Genome-Wide Association Analysis Reveals QTL and Candidate Mutations Structure, Function, and Regulation

Total Page:16

File Type:pdf, Size:1020Kb

Genome-Wide Association Analysis Reveals QTL and Candidate Mutations Structure, Function, and Regulation Jivanji et al. Genet Sel Evol (2019) 51:62 https://doi.org/10.1186/s12711-019-0506-2 Genetics Selection Evolution RESEARCH ARTICLE Open Access Genome-wide association analysis reveals QTL and candidate mutations involved in white spotting in cattle Swati Jivanji1* , Gemma Worth2, Thomas J. Lopdell2, Anna Yeates2, Christine Couldrey2, Edwardo Reynolds1, Kathryn Tiplady2, Lorna McNaughton2, Thomas J. J. Johnson2, Stephen R. Davis2, Bevin Harris2, Richard Spelman2, Russell G. Snell3, Dorian Garrick1 and Mathew D. Littlejohn2 Abstract Background: White spotting of the coat is a characteristic trait of various domestic species including cattle and other mammals. It is a hallmark of Holstein–Friesian cattle, and several previous studies have detected genetic loci with major efects for white spotting in animals with Holstein–Friesian ancestry. Here, our aim was to better understand the underlying genetic and molecular mechanisms of white spotting, by conducting the largest mapping study for this trait in cattle, to date. Results: Using imputed whole-genome sequence data, we conducted a genome-wide association analysis in 2973 mixed-breed cows and bulls. Highly signifcant quantitative trait loci (QTL) were found on chromosomes 6 and 22, highlighting the well-established coat color genes KIT and MITF as likely responsible for these efects. These results are in broad agreement with previous studies, although we also report a third signifcant QTL on chromosome 2 that appears to be novel. This signal maps immediately adjacent to the PAX3 gene, which encodes a known transcrip- tion factor that controls MITF expression and is the causal locus for white spotting in horses. More detailed exami- nation of these loci revealed a candidate causal mutation in PAX3 (p.Thr424Met), and another candidate mutation (rs209784468) within a conserved element in intron 2 of MITF transcripts expressed in the skin. These analyses also revealed a mechanistic ambiguity at the chromosome 6 locus, where highly dispersed association signals suggested multiple or multiallelic QTL involving KIT and/or other genes in this region. Conclusions: Our fndings extend those of previous studies that reported KIT as a likely causal gene for white spot- ting, and report novel associations between candidate causal mutations in both the MITF and PAX3 genes. The sizes of the efects of these QTL are substantial, and could be used to select animals with darker, or conversely whiter, coats depending on the desired characteristics. Background several genes with major efects have been described and Coat patterning traits provide visual characteristics are relevant across species, as well as many other loci that allow diferentiation between domesticated animal with small efects that account for the remaining genetic breeds and between strains within breeds. White spotting variance [1]. Tis oligogenic architecture derives from the is one of these phenotypes, and is a feature of a variety of multifaceted biology that contributes to white spotting of mammals including cattle, horses, dogs, cats and mice. the coat, which is hypothesised to arise from abnormal White spotting is a complex quantitative trait, for which melanocyte precursor migration and/or development. Mouse models have demonstrated that pigment cells *Correspondence: [email protected] originate from the neural crest cells via the SOX10 posi- 1 Massey University Manawatu, Private Bag 11 222, Palmerston tive glial bipotent progenitor cells during embryogenesis, North 4442, New Zealand and migrate dorsally via the neural tube [2]. Tese cells Full list of author information is available at the end of the article © The Author(s) 2019. This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creat iveco mmons .org/licen ses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/ publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated. Jivanji et al. Genet Sel Evol (2019) 51:62 Page 2 of 18 proceed to diferentiate into melanoblasts by acquiring trait [1]. Together these studies converge on the involve- expression of the genes micropthalmia-associated tran- ment of KIT and MITF gene expression in white spot- scription factor (MITF), proto-oncogene receptor tyrosine ting in dairy cattle, however the causal variants that drive kinase (KIT) and dopachrome tautomerase (DCT), and these efects have yet to be defnitively identifed and may migrate down the ventral axis of the body. When the cells be breed-specifc. reach their destination, they migrate into the epidermis Here, our aim was to investigate white spotting in New where some melanoblasts localise to the hair follicle and Zealand dairy cattle, by using whole-genome sequence diferentiate into melanocytes. A subset of melanoblasts genotype data to conduct the largest GWAS of white dediferentiate, losing MITF and KIT gene expression, spotting to date. We report three genome-wide signif- and colonise the hair follicle bulge where they act as mel- cant QTL for white spotting. Efects on chromosomes 6 anocyte stem cells and replenish diferentiated melano- and 22 extend on previous associations at these loci, and cytes during subsequent hair cycles [2]. Disruption of any further implicate the KIT and MITF genes as responsible of the above processes is expected to result in parts of the for these efects. For the frst time, we also report a QTL body lacking mature melanocytes, and thus regions of on chromosome 2 that implicates the PAX3 gene in white abnormal pigmentation in the hair coat. spotting of dairy cattle and highlight an amino acid sub- Quantitative trait loci (QTL) and mutations that cause stitution that may underlie this efect. white spotting have been described for a variety of spe- cies. Genetic studies in the horse revealed an inversion in Methods the KIT gene associated with the Tobiano white-spotting Study population [3], and a mutation in the PAX3 gene associated with a White spotting data were derived from several cohorts of splashed white pattern [4, 5]. Several mutations in the animals that included: 885 outbred dairy bulls (223 J, 327 KIT gene have also been associated with complete white HF, and 335 HF × J), 1389 outbred dairy cows (51 J, 265 [6] or roan coat phenotypes [7]. Studies on white spotting HF, and 1073 HF × J), and 699 HF × J F2 cross cows from in dogs have revealed associations with the MITF gene an experimental pedigree. Breed defnitions, in these [8], and in mice more than 10 genes have been reported cases, defne animals from a 4-generation pedigree that 16 15 to be associated with white spotting traits, including the were /16 J or HF as purebreds, with /16 animals defned KIT and MITF genes [9]. Comparatively few studies have as crossbreeds. Te F2 animals were ½ HF × ½ J, repre- investigated the genetics of white spotting in cattle. Liu senting a study population that was previously described et al. [10] found signifcant QTL on chromosomes 6, 18 in several publications [10, 13–15]. Genotyping data were and 22 using linkage analysis within Holstein–Friesian available for 2973 animals, with genotype and pheno- (HF) × Jersey (J) crossbred cows. It has been suggested type information derived as described in the following that the QTL on chromosomes 6 and 22 might be under- sections. pinned by the KIT and MITF genes, respectively [10]. Fontanesi et al. [11] compared the sequences of the MITF Measurements of white spotting in our study population gene in white spotted Italian Holstein and Simmental cat- For animals in the F2 population, proportion of white tle, and solid coloured Italian Brown and Reggiana cat- spotting values that had been derived for a previous study tle, and found a haplotype (carrying allele g.31831615T) [10] were used directly in the current study. Video foot- that is associated with white spotting. Tis haplotype age was recorded on 1389 cows walking single fle either accounts for some, but not all of the variation observed into or out of the milking shed using a GoPro HERO4 in the white spotting phenotype [11]. More recently, camera, at a 4000 pixel horizontal resolution. Still images Hofstetter et al. [12] investigated atypical white spotting that provide a clear side-on view of each animal were cap- in Brown Swiss cattle. Tey identifed two completely tured from the video footage using VideoPad Video Edi- linked single nucleotide variants within the 5′ regula- tor (v5.3). Additional side-on images representing either tory region of the MITF gene associated with white spot- the right or left profle of 885 bulls were made available ting, and although these variants largely account for the by LIC and incorporated into the dataset. First, cows and manifestation of white spotting, they do not account for bulls were scored for the presence or absence of white the variability between individuals, which provides fur- on their coat and, then, the proportion of white spotting ther evidence for a polygenic trait [12]. Hayes et al. [1] was quantifed. Quantifcation was carried out manually detected the MITF and KIT genes in a genome-wide using the image processing software, GNU Image Manip- association study (GWAS) that investigated the pro- ulation Program (GIMP, v2.9.8), to generate an objective portion of black in black and white Holstein cows, and measurement of the proportion of white color. Te free- reported an additional signal on chromosome 8, which hand tool was used to trace each animal and remove the carries PAX5 i.e. another potential candidate gene for this background. Te pixel count from the remaining image, Jivanji et al.
Recommended publications
  • FARSB Antibody (C-Term) Affinity Purified Rabbit Polyclonal Antibody (Pab) Catalog # Ap14090b
    10320 Camino Santa Fe, Suite G San Diego, CA 92121 Tel: 858.875.1900 Fax: 858.622.0609 FARSB Antibody (C-term) Affinity Purified Rabbit Polyclonal Antibody (Pab) Catalog # AP14090b Specification FARSB Antibody (C-term) - Product Information Application WB, IHC-P,E Primary Accession Q9NSD9 Other Accession NP_005678.3 Reactivity Human Host Rabbit Clonality Polyclonal Isotype Rabbit Ig Calculated MW 66116 Antigen Region 535-564 FARSB Antibody (C-term) - Additional Information Gene ID 10056 FARSB Antibody (C-term) (Cat. #AP14090b) western blot analysis in Y79 cell line lysates Other Names (35ug/lane).This demonstrates the FARSB Phenylalanine--tRNA ligase beta subunit, antibody detected the FARSB protein (arrow). Phenylalanyl-tRNA synthetase beta subunit, PheRS, FARSB, FARSLB, FRSB Target/Specificity This FARSB antibody is generated from rabbits immunized with a KLH conjugated synthetic peptide between 535-564 amino acids from the C-terminal region of human FARSB. Dilution WB~~1:1000 IHC-P~~1:10~50 Format Purified polyclonal antibody supplied in PBS with 0.09% (W/V) sodium azide. This antibody is purified through a protein A column, followed by peptide affinity purification. FARSB Antibody (C-term) (Cat. #AP14090b)immunohistochemistry analysis Storage in formalin fixed and paraffin embedded Maintain refrigerated at 2-8°C for up to 2 human cerebellum tissue followed by weeks. For long term storage store at -20°C peroxidase conjugation of the secondary in small aliquots to prevent freeze-thaw antibody and DAB staining.This data cycles. demonstrates the use of ANKS1B FARSB Antibody (C-term) for immunohistochemistry. Precautions Clinical relevance has not been evaluated. FARSB Antibody (C-term) is for research use Page 1/2 10320 Camino Santa Fe, Suite G San Diego, CA 92121 Tel: 858.875.1900 Fax: 858.622.0609 only and not for use in diagnostic or FARSB Antibody (C-term) - Background therapeutic procedures.
    [Show full text]
  • Natural Selection Has Shaped Coding and Non-Coding Transcription in Primate CD4+ T-Cells 2 3 Charles G
    bioRxiv preprint doi: https://doi.org/10.1101/083212; this version posted October 25, 2016. 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 Natural Selection has Shaped Coding and Non-coding Transcription in Primate CD4+ T-cells 2 3 Charles G. Danko1,2,*, Zhong Wang1, Edward J. Rice1, Tinyi Chu1,3, Andre L. Martins1, 4 Elia Tait Wojno1,4, John T. Lis5, W. Lee Kraus6,7, & Adam Siepel8,* 5 6 1 Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853. 7 2 Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853. 8 3 Graduate field of Computational Biology, Cornell University, Ithaca, NY 14853. 9 4 Department of Microbiology & Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853. 10 5 Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853. 11 6 Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, 12 University of Texas Southwestern Medical Center, Dallas, TX 75390. 13 7 Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical 14 Center, Dallas, TX 75390. 15 8 Simons Center for Quantitative Biology, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724. 16 17 * Address correspondence to: 18 Charles G. Danko, Ph.D. Adam Siepel, Ph.D. 19 Baker Institute for Animal Health Simons Center for Quantitative Biology 20 Cornell University Cold Spring Harbor Laboratory 21 Hungerford Hill Rd.
    [Show full text]
  • 1 Supporting Information for a Microrna Network Regulates
    Supporting Information for A microRNA Network Regulates Expression and Biosynthesis of CFTR and CFTR-ΔF508 Shyam Ramachandrana,b, Philip H. Karpc, Peng Jiangc, Lynda S. Ostedgaardc, Amy E. Walza, John T. Fishere, Shaf Keshavjeeh, Kim A. Lennoxi, Ashley M. Jacobii, Scott D. Rosei, Mark A. Behlkei, Michael J. Welshb,c,d,g, Yi Xingb,c,f, Paul B. McCray Jr.a,b,c Author Affiliations: Department of Pediatricsa, Interdisciplinary Program in Geneticsb, Departments of Internal Medicinec, Molecular Physiology and Biophysicsd, Anatomy and Cell Biologye, Biomedical Engineeringf, Howard Hughes Medical Instituteg, Carver College of Medicine, University of Iowa, Iowa City, IA-52242 Division of Thoracic Surgeryh, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Canada-M5G 2C4 Integrated DNA Technologiesi, Coralville, IA-52241 To whom correspondence should be addressed: Email: [email protected] (M.J.W.); yi- [email protected] (Y.X.); Email: [email protected] (P.B.M.) This PDF file includes: Materials and Methods References Fig. S1. miR-138 regulates SIN3A in a dose-dependent and site-specific manner. Fig. S2. miR-138 regulates endogenous SIN3A protein expression. Fig. S3. miR-138 regulates endogenous CFTR protein expression in Calu-3 cells. Fig. S4. miR-138 regulates endogenous CFTR protein expression in primary human airway epithelia. Fig. S5. miR-138 regulates CFTR expression in HeLa cells. Fig. S6. miR-138 regulates CFTR expression in HEK293T cells. Fig. S7. HeLa cells exhibit CFTR channel activity. Fig. S8. miR-138 improves CFTR processing. Fig. S9. miR-138 improves CFTR-ΔF508 processing. Fig. S10. SIN3A inhibition yields partial rescue of Cl- transport in CF epithelia.
    [Show full text]
  • Exercise, Skeletal Muscle and Inflammation
    EEXXEERRCCIISSEE IIMMMMUUNNOOLLOOGGYY RREEVVIIEEWW VOLUME 21 • 2015 The International Society of Exercise and Immunology EXERCISEEXERCISE IMMUNOLOGY IMMUNOLOGY REVIEW REVIEW An official Publication of ISEI and DGSP We gratefully acknowledge and thank for years of sponsoring by the Verein zurK Förderungarsten K derrüg Sportmediziner for the editorial team and all our readers Hinnak Northoff for the editorial team and all our readers EXERCISE VOLUME 21 • 2015 IMMUN OLOGY REVIEW CONTENTS From the Editors 7 Reviews Cytokine expression and secretion by skeletal muscle cells: regulatory mechanisms and exercise effects Jonathan M. Peake, Paul Della Gatta, Katsuhiko Suzuki, David C. Nieman 8 Exercise and inflammation-related epigenetic modifications: focus on DNA methylation. Steven Horsburgh, Paula Robson-Ansley, Rozanne Adams, Carine Smith 26 Exercise, skeletal muscle and inflammation: ARE-binding proteins as key regulators in inflammatory and adaptive networks Thomas Beiter, Miriam Hoene, Frauke Prenzler, Frank C. Mooren, Jürgen M. Steinacker, Cora Weigert, Andreas M. Nieß, Barbara Munz 42 Metabolic signals and innate immune activation in obesity and exercise Robert Ringseis, Klaus Eder, Frank C. Mooren, Karsten Krüger 58 The microbiota: An exercise immunology perspective Stéphane Bermon, Bernardo Petriz, Alma Kajėnienė, Jonato Prestes, Lindy Castell, Octavio L. Franco 70 Understanding graft-versus-host disease. Preliminary findings regarding the effects of exercise in affected patients Carmen Fiuza-Luces, Nuria Garatachea, Richard J. Simpson,
    [Show full text]
  • Downloaded from the App Store and Nucleobase, Nucleotide and Nucleic Acid Metabolism 7 Google Play
    Hoytema van Konijnenburg et al. Orphanet J Rare Dis (2021) 16:170 https://doi.org/10.1186/s13023-021-01727-2 REVIEW Open Access Treatable inherited metabolic disorders causing intellectual disability: 2021 review and digital app Eva M. M. Hoytema van Konijnenburg1†, Saskia B. Wortmann2,3,4†, Marina J. Koelewijn2, Laura A. Tseng1,4, Roderick Houben6, Sylvia Stöckler‑Ipsiroglu5, Carlos R. Ferreira7 and Clara D. M. van Karnebeek1,2,4,8* Abstract Background: The Treatable ID App was created in 2012 as digital tool to improve early recognition and intervention for treatable inherited metabolic disorders (IMDs) presenting with global developmental delay and intellectual disabil‑ ity (collectively ‘treatable IDs’). Our aim is to update the 2012 review on treatable IDs and App to capture the advances made in the identifcation of new IMDs along with increased pathophysiological insights catalyzing therapeutic development and implementation. Methods: Two independent reviewers queried PubMed, OMIM and Orphanet databases to reassess all previously included disorders and therapies and to identify all reports on Treatable IDs published between 2012 and 2021. These were included if listed in the International Classifcation of IMDs (ICIMD) and presenting with ID as a major feature, and if published evidence for a therapeutic intervention improving ID primary and/or secondary outcomes is avail‑ able. Data on clinical symptoms, diagnostic testing, treatment strategies, efects on outcomes, and evidence levels were extracted and evaluated by the reviewers and external experts. The generated knowledge was translated into a diagnostic algorithm and updated version of the App with novel features. Results: Our review identifed 116 treatable IDs (139 genes), of which 44 newly identifed, belonging to 17 ICIMD categories.
    [Show full text]
  • © 2016 Hillary Stires ALL RIGHTS RESERVED
    © 2016 Hillary Stires ALL RIGHTS RESERVED ELUCIDATING MECHANISMS THAT DRIVE INCREASED TUMORIGENESIS IN RATS EXPOSED TO ALCOHOL IN UTERO By HILLARY STIRES A dissertation submitted to the Graduate School – New Brunswick Rutgers, The State University of New Jersey in partial fulfillment of the requirements for the degree of Doctor of Philosophy Graduate Program in Endocrinology and Animal Biosciences written under the direction of Dr. Wendie S. Cohick and approved by ______________________________ ______________________________ ______________________________ ______________________________ New Brunswick, New Jersey May 2016 ABSTRACT OF THE DISSERTATION Elucidating Mechanisms That Drive Increased Tumorigenesis in Rats Exposed to Alcohol in Utero By HILLARY STIRES Dissertation Director: Dr. Wendie S. Cohick Alcohol exposure during gestation increases breast cancer risk in offspring. While the mechanisms that underlie this effect are not fully understood, serum estradiol (E2) is increased in these animals during proestrus, suggesting a role for the estrogen axis. To test this hypothesis, it was necessary to develop a stress-free method of hormone administration that could be used in long-term studies of carcinogen exposure to ovariectomized (OVX) rats. Rats were OVX on post- natal day (PND) 40 then treated with daily peroral E2 with or without P4 by adding hormones to peanut butter. On PND 50 rats were injected with nitrosomethylurea (NMU) to induce mammary tumor development. After 26 weeks, there was no difference in tumor incidence suggesting that E2 alone at a normal physiological level can result in tumor development. These results indicate that this may be a useful method to examine the mechanisms of steroid action in mammary tumorigenesis. In addition to changes in circulating E2, local estrogen signaling may be altered in the mammary gland in response to alcohol in utero.
    [Show full text]
  • Hybrid Versus Autochthonous Turkey Populations: Homozygous Genomic Regions Occurrences Due to Artificial and Natural Selection
    animals Article Hybrid Versus Autochthonous Turkey Populations: Homozygous Genomic Regions Occurrences Due to Artificial and Natural Selection Maria Giuseppina Strillacci 1,* , Stefano Paolo Marelli 1 and Guillermo Martinez-Velazquez 2 1 Department of Veterinary Medicine, University of Milan, Via Festa del Perdono, 7, 20122 Milano, Italy; [email protected] 2 INIFAP-Campo Experimental Santiago Ixcuintla, Apdo. Postal 100, Santiago Ixcuintla 63300, Mexico; [email protected] * Correspondence: [email protected]; Tel.: +39-025-033-4582 Received: 15 June 2020; Accepted: 28 July 2020; Published: 30 July 2020 Simple Summary: In this study we investigate the genomic differentiation of traditional Mexican turkey breeds and commercial hybrid strains. The analysis aimed to identify the effects of different types of selection on the birds’ genome structure. Mexican turkeys are characterized by an adaptive selection to their specific original environment; on the other hand, commercial hybrid strains are directionally selected to maximize productive traits and to reduce production costs. The Mexican turkeys were grouped in two geographic subpopulations, while high genomic homogeneity was found in hybrid birds. Traditional breeds and commercial strains are clearly differentiated from a genetic point of view. Inbreeding coefficients were here calculated with different approaches. A clear effect of selection for productive traits was recorded. Abstract: The Mexican turkey population is considered to be the descendant of the original domesticated wild turkey and it is distinct from hybrid strains obtained by the intense artificial selection activity that has occurred during the last 40 years. In this study 30 Mexican turkeys were genomically compared to 38 commercial hybrids using 327,342 SNP markers in order to elucidate the differences in genome variability resulting from different types of selection, i.e., only adaptive for Mexican turkey, and strongly directional for hybrids.
    [Show full text]
  • Genome-Wide CRISPR-Cas9 Screens Reveal Loss of Redundancy Between PKMYT1 and WEE1 in Glioblastoma Stem-Like Cells
    Article Genome-wide CRISPR-Cas9 Screens Reveal Loss of Redundancy between PKMYT1 and WEE1 in Glioblastoma Stem-like Cells Graphical Abstract Authors Chad M. Toledo, Yu Ding, Pia Hoellerbauer, ..., Bruce E. Clurman, James M. Olson, Patrick J. Paddison Correspondence [email protected] (J.M.O.), [email protected] (P.J.P.) In Brief Patient-derived glioblastoma stem-like cells (GSCs) can be grown in conditions that preserve patient tumor signatures and their tumor initiating capacity. Toledo et al. use these conditions to perform genome-wide CRISPR-Cas9 lethality screens in both GSCs and non- transformed NSCs, revealing PKMYT1 as a candidate GSC-lethal gene. Highlights d CRISPR-Cas9 lethality screens performed in patient brain- tumor stem-like cells d PKMYT1 is identified in GSCs, but not NSCs, as essential for facilitating mitosis d PKMYT1 and WEE1 act redundantly in NSCs, where their inhibition is synthetic lethal d PKMYT1 and WEE1 redundancy can be broken by over- activation of EGFR and AKT Toledo et al., 2015, Cell Reports 13, 2425–2439 December 22, 2015 ª2015 The Authors http://dx.doi.org/10.1016/j.celrep.2015.11.021 Cell Reports Article Genome-wide CRISPR-Cas9 Screens Reveal Loss of Redundancy between PKMYT1 and WEE1 in Glioblastoma Stem-like Cells Chad M. Toledo,1,2,14 Yu Ding,1,14 Pia Hoellerbauer,1,2 Ryan J. Davis,1,2,3 Ryan Basom,4 Emily J. Girard,3 Eunjee Lee,5 Philip Corrin,1 Traver Hart,6,7 Hamid Bolouri,1 Jerry Davison,4 Qing Zhang,4 Justin Hardcastle,1 Bruce J. Aronow,8 Christopher L.
    [Show full text]
  • Primate Skeletal Epigenetics
    Primate Skeletal Epigenetics: Evolutionary Implications of DNA Methylation Patterns in the Skeletal Tissues of Human and Nonhuman Primates by Genevieve Housman A Dissertation Presented in Partial Fulfillment of the Requirements for the Degree Doctor of Philosophy Approved April 2017 by the Graduate Supervisory Committee: Anne Stone, Chair Ellen Quillen Kenro Kusumi Christopher Stojanowski ARIZONA STATE UNIVERSITY May 2017 ABSTRACT Within the primate lineage, skeletal traits that contribute to inter-specific anatomical variation and enable varied niche occupations and forms of locomotion are often described as the result of environmental adaptations. However, skeletal phenotypes are more accurately defined as complex traits, and environmental, genetic, and epigenetic mechanisms, such as DNA methylation which regulates gene expression, all contribute to these phenotypes. Nevertheless, skeletal complexity in relation to epigenetic variation has not been assessed across the primate order. In order to gain a complete understanding of the evolution of skeletal phenotypes across primates, it is necessary to study skeletal epigenetics in primates. This study attempts to fill this gap by identifying intra- and inter- specific variation in primate skeletal tissue methylation in order to test whether specific features of skeletal form are related to specific variations in methylation. Specifically, methylation arrays and gene-specific methylation sequencing are used to identify DNA methylation patterns in femoral trabecular bone and cartilage of several nonhuman primate species. Samples include baboons (Papio spp.), macaques (Macaca mulatta), vervets (Chlorocebus aethiops), chimpanzees (Pan troglodytes), and marmosets (Callithrix jacchus), and the efficiencies of these methods are validated in each taxon. Within one nonhuman primate species (baboons), intra-specific variations in methylation patterns are identified across a range of comparative levels, including skeletal tissue differences (bone vs.
    [Show full text]
  • Variation in the TLR10/TLR1/TLR6 Locus Is the Major Genetic Determinant of Interindividual Difference in TLR1/2-Mediated Responses
    Genes and Immunity (2013) 14, 52–57 & 2013 Macmillan Publishers Limited All rights reserved 1466-4879/13 www.nature.com/gene ORIGINAL ARTICLE Variation in the TLR10/TLR1/TLR6 locus is the major genetic determinant of interindividual difference in TLR1/2-mediated responses C Mikacenic1, AP Reiner2, TD Holden1, DA Nickerson3 and MM Wurfel1 Toll-like receptor (TLR)-mediated innate immune responses are important in early host defense. Using a candidate gene approach, we previously identified genetic variation within TLR1 that is associated with hyper-responsiveness to a TLR1/2 agonist in vitro and with death and organ dysfunction in patients with sepsis. Here we report a genome-wide association study (GWAS) designed to identify genetic loci controlling whole blood cytokine responses to the TLR1/2 lipopeptide agonist, Pam3CSK4 (N-palmitoyl-S- À 27 dipalmitoylglyceryl Cys-Ser-(Lys)4) ex vivo. We identified a very strong association (Po1 Â 10 ) between genetic variation within the TLR10/1/6 locus on chromosome 4, and Pam3CSK4-induced cytokine responses. This was the predominant association explaining over 35% of the population variance for this phenotype. Notably, strong associations were observed within TLR10, suggesting that genetic variation in TLR10 may influence bacterial lipoprotein-induced responses. These findings establish the TLR10/1/6 locus as the dominant common genetic factor controlling interindividual variability in Pam3CSK4-induced whole blood responses in the healthy population. Genes and Immunity (2013) 14, 52–57; doi:10.1038/gene.2012.53; published online 15 November 2012 Keywords: TLR; polymorphism; genomics; innate immunity INTRODUCTION variability is attributable to common genetic variants.
    [Show full text]
  • A Rare Variant of ANK3 Is Associated with Intracranial Aneurysm
    ORIGINAL RESEARCH published: 25 June 2021 doi: 10.3389/fneur.2021.672570 A Rare Variant of ANK3 Is Associated With Intracranial Aneurysm Junyu Liu 1, Xin Liao 2,3, Jilin Zhou 1, Bingyang Li 2, Lu Xu 1, Songlin Liu 1, Yifeng Li 1, Dun Yuan 1, Chongyu Hu 4, Weixi Jiang 1* and Junxia Yan 2,5* 1 Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China, 2 Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China, 3 The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China, 4 Department of Neurology, Hunan People’s Hospital, Changsha, China, 5 Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, China Intracranial aneurysm (IA) is a cerebrovascular disorder in which abnormal dilation of a blood vessel results from weakening of the blood vessel wall. The aneurysm may rupture, leading to subarachnoid hemorrhage with severe outcomes. This study was conducted to identify the genetic factors involved in the etiology of IA. Whole-exome sequencing was performed in three IA-aggregate families to identify candidate variants. Further association studies of candidate variants were performed among sporadic cases Edited by: and controls. Bioinformatic analysis was used to predict the functions of candidate Osama O. Zaidat, genes and variants. Twenty variants were identified after whole-exome sequencing, Northeast Ohio Medical University, among which eight were selected for replicative association studies. ANK3 c.4403G>A United States (p.R1468H) was significantly associated with IA (odds ratio 4.77; 95% confidence interval Reviewed by: Basil Erwin Grüter, 1.94–11.67; p-value = 0.00019).
    [Show full text]
  • The Binding Sites of Mir-619-5P in the Mrnas Of
    Atambayeva et al. BMC Genomics (2017) 18:428 DOI 10.1186/s12864-017-3811-6 RESEARCH ARTICLE Open Access The Binding Sites of miR-619-5p in the mRNAs of Human and Orthologous Genes Shara Atambayeva1*, Raigul Niyazova1, Anatoliy Ivashchenko1, Anna Pyrkova1, Ilya Pinsky1, Aigul Akimniyazova1 and Siegfried Labeit2 Abstract Background: Normally, one miRNA interacts with the mRNA of one gene. However, there are miRNAs that can bind to many mRNAs, and one mRNA can be the target of many miRNAs. This significantly complicates the study of the properties of miRNAs and their diagnostic and medical applications. Results: The search of 2,750 human microRNAs (miRNAs) binding sites in 12,175 mRNAs of human genes using the MirTarget program has been completed. For the binding sites of the miR-619-5p the hybridization free energy of the bonds was equal to 100% of the maximum potential free energy. The mRNAs of 201 human genes have complete complementary binding sites of miR-619-5p in the 3’UTR (214 sites), CDS (3 sites), and 5’UTR (4 sites). The mRNAs of CATAD1, ICA1L, GK5, POLH, and PRR11 genes have six miR-619-5p binding sites, and the mRNAs of OPA3 and CYP20A1 genes have eight and ten binding sites, respectively. All of these miR-619-5p binding sites are located in the 3’UTRs. The miR-619-5p binding site in the 5’UTR of mRNA of human USP29 gene is found in the mRNAs of orthologous genes of primates. Binding sites of miR-619-5p in the coding regions of mRNAs of C8H8orf44, C8orf44, and ISY1 genes encode the WLMPVIP oligopeptide, which is present in the orthologous proteins.
    [Show full text]