Maternal Pregravid Obesity Remodels the DNA Methylation Landscape Of

Total Page:16

File Type:pdf, Size:1020Kb

Maternal Pregravid Obesity Remodels the DNA Methylation Landscape Of Maternal Pregravid Obesity Remodels the DNA Methylation Landscape of Cord Blood Monocytes Disrupting Their Inflammatory Program This information is current as of September 26, 2021. Suhas Sureshchandra, Randall M. Wilson, Maham Rais, Nicole E. Marshall, Jonathan Q. Purnell, Kent L. Thornburg and Ilhem Messaoudi J Immunol published online 8 September 2017 http://www.jimmunol.org/content/early/2017/09/07/jimmun Downloaded from ol.1700434 Supplementary http://www.jimmunol.org/content/suppl/2017/09/08/jimmunol.170043 http://www.jimmunol.org/ Material 4.DCSupplemental Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists by guest on September 26, 2021 • Fast Publication! 4 weeks from acceptance to publication *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2017 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. Published September 8, 2017, doi:10.4049/jimmunol.1700434 The Journal of Immunology Maternal Pregravid Obesity Remodels the DNA Methylation Landscape of Cord Blood Monocytes Disrupting Their Inflammatory Program Suhas Sureshchandra,* Randall M. Wilson,† Maham Rais,† Nicole E. Marshall,‡ Jonathan Q. Purnell,x Kent L. Thornburg,x and Ilhem Messaoudi* Prepregnancy maternal obesity is associated with adverse outcomes for the offspring, including increased incidence of neonatal bacterial sepsis and necrotizing enterocolitis. We recently reported that umbilical cord blood (UCB) monocytes from babies born to obese mothers generate a reduced IL-6/TNF-a response to TLR 1/2 and 4 ligands compared to those collected from lean mothers. These observations suggest altered development of the offspring’s immune system, which in turn results in dysregulated function. We therefore investigated transcriptional and epigenetic differences within UCB monocytes stratified by prepregnancy Downloaded from maternal body mass index. We show that UCB monocytes from babies born to obese mothers generate a dampened response to LPS stimulation compared with those born to lean mothers, at the level of secreted immune mediators and transcription. Because gene expression profiles of resting UCB monocytes from both groups were comparable, we next investigated the role of epigenetic differences. Indeed, we detected stark differences in methylation levels within promoters and regulatory regions of genes involved in TLR signaling in resting UCB monocytes. Interestingly, the DNA methylation status of resting cells was highly predictive of transcriptional changes post-LPS stimulation, suggesting that cytosine methylation is one of the dominant mechanisms driving http://www.jimmunol.org/ functional inadequacy in UCB monocytes obtained from babies born to obese mothers. These data highlight a potentially critical role of maternal pregravid obesity-associated epigenetic changes in influencing the function of an offspring’s monocytes at birth. These findings further our understanding of mechanisms that explain the increased risk of infection in neonates born to mothers with high prepregnancy body mass index. The Journal of Immunology, 2017, 199: 000–000. lmost 37% of women of childbearing age are categorized obesity include increased rates of preeclampsia, gestational hyper- as obese, making obesity one of the most common tension, gestational diabetes, placental abruption, preterm delivery, comorbidities during pregnancy (1). It is well estab- and cesarean delivery (1). For the fetus, complications include A by guest on September 26, 2021 lished that a high prepregnancy (pregravid) body mass index increased risk of stillbirth, abnormal growth, and cardiac or neural (BMI) is associated with detrimental health outcomes for both tube defects (1, 4). Moreover, neonates born to obese mothers are mother and child (2–4). Maternal complications of pregravid at increased risk of bacterial sepsis and necrotizing enterocolitis, requiring admission to the neonatal intensive care unit (5, 6). *Department of Molecular Biology and Biochemistry, School of Biological Sciences, Some adverse health outcomes for the offspring persist into University of California Irvine, Irvine, CA 92697; †Division of Biomedical Sciences, Uni- adulthood, including increased susceptibility to respiratory in- versity of California Riverside, Riverside, CA 92521; ‡Maternal-Fetal Medicine, x fections such as respiratory syncytial virus (7, 8), asthma (9), Oregon Health and Science University, Portland, OR 97239; and Department of Medicine, The Knight Cardiovascular Institute, Oregon Health and Science Uni- wheezing (10), cancer (11), type 2 diabetes (12), and cardiovas- versity, Portland, OR 97239 cular disease (13), culminating in an increased risk for all-cause ORCIDs: 0000-0003-2555-2111 (M.R.); 0000-0003-3105-555X (N.E.M.). offspring mortality (14). Received for publication March 24, 2017. Accepted for publication August 7, 2017. The aforementioned observations strongly suggest that pregravid This work was supported by National Institutes of Health Grants KL2TR000152 (to obesity disrupts the development and maturation of the offspring’s N.E.M.) and R03AI112808 (to I.M.) and the National Center for Advancing Trans- immune system in utero. This hypothesis is supported by studies lational Sciences of the National Institutes of Health under Award UL1TR000128. in murine models that have shown detrimental effects on the Gene expression and methylation data have been submitted to the National Center for Biotechnology Information Sequence Read Archive (http://www.ncbi.nlm.nih.gov/ immune system of pups born to obese compared with lean dams sra) under accession number PRJNA398556. (15, 16). Specifically, pups born to obese dams generated disparate S.S., N.E.M., J.Q.P., K.L.T., and I.M. conceived and designed the experiments; S.S., IgG (smaller) and IgE (larger) Ab responses following vaccination R.M.W., and M.R. performed the experiments; S.S. and R.M.W. analyzed the data; with OVA (15). If these results are true in humans, they could S.S. and I.M. wrote the paper with input from N.E.M., J.Q.P., and K.L.T. explain the increased incidence of asthma (9) and wheezing (10) Address correspondence and reprint requests to Dr. Ilhem Messaoudi, 2400 Biolog- ical Sciences III, University of California Irvine, Irvine, CA 92697-3900. E-mail in children born to obese mothers. Additional rodent studies address: [email protected] reported greater morbidity and mortality following bacterial The online version of this article contains supplemental material. infection (Escherichia coli sepsis and methicillin-resistant Staphy- Abbreviations used in this article: BMI, body mass index; DEG, differentially expressed lococcus aureus infection) and greater susceptibility to auto- gene; DMC, differentially methylated cytosine; DMR, differentially methylated region; immune encephalitis in pups born to obese dams (16). Lastly, ex eDMR, empirical DMR; FC, fold change; FDR, false discovery rate; GO, gene ontology; RPKM, read per kilobase of transcript per million mapped reads; TF, tran- vivo LPS stimulation of colonic lamina propria lymphocytes scription factor; TSS, transcription start site; UCB, umbilical cord blood; UCBMC, resulted in increased secretion of inflammatory cytokines IL-6, UCB mononuclear cell. IL-1b, and IL-17, whereas LPS stimulation of splenocytes resulted Copyright Ó 2017 by The American Association of Immunologists, Inc. 0022-1767/17/$35.00 in decreased levels of TNF-a and IL-6 in pups born to obese dams www.jimmunol.org/cgi/doi/10.4049/jimmunol.1700434 2 EPIGENETIC IMPACT OF MATERNAL OBESITY ON CORD BLOOD MONOCYTE (16). Collectively, these data strongly suggest dysregulated im- Abs conjugated to magnetic microbeads per the manufacturer’s recom- munity in pups born to obese dams where aberrant responses (such mendations (Miltenyi Biotec, San Diego, CA). Magnetically bound UCB as to auto-antigens) are exaggerated, whereas protective anti- monocytes were washed and eluted for collection. Positive selection of UCB monocytes was chosen to ensure high purity of the population, microbial responses are reduced. Similarly, a baboon study which was assessed using flow cytometry and was $90% for all samples reported significant changes in the expression of genes involved in analyzed. Ag presentation, complement and coagulation cascade, leukocyte LPS stimulation and detection of soluble mediators migration, and BCR signaling in PBMCs collected from infants born to obese compared with lean dams (17). Purified UCB monocytes were plated at 2 3 105 per well and stimulated However, only a few studies have explored the impact of ma- with 100 ng (1 mg/ml) of LPS (TLR4 ligand, E. coli 055:B5; InvivoGen, ternal pregravid obesity on neonatal immune function in humans. San Diego, CA) or left untreated for 16 h at 37˚C and 5% CO2. The above concentration of LPS was previously standardized for robust detection of One study reported that children born to obese mothers have a 16- gene expression in primary human monocytes
Recommended publications
  • Architecture of a Lymphomyeloid Developmental Switch Controlled by PU.1, Notch and Gata3 Marissa Morales Del Real and Ellen V
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Caltech Authors DEVELOPMENT AND STEM CELLS RESEARCH ARTICLE 1207 Development 140, 1207-1219 (2013) doi:10.1242/dev.088559 © 2013. Published by The Company of Biologists Ltd Architecture of a lymphomyeloid developmental switch controlled by PU.1, Notch and Gata3 Marissa Morales Del Real and Ellen V. Rothenberg* SUMMARY Hematopoiesis is a classic system with which to study developmental potentials and to investigate gene regulatory networks that control choices among alternate lineages. T-cell progenitors seeding the thymus retain several lineage potentials. The transcription factor PU.1 is involved in the decision to become a T cell or a myeloid cell, and the developmental outcome of expressing PU.1 is dependent on exposure to Notch signaling. PU.1-expressing T-cell progenitors without Notch signaling often adopt a myeloid program, whereas those exposed to Notch signals remain in a T-lineage pathway. Here, we show that Notch signaling does not alter PU.1 transcriptional activity by degradation/alteration of PU.1 protein. Instead, Notch signaling protects against the downregulation of T-cell factors so that a T-cell transcriptional network is maintained. Using an early T-cell line, we describe two branches of this network. The first involves inhibition of E-proteins by PU.1 and the resulting inhibition of Notch signaling target genes. Effects of E- protein inhibition can be reversed by exposure to Notch signaling. The second network is dependent on the ability of PU.1 to inhibit important T-cell transcription factor genes such as Myb, Tcf7 and Gata3 in the absence of Notch signaling.
    [Show full text]
  • Supplementary Figure 1. Qrt-PCR Analyses for Measuring the Knockdown Efficiency of Transcription Factors. A549 and U937 Cells Kn
    Supplementary Figure 1. qRT-PCR analyses for measuring the knockdown efficiency of transcription factors. A549 and U937 cells knocking down or overexpressing different transcription factors including NF-κB subunits p65 and p50 (A), HIF1 (B), TCF4 (C), AP-1 subunits c-Jun and c-FOS (D), or FOXP3 (E) were subjected to RNA isolation, followed by qRT-PCR analyses to examine the expression of these transcription factors. **P<0.01 and ***P<0.001. 1 Supplementary Figure 2. Effects of FOXP3 downregulation and overexpression on the expression of NLRP1, IL1B and IL18. A549 and U937 cells were transfected with si-FOXP3 or pCDNA3-2×Flag-FOXP3. After 24 h, cells were subjected to RNA isolation, followed by qRT-PCR analyses to examine the expression of NLRP1 (A), IL1B (B) and IL18 (C). ***P<0.001. 2 Supplementary Figure 3. Effects of CtBP2 downregulation and overexpression on the expression of miR-199a-3p, NLRP1, IL1B and IL18. A549 and U937 cells were transfected with si-CtBP2 or pCDNA3-2×Flag-CtBP2. After 24 h, cells were subjected to RNA isolation, followed by qRT-PCR analyses to examine the expression of miR-199a-3p (A), NLRP1 (B), IL1B (C) and IL18 (D). ***P<0.001. 3 Supplementary Figure 4. CHFTC specifically bond to the promoter of miR-199a-3p. (A and B) Effects of CtBP2 knockdown and overexpression on HDAC1 and FOXP3 protein levels. A549 cells were transfected with two CtBP2-specific shRNAs and pCDNA3-2×Flag-CtBP2 to obtain two CtBP2-knockdown cell lines (KD-1 and KD-2) (A) and two CtBP2-overexpression cells lines (OE-1 and OE-2) (B), respectively.
    [Show full text]
  • Gene PMID WBS Locus ABR 26603386 AASDH 26603386
    Supplementary material J Med Genet Gene PMID WBS Locus ABR 26603386 AASDH 26603386 ABCA1 21304579 ABCA13 26603386 ABCA3 25501393 ABCA7 25501393 ABCC1 25501393 ABCC3 25501393 ABCG1 25501393 ABHD10 21304579 ABHD11 25501393 yes ABHD2 25501393 ABHD5 21304579 ABLIM1 21304579;26603386 ACOT12 25501393 ACSF2,CHAD 26603386 ACSL4 21304579 ACSM3 26603386 ACTA2 25501393 ACTN1 26603386 ACTN3 26603386;25501393;25501393 ACTN4 21304579 ACTR1B 21304579 ACVR2A 21304579 ACY3 19897463 ACYP1 21304579 ADA 25501393 ADAM12 21304579 ADAM19 25501393 ADAM32 26603386 ADAMTS1 25501393 ADAMTS10 25501393 ADAMTS12 26603386 ADAMTS17 26603386 ADAMTS6 21304579 ADAMTS7 25501393 ADAMTSL1 21304579 ADAMTSL4 25501393 ADAMTSL5 25501393 ADCY3 25501393 ADK 21304579 ADRBK2 25501393 AEBP1 25501393 AES 25501393 AFAP1,LOC84740 26603386 AFAP1L2 26603386 AFG3L1 21304579 AGAP1 26603386 AGAP9 21304579 Codina-Sola M, et al. J Med Genet 2019; 56:801–808. doi: 10.1136/jmedgenet-2019-106080 Supplementary material J Med Genet AGBL5 21304579 AGPAT3 19897463;25501393 AGRN 25501393 AGXT2L2 25501393 AHCY 25501393 AHDC1 26603386 AHNAK 26603386 AHRR 26603386 AIDA 25501393 AIFM2 21304579 AIG1 21304579 AIP 21304579 AK5 21304579 AKAP1 25501393 AKAP6 21304579 AKNA 21304579 AKR1E2 26603386 AKR7A2 21304579 AKR7A3 26603386 AKR7L 26603386 AKT3 21304579 ALDH18A1 25501393;25501393 ALDH1A3 21304579 ALDH1B1 21304579 ALDH6A1 21304579 ALDOC 21304579 ALG10B 26603386 ALG13 21304579 ALKBH7 25501393 ALPK2 21304579 AMPH 21304579 ANG 21304579 ANGPTL2,RALGPS1 26603386 ANGPTL6 26603386 ANK2 21304579 ANKMY1 26603386 ANKMY2
    [Show full text]
  • Supplemental Materials ZNF281 Enhances Cardiac Reprogramming
    Supplemental Materials ZNF281 enhances cardiac reprogramming by modulating cardiac and inflammatory gene expression Huanyu Zhou, Maria Gabriela Morales, Hisayuki Hashimoto, Matthew E. Dickson, Kunhua Song, Wenduo Ye, Min S. Kim, Hanspeter Niederstrasser, Zhaoning Wang, Beibei Chen, Bruce A. Posner, Rhonda Bassel-Duby and Eric N. Olson Supplemental Table 1; related to Figure 1. Supplemental Table 2; related to Figure 1. Supplemental Table 3; related to the “quantitative mRNA measurement” in Materials and Methods section. Supplemental Table 4; related to the “ChIP-seq, gene ontology and pathway analysis” and “RNA-seq” and gene ontology analysis” in Materials and Methods section. Supplemental Figure S1; related to Figure 1. Supplemental Figure S2; related to Figure 2. Supplemental Figure S3; related to Figure 3. Supplemental Figure S4; related to Figure 4. Supplemental Figure S5; related to Figure 6. Supplemental Table S1. Genes included in human retroviral ORF cDNA library. Gene Gene Gene Gene Gene Gene Gene Gene Symbol Symbol Symbol Symbol Symbol Symbol Symbol Symbol AATF BMP8A CEBPE CTNNB1 ESR2 GDF3 HOXA5 IL17D ADIPOQ BRPF1 CEBPG CUX1 ESRRA GDF6 HOXA6 IL17F ADNP BRPF3 CERS1 CX3CL1 ETS1 GIN1 HOXA7 IL18 AEBP1 BUD31 CERS2 CXCL10 ETS2 GLIS3 HOXB1 IL19 AFF4 C17ORF77 CERS4 CXCL11 ETV3 GMEB1 HOXB13 IL1A AHR C1QTNF4 CFL2 CXCL12 ETV7 GPBP1 HOXB5 IL1B AIMP1 C21ORF66 CHIA CXCL13 FAM3B GPER HOXB6 IL1F3 ALS2CR8 CBFA2T2 CIR1 CXCL14 FAM3D GPI HOXB7 IL1F5 ALX1 CBFA2T3 CITED1 CXCL16 FASLG GREM1 HOXB9 IL1F6 ARGFX CBFB CITED2 CXCL3 FBLN1 GREM2 HOXC4 IL1F7
    [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]
  • Determining HDAC8 Substrate Specificity by Noah Ariel Wolfson A
    Determining HDAC8 substrate specificity by Noah Ariel Wolfson A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy (Biological Chemistry) in the University of Michigan 2014 Doctoral Committee: Professor Carol A. Fierke, Chair Professor Robert S. Fuller Professor Anna K. Mapp Associate Professor Patrick J. O’Brien Associate Professor Raymond C. Trievel Dedication My thesis is dedicated to all my family, mentors, and friends who made getting to this point possible. ii Table of Contents Dedication ....................................................................................................................................... ii List of Figures .............................................................................................................................. viii List of Tables .................................................................................................................................. x List of Appendices ......................................................................................................................... xi Abstract ......................................................................................................................................... xii Chapter 1 HDAC8 substrates: Histones and beyond ...................................................................... 1 Overview ..................................................................................................................................... 1 HDAC introduction
    [Show full text]
  • A Private 16Q24.2Q24.3 Microduplication in a Boy with Intellectual Disability, Speech Delay and Mild Dysmorphic Features
    G C A T T A C G G C A T genes Article A Private 16q24.2q24.3 Microduplication in a Boy with Intellectual Disability, Speech Delay and Mild Dysmorphic Features Orazio Palumbo * , Pietro Palumbo , Ester Di Muro, Luigia Cinque, Antonio Petracca, Massimo Carella and Marco Castori Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; [email protected] (P.P.); [email protected] (E.D.M.); [email protected] (L.C.); [email protected] (A.P.); [email protected] (M.C.); [email protected] (M.C.) * Correspondence: [email protected]; Tel.: +39-088-241-6350 Received: 5 June 2020; Accepted: 24 June 2020; Published: 26 June 2020 Abstract: No data on interstitial microduplications of the 16q24.2q24.3 chromosome region are available in the medical literature and remain extraordinarily rare in public databases. Here, we describe a boy with a de novo 16q24.2q24.3 microduplication at the Single Nucleotide Polymorphism (SNP)-array analysis spanning ~2.2 Mb and encompassing 38 genes. The patient showed mild-to-moderate intellectual disability, speech delay and mild dysmorphic features. In DECIPHER, we found six individuals carrying a “pure” overlapping microduplication. Although available data are very limited, genomic and phenotype comparison of our and previously annotated patients suggested a potential clinical relevance for 16q24.2q24.3 microduplication with a variable and not (yet) recognizable phenotype predominantly affecting cognition. Comparing the cytogenomic data of available individuals allowed us to delineate the smallest region of overlap involving 14 genes. Accordingly, we propose ANKRD11, CDH15, and CTU2 as candidate genes for explaining the related neurodevelopmental manifestations shared by these patients.
    [Show full text]
  • Supplementary Table S4. FGA Co-Expressed Gene List in LUAD
    Supplementary Table S4. FGA co-expressed gene list in LUAD tumors Symbol R Locus Description FGG 0.919 4q28 fibrinogen gamma chain FGL1 0.635 8p22 fibrinogen-like 1 SLC7A2 0.536 8p22 solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 DUSP4 0.521 8p12-p11 dual specificity phosphatase 4 HAL 0.51 12q22-q24.1histidine ammonia-lyase PDE4D 0.499 5q12 phosphodiesterase 4D, cAMP-specific FURIN 0.497 15q26.1 furin (paired basic amino acid cleaving enzyme) CPS1 0.49 2q35 carbamoyl-phosphate synthase 1, mitochondrial TESC 0.478 12q24.22 tescalcin INHA 0.465 2q35 inhibin, alpha S100P 0.461 4p16 S100 calcium binding protein P VPS37A 0.447 8p22 vacuolar protein sorting 37 homolog A (S. cerevisiae) SLC16A14 0.447 2q36.3 solute carrier family 16, member 14 PPARGC1A 0.443 4p15.1 peroxisome proliferator-activated receptor gamma, coactivator 1 alpha SIK1 0.435 21q22.3 salt-inducible kinase 1 IRS2 0.434 13q34 insulin receptor substrate 2 RND1 0.433 12q12 Rho family GTPase 1 HGD 0.433 3q13.33 homogentisate 1,2-dioxygenase PTP4A1 0.432 6q12 protein tyrosine phosphatase type IVA, member 1 C8orf4 0.428 8p11.2 chromosome 8 open reading frame 4 DDC 0.427 7p12.2 dopa decarboxylase (aromatic L-amino acid decarboxylase) TACC2 0.427 10q26 transforming, acidic coiled-coil containing protein 2 MUC13 0.422 3q21.2 mucin 13, cell surface associated C5 0.412 9q33-q34 complement component 5 NR4A2 0.412 2q22-q23 nuclear receptor subfamily 4, group A, member 2 EYS 0.411 6q12 eyes shut homolog (Drosophila) GPX2 0.406 14q24.1 glutathione peroxidase
    [Show full text]
  • Systematic Integration of GATA Transcription Factors and Epigenomes Via IDEAS Paints the Regulatory Landscape of Hematopoietic Cells
    Received: 2 July 2019 Accepted: 17 October 2019 DOI: 10.1002/iub.2195 CRITICAL REVIEW Systematic integration of GATA transcription factors and epigenomes via IDEAS paints the regulatory landscape of hematopoietic cells Ross C. Hardison1 | Yu Zhang1 | Cheryl A. Keller1 | Guanjue Xiang1 | Elisabeth F. Heuston2 | Lin An1 | Jens Lichtenberg2 | Belinda M. Giardine1 | David Bodine2 | Shaun Mahony1 | Qunhua Li1 | Feng Yue3 | Mitchell J. Weiss4 | Gerd A. Blobel5 | James Taylor6 | Jim Hughes7 | Douglas R. Higgs7 | Berthold Göttgens8 1Departments of Biochemistry and Molecular Biology and of Statistics, The Abstract Pennsylvania State University, University Members of the GATA family of transcription factors play key roles in the dif- Park, PA ferentiation of specific cell lineages by regulating the expression of target 2 Genetics and Molecular Biology Branch, genes. Three GATA factors play distinct roles in hematopoietic differentiation. Hematopoiesis Section, National Institutes of Health, NHGRI, Bethesda, MD In order to better understand how these GATA factors function to regulate 3Department of Biochemistry and genes throughout the genome, we are studying the epigenomic and transcrip- Molecular Biology, The Pennsylvania State tional landscapes of hematopoietic cells in a model-driven, integrative fashion. University College of Medicine, Hershey, PA We have formed the collaborative multi-lab VISION project to conduct ValI- 4Hematology Department, St. Jude dated Systematic IntegratiON of epigenomic data in mouse and human hema- Children's Research Hospital, topoiesis. The epigenomic data included nuclease accessibility in chromatin, Memphis, TN CTCF occupancy, and histone H3 modifications for 20 cell types covering 5Children's Hospital of Philadelphia, hematopoietic stem cells, multilineage progenitor cells, and mature cells across Philadelphia, PA 6Departments of Biology and of Computer the blood cell lineages of mouse.
    [Show full text]
  • Genomic Profiling of Short- and Long-Term Caloric Restriction Effects in the Liver of Aging Mice
    Genomic profiling of short- and long-term caloric restriction effects in the liver of aging mice Shelley X. Cao, Joseph M. Dhahbi, Patricia L. Mote, and Stephen R. Spindler* Department of Biochemistry, University of California, Riverside, CA 92521 Edited by Bruce N. Ames, University of California, Berkeley, CA, and approved July 11, 2001 (received for review June 19, 2001) We present genome-wide microarray expression analysis of 11,000 aging and CR on gene expression. Control young (7-month-old; n ϭ genes in an aging potentially mitotic tissue, the liver. This organ has 3) and old (27-month-old; n ϭ 3) mice were fed 95 kcal of a a major impact on health and homeostasis during aging. The effects semipurified control diet (Harlan Teklad, Madison, WI; no. of life- and health-span-extending caloric restriction (CR) on gene TD94145) per week after weaning. Long-term CR (LT-CR) young expression among young and old mice and between long-term CR (7-month-old; n ϭ 3) and old (27-month-old; n ϭ 3) mice were fed (LT-CR) and short-term CR (ST-CR) were examined. This experimental 53 kcal of a semipurified CR diet (Harlan Teklad; no. TD94146) per design allowed us to accurately distinguish the effects of aging from week after weaning. Short-term CR (ST-CR) mice were 34-month- those of CR on gene expression. Aging was accompanied by changes old control mice that were switched to 80 kcal of CR diet for 2 in gene expression associated with increased inflammation, cellular weeks, followed by 53 kcal for 2 weeks (n ϭ 3).
    [Show full text]
  • Classification and Evolution of P-Loop Gtpases and Related Atpases Detlefd.Leipe,Yurii.Wolf,Eugenev.Koonin*Andl.Aravind
    doi:10.1006/jmbi.2001.5378availableonlineathttp://www.idealibrary.comon J. Mol. Biol. (2002) 317, 41±72 Classification and Evolution of P-loop GTPases and Related ATPases DetlefD.Leipe,YuriI.Wolf,EugeneV.Koonin*andL.Aravind National Center for Sequences and available structures were compared for all the widely dis- Biotechnology Information tributed representatives of the P-loop GTPases and GTPase-related pro- National Library of Medicine teins with the aim of constructing an evolutionary classi®cation for this National Institutes of Health superclass of proteins and reconstructing the principal events in their Bethesda, MD 20894, USA evolution. The GTPase superclass can be divided into two large classes, each of which has a unique set of sequence and structural signatures (synapomorphies). The ®rst class, designated TRAFAC (after translation factors) includes enzymes involved in translation (initiation, elongation, and release factors), signal transduction (in particular, the extended Ras- like family), cell motility, and intracellular transport. The second class, designated SIMIBI (after signal recognition particle, MinD, and BioD), consists of signal recognition particle (SRP) GTPases, the assemblage of MinD-like ATPases, which are involved in protein localization, chromo- some partitioning, and membrane transport, and a group of metabolic enzymes with kinase or related phosphate transferase activity. These two classes together contain over 20 distinct families that are further subdi- vided into 57 subfamilies (ancient lineages) on the basis of conserved sequence motifs, shared structural features, and domain architectures. Ten subfamilies show a universal phyletic distribution compatible with presence in the last universal common ancestor of the extant life forms (LUCA). These include four translation factors, two OBG-like GTPases, the YawG/YlqF-like GTPases (these two subfamilies also consist of pre- dicted translation factors), the two signal-recognition-associated GTPases, and the MRP subfamily of MinD-like ATPases.
    [Show full text]
  • ZNF19 (NM 006961) Human Tagged ORF Clone Lentiviral Particle Product Data
    OriGene Technologies, Inc. 9620 Medical Center Drive, Ste 200 Rockville, MD 20850, US Phone: +1-888-267-4436 [email protected] EU: [email protected] CN: [email protected] Product datasheet for RC208208L3V ZNF19 (NM_006961) Human Tagged ORF Clone Lentiviral Particle Product data: Product Type: Lentiviral Particles Product Name: ZNF19 (NM_006961) Human Tagged ORF Clone Lentiviral Particle Symbol: ZNF19 Synonyms: KOX12 Vector: pLenti-C-Myc-DDK-P2A-Puro (PS100092) ACCN: NM_006961 ORF Size: 1374 bp ORF Nucleotide The ORF insert of this clone is exactly the same as(RC208208). Sequence: OTI Disclaimer: The molecular sequence of this clone aligns with the gene accession number as a point of reference only. However, individual transcript sequences of the same gene can differ through naturally occurring variations (e.g. polymorphisms), each with its own valid existence. This clone is substantially in agreement with the reference, but a complete review of all prevailing variants is recommended prior to use. More info OTI Annotation: This clone was engineered to express the complete ORF with an expression tag. Expression varies depending on the nature of the gene. RefSeq: NM_006961.3 RefSeq Size: 2743 bp RefSeq ORF: 1377 bp Locus ID: 7567 UniProt ID: P17023 Protein Families: Transcription Factors MW: 52.5 kDa Gene Summary: The protein encoded by this gene contains a zinc finger, a nucleic acid-binding domain present in many transcription factors. This gene is located in a region next to ZNF23, a gene also encoding a zinc finger protein, on chromosome 16. [provided by RefSeq, Jul 2008] This product is to be used for laboratory only.
    [Show full text]