Recalibrating the Genome-Wide Association Null Hypothesis

Total Page:16

File Type:pdf, Size:1020Kb

Recalibrating the Genome-Wide Association Null Hypothesis bioRxiv preprint doi: https://doi.org/10.1101/597484; this version posted October 4, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 1 1 Recalibrating the Genome-wide Association Null Hypothesis 2 Enables the Detection of Core Genes Underlying Complex 3 Traits 4 1,2 1,2 2-4 5 Wei Cheng , Sohini Ramachandran y, and Lorin Crawford y 6 1 Department of Ecology and Evolutionary Biology, Brown University, Providence, RI, 7 USA 8 2 Center for Computational Molecular Biology, Brown University, Providence, RI, USA 9 3 Department of Biostatistics, Brown University, Providence, RI, USA 10 4 Center for Statistical Sciences, Brown University, Providence, RI, USA 11 E-mail: [email protected]; lorin [email protected] y 12 Abstract 13 Traditional univariate genome-wide association studies generate false positives and negatives due to 14 difficulties distinguishing causal variants from \interactive" variants (i.e., variants correlated with causal 15 variants without directly influencing the trait). Recent efforts have been directed at identifying gene or 16 pathway associations, but these can be computationally costly and hampered by strict model assumptions. 17 Here, we present gene-", a new approach for identifying statistical associations between sets of variants 18 and quantitative traits. Our key innovation is a recalibration of the genome-wide null hypothesis to treat 19 small-yet-nonzero associations emitted by interactive variants as non-causal. gene-" efficiently identifies 20 core genes under a variety of simulated genetic architectures, achieving greater than a 90% true positive 21 rate at 1% false positive rate for polygenic traits. Lastly, we apply gene-" to summary statistics derived 22 from six quantitative traits using European-ancestry individuals in the UK Biobank, and identify genes 23 that are enriched in biological relevant pathways. 24 Introduction 25 Over the last decade, there has been considerable debate surrounding whether genome-wide single- 26 nucleotide polymorphism (SNP) genotype data can offer insight into the genetic architecture of com- 27 plex traits [1{5]. In the traditional genome-wide association (GWA) framework, individual SNPs are 28 tested independently for association with a trait of interest. While this approach has drawbacks [2,3,6], 29 more recent approaches that combine SNPs within a region have gained power to detect biologically 30 relevant genes and pathways enriched for correlations with complex traits [7{14]. Reconciling these two 31 observations is crucial for biomedical genomics. 32 In the traditional GWA model, each SNP is assumed to either (i) directly influence (or perfectly 33 tag a variant that directly influences) the trait of interest; or (ii) have no affect on the trait at all (see 34 Fig. 1a). Throughout this manuscript, for simplicity, we refer to SNPs under the former as \causal" and 35 those under latter as \non-causal". These classifications are based on ordinary least squares (OLS) effect 36 size estimates βj for each j-th SNP in a regression framework, where the null hypothesis assumes no 37 association for the true effects of non-causal SNPs (H0 : βj = 0). The traditional GWA model is agnostic 38 to trait architecture,b and is underpowered with a high false-positive rate for \polygenic" traits or traits 39 which are generated by many mutations of small effect [5, 15{17]. 40 Suppose that in truth each SNP in a GWA dataset instead belongs to one of three categories: (i) 41 causal or truly associated; (ii) statistically associated with the trait but not directly influencing it, which 42 we refer to as \interactive"; and (iii) non-causal (Fig. 1b) [18]. Equivalently, these three categories can be bioRxiv preprint doi: https://doi.org/10.1101/597484; this version posted October 4, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 2 43 defined using the relationship between the true SNP effects β and the observed effect size estimates β:(i) 44 for causal SNPs, the true effects β = 0 and the observed effect size estimates β = 0; (ii) for \interactive" 6 6 45 SNPs, β = 0 or β 0, and β = 0; and (iii) for non-causal SNPs, β = 0 and β = 0. More specifically,b ≈ 6 46 causal SNPs lie in core genes that directly influence the trait of interest. Interactiveb SNPs then follow 47 one of two definitions. Theyb are either non-causal SNPs that are statisticallyb associated with the trait 48 due to some varying degree of linkage disequilibrium (LD) with causal SNPs (β = 0); or they are SNPs 49 that are not directly causal, but are located within a gene that covaries with a core gene (β 0) [19]. ≈ 50 In either setting, interactive SNPs will emit intermediate statistical noise (in some cases, even appearing 51 indistinguishable from causal SNPs), thereby confounding traditional GWA tests (Fig. 1b). Hereafter, 52 we refer to this noise as \epsilon-genic effects” (denoted in shorthand as \"-genic effects”). There is a 53 need for a computational framework that has the ability to identify mutations associated with a wide 54 range of traits, regardless of whether heritability is sparsely or uniformly distributed across the genome. 55 Here, we develop a new and scalable quantitative approach for testing aggregated sets of SNP-level 56 GWA summary statistics. In practice, our approach can be applied to any user-specified set of genomic 57 regions, such as regulatory elements, intergenic regions, or gene sets. In this study, for simplicity, we refer 58 to our method as a gene-level test (i.e., an annotated collection of SNPs within the boundary of a gene). 59 The key contribution for our approaches is that gene-level association tests should treat interactive SNPs 60 with "-genic effects as non-causal. Conceptually, this requires assessing whether a given SNP explains 2 61 more than an \epsilon" proportion of narrow-sense heritability, h . In this generalized model, we assume 62 that true SNP-level effects for complex traits are jointly distributed as 2 63 β (0; Ω); Ω = σ ; Ω = σ ρ(x ; x )σ ; (1) ∼ N jj j jl j j l l 1=2 1=2 2 2 64 where Ω = H ΣH is a J J variance-covariance matrix, and H = diag(σ1; : : : ; σJ ) is a J J diagonal 2 × 2 × 65 matrix with elements σj denoting the proportion of narrow-sense heritability h contributed by the j-th 2 2 66 SNP (with the constraint σj = h ). Under our framework, we recalibrate the GWA null hypothesis 67 to assume approximately no association for interactive and non-causal SNPs, H0 : βj 0. This can be P 2 2 ≈ 2 68 formally restated using the variance-component-based null hypothesis, H : σ σ , where σ represents 0 j ≤ " " 69 the maximum proportion-of-variance-explained (PVE) by an interactive or non-causal SNP (Fig. 1b). 2 2 70 Non-core genes are then defined as genes that only contain SNPs with "-genic effects (i.e. 0 σ σ ≤ j ≤ " 71 for every j-th SNP within that region). Core genes, on the other hand, are genes that contain at least 2 2 72 one causal SNP (i.e. σj > σ" for at least one SNP j within that region). By modeling "-genic effects 2 73 (i.e., different values of σ" for interactive SNPs), our approach flexibly constructs an appropriate null 74 hypothesis for a wide range of traits with genetic architectures that land anywhere on the polygenic 75 spectrum (see Methods). 76 We refer to our gene-level association framework as \gene-"" (pronounced \genie"). gene-" lowers 77 false positive rates and increases power for identifying gene-level associations from GWA studies via 78 two key conceptual insights. First, gene-" regularizes observed (and inflated) GWA summary statistics 79 so that SNP-level effect size estimates are positively correlated with the assumed generative model of 80 complex traits. Second, it examines the distribution of regularized effect sizes to determine the "-genic 2 81 threshold σ" of interactive and non-causal SNPs. This makes for an improved hypothesis testing strategy 82 for identifying core genes associated with complex traits. With detailed simulations, we assess the power 83 of gene-" to identify core genes under a variety of genetic architectures, and compare its performance 84 against multiple competing approaches [7,10,12,14,20]. We also apply gene-" to the SNP-level summary 85 statistics of six quantitative traits assayed in individuals of European ancestry from the UK Biobank [21]. bioRxiv preprint doi: https://doi.org/10.1101/597484; this version posted October 4, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 3 86 Results 87 Overview of gene-" 88 The gene-" framework requires two inputs: GWA SNP-level effect size estimates, and an empirical linkage 89 disequilibrium (LD, or variance-covariance) matrix. The LD matrix can be estimated directly from 90 genotype data, or from an ancestry-matched set of samples if genotype data are not available to the 2 91 user. We use these inputs to both estimate gene-level contributions to narrow-sense heritability h , and 92 perform gene-level association tests.
Recommended publications
  • Proquest Dissertations
    nm u Ottawa L'Universite canadienne Canada's university run FACULTE DES ETUDES SUPERIEURES ^=1 FACULTY OF GRADUATE AND ET POSTOCTORALES U Ottawa POSDOCTORAL STUDIES L'Univeisittf canathenne Canada's university Anna Francina Jackson „_„^^.^^_^^^.„^ M.Sc. (Cellular and Molecular Medicine) __„.___._ Department of Cellular and Molecular Medicine The Functional Characterization of Wdr68 Regulation of Pax7 Activity in Myogenesis TITRE DE LA THESE / TITLE OF THESIS M. Rudnicki „„„„_.___ N;LWij3CT:Berg©ron RJjcreaton_ Gary W. Slater Le Doyen de la Faculte des etudes superieures et postdoctorales / Dean of the Faculty of Graduate and Postdoctoral Studies The Functional Characterization of Wdr68 Regulation of Pax7 Activity in Myogenesis Anna Francina Jackson This thesis is submitted to the Faculty of Graduate and Postdoctoral Studies in partial fulfillment of the requirements for a Master of Science degree in Cellular and Molecular Medicine. Department of Cellular and Molecular Medicine Faculty of Medicine University of Ottawa May 14, 2010 ©Anna Francina Jackson, Ottawa, Canada, 2010 Library and Archives Bibliotheque et 1*1 Canada Archives Canada Published Heritage Direction du Branch Patrimoine de I'edition 395 Wellington Street 395, rue Wellington OttawaONK1A0N4 OttawaONK1A0N4 Canada Canada Your file Votre reference ISBN: 978-0-494-74221-1 Our file Notre reference ISBN: 978-0-494-74221-1 NOTICE: AVIS: The author has granted a non­ L'auteur a accorde une licence non exclusive exclusive license allowing Library and permettant a la Bibliotheque
    [Show full text]
  • Multidrug Transporter MRP4/ABCC4 As a Key Determinant of Pancreatic
    www.nature.com/scientificreports OPEN Multidrug transporter MRP4/ ABCC4 as a key determinant of pancreatic cancer aggressiveness A. Sahores1, A. Carozzo1, M. May1, N. Gómez1, N. Di Siervi1, M. De Sousa Serro1, A. Yanef1, A. Rodríguez‑González2, M. Abba3, C. Shayo2 & C. Davio1* Recent fndings show that MRP4 is critical for pancreatic ductal adenocarcinoma (PDAC) cell proliferation. Nevertheless, the signifcance of MRP4 protein levels and function in PDAC progression is still unclear. The aim of this study was to determine the role of MRP4 in PDAC tumor aggressiveness. Bioinformatic studies revealed that PDAC samples show higher MRP4 transcript levels compared to normal adjacent pancreatic tissue and circulating tumor cells express higher levels of MRP4 than primary tumors. Also, high levels of MRP4 are typical of high-grade PDAC cell lines and associate with an epithelial-mesenchymal phenotype. Moreover, PDAC patients with high levels of MRP4 depict dysregulation of pathways associated with migration, chemotaxis and cell adhesion. Silencing MRP4 in PANC1 cells reduced tumorigenicity and tumor growth and impaired cell migration. Transcriptomic analysis revealed that MRP4 silencing alters PANC1 gene expression, mainly dysregulating pathways related to cell-to-cell interactions and focal adhesion. Contrarily, MRP4 overexpression signifcantly increased BxPC-3 growth rate, produced a switch in the expression of EMT markers, and enhanced experimental metastatic incidence. Altogether, our results indicate that MRP4 is associated with a more aggressive phenotype in PDAC, boosting pancreatic tumorigenesis and metastatic capacity, which could fnally determine a fast tumor progression in PDAC patients. Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal human malignancies, due to its late diag- nosis, inherent resistance to treatment and early dissemination 1.
    [Show full text]
  • DYRK1A Binds to an Evolutionarily Conserved WD40-Repeat Title Protein WDR68 and Induces Its Nuclear Translocation
    DYRK1A binds to an evolutionarily conserved WD40-repeat Title protein WDR68 and induces its nuclear translocation. Author(s) Miyata, Yoshihiko; Nishida, Eisuke Citation Biochimica et biophysica acta (2011), 1813(10): 1728-1739 Issue Date 2011-10 URL http://hdl.handle.net/2433/148020 © 2011 Elsevier B.V.; This is not the published version. Please cite only the published version.; この論文は出版社版であり Right ません。引用の際には出版社版をご確認ご利用ください 。 Type Journal Article Textversion author Kyoto University *REVISED Manuscript (text UNmarked) Click here to view linked References DYRK1A binds to an evolutionarily conserved WD40-repeat protein WDR68 and induces its nuclear translocation Yoshihiko Miyata*, Eisuke Nishida Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Kitashirakawa Oiwake-cho, Kyoto 606-8502, Japan * Corresponding author. Department of Cell & Developmental Biology, Graduate School of Biostudies, Kyoto University, Kitashirakawa Oiwake-cho, Sakyo-ku, Kyoto 606-8502, Japan. Tel.: +81-75-753-4231; fax: +81-75-753-4235. 1 ABSTRACT DYRK1A is encoded in the Down’s syndrome critical region on human chromosome 21, and plays an important role in the functional and developmental regulation of many types of cells, including neuronal cells. Here we have identified WDR68, an evolutionarily conserved protein with WD40-repeat domains, as a cellular binding partner of DYRK1A. WDR68 was originally identified in petunia as AN11 that controls the pigmentation of flowers by stimulating the transcription of anthocyanin biosynthetic genes. Experiments with RNA interference showed that WDR68 was indispensable for the optimal proliferation and survival of mammalian cultured cell, and WDR68 depletion induced cell apoptosis. DYRK1A and DYRK1B, but not DYRK2, DYRK3, or DYRK4, bound to endogenous and expressed WDR68.
    [Show full text]
  • Estimation of Non-Null SNP Effect Size Distributions Enables the Detection
    bioRxiv preprint doi: https://doi.org/10.1101/597484; this version posted May 13, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 1 1 Estimation of Non-null SNP Effect Size Distributions Enables 2 the Detection of Enriched Genes Underlying Complex Traits 3 1,2 1,2 2-4 4 Wei Cheng , Sohini Ramachandran y, and Lorin Crawford y 5 1 Department of Ecology and Evolutionary Biology, Brown University, Providence, RI, 6 USA 7 2 Center for Computational Molecular Biology, Brown University, Providence, RI, USA 8 3 Department of Biostatistics, Brown University, Providence, RI, USA 9 4 Center for Statistical Sciences, Brown University, Providence, RI, USA 10 Corresponding E-mail: [email protected]; lorin [email protected] y 11 Abstract 12 Traditional univariate genome-wide association studies generate false positives and negatives due to 13 difficulties distinguishing associated variants from variants with spurious nonzero effects that do not 14 directly influence the trait. Recent efforts have been directed at identifying genes or signaling pathways 15 enriched for mutations in quantitative traits or case-control studies, but these can be computationally 16 costly and hampered by strict model assumptions. Here, we present gene-", a new approach for identifying 17 statistical associations between sets of variants and quantitative traits. Our key insight is that enrichment 18 studies on the gene-level are improved when we reformulate the genome-wide SNP-level null hypothesis 19 to identify spurious small-to-intermediate SNP effects and classify them as non-causal.
    [Show full text]
  • Noncoding Rnas As Novel Pancreatic Cancer Targets
    NONCODING RNAS AS NOVEL PANCREATIC CANCER TARGETS by Amy Makler A Thesis Submitted to the Faculty of The Charles E. Schmidt College of Science In Partial Fulfillment of the Requirements for the Degree of Master of Science Florida Atlantic University Boca Raton, FL August 2018 Copyright 2018 by Amy Makler ii ACKNOWLEDGEMENTS I would first like to thank Dr. Narayanan for his continuous support, constant encouragement, and his gentle, but sometimes critical, guidance throughout the past two years of my master’s education. His faith in my abilities and his belief in my future success ensured I continue down this path of research. Working in Dr. Narayanan’s lab has truly been an unforgettable experience as well as a critical step in my future endeavors. I would also like to extend my gratitude to my committee members, Dr. Binninger and Dr. Jia, for their support and suggestions regarding my thesis. Their recommendations added a fresh perspective that enriched our initial hypothesis. They have been indispensable as members of my committee, and I thank them for their contributions. My parents have been integral to my successes in life and their support throughout my education has been crucial. They taught me to push through difficulties and encouraged me to pursue my interests. Thank you, mom and dad! I would like to thank my boyfriend, Joshua Disatham, for his assistance in ensuring my writing maintained a logical progression and flow as well as his unwavering support. He was my rock when the stress grew unbearable and his encouraging words kept me pushing along.
    [Show full text]
  • Mammalian PRC1 Complexes: Compositional Complexity and Diverse Molecular Mechanisms
    International Journal of Molecular Sciences Review Mammalian PRC1 Complexes: Compositional Complexity and Diverse Molecular Mechanisms Zhuangzhuang Geng 1 and Zhonghua Gao 1,2,3,* 1 Departments of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA 17033, USA; [email protected] 2 Penn State Hershey Cancer Institute, Hershey, PA 17033, USA 3 The Stem Cell and Regenerative Biology Program, Penn State College of Medicine, Hershey, PA 17033, USA * Correspondence: [email protected] Received: 6 October 2020; Accepted: 5 November 2020; Published: 14 November 2020 Abstract: Polycomb group (PcG) proteins function as vital epigenetic regulators in various biological processes, including pluripotency, development, and carcinogenesis. PcG proteins form multicomponent complexes, and two major types of protein complexes have been identified in mammals to date, Polycomb Repressive Complexes 1 and 2 (PRC1 and PRC2). The PRC1 complexes are composed in a hierarchical manner in which the catalytic core, RING1A/B, exclusively interacts with one of six Polycomb group RING finger (PCGF) proteins. This association with specific PCGF proteins allows for PRC1 to be subdivided into six distinct groups, each with their own unique modes of action arising from the distinct set of associated proteins. Historically, PRC1 was considered to be a transcription repressor that deposited monoubiquitylation of histone H2A at lysine 119 (H2AK119ub1) and compacted local chromatin. More recently, there is increasing evidence that demonstrates the transcription activation role of PRC1. Moreover, studies on the higher-order chromatin structure have revealed a new function for PRC1 in mediating long-range interactions. This provides a different perspective regarding both the transcription activation and repression characteristics of PRC1.
    [Show full text]
  • THE OLD and NEW FACE of CRANIOFACIAL RESEARCH: How Animal Models Inform Human Craniofacial Genetic and Clinical Data
    HHS Public Access Author manuscript Author ManuscriptAuthor Manuscript Author Dev Biol Manuscript Author . Author manuscript; Manuscript Author available in PMC 2017 July 15. Published in final edited form as: Dev Biol. 2016 July 15; 415(2): 171–187. doi:10.1016/j.ydbio.2016.01.017. THE OLD AND NEW FACE OF CRANIOFACIAL RESEARCH: How animal models inform human craniofacial genetic and clinical data Eric Van Otterloo, Trevor Williams, and Kristin B. Artinger Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA Kristin B. Artinger: [email protected] Abstract The craniofacial skeletal structures that comprise the human head develop from multiple tissues that converge to form the bones and cartilage of the face. Because of their complex development and morphogenesis, many human birth defects arise due to disruptions in these cellular populations. Thus, determining how these structures normally develop is vital if we are to gain a deeper understanding of craniofacial birth defects and devise treatment and prevention options. In this review, we will focus on how animal model systems have been used historically and in an ongoing context to enhance our understanding of human craniofacial development. We do this by first highlighting “animal to man” approaches: that is, how animal models are being utilized to understand fundamental mechanisms of craniofacial development. We discuss emerging technologies, including high throughput sequencing and genome editing, and new animal repository resources, and how their application can revolutionize the future of animal models in craniofacial research. Secondly, we highlight “man to animal” approaches, including the current use of animal models to test the function of candidate human disease variants.
    [Show full text]
  • Dual-Specificity, Tyrosine Phosphorylation-Regulated Kinases
    International Journal of Molecular Sciences Review Dual-Specificity, Tyrosine Phosphorylation-Regulated Kinases (DYRKs) and cdc2-Like Kinases (CLKs) in Human Disease, an Overview Mattias F. Lindberg and Laurent Meijer * Perha Pharmaceuticals, Perharidy Peninsula, 29680 Roscoff, France; [email protected] * Correspondence: [email protected] Abstract: Dual-specificity tyrosine phosphorylation-regulated kinases (DYRK1A, 1B, 2-4) and cdc2- like kinases (CLK1-4) belong to the CMGC group of serine/threonine kinases. These protein ki- nases are involved in multiple cellular functions, including intracellular signaling, mRNA splicing, chromatin transcription, DNA damage repair, cell survival, cell cycle control, differentiation, ho- mocysteine/methionine/folate regulation, body temperature regulation, endocytosis, neuronal development, synaptic plasticity, etc. Abnormal expression and/or activity of some of these kinases, DYRK1A in particular, is seen in many human nervous system diseases, such as cognitive deficits associated with Down syndrome, Alzheimer’s disease and related diseases, tauopathies, demen- tia, Pick’s disease, Parkinson’s disease and other neurodegenerative diseases, Phelan-McDermid syndrome, autism, and CDKL5 deficiency disorder. DYRKs and CLKs are also involved in dia- betes, abnormal folate/methionine metabolism, osteoarthritis, several solid cancers (glioblastoma, breast, and pancreatic cancers) and leukemias (acute lymphoblastic leukemia, acute megakaryoblas- Citation: Lindberg, M.F.; Meijer, L. tic leukemia), viral infections (influenza, HIV-1, HCMV, HCV, CMV, HPV), as well as infections Dual-Specificity, Tyrosine caused by unicellular parasites (Leishmania, Trypanosoma, Plasmodium). This variety of pathological Phosphorylation-Regulated Kinases implications calls for (1) a better understanding of the regulations and substrates of DYRKs and (DYRKs) and cdc2-Like Kinases CLKs and (2) the development of potent and selective inhibitors of these kinases and their evaluation (CLKs) in Human Disease, an as therapeutic drugs.
    [Show full text]
  • Loss-Of-Function Mutations in the CABLES1 Gene Are a Novel Cause
    248 L C Hernández- CABLES1 mutations in 24:8 379–392 Research Ramírez et al. Cushing’s disease Open Access Loss-of-function mutations in the CABLES1 gene are a novel cause of Cushing’s disease Laura C Hernández-Ramírez1, Ryhem Gam2, Nuria Valdés1,3, Maya B Lodish1, Nathan Pankratz4, Aurelio Balsalobre2, Yves Gauthier2, Fabio R Faucz1, Giampaolo Trivellin1, Prashant Chittiboina5, John Lane4, Denise M Kay6 Aggeliki Dimopoulos7, Stephan Gaillard8,9, Mario Neou8, Jérôme Bertherat8,10, Guillaume Assié8,10, Chiara Villa8,11,12, James L Mills7,*, Jacques Drouin2,*, and Constantine A Stratakis1,* 1Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, Maryland, USA 2Laboratoire de Génétique Moléculaire, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, Québec, Canada 3Service of Endocrinology and Nutrition, Hospital Universitario Central de Asturias, Instituto Universita- rio de Oncología del Principado de Asturias, Universidad de Oviedo, Oviedo, Spain 4Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, Minnesota, USA 5Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, Maryland, USA 6Newborn Screening Program, Wadsworth Center, New York State Department of Health, Albany, New York, USA 7Division of Intramural Population Health Research, Epidemiology Branch, Eunice Kennedy Shriver
    [Show full text]
  • Molecular Characterization of Selectively Vulnerable Neurons in Alzheimer’S Disease
    RESOURCE https://doi.org/10.1038/s41593-020-00764-7 Molecular characterization of selectively vulnerable neurons in Alzheimer’s disease Kun Leng1,2,3,4,13, Emmy Li1,2,3,13, Rana Eser 5, Antonia Piergies5, Rene Sit2, Michelle Tan2, Norma Neff 2, Song Hua Li5, Roberta Diehl Rodriguez6, Claudia Kimie Suemoto7,8, Renata Elaine Paraizo Leite7, Alexander J. Ehrenberg 5, Carlos A. Pasqualucci7, William W. Seeley5,9, Salvatore Spina5, Helmut Heinsen7,10, Lea T. Grinberg 5,7,11 ✉ and Martin Kampmann 1,2,12 ✉ Alzheimer’s disease (AD) is characterized by the selective vulnerability of specific neuronal populations, the molecular sig- natures of which are largely unknown. To identify and characterize selectively vulnerable neuronal populations, we used single-nucleus RNA sequencing to profile the caudal entorhinal cortex and the superior frontal gyrus—brain regions where neurofibrillary inclusions and neuronal loss occur early and late in AD, respectively—from postmortem brains spanning the progression of AD-type tau neurofibrillary pathology. We identified RORB as a marker of selectively vulnerable excitatory neu- rons in the entorhinal cortex and subsequently validated their depletion and selective susceptibility to neurofibrillary inclusions during disease progression using quantitative neuropathological methods. We also discovered an astrocyte subpopulation, likely representing reactive astrocytes, characterized by decreased expression of genes involved in homeostatic functions. Our characterization of selectively vulnerable neurons in AD paves the way for future mechanistic studies of selective vulnerability and potential therapeutic strategies for enhancing neuronal resilience. elective vulnerability is a fundamental feature of neurodegen- Here, we performed snRNA-seq on postmortem brain tissue erative diseases, in which different neuronal populations show from a cohort of individuals spanning the progression of AD-type a gradient of susceptibility to degeneration.
    [Show full text]
  • Peri-Implantation Lethality in Mice Carrying Megabase-Scale Deletion
    www.nature.com/scientificreports OPEN Peri-implantation lethality in mice carrying megabase-scale deletion on 5qc3.3 is caused by Exoc1 null Received: 12 June 2015 Accepted: 03 August 2015 mutation Published: 08 September 2015 Seiya Mizuno*, Kohei Takami*, Yoko Daitoku, Yoko Tanimoto, Tra Thi Huong Dinh, Saori Mizuno-Iijima, Yoshikazu Hasegawa, Satoru Takahashi, Fumihiro Sugiyama & Ken-ichi Yagami We found a novel spontaneous mouse mutant with depigmentation in the ventral body, which we called White Spotting (WS) mouse. Genetic investigation revealed deletion of a > 1.2-Mb genomic region containing nine genes (Kit, Kdr, Srd5a3, Tmeme165, Clock, Pdcl2, Nmu, Exoc1, and Cep135). We designated this mutant allele KitWS. Interestingly, homozygous mutants (KitWS/WS) showed a peri- implantation lethal phenotype. Expression analyses of these nine genes in blastocysts suggested that Exoc1 was a prime candidate for this phenotype. We produced Exoc1 knockout mice, and the same peri-implantation lethal phenotype was seen in Exoc1−/− embryos. In addition, the polygenic effect without Exoc1 was investigated in genome-edited KitWE mice carrying the Mb-scale deletion induced by the CRISPR/Cas9 system. As KitWE/WE embryos did not exhibit the abnormal phenotype, which was seen in KitWS/WS. We concluded that peri-implantation lethality in KitWS/WS was caused by a monogenic defect of Exoc1. There have been many reports of spontaneous mouse mutants with depigmentation phenotypes1, among which Kit gene mutant mouse strains have been investigated in detail. More than 150 Kit mutant strains, including 72 spontaneous mutants, are listed in the Mouse Genome Informatics (MGI) database (http:// www.informatics.jax.org).
    [Show full text]
  • AUTS2 Controls Neuronal Lineage Choice Through a Novel PRC1-Independent Complex And
    bioRxiv preprint doi: https://doi.org/10.1101/2021.06.29.450402; this version posted June 29, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. AUTS2 controls neuronal lineage choice through a novel PRC1-independent complex and BMP inhibition Zhuangzhuang Geng1, Qiang Wang1, Weili Miao2, Trevor Wolf3, Jessenia Chavez1, Emily Giddings3, Ryan Hobbs4, David J. DeGraff5,6, Yinsheng Wang2, James Stafford3, Zhonghua Gao1,6,7,# 1, Departments of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA 17033, USA 2, Department of Chemistry, University of California at Riverside, Riverside, CA 92521, USA 3, Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA 4, Department of Dermatology, Penn State College of Medicine, Hershey, PA 17033, USA 5, Department of Pathology and Laboratory Medicine, Penn State College of Medicine, Hershey, PA 17033, USA 6, Penn State Hershey Cancer Institute, Hershey, PA 17033, USA 7, The Stem Cell and Regenerative Biology Program, Penn State College of Medicine, Hershey, PA 17033, USA #, correspondence author, [email protected] bioRxiv preprint doi: https://doi.org/10.1101/2021.06.29.450402; this version posted June 29, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Summary Despite a prominent risk factor for Neurodevelopmental disorders (NDD), it remains unclear how Autism Susceptibility Candidate 2 (AUTS2) controls the neurodevelopmental program. Our studies investigated the role of AUTS2 in neuronal differentiation and discovered that AUTS2, together with WDR68 and SKI, forms a novel protein complex (AWS) specifically in neuronal progenitors and promotes neuronal differentiation through inhibiting BMP signaling.
    [Show full text]