251
|
Abstract
PURPOSE OF REVIEW The goal of the review is to provide a comprehensive overview of the current understanding of the mechanisms underlying variation in human stature. RECENT FINDINGS Human height is an anthropometric trait that varies considerably within human populations as well as across the globe. Historically, much research focus was placed on understanding the biology of growth plate chondrocytes and how modifications to core chondrocyte proliferation and differentiation pathways potentially shaped height attainment in normal as well as pathological contexts. Recently, much progress has been made to improve our understanding regarding the mechanisms underlying the normal and pathological range of height variation within as well as between human populations, and today, it is understood to reflect complex interactions among a myriad of genetic, environmental, and evolutionary factors. Indeed, recent improvements in genetics (e.g., GWAS) and breakthroughs in functional genomics (e.g., whole exome sequencing, DNA methylation analysis, ATAC-sequencing, and CRISPR) have shed light on previously unknown pathways/mechanisms governing pathological and common height variation. Additionally, the use of an evolutionary perspective has also revealed important mechanisms that have shaped height variation across the planet. This review provides an overview of the current knowledge of the biological mechanisms underlying height variation by highlighting new research findings on skeletal growth control with an emphasis on previously unknown pathways/mechanisms influencing pathological and common height variation. In this context, this review also discusses how evolutionary forces likely shaped the genomic architecture of height across the globe.
Collapse
Affiliation(s)
| | - Terence D Capellini
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
252
|
Young AI, Benonisdottir S, Przeworski M, Kong A. Deconstructing the sources of genotype-phenotype associations in humans. Science 2019; 365:1396-1400. [PMID: 31604265 PMCID: PMC6894903 DOI: 10.1126/science.aax3710] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Efforts to link variation in the human genome to phenotypes have progressed at a tremendous pace in recent decades. Most human traits have been shown to be affected by a large number of genetic variants across the genome. To interpret these associations and to use them reliably-in particular for phenotypic prediction-a better understanding of the many sources of genotype-phenotype associations is necessary. We summarize the progress that has been made in this direction in humans, notably in decomposing direct and indirect genetic effects as well as population structure confounding. We discuss the natural next steps in data collection and methodology development, with a focus on what can be gained by analyzing genotype and phenotype data from close relatives.
Collapse
Affiliation(s)
- Alexander I Young
- Big Data Institute, Li Ka Shing Centre for Health Information Discovery, University of Oxford, Oxford, UK.
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Stefania Benonisdottir
- Big Data Institute, Li Ka Shing Centre for Health Information Discovery, University of Oxford, Oxford, UK
| | - Molly Przeworski
- Department of Biological Sciences, Columbia University, New York, NY, USA.
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Augustine Kong
- Big Data Institute, Li Ka Shing Centre for Health Information Discovery, University of Oxford, Oxford, UK.
| |
Collapse
|
253
|
Akiyama M, Ishigaki K, Sakaue S, Momozawa Y, Horikoshi M, Hirata M, Matsuda K, Ikegawa S, Takahashi A, Kanai M, Suzuki S, Matsui D, Naito M, Yamaji T, Iwasaki M, Sawada N, Tanno K, Sasaki M, Hozawa A, Minegishi N, Wakai K, Tsugane S, Shimizu A, Yamamoto M, Okada Y, Murakami Y, Kubo M, Kamatani Y. Characterizing rare and low-frequency height-associated variants in the Japanese population. Nat Commun 2019; 10:4393. [PMID: 31562340 PMCID: PMC6764965 DOI: 10.1038/s41467-019-12276-5] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 08/27/2019] [Indexed: 12/29/2022] Open
Abstract
Human height is a representative phenotype to elucidate genetic architecture. However, the majority of large studies have been performed in European population. To investigate the rare and low-frequency variants associated with height, we construct a reference panel (N = 3,541) for genotype imputation by integrating the whole-genome sequence data from 1,037 Japanese with that of the 1000 Genomes Project, and perform a genome-wide association study in 191,787 Japanese. We report 573 height-associated variants, including 22 rare and 42 low-frequency variants. These 64 variants explain 1.7% of the phenotypic variance. Furthermore, a gene-based analysis identifies two genes with multiple height-increasing rare and low-frequency nonsynonymous variants (SLC27A3 and CYP26B1; PSKAT-O < 2.5 × 10−6). Our analysis shows a general tendency of the effect sizes of rare variants towards increasing height, which is contrary to findings among Europeans, suggesting that height-associated rare variants are under different selection pressure in Japanese and European populations. Thousands of genetic loci are known to associate with human height, but these are mainly based on studies in European ancestry populations. Here, Akiyama et al. construct a genotype reference panel for the Japanese population followed by GWAS and report 573 height associated variants in 191,787 Japanese.
Collapse
Affiliation(s)
- Masato Akiyama
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan.,Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.,Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Kazuyoshi Ishigaki
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Saori Sakaue
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan.,Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Yukihide Momozawa
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Momoko Horikoshi
- Laboratory for Endocrinology, Metabolism and Kidney Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Makoto Hirata
- Laboratory of Genome Technology, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Koichi Matsuda
- Laboratory of Clinical Genome Sequencing, Department of Computational Biology and Medical Sciences, Graduate school of Frontier Sciences, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Shiro Ikegawa
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, 108-8639, Japan
| | - Atsushi Takahashi
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan.,Department of Genomic Medicine, Research Institute, National Cerebral and Cardiovascular Center, Osaka, 565-8565, Japan
| | - Masahiro Kanai
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan.,Department of Biomedical Informatics, Harvard Medical School, Boston, MA02115, USA
| | - Sadao Suzuki
- Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Daisuke Matsui
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, 602-8566, Japan
| | - Mariko Naito
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan.,Department of Oral Epidemiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Taiki Yamaji
- Division of Epidemiology, Center for Public Health Sciences, National Cancer Center, Tokyo, 104-0045, Japan
| | - Motoki Iwasaki
- Division of Epidemiology, Center for Public Health Sciences, National Cancer Center, Tokyo, 104-0045, Japan
| | - Norie Sawada
- Division of Epidemiology, Center for Public Health Sciences, National Cancer Center, Tokyo, 104-0045, Japan
| | - Kozo Tanno
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Iwate, 028-3694, Japan.,Department of Hygiene and Preventive Medicine, School of Medicine, Iwate Medical University, Iwate, 028-3694, Japan
| | - Makoto Sasaki
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Iwate, 028-3694, Japan
| | - Atsushi Hozawa
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, 980-8573, Japan.,Graduate School of Medicine, Tohoku University, Sendai, 980-8575, Japan
| | - Naoko Minegishi
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, 980-8573, Japan.,Graduate School of Medicine, Tohoku University, Sendai, 980-8575, Japan
| | - Kenji Wakai
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Shoichiro Tsugane
- Center for Public Health Sciences, National Cancer Center, Tokyo, 104-0045, Japan
| | - Atsushi Shimizu
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Iwate, 028-3694, Japan
| | - Masayuki Yamamoto
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, 980-8573, Japan.,Graduate School of Medicine, Tohoku University, Sendai, 980-8575, Japan
| | - Yukinori Okada
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan.,Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan.,Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Osaka, 565-0871, Japan
| | - Yoshinori Murakami
- Division of Molecular Pathology, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Michiaki Kubo
- RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan. .,Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan. .,Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, 108-8639, Japan.
| |
Collapse
|
254
|
Bøtkjær JA, Noer PR, Oxvig C, Yding Andersen C. A common variant of the pregnancy-associated plasma protein-A (PAPPA) gene encodes a protein with reduced proteolytic activity towards IGF-binding proteins. Sci Rep 2019; 9:13231. [PMID: 31519945 PMCID: PMC6744435 DOI: 10.1038/s41598-019-49626-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 07/05/2019] [Indexed: 02/03/2023] Open
Abstract
Pregnancy-associated plasma protein-A (PAPP-A) is a key regulator of insulin-like growth factor (IGF) bioactivity, by releasing the IGFs from their corresponding IGF-binding proteins (IGFBPs). The minor allele of the single nucleotide polymorphism (SNP), rs7020782 (serine < tyrosine), in PAPPA has previously been associated with recurrent pregnancy loss as well as with significant reduced levels of PAPP-A protein in human ovarian follicles. The aim of the present study was to reveal a possible functional effect of the rs7020782 SNP in PAPPA by comparing recombinant PAPP-A proteins from transfected human embryonic kidney 293 T cells. The proteolytic cleavage of IGFBP-4 was shown to be affected by the rs7020782 SNP in PAPPA, showing a significantly reduced cleavage rate for the serine variant compared to the tyrosine variant (p-value < 0.001). The serine variant also showed a trend towards reduced cleavage rates, that was not significant, towards IGFBP-2 and IGFBP-5 compared to the tyrosine variant. No differences were found when analysing cell surface binding, complex formation between PAPP-A and STC2 or proMBP, nor when analysing STC1 inhibition of PAPP-A-mediated IGFBP-4 cleavage. Regulation of IGF bioactivity in reproductive tissues is important and the rs7020782 SNP in PAPPA may disturb this regulation by altering the specific activity of PAPP-A.
Collapse
Affiliation(s)
- Jane Alrø Bøtkjær
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Rigshospitalet, Copenhagen University Hospital, Copenhagen University, Copenhagen, DK-2100, Denmark.
| | - Pernille Rimmer Noer
- Department of Molecular Biology and Genetics, University of Aarhus, Aarhus, DK-8000, Denmark
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, University of Aarhus, Aarhus, DK-8000, Denmark
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Rigshospitalet, Copenhagen University Hospital, Copenhagen University, Copenhagen, DK-2100, Denmark
| |
Collapse
|
255
|
Choobdar S, Ahsen ME, Crawford J, Tomasoni M, Fang T, Lamparter D, Lin J, Hescott B, Hu X, Mercer J, Natoli T, Narayan R, DREAM Module Identification Challenge Consortium, Subramanian A, Zhang JD, Stolovitzky G, Kutalik Z, Lage K, Slonim DK, Saez-Rodriguez J, Cowen LJ, Bergmann S, Marbach D. Assessment of network module identification across complex diseases. Nat Methods 2019; 16:843-852. [PMID: 31471613 PMCID: PMC6719725 DOI: 10.1038/s41592-019-0509-5] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 07/10/2019] [Indexed: 12/11/2022]
Abstract
Many bioinformatics methods have been proposed for reducing the complexity of large gene or protein networks into relevant subnetworks or modules. Yet, how such methods compare to each other in terms of their ability to identify disease-relevant modules in different types of network remains poorly understood. We launched the 'Disease Module Identification DREAM Challenge', an open competition to comprehensively assess module identification methods across diverse protein-protein interaction, signaling, gene co-expression, homology and cancer-gene networks. Predicted network modules were tested for association with complex traits and diseases using a unique collection of 180 genome-wide association studies. Our robust assessment of 75 module identification methods reveals top-performing algorithms, which recover complementary trait-associated modules. We find that most of these modules correspond to core disease-relevant pathways, which often comprise therapeutic targets. This community challenge establishes biologically interpretable benchmarks, tools and guidelines for molecular network analysis to study human disease biology.
Collapse
Affiliation(s)
- Sarvenaz Choobdar
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Mehmet E Ahsen
- Icahn Institute for Genomics and Multiscale Biology and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jake Crawford
- Department of Computer Science, Tufts University, Medford, MA, USA
| | - Mattia Tomasoni
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Tao Fang
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - David Lamparter
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Verge Genomics, San Francisco, CA, USA
| | - Junyuan Lin
- Department of Mathematics, Tufts University, Medford, MA, USA
| | - Benjamin Hescott
- College of Computer and Information Science, Northeastern University, Boston, MA, USA
| | - Xiaozhe Hu
- Department of Mathematics, Tufts University, Medford, MA, USA
| | - Johnathan Mercer
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Stanley Center at the Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ted Natoli
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Rajiv Narayan
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Jitao D Zhang
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Gustavo Stolovitzky
- Icahn Institute for Genomics and Multiscale Biology and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- IBM T.J. Watson Research Center, Yorktown Heights, NY, USA
| | - Zoltán Kutalik
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- University Institute of Primary Care and Public Health, University of Lausanne, Lausanne, Switzerland
| | - Kasper Lage
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Stanley Center at the Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Biological Psychiatry, Mental Health Center Sct. Hans, University of Copenhagen, Roskilde, Denmark
| | - Donna K Slonim
- Department of Computer Science, Tufts University, Medford, MA, USA
- Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| | - Julio Saez-Rodriguez
- Institute for Computational Biomedicine, Faculty of Medicine, Heidelberg University, Bioquant, Heidelberg, Germany
- RWTH Aachen University, Faculty of Medicine, Joint Research Center for Computational Biomedicine, Aachen, Germany
| | - Lenore J Cowen
- Department of Computer Science, Tufts University, Medford, MA, USA
- Department of Mathematics, Tufts University, Medford, MA, USA
| | - Sven Bergmann
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
- Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa.
| | - Daniel Marbach
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland.
| |
Collapse
Collaborators
Fabian Aicheler, Nicola Amoroso, Alex Arenas, Karthik Azhagesan, Aaron Baker, Michael Banf, Serafim Batzoglou, Anaïs Baudot, Roberto Bellotti, Sven Bergmann, Keith A Boroevich, Christine Brun, Stanley Cai, Michael Caldera, Alberto Calderone, Gianni Cesareni, Weiqi Chen, Christine Chichester, Sarvenaz Choobdar, Lenore Cowen, Jake Crawford, Hongzhu Cui, Phuong Dao, Manlio De Domenico, Andi Dhroso, Gilles Didier, Mathew Divine, Antonio Del Sol, Tao Fang, Xuyang Feng, Jose C Flores-Canales, Santo Fortunato, Anthony Gitter, Anna Gorska, Yuanfang Guan, Alain Guénoche, Sergio Gómez, Hatem Hamza, András Hartmann, Shan He, Anton Heijs, Julian Heinrich, Benjamin Hescott, Xiaozhe Hu, Ying Hu, Xiaoqing Huang, V Keith Hughitt, Minji Jeon, Lucas Jeub, Nathan T Johnson, Keehyoung Joo, InSuk Joung, Sascha Jung, Susana G Kalko, Piotr J Kamola, Jaewoo Kang, Benjapun Kaveelerdpotjana, Minjun Kim, Yoo-Ah Kim, Oliver Kohlbacher, Dmitry Korkin, Kiryluk Krzysztof, Khalid Kunji, Zoltàn Kutalik, Kasper Lage, David Lamparter, Sean Lang-Brown, Thuc Duy Le, Jooyoung Lee, Sunwon Lee, Juyong Lee, Dong Li, Jiuyong Li, Junyuan Lin, Lin Liu, Antonis Loizou, Zhenhua Luo, Artem Lysenko, Tianle Ma, Raghvendra Mall, Daniel Marbach, Tomasoni Mattia, Mario Medvedovic, Jörg Menche, Johnathan Mercer, Elisa Micarelli, Alfonso Monaco, Felix Müller, Rajiv Narayan, Oleksandr Narykov, Ted Natoli, Thea Norman, Sungjoon Park, Livia Perfetto, Dimitri Perrin, Stefano Pirrò, Teresa M Przytycka, Xiaoning Qian, Karthik Raman, Daniele Ramazzotti, Emilie Ramsahai, Balaraman Ravindran, Philip Rennert, Julio Saez-Rodriguez, Charlotta Schärfe, Roded Sharan, Ning Shi, Wonho Shin, Hai Shu, Himanshu Sinha, Donna K Slonim, Lionel Spinelli, Suhas Srinivasan, Aravind Subramanian, Christine Suver, Damian Szklarczyk, Sabina Tangaro, Suresh Thiagarajan, Laurent Tichit, Thorsten Tiede, Beethika Tripathi, Aviad Tsherniak, Tatsuhiko Tsunoda, Dénes Türei, Ehsan Ullah, Golnaz Vahedi, Alberto Valdeolivas, Jayaswal Vivek, Christian von Mering, Andra Waagmeester, Bo Wang, Yijie Wang, Barbara A Weir, Shana White, Sebastian Winkler, Ke Xu, Taosheng Xu, Chunhua Yan, Liuqing Yang, Kaixian Yu, Xiangtian Yu, Gaia Zaffaroni, Mikhail Zaslavskiy, Tao Zeng, Jitao D Zhang, Lu Zhang, Weijia Zhang, Lixia Zhang, Xinyu Zhang, Junpeng Zhang, Xin Zhou, Jiarui Zhou, Hongtu Zhu, Junjie Zhu, Guido Zuccon,
Collapse
|
256
|
Coupé C, Oh YM, Dediu D, Pellegrino F. Different languages, similar encoding efficiency: Comparable information rates across the human communicative niche. SCIENCE ADVANCES 2019; 5:eaaw2594. [PMID: 32047854 PMCID: PMC6984970 DOI: 10.1126/sciadv.aaw2594] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 08/02/2019] [Indexed: 05/18/2023]
Abstract
Language is universal, but it has few indisputably universal characteristics, with cross-linguistic variation being the norm. For example, languages differ greatly in the number of syllables they allow, resulting in large variation in the Shannon information per syllable. Nevertheless, all natural languages allow their speakers to efficiently encode and transmit information. We show here, using quantitative methods on a large cross-linguistic corpus of 17 languages, that the coupling between language-level (information per syllable) and speaker-level (speech rate) properties results in languages encoding similar information rates (~39 bits/s) despite wide differences in each property individually: Languages are more similar in information rates than in Shannon information or speech rate. These findings highlight the intimate feedback loops between languages' structural properties and their speakers' neurocognition and biology under communicative pressures. Thus, language is the product of a multiscale communicative niche construction process at the intersection of biology, environment, and culture.
Collapse
Affiliation(s)
- Christophe Coupé
- Laboratoire Dynamique du Langage UMR5596, CNRS, University of Lyon, 14 Avenue Berthelot, 69007 Lyon, France
- Department of Linguistics, The University of Hong Kong, Pokfulam Road, Hong Kong
| | - Yoon Mi Oh
- New Zealand Institute of Language, Brain and Behaviour, University of Canterbury, Private Bag 4800, Christchurch 8140, New Zealand
- Department of French Language and Literature, Ajou University, Suwon, 16499 Gyeonggi-do, South Korea
| | - Dan Dediu
- Laboratoire Dynamique du Langage UMR5596, CNRS, University of Lyon, 14 Avenue Berthelot, 69007 Lyon, France
- Collegium de Lyon, University of Lyon, 24 rue Baldassini, 69007 Lyon, France
| | - François Pellegrino
- Laboratoire Dynamique du Langage UMR5596, CNRS, University of Lyon, 14 Avenue Berthelot, 69007 Lyon, France
- Corresponding author.
| |
Collapse
|
257
|
Abstract
BACKGROUND Obesity and type 2 diabetes (T2D) are major public health issues worldwide, and put a significant burden on the healthcare system. Genetic variants, along with traditional risk factors such as diet and physical activity, could account for up to approximately a quarter of disease risk. Epigenetic factors have demonstrated potential in accounting for additional phenotypic variation, along with providing insights into the causal relationship linking genetic variants to phenotypes. SCOPE OF REVIEW In this review article, we discuss the epidemiological and functional insights into epigenetic disturbances in obesity and diabetes, along with future research directions and approaches, with a focus on DNA methylation. MAJOR CONCLUSIONS Epigenetic mechanisms have been shown to contribute to obesity and T2D disease development, as well as potential differences in disease risks between ethnic populations. Technology to investigate epigenetic profiles in diseased individuals and tissues has advanced significantly in the last years, and suggests potential in application of epigenetic factors in clinical monitoring and as therapeutic options.
Collapse
Affiliation(s)
- Marie Loh
- Lee Kong Chian School of Medicine, Nanyang Technological University 308232, Singapore; Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore (A*STAR) 138648, Singapore; Yong Loo Lin School of Medicine, National University of Singapore 117596, Singapore; Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK
| | - Li Zhou
- Lee Kong Chian School of Medicine, Nanyang Technological University 308232, Singapore
| | - Hong Kiat Ng
- Lee Kong Chian School of Medicine, Nanyang Technological University 308232, Singapore
| | - John Campbell Chambers
- Lee Kong Chian School of Medicine, Nanyang Technological University 308232, Singapore; Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK; Department of Cardiology, Ealing Hospital, London North West Healthcare NHS Trust, Southall UB1 3HW, UK; Imperial College Healthcare NHS Trust, London W12 0HS, UK.
| |
Collapse
|
258
|
Zajac GJM, Fritsche LG, Weinstock JS, Dagenais SL, Lyons RH, Brummett CM, Abecasis GR. Estimation of DNA contamination and its sources in genotyped samples. Genet Epidemiol 2019; 43:980-995. [PMID: 31452258 DOI: 10.1002/gepi.22257] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 07/11/2019] [Accepted: 08/09/2019] [Indexed: 11/11/2022]
Abstract
Array genotyping is a cost-effective and widely used tool that enables assessment of up to millions of genetic markers in hundreds of thousands of individuals. Genotyping array data are typically highly accurate but sensitive to mixing of DNA samples from multiple individuals before or during genotyping. Contaminated samples can lead to genotyping errors and consequently cause false positive signals or reduce power of association analyses. Here, we propose a new method to identify contaminated samples and the sources of contamination within a genotyping batch. Through analysis of array intensity and genotype data from intentionally mixed samples and 22,366 samples of the Michigan Genomics Initiative, an ongoing biobank-based study, we show that our method can reliably estimate contamination. We also show that identifying sources of contamination can implicate problematic sample processing steps and guide process improvements. Compared to existing methods, our approach can estimate the proportion of contaminating DNA more accurately, eliminate the need for external databases of allele frequencies, and provide contamination estimates that are more robust to the ancestral origin of the contaminating sample.
Collapse
Affiliation(s)
- Gregory J M Zajac
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Lars G Fritsche
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Joshua S Weinstock
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Susan L Dagenais
- Department of Biological Chemistry and DNA Sequencing Core, University of Michigan, Ann Arbor, Michigan
| | - Robert H Lyons
- Department of Biological Chemistry and DNA Sequencing Core, University of Michigan, Ann Arbor, Michigan
| | - Chad M Brummett
- Department of Anesthesiology, Division of Pain Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Gonçalo R Abecasis
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, Michigan
| |
Collapse
|
259
|
Jiang P, Hu Y, Wang Y, Zhang J, Zhu Q, Bai L, Tong Q, Li T, Zhao L. Efficient Mining of Variants From Trios for Ventricular Septal Defect Association Study. Front Genet 2019; 10:670. [PMID: 31440271 PMCID: PMC6694746 DOI: 10.3389/fgene.2019.00670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 06/27/2019] [Indexed: 11/28/2022] Open
Abstract
Ventricular septal defect (VSD) is a fatal congenital heart disease showing severe consequence in affected infants. Early diagnosis plays an important role, particularly through genetic variants. Existing panel-based approaches of variants mining suffer from shortage of large panels, costly sequencing, and missing rare variants. Although a trio-based method alleviates these limitations to some extent, it is agnostic to novel mutations and computational intensive. Considering these limitations, we are studying a novel variants mining algorithm from trio-based sequencing data and apply it on a VSD trio to identify associated mutations. Our approach starts with irrelevant k-mer filtering from sequences of a trio via a newly conceived coupled Bloom Filter, then corrects sequencing errors by using a statistical approach and extends kept k-mers into long sequences. These extended sequences are used as input for variants needed. Later, the obtained variants are comprehensively analyzed against existing databases to mine VSD-related mutations. Experiments show that our trio-based algorithm narrows down candidate coding genes and lncRNAs by about 10- and 5-folds comparing with single sequence-based approaches, respectively. Meanwhile, our algorithm is 10 times faster and 2 magnitudes memory-frugal compared with existing state-of-the-art approach. By applying our approach to a VSD trio, we fish out an unreported gene—CD80, a combination of two genes—MYBPC3 and TRDN and a lncRNA—NONHSAT096266.2, which are highly likely to be VSD-related.
Collapse
Affiliation(s)
- Peng Jiang
- Precision Medicine Research Center, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Yaofei Hu
- Precision Medicine Research Center, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Yiqi Wang
- Precision Medicine Research Center, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Jin Zhang
- Precision Medicine Research Center, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Qinghong Zhu
- Precision Medicine Research Center, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Lin Bai
- School of Computing and Electronic Information, Guangxi University, Nanning, China
| | - Qiang Tong
- Precision Medicine Research Center, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Tao Li
- Precision Medicine Research Center, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Liang Zhao
- Precision Medicine Research Center, Taihe Hospital, Hubei University of Medicine, Shiyan, China.,School of Computing and Electronic Information, Guangxi University, Nanning, China
| |
Collapse
|
260
|
Rosário R, Olsen NJ, Rohde JF, Händel MN, Santos R, Heitmann BL. Longitudinal associations between body composition and regional fat distribution and later attained height at school entry among preschool children predisposed to overweight. Eur J Clin Nutr 2019; 74:465-471. [PMID: 31444466 DOI: 10.1038/s41430-019-0494-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/19/2019] [Accepted: 08/01/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND/OBJECTIVES To investigate the associations between indicators of obesity and fat distribution, such as body mass index (BMI), fat mass, and skinfold measures during preschool age, and attained height at school entry. SUBJECTS/METHODS The Healthy Start primary intervention study comprised 1100 obesity-prone preschool children from the greater Copenhagen area, with a mean [standard deviation (SD)] age of 4.0 (1.1) years at baseline. Anthropometry was measured by trained health professionals at baseline (preschool age) and follow-up height at school entry was gathered by school nurses. Prospective associations between body fat measures and later attained height were examined using generalized linear models with adjustments for potential confounders. RESULTS Greater adiposity at preschool age was directly associated with a higher attained height at follow-up at school-age, when adjusting for confounders. A baseline difference of one BMI unit was associated with a greater attained height of 0.8 cm [(95% confidence interval (CI) 0.5; 1.2]. Furthermore, a difference of 1 mm in the sum of four skinfolds measured at baseline was associated with a greater attained height of 0.1 cm (95% CI 0.03; 0.2) at follow-up. Children with overweight or obesity at baseline attained a significantly higher height of 2.9 (95% CI 1.6; 4.1) cm at follow-up after full adjustment than normal weight children. CONCLUSIONS Our results supports that greater adiposity at preschool age is associated with greater tallness. Although a greater height is assumed to be desirable, accelerated growth in childhood may in itself be a risk factor for obesity later in life.
Collapse
Affiliation(s)
- Rafaela Rosário
- School of Nursing, University of Minho, Minho, Portugal. .,Health Sciences Research Unit: Nursing (UICISA: E), Nursing School of Coimbra (ESEnfC), Coimbra, Portugal.
| | - Nanna Julie Olsen
- Research Unit for Dietary Studies, The Parker Institute, Bispebjerg and Frederiksberg Hospital, The Capital Region, Copenhagen, Denmark
| | - Jeanett Friis Rohde
- Research Unit for Dietary Studies, The Parker Institute, Bispebjerg and Frederiksberg Hospital, The Capital Region, Copenhagen, Denmark
| | - Mina Nicole Händel
- Research Unit for Dietary Studies, The Parker Institute, Bispebjerg and Frederiksberg Hospital, The Capital Region, Copenhagen, Denmark
| | - Rute Santos
- Research Centre in Physical Activity, Health and Leisure, Faculty of Sport, University of Porto, Porto, Portugal.,Early Start and Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
| | - Berit Lilienthal Heitmann
- Research Unit for Dietary Studies, The Parker Institute, Bispebjerg and Frederiksberg Hospital, The Capital Region, Copenhagen, Denmark.,The Boden Institute of Obesity, Nutrition, Exercise & Eating Disorders, University of Sydney, Sydney, Australia.,Department of Public Health, Section for General Practice, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
261
|
Yoon S, Nguyen HCT, Yoo YJ, Kim J, Baik B, Kim S, Kim J, Kim S, Nam D. Efficient pathway enrichment and network analysis of GWAS summary data using GSA-SNP2. Nucleic Acids Res 2019; 46:e60. [PMID: 29562348 PMCID: PMC6007455 DOI: 10.1093/nar/gky175] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 03/13/2018] [Indexed: 01/19/2023] Open
Abstract
Pathway-based analysis in genome-wide association study (GWAS) is being widely used to uncover novel multi-genic functional associations. Many of these pathway-based methods have been used to test the enrichment of the associated genes in the pathways, but exhibited low powers and were highly affected by free parameters. We present the novel method and software GSA-SNP2 for pathway enrichment analysis of GWAS P-value data. GSA-SNP2 provides high power, decent type I error control and fast computation by incorporating the random set model and SNP-count adjusted gene score. In a comparative study using simulated and real GWAS data, GSA-SNP2 exhibited high power and best prioritized gold standard positive pathways compared with six existing enrichment-based methods and two self-contained methods (alternative pathway analysis approach). Based on these results, the difference between pathway analysis approaches was investigated and the effects of the gene correlation structures on the pathway enrichment analysis were also discussed. In addition, GSA-SNP2 is able to visualize protein interaction networks within and across the significant pathways so that the user can prioritize the core subnetworks for further studies. GSA-SNP2 is freely available at https://sourceforge.net/projects/gsasnp2.
Collapse
Affiliation(s)
- Sora Yoon
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Hai C T Nguyen
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Yun J Yoo
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul 08826, South Korea.,Department of Mathematics Education, Seoul National University, Seoul 08826, Republic of Korea
| | - Jinhwan Kim
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Bukyung Baik
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Sounkou Kim
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Jin Kim
- SK Telecom, Seoul 04539, Republic of Korea
| | - Sangsoo Kim
- School of Systems Biomedical Science, Soongsil University, Seoul 06978, Republic of Korea
| | - Dougu Nam
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea.,Department of Mathematical Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| |
Collapse
|
262
|
Locke AE, Steinberg KM, Chiang CWK, Service SK, Havulinna AS, Stell L, Pirinen M, Abel HJ, Chiang CC, Fulton RS, Jackson AU, Kang CJ, Kanchi KL, Koboldt DC, Larson DE, Nelson J, Nicholas TJ, Pietilä A, Ramensky V, Ray D, Scott LJ, Stringham HM, Vangipurapu J, Welch R, Yajnik P, Yin X, Eriksson JG, Ala-Korpela M, Järvelin MR, Männikkö M, Laivuori H, Dutcher SK, Stitziel NO, Wilson RK, Hall IM, Sabatti C, Palotie A, Salomaa V, Laakso M, Ripatti S, Boehnke M, Freimer NB. Exome sequencing of Finnish isolates enhances rare-variant association power. Nature 2019; 572:323-328. [PMID: 31367044 PMCID: PMC6697530 DOI: 10.1038/s41586-019-1457-z] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 07/02/2019] [Indexed: 12/30/2022]
Abstract
Exome-sequencing studies have generally been underpowered to identify deleterious alleles with a large effect on complex traits as such alleles are mostly rare. Because the population of northern and eastern Finland has expanded considerably and in isolation following a series of bottlenecks, individuals of these populations have numerous deleterious alleles at a relatively high frequency. Here, using exome sequencing of nearly 20,000 individuals from these regions, we investigate the role of rare coding variants in clinically relevant quantitative cardiometabolic traits. Exome-wide association studies for 64 quantitative traits identified 26 newly associated deleterious alleles. Of these 26 alleles, 19 are either unique to or more than 20 times more frequent in Finnish individuals than in other Europeans and show geographical clustering comparable to Mendelian disease mutations that are characteristic of the Finnish population. We estimate that sequencing studies of populations without this unique history would require hundreds of thousands to millions of participants to achieve comparable association power.
Collapse
Affiliation(s)
- Adam E Locke
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Karyn Meltz Steinberg
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA
| | - Charleston W K Chiang
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Quantitative and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Susan K Service
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA
| | - Aki S Havulinna
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- National Institute for Health and Welfare, Helsinki, Finland
| | - Laurel Stell
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Matti Pirinen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Helsinki Institute for Information Technology HIIT and Department of Mathematics and Statistics, University of Helsinki, Helsinki, Finland
| | - Haley J Abel
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
| | - Colby C Chiang
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
| | - Robert S Fulton
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
| | - Anne U Jackson
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Chul Joo Kang
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
| | - Krishna L Kanchi
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
| | - Daniel C Koboldt
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
- The Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - David E Larson
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
| | - Joanne Nelson
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
| | - Thomas J Nicholas
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
- USTAR Center for Genetic Discovery and Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Arto Pietilä
- National Institute for Health and Welfare, Helsinki, Finland
| | - Vasily Ramensky
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA
- Federal State Institution "National Medical Research Center for Preventive Medicine" of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Debashree Ray
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Departments of Epidemiology and Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Laura J Scott
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Heather M Stringham
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Jagadish Vangipurapu
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Ryan Welch
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Pranav Yajnik
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Xianyong Yin
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Johan G Eriksson
- Department of Public Health Solutions, National Institute for Health and Welfare, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mika Ala-Korpela
- Systems Epidemiology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, University of Oulu, Oulu, Finland
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia
| | - Marjo-Riitta Järvelin
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Unit of Primary Health Care, Oulu University Hospital, Oulu, Finland
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, London, UK
| | - Minna Männikkö
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Northern Finland Birth Cohorts, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Hannele Laivuori
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Obstetrics and Gynecology, Tampere University Hospital and University of Tampere, Faculty of Medicine and Health Technology, Tampere, Finland
| | - Susan K Dutcher
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
| | - Nathan O Stitziel
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Richard K Wilson
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
- The Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Ira M Hall
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
| | - Chiara Sabatti
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
- Department of Statistics, Stanford University, Stanford, CA, USA
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Analytical and Translational Genetics Unit (ATGU), Psychiatric & Neurodevelopmental Genetics Unit, Departments of Psychiatry and Neurology, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Veikko Salomaa
- National Institute for Health and Welfare, Helsinki, Finland
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA.
| | - Nelson B Freimer
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
263
|
Porcu E, Rüeger S, Lepik K, Santoni FA, Reymond A, Kutalik Z. Mendelian randomization integrating GWAS and eQTL data reveals genetic determinants of complex and clinical traits. Nat Commun 2019; 10:3300. [PMID: 31341166 PMCID: PMC6656778 DOI: 10.1038/s41467-019-10936-0] [Citation(s) in RCA: 183] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 06/11/2019] [Indexed: 01/21/2023] Open
Abstract
Genome-wide association studies (GWAS) have identified thousands of variants associated with complex traits, but their biological interpretation often remains unclear. Most of these variants overlap with expression QTLs, indicating their potential involvement in regulation of gene expression. Here, we propose a transcriptome-wide summary statistics-based Mendelian Randomization approach (TWMR) that uses multiple SNPs as instruments and multiple gene expression traits as exposures, simultaneously. Applied to 43 human phenotypes, it uncovers 3,913 putatively causal gene-trait associations, 36% of which have no genome-wide significant SNP nearby in previous GWAS. Using independent association summary statistics, we find that the majority of these loci were missed by GWAS due to power issues. Noteworthy among these links is educational attainment-associated BSCL2, known to carry mutations leading to a Mendelian form of encephalopathy. We also find pleiotropic causal effects suggestive of mechanistic connections. TWMR better accounts for pleiotropy and has the potential to identify biological mechanisms underlying complex traits.
Collapse
Affiliation(s)
- Eleonora Porcu
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland. .,Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| | - Sina Rüeger
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.,University Center for Primary Care and Public Health, University of Lausanne, Switzerland, Lausanne, Switzerland
| | - Kaido Lepik
- University Center for Primary Care and Public Health, University of Lausanne, Switzerland, Lausanne, Switzerland.,Institute of Computer Science, University of Tartu, Tartu, Estonia
| | | | | | - Federico A Santoni
- Endocrine, Diabetes, and Metabolism Service, CHUV and University of Lausanne, Lausanne, Switzerland
| | - Alexandre Reymond
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Zoltán Kutalik
- Swiss Institute of Bioinformatics, Lausanne, Switzerland. .,University Center for Primary Care and Public Health, University of Lausanne, Switzerland, Lausanne, Switzerland.
| |
Collapse
|
264
|
Mortzfeld BM, Taubenheim J, Klimovich AV, Fraune S, Rosenstiel P, Bosch TCG. Temperature and insulin signaling regulate body size in Hydra by the Wnt and TGF-beta pathways. Nat Commun 2019; 10:3257. [PMID: 31332174 PMCID: PMC6646324 DOI: 10.1038/s41467-019-11136-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 06/07/2019] [Indexed: 02/03/2023] Open
Abstract
How multicellular organisms assess and control their size is a fundamental question in biology, yet the molecular and genetic mechanisms that control organ or organism size remain largely unsolved. The freshwater polyp Hydra demonstrates a high capacity to adapt its body size to different temperatures. Here we identify the molecular mechanisms controlling this phenotypic plasticity and show that temperature-induced cell number changes are controlled by Wnt- and TGF-β signaling. Further we show that insulin-like peptide receptor (INSR) and forkhead box protein O (FoxO) are important genetic drivers of size determination controlling the same developmental regulators. Thus, environmental and genetic factors directly affect developmental mechanisms in which cell number is the strongest determinant of body size. These findings identify the basic mechanisms as to how size is regulated on an organismic level and how phenotypic plasticity is integrated into conserved developmental pathways in an evolutionary informative model organism.
Collapse
Affiliation(s)
- Benedikt M Mortzfeld
- Zoological Institute, Christian-Albrechts University Kiel, Am Botanischen Garten 1-9, 24118, Kiel, Germany
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Rd, Dartmouth, MA, 02747, USA
| | - Jan Taubenheim
- Zoological Institute, Christian-Albrechts University Kiel, Am Botanischen Garten 1-9, 24118, Kiel, Germany
- Institute for Zoology and Organismic Interactions, Heinrich-Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Alexander V Klimovich
- Zoological Institute, Christian-Albrechts University Kiel, Am Botanischen Garten 1-9, 24118, Kiel, Germany
| | - Sebastian Fraune
- Zoological Institute, Christian-Albrechts University Kiel, Am Botanischen Garten 1-9, 24118, Kiel, Germany
- Institute for Zoology and Organismic Interactions, Heinrich-Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian-Albrechts University Kiel, University Hospital Schleswig-Holstein, Rosalind-Franklin-Straße 12, 24105, Kiel, Germany
| | - Thomas C G Bosch
- Zoological Institute, Christian-Albrechts University Kiel, Am Botanischen Garten 1-9, 24118, Kiel, Germany.
| |
Collapse
|
265
|
Abstract
Zusammenfassung
Häufige Krankheiten, die sog. Volkskrankheiten, sind in der Regel multifaktoriell verursacht, d. h. zu ihrer Entwicklung tragen sowohl genetische Faktoren als auch nicht-genetische Umgebungseinflüsse bei. Die geschätzte Gesamterblichkeit (‑heritabilität) reicht von moderat bis vergleichsweise hoch. Die genetische Architektur ist komplex und kann das gesamte allelische Spektrum, von häufigen Varianten mit niedriger Penetranz bis hin zu seltenen Varianten mit höherer Penetranz, sowie alle möglichen Kombinationen umfassen. Während häufige Varianten seit mehreren Jahren mit großem Erfolg durch genomweite Assoziationsstudien (GWAS) identifiziert werden, war bisher die Identifizierung seltener Varianten, insbesondere aufgrund der großen Zahl beitragender Gene, nur begrenzt erfolgreich. Dies ändert sich derzeit dank der Anwendung von Hochdurchsatz-Sequenziertechnologien („next-generation sequencing“, NGS) und der daraus resultierenden zunehmenden Verfügbarkeit von exom- und genomweiten Sequenzdaten großer Kollektive. In diesem Artikel geben wir einen Überblick über die Bedeutung seltener Varianten bei häufigen Erkrankungen sowie den aktuellen Stand in Bezug auf deren Identifizierung mittels NGS. Wir betrachten insbesondere die folgenden Fragen: Bei welchen häufigen Krankheiten ist ein Beitrag seltener Varianten zu erwarten, wie können diese Varianten identifiziert werden, und welches Potenzial bieten seltene Varianten für das Verständnis biologischer Prozesse bzw. für die Translation in die klinische Praxis?
Collapse
Affiliation(s)
- Kerstin U. Ludwig
- Aff2 0000 0000 8786 803X grid.15090.3d Emmy-Noether-Gruppe „Kraniofaziale Genomik“, Institut für Humangenetik U ni ver si täts kli ni kum Bonn Venusberg-Campus 1, Gebäude 76 53127 Bonn Deutschland
| | - Franziska Degenhardt
- Aff1 0000 0000 8786 803X grid.15090.3d Institut für Humangenetik Universitätsklinikum Bonn Bonn Deutschland
| | - Markus M. Nöthen
- Aff1 0000 0000 8786 803X grid.15090.3d Institut für Humangenetik Universitätsklinikum Bonn Bonn Deutschland
| |
Collapse
|
266
|
Analysis of the genetic basis of height in large Jewish nuclear families. PLoS Genet 2019; 15:e1008082. [PMID: 31283753 PMCID: PMC6638967 DOI: 10.1371/journal.pgen.1008082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 07/18/2019] [Accepted: 03/11/2019] [Indexed: 12/26/2022] Open
Abstract
Despite intensive study, most of the specific genetic factors that contribute to variation in human height remain undiscovered. We conducted a family-based linkage study of height in a unique cohort of very large nuclear families from a founder (Jewish) population. This design allowed for increased power to detect linkage, compared to previous family-based studies. Loci we identified in discovery families could explain an estimated lower bound of 6% of the variance in height in validation families. We showed that these loci are not tagging known common variants associated with height. Rather, we suggest that the observed signals arise from variants with large effects that are rare globally but elevated in frequency in the Jewish population. Rare variants with large effects have been suggested to account for some of the missing heritability of height, but detection of such variants in genome-wide association studies (GWAS) requires very large sample sizes due to their low frequencies. Here, we designed a unique study of height, in which we sought to increase the effective frequency of rare variants in our sample. This was done by assembling a cohort of very large nuclear families, with an average of 12 and a maximum of 20 siblings per nuclear family. In this design, any variant segregating in our cohort has a minimum expected frequency of ~1%, regardless of its frequency in the general population. In addition, we recruited all participants from a founder (Jewish) population, in which some variants that are rare in a cosmopolitan population can rise to high frequencies. To further increase power, we developed methods to obtain highly accurate height measurements, genotype calls, and inheritance pattern reconstructions. These factors allowed us to detect many more QTLs than previous family-based studies of height, despite a modest sample size of only 397 participants. The approach described in this paper provides insights into the genetic basis of height and the roles of population-specific vs. cosmopolitan variants, and may serve as a complement to GWAS for genetic investigations of other complex traits.
Collapse
|
267
|
Spracklen CN, Karaderi T, Yaghootkar H, Schurmann C, Fine RS, Kutalik Z, Preuss MH, Lu Y, Wittemans LBL, Adair LS, Allison M, Amin N, Auer PL, Bartz TM, Blüher M, Boehnke M, Borja JB, Bork-Jensen J, Broer L, Chasman DI, Chen YDI, Chirstofidou P, Demirkan A, van Duijn CM, Feitosa MF, Garcia ME, Graff M, Grallert H, Grarup N, Guo X, Haesser J, Hansen T, Harris TB, Highland HM, Hong J, Ikram MA, Ingelsson E, Jackson R, Jousilahti P, Kähönen M, Kizer JR, Kovacs P, Kriebel J, Laakso M, Lange LA, Lehtimäki T, Li J, Li-Gao R, Lind L, Luan J, Lyytikäinen LP, MacGregor S, Mackey DA, Mahajan A, Mangino M, Männistö S, McCarthy MI, McKnight B, Medina-Gomez C, Meigs JB, Molnos S, Mook-Kanamori D, Morris AP, de Mutsert R, Nalls MA, Nedeljkovic I, North KE, Pennell CE, Pradhan AD, Province MA, Raitakari OT, Raulerson CK, Reiner AP, Ridker PM, Ripatti S, Roberston N, Rotter JI, Salomaa V, Sandoval-Zárate AA, Sitlani CM, Spector TD, Strauch K, Stumvoll M, Taylor KD, Thuesen B, Tönjes A, Uitterlinden AG, Venturini C, Walker M, Wang CA, Wang S, Wareham NJ, Willems SM, Willems van Dijk K, Wilson JG, Wu Y, Yao J, Young KL, Langenberg C, Frayling TM, et alSpracklen CN, Karaderi T, Yaghootkar H, Schurmann C, Fine RS, Kutalik Z, Preuss MH, Lu Y, Wittemans LBL, Adair LS, Allison M, Amin N, Auer PL, Bartz TM, Blüher M, Boehnke M, Borja JB, Bork-Jensen J, Broer L, Chasman DI, Chen YDI, Chirstofidou P, Demirkan A, van Duijn CM, Feitosa MF, Garcia ME, Graff M, Grallert H, Grarup N, Guo X, Haesser J, Hansen T, Harris TB, Highland HM, Hong J, Ikram MA, Ingelsson E, Jackson R, Jousilahti P, Kähönen M, Kizer JR, Kovacs P, Kriebel J, Laakso M, Lange LA, Lehtimäki T, Li J, Li-Gao R, Lind L, Luan J, Lyytikäinen LP, MacGregor S, Mackey DA, Mahajan A, Mangino M, Männistö S, McCarthy MI, McKnight B, Medina-Gomez C, Meigs JB, Molnos S, Mook-Kanamori D, Morris AP, de Mutsert R, Nalls MA, Nedeljkovic I, North KE, Pennell CE, Pradhan AD, Province MA, Raitakari OT, Raulerson CK, Reiner AP, Ridker PM, Ripatti S, Roberston N, Rotter JI, Salomaa V, Sandoval-Zárate AA, Sitlani CM, Spector TD, Strauch K, Stumvoll M, Taylor KD, Thuesen B, Tönjes A, Uitterlinden AG, Venturini C, Walker M, Wang CA, Wang S, Wareham NJ, Willems SM, Willems van Dijk K, Wilson JG, Wu Y, Yao J, Young KL, Langenberg C, Frayling TM, Kilpeläinen TO, Lindgren CM, Loos RJF, Mohlke KL. Exome-Derived Adiponectin-Associated Variants Implicate Obesity and Lipid Biology. Am J Hum Genet 2019; 105:15-28. [PMID: 31178129 DOI: 10.1016/j.ajhg.2019.05.002] [Show More Authors] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/30/2019] [Indexed: 12/25/2022] Open
Abstract
Circulating levels of adiponectin, an adipocyte-secreted protein associated with cardiovascular and metabolic risk, are highly heritable. To gain insights into the biology that regulates adiponectin levels, we performed an exome array meta-analysis of 265,780 genetic variants in 67,739 individuals of European, Hispanic, African American, and East Asian ancestry. We identified 20 loci associated with adiponectin, including 11 that had been reported previously (p < 2 × 10-7). Comparison of exome array variants to regional linkage disequilibrium (LD) patterns and prior genome-wide association study (GWAS) results detected candidate variants (r2 > .60) spanning as much as 900 kb. To identify potential genes and mechanisms through which the previously unreported association signals act to affect adiponectin levels, we assessed cross-trait associations, expression quantitative trait loci in subcutaneous adipose, and biological pathways of nearby genes. Eight of the nine loci were also associated (p < 1 × 10-4) with at least one obesity or lipid trait. Candidate genes include PRKAR2A, PTH1R, and HDAC9, which have been suggested to play roles in adipocyte differentiation or bone marrow adipose tissue. Taken together, these findings provide further insights into the processes that influence circulating adiponectin levels.
Collapse
Affiliation(s)
- Cassandra N Spracklen
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tugce Karaderi
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7FZ, UK; Department of Biological Sciences, Faculty of Arts and Sciences, Eastern Mediterranean University, Famagusta, Cyprus; Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; DTU Health Technology, Technical University of Denmark, Lyngby 2800, Denmark
| | - Hanieh Yaghootkar
- Genetics of Complex Traits, University of Exeter Medical School, Royal Devon & Exeter Hospital, Exeter EX2 5DW, UK; Research Centre for Optimal Health, School of Life Sciences, University of Westminster, London, UK
| | - Claudia Schurmann
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rebecca S Fine
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Division of Endocrinology and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Zoltan Kutalik
- Genetics of Complex Traits, University of Exeter Medical School, Royal Devon & Exeter Hospital, Exeter EX2 5DW, UK; University Center for Primary Care and Public Health, University of Lausanne, Lausanne 1010, Switzerland; Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| | - Michael H Preuss
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yingchang Lu
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt Epidemiology Center, Vanderbilt University School of Medicine, Nashville, TN 37203-1738, USA; Genetics of Obesity and Related Metabolic Traits Program, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Laura B L Wittemans
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7FZ, UK; MRC Epidemiology Unit, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Linda S Adair
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27516, USA
| | - Matthew Allison
- Department of Family Medicine and Public Health, University of California, San Diego, CA 92093, USA
| | - Najaf Amin
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam 3015CN, the Netherlands
| | - Paul L Auer
- Joseph J. Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI 53201, USA
| | - Traci M Bartz
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA 98101, USA; Department of Biostatistics, University of Washington, Seattle, WA 98101, USA
| | - Matthias Blüher
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig, Leipzig 4103, Germany
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Judith B Borja
- Office of Population Studies Foundation, Inc, Cebu City, Philippines; Department of Nutrition and Dietetics, University of San Carlos, Cebu City, Philippines
| | - Jette Bork-Jensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Linda Broer
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam 3000 CA, the Netherlands
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, LABioMed at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Paraskevi Chirstofidou
- Department of Twin Research and Genetic Epidemiology, Kings College London, London SE1 7EH, UK
| | - Ayse Demirkan
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam 3015CN, the Netherlands
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam 3015CN, the Netherlands
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Melissa E Garcia
- National Heart, Lung, and Blood Institute, Bethesda, MD 20892, USA
| | - Mariaelisa Graff
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Carolina Center for Genome Sciences, Chapel Hill, NC 27599, USA
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München Research Center for Environmental Health, München-Neuherberg 85764, Germany; German Center for Diabetes Research, München-Neuherberg 85765, Germany
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, LABioMed at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Jeffrey Haesser
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIH, Bethesda, MD 20892, USA
| | - Heather M Highland
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jaeyoung Hong
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 2118, USA
| | - M Arfan Ikram
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam 3000 CA, the Netherlands; Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam 3000 CA, the Netherlands
| | - Erik Ingelsson
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University, Palo Alto, CA 94304, USA; Stanford Cardiovascular Institute, Stanford University of Medicine, Palo Alto, CA 94304, USA; Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala 75185, Sweden; Stanford Diabetes Research Center, Stanford University, Stanford, CA 94305, USA
| | - Rebecca Jackson
- Division of Endocrinology, Diabetes, and Metabolism, Ohio State University, Columbus, OH 43210, USA
| | - Pekka Jousilahti
- Department of Public Health Solutions, National Institute for Health and Welfare, Helsinki 00271, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, Tampere 33522, Finland; Department of Clinical Physiology, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere 33522, Finland
| | - Jorge R Kizer
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Peter Kovacs
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig, Leipzig 4103, Germany
| | - Jennifer Kriebel
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München Research Center for Environmental Health, München-Neuherberg 85764, Germany; German Center for Diabetes Research, München-Neuherberg 85765, Germany
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University of Hospital, Kuopio 70029 KYS, Finland
| | - Leslie A Lange
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado-Denver, Denver, CO 80045, USA
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere 33520, Finland; Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere 33522, Finland
| | - Jin Li
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Ruifang Li-Gao
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala 75185, Sweden
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere 33522, Finland; Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere 33521, Finland
| | - Stuart MacGregor
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - David A Mackey
- Faculty of Health and Medical Sciences, The University of Western Australia, Perth, WA 6009, Australia; Centre for Ophthalmology and Visual Science, Lions Eye Institute, The University of Western Australia, Perth, WA 6009, Australia
| | - Anubha Mahajan
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7FZ, UK; Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, Kings College London, London SE1 7EH, UK; NIHR Biomedical Research Centre, Guy's and St Thomas' Foundation Trust, London SE1 9RT, UK
| | - Satu Männistö
- Department of Public Health Solutions, National Institute for Health and Welfare, Helsinki 00271, Finland
| | - Mark I McCarthy
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7FZ, UK; Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK; Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford OX3 7FZ, UK
| | - Barbara McKnight
- Department of Biostatistics, University of Washington, Seattle, WA 98101, USA
| | - Carolina Medina-Gomez
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam 3000 CA, the Netherlands; Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam 3000 CA, the Netherlands
| | - James B Meigs
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Program in Population and Medical Genetics, Broad Institute, Cambridge, MA 02114, USA
| | - Sophie Molnos
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München Research Center for Environmental Health, München-Neuherberg 85764, Germany; German Center for Diabetes Research, München-Neuherberg 85765, Germany
| | - Dennis Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands; Department of Public Health and Primary Care, Leiden University Medical Center, Leiden 2334 ZA, the Netherlands
| | - Andrew P Morris
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7FZ, UK; Department of Biostatistics, University of Liverpool, Liverpool L69 3GL, UK
| | - Renee de Mutsert
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Mike A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD 20892, USA; Data Tecnica International, Glen Echo, MD 20812, USA
| | - Ivana Nedeljkovic
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam 3015CN, the Netherlands
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Craig E Pennell
- School of Medicine and Public Health, Faculty of Medicine and Health, The University of Newcastle, Newcastle, NSW 2305, Australia
| | - Aruna D Pradhan
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Michael A Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Olli T Raitakari
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland; Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland; Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - Chelsea K Raulerson
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Alex P Reiner
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Samuli Ripatti
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Public Health, University of Helsinki, Helsinki 00014, Finland; Institute for Molecular Medicine Finland, Helsinki 00014, Finland
| | - Neil Roberston
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7FZ, UK; Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, LABioMed at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Veikko Salomaa
- Department of Public Health Solutions, National Institute for Health and Welfare, Helsinki 00271, Finland
| | | | - Colleen M Sitlani
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA 98101, USA
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, Kings College London, London SE1 7EH, UK
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg 85764, Germany; Chair of Genetic Epidemiology, Institute of Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-Universität, Munich 81377, Germany
| | - Michael Stumvoll
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig, Leipzig 4103, Germany
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, LABioMed at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Betina Thuesen
- Center for Clinical Research and Disease Prevention, Bispebjerg and Frederiksberg Hospital, The Capital Region, Copenhagen 2400, Denmark
| | - Anke Tönjes
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig, Leipzig 4103, Germany
| | - Andre G Uitterlinden
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam 3000 CA, the Netherlands; Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam 3000 CA, the Netherlands
| | - Cristina Venturini
- Department of Twin Research and Genetic Epidemiology, Kings College London, London SE1 7EH, UK
| | - Mark Walker
- Institute of Cellular Medicine, The Medical School, Newcastle University, Newcastle, UK
| | - Carol A Wang
- School of Medicine and Public Health, Faculty of Medicine and Health, The University of Newcastle, Newcastle, NSW 2305, Australia
| | - Shuai Wang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 2118, USA
| | | | - Sara M Willems
- MRC Epidemiology Unit, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Ko Willems van Dijk
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden 2333 ZA, the Netherlands; Department of Human Genetics, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - James G Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Ying Wu
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, LABioMed at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Kristin L Young
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | - Timothy M Frayling
- Genetics of Complex Traits, University of Exeter Medical School, Royal Devon & Exeter Hospital, Exeter EX2 5DW, UK
| | - Tuomas O Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Cecilia M Lindgren
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7FZ, UK; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7LF, UK
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Mindich Child Health and Development Institute, Ichan School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
268
|
Dauber A. Genetic Testing for the Child With Short Stature-Has the Time Come To Change Our Diagnostic Paradigm? J Clin Endocrinol Metab 2019; 104:2766-2769. [PMID: 30753512 DOI: 10.1210/jc.2019-00019] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 02/05/2019] [Indexed: 12/29/2022]
Affiliation(s)
- Andrew Dauber
- Division of Endocrinology, Children's National Health System, Washington, DC
| |
Collapse
|
269
|
Milanesi E, Voinsky I, Hadar A, Srouji A, Maj C, Shekhtman T, Gershovits M, Gilad S, Chillotti C, Squassina A, Potash JB, Schulze TG, Goes FS, Zandi P, Kelsoe JR, Gurwitz D. RNA sequencing of bipolar disorder lymphoblastoid cell lines implicates the neurotrophic factor HRP-3 in lithium's clinical efficacy. World J Biol Psychiatry 2019; 20:449-461. [PMID: 28854847 DOI: 10.1080/15622975.2017.1372629] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Objectives: Lithium remains the oldest and most effective treatment for mood stabilisation in bipolar disorder (BD), even though at least half of patients are only partially responsive or do not respond. This study aimed to identify biomarkers associated with lithium response in BD, based on comparing RNA sequencing information derived from lymphoblastoid cell lines (LCLs) of lithium-responsive (LR) versus lithium non-responsive (LNR) BD patients, to assess gene expression variations that might bear on treatment outcome. Methods: RNA sequencing was carried out on 24 LCLs from female BD patients (12 LR and 12 LNR) followed by qPCR validation in two additional independent cohorts (41 and 17 BD patients, respectively). Results: Fifty-six genes showed nominal differential expression comparing LR and LNR (FC ≥ |1.3|, P ≤ 0.01). The differential expression of HDGFRP3 and ID2 was validated by qPCR in the independent cohorts. Conclusions: We observed higher expression levels of HDGFRP3 and ID2 in BD patients who favourably respond to lithium. Both of these genes are involved in neurogenesis, and HDGFRP3 has been suggested to be a neurotrophic factor. Additional studies in larger BD cohorts are needed to confirm the potential of HDGFRP3 and ID2 expression levels in blood cells as tentative favourable lithium response biomarkers.
Collapse
Affiliation(s)
- Elena Milanesi
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University , Tel Aviv , Israel.,Genetics Unit, IRCCS, San Giovanni di Dio, Fatebenefratelli , Brescia , Italy
| | - Irena Voinsky
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University , Tel Aviv , Israel
| | - Adva Hadar
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University , Tel Aviv , Israel
| | - Ala Srouji
- Institute of Psychiatric Phenomics and Genomics, Ludwig-Maximilians-University Munich , Munich , Germany.,Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health , Mannheim , Germany
| | - Carlo Maj
- Genetics Unit, IRCCS, San Giovanni di Dio, Fatebenefratelli , Brescia , Italy
| | - Tatyana Shekhtman
- Department of Psychiatry, University of California , San Diego , CA , USA
| | - Michael Gershovits
- The Nancy & Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science , Rehovot , Israel
| | - Shlomit Gilad
- The Nancy & Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science , Rehovot , Israel
| | - Caterina Chillotti
- Unit of Clinical Pharmacology, University Hospital of Cagliari , Cagliari , Italy
| | - Alessio Squassina
- Section of Neurosciences and Clinical Pharmacology, Department of Biomedical Sciences, School of Medicine, University of Cagliari , Cagliari , Italy
| | - James B Potash
- Department of Psychiatry, University of Iowa Carver College of Medicine , Iowa City , IA , USA
| | - Thomas G Schulze
- Institute of Psychiatric Phenomics and Genomics, Ludwig-Maximilians-University Munich , Munich , Germany.,Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health , Mannheim , Germany.,Department of Psychiatry and Psychotherapy, University Medical Center Georg-August-University , Göttingen , Germany
| | - Fernando S Goes
- Department of Psychiatry, Johns Hopkins University , Baltimore , MD , USA
| | - Peter Zandi
- Department of Psychiatry, Johns Hopkins University , Baltimore , MD , USA
| | - John R Kelsoe
- Department of Psychiatry, University of California , San Diego , CA , USA
| | - David Gurwitz
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University , Tel Aviv , Israel
| |
Collapse
|
270
|
Baral K, Rotwein P. The insulin-like growth factor 2 gene in mammals: Organizational complexity within a conserved locus. PLoS One 2019; 14:e0219155. [PMID: 31251794 PMCID: PMC6599137 DOI: 10.1371/journal.pone.0219155] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 06/17/2019] [Indexed: 01/10/2023] Open
Abstract
The secreted protein, insulin-like growth factor 2 (IGF2), plays a central role in fetal and prenatal growth and development, and is regulated at the genetic level by parental imprinting, being expressed predominantly from the paternally derived chromosome in mice and humans. Here, IGF2/Igf2 and its locus has been examined in 19 mammals from 13 orders spanning ~166 million years of evolutionary development. By using human or mouse DNA segments as queries in genome analyses, and by assessing gene expression using RNA-sequencing libraries, more complexity was identified within IGF2/Igf2 than was annotated previously. Multiple potential 5’ non-coding exons were mapped in most mammals and are presumably linked to distinct IGF2/Igf2 promoters, as shown for several species by interrogating RNA-sequencing libraries. DNA similarity was highest in IGF2/Igf2 coding exons; yet, even though the mature IGF2 protein was conserved, versions of 67 or 70 residues are produced secondary to species-specific maintenance of alternative RNA splicing at a variable intron-exon junction. Adjacent H19 was more divergent than IGF2/Igf2, as expected in a gene for a noncoding RNA, and was identified in only 10/19 species. These results show that common features, including those defining IGF2/Igf2 coding and several non-coding exons, were likely present at the onset of the mammalian radiation, but that others, such as a putative imprinting control region 5’ to H19 and potential enhancer elements 3’ to H19, diversified with speciation. This study also demonstrates that careful analysis of genomic and gene expression repositories can provide new insights into gene structure and regulation.
Collapse
Affiliation(s)
- Kabita Baral
- Graduate School, College of Science, University of Texas at El Paso, El Paso, Texas
| | - Peter Rotwein
- Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech Health University Health Sciences Center, El Paso, Texas
- * E-mail:
| |
Collapse
|
271
|
Nikopoulos K, Cisarova K, Quinodoz M, Koskiniemi-Kuendig H, Miyake N, Farinelli P, Rehman AU, Khan MI, Prunotto A, Akiyama M, Kamatani Y, Terao C, Miya F, Ikeda Y, Ueno S, Fuse N, Murakami A, Wada Y, Terasaki H, Sonoda KH, Ishibashi T, Kubo M, Cremers FPM, Kutalik Z, Matsumoto N, Nishiguchi KM, Nakazawa T, Rivolta C. A frequent variant in the Japanese population determines quasi-Mendelian inheritance of rare retinal ciliopathy. Nat Commun 2019; 10:2884. [PMID: 31253780 PMCID: PMC6599023 DOI: 10.1038/s41467-019-10746-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 05/23/2019] [Indexed: 12/21/2022] Open
Abstract
Hereditary retinal degenerations (HRDs) are Mendelian diseases characterized by progressive blindness and caused by ultra-rare mutations. In a genomic screen of 331 unrelated Japanese patients, we identify a disruptive Alu insertion and a nonsense variant (p.Arg1933*) in the ciliary gene RP1, neither of which are rare alleles in Japan. p.Arg1933* is almost polymorphic (frequency = 0.6%, amongst 12,000 individuals), does not cause disease in homozygosis or heterozygosis, and yet is significantly enriched in HRD patients (frequency = 2.1%, i.e., a 3.5-fold enrichment; p-value = 9.2 × 10-5). Familial co-segregation and association analyses show that p.Arg1933* can act as a Mendelian mutation in trans with the Alu insertion, but might also associate with disease in combination with two alleles in the EYS gene in a non-Mendelian pattern of heredity. Our results suggest that rare conditions such as HRDs can be paradoxically determined by relatively common variants, following a quasi-Mendelian model linking monogenic and complex inheritance.
Collapse
Affiliation(s)
- Konstantinos Nikopoulos
- Unit of Medical Genetics, Department of Computational Biology, University of Lausanne, 1015, Lausanne, Switzerland
- Service of Medical Genetics, Lausanne University Hospital (CHUV), 1011, Lausanne, Switzerland
| | - Katarina Cisarova
- Unit of Medical Genetics, Department of Computational Biology, University of Lausanne, 1015, Lausanne, Switzerland
| | - Mathieu Quinodoz
- Unit of Medical Genetics, Department of Computational Biology, University of Lausanne, 1015, Lausanne, Switzerland
| | - Hanna Koskiniemi-Kuendig
- Unit of Medical Genetics, Department of Computational Biology, University of Lausanne, 1015, Lausanne, Switzerland
| | - Noriko Miyake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Pietro Farinelli
- Unit of Medical Genetics, Department of Computational Biology, University of Lausanne, 1015, Lausanne, Switzerland
| | - Atta Ur Rehman
- Unit of Medical Genetics, Department of Computational Biology, University of Lausanne, 1015, Lausanne, Switzerland
| | - Muhammad Imran Khan
- Department of Human Genetics, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, 6525 GA, Nijmegen, The Netherlands
| | - Andrea Prunotto
- Unit of Medical Genetics, Department of Computational Biology, University of Lausanne, 1015, Lausanne, Switzerland
| | - Masato Akiyama
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Chikashi Terao
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Fuyuki Miya
- Department of Medical Science Mathematics, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan
| | - Yasuhiro Ikeda
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Shinji Ueno
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Nobuo Fuse
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Sendai, 980-8573, Japan
| | - Akira Murakami
- Department of Ophthalmology, Juntendo University School of Medicine, Tokyo, 113-8421, Japan
| | - Yuko Wada
- Yuko Wada Eye Clinic, Sendai, 980-0011, Japan
| | - Hiroko Terasaki
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Koh-Hei Sonoda
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Tatsuro Ishibashi
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Michiaki Kubo
- RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Frans P M Cremers
- Department of Human Genetics, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, 6525 GA, Nijmegen, The Netherlands
| | - Zoltán Kutalik
- Institute of Social and Preventive Medicine, Lausanne University Hospital, 1011, Lausanne, Switzerland
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Koji M Nishiguchi
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
- Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Toru Nakazawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
- Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Carlo Rivolta
- Unit of Medical Genetics, Department of Computational Biology, University of Lausanne, 1015, Lausanne, Switzerland.
- Department of Genetics and Genome Biology, University of Leicester, Leicester, LE1 7RH, UK.
- Institute of Molecular and Clinical Ophthalmology Basel (IOB), 4031, Basel, Switzerland.
- University of Basel, 4001, Basel, Switzerland.
| |
Collapse
|
272
|
McInnes G, Daneshjou R, Katsonis P, Lichtarge O, Srinivasan R, Rana S, Radivojac P, Mooney SD, Pagel KA, Stamboulian M, Jiang Y, Capriotti E, Wang Y, Bromberg Y, Bovo S, Savojardo C, Martelli PL, Casadio R, Pal LR, Moult J, Brenner SE, Altman R. Predicting venous thromboembolism risk from exomes in the Critical Assessment of Genome Interpretation (CAGI) challenges. Hum Mutat 2019; 40:1314-1320. [PMID: 31140652 DOI: 10.1002/humu.23825] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/07/2019] [Accepted: 05/27/2019] [Indexed: 01/14/2023]
Abstract
Genetics play a key role in venous thromboembolism (VTE) risk, however established risk factors in European populations do not translate to individuals of African descent because of the differences in allele frequencies between populations. As part of the fifth iteration of the Critical Assessment of Genome Interpretation, participants were asked to predict VTE status in exome data from African American subjects. Participants were provided with 103 unlabeled exomes from patients treated with warfarin for non-VTE causes or VTE and asked to predict which disease each subject had been treated for. Given the lack of training data, many participants opted to use unsupervised machine learning methods, clustering the exomes by variation in genes known to be associated with VTE. The best performing method using only VTE related genes achieved an area under the ROC curve of 0.65. Here, we discuss the range of methods used in the prediction of VTE from sequence data and explore some of the difficulties of conducting a challenge with known confounders. In addition, we show that an existing genetic risk score for VTE that was developed in European subjects works well in African Americans.
Collapse
Affiliation(s)
- Gregory McInnes
- Biomedical Informatics Training Program, Stanford University, Stanford, California
| | - Roxana Daneshjou
- Department of Dermatology, Stanford School of Medicine, Stanford, California
| | - Panagiostis Katsonis
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Olivier Lichtarge
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.,Department of Biochemistry & Molecular Biology, Baylor College of Medicine, Houston, Texas.,Department of Pharmacology, Baylor College of Medicine, Houston, Texas.,Computational and Integrative Biomedical Research Center, Baylor College of Medicine, Houston, Texas
| | | | - Sadhna Rana
- Innovations Labs, Tata Consultancy Services, Hyderabad, India
| | - Predrag Radivojac
- Khoury College of Computer and Information Sciences, Northeastern University, Boston, Massachusetts
| | - Sean D Mooney
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, Washington
| | - Kymberleigh A Pagel
- Department of Computer Science and Informatics, Indiana University, Bloomington, Indiana
| | - Moses Stamboulian
- Department of Computer Science and Informatics, Indiana University, Bloomington, Indiana
| | - Yuxiang Jiang
- Department of Computer Science and Informatics, Indiana University, Bloomington, Indiana
| | - Emidio Capriotti
- BioFolD Unit, Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Bologna, Italy
| | - Yanran Wang
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, New Jersey
| | - Yana Bromberg
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, New Jersey
| | - Samuele Bovo
- Department of Pharmacy and Biotechnology, Bologna Biocomputing Group, University of Bologna, Italy
| | - Castrense Savojardo
- Department of Pharmacy and Biotechnology, Bologna Biocomputing Group, University of Bologna, Italy
| | - Pier Luigi Martelli
- Department of Pharmacy and Biotechnology, Bologna Biocomputing Group, University of Bologna, Italy
| | - Rita Casadio
- Department of Pharmacy and Biotechnology, Bologna Biocomputing Group, University of Bologna, Italy.,Institute of Biomembrane and Bioenergetics, Consiglio Nazionale delle Ricerche, Bari, Italy
| | - Lipika R Pal
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland
| | - John Moult
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland.,Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland
| | - Steven E Brenner
- Department of Plant and Microbial biology, University of California Berkeley, Berkeley, California
| | - Russ Altman
- Departments of Bioengineering, Biomedical Data Science, Genetics, and Medicine, Stanford University, Stanford, California
| |
Collapse
|
273
|
Sutherland HG, Albury CL, Griffiths LR. Advances in genetics of migraine. J Headache Pain 2019; 20:72. [PMID: 31226929 PMCID: PMC6734342 DOI: 10.1186/s10194-019-1017-9] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 05/24/2019] [Indexed: 02/06/2023] Open
Abstract
Background Migraine is a complex neurovascular disorder with a strong genetic component. There are rare monogenic forms of migraine, as well as more common polygenic forms; research into the genes involved in both types has provided insights into the many contributing genetic factors. This review summarises advances that have been made in the knowledge and understanding of the genes and genetic variations implicated in migraine etiology. Findings Migraine is characterised into two main types, migraine without aura (MO) and migraine with aura (MA). Hemiplegic migraine is a rare monogenic MA subtype caused by mutations in three main genes - CACNA1A, ATP1A2 and SCN1A - which encode ion channel and transport proteins. Functional studies in cellular and animal models show that, in general, mutations result in impaired glutamatergic neurotransmission and cortical hyperexcitability, which make the brain more susceptible to cortical spreading depression, a phenomenon thought to coincide with aura symptoms. Variants in other genes encoding ion channels and solute carriers, or with roles in regulating neurotransmitters at neuronal synapses, or in vascular function, can also cause monogenic migraine, hemiplegic migraine and related disorders with overlapping symptoms. Next-generation sequencing will accelerate the finding of new potentially causal variants and genes, with high-throughput bioinformatics analysis methods and functional analysis pipelines important in prioritising, confirming and understanding the mechanisms of disease-causing variants. With respect to common migraine forms, large genome-wide association studies (GWAS) have greatly expanded our knowledge of the genes involved, emphasizing the role of both neuronal and vascular pathways. Dissecting the genetic architecture of migraine leads to greater understanding of what underpins relationships between subtypes and comorbid disorders, and may have utility in diagnosis or tailoring treatments. Further work is required to identify causal polymorphisms and the mechanism of their effect, and studies of gene expression and epigenetic factors will help bridge the genetics with migraine pathophysiology. Conclusions The complexity of migraine disorders is mirrored by their genetic complexity. A comprehensive knowledge of the genetic factors underpinning migraine will lead to improved understanding of molecular mechanisms and pathogenesis, to enable better diagnosis and treatments for migraine sufferers.
Collapse
Affiliation(s)
- Heidi G Sutherland
- Genomics Research Centre, Institute of Health and Biomedical Innovation. School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Cassie L Albury
- Genomics Research Centre, Institute of Health and Biomedical Innovation. School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Lyn R Griffiths
- Genomics Research Centre, Institute of Health and Biomedical Innovation. School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD, Australia.
| |
Collapse
|
274
|
Li JB, Liu ZX, Zhang R, Ma SP, Lin T, Li YX, Yang SH, Zhang WC, Wang YP. Sp1 contributes to overexpression of stanniocalcin 2 through regulation of promoter activity in colon adenocarcinoma. World J Gastroenterol 2019; 25:2776-2787. [PMID: 31236000 PMCID: PMC6580349 DOI: 10.3748/wjg.v25.i22.2776] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/22/2019] [Accepted: 04/29/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Aberrant expression of stanniocalcin 2 (STC2) is implicated in colon adenocarcinoma (COAD). A previous study identified that STC2 functions as a tumor promoter to drive development of some cancers, but the role of its overexpression in the development of COAD remains unclear. AIM To evaluate the regulation mechanism of STC2 overexpression in COAD. METHODS The expression of STC2 in COAD was assessed by TCGA COAD database and GEO (GSE50760). Methylation level of the STC2 promoter was evaluated with beta value in UALCAN platform, and the correlation between STC2 expression and survival rate was investigated with TCGA COAD. Transcription binding site prediction was conducted by TRANSFAC and LASAGNA, and a luciferase reporter system was used to identify STC2 promoter activity in several cell lines, including HEK293T, NCM460, HT29, SW480, and HCT116. Western blotting was performed to evaluate the role of Sp1 on the expression of STC2. RESULTS The central finding of this work is that STC2 is overexpressed in COAD tissues and positively correlated with poor prognosis. Importantly, the binding site of the transcription factor Sp1 is widely located in the promoter region of STC2. A luciferase reporter system was successfully constructed to analyze the transcription activity of STC2, and knocking down the expression of Sp1 significantly inhibited the transcription activity of STC2. Furthermore, inhibition of Sp1 remarkably decreased protein levels of STC2. CONCLUSION Our data provide evidence that the transcription factor Sp1 is essential for the overexpression of STC2 in COAD through activation of promoter activity. Taken together, our finding provides new insights into the mechanism of oncogenic function of COAD by STC2.
Collapse
Affiliation(s)
- Ji-Bin Li
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, China
| | - Zhe-Xian Liu
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, China
| | - Rui Zhang
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, China
| | - Si-Ping Ma
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, China
| | - Tao Lin
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, China
| | - Yan-Xi Li
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, China
| | - Shi-Hua Yang
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, China
- China Medical University, Shenyang 110000, Liaoning Province, China
| | - Wan-Chuan Zhang
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, China
- China Medical University, Shenyang 110000, Liaoning Province, China
| | - Yong-Peng Wang
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, China
| |
Collapse
|
275
|
Gervais L, Perrier C, Bernard M, Merlet J, Pemberton JM, Pujol B, Quéméré E. RAD-sequencing for estimating genomic relatedness matrix-based heritability in the wild: A case study in roe deer. Mol Ecol Resour 2019; 19:1205-1217. [PMID: 31058463 DOI: 10.1111/1755-0998.13031] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 04/19/2019] [Accepted: 04/23/2019] [Indexed: 01/02/2023]
Abstract
Estimating the evolutionary potential of quantitative traits and reliably predicting responses to selection in wild populations are important challenges in evolutionary biology. The genomic revolution has opened up opportunities for measuring relatedness among individuals with precision, enabling pedigree-free estimation of trait heritabilities in wild populations. However, until now, most quantitative genetic studies based on a genomic relatedness matrix (GRM) have focused on long-term monitored populations for which traditional pedigrees were also available, and have often had access to knowledge of genome sequence and variability. Here, we investigated the potential of RAD-sequencing for estimating heritability in a free-ranging roe deer (Capreolous capreolus) population for which no prior genomic resources were available. We propose a step-by-step analytical framework to optimize the quality and quantity of the genomic data and explore the impact of the single nucleotide polymorphism (SNP) calling and filtering processes on the GRM structure and GRM-based heritability estimates. As expected, our results show that sequence coverage strongly affects the number of recovered loci, the genotyping error rate and the amount of missing data. Ultimately, this had little effect on heritability estimates and their standard errors, provided that the GRM was built from a minimum number of loci (above 7,000). Genomic relatedness matrix-based heritability estimates thus appear robust to a moderate level of genotyping errors in the SNP data set. We also showed that quality filters, such as the removal of low-frequency variants, affect the relatedness structure of the GRM, generating lower h2 estimates. Our work illustrates the huge potential of RAD-sequencing for estimating GRM-based heritability in virtually any natural population.
Collapse
Affiliation(s)
- Laura Gervais
- CEFS, INRA, Université de Toulouse, Castanet-Tolosan, Cedex, France.,Laboratoire Évolution & Diversité Biologique (EDB UMR 5174), CNRS, IRD, UPS, Université Fédérale de Toulouse Midi-Pyrénées, Toulouse, France
| | | | - Maria Bernard
- SIGENAE, INRA, Jouy-en-Josas, France.,GABI, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Joël Merlet
- CEFS, INRA, Université de Toulouse, Castanet-Tolosan, Cedex, France
| | - Josephine M Pemberton
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Benoit Pujol
- Laboratoire Évolution & Diversité Biologique (EDB UMR 5174), CNRS, IRD, UPS, Université Fédérale de Toulouse Midi-Pyrénées, Toulouse, France.,PSL Université Paris: EPHE-UPVD-CNRS, Université de Perpignan, Perpignan, France
| | - Erwan Quéméré
- CEFS, INRA, Université de Toulouse, Castanet-Tolosan, Cedex, France
| |
Collapse
|
276
|
Vasques GA, Andrade NLM, Jorge AAL. Genetic causes of isolated short stature. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2019; 63:70-78. [PMID: 30864634 PMCID: PMC10118839 DOI: 10.20945/2359-3997000000105] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 02/22/2019] [Indexed: 11/23/2022]
Abstract
Short stature is a common feature, and frequently remains without a specific diagnosis after conventional clinical and laboratorial evaluation. Longitudinal growth is mainly determined by genetic factors, and hundreds of common variants have been associated to height variability among healthy individuals. Although isolated short stature may be caused by the combination of variants, with a deleterious impact on the growth of individuals with polygenic inheritance, recent studies have pointed out some monogenic defects as the cause of the growth disorder observed in nonsyndromic children. The majority of these defects are in genes related to the growth plate cartilage and in the growth hormone (GH) - insulin-like growth factor 1 (IGF-1) axis. Affected patients usually present the mildest spectrum of some forms of skeletal dysplasia, or subtle abnormalities of laboratory tests, suggesting hormonal resistance or insensibility. The lack of specific characteristics, however, does not allow formulation of a definitive diagnosis without the use of broad genetic studies. Thus, molecular genetic studies including panels of genes or exome analysis will become essential in investigating and identifying the causes of isolated short stature in children, with a crucial impact on treatment and follow-up.
Collapse
Affiliation(s)
- Gabriela A Vasques
- Unidade de Endocrinologia Genética (LIM25), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brasil.,Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM42), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brasil
| | - Nathalia L M Andrade
- Unidade de Endocrinologia Genética (LIM25), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brasil.,Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM42), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brasil
| | - Alexander A L Jorge
- Unidade de Endocrinologia Genética (LIM25), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brasil.,Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM42), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brasil
| |
Collapse
|
277
|
Missing heritability of complex diseases: case solved? Hum Genet 2019; 139:103-113. [DOI: 10.1007/s00439-019-02034-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 05/28/2019] [Indexed: 10/26/2022]
|
278
|
Liu F, Zhong K, Jing X, Uitterlinden AG, Hendriks AEJ, Drop SLS, Kayser M. Update on the predictability of tall stature from DNA markers in Europeans. Forensic Sci Int Genet 2019; 42:8-13. [PMID: 31207428 DOI: 10.1016/j.fsigen.2019.05.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 05/08/2019] [Accepted: 05/27/2019] [Indexed: 10/26/2022]
Abstract
Predicting adult height from DNA has important implications in forensic DNA phenotyping. In 2014, we introduced a prediction model consisting of 180 height-associated SNPs based on data from 10,361 Northwestern Europeans enriched with tall individuals (770 > 1.88 standard deviation), which yielded a mid-ranged accuracy (AUC = 0.75 for binary prediction of tall stature and R2 = 0.12 for quantitative prediction of adult height). Here, we provide an update on DNA-based height predictability considering an enlarged list of subsequently-published height-associated SNPs using data from the same set of 10,361 Europeans. A prediction model based on the full set of 689 SNPs showed an improved accuracy relative to previous models for both tall stature (AUC = 0.79) and quantitative height (R2 = 0.21). A feature selection analysis revealed a subset of 412 most informative SNPs while the corresponding prediction model retained most of the accuracy (AUC = 0.76 and R2 = 0.19) achieved with the full model. Over all, our study empirically exemplifies that the accuracy for predicting human appearance phenotypes with very complex underlying genetic architectures, such as adult height, can be improved by increasing the number of phenotype-associated DNA variants. Our work also demonstrates that a careful sub-selection allows for a considerable reduction of the number of DNA predictors that achieve similar prediction accuracy as provided by the full set. This is forensically relevant due to restrictions in the number of SNPs simultaneously analyzable with forensically suitable DNA technologies in the current days of targeted massively parallel sequencing in forensic genetics.
Collapse
Affiliation(s)
- Fan Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China; Department of Genetic Identification, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - Kaiyin Zhong
- Department of Genetic Identification, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - Xiaoxi Jing
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China.
| | - André G Uitterlinden
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - A Emile J Hendriks
- Department of Pediatrics, Pediatric Endocrinology and Diabetes, University of Cambridge, United Kingdom.
| | - Stenvert L S Drop
- Department of Pediatrics, Division of Endocrinology, Sophia Children's Hospital, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - Manfred Kayser
- Department of Genetic Identification, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands.
| |
Collapse
|
279
|
Freire BL, Homma TK, Funari MFA, Lerario AM, Vasques GA, Malaquias AC, Arnhold IJP, Jorge AAL. Multigene Sequencing Analysis of Children Born Small for Gestational Age With Isolated Short Stature. J Clin Endocrinol Metab 2019; 104:2023-2030. [PMID: 30602027 DOI: 10.1210/jc.2018-01971] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 12/27/2018] [Indexed: 02/04/2023]
Abstract
CONTEXT Patients born small for gestational age (SGA) who present with persistent short stature could have an underlying genetic etiology that will account for prenatal and postnatal growth impairment. We applied a unique massive parallel sequencing approach in cohort of patients with exclusively nonsyndromic SGA to simultaneously interrogate for clinically substantial genetic variants. OBJECTIVE To perform a genetic investigation of children with isolated short stature born SGA. DESIGN Screening by exome (n = 16) or targeted gene panel (n = 39) sequencing. SETTING Tertiary referral center for growth disorders. PATIENTS AND METHODS We selected 55 patients born SGA with persistent short stature without an identified cause of short stature. MAIN OUTCOME MEASURES Frequency of pathogenic findings. RESULTS We identified heterozygous pathogenic or likely pathogenic genetic variants in 8 of 55 patients, all in genes already associated with growth disorders. Four of the genes are associated with growth plate development, IHH (n = 2), NPR2 (n = 2), SHOX (n = 1), and ACAN (n = 1), and two are involved in the RAS/MAPK pathway, PTPN11 (n = 1) and NF1 (n = 1). None of these patients had clinical findings that allowed for a clinical diagnosis. Seven patients were SGA only for length and one was SGA for both length and weight. CONCLUSION These genomic approaches identified pathogenic or likely pathogenic genetic variants in 8 of 55 patients (15%). Six of the eight patients carried variants in genes associated with growth plate development, indicating that mild forms of skeletal dysplasia could be a cause of growth disorders in this group of patients.
Collapse
Affiliation(s)
- Bruna L Freire
- Unidade de Endocrinologia Genética, Laboratório de Endocrinologia Celular e Molecular LIM25, Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de São Paulo, São Paulo CEP, Brazil
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo CEP, Brazil
| | - Thais K Homma
- Unidade de Endocrinologia Genética, Laboratório de Endocrinologia Celular e Molecular LIM25, Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de São Paulo, São Paulo CEP, Brazil
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo CEP, Brazil
| | - Mariana F A Funari
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo CEP, Brazil
| | - Antônio M Lerario
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, Michigan
| | - Gabriela A Vasques
- Unidade de Endocrinologia Genética, Laboratório de Endocrinologia Celular e Molecular LIM25, Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de São Paulo, São Paulo CEP, Brazil
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo CEP, Brazil
| | - Alexsandra C Malaquias
- Unidade de Endocrinologia Genética, Laboratório de Endocrinologia Celular e Molecular LIM25, Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de São Paulo, São Paulo CEP, Brazil
- Unidade de Endocrinologia Pediátrica, Departamento de Pediatria, Irmandade da Santa Casa de Misericórdia de São Paulo, Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, Brazil
| | - Ivo J P Arnhold
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo CEP, Brazil
| | - Alexander A L Jorge
- Unidade de Endocrinologia Genética, Laboratório de Endocrinologia Celular e Molecular LIM25, Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de São Paulo, São Paulo CEP, Brazil
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo CEP, Brazil
| |
Collapse
|
280
|
Wünnemann F, Sin Lo K, Langford-Avelar A, Busseuil D, Dubé MP, Tardif JC, Lettre G. Validation of Genome-Wide Polygenic Risk Scores for Coronary Artery Disease in French Canadians. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2019; 12:e002481. [PMID: 31184202 PMCID: PMC6587223 DOI: 10.1161/circgen.119.002481] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 05/17/2019] [Indexed: 11/16/2022]
Abstract
BACKGROUND Coronary artery disease (CAD) represents one of the leading causes of morbidity and mortality worldwide. Given the healthcare risks and societal impacts associated with CAD, their clinical management would benefit from improved prevention and prediction tools. Polygenic risk scores (PRS) based on an individual's genome sequence are emerging as potentially powerful biomarkers to predict the risk to develop CAD. Two recently derived genome-wide PRS have shown high specificity and sensitivity to identify CAD cases in European-ancestry participants from the UK Biobank. However, validation of the PRS predictive power and transferability in other populations is now required to support their clinical utility. METHODS We calculated both PRS (GPSCAD and metaGRSCAD) in French-Canadian individuals from 3 cohorts totaling 3639 prevalent CAD cases and 7382 controls and tested their power to predict prevalent, incident, and recurrent CAD. We also estimated the impact of the founder French-Canadian familial hypercholesterolemia deletion ( LDLR delta >15 kb deletion) on CAD risk in one of these cohorts and used this estimate to calibrate the impact of the PRS. RESULTS Our results confirm the ability of both PRS to predict prevalent CAD comparable to the original reports (area under the curve=0.72-0.89). Furthermore, the PRS identified about 6% to 7% of individuals at CAD risk similar to carriers of the LDLR delta >15 kb mutation, consistent with previous estimates. However, the PRS did not perform as well in predicting an incident or recurrent CAD (area under the curve=0.56-0.60), maybe because of confounding because 76% of the participants were on statin treatment. This result suggests that additional work is warranted to better understand how ascertainment biases and study design impact PRS for CAD. CONCLUSIONS Collectively, our results confirm that novel, genome-wide PRS is able to predict CAD in French Canadians; with further improvements, this is likely to pave the way towards more targeted strategies to predict and prevent CAD-related adverse events.
Collapse
Affiliation(s)
- Florian Wünnemann
- Montreal Heart Institute (F.W., K.S.L., A.L.-A., D.B., M.-P.D., J.-C.T., G.L.), Université de Montréal, Québec, Canada
| | - Ken Sin Lo
- Montreal Heart Institute (F.W., K.S.L., A.L.-A., D.B., M.-P.D., J.-C.T., G.L.), Université de Montréal, Québec, Canada
| | - Alexandra Langford-Avelar
- Montreal Heart Institute (F.W., K.S.L., A.L.-A., D.B., M.-P.D., J.-C.T., G.L.), Université de Montréal, Québec, Canada
| | - David Busseuil
- Montreal Heart Institute (F.W., K.S.L., A.L.-A., D.B., M.-P.D., J.-C.T., G.L.), Université de Montréal, Québec, Canada
| | - Marie-Pierre Dubé
- Montreal Heart Institute (F.W., K.S.L., A.L.-A., D.B., M.-P.D., J.-C.T., G.L.), Université de Montréal, Québec, Canada
- Faculté de Médecine (M.-P.D., J-C.T., G.L.), Université de Montréal, Québec, Canada
| | - Jean-Claude Tardif
- Montreal Heart Institute (F.W., K.S.L., A.L.-A., D.B., M.-P.D., J.-C.T., G.L.), Université de Montréal, Québec, Canada
- Faculté de Médecine (M.-P.D., J-C.T., G.L.), Université de Montréal, Québec, Canada
| | - Guillaume Lettre
- Montreal Heart Institute (F.W., K.S.L., A.L.-A., D.B., M.-P.D., J.-C.T., G.L.), Université de Montréal, Québec, Canada
- Faculté de Médecine (M.-P.D., J-C.T., G.L.), Université de Montréal, Québec, Canada
| |
Collapse
|
281
|
Brazel DM, Jiang Y, Hughey JM, Turcot V, Zhan X, Gong J, Batini C, Weissenkampen JD, Liu M, Barnes DR, Bertelsen S, Chou YL, Erzurumluoglu AM, Faul JD, Haessler J, Hammerschlag AR, Hsu C, Kapoor M, Lai D, Le N, de Leeuw CA, Loukola A, Mangino M, Melbourne CA, Pistis G, Qaiser B, Rohde R, Shao Y, Stringham H, Wetherill L, Zhao W, Agrawal A, Bierut L, Chen C, Eaton CB, Goate A, Haiman C, Heath A, Iacono WG, Martin NG, Polderman TJ, Reiner A, Rice J, Schlessinger D, Scholte HS, Smith JA, Tardif JC, Tindle HA, van der Leij AR, Boehnke M, Chang-Claude J, Cucca F, David SP, Foroud T, Howson JMM, Kardia SLR, Kooperberg C, Laakso M, Lettre G, Madden P, McGue M, North K, Posthuma D, Spector T, Stram D, Tobin MD, Weir DR, Kaprio J, Abecasis GR, Liu DJ, Vrieze S. Exome Chip Meta-analysis Fine Maps Causal Variants and Elucidates the Genetic Architecture of Rare Coding Variants in Smoking and Alcohol Use. Biol Psychiatry 2019; 85:946-955. [PMID: 30679032 PMCID: PMC6534468 DOI: 10.1016/j.biopsych.2018.11.024] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 11/05/2018] [Accepted: 11/29/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Smoking and alcohol use have been associated with common genetic variants in multiple loci. Rare variants within these loci hold promise in the identification of biological mechanisms in substance use. Exome arrays and genotype imputation can now efficiently genotype rare nonsynonymous and loss of function variants. Such variants are expected to have deleterious functional consequences and to contribute to disease risk. METHODS We analyzed ∼250,000 rare variants from 16 independent studies genotyped with exome arrays and augmented this dataset with imputed data from the UK Biobank. Associations were tested for five phenotypes: cigarettes per day, pack-years, smoking initiation, age of smoking initiation, and alcoholic drinks per week. We conducted stratified heritability analyses, single-variant tests, and gene-based burden tests of nonsynonymous/loss-of-function coding variants. We performed a novel fine-mapping analysis to winnow the number of putative causal variants within associated loci. RESULTS Meta-analytic sample sizes ranged from 152,348 to 433,216, depending on the phenotype. Rare coding variation explained 1.1% to 2.2% of phenotypic variance, reflecting 11% to 18% of the total single nucleotide polymorphism heritability of these phenotypes. We identified 171 genome-wide associated loci across all phenotypes. Fine mapping identified putative causal variants with double base-pair resolution at 24 of these loci, and between three and 10 variants for 65 loci. Twenty loci contained rare coding variants in the 95% credible intervals. CONCLUSIONS Rare coding variation significantly contributes to the heritability of smoking and alcohol use. Fine-mapping genome-wide association study loci identifies specific variants contributing to the biological etiology of substance use behavior.
Collapse
Affiliation(s)
- David M Brazel
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, Colorado; Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado
| | - Yu Jiang
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania
| | - Jordan M Hughey
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania
| | - Valérie Turcot
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada; Montreal Heart Institute, Montreal, Quebec, Canada
| | - Xiaowei Zhan
- Department of Clinical Science, Center for Genetics of Host Defense, University of Texas Southwestern, Dallas, Texas
| | - Jian Gong
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Chiara Batini
- Department of Health Sciences, University of Leicester, Leicester, United Kingdom
| | - J Dylan Weissenkampen
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania
| | - MengZhen Liu
- Department of Psychology, University of Minnesota, Minneapolis, Minnesota
| | - Daniel R Barnes
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Sarah Bertelsen
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yi-Ling Chou
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri
| | | | - Jessica D Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, Michigan
| | - Jeff Haessler
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Anke R Hammerschlag
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, University of Amsterdam, Amsterdam, the Netherlands
| | - Chris Hsu
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Manav Kapoor
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Dongbing Lai
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Nhung Le
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, Illinois
| | - Christiaan A de Leeuw
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, University of Amsterdam, Amsterdam, the Netherlands
| | - Anu Loukola
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland; Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom; National Institute for Health Research Biomedical Research Centre at Guy's and St Thomas' Foundation Trust, London, United Kingdom
| | - Carl A Melbourne
- Department of Health Sciences, University of Leicester, Leicester, United Kingdom
| | - Giorgio Pistis
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, Italy
| | - Beenish Qaiser
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland; Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Rebecca Rohde
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Yaming Shao
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Heather Stringham
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | - Leah Wetherill
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Wei Zhao
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan
| | - Arpana Agrawal
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri
| | - Laura Bierut
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri
| | - Chu Chen
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Epidemiology, Head and Neck Surgery Center, University of Washington, Seattle, Washington; Department of Otolaryngology, Head and Neck Surgery Center, University of Washington, Seattle, Washington
| | - Charles B Eaton
- Department of Family Medicine, Brown University, Providence, Rhode Island
| | - Alison Goate
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Christopher Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Andrew Heath
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri
| | - William G Iacono
- Department of Psychology, University of Minnesota, Minneapolis, Minnesota
| | | | - Tinca J Polderman
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, University of Amsterdam, Amsterdam, the Netherlands
| | - Alex Reiner
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Epidemiology, Head and Neck Surgery Center, University of Washington, Seattle, Washington
| | - John Rice
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri; Department of Mathematics, Washington University in St. Louis, St. Louis, Missouri
| | - David Schlessinger
- National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - H Steven Scholte
- Department of Psychology, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Brain and Cognition, University of Amsterdam, Amsterdam, the Netherlands
| | - Jennifer A Smith
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan
| | - Jean-Claude Tardif
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada; Montreal Heart Institute, Montreal, Quebec, Canada
| | - Hilary A Tindle
- Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Andries R van der Leij
- Department of Psychology, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Brain and Cognition, University of Amsterdam, Amsterdam, the Netherlands
| | - Michael Boehnke
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, Italy
| | - Sean P David
- Department of Medicine, Stanford University, Stanford, California
| | - Tatiana Foroud
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Joanna M M Howson
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Sharon L R Kardia
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan
| | - Charles Kooperberg
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Markku Laakso
- Department of Internal Medicine, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Guillaume Lettre
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada; Montreal Heart Institute, Montreal, Quebec, Canada
| | - Pamela Madden
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri
| | - Matt McGue
- Department of Psychology, University of Minnesota, Minneapolis, Minnesota
| | - Kari North
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Danielle Posthuma
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, University of Amsterdam, Amsterdam, the Netherlands; Department of Clinical Genetics, VU University Medical Centre, University of Amsterdam, Amsterdam, the Netherlands
| | - Timothy Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Daniel Stram
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Martin D Tobin
- Department of Health Sciences, University of Leicester, Leicester, United Kingdom
| | - David R Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, Michigan
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland; Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Gonçalo R Abecasis
- Regeneron Pharmaceuticals, Tarrytown, New York; Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | - Dajiang J Liu
- Institute of Personalized Medicine, Penn State College of Medicine, Hershey, Pennsylvania.
| | - Scott Vrieze
- Department of Psychology, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
282
|
Bonhomme M, Fariello MI, Navier H, Hajri A, Badis Y, Miteul H, Samac DA, Dumas B, Baranger A, Jacquet C, Pilet-Nayel ML. A local score approach improves GWAS resolution and detects minor QTL: application to Medicago truncatula quantitative disease resistance to multiple Aphanomyces euteiches isolates. Heredity (Edinb) 2019; 123:517-531. [PMID: 31138867 DOI: 10.1038/s41437-019-0235-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 04/19/2019] [Accepted: 05/08/2019] [Indexed: 12/31/2022] Open
Abstract
Quantitative trait loci (QTL) with small effects, which are pervasive in quantitative phenotypic variation, are difficult to detect in genome-wide association studies (GWAS). To improve their detection, we propose to use a local score approach that accounts for the surrounding signal due to linkage disequilibrium, by accumulating association signals from contiguous single markers. Simulations revealed that, in a GWAS context with high marker density, the local score approach outperforms single SNP p-value-based tests for detecting minor QTL (heritability of 5-10%) and is competitive with regard to alternative methods, which also aggregate p-values. Using more than five million SNPs, this approach was applied to identify loci involved in Quantitative Disease Resistance (QDR) to different isolates of the plant root rot pathogen Aphanomyces euteiches, from a GWAS performed on a collection of 174 accessions of the model legume Medicago truncatula. We refined the position of a previously reported major locus, underlying MYB/NB-ARC/tyrosine kinase candidate genes conferring resistance to two closely related A. euteiches isolates belonging to pea pathotype I. We also discovered a diversity of minor resistance QTL, not detected using p-value-based tests, some of which being putatively shared in response to pea (pathotype I and III) and/or alfalfa (race 1 and 2) isolates. Candidate genes underlying these QTL suggest pathogen effector recognition and plant proteasome as key functions associated with M. truncatula resistance to A. euteiches. GWAS on any organism can benefit from the local score approach to uncover many weak-effect QTL.
Collapse
Affiliation(s)
- Maxime Bonhomme
- Laboratoire de Recherche en Sciences Végétales, Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier (UPS), Castanet Tolosan, France.
| | - Maria Inés Fariello
- Universidad de la República, UdelaR, Facultad de Ingeniería, IMERL, Montevideo, Uruguay
| | - Hélène Navier
- IGEPP, INRA, Agrocampus Ouest, Université de Rennes 1, F-35650, Le Rheu, France
| | - Ahmed Hajri
- IGEPP, INRA, Agrocampus Ouest, Université de Rennes 1, F-35650, Le Rheu, France
| | - Yacine Badis
- Laboratoire de Recherche en Sciences Végétales, Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier (UPS), Castanet Tolosan, France
| | - Henri Miteul
- IGEPP, INRA, Agrocampus Ouest, Université de Rennes 1, F-35650, Le Rheu, France
| | | | - Bernard Dumas
- Laboratoire de Recherche en Sciences Végétales, Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier (UPS), Castanet Tolosan, France
| | - Alain Baranger
- IGEPP, INRA, Agrocampus Ouest, Université de Rennes 1, F-35650, Le Rheu, France
| | - Christophe Jacquet
- Laboratoire de Recherche en Sciences Végétales, Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier (UPS), Castanet Tolosan, France
| | | |
Collapse
|
283
|
Jiang SH, Athanasopoulos V, Ellyard JI, Chuah A, Cappello J, Cook A, Prabhu SB, Cardenas J, Gu J, Stanley M, Roco JA, Papa I, Yabas M, Walters GD, Burgio G, McKeon K, Byers JM, Burrin C, Enders A, Miosge LA, Canete PF, Jelusic M, Tasic V, Lungu AC, Alexander SI, Kitching AR, Fulcher DA, Shen N, Arsov T, Gatenby PA, Babon JJ, Mallon DF, de Lucas Collantes C, Stone EA, Wu P, Field MA, Andrews TD, Cho E, Pascual V, Cook MC, Vinuesa CG. Functional rare and low frequency variants in BLK and BANK1 contribute to human lupus. Nat Commun 2019; 10:2201. [PMID: 31101814 PMCID: PMC6525203 DOI: 10.1038/s41467-019-10242-9] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 04/25/2019] [Indexed: 11/21/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is the prototypic systemic autoimmune disease. It is thought that many common variant gene loci of weak effect act additively to predispose to common autoimmune diseases, while the contribution of rare variants remains unclear. Here we describe that rare coding variants in lupus-risk genes are present in most SLE patients and healthy controls. We demonstrate the functional consequences of rare and low frequency missense variants in the interacting proteins BLK and BANK1, which are present alone, or in combination, in a substantial proportion of lupus patients. The rare variants found in patients, but not those found exclusively in controls, impair suppression of IRF5 and type-I IFN in human B cell lines and increase pathogenic lymphocytes in lupus-prone mice. Thus, rare gene variants are common in SLE and likely contribute to genetic risk. Function-altering variants of immune-related genes cause rare autoimmune syndromes, whereas their contribution to common autoimmune diseases remains uncharacterized. Here the authors show that rare variants of lupus-associated genes are present in the majority of lupus patients and healthy controls, but only the variants found in lupus patients alter gene function.
Collapse
Affiliation(s)
- Simon H Jiang
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Acton, 2601, ACT, Australia. .,Centre for Personalised Immunology, NHMRC Centre for Research Excellence, Acton, 2601, Australia. .,Department of Renal Medicine, The Canberra Hospital, Garran, 2601, ACT, Australia.
| | - Vicki Athanasopoulos
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Acton, 2601, ACT, Australia.,Centre for Personalised Immunology, NHMRC Centre for Research Excellence, Acton, 2601, Australia
| | - Julia I Ellyard
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Acton, 2601, ACT, Australia.,Centre for Personalised Immunology, NHMRC Centre for Research Excellence, Acton, 2601, Australia
| | - Aaron Chuah
- Centre for Personalised Immunology, NHMRC Centre for Research Excellence, Acton, 2601, Australia.,Genome Informatics Laboratory, John Curtin School of Medical Research, Acton, 2601, ACT, Australia
| | - Jean Cappello
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Acton, 2601, ACT, Australia.,Centre for Personalised Immunology, NHMRC Centre for Research Excellence, Acton, 2601, Australia
| | - Amelia Cook
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Acton, 2601, ACT, Australia.,Centre for Personalised Immunology, NHMRC Centre for Research Excellence, Acton, 2601, Australia
| | - Savit B Prabhu
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Acton, 2601, ACT, Australia.,Centre for Personalised Immunology, NHMRC Centre for Research Excellence, Acton, 2601, Australia.,Paediatric Biology Center, Translational Health Science and Technology Institute, Faridabad, 121001, Haryana, India
| | | | - Jinghua Gu
- Baylor Medical Institute, Houston, 77030, Texas, USA
| | - Maurice Stanley
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Acton, 2601, ACT, Australia.,Centre for Personalised Immunology, NHMRC Centre for Research Excellence, Acton, 2601, Australia
| | - Jonathan A Roco
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Acton, 2601, ACT, Australia.,Centre for Personalised Immunology, NHMRC Centre for Research Excellence, Acton, 2601, Australia
| | - Ilenia Papa
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Acton, 2601, ACT, Australia
| | - Mehmet Yabas
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Acton, 2601, ACT, Australia.,Department of Genetics and Bioengineering, Trakya University, Edirne, 22030, Turkey
| | - Giles D Walters
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Acton, 2601, ACT, Australia.,Centre for Personalised Immunology, NHMRC Centre for Research Excellence, Acton, 2601, Australia.,Department of Renal Medicine, The Canberra Hospital, Garran, 2601, ACT, Australia
| | - Gaetan Burgio
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Acton, 2601, ACT, Australia
| | - Kathryn McKeon
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Acton, 2601, ACT, Australia.,Centre for Personalised Immunology, NHMRC Centre for Research Excellence, Acton, 2601, Australia
| | - James M Byers
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Acton, 2601, ACT, Australia.,Centre for Personalised Immunology, NHMRC Centre for Research Excellence, Acton, 2601, Australia
| | - Charlotte Burrin
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Acton, 2601, ACT, Australia
| | - Anselm Enders
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Acton, 2601, ACT, Australia.,Centre for Personalised Immunology, NHMRC Centre for Research Excellence, Acton, 2601, Australia
| | - Lisa A Miosge
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Acton, 2601, ACT, Australia
| | - Pablo F Canete
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Acton, 2601, ACT, Australia.,Centre for Personalised Immunology, NHMRC Centre for Research Excellence, Acton, 2601, Australia
| | - Marija Jelusic
- Department of Paediatric Rheumatology and Immunology, University of Zagreb School of Medicine, Zagreb, 10000, Croatia
| | - Velibor Tasic
- University Children's Hospital, Medical School, Skopje, 1000, Macedonia
| | - Adrian C Lungu
- Department of Pediatric Nephrology, Fundeni Clinical Institute, Bucharest, 022328, Romania
| | - Stephen I Alexander
- Centre for Personalised Immunology, NHMRC Centre for Research Excellence, Acton, 2601, Australia.,Westmead Children's Hospital, Westmead, 2145, NSW, Australia
| | - Arthur R Kitching
- Centre for Personalised Immunology, NHMRC Centre for Research Excellence, Acton, 2601, Australia.,Centre for Inflammatory Diseases, Department of Medicine, Monash University, Clayton, 3168, VIC, Australia
| | - David A Fulcher
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Acton, 2601, ACT, Australia.,Centre for Personalised Immunology, NHMRC Centre for Research Excellence, Acton, 2601, Australia.,Department of Immunology, The Canberra Hospital, Garran, 2601, ACT, Australia
| | - Nan Shen
- China Australia Centre for Personalised Immunology, Renji Hospital Shanghai, JiaoTong University Shanghai, Huangpu Qu, 200333, China
| | - Todor Arsov
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Acton, 2601, ACT, Australia.,Centre for Personalised Immunology, NHMRC Centre for Research Excellence, Acton, 2601, Australia.,China Australia Centre for Personalised Immunology, Renji Hospital Shanghai, JiaoTong University Shanghai, Huangpu Qu, 200333, China
| | - Paul A Gatenby
- Department of Immunology, The Canberra Hospital, Garran, 2601, ACT, Australia
| | - Jeff J Babon
- Walter and Eliza Hall Institute, Parkville, 3052, VIC, Australia
| | - Dominic F Mallon
- Immunology PathWest Fiona Stanley Hospital, Murdoch, 6150, WA, Australia
| | | | - Eric A Stone
- Research School of Biology and Research School of Finance, Actuarial Studies and Statistics, Acton, 2601, ACT, Australia
| | - Philip Wu
- Centre for Personalised Immunology, NHMRC Centre for Research Excellence, Acton, 2601, Australia.,Australian Phenomics Facility, ANU, Acton, 2601, ACT, Australia
| | - Matthew A Field
- Centre for Personalised Immunology, NHMRC Centre for Research Excellence, Acton, 2601, Australia.,Genome Informatics Laboratory, John Curtin School of Medical Research, Acton, 2601, ACT, Australia
| | - Thomas D Andrews
- Centre for Personalised Immunology, NHMRC Centre for Research Excellence, Acton, 2601, Australia.,Genome Informatics Laboratory, John Curtin School of Medical Research, Acton, 2601, ACT, Australia.,National Computational Infrastructure, ANU, Acton, 2601, ACT, Australia
| | - Eun Cho
- Centre for Personalised Immunology, NHMRC Centre for Research Excellence, Acton, 2601, Australia.,Genome Informatics Laboratory, John Curtin School of Medical Research, Acton, 2601, ACT, Australia
| | | | - Matthew C Cook
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Acton, 2601, ACT, Australia.,Centre for Personalised Immunology, NHMRC Centre for Research Excellence, Acton, 2601, Australia.,Department of Immunology, The Canberra Hospital, Garran, 2601, ACT, Australia.,China Australia Centre for Personalised Immunology, Renji Hospital Shanghai, JiaoTong University Shanghai, Huangpu Qu, 200333, China
| | - Carola G Vinuesa
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Acton, 2601, ACT, Australia. .,Centre for Personalised Immunology, NHMRC Centre for Research Excellence, Acton, 2601, Australia. .,China Australia Centre for Personalised Immunology, Renji Hospital Shanghai, JiaoTong University Shanghai, Huangpu Qu, 200333, China.
| |
Collapse
|
284
|
Cai Z, Guldbrandtsen B, Lund MS, Sahana G. Weighting sequence variants based on their annotation increases the power of genome-wide association studies in dairy cattle. Genet Sel Evol 2019; 51:20. [PMID: 31077144 PMCID: PMC6511139 DOI: 10.1186/s12711-019-0463-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 05/03/2019] [Indexed: 01/09/2023] Open
Abstract
Background Genome-wide association studies (GWAS) are widely used to identify regions of the genome that harbor genetic determinants of quantitative traits. However, the multiple-testing burden from scanning tens of millions of whole-genome sequence variants reduces the power to identify associated variants, especially if sample size is limited. In addition, factors such as inaccuracy of imputation, complex linkage disequilibrium structures, and multiple closely-located causal variants may result in an identified causative mutation not being the most significant single nucleotide polymorphism in a particular genomic region. Therefore, the use of information from different sources, particularly variant annotations, was proposed to enhance the fine-mapping of causal variants. Here, we tested whether applying significance thresholds based on variant annotation categories increases the power of GWAS compared with a flat Bonferroni multiple-testing correction. Results Whole-genome sequence variants in dairy cattle were categorized according to type and predicted impact. Then, GWAS between markers and 17 quantitative traits were analyzed for enrichment for association of each annotation category. By using annotation categories that were determined with the variants effect predictor software and datasets indicating regions of open chromatin, “low impact” variants were found to be highly enriched. Moreover, when the variants annotated as “modifier” and not located at open chromatin regions were further classified into different types of potential regulatory elements, the high impact variants, moderate impact variants, variants located in the 3′ and 5′ untranslated regions, and variants located in potential non-coding RNA regions exhibited relatively more enrichment. In contrast, a similar study on human GWAS data reported that enrichment of association signals was highest with high impact variants. We observed an increase in power when these variant category-based significance thresholds were applied for GWAS results on stature in Nordic Holstein cattle, as more candidate genes from previous large GWAS meta-analysis for cattle stature were confirmed. Conclusions Use of variant category-based genome-wide significance thresholds can marginally increase the power to detect the candidate genes in cattle. With the continued improvements in annotation of the bovine genome, we anticipate that the growing usefulness of variant category-based significance thresholds will be demonstrated. Electronic supplementary material The online version of this article (10.1186/s12711-019-0463-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zexi Cai
- Department of Molecular Biology and Genetics, Center for Quantitative Genetics and Genomics, Aarhus University, 8830, Tjele, Denmark.
| | - Bernt Guldbrandtsen
- Department of Molecular Biology and Genetics, Center for Quantitative Genetics and Genomics, Aarhus University, 8830, Tjele, Denmark
| | - Mogens Sandø Lund
- Department of Molecular Biology and Genetics, Center for Quantitative Genetics and Genomics, Aarhus University, 8830, Tjele, Denmark
| | - Goutam Sahana
- Department of Molecular Biology and Genetics, Center for Quantitative Genetics and Genomics, Aarhus University, 8830, Tjele, Denmark
| |
Collapse
|
285
|
Styrkarsdottir U, Stefansson OA, Gunnarsdottir K, Thorleifsson G, Lund SH, Stefansdottir L, Juliusson K, Agustsdottir AB, Zink F, Halldorsson GH, Ivarsdottir EV, Benonisdottir S, Jonsson H, Gylfason A, Norland K, Trajanoska K, Boer CG, Southam L, Leung JCS, Tang NLS, Kwok TCY, Lee JSW, Ho SC, Byrjalsen I, Center JR, Lee SH, Koh JM, Lohmander LS, Ho-Pham LT, Nguyen TV, Eisman JA, Woo J, Leung PC, Loughlin J, Zeggini E, Christiansen C, Rivadeneira F, van Meurs J, Uitterlinden AG, Mogensen B, Jonsson H, Ingvarsson T, Sigurdsson G, Benediktsson R, Sulem P, Jonsdottir I, Masson G, Holm H, Norddahl GL, Thorsteinsdottir U, Gudbjartsson DF, Stefansson K. GWAS of bone size yields twelve loci that also affect height, BMD, osteoarthritis or fractures. Nat Commun 2019; 10:2054. [PMID: 31053729 PMCID: PMC6499783 DOI: 10.1038/s41467-019-09860-0] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 04/03/2019] [Indexed: 12/12/2022] Open
Abstract
Bone area is one measure of bone size that is easily derived from dual-energy X-ray absorptiometry (DXA) scans. In a GWA study of DXA bone area of the hip and lumbar spine (N ≥ 28,954), we find thirteen independent association signals at twelve loci that replicate in samples of European and East Asian descent (N = 13,608 - 21,277). Eight DXA area loci associate with osteoarthritis, including rs143384 in GDF5 and a missense variant in COL11A1 (rs3753841). The strongest DXA area association is with rs11614913[T] in the microRNA MIR196A2 gene that associates with lumbar spine area (P = 2.3 × 10-42, β = -0.090) and confers risk of hip fracture (P = 1.0 × 10-8, OR = 1.11). We demonstrate that the risk allele is less efficient in repressing miR-196a-5p target genes. We also show that the DXA area measure contributes to the risk of hip fracture independent of bone density.
Collapse
Affiliation(s)
| | | | | | | | - Sigrun H Lund
- deCODE genetics/Amgen Inc., Reykjavik, 101, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| | | | | | | | - Florian Zink
- deCODE genetics/Amgen Inc., Reykjavik, 101, Iceland
| | | | | | | | | | | | | | - Katerina Trajanoska
- Department of Epidemiology, ErasmusMC, 3015 GD, Rotterdam, The Netherlands
- Department of Internal Medicine, ErasmusMC, 3015 GD, Rotterdam, the Netherlands
| | - Cindy G Boer
- Department of Internal Medicine, ErasmusMC, 3015 GD, Rotterdam, the Netherlands
| | - Lorraine Southam
- Wellcome Trust Sanger Institute, Hinxton, CB10 1SA, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Jason C S Leung
- Jockey Club Centre for Osteoporosis Care and Control, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Nelson L S Tang
- Faculty of Medicine, Department of Chemical Pathology and Laboratory for Genetics of Disease Susceptibility, Li Ka Shing Institute of Health Sciences,, The Chinese University of Hong Kong, Hong Kong, China
- CUHK Shenzhen Research Institute, Shenzhen, 518000, China
| | - Timothy C Y Kwok
- Jockey Club Centre for Osteoporosis Care and Control, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Hong Kong, China
| | - Jenny S W Lee
- Faculty of Medicine, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Department of Medicine, Alice Ho Miu Ling Nethersole Hospital and Tai Po Hospital, Hong Kong, China
| | - Suzanne C Ho
- JC School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | | | - Jacqueline R Center
- Bone Biology Division, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
- School of Medicine Sydney, University of Notre Dame Australia, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW, 2010, Australia
| | - Seung Hun Lee
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - Jung-Min Koh
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - L Stefan Lohmander
- Orthopaedics, Department of Clinical Sciences Lund, Lund University, SE-22 100, Lund, Sweden
| | - Lan T Ho-Pham
- Bone and Muscle Research Lab, Ton Duc Thang University, Ho Chi Minh City, 700000, Vietnam
| | - Tuan V Nguyen
- Bone Biology Division, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW, 2010, Australia
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - John A Eisman
- Bone Biology Division, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
- School of Medicine Sydney, University of Notre Dame Australia, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW, 2010, Australia
- Clinical Translation and Advanced Education, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
| | - Jean Woo
- Faculty of Medicine, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Ping-C Leung
- Jockey Club Centre for Osteoporosis Care and Control, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - John Loughlin
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, NE1 7RU, UK
| | - Eleftheria Zeggini
- Wellcome Trust Sanger Institute, Hinxton, CB10 1SA, UK
- Institute of Translational Genomics, Helmholtz Zentrum München, 85764, München, Germany
| | | | - Fernando Rivadeneira
- Department of Epidemiology, ErasmusMC, 3015 GD, Rotterdam, The Netherlands
- Department of Internal Medicine, ErasmusMC, 3015 GD, Rotterdam, the Netherlands
| | - Joyce van Meurs
- Department of Internal Medicine, ErasmusMC, 3015 GD, Rotterdam, the Netherlands
| | | | - Brynjolfur Mogensen
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
- Department of Emergengy Medicine, Landspitali, The National University Hospital of Iceland, 101, Reykjavik, Iceland
- Research Institute in Emergency Medicine, Landspitali, The National University Hospital of Iceland, and University of Iceland, 101, Reykjavik, Iceland
| | - Helgi Jonsson
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
- Department of Medicine, Landspitali-The National University Hospital of Iceland, 101, Reykjavik, Iceland
| | - Thorvaldur Ingvarsson
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
- Department of Orthopedic Surgery, Akureyri Hospital, 600, Akureyri, Iceland
- Institution of Health Science, University of Akureyri, 600, Akureyri, Iceland
| | - Gunnar Sigurdsson
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
- Research Service Center, Reykjavik, 201, Iceland
- Department of Endocrinology and Metabolism, Landspitali, The National University Hospital of Iceland, 101, Reykjavik, Iceland
| | - Rafn Benediktsson
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
- Department of Endocrinology and Metabolism, Landspitali, The National University Hospital of Iceland, 101, Reykjavik, Iceland
| | | | - Ingileif Jonsdottir
- deCODE genetics/Amgen Inc., Reykjavik, 101, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
- Department of Immunology, Landspitali-The National University Hospital of Iceland, 101, Reykjavik, Iceland
| | - Gisli Masson
- deCODE genetics/Amgen Inc., Reykjavik, 101, Iceland
| | - Hilma Holm
- deCODE genetics/Amgen Inc., Reykjavik, 101, Iceland
| | | | - Unnur Thorsteinsdottir
- deCODE genetics/Amgen Inc., Reykjavik, 101, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| | - Daniel F Gudbjartsson
- deCODE genetics/Amgen Inc., Reykjavik, 101, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, 107, Iceland
| | - Kari Stefansson
- deCODE genetics/Amgen Inc., Reykjavik, 101, Iceland.
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland.
| |
Collapse
|
286
|
Tiosano D, Baris HN, Chen A, Hitzert MM, Schueler M, Gulluni F, Wiesener A, Bergua A, Mory A, Copeland B, Gleeson JG, Rump P, van Meer H, Sival DA, Haucke V, Kriwinsky J, Knaup KX, Reis A, Hauer NN, Hirsch E, Roepman R, Pfundt R, Thiel CT, Wiesener MS, Aslanyan MG, Buchner DA. Mutations in PIK3C2A cause syndromic short stature, skeletal abnormalities, and cataracts associated with ciliary dysfunction. PLoS Genet 2019; 15:e1008088. [PMID: 31034465 PMCID: PMC6508738 DOI: 10.1371/journal.pgen.1008088] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 05/09/2019] [Accepted: 03/12/2019] [Indexed: 02/07/2023] Open
Abstract
PIK3C2A is a class II member of the phosphoinositide 3-kinase (PI3K) family that catalyzes the phosphorylation of phosphatidylinositol (PI) into PI(3)P and the phosphorylation of PI(4)P into PI(3,4)P2. At the cellular level, PIK3C2A is critical for the formation of cilia and for receptor mediated endocytosis, among other biological functions. We identified homozygous loss-of-function mutations in PIK3C2A in children from three independent consanguineous families with short stature, coarse facial features, cataracts with secondary glaucoma, multiple skeletal abnormalities, neurological manifestations, among other findings. Cellular studies of patient-derived fibroblasts found that they lacked PIK3C2A protein, had impaired cilia formation and function, and demonstrated reduced proliferative capacity. Collectively, the genetic and molecular data implicate mutations in PIK3C2A in a new Mendelian disorder of PI metabolism, thereby shedding light on the critical role of a class II PI3K in growth, vision, skeletal formation and neurological development. In particular, the considerable phenotypic overlap, yet distinct features, between this syndrome and Lowe’s syndrome, which is caused by mutations in the PI-5-phosphatase OCRL, highlight the key role of PI metabolizing enzymes in specific developmental processes and demonstrate the unique non-redundant functions of each enzyme. This discovery expands what is known about disorders of PI metabolism and helps unravel the role of PIK3C2A and class II PI3Ks in health and disease. Identifying the genetic basis of rare disorders can provide insight into gene function, susceptibility to disease, guide the development of new therapeutics, improve opportunities for genetic counseling, and help clinicians evaluate and potentially treat complicated clinical presentations. However, it is estimated that the genetic basis of approximately one-half of all rare genetic disorders remains unknown. We describe one such rare disorder based on genetic and clinical evaluations of individuals from 3 unrelated consanguineous families with a similar constellation of features including short stature, coarse facial features, cataracts with secondary glaucoma, multiple skeletal abnormalities, neurological manifestations including stroke, among other findings. We discovered that these features were due to deficiency of the PIK3C2A enzyme. PIK3C2A is a class II member of the phosphoinositide 3-kinase (PI3K) family that catalyzes the phosphorylation of the lipids phosphatidylinositol (PI) into PI(3)P and the phosphorylation of PI(4)P into PI(3,4)P2 that are essential for a variety of cellular processes including cilia formation and vesicle trafficking. This syndrome is the first monogenic disorder caused by mutations in a class II PI3K family member and thus sheds new light on their role in human development.
Collapse
Affiliation(s)
- Dov Tiosano
- Division of Pediatric Endocrinology, Ruth Children's Hospital, Rambam Medical Center, Haifa, Israel
- Rappaport Family Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
| | - Hagit N. Baris
- Rappaport Family Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
- The Genetics Institute, Rambam Health Care Campus, Haifa, Israel
| | - Anlu Chen
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Marrit M. Hitzert
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Markus Schueler
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Federico Gulluni
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Torino, Italy
| | - Antje Wiesener
- Institute of Human Genetics, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Antonio Bergua
- Department of Ophthalmology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Adi Mory
- The Genetics Institute, Rambam Health Care Campus, Haifa, Israel
| | - Brett Copeland
- Laboratory of Pediatric Brain Diseases, Rockefeller University, New York, New York, United States of America
| | - Joseph G. Gleeson
- Laboratory of Pediatric Brain Diseases, Rockefeller University, New York, New York, United States of America
- Department of Neurosciences, University of California, San Diego, La Jolla, California, United States of America
| | - Patrick Rump
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hester van Meer
- Department of Pediatrics, Beatrix Children’s Hospital, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Deborah A. Sival
- Department of Pediatrics, Beatrix Children’s Hospital, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Volker Haucke
- Leibniz-Institut für Molekulare Pharmakologie, Berlin Faculty of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Josh Kriwinsky
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Karl X. Knaup
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - André Reis
- Institute of Human Genetics, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Nadine N. Hauer
- Institute of Human Genetics, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Torino, Italy
| | - Ronald Roepman
- Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rolph Pfundt
- Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Christian T. Thiel
- Institute of Human Genetics, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Michael S. Wiesener
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Mariam G. Aslanyan
- Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - David A. Buchner
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio, United States of America
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
- Research Institute for Children’s Health, Case Western Reserve University, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
287
|
Njølstad PR, Andreassen OA, Brunak S, Børglum AD, Dillner J, Esko T, Franks PW, Freimer N, Groop L, Heimer H, Hougaard DM, Hovig E, Hveem K, Jalanko A, Kaprio J, Knudsen GP, Melbye M, Metspalu A, Mortensen PB, Palmgren J, Palotie A, Reed W, Stefánsson H, Stitziel NO, Sullivan PF, Thorsteinsdóttir U, Vaudel M, Vuorio E, Werge T, Stoltenberg C, Stefánsson K. Roadmap for a precision-medicine initiative in the Nordic region. Nat Genet 2019; 51:924-930. [DOI: 10.1038/s41588-019-0391-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
288
|
Predicting adult height from DNA variants in a European-Asian admixed population. Int J Legal Med 2019; 133:1667-1679. [PMID: 30976986 DOI: 10.1007/s00414-019-02039-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 03/05/2019] [Indexed: 01/12/2023]
Abstract
Accurate genomic profiling for adult height is of high practical relevance in forensics genetics. Adult height is a classical reference trait in the field of human complex trait genetics characterized by highly polygenic nature and relatively high heritability. A meta-analysis of genome-wide association studies by the Genetic Investigation of Anthropocentric Traits (GIANT) consortium has identified 697 DNA variants associated with adult height in Europeans; however, whether these variants will still be informative in non-Europeans is still in question. The present study investigated the predictive power of these 697 height-associated SNPs in 687 Uyghurs of European-Asian admixed origin. Among all GIANT SNPs, 11% showed nominally significant association (6.78 × 10-4 < p < 0.05) with adult height in the Uyghur population and among the significant SNPs 77% of allele effects were in the same direction as those in Europeans reported in the GIANT study. Fitting linear and logistic models using a polygenic score consisting of all GIANT SNPs resulted in an 80-20 cross-validated mean R2 of 10.08% (95% CI 3.16-18.40%) for quantitative height prediction and a mean AUC value of 0.65 (95% CI 0.57-0.72%) for qualitative "above average" prediction. Fine-tuning the SNP set using their association p values considerably improved the prediction results (number of SNPs = 62, R2 = 15.59%, 95% CI 6.80-25.71%; AUC = 0.70, 95% CI 62-0.77) in the Uyghurs. Overall, our findings demonstrate substantial differences between the European and Asian populations in the genetics of adult height, emphasizing the importance of population heterogeneity underlying the genetic architecture of adult height.
Collapse
|
289
|
Zhao J, Sauvage C, Zhao J, Bitton F, Bauchet G, Liu D, Huang S, Tieman DM, Klee HJ, Causse M. Meta-analysis of genome-wide association studies provides insights into genetic control of tomato flavor. Nat Commun 2019; 10:1534. [PMID: 30948717 PMCID: PMC6449550 DOI: 10.1038/s41467-019-09462-w] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 03/04/2019] [Indexed: 12/30/2022] Open
Abstract
Tomato flavor has changed over the course of long-term domestication and intensive breeding. To understand the genetic control of flavor, we report the meta-analysis of genome-wide association studies (GWAS) using 775 tomato accessions and 2,316,117 SNPs from three GWAS panels. We discover 305 significant associations for the contents of sugars, acids, amino acids, and flavor-related volatiles. We demonstrate that fruit citrate and malate contents have been impacted by selection during domestication and improvement, while sugar content has undergone less stringent selection. We suggest that it may be possible to significantly increase volatiles that positively contribute to consumer preferences while reducing unpleasant volatiles, by selection of the relevant allele combinations. Our results provide genetic insights into the influence of human selection on tomato flavor and demonstrate the benefits obtained from meta-analysis. Flavor is one of the most important traits for improving tomato sensory quality and consumer acceptability. Here, the authors report meta-analysis of genome-wide association studies of flavor related traits and show genetic insights into the influence of human selection during domestication and improvement.
Collapse
Affiliation(s)
- Jiantao Zhao
- INRA, UR1052, Génétique et Amélioration des Fruits et Légumes, Domaine Saint Maurice, 67 Allée des Chênes CS 60094, 84143, Montfavet Cedex, France
| | - Christopher Sauvage
- INRA, UR1052, Génétique et Amélioration des Fruits et Légumes, Domaine Saint Maurice, 67 Allée des Chênes CS 60094, 84143, Montfavet Cedex, France.,Syngenta, 12 Chemin de l'Hobit, Saint Sauveur, 31790, France
| | - Jinghua Zhao
- MRC Epidemiology Unit & Institute of Metabolic Science, University of Cambridge, Addrenbrooke's Hospital, Box 285, Hills Road, Cambridge, CB2 0QQ, UK.,Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, Strangeways Research Laboratory, University of Cambridge, Wort's Causeway, Cambridge, CB1 8RN, UK
| | - Frédérique Bitton
- INRA, UR1052, Génétique et Amélioration des Fruits et Légumes, Domaine Saint Maurice, 67 Allée des Chênes CS 60094, 84143, Montfavet Cedex, France
| | - Guillaume Bauchet
- INRA, UR1052, Génétique et Amélioration des Fruits et Légumes, Domaine Saint Maurice, 67 Allée des Chênes CS 60094, 84143, Montfavet Cedex, France.,Boyce Thompson Institute, Cornell University, 533 Tower Rd, Ithaca, NY, 14853, USA
| | - Dan Liu
- Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, 518124, Shenzhen, Guangdong, China
| | - Sanwen Huang
- Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, 518124, Shenzhen, Guangdong, China.,Key Laboratory of Biology and Genetic Improvement of Horticultural Crops of the Ministry of Agriculture, Sino-Dutch Joint Laboratory of Horticultural Genomics, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, 100081, Beijing, China
| | - Denise M Tieman
- Horticultural Sciences, Plant Innovation Center, University of Florida, Post Office Box 110690, Gainesville, FL, 32611, USA
| | - Harry J Klee
- Horticultural Sciences, Plant Innovation Center, University of Florida, Post Office Box 110690, Gainesville, FL, 32611, USA
| | - Mathilde Causse
- INRA, UR1052, Génétique et Amélioration des Fruits et Légumes, Domaine Saint Maurice, 67 Allée des Chênes CS 60094, 84143, Montfavet Cedex, France.
| |
Collapse
|
290
|
Petty LE, Highland HM, Gamazon ER, Hu H, Karhade M, Chen HH, de Vries PS, Grove ML, Aguilar D, Bell GI, Huff CD, Hanis CL, Doddapaneni H, Munzy DM, Gibbs RA, Ma J, Parra EJ, Cruz M, Valladares-Salgado A, Arking DE, Barbeira A, Im HK, Morrison AC, Boerwinkle E, Below JE. Functionally oriented analysis of cardiometabolic traits in a trans-ethnic sample. Hum Mol Genet 2019; 28:1212-1224. [PMID: 30624610 PMCID: PMC6423424 DOI: 10.1093/hmg/ddy435] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 11/13/2018] [Accepted: 11/20/2018] [Indexed: 01/02/2023] Open
Abstract
Interpretation of genetic association results is difficult because signals often lack biological context. To generate hypotheses of the functional genetic etiology of complex cardiometabolic traits, we estimated the genetically determined component of gene expression from common variants using PrediXcan (1) and determined genes with differential predicted expression by trait. PrediXcan imputes tissue-specific expression levels from genetic variation using variant-level effect on gene expression in transcriptome data. To explore the value of imputed genetically regulated gene expression (GReX) models across different ancestral populations, we evaluated imputed expression levels for predictive accuracy genome-wide in RNA sequence data in samples drawn from European-ancestry and African-ancestry populations and identified substantial predictive power using European-derived models in a non-European target population. We then tested the association of GReX on 15 cardiometabolic traits including blood lipid levels, body mass index, height, blood pressure, fasting glucose and insulin, RR interval, fibrinogen level, factor VII level and white blood cell and platelet counts in 15 755 individuals across three ancestry groups, resulting in 20 novel gene-phenotype associations reaching experiment-wide significance across ancestries. In addition, we identified 18 significant novel gene-phenotype associations in our ancestry-specific analyses. Top associations were assessed for additional support via query of S-PrediXcan (2) results derived from publicly available genome-wide association studies summary data. Collectively, these findings illustrate the utility of transcriptome-based imputation models for discovery of cardiometabolic effect genes in a diverse dataset.
Collapse
Affiliation(s)
- Lauren E Petty
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Heather M Highland
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Eric R Gamazon
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Clare Hall, University of Cambridge, Cambridge, UK
| | - Hao Hu
- Department of Epidemiology, MD Anderson Cancer Center, Houston, TX, USA
| | - Mandar Karhade
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Hung-Hsin Chen
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Paul S de Vries
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Megan L Grove
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - David Aguilar
- Department of Cardiology, Baylor College of Medicine Houston, TX, USA
| | - Graeme I Bell
- Departments of Medicine and Human Genetics, The University of Chicago, Chicago, IL, USA
| | - Chad D Huff
- Department of Epidemiology, MD Anderson Cancer Center, Houston, TX, USA
| | - Craig L Hanis
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | | | - Donna M Munzy
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Richard A Gibbs
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Jianzhong Ma
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Esteban J Parra
- Department of Anthropology, University of Toronto at Mississauga, Mississauga, Ontario, Canada
| | - Miguel Cruz
- Unidad de Investigación Médica en Bioquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, IMSS, Mexico City, Mexico
| | - Adan Valladares-Salgado
- Unidad de Investigación Médica en Bioquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, IMSS, Mexico City, Mexico
| | - Dan E Arking
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alvaro Barbeira
- Section of Genetic Medicine, Department of Medicine, University of Chicago, IL, USA
| | - Hae Kyung Im
- Section of Genetic Medicine, Department of Medicine, University of Chicago, IL, USA
| | - Alanna C Morrison
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Eric Boerwinkle
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jennifer E Below
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
291
|
Rediscovering the value of families for psychiatric genetics research. Mol Psychiatry 2019; 24:523-535. [PMID: 29955165 PMCID: PMC7028329 DOI: 10.1038/s41380-018-0073-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 01/11/2018] [Accepted: 03/26/2018] [Indexed: 01/09/2023]
Abstract
As it is likely that both common and rare genetic variation are important for complex disease risk, studies that examine the full range of the allelic frequency distribution should be utilized to dissect the genetic influences on mental illness. The rate limiting factor for inferring an association between a variant and a phenotype is inevitably the total number of copies of the minor allele captured in the studied sample. For rare variation, with minor allele frequencies of 0.5% or less, very large samples of unrelated individuals are necessary to unambiguously associate a locus with an illness. Unfortunately, such large samples are often cost prohibitive. However, by using alternative analytic strategies and studying related individuals, particularly those from large multiplex families, it is possible to reduce the required sample size while maintaining statistical power. We contend that using whole genome sequence (WGS) in extended pedigrees provides a cost-effective strategy for psychiatric gene mapping that complements common variant approaches and WGS in unrelated individuals. This was our impetus for forming the "Pedigree-Based Whole Genome Sequencing of Affective and Psychotic Disorders" consortium. In this review, we provide a rationale for the use of WGS with pedigrees in modern psychiatric genetics research. We begin with a focused review of the current literature, followed by a short history of family-based research in psychiatry. Next, we describe several advantages of pedigrees for WGS research, including power estimates, methods for studying the environment, and endophenotypes. We conclude with a brief description of our consortium and its goals.
Collapse
|
292
|
Murakami M, Kamimura D, Hirano T. Pleiotropy and Specificity: Insights from the Interleukin 6 Family of Cytokines. Immunity 2019; 50:812-831. [DOI: 10.1016/j.immuni.2019.03.027] [Citation(s) in RCA: 231] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 02/08/2023]
|
293
|
Marouli E, Del Greco MF, Astley CM, Yang J, Ahmad S, Berndt SI, Caulfield MJ, Evangelou E, McKnight B, Medina-Gomez C, van Vliet-Ostaptchouk JV, Warren HR, Zhu Z, Hirschhorn JN, Loos RJF, Kutalik Z, Deloukas P. Mendelian randomisation analyses find pulmonary factors mediate the effect of height on coronary artery disease. Commun Biol 2019; 2:119. [PMID: 30937401 PMCID: PMC6437163 DOI: 10.1038/s42003-019-0361-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 02/22/2019] [Indexed: 01/06/2023] Open
Abstract
There is evidence that lower height is associated with a higher risk of coronary artery disease (CAD) and increased risk of type 2 diabetes (T2D). It is not clear though whether these associations are causal, direct or mediated by other factors. Here we show that one standard deviation higher genetically determined height (~6.5 cm) is causally associated with a 16% decrease in CAD risk (OR = 0.84, 95% CI 0.80-0.87). This causal association remains after performing sensitivity analyses relaxing pleiotropy assumptions. The causal effect of height on CAD risk is reduced by 1-3% after adjustment for potential mediators (lipids, blood pressure, glycaemic traits, body mass index, socio-economic status). In contrast, our data suggest that lung function (measured by forced expiratory volume [FEV1] and forced vital capacity [FVC]) is a mediator of the effect of height on CAD. We observe no direct causal effect of height on the risk of T2D.
Collapse
Affiliation(s)
- Eirini Marouli
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ UK
- Centre for Genomic Health, Life Sciences, Queen Mary University of London, London, EC1M 6BQ UK
| | - M. Fabiola Del Greco
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lubeck, Bolzano, 39100 Italy
| | - Christina M. Astley
- Boston Children’s Hospital, Boston, MA 02115 USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142 USA
| | - Jian Yang
- Institute for Molecular Bioscience, University of Queensland, Brisbane, 4072 QLD Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, 4072 QLD Australia
| | - Shafqat Ahmad
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA 02115 USA
- Division of Preventive Medicine, Harvard Medical School, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02215 USA
- Department of Medical Sciences, Molecular Epidemiology, Uppsala University, Uppsala, 751 41 Sweden
| | - Sonja I. Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892 USA
| | - Mark J. Caulfield
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ UK
- National Institute for Health Research, Barts Cardiovascular Biomedical Research Center, Queen Mary University of London, London, EC1M 6BQ UK
| | - Evangelos Evangelou
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, W2 1PG UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, 45110 Greece
| | - Barbara McKnight
- Department of Biostatistics, University of Washington, Seattle, WA 98101 USA
| | - Carolina Medina-Gomez
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, 3015 GE The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, 3015 GE The Netherlands
| | - Jana V. van Vliet-Ostaptchouk
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, 9713 GZ The Netherlands
| | - Helen R. Warren
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ UK
- National Institute for Health Research, Barts Cardiovascular Biomedical Research Center, Queen Mary University of London, London, EC1M 6BQ UK
| | - Zhihong Zhu
- Institute for Molecular Bioscience, University of Queensland, Brisbane, 4072 QLD Australia
| | - Joel N. Hirschhorn
- Boston Children’s Hospital, Boston, MA 02115 USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142 USA
| | - Ruth J. F. Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Zoltan Kutalik
- Institute of Social and Preventive Medicine, Lausanne University Hospital, Lausanne, 1010 Switzerland
- Swiss Institute of Bioinformatics, Lausanne, 1015 Switzerland
| | - Panos Deloukas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ UK
- Centre for Genomic Health, Life Sciences, Queen Mary University of London, London, EC1M 6BQ UK
- Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, 21589 Saudi Arabia
| |
Collapse
|
294
|
Sullivan PF, Geschwind DH. Defining the Genetic, Genomic, Cellular, and Diagnostic Architectures of Psychiatric Disorders. Cell 2019; 177:162-183. [PMID: 30901538 PMCID: PMC6432948 DOI: 10.1016/j.cell.2019.01.015] [Citation(s) in RCA: 281] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/03/2019] [Accepted: 01/04/2019] [Indexed: 01/01/2023]
Abstract
Studies of the genetics of psychiatric disorders have become one of the most exciting and fast-moving areas in human genetics. A decade ago, there were few reproducible findings, and now there are hundreds. In this review, we focus on the findings that have illuminated the genetic architecture of psychiatric disorders and the challenges of using these findings to inform our understanding of pathophysiology. The evidence is now overwhelming that psychiatric disorders are "polygenic"-that many genetic loci contribute to risk. With the exception of a subset of those with ASD, few individuals with a psychiatric disorder have a single, deterministic genetic cause; rather, developing a psychiatric disorder is influenced by hundreds of different genetic variants, consistent with a polygenic model. As progressively larger studies have uncovered more about their genetic architecture, the need to elucidate additional architectures has become clear. Even if we were to have complete knowledge of the genetic architecture of a psychiatric disorder, full understanding requires deep knowledge of the functional genomic architecture-the implicated loci impact regulatory processes that influence gene expression and the functional coordination of genes that control biological processes. Following from this is cellular architecture: of all brain regions, cell types, and developmental stages, where and when are the functional architectures operative? Given that the genetic architectures of different psychiatric disorders often strongly overlap, we are challenged to re-evaluate and refine the diagnostic architectures of psychiatric disorders using fundamental genetic and neurobiological data.
Collapse
Affiliation(s)
- Patrick F Sullivan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden; Departments of Genetics and Psychiatry, University of North Carolina, Chapel Hill, NC, USA.
| | - Daniel H Geschwind
- Departments of Neurology, Psychiatry, and Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Institute for Precision Health, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| |
Collapse
|
295
|
Breitfeld J, Wiele N, Gutsmann B, Stumvoll M, Blüher M, Scholz M, Kovacs P, Tönjes A. Circulating Adipokine
VASPIN
Is Associated with Serum Lipid Profiles in Humans. Lipids 2019; 54:203-210. [DOI: 10.1002/lipd.12139] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/08/2019] [Accepted: 02/09/2019] [Indexed: 01/14/2023]
Affiliation(s)
- Jana Breitfeld
- IFB Adiposity DiseasesUniversity of Leipzig Liebigstrasse 21, 04103, Leipzig Germany
| | - Norman Wiele
- Department of Medicine, Division of Endocrinology and NephrologyUniversity of Leipzig Liebigstrasse 21, 04103, Leipzig Germany
| | - Beate Gutsmann
- Department of Medicine, Division of Endocrinology and NephrologyUniversity of Leipzig Liebigstrasse 21, 04103, Leipzig Germany
| | - Michael Stumvoll
- IFB Adiposity DiseasesUniversity of Leipzig Liebigstrasse 21, 04103, Leipzig Germany
- Department of Medicine, Division of Endocrinology and NephrologyUniversity of Leipzig Liebigstrasse 21, 04103, Leipzig Germany
| | - Matthias Blüher
- IFB Adiposity DiseasesUniversity of Leipzig Liebigstrasse 21, 04103, Leipzig Germany
- Department of Medicine, Division of Endocrinology and NephrologyUniversity of Leipzig Liebigstrasse 21, 04103, Leipzig Germany
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and EpidemiologyUniversity of Leipzig Härtelstrasse 16‐18, 04103, Leipzig Germany
- LIFE Research CenterUniversity of Leipzig Philipp‐Rosenthal‐Straβe 27, 04103, Leipzig Germany
| | - Peter Kovacs
- Department of Medicine, Division of Endocrinology and NephrologyUniversity of Leipzig Liebigstrasse 21, 04103, Leipzig Germany
| | - Anke Tönjes
- Department of Medicine, Division of Endocrinology and NephrologyUniversity of Leipzig Liebigstrasse 21, 04103, Leipzig Germany
| |
Collapse
|
296
|
Jakobson CM, She R, Jarosz DF. Pervasive function and evidence for selection across standing genetic variation in S. cerevisiae. Nat Commun 2019; 10:1222. [PMID: 30874558 PMCID: PMC6420628 DOI: 10.1038/s41467-019-09166-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 02/21/2019] [Indexed: 02/06/2023] Open
Abstract
Quantitative genetics aims to map genotype to phenotype, often with the goal of understanding how organisms evolved. However, it remains unclear whether the genetic variants identified are exemplary of evolution. Here we analyzed progeny of two wild Saccharomyces cerevisiae isolates to identify 195 loci underlying complex metabolic traits, resolving 107 to single polymorphisms with diverse molecular mechanisms. More than 20% of causal variants exhibited patterns of emergence inconsistent with neutrality. Moreover, contrary to drift-centric expectation, variation in diverse wild yeast isolates broadly exhibited this property: over 30% of shared natural variants exhibited phylogenetic signatures suggesting that they are not neutral. This pattern is likely attributable to both homoplasy and balancing selection on ancestral polymorphism. Variants that emerged repeatedly were more likely to have done so in isolates from the same ecological niche. Our results underscore the power of super-resolution mapping of ecologically relevant traits in understanding adaptation and evolution.
Collapse
Affiliation(s)
- Christopher M Jakobson
- Department of Chemical & Systems Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Richard She
- Department of Chemical & Systems Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Daniel F Jarosz
- Department of Chemical & Systems Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
297
|
Ormel J, Hartman CA, Snieder H. The genetics of depression: successful genome-wide association studies introduce new challenges. Transl Psychiatry 2019; 9:114. [PMID: 30877272 PMCID: PMC6420566 DOI: 10.1038/s41398-019-0450-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 02/13/2019] [Indexed: 12/15/2022] Open
Abstract
The recent successful genome-wide association studies (GWASs) for depression have yielded more than 80 replicated loci and brought back the excitement that had evaporated during the years of negative GWAS findings. The identified loci provide anchors to explore their relevance for depression, but this comes with new challenges. Using the watershed model of genotype-phenotype relationships as a conceptual aid and recent genetic findings on other complex phenotypes, we discuss why it took so long and identify seven future challenges. The biggest challenge involves the identification of causal mechanisms since GWAS associations merely flag genomic regions without a direct link to underlying biological function. Furthermore, the genetic association with the index phenotype may also be part of a more extensive causal pathway (e.g., from variant to comorbid condition) or be due to indirect influences via intermediate traits located in the causal pathways to the final outcome. This challenge is highly relevant for depression because even its narrow definition of major depressive disorder captures a heterogeneous set of phenotypes which are often measured by even more broadly defined operational definitions consisting of a few questions (minimal phenotyping). Here, Mendelian randomization and future discovery of additional genetic variants for depression and related phenotypes will be of great help. In addition, reduction of phenotypic heterogeneity may also be worthwhile. Other challenges include detecting rare variants, determining the genetic architecture of depression, closing the "heritability gap", and realizing the potential for personalized treatment. Along the way, we identify pertinent open questions that, when addressed, will advance the field.
Collapse
Affiliation(s)
- Johan Ormel
- Departments of Epidemiology and Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Catharina A Hartman
- Departments of Epidemiology and Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Harold Snieder
- Departments of Epidemiology and Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
298
|
Wray NR, Wijmenga C, Sullivan PF, Yang J, Visscher PM. Common Disease Is More Complex Than Implied by the Core Gene Omnigenic Model. Cell 2019; 173:1573-1580. [PMID: 29906445 DOI: 10.1016/j.cell.2018.05.051] [Citation(s) in RCA: 177] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 02/19/2018] [Accepted: 05/24/2018] [Indexed: 12/19/2022]
Abstract
The evidence that most adult-onset common diseases have a polygenic genetic architecture fully consistent with robust biological systems supported by multiple back-up mechanisms is now overwhelming. In this context, we consider the recent "omnigenic" or "core genes" model. A key assumption of the model is that there is a relatively small number of core genes relevant to any disease. While intuitively appealing, this model may underestimate the biological complexity of common disease, and therefore, the goal to discover core genes should not guide experimental design. We consider other implications of polygenicity, concluding that a focus on patient stratification is needed to achieve the goals of precision medicine.
Collapse
Affiliation(s)
- Naomi R Wray
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, Australia.
| | - Cisca Wijmenga
- University of Groningen, University Medical Center Groningen, Department of Genetics, P.O. Box 30001, 9700 RB Groningen, Netherlands
| | - Patrick F Sullivan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jian Yang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Peter M Visscher
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| |
Collapse
|
299
|
Genomic basis of delayed reward discounting. Behav Processes 2019; 162:157-161. [PMID: 30876880 DOI: 10.1016/j.beproc.2019.03.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 03/04/2019] [Accepted: 03/09/2019] [Indexed: 02/07/2023]
Abstract
Delayed reward discounting (DRD) is a behavioral economic measure of impulsivity, reflecting how rapidly a reward loses value based on its temporal distance. In humans, more impulsive DRD is associated with susceptibility to a number of psychiatric diseases (e.g., addiction, ADHD), health outcomes (e.g., obesity), and lifetime outcomes (e.g., educational attainment). Although the determinants of DRD are both genetic and environmental, this review focuses on its genetic basis. Both rodent studies using inbred strains and human twin studies indicate that DRD is moderately heritable, a conclusion that was further supported by a recent human genome-wide association study (GWAS) that used single nucleotide polymorphisms (SNP) to estimate heritability. The GWAS of DRD also identified genetic correlations with psychiatric diagnoses, health outcomes, and measures of cognitive performance. Future research priorities include rodent studies probing putative genetic mechanisms of DRD and human GWASs using larger samples and non-European cohorts. Continuing to characterize genomic influences on DRD has the potential to yield important biological insights with implications for a variety of medically and socially important outcomes.
Collapse
|
300
|
Statistical power in genome-wide association studies and quantitative trait locus mapping. Heredity (Edinb) 2019; 123:287-306. [PMID: 30858595 DOI: 10.1038/s41437-019-0205-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 02/22/2019] [Accepted: 02/24/2019] [Indexed: 12/16/2022] Open
Abstract
Power calculation prior to a genetic experiment can help investigators choose the optimal sample size to detect a quantitative trait locus (QTL). Without the guidance of power analysis, an experiment may be underpowered or overpowered. Either way will result in wasted resource. QTL mapping and genome-wide association studies (GWAS) are often conducted using a linear mixed model (LMM) with controls of population structure and polygenic background using markers of the whole genome. Power analysis for such a mixed model is often conducted via Monte Carlo simulations. In this study, we derived a non-centrality parameter for the Wald test statistic for association, which allows analytical power analysis. We show that large samples are not necessary to detect a biologically meaningful QTL, say explaining 5% of the phenotypic variance. Several R functions are provided so that users can perform power analysis to determine the minimum sample size required to detect a given QTL with a certain statistical power or calculate the statistical power with given sample size and known values of other population parameters.
Collapse
|