Chapter 1 Introduction

Total Page:16

File Type:pdf, Size:1020Kb

Chapter 1 Introduction University of Southampton Human Development and Health Faculty of Medicine CDKN2A: A Predictive Marker for Bone Mineral Density or Direct Role in Osteoblast Function? Eloïse Cook PhD Thesis July 2016 Supervisors: Prof Karen Lillycrop, Prof Nicholas Harvey and Dr Stuart Lanham UNIVERSITY OF SOUTHAMPTON ABSTRACT FACULTY OF MEDICINE ACADEMIC UNIT OF HUMAN DEVELOPMENT AND HEALTH Doctor of Philosophy CDKN2A: A Predictive Marker for Bone Mineral Density or Direct Role in Osteoblast Function? By Eloïse Cook As the population is becoming more aged, osteoporosis is becoming more prevalent and the number of fragility fractures that are occurring is increasing. One of the main predictors of developing osteoporosis in later life is a bone mineral density that is greater than 2.5 standard deviations below the young adult sex-matched mean, though studies have only been able to explain 5% of the variance seen in bone mineral densities between individuals. There is now increasing evidence that the development of osteoporosis can begin in utero and that epigenetic processes, such as DNA methylation, may be central to the mechanism by which early development influences bone mineral density and later bone health. Previous work within the group has identified associations of a 300bp differentially methylated region within the CDKN2A locus with bone mineral content, bone area and areal bone mineral density in offspring from the Southampton Women’s Survey (SWS) cohort. As methylation of CDKN2A increases, bone mineral content, bone area and areal bone mineral density all decrease at both 4 and 6 years of age. The two cell cycle regulators p16INK4A and p14ARF are transcribed from this gene as well as the lncRNA ANRIL. Various techniques were used to determine whether this differentially methylated region has a functional role in osteoblasts or whether it is just a potential marker for predicting bone mineral density. It was shown that there was specific binding in the osteoblast-like SaOS-2 cell line to this region and that methylation of these CpGs led to a decrease in transcription factor binding. Treatment of both SaOS-2 cells and primary osteoblasts with siRNAs against ANRIL caused a decrease in cell number, a decrease in cell proliferation, an increase in cells in G0/G1 phase of the cell cycle and, in SaOS-2 cells, caused an increase in the expression of bone differentiation markers RUNX2 and ALP. Knock down of ANRIL expression also caused a change in genes that have roles in osteoblast specific pathways as well as genes that were involved in molecular functions such as nuclear regulation and acetylation. Associations were found between CpG methylation in RXRA and offspring bone outcomes at birth in both the SWS and MAVIDOS cohorts. Daily maternal cholecalciferol supplementation was also shown to decrease the methylation of CpG loci within RXRA in offspring umbilical cord and further investigation showed that this effect of maternal supplementation was only seen in children born during the winter months. Together, these findings suggest that the association between CDKN2A methylation at birth and later bone health could indicate a potential role for CDKN2A in bone development and function and raises the possibility that differential methylation of CDKN2A may allow the identification of individuals at increased risk of osteoporosis in later life. Due to the effect of ANRIL knock down in primary osteoblasts, which could not be explained through normal cell cycle pathways, results suggest that ANRIL has potential trans-regulatory functions in these cells. These findings suggest that ANRIL could potentially play a role in bone differentiation, cell proliferation and metabolism and that dysregulation of the expression of this lncRNA could have adverse effects on bone health, potentially increasing the risk of individuals developing osteoporosis in later life. I Table of Contents Abstract – I Table of contents – II List of figures – VII List of tables – X Declaration of authorship – XI Acknowledgements – XII Abbreviations – XIII Page Number 1.0 Introduction 1 1.1 Osteoporosis 3 1.1.1 Epidemiology of osteoporosis 3 1.1.1.1 Fracture 3 1.1.1.2 Morbidity 4 1.1.1.3 Mortality 5 1.1.1.4 Economic cost 6 1.2 Bone formation 6 1.2.1 Osteoblast differentiation 7 1.2.1.1 Transcription factors 9 1.2.1.2 Signalling pathways 10 1.2.1.3 Systemic regulation 16 1.2.2 Osteoclasts 17 1.2.3 Bone remodelling 18 1.2.4 Genome-wide association studies (GWASs) 19 1.2.5 Pathogenesis of osteoporosis 19 1.3 Developmental origins of health and disease (DOHaD) 21 1.3.1 Human studies 22 1.3.2 Animal studies 24 1.3.3 Evidence for the developmental origins of osteoporosis 25 1.3.3.1 Population studies 25 1.3.1.2 Physiological studies 26 1.3.3.3 Maternal lifestyle 27 1.3.3.4 Childhood growth 27 1.3.4 Vitamin D 28 1.4 Epigenetics 29 1.4.1 Epigenetic mechanisms 30 1.4.1.1 DNA methylation 30 1.4.1.2 Histone modifications 31 1.4.1.3 microRNAs (miRNAs) 31 1.4.1.4 Long non-coding RNAs (lncRNAs) 32 1.4.1.5 Evidence epigenetic processes mediate developmental origins of adult disease using animal models 33 1.4.1.6 Vitamin D and epigenetic mechanisms 33 1.4.2 Epigenetics and early life environment 34 1.4.3 Epigenetics and bone 36 1.5 Identification of potential predictive markers of offspring bone outcomes 38 1.5.1 Epigenetic biomarkers of later health 38 1.5.2 Epigenetic markers of later bone health 39 II 1.5.2.1 RXRα 39 1.5.2.2 Endothelial nitric oxide synthase (eNOS) 40 1.5.2.3 CDKN2A 41 1.6 Aims and hypotheses 43 2.0 Materials and Methods 45 2.1 Materials 47 2.2 Methods 50 2.2.1 Cell culture 50 2.2.1.1 Osteosarcoma SaOS-2 cell line 50 2.2.1.2 Human bone marrow stromal cells (hBMSCs) 50 2.2.2 Transfection of SaOS-2 with luciferase reporter constructs 52 2.2.2.1 ANRIL/p14ARF luciferase reporter constructs 52 2.2.2.2 Inoculation and minipreparation of plasmid DNA 52 2.2.2.3 Preparing plasmid DNA for transfection 53 2.2.2.4 Transfection of plasmid DNA 53 2.2.2.5 Preparation of cells for Dual-Luciferase assay 54 2.2.3 Isolation of nuclear extract 54 2.2.4 Analysis of transcription factor binding by electrophoretic mobility shift assays 54 2.2.4.1 Annealing oligonucleotides 54 2.2.4.2 Electrophoretic mobility shift assay (EMSA) 55 2.2.5 Transfection with siRNA against ANRIL 57 2.2.5.1 SaOS-2 57 2.2.5.2 hBMSCs and primary osteoblasts 59 2.2.6 Isolation of total RNA 59 2.2.6.1 TRI Reagent 59 2.2.6.2 RNeasy kit 60 2.2.7 Illumina Human H12 Gene Expression Array 60 2.2.8 DNase I treatment 60 2.2.9 cDNA synthesis 61 2.2.10 Real time quantitative polymerase chain reaction (RT-qPCR) 61 2.2.10.1 GeNorm assay 61 2.2.10.2 TaqMan assays 61 2.2.11 Fluorescence-activated cell sorting (FACS) 62 2.2.12 Measuring methylation levels of MAVIDOS umbilical cord 63 2.2.12.1 Isolation of genomic DNA from umbilical cord 63 2.2.12.2 Bisulfite conversion of genomic DNA 66 2.2.12.3 Polymerase chain reaction (PCR) 66 2.2.12.4 Pyrosequencing 68 2.2.13 Statistical analysis 68 2.2.13.1 Statistical analysis of siRNA transfected cells 68 2.2.13.2 Statistical analysis of MAVIDOS cohort 68 3.0 Identification of CDKN2A as a predictive marker of bone mineral density 71 3.1 Introduction 73 3.1.1 CDKN2A gene locus 73 3.2 Aims 76 3.3 Results 77 3.3.1 Does the DMR control CDKN2A expression? 77 III 3.3.2 Is there specific binding to the CpGs within the CDKN2A DMR in bone cells? 79 3.3.3 Does the methylation of CpGs -887 and -907 in the CDKN2A DMR alter the binding of transcription factors? 82 3.3.4 What transcription factors bind to CpGs -887 and -907? 85 3.4 Discussion 89 3.5 Summary 93 3.6 Future work 93 4.0 Functional significance of ANRIL in SaOS-2, an osteoblast-like cell line 95 4.1 Introduction 97 4.2 Aims 99 4.3 Results 100 4.3.1 Is ANRIL expressed in bone-related cells? 100 4.3.2 Does ANRIL knock down affect SaOS-2 cell number? 103 4.3.3 Does knock down of ANRIL expression cause cell cycle arrest? 106 4.3.4 Does ANRIL knock down have an effect on osteoblast differentiation? 108 4.3.5 Does ANRIL knock down have an effect on mineralisation? 110 4.3.6 Does ANRIL mediate its affect through altered expression of the CDKN2A/B locus? 111 4.4 Discussion 113 4.5 Summary 118 5.0 Characterisation of ANRIL in human bone marrow stromal cells and primary osteoblasts 121 5.1 Introduction 123 5.2 Aims 123 5.3 Results 124 5.3.1 ANRIL knock down causes a decrease in hBMSC and primary osteoblast cell number 124 5.3.2 ANRIL knock down does not have an effect on the viability of primary cells 129 5.3.3 Knock down of ANRIL expression causes a decrease in the proliferation of primary cells 131 5.3.4 Knock down of ANRIL expression causes cell cycle arrest in hBMSCs and primary osteoblasts 133 5.3.5 Does ANRIL mediate its affect through altered expression of the CDKN2A/B locus in primary cells? 135 5.3.6 hBMSCs and primary osteoblasts treated with siRNAs against ANRIL are not undergoing replicative senescence 138 5.4 Discussion 140 5.5 Summary 142 5.6 Future work 143 6.0 Effects of ANRIL knock down on gene expression in human primary osteoblasts 145 6.1 Introduction 147 6.2 Aims 148 6.3 Results 149 6.3.1 Validation of Illumina Human H12 Gene Expression Array results 149 IV 6.3.2 ANRIL knock down in primary osteoblasts alters osteoblast- specific pathways 154 6.4 Discussion 163 6.5 Summary 166 6.6 Future work 167 7.0 The association between the methylation status of CDKN2A, RXRA and NOS3 with bone outcomes at birth 169 7.1 Introduction 171 7.1.1 DNA methylation as a marker for a later phenotype 171 7.1.2 Maternal vitamin D and offspring bone health
Recommended publications
  • Identification and Developmental Expression of the Full Complement Of
    Goldstone et al. BMC Genomics 2010, 11:643 http://www.biomedcentral.com/1471-2164/11/643 RESEARCH ARTICLE Open Access Identification and developmental expression of the full complement of Cytochrome P450 genes in Zebrafish Jared V Goldstone1, Andrew G McArthur2, Akira Kubota1, Juliano Zanette1,3, Thiago Parente1,4, Maria E Jönsson1,5, David R Nelson6, John J Stegeman1* Abstract Background: Increasing use of zebrafish in drug discovery and mechanistic toxicology demands knowledge of cytochrome P450 (CYP) gene regulation and function. CYP enzymes catalyze oxidative transformation leading to activation or inactivation of many endogenous and exogenous chemicals, with consequences for normal physiology and disease processes. Many CYPs potentially have roles in developmental specification, and many chemicals that cause developmental abnormalities are substrates for CYPs. Here we identify and annotate the full suite of CYP genes in zebrafish, compare these to the human CYP gene complement, and determine the expression of CYP genes during normal development. Results: Zebrafish have a total of 94 CYP genes, distributed among 18 gene families found also in mammals. There are 32 genes in CYP families 5 to 51, most of which are direct orthologs of human CYPs that are involved in endogenous functions including synthesis or inactivation of regulatory molecules. The high degree of sequence similarity suggests conservation of enzyme activities for these CYPs, confirmed in reports for some steroidogenic enzymes (e.g. CYP19, aromatase; CYP11A, P450scc; CYP17, steroid 17a-hydroxylase), and the CYP26 retinoic acid hydroxylases. Complexity is much greater in gene families 1, 2, and 3, which include CYPs prominent in metabolism of drugs and pollutants, as well as of endogenous substrates.
    [Show full text]
  • (12) Patent Application Publication (10) Pub. No.: US 2011/0190389 A1 Arterburn Et Al
    US 2011 0190389A1 (19) United States (12) Patent Application Publication (10) Pub. No.: US 2011/0190389 A1 Arterburn et al. (43) Pub. Date: Aug. 4, 2011 (54) OXYLIPINS FROM LONG CHAIN C07C 57/03 (2006.01) POLYUNSATURATED FATTY ACDS AND A6IP 29/00 (2006.01) METHODS OF MAKING AND USING THE CI2P 7/64 (2006.01) SAME CI2P I 7/02 (2006.01) C07D 303/38 (2006.01) (76) Inventors: Linda Arterburn, Ellicott City, A6IP 25/28 (2006.01) MD (US); William Barclay, Boulder, CO (US); Bindi Dangi, (52) U.S. Cl. ......... 514/475: 514/560; 554/124; 435/134; Elkridge, MD (US); James Flatt, 435/123:549/561 Colorado Springs, CO (US); Jung Lee, McLean, VA (US); Dutt (57) ABSTRACT Vinjamoori, Chesterfield, MO Disclosed are novel oxylipins, referred to herein as (US); Mary Van Elswyk, docosanoids and eicosanoids, that are derived from C22 poly Longmont, CO (US) unsaturated fatty acids and from C20 polyunsaturated fatty acids, respectively, and methods of making and using Such (21) Appl. No.: 12/531,344 oxylipins. Also disclosed is the use of docosapentaenoic acid 1-1. (C22:5n-6) (DPAn-6), docosapentaenoic acid (C22:5n-3) (22) PCT Filed: Feb. 20, 2008 (DPAn-3), and docosatetraenoic acid (DTAn-6: C22:4n-6), (86). PCT No.: PCT/USO8/54.456 docosatrienoic acid (C22:3n-3) (DTrAn-3), docosadienoic acid (C22:2n-6) (DDAn-6), eicosatrienoic acid (C20:3n-3) S371 (c)(1), (ETrAn-3) eicosapentaenoic acid and arachidonic acid as (2), (4) Date: Feb. 15, 2011 substrates for the production of novel oxylipins, and to the oxylipins produced thereby.
    [Show full text]
  • Tumor-Associated Antigens Identified Early in Mouse Mammary Tumor Development Can Be Effective Vaccine Targets
    Vaccine 37 (2019) 3552–3561 Contents lists available at ScienceDirect Vaccine journal homepage: www.elsevier.com/locate/vaccine Tumor-associated antigens identified early in mouse mammary tumor development can be effective vaccine targets ⇑ Sasha E. Stanton a, , Ekram Gad a, Lauren R. Corulli a, Hailing Lu a,1, Mary L. Disis a a Cancer Vaccine Institute, University of Washington, Seattle WA, 98109, USA article info abstract Article history: Breast cancer vaccines composed of antigens identified by serological analysis of cDNA expression Received 25 June 2018 libraries (SEREX) induce antigen specific immune responses in patients but have had disappointing clin- Received in revised form 5 April 2019 ical benefits. While many attempts to modify the adjuvants and vaccine method have been tried, one Accepted 9 May 2019 issue not addressed was whether the SEREX tumor-associated antigens identified from late stages of dis- Available online 21 May 2019 ease were ideal targets. We questioned in the transgenic TgMMTV-neu mouse model whether the antigen repertoire is distinct between early and late stage breast cancer and whether the antigens identified via Keywords: SEREX from transgenic mice with early or late stage tumors would elicit differential anti-tumor effects to Breast cancer prevention address this question. Vaccine antigens Th1 Three early stage antigens, Pdhx, Stk39, and Otud6B, were identified from a SEREX screen of mice prior DNA vaccines to development of palpable lesions. Formulated into a vaccine, each early antigen inhibited tumor growth Mouse mammary tumor models (p < 0.0001). The antigens identified from mice with late stage tumors (Swap70, Gsn, and Arhgef2) were unable to inhibit tumor growth when used as vaccines (for example Gsn p = 0.26).
    [Show full text]
  • Synonymous Single Nucleotide Polymorphisms in Human Cytochrome
    DMD Fast Forward. Published on February 9, 2009 as doi:10.1124/dmd.108.026047 DMD #26047 TITLE PAGE: A BIOINFORMATICS APPROACH FOR THE PHENOTYPE PREDICTION OF NON- SYNONYMOUS SINGLE NUCLEOTIDE POLYMORPHISMS IN HUMAN CYTOCHROME P450S LIN-LIN WANG, YONG LI, SHU-FENG ZHOU Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, P. R. China (LL Wang & Y Li) Discipline of Chinese Medicine, School of Health Sciences, RMIT University, Bundoora, Victoria 3083, Australia (LL Wang & SF Zhou). 1 Copyright 2009 by the American Society for Pharmacology and Experimental Therapeutics. DMD #26047 RUNNING TITLE PAGE: a) Running title: Prediction of phenotype of human CYPs. b) Author for correspondence: A/Prof. Shu-Feng Zhou, MD, PhD Discipline of Chinese Medicine, School of Health Sciences, RMIT University, WHO Collaborating Center for Traditional Medicine, Bundoora, Victoria 3083, Australia. Tel: + 61 3 9925 7794; fax: +61 3 9925 7178. Email: [email protected] c) Number of text pages: 21 Number of tables: 10 Number of figures: 2 Number of references: 40 Number of words in Abstract: 249 Number of words in Introduction: 749 Number of words in Discussion: 1459 d) Non-standard abbreviations: CYP, cytochrome P450; nsSNP, non-synonymous single nucleotide polymorphism. 2 DMD #26047 ABSTRACT Non-synonymous single nucleotide polymorphisms (nsSNPs) in coding regions that can lead to amino acid changes may cause alteration of protein function and account for susceptivity to disease. Identification of deleterious nsSNPs from tolerant nsSNPs is important for characterizing the genetic basis of human disease, assessing individual susceptibility to disease, understanding the pathogenesis of disease, identifying molecular targets for drug treatment and conducting individualized pharmacotherapy.
    [Show full text]
  • Colorectal Cancer and Omega Hydroxylases
    1 The differential expression of omega-3 and omega-6 fatty acid metabolising enzymes in colorectal cancer and its prognostic significance Abdo Alnabulsi1,2, Rebecca Swan1, Beatriz Cash2, Ayham Alnabulsi2, Graeme I Murray1 1Pathology, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, AB25, 2ZD, UK. 2Vertebrate Antibodies, Zoology Building, Tillydrone Avenue, Aberdeen, AB24 2TZ, UK. Address correspondence to: Professor Graeme I Murray Email [email protected] Phone: +44(0)1224 553794 Fax: +44(0)1224 663002 Running title: omega hydroxylases and colorectal cancer 2 Abstract Background: Colorectal cancer is a common malignancy and one of the leading causes of cancer related deaths. The metabolism of omega fatty acids has been implicated in tumour growth and metastasis. Methods: This study has characterised the expression of omega fatty acid metabolising enzymes CYP4A11, CYP4F11, CYP4V2 and CYP4Z1 using monoclonal antibodies we have developed. Immunohistochemistry was performed on a tissue microarray containing 650 primary colorectal cancers, 285 lymph node metastasis and 50 normal colonic mucosa. Results: The differential expression of CYP4A11 and CYP4F11 showed a strong association with survival in both the whole patient cohort (HR=1.203, 95% CI=1.092-1.324, χ2=14.968, p=0.001) and in mismatch repair proficient tumours (HR=1.276, 95% CI=1.095-1.488, χ2=9.988, p=0.007). Multivariate analysis revealed that the differential expression of CYP4A11 and CYP4F11 was independently prognostic in both the whole patient cohort (p = 0.019) and in mismatch repair proficient tumours (p=0.046). Conclusions: A significant and independent association has been identified between overall survival and the differential expression of CYP4A11 and CYP4F11 in the whole patient cohort and in mismatch repair proficient tumours.
    [Show full text]
  • Type of the Paper (Article
    Supplementary Material A Proteomics Study on the Mechanism of Nutmeg-induced Hepatotoxicity Wei Xia 1, †, Zhipeng Cao 1, †, Xiaoyu Zhang 1 and Lina Gao 1,* 1 School of Forensic Medicine, China Medical University, Shenyang 110122, P. R. China; lessen- [email protected] (W.X.); [email protected] (Z.C.); [email protected] (X.Z.) † The authors contributed equally to this work. * Correspondence: [email protected] Figure S1. Table S1. Peptide fraction separation liquid chromatography elution gradient table. Time (min) Flow rate (mL/min) Mobile phase A (%) Mobile phase B (%) 0 1 97 3 10 1 95 5 30 1 80 20 48 1 60 40 50 1 50 50 53 1 30 70 54 1 0 100 1 Table 2. Liquid chromatography elution gradient table. Time (min) Flow rate (nL/min) Mobile phase A (%) Mobile phase B (%) 0 600 94 6 2 600 83 17 82 600 60 40 84 600 50 50 85 600 45 55 90 600 0 100 Table S3. The analysis parameter of Proteome Discoverer 2.2. Item Value Type of Quantification Reporter Quantification (TMT) Enzyme Trypsin Max.Missed Cleavage Sites 2 Precursor Mass Tolerance 10 ppm Fragment Mass Tolerance 0.02 Da Dynamic Modification Oxidation/+15.995 Da (M) and TMT /+229.163 Da (K,Y) N-Terminal Modification Acetyl/+42.011 Da (N-Terminal) and TMT /+229.163 Da (N-Terminal) Static Modification Carbamidomethyl/+57.021 Da (C) 2 Table S4. The DEPs between the low-dose group and the control group. Protein Gene Fold Change P value Trend mRNA H2-K1 0.380 0.010 down Glutamine synthetase 0.426 0.022 down Annexin Anxa6 0.447 0.032 down mRNA H2-D1 0.467 0.002 down Ribokinase Rbks 0.487 0.000
    [Show full text]
  • Myopia Genetics Report
    Special Issue IMI – Myopia Genetics Report Milly S. Tedja,1,2 Annechien E. G. Haarman,1,2 Magda A. Meester-Smoor,1,2 Jaakko Kaprio,3,4 David A. Mackey,5–7 Jeremy A. Guggenheim,8 Christopher J. Hammond,9 Virginie J. M. Verhoeven,1,2,10 and Caroline C. W. Klaver1,2,11; for the CREAM Consortium 1Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands 2Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands 3Institute for Molecular Medicine, University of Helsinki, Helsinki, Finland 4Department of Public Health, University of Helsinki, Helsinki, Finland 5Centre for Eye Research Australia, Ophthalmology, Department of Surgery, University of Melbourne, Royal Victorian Eye and Ear Hospital, Melbourne, Victoria, Australia 6Department of Ophthalmology, Menzies Institute of Medical Research, University of Tasmania, Hobart, Tasmania, Australia 7Centre for Ophthalmology and Visual Science, Lions Eye Institute, University of Western Australia, Perth, Western Australia, Australia 8School of Optometry and Vision Sciences, Cardiff University, Cardiff, United Kingdom 9Section of Academic Ophthalmology, School of Life Course Sciences, King’s College London, London, United Kingdom 10Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, the Netherlands 11Department of Ophthalmology, Radboud University Medical Center, Nijmegen, the Netherlands Correspondence: Caroline C. W. The knowledge on the genetic background of refractive error and myopia has expanded Klaver, Erasmus Medical Center, dramatically in the past few years. This white paper aims to provide a concise summary of Room Na-2808, P.O. Box 2040, 3000 current genetic findings and defines the direction where development is needed. CA, Rotterdam, the Netherlands; [email protected]. We performed an extensive literature search and conducted informal discussions with key MST and AEGH contributed equally to stakeholders.
    [Show full text]
  • Reduction of Lipid Accumulation Rescues Bietti's Crystalline Dystrophy
    Reduction of lipid accumulation rescues Bietti’s crystalline dystrophy phenotypes Masayuki Hataa,b, Hanako O. Ikedaa,b,1, Sachiko Iwaia, Yuto Iidaa, Norimoto Gotoha, Isao Asakac, Kazutaka Ikedad,e, Yosuke Isobed, Aya Horid, Saori Nakagawaf, Susumu Yamatof, Makoto Aritad,e,g, Nagahisa Yoshimuraa,b, and Akitaka Tsujikawaa aDepartment of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan; bNeuroprotective Treatment Project for Ocular Diseases, Institute for Advancement of Clinical and Translational Science, Kyoto University Hospital, Kyoto 6068507, Japan; cDepartment of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, Kyoto 6068507, Japan; dLaboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Kanagawa 2300045, Japan; eGraduate School of Medical Life Science, Yokohama City University, Kanagawa 2300045, Japan; fDepartment of Bioanalytical Chemistry, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, Niigata 9568603, Japan; and gDivision of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo 1058512, Japan Edited by Paul S. Bernstein, University of Utah School of Medicine, Salt Lake City, UT, and accepted by Editorial Board Member Jeremy Nathans March 9, 2018 (received for review October 4, 2017) Bietti’s crystalline dystrophy (BCD) is an intractable and progres- predicted to be a member of the cytochrome P450 superfamily and sive chorioretinal degenerative disease caused by mutations in the may be involved in the metabolism of lipids (3, 4, 8–11). However, CYP4V2 gene, resulting in blindness in most patients. Although we the mechanisms of RPE damage in BCD remain largely unknown and others have shown that retinal pigment epithelium (RPE) cells because of several problems associated with the research into are primarily impaired in patients with BCD, the underlying mech- BCD.
    [Show full text]
  • Supplementary Materials
    Supplementary materials Supplementary Table S1: MGNC compound library Ingredien Molecule Caco- Mol ID MW AlogP OB (%) BBB DL FASA- HL t Name Name 2 shengdi MOL012254 campesterol 400.8 7.63 37.58 1.34 0.98 0.7 0.21 20.2 shengdi MOL000519 coniferin 314.4 3.16 31.11 0.42 -0.2 0.3 0.27 74.6 beta- shengdi MOL000359 414.8 8.08 36.91 1.32 0.99 0.8 0.23 20.2 sitosterol pachymic shengdi MOL000289 528.9 6.54 33.63 0.1 -0.6 0.8 0 9.27 acid Poricoic acid shengdi MOL000291 484.7 5.64 30.52 -0.08 -0.9 0.8 0 8.67 B Chrysanthem shengdi MOL004492 585 8.24 38.72 0.51 -1 0.6 0.3 17.5 axanthin 20- shengdi MOL011455 Hexadecano 418.6 1.91 32.7 -0.24 -0.4 0.7 0.29 104 ylingenol huanglian MOL001454 berberine 336.4 3.45 36.86 1.24 0.57 0.8 0.19 6.57 huanglian MOL013352 Obacunone 454.6 2.68 43.29 0.01 -0.4 0.8 0.31 -13 huanglian MOL002894 berberrubine 322.4 3.2 35.74 1.07 0.17 0.7 0.24 6.46 huanglian MOL002897 epiberberine 336.4 3.45 43.09 1.17 0.4 0.8 0.19 6.1 huanglian MOL002903 (R)-Canadine 339.4 3.4 55.37 1.04 0.57 0.8 0.2 6.41 huanglian MOL002904 Berlambine 351.4 2.49 36.68 0.97 0.17 0.8 0.28 7.33 Corchorosid huanglian MOL002907 404.6 1.34 105 -0.91 -1.3 0.8 0.29 6.68 e A_qt Magnogrand huanglian MOL000622 266.4 1.18 63.71 0.02 -0.2 0.2 0.3 3.17 iolide huanglian MOL000762 Palmidin A 510.5 4.52 35.36 -0.38 -1.5 0.7 0.39 33.2 huanglian MOL000785 palmatine 352.4 3.65 64.6 1.33 0.37 0.7 0.13 2.25 huanglian MOL000098 quercetin 302.3 1.5 46.43 0.05 -0.8 0.3 0.38 14.4 huanglian MOL001458 coptisine 320.3 3.25 30.67 1.21 0.32 0.9 0.26 9.33 huanglian MOL002668 Worenine
    [Show full text]
  • Deubiquitylases in Developmental Ubiquitin Signaling and Congenital Diseases
    Cell Death & Differentiation (2021) 28:538–556 https://doi.org/10.1038/s41418-020-00697-5 REVIEW ARTICLE Deubiquitylases in developmental ubiquitin signaling and congenital diseases 1 1,2 1 Mohammed A. Basar ● David B. Beck ● Achim Werner Received: 16 October 2020 / Revised: 20 November 2020 / Accepted: 24 November 2020 / Published online: 17 December 2020 This is a U.S. government work and not under copyright protection in the U.S.; foreign copyright protection may apply 2020 Abstract Metazoan development from a one-cell zygote to a fully formed organism requires complex cellular differentiation and communication pathways. To coordinate these processes, embryos frequently encode signaling information with the small protein modifier ubiquitin, which is typically attached to lysine residues within substrates. During ubiquitin signaling, a three-step enzymatic cascade modifies specific substrates with topologically unique ubiquitin modifications, which mediate changes in the substrate’s stability, activity, localization, or interacting proteins. Ubiquitin signaling is critically regulated by deubiquitylases (DUBs), a class of ~100 human enzymes that oppose the conjugation of ubiquitin. DUBs control many essential cellular functions and various aspects of human physiology and development. Recent genetic studies have fi 1234567890();,: 1234567890();,: identi ed mutations in several DUBs that cause developmental disorders. Here we review principles controlling DUB activity and substrate recruitment that allow these enzymes to regulate ubiquitin signaling during development. We summarize key mechanisms of how DUBs control embryonic and postnatal differentiation processes, highlight developmental disorders that are caused by mutations in particular DUB members, and describe our current understanding of how these mutations disrupt development. Finally, we discuss how emerging tools from human disease genetics will enable the identification and study of novel congenital disease-causing DUBs.
    [Show full text]
  • Metabolic Activation and Toxicological Evaluation of Polychlorinated Biphenyls in Drosophila Melanogaster T
    www.nature.com/scientificreports OPEN Metabolic activation and toxicological evaluation of polychlorinated biphenyls in Drosophila melanogaster T. Idda1,7, C. Bonas1,7, J. Hofmann1, J. Bertram1, N. Quinete1,2, T. Schettgen1, K. Fietkau3, A. Esser1, M. B. Stope4, M. M. Leijs3, J. M. Baron3, T. Kraus1, A. Voigt5,6 & P. Ziegler1* Degradation of polychlorinated biphenyls (PCBs) is initiated by cytochrome P450 (CYP) enzymes and includes PCB oxidation to OH-metabolites, which often display a higher toxicity than their parental compounds. In search of an animal model refecting PCB metabolism and toxicity, we tested Drosophila melanogaster, a well-known model system for genetics and human disease. Feeding Drosophila with lower chlorinated (LC) PCB congeners 28, 52 or 101 resulted in the detection of a human-like pattern of respective OH-metabolites in fy lysates. Feeding fies high PCB 28 concentrations caused lethality. Thus we silenced selected CYPs via RNA interference and analyzed the efect on PCB 28-derived metabolite formation by assaying 3-OH-2′,4,4′-trichlorobiphenyl (3-OHCB 28) and 3′-OH-4′,4,6′-trichlorobiphenyl (3′-OHCB 28) in fy lysates. We identifed several drosophila CYPs (dCYPs) whose knockdown reduced PCB 28-derived OH-metabolites and suppressed PCB 28 induced lethality including dCYP1A2. Following in vitro analysis using a liver-like CYP-cocktail, containing human orthologues of dCYP1A2, we confrm human CYP1A2 as a PCB 28 metabolizing enzyme. PCB 28-induced mortality in fies was accompanied by locomotor impairment, a common phenotype of neurodegenerative disorders. Along this line, we show PCB 28-initiated caspase activation in diferentiated fy neurons.
    [Show full text]
  • Structural Variants in Mongolian Originated Ruminant: Role in Adaptation of Extreme-Environment
    Structural variants in Mongolian originated ruminant: role in adaptation of extreme-environment Yanping Xing Inner Mongolia Agricultural University https://orcid.org/0000-0001-6416-7633 Yu Qi Inner Mongolia Agricultural University Chimgee Purev Inner Mongolia Agricultural University Shengyuan Wang Inner Mongolia Agricultural University Hongbing Wang Hunan Agricultural University Kaifeng Wu Inner Mongolia Agricultural University Junwei Cao Inner Mongolia Agricultural University Chunxia Liu Inner Mongolia Agricultural University Yiyi Liu Inner Mongolia Agricultural University Lu Li Inner Mongolia Agricultural University Yanru Zhang ( [email protected] ) Inner Mongolia Agricultural University Huanmin Zhou ( [email protected] ) Inner Mongolia Agricultural University https://orcid.org/0000-0003-3852-0071 Research article Keywords: Mongolian cattle, Genome-sequencing, transcriptome-sequencing, extreme-environment, adaptation Posted Date: March 17th, 2020 DOI: https://doi.org/10.21203/rs.3.rs-17431/v1 License: This work is licensed under a Creative Commons Attribution 4.0 International License. Read Full License 1 Structural variants in Mongolian originated ruminant: role in adaptation of extreme-environment 2 Yanping Xing1#, Yu Qi1,2#, Chimgee Purev3, Shenyuan Wang1, Hongbing Wang4, Kaifeng Wu1, Junwei Cao1, Chunxia Liu1, Yiyi Liu1, 3 Lu Li1, Yanru Zhang1* & Huanmin Zhou1* 4 5 1. Inner Mongolia Key Laboratory of Bio-Manufacture, Inner Mongolian Agriculture University, Huhhot, 010018, the People’s 6 Republic of China. 7 2. College of Animal Science and Technology, Inner Mongolian University for Nationalities, Tongliao, 028000, the People’s Republic of 8 China. 9 3. Mongolia China Joint Laboratory of Applied Molecular Biology, Mongolian National University, Ulan Bator, 999097, Mongolian. 10 4. Institute of Animal and Veterinary Science, Department of Agriculture and Rural Areas of Hunan Province, Changsha, 410131, the 11 People’s Republic of China.
    [Show full text]