weiss, Mary K. Wojczynski, Diane M. Becker, Lewis C. Becker, Eric Boerwinkle, Donald W. Bowden, Ian J. Deary, Abbas Dehghan, Stephan B. Felix, Vilmundur Gudnason, Terho Lehtimaki, Rasika Mathias, Dennis O. Mook-Kanamori, Bruce M. Psaty, Daniel J. Rader, Jerome I. Rotter, James G. Wilson, Cornelia M. van Duijn, Henry Volzke, Sekar Kathiresan, Patricia A. Peyser, Christopher J. O'Donnell (2016)Multiethnic Exome-Wide Association Study of Subclinical Atherosclerosis, In: Circulation. Cardiovascular genetics9(6)511pp. 511-520 Lippincott Williams & Wilkins

Background-The burden of subclinical atherosclerosis in asymptomatic individuals is heritable and associated with elevated risk of developing clinical coronary heart disease. We sought to identify genetic variants in protein-coding regions associated with subclinical atherosclerosis and the risk of subsequent coronary heart disease. Methods and Results-We studied a total of 25 109 European ancestry and African ancestry participants with coronary artery calcification (CAC) measured by cardiac computed tomography and 52 869 participants with common carotid intima-media thickness measured by ultrasonography within the CHARGE Consortium (Cohorts for Heart and Aging Research in Genomic Epidemiology). Participants were genotyped for 247 870 DNA sequence variants (231 539 in exons) across the genome. A meta-analysis of exome-wide association studies was performed across cohorts for CAC and carotid intima-media thickness. APOB p.Arg3527Gln was associated with 4-fold excess CAC (P=3x10(-10)). The APOE epsilon 2 allele (p.Arg176Cys) was associated with both 22.3% reduced CAC (P=1x10(-12)) and 1.4% reduced carotid intima-media thickness (P=4x10(-14)) in carriers compared with noncarriers. In secondary analyses conditioning on low-density lipoprotein cholesterol concentration, the epsilon 2 protective association with CAC, although attenuated, remained strongly significant. Additionally, the presence of epsilon 2 was associated with reduced risk for coronary heart disease (odds ratio 0.77; P=1x10(-11)). Conclusions-Exome-wide association meta-analysis demonstrates that protein-coding variants in APOB and APOE associate with subclinical atherosclerosis. APOE epsilon 2 represents the first significant association for multiple subclinical atherosclerosis traits across multiple ethnicities, as well as clinical coronary heart disease.

Jun Liu, Jan Bert van Klinken, Sabina Semiz, Ko Willems van Dijk, Aswin Verhoeven, Thomas Hankemeier, Amy C. Harms, Eric Sijbrands, Nuala A. Sheehan, Cornelia M. van Duijn, Ayse Demirkan (2017)A Mendelian Randomization Study of Metabolite Profiles, Fasting Glucose, and Type 2 Diabetes, In: Diabetes (New York, N.Y.)66(11)pp. 2915-2926 Amer Diabetes Assoc

Mendelian randomization (MR) provides us the opportunity to investigate the causal paths of metabolites in type 2 diabetes and glucose homeostasis. We developed and tested an MR approach based on genetic risk scoring for plasma metabolite levels, utilizing a pathway-based sensitivity analysis to control for nonspecific effects. We focused on 124 circulating metabolites that correlate with fasting glucose in the Erasmus Rucphen Family (ERF) study (n = 2,564) and tested the possible causal effect of each metabolite with glucose and type 2 diabetes and vice versa. We detected 14 paths with potential causal effects by MR, following pathway-based sensitivity analysis. Our results suggest that elevated plasma triglycerides might be partially responsible for increased glucose levels and type 2 diabetes risk, which is consistent with previous reports. Additionally, elevated HDL components, i.e., small HDL triglycerides, might have a causal role of elevating glucose levels. In contrast, large (L) and extra large (XL) HDL lipid components, i.e., XL-HDL cholesterol, XL-HDL-free cholesterol, XL-HDL phospholipids, L-HDL cholesterol, and L-HDL-free cholesterol, as well as HDL cholesterol seem to be protective against increasing fasting glucose but not against type 2 diabetes. Finally, we demonstrate that genetic predisposition to type 2 diabetes associates with increased levels of alanine and decreased levels of phosphatidylcholine alkyl-acyl C42:5 and phosphatidylcholine alkyl-acyl C44:4. Our MR results provide novel insight into promising causal paths to and from glucose and type 2 diabetes and underline the value of additional information from high-resolution metabolomics over classic biochemistry.

Claudia T. Silva, Irina V. Zorkoltseva, Najaf Amin, Ayse Demirkan, Elisabeth M. van Leeuwen, Jan A. Kors, Marten van den Berg, Bruno H. Stricker, Andre G. Uitterlinden, Anatoly V. Kirichenko, Jacqueline C. M. Witteman, Rob Willemsen, Ben A. Oostra, Tatiana I. Axenovich, Cornelia M. van Duijn, Aaron Isaacs (2016)A Combined Linkage and Exome Sequencing Analysis for Electrocardiogram Parameters in the Erasmus Rucphen Family Study, In: Frontiers in genetics7190pp. 190-190 Frontiers Media Sa

Electrocardiogram (ECG) measurements play a key role in the diagnosis and prediction of cardiac arrhythmias and sudden cardiac death. ECG parameters, such as the PR, QRS, and QT intervals, are known to be heritable and genome-wide association studies of these phenotypes have been successful in identifying common variants; however, a large proportion of the genetic variability of these traits remains to be elucidated. The aim of this study was to discover loci potentially harboring rare variants utilizing variance component linkage analysis in 1547 individuals from a large family-based study, the Erasmus Rucphen Family Study (ERF). Linked regions were further explored using exome sequencing. Five suggestive linkage peaks were identified: two for QT interval (1q24, LOD = 2.63; 2q34, LOD = 2.05), one for QRS interval (1p35, LOD = 2.52) and two for PR interval (9p22, LOD = 2.20; 14q11, LOD = 2.29). Fine-mapping using exome sequence data identified a C > G missense variant (c.713C > G, p.Ser238Cys) in the FCRL2 gene associated with QT (rs74608430; P = 2.8 x 10(-4), minor allele frequency = 0.019). Heritability analysis demonstrated that the SNP explained 2.42% of the trait's genetic variability in ERF (P = 0.02). Pathway analysis suggested that the gene is involved in cytosolic Ca2+ levels (P = 3.3 x 10(-3)) and AMPK stimulated fatty acid oxidation in muscle (P = 4.1 x 10(-3)). Look-ups in bioinformatics resources showed that expression of FCRL2 is associated with ARHGAP24 and SETBP1 expression. This finding was not replicated in the Rotterdam study. Combining the bioinformatics information with the association and linkage analyses, FCRL2 emerges as a strong candidate gene for QT interval.

Wishal D. Ramdas, Najaf Amin, Leonieke M. E. van Koolwijk, A. Cecile J. W. Janssens, Ayse Demirkan, Paulus T. V. M. de Jong, Yurii S. Aulchenko, Roger C. W. Wolfs, Albert Hofman, Fernando Rivadeneira, Andre G. Uitterlinden, Ben A. Oostra, Hans G. Lemij, Caroline C. W. Klaver, Johannes R. Vingerling, Nomdo M. Jansonius, Cornelia M. van Duijn (2011)Genetic architecture of open angle glaucoma and related determinants, In: Journal of medical genetics48(3)190pp. 190-196 Bmj Publishing Group

Background Although the vertical cup-disc ratio (VCDR) and intraocular pressure (IOP) are important determinants of open angle glaucoma (OAG), it is unclear to what extent the genetic origin of these traits overlap with those of OAG. We evaluated whether the same genes that determine VCDR and IOP also predict OAG. Methods Genetic risk scores were constructed from single nucleotide polymorphisms (SNPs) using genome wide association data of 9326 participants from the Rotterdam Study cohorts (mean +/- SD age: 64.6 +/- 9.1 years). These risk scores were used to calculate the explained variance of VCDR and IOP in an independent cohort (Erasmus Rucphen Family study) consisting of 1646 participants (mean +/- SD age: 46.8 +/- 14.1 years) and the OAG risk in a subset of the Rotterdam Study cohorts. To evaluate false positive findings, we generated two new variables containing randomly sampled values to serve as a negative control. Results The explained variance of VCDR increased when increasing the number of SNPs included in the risk score, suggesting a polygenic model. We found no clear evidence for a similar model for IOP, suggesting that a small number of SNPs determine the susceptibility to IOP. The SNPs related to IOP in terms of p values contributed little to VCDR. The risk scores associated with VCDR were also associated significantly with OAG. This suggests a common polygenic background for VCDR and OAG Conclusions We found evidence for a polygenic model underlying one of the major traits of OAG, VCDR, and OAG itself. The IOP did not show any evidence for such a model.

Ayse Demirkan, Aaron Isaacs, Peter Ugocsai, Gerhard Liebisch, Maksim Struchalin, Igor Rudan, James F. Wilson, Peter P. Pramstaller, Ulf Gyllensten, Harry Campbell, Gerd Schmitz, Ben A. Oostra, Cornelia M. van Duijn (2013)Plasma phosphatidylcholine and sphingomyelin concentrations are associated with depression and anxiety symptoms in a Dutch family-based lipidomics study, In: Journal of psychiatric research47(3)357pp. 357-362 Elsevier

The central nervous system has the second highest concentration of lipids after adipose tissue. Alterations in neural membrane phospho- and sphingolipid composition can influence crucial intra- and intercellular signalling and alter the membrane's properties. Recently, the polyunsaturated fatty acids (PUFA) hypothesis for depression suggests that phospho- and sphingolipid metabolism includes potential pathways for the disease. In 742 people from a Dutch family-based study, we assessed the relationships between 148 different plasma phospho- and sphingolipid species and depression/anxiety symptoms as measured by the Hospital Anxiety and Depression Scales (HADS-A and HADS-D) and the Centre for Epidemiological Studies Depression Scale (CES-D). We observed significant differences in plasma sphingomyelins (SPM), particularly the SPM 23:1/SPM 16:0 ratio, which was inversely correlated with depressive symptom scores. We observed a similar trend for plasma phosphatidylcholines (PC), particularly the molar proportion of PC O 36:4 and its ratio to ceramide CER 20:0. Absolute levels of PC O 36:4 were also associated with depression symptoms in an independent replication. To our knowledge this is the first study on depressive symptoms that focuses on specific phospho- and sphingolipid molecules in plasma rather than total PUFA concentrations. The findings of this lipidomic study suggests that plasma sphingomyelins and ether phospholipids should be further studied for their potential as biomarkers and for a better understanding of the underlying mechanisms of this systemic disease. (C) 2012 Published by Elsevier Ltd.

Burcak Vural, Fatih Yakar, Duygu Derin, Pinar Saip, Aysun Yakar, Ayse Demirkan, Aydin Karabulut, Elif Ugurel, Naci Cine, Zeki Kilicaslan, Erdem Tuzun, Ugur Ozbek (2012)Evaluation of Glutathione S-Transferase P1 Polymorphisms (Ile105Val and Ala114Val) in Patients with Small Cell Lung Cancer, In: Genetic testing and molecular biomarkers16(7)701pp. 701-706 Mary Ann Liebert, Inc

Aims: Glutathione S-transferase P1 (GSTP1) plays an important role in cellular protection against oxidative stress and toxic chemicals. Polymorphisms within GSTP1 are associated with alterations in enzyme activity, which may lead to development of lung disease and cancer. In this study, we aimed to investigate the GSTP1 Ile105Val and Ala114Val polymorphisms in patients with small cell lung cancer (SCLC). Patients/Methods: GSTP1 Ile105Val polymorphism in exon 5 and GSTP1 Ala114Val polymorphism in exon 6 were determined by using polymerase chain reaction-restriction fragment length polymorphism techniques in 89 patients with SCLC and 108 control patients with chronic obstructive pulmonary disease (COPD). Genotype frequencies and cigarette smoking intensities were compared among SCLC and COPD patients. Results: There were significantly less SCLC patients with variant exon 6 genotypes than COPD patients (7.9% vs. 20.4%, p = 0.007), while the number of patients with variant exon 5 genotypes were comparable among groups. SCLC and COPD patients with variant exon 6 genotype showed trends toward exhibiting reduced cigarette consumption. Conclusions: The variant GSTP1 exon 6 genotype might be conferring protection against SCLC development. Whether this effect is associated with exposure to cigarette smoking needs to be clarified.

Dmitry Shungin, Thomas W. Winkler, Damien C. Croteau-Chonka, Teresa Ferreira, Adam E. Lockes, Reedik Maegi, Rona J. Strawbridge, Tune H. Pers, Krista Fischer, Anne E. Justice, Tsegaselassie Workalemahu, Joseph M. W. Wu, Martin L. Buchkovich, Nancy L. Heard-Costa, Tamara S. Roman, Alexander W. Drong, Ci Song, Stefan Gustafsson, Felix R. Day, Tonu Esko, Tove Fall, Zoltan Kutalik, Jian'an Luan, Joshua C. Randall, Andre Scherag, Sailaja Vedantam, Andrew R. Wood, Jin Chen, Rudolf Fehrmann, Juha Karjalainen, Bratati Kahali, Ching-Ti Liu, Ellen M. Schmidt, Devin Absher, Najaf Amin, Denise Anderson, Marian Beekman, Jennifer L. Bragg-Gresham, Steven Buyske, Ayse Demirkan, Georg B. Ehret, Mary F. Feitosa, Anuj Goel, Anne U. Jackson, Toby Johnson, Marcus E. Kleber, Kati Kristiansson, Massimo Mangino, Irene Mateo Leach, Carolina Medina-Gomez, Cameron D. Palmer, Dorota Pasko, Sonali Pechlivaniss, Marjolein J. Peters, Inga Prokopenko, Alena Stancakova, Yun Ju Sung, Toshiko Tanakam, Alexander Teumer, Jana V. Van Vliet-Ostaptchouk, Loic Yengo, Weihua Zhang, Eva Albrecht, Johan Arnlov, Gillian M. Arscott, Stefania Bandinelli, Amy Barrett, Claire Bellis, Amanda J. Bennett, Christian Berne, Matthias Blueher, Stefan Buhringer, Fabrice Bonnet, Yvonne Boettcher, Marcel Bruinenberg, Delia B. Carba, Ida H. Caspersen, Robert Clarke, E. Warwick Daw, Joris Deelen, Ewa Deelman, Graciela Delgado, Alex S. F. Doney, Niina Eklund, Michael R. Erdos, Karol Estrada, Elodie Eury, Nele Friedrichs, Melissa E. Garcia, Vilmantas Giedraitis, Bruna Gigante, Alan S. Go, Alain Golay, Harald Grallert, Tanja B. Grammer, Juergen Graessler, Jagvir Grewal, Christopher J. Groves, Toomas Haller, Goran Hallmans (2015)New genetic loci link adipose and insulin biology to body fat distribution, In: Nature (London)518(7538)187pp. 187-U378 NATURE PORTFOLIO

Body fat distribution is a heritable trait and a well-established predictor of adverse metabolic outcomes, independent of overall adiposity. To increase our understanding of the genetic basis of body fat distribution and its molecular links to cardiometabolic traits, here we conduct genome-wide association meta-analyses of traits related to waist and hip circumferences in up to 224,459 individuals. We identify 49 loci (33 new) associated with waist-to-hip ratio adjusted for body mass index (BMI), and an additional 19 loci newly associated with related waist and hip circumference measures (P < 5 x 10(-8)). In total, 20 of the 49 waist-to-hip ratio adjusted for BMI loci show significant sexual dimorphism, 19 of which display a stronger effect in women. The identified loci were enriched for genes expressed in adipose tissue and for putative regulatory elements in adipocytes. Pathway analyses implicated adipogenesis, angiogenesis, transcriptional regulation and insulin resistance as processes affecting fat distribution, providing insight into potential pathophysiological mechanisms.

Canbahar Sezgin, Muge Gunalp, Sinan Genc, Nurdan Acar, Evren Ustuner, Ahmet Burak Oguz, Ayca Koca Tanriverdi, Arda Demirkan, Onur Polat, Ayse Demirkan (2020)Diagnostic Value of Bedside Lung Ultrasonography in Pneumonia, In: Ultrasound in medicine & biology46(5)pp. 1189-1196 Elsevier Inc

Bedside lung ultrasonography (LUS) is a reliable method for the diagnosis of pneumonia. No common consensus exists in the literature regarding the effectiveness of LUS findings, except consolidation, for the diagnosis of pneumonia. The primary objective of this study was to investigate the effectiveness of LUS for the diagnosis of pneumonia, and the secondary objective was to investigate the use of LUS findings, except consolidation, for the diagnosis of pneumonia. A total of 127 patients with clinically suspected pneumonia were evaluated in the study. The sensitivity and specificity of LUS were 98.0% and 95.8%, respectively. In the cases where consolidation was not determined in LUS but B-3 line positivity or a diffuse interstitial pattern was present, the sensitivity and specificity were 93.3% and 88.2%, respectively. When consolidation was not observed in LUS, the presence of B-3 line positivity or diffuse interstitial pattern could be used for the diagnosis of pneumonia.

Henriet Springelkamp, Aniket Mishra, Pirro G. Hysi, Puya Gharahkhani, Rene Hoehn, Chiea-Chuen Khor, Jessica N. Cooke Bailey, Xiaoyan Luo, Wishal D. Ramdas, Eranga Vithana, Victor Koh, Seyhan Yazar, Liang Xu, Hannah Forward, Lisa S. Kearns, Najaf Amin, Adriana I. Iglesias, Kar-Seng Sim, Elisabeth M. van Leeuwen, Ayse Demirkan, Sven van der Lee, Seng-Chee Loon, Fernando Rivadeneira, Abhishek Nag, Paul G. Sanfilippo, Arne Schillert, Paulus T. V. M. de Jong, Ben A. Oostra, Andre G. Uitterlinden, Albert Hofman, Tiger Zhou, Kathryn P. Burdon, Timothy D. Spector, Karl J. Lackner, Seang-Mei Saw, Johannes R. Vingerling, Yik-Ying Teo, Louis R. Pasquale, Roger C. W. Wolfs, Hans G. Lemij, E-Shyong Tai, Jost B. Jonas, Ching-Yu Cheng, Tin Aung, Nomdo M. Jansonius, Caroline C. W. Klaver, Jamie E. Craig, Terri L. Young, Jonathan L. Haines, Stuart MacGregor, David A. Mackey, Norbert Pfeiffer, Tien-Yin Wong, Janey L. Wiggs, Alex W. Hewitt, Cornelia M. van Duijn, Christopher J. Hammond (2015)Meta-analysis of Genome-Wide Association Studies Identifies Novel Loci Associated With Optic Disc Morphology, In: Genetic epidemiology39(3)pp. 207-216 Wiley

Primary open-angle glaucoma is the most common optic neuropathy and an important cause of irreversible blindness worldwide. The optic nerve head or optic disc is divided in two parts: a central cup (without nerve fibers) surrounded by the neuroretinal rim (containing axons of the retinal ganglion cells). The International Glaucoma Genetics Consortium conducted a meta-analysis of genome-wide association studies consisting of 17,248 individuals of European ancestry and 6,841 individuals of Asian ancestry. The outcomes of the genome-wide association studies were disc area and cup area. These specific measurements describe optic nerve morphology in another way than the vertical cup-disc ratio, which is a clinically used measurement, and may shed light on new glaucoma mechanisms. We identified 10 new loci associated with disc area (CDC42BPA, F5, DIRC3, RARB, ABI3BP, DCAF4L2, ELP4, TMTC2, NR2F2, and HORMAD2) and another 10 new loci associated with cup area (DHRS3, TRIB2, EFEMP1, FLNB, FAM101, DDHD1, ASB7, KPNB1, BCAS3, and TRIOBP). The new genes participate in a number of pathways and future work is likely to identify more functions related to the pathogenesis of glaucoma.

Cristen J Willer, Ellen M Schmidt, Sebanti Sengupta, Gina M Peloso, Stefan Gustafsson, Stavroula Kanoni, Andrea Ganna, Jin Chen, Martin L Buchkovich, Samia Mora, Jacques S Beckmann, Jennifer L Bragg-Gresham, Hsing-Yi Chang, Ayşe Demirkan, Heleen M Den Hertog, Ron Do, Louise A Donnelly, Georg B Ehret, Tõnu Esko, Mary F Feitosa, Teresa Ferreira, Krista Fischer, Pierre Fontanillas, Ross M Fraser, Daniel F Freitag, Deepti Gurdasani, Kauko Heikkilä, Elina Hyppönen, Aaron Isaacs, Anne U Jackson, Åsa Johansson, Toby Johnson, Marika Kaakinen, Johannes Kettunen, Marcus E Kleber, Xiaohui Li, Jian'an Luan, Leo-Pekka Lyytikäinen, Patrik K E Magnusson, Massimo Mangino, Evelin Mihailov, May E Montasser, Martina Müller-Nurasyid, Ilja M Nolte, Jeffrey R O'Connell, Cameron D Palmer, Markus Perola, Ann-Kristin Petersen, Serena Sanna, Richa Saxena, Susan K Service, Sonia Shah, Dmitry Shungin, Carlo Sidore, Ci Song, Rona J Strawbridge, Ida Surakka, Toshiko Tanaka, Tanya M Teslovich, Gudmar Thorleifsson, Evita G Van den Herik, Benjamin F Voight, Kelly A Volcik, Lindsay L Waite, Andrew Wong, Ying Wu, Weihua Zhang, Devin Absher, Gershim Asiki, Inês Barroso, Latonya F Been, Jennifer L Bolton, Lori L Bonnycastle, Paolo Brambilla, Mary S Burnett, Giancarlo Cesana, Maria Dimitriou, Alex S F Doney, Angela Döring, Paul Elliott, Stephen E Epstein, Gudmundur Ingi Eyjolfsson, Bruna Gigante, Mark O Goodarzi, Harald Grallert, Martha L Gravito, Christopher J Groves, Göran Hallmans, Anna-Liisa Hartikainen, Caroline Hayward, Dena Hernandez, Andrew A Hicks, Hilma Holm, Yi-Jen Hung, Thomas Illig, Michelle R Jones, Pontiano Kaleebu, John J P Kastelein, Kay-Tee Khaw, Eric Kim (2013)Discovery and refinement of loci associated with lipid levels, In: Nature genetics45(11)1274pp. 1274-1283

Levels of low-density lipoprotein (LDL) cholesterol, high-density lipoprotein (HDL) cholesterol, triglycerides and total cholesterol are heritable, modifiable risk factors for coronary artery disease. To identify new loci and refine known loci influencing these lipids, we examined 188,577 individuals using genome-wide and custom genotyping arrays. We identify and annotate 157 loci associated with lipid levels at P < 5 × 10(-8), including 62 loci not previously associated with lipid levels in humans. Using dense genotyping in individuals of European, East Asian, South Asian and African ancestry, we narrow association signals in 12 loci. We find that loci associated with blood lipid levels are often associated with cardiovascular and metabolic traits, including coronary artery disease, type 2 diabetes, blood pressure, waist-hip ratio and body mass index. Our results demonstrate the value of using genetic data from individuals of diverse ancestry and provide insights into the biological mechanisms regulating blood lipids to guide future genetic, biological and therapeutic research.

Estefania Moreno-Gordaliza, Sven J. van der Lee, Ayse Demirkan, Cornelia M. van Duijn, Johan Kuiper, Petrus W. Lindenburg, Thomas Hankemeier (2016)A novel method for serum lipoprotein profiling using high performance capillary isotachophoresis, In: Analytica chimica acta944pp. 57-69 Elsevier

A new capillary isotachophoresis (cITP) method for lipoprotein profiling with superior lipoprotein coverage compared to previous methods has been developed, resolving twice as many lipoprotein species (18 peaks/fractions) in serum or plasma in less than 9.5 min. For this, a novel mixture of 24 spacers, including amino acids, dipeptides and sulfonic acids, was developed and fine-tuned, using predictive software (PeakMaster) and testing of spiked serum samples. Lipoprotein peaks were identified by serum-spiking with reference lipoproteins. Compatibility with common lipophilic stains for selective lipoprotein detection with either UV/Vis or laser-induced fluorescence was demonstrated. A special new capillary with a neutral coating (combining water-compatible OV1701-OH deactivation and methylation) was used for the first time for electrodriven separations, allowing very stable separations in a pH 8.8-9.4 gradient system, being functional for more than 100 injections. Excellent reproducibility was achieved, with coefficients of variation lower than 2.6% for absolute migration times. Comparison was performed with human plasma samples analyzed by NMR, leading to similar results with cITP after multivariate statistics, regarding group-clustering and lipoprotein species correlation. The new cITP method was applied to the analysis of serum samples from a LDL receptor knock-out mice model fed either a normal diet or a western-type diet. Differences in the lipoprotein levels and in the sublipoprotein types were detected, showing a shift to more atherogenic particles due to the high cholesterol diet. In summary, this novel method will allow more detailed and informative profiling of lipoprotein particle subtypes for cardiovascular disease research. (C) 2016 Elsevier B.V. All rights reserved.

Juho Tynkkynen, Vincent Chouraki, Sven J. van der Lee, Jussi Hernesniemi, Qiong Yang, Shuo Li, Alexa Beiser, Martin G. Larson, Katri Sääksjärvi, Martin J. Shipley, Archana Singh-Manoux, Robert E. Gerszten, Thomas J. Wang, Aki S. Havulinna, Peter Würtz, Krista Fischer, Ayse Demirkan, M. Arfan Ikram, Najaf Amin, Terho Lehtimäki, Mika Kähönen, Markus Perola, Andres Metspalu, Antti J. Kangas, Pasi Soininen, Mika Ala-Korpela, Ramachandran S. Vasan, Mika Kivimäki, Cornelia M. van Duijn, Sudha Seshadri, Veikko Salomaa (2018)Association of branched-chain amino acids and other circulating metabolites with risk of incident dementia and Alzheimer's disease: A prospective study in eight cohorts, In: Alzheimer's & dementia14(6)pp. 723-733 Elsevier Inc

Metabolite, lipid, and lipoprotein lipid profiling can provide novel insights into mechanisms underlying incident dementia and Alzheimer's disease. We studied eight prospective cohorts with 22,623 participants profiled by nuclear magnetic resonance or mass spectrometry metabolomics. Four cohorts were used for discovery with replication undertaken in the other four to avoid false positives. For metabolites that survived replication, combined association results are presented. Over 246,698 person-years, 995 and 745 cases of incident dementia and Alzheimer's disease were detected, respectively. Three branched-chain amino acids (isoleucine, leucine, and valine), creatinine and two very low density lipoprotein (VLDL)-specific lipoprotein lipid subclasses were associated with lower dementia risk. One high density lipoprotein (HDL; the concentration of cholesterol esters relative to total lipids in large HDL) and one VLDL (total cholesterol to total lipids ratio in very large VLDL) lipoprotein lipid subclass was associated with increased dementia risk. Branched-chain amino acids were also associated with decreased Alzheimer's disease risk and the concentration of cholesterol esters relative to total lipids in large HDL with increased Alzheimer's disease risk. Further studies can clarify whether these molecules play a causal role in dementia pathogenesis or are merely markers of early pathology.

Cassandra N Spracklen, Tugce Karaderi, Hanieh Yaghootkar, Claudia Schurmann, Rebecca S Fine, Zoltan Kutalik, Michael H Preuss, Yingchang Lu, Laura B L Wittemans, Linda S Adair, Matthew Allison, Najaf Amin, Paul L Auer, Traci M Bartz, Matthias Blüher, Michael Boehnke, Judith B Borja, Jette Bork-Jensen, Linda Broer, Daniel I Chasman, Yii-Der Ida Chen, Paraskevi Chirstofidou, Ayse Demirkan, Cornelia M van Duijn, Mary F Feitosa, Melissa E Garcia, Mariaelisa Graff, Harald Grallert, Niels Grarup, Xiuqing Guo, Jeffrey Haesser, Torben Hansen, Tamara B Harris, Heather M Highland, Jaeyoung Hong, M Arfan Ikram, Erik Ingelsson, Rebecca Jackson, Pekka Jousilahti, Mika Kähönen, Jorge R Kizer, Peter Kovacs, Jennifer Kriebel, Markku Laakso, Leslie A Lange, Terho Lehtimäki, Jin Li, Ruifang Li-Gao, Lars Lind, Jian'an Luan, Leo-Pekka Lyytikäinen, Stuart MacGregor, David A Mackey, Anubha Mahajan, Massimo Mangino, Satu Männistö, Mark I McCarthy, Barbara McKnight, Carolina Medina-Gomez, James B Meigs, Sophie Molnos, Dennis Mook-Kanamori, Andrew P Morris, Renee de Mutsert, Mike A Nalls, Ivana Nedeljkovic, Kari E North, Craig E Pennell, Aruna D Pradhan, Michael A Province, Olli T Raitakari, Chelsea K Raulerson, Alex P Reiner, Paul M Ridker, Samuli Ripatti, Neil Roberston, Jerome I Rotter, Veikko Salomaa, America A Sandoval-Zárate, Colleen M Sitlani, Tim D Spector, Konstantin Strauch, Michael Stumvoll, Kent D Taylor, Betina Thuesen, Anke Tönjes, Andre G Uitterlinden, Cristina Venturini, Mark Walker, Carol A Wang, Shuai Wang, Nicholas J Wareham, Sara M Willems, Ko Willems van Dijk, James G Wilson, Ying Wu, Jie Yao, Kristin L Young, Claudia Langenberg, Timothy M Frayling, Tuomas O Kilpeläinen, Cecilia M Lindgren, Ruth J F Loos, Karen L Mohlke (2019)Exome-Derived Adiponectin-Associated Variants Implicate Obesity and Lipid Biology, In: American journal of human genetics105(3)pp. 670-671
Ji Chen, Ayse Demirkan The Trans-Ancestral Genomic Architecture of Glycaemic Traits, In: bioRxiv Cold Spring Harbor Laboratory Press

Glycaemic traits are used to diagnose and monitor type 2 diabetes, and cardiometabolic health. To date, most genetic studies of glycaemic traits have focused on individuals of European ancestry. Here, we aggregated genome-wide association studies in up to 281,416 individuals without diabetes (30% non-European ancestry) with fasting glucose, 2h-glucose post-challenge, glycated haemoglobin, and fasting insulin data. Trans-ancestry and single-ancestry meta-analyses identified 242 loci (99 novel; P

Harish Dharuri, Peter Henneman, Ayse Demirkan, Jan Bert van Klinken, Dennis Owen Mook-Kanamori, Rui Wang-Sattler, Christian Gieger, Jerzy Adamski, Kristina Hettne, Marco Roos, Karsten Suhre, Cornelia M Van Duijn, Ko Willems van Dijk, Peter AC 't Hoen (2013)Automated workflow-based exploitation of pathway databases provides new insights into genetic associations of metabolite profiles, In: BMC genomics14(1)865pp. 865-865 BioMed Central
Ashley van der Spek, Linda Broer, Harmen H. M. Draisma, Rene Pool, Eva Albrecht, Marian Beekman, Massimo Mangino, Mait Raag, Dale R. Nyholt, Harish K. Dharuri, Veryan Codd, Najaf Amin, Eco J. C. de Geus, Joris Deelen, Ayse Demirkan, Idil Yet, Krista Fischer, Toomas Haller, Anjali K. Henders, Aaron Isaacs, Sarah E. Medland, Grant W. Montgomery, Simon P. Mooijaart, Konstantin Strauch, H. Eka D. Suchiman, Anika A. M. Vaarhorst, Diana van Heemst, Rui Wang-Sattler, John B. Whitfield, Gonneke Willemsen, Margaret J. Wright, Nicholas G. Martin, Nilesh J. Samani, Andres Metspalu, P. Eline Slagboom, Tim D. Spector, Dorret Boomsma, Cornelia M. van Duijn, Christian Gieger (2019)Metabolomics reveals a link between homocysteine and lipid metabolism and leukocyte telomere length: the ENGAGE consortium, In: Scientific reports9(Aug (E-published))11623pp. 11623-12 NATURE PORTFOLIO

Telomere shortening has been associated with multiple age-related diseases such as cardiovascular disease, diabetes, and dementia. However, the biological mechanisms responsible for these associations remain largely unknown. In order to gain insight into the metabolic processes driving the association of leukocyte telomere length (LTL) with age-related diseases, we investigated the association between LTL and serum metabolite levels in 7,853 individuals from seven independent cohorts. LTL was determined by quantitative polymerase chain reaction and the levels of 131 serum metabolites were measured with mass spectrometry in biological samples from the same blood draw. With partial correlation analysis, we identified six metabolites that were significantly associated with LTL after adjustment for multiple testing: lysophosphatidylcholine acyl C17:0 (lysoPC a C17:0, p-value=7.1 x 10(-6)), methionine (p-value=9.2 x 10(-5)), tyrosine (p-value=2.1 x 10(-4)), phosphatidylcholine diacyl C32:1 (PC aa C32:1, p-value=2.4 x 10(-4)), hydroxypropionylcarnitine (C3-OH, p-value=2.6 x 10(-4)), and phosphatidylcholine acyl-alkyl C38:4 (PC ae C38:4, p-value=9.0 x 10(-4)). Pathway analysis showed that the three phosphatidylcholines and methionine are involved in homocysteine metabolism and we found supporting evidence for an association of lipid metabolism with LTL. In conclusion, we found longer LTL associated with higher levels of lysoPC a C17:0 and PC ae C38:4, and with lower levels of methionine, tyrosine, PC aa C32:1, and C3-OH. These metabolites have been implicated in inflammation, oxidative stress, homocysteine metabolism, and in cardiovascular disease and diabetes, two major drivers of morbidity and mortality.

Andrew A. Hicks, Peter P. Pramstaller, Åsa Johansson, Veronique Vitart, Igor Rudan, Peter Ugocsai, Yurii Aulchenko, Christopher S. Franklin, Gerhard Liebisch, Jeanette Erdmann, Inger Jonasson, Irina V. Zorkoltseva, Cristian Pattaro, Caroline Hayward, Aaron Isaacs, Christian Hengstenberg, Susan Campbell, Carsten Gnewuch, A. CecileJ.W. Janssens, Anatoly V. Kirichenko, Inke R. König, Fabio Marroni, Ozren Polasek, Ayse Demirkan, Ivana Kolcic, Christine Schwienbacher, Wilmar Igl, Zrinka Biloglav, Jacqueline C. M. Witteman, Irene Pichler, Ghazal Zaboli, Tatiana I. Axenovich, Annette Peters, Stefan Schreiber, H.-Erich Wichmann, Heribert Schunkert, Nick Hastie, Ben A. Oostra, Sarah H. Wild, Thomas Meitinger, Ulf Gyllensten, Cornelia M. van Duijn, James F. Wilson, Alan Wright, Gerd Schmitz, Harry Campbell (2009)Genetic Determinants of Circulating Sphingolipid Concentrations in European Populations, In: PLoS genetics5(10)e1000672pp. e1000672-e1000672 Public Library of Science

Sphingolipids have essential roles as structural components of cell membranes and in cell signalling, and disruption of their metabolism causes several diseases, with diverse neurological, psychiatric, and metabolic consequences. Increasingly, variants within a few of the genes that encode enzymes involved in sphingolipid metabolism are being associated with complex disease phenotypes. Direct experimental evidence supports a role of specific sphingolipid species in several common complex chronic disease processes including atherosclerotic plaque formation, myocardial infarction (MI), cardiomyopathy, pancreatic β-cell failure, insulin resistance, and type 2 diabetes mellitus. Therefore, sphingolipids represent novel and important intermediate phenotypes for genetic analysis, yet little is known about the major genetic variants that influence their circulating levels in the general population. We performed a genome-wide association study (GWAS) between 318,237 single-nucleotide polymorphisms (SNPs) and levels of circulating sphingomyelin (SM), dihydrosphingomyelin (Dih-SM), ceramide (Cer), and glucosylceramide (GluCer) single lipid species (33 traits); and 43 matched metabolite ratios measured in 4,400 subjects from five diverse European populations. Associated variants (32) in five genomic regions were identified with genome-wide significant corrected p -values ranging down to 9.08×10 −66 . The strongest associations were observed in or near 7 genes functionally involved in ceramide biosynthesis and trafficking: SPTLC3 , LASS4 , SGPP1 , ATP10D , and FADS1–3 . Variants in 3 loci ( ATP10D , FADS3 , and SPTLC3 ) associate with MI in a series of three German MI studies. An additional 70 variants across 23 candidate genes involved in sphingolipid-metabolizing pathways also demonstrate association ( p  = 10 −4 or less). Circulating concentrations of several key components in sphingolipid metabolism are thus under strong genetic control, and variants in these loci can be tested for a role in the development of common cardiovascular, metabolic, neurological, and psychiatric diseases. Although several rare monogenic diseases are caused by defects in enzymes involved in sphingolipid biosynthesis and metabolism, little is known about the major variants that control the circulating levels of these important bioactive molecules. As well as being essential components of plasma membranes and endosomes, sphingolipids play critical roles in cell surface protection, protein and lipid transport and sorting, and cellular signalling cascades. Experimental evidence supports a role for sphingolipids in several common complex chronic metabolic, cardiovascular, or neurological disease processes. Therefore, sphingolipids represent novel and important intermediate phenotypes for genetic analysis, and discovering the genetic variants that influence their circulating concentrations is an important step towards understanding how the genetic control of sphingolipids might contribute to common human disease. We have identified 32 variants in 7 genes that have a strong effect on the circulating plasma levels of 33 distinct sphingolipids, and 43 matched metabolite ratios. In a series of 3 German MI studies, we see association with MI for variants in 3 of the genes tested. Further cardiovascular, metabolic, neurological, and psychiatric disease associations can be tested with the variants described here, which may identify additional disease risk and potentially useful therapeutic targets.

Najaf Amin, Karla V. Allebrandt, Ashley van der Spek, Bertram Mueller-Myhsok, Karin Hek, Maris Teder-Laving, Caroline Hayward, Tonu Esko, Josine G. van Mill, Hamdi Mbarek, Nathaniel F. Watson, Scott A. Melville, Fabiola M. Del Greco, Enda M. Byrne, Edwin Oole, Ivana Kolcic, Ting-hsu Chen, Daniel S. Evans, Josef Coresh, Nicole Vogelzangs, Juha Karjalainen, Gonneke Willemsen, Sina A. Gharib, Lina Zgaga, Evelin Mihailov, Katie L. Stone, Harry Campbell, Rutger Ww Brouwer, Ayse Demirkan, Aaron Isaacs, Zoran Dogas, Kristin D. Marciante, Susan Campbell, Fran Borovecki, Annemarie I. Luik, Man Li, Jouke Jan Hottenga, Jennifer E. Huffman, Mirjam C. G. N. van den Hout, Steven R. Cummings, Yuru S. Aulchenko, Philip R. Gehrman, Andre G. Uitterlinden, Heinz-Erich Wichmann, Martina Muller-Nurasyid, Rudolf S. N. Fehrmann, Grant W. Montgomery, Albert Hofman, Wen Hong Linda Kao, Ben A. Oostra, Alan F. Wright, Jacqueline M. Vink, James F. Wilson, Peter P. Pramstaller, Andrew A. Hicks, Ozren Polasek, Naresh M. Punjabi, Susan Redline, Bruce M. Psaty, Andrew C. Heath, Martha Merrow, Gregory J. Tranah, Daniel J. Gottlieb, Dorret I. Boomsma, Nicholas G. Martin, Igor Rudan, Henning Tiemeier, Wilfred F. J. van IJcken, Brenda W. Penninx, Andres Metspalu, Thomas Meitinger, Lude Franke, Till Roenneberg, Cornelia M. van Duijn (2016)Genetic variants in RBFOX3 are associated with sleep latency, In: European journal of human genetics : EJHG24(10)1488pp. 1488-1495 Springer Nature

Time to fall asleep (sleep latency) is a major determinant of sleep quality. Chronic, long sleep latency is a major characteristic of sleep-onset insomnia and/or delayed sleep phase syndrome. In this study we aimed to discover common polymorphisms that contribute to the genetics of sleep latency. We performed a meta-analysis of genome-wide association studies (GWAS) including 2 572 737 single nucleotide polymorphisms (SNPs) established in seven European cohorts including 4242 individuals. We found a cluster of three highly correlated variants (rs9900428, rs9907432 and rs7211029) in the RNA-binding protein fox-1 homolog 3 gene (RBFOX3) associated with sleep latency (P-values = 5.77 x 10(-08), 6.59 x 10(-08) and 9.17 x 10(-08)). These SNPs were replicated in up to 12 independent populations including 30 377 individuals (P-values = 1.5 x 10(-02), 7.0 x 10(-03) and 2.5 x 10(-03); combined meta-analysis P-values = 5.5 x 10(-07), 5.4 x 10(-07) and 1.0 x 10(-07)). A functional prediction of RBFOX3 based on co-expression with other genes shows that this gene is predominantly expressed in brain (P-value = 1.4 x 10(-316)) and the central nervous system (P-value = 7.5 x 10(-321)). The predicted function of RBFOX3 based on co-expression analysis with other genes shows that this gene is significantly involved in the release cycle of neurotransmitters including gamma-aminobutyric acid and various monoamines (P-values

Dina Vojinovic, Sven J. van der Lee, Cornelia M. van Duijn, Meike W. Vernooij, Maryam Kavousi, Najaf Amin, Ayse Demirkan, M. Arfan Ikram, Aad van der Lugt, Daniel Bos (2018)Metabolic profiling of intra- and extracranial carotid artery atherosclerosis, In: Atherosclerosis27260pp. 60-65 Elsevier

Background and aims: Increasing evidence shows that intracranial carotid artery atherosclerosis may develop under the influence of a differential metabolic risk factor profile than atherosclerosis in the extracranial part of the carotid artery. To further elucidate these differences, we investigated associations of a wide range of circulating metabolites with intracranial and extracranial carotid artery atherosclerosis.& para;& para;Methods: From the population-based Rotterdam Study, blood samples from 1111 participants were used to determine a wide range of metabolites by proton nuclear magnetic resonance (NMR). Moreover, these participants underwent non-contrast computed tomography of the neck and head to quantify the amount of extra-and intracranial carotid artery calcification (ECAC and ICAC), as a proxy of atherosclerosis. We assessed associations of the metabolites with ICAC and ECAC and compared the metabolic association patterns of the two.& para;& para;Results: We found that one standard deviation (SD) increase in concentration of 3-hydroxybutyrate, a ketone body, was significantly associated with a 0.11 SD increase in ICAC volume (p = 1.8 x 10(-4)). When we compared the metabolic association pattern of ICAC with that of ECAC, we observed differences in glycolysis-related metabolite measures, lipoprotein subfractions, and amino acids. Interestingly, glyco-protein acetyls were associated with calcification in both studied vessel beds. These associations were most prominent in men.& para;& para;Conclusions: We found that a higher circulating level of 3-hydroxybutyrate was associated with an increase in ICAC. Furthermore, we found differences in metabolic association patterns of ICAC and ECAC, providing further evidence for location-specific differences in the etiology of atherosclerosis. (C) 2018 Elsevier B.V. All rights reserved.

Gerrit L J Onderwater, Lannie Ligthart, Mariska Bot, Ayse Demirkan, Jingyuan Fu, Carla J H van der Kallen, Lisanne S Vijfhuizen, René Pool, Jun Liu, Floris H M Vanmolkot, Marian Beekman, Ke-Xin Wen, Najaf Amin, Carisha S Thesing, Judith A Pijpers, Dennis A Kies, Ronald Zielman, Irene de Boer, Marleen M J van Greevenbroek, Ilja C W Arts, Yuri Milaneschi, Miranda T Schram, Pieter C Dagnelie, Lude Franke, M Arfan Ikram, Michel D Ferrari, Jelle J Goeman, P Eline Slagboom, Cisca Wijmenga, Coen D A Stehouwer, Dorret I Boomsma, Cornelia M van Duijn, Brenda W Penninx, Peter A C 't Hoen, Gisela M Terwindt, Arn M J M van den Maagdenberg (2019)Large-scale plasma metabolome analysis reveals alterations in HDL metabolism in migraine, In: Neurology92(16)e1899pp. e1899-e1911

To identify a plasma metabolomic biomarker signature for migraine. Plasma samples from 8 Dutch cohorts (n = 10,153: 2,800 migraine patients and 7,353 controls) were profiled on a H-NMR-based metabolomics platform, to quantify 146 individual metabolites (e.g., lipids, fatty acids, and lipoproteins) and 79 metabolite ratios. Metabolite measures associated with migraine were obtained after single-metabolite logistic regression combined with a random-effects meta-analysis performed in a nonstratified and sex-stratified manner. Next, a global test analysis was performed to identify sets of related metabolites associated with migraine. The Holm procedure was applied to control the family-wise error rate at 5% in single-metabolite and global test analyses. Decreases in the level of apolipoprotein A1 (β -0.10; 95% confidence interval [CI] -0.16, -0.05; adjusted = 0.029) and free cholesterol to total lipid ratio present in small high-density lipoprotein subspecies (HDL) (β -0.10; 95% CI -0.15, -0.05; adjusted = 0.029) were associated with migraine status. In addition, only in male participants, a decreased level of omega-3 fatty acids (β -0.24; 95% CI -0.36, -0.12; adjusted = 0.033) was associated with migraine. Global test analysis further supported that HDL traits (but not other lipoproteins) were associated with migraine status. Metabolic profiling of plasma yielded alterations in HDL metabolism in migraine patients and decreased omega-3 fatty acids only in male migraineurs.

Ayse Demirkan, Cornelia M. van Duijn, Peter Ugocsai, Aaron Isaacs, Peter P. Pramstaller, Gerhard Liebisch, James F. Wilson, Asa Johansson, Igor Rudan, Yurii S. Aulchenko, Anatoly V. Kirichenko, A. Cecile J. W. Janssens, Ritsert C. Jansen, Carsten Gnewuch, Francisco S. Domingues, Cristian Pattaro, Sarah H. Wild, Inger Jonasson, Ozren Polasek, Irina V. Zorkoltseva, Albert Hofman, Lennart C. Karssen, Maksim Struchalin, James Floyd, Wilmar Igl, Zrinka Biloglav, Linda Broer, Arne Pfeufer, Irene Pichler, Susan Campbell, Ghazal Zaboli, Ivana Kolcic, Fernando Rivadeneira, Jennifer Huffman, Nicholas D. Hastie, Andre Uitterlinden, Lude Franke, Christopher S. Franklin, Veronique Vitart, Christopher P. Nelson, Michael Preuss, Joshua C. Bis, Christopher J. O'Donnell, Nora Franceschini, Jacqueline C. M. Witteman, Tatiana Axenovich, Ben A. Oostra, Thomas Meitinger, Andrew A. Hicks, Caroline Hayward, Alan F. Wright, Ulf Gyllensten, Harry Campbell, Gerd Schmitz (2012)Genome-Wide Association Study Identifies Novel Loci Associated with Circulating Phospho- and Sphingolipid Concentrations, In: PLoS genetics8(2)1002490pp. e1002490-e1002490 Public Library Science

Phospho- and sphingolipids are crucial cellular and intracellular compounds. These lipids are required for active transport, a number of enzymatic processes, membrane formation, and cell signalling. Disruption of their metabolism leads to several diseases, with diverse neurological, psychiatric, and metabolic consequences. A large number of phospholipid and sphingolipid species can be detected and measured in human plasma. We conducted a meta-analysis of five European family-based genome-wide association studies (N = 4034) on plasma levels of 24 sphingomyelins (SPM), 9 ceramides (CER), 57 phosphatidylcholines (PC), 20 lysophosphatidylcholines (LPC), 27 phosphatidylethanolamines (PE), and 16 PE-based plasmalogens (PLPE), as well as their proportions in each major class. This effort yielded 25 genome-wide significant loci for phospholipids (smallest P-value = 9.88 x 10(-204)) and 10 loci for sphingolipids (smallest P-value = 3.10 x 10(-57)). After a correction for multiple comparisons (P-value, 2.2 x 10(-9)), we observed four novel loci significantly associated with phospholipids (PAQR9, AGPAT1, PKD2L1, PDXDC1) and two with sphingolipids (PLD2 and APOE) explaining up to 3.1% of the variance. Further analysis of the top findings with respect to within class molar proportions uncovered three additional loci for phospholipids (PNLIPRP2, PCDH20, and ABDH3) suggesting their involvement in either fatty acid elongation/saturation processes or fatty acid specific turnover mechanisms. Among those, 14 loci (KCNH7, AGPAT1, PNLIPRP2, SYT9, FADS1-2-3, DLG2, APOA1, ELOVL2, CDK17, LIPC, PDXDC1, PLD2, LASS4, and APOE) mapped into the glycerophospholipid and 12 loci (ILKAP, ITGA9, AGPAT1, FADS1-2-3, APOA1, PCDH20, LIPC, PDXDC1, SGPP1, APOE, LASS4, and PLD2) to the sphingolipid pathways. In large meta-analyses, associations between FADS1-2-3 and carotid intima media thickness, AGPAT1 and type 2 diabetes, and APOA1 and coronary artery disease were observed. In conclusion, our study identified nine novel phospho- and sphingolipid loci, substantially increasing our knowledge of the genetic basis for these traits.

Mariaelisa Graff, Robert A Scott, Anne E Justice, Kristin L Young, Mary F Feitosa, Llilda Barata, Thomas W Winkler, Audrey Y Chu, Anubha Mahajan, David Hadley, Luting Xue, Tsegaselassie Workalemahu, Nancy L Heard-Costa, Marcel den Hoed, Tarunveer S Ahluwalia, Qibin Qi, Julius S Ngwa, Frida Renström, Lydia Quaye, John D Eicher, James E Hayes, Marilyn Cornelis, Zoltan Kutalik, Elise Lim, Jian'an Luan, Jennifer E Huffman, Weihua Zhang, Wei Zhao, Paula J Griffin, Toomas Haller, Shafqat Ahmad, Pedro M Marques-Vidal, Stephanie Bien, Loic Yengo, Alexander Teumer, Albert Vernon Smith, Meena Kumari, Marie Neergaard Harder, Johanne Marie Justesen, Marcus E Kleber, Mette Hollensted, Kurt Lohman, Natalia V Rivera, John B Whitfield, Jing Hua Zhao, Heather M Stringham, Leo-Pekka Lyytikäinen, Charlotte Huppertz, Gonneke Willemsen, Wouter J Peyrot, Ying Wu, Kati Kristiansson, Ayse Demirkan, Myriam Fornage, Maija Hassinen, Lawrence F Bielak, Gemma Cadby, Toshiko Tanaka, Reedik Mägi, Peter J van der Most, Anne U Jackson, Jennifer L Bragg-Gresham, Veronique Vitart, Jonathan Marten, Pau Navarro, Claire Bellis, Dorota Pasko, Åsa Johansson, Søren Snitker, Yu-Ching Cheng, Joel Eriksson, Unhee Lim, Mette Aadahl, Linda S Adair, Najaf Amin, Beverley Balkau, Juha Auvinen, John Beilby, Richard N Bergman, Sven Bergmann, Alain G Bertoni, John Blangero, Amélie Bonnefond, Lori L Bonnycastle, Judith B Borja, Søren Brage, Fabio Busonero, Steve Buyske, Harry Campbell, Peter S Chines, Francis S Collins, Tanguy Corre, George Davey Smith, Graciela E Delgado, Nicole Dueker, Marcus Dörr, Tapani Ebeling, Gudny Eiriksdottir, Tõnu Esko, Jessica D Faul, Mao Fu, Kristine Færch, Christian Gieger, Sven Gläser, Jian Gong, Penny Gordon-Larsen, Harald Grallert, Tanja B Grammer, Niels Grarup, Gerard van Grootheest, Kennet Harald, Nicholas D Hastie, Aki S Havulinna, Dena Hernandez, Lucia Hindorff, Lynne J Hocking, Oddgeir L Holmens, Christina Holzapfel, Jouke Jan Hottenga, Jie Huang, Tao Huang, Jennie Hui, Cornelia Huth, Nina Hutri-Kähönen, Alan L James, John-Olov Jansson, Min A Jhun, Markus Juonala, Leena Kinnunen, Heikki A Koistinen, Ivana Kolcic, Pirjo Komulainen, Johanna Kuusisto, Kirsti Kvaløy, Mika Kähönen, Timo A Lakka, Lenore J Launer, Benjamin Lehne, Cecilia M Lindgren, Mattias Lorentzon, Robert Luben, Michel Marre, Yuri Milaneschi, Keri L Monda, Grant W Montgomery, Marleen H M De Moor, Antonella Mulas, Martina Müller-Nurasyid, A W Musk, Reija Männikkö, Satu Männistö, Narisu Narisu, Matthias Nauck, Jennifer A Nettleton, Ilja M Nolte, Albertine J Oldehinkel, Matthias Olden, Ken K Ong, Sandosh Padmanabhan, Lavinia Paternoster, Jeremiah Perez, Markus Perola, Annette Peters, Ulrike Peters, Patricia A Peyser, Inga Prokopenko, Hannu Puolijoki, Olli T Raitakari, Tuomo Rankinen, Laura J Rasmussen-Torvik, Rajesh Rawal, Paul M Ridker, Lynda M Rose, Igor Rudan, Cinzia Sarti, Mark A Sarzynski, Kai Savonen, William R Scott, Serena Sanna, Alan R Shuldiner, Steve Sidney, Günther Silbernagel, Blair H Smith, Jennifer A Smith, Harold Snieder, Alena Stančáková, Barbara Sternfeld, Amy J Swift, Tuija Tammelin, Sian-Tsung Tan, Barbara Thorand, Dorothée Thuillier, Liesbeth Vandenput, Henrik Vestergaard, Jana V van Vliet-Ostaptchouk, Marie-Claude Vohl, Uwe Völker, Gérard Waeber, Mark Walker, Sarah Wild, Andrew Wong, Alan F Wright, M Carola Zillikens, Niha Zubair, Christopher A Haiman, Loic Lemarchand, Ulf Gyllensten, Claes Ohlsson, Albert Hofman, Fernando Rivadeneira, André G Uitterlinden, Louis Pérusse, James F Wilson, Caroline Hayward, Ozren Polasek, Francesco Cucca, Kristian Hveem, Catharina A Hartman, Anke Tönjes, Stefania Bandinelli, Lyle J Palmer, Sharon L R Kardia, Rainer Rauramaa, Thorkild I A Sørensen, Jaakko Tuomilehto, Veikko Salomaa, Brenda W J H Penninx, Eco J C de Geus, Dorret I Boomsma, Terho Lehtimäki, Massimo Mangino, Markku Laakso, Claude Bouchard, Nicholas G Martin, Diana Kuh, Yongmei Liu, Allan Linneberg, Winfried März, Konstantin Strauch, Mika Kivimäki, Tamara B Harris, Vilmundur Gudnason, Henry Völzke, Lu Qi, Marjo-Riitta Järvelin, John C Chambers, Jaspal S Kooner, Philippe Froguel, Charles Kooperberg, Peter Vollenweider, Göran Hallmans, Torben Hansen, Oluf Pedersen, Andres Metspalu, Nicholas J Wareham, Claudia Langenberg, David R Weir, David J Porteous, Eric Boerwinkle, Daniel I Chasman, Gonçalo R Abecasis, Inês Barroso, Mark I McCarthy, Timothy M Frayling, Jeffrey R O'Connell, Cornelia M van Duijn, Michael Boehnke, Iris M Heid, Karen L Mohlke, David P Strachan, Caroline S Fox, Ching-Ti Liu, Joel N Hirschhorn, Robert J Klein, Andrew D Johnson, Ingrid B Borecki, Paul W Franks, Kari E North, L Adrienne Cupples, Ruth J F Loos, Tuomas O Kilpeläinen (2017)Correction: Genome-wide physical activity interactions in adiposity - A meta-analysis of 200,452 adults, In: PLoS genetics13(8)pp. e1006972-e1006972

[This corrects the article DOI: 10.1371/journal.pgen.1006528.].

Mariaelisa Graff, Robert A Scott, Anne E Justice, Kristin L Young, Mary F Feitosa, Llilda Barata, Thomas W Winkler, Audrey Y Chu, Anubha Mahajan, David Hadley, Luting Xue, Tsegaselassie Workalemahu, Nancy L Heard-Costa, Marcel den Hoed, Tarunveer S Ahluwalia, Qibin Qi, Julius S Ngwa, Frida Renström, Lydia Quaye, John D Eicher, James E Hayes, Marilyn Cornelis, Zoltan Kutalik, Elise Lim, Jian'an Luan, Jennifer E Huffman, Weihua Zhang, Wei Zhao, Paula J Griffin, Toomas Haller, Shafqat Ahmad, Pedro M Marques-Vidal, Stephanie Bien, Loic Yengo, Alexander Teumer, Albert Vernon Smith, Meena Kumari, Marie Neergaard Harder, Johanne Marie Justesen, Marcus E Kleber, Mette Hollensted, Kurt Lohman, Natalia V Rivera, John B Whitfield, Jing Hua Zhao, Heather M Stringham, Leo-Pekka Lyytikäinen, Charlotte Huppertz, Gonneke Willemsen, Wouter J Peyrot, Ying Wu, Kati Kristiansson, Ayse Demirkan, Myriam Fornage, Maija Hassinen, Lawrence F Bielak, Gemma Cadby, Toshiko Tanaka, Reedik Mägi, Peter J van der Most, Anne U Jackson, Jennifer L Bragg-Gresham, Veronique Vitart, Jonathan Marten, Pau Navarro, Claire Bellis, Dorota Pasko, Åsa Johansson, Søren Snitker, Yu-Ching Cheng, Joel Eriksson, Unhee Lim, Mette Aadahl, Linda S Adair, Najaf Amin, Beverley Balkau, Juha Auvinen, John Beilby, Richard N Bergman, Sven Bergmann, Alain G Bertoni, John Blangero, Amélie Bonnefond, Lori L Bonnycastle, Judith B Borja, Søren Brage, Fabio Busonero, Steve Buyske, Harry Campbell, Peter S Chines, Francis S Collins, Tanguy Corre, George Davey Smith, Graciela E Delgado, Nicole Dueker, Marcus Dörr, Tapani Ebeling, Gudny Eiriksdottir, Tõnu Esko, Jessica D Faul, Mao Fu, Kristine Færch, Christian Gieger, Sven Gläser, Jian Gong, Penny Gordon-Larsen, Harald Grallert, Tanja B Grammer, Niels Grarup, Gerard van Grootheest, Kennet Harald, Nicholas D Hastie, Aki S Havulinna, Dena Hernandez, Lucia Hindorff, Lynne J Hocking, Oddgeir L Holmens, Christina Holzapfel, Jouke Jan Hottenga, Jie Huang, Tao Huang, Jennie Hui, Cornelia Huth, Nina Hutri-Kähönen, Alan L James, John-Olov Jansson, Min A Jhun, Markus Juonala, Leena Kinnunen, Heikki A Koistinen, Ivana Kolcic, Pirjo Komulainen, Johanna Kuusisto, Kirsti Kvaløy, Mika Kähönen, Timo A Lakka, Lenore J Launer, Benjamin Lehne, Cecilia M Lindgren, Mattias Lorentzon, Robert Luben, Michel Marre, Yuri Milaneschi, Keri L Monda, Grant W Montgomery, Marleen H M De Moor, Antonella Mulas, Martina Müller-Nurasyid, A W Musk, Reija Männikkö, Satu Männistö, Narisu Narisu, Matthias Nauck, Jennifer A Nettleton, Ilja M Nolte, Albertine J Oldehinkel, Matthias Olden, Ken K Ong, Sandosh Padmanabhan, Lavinia Paternoster, Jeremiah Perez, Markus Perola, Annette Peters, Ulrike Peters, Patricia A Peyser, Inga Prokopenko, Hannu Puolijoki, Olli T Raitakari, Tuomo Rankinen, Laura J Rasmussen-Torvik, Rajesh Rawal, Paul M Ridker, Lynda M Rose, Igor Rudan, Cinzia Sarti, Mark A Sarzynski, Kai Savonen, William R Scott, Serena Sanna, Alan R Shuldiner, Steve Sidney, Günther Silbernagel, Blair H Smith, Jennifer A Smith, Harold Snieder, Alena Stančáková, Barbara Sternfeld, Amy J Swift, Tuija Tammelin, Sian-Tsung Tan, Barbara Thorand, Dorothée Thuillier, Liesbeth Vandenput, Henrik Vestergaard, Jana V van Vliet-Ostaptchouk, Marie-Claude Vohl, Uwe Völker, Gérard Waeber, Mark Walker, Sarah Wild, Andrew Wong, Alan F Wright, M Carola Zillikens, Niha Zubair, Christopher A Haiman, Loic Lemarchand, Ulf Gyllensten, Claes Ohlsson, Albert Hofman, Fernando Rivadeneira, André G Uitterlinden, Louis Pérusse, James F Wilson, Caroline Hayward, Ozren Polasek, Francesco Cucca, Kristian Hveem, Catharina A Hartman, Anke Tönjes, Stefania Bandinelli, Lyle J Palmer, Sharon L R Kardia, Rainer Rauramaa, Thorkild I A Sørensen, Jaakko Tuomilehto, Veikko Salomaa, Brenda W J H Penninx, Eco J C de Geus, Dorret I Boomsma, Terho Lehtimäki, Massimo Mangino, Markku Laakso, Claude Bouchard, Nicholas G Martin, Diana Kuh, Yongmei Liu, Allan Linneberg, Winfried März, Konstantin Strauch, Mika Kivimäki, Tamara B Harris, Vilmundur Gudnason, Henry Völzke, Lu Qi, Marjo-Riitta Järvelin, John C Chambers, Jaspal S Kooner, Philippe Froguel, Charles Kooperberg, Peter Vollenweider, Göran Hallmans, Torben Hansen, Oluf Pedersen, Andres Metspalu, Nicholas J Wareham, Claudia Langenberg, David R Weir, David J Porteous, Eric Boerwinkle, Daniel I Chasman, Gonçalo R Abecasis, Inês Barroso, Mark I McCarthy, Timothy M Frayling, Jeffrey R O'Connell, Cornelia M van Duijn, Michael Boehnke, Iris M Heid, Karen L Mohlke, David P Strachan, Caroline S Fox, Ching-Ti Liu, Joel N Hirschhorn, Robert J Klein, Andrew D Johnson, Ingrid B Borecki, Paul W Franks, Kari E North, L Adrienne Cupples, Ruth J F Loos, Tuomas O Kilpeläinen (2017)Correction: Genome-wide physical activity interactions in adiposity - A meta-analysis of 200,452 adults, In: PLoS genetics13(8)pp. e1006972-e1006972

[This corrects the article DOI: 10.1371/journal.pgen.1006528.].

A Mahajan, J Wessel, S M Willems, W Zhao, N R Robertson, A Y Chu, W Gan, H Kitajima, D Taliun, N W Rayner, X Guo, Y Lu, M Li, R A Jensen, Y Hu, S Huo, K K Lohman, W Zhang, J P Cook, B P Prins, J Flannick, N Grarup, V V Trubetskoy, J Kravic, Y J Kim, D V Rybin, H Yaghootkar, M Mueller-Nurasyid, K Meidtner, R Li-Gao, T V Varga, J Marten, J Li, A V Smith, P An, S Ligthart, S Gustafsson, G Malerba, A Demirkan, J F Tajes, V Steinthorsdottir, M Wuttke, C Lecoeur, M Preuss, L F Bielak, M Graff, H M Highland, A E Justice, D J Liu, E Marouli, G M Peloso, H R Warren, S Afaq, S Afzal, E Ahlqvist, P Almgren, N Amin, L B Bang, A G Bertoni, C Bombieri, J Bork-Jensen, I Brandslund, J A Brody, N P Burtt, M Canouil, Y-DI Chen, Y S Cho, C Christensen, S V Eastwood, K-U Eckardt, K Fischer, G Gambaro, V Giedraitis, M L Grove, H G de Haan, S Hackinger, Y Hai, S Han, A Tybjaerg-Hansen, M-F Hivert, B Isomaa, S Jager, M E Jorgensen, T Jorgensen, A Karajamaki, B-J Kim, S S Kim, H A Koistinen, P Kovacs, J Kriebel, F Kronenberg, K Lall, L A Lange, J-J Lee, B Lehne, H Li, K-H Lin, A Linneberg, C-T Liu, J Liu, M Loh, R Magi, V Mamakou, R McKean-Cowdin, G Nadkarni, M Neville, S F Nielsen, I Ntalla, P A Peyser, W Rathmann, K Rice, S S Rich, L Rode, O Rolandsson, S Schonherr, E Selvin, K S Small, A Stancakova, P Surendran, K D Taylor, T M Teslovich, B Thorand, G Thorleifsson, A Tin, A Tonjes, A Varbo, D R Witte, A R Wood, P Yajnik, J Yao, L Yengo, R Young, P Amouyel, H Boeing, E Boerwinkle, E P Bottinger, R Chowdhury, F S Collins, G Dedoussis, A Dehghan, P Deloukas, M M Ferrario, J Ferrieres, J C Florez, P Frossard, V Gudnason, T B Harris, S R Heckbert, JMM Howson, M Ingelsson, S Kathiresan, F Kee, J Kuusisto, C Langenberg, L J Launer, C M Lindgren, S Mannisto, T Meitinger, O Melander, K L Mohlke, M Moitry, A D Morris, A D Murray, R de Mutsert, M Orho-Melander, K R Owen, M Perola, A Peters, M A Province, A Rasheed, P M Ridker, F Rivadineira, F R Rosendaal, A H Rosengren, V Salomaa, WH-H Sheu, R Sladek, B H Smith, K Strauch, A G Uitterlinden, R Varma, C J Willer, M Bluher, A S Butterworth, J C Chambers, D I Chasman, J Danesh, C van Duijn, J Dupuis, O H Franco, P W Franks, P Froguel, H Grallert, L Groop, B-G Han, T Hansen, A T Hattersley, C Hayward, E Ingelsson, SLR Kardia, F Karpe, J S Kooner, A Kottgen, K Kuulasmaa, M Laakso, X Lin, L Lind, Y Liu, RJF Loos, J Marchini, A Metspalu, D Mook-Kanamori, B G Nordestgaard, CNA Palmer, J S Pankow, O Pedersen, B M Psaty, R Rauramaa, N Sattar, M B Schulze, N Soranzo, T D Spector, K Stefansson, M Stumvoll, U Thorsteinsdottir, T Tuomi, J Tuomilehto, N J Wareham, J G Wilson, E Zeggini, R A Scott, I Barroso, T M Frayling, M O Goodarzi, J B Meigs, M Boehnke, D Saleheen, A P Morris, J I Rotter, M I McCarthy, E Consortium, MAGIC Consortium, GIANT Consortium (2018)Refining the accuracy of validated target identification through coding variant fine-mapping in type 2 diabetes Nature Research (part of Springer Nature)

We aggregated coding variant data for 81,412 type 2 diabetes cases and 370,832 controls of diverse ancestry, identifying 40 coding variant association signals (P 

Joris Deelen, Johannes Kettunen, Krista Fischer, Ashley van der Spek, Stella Trompet, Gabi Kastenmueller, Andy Boyd, Jonas Zierer, Erik B. van den Akker, Mika Ala-Korpela, Najaf Amin, Ayse Demirkan, Mohsen Ghanbari, Diana van Heemst, M. Arfan Ikram, Jan Bert van Klinken, Simon P. Mooijaart, Annette Peters, Veikko Salomaa, Naveed Sattar, Tim D. Spector, Henning Tiemeier, Aswin Verhoeven, Melanie Waldenberger, Peter Wurtz, George Davey Smith, Andres Metspalu, Markus Perola, Cristina Menni, Johanna M. Geleijnse, Fotios Drenos, Marian Beekman, J. Wouter Jukema, Cornelia M. van Duijn, P. Eline Slagboom (2019)A metabolic profile of all-cause mortality risk identified in an observational study of 44,168 individuals, In: Nature communications10(1)3346pp. 3346-8 NATURE PORTFOLIO

Predicting longer-term mortality risk requires collection of clinical data, which is often cumbersome. Therefore, we use a well-standardized metabolomics platform to identify metabolic predictors of long-term mortality in the circulation of 44,168 individuals (age at baseline 18-109), of whom 5512 died during follow-up. We apply a stepwise (forward-backward) procedure based on meta-analysis results and identify 14 circulating biomarkers independently associating with all-cause mortality. Overall, these associations are similar in men and women and across different age strata. We subsequently show that the prediction accuracy of 5- and 10-year mortality based on a model containing the identified biomarkers and sex (C-statistic = 0.837 and 0.830, respectively) is better than that of a model containing conventional risk factors for mortality (C-statistic = 0.772 and 0.790, respectively). The use of the identified metabolic profile as a predictor of mortality or surrogate endpoint in clinical studies needs further investigation.

Daniel I. Chasman, Christian Fuchsberger, Cristian Pattaro, Alexander Teumer, Carsten A. Boeger, Karlhans Endlich, Matthias Olden, Ming-Huei Chen, Adrienne Tin, Daniel Taliun, Man Li, Xiaoyi Gao, Mathias Gorski, Qiong Yang, Claudia Hundertmark, Meredith C. Foster, Conall M. O'Seaghdha, Nicole Glazer, Aaron Isaacs, Ching-Ti Liu, Albert V. Smith, Jeffrey R. O'Connell, Maksim Struchalin, Toshiko Tanaka, Guo Li, Andrew D. Johnson, Hinco J. Gierman, Mary F. Feitosa, Shih-Jen Hwang, Elizabeth J. Atkinson, Kurt Lohman, Marilyn C. Cornelis, Asa Johansson, Anke Toenjes, Abbas Dehghan, Jean-Charles Lambert, Elizabeth G. Holliday, Rossella Sorice, Zoltan Kutalik, Terho Lehtimaeki, Tonu Esko, Harshal Deshmukh, Sheila Ulivi, Audrey Y. Chu, Federico Murgia, Stella Trompet, Medea Imboden, Stefan Coassin, Giorgio Pistis, Tamara B. Harris, Lenore J. Launer, Thor Aspelund, Gudny Eiriksdottir, Braxton D. Mitchell, Eric Boerwinkle, Helena Schmidt, Margherita Cavalieri, Madhumathi Rao, Frank Hu, Ayse Demirkan, Ben A. Oostra, Mariza de Andrade, Stephen T. Turner, Jingzhong Ding, Jeanette S. Andrews, Barry I. Freedman, Franco Giulianini, Wolfgang Koenig, Thomas Illig, Christa Meisinger, Christian Gieger, Lina Zgaga, Tatijana Zemunik, Mladen Boban, Cosetta Minelli, Heather E. Wheeler, Wilmar Igl, Ghazal Zaboli, Sarah H. Wild, Alan F. Wright, Harry Campbell, David Ellinghaus, Ute Noethlings, Gunnar Jacobs, Reiner Biffar, Florian Ernst, Georg Homuth, Heyo K. Kroemer, Matthias Nauck, Sylvia Stracke, Uwe Voelker, Henry Voelzke, Peter Kovacs, Michael Stumvoll, Reedik Maegi, Albert Hofman, Andre G. Uitterlinden, Fernando Rivadeneira, Yurii S. Aulchenko, Ozren Polasek, Inga Prokopenko (2012)Integration of genome-wide association studies with biological knowledge identifies six novel genes related to kidney function, In: Human molecular genetics21(24)pp. 5329-5343 Oxford Univ Press

In conducting genome-wide association studies (GWAS), analytical approaches leveraging biological information may further understanding of the pathophysiology of clinical traits. To discover novel associations with estimated glomerular filtration rate (eGFR), a measure of kidney function, we developed a strategy for integrating prior biological knowledge into the existing GWAS data for eGFR from the CKDGen Consortium. Our strategy focuses on single nucleotide polymorphism (SNPs) in genes that are connected by functional evidence, determined by literature mining and gene ontology (GO) hierarchies, to genes near previously validated eGFR associations. It then requires association thresholds consistent with multiple testing, and finally evaluates novel candidates by independent replication. Among the samples of European ancestry, we identified a genome-wide significant SNP in FBXL20 (P 5.6 10(9)) in meta-analysis of all available data, and additional SNPs at the INHBC, LRP2, PLEKHA1, SLC3A2 and SLC7A6 genes meeting multiple-testing corrected significance for replication and overall P-values of 4.5 10(4)2.2 10(7). Neither the novel PLEKHA1 nor FBXL20 associations, both further supported by association with eGFR among African Americans and with transcript abundance, would have been implicated by eGFR candidate gene approaches. LRP2, encoding the megalin receptor, was identified through connection with the previously known eGFR gene DAB2 and extends understanding of the megalin system in kidney function. These findings highlight integration of existing genome-wide association data with independent biological knowledge to uncover novel candidate eGFR associations, including candidates lacking known connections to kidney-specific pathways. The strategy may also be applicable to other clinical phenotypes, although more testing will be needed to assess its potential for discovery in general.

Jun Liu, Sabina Semiz, Sven J. van der Lee, Ashley van der Spek, Aswin Verhoeven, Jan B. van Klinken, Eric Sijbrands, Amy C. Harms, Thomas Hankemeier, Ko Willems van Dijk, Cornelia M. van Duijn, Ayse Demirkan (2017)Metabolomics based markers predict type 2 diabetes in a 14-year follow-up study, In: Metabolomics13(9)104pp. 104-104 Springer Nature

Background The growing field of metabolomics has opened up new opportunities for prediction of type 2 diabetes (T2D) going beyond the classical biochemistry assays. Objectives We aimed to identify markers from different pathways which represent early metabolic changes and test their predictive performance for T2D, as compared to the performance of traditional risk factors (TRF). Methods We analyzed 2776 participants from the Erasmus Rucphen Family study from which 1571 disease free individuals were followed up to 14-years. The targeted metabolomics measurements at baseline were performed by three different platforms using either nuclear magnetic resonance spectroscopy or mass spectrometry. We selected 24 T2D markers by using Least Absolute Shrinkage and Selection operator (LASSO) regression and tested their association to incidence of disease during follow-up. Results The 24 markers i.e. high-density, low-density and very low-density lipoprotein sub-fractions, certain triglycerides, amino acids, and small intermediate compounds predicted future T2D with an area under the curve (AUC) of 0.81. The performance of the metabolic markers compared to glucose was significantly higher among the young (age < 50 years) (0.86 vs. 0.77, p-value

Hendrik R. Taal, Germaine C. Verwoert, Ayse Demirkan, A. Cecile J. W. Janssens, Kenneth Rice, Georg Ehret, Albert V. Smith, Ben F. J. Verhaaren, Jacqueline C. M. Witteman, Albert Hofman, Meike W. Vernooij, Andre G. Uitterlinden, Fernando Rivadeneira, M. Arfan Ikram, Daniel Levy, Albert J. van der Heijden, Vincent W. V. Jaddoe, Cornelia M. van Duijn (2012)Genome-Wide Profiling of Blood Pressure in Adults and Children, In: Hypertension (Dallas, Tex. 1979)59(2)241pp. 241-247 Lippincott Williams & Wilkins

Hypertension is an important determinant of cardiovascular morbidity and mortality and has a substantial heritability, which is likely of polygenic origin. The aim of this study was to assess to what extent multiple common genetic variants contribute to blood pressure regulation in both adults and children and to assess overlap in variants between different age groups, using genome-wide profiling. Single nucleotide polymorphism sets were defined based on a meta-analysis of genome-wide association studies on systolic blood pressure and diastolicblood pressure performed by the Cohort for Heart and Aging Research in Genome Epidemiology (n = 29 136), using different P value thresholds for selecting single nucleotide polymorphisms. Subsequently, genetic risk scores for systolic blood pressure and diastolic blood pressure were calculated in an independent adult population (n = 2072) and a child population (n = 1034). The explained variance of the genetic risk scores was evaluated using linear regression models, including sex, age, and body mass index. Genetic risk scores, including also many nongenome-wide significant single nucleotide polymorphisms, explained more of the variance than scores based only on very significant single nucleotide polymorphisms in adults and children. Genetic risk scores significantly explained

Simon Heath, Mark Lathrop, Diana Zelenika, Aravinda Chakravarti, Mark J. Caulfield, Daniel Levy, Patricia B. Munroe, Christopher Newton-Cheh, Ayse Demirkan (2011)Genetic variants in novel pathways influence blood pressure and cardiovascular disease risk, In: Nature (London)478(7367)103pp. 103-109 Nature Publishing Group

Blood pressure is a heritable trait1 influenced by several biological pathways and responsive to environmental stimuli. Over one billion people worldwide have hypertension (≥140 mm Hg systolic blood pressure or ≥90 mm Hg diastolic blood pressure)2. Even small increments in blood pressure are associated with an increased risk of cardiovascular events3. This genome-wide association study of systolic and diastolic blood pressure, which used a multi-stage design in 200,000 individuals of European descent, identified sixteen novel loci: six of these loci contain genes previously known or suspected to regulate blood pressure (GUCY1A3-GUCY1B3, NPR3-C5orf23, ADM, FURIN-FES, GOSR2, GNAS-EDN3); the other ten provide new clues to blood pressure physiology. A genetic risk score based on 29 genome-wide significant variants was associated with hypertension, left ventricular wall thickness, stroke and coronary artery disease, but not kidney disease or kidney function. We also observed associations with blood pressure in East Asian, South Asian and African ancestry individuals. Our findings provide new insights into the genetics and biology of blood pressure, and suggest potential novel therapeutic pathways for cardiovascular disease prevention

Sara M Willems, Daniel J Wright, Felix R Day, Katerina Trajanoska, Peter K Joshi, John A Morris, Amy M Matteini, Fleur C Garton, Niels Grarup, Nikolay Oskolkov, Anbupalam Thalamuthu, Massimo Mangino, Jun Liu, Ayse Demirkan, Monkol Lek, Liwen Xu, Guan Wang, Christopher Oldmeadow, Kyle J Gaulton, Luca A Lotta, Eri Miyamoto-Mikami, Manuel A Rivas, Tom White, Po-Ru Loh, Mette Aadahl, Najaf Amin, John R Attia, Krista Austin, Beben Benyamin, Søren Brage, Yu-Ching Cheng, Paweł Cięszczyk, Wim Derave, Karl-Fredrik Eriksson, Nir Eynon, Allan Linneberg, Alejandro Lucia, Myosotis Massidda, Braxton D Mitchell, Motohiko Miyachi, Haruka Murakami, Sandosh Padmanabhan, Ashutosh Pandey, Ioannis Papadimitriou, Deepak K Rajpal, Craig Sale, Theresia M Schnurr, Francesco Sessa, Nick Shrine, Martin D Tobin, Ian Varley, Louise V Wain, Naomi R Wray, Cecilia M Lindgren, Daniel G MacArthur, Dawn M Waterworth, Mark I McCarthy, Oluf Pedersen, Kay-Tee Khaw, Douglas P Kiel, Yannis Pitsiladis, Noriyuki Fuku, Paul W Franks, Kathryn N North, Cornelia M van Duijn, Karen A Mather, Torben Hansen, Ola Hansson, Tim Spector, Joanne M Murabito, J Brent Richards, Fernando Rivadeneira, Claudia Langenberg, John R B Perry, Nick J Wareham, Robert A Scott (2017)Large-scale GWAS identifies multiple loci for hand grip strength providing biological insights into muscular fitness, In: Nature communications8(1)16015pp. 16015-16015

Hand grip strength is a widely used proxy of muscular fitness, a marker of frailty, and predictor of a range of morbidities and all-cause mortality. To investigate the genetic determinants of variation in grip strength, we perform a large-scale genetic discovery analysis in a combined sample of 195,180 individuals and identify 16 loci associated with grip strength (P

Claudia Tamar Silva, Irina V. Zorkoltseva, Maartje N. Niemeijer, Marten E. van den Berg, Najaf Amin, Ayşe Demirkan, Elisa van Leeuwen, Adriana I. Iglesias, Laura B. Piñeros-Hernández, Carlos M. Restrepo, Jan A. Kors, Anatoly V. Kirichenko, Rob Willemsen, Ben A. Oostra, Bruno H. Stricker, André G. Uitterlinden, Tatiana I. Axenovich, Cornelia M. van Duijn, Aaron Isaacs (2018)A combined linkage, microarray and exome analysis suggests MAP3K11 as a candidate gene for left ventricular hypertrophy, In: BMC medical genomics11(1)22pp. 22-22 BioMed Central
Burcak Vural, Fatmahan Atalar, Cavlan Ciftci, Ayse Demirkan, Belgin Susleyici-Duman, Demet Gunay, Belhhan Akpinar, Ertan Sagbas, Ugur Ozbek, Ahmet Sevim Buyukdevrim (2008)Presence of fatty-acid-binding protein 4 expression in human epicardial adipose tissue in metabolic syndrome, In: Cardiovascular pathology17(6)392pp. 392-398 Elsevier Inc

Metabolic syndrome is a cluster of different clinical manifestations that are risk factors for atherothrombotic cardiovascular disorders. Fatty-acid-binding protein 4 (FABP4/aP2), which is highly expressed in adipocytes, specifically exerts intracellular lipid trafficking. A high level of fatty-acid-binding protein 4 expression present in obese subjects has also been found in mice and humans, especially in macrophages at atherosclerotic lesions. An in vivo study demonstrated that the inhibitor of aP2 would be a new therapeutic agent for treating metabolic diseases in mice. We have investigated the mRNA expression of fatty-acid-binding protein 4 in human epicardial adipose and ascending aorta tissues of metabolic syndrome and nonmetabolic syndrome patients. Paired epicardial adipose and ascending aorta tissue samples were obtained from 10 metabolic syndrome patients and 4 nonmetabolic syndrome patients during coronary bypass grafting and aortic valve replacement therapy, respectively. Fatty-acid-binding protein 4 gene expression was determined by quantitative real-time polymerase chain reaction. Fatty-acid-binding protein 4 expression of epicardial adipose tissue was significantly higher in metabolic syndrome patients than in nonmetabolic syndrome controls ( P

Jon B. Toledo, Matthias Arnold, Gabi Kastenmueller, Rui Chang, Rebecca A. Baillie, Xianlin Han, Madhav Thambisetty, Jessica D. Tenenbaum, Karsten Suhre, J. Will Thompson, Lisa St John-Williams, Siamak MahmoudianDehkordi, Daniel M. Rotroff, John R. Jack, Alison Motsinger-Reif, Shannon L. Risacher, Colette Blach, Joseph E. Lucas, Tyler Massaro, Gregory Louie, Hongjie Zhu, Guido Dallmann, Kristaps Klavins, Therese Koal, Sungeun Kim, Kwangsik Nho, Li Shen, Ramon Casanova, Sudhir Varma, Cristina Legido-Quigley, M. Arthur Moseley, Kuixi Zhu, Marc Y. R. Henrion, Sven J. van der Lee, Amy C. Harms, Ayse Demirkan, Thomas Hankemeier, Cornelia M. van Duijn, John Q. Trojanowski, Leslie M. Shaw, Andrew J. Saykin, Michael W. Weiner, P. Murali Doraiswamy, Rima Kaddurah-Daouk (2017)Metabolic network failures in Alzheimer's disease: A biochemical road map, In: Alzheimer's & dementia13(9)965pp. 965-984 Wiley

Introduction: The Alzheimer's Disease Research Summits of 2012 and 2015 incorporated experts from academia, industry, and nonprofit organizations to develop new research directions to transform our understanding of Alzheimer's disease (AD) and propel the development of critically needed therapies. In response to their recommendations, big data at multiple levels are being generated and integrated to study network failures in disease. We used metabolomics as a global biochemical approach to identify peripheral metabolic changes in AD patients and correlate them to cerebrospinal fluid pathology markers, imaging features, and cognitive performance. Methods: Fasting serum samples from the Alzheimer's Disease Neuroimaging Initiative (199 control, 356 mild cognitive impairment, and 175 AD participants) were analyzed using the AbsoluteIDQ-p180 kit. Performance was validated in blinded replicates, and values were medication adjusted. Results: Multivariable-adjusted analyses showed that sphingomyelins and ether-containing phosphatidylcholines were altered in preclinical biomarker-defined AD stages, whereas acylcarnitines and several amines, including the branched-chain amino acid valine and alpha-aminoadipic acid, changed in symptomatic stages. Several of the analytes showed consistent associations in the Rotterdam, Erasmus Rucphen Family, and Indiana Memory and Aging Studies. Partial correlation networks constructed for A beta(1-42), tau, imaging, and cognitive changes provided initial biochemical insights for disease-related processes. Coexpression networks interconnected key metabolic effectors of disease. Discussion: Metabolomics identified key disease-related metabolic changes and disease-progression-related changes. Defining metabolic changes during AD disease trajectory and its relationship to clinical phenotypes provides a powerful roadmap for drug and biomarker discovery. (C) 2017 the Alzheimer's Association. Published by Elsevier Inc. All rights reserved.

Jun Liu, Elena Carnero-Montoro, Jenny van Dongen, Samantha Lent, Ivana Nedeljkovic, Symen Ligthart, Pei-Chien Tsai, Tiphaine C. Martin, Pooja R. Mandaviya, Rick Jansen, Marjolein J. Peters, Liesbeth Duijts, Vincent W. V. Jaddoe, Henning Tiemeier, Janine F. Felix, Gonneke Willemsen, Eco J. C. de Geus, Audrey Y. Chu, Daniel Levy, Shih-Jen Hwang, Jan Bressler, Rahul Gondalia, Elias L. Salfati, Christian Herder, Bertha A. Hidalgo, Toshiko Tanaka, Ann Zenobia Moore, Rozenn N. Lemaitre, Min A. Jhun, Jennifer A. Smith, Nona Sotoodehnia, Stefania Bandinelli, Luigi Ferrucci, Donna K. Arnett, Harald Grallert, Themistocles L. Assimes, Lifang Hou, Andrea Baccarelli, Eric A. Whitsel, Ko Willems van Dijk, Najaf Amin, Andre G. Uitterlinden, Eric J. G. Sijbrands, Oscar H. Franco, Abbas Dehghan, Tim D. Spector, Josee Dupuis, Marie-France Hivert, Jerome Rotter, James B. Meigs, James S. Pankow, Joyce B. J. van Meurs, Aaron Isaacs, Dorret Boomsma, Jordana T. Bell, Ayse Demirkan, Cornelia M. van Duijn (2019)An integrative cross-omics analysis of DNA methylation sites of glucose and insulin homeostasis, In: Nature communications10(1)2581pp. 2581-11 NATURE PORTFOLIO

Despite existing reports on differential DNA methylation in type 2 diabetes (T2D) and obesity, our understanding of its functional relevance remains limited. Here we show the effect of differential methylation in the early phases of T2D pathology by a blood-based epigenome-wide association study of 4808 non-diabetic Europeans in the discovery phase and 11,750 individuals in the replication. We identify CpGs in LETM1, RBM20, IRS2, MAN2A2 and the 1q25.3 region associated with fasting insulin, and in FCRL6, SLAMF1, APOBEC3H and the 15q26.1 region with fasting glucose. In silico cross-omics analyses highlight the role of differential methylation in the crosstalk between the adaptive immune system and glucose homeostasis. The differential methylation explains at least 16.9% of the association between obesity and insulin. Our study sheds light on the biological interactions between genetic variants driving differential methylation and gene expression in the early pathogenesis of T2D.

Pervin Vural, Sevgin Degirmencioglu, Neslihan Y. Saral, Ayse Demirkan, Cemil Akgul, Gokhan Yildirim, Halim Issever, Hacer Eroglu (2010)Tumor necrosis factor alpha, interleukin-6 and interleukin-10 polymorphisms in preeclampsia, In: The journal of obstetrics and gynaecology research36(1)64pp. 64-71 Wiley

Aim: Preeclampsia (PE) is one of the most serious disorders of pregnancy. The imbalance between pro- and anti-inflammatory cytokines may play a role in its etiology. The aim of the present study was to investigate whether cytokine gene polymorphism is associated with PE, and to evaluate the relationship between genotypes and clinical/laboratory manifestation of PE. Methods: We investigated single nucleotide polymorphisms of tumor necrosis factor (TNF)alpha(-308 G/A), interleukin (IL)-6 (-174 G/C), IL-10 (-1082 G/A) genes in DNA from peripheral blood leukocytes of 101 PE patients and 95 healthy control women. Results: In PE, there was a significant increase of the IL-10 (-1082) A allele frequency (P = 0.04). No significant differences were found in genotypes or allele frequencies of TNF alpha(-308) and IL-6 (-174) genes between PE women and controls. While TNF alpha(-308) and IL-6 (-174) genotypes did not influence clinical/laboratory parameters in PE, IL-10 (-1082) A allele carrying genotypes (AG + AA) were associated with higher glucose and lower HDL-cholesterol levels. Conclusion: Because women with IL-10 (-1082) AA genotype have 3.38-fold increased risk of developing PE according to GG genotype (95% CI 1.21-9.4, P = 0.01), we suggest that IL-10 (-1082) variant A allele is associated with an increased risk of preeclampsia, which is independent from its metabolic effects.

Harmen H. M. Draisma, René Pool, Michael Kobl, Rick Jansen, Ann-Kristin Petersen, Anika A. M. Vaarhorst, Idil Yet, Toomas Haller, Ayşe Demirkan, Tõnu Esko, Gu Zhu, Stefan Böhringer, Marian Beekman, Jan Bert van Klinken, Werner Römisch-Margl, Cornelia Prehn, Jerzy Adamski, Anton J. M. de Craen, Elisabeth M. van Leeuwen, Najaf Amin, Harish Dharuri, Harm-Jan Westra, Lude Franke, Eco J. C. de Geus, Jouke Jan Hottenga, Gonneke Willemsen, Anjali K. Henders, Grant W. Montgomery, Dale R. Nyholt, John B. Whitfield, Brenda W. Penninx, Tim D. Spector, Andres Metspalu, P. Eline Slagboom, Ko Willems van Dijk, Peter A. C. ‘t Hoen, Konstantin Strauch, Nicholas G. Martin, Gert-Jan B. van Ommen, Thomas Illig, Jordana T. Bell, Massimo Mangino, Karsten Suhre, Mark I. McCarthy, Christian Gieger, Aaron Isaacs, Cornelia M. van Duijn, Dorret I. Boomsma (2015)Genome-wide association study identifies novel genetic variants contributing to variation in blood metabolite levels, In: Nature communications6(1)7208pp. 7208-7208 Nature Publishing Group UK

Metabolites are small molecules involved in cellular metabolism, which can be detected in biological samples using metabolomic techniques. Here we present the results of genome-wide association and meta-analyses for variation in the blood serum levels of 129 metabolites as measured by the Biocrates metabolomic platform. In a discovery sample of 7,478 individuals of European descent, we find 4,068 genome- and metabolome-wide significant ( Z -test, P

Ervin R. Fox, J. Hunter Young, Yali Li, Albert W. Dreisbach, Brendan J. Keating, Solomon K. Musani, Kiang Liu, Alanna C. Morrison, Santhi Ganesh, Abdullah Kutlar, Vasan S. Ramachandran, Josef F. Polak, Richard R. Fabsitz, Daniel L. Dries, Deborah N. Farlow, Susan Redline, Adebowale Adeyemo, Joel N. Hirschorn, Yan V. Sun, Sharon B. Wyatt, Alan D. Penman, Walter Palmas, Jerome I. Rotter, Raymond R. Townsend, Ayo P. Doumatey, Bamidele O. Tayo, Thomas H. Mosley, Helen N. Lyon, Sun J. Kang, Charles N. Rotimi, Richard S. Cooper, Nora Franceschini, J. David Curb, Lisa W. Martin, Charles B. Eaton, Sharon L.R. Kardia, Herman A. Taylor, Mark J. Caulfield, Georg B. Ehret, Toby Johnson, Aravinda Chakravarti, Xiaofeng Zhu, Daniel Levy, Patricia B. Munroe, Kenneth M. Rice, Murielle Bochud, Andrew D. Johnson, Daniel I. Chasman, Albert V. Smith, Martin D. Tobin, Germaine C. Verwoert, Shih-Jen Hwang, Vasyl Pihur, Peter Vollenweider, Paul F. O'Reilly, Najaf Amin, Jennifer L. Bragg-Gresham, Alexander Teumer, Nicole L. Glazer, Lenore Launer, Jing Hua Zhao, Yurii Aulchenko, Simon Heath, Siim Sõber, Afshin Parsa, Jian'an Luan, Pankaj Arora, Abbas Dehghan, Feng Zhang, Gavin Lucas, Andrew A. Hicks, Anne U. Jackson, John F. Peden, Toshiko Tanaka, Sarah H. Wild, Igor Rudan, Wilmar Igl, Yuri Milaneschi, Alex N. Parker, Cristiano Fava, John C. Chambers, Meena Kumari, Min JinGo, Pim van der Harst, Wen Hong Linda Kao, Marketa Sjögren, D.G. Vinay, Myriam Alexander, Yasuharu Tabara, Sue Shaw-Hawkins, Peter H. Whincup, Yongmei Liu, Gang Shi, Johanna Kuusisto, Mark Seielstad, Xueling Sim, Khanh-Dung Hoang Nguyen, Terho Lehtimäki, Giuseppe Matullo, Ayse Demirkan (2011)Association of genetic variation with systolic and diastolic blood pressure among African Americans: the Candidate Gene Association Resource study, In: Human molecular genetics20(11)ddr092pp. 2273-2284 Oxford University Press

The prevalence of hypertension in African Americans (AAs) is higher than in other US groups; yet, few have performed genome-wide association studies (GWASs) in AA. Among people of European descent, GWASs have identified genetic variants at 13 loci that are associated with blood pressure. It is unknown if these variants confer susceptibility in people of African ancestry. Here, we examined genome-wide and candidate gene associations with systolic blood pressure (SBP) and diastolic blood pressure (DBP) using the Candidate Gene Association Resource (CARe) consortium consisting of 8591 AAs. Genotypes included genome-wide single-nucleotide polymorphism (SNP) data utilizing the Affymetrix 6.0 array with imputation to 2.5 million HapMap SNPs and candidate gene SNP data utilizing a 50K cardiovascular gene-centric array (ITMAT-Broad-CARe [IBC] array). For Affymetrix data, the strongest signal for DBP was rs10474346 ( P = 3.6 × 10 −8 ) located near GPR98 and ARRDC3 . For SBP, the strongest signal was rs2258119 in C21orf91 ( P = 4.7 × 10 −8 ). The top IBC association for SBP was rs2012318 ( P = 6.4 × 10 −6 ) near SLC25A42 and for DBP was rs2523586 ( P = 1.3 × 10 −6 ) near HLA-B . None of the top variants replicated in additional AA ( n = 11 882) or European-American ( n = 69 899) cohorts. We replicated previously reported European-American blood pressure SNPs in our AA samples ( SH2B3 , P = 0.009; TBX3-TBX5 , P = 0.03; and CSK-ULK3 , P = 0.0004). These genetic loci represent the best evidence of genetic influences on SBP and DBP in AAs to date. More broadly, this work supports that notion that blood pressure among AAs is a trait with genetic underpinnings but also with significant complexity.

P. Saip, F. Sen, B. Vural, E. Ugurel, A. Demirkan, D. Derin, Y. Eralp, H. Camlica, Z. Ustuner, U. Ozbek (2011)Glutathione S-transferase P1 polymorphisms are associated with time to tumor progression in small cell lung cancer patients, In: Journal of B.U. ON16(2)pp. 241-246 ZERBINIS MEDICAL PUBL

Purpose: Many of commonly used chemotherapeutics in lung cancer treatment are metabolized by glutathione-S transferases (GSTs). The placental isoform of GST (GSTP1) is the most abundant isoform in the lung. Polymorphisms within the GSTP1 may result in alterations in enzyme activity and change sensitivity to platinum-based chemotherapy. We investigated whether the polymorphism within the exons 5 and 6 of GSTP1 gene may change response to therapy, time to tumor progression (TTP) and overall survival in small cell lung cancer (SCLC) patients. Methods: Ninety-four histologically confirmed patients with SCLC were enrolled in this study during 1995-2006. GSTP1 Ile105Val polymorphism in exon 5 and GSTP1 Ala114Val polymorphism in exon 6 were determined by using PCR-RFLP techniques. Associations between the GSTP1 polymorphisms and treatment response were evaluated using the chi-square test. Associations between the GSTP1 polymorphisms and TTP and overall survival were compared using Kaplan-Meier survival curves. Results: We found no significant associations between exon 5 and exon 6 GSTP1 gene polymorphisms and response to therapy or overall survival. Patients carrying both variant exon 5 (Ile/Val or Val/Val) and variant exon 6 (Ala/Val) genotypes had significantly shorter TTP (5 vs. 8 months, p = 0.04). Moreover, patients with heterozygote exon 6 variant had presented with extensive-stage disease. Conclusion: No individual effect of variant alleles was found in relation to chemotherapy response, median TTP and overall survival. The carriage of both types of variant alleles may predict worse outcome.

Vincent Pascat, Liudmila Zudina, Anna Ulrich, Jared G. Maina, Marika Kaakinen, Igors Pupko, Amélie Bonnefond, Ayse Demirkan, Zhanna Balkhiyarova, Philippe Froguel, Inga Prokopenko (2024)comorbidPGS: an R package assessing shared predisposition between Phenotypes using Polygenic Scores, In: Human Heredity1 Karger Publishers

Introduction Polygenic Score (PGS) is a valuable method for assessing the estimated genetic liability to a given outcome or genetic variability contributing to a quantitative trait. While PRSs are widely used for complex traits, their application in uncovering shared genetic predisposition between phenotypes, i.e. when genetic variants influence more than one phenotype, remains limited. Methods We developed an R package, comorbidPGS, which facilitates a systematic evaluation of shared genetic effects among (cor)related phenotypes using PGSs. The comorbidPGS package takes as input a set of Single Nucleotide Polymorphisms (SNPs) along with their established effects on the original phenotype (Po), referred to as Po-PGS. It generates a comprehensive summary of effect(s) of Po-PGS on target phenotype(s) (Pt) with customisable graphical features. Results We applied comorbidPGS to investigate the shared genetic predisposition between phenotypes defining elevated blood pressure (Systolic Blood Pressure, SBP; Diastolic Blood Pressure, DBP; Pulse Pressure, PP) and several cancers (Breast Cancer, BrC; Pancreatic Cancer, PanC; Kidney Cancer, KidC; Prostate Cancer, PrC; Colorectal Cancer, CrC) using the European ancestry UK Biobank individuals and GWAS meta-analyses summary statistics from independent set of European ancestry individuals. We report a significant association between elevated DBP and the genetic risk of PrC (β (SE)=0.066 (0.017), P-value=9.64×10^(-5)), as well as between CrC PGS and both, lower SBP (β (SE)=-0.10 [0.029], P-value=3.83×10^(-4))) and lower DBP (β (SE)=-0.055 [0.017], P-value=1.05×10^(-3)). Our analysis highlights two nominally significant relationships for individuals with genetic predisposition to elevated SBP leading to higher risk of KidC (OR [95%CI]=1.04 [1.0039-1.087], P-value=2.82×10^(-2)) and PrC (OR [95%CI]=1.02 [1.003-1.041], P-value=2.22×10^(-2)). Conclusion Using comorbidPGS, we underscore mechanistic relationships between blood pressure regulation and susceptibility to three comorbid malignancies. This package offers valuable means to evaluate shared genetic susceptibility between (cor)related phenotypes through polygenic scores.

Nabeel Merali, Julien Terroire, Maria Danae Jessel, Ayse Demirkan, Nicola Annells, Adam Frampton (2024)Bile Microbiome Signatures as Biomarkers for Differentiating Pancreatic Ductal Adenocarcinoma from Benign Disease: Discovery of novel microbial signatures in a UK pilot study, In: European journal of surgical oncology50(1)107154

Background: The intra-tumoural microbiome can influence pancreatic tumourigenesis and chemoresistance, and therefore patient survival. The role played by bile microbiota in PDAC is unknown. We aimed to define bile microbiome signatures in patients presenting with obstructive jaundice caused by benign and malignant pancreaticobiliary disease to develop novel cancer biomarkers. Methods: Prospective bile samples were obtained from 37 patients who underwent either endoscopic retrograde cholangiopancreatography (ERCP) or percutaneous transhepatic cholangiography (PTC). Variable regions (V3–V4) of the 16S rRNA genes were amplified by PCR and next generation sequencing was performed. The cohort consisted of 12 PDAC, 6 cholangiocarcinoma, 10 choledocholithiasis, 7 gallstone pancreatitis and 2 primary sclerosing cholangitis patients. Bile samples from 8 patients were excluded from the analysis because of low read count. Results: Using the 16S rRNA method, we identified a total of 108 genera from 29 individuals (12 PDAC and 17 benign). Bile microbial diversity significantly differed between patients with PDAC vs. benign disease (p=0.0173). The separation of PDAC from benign samples is clearly seen through unsupervised clustering based on Canberra distances. We found 4 genera to be of significantly different abundance between PDAC vs. benign groups by association p-value and supported by false discovery rate (fdr). These were Escherichia, Rothia, Streptococcus and Prevotella. Conclusion: We show that patients with obstructive jaundice caused by PDAC have an altered microbiome composition in the bile, compared to those with benign disease. These bile-based microbes could be developed into potential diagnostic and prognostic biomarkers for PDAC and warrant further investigation.

Zhanna Balkhiiarova, Saqib Hassan, Marika Kaakinen, Harmen Draisma, Liudmila Zudina, Mohd A Ganie, Aafia Rashid, Zhanna Balkhiyarova, George S Kiran, Paris Vogazianos, Christos Shammas, Joseph Selvin, Athos Antoniades, Ayse Demirkan, Inga Prokopenko (2022)Bifidobacterium Is Enriched in Gut Microbiome of Kashmiri Women with Polycystic Ovary Syndrome, In: Genes13(2)

Polycystic ovary syndrome (PCOS) is a very common endocrine condition in women in India. Gut microbiome alterations were shown to be involved in PCOS, yet it is remarkably understudied in Indian women who have a higher incidence of PCOS as compared to other ethnic populations. During the regional PCOS screening program among young women, we recruited 19 drug naive women with PCOS and 20 control women at the Sher-i-Kashmir Institute of Medical Sciences, Kashmir, North India. We profiled the gut microbiome in faecal samples by 16S rRNA sequencing and included 40/58 operational taxonomic units (OTUs) detected in at least 1/3 of the subjects with relative abundance (RA) ≥ 0.1%. We compared the RAs at a family/genus level in PCOS/non-PCOS groups and their correlation with 33 metabolic and hormonal factors, and corrected for multiple testing, while taking the variation in day of menstrual cycle at sample collection, age and BMI into account. Five genera were significantly enriched in PCOS cases: , , and previously reported for PCOS , and confirmed by different statistical models. At the family level, the relative abundance of was enriched, whereas was decreased among cases. We observed increased relative abundance of and with higher fasting blood glucose levels, and and with larger hip, waist circumference, weight, and with lower prolactin levels. We also detected a novel association between and follicle-stimulating hormone levels and between and alkaline phosphatase, independently of the BMI of the participants. Our report supports that there is a relationship between gut microbiome composition and PCOS with links to specific reproductive health metabolic and hormonal predictors in Indian women.

Jun Liu, Lies Lahousse, Michel G Nivard, Mariska Bot, Lianmin Chen, Jan Bert van Klinken, Carisha S Thesing, Marian Beekman, Erik Ben van den Akker, Roderick C Slieker, Eveline Waterham, Carla J H van der Kallen, Irene de Boer, Ruifang Li-Gao, Dina Vojinovic, Najaf Amin, Djawad Radjabzadeh, Robert Kraaij, Louise J M Alferink, Sarwa Darwish Murad, André G Uitterlinden, Gonneke Willemsen, Rene Pool, Yuri Milaneschi, Diana van Heemst, H Eka D Suchiman, Femke Rutters, Petra J M Elders, Joline W J Beulens, Amber A W A van der Heijden, Marleen M J van Greevenbroek, Ilja C W Arts, Gerrit L J Onderwater, Arn M J M van den Maagdenberg, Dennis O Mook-Kanamori, Thomas Hankemeier, Gisela M Terwindt, Coen D A Stehouwer, Johanna M Geleijnse, Leen M 't Hart, P Eline Slagboom, Ko Willems van Dijk, Alexandra Zhernakova, Jingyuan Fu, Brenda W J H Penninx, Dorret I Boomsma, Ayşe Demirkan, Bruno H C Stricker, Cornelia M van Duijn (2020)Integration of epidemiologic, pharmacologic, genetic and gut microbiome data in a drug-metabolite atlas, In: Nature medicine26(1)110pp. 110-117

Progress in high-throughput metabolic profiling provides unprecedented opportunities to obtain insights into the effects of drugs on human metabolism. The Biobanking BioMolecular Research Infrastructure of the Netherlands has constructed an atlas of drug-metabolite associations for 87 commonly prescribed drugs and 150 clinically relevant plasma-based metabolites assessed by proton nuclear magnetic resonance. The atlas includes a meta-analysis of ten cohorts (18,873 persons) and uncovers 1,071 drug-metabolite associations after evaluation of confounders including co-treatment. We show that the effect estimates of statins on metabolites from the cross-sectional study are comparable to those from intervention and genetic observational studies. Further data integration links proton pump inhibitors to circulating metabolites, liver function, hepatic steatosis and the gut microbiome. Our atlas provides a tool for targeted experimental pharmaceutical research and clinical trials to improve drug efficacy, safety and repurposing. We provide a web-based resource for visualization of the atlas (http://bbmri.researchlumc.nl/atlas/).

Shahzad Ahmad, Marta Del Campo Milan, Oskar Hansson, Ayse Demirkan, Ruiz Agustin, Maria E Sáez, Nikolaos Giagtzoglou, Alfredo Cabrera-Socorro, Margot H M Bakker, Alfredo Ramirez, Thomas Hankemeier, Erik Stomrud, Niklas Mattsson-Carlgren, Philip Scheltens, Wiesje M van der Flier, M Arfan Ikram, Anders Malarstig, Charlotte E Teunissen, Najaf Amin, Cornelia M van Duijn (2020)CDH6 and HAGH protein levels in plasma associate with Alzheimer's disease in APOE ε4 carriers, In: Scientific reports10(1)8233pp. 8233-8233

Many Alzheimer's disease (AD) genes including Apolipoprotein E (APOE) are found to be expressed in blood-derived macrophages and thus may alter blood protein levels. We measured 91 neuro-proteins in plasma from 316 participants of the Rotterdam Study (incident AD = 161) using Proximity Extension Ligation assay. We studied the association of plasma proteins with AD in the overall sample and stratified by APOE. Findings from the Rotterdam study were replicated in 186 AD patients of the BioFINDER study. We further evaluated the correlation of these protein biomarkers with total tau (t-tau), phosphorylated tau (p-tau) and amyloid-beta (Aβ) 42 levels in cerebrospinal fluid (CSF) in the Amsterdam Dementia Cohort (N = 441). Finally, we conducted a genome-wide association study (GWAS) to identify the genetic variants determining the blood levels of AD-associated proteins. Plasma levels of the proteins, CDH6 (β = 0.638, P = 3.33 × 10 ) and HAGH (β = 0.481, P = 7.20 × 10 ), were significantly elevated in APOE ε4 carrier AD patients. The findings in the Rotterdam Study were replicated in the BioFINDER study for both CDH6 (β = 1.365, P = 3.97 × 10 ) and HAGH proteins (β = 0.506, P = 9.31 × 10 ) when comparing cases and controls in APOE ε4 carriers. In the CSF, CDH6 levels were positively correlated with t-tau and p-tau in the total sample as well as in APOE ε4 stratum (P 

Josephine H. Li, Laura N. Brenner, Varinderpal Kaur, Katherine Figueroa, Philip Schroeder, Alicia Huerta-Chagoya, Miriam S. Udler, Aaron Leong, Josep M. Mercader, Jose C. Florez, Magic Investigators, Diab Prevent Program DPP Res Grp, Ayse Demirkan (2023)Genome-wide association analysis identifies ancestry-specific genetic variation associated with acute response to metformin and glipizide in SUGAR-MGH, In: Diabetologia66(7)pp. 1260-1272 Springer Nature

Aims/hypothesis Characterisation of genetic variation that influences the response to glucose-lowering medications is instrumental to precision medicine for treatment of type 2 diabetes. The Study to Understand the Genetics of the Acute Response to Metformin and Glipizide in Humans (SUGAR-MGH) examined the acute response to metformin and glipizide in order to identify new pharmacogenetic associations for the response to common glucose-lowering medications in individuals at risk of type 2 diabetes.Methods One thousand participants at risk for type 2 diabetes from diverse ancestries underwent sequential glipizide and metformin challenges. A genome-wide association study was performed using the Illumina Multi-Ethnic Genotyping Array. Imputation was performed with the TOPMed reference panel. Multiple linear regression using an additive model tested for association between genetic variants and primary endpoints of drug response. In a more focused analysis, we evaluated the influence of 804 unique type 2 diabetes- and glycaemic trait-associated variants on SUGAR-MGH outcomes and performed colocalisation analyses to identify shared genetic signals.Results Five genome-wide significant variants were associated with metformin or glipizide response. The strongest association was between an African ancestry-specific variant (minor allele frequency [MAF(Afr)]=0.0283) at rs149403252 and lower fasting glucose at Visit 2 following metformin (p=1.9x10(-9)); carriers were found to have a 0.94 mmol/l larger decrease in fasting glucose. rs111770298, another African ancestry-specific variant (MAF(Afr)=0.0536), was associated with a reduced response to metformin (p=2.4x10(-8)), where carriers had a 0.29 mmol/l increase in fasting glucose compared with non-carriers, who experienced a 0.15 mmol/l decrease. This finding was validated in the Diabetes Prevention Program, where rs111770298 was associated with a worse glycaemic response to metformin: heterozygous carriers had an increase in HbA(1c) of 0.08% and non-carriers had an HbA(1c) increase of 0.01% after 1 year of treatment (p=3.3x10(-3)). We also identified associations between type 2 diabetes-associated variants and glycaemic response, including the type 2 diabetes-protective C allele of rs703972 near ZMIZ1 and increased levels of active glucagon-like peptide 1 (GLP-1) (p=1.6x10(-5)), supporting the role of alterations in incretin levels in type 2 diabetes pathophysiology.Conclusions/interpretation We present a well-phenotyped, densely genotyped, multi-ancestry resource to study gene-drug interactions, uncover novel variation associated with response to common glucose-lowering medications and provide insight into mechanisms of action of type 2 diabetes-related variation.

Peitao Wu, Denis Rybin, Lawrence F. Bielak, Mary F. Feitosa, Nora Franceschini, Yize Li, Yingchang Lu, Jonathan Marten, Solomon K. Musani, Raymond Noordam, Sridharan Raghavan, Lynda M. Rose, Karen Schwander, Albert Smith, Salman M. Tajuddin, Dina Vojinovic, Najaf Amin, Donna K. Arnett, Erwin P. Bottinger, Ayse Demirkan, Jose C. Florez, Mohsen Ghanbari, Tamara B. Harris, Lenore J. Launer, Jingmin Liu, Jun Liu, Dennis O. Mook-Kanamori, Alison D. Murray, Mike A. Nalls, Patricia A. Peyser, Andre G. Uitterlinden, Trudy Voortman, Claude Bouchard, Daniel Chasman, Adolfo Correa, Renee de Mutsert, Michele K. Evans, Vilmundur Gudnason, Caroline Hayward, Linda Kao, Sharon L. R. Kardia, Charles Kooperberg, Ruth J. F. Loos, Michael M. Province, Tuomo Rankinen, Susan Redline, Paul M. Ridker, Jerome Rotter, David Siscovick, Blair H. Smith, Cornelia van Duijn, Alan B. Zonderman, D. C. Rao, James G. Wilson, Josee Dupuis, James B. Meigs, Ching-Ti Liu, Jason L. Vassy (2020)Smoking-by-genotype interaction in type 2 diabetes risk and fasting glucose, In: PloS one15(5)0230815pp. e0230815-e0230815 Public Library Science

Smoking is a potentially causal behavioral risk factor for type 2 diabetes (T2D), but not all smokers develop T2D. It is unknown whether genetic factors partially explain this variation. We performed genome-environment-wide interaction studies to identify loci exhibiting potential interaction with baseline smoking status (ever vs. never) on incident T2D and fasting glucose (FG). Analyses were performed in participants of European (EA) and African ancestry (AA) separately. Discovery analyses were conducted using genotype data from the 50,000-single-nucleotide polymorphism (SNP) ITMAT-Broad-CARe (IBC) array in 5 cohorts from from the Candidate Gene Association Resource Consortium (n = 23,189). Replication was performed in up to 16 studies from the Cohorts for Heart Aging Research in Genomic Epidemiology Consortium (n = 74,584). In meta-analysis of discovery and replication estimates, 5 SNPs met at least one criterion for potential interaction with smoking on incident T2D at p< 1x10(-7) (adjusted for multiple hypothesis-testing with the IBC array). Two SNPs had significant joint effects in the overall model and significant main effects only in one smoking stratum: rs140637 (FBN1) in AA individuals had a significant main effect only among smokers, and rs1444261 (closest gene C2orf63) in EA individuals had a significant main effect only among nonsmokers. Three additional SNPs were identified as having potential interaction by exhibiting a significant main effects only in smokers: rs1801232 (CUBN) in AA individuals, rs12243326 (TCF7L2) in EA individuals, and rs4132670 (TCF7L2) in EA individuals. No SNP met significance for potential interaction with smoking on baseline FG. The identification of these loci provides evidence for genetic interactions with smoking exposure that may explain some of the heterogeneity in the association between smoking and T2D.

Xiaoyan Yin, Christine M. Willinger, Joshua Keefe, Jun Liu, Antonio Fernández-Ortiz, Borja Ibáñez, José Peñalvo, Aram Adourian, George Chen, Dolores Corella, Reinald Pamplona, Manuel Portero-Otin, Mariona Jove, Paul Courchesne, Cornelia M. van Duijn, Valentín Fuster, José M. Ordovás, Ayşe Demirkan, Martin G. Larson, Daniel Levy (2020)Lipidomic profiling identifies signatures of metabolic risk, In: EBioMedicine51102520pp. 102520-102520 Elsevier B.V

Metabolic syndrome (MetS), the clustering of metabolic risk factors, is associated with cardiovascular disease risk. We sought to determine if dysregulation of the lipidome may contribute to metabolic risk factors. We measured 154 circulating lipid species in 658 participants from the Framingham Heart Study (FHS) using liquid chromatography-tandem mass spectrometry and tested for associations with obesity, dysglycemia, and dyslipidemia. Independent external validation was sought in three independent cohorts. Follow-up data from the FHS were used to test for lipid metabolites associated with longitudinal changes in metabolic risk factors. Thirty-nine lipids were associated with obesity and eight with dysglycemia in the FHS. Of 32 lipids that were available for replication for obesity and six for dyslipidemia, 28 (88%) replicated for obesity and five (83%) for dysglycemia. Four lipids were associated with longitudinal changes in body mass index and four were associated with changes in fasting blood glucose in the FHS. We identified and replicated several novel lipid biomarkers of key metabolic traits. The lipid moieties identified in this study are involved in biological pathways of metabolic risk and can be explored for prognostic and therapeutic utility.

Fiona A Hagenbeek, René Pool, Jenny van Dongen, Harmen H M Draisma, Jouke Jan Hottenga, Gonneke Willemsen, Abdel Abdellaoui, Iryna O Fedko, Anouk den Braber, Pieter Jelle Visser, Eco J C N de Geus, Ko Willems van Dijk, Aswin Verhoeven, H Eka Suchiman, Marian Beekman, P Eline Slagboom, Cornelia M van Duijn, Amy C Harms, Thomas Hankemeier, Meike Bartels, Michel G Nivard, Dorret I Boomsma, Ayse Demirkan (2020)Heritability estimates for 361 blood metabolites across 40 genome-wide association studies, In: Nature communications11(1)39pp. 39-39

Metabolomics examines the small molecules involved in cellular metabolism. Approximately 50% of total phenotypic differences in metabolite levels is due to genetic variance, but heritability estimates differ across metabolite classes. We perform a review of all genome-wide association and (exome-) sequencing studies published between November 2008 and October 2018, and identify >800 class-specific metabolite loci associated with metabolite levels. In a twin-family cohort (N = 5117), these metabolite loci are leveraged to simultaneously estimate total heritability (h ), and the proportion of heritability captured by known metabolite loci (h ) for 309 lipids and 52 organic acids. Our study reveals significant differences in h among different classes of lipids and organic acids. Furthermore, phosphatidylcholines with a high degree of unsaturation have higher h estimates than phosphatidylcholines with low degrees of unsaturation. This study highlights the importance of common genetic variants for metabolite levels, and elucidates the genetic architecture of metabolite classes.

Alexander Kurilshikov, Carolina Medina-Gomez, Rodrigo Bacigalupe, Djawad Radjabzadeh, Jun Wang, Ayse Demirkan, Caroline Le Roy, Juan Antonio Raygoza Garay, Casey T. Finnicum, Xingrong Liu, Daria Zhernakova, Marc Jan Bonder, Tue H. Hansen, Fabian Frost, Malte C. Ruhlemann, Williams Turpin, Jee-Young Moon, Han-Na Kim, Kreete Lull, Elad Barkan, Shiraz A. Shah, Myriam Fornage, Joanna Szopinska-Tokov, Zachary D. Wallen, Dmitrii Borisevich, Lars Agreus, Anna Andreasson, Corinna Bang, Larbi Bedrani, Jordana T. Bell, Hans Bisgaard, Michael Boehnke, Dorret Boomsma, Robert D. Burk, Annique Claringbould, Kenneth Croitoru, Gareth E. Davies, Cornelia M. van Duijn, Liesbeth Duijts, Gwen Falony, Jingyuan Fu, Adriaan van der Graaf, Torben Hansen, Georg Homuth, David A. Hughes, Richard G. Ijzerman, Matthew A. Jackson, Vincent W. V. Jaddoe, Marie Joossens, Torben Jorgensen, Daniel Keszthelyi, Rob Knight, Markku Laakso, Matthias Laudes, Lenore J. Launer, Wolfgang Lieb, Aldons J. Lusis, Ad A. M. Masclee, Henriette A. Moll, Zlatan Mujagic, Qi Qibin, Daphna Rothschild, Hocheol Shin, Soren J. Sorensen, Claire J. Steves, Jonathan Thorsen, Nicholas J. Timpson, Raul Y. Tito, Sara Vieira-Silva, Uwe Volker, Henry Volzke, Urmo Vosa, Kaitlin H. Wade, Susanna Walter, Kyoko Watanabe, Stefan Weiss, Frank U. Weiss, Omer Weissbrod, Harm-Jan Westra, Gonneke Willemsen, Haydeh Payami, Daisy M. A. E. Jonkers, Alejandro Arias Vasquez, Eco J. C. de Geus, Katie A. Meyer, Jakob Stokholm, Eran Segal, Elin Org, Cisca Wijmenga, Hyung-Lae Kim, Robert C. Kaplan, Tim D. Spector, Andre G. Uitterlinden, Fernando Rivadeneira, Andre Franke, Markus M. Lerch, Lude Franke, Serena Sanna, Mauro D'Amato, Oluf Pedersen, Andrew D. Paterson, Robert Kraaij, Jeroen Raes, Alexandra Zhernakova (2021)Large-scale association analyses identify host factors influencing human gut microbiome composition, In: Nature genetics53(2)pp. 156-165 NATURE PORTFOLIO

To study the effect of host genetics on gut microbiome composition, the MiBioGen consortium curated and analyzed genome-wide genotypes and 16S fecal microbiome data from 18,340 individuals (24 cohorts). Microbial composition showed high variability across cohorts: only 9 of 410 genera were detected in more than 95% of samples. A genome-wide association study of host genetic variation regarding microbial taxa identified 31 loci affecting the microbiome at a genome-wide significant (P < 5 x 10(-8)) threshold. One locus, the lactase (LCT) gene locus, reached study-wide significance (genome-wide association study signal: P = 1.28 x 10(-20)), and it showed an age-dependent association with Bifidobacterium abundance. Other associations were suggestive (1.95 x 10(-10) < P < 5 x 10(-8)) but enriched for taxa showing high heritability and for genes expressed in the intestine and brain. A phenome-wide association study and Mendelian randomization identified enrichment of microbiome trait loci in the metabolic, nutrition and environment domains and suggested the microbiome might have causal effects in ulcerative colitis and rheumatoid arthritis.

Zachary D Wallen, Ayse Demirkan, Guy Twa, Gwendolyn Cohen, Marissa N Dean, David G Standaert, Timothy R Sampson, Haydeh Payami (2022)Metagenomics of Parkinson's disease implicates the gut microbiome in multiple disease mechanisms, In: Nature communications13(1)6958pp. 6958-6958

Parkinson's disease (PD) may start in the gut and spread to the brain. To investigate the role of gut microbiome, we conducted a large-scale study, at high taxonomic resolution, using uniform standardized methods from start to end. We enrolled 490 PD and 234 control individuals, conducted deep shotgun sequencing of fecal DNA, followed by metagenome-wide association studies requiring significance by two methods (ANCOM-BC and MaAsLin2) to declare disease association, network analysis to identify polymicrobial clusters, and functional profiling. Here we show that over 30% of species, genes and pathways tested have altered abundances in PD, depicting a widespread dysbiosis. PD-associated species form polymicrobial clusters that grow or shrink together, and some compete. PD microbiome is disease permissive, evidenced by overabundance of pathogens and immunogenic components, dysregulated neuroactive signaling, preponderance of molecules that induce alpha-synuclein pathology, and over-production of toxicants; with the reduction in anti-inflammatory and neuroprotective factors limiting the capacity to recover. We validate, in human PD, findings that were observed in experimental models; reconcile and resolve human PD microbiome literature; and provide a broad foundation with a wealth of concrete testable hypotheses to discern the role of the gut microbiome in PD.

Stavroula Kanoni, Sarah E Graham, Yuxuan Wang, Ida Surakka, Shweta Ramdas, Xiang Zhu, Shoa L Clarke, Konain Fatima Bhatti, Sailaja Vedantam, Thomas W Winkler, Adam E Locke, Eirini Marouli, Greg J M Zajac, Kuan-Han H Wu, Ioanna Ntalla, Qin Hui, Derek Klarin, Austin T Hilliard, Zeyuan Wang, Chao Xue, Gudmar Thorleifsson, Anna Helgadottir, Daniel F Gudbjartsson, Hilma Holm, Isleifur Olafsson, Mi Yeong Hwang, Sohee Han, Masato Akiyama, Saori Sakaue, Chikashi Terao, Masahiro Kanai, Wei Zhou, Ben M Brumpton, Humaira Rasheed, Aki S Havulinna, Yogasudha Veturi, Jennifer Allen Pacheco, Elisabeth A Rosenthal, Todd Lingren, QiPing Feng, Iftikhar J Kullo, Akira Narita, Jun Takayama, Hilary C Martin, Karen A Hunt, Bhavi Trivedi, Jeffrey Haessler, Franco Giulianini, Yuki Bradford, Jason E Miller, Archie Campbell, Kuang Lin, Iona Y Millwood, Asif Rasheed, George Hindy, Jessica D Faul, Wei Zhao, David R Weir, Constance Turman, Hongyan Huang, Mariaelisa Graff, Ananyo Choudhury, Dhriti Sengupta, Anubha Mahajan, Michael R Brown, Weihua Zhang, Ketian Yu, Ellen M Schmidt, Anita Pandit, Stefan Gustafsson, Xianyong Yin, Jian'an Luan, Jing-Hua Zhao, Fumihiko Matsuda, Hye-Mi Jang, Kyungheon Yoon, Carolina Medina-Gomez, Achilleas Pitsillides, Jouke Jan Hottenga, Andrew R Wood, Yingji Ji, Zishan Gao, Simon Haworth, Noha A Yousri, Ruth E Mitchell, Jin Fang Chai, Mette Aadahl, Anne A Bjerregaard, Jie Yao, Ani Manichaikul, Chii-Min Hwu, Yi-Jen Hung, Helen R Warren, Julia Ramirez, Jette Bork-Jensen, Line L Kårhus, Anuj Goel, Maria Sabater-Lleal, Raymond Noordam, Pala Mauro, Floris Matteo, Aaron F McDaid, Pedro Marques-Vidal, Matthias Wielscher, Stella Trompet, Naveed Sattar, Line T Møllehave, Matthias Munz, Lingyao Zeng, Jianfeng Huang, Bin Yang, Alaitz Poveda, Azra Kurbasic, Claudia Lamina, Lukas Forer, Markus Scholz, Tessel E Galesloot, Jonathan P Bradfield, Sanni E Ruotsalainen, EWarwick Daw, Joseph M Zmuda, Jonathan S Mitchell, Christian Fuchsberger, Henry Christensen, Jennifer A Brody, Miguel Vazquez-Moreno, Mary F Feitosa, Mary K Wojczynski, Zhe Wang, Michael H Preuss, Massimo Mangino, Paraskevi Christofidou, Niek Verweij, Jan W Benjamins, Jorgen Engmann, Noah L Tsao, Anurag Verma, Roderick C Slieker, Ken Sin Lo, Nuno R Zilhao, Phuong Le, Marcus E Kleber, Graciela E Delgado, Shaofeng Huo, Daisuke D Ikeda, Hiroyuki Iha, Jian Yang, Jun Liu, Ayşe Demirkan, Hampton L Leonard, Jonathan Marten, Mirjam Frank, Börge Schmidt, Laura J Smyth, Marisa Cañadas-Garre, Chaolong Wang, Masahiro Nakatochi, Andrew Wong, Nina Hutri-Kähönen, Xueling Sim, Rui Xia, Alicia Huerta-Chagoya, Juan Carlos Fernandez-Lopez, Valeriya Lyssenko, Suraj S Nongmaithem, Swati Bayyana, Heather M Stringham, Marguerite R Irvin, Christopher Oldmeadow, Han-Na Kim, Seungho Ryu, Paul R H J Timmers, Liubov Arbeeva, Rajkumar Dorajoo, Leslie A Lange, Gauri Prasad, Laura Lorés-Motta, Marc Pauper, Jirong Long, Xiaohui Li, Elizabeth Theusch, Fumihiko Takeuchi, Cassandra N Spracklen, Anu Loukola, Sailalitha Bollepalli, Sophie C Warner, Ya Xing Wang, Wen B Wei, Teresa Nutile, Daniela Ruggiero, Yun Ju Sung, Shufeng Chen, Fangchao Liu, Jingyun Yang, Katherine A Kentistou, Bernhard Banas, Giuseppe Giovanni Nardone, Karina Meidtner, Lawrence F Bielak, Jennifer A Smith, Prashantha Hebbar, Aliki-Eleni Farmaki, Edith Hofer, Maoxuan Lin, Maria Pina Concas, Simona Vaccargiu, Peter J van der Most, Niina Pitkänen, Brian E Cade, Sander W van der Laan, Kumaraswamy Naidu Chitrala, Stefan Weiss, Amy R Bentley, Ayo P Doumatey, Adebowale A Adeyemo, Jong Young Lee, Eva R B Petersen, Aneta A Nielsen, Hyeok Sun Choi, Maria Nethander, Sandra Freitag-Wolf, Lorraine Southam, Nigel W Rayner, Carol A Wang, Shih-Yi Lin, Jun-Sing Wang, Christian Couture, Leo-Pekka Lyytikäinen, Kjell Nikus, Gabriel Cuellar-Partida, Henrik Vestergaard, Bertha Hidalgo, Olga Giannakopoulou, Qiuyin Cai, Morgan O Obura, Jessica van Setten, Xiaoyin Li, Jingjing Liang, Hua Tang, Natalie Terzikhan, Jae Hun Shin, Rebecca D Jackson, Alexander P Reiner, Lisa Warsinger Martin, Zhengming Chen, Liming Li, Takahisa Kawaguchi, Joachim Thiery, Joshua C Bis, Lenore J Launer, Huaixing Li, Mike A Nalls, Olli T Raitakari, Sahoko Ichihara, Sarah H Wild, Christopher P Nelson, Harry Campbell, Susanne Jäger, Toru Nabika, Fahd Al-Mulla, Harri Niinikoski, Peter S Braund, Ivana Kolcic, Peter Kovacs, Tota Giardoglou, Tomohiro Katsuya, Dominique de Kleijn, Gert J de Borst, Eung Kweon Kim, Hieab H H Adams, M Arfan Ikram, Xiaofeng Zhu, Folkert W Asselbergs, Adriaan O Kraaijeveld, Joline W J Beulens, Xiao-Ou Shu, Loukianos S Rallidis, Oluf Pedersen, Torben Hansen, Paul Mitchell, Alex W Hewitt, Mika Kähönen, Louis Pérusse, Claude Bouchard, Anke Tönjes, Yii-Der Ida Chen, Craig E Pennell, Trevor A Mori, Wolfgang Lieb, Andre Franke, Claes Ohlsson, Dan Mellström, Yoon Shin Cho, Hyejin Lee, Jian-Min Yuan, Woon-Puay Koh, Sang Youl Rhee, Jeong-Taek Woo, Iris M Heid, Klaus J Stark, Martina E Zimmermann, Henry Völzke, Georg Homuth, Michele K Evans, Alan B Zonderman, Ozren Polasek, Gerard Pasterkamp, Imo E Hoefer, Susan Redline, Katja Pahkala, Albertine J Oldehinkel, Harold Snieder, Ginevra Biino, Reinhold Schmidt, Helena Schmidt, Stefania Bandinelli, George Dedoussis, Thangavel Alphonse Thanaraj, Sharon L R Kardia, Patricia A Peyser, Norihiro Kato, Matthias B Schulze, Giorgia Girotto, Carsten A Böger, Bettina Jung, Peter K Joshi, David A Bennett, Philip L De Jager, Xiangfeng Lu, Vasiliki Mamakou, Morris Brown, Mark J Caulfield, Patricia B Munroe, Xiuqing Guo, Marina Ciullo, Jost B Jonas, Nilesh J Samani, Jaakko Kaprio, Päivi Pajukanta, Teresa Tusié-Luna, Carlos A Aguilar-Salinas, Linda S Adair, Sonny Augustin Bechayda, H Janaka de Silva, Ananda R Wickremasinghe, Ronald M Krauss, Jer-Yuarn Wu, Wei Zheng, Anneke Iden Hollander, Dwaipayan Bharadwaj, Adolfo Correa, James G Wilson, Lars Lind, Chew-Kiat Heng, Amanda E Nelson, Yvonne M Golightly, James F Wilson, Brenda Penninx, Hyung-Lae Kim, John Attia, Rodney J Scott, D C Rao, Donna K Arnett, Steven C Hunt, Mark Walker, Heikki A Koistinen, Giriraj R Chandak, Josep M Mercader, Maria C Costanzo, Dongkeun Jang, Noël P Burtt, Clicerio Gonzalez Villalpando, Lorena Orozco, Myriam Fornage, EShyong Tai, Rob M van Dam, Terho Lehtimäki, Nish Chaturvedi, Mitsuhiro Yokota, Jianjun Liu, Dermot F Reilly, Amy Jayne McKnight, Frank Kee, Karl-Heinz Jöckel, Mark I McCarthy, Colin N A Palmer, Veronique Vitart, Caroline Hayward, Eleanor Simonsick, Cornelia M van Duijn, Zi-Bing Jin, Jia Qu, Haretsugu Hishigaki, Xu Lin, Winfried März, Vilmundur Gudnason, Jean-Claude Tardif, Guillaume Lettre, Leen M 't Hart, Petra J M Elders, Scott M Damrauer, Meena Kumari, Mika Kivimaki, Pim van der Harst, Tim D Spector, Ruth J F Loos, Michael A Province, Esteban J Parra, Miguel Cruz, Bruce M Psaty, Ivan Brandslund, Peter P Pramstaller, Charles N Rotimi, Kaare Christensen, Samuli Ripatti, Elisabeth Widén, Hakon Hakonarson, Struan F A Grant, Lambertus A L M Kiemeney, Jacqueline de Graaf, Markus Loeffler, Florian Kronenberg, Dongfeng Gu, Jeanette Erdmann, Heribert Schunkert, Paul W Franks, Allan Linneberg, J Wouter Jukema, Amit V Khera, Minna Männikkö, Marjo-Riitta Jarvelin, Zoltan Kutalik, Cucca Francesco, Dennis O Mook-Kanamori, Ko Willems van Dijk, Hugh Watkins, David P Strachan, Niels Grarup, Peter Sever, Neil Poulter, Lee-Ming Chuang, Jerome I Rotter, Thomas M Dantoft, Fredrik Karpe, Matt J Neville, Nicholas J Timpson, Ching-Yu Cheng, Tien-Yin Wong, Chiea Chuen Khor, Hengtong Li, Charumathi Sabanayagam, Annette Peters, Christian Gieger, Andrew T Hattersley, Nancy L Pedersen, Patrik K E Magnusson, Dorret I Boomsma, Allegonda H M Willemsen, LAdrienne Cupples, Joyce B J van Meurs, Mohsen Ghanbari, Penny Gordon-Larsen, Wei Huang, Young Jin Kim, Yasuharu Tabara, Nicholas J Wareham, Claudia Langenberg, Eleftheria Zeggini, Johanna Kuusisto, Markku Laakso, Erik Ingelsson, Goncalo Abecasis, John C Chambers, Jaspal S Kooner, Paul S de Vries, Alanna C Morrison, Scott Hazelhurst, Michèle Ramsay, Kari E North, Martha Daviglus, Peter Kraft, Nicholas G Martin, John B Whitfield, Shahid Abbas, Danish Saleheen, Robin G Walters, Michael V Holmes, Corri Black, Blair H Smith, Aris Baras, Anne E Justice, Julie E Buring, Paul M Ridker, Daniel I Chasman, Charles Kooperberg, Gen Tamiya, Masayuki Yamamoto, David A van Heel, Richard C Trembath, Wei-Qi Wei, Gail P Jarvik, Bahram Namjou, M Geoffrey Hayes, Marylyn D Ritchie, Pekka Jousilahti, Veikko Salomaa, Kristian Hveem, Bjørn Olav Åsvold, Michiaki Kubo, Yoichiro Kamatani, Yukinori Okada, Yoshinori Murakami, Bong-Jo Kim, Unnur Thorsteinsdottir, Kari Stefansson, Jifeng Zhang, YEugene Chen, Yuk-Lam Ho, Julie A Lynch, Daniel J Rader, Philip S Tsao, Kyong-Mi Chang, Kelly Cho, Christopher J O'Donnell, John M Gaziano, Peter W F Wilson, Timothy M Frayling, Joel N Hirschhorn, Sekar Kathiresan, Karen L Mohlke, Yan V Sun, Andrew P Morris, Michael Boehnke, Christopher D Brown, Pradeep Natarajan, Panos Deloukas, Cristen J Willer, Themistocles L Assimes, Gina M Peloso (2022)Implicating genes, pleiotropy, and sexual dimorphism at blood lipid loci through multi-ancestry meta-analysis, In: Genome biology23(1)268pp. 268-268

Genetic variants within nearly 1000 loci are known to contribute to modulation of blood lipid levels. However, the biological pathways underlying these associations are frequently unknown, limiting understanding of these findings and hindering downstream translational efforts such as drug target discovery. To expand our understanding of the underlying biological pathways and mechanisms controlling blood lipid levels, we leverage a large multi-ancestry meta-analysis (N = 1,654,960) of blood lipids to prioritize putative causal genes for 2286 lipid associations using six gene prediction approaches. Using phenome-wide association (PheWAS) scans, we identify relationships of genetically predicted lipid levels to other diseases and conditions. We confirm known pleiotropic associations with cardiovascular phenotypes and determine novel associations, notably with cholelithiasis risk. We perform sex-stratified GWAS meta-analysis of lipid levels and show that 3-5% of autosomal lipid-associated loci demonstrate sex-biased effects. Finally, we report 21 novel lipid loci identified on the X chromosome. Many of the sex-biased autosomal and X chromosome lipid loci show pleiotropic associations with sex hormones, emphasizing the role of hormone regulation in lipid metabolism. Taken together, our findings provide insights into the biological mechanisms through which associated variants lead to altered lipid levels and potentially cardiovascular disease risk.

Hanieh Yaghootkar, Yiying Zhang, Cassandra N Spracklen, Tugce Karaderi, Lam Opal Huang, Jonathan Bradfield, Claudia Schurmann, Rebecca S Fine, Michael H Preuss, Zoltan Kutalik, Laura B L Wittemans, Yingchang Lu, Sophia Metz, Sara M Willems, Ruifang Li-Gao, Niels Grarup, Shuai Wang, Sophie Molnos, América A Sandoval-Zárate, Mike A Nalls, Leslie A Lange, Jeffrey Haesser, Xiuqing Guo, Leo-Pekka Lyytikäinen, Mary F Feitosa, Colleen M Sitlani, Cristina Venturini, Anubha Mahajan, Tim Kacprowski, Carol A Wang, Daniel I Chasman, Najaf Amin, Linda Broer, Neil Robertson, Kristin L Young, Matthew Allison, Paul L Auer, Matthias Blüher, Judith B Borja, Jette Bork-Jensen, Germán D Carrasquilla, Paraskevi Christofidou, Ayse Demirkan, Claudia A Doege, Melissa E Garcia, Mariaelisa Graff, Kaiying Guo, Hakon Hakonarson, Jaeyoung Hong, Yii-Der Ida Chen, Rebecca Jackson, Hermina Jakupović, Pekka Jousilahti, Anne E Justice, Mika Kähönen, Jorge R Kizer, Jennifer Kriebel, Charles A LeDuc, Jin Li, Lars Lind, Jian'an Luan, David A Mackey, Massimo Mangino, Satu Männistö, Jayne F Martin Carli, Carolina Medina-Gomez, Dennis O Mook-Kanamori, Andrew P Morris, Renée de Mutsert, Matthias Nauck, Ivana Prokic, Craig E Pennell, Arund D Pradhan, Bruce M Psaty, Olli T Raitakari, Robert A Scott, Tea Skaaby, Konstantin Strauch, Kent D Taylor, Alexander Teumer, Andre G Uitterlinden, Ying Wu, Jie Yao, Mark Walker, Kari E North, Peter Kovacs, M Arfan Ikram, Cornelia M van Duijn, Paul M Ridker, Stephen Lye, Georg Homuth, Erik Ingelsson, Tim D Spector, Barbara McKnight, Michael A Province, Terho Lehtimäki, Linda S Adair, Jerome I Rotter, Alexander P Reiner, James G Wilson, Tamara B Harris, Samuli Ripatti, Harald Grallert, James B Meigs, Veikko Salomaa, Torben Hansen, Ko Willems van Dijk, Nicholas J Wareham, Struan F A Grant, Claudia Langenberg, Timothy M Frayling, Cecilia M Lindgren, Karen L Mohlke, Rudolph L Leibel, Ruth J F Loos, Tuomas O Kilpeläinen (2020)Genetic Studies of Leptin Concentrations Implicate Leptin in the Regulation of Early Adiposity, In: Diabetes (New York, N.Y.)69(12)2806pp. 2806-2818

Leptin influences food intake by informing the brain about the status of body fat stores. Rare mutations associated with congenital leptin deficiency cause severe early-onset obesity that can be mitigated by administering leptin. However, the role of genetic regulation of leptin in polygenic obesity remains poorly understood. We performed an exome-based analysis in up to 57,232 individuals of diverse ancestries to identify genetic variants that influence adiposity-adjusted leptin concentrations. We identify five novel variants, including four missense variants, in , , , and , and one intergenic variant near The missense variant Val94Met (rs17151919) in was common in individuals of African ancestry only, and its association with lower leptin concentrations was specific to this ancestry ( = 2 × 10 , = 3,901). Using in vitro analyses, we show that the Met94 allele decreases leptin secretion. We also show that the Met94 allele is associated with higher BMI in young African-ancestry children but not in adults, suggesting that leptin regulates early adiposity.

Vasiliki Lagou, Longda Jiang, Anna Ulrich, Liudmila Zudina, Ayse Demirkan, Karla Sofia Gutiérrez González, Marika Kaakinen, Zhanna Balkhiiarova, Inga Prokopenko, Alessia Faggian, Jared G. Maina, Shiqian Chen, Petar V. Todorov, Sodbo Sharapov, Alessia David, Letizia Marullo, Reedik Magi, Gudmar Thorleifsson, He Gao, Roxana-Maria Rujan, Emma Ahlqvist, Evangelos Evangelou, Beben Benyamin, Robert A Scott, Aaron Isaacs, Jing Hua Zhao, Sara M. Willems, Toby Johnson, Christian Gieger, Harald Grallert, Christa Meisinger, Martina Mueller-Nurasyid, Rona J Strawbridge, Anuj Goel, Denis Rybin, Eva Albrecht, Anne U Jackson, Heather M Stringham, Ivan R., Jr Correa, Eric Farber-Eger, Valgerdur Steinthorsdottir, Andre G. Uitterlinden, Patricia B. Munroe, Morris J. Brown, Julian Schmidberger, Oddgeir Holmen, Barbara Thorand, Kristian Hveem, Tom Wilsgaard, Karen L Mohlke, Zhe Wang, Aleksey Shmeliov, Marcel den Hoed, Ruth J F Loos, Wolfgang Kratzer, Mark Haenle, Wolfgang Koenig, Bernhard O. Boehm, Tricia M. Tan, Alejandra Tomas, Victoria Salem, Inês Barroso, Jaakko Tuomilehto, Michael Boehnke, Jose C. Florez, Anders Hamsten, Hugh Watkins, Inger Njolstad, H-Erich Wichmann, Mark J Caulfield, Kay-Tee Khaw, Cornelia van Duijn, Albert Hofman, Nicholas J. Wareham, Claudia Langenberg, John B. Whitfield, Nicholas G. Martin, Grant Montgomery, Chiara Scapoli, Ioanna Tzoulaki, Paul Elliott, Unnur Thorsteinsdottir, Kari Stefansson, Evan L. Brittain, MI McCarthy, Philippe Froguel, Patrick M. Sexton, Denise Wootten, Leif Groop, Josée Dupuis, James B Meigs, Giuseppe Deganutti, Tune H. Pers, Christopher A. Reynolds, Yurii S. Aulchenko, Ben Jones (2023)GWAS of random glucose in 476,326 individuals provide insights into diabetes pathophysiology, complications and treatment stratification, In: Nature Genetics55(9)pp. 1448-1461 Nature Research

Conventional measurements of fasting and postprandial blood glucose levels investigated in genome-wide association studies (GWAS) cannot capture the effects of DNA variability on 'around the clock' glucoregulatory processes. Here we show that GWAS meta-analysis of glucose measurements under nonstandardized conditions (random glucose (RG)) in 476,326 individuals of diverse ancestries and without diabetes enables locus discovery and innovative pathophysiological observations. We discovered 120 RG loci represented by 150 distinct signals, including 13 with sex-dimorphic effects, two cross-ancestry and seven rare frequency signals. Of these, 44 loci are new for glycemic traits. Regulatory, glycosylation and metagenomic annotations highlight ileum and colon tissues, indicating an underappreciated role of the gastrointestinal tract in controlling blood glucose. Functional follow-up and molecular dynamics simulations of lower frequency coding variants in glucagon-like peptide-1 receptor (GLP1R), a type 2 diabetes treatment target, reveal that optimal selection of GLP-1R agonist therapy will benefit from tailored genetic stratification. We also provide evidence from Mendelian randomization that lung function is modulated by blood glucose and that pulmonary dysfunction is a diabetes complication. Our investigation yields new insights into the biology of glucose regulation, diabetes complications and pathways for treatment stratification. Genome-wide association analyses of blood glucose measurements under nonstandardized conditions provide insights into the biology of glucose regulation, diabetes complications and pathways for treatment stratification.

Ji Chen, Cassandra N Spracklen, Gaëlle Marenne, Arushi Varshney, Laura J Corbin, Jian'an Luan, Sara M Willems, Ying Wu, Xiaoshuai Zhang, Momoko Horikoshi, Thibaud S Boutin, Reedik Mägi, Johannes Waage, Ruifang Li-Gao, Kei Hang Katie Chan, Jie Yao, Mila D Anasanti, Audrey Y Chu, Annique Claringbould, Jani Heikkinen, Jaeyoung Hong, Jouke-Jan Hottenga, Shaofeng Huo, Marika A Kaakinen, Tin Louie, Winfried März, Hortensia Moreno-Macias, Anne Ndungu, Sarah C Nelson, Ilja M Nolte, Kari E North, Chelsea K Raulerson, Debashree Ray, Rebecca Rohde, Denis Rybin, Claudia Schurmann, Xueling Sim, Lorraine Southam, Isobel D Stewart, Carol A Wang, Yujie Wang, Peitao Wu, Weihua Zhang, Tarunveer S Ahluwalia, Emil V R Appel, Lawrence F Bielak, Jennifer A Brody, Noël P Burtt, Claudia P Cabrera, Brian E Cade, Jin Fang Chai, Xiaoran Chai, Li-Ching Chang, Chien-Hsiun Chen, Brian H Chen, Kumaraswamy Naidu Chitrala, Yen-Feng Chiu, Hugoline G de Haan, Graciela E Delgado, Ayse Demirkan, Qing Duan, Jorgen Engmann, Segun A Fatumo, Javier Gayán, Franco Giulianini, Jung Ho Gong, Stefan Gustafsson, Yang Hai, Fernando P Hartwig, Jing He, Yoriko Heianza, Tao Huang, Alicia Huerta-Chagoya, Mi Yeong Hwang, Richard A Jensen, Takahisa Kawaguchi, Katherine A Kentistou, Young Jin Kim, Marcus E Kleber, Ishminder K Kooner, Shuiqing Lai, Leslie A Lange, Carl D Langefeld, Marie Lauzon, Man Li, Symen Ligthart, Jun Liu, Marie Loh, Jirong Long, Valeriya Lyssenko, Massimo Mangino, Carola Marzi, May E Montasser, Abhishek Nag, Masahiro Nakatochi, Damia Noce, Raymond Noordam, Giorgio Pistis, Michael Preuss, Laura Raffield, Laura J Rasmussen-Torvik, Stephen S Rich, Neil R Robertson, Rico Rueedi, Kathleen Ryan, Serena Sanna, Richa Saxena, Katharina E Schraut, Bengt Sennblad, Kazuya Setoh, Albert V Smith, Thomas Sparsø, Rona J Strawbridge, Fumihiko Takeuchi, Jingyi Tan, Stella Trompet, Erik van den Akker, Peter J van der Most, Niek Verweij, Mandy Vogel, Heming Wang, Chaolong Wang, Nan Wang, Helen R Warren, Wanqing Wen, Tom Wilsgaard, Andrew Wong, Andrew R Wood, Tian Xie, Mohammad Hadi Zafarmand, Jing-Hua Zhao, Wei Zhao, Najaf Amin, Zorayr Arzumanyan, Arne Astrup, Stephan J L Bakker, Damiano Baldassarre, Marian Beekman, Richard N Bergman, Alain Bertoni, Matthias Blüher, Lori L Bonnycastle, Stefan R Bornstein, Donald W Bowden, Qiuyin Cai, Archie Campbell, Harry Campbell, Yi Cheng Chang, Eco J C de Geus, Abbas Dehghan, Shufa Du, Gudny Eiriksdottir, Aliki Eleni Farmaki, Mattias Frånberg, Christian Fuchsberger, Yutang Gao, Anette P Gjesing, Anuj Goel, Sohee Han, Catharina A Hartman, Christian Herder, Andrew A Hicks, Chang-Hsun Hsieh, Willa A Hsueh, Sahoko Ichihara, Michiya Igase, M Arfan Ikram, W Craig Johnson, Marit E Jørgensen, Peter K Joshi, Rita R Kalyani, Fouad R Kandeel, Tomohiro Katsuya, Chiea Chuen Khor, Wieland Kiess, Ivana Kolcic, Teemu Kuulasmaa, Johanna Kuusisto, Kristi Läll, Kelvin Lam, Deborah A Lawlor, Nanette R Lee, Rozenn N Lemaitre, Honglan Li, Shih-Yi Lin, Jaana Lindström, Allan Linneberg, Jianjun Liu, Carlos Lorenzo, Tatsuaki Matsubara, Fumihiko Matsuda, Geltrude Mingrone, Simon Mooijaart, Sanghoon Moon, Toru Nabika, Girish N Nadkarni, Jerry L Nadler, Mari Nelis, Matt J Neville, Jill M Norris, Yasumasa Ohyagi, Annette Peters, Patricia A Peyser, Ozren Polasek, Qibin Qi, Dennis Raven, Dermot F Reilly, Alex Reiner, Fernando Rivideneira, Kathryn Roll, Igor Rudan, Charumathi Sabanayagam, Kevin Sandow, Naveed Sattar, Annette Schürmann, Jinxiu Shi, Heather M Stringham, Kent D Taylor, Tanya M Teslovich, Betina Thuesen, Paul R H J Timmers, Elena Tremoli, Michael Y Tsai, Andre Uitterlinden, Rob M van Dam, Diana van Heemst, Astrid van Hylckama Vlieg, Jana V van Vliet-Ostaptchouk, Jagadish Vangipurapu, Henrik Vestergaard, Tao Wang, Ko Willems van Dijk, Tatijana Zemunik, Gonçalo R Abecasis, Linda S Adair, Carlos Alberto Aguilar-Salinas, Marta E Alarcón-Riquelme, Ping An, Larissa Aviles-Santa, Diane M Becker, Lawrence J Beilin, Sven Bergmann, Hans Bisgaard, Corri Black, Michael Boehnke, Eric Boerwinkle, Bernhard O Böhm, Klaus Bønnelykke, D I Boomsma, Erwin P Bottinger, Thomas A Buchanan, Mickaël Canouil, Mark J Caulfield, John C Chambers, Daniel I Chasman, Yii-Der Ida Chen, Ching-Yu Cheng, Francis S Collins, Adolfo Correa, Francesco Cucca, H Janaka de Silva, George Dedoussis, Sölve Elmståhl, Michele K Evans, Ele Ferrannini, Luigi Ferrucci, Jose C Florez, Paul W Franks, Timothy M Frayling, Philippe Froguel, Bruna Gigante, Mark O Goodarzi, Penny Gordon-Larsen, Harald Grallert, Niels Grarup, Sameline Grimsgaard, Leif Groop, Vilmundur Gudnason, Xiuqing Guo, Anders Hamsten, Torben Hansen, Caroline Hayward, Susan R Heckbert, Bernardo L Horta, Wei Huang, Erik Ingelsson, Pankow S James, Marjo-Ritta Jarvelin, Jost B Jonas, J Wouter Jukema, Pontiano Kaleebu, Robert Kaplan, Sharon L R Kardia, Norihiro Kato, Sirkka M Keinanen-Kiukaanniemi, Bong-Jo Kim, Mika Kivimaki, Heikki A Koistinen, Jaspal S Kooner, Antje Körner, Peter Kovacs, Diana Kuh, Meena Kumari, Zoltan Kutalik, Markku Laakso, Timo A Lakka, Lenore J Launer, Karin Leander, Huaixing Li, Xu Lin, Lars Lind, Cecilia Lindgren, Simin Liu, Ruth J F Loos, Patrik K E Magnusson, Anubha Mahajan, Andres Metspalu, Dennis O Mook-Kanamori, Trevor A Mori, Patricia B Munroe, Inger Njølstad, Jeffrey R O'Connell, Albertine J Oldehinkel, Ken K Ong, Sandosh Padmanabhan, Colin N A Palmer, Nicholette D Palmer, Oluf Pedersen, Craig E Pennell, David J Porteous, Peter P Pramstaller, Michael A Province, Bruce M Psaty, Lu Qi, Leslie J Raffel, Rainer Rauramaa, Susan Redline, Paul M Ridker, Frits R Rosendaal, Timo E Saaristo, Manjinder Sandhu, Jouko Saramies, Neil Schneiderman, Peter Schwarz, Laura J Scott, Elizabeth Selvin, Peter Sever, Xiao-Ou Shu, P Eline Slagboom, Kerrin S Small, Blair H Smith, Harold Snieder, Tamar Sofer, Thorkild I A Sørensen, Tim D Spector, Alice Stanton, Claire J Steves, Michael Stumvoll, Liang Sun, Yasuharu Tabara, E Shyong Tai, Nicholas J Timpson, Anke Tönjes, Jaakko Tuomilehto, Teresa Tusie, Matti Uusitupa, Pim van der Harst, Cornelia van Duijn, Veronique Vitart, Peter Vollenweider, Tanja G M Vrijkotte, Lynne E Wagenknecht, Mark Walker, Ya X Wang, Nick J Wareham, Richard M Watanabe, Hugh Watkins, Wen B Wei, Ananda R Wickremasinghe, Gonneke Willemsen, James F Wilson, Tien-Yin Wong, Jer-Yuarn Wu, Anny H Xiang, Lisa R Yanek, Loïc Yengo, Mitsuhiro Yokota, Eleftheria Zeggini, Wei Zheng, Alan B Zonderman, Jerome I Rotter, Anna L Gloyn, Mark I McCarthy, Josée Dupuis, James B Meigs, Robert A Scott, Inga Prokopenko, Aaron Leong, Ching-Ti Liu, Stephen C J Parker, Karen L Mohlke, Claudia Langenberg, Eleanor Wheeler, Andrew P Morris, Inês Barroso (2021)The trans-ancestral genomic architecture of glycemic traits, In: Nature genetics53(6)pp. 840-860

Glycemic traits are used to diagnose and monitor type 2 diabetes and cardiometabolic health. To date, most genetic studies of glycemic traits have focused on individuals of European ancestry. Here we aggregated genome-wide association studies comprising up to 281,416 individuals without diabetes (30% non-European ancestry) for whom fasting glucose, 2-h glucose after an oral glucose challenge, glycated hemoglobin and fasting insulin data were available. Trans-ancestry and single-ancestry meta-analyses identified 242 loci (99 novel; P 

Sara M. Willems, Natasha H. J. Ng, Juan Fernandez, Rebecca S. Fine, Eleanor Wheeler, Jennifer Wessel, Hidetoshi Kitajima, Gaelle Marenne, Xueling Sim, Hanieh Yaghootkar, Shuai Wang, Sai Chen, Yuning Chen, Yii-Der Ida Chen, Niels Grarup, Ruifang Li-Gao, Tibor V. Varga, Jennifer L. Asimit, Shuang Feng, Rona J. Strawbridge, Erica L. Kleinbrink, Tarunveer S. Ahluwalia, Ping An, Emil V. Appel, Dan E. Arking, Juha Auvinen, Lawrence F. Bielak, Nathan A. Bihlmeyer, Jette Bork-Jensen, Jennifer A. Brody, Archie Campbell, Audrey Y. Chu, Gail Davies, Ayse Demirkan, James S. Floyd, Franco Giulianini, Xiuqing Guo, Stefan Gustafsson, Anne U. Jackson, Johanna Jakobsdottir, Marjo-Riitta Jarvelin, Richard A. Jensen, Stavroula Kanoni, Sirkka Keinanen-Kiukaanniemi, Man Li, Yingchang Lu, Jian'an Luan, Alisa K. Manning, Jonathan Marten, Karina Meidtner, Dennis O. Mook-Kanamori, Taulant Muka, Giorgio Pistis, Bram Prins, Kenneth M. Rice, Serena Sanna, Albert Vernon Smith, Jennifer A. Smith, Lorraine Southam, Heather M. Stringham, Vinicius Tragante, Sander W. van der Laan, Helen R. Warren, Jie Yao, Andrianos M. Yiorkas, Weihua Zhang, Wei Zhao, Mariaelisa Graff, Heather M. Highland, Anne E. Justice, Eirini Marouli, Carolina Medina-Gomez, Saima Afaq, Wesam A. Alhejily, Najaf Amin, Folkert W. Asselbergs, Lori L. Bonnycastle, Michiel L. Bots, Ivan Brandslund, Ji Chen, John Danesh, Renée de Mutsert, Abbas Dehghan, Tapani Ebeling, Paul Elliott, Aliki-Eleni Farmaki, Jessica D. Faul, Paul W. Franks, Steve Franks, Andreas Fritsche, Anette P. Gjesing, Mark O. Goodarzi, Vilmundur Gudnason, Göran Hallmans, Tamara B. Harris, Karl-Heinz Herzig, Marie-France Hivert, Min A Jhun, Torben Jørgensen, Marit E. Jørgensen, Pekka Jousilahti, Eero Kajantie, Maria Karaleftheri, Sharon L.R. Kardia, Leena Kinnunen, Heikki A. Koistinen, Pirjo Komulainen, Peter Kovacs, Johanna Kuusisto, Markku Laakso, Leslie A. Lange, Lenore J. Launer, Aaron Leong, Jaana Lindström, Jocelyn E. Manning Fox, Satu Männistö, Nisa M. Maruthur, Leena Moilanen, Antonella Mulas, Mike A. Nalls, Matthew Neville, James S. Pankow, Alison Pattie, Eva R.B. Petersen, Hannu Puolijoki, Asif Rasheed, Paul Redmond, Frida Renström, Michael Roden, Danish Saleheen, Juha Saltevo, Kai Savonen, Sylvain Sebert, Tea Skaaby, Kerrin S. Small, Alena Stančáková, Jakob Stokholm, Konstantin Strauch, E-Shyong Tai, Kent D. Taylor, Betina H. Thuesen, Anke Tönjes, Emmanouil Tsafantakis, Tiinamaija Tuomi, Jaakko Tuomilehto, Matti Uusitupa, Marja Vääräsmäki, Ilonca Vaartjes, Magdalena Zoledziewska, Goncalo Abecasis, Beverley Balkau, Hans Bisgaard, Alexandra I. Blakemore, Matthias Blüher, Heiner Boeing, Eric Boerwinkle, Klaus Bønnelykke, Erwin P. Bottinger, Mark J. Caulfield, John C. Chambers, Daniel I. Chasman, Ching-Yu Cheng, Francis S. Collins, Josef Coresh, Francesco Cucca, Gert J. de Borst, Ian J. Deary, George Dedoussis, Panos Deloukas, Hester M. den Ruijter, Josée Dupuis, Michele K. Evans, Ele Ferrannini, Oscar H. Franco, Harald Grallert, Torben Hansen, Andrew T. Hattersley, Caroline Hayward, Joel N. Hirschhorn, Arfan Ikram, Erik Ingelsson, Fredrik Karpe, Kay-Tee Kaw, Wieland Kiess, Jaspal S. Kooner, Antje Körner, Timo Lakka, Claudia Langenberg, Lars Lind, Cecilia M. Lindgren, Allan Linneberg, Leonard Lipovich, Ching-Ti Liu, Jun Liu, Yongmei Liu, Ruth J.F. Loos, Patrick E. MacDonald, Karen L. Mohlke, Andrew D. Morris, Patricia B. Munroe, Alison Murray, Sandosh Padmanabhan, Colin N. A . Palmer, Gerard Pasterkamp, Oluf Pedersen, Patricia A. Peyser, Ozren Polasek, David Porteous, Michael A. Province, Bruce M. Psaty, Rainer Rauramaa, Paul M. Ridker, Olov Rolandsson, Patrik Rorsman, Frits R. Rosendaal, Igor Rudan, Veikko Salomaa, Matthias B. Schulze, Robert Sladek, Blair H. Smith, Timothy D. Spector, John M. Starr, Michael Stumvoll, Cornelia M. van Duijn, Mark Walker, Nick J. Wareham, David R. Weir, James G. Wilson, Tien Yin Wong, Eleftheria Zeggini, Alan B. Zonderman, Jerome I. Rotter, Andrew P. Morris, Michael Boehnke, Jose C. Florez, Mark I. McCarthy, James B. Meigs, Anubha Mahajan, Robert A. Scott, Anna L. Gloyn, Inês Barroso (2023)Large-scale exome array summary statistics resources for glycemic traits to aid effector gene prioritization, In: Wellcome open research8

Background Genome-wide association studies for glycemic traits have identified hundreds of loci associated with these biomarkers of glucose homeostasis. Despite this success, the challenge remains to link variant associations to genes, and underlying biological pathways. Methods To identify coding variant associations which may pinpoint effector genes at both novel and previously established genome-wide association loci, we performed meta-analyses of exome-array studies for four glycemic traits: glycated hemoglobin (HbA1c, up to 144,060 participants), fasting glucose (FG, up to 129,665 participants), fasting insulin (FI, up to 104,140) and 2hr glucose post-oral glucose challenge (2hGlu, up to 57,878). In addition, we performed network and pathway analyses. Results Single-variant and gene-based association analyses identified coding variant associations at more than 60 genes, which when combined with other datasets may be useful to nominate effector genes. Network and pathway analyses identified pathways related to insulin secretion, zinc transport and fatty acid metabolism. HbA1c associations were strongly enriched in pathways related to blood cell biology. Conclusions Our results provided novel glycemic trait associations and highlighted pathways implicated in glycemic regulation. Exome-array summary statistic results are being made available to the scientific community to enable further discoveries.

Jun Liu, Paul S de Vries, Fabiola Del Greco M, Åsa Johansson, Katharina E Schraut, Caroline Hayward, Ko Willems van Dijk, Oscar H Franco, Andrew A Hicks, Veronique Vitart, Igor Rudan, Harry Campbell, Ozren Polašek, Peter P Pramstaller, James F Wilson, Ulf Gyllensten, Cornelia M van Duijn, Abbas Dehghan, Ayşe Demirkan (2022)A multi-omics study of circulating phospholipid markers of blood pressure, In: Scientific reports12(1)574pp. 574-574

High-throughput techniques allow us to measure a wide-range of phospholipids which can provide insight into the mechanisms of hypertension. We aimed to conduct an in-depth multi-omics study of various phospholipids with systolic blood pressure (SBP) and diastolic blood pressure (DBP). The associations of blood pressure and 151 plasma phospholipids measured by electrospray ionization tandem mass spectrometry were performed by linear regression in five European cohorts (n = 2786 in discovery and n = 1185 in replication). We further explored the blood pressure-related phospholipids in Erasmus Rucphen Family (ERF) study by associating them with multiple cardiometabolic traits (linear regression) and predicting incident hypertension (Cox regression). Mendelian Randomization (MR) and phenome-wide association study (Phewas) were also explored to further investigate these association results. We identified six phosphatidylethanolamines (PE 38:3, PE 38:4, PE 38:6, PE 40:4, PE 40:5 and PE 40:6) and two phosphatidylcholines (PC 32:1 and PC 40:5) which together predicted incident hypertension with an area under the ROC curve (AUC) of 0.61. The identified eight phospholipids are strongly associated with triglycerides, obesity related traits (e.g. waist, waist-hip ratio, total fat percentage, body mass index, lipid-lowering medication, and leptin), diabetes related traits (e.g. glucose, insulin resistance and insulin) and prevalent type 2 diabetes. The genetic determinants of these phospholipids also associated with many lipoproteins, heart rate, pulse rate and blood cell counts. No significant association was identified by bi-directional MR approach. We identified eight blood pressure-related circulating phospholipids that have a predictive value for incident hypertension. Our cross-omics analyses show that phospholipid metabolites in the circulation may yield insight into blood pressure regulation and raise a number of testable hypothesis for future research.

Shweta Ramdas, Jonathan Judd, Sarah E. Graham, Stavroula Kanoni, Yuxuan Wang, Ida Surakka, Brandon Wenz, Shoa L. Clarke, Alessandra Chesi, Andrew Wells, Konain Fatima Bhatti, Sailaja Vedantam, Thomas W. Winkler, Adam E. Locke, Eirini Marouli, Greg J.M. Zajac, Kuan-Han H. Wu, Ioanna Ntalla, Qin Hui, Derek Klarin, Austin T. Hilliard, Zeyuan Wang, Chao Xue, Gudmar Thorleifsson, Anna Helgadottir, Daniel F. Gudbjartsson, Hilma Holm, Isleifur Olafsson, Mi Yeong Hwang, Sohee Han, Masato Akiyama, Saori Sakaue, Chikashi Terao, Masahiro Kanai, Wei Zhou, Ben M. Brumpton, Humaira Rasheed, Aki S. Havulinna, Yogasudha Veturi, Jennifer Allen Pacheco, Elisabeth A. Rosenthal, Todd Lingren, QiPing Feng, Iftikhar J. Kullo, Akira Narita, Jun Takayama, Hilary C. Martin, Karen A. Hunt, Bhavi Trivedi, Jeffrey Haessler, Franco Giulianini, Yuki Bradford, Jason E. Miller, Archie Campbell, Kuang Lin, Iona Y. Millwood, Asif Rasheed, George Hindy, Jessica D. Faul, Wei Zhao, David R. Weir, Constance Turman, Hongyan Huang, Mariaelisa Graff, Ananyo Choudhury, Dhriti Sengupta, Anubha Mahajan, Michael R. Brown, Weihua Zhang, Ketian Yu, Ellen M. Schmidt, Anita Pandit, Stefan Gustafsson, Xianyong Yin, Jian’an Luan, Jing-Hua Zhao, Fumihiko Matsuda, Hye-Mi Jang, Kyungheon Yoon, Carolina Medina-Gomez, Achilleas Pitsillides, Jouke Jan Hottenga, Andrew R. Wood, Yingji Ji, Zishan Gao, Simon Haworth, Ruth E. Mitchell, Jin Fang Chai, Mette Aadahl, Anne A. Bjerregaard, Jie Yao, Ani Manichaikul, Wen-Jane Lee, Chao Agnes Hsiung, Helen R. Warren, Julia Ramirez, Jette Bork-Jensen, Line L. Kårhus, Anuj Goel, Maria Sabater-Lleal, Raymond Noordam, Pala Mauro, Floris Matteo, Aaron F. McDaid, Pedro Marques-Vidal, Matthias Wielscher, Stella Trompet, Naveed Sattar, Line T. Møllehave, Matthias Munz, Lingyao Zeng, Jianfeng Huang, Bin Yang, Alaitz Poveda, Azra Kurbasic, Sebastian Schönherr, Lukas Forer, Markus Scholz, Tessel E. Galesloot, Jonathan P. Bradfield, Sanni E. Ruotsalainen, E. Warwick Daw, Joseph M. Zmuda, Jonathan S. Mitchell, Christian Fuchsberger, Henry Christensen, Jennifer A. Brody, Phuong Le, Mary F. Feitosa, Mary K. Wojczynski, Daiane Hemerich, Michael Preuss, Massimo Mangino, Paraskevi Christofidou, Niek Verweij, Jan W. Benjamins, Jorgen Engmann, Tsao L. Noah, Anurag Verma, Roderick C. Slieker, Ken Sin Lo, Nuno R. Zilhao, Marcus E. Kleber, Graciela E. Delgado, Shaofeng Huo, Daisuke D. Ikeda, Hiroyuki Iha, Jian Yang, Jun Liu, Ayşe Demirkan, Hampton L. Leonard, Jonathan Marten, Carina Emmel, Börge Schmidt, Laura J. Smyth, Marisa Cañadas-Garre, Chaolong Wang, Masahiro Nakatochi, Andrew Wong, Nina Hutri-Kähönen, Xueling Sim, Rui Xia, Alicia Huerta-Chagoya, Juan Carlos Fernandez-Lopez, Valeriya Lyssenko, Suraj S. Nongmaithem, Alagu Sankareswaran, Marguerite R. Irvin, Christopher Oldmeadow, Han-Na Kim, Seungho Ryu, Paul R.H.J. Timmers, Liubov Arbeeva, Rajkumar Dorajoo, Leslie A. Lange, Gauri Prasad, Laura Lorés-Motta, Marc Pauper, Jirong Long, Xiaohui Li, Elizabeth Theusch, Fumihiko Takeuchi, Cassandra N. Spracklen, Anu Loukola, Sailalitha Bollepalli, Sophie C. Warner, Ya Xing Wang, Wen B. Wei, Teresa Nutile, Daniela Ruggiero, Yun Ju Sung, Shufeng Chen, Fangchao Liu, Jingyun Yang, Katherine A. Kentistou, Bernhard Banas, Anna Morgan, Karina Meidtner, Lawrence F. Bielak, Jennifer A. Smith, Prashantha Hebbar, Aliki-Eleni Farmaki, Edith Hofer, Maoxuan Lin, Maria Pina Concas, Simona Vaccargiu, Peter J. van der Most, Niina Pitkänen, Brian E. Cade, Sander W. van der Laan, Kumaraswamy Naidu Chitrala, Stefan Weiss, Amy R. Bentley, Ayo P. Doumatey, Adebowale A. Adeyemo, Jong Young Lee, Eva R.B. Petersen, Aneta A. Nielsen, Hyeok Sun Choi, Maria Nethander, Sandra Freitag-Wolf, Lorraine Southam, Nigel W. Rayner, Carol A. Wang, Shih-Yi Lin, Jun-Sing Wang, Christian Couture, Leo-Pekka Lyytikäinen, Kjell Nikus, Gabriel Cuellar-Partida, Henrik Vestergaard, Bertha Hidalgo, Olga Giannakopoulou, Qiuyin Cai, Morgan O. Obura, Jessica van Setten, Karen Y. He, Hua Tang, Natalie Terzikhan, Jae Hun Shin, Rebecca D. Jackson, Alexander P. Reiner, Lisa Warsinger Martin, Zhengming Chen, Liming Li, Takahisa Kawaguchi, Joachim Thiery, Joshua C. Bis, Lenore J. Launer, Huaixing Li, Mike A. Nalls, Olli T. Raitakari, Sahoko Ichihara, Sarah H. Wild, Christopher P. Nelson, Harry Campbell, Susanne Jäger, Toru Nabika, Fahd Al-Mulla, Harri Niinikoski, Peter S. Braund, Ivana Kolcic, Peter Kovacs, Tota Giardoglou, Tomohiro Katsuya, Dominique de Kleijn, Gert J. de Borst, Eung Kweon Kim, Hieab H.H. Adams, M. Arfan Ikram, Xiaofeng Zhu, Folkert W. Asselbergs, Adriaan O. Kraaijeveld, Joline W.J. Beulens, Xiao-Ou Shu, Loukianos S. Rallidis, Oluf Pedersen, Torben Hansen, Paul Mitchell, Alex W. Hewitt, Mika Kähönen, Louis Pérusse, Claude Bouchard, Anke Tönjes, Yii-Der Ida Chen, Craig E. Pennell, Trevor A. Mori, Wolfgang Lieb, Andre Franke, Claes Ohlsson, Dan Mellström, Yoon Shin Cho, Hyejin Lee, Jian-Min Yuan, Woon-Puay Koh, Sang Youl Rhee, Jeong-Taek Woo, Iris M. Heid, Klaus J. Stark, Martina E. Zimmermann, Henry Völzke, Georg Homuth, Michele K. Evans, Alan B. Zonderman, Ozren Polasek, Gerard Pasterkamp, Imo E. Hoefer, Susan Redline, Katja Pahkala, Albertine J. Oldehinkel, Harold Snieder, Ginevra Biino, Reinhold Schmidt, Helena Schmidt, Stefania Bandinelli, George Dedoussis, Thangavel Alphonse Thanaraj, Patricia A. Peyser, Norihiro Kato, Matthias B. Schulze, Giorgia Girotto, Carsten A. Böger, Bettina Jung, Peter K. Joshi, David A. Bennett, Philip L. De Jager, Xiangfeng Lu, Vasiliki Mamakou, Morris Brown, Mark J. Caulfield, Patricia B. Munroe, Xiuqing Guo, Marina Ciullo, Jost B. Jonas, Nilesh J. Samani, Jaakko Kaprio, Päivi Pajukanta, Teresa Tusié-Luna, Carlos A. Aguilar-Salinas, Linda S. Adair, Sonny Augustin Bechayda, H. Janaka de Silva, Ananda R. Wickremasinghe, Ronald M. Krauss, Jer-Yuarn Wu, Wei Zheng, Anneke I. den Hollander, Dwaipayan Bharadwaj, Adolfo Correa, James G. Wilson, Lars Lind, Chew-Kiat Heng, Amanda E. Nelson, Yvonne M. Golightly, James F. Wilson, Brenda Penninx, Hyung-Lae Kim, John Attia, Rodney J. Scott, D.C. Rao, Donna K. Arnett, Mark Walker, Laura J. Scott, Heikki A. Koistinen, Giriraj R. Chandak, Josep M. Mercader, Clicerio Gonzalez Villalpando, Lorena Orozco, Myriam Fornage, E. Shyong Tai, Rob M. van Dam, Terho Lehtimäki, Nish Chaturvedi, Mitsuhiro Yokota, Jianjun Liu, Dermot F. Reilly, Amy Jayne McKnight, Frank Kee, Karl-Heinz Jöckel, Mark I. McCarthy, Colin N.A. Palmer, Veronique Vitart, Caroline Hayward, Eleanor Simonsick, Cornelia M. van Duijn, Zi-Bing Jin, Fan Lu, Haretsugu Hishigaki, Xu Lin, Winfried März, Vilmundur Gudnason, Jean-Claude Tardif, Guillaume Lettre, Leen M. t Hart, Petra J.M. Elders, Daniel J. Rader, Scott M. Damrauer, Meena Kumari, Mika Kivimaki, Pim van der Harst, Tim D. Spector, Ruth J.F. Loos, Michael A. Province, Esteban J. Parra, Miguel Cruz, Bruce M. Psaty, Ivan Brandslund, Peter P. Pramstaller, Charles N. Rotimi, Kaare Christensen, Samuli Ripatti, Elisabeth Widén, Hakon Hakonarson, Struan F.A. Grant, Lambertus Kiemeney, Jacqueline de Graaf, Markus Loeffler, Florian Kronenberg, Dongfeng Gu, Jeanette Erdmann, Heribert Schunkert, Paul W. Franks, Allan Linneberg, J. Wouter Jukema, Amit V. Khera, Minna Männikkö, Marjo-Riitta Jarvelin, Zoltan Kutalik, Cucca Francesco, Dennis O. Mook-Kanamori, Ko Willems van Dijk, Hugh Watkins, David P. Strachan, Niels Grarup, Peter Sever, Neil Poulter, Wayne Huey-Herng Sheu, Jerome I. Rotter, Thomas M. Dantoft, Fredrik Karpe, Matt J. Neville, Nicholas J. Timpson, Ching-Yu Cheng, Tien-Yin Wong, Chiea Chuen Khor, Hengtong Li, Charumathi Sabanayagam, Annette Peters, Christian Gieger, Andrew T. Hattersley, Nancy L. Pedersen, Patrik K.E. Magnusson, Dorret I. Boomsma, Eco J.C. de Geus, L. Adrienne Cupples, Joyce B.J. van Meurs, Arfan Ikram, Mohsen Ghanbari, Penny Gordon-Larsen, Wei Huang, Young Jin Kim, Yasuharu Tabara, Nicholas J. Wareham, Claudia Langenberg, Eleftheria Zeggini, Jaakko Tuomilehto, Johanna Kuusisto, Markku Laakso, Erik Ingelsson, Goncalo Abecasis, John C. Chambers, Jaspal S. Kooner, Paul S. de Vries, Alanna C. Morrison, Scott Hazelhurst, Michèle Ramsay, Kari E. North, Martha Daviglus, Peter Kraft, Nicholas G. Martin, John B. Whitfield, Shahid Abbas, Danish Saleheen, Robin G. Walters, Michael V. Holmes, Corri Black, Blair H. Smith, Aris Baras, Anne E. Justice, Julie E. Buring, Paul M. Ridker, Daniel I. Chasman, Charles Kooperberg, Gen Tamiya, Masayuki Yamamoto, David A. van Heel, Richard C. Trembath, Wei-Qi Wei, Gail P. Jarvik, Bahram Namjou, M. Geoffrey Hayes, Marylyn D. Ritchie, Pekka Jousilahti, Veikko Salomaa, Kristian Hveem, Bjørn Olav Åsvold, Michiaki Kubo, Yoichiro Kamatani, Yukinori Okada, Yoshinori Murakami, Bong-Jo Kim, Unnur Thorsteinsdottir, Kari Stefansson, Jifeng Zhang, Y. Eugene Chen, Yuk-Lam Ho, Julie A. Lynch, Philip S. Tsao, Kyong-Mi Chang, Kelly Cho, Christopher J. O'Donnell, John M. Gaziano, Peter Wilson, Karen L. Mohlke, Timothy M. Frayling, Joel N. Hirschhorn, Sekar Kathiresan, Michael Boehnke, Struan Grant, Pradeep Natarajan, Yan V. Sun, Andrew P. Morris, Panos Deloukas, Gina Peloso, Themistocles L. Assimes, Cristen J. Willer, Xiang Zhu, Christopher D. Brown (2022)A multi-layer functional genomic analysis to understand noncoding genetic variation in lipids, In: American journal of human genetics109(8)1366pp. 1366-1387 Elsevier Inc

A major challenge of genome-wide association studies (GWASs) is to translate phenotypic associations into biological insights. Here, we integrate a large GWAS on blood lipids involving 1.6 million individuals from five ancestries with a wide array of functional genomic datasets to discover regulatory mechanisms underlying lipid associations. We first prioritize lipid-associated genes with expression quantitative trait locus (eQTL) colocalizations and then add chromatin interaction data to narrow the search for functional genes. Polygenic enrichment analysis across 697 annotations from a host of tissues and cell types confirms the central role of the liver in lipid levels and highlights the selective enrichment of adipose-specific chromatin marks in high-density lipoprotein cholesterol and triglycerides. Overlapping transcription factor (TF) binding sites with lipid-associated loci identifies TFs relevant in lipid biology. In addition, we present an integrative framework to prioritize causal variants at GWAS loci, producing a comprehensive list of candidate causal genes and variants with multiple layers of functional evidence. We highlight two of the prioritized genes, CREBRF and RRBP1, which show convergent evidence across functional datasets supporting their roles in lipid biology. In this study, we present a multi-layer framework to combine the largest multi-ancestry GWAS to date on lipid levels with both transcriptomic and epigenomic datasets to prioritize regulatory variants, effector genes, cell types, and tissues with strong functional relevance to lipid biology.

Stefania Boccia, Jun Liu, Ayşe Demirkan, Cornelia van Duijn, Marco Mariani, Carolina Castagna, Roberta Pastorino, Szilvia Fiatal, Péter Pikó, Róza Ádány, Giordano Bottà (2021)Identification of Biomarkers for the Prevention of Chronic Disease, In: Personalised Health Carepp. 9-32 Springer International Publishing

One of the goals of personalised medicine (PM) is to use the ever-growing understanding of biology to provide a higher level of precision in disease prevention and patient care. PM strategies include the use of decision-making processes based on biomarker-driven approaches. Genes, gene expression products (i.e. transcripts and proteins) and metabolites are the main biomarker families. Given this molecular diversity of biomarkers, the increase in high-throughput omics technologies offers an amazing opportunity to capture the whole picture of biological systems in a hypothesis-free and unbiased model. This chapter examines as the high-throughput era in omics is progressing and as genomics and other omics will be effective in disentangling the aetiology and progression of the diseases.

L. Yengo, S. Vedantam, E. Marouli, J. Sidorenko, E. Bartell, S. Sakaue, M. Graff, A.U. Eliasen, Y.X. Jiang, S. Raghavan, J.K. Miao, J.D. Arias, S.E. Graham, R.E. Mukamel, C.N. Spracklen, X.Y. Yin, S.H. Chen, T. Ferreira, H.H. Highland, Y.J. Ji, T. Karaderi, K. Lin, K. Lull, D.E. Malden, C. Medina-Gomez, M. Machado, A. Moore, S. Rueger, X. Sim, S. Vrieze, T.S. Ahluwalia, M. Akiyama, M.A. Allison, M. Alvarez, M.K. Andersen, A. Ani, V. Appadurai, L. Arbeeva, S. Bhaskar, L.F. Bielak, S. Bollepalli, L.L. Bonnycastle, J. Bork-Jensen, J.P. Bradfield, Y. Bradford, P.S. Braund, J.A. Brody, K.S. Burgdorf, B.E. Cade, H. Cai, Q.Y. Cai, A. Campbell, M. Canadas-Garre, E. Catamo, J.F. Chai, X.R. Chai, L.C. Chang, Y.C. Chang, C.H. Chen, A. Chesi, S.H. Choi, R.H. Chung, M. Cocca, M.P. Concas, C. Couture, G. Cuellar-Partida, R. Danning, E.W. Daw, F. Degenhard, G.E. Delgado, A. Delitala, A. Demirkan, X. Deng, P. Devineni, A. Dietl, M. Dimitriou, L. Dimitrov, R. Dorajoo, A.B. Ekici, J.E. Engmann, Z. Fairhurst-Hunter, A.E. Farmaki, J.D. Faul, J.C. Fernandez-Lopez, L. Forer, M. Francescatto, S. Freitag-Wolf, C. Fuchsberger, T.E. Galesloot, Y. Gao, Z.S. Gao, F. Geller, O. Giannakopoulou, F. Giulianini, A.P. Gjesing, A. Goel, S.D. Gordon, M. Gorski, J. Grove, X.Q. Guo, S. Gustafsson, J. Haessler, T.F. Hansen, A.S. Havulinna, S.J. Haworth, J. He, N. Heard-Costa, P. Hebbar, G. Hindy, Y.L.A. Ho, E. Hofer, E. Holliday, K. Horn, W.E. Hornsby, J.J. Hottenga, H.Y. Huang, J. Huang, A. Huerta-Chagoya, J.E. Huffman, Y.J. Hung, S.F. Huo, M.Y. Hwang, H. Iha, D.D. Ikeda, M. Isono, A.U. Jackson, S. Jager, I.E. Jansen, I. Johansson, J.B. Jonas, A. Jonsson, T. Jorgensen, I.P. Kalafati, M. Kanai, S. Kanoni, L.L. Karhus, A. Kasturiratne, T. Katsuya, T. Kawaguchi, R.L. Kember, K.A. Kentistou, H.N. Kim, Y.J. Kim, M.E. Kleber, M.J. Knol, A. Kurbasic, M. Lauzon, R. Lea, J.Y. Lee, H.L. Leonard, S.C.A. Li, X.H. Li, X.Y. Li, J.J. Liang, H.H. Lin, S.Y. Lin, J. Liu, X.P. Liu, J.R. Long, L. Lores-Motta, J.A. Luan, V. Lyssenko, L.P. Lyytikainen, A. Mahajan, V. Mamakou, M. Mangino, A. Manichaikul, J. Marten, M. Mattheisen, L. Mavarani, A.F. McDaid, K. Meidtner, T.L. Melendez, J.M. Mercader, Y. Milaneschi, J.E. Miller, I.Y. Millwood, P.P. Mishra, R.E. Mitchell, L.T. Mollehave, A. Morgan, S. Mucha, M. Munz, M. Nakatochi, C.P. Nelson, M. Nethander, C.W. Nho, A.A. Nielsen, I.M. Nolte, S.S. Nongmaithem, R. Noordam, I. Ntalla, T. Nutile, A. Pandit, P. Christofidou, K. Parna, M. Pauper, E.R.B. Petersen, L.V. Petersen, N. Pitkanen, O. Polasek, A. Poveda, M.H. Preuss, S. Pyarajan, L.M. Raffield, H. Rakugi, J. Ramirez, A. Rasheed, D. Raven, N.W. Rayner, C. Riveros, R. Rohde, D. Ruggiero, S.E. Ruotsalainen, K.A. Ryan, M. Sabater-Lleal, R. Saxena, M. Scholz, A. Sendamarai, B.T. Shen, J.C.Z. Shi, J.H. Shin, C. Sidore, C.M. Sitlani, R.K.C. Slieker, R.A.J. Smit, A.V. Smith, J.A. Smith, L.J. Smyth, L.E. Southam, V. Steinthorsdottir, L. Sun, F. Takeuchi, D. Tallapragada, K.D. Taylor, B.O. Tayo, C. Tcheandjieu, N. Terzikhan, P. Tesolin, A. Teumer, E. Theusch, D.J. Thompson, G. Thorleifsson, P.R.H.J. Timmers, S. Trompet, C. Turman, S. Vaccargiu, S.W. van der Laan, P.J. van der Most, J.B. van Klinken, J. van Setten, S.S. Verma, N. Verweij, Y. Veturi, C.A. Wang, C.L. Wang, L.H. Wang, Z. Wang, H.R. Warren, W.B. Wei, A.R. Wickremasinghe, M. Wielscher, K.L. Wiggins, B.S. Winsvold, A. Wong, Y. Wu, M. Wuttke, R. Xia, T. Xie, K. Yamamoto, J.Y. Yang, J. Yao, H. Young, N.A. Yousri, L. Yu, L.Y. Zeng, W.H. Zhang, X.Y. Zhang, J.H. Zhao, W. Zhao, W. Zhou, M.E. Zimmermann, M. Zoledziewska, L.S. Adair, H.H.H. Adams, C.A. Aguilar-Salinas, F. Al-Mulla, D.K. Arnett (2022)A saturated map of common genetic variants associated with human height, In: Nature610(7933)pp. 704-712

Common single-nucleotide polymorphisms (SNPs) are predicted to collectively explain 40-50% of phenotypic variation in human height, but identifying the specific variants and associated regions requires huge sample sizes1. Here, using data from a genome-wide association study of 5.4 million individuals of diverse ancestries, we show that 12,111 independent SNPs that are significantly associated with height account for nearly all of the common SNP-based heritability. These SNPs are clustered within 7,209 non-overlapping genomic segments with a mean size of around 90 kb, covering about 21% of the genome. The density of independent associations varies across the genome and the regions of increased density are enriched for biologically relevant genes. In out-of-sample estimation and prediction, the 12,111 SNPs (or all SNPs in the HapMap 3 panel2) account for 40% (45%) of phenotypic variance in populations of European ancestry but only around 10-20% (14-24%) in populations of other ancestries. Effect sizes, associated regions and gene prioritization are similar across ancestries, indicating that reduced prediction accuracy is likely to be explained by linkage disequilibrium and differences in allele frequency within associated regions. Finally, we show that the relevant biological pathways are detectable with smaller sample sizes than are needed to implicate causal genes and variants. Overall, this study provides a comprehensive map of specific genomic regions that contain the vast majority of common height-associated variants. Although this map is saturated for populations of European ancestry, further research is needed to achieve equivalent saturation in other ancestries.

Natalia Pervjakova, Gunn-Helen Moen, Maria-Carolina Borges, Teresa Ferreira, James P Cook, Catherine Allard, Robin N Beaumont, Mickaël Canouil, Gad Hatem, Anni Heiskala, Anni Joensuu, Ville Karhunen, Soo Heon Kwak, Frederick T J Lin, Jun Liu, Sheryl Rifas-Shiman, Claudia H Tam, Wing Hung Tam, Gudmar Thorleifsson, Toby Andrew, Juha Auvinen, Bishwajit Bhowmik, Amélie Bonnefond, Fabien Delahaye, Ayse Demirkan, Philippe Froguel, Kadri Haller-Kikkatalo, Hildur Hardardottir, Sandra Hummel, Akhtar Hussain, Eero Kajantie, Elina Keikkala, Amna Khamis, Jari Lahti, Tove Lekva, Sanna Mustaniemi, Christine Sommer, Aili Tagoma, Evangelia Tzala, Raivo Uibo, Marja Vääräsmäki, Pia M Villa, Kåre I Birkeland, Luigi Bouchard, Cornelia M Duijn, Sarah Finer, Leif Groop, Esa Hämäläinen, Geoffrey M Hayes, Graham A Hitman, Hak C Jang, Marjo-Riitta Järvelin, Anne Karen Jenum, Hannele Laivuori, Ronald C Ma, Olle Melander, Emily Oken, Kyong Soo Park, Patrice Perron, Rashmi B Prasad, Elisabeth Qvigstad, Sylvain Sebert, Kari Stefansson, Valgerdur Steinthorsdottir, Tiinamaija Tuomi, Marie-France Hivert, Paul W Franks, Mark I McCarthy, Cecilia M Lindgren, Rachel M Freathy, Deborah A Lawlor, Andrew P Morris, Reedik Mägi (2022)Multi-ancestry genome-wide association study of gestational diabetes mellitus highlights genetic links with type 2 diabetes, In: Human molecular genetics31(19)pp. 3377-3391

Gestational diabetes mellitus (GDM) is associated with increased risk of pregnancy complications and adverse perinatal outcomes. GDM often reoccurs and is associated with increased risk of subsequent diagnosis of type 2 diabetes (T2D). To improve our understanding of the aetiological factors and molecular processes driving the occurrence of GDM, including the extent to which these overlap with T2D pathophysiology, the GENetics of Diabetes In Pregnancy Consortium assembled genome-wide association studies of diverse ancestry in a total of 5485 women with GDM and 347 856 without GDM. Through multi-ancestry meta-analysis, we identified five loci with genome-wide significant association (P 

Nabeel Merali, Tarak Chouari, Julien Marc Terroire, Maria-Danae Jessel, Daniel S K Liu, James-Halle Smith, Tyler Wooldridge, Tony Singh Dhillon, Jose I Jimenez, Jonathan Krell, Keith J. Roberts, Timothy A Rockall, Eirini Velliou, Shivan Sivakumar, Elisa Giovannetti, Ayse Demirkan, Nicola E. Annels, Adam E. Frampton (2023)Bile Microbiome Signatures Associated with Pancreatic Ductal Adenocarcinoma Compared to Benign Disease: A UK Pilot Study, In: International journal of molecular sciences24(23)16888 MDPI

Pancreatic ductal adenocarcinoma (PDAC) has a very poor survival. The intra-tumoural microbiome can influence pancreatic tumourigenesis and chemoresistance and, therefore, patient survival. The role played by bile microbiota in PDAC is unknown. We aimed to define bile microbiome signatures that can effectively distinguish malignant from benign tumours in patients presenting with obstructive jaundice caused by benign and malignant pancreaticobiliary disease. Prospective bile samples were obtained from 31 patients who underwent either Endoscopic Retrograde Cholangiopancreatography (ERCP) or Percutaneous Transhepatic Cholangiogram (PTC). Variable regions (V3–V4) of the 16S rRNA genes of microorganisms present in the samples were amplified by Polymerase Chain Reaction (PCR) and sequenced. The cohort consisted of 12 PDAC, 10 choledocholithiasis, seven gallstone pancreatitis and two primary sclerosing cholangitis patients. Using the 16S rRNA method, we identified a total of 135 genera from 29 individuals (12 PDAC and 17 benign). The bile microbial beta diversity significantly differed between patients with PDAC vs. benign disease (Permanova p = 0.0173). The separation of PDAC from benign samples is clearly seen through unsupervised clustering of Aitchison distance. We found three genera to be of significantly lower abundance among PDAC samples vs. benign, adjusting for false discovery rate (FDR). These were Escherichia (FDR = 0.002) and two unclassified genera, one from Proteobacteria (FDR = 0.002) and one from Enterobacteriaceae (FDR = 0.011). In the same samples, the genus Streptococcus (FDR = 0.033) was found to be of increased abundance in the PDAC group. We show that patients with obstructive jaundice caused by PDAC have an altered microbiome composition in the bile compared to those with benign disease. These bile-based microbes could be developed into potential diagnostic and prognostic biomarkers for PDAC and warrant further investigation.

Mariska Bot, Yuri Milaneschi, Tahani Al-Shehri, Najaf Amin, Sanzhima Garmaeva, Gerrit L.J. Onderwater, Rene Pool, Carisha S. Thesing, Lisanne S. Vijfhuizen, Nicole Vogelzangs, Ilja C.W. Arts, Ayse Demirkan, Cornelia van Duijn, Marleen van Greevenbroek, Carla J.H. van der Kallen, Sebastian Köhler, Lannie Ligthart, Arn M.J.M. van den Maagdenberg, Dennis O. Mook-Kanamori, Renée de Mutsert, Henning Tiemeier, Miranda T. Schram, Coen D.A. Stehouwer, Gisela M. Terwindt, Ko Willems van Dijk, Jingyuan Fu, Alexandra Zhernakova, Marian Beekman, P. Eline Slagboom, Dorret I. Boomsma, Brenda W.J.H. Penninx, M. Beekman, H.E.D. Suchiman, J. Deelen, N. Amin, J.W. Beulens, J.A. van der Bom, N. Bomer, A. Demirkan, J.A. van Hilten, J.M.T.A. Meessen, R. Pool, M.H. Moed, J. Fu, G.L.J. Onderwater, F. Rutters, C. So-Osman, W.M. van der Flier, A.A.W.A. van der Heijden, A. van der Spek, F.W. Asselbergs, E. Boersma, P.M. Elders, J.M. Geleijnse, M.A. Ikram, M. Kloppenburg, I. Meulenbelt, S.P. Mooijaart, R.G.H.H. Nelissen, M.G. Netea, B.W.J.H. Penninx, C.D.A. Stehouwer, C.E. Teunissen, G.M. Terwindt, L.M. ’t Hart, A.M.J.M. van den Maagdenberg, P. van der Harst, I.C.C. van der Horst, C.J.H. van der Kallen, M.M.J. van Greevenbroek, W.E. van Spil, C. Wijmenga, A.H. Zwinderman, A. Zhernikova, J.W. Jukema, N. Sattar (2020)Metabolomics Profile in Depression: A Pooled Analysis of 230 Metabolic Markers in 5283 Cases With Depression and 10,145 Controls, In: Biological psychiatry (1969)87(5)409pp. 409-418 Elsevier Inc

Depression has been associated with metabolic alterations, which adversely impact cardiometabolic health. Here, a comprehensive set of metabolic markers, predominantly lipids, was compared between depressed and nondepressed persons. Nine Dutch cohorts were included, comprising 10,145 control subjects and 5283 persons with depression, established with diagnostic interviews or questionnaires. A proton nuclear magnetic resonance metabolomics platform provided 230 metabolite measures: 51 lipids, fatty acids, and low-molecular-weight metabolites; 98 lipid composition and particle concentration measures of lipoprotein subclasses; and 81 lipid and fatty acids ratios. For each metabolite measure, logistic regression analyses adjusted for gender, age, smoking, fasting status, and lipid-modifying medication were performed within cohort, followed by random-effects meta-analyses. Of the 51 lipids, fatty acids, and low-molecular-weight metabolites, 21 were significantly related to depression (false discovery rate q < .05). Higher levels of apolipoprotein B, very-low-density lipoprotein cholesterol, triglycerides, diglycerides, total and monounsaturated fatty acids, fatty acid chain length, glycoprotein acetyls, tyrosine, and isoleucine and lower levels of high-density lipoprotein cholesterol, acetate, and apolipoprotein A1 were associated with increased odds of depression. Analyses of lipid composition indicators confirmed a shift toward less high-density lipoprotein and more very-low-density lipoprotein and triglyceride particles in depression. Associations appeared generally consistent across gender, age, and body mass index strata and across cohorts with depressive diagnoses versus symptoms. This large-scale meta-analysis indicates a clear distinctive profile of circulating lipid metabolites associated with depression, potentially opening new prevention or treatment avenues for depression and its associated cardiometabolic comorbidity.

Fiona A. Hagenbeek, René Pool, Jenny van Dongen, H. M. Draisma, Jouke Jan Hottenga, Gonneke Willemsen, Abdel Abdellaoui, Iryna O. Fedko, Anouk den Braber, Pieter Jelle Visser, Eco J. C. N. de Geus, Ko Willems van Dijk, Aswin Verhoeven, H. Eka Suchiman, Marian Beekman, P. Eline Slagboom, Cornelia M. van Duijn, Amy C. Harms, Thomas Hankemeier, Meike Bartels, Michel G. Nivard, Dorret I. Boomsma, Ayse Demirkan (2020)Author Correction: Heritability estimates for 361 blood metabolites across 40 genome-wide association studies, In: Nature communications11(1)1702pp. 1702-1702 Nature Publishing Group UK