Supplementary Table 1: Gene List of 44 Upregulated Enzymes in Transformed Mesenchymal Stem Cell Cancer Model

Total Page:16

File Type:pdf, Size:1020Kb

Supplementary Table 1: Gene List of 44 Upregulated Enzymes in Transformed Mesenchymal Stem Cell Cancer Model Supplementary Table 1: Gene list of 44 upregulated enzymes in transformed mesenchymal stem cell cancer model. Gene expression values for parental MSC (MSC 0) and transformed MSC (MSC5) are an average of three replicate log-2 transformed expression values from affymetrix U133 plus 2 genechip experiments with the log-fold change (LFC) indicating the difference (MSC5-MSC0). Supplementary Table 1 HGNC Symbol Alias Enzyme ID U133 plus2 probe set MSC0 MSC5 LFC Ttest pval # Gene rifs # Pubmed cites from Genecards (Sep 2007) Pathway / Function PharmGKB Drugs? Drug pathways? Therapeutic Target Database Thomson Pharma RNASEH2A AGS4; JUNB; RNHL; RNHIA; RNASEHI 3.1.26.- 203022_at 8.56 9.87 1.31 1.56E-05 0 11 RNA degradation none none none PPAP2C LPP2; PAP-2c; PAP2-g 3.1.3.4 209529_at 6.48 8.63 2.16 5.74E-03 2 13 Glycerolipid synthesis none none none ADARB1 ADAR2, ADAR2a, ADAR2a-L1, ADAR2a-L2, ADAR2a-L3, ADAR2b, ADAR2c 3.5.-.- 234799_at 6.36 8.15 1.79 2.03E-04 10 58 RNA pre-mRNA editing none none none ADARB1 3.5.-.- 203865_s_at 6.99 8.42 1.43 6.94E-03 10 58 RNA pre-mRNA editing none none none UAP1 AgX; AGX1; SPAG2 2.7.7.23 209340_at 11.17 12.45 1.28 2.46E-07 0 37 polysaccharide synthesis none none RNMT MET; RG7MT1; hCMT1c; KIAA0398; DKFZp686H1252 2.1.1.56 202684_s_at 5.70 6.78 1.08 8.15E-03 1 24 RNA (mRNA) capping none none GPD2 GDH2, mGPDH 1.1.1.8 211613_s_at 5.71 6.73 1.02 7.02E-03 2 37 glycolysis none none GCDH ACAD5, GCD 1.3.99.7 237304_at 5.38 6.39 1.01 2.44E-02 4 63 lys, hydroxy-lys, and trp metabolism none none ESPL1 3.4.22.49 38158_at 8.03 9.08 1.05 1.38E-03 6 85 mitotic chromatid separation none none BCAT1 2.6.1.26 214452_at 7.20 8.51 1.31 7.80E-03 1 20 amino acid transamination none none BCAT1 2.6.1.26 214390_s_at 7.72 8.79 1.07 6.78E-03 1 20 amino acid transamination none none PRSS3 RP11-176F3.3, MTG, PRSS4, TRY3, TRY4 3.4.21.4 207463_x_at 8.43 10.02 1.59 1.06E-04 8 47 trypsin inhibitor degradation none none PRSS3 RP11-176F3.3, MTG, PRSS4, TRY3, TRY4 3.4.21.4 213421_x_at 8.59 10.13 1.53 1.10E-05 8 47 trypsin inhibitor degradation none none OAS1 IFI-4, OIAS, OIASI 2.7.7.- 205552_s_at 5.37 6.92 1.55 5.95E-03 13 47 viral immune response none none ANXA3 ANX3 3.1.4.36 209369_at 4.10 5.82 1.72 3.47E-02 1 53 Ca++ dependent signal transduction none none CTSH ACC-4, ACC-5, CPSB, DKFZp686B24257, MGC1519, minichain 3.4.22.16 202295_s_at 6.35 7.67 1.31 3.14E-05 6 117 lysosomal degradation none none MTAP MSAP, c86fus 2.4.2.28 211363_s_at 6.10 7.15 1.05 2.90E-03 12 108 polyamine metabolism none Research target ODC1 4.1.1.17 200790_at 11.35 13.01 1.66 2.57E-05 9 128 polyamine biosynthesis none Eflornithine PGM2 FLJ10983, MSTP006 5.4.2.2 225367_at 8.42 9.78 1.37 1.66E-04 0 23 Glucose metabolism none Research target PGM2 FLJ10983, MSTP006 5.4.2.2 225366_at 7.44 8.59 1.15 2.81E-05 0 23 Glucose metabolism none Research target PGM2 FLJ10983, MSTP006 5.4.2.2 223738_s_at 7.99 9.08 1.09 7.60E-05 0 23 Glucose metabolism none Research target CTPS 6.3.4.2 202613_at 8.86 10.30 1.44 2.88E-05 1 25 nucleotide synthesis none Research target CPE 3.4.17.10 201117_s_at 3.36 4.43 1.06 4.01E-03 4 117 neuropeptide cleavage none Research target MME CALLA, CD10, DKFZp686O16152, MGC126681, M3.4.24.11 203434_s_at 4.75 8.60 3.84 3.21E-05 9 178 proteolytic cleavage none Neprilysin MME 3.4.24.11 203435_s_at 7.09 8.58 1.48 6.88E-03 9 178 proteolytic cleavage none Neprilysin NP 3.4.2.1 201695_s_at 8.75 10.34 1.59 5.09E-04 9 118 Purine metabolism Methotrexate* Research target NT5C3 3.1.3.5 223298_s_at 8.00 9.13 1.13 4.20E-03 2 26 Pyrimidine Biosynthesis Methotrexate* Pentoxifylline PFAS PURL; FGAMS; FGARAT; KIAA0361 6.3.5.3 213302_at 7.87 8.94 1.07 1.61E-05 0 10 Purine Biosynthesis Methotrexate* none ALDH1A3 ALDH6; RALDH3; ALDH1A6 1.2.1.3 222168_at 5.53 6.57 1.03 5.79E-02 1 13 alcohol metabolism Ifosamide; Cyclophosphamide pathways Disulfiram UPP1 UDRPASE, UP, UPASE, UPP 2.4.2.3 203234_at 6.52 7.73 1.21 6.24E-03 4 31 Pyrimidine Biosynthesis 5-fluorouracil pathway none UCK2 TSA903, UK, UMPK 2.7.4.- 209825_s_at 8.74 10.25 1.51 9.87E-05 3 46 Pyrimidine Biosynthesis 5-fluorouracil ; Methotrexate none GMPS 6.3.5.2 214431_at 9.40 10.44 1.04 2.49E-05 0 46 Purine Biosynthesis Methotrexate* none PAICS ADE2, ADE2H1, AIRC, DKFZp781N1372, MGC1344.1.1.21, 6.3.2.- 201014_s_at 9.75 10.97 1.23 2.19E-05 0 47 Purine Biosynthesis Methotrexate* none GART AIRS, GARS, GARTF, MGC47764, PAIS, PGFT, PRGS 2.1.2.2, 6.3.1.- 210005_at 5.79 7.37 1.58 3.59E-03 4 58 Purine Biosynthesis Methotrexate pemetrexed GART AIRS, GARS, GARTF, MGC47764, PAIS, PGFT, PRGS 2.1.2.2, 6.3.1.- 217445_s_at 6.17 7.54 1.37 1.07E-03 4 58 Purine Biosynthesis Methotrexate pemetrexed ADCY3 AC3, KIAA0511 4.6.1.1 226118_at 7.88 9.04 1.15 4.30E-03 3 66 cAMP synthesis Mercaptopurine, Methotrexate Research target RRM2 R2, RR2M 1.17.4.1 201890_at 10.05 12.19 2.15 4.30E-05 10 69 nucleotide synthesis Gemcitabine and Fluorouracil pathways none RRM2 R2, RR2M 1.17.4.1 209773_s_at 11.04 12.77 1.73 1.10E-06 10 69 nucleotide synthesis Gemcitabine and Fluorouracil pathways none RRM1 R1, RIR1, RR1 1.7.14.1 201476_s_at 9.42 10.61 1.19 8.69E-05 11 93 nucleotide synthesis Gemcitabine none RRM1 R1, RIR1, RR1 1.7.14.1 201477_s_at 10.17 11.18 1.01 1.05E-05 11 93 nucleotide synthesis Gemcitabine none ASNS TS11 6.3.5.4 205047_s_at 8.48 9.72 1.25 2.62E-04 8 95 asparagine synthesis Asparaginase none PPAT ATASE, GPAT, PRAT 2.4.2.14 209434_s_at 7.72 9.21 1.49 3.78E-04 0 107 Purine Biosynthesis Methotrexate none PPAT ATASE, GPAT, PRAT 2.4.2.14 209433_s_at 7.59 9.00 1.41 9.83E-05 0 107 Purine Biosynthesis Methotrexate none UMPS OPRT 2.4.2.10, 4.1.1.23 202706_s_at 8.40 9.76 1.35 2.95E-03 6 109 Pyrimidine Biosynthesis 5-fluorouracil pathway none UMPS OPRT 2.4.2.10, 4.1.1.23 215165_x_at 8.50 9.79 1.30 9.10E-05 6 109 Pyrimidine Biosynthesis 5-fluorouracil pathway none SOAT1 RP11-215I23.1, ACACT, ACAT, ACAT1, RP11-215I23.2, SOAT, STAT 2.3.1.26 221561_at 8.73 9.87 1.14 4.17E-03 22 114 cholesterol metabolism Statin pathway none 1-beta-arabinofuranosylcytosine (AraC), DCK MGC117410, MGC138632 2.7.1.74 203302_at 8.40 9.42 1.01 5.97E-03 18 117 deoxyribonucleoside phosphorylation Gemcitabine none XDH XO, XOR 1.1.1.204 241994_at 5.94 6.97 1.03 1.22E-02 7 122 Purine metabolism doxorubicin, methotrexate, allopurinol, menadione Allopurinol UNG DGU, DKFZp781L1143, HIGM4, UDG, UNG1, UNG15, UNG2 3.2.2.3 202330_s_at 7.77 9.02 1.25 1.04E-03 12 130 base excision repair pathway 5-fluoroucil pathway, antineoplastic agents none GCH1 DYT5, GCH, GTP-CH-1, GTPCH1 3.5.4.16 204224_s_at 4.32 5.36 1.04 2.49E-02 12 133 Purine Biosynthesis Methotrexate Research target 5-fluorouracil, antimetabolite, antineoplastic agents, fluorouracil, Lamotrigine ; Malarone; Pyrimethamine; pemetrexed; proguanil; DHFR 1.5.1.3 202533_s_at 8.10 9.27 1.17 6.26E-05 22 145 Purine Biosynthesis methotrexate trimetrexate 45 DPYD 1.3.1.2 1554536_at 3.39 4.41 1.02 1.94E-02 31 147 pyrimidine metabolism 5-fluorouracil, capecitabine, 5-fluorouracil, methotrexate, raltitrexed Enfuvirtide 3 MMP3 MGC126102, MGC126103, MGC126104, MMP-3, SL-1, STMY, STMY1, STR 3.4.24.17 205828_at 5.65 7.79 2.15 1.08E-03 95 257 extracellular matrix degradation pravastatin Research target 237(9) MMP1 CLG, CLGN 3.4.24.7 204475_at 11.75 13.89 2.14 7.86E-04 148 311 extracellular matrix degradation doxorubicin Research target 237(8) aspirin, celecoxib, clomipramine, glucocorticoids, prostaglandins, PTGS2 COX-2, COX2, PGG/HS, PGHS-2, PHS-2, hCox-2 1.14.99.1 204748_at 7.05 8.44 1.39 6.96E-03 549 616 prostaglandin biosynthesis rofecoxib, statin, valdecoxib Celecoxib; Rofecoxib; Valdecoxib 124 Patents? Druggable Target (from Hajduk et al 2005) Successful anti-cancer Target (from Mayburd et al 2007} Mayburd Rank Previous evidence for a role in cancer? Reference No Yes 0.9845 No 0.8114 No 0.3103 No 0.3103 No 0.8614 overexpression not validated in MSCs No 0.4845 this probe is not expressed in any cancers Chowdhury SK Biochem Biophys Res Commun. 2005 ; MacDonald MJ Cancer Res.
Recommended publications
  • A Genetic Variant Protective Against Severe COVID-19 Is Inherited from Neandertals
    bioRxiv preprint doi: https://doi.org/10.1101/2020.10.05.327197; this version posted October 9, 2020. 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 4.0 International license. A genetic variant protective against severe COVID-19 is inherited from Neandertals Authors Hugo Zeberg1,2* and Svante Pääbo1,3* Affiliations 1 Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, D-04103 Leipzig, Germany. 2 Department of Neuroscience, Karolinska Institutet, SE-17177 Stockholm, Sweden. 3 Okinawa Institute of Science and Technology, Onna-son, Okinawa 904-0495, Japan. *Corresponding authors: [email protected], [email protected] Abstract It was recently shown that the major genetic risk factor associated with becoming severely ill with COVID-19 when infected by SARS-CoV-2 is inherited from Neandertals. Thanks to new genetic association studies additional risk factors are now being discovered. Using data from a recent genome- wide associations from the Genetics of Mortality in Critical Care (GenOMICC) consortium, we show that a haplotype at a region associated with requiring intensive care is inherited from Neandertals. It encodes proteins that activate enzymes that are important during infections with RNA viruses. As compared to the previously described Neandertal risk haplotype, this Neandertal haplotype is protective against severe COVID-19, is of more moderate effect, and is found at substantial frequencies in all regions of the world outside Africa. 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.10.05.327197; this version posted October 9, 2020.
    [Show full text]
  • Newly Developed Serine Protease Inhibitors Decrease Visceral Hypersensitivity in a Post-Inflammatory Rat Model for Irritable Bowel Syndrome
    This item is the archived peer-reviewed author-version of: Newly developed serine protease inhibitors decrease visceral hypersensitivity in a post-inflammatory rat model for irritable bowel syndrome Reference: Ceuleers Hannah, Hanning Nikita, Heirbaut Leen, Van Remoortel Samuel, Joossens Jurgen, van der Veken Pieter, Francque Sven, De Bruyn Michelle, Lambeir Anne-Marie, de Man Joris, ....- New ly developed serine protease inhibitors decrease visceral hypersensitivity in a post-inflammatory rat model for irritable bow el syndrome British journal of pharmacology - ISSN 0007-1188 - 175:17(2018), p. 3516-3533 Full text (Publisher's DOI): https://doi.org/10.1111/BPH.14396 To cite this reference: https://hdl.handle.net/10067/1530780151162165141 Institutional repository IRUA NEWLY DEVELOPED SERINE PROTEASE INHIBITORS DECREASE VISCERAL HYPERSENSITIVITY IN A POST-INFLAMMATORY RAT MODEL FOR IRRITABLE BOWEL SYNDROME. Running title: Serine proteases in visceral hypersensitivity Hannah Ceuleers, Nikita Hanning, Jelena Heirbaut, Samuel Van Remoortel, Michelle De bruyn, Jurgen Joossens, Pieter van der Veken, Anne-Marie Lambeir, Sven M Francque, Joris G De Man, Jean-Pierre Timmermans, Koen Augustyns, Ingrid De Meester, Benedicte Y De Winter Hannah Ceuleers, Nikita Hanning, Jelena Heirbaut, Sven Francque, Joris G De Man, Benedicte Y De Winter, Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium. Samuel Van Remoortel, Jean-Pierre Timmermans, Laboratory of Cell Biology and Histology, University of Antwerp, Antwerp, Belgium. Jurgen Joossens, Pieter van der Veken, Koen Augustyns, Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium. Sven Francque, Antwerp University Hospital, Antwerp, Belgium. Michelle De bruyn, Anne-Marie Lambeir, Ingrid De Meester, Laboratory of Medical Biochemistry, University of Antwerp, Antwerp, Belgium.
    [Show full text]
  • DNA Methylation Analysis Identifies Patterns in Progressive Glioma Grades to Predict Patient Survival
    International Journal of Molecular Sciences Article DNA Methylation Analysis Identifies Patterns in Progressive Glioma Grades to Predict Patient Survival Jingyin Weng and Nicole Salazar * Department of Biology, San Francisco State University, San Francisco, CA 94132, USA; [email protected] * Correspondence: [email protected] Abstract: DNA methylation is an epigenetic change to the genome that impacts gene activities with- out modification to the DNA sequence. Alteration in the methylation pattern is a naturally occurring event throughout the human life cycle which may result in the development of diseases such as cancer. In this study, we analyzed methylation data from The Cancer Genome Atlas, under the Lower-Grade Glioma (LGG) and Glioblastoma Multiforme (GBM) projects, to identify methylation markers that exhibit unique changes in DNA methylation pattern along with tumor grade progression, to predict patient survival. We found ten glioma grade-associated Cytosine-phosphate-Guanine (CpG) sites that targeted four genes (SMOC1, KCNA4, SLC25A21, and UPP1) and the methylation pattern is strongly associated with glioma specific molecular alterations, primarily isocitrate dehydrogenase (IDH) mutation and chromosome 1p/19q codeletion. The ten CpG sites collectively distinguished a cohort of diffuse glioma patients with remarkably poor survival probability. Our study highlights genes (KCNA4 and SLC25A21) that were not previously associated with gliomas to have contributed to the poorer patient outcome. These CpG sites can aid glioma tumor progression monitoring and serve as prognostic markers to identify patients diagnosed with less aggressive and malignant gliomas that exhibit similar survival probability to GBM patients. Keywords: glioma; glioblastoma; DNA methylation; progression; TCGA; WGCNA; prognosis Citation: Weng, J.; Salazar, N.
    [Show full text]
  • CD29 Identifies IFN-Γ–Producing Human CD8+ T Cells With
    + CD29 identifies IFN-γ–producing human CD8 T cells with an increased cytotoxic potential Benoît P. Nicoleta,b, Aurélie Guislaina,b, Floris P. J. van Alphenc, Raquel Gomez-Eerlandd, Ton N. M. Schumacherd, Maartje van den Biggelaarc,e, and Monika C. Wolkersa,b,1 aDepartment of Hematopoiesis, Sanquin Research, 1066 CX Amsterdam, The Netherlands; bLandsteiner Laboratory, Oncode Institute, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; cDepartment of Research Facilities, Sanquin Research, 1066 CX Amsterdam, The Netherlands; dDivision of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; and eDepartment of Molecular and Cellular Haemostasis, Sanquin Research, 1066 CX Amsterdam, The Netherlands Edited by Anjana Rao, La Jolla Institute for Allergy and Immunology, La Jolla, CA, and approved February 12, 2020 (received for review August 12, 2019) Cytotoxic CD8+ T cells can effectively kill target cells by producing therefore developed a protocol that allowed for efficient iso- cytokines, chemokines, and granzymes. Expression of these effector lation of RNA and protein from fluorescence-activated cell molecules is however highly divergent, and tools that identify and sorting (FACS)-sorted fixed T cells after intracellular cytokine + preselect CD8 T cells with a cytotoxic expression profile are lacking. staining. With this top-down approach, we performed an un- + Human CD8 T cells can be divided into IFN-γ– and IL-2–producing biased RNA-sequencing (RNA-seq) and mass spectrometry cells. Unbiased transcriptomics and proteomics analysis on cytokine- γ– – + + (MS) analyses on IFN- and IL-2 producing primary human producing fixed CD8 T cells revealed that IL-2 cells produce helper + + + CD8 Tcells.
    [Show full text]
  • Protein Interactions in the Cancer Proteome† Cite This: Mol
    Molecular BioSystems View Article Online PAPER View Journal | View Issue Small-molecule binding sites to explore protein– protein interactions in the cancer proteome† Cite this: Mol. BioSyst., 2016, 12,3067 David Xu,ab Shadia I. Jalal,c George W. Sledge Jr.d and Samy O. Meroueh*aef The Cancer Genome Atlas (TCGA) offers an unprecedented opportunity to identify small-molecule binding sites on proteins with overexpressed mRNA levels that correlate with poor survival. Here, we analyze RNA-seq and clinical data for 10 tumor types to identify genes that are both overexpressed and correlate with patient survival. Protein products of these genes were scanned for binding sites that possess shape and physicochemical properties that can accommodate small-molecule probes or therapeutic agents (druggable). These binding sites were classified as enzyme active sites (ENZ), protein–protein interaction sites (PPI), or other sites whose function is unknown (OTH). Interestingly, the overwhelming majority of binding sites were classified as OTH. We find that ENZ, PPI, and OTH binding sites often occurred on the same structure suggesting that many of these OTH cavities can be used for allosteric modulation of Creative Commons Attribution 3.0 Unported Licence. enzyme activity or protein–protein interactions with small molecules. We discovered several ENZ (PYCR1, QPRT,andHSPA6)andPPI(CASC5, ZBTB32,andCSAD) binding sites on proteins that have been seldom explored in cancer. We also found proteins that have been extensively studied in cancer that have not been previously explored with small molecules that harbor ENZ (PKMYT1, STEAP3,andNNMT) and PPI (HNF4A, MEF2B,andCBX2) binding sites. All binding sites were classified by the signaling pathways to Received 29th March 2016, which the protein that harbors them belongs using KEGG.
    [Show full text]
  • 1A Multiple Sclerosis Treatment
    The Pharmacogenomics Journal (2012) 12, 134–146 & 2012 Macmillan Publishers Limited. All rights reserved 1470-269X/12 www.nature.com/tpj ORIGINAL ARTICLE Network analysis of transcriptional regulation in response to intramuscular interferon-b-1a multiple sclerosis treatment M Hecker1,2, RH Goertsches2,3, Interferon-b (IFN-b) is one of the major drugs for multiple sclerosis (MS) 3 2 treatment. The purpose of this study was to characterize the transcriptional C Fatum , D Koczan , effects induced by intramuscular IFN-b-1a therapy in patients with relapsing– 2 1 H-J Thiesen , R Guthke remitting form of MS. By using Affymetrix DNA microarrays, we obtained and UK Zettl3 genome-wide expression profiles of peripheral blood mononuclear cells of 24 MS patients within the first 4 weeks of IFN-b administration. We identified 1Leibniz Institute for Natural Product Research 121 genes that were significantly up- or downregulated compared with and Infection Biology—Hans-Knoell-Institute, baseline, with stronger changed expression at 1 week after start of therapy. Jena, Germany; 2University of Rostock, Institute of Immunology, Rostock, Germany and Eleven transcription factor-binding sites (TFBS) are overrepresented in the 3University of Rostock, Department of Neurology, regulatory regions of these genes, including those of IFN regulatory factors Rostock, Germany and NF-kB. We then applied TFBS-integrating least angle regression, a novel integrative algorithm for deriving gene regulatory networks from gene Correspondence: M Hecker, Leibniz Institute for Natural Product expression data and TFBS information, to reconstruct the underlying network Research and Infection Biology—Hans-Knoell- of molecular interactions. An NF-kB-centered sub-network of genes was Institute, Beutenbergstr.
    [Show full text]
  • A Novel Resveratrol Analog: Its Cell Cycle Inhibitory, Pro-Apoptotic and Anti-Inflammatory Activities on Human Tumor Cells
    A NOVEL RESVERATROL ANALOG : ITS CELL CYCLE INHIBITORY, PRO-APOPTOTIC AND ANTI-INFLAMMATORY ACTIVITIES ON HUMAN TUMOR CELLS A dissertation submitted to Kent State University in partial fulfillment of the requirements for the degree of Doctor of Philosophy by Boren Lin May 2006 Dissertation written by Boren Lin B.S., Tunghai University, 1996 M.S., Kent State University, 2003 Ph. D., Kent State University, 2006 Approved by Dr. Chun-che Tsai , Chair, Doctoral Dissertation Committee Dr. Bryan R. G. Williams , Co-chair, Doctoral Dissertation Committee Dr. Johnnie W. Baker , Members, Doctoral Dissertation Committee Dr. James L. Blank , Dr. Bansidhar Datta , Dr. Gail C. Fraizer , Accepted by Dr. Robert V. Dorman , Director, School of Biomedical Sciences Dr. John R. Stalvey , Dean, College of Arts and Sciences ii TABLE OF CONTENTS LIST OF FIGURES……………………………………………………………….………v LIST OF TABLES……………………………………………………………………….vii ACKNOWLEDGEMENTS….………………………………………………………….viii I INTRODUCTION….………………………………………………….1 Background and Significance……………………………………………………..1 Specific Aims………………………………………………………………………12 II MATERIALS AND METHODS.…………………………………………….16 Cell Culture and Compounds…….……………….…………………………….….16 MTT Cell Viability Assay………………………………………………………….16 Trypan Blue Exclusive Assay……………………………………………………...18 Flow Cytometry for Cell Cycle Analysis……………..……………....……………19 DNA Fragmentation Assay……………………………………………...…………23 Caspase-3 Activity Assay………………………………...……….….…….………24 Annexin V-FITC Staining Assay…………………………………..…...….………28 NF-kappa B p65 Activity Assay……………………………………..………….…29
    [Show full text]
  • PRSS3 Monoclonal Antibody (419911) Catalog Number MA5-24156 Product Data Sheet
    Lot Number: TC2545731D Website: thermofisher.com Customer Service (US): 1 800 955 6288 ext. 1 Technical Support (US): 1 800 955 6288 ext. 441 thermofisher.com/contactus PRSS3 Monoclonal Antibody (419911) Catalog Number MA5-24156 Product Data Sheet Details Species Reactivity Size 100 µg Tested species reactivity Mouse Host / Isotype Rat IgG1 Tested Applications Dilution * Class Monoclonal Immunohistochemistry (Frozen) 8-25 µg/ml Type Antibody (IHC (F)) Clone 419911 * Suggested working dilutions are given as a guide only. It is recommended that the user titrate the product for use in their own experiment using appropriate negative and positive controls. Mouse myeloma cell line Immunogen NS0-derived recombinant mouse Trypsin 3/PRSS3 Phe16-Asn246 Conjugate Unconjugated Form Lyophilized Concentration 0.5mg/ml Purification Protein A/G Storage Buffer PBS with 5% trehalose Contains No Preservative Storage Conditions -20° C, Avoid Freeze/Thaw Cycles Product Specific Information Reconstitute at 0.5 mg/mL in sterile PBS. Background/Target Information PRSS3 encodes a trypsinogen, which is a member of the trypsin family of serine proteases. This enzyme is expressed in the brain and pancreas and is resistant to common trypsin inhibitors. It is active on peptide linkages involving the carboxyl group of lysine or arginine. PRSS3 is localized to the locus of T cell receptor beta variable orphans on chromosome 9. Four transcript variants encoding different isoforms have been described for this gene. For Research Use Only. Not for use in diagnostic procedures. Not for resale without express authorization. For Research Use Only. Not for use in diagnostic procedures. Not for resale without express authorization.
    [Show full text]
  • Structural Basis for Cytosolic Double-Stranded RNA Surveillance by Human Oligoadenylate Synthetase 1
    Structural basis for cytosolic double-stranded RNA surveillance by human oligoadenylate synthetase 1 Jesse Donovan, Matthew Dufner, and Alexei Korennykh1 Department of Molecular Biology, Princeton University, Princeton, NJ 08540 Edited by Jennifer A. Doudna, University of California, Berkeley, CA, and approved December 19, 2012 (received for review October 23, 2012) The human sensor of double-stranded RNA (dsRNA) oligoadenylate Results and Discussion synthetase 1 (hOAS1) polymerizes ATP into 2′,5′-linked iso-RNA (2- Overview of the hOAS1•dsRNA•dATP Ternary Complex. To un- 5A) involved in innate immunity, cell cycle, and differentiation. We derstand how OAS1 recognizes dsRNA, we conducted cocrys- report the crystal structure of hOAS1 in complex with dsRNA and tallization screening with hOAS1 and dsRNA sequences derived 2′-deoxy ATP at 2.7 Å resolution, which reveals the mechanism of from RNA constructs known to activate the sensor (2). Single cytoplasmic dsRNA recognition and activation of oligoadenylate cocrystals were obtained only with dsRNA having 18 bp and a ser- synthetases. Human OAS1 recognizes dsRNA using a previously endipitously constructed sequence GGCUUUUGACCUUUAU- uncharacterized protein/RNA interface that forms via a conforma- GC. The structure of the ternary complex was determined at 2.7 Å tional change induced by binding of dsRNA. The protein/RNA in- resolution (Table S1). In the cocrystal structure, hOAS1 is bound to fi terface involves two minor grooves and has no sequence-speci c one face of the RNA double-helix (Fig. 1A and Figs. S1 and S2)and contacts, with the exception of a single hydrogen bond between the dsRNA termini are unobstructed by the protein (Table S2).
    [Show full text]
  • Irf2)Intrypsinogen5 Gene Transcription
    Characterization of dsRNA-induced pancreatitis model reveals the regulatory role of IFN regulatory factor 2 (Irf2)intrypsinogen5 gene transcription Hideki Hayashia, Tomoko Kohnoa, Kiyoshi Yasuia, Hiroyuki Murotab, Tohru Kimurac, Gordon S. Duncand, Tomoki Nakashimae, Kazuo Yamamotod, Ichiro Katayamab, Yuhua Maa, Koon Jiew Chuaa, Takashi Suematsua, Isao Shimokawaf, Shizuo Akirag, Yoshinao Kuboa, Tak Wah Makd,1, and Toshifumi Matsuyamaa,h,1 aDivision of Cytokine Signaling, Department of Molecular Biology and Immunology and fDepartment of Investigative Pathology, Nagasaki University Graduate School of Biomedical Science, Nagasaki 852-8523, Japan; Departments of bDermatology and cPathology, Graduate School of Medicine and gDepartment of Host Defense, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan; dCampbell Family Cancer Research Institute, Princess Margaret Hospital, Toronto, ON, Canada M5G 2M9; eDepartment of Cell Signaling, Tokyo Medical and Dental University, Tokyo 113-8549, Japan; and hGlobal Center of Excellence Program, Nagasaki University, Nagasaki 852-8523, Japan Contributed by Tak Wah Mak, October 5, 2011 (sent for review September 8, 2011) − − Mice deficient for interferon regulatory factor (Irf)2 (Irf2 / mice) transcriptional activation of dsRNA-sensing PRRs were critical for exhibit immunological abnormalities and cannot survive lympho- the pIC-induced death. cytic choriomeningitis virus infection. The pancreas of these ani- mals is highly inflamed, a phenotype replicated by treatment with Results and Discussion − − poly(I:C), a synthetic double-stranded RNA. Trypsinogen5 mRNA Irf2 / Mice Show IFN-Dependent Poly(I:C)-Induced Pancreatitis and −/− IFN-Independent Secretory Dysfunction in Pancreatic Acinar Cells. was constitutively up-regulated about 1,000-fold in Irf2 mice − − LCMV-infected Irf2 / mice die within 4 wk postinfection (3), compared with controls as assessed by quantitative RT-PCR.
    [Show full text]
  • The Evolution of the Viral RNA Sensor OAS1 in Old World Monkeys and Cetartiodactyls
    City University of New York (CUNY) CUNY Academic Works All Dissertations, Theses, and Capstone Projects Dissertations, Theses, and Capstone Projects 2-2016 The Evolution of the Viral RNA Sensor OAS1 in Old World Monkeys and Cetartiodactyls Ian Fish Graduate Center, City University of New York How does access to this work benefit ou?y Let us know! More information about this work at: https://academicworks.cuny.edu/gc_etds/759 Discover additional works at: https://academicworks.cuny.edu This work is made publicly available by the City University of New York (CUNY). Contact: [email protected] The Evolution of the Viral RNA Sensor OAS1 in Old World Monkeys and Cetartiodactyls by Ian Fish The City University of New York 2016 i Copyright 2016 by Fish, Ian All rights reserved ii This manuscript has been read and accepted for the Graduate Faculty in Biology in satisfaction of the dissertation requirement for the degree of Doctor of Philosophy. ______________ ______________________________ Date Chair of Examining Committee Dr. Stéphane Boissinot ______________ ______________________________ Date Executive Officer Dr. Laurel Eckhardt Supervising Committee Members: ____________________________ Dr. Cathy Savage-Dunn, Queens College ____________________________ Dr. Susan Rotenberg, Queens College ____________________________ Dr. Shaneen Singh, Brooklyn College ____________________________ Dr. Margaret MacDonald, The Rockefeller University iii Abstract The Evolution of the Viral RNA Sensor OAS1 in Old World Monkeys and Cetartiodactyls author: Ian Fish advisor: Dr. Stéphane Boissinot Animals produce an array of sensors patrolling the intracellular environment poised to detect and respond to viral infection. The oligoadenylate synthetase family of enzymes comprises a crucial part of this innate immune response, directly signaling endonuclease activity responsible for inhibiting viral replication.
    [Show full text]
  • PRSS3 Is a Prognostic Marker in Invasive Ductal Carcinoma of the Breast
    www.impactjournals.com/oncotarget/ Oncotarget, 2017, Vol. 8, (No. 13), pp: 21444-21453 Research Paper PRSS3 is a prognostic marker in invasive ductal carcinoma of the breast Li Qian1,*, Xiangxiang Gao2,*, Hua Huang1, Shumin Lu3, Yin Cai3, Yu Hua3, Yifei Liu1, Jianguo Zhang1 1Department of Clinical Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China 2Department of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, Nantong, Jiangsu, China 3Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China *These authors contributed equally to this work Correspondence to: Jianguo Zhang, email: [email protected] Yifei Liu, email: [email protected] Keywords: invasive ductal carcinoma, immunohistochemistry, PRSS3, prognosis Received: November 08, 2016 Accepted: January 27, 2017 Published: February 21, 2017 ABSTRACT Objective: Serine protease 3 (PRSS3) is an isoform of trypsinogen, and plays an important role in the development of many malignancies. The objective of this study was to determine PRSS3 mRNA and protein expression levels in invasive ductal carcinoma of the breast and normal surrounding tissue samples. Results: Both PRSS3 mRNA and protein levels were significantly higher in invasive ductal carcinoma of the breast tissues than in normal or benign tissues (all P < 0.05). High PRSS3 protein levels were associated with patients’ age, histological grade, Her-2 expression level, ki-67 expression, and the 5.0-year survival rate. These high protein levels are independent prognostic markers in invasive ductal carcinoma of the breast. Materials and Methods: We used real-time quantitative polymerase chain reactions (N = 40) and tissue microarray immunohistochemistry analysis (N = 286) to determine PRSS3 mRNA and protein expression, respectively.
    [Show full text]