Supplement Table 1: Genes Downregualted by CSA in Skin And

Total Page:16

File Type:pdf, Size:1020Kb

Supplement Table 1: Genes Downregualted by CSA in Skin And Supplement Table 1: Genes downregualted by CSA in skin and associated cell types Associated cell types: Symbol resting cells cytokine activation T cell activation Description PITPNB _ T cell+PBMC act. phosphatidylinositol transfer protein, beta IDH3A _ T cell+PBMC act. isocitrate dehydrogenase 3 (NAD+) alpha RCC1 _ T cell+PBMC act. regulator of chromosome condensation 1 BZW1 _ T cell+PBMC act. basic leucine zipper and W2 domains 1 SYNCRIP _ T cell+PBMC act. synaptotagmin binding, cytoplasmic RNA interacting protein CXCL1 _ IL-17 *, TNF PBMC act. chemokine (C-X-C motif) ligand 1 CXCL11 _ IFN PBMC act. chemokine (C-X-C motif) ligand 11 CXCL9 _ IFN PBMC act. chemokine (C-X-C motif) ligand 9 CXCL10 _ IFN PBMC act. chemokine (C-X-C motif) ligand 10 MMP12 _ PBMC act. matrix metallopeptidase 12 (macrophage elastase) TGM3 _ PBMC act. transglutaminase 3 CCNE2 _ PBMC act. cyclin E2 RRM2 _ PBMC act. ribonucleotide reductase M2 polypeptide DHFR _ PBMC act. dihydrofolate reductase RABGGTA _ TNF Rab geranylgeranyltransferase, alpha subunit DEFB4 _ IFN defensin, beta 4 TAP2 _ IFN transporter 2, ATP-binding cassette, sub-family B (MDR/TAP) LCN2 _ IL-17* lipocalin 2 (oncogene 24p3) TRIP13 common Fb+KC T cell+PBMC act. thyroid hormone receptor interactor 13 RPP40 common Fb+KC T cell+PBMC act. ribonuclease P 40kDa subunit TYMS common Fb+KC PBMC act. thymidylate synthetase CDC6 common Fb+KC PBMC act. cell division cycle 6 homolog (S. cerevisiae) TOP2A common Fb+KC PBMC act. topoisomerase (DNA) II alpha 170kDa CDC2 common Fb+KC PBMC act. cell division cycle 2, G1 to S and G2 to M CCNA2 common Fb+KC PBMC act. cyclin A2 CCNB1 common Fb+KC PBMC act. cyclin B1 PHLDA2 common Fb+KC PBMC act. pleckstrin homology-like domain, family A, member 2 ZWINT common Fb+KC PBMC act. ZW10 interactor KIAA0101 common Fb+KC PBMC act. KIAA0101 KIF11 common Fb+KC PBMC act. kinesin family member 11 TK1 common Fb+KC PBMC act. thymidine kinase 1, soluble MCM4 common Fb+KC PBMC act. MCM4 minichromosome maintenance deficient 4 (S. cerevisiae) OSMR common Fb+KC IFN KC+IFN oncostatin M receptor SERPINB7 common Fb+KC serpin peptidase inhibitor, clade B (ovalbumin), member 7 ALDH1A3 common Fb+KC aldehyde dehydrogenase 1 family, member A3 CASP7 common iDC+mDC TNF PBMC act. caspase 7, apoptosis-related cysteine peptidase NOS2A common iDC+mDC nitric oxide synthase 2A (inducible, hepatocytes) BCL2A1 common myeloid T cell+PBMC act. BCL2-related protein A1 NP common myeloid T cell+PBMC act. nucleoside phosphorylase ECGF1 common myeloid TNF, IFN PBMC act. endothelial cell growth factor 1 (platelet-derived) IL1B common myeloid IL-17^, TNF PBMC act. interleukin 1, beta MMP9 common myeloid TNF PBMC act. matrix metallopeptidase 9 SOD2 common myeloid TNF PBMC act. superoxide dismutase 2, mitochondrial PLAUR common myeloid IFN PBMC act. plasminogen activator, urokinase receptor CTSL1 common myeloid IFN PBMC act. cathepsin L1 ALAS1 common myeloid TNF PBMC act. aminolevulinate, delta-, synthase 1 CCL18 common myeloid PBMC act. chemokine (C-C motif) ligand 18 PTPRE common myeloid PBMC act. protein tyrosine phosphatase, receptor type, E GM2A common myeloid PBMC act. GM2 ganglioside activator LILRB4 common myeloid PBMC act. leukocyte immunoglobulin-like receptor, subfamily B UPP1 common myeloid PBMC act. uridine phosphorylase 1 GK common myeloid PBMC act. glycerol kinase GK3P common myeloid PBMC act. glycerol kinase 3 pseudogene KYNU common myeloid PBMC act. kynureninase (L-kynurenine hydrolase) GPX3 common myeloid PBMC act. glutathione peroxidase 3 (plasma) CTSC common myeloid TNF, IFN cathepsin C LYN common myeloid TNF, IFN v-yes-1 Yamaguchi sarcoma viral related oncogene homolog CXCL2 common myeloid TNF, IFN chemokine (C-X-C motif) ligand 2 MALT1 common myeloid IFN mucosa associated lymphoid tissue lymphoma translocation gene 1 CASP1 common myeloid IFN caspase 1, apoptosis-related cysteine peptidase ACP5 common myeloid IFN acid phosphatase 5, tartrate resistant CD14 common myeloid CD14 molecule SERPINA1 common myeloid serpin peptidase inhibitor, clade A FCER1G common myeloid Fc fragment of IgE, high affinity I, receptor for; gamma polypeptide CYBB common myeloid cytochrome b-245, beta polypeptide AMPD3 common myeloid adenosine monophosphate deaminase (isoform E) C1QB common myeloid complement component 1, q subcomponent, B chain MYO5A common myeloid myosin VA (heavy chain 12, myoxin) S100A9 common myeloid S100 calcium binding protein A9 LPHN2 Fb latrophilin 2 PLAT Fb PBMC act. plasminogen activator, tissue HSPA4 housekeeping T cell+PBMC act. heat shock 70kDa protein 4 housekeeping T cell+PBMC act. MRPS12 housekeeping T cell+PBMC act. mitochondrial ribosomal protein S12 UCK2 housekeeping T cell+PBMC act. uridine-cytidine kinase 2 AHCY housekeeping T cell+PBMC act. S-adenosylhomocysteine hydrolase TSR1 housekeeping T cell+PBMC act. TSR1, 20S rRNA accumulation, homolog (S. cerevisiae) YKT6 housekeeping T cell+PBMC act. YKT6 v-SNARE homolog (S. cerevisiae) CD47 housekeeping IFN T cell act. CD47 molecule CEBPG housekeeping T cell act. CCAAT/enhancer binding protein (C/EBP), gamma PGK1 housekeeping IFN PBMC act. phosphoglycerate kinase 1 WARS housekeeping IFN PBMC act. tryptophanyl-tRNA synthetase APOL1 housekeeping IFN PBMC act. apolipoprotein L, 1 CSTB housekeeping PBMC act. cystatin B (stefin B) CCNE1 housekeeping PBMC act. cyclin E1 PCNA housekeeping proliferating cell nuclear antigen CHI3L2 housekeeping chitinase 3-like 2 VRK2 housekeeping vaccinia related kinase 2 SSR1 housekeeping signal sequence receptor, alpha DPAGT1 housekeeping dolichyl-phosphate (UDP-N-acetylglucosamine) KLK13 housekeeping kallikrein-related peptidase 13 SLC23A2 housekeeping solute carrier family 23 (nucleobase transporters), member 2 SLC25A13 housekeeping solute carrier family 25, member 13 (citrin) DUSP3 housekeeping dual specificity phosphatase 3 HIPK3 housekeeping homeodomain interacting protein kinase 3 PDXK iDC T cell+PBMC act. pyridoxal (pyridoxine, vitamin B6) kinase CD53 iDC CD53 molecule IL1RN iDC PBMC act. interleukin 1 receptor antagonist HMGCS1 KC T cell+PBMC act. 3-hydroxy-3-methylglutaryl-Coenzyme A synthase 1 (soluble) SLC16A1 KC T cell+PBMC act. solute carrier family 16, member 1 FEN1 KC T cell+PBMC act. flap structure-specific endonuclease 1 EVA1 KC T cell act. epithelial V-like antigen 1 MCM2 KC PBMC act. MCM2 minichromosome maintenance deficient 2, mitotin KIF2C KC PBMC act. kinesin family member 2C PTTG1 KC PBMC act. pituitary tumor-transforming 1 MALL KC IFN mal, T-cell differentiation protein-like XDH KC IFN xanthine dehydrogenase VSNL1 KC IFN visinin-like 1 SERPINB3 KC serpin peptidase inhibitor, clade B (ovalbumin), member 3 EPHA2 KC EPH receptor A2 CDH3 KC cadherin 3, type 1, P-cadherin (placental) S100A2 KC S100 calcium binding protein A2 ABCA12 KC ATP-binding cassette, sub-family A (ABC1), member 12 CKMT1B KC creatine kinase, mitochondrial 1B SLPI KC secretory leukocyte peptidase inhibitor PKP1 KC plakophilin 1 (ectodermal dysplasia/skin fragility syndrome) ATP12A KC ATPase, H+/K+ transporting, nongastric, alpha polypeptide PRSS1 KC protease, serine, 1 (trypsin 1) ALOX12B KC arachidonate 12-lipoxygenase, 12R type C14orf147 KC chromosome 14 open reading frame 147 SPRR1A KC small proline-rich protein 1A DSG3 KC desmoglein 3 (pemphigus vulgaris antigen) KRT17 KC keratin 17 AASS KC aminoadipate-semialdehyde synthase TGM1 KC transglutaminase 1 ETHE1 KC ethylmalonic encephalopathy 1 KLK10 KC kallikrein-related peptidase 10 SPRR1B KC small proline-rich protein 1B (cornifin) KRT16 KC keratin 16 (focal non-epidermolytic palmoplantar keratoderma) AKR1B10 KC aldo-keto reductase family 1, member B10 (aldose reductase) MDFI KC MyoD family inhibitor FGFBP1 KC fibroblast growth factor binding protein 1 KRT6C KC keratin 6C DSC2 KC desmocollin 2 PRSS3 KC protease, serine, 3 (mesotrypsin) PI3 KC peptidase inhibitor 3, skin-derived (SKALP) SPRR2C KC small proline-rich protein 2C SPRR2D KC small proline-rich protein 2D CFLAR leukocytes TNF, IFN T cell+PBMC act. CASP8 and FADD-like apoptosis regulator CCL20 leukocytes TNF T cell+PBMC act. chemokine (C-C motif) ligand 20 PBEF1 leukocytes T cell+PBMC act. pre-B-cell colony enhancing factor 1 CCL4 leukocytes T cell+PBMC act. chemokine (C-C motif) ligand 4 GZMB leukocytes T cell+PBMC act. granzyme B LCP2 leukocytes T cell+PBMC act. lymphocyte cytosolic protein 2 PSMB10 leukocytes TNF, IFN PBMC act. proteasome (prosome, macropain) subunit, beta type, 10 IRF1 leukocytes TNF, IFN PBMC act. interferon regulatory factor 1 IL8 leukocytes TNF PBMC act. interleukin 8 GBP1 leukocytes IFN PBMC act. guanylate binding protein 1, interferon-inducible, 67kDa RAB27A leukocytes IFN PBMC act. RAB27A, member RAS oncogene family SUB1 leukocytes PBMC act. SUB1 homolog (S. cerevisiae) RGS1 leukocytes PBMC act. regulator of G-protein signalling 1 KIF2A leukocytes PBMC act. kinesin heavy chain member 2A STAT3 leukocytes IFN signal transducer and activator of transcription 3 ZFP36 leukocytes IFN zinc finger protein 36, C3H type, homolog (mouse) IL7R leukocytes interleukin 7 receptor PTPN22 leukocytes protein tyrosine phosphatase, non-receptor type 22 (lymphoid) CD3D leukocytes CD3d molecule, delta (CD3-TCR complex) C1orf9 leukocytes chromosome 1 open reading frame 9 STK17A leukocytes serine/threonine kinase 17a AMD1 macrophages T-cell+PBMCact adenosylmethionine decarboxylase 1 AGPS macrophages T-cell+PBMCact alkylglycerone phosphate synthase HK2 macrophages T-cell+PBMCact hexokinase 2 RALA macrophages IFN PBMCact,KC+IFN v-ral simian leukemia viral oncogene homolog A (ras related) RAC2 macrophages PBMCact ras-related C3 botulinum toxin substrate 2 RAB31 macrophages RAB31, member RAS oncogene family ADAMDEC1 macrophages ADAM-like, decysin 1 CD83 mDC TNF T cell+PBMC act. CD83 molecule CCL3 mDC T cell+PBMC act. chemokine (C-C motif) ligand 3 MPHOSPH6 mDC T cell+PBMC act. M-phase phosphoprotein 6 N4BP1 mDC TNF, IFN PBMCact,KC+TNFand IFN Nedd4 binding protein 1 UBD mDC IFN PBMC act. ubiquitin D IL13RA1 mDC TNF interleukin 13 receptor, alpha 1 CFB mDC IFN complement factor B IL4R mDC IFN interleukin 4 receptor TOP1 monocytes IFN T cell+PBMC act. topoisomerase (DNA) I PTP4A1 monocytes T cell+PBMC act.
Recommended publications
  • The Role of Nuclear Lamin B1 in Cell Proliferation and Senescence
    Downloaded from genesdev.cshlp.org on September 29, 2021 - Published by Cold Spring Harbor Laboratory Press The role of nuclear lamin B1 in cell proliferation and senescence Takeshi Shimi,1 Veronika Butin-Israeli,1 Stephen A. Adam,1 Robert B. Hamanaka,2 Anne E. Goldman,1 Catherine A. Lucas,1 Dale K. Shumaker,1 Steven T. Kosak,1 Navdeep S. Chandel,2 and Robert D. Goldman1,3 1Department of Cell and Molecular Biology, 2Department of Medicine, Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA Nuclear lamin B1 (LB1) is a major structural component of the nucleus that appears to be involved in the regulation of many nuclear functions. The results of this study demonstrate that LB1 expression in WI-38 cells decreases during cellular senescence. Premature senescence induced by oncogenic Ras also decreases LB1 expression through a retinoblastoma protein (pRb)-dependent mechanism. Silencing the expression of LB1 slows cell proliferation and induces premature senescence in WI-38 cells. The effects of LB1 silencing on proliferation require the activation of p53, but not pRb. However, the induction of premature senescence requires both p53 and pRb. The proliferation defects induced by silencing LB1 are accompanied by a p53-dependent reduction in mitochondrial reactive oxygen species (ROS), which can be rescued by growth under hypoxic conditions. In contrast to the effects of LB1 silencing, overexpression of LB1 increases the proliferation rate and delays the onset of senescence of WI-38 cells. This overexpression eventually leads to cell cycle arrest at the G1/S boundary.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Supplementary Materials
    1 Supplementary Materials: Supplemental Figure 1. Gene expression profiles of kidneys in the Fcgr2b-/- and Fcgr2b-/-. Stinggt/gt mice. (A) A heat map of microarray data show the genes that significantly changed up to 2 fold compared between Fcgr2b-/- and Fcgr2b-/-. Stinggt/gt mice (N=4 mice per group; p<0.05). Data show in log2 (sample/wild-type). 2 Supplemental Figure 2. Sting signaling is essential for immuno-phenotypes of the Fcgr2b-/-lupus mice. (A-C) Flow cytometry analysis of splenocytes isolated from wild-type, Fcgr2b-/- and Fcgr2b-/-. Stinggt/gt mice at the age of 6-7 months (N= 13-14 per group). Data shown in the percentage of (A) CD4+ ICOS+ cells, (B) B220+ I-Ab+ cells and (C) CD138+ cells. Data show as mean ± SEM (*p < 0.05, **p<0.01 and ***p<0.001). 3 Supplemental Figure 3. Phenotypes of Sting activated dendritic cells. (A) Representative of western blot analysis from immunoprecipitation with Sting of Fcgr2b-/- mice (N= 4). The band was shown in STING protein of activated BMDC with DMXAA at 0, 3 and 6 hr. and phosphorylation of STING at Ser357. (B) Mass spectra of phosphorylation of STING at Ser357 of activated BMDC from Fcgr2b-/- mice after stimulated with DMXAA for 3 hour and followed by immunoprecipitation with STING. (C) Sting-activated BMDC were co-cultured with LYN inhibitor PP2 and analyzed by flow cytometry, which showed the mean fluorescence intensity (MFI) of IAb expressing DC (N = 3 mice per group). 4 Supplemental Table 1. Lists of up and down of regulated proteins Accession No.
    [Show full text]
  • To Study Mutant P53 Gain of Function, Various Tumor-Derived P53 Mutants
    Differential effects of mutant TAp63γ on transactivation of p53 and/or p63 responsive genes and their effects on global gene expression. A thesis submitted in partial fulfillment of the requirements for the degree of Master of Science By Shama K Khokhar M.Sc., Bilaspur University, 2004 B.Sc., Bhopal University, 2002 2007 1 COPYRIGHT SHAMA K KHOKHAR 2007 2 WRIGHT STATE UNIVERSITY SCHOOL OF GRADUATE STUDIES Date of Defense: 12-03-07 I HEREBY RECOMMEND THAT THE THESIS PREPARED UNDER MY SUPERVISION BY SHAMA KHAN KHOKHAR ENTITLED Differential effects of mutant TAp63γ on transactivation of p53 and/or p63 responsive genes and their effects on global gene expression BE ACCEPTED IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF Master of Science Madhavi P. Kadakia, Ph.D. Thesis Director Daniel Organisciak , Ph.D. Department Chair Committee on Final Examination Madhavi P. Kadakia, Ph.D. Steven J. Berberich, Ph.D. Michael Leffak, Ph.D. Joseph F. Thomas, Jr., Ph.D. Dean, School of Graduate Studies 3 Abstract Khokhar, Shama K. M.S., Department of Biochemistry and Molecular Biology, Wright State University, 2007 Differential effect of TAp63γ mutants on transactivation of p53 and/or p63 responsive genes and their effects on global gene expression. p63, a member of the p53 gene family, known to play a role in development, has more recently also been implicated in cancer progression. Mice lacking p63 exhibit severe developmental defects such as limb truncations, abnormal skin, and absence of hair follicles, teeth, and mammary glands. Germline missense mutations of p63 have been shown to be responsible for several human developmental syndromes including SHFM, EEC and ADULT syndromes and are associated with anomalies in the development of organs of epithelial origin.
    [Show full text]
  • 4-6 Weeks Old Female C57BL/6 Mice Obtained from Jackson Labs Were Used for Cell Isolation
    Methods Mice: 4-6 weeks old female C57BL/6 mice obtained from Jackson labs were used for cell isolation. Female Foxp3-IRES-GFP reporter mice (1), backcrossed to B6/C57 background for 10 generations, were used for the isolation of naïve CD4 and naïve CD8 cells for the RNAseq experiments. The mice were housed in pathogen-free animal facility in the La Jolla Institute for Allergy and Immunology and were used according to protocols approved by the Institutional Animal Care and use Committee. Preparation of cells: Subsets of thymocytes were isolated by cell sorting as previously described (2), after cell surface staining using CD4 (GK1.5), CD8 (53-6.7), CD3ε (145- 2C11), CD24 (M1/69) (all from Biolegend). DP cells: CD4+CD8 int/hi; CD4 SP cells: CD4CD3 hi, CD24 int/lo; CD8 SP cells: CD8 int/hi CD4 CD3 hi, CD24 int/lo (Fig S2). Peripheral subsets were isolated after pooling spleen and lymph nodes. T cells were enriched by negative isolation using Dynabeads (Dynabeads untouched mouse T cells, 11413D, Invitrogen). After surface staining for CD4 (GK1.5), CD8 (53-6.7), CD62L (MEL-14), CD25 (PC61) and CD44 (IM7), naïve CD4+CD62L hiCD25-CD44lo and naïve CD8+CD62L hiCD25-CD44lo were obtained by sorting (BD FACS Aria). Additionally, for the RNAseq experiments, CD4 and CD8 naïve cells were isolated by sorting T cells from the Foxp3- IRES-GFP mice: CD4+CD62LhiCD25–CD44lo GFP(FOXP3)– and CD8+CD62LhiCD25– CD44lo GFP(FOXP3)– (antibodies were from Biolegend). In some cases, naïve CD4 cells were cultured in vitro under Th1 or Th2 polarizing conditions (3, 4).
    [Show full text]
  • Santa Cruz Biotechnology, Inc. 1.800.457.3801 831.457.3800 Fax 831.457.3801 Europe +00800 4573 8000 49 6221 4503 0
    Produktinformation Diagnostik & molekulare Diagnostik Laborgeräte & Service Zellkultur & Verbrauchsmaterial Forschungsprodukte & Biochemikalien Weitere Information auf den folgenden Seiten! See the following pages for more information! Lieferung & Zahlungsart Lieferung: frei Haus Bestellung auf Rechnung SZABO-SCANDIC Lieferung: € 10,- HandelsgmbH & Co KG Erstbestellung Vorauskassa Quellenstraße 110, A-1100 Wien T. +43(0)1 489 3961-0 Zuschläge F. +43(0)1 489 3961-7 [email protected] • Mindermengenzuschlag www.szabo-scandic.com • Trockeneiszuschlag • Gefahrgutzuschlag linkedin.com/company/szaboscandic • Expressversand facebook.com/szaboscandic SAN TA C RUZ BI OTEC HNOL OG Y, INC . BZW1 shRNA (h) Lentiviral Particles: sc-94750-V BACKGROUND APPLICATIONS BZW1 (basic leucine zipper and W2 domains 1), also known as BZAP45, is a BZW1 shRNA (h) Lentiviral Particles is recommended for the inhibition of 419 amino acid member of the BZW family that contains one W2 domain and BZW1 expression in human cells. enhances histone H4 gene transcription. Existing as two alternatively spliced isoforms, BZW1 is expressed in day 3 embryos and is encoded by a gene that SUPPORT REAGENTS maps to human chromosome 2q33.1. Chromosome 2 consists of 237 mil lion Control shRNA Lentiviral Particles: sc-108080. Available as 200 µl frozen bases, encodes over 1,400 genes and makes up approximately 8% of the viral stock containing 1.0 x 10 6 infectious units of virus (IFU); contains an human genome. A number of genetic diseases are linked to genes on chro - shRNA construct encoding a scrambled sequence that will not lead to the mosome 2. Harlequin icthyosis, a rare and morbid skin deformity, is associat - specific degradation of any known cellular mRNA.
    [Show full text]
  • Vitellogenic Oocytes
    CELL STRUCTURE AND FUNCTION 26: 693–703 (2001) © 2001 by Japan Society for Cell Biology Somatic Lamins in Germinal Vesicles of Goldfish (Carassius auratus) Vitellogenic Oocytes Akihiko Yamaguchi1 and Yoshitaka Nagahama Laboratory of Reproductive Biology, Department of Developmental Biology, National Institute for Basic Biology, Okazaki 444-8585, Japan 1Present address: Laboratory of Marine Biology, Department of Animal and Marine Bioresouruce Science, Faculty of Agriculture, Kyushu Univsity, Fukuoka, Japan ABSTRACT. In fish and amphibians, B-type lamins are divided into somatic (B1, B2) and oocyte-type (B3) lamins. In this study, we purified nuclear lamins from rainbow trout erythrocytes, raised an anti-lamin mono- clonal antibody (L-200) that recognizes goldfish somatic-lamins, and isolated cDNAs encoding goldfish B-type lamins (B1 and B2) from a goldfish cell culture cDNA library. Goldfish B-type lamins are structurally similar to lamins found in other vertebrates with minor amino acid substitutions in the conserved region. Western blot anal- ysis showed that goldfish oocytes contained mainly GV-lamin B3 as well as some somatic lamins. Laser-confocal microscope observations revealed that lamin B3 was present only in GV nuclear lamina, whereas somatic lamins were present in dense fibrillar structures throughout nuclear gels of isolated GVs. Similar nuclear filamentous structures were also observed in GVs of paraffin embedded oocytes. Epitope mapping indicated that L-200 recognized a conserved region containing a short stretch of the -helix coiled-coil rod domain (Y(E/Q)(Q/E)LL). A similar motif is also present in other cytoplasmic intermediate filaments (i.e., vimentin, desmin, peripherin and GFAP). Taken together, these findings suggest that lamins or lamin-related intermediate filaments are an impor- tant component of the interior architecture of goldfish vitellogenic oocyte nuclei (GVs).
    [Show full text]
  • The Capacity of Long-Term in Vitro Proliferation of Acute Myeloid
    The Capacity of Long-Term in Vitro Proliferation of Acute Myeloid Leukemia Cells Supported Only by Exogenous Cytokines Is Associated with a Patient Subset with Adverse Outcome Annette K. Brenner, Elise Aasebø, Maria Hernandez-Valladares, Frode Selheim, Frode Berven, Ida-Sofie Grønningsæter, Sushma Bartaula-Brevik and Øystein Bruserud Supplementary Material S2 of S31 Table S1. Detailed information about the 68 AML patients included in the study. # of blasts Viability Proliferation Cytokine Viable cells Change in ID Gender Age Etiology FAB Cytogenetics Mutations CD34 Colonies (109/L) (%) 48 h (cpm) secretion (106) 5 weeks phenotype 1 M 42 de novo 241 M2 normal Flt3 pos 31.0 3848 low 0.24 7 yes 2 M 82 MF 12.4 M2 t(9;22) wt pos 81.6 74,686 low 1.43 969 yes 3 F 49 CML/relapse 149 M2 complex n.d. pos 26.2 3472 low 0.08 n.d. no 4 M 33 de novo 62.0 M2 normal wt pos 67.5 6206 low 0.08 6.5 no 5 M 71 relapse 91.0 M4 normal NPM1 pos 63.5 21,331 low 0.17 n.d. yes 6 M 83 de novo 109 M1 n.d. wt pos 19.1 8764 low 1.65 693 no 7 F 77 MDS 26.4 M1 normal wt pos 89.4 53,799 high 3.43 2746 no 8 M 46 de novo 26.9 M1 normal NPM1 n.d. n.d. 3472 low 1.56 n.d. no 9 M 68 MF 50.8 M4 normal D835 pos 69.4 1640 low 0.08 n.d.
    [Show full text]
  • Methods in and Applications of the Sequencing of Short Non-Coding Rnas" (2013)
    University of Pennsylvania ScholarlyCommons Publicly Accessible Penn Dissertations 2013 Methods in and Applications of the Sequencing of Short Non- Coding RNAs Paul Ryvkin University of Pennsylvania, [email protected] Follow this and additional works at: https://repository.upenn.edu/edissertations Part of the Bioinformatics Commons, Genetics Commons, and the Molecular Biology Commons Recommended Citation Ryvkin, Paul, "Methods in and Applications of the Sequencing of Short Non-Coding RNAs" (2013). Publicly Accessible Penn Dissertations. 922. https://repository.upenn.edu/edissertations/922 This paper is posted at ScholarlyCommons. https://repository.upenn.edu/edissertations/922 For more information, please contact [email protected]. Methods in and Applications of the Sequencing of Short Non-Coding RNAs Abstract Short non-coding RNAs are important for all domains of life. With the advent of modern molecular biology their applicability to medicine has become apparent in settings ranging from diagonistic biomarkers to therapeutics and fields angingr from oncology to neurology. In addition, a critical, recent technological development is high-throughput sequencing of nucleic acids. The convergence of modern biotechnology with developments in RNA biology presents opportunities in both basic research and medical settings. Here I present two novel methods for leveraging high-throughput sequencing in the study of short non- coding RNAs, as well as a study in which they are applied to Alzheimer's Disease (AD). The computational methods presented here include High-throughput Annotation of Modified Ribonucleotides (HAMR), which enables researchers to detect post-transcriptional covalent modifications ot RNAs in a high-throughput manner. In addition, I describe Classification of RNAs by Analysis of Length (CoRAL), a computational method that allows researchers to characterize the pathways responsible for short non-coding RNA biogenesis.
    [Show full text]
  • Supplemental Information
    Supplemental information Dissection of the genomic structure of the miR-183/96/182 gene. Previously, we showed that the miR-183/96/182 cluster is an intergenic miRNA cluster, located in a ~60-kb interval between the genes encoding nuclear respiratory factor-1 (Nrf1) and ubiquitin-conjugating enzyme E2H (Ube2h) on mouse chr6qA3.3 (1). To start to uncover the genomic structure of the miR- 183/96/182 gene, we first studied genomic features around miR-183/96/182 in the UCSC genome browser (http://genome.UCSC.edu/), and identified two CpG islands 3.4-6.5 kb 5’ of pre-miR-183, the most 5’ miRNA of the cluster (Fig. 1A; Fig. S1 and Seq. S1). A cDNA clone, AK044220, located at 3.2-4.6 kb 5’ to pre-miR-183, encompasses the second CpG island (Fig. 1A; Fig. S1). We hypothesized that this cDNA clone was derived from 5’ exon(s) of the primary transcript of the miR-183/96/182 gene, as CpG islands are often associated with promoters (2). Supporting this hypothesis, multiple expressed sequences detected by gene-trap clones, including clone D016D06 (3, 4), were co-localized with the cDNA clone AK044220 (Fig. 1A; Fig. S1). Clone D016D06, deposited by the German GeneTrap Consortium (GGTC) (http://tikus.gsf.de) (3, 4), was derived from insertion of a retroviral construct, rFlpROSAβgeo in 129S2 ES cells (Fig. 1A and C). The rFlpROSAβgeo construct carries a promoterless reporter gene, the β−geo cassette - an in-frame fusion of the β-galactosidase and neomycin resistance (Neor) gene (5), with a splicing acceptor (SA) immediately upstream, and a polyA signal downstream of the β−geo cassette (Fig.
    [Show full text]
  • Depletion of the Third Complement Component Ameliorates Age- Dependent Oxidative Stress and Positively Modulates Autophagic Activity in Aged Retinas in a Mouse Model
    Hindawi Oxidative Medicine and Cellular Longevity Volume 2017, Article ID 5306790, 17 pages https://doi.org/10.1155/2017/5306790 Research Article Depletion of the Third Complement Component Ameliorates Age- Dependent Oxidative Stress and Positively Modulates Autophagic Activity in Aged Retinas in a Mouse Model 1 1 1 1 Dorota Rogińska, Miłosz P. Kawa, Ewa Pius-Sadowska, Renata Lejkowska, 1 2 3,4 3,4 Karolina Łuczkowska, Barbara Wiszniewska, Kai Kaarniranta, Jussi J. Paterno, 5 1 2,6 Christian A. Schmidt, Bogusław Machaliński, and Anna Machalińska 1Department of General Pathology, Pomeranian Medical University, Al. Powstancow Wlkp. 72, 70-111 Szczecin, Poland 2Department of Histology and Embryology, Pomeranian Medical University, Al. Powstancow Wlkp. 72, 70-111 Szczecin, Poland 3Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, 70211 Kuopio, Finland 4Department of Ophthalmology, Kuopio University Hospital, 70211 Kuopio, Finland 5Clinic for Internal Medicine C, University of Greifswald, 17475 Greifswald, Germany 6Department of Ophthalmology, Pomeranian Medical University, Al. Powstancow Wlkp. 72, 70-111 Szczecin, Poland Correspondence should be addressed to Anna Machalińska; [email protected] Received 25 April 2017; Revised 28 June 2017; Accepted 9 July 2017; Published 8 August 2017 Academic Editor: Kota V. Ramana Copyright © 2017 Dorota Rogińska et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. The aim of the study was to investigate the influence of complement component C3 global depletion on the biological structure and function of the aged retina. In vivo morphology (OCT), electrophysiological function (ERG), and the expression of selected oxidative stress-, apoptosis-, and autophagy-related proteins were assessed in retinas of 12-month-old C3-deficient and WT mice.
    [Show full text]
  • Downloaded from Genomic Data Common Website (GDC at Accessed on 2019)
    G C A T T A C G G C A T genes Article Molecular Pathways Associated with Kallikrein 6 Overexpression in Colorectal Cancer Ritu Pandey 1,2,*, Muhan Zhou 3, Yuliang Chen 3, Dalila Darmoul 4 , Conner C. Kisiel 2, Valentine N. Nfonsam 5 and Natalia A. Ignatenko 1,2 1 Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA; [email protected] 2 University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA; [email protected] 3 Bioinformatics Shared Resource, University of Arizona Cancer Center, Tucson, AZ 85724, USA; [email protected] (M.Z.); [email protected] (Y.C.) 4 Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Paris, Lariboisière Hospital, 75010 Paris, France; [email protected] 5 Department of Surgery, Section of Surgical Oncology, University of Arizona, Tucson, AZ 85724, USA; [email protected] * Correspondence: [email protected] Abstract: Colorectal cancer (CRC) remains one of the leading causes of cancer-related death world- wide. The high mortality of CRC is related to its ability to metastasize to distant organs. The kallikrein-related peptidase Kallikrein 6 (KLK6) is overexpressed in CRC and contributes to cancer cell invasion and metastasis. The goal of this study was to identify KLK6-associated markers for the CRC prognosis and treatment. Tumor Samples from the CRC patients with significantly elevated Citation: Pandey, R.; Zhou, M.; Chen, KLK6 transcript levels were identified in the RNA-Seq data from Cancer Genome Atlas (TCGA) Y.; Darmoul, D.; Kisiel, C.C.; and their expression profiles were evaluated using Gene Ontology (GO), Phenotype and Reactome Nfonsam, V.N.; Ignatenko, N.A.
    [Show full text]