The Genetics of Physiological Dispersion in Signs of Diabetes Using Murine Models

Total Page:16

File Type:pdf, Size:1020Kb

The Genetics of Physiological Dispersion in Signs of Diabetes Using Murine Models The Genetics of Physiological Dispersion in Signs of Diabetes Using Murine Models Dayna Brown Department of Biology University of Prince Edward Island Charlottetown, PEI, Canada A thesis submitted in partial fulfilment of the requirements of the Honours Programme in the Department of Biology This Thesis is ___________________________________ Accepted Dean of Science University of Prince Edward Island May 2018 ii ABSTRACT Diabetes is a collection of diseases that affects hundreds of millions of people, with type 2 (T2D) being the most prevalent. The heritability of T2D has yet to be explained, despite many studies. This study aims to determine a piece of the missing heritability by using the newly emerging phenomenon of ‘phenotypic dispersion’ (PD) which refers to genes affecting not only mean effects but also the residual variance within genotypes. The association of loci to residual variance of traits is the method used to determine genes that may contribute to T2D development. Eight in silico mouse data sets were analyzed to test for relationships between mapped microsatellite markers and the PD of diabetic plasma variables (cholesterol, high and low-density lipoprotein, and triglyceride). 29 loci were found to be significantly associated with PD of traits within the data sets. Single nucleotide polymorphisms (SNPs) in genes nearby the associated loci were identified, with Fto, Apoa2, Foxa2, Bpifb6, and Apt1a4 being the most notable genes involved. The association between genotype and PD at all associated loci showed that when the genotype was dominant that it was dominant for low dispersion, which is suggestive of a genetic mechanism controlling the dispersion of traits. These findings suggest that there may be genes that influence the dispersion of traits associated with developing T2D. iii ACKNOWLEDGEMENTS I would like to acknowledge my co-supervisors, Dr. Guy Perry for his continued support and effort throughout the duration of my honours, and Dr. Larry Hale for his guidance and assistance. I truly appreciate the skills and abilities I have gained conducting research under their supervision. My father, Stephen Brown, and my grandmother, Rowena Brown, deserve significant recognition for their unconditional encouragement and support in my education. I am eternally grateful to them for all that they have done for me. I would also like to thank the Biology Department for allowing me to do this honours research, and for making my undergraduate degree the great experience that it was. iv Table of Contents Abstract................................................................................................................................ii Acknowledgments...............................................................................................................iii Table of Contents................................................................................................................iv List of Tables.......................................................................................................................vi List of Figures.....................................................................................................................ix Introduction and Literature Review................................................................................1 Diabetes as a Disease...............................................................................................1 Genetics of Diabetes.................................................................................................2 Materials and Methods......................................................................................................6 In silico data.............................................................................................................7 Ishimori et al. 2004......................................................................................7 Kim et al. 2001............................................................................................8 Li et al. 2008..............................................................................................10 Smith Richards et al. 2002.........................................................................11 Stylianou et al. 2006b.................................................................................13 Stylianou et al. 2006c-e..............................................................................13 Comparison Between Studies.....................................................................15 Statistical Analysis.................................................................................................15 Genomic SNP Targets............................................................................................17 Results................................................................................................................................19 Covariates...............................................................................................................19 Qualitative and Limited Dependent Variable Model.............................................26 Ishimori et al. 2004....................................................................................26 Kim et al. 2001...........................................................................................36 Li et al. 2008..............................................................................................36 Smith Richards et al. 2002.........................................................................40 v Table of Contents Continued Stylianou et al. 2006b.................................................................................40 Stylianou et al. 2006c.................................................................................47 Stylianou et al. 2006d.................................................................................47 Stylianou et al. 20006e...............................................................................47 Gene Candidates.....................................................................................................50 Ishimori et al. 2004....................................................................................50 Li et al. 2008..............................................................................................59 Smith Richards et al. 2002.........................................................................59 Stylianou et al. 2006b.................................................................................68 Discussion........................................................................................................................100 Summary..............................................................................................................104 Literature Cited.................................................................................................................105 vi List of Tables Table 1. Association of multi-locus heterozygosity (MLH) with original and dispersed plasma traits (plasma high-density lipoprotein (pHDL; mg/dl), plasma non-HDL (pnonHDL; mg/dl), plasma cholesterol (pChol; mg/dl) and plasma triglyceride (pTG; mg/dl) in general linear modeling (GLM) in Ishimori et al. 2004........................................................................................................................20 Table 2. Influence of multi-locus heterozygosity (MLH) on original (general linear modeling, GLM) and phenotypic dispersion (PD) (qualitative and limited dependent modeling; QLIM) analysis in traits from Kim et al. 2001. pGlu = plasma glucose (mg/dl), pIns = plasma insulin (ng/ml).........................................21 Table 3. The effects of multi-locus heterozygosity (MLH) on original and phenotypic dispersion (PD) in plasma cholesterol (pChol; mg/dl), plasma high-density lipoprotein (pHDL; mg/dl) and plasma triglyceride (pTG; mg/dl) in Li et al. 2008........................................................................................................................22 Table 4. The association of multi-locus heterozygosity (MLH), paternal grandmother (pgm), and F1 sire with original traits and phenotypic dispersion (PD) in plasma glucose (pGlu; mg/dl) and plasma triglyceride (pTG; mg/dl) from Smith Richards et al. 2002........................................................................................................................23 Table 5. The influence of multi-locus heterozygosity (MLH) and sex on original phenotypic dispersion (PD) traits (plasma triglyceride (pTG; mg/dl), plasma high- density lipoprotein (pHDL; mg/dl) and plasma cholesterol (pChol; mg/dl)) in Stylianou et al. 2006b.............................................................................................24 Table 6. The association of sex and multi-locus heterozygosity (MLH) original traits and phenotypic dispersion (PD) for plasma cholesterol (pChol; mg/dl), plasma high- density lipoprotein (pHDL; mg/dl), plasma triglyceride (pTG; mg/dl) and plasma nonHDL (pnonHDL; mg/dl) from Stylianou et al. 2006c......................................25 Table 7. The association of sex and multi-locus heterozygosity (MLH) with original and dispersive plasma cholesterol (pChol; mg/dl), plasma triglyceride (pTG; mg/dl), plasma high-density lipoprotein (pHDL; mg/dl) and plasma nonHDL (pnonHDL; mg/dl) from Stylianou et al. 2006d........................................................................27 Table 8. The association of sex and multi-locus heterozygosity (MLH) with
Recommended publications
  • IQSEC2 Antibody Cat
    IQSEC2 Antibody Cat. No.: 8011 Western blot analysis of IQSEC2 in SK-N-SH cell lysate with IQSEC2 antibody at 1 μg/ml. Immunohistochemistry of IQSEC2 in mouse brain tissue with IQSEC2 antibody at 5 μg/ml. Specifications HOST SPECIES: Rabbit SPECIES REACTIVITY: Human, Mouse, Rat IQSEC2 antibody was raised against an 18 amino acid peptide near the carboxy terminus of human IQSEC2. IMMUNOGEN: The immunogen is located within amino acids 1380 - 1430 of IQSEC2. TESTED APPLICATIONS: ELISA, IHC-P, WB IQSEC2 antibody can be used for detection of IQSEC2 by Western blot at 1 - 2 μg/ml. Antibody can also be used for immunohistochemistry starting at 5 μg/mL. APPLICATIONS: Antibody validated: Western Blot in human samples and Immunohistochemistry in mouse samples. All other applications and species not yet tested. September 27, 2021 1 https://www.prosci-inc.com/iqsec2-antibody-8011.html IQSEC2 antibody is human, mouse and rat reactive. At least two isoforms of IQSEC2 are SPECIFICITY: known to exist; this antibody will only detect the larger isoform. IQSEC2 antibody is predicted to not cross-react with IQSEC1. POSITIVE CONTROL: 1) Cat. No. 1220 - SK-N-SH Cell Lysate Predicted: 104, 133, 141, 164 kDa PREDICTED MOLECULAR WEIGHT: Observed: 140 kDa Properties PURIFICATION: IQSEC2 antibody is affinity chromatography purified via peptide column. CLONALITY: Polyclonal ISOTYPE: IgG CONJUGATE: Unconjugated PHYSICAL STATE: Liquid BUFFER: IQSEC2 antibody is supplied in PBS containing 0.02% sodium azide. CONCENTRATION: 1 mg/mL IQSEC2 antibody can be stored at 4˚C for three months and -20˚C, stable for up to one STORAGE CONDITIONS: year.
    [Show full text]
  • Seq2pathway Vignette
    seq2pathway Vignette Bin Wang, Xinan Holly Yang, Arjun Kinstlick May 19, 2021 Contents 1 Abstract 1 2 Package Installation 2 3 runseq2pathway 2 4 Two main functions 3 4.1 seq2gene . .3 4.1.1 seq2gene flowchart . .3 4.1.2 runseq2gene inputs/parameters . .5 4.1.3 runseq2gene outputs . .8 4.2 gene2pathway . 10 4.2.1 gene2pathway flowchart . 11 4.2.2 gene2pathway test inputs/parameters . 11 4.2.3 gene2pathway test outputs . 12 5 Examples 13 5.1 ChIP-seq data analysis . 13 5.1.1 Map ChIP-seq enriched peaks to genes using runseq2gene .................... 13 5.1.2 Discover enriched GO terms using gene2pathway_test with gene scores . 15 5.1.3 Discover enriched GO terms using Fisher's Exact test without gene scores . 17 5.1.4 Add description for genes . 20 5.2 RNA-seq data analysis . 20 6 R environment session 23 1 Abstract Seq2pathway is a novel computational tool to analyze functional gene-sets (including signaling pathways) using variable next-generation sequencing data[1]. Integral to this tool are the \seq2gene" and \gene2pathway" components in series that infer a quantitative pathway-level profile for each sample. The seq2gene function assigns phenotype-associated significance of genomic regions to gene-level scores, where the significance could be p-values of SNPs or point mutations, protein-binding affinity, or transcriptional expression level. The seq2gene function has the feasibility to assign non-exon regions to a range of neighboring genes besides the nearest one, thus facilitating the study of functional non-coding elements[2]. Then the gene2pathway summarizes gene-level measurements to pathway-level scores, comparing the quantity of significance for gene members within a pathway with those outside a pathway.
    [Show full text]
  • DNA Topoisomerases and Cancer Topoisomerases and TOP Genes in Humans Humans Vs
    DNA Topoisomerases Review DNA Topoisomerases And Cancer Topoisomerases and TOP Genes in Humans Humans vs. Escherichia Coli Topoisomerase differences Comparisons Topoisomerase and genomes Top 1 Top1 and Top2 differences Relaxation of DNA Top1 DNA supercoiling DNA supercoiling In the context of chromatin, where the rotation of DNA is constrained, DNA supercoiling (over- and under-twisting and writhe) is readily generated. TOP1 and TOP1mt remove supercoiling by DNA untwisting, acting as “swivelases”, whereas TOP2a and TOP2b remove writhe, acting as “writhases” at DNA crossovers (see TOP2 section). Here are some basic facts concerning DNA supercoiling that are relevant to topoisomerase activity: • Positive supercoiling (Sc+) tightens the DNA helix whereas negative supercoiling (Sc-) facilitates the opening of the duplex and the generation of single-stranded segments. • Nucleosome formation and disassembly absorbs and releases Sc-, respectively. • Polymerases generate Sc+ ahead and Sc- behind their tracks. • Excess of Sc+ arrests DNA tracking enzymes (helicases and polymerases), suppresses transcription elongation and initiation, and destabilizes nucleosomes. • Sc- facilitates DNA melting during the initiation of replication and transcription, D-loop formation and homologous recombination and nucleosome formation. • Excess of Sc- favors the formation of alternative DNA structures (R-loops, guanine quadruplexes, right-handed DNA (Z-DNA), plectonemic structures), which then absorb Sc- upon their formation and attract regulatory proteins. The
    [Show full text]
  • RNA Epigenetics: Fine-Tuning Chromatin Plasticity and Transcriptional Regulation, and the Implications in Human Diseases
    G C A T T A C G G C A T genes Review RNA Epigenetics: Fine-Tuning Chromatin Plasticity and Transcriptional Regulation, and the Implications in Human Diseases Amber Willbanks, Shaun Wood and Jason X. Cheng * Department of Pathology, Hematopathology Section, University of Chicago, Chicago, IL 60637, USA; [email protected] (A.W.); [email protected] (S.W.) * Correspondence: [email protected] Abstract: Chromatin structure plays an essential role in eukaryotic gene expression and cell identity. Traditionally, DNA and histone modifications have been the focus of chromatin regulation; however, recent molecular and imaging studies have revealed an intimate connection between RNA epigenetics and chromatin structure. Accumulating evidence suggests that RNA serves as the interplay between chromatin and the transcription and splicing machineries within the cell. Additionally, epigenetic modifications of nascent RNAs fine-tune these interactions to regulate gene expression at the co- and post-transcriptional levels in normal cell development and human diseases. This review will provide an overview of recent advances in the emerging field of RNA epigenetics, specifically the role of RNA modifications and RNA modifying proteins in chromatin remodeling, transcription activation and RNA processing, as well as translational implications in human diseases. Keywords: 5’ cap (5’ cap); 7-methylguanosine (m7G); R-loops; N6-methyladenosine (m6A); RNA editing; A-to-I; C-to-U; 2’-O-methylation (Nm); 5-methylcytosine (m5C); NOL1/NOP2/sun domain Citation: Willbanks, A.; Wood, S.; (NSUN); MYC Cheng, J.X. RNA Epigenetics: Fine-Tuning Chromatin Plasticity and Transcriptional Regulation, and the Implications in Human Diseases. Genes 2021, 12, 627.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Deoxyguanosine Depurination and Yield DNA–Protein Cross-Links in Nucleosome Core Particles and Cells
    Histone tails decrease N7-methyl-2′-deoxyguanosine depurination and yield DNA–protein cross-links in nucleosome core particles and cells Kun Yanga, Daeyoon Parkb, Natalia Y. Tretyakovac, and Marc M. Greenberga,1 aDepartment of Chemistry, Johns Hopkins University, Baltimore, MD 21218; bDepartment of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Twin Cities, Minneapolis, MN 55417; and cDepartment of Medicinal Chemistry, University of Minnesota, Twin Cities, Minneapolis, MN 55417 Edited by Cynthia J. Burrows, University of Utah, Salt Lake City, UT, and approved October 18, 2018 (received for review August 6, 2018) Monofunctional alkylating agents preferentially react at the adducts. DPC formation between histone proteins and MdG was N7 position of 2′-deoxyguanosine in duplex DNA. Methylated also observed in MMS-treated V79 Chinese hamster lung cells. DNA, such as that produced by methyl methanesulfonate (MMS) Intracellular DPC formation from MdG could have significant and temozolomide, exists for days in organisms. The predominant biological effects. consequence of N7-methyl-2′-deoxyguanosine (MdG) is widely be- Compared with other DNA lesions, MdG minimally perturbs lieved to be abasic site (AP) formation via hydrolysis, a process duplex structure (12). The steric effects of N7-methylation within that is slow in free DNA. Examination of MdG reactivity within the major groove are minimal, and the aromatic planar structure nucleosome core particles (NCPs) provided two general observa- is maintained. Although the Watson–Crick hydrogen bonding tions. MdG depurination rate constants are reduced in NCPs com- pattern of dG is retained, recent structural studies raise the pared with when the identical DNA sequence is free in solution.
    [Show full text]
  • Integrative Differential Expression and Gene Set Enrichment Analysis Using Summary Statistics for Scrna-Seq Studies
    ARTICLE https://doi.org/10.1038/s41467-020-15298-6 OPEN Integrative differential expression and gene set enrichment analysis using summary statistics for scRNA-seq studies ✉ Ying Ma 1,7, Shiquan Sun 1,7, Xuequn Shang2, Evan T. Keller 3, Mengjie Chen 4,5 & Xiang Zhou 1,6 Differential expression (DE) analysis and gene set enrichment (GSE) analysis are commonly applied in single cell RNA sequencing (scRNA-seq) studies. Here, we develop an integrative 1234567890():,; and scalable computational method, iDEA, to perform joint DE and GSE analysis through a hierarchical Bayesian framework. By integrating DE and GSE analyses, iDEA can improve the power and consistency of DE analysis and the accuracy of GSE analysis. Importantly, iDEA uses only DE summary statistics as input, enabling effective data modeling through com- plementing and pairing with various existing DE methods. We illustrate the benefits of iDEA with extensive simulations. We also apply iDEA to analyze three scRNA-seq data sets, where iDEA achieves up to five-fold power gain over existing GSE methods and up to 64% power gain over existing DE methods. The power gain brought by iDEA allows us to identify many pathways that would not be identified by existing approaches in these data. 1 Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA. 2 School of Computer Science, Northwestern Polytechnical University, Xi’an, Shaanxi 710072, P.R. China. 3 Department of Urology, University of Michigan, Ann Arbor, MI 48109, USA. 4 Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA. 5 Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA.
    [Show full text]
  • Integrating Single-Step GWAS and Bipartite Networks Reconstruction Provides Novel Insights Into Yearling Weight and Carcass Traits in Hanwoo Beef Cattle
    animals Article Integrating Single-Step GWAS and Bipartite Networks Reconstruction Provides Novel Insights into Yearling Weight and Carcass Traits in Hanwoo Beef Cattle Masoumeh Naserkheil 1 , Abolfazl Bahrami 1 , Deukhwan Lee 2,* and Hossein Mehrban 3 1 Department of Animal Science, University College of Agriculture and Natural Resources, University of Tehran, Karaj 77871-31587, Iran; [email protected] (M.N.); [email protected] (A.B.) 2 Department of Animal Life and Environment Sciences, Hankyong National University, Jungang-ro 327, Anseong-si, Gyeonggi-do 17579, Korea 3 Department of Animal Science, Shahrekord University, Shahrekord 88186-34141, Iran; [email protected] * Correspondence: [email protected]; Tel.: +82-31-670-5091 Received: 25 August 2020; Accepted: 6 October 2020; Published: 9 October 2020 Simple Summary: Hanwoo is an indigenous cattle breed in Korea and popular for meat production owing to its rapid growth and high-quality meat. Its yearling weight and carcass traits (backfat thickness, carcass weight, eye muscle area, and marbling score) are economically important for the selection of young and proven bulls. In recent decades, the advent of high throughput genotyping technologies has made it possible to perform genome-wide association studies (GWAS) for the detection of genomic regions associated with traits of economic interest in different species. In this study, we conducted a weighted single-step genome-wide association study which combines all genotypes, phenotypes and pedigree data in one step (ssGBLUP). It allows for the use of all SNPs simultaneously along with all phenotypes from genotyped and ungenotyped animals. Our results revealed 33 relevant genomic regions related to the traits of interest.
    [Show full text]
  • Genetic and Genomic Analysis of Hyperlipidemia, Obesity and Diabetes Using (C57BL/6J × TALLYHO/Jngj) F2 Mice
    University of Tennessee, Knoxville TRACE: Tennessee Research and Creative Exchange Nutrition Publications and Other Works Nutrition 12-19-2010 Genetic and genomic analysis of hyperlipidemia, obesity and diabetes using (C57BL/6J × TALLYHO/JngJ) F2 mice Taryn P. Stewart Marshall University Hyoung Y. Kim University of Tennessee - Knoxville, [email protected] Arnold M. Saxton University of Tennessee - Knoxville, [email protected] Jung H. Kim Marshall University Follow this and additional works at: https://trace.tennessee.edu/utk_nutrpubs Part of the Animal Sciences Commons, and the Nutrition Commons Recommended Citation BMC Genomics 2010, 11:713 doi:10.1186/1471-2164-11-713 This Article is brought to you for free and open access by the Nutrition at TRACE: Tennessee Research and Creative Exchange. It has been accepted for inclusion in Nutrition Publications and Other Works by an authorized administrator of TRACE: Tennessee Research and Creative Exchange. For more information, please contact [email protected]. Stewart et al. BMC Genomics 2010, 11:713 http://www.biomedcentral.com/1471-2164/11/713 RESEARCH ARTICLE Open Access Genetic and genomic analysis of hyperlipidemia, obesity and diabetes using (C57BL/6J × TALLYHO/JngJ) F2 mice Taryn P Stewart1, Hyoung Yon Kim2, Arnold M Saxton3, Jung Han Kim1* Abstract Background: Type 2 diabetes (T2D) is the most common form of diabetes in humans and is closely associated with dyslipidemia and obesity that magnifies the mortality and morbidity related to T2D. The genetic contribution to human T2D and related metabolic disorders is evident, and mostly follows polygenic inheritance. The TALLYHO/ JngJ (TH) mice are a polygenic model for T2D characterized by obesity, hyperinsulinemia, impaired glucose uptake and tolerance, hyperlipidemia, and hyperglycemia.
    [Show full text]
  • Supplementary Table 1: Adhesion Genes Data Set
    Supplementary Table 1: Adhesion genes data set PROBE Entrez Gene ID Celera Gene ID Gene_Symbol Gene_Name 160832 1 hCG201364.3 A1BG alpha-1-B glycoprotein 223658 1 hCG201364.3 A1BG alpha-1-B glycoprotein 212988 102 hCG40040.3 ADAM10 ADAM metallopeptidase domain 10 133411 4185 hCG28232.2 ADAM11 ADAM metallopeptidase domain 11 110695 8038 hCG40937.4 ADAM12 ADAM metallopeptidase domain 12 (meltrin alpha) 195222 8038 hCG40937.4 ADAM12 ADAM metallopeptidase domain 12 (meltrin alpha) 165344 8751 hCG20021.3 ADAM15 ADAM metallopeptidase domain 15 (metargidin) 189065 6868 null ADAM17 ADAM metallopeptidase domain 17 (tumor necrosis factor, alpha, converting enzyme) 108119 8728 hCG15398.4 ADAM19 ADAM metallopeptidase domain 19 (meltrin beta) 117763 8748 hCG20675.3 ADAM20 ADAM metallopeptidase domain 20 126448 8747 hCG1785634.2 ADAM21 ADAM metallopeptidase domain 21 208981 8747 hCG1785634.2|hCG2042897 ADAM21 ADAM metallopeptidase domain 21 180903 53616 hCG17212.4 ADAM22 ADAM metallopeptidase domain 22 177272 8745 hCG1811623.1 ADAM23 ADAM metallopeptidase domain 23 102384 10863 hCG1818505.1 ADAM28 ADAM metallopeptidase domain 28 119968 11086 hCG1786734.2 ADAM29 ADAM metallopeptidase domain 29 205542 11085 hCG1997196.1 ADAM30 ADAM metallopeptidase domain 30 148417 80332 hCG39255.4 ADAM33 ADAM metallopeptidase domain 33 140492 8756 hCG1789002.2 ADAM7 ADAM metallopeptidase domain 7 122603 101 hCG1816947.1 ADAM8 ADAM metallopeptidase domain 8 183965 8754 hCG1996391 ADAM9 ADAM metallopeptidase domain 9 (meltrin gamma) 129974 27299 hCG15447.3 ADAMDEC1 ADAM-like,
    [Show full text]
  • Análise Integrativa De Perfis Transcricionais De Pacientes Com
    UNIVERSIDADE DE SÃO PAULO FACULDADE DE MEDICINA DE RIBEIRÃO PRETO PROGRAMA DE PÓS-GRADUAÇÃO EM GENÉTICA ADRIANE FEIJÓ EVANGELISTA Análise integrativa de perfis transcricionais de pacientes com diabetes mellitus tipo 1, tipo 2 e gestacional, comparando-os com manifestações demográficas, clínicas, laboratoriais, fisiopatológicas e terapêuticas Ribeirão Preto – 2012 ADRIANE FEIJÓ EVANGELISTA Análise integrativa de perfis transcricionais de pacientes com diabetes mellitus tipo 1, tipo 2 e gestacional, comparando-os com manifestações demográficas, clínicas, laboratoriais, fisiopatológicas e terapêuticas Tese apresentada à Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo para obtenção do título de Doutor em Ciências. Área de Concentração: Genética Orientador: Prof. Dr. Eduardo Antonio Donadi Co-orientador: Prof. Dr. Geraldo A. S. Passos Ribeirão Preto – 2012 AUTORIZO A REPRODUÇÃO E DIVULGAÇÃO TOTAL OU PARCIAL DESTE TRABALHO, POR QUALQUER MEIO CONVENCIONAL OU ELETRÔNICO, PARA FINS DE ESTUDO E PESQUISA, DESDE QUE CITADA A FONTE. FICHA CATALOGRÁFICA Evangelista, Adriane Feijó Análise integrativa de perfis transcricionais de pacientes com diabetes mellitus tipo 1, tipo 2 e gestacional, comparando-os com manifestações demográficas, clínicas, laboratoriais, fisiopatológicas e terapêuticas. Ribeirão Preto, 2012 192p. Tese de Doutorado apresentada à Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo. Área de Concentração: Genética. Orientador: Donadi, Eduardo Antonio Co-orientador: Passos, Geraldo A. 1. Expressão gênica – microarrays 2. Análise bioinformática por module maps 3. Diabetes mellitus tipo 1 4. Diabetes mellitus tipo 2 5. Diabetes mellitus gestacional FOLHA DE APROVAÇÃO ADRIANE FEIJÓ EVANGELISTA Análise integrativa de perfis transcricionais de pacientes com diabetes mellitus tipo 1, tipo 2 e gestacional, comparando-os com manifestações demográficas, clínicas, laboratoriais, fisiopatológicas e terapêuticas.
    [Show full text]
  • Binding Specificities of Human RNA Binding Proteins Towards Structured
    bioRxiv preprint doi: https://doi.org/10.1101/317909; this version posted March 1, 2019. 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. 1 Binding specificities of human RNA binding proteins towards structured and linear 2 RNA sequences 3 4 Arttu Jolma1,#, Jilin Zhang1,#, Estefania Mondragón4,#, Teemu Kivioja2, Yimeng Yin1, 5 Fangjie Zhu1, Quaid Morris5,6,7,8, Timothy R. Hughes5,6, Louis James Maher III4 and Jussi 6 Taipale1,2,3,* 7 8 9 AUTHOR AFFILIATIONS 10 11 1Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden 12 2Genome-Scale Biology Program, University of Helsinki, Helsinki, Finland 13 3Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom 14 4Department of Biochemistry and Molecular Biology and Mayo Clinic Graduate School of 15 Biomedical Sciences, Mayo Clinic College of Medicine and Science, Rochester, USA 16 5Department of Molecular Genetics, University of Toronto, Toronto, Canada 17 6Donnelly Centre, University of Toronto, Toronto, Canada 18 7Edward S Rogers Sr Department of Electrical and Computer Engineering, University of 19 Toronto, Toronto, Canada 20 8Department of Computer Science, University of Toronto, Toronto, Canada 21 #Authors contributed equally 22 *Correspondence: [email protected] 23 24 25 SUMMARY 26 27 Sequence specific RNA-binding proteins (RBPs) control many important 28 processes affecting gene expression. They regulate RNA metabolism at multiple 29 levels, by affecting splicing of nascent transcripts, RNA folding, base modification, 30 transport, localization, translation and stability. Despite their central role in most 31 aspects of RNA metabolism and function, most RBP binding specificities remain 32 unknown or incompletely defined.
    [Show full text]