Heme, a Metabolic Sensor, Directly Regulates the Activity of the KDM4 Histone Demethylase Family and Their Interactions with Partner Proteins

Total Page:16

File Type:pdf, Size:1020Kb

Heme, a Metabolic Sensor, Directly Regulates the Activity of the KDM4 Histone Demethylase Family and Their Interactions with Partner Proteins cells Article Heme, A Metabolic Sensor, Directly Regulates the Activity of the KDM4 Histone Demethylase Family and Their Interactions with Partner Proteins Purna Chaitanya Konduri y , Tianyuan Wang y, Narges Salamat and Li Zhang * Department of Biological Sciences, University of Texas at Dallas, Mail Stop RL11, 800 W. Campbell Road, Richardson, TX 75080, USA; [email protected] (P.C.K.); [email protected] (T.W.); [email protected] (N.S.) * Correspondence: [email protected]; Tel.: +1-972-883-5757 These authors contributed equally to this paper. y Received: 7 February 2020; Accepted: 19 March 2020; Published: 22 March 2020 Abstract: The KDM4 histone demethylase subfamily is constituted of yeast JmjC domain-containing proteins, such as Gis1, and human Gis1 orthologues, such as KDM4A/B/C. KDM4 proteins have important functions in regulating chromatin structure and gene expression in response to metabolic and nutritional stimuli. Heme acts as a versatile signaling molecule to regulate important cellular functions in diverse organisms ranging from bacteria to humans. Here, using purified KDM4 proteins containing the JmjN/C domain, we showed that heme stimulates the histone demethylase activity of the JmjN/C domains of KDM4A and Cas well as full-length Gis1. Furthermore, we found that the C-terminal regions of KDM4 proteins, like that of Gis1, can confer heme regulation when fused to an unrelated transcriptional activator. Interestingly, biochemical pull-down of Gis1-interacting proteins followed by mass spectrometry identified 147 unique proteins associated with Gis1 under heme-sufficient and/or heme-deficient conditions. These 147 proteins included a significant number of heterocyclic compound-binding proteins, Ubl-conjugated proteins, metabolic enzymes/proteins, and acetylated proteins. These results suggested that KDM4s interact with diverse cellular proteins to form a complex network to sense metabolic and nutritional conditions like heme levels and respond by altering their interactions with other proteins and functional activities, such as histone demethylation. Keywords: Gis1; KDM4; heme; histone demethylase; protein interaction; JmjC domain 1. Introduction Histone lysyl demethylases (KDMs) play important roles in chromatin remodeling and gene regulation by catalyzing the demethylation of methylated lysine residues on the N-terminal tails of histones [1]. The majority of KDMs are JmjC domain-containing proteins, which are dioxygenases that use α-ketoglutarate and Fe2+ to oxidize various substrates [2–4]. The KDM4 family, containing five members, i.e., KDM4A-E, accepts mono-, di- and trimethylated lysines as substrates [5,6]. KDM4A/B/C (Figure1), but not KDM4D or E, contain the JmjC domain in the N-terminus as well as Tudor and plant homeodomain (PHD) domains in the C-terminus. KDM4A/B/C are widely expressed, and their orthologues are found among all vertebrates [7,8]. KDM4A/B/C share more than 50% sequence identity (Figure1) and catalyze the demethylation of tri- and di-methylated forms of both histone H3 lysine 9 (H3K9me3/me2) and lysine 36 (H3K36me3/me2) [3,9–13]. KDM4 proteins play important roles in diverse biological processes, including oncogenesis, inflammation, and hematopoiesis [8,14–16]. It is 2+ worth noting that the histone demethylase activity of KDM4s requires α-ketoglutarate, O2, and Fe , which are important nutritional and metabolic indicators. Thus, histone demethylase activity is linked to and is highly responsive to nutrient status [17–22]. Cells 2020, 9, 773; doi:10.3390/cells9030773 www.mdpi.com/journal/cells Cells 2020, 9, x FOR PEER REVIEW 2 of 24 Cellsα-ketoglutarate,2020, 9, 773 O2, and Fe2+, which are important nutritional and metabolic indicators. Thus, histone2 of 24 demethylase activity is linked to and is highly responsive to nutrient status [17–22]. Figure 1. The structure and alignment of Gis1 and KDM4AKDM4A/B/C/B/C domains.domains. The domains of Gis1 and KDM4s are are shown. shown. Gis1 Gis1 contains contains the the JmjN JmjN + JmjC+ JmjC (JmjN/C) (JmjN /domain,C) domain, the coiled-coil the coiled-coil domain domain (C/C), (C two/C), twoC2H2 C2H2 type typezinc zincfingers fingers (ZnFs), (ZnFs), and and two two transcr transcriptioniption activation activation domains domains (TAD1 (TAD1 and and TAD2). TAD2). KDM4AKDM4A/B/C/B/C proteins contain the JmjNJmjN/C/C domain, two PHD domains (each containing a conserved C4HC3 or C4HC2H motif), and two Tudor domains.domains. The red lines represent cysteine-proline (CP) motifs, black lines indicate the start and end of the shown domains, and the pink box in the Gis1 structure denotes the coiled-coil domain. The % amino acid identity and similarity between Gis1 and KDM4s in JmjNJmjN/C/C and zinc fingerfinger domainsdomains areare shownshown inin parentheses.parentheses. The yeast JmjC domain-containing protein Gis1 is anan orthologueorthologue ofof KDM4KDM4 proteins.proteins. Previous data, including our own recent data, showed that Gis1 possesses histone demethylase activity at H3K36 [[23–26].23–26]. In yeast,yeast, Gis1Gis1 isis oneone ofof the the key key regulators regulators mediating mediating transcriptional transcriptional reprogramming reprogramming of carbonof carbon metabolism metabolism and and stress stress responses responses during during diauxic diauxic and and post-diauxic post-diauxic shifts shifts [[27–32].27–32]. Gis1Gis1 binds to the post-diauxic shift (PDS) DNA DNA element element and and ca cann both both activate activate and and repress repress transcription transcription [33–35].[33–35]. It acts downstream of TOR, RASRAS/cAMP,/cAMP, andand AKTAKT/Sc/Sch9h9 signalingsignaling pathways in response to nutrient status [[28,30,32].28,30,32]. Gis1Gis1 containscontains multiple multiple domains, domains, including including a JmjNa JmjN region, region, a JmjC a JmjC region, region, a coiled-coil a coiled- domain,coil domain, two two C2H2 C2H2 type type zinc zinc fingers fingers (ZnFs), (ZnFs), and and two two transcription transcription activationactivation domains (TADs) (TADs) (Figure1 1))[ [30,36,37].30,36,37]. JmjN JmjN and and JmjC JmjC interact interact physica physicallylly to form to form a structural a structural unit unit or a ordomain a domain [37]. [The37]. TheJmjN/C JmjN domain/C domain presumably presumably confers confers histone histone demethylase demethylase activity, activity, but but is is dispensable dispensable for transcriptional activation activation by by Gis1 Gis1 [36]. [36]. This This Gis1 Gis1 JmjN/C JmjN/ Cdomain domain is ishighly highly homologous homologous to tothose those of ofKDM4A/B/C, KDM4A/B/C, while while the the C-terminal C-terminal regions regions of of Gis1 Gis1 and and KDM4A/B/C KDM4A/B/C share share low low homology homology (Figure (Figure 11).). The Gis1 zinc fingersfingers allow Gis1 to bind to DNA an andd regulate transcription [[30,36,37].30,36,37]. The C-terminal regions of KDM4AKDM4A/B/C/B/C proteins contain two PHDsPHDs and twotwo TudorTudor domainsdomains (Figure(Figure1 ).1). TheThe PHDPHD domain is a protein module containing ~50 amino acid residues with a conserved C4HC3 or C4HC2H motif that coordinates two zinc ions in a cross-brace configurationconfiguration [[38].38]. The Tudor domain typically contains ~60 amino amino acids acids that that comprise comprise 4–5 4–5 antiparallel antiparallel ββ-strands-strands to to form form a a barrel-like barrel-like structure structure [39]. [39]. Both PHDPHD and and Tudor Tudor domains domains are histoneare histone reader reader domains domains that recognize that recognize post-translationally post-translationally modified histones,modified suchhistones, as methylated such as methylated H3 [40–43 ].H3 Although [40–43]. Although the JmjN/C the domains JmjN/C of domains Gis1 and of KDM4s Gis1 and are KDM4s highly conserved,are highly conserved, the C-terminal the C-terminal domain sequences domain sequen of Gis1ces are of quite Gis1 di arefferent, quite anddifferent, there isand no there homology is no betweenhomology the between TADs of the Gis1 TADs and of the Gis1 corresponding and the corresp regionsonding of KDM4s regions (Figure of KDM4s1). Thus, (Figure we do 1). not Thus, expect we thatdo not KDM4s expect can that substitute KDM4s can for Gis1substitute in yeast. for Gis1 in yeast. Interestingly, ourour recentrecent studiesstudies showedshowed thatthat hemeheme promotespromotes both transcriptionaltranscriptional and histone demethylase activities of Gis1 [23]. [23]. Heme Heme can can bind bind to to both both the the JmjN/C JmjN/C and ZnF domains, however, however, heme binding to the JmjN/C JmjN/C domain is not necessary for heme regulation regulation of Gis1 Gis1 activity. activity. In In contrast, contrast, heme binding to the ZnF domain promotes heme activation of both transcriptional and histone demethylase activities [23]. Under heme-deficient conditions, Gis1 is free of heme and is inactive. Its Cells 2020, 9, 773 3 of 24 activities are repressed by the ZnF domain, which likely cooperates with other cellular proteins. When heme levels increase, heme binds to the JmjN/C and ZnF domains, presumably causing other cellular proteins to dissociate from Gis1 and alter Gis1 conformation, thereby leading to full activation of Gis1 transcriptional and demethylase activities. Heme acts as a prosthetic group or cofactor in proteins and enzymes required for oxygen utilization and metabolism, such as globins and cytochromes [44–47]. Heme serves as a signaling molecule regulating diverse processes ranging from gene transcription
Recommended publications
  • Investigation of Histone Lysine-Specific Demethylase 5D
    FULL LENGTH Iranian Biomedical Journal 20(2): 117-121 April 2016 Investigation of Histone Lysine-Specific Demethylase 5D (KDM5D) Isoform Expression in Prostate Cancer Cell Lines: a System Approach Zohreh Jangravi1, 2, Mohammad Najafi1,3 and Mohammd Shabani*1 1Dept. of Biochemistry, Iran University of Medical Sciences, Tehran, Iran; 2Dept. of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; 3Dept. of Biochemistry, Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran Received 2 September 2014; revised 3 December 2014; accepted 7 December 2014 ABSTRACT Background: It is now well-demonstrated that histone demethylases play an important role in developmental controls, cell-fate decisions, and a variety of diseases such as cancer. Lysine-specific demethylase 5D (KDM5D) is a male-specific histone demethylase that specifically demethylates di- and tri-methyl H3K4 at the start site of active gene. In this light, the aim of this study was to investigate isoform/transcript-specific expression profiles of KDM5D in three prostate cancer cell lines, Du-145, LNCaP, and PC3. Methods: Real-time PCR analysis was performed to determine the expression levels of different KDM5D transcripts in the prostate cell lines. A gene regulatory network was established to analyze the gene expression profile. Results: Significantly different expression levels of both isoforms were found among the three cell lines. Interestingly, isoform I was expressed in three cell lines while isoform III did only in DU-145. The expression levels of both isoforms were higher in DU-145 when compared to other cell lines (P<0.0001).
    [Show full text]
  • Lanosterol 14Α-Demethylase (CYP51)
    463 Lanosterol 14-demethylase (CYP51), NADPH–cytochrome P450 reductase and squalene synthase in spermatogenesis: late spermatids of the rat express proteins needed to synthesize follicular fluid meiosis activating sterol G Majdicˇ, M Parvinen1, A Bellamine2, H J Harwood Jr3, WWKu3, M R Waterman2 and D Rozman4 Veterinary Faculty, Clinic of Reproduction, Cesta v Mestni log 47a, 1000 Ljubljana, Slovenia 1Institute of Biomedicine, Department of Anatomy, University of Turku, Kiinamyllynkatu 10, FIN-20520 Turku, Finland 2Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232–0146, USA 3Pfizer Central Research, Department of Metabolic Diseases, Box No. 0438, Eastern Point Road, Groton, Connecticut 06340, USA 4Institute of Biochemistry, Medical Center for Molecular Biology, Medical Faculty University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia (Requests for offprints should be addressed to D Rozman; Email: [email protected]) (G Majdicˇ is now at Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas 75235–8857, USA) Abstract Lanosterol 14-demethylase (CYP51) is a cytochrome detected in step 3–19 spermatids, with large amounts in P450 enzyme involved primarily in cholesterol biosynthe- the cytoplasm/residual bodies of step 19 spermatids, where sis. CYP51 in the presence of NADPH–cytochrome P450 P450 reductase was also observed. Squalene synthase was reductase converts lanosterol to follicular fluid meiosis immunodetected in step 2–15 spermatids of the rat, activating sterol (FF-MAS), an intermediate of cholesterol indicating that squalene synthase and CYP51 proteins are biosynthesis which accumulates in gonads and has an not equally expressed in same stages of spermatogenesis. additional function as oocyte meiosis-activating substance.
    [Show full text]
  • Table S1. 49 Histone Variants Were Identified with High Sequence Coverage Through LC-MS/MS Analysis Electronic Supplementary
    Electronic Supplementary Material (ESI) for Analytical Methods. This journal is © The Royal Society of Chemistry 2020 Table S1. 49 histone variants were identified with high sequence coverage through LC-MS/MS analysis Sequence Uniprot IDs Protein Name Protein Description Coverage Ratio E2+/E2- RSD P07305 H10_HUMAN 67.5% Histone H1.0 OS=Homo sapiens GN=H1F0 PE=1 SV=3 4.85 23.3% Histone H1.1 OS=Homo sapiens GN=HIST1H1A PE=1 Q02539 H11_HUMAN 74.4% SV=3 0.35 92.6% Histone H1.2 OS=Homo sapiens GN=HIST1H1C PE=1 P16403 H12_HUMAN 67.1% SV=2 0.73 80.6% Histone H1.3 OS=Homo sapiens GN=HIST1H1D PE=1 P16402 H13_HUMAN 63.8% SV=2 0.75 77.7% Histone H1.4 OS=Homo sapiens GN=HIST1H1E PE=1 P10412 H14_HUMAN 69.0% SV=2 0.70 80.3% Histone H1.5 OS=Homo sapiens GN=HIST1H1B PE=1 P16401 H15_HUMAN 79.6% SV=3 0.29 98.3% Testis-specific H1 histone OS=Homo sapiens GN=H1FNT Q75WM6 H1FNT_HUMAN 7.8% \ \ PE=2 SV=3 Histone H1oo OS=Homo sapiens GN=H1FOO PE=2 Q8IZA3 H1FOO_HUMAN 5.2% \ \ SV=1 Histone H1t OS=Homo sapiens GN=HIST1H1T PE=2 P22492 H1T_HUMAN 31.4% SV=4 1.42 65.0% Q92522 H1X_HUMAN 82.6% Histone H1x OS=Homo sapiens GN=H1FX PE=1 SV=1 1.15 33.2% Histone H2A type 1 OS=Homo sapiens GN=HIST1H2AG P0C0S8 H2A1_HUMAN 99.2% PE=1 SV=2 0.57 26.8% Q96QV6 H2A1A_HUMAN 58.0% Histone H2A type 1-A OS=Homo sapiens 0.90 11.2% GN=HIST1H2AA PE=1 SV=3 Histone H2A type 1-B/E OS=Homo sapiens P04908 H2A1B_HUMAN 99.2% GN=HIST1H2AB PE=1 SV=2 0.92 30.2% Histone H2A type 1-C OS=Homo sapiens Q93077 H2A1C_HUMAN 100.0% GN=HIST1H2AC PE=1 SV=3 0.76 27.6% Histone H2A type 1-D OS=Homo sapiens P20671
    [Show full text]
  • A Cell Line P53 Mutation Type UM
    A Cell line p53 mutation Type UM-SCC 1 wt UM-SCC5 Exon 5, 157 GTC --> TTC Missense mutation by transversion (Valine --> Phenylalanine UM-SCC6 wt UM-SCC9 wt UM-SCC11A wt UM-SCC11B Exon 7, 242 TGC --> TCC Missense mutation by transversion (Cysteine --> Serine) UM-SCC22A Exon 6, 220 TAT --> TGT Missense mutation by transition (Tyrosine --> Cysteine) UM-SCC22B Exon 6, 220 TAT --> TGT Missense mutation by transition (Tyrosine --> Cysteine) UM-SCC38 Exon 5, 132 AAG --> AAT Missense mutation by transversion (Lysine --> Asparagine) UM-SCC46 Exon 8, 278 CCT --> CGT Missense mutation by transversion (Proline --> Alanine) B 1 Supplementary Methods Cell Lines and Cell Culture A panel of ten established HNSCC cell lines from the University of Michigan series (UM-SCC) was obtained from Dr. T. E. Carey at the University of Michigan, Ann Arbor, MI. The UM-SCC cell lines were derived from eight patients with SCC of the upper aerodigestive tract (supplemental Table 1). Patient age at tumor diagnosis ranged from 37 to 72 years. The cell lines selected were obtained from patients with stage I-IV tumors, distributed among oral, pharyngeal and laryngeal sites. All the patients had aggressive disease, with early recurrence and death within two years of therapy. Cell lines established from single isolates of a patient specimen are designated by a numeric designation, and where isolates from two time points or anatomical sites were obtained, the designation includes an alphabetical suffix (i.e., "A" or "B"). The cell lines were maintained in Eagle's minimal essential media supplemented with 10% fetal bovine serum and penicillin/streptomycin.
    [Show full text]
  • Environmental Influences on Endothelial Gene Expression
    ENDOTHELIAL CELL GENE EXPRESSION John Matthew Jeff Herbert Supervisors: Prof. Roy Bicknell and Dr. Victoria Heath PhD thesis University of Birmingham August 2012 University of Birmingham Research Archive e-theses repository This unpublished thesis/dissertation is copyright of the author and/or third parties. The intellectual property rights of the author or third parties in respect of this work are as defined by The Copyright Designs and Patents Act 1988 or as modified by any successor legislation. Any use made of information contained in this thesis/dissertation must be in accordance with that legislation and must be properly acknowledged. Further distribution or reproduction in any format is prohibited without the permission of the copyright holder. ABSTRACT Tumour angiogenesis is a vital process in the pathology of tumour development and metastasis. Targeting markers of tumour endothelium provide a means of targeted destruction of a tumours oxygen and nutrient supply via destruction of tumour vasculature, which in turn ultimately leads to beneficial consequences to patients. Although current anti -angiogenic and vascular targeting strategies help patients, more potently in combination with chemo therapy, there is still a need for more tumour endothelial marker discoveries as current treatments have cardiovascular and other side effects. For the first time, the analyses of in-vivo biotinylation of an embryonic system is performed to obtain putative vascular targets. Also for the first time, deep sequencing is applied to freshly isolated tumour and normal endothelial cells from lung, colon and bladder tissues for the identification of pan-vascular-targets. Integration of the proteomic, deep sequencing, public cDNA libraries and microarrays, delivers 5,892 putative vascular targets to the science community.
    [Show full text]
  • Download Download
    Supplementary Figure S1. Results of flow cytometry analysis, performed to estimate CD34 positivity, after immunomagnetic separation in two different experiments. As monoclonal antibody for labeling the sample, the fluorescein isothiocyanate (FITC)- conjugated mouse anti-human CD34 MoAb (Mylteni) was used. Briefly, cell samples were incubated in the presence of the indicated MoAbs, at the proper dilution, in PBS containing 5% FCS and 1% Fc receptor (FcR) blocking reagent (Miltenyi) for 30 min at 4 C. Cells were then washed twice, resuspended with PBS and analyzed by a Coulter Epics XL (Coulter Electronics Inc., Hialeah, FL, USA) flow cytometer. only use Non-commercial 1 Supplementary Table S1. Complete list of the datasets used in this study and their sources. GEO Total samples Geo selected GEO accession of used Platform Reference series in series samples samples GSM142565 GSM142566 GSM142567 GSM142568 GSE6146 HG-U133A 14 8 - GSM142569 GSM142571 GSM142572 GSM142574 GSM51391 GSM51392 GSE2666 HG-U133A 36 4 1 GSM51393 GSM51394 only GSM321583 GSE12803 HG-U133A 20 3 GSM321584 2 GSM321585 use Promyelocytes_1 Promyelocytes_2 Promyelocytes_3 Promyelocytes_4 HG-U133A 8 8 3 GSE64282 Promyelocytes_5 Promyelocytes_6 Promyelocytes_7 Promyelocytes_8 Non-commercial 2 Supplementary Table S2. Chromosomal regions up-regulated in CD34+ samples as identified by the LAP procedure with the two-class statistics coded in the PREDA R package and an FDR threshold of 0.5. Functional enrichment analysis has been performed using DAVID (http://david.abcc.ncifcrf.gov/)
    [Show full text]
  • Sterol Homeostasis Requires Regulated Degradation of Squalene
    RESEARCH ARTICLE elife.elifesciences.org Sterol homeostasis requires regulated degradation of squalene monooxygenase by the ubiquitin ligase Doa10/Teb4 Ombretta Foresti1,2, Annamaria Ruggiano1,2, Hans K Hannibal-Bach3, Christer S Ejsing3, Pedro Carvalho1,2* 1Cell and Developmental Biology Programme, Center for Genomic Regulation (CRG), Barcelona, Spain; 2Universitat Pompeu Fabra, Barcelona, Spain; 3Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark Abstract Sterol homeostasis is essential for the function of cellular membranes and requires feedback inhibition of HMGR, a rate-limiting enzyme of the mevalonate pathway. As HMGR acts at the beginning of the pathway, its regulation affects the synthesis of sterols and of other essential mevalonate-derived metabolites, such as ubiquinone or dolichol. Here, we describe a novel, evolutionarily conserved feedback system operating at a sterol-specific step of the mevalonate pathway. This involves the sterol-dependent degradation of squalene monooxygenase mediated by the yeast Doa10 or mammalian Teb4, a ubiquitin ligase implicated in a branch of the endoplasmic reticulum (ER)-associated protein degradation (ERAD) pathway. Since the other branch of ERAD is required for HMGR regulation, our results reveal a fundamental role for ERAD in sterol homeostasis, with the two branches of this pathway acting together to control sterol biosynthesis at different levels and thereby allowing independent regulation of multiple products of the mevalonate pathway. DOI: 10.7554/eLife.00953.001 *For correspondence: pedro. [email protected] Introduction Sterols, such as cholesterol in animals or ergosterol in yeast, are essential components of cellular Competing interests: The authors membranes and their concentration to a large extent determines many of the membrane properties, declare that no competing interests exist.
    [Show full text]
  • Identification of Reproduction-Related Gene Polymorphisms Using Whole
    INVESTIGATION Identification of Reproduction-Related Gene Polymorphisms Using Whole Transcriptome Sequencing in the Large White Pig Population Daniel Fischer,* Asta Laiho,† Attila Gyenesei,‡ and Anu Sironen*,1 *Natural Resources Institute Finland (Luke), Green Technology, Animal and Plant Genomics and Breeding, FI-31600 Jokioinen, Finland, †The Finnish Microarray and Sequencing Centre, Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Tykistökatu 6, FI-20520 Turku, Finland, and ‡Campus Science Support Facilities, Vienna Biocenter, A-1030 Vienna, Austria ORCID ID: 0000-0003-2064-6960 (D.F.) ABSTRACT Recent developments in high-throughput sequencing techniques have enabled large-scale KEYWORDS analysis of genetic variations and gene expression in different tissues and species, but gene expression oviduct patterns and genetic variations in livestock are not well-characterized. In this study, we have used high- testis throughput transcriptomic sequencing of the Finnish Large White to identify gene expression patterns and gene expression coding polymorphisms within the breed in the testis and oviduct. The main objective of this study was to polymorphism identify polymorphisms within genes that are highly and specifically expressed in male and/or female SNP reproductive organs. The differential expression (DE) analysis underlined 1234 genes highly expressed in pig the testis and 1501 in the oviduct. Furthermore, we used a novel in-house R-package hoardeR for the transcriptome identification of novel genes and their orthologs, which underlined 55 additional DE genes based on RNAseq orthologs in the human, cow, and sheep. Identification of polymorphisms in the dataset resulted in a total of reproduction 29,973 variants, of which 10,704 were known coding variants.
    [Show full text]
  • Genome-Wide Association Study of Dietary Intake in the UK Biobank Study and Its Associations with Schizophrenia and Other Traits Maria Niarchou1,2,3, Enda M
    Niarchou et al. Translational Psychiatry (2020) 10:51 https://doi.org/10.1038/s41398-020-0688-y Translational Psychiatry ARTICLE Open Access Genome-wide association study of dietary intake in the UK biobank study and its associations with schizophrenia and other traits Maria Niarchou1,2,3, Enda M. Byrne1, Maciej Trzaskowski4, Julia Sidorenko 1,5, Kathryn E. Kemper1, John J. McGrath 6,7,8,MichaelC.O’ Donovan 2,MichaelJ.Owen2 and Naomi R. Wray 1,6 Abstract Motivated by observational studies that report associations between schizophrenia and traits, such as poor diet, increased body mass index and metabolic disease, we investigated the genetic contribution to dietary intake in a sample of 335,576 individuals from the UK Biobank study. A principal component analysis applied to diet question item responses generated two components: Diet Component 1 (DC1) represented a meat-related diet and Diet Component 2 (DC2) a fish and plant-related diet. Genome-wide association analysis identified 29 independent single- nucleotide polymorphisms (SNPs) associated with DC1 and 63 SNPs with DC2. Estimated from over 35,000 3rd-degree relative pairs that are unlikely to share close family environments, heritabilities for both DC1 and DC2 were 0.16 (standard error (s.e.) = 0.05). SNP-based heritability was 0.06 (s.e. = 0.003) for DC1 and 0.08 (s.e = 0.004) for DC2. We estimated significant genetic correlations between both DCs and schizophrenia, and several other traits. Mendelian randomisation analyses indicated a negative uni-directional relationship between liability to schizophrenia and tendency towards selecting a meat-based diet (which could be direct or via unidentified correlated variables), but a bi- fi 1234567890():,; 1234567890():,; 1234567890():,; 1234567890():,; directional relationship between liability to schizophrenia and tendency towards selecting a sh and plant-based diet consistent with genetic pleiotropy.
    [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]
  • The UVB-Induced Gene Expression Profile of Human Epidermis in Vivo Is Different from That of Cultured Keratinocytes
    Oncogene (2006) 25, 2601–2614 & 2006 Nature Publishing Group All rights reserved 0950-9232/06 $30.00 www.nature.com/onc ORIGINAL ARTICLE The UVB-induced gene expression profile of human epidermis in vivo is different from that of cultured keratinocytes CD Enk1, J Jacob-Hirsch2, H Gal3, I Verbovetski4, N Amariglio2, D Mevorach4, A Ingber1, D Givol3, G Rechavi2 and M Hochberg1 1Department of Dermatology, The Hadassah-Hebrew University Medical Center, Jerusalem, Israel; 2Department of Pediatric Hemato-Oncology and Functional Genomics, Safra Children’s Hospital, Sheba Medical Center and Sackler School of Medicine, Tel-Aviv University,Tel Aviv, Israel; 3Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel and 4The Laboratory for Cellular and Molecular Immunology, Department of Medicine, The Hadassah-Hebrew University Medical Center, Jerusalem, Israel In order to obtain a comprehensive picture of the radiation. UVB, with a wavelength range between 290 molecular events regulating cutaneous photodamage of and 320 nm, represents one of the most important intact human epidermis, suction blister roofs obtained environmental hazards affectinghuman skin (Hahn after a single dose of in vivo ultraviolet (UV)B exposure and Weinberg, 2002). To protect itself against the were used for microarray profiling. We found a changed DNA-damaging effects of sunlight, the skin disposes expression of 619 genes. Half of the UVB-regulated genes over highly complicated cellular programs, including had returned to pre-exposure baseline levels at 72 h, cell-cycle arrest, DNA repair and apoptosis (Brash et al., underscoring the transient character of the molecular 1996). Failure in selected elements of these defensive cutaneous UVB response.
    [Show full text]
  • Genome-Wide Screen of Cell-Cycle Regulators in Normal and Tumor Cells
    bioRxiv preprint doi: https://doi.org/10.1101/060350; this version posted June 23, 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. Genome-wide screen of cell-cycle regulators in normal and tumor cells identifies a differential response to nucleosome depletion Maria Sokolova1, Mikko Turunen1, Oliver Mortusewicz3, Teemu Kivioja1, Patrick Herr3, Anna Vähärautio1, Mikael Björklund1, Minna Taipale2, Thomas Helleday3 and Jussi Taipale1,2,* 1Genome-Scale Biology Program, P.O. Box 63, FI-00014 University of Helsinki, Finland. 2Science for Life laboratory, Department of Biosciences and Nutrition, Karolinska Institutet, SE- 141 83 Stockholm, Sweden. 3Science for Life laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, S-171 21 Stockholm, Sweden To identify cell cycle regulators that enable cancer cells to replicate DNA and divide in an unrestricted manner, we performed a parallel genome-wide RNAi screen in normal and cancer cell lines. In addition to many shared regulators, we found that tumor and normal cells are differentially sensitive to loss of the histone genes transcriptional regulator CASP8AP2. In cancer cells, loss of CASP8AP2 leads to a failure to synthesize sufficient amount of histones in the S-phase of the cell cycle, resulting in slowing of individual replication forks. Despite this, DNA replication fails to arrest, and tumor cells progress in an elongated S-phase that lasts several days, finally resulting in death of most of the affected cells.
    [Show full text]