2011 ADA Posters 1261-2041.Indd

Total Page:16

File Type:pdf, Size:1020Kb

2011 ADA Posters 1261-2041.Indd INTEGRATED PHYSIOLOGY—INSULINCATEGORY SECRETION IN VIVO 1622-P 1624-P Hyperglycemia Mediated Accelerates Aging in Vascular Endothelial Unique Cellular Responses of Adult Blood-Derived Endothelial Pro- Cells genitor Cells and Mature Endothelial Cells to High Glucose ROKHSANA MORTUZA, SHALI CHEN, BIAO FENG, SUBRATA CHAKRABARTI, EMILY KEATS, ZIA A. KHAN, London, ON, Canada London, ON, Canada, Lindon, ON, Canada Diabetes leads to dysfunction of selected organ systems, and vascular Glucose-induced endothelial cell dysfunction is a main factor causing endothelial cell (EC) dysfunction and loss is the key initiating and perpetuating tissue damage in the eye, kidney, heart and nerve in Diabetes. We step in the development of secondary diabetic complications. A number hypothesized that hyperglycemia induced oxidative stress causes an of studies have investigated the effect of diabetes on non-vasculogenic accelerated aging process and that such process is mediated through precursor cells (also known as early endothelial progenitor cells). However, a alterations of silent information regulator proteins or sirtuins(SIRTs) through comparative study investigating the effects of high levels of glucose on the epigenetic mechanisms. proliferative/vasculogenic endothelial progenitor cells (EPCs; late EPCs) and We investigated glucose induced aging and alterations of SIRTs in two mature ECs is lacking. We herein determined the response of adult blood- types of endothelial cells, human umbilical vein endothelial cells(HUVECs) derived EPCs and mature ECs to high levels glucose. and human microvascular endothelial cells(HMECs). The cells were grown Mononuclear cells (MNCs) from normal adult blood were successfully and propagated in high glucose (25 mmol, HG) and low glucose(5 mmol, NG). differentiated into ECs and characterized through our established protocol. Cells were observed daily and images taken to analyze their rate of growth Mature ECs were obtained from neonatal foreskin. Cells were then subject to and confl uency. At each passage subculture, some cells were collected 5 mmol/L or 25 mmol/L glucose and cellular activity assays were performed. for analysis of SIRTs, excision repair cross complimenting 1 and 4(ERCC1, We further investigated the mechanisms of increased oxidative stress in ERCC4) by RT-PCR. Cells from each passage were stained for senescence these EC types. activated beta galactosidase(SA-βgal), a known biomarker of cellular aging. Blood-derived EPCs were characterized by fl ow cytometry, cell staining, Morphometric analyses were performed. and RT-PCR. The progenitor phenotype was confi rmed by our recently Both types of cells grew faster in NG compared to HG. When exposed to discovered endostatin response assay. Following characterization, the HG, both cell types showed evidence of early senescence as evidenced by cells were exposed to high levels of glucose for 6 and 12 days. Our results increased SA-βgal expression. The replicative capacities of these cells were show that short term exposure (6-day) to glucose increased endothelin-1 diminished and they developed an irregular and hypertrophic phenotype. while decreasing the endothelin receptors in both EPCs and mature ECs. Interestingly, we found that HMEC cells started showing such changes after Interestingly, long-term exposure of cells to glucose (12 days) increased passage 1 in HG(compared to passage 4 in NG), whereas such changes were endothelin receptors in mature ECs but not EPCs. These results suggest seen in HUVECs at passage 4 in HG(compared to passage 7 in NG). Almost that mature ECs are more susceptible to the adverse effect of high glucose. all HMECs in HG at passage 5(compared to only 1.5% in NG) and HUVECs Our results also show high glucose-induced E-selectin expression in EPCs in HG at passage 11(compared to 61.4% in NG) showed β-gal positivity. suggesting an angiogenic phenotype which was not evident in mature ECs. Quantitative RT-PCR analyses showed this aging process was associated Lastly, our data illustrate NADPH oxidase was increased in mature ECs but with decreased SIRT 1, 2, 5 expressions in both cell lines. ERCC1, ERCC4 remained unaltered in the EPCs. mRNA expression was found to be decreased in HMEC but increased in In conclusion, our results show differential response of adult blood EPCs HUVEC in earlier passages. Such changes were prevented by treatment with and mature ECs to high levels of glucose and evidence for the therapeutic resveratrol. potential of vasculogenic EPCs. Data from this study suggests that hyperglycemia leads to accelerated Supported by: Canadian Diabetes Association and the Lawson Health Research aging in the endothelial cells. Furthermore, such aging process is mediated Institute through SIRTs, ERCC1 and 4 and varies depending on the cell type. 1623-P INTEGRATED PHYSIOLOGY—INSULIN SECRETION Transcription Factor mafB and a Functional Link to E-cadherin IN VIVO Expression in β Cell Differentiation MARIKO TSUCHIYA, KEN TSUCHIYA, KOSAKU NITTA, ATSUSHI MAEDA, Tokyo, [See also: Presidents Posters 451-PP to 452-PP, page A125.] Japan It is essential to clarify the mechanism by which endocrine cells including β-cells differentiate or transform during development or regeneration in Guided Audio Tour: Insulin Secretion—In Vivo Physiology (Posters 1625-P the pancreas. First, we identifi ed a few cells in the ductal epithelium of to 1634-P), see page 13. the pancreas of newborn that stained positive for insulin and mafB and & 1625-P negative for E-cadherin, whereas almost all the surrounding duct cells Short-Term Administration of the Atypical Antipsychotic Olanzapine stained positive for E-cadherin, suggesting that there may be cross-talk Induces Meal-Specifi c Impairments in Glucose and Lipid Metabolism, between mafB and E-cadherin. The purpose of the present study was to Independent of Weight Gain or Psychiatric Disease elucidate the consequences of E-cadherin and mafB expression in relation KAREN L. TEFF, MICHAEL R. RICKELS, JOANNE GRUDZIAK, KARL RICKELS, Phila- to induction of ductal epithelial cell (β-cell progenitor) differentiation into delphia, PA endocrine cells by mafB. We used the technique to intravenously inject The atypical antipsychotics are associated with an increased incidence mice and transfect cultured cells with synthetic mafB-siRNA and analyzed of diabetes. The mechanisms underlying these metabolic defects are not the resulting alterations in gene and protein expression level by real-time understood, although it has been speculated that the initiating pathophysiology PCR or immunohistochemistry. To observe the precise mechanism of this is weight gain, secondary to centrally-mediated increases in appetite. Our process, we suppressed mafB gene expression to 40% of the control level objectives were to determine if olanzapine administration impairs insulin in AsPc1 cells, a cell line derived from a pancreatic carcinoma in which sensitivity and post-prandial responses to the physiologically-relevant stimulus maf expression has been confi rmed by real-time PCR. Suppression of mafB of a meal, independent of weight gain or psychiatric disease. Healthy male 2 Obesity expression by siRNA in AsPC1 cells resulted in upregulation of mRNA (1.4 subjects (BMI=69.8+2.6 Kg/m ) were admitted into the CTRC for 12-days. POSTERS fold) and immunostaining intensity (1.4 fold) of E-cadherin. Expression of Activity was maintained for each subject at a level matched to their free α-catenin (1.2 fold), which is indispensable for cadherin-catenin-complex- living levels. On Days 1 and 3 in a randomized order, subjects underwent a 600 Integrated Physiology/ mediated cell adhesion, was also increased, and β-catenin was increased kcal mixed nutrient meal challenge (n=10) and a euglycemic, hyperinsulinemic by 3.5 fold and GSK3 was slightly suppressed (0.8 fold), suggesting that clamp (7/10 subjects). The meal was labeled with [1-13C] glucose to monitor mafB may regulate ductal epithelial progenitor differentiation by expression 2 glucose appearance and 6,6-[ H2]-glucose was infused to measure glucose of the cadherin-catenin complex in the WNT signaling pathway. In the in disposal and endogenous glucose production during both the meal challenge vivo study mafB-siRNA treatment in the mouse pancreas resulted in similar and the clamp. Subjects were then administered olanzapine (5 mg/d, Days 4-5; changes. Moreover, mafB suppression downregulated expression of γ-actin 10 mg/d, Days 6-12). On Days 10 and 12, the metabolic tests were repeated. by the results of microarray profi ling. Taken together, the fi ndings in this Weight remained stable throughout the study (69.8+2.6,pre:70.1+2.9 Kg/ study suggested that mafB, whose expression is modifi ed by other mafs or m2, post-olanzapine) and no increase in food intake was found. Olanzapine other genes, even in stress or metabolic conditions in the mature pancreas increased post-prandial glucose (area under the curve: pre, 4363+1981 vs may regulate the E-cadherin-catenin complex in the WNT signaling pathway, post, 6579+1838, P<0.02), insulin (6824+4162 vs 13676+6739, P<0.001) and resulting in cell adherence and endocrine cell differentiation. triglyceride (1962+3995 vs. 4200+3938, P<0.01) concentrations. Small but For author disclosure information, see page 785. & Guided Audio Tour poster ADA-Funded Research A442 INTEGRATED PHYSIOLOGY—INSULINCATEGORY SECRETION IN VIVO signifi cant decreases in glucose disposal were found during the meal and the clamp (P<0.01) but no ef fects were obser ved on endogenous glucose production. These data demonstrate that short-term administration of olanzapine to normal subjects induces meal specifi c impairments in post-prandial glucose 1627-P and lipid metabolism, independent of weight gain or psychiatric disease. Hyperglycemic Clamp EvaluationWITHDRAWN of First and Second Phase Insulin The metabolic dysregulation likely contributes to the increased incidence of Secretion in Type 2 Diabetic Patients with BMI 24 - 34 Kg/m2 Submitted metabolic disease in patients treated with this and similar medications. to the Ileal interposition Associated with Sleeve Gastrectomy Supported by: NIH DK084383 (KLT).
Recommended publications
  • Hypoxia and Oxygen-Sensing Signaling in Gene Regulation and Cancer Progression
    International Journal of Molecular Sciences Review Hypoxia and Oxygen-Sensing Signaling in Gene Regulation and Cancer Progression Guang Yang, Rachel Shi and Qing Zhang * Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; [email protected] (G.Y.); [email protected] (R.S.) * Correspondence: [email protected]; Tel.: +1-214-645-4671 Received: 6 October 2020; Accepted: 29 October 2020; Published: 31 October 2020 Abstract: Oxygen homeostasis regulation is the most fundamental cellular process for adjusting physiological oxygen variations, and its irregularity leads to various human diseases, including cancer. Hypoxia is closely associated with cancer development, and hypoxia/oxygen-sensing signaling plays critical roles in the modulation of cancer progression. The key molecules of the hypoxia/oxygen-sensing signaling include the transcriptional regulator hypoxia-inducible factor (HIF) which widely controls oxygen responsive genes, the central members of the 2-oxoglutarate (2-OG)-dependent dioxygenases, such as prolyl hydroxylase (PHD or EglN), and an E3 ubiquitin ligase component for HIF degeneration called von Hippel–Lindau (encoding protein pVHL). In this review, we summarize the current knowledge about the canonical hypoxia signaling, HIF transcription factors, and pVHL. In addition, the role of 2-OG-dependent enzymes, such as DNA/RNA-modifying enzymes, JmjC domain-containing enzymes, and prolyl hydroxylases, in gene regulation of cancer progression, is specifically reviewed. We also discuss the therapeutic advancement of targeting hypoxia and oxygen sensing pathways in cancer. Keywords: hypoxia; PHDs; TETs; JmjCs; HIFs 1. Introduction Molecular oxygen serves as a co-factor in many biochemical processes and is fundamental for aerobic organisms to maintain intracellular ATP levels [1,2].
    [Show full text]
  • Mutant IDH, (R)-2-Hydroxyglutarate, and Cancer
    Downloaded from genesdev.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press REVIEW What a difference a hydroxyl makes: mutant IDH, (R)-2-hydroxyglutarate, and cancer Julie-Aurore Losman1 and William G. Kaelin Jr.1,2,3 1Department of Medical Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02215, USA; 2Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA Mutations in metabolic enzymes, including isocitrate whether altered cellular metabolism is a cause of cancer dehydrogenase 1 (IDH1) and IDH2, in cancer strongly or merely an adaptive response of cancer cells in the face implicate altered metabolism in tumorigenesis. IDH1 of accelerated cell proliferation is still a topic of some and IDH2 catalyze the interconversion of isocitrate and debate. 2-oxoglutarate (2OG). 2OG is a TCA cycle intermediate The recent identification of cancer-associated muta- and an essential cofactor for many enzymes, including tions in three metabolic enzymes suggests that altered JmjC domain-containing histone demethylases, TET cellular metabolism can indeed be a cause of some 5-methylcytosine hydroxylases, and EglN prolyl-4-hydrox- cancers (Pollard et al. 2003; King et al. 2006; Raimundo ylases. Cancer-associated IDH mutations alter the enzymes et al. 2011). Two of these enzymes, fumarate hydratase such that they reduce 2OG to the structurally similar (FH) and succinate dehydrogenase (SDH), are bone fide metabolite (R)-2-hydroxyglutarate [(R)-2HG]. Here we tumor suppressors, and loss-of-function mutations in FH review what is known about the molecular mechanisms and SDH have been identified in various cancers, in- of transformation by mutant IDH and discuss their im- cluding renal cell carcinomas and paragangliomas.
    [Show full text]
  • Protein Identities in Evs Isolated from U87-MG GBM Cells As Determined by NG LC-MS/MS
    Protein identities in EVs isolated from U87-MG GBM cells as determined by NG LC-MS/MS. No. Accession Description Σ Coverage Σ# Proteins Σ# Unique Peptides Σ# Peptides Σ# PSMs # AAs MW [kDa] calc. pI 1 A8MS94 Putative golgin subfamily A member 2-like protein 5 OS=Homo sapiens PE=5 SV=2 - [GG2L5_HUMAN] 100 1 1 7 88 110 12,03704523 5,681152344 2 P60660 Myosin light polypeptide 6 OS=Homo sapiens GN=MYL6 PE=1 SV=2 - [MYL6_HUMAN] 100 3 5 17 173 151 16,91913397 4,652832031 3 Q6ZYL4 General transcription factor IIH subunit 5 OS=Homo sapiens GN=GTF2H5 PE=1 SV=1 - [TF2H5_HUMAN] 98,59 1 1 4 13 71 8,048185945 4,652832031 4 P60709 Actin, cytoplasmic 1 OS=Homo sapiens GN=ACTB PE=1 SV=1 - [ACTB_HUMAN] 97,6 5 5 35 917 375 41,70973209 5,478027344 5 P13489 Ribonuclease inhibitor OS=Homo sapiens GN=RNH1 PE=1 SV=2 - [RINI_HUMAN] 96,75 1 12 37 173 461 49,94108966 4,817871094 6 P09382 Galectin-1 OS=Homo sapiens GN=LGALS1 PE=1 SV=2 - [LEG1_HUMAN] 96,3 1 7 14 283 135 14,70620005 5,503417969 7 P60174 Triosephosphate isomerase OS=Homo sapiens GN=TPI1 PE=1 SV=3 - [TPIS_HUMAN] 95,1 3 16 25 375 286 30,77169764 5,922363281 8 P04406 Glyceraldehyde-3-phosphate dehydrogenase OS=Homo sapiens GN=GAPDH PE=1 SV=3 - [G3P_HUMAN] 94,63 2 13 31 509 335 36,03039959 8,455566406 9 Q15185 Prostaglandin E synthase 3 OS=Homo sapiens GN=PTGES3 PE=1 SV=1 - [TEBP_HUMAN] 93,13 1 5 12 74 160 18,68541938 4,538574219 10 P09417 Dihydropteridine reductase OS=Homo sapiens GN=QDPR PE=1 SV=2 - [DHPR_HUMAN] 93,03 1 1 17 69 244 25,77302971 7,371582031 11 P01911 HLA class II histocompatibility antigen,
    [Show full text]
  • Egln2 Associates with the NRF1PGC1 Complex and Controls
    Article EglN2 associates with the NRF1-PGC1a complex and controls mitochondrial function in breast cancer Jing Zhang1,†, Chengyang Wang2,†, Xi Chen3, Mamoru Takada1, Cheng Fan1, Xingnan Zheng1, Haitao Wen1,4, Yong Liu1, Chenguang Wang5, Richard G Pestell6, Katherine M Aird7, William G Kaelin Jr8,9, Xiaole Shirley Liu10 & Qing Zhang1,11,* Abstract Introduction The EglN2/PHD1 prolyl hydroxylase is an important oxygen sensor The presence of hypoxic cells in the tumor microenvironment was contributing to breast tumorigenesis. Emerging studies suggest proposed by Thomlinson and Gray more than 50 years ago that there is functional cross talk between oxygen sensing and (Thomlinson & Gray, 1955). These hypoxic cells confer radio- or mitochondrial function, both of which play an essential role for chemotherapeutic resistance and therefore are hypothesized to be sustained tumor growth. However, the potential link between under selection for aggressive malignancy during the course of EglN2 and mitochondrial function remains largely undefined. cancer development (Brown & Wilson, 2004). One central question Here, we show that EglN2 depletion decreases mitochondrial is how hypoxic cancer cells sense their oxygen availability, adapt to respiration in breast cancer under normoxia and hypoxia, which the stressful environment, and proliferate out of control. The key correlates with decreased mitochondrial DNA in a HIF1/2a- proteins mediating oxygen sensing in these cells mainly involve independent manner. Integrative analyses of gene expression proteins that are responsible for the hydroxylation of hypoxia- profile and genomewide binding of EglN2 under hypoxic condi- inducible factor (HIF), namely the prolyl hydroxylases EglN1-3. As a tions reveal nuclear respiratory factor 1 (NRF1) motif enrichment key EglN enzyme substrate, HIF1a is hydroxylated on prolines 402 in EglN2-activated genes, suggesting NRF1 as an EglN2 binding and 564 under normoxic conditions.
    [Show full text]
  • Integrative Genomics Identifies New Genes Associated with Severe COPD and Emphysema Phuwanat Sakornsakolpat1,2, Jarrett D
    Sakornsakolpat et al. Respiratory Research (2018) 19:46 https://doi.org/10.1186/s12931-018-0744-9 RESEARCH Open Access Integrative genomics identifies new genes associated with severe COPD and emphysema Phuwanat Sakornsakolpat1,2, Jarrett D. Morrow1, Peter J. Castaldi1,3, Craig P. Hersh1,4, Yohan Bossé5, Edwin K. Silverman1,4, Ani Manichaikul6 and Michael H. Cho1,4* Abstract Background: Genome-wide association studies have identified several genetic risk loci for severe chronic obstructive pulmonary disease (COPD) and emphysema. However, these studies do not fully explain disease heritability and in most cases, fail to implicate specific genes. Integrative methods that combine gene expression data with GWAS can provide more power in discovering disease-associated genes and give mechanistic insight into regulated genes. Methods: We applied a recently described method that imputes gene expression using reference transcriptome data to genome-wide association studies for two phenotypes (severe COPD and quantitative emphysema) and blood and lung tissue gene expression datasets. We further tested the potential causality of individual genes using multi-variant colocalization. Results: We identified seven genes significantly associated with severe COPD, and five genes significantly associated with quantitative emphysema in whole blood or lung. We validated results in independent transcriptome databases and confirmed colocalization signals for PSMA4, EGLN2, WNT3, DCBLD1, and LILRA3. Three of these genes were not located within previously reported GWAS loci for either phenotype. We also identified genetically driven pathways, including those related to immune regulation. Conclusions: An integrative analysis of GWAS and gene expression identified novel associations with severe COPD and quantitative emphysema, and also suggested disease-associated genes in known COPD susceptibility loci.
    [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]
  • Electronic Supplementary Material (ESI) for Metallomics
    Electronic Supplementary Material (ESI) for Metallomics. This journal is © The Royal Society of Chemistry 2018 Uniprot Entry name Gene names Protein names Predicted Pattern Number of Iron role EC number Subcellular Membrane Involvement in disease Gene ontology (biological process) Id iron ions location associated 1 P46952 3HAO_HUMAN HAAO 3-hydroxyanthranilate 3,4- H47-E53-H91 1 Fe cation Catalytic 1.13.11.6 Cytoplasm No NAD biosynthetic process [GO:0009435]; neuron cellular homeostasis dioxygenase (EC 1.13.11.6) (3- [GO:0070050]; quinolinate biosynthetic process [GO:0019805]; response to hydroxyanthranilate oxygenase) cadmium ion [GO:0046686]; response to zinc ion [GO:0010043]; tryptophan (3-HAO) (3-hydroxyanthranilic catabolic process [GO:0006569] acid dioxygenase) (HAD) 2 O00767 ACOD_HUMAN SCD Acyl-CoA desaturase (EC H120-H125-H157-H161; 2 Fe cations Catalytic 1.14.19.1 Endoplasmic Yes long-chain fatty-acyl-CoA biosynthetic process [GO:0035338]; unsaturated fatty 1.14.19.1) (Delta(9)-desaturase) H160-H269-H298-H302 reticulum acid biosynthetic process [GO:0006636] (Delta-9 desaturase) (Fatty acid desaturase) (Stearoyl-CoA desaturase) (hSCD1) 3 Q6ZNF0 ACP7_HUMAN ACP7 PAPL PAPL1 Acid phosphatase type 7 (EC D141-D170-Y173-H335 1 Fe cation Catalytic 3.1.3.2 Extracellular No 3.1.3.2) (Purple acid space phosphatase long form) 4 Q96SZ5 AEDO_HUMAN ADO C10orf22 2-aminoethanethiol dioxygenase H112-H114-H193 1 Fe cation Catalytic 1.13.11.19 Unknown No oxidation-reduction process [GO:0055114]; sulfur amino acid catabolic process (EC 1.13.11.19) (Cysteamine
    [Show full text]
  • Chapter 2 2. Q Labelling in Hyoscyamine
    Durham E-Theses The biosynthesis of the tropane alkaloid hyoscyamine in datura stramonium Wong, Chi W. How to cite: Wong, Chi W. (1999) The biosynthesis of the tropane alkaloid hyoscyamine in datura stramonium, Durham theses, Durham University. Available at Durham E-Theses Online: http://etheses.dur.ac.uk/4310/ Use policy The full-text may be used and/or reproduced, and given to third parties in any format or medium, without prior permission or charge, for personal research or study, educational, or not-for-prot purposes provided that: • a full bibliographic reference is made to the original source • a link is made to the metadata record in Durham E-Theses • the full-text is not changed in any way The full-text must not be sold in any format or medium without the formal permission of the copyright holders. Please consult the full Durham E-Theses policy for further details. Academic Support Oce, Durham University, University Oce, Old Elvet, Durham DH1 3HP e-mail: [email protected] Tel: +44 0191 334 6107 http://etheses.dur.ac.uk COPYRIGHT The copyright of this thesis rests with the author. No quotation form it should be published without prior consent, and any information derived from this thesis should be acknowledged. DECLARATION The work contained in this thesis was carried out in the Department of Chemistry at the University of Durham between October 1995 and September 1998. All the work was carried out by the author, unless otherwise indicated. It has not been previously submitted for a degree at this or any other university.
    [Show full text]
  • Downloaded from the UCSC PRO-Seq Library Preparation and Sequencing
    ARTICLE https://doi.org/10.1038/s41467-021-21687-2 OPEN Multi-omics analysis reveals contextual tumor suppressive and oncogenic gene modules within the acute hypoxic response ✉ ✉ Zdenek Andrysik1,2, Heather Bender1,2, Matthew D. Galbraith 1,2 & Joaquin M. Espinosa 1,2,3 Cellular adaptation to hypoxia is a hallmark of cancer, but the relative contribution of hypoxia- inducible factors (HIFs) versus other oxygen sensors to tumorigenesis is unclear. We employ 1234567890():,; a multi-omics pipeline including measurements of nascent RNA to characterize transcrip- tional changes upon acute hypoxia. We identify an immediate early transcriptional response that is strongly dependent on HIF1A and the kinase activity of its cofactor CDK8, includes indirect repression of MYC targets, and is highly conserved across cancer types. HIF1A drives this acute response via conserved high-occupancy enhancers. Genetic screen data indicates that, in normoxia, HIF1A displays strong cell-autonomous tumor suppressive effects through a gene module mediating mTOR inhibition. Conversely, in advanced malignancies, expression of a module of HIF1A targets involved in collagen remodeling is associated with poor prog- nosis across diverse cancer types. In this work, we provide a valuable resource for investi- gating context-dependent roles of HIF1A and its targets in cancer biology. 1 Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA. 2 Linda Crnic Institute for Down Syndrome, School of Medicine, University
    [Show full text]
  • Copyright © 2019 by Yue Wu
    Studies in Using Gold Nanoparticles in Treating Cancer and Inhibiting Metastasis A Dissertation Presented to The Academic Faculty by Yue Wu In Partial Fulfillment of the Requirements for the Degree Doctor of Philosophy in the School of Chemistry and Biochemistry Georgia Institute of Technology May 2019 COPYRIGHT © 2019 BY YUE WU Studies in Using Gold Nanoparticles in Treating Cancer and Inhibiting Metastasis Approved by: Dr. Mostafa A. El-Sayed, Advisor Dr. Robert Dickson School of Chemistry and Biochemistry School of Chemistry and Biochemistry Georgia Institute of Technology Georgia Institute of Technology Dr. Ingeborg Schmidt-Krey Dr. Zhong Lin Wang School of Chemistry and Biochemistry School of Material Science and Georgia Institute of Technology Engineering Georgia Institute of Technology Dr. Ronghu Wu School of Chemistry and Biochemistry Georgia Institute of Technology Date Approved: March 26, 2019 To My Family ACKNOWLEDGEMENTS First, I would like to thank my advisor, Prof. Mostafa A. El-Sayed, for his guidance, encouragement, support, sharing with me his life-long enthusiasm and dedication towards scientific research, and the opportunity he provided me to move into the nanotechnology and nanomedicine field. His optimistic, positive, modest, and humorous personality is always inspiring me. I would also like to acknowledge the professors during my PhD time, including Prof. Ning Fang, for his guidance of optical imaging and great support for my research, Prof. Todd Sulchek for his help with cell mechanical measurement, Profs. Ronghu Wu, Fangjun Wang, and Facundo Fernandez for their help with mass spectrometry based proteomics and metabolomics, Profs. Ivan El-Sayed and Dong Shin for their guidance on animal work or clinical applications, and Prof.
    [Show full text]
  • NIH Public Access Author Manuscript Curr Cancer Drug Targets
    NIH Public Access Author Manuscript Curr Cancer Drug Targets. Author manuscript; available in PMC 2013 July 06. NIH-PA Author ManuscriptPublished NIH-PA Author Manuscript in final edited NIH-PA Author Manuscript form as: Curr Cancer Drug Targets. 2013 June 10; 13(5): 558–579. Histone Lysine-Specific Methyltransferases and Demethylases in Carcinogenesis: New Targets for Cancer Therapy and Prevention Xuejiao Tian1,2,3, Saiyang Zhang4, Hong-Min Liu4, Yan-Bing Zhang4, Christopher A Blair1,2, Dan Mercola5, Paolo Sassone-Corsi6, and Xiaolin Zi1,2,3,* 1Department of Urology, University of California, Irvine, Orange, CA 92868, USA 2Department of Pharmaceutical Sciences, University of California, Irvine, Orange, CA 92868, USA 3Department of Pharmacology, University of California, Irvine, Orange, CA 92868, USA 4School of Pharmaceutical Sciences, Zhengzhou University, No.100, Science Avenue, Zhengzhou 450001, Henan, China 5Department of Pathology and Laboratory medicine, University of California, Irvine, Orange, CA 92868, USA 6Department of Biological Chemistry, University of California, Irvine, Orange, CA 92868, USA Abstract Aberrant histone lysine methylation that is controlled by histone lysine methyltransferases (KMTs) and demethylases (KDMs) plays significant roles in carcinogenesis. Infections by tumor viruses or parasites and exposures to chemical carcinogens can modify the process of histone lysine methylation. Many KMTs and KDMs contribute to malignant transformation by regulating the expression of human telomerase reverse transcriptase (hTERT), forming a fused gene, interacting with proto-oncogenes or being up-regulated in cancer cells. In addition, histone lysine methylation participates in tumor suppressor gene inactivation during the early stages of carcinogenesis by regulating DNA methylation and/or by other DNA methylation independent mechanisms.
    [Show full text]
  • UCLA Electronic Theses and Dissertations
    UCLA UCLA Electronic Theses and Dissertations Title A Liquid Chromatography-Mass Spectrometry Platform for Identifying Protein Targets of Small-Molecule Binding Relevant to Disease and Metabolism Permalink https://escholarship.org/uc/item/8pn170t7 Author O'Brien Johnson, Reid Publication Date 2018 Peer reviewed|Thesis/dissertation eScholarship.org Powered by the California Digital Library University of California UNIVERSITY OF CALIFORNIA Los Angeles A Liquid Chromatography-Mass Spectrometry Platform for Identifying Protein Targets of Small-Molecule Binding Relevant to Disease and Metabolism A dissertation submitted in partial satisfaction of the requirements for the degree Doctor of Philosophy in Biochemistry and Molecular Biology by Reid Lee O’Brien Johnson 2018 ABSTRACT OF THE DISSERTATION A Liquid Chromatography-Mass Spectrometry Platform for Identifying Protein Targets of Small-Molecule Binding Relevant to Disease and Metabolism by Reid Lee O’Brien Johnson Doctor of Philosophy in Biochemistry and Molecular Biology University of California, Los Angeles, 2018 Professor Joseph Ambrose Loo, Chair Identifying small-molecule binders to protein targets remains a daunting task due to the huge diversity in compound structure, activity, and mechanisms of action. Affinity- based target identification techniques are limited by the necessity to modify each drug individually (without losing bioactivity), while non-affinity based approaches are dependent on the drug’s ability to induce specific biochemical/cellular readouts. To overcome these limitations, we have developed a high-throughput liquid chromatography- mass spectrometry (LC-MS) platform coupled to a universally applicable target identification approach that analyzes direct small-molecule binding to its protein target(s). DARTS (drug affinity responsive target stability) relies on a well-known phenomenon in which ligand binding causes thermodynamic stabilization of its target protein’s structure such that the protein becomes resistant to a variety of insults, including proteolysis.
    [Show full text]