New Approaches to Improve Prediction of Drug-Induced Liver Injury

Total Page:16

File Type:pdf, Size:1020Kb

New Approaches to Improve Prediction of Drug-Induced Liver Injury New approaches to improve prediction of drug-induced liver injury Dissertation zur Erlangung des naturwissenschafltichen Doktorgrades der Julius-Maximilians-Universität Würzburg vorgelegt von Melanie Adler aus Greiz Würzburg 2012 Eingereicht am 02.02.2012 bei der Fakultät für Chemie und Pharmazie Gutachter der schriftlichen Arbeit 1. Gutachter PD. Dr. Angela Mally 2. Gutachter Prof. Dr. Ulrike Holzgrabe 1. Prüfer PD Dr. Angela Mally 2. Prüfer Prof. Dr. Ulrike Holzgrabe 3. Prüfer Prof. Dr. Wolfgang Dekant des öffentlichen Promotionskolloquiums Datum des öffentlichen Promotionskolloquiums: 27.02.2012 Doktorurkunde ausgehändigt am ………………………………………………. Für meine Eltern "Wer immer tut, was er schon kann, bleibt immer das, was er schon ist." Henry Ford INDEX INDEX Index .............................................................................................................. I Abbreviations ............................................................................................... V 1 Introduction .......................................................................................... 1 1.1 The liver as a critical target organ of drug toxicity ................................ 1 1.1.1 Liver morphology and function ............................................................................ 3 1.1.2 Xenobiotic metabolism ........................................................................................ 4 1.1.3 Types and mechanisms of drug-induced liver injury ........................................... 6 1.2 Methods for detection of hepatotoxicity ................................................10 1.2.1 Primary hepatocytes as an in vitro model to study hepatotoxicity ..................... 10 1.2.2 Traditional approaches ...................................................................................... 11 1.2.3 Application of omics technologies ..................................................................... 12 1.2.3.1 Novel biomarker candidates for hepatotoxicity ........................................................ 13 1.2.3.2 Mechanistic understanding of hepatotoxicity ........................................................... 15 1.2.4 RNA interference as an investigative tool in mechanistic toxicology................. 17 1.2.4.1 Mechanism of siRNA-mediated gene silencing ....................................................... 18 1.3 Aims of this work .....................................................................................21 2 Materials and Methods ...................................................................... 22 2.1 Materials ...................................................................................................22 2.1.1 Technical equipment ......................................................................................... 22 2.1.2 Chemicals and reagents ................................................................................... 23 2.1.3 Culture media and supplements ....................................................................... 25 2.1.4 Kits .................................................................................................................... 25 2.1.5 Buffers and solutions ......................................................................................... 26 2.1.6 Antibodies ......................................................................................................... 28 2.1.7 Oligonucleotides ................................................................................................ 29 2.1.8 Software ............................................................................................................ 30 2.2 In vivo studies ..........................................................................................30 2.2.1 Standardized study design of the InnoMed Predictive Toxicology Project ........ 30 2.2.2 Clinical examination .......................................................................................... 32 2.2.3 Construction of tissue microarrays .................................................................... 33 2.3 In vitro studies .........................................................................................33 2.3.1 Isolation of primary rat hepatocytes .................................................................. 34 2.3.2 Trypan Blue exclusion test ................................................................................ 35 2.3.3 Culture conditions of hepatocytes ..................................................................... 36 I INDEX 2.3.4 Transfection of siRNA duplex ........................................................................... 36 2.3.5 Treatment with compound BAY16 .................................................................... 37 2.3.6 WST-1 assay .................................................................................................... 38 2.3.7 CellTiter-Glo® Luminescent cell viability assay ................................................. 39 2.4 Molecular biological methods ............................................................... 40 2.4.1 Isolation of total RNA from tissues .................................................................... 40 2.4.2 Isolation of total RNA from rat hepatocytes ...................................................... 41 2.4.3 RNA quantification and quality control .............................................................. 41 2.4.4 cDNA synthesis and qRT- PCR using SYBR® Green ....................................... 42 2.4.5 cDNA synthesis and qRT- PCR using hydrolysis probes ................................. 45 2.4.6 Microarray experiments .................................................................................... 46 2.4.6.1 Preparation of labeled aRNA ................................................................................... 48 2.4.6.2 Hybridization and microarray processing ................................................................. 49 2.4.6.3 Data extraction and analysis .................................................................................... 50 2.5 Biochemical methods ............................................................................. 51 2.5.1 Protein preparation from tissue ......................................................................... 51 2.5.2 Protein preparation from rat hepatocytes .......................................................... 52 2.5.3 Protein separation by SDS polyacrylamid gel electrophoresis ......................... 52 2.5.4 Protein detection by western blot analysis and immune detection ................... 53 2.5.5 Immunofluorescence ........................................................................................ 55 2.5.6 Immunohistochemical analyses ........................................................................ 55 2.5.7 Quantification of proteins by ELISA .................................................................. 57 2.5.7.1 Development and validation of the rat NGAL ELISA ............................................... 58 2.5.7.2 Thiostatin ELISA ....................................................................................................... 59 2.5.7.3 Clusterin ELISA ........................................................................................................ 60 2.5.8 PON1 activity assay .......................................................................................... 60 3 Assessment of candidate biomarkers of drug induced liver injury in preclinical toxicity studies ................................................. 61 3.1 Background ............................................................................................. 61 3.2 Results ..................................................................................................... 62 3.2.1 Biomarker responses to BAY16 in relation to traditional endpoints of toxicity .. 62 3.2.1.1 Clinicopathological and histopathological observations ........................................... 62 3.2.1.2 Gene expression of biomarkers ............................................................................... 65 3.2.1.3 Detection and quantification of biomarkers in serum and urine ............................... 67 3.2.1.4 Immunolocalization of putative biomarkers .............................................................. 70 3.2.1.5 Summary .................................................................................................................. 72 3.2.2 Biomarker responses to EMD335823 in relation to traditional endpoints of toxicity ............................................................................................................... 73 3.2.2.1 Histopathological and clinicopathological observations ........................................... 73 II INDEX 3.2.2.2 Gene expression of biomarkers ............................................................................... 76 3.2.2.3 Detection and quantification of biomarkers in serum and urine ............................... 77 3.2.2.4 Immunolocalization of putative biomarkers .............................................................. 79 3.2.2.5 Summary .................................................................................................................. 80 3.2.3 Biomarker responses to BI-3 in relation to traditional endpoints of toxicity ....... 81 3.2.3.1 Clinicopathological and histopathological observations ........................................... 81 3.2.3.2 Gene expression of biomarkers ..............................................................................
Recommended publications
  • Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model
    Downloaded from http://www.jimmunol.org/ by guest on September 25, 2021 T + is online at: average * The Journal of Immunology , 34 of which you can access for free at: 2016; 197:1477-1488; Prepublished online 1 July from submission to initial decision 4 weeks from acceptance to publication 2016; doi: 10.4049/jimmunol.1600589 http://www.jimmunol.org/content/197/4/1477 Molecular Profile of Tumor-Specific CD8 Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A. Waugh, Sonia M. Leach, Brandon L. Moore, Tullia C. Bruno, Jonathan D. Buhrman and Jill E. Slansky J Immunol cites 95 articles Submit online. Every submission reviewed by practicing scientists ? is published twice each month by Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts http://jimmunol.org/subscription Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html http://www.jimmunol.org/content/suppl/2016/07/01/jimmunol.160058 9.DCSupplemental This article http://www.jimmunol.org/content/197/4/1477.full#ref-list-1 Information about subscribing to The JI No Triage! Fast Publication! Rapid Reviews! 30 days* Why • • • Material References Permissions Email Alerts Subscription Supplementary The Journal of Immunology The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2016 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. This information is current as of September 25, 2021. The Journal of Immunology Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A.
    [Show full text]
  • Examination of the Transcription Factors Acting in Bone Marrow
    THESIS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY (PHD) Examination of the transcription factors acting in bone marrow derived macrophages by Gergely Nagy Supervisor: Dr. Endre Barta UNIVERSITY OF DEBRECEN DOCTORAL SCHOOL OF MOLECULAR CELL AND IMMUNE BIOLOGY DEBRECEN, 2016 Table of contents Table of contents ........................................................................................................................ 2 1. Introduction ............................................................................................................................ 5 1.1. Transcriptional regulation ................................................................................................... 5 1.1.1. Transcriptional initiation .................................................................................................. 5 1.1.2. Co-regulators and histone modifications .......................................................................... 8 1.2. Promoter and enhancer sequences guiding transcription factors ...................................... 11 1.2.1. General transcription factors .......................................................................................... 11 1.2.2. The ETS superfamily ..................................................................................................... 17 1.2.3. The AP-1 and CREB proteins ........................................................................................ 20 1.2.4. Other promoter specific transcription factor families ...................................................
    [Show full text]
  • Effect of a Caloric Restriction Based On
    UNIVERSIDAD AUTÓNOMA DE MADRID FACULTAD DE CIENCIAS DEPARTAMENTO DE BIOLOGÍA DOCTORAL THESIS Biology PhD “Effect of a caloric restriction based on the Mediterranean diet and intake of traditional Mediterranean foods on the expression of microRNAs regulating molecular processes associated with aging” INSTITUTO MADRILEÑO DE ESTUDIOS AVANZADOS EN ALIMENTACIÓN (IMDEA FOOD INSTITUTE) VÍCTOR MICÓ MORENO Madrid, 2018 UNIVERSIDAD AUTÓNOMA DE MADRID FACULTAD DE CIENCIAS DEPARTAMENTO DE BIOLOGÍA DOCTORAL THESIS Biology PhD “Effect of a caloric restriction based on the Mediterranean diet and intake of traditional Mediterranean foods on the expression of microRNAs regulating molecular processes associated with aging” INSTITUTO MADRILEÑO DE ESTUDIOS AVANZADOS EN ALIMENTACIÓN (IMDEA FOOD INSTITUTE) Memoria presentada por: Víctor Micó Moreno Para optar al grado de: DOCTOR EN BIOLOGÍA Doña Lidia Ángeles Daimiel Ruíz, Doctora en Biología Celular y Genética por la Universidad Autónoma de Madrid, investigadora del Instituto IMDEA Alimentación, informa favorablemente la solicitud de autorización de defensa de la tesis doctoral con el Título: “Effect of a caloric restriction based on the Mediterranean diet and intake of traditional Mediterranean foods on the expression of microRNAs regulating molecular processes associated with aging”, presentada por Don Víctor Micó Moreno para optar al grado de Doctor en Biología. Este trabajo ha sido realizado en el Instituto Madrileño de Estudios Avanzados en Alimentación (IMDEA Alimentación) bajo su dirección, y cumple satisfactoriamente las condiciones requeridas por el Departamento de Biología de la Universidad Autónoma de Madrid para optar al Título de Doctor. Ha actuado como tutor académico, y presenta su conformidad el Dr. Carlos Francisco Sentís Castaño, vicedecano de Personal Docente e Investigador y profesor titular del Departamento de Biología de la Facultad de Ciencias de la Universidad Autónoma de Madrid.
    [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]
  • Supplemental Materials ZNF281 Enhances Cardiac Reprogramming
    Supplemental Materials ZNF281 enhances cardiac reprogramming by modulating cardiac and inflammatory gene expression Huanyu Zhou, Maria Gabriela Morales, Hisayuki Hashimoto, Matthew E. Dickson, Kunhua Song, Wenduo Ye, Min S. Kim, Hanspeter Niederstrasser, Zhaoning Wang, Beibei Chen, Bruce A. Posner, Rhonda Bassel-Duby and Eric N. Olson Supplemental Table 1; related to Figure 1. Supplemental Table 2; related to Figure 1. Supplemental Table 3; related to the “quantitative mRNA measurement” in Materials and Methods section. Supplemental Table 4; related to the “ChIP-seq, gene ontology and pathway analysis” and “RNA-seq” and gene ontology analysis” in Materials and Methods section. Supplemental Figure S1; related to Figure 1. Supplemental Figure S2; related to Figure 2. Supplemental Figure S3; related to Figure 3. Supplemental Figure S4; related to Figure 4. Supplemental Figure S5; related to Figure 6. Supplemental Table S1. Genes included in human retroviral ORF cDNA library. Gene Gene Gene Gene Gene Gene Gene Gene Symbol Symbol Symbol Symbol Symbol Symbol Symbol Symbol AATF BMP8A CEBPE CTNNB1 ESR2 GDF3 HOXA5 IL17D ADIPOQ BRPF1 CEBPG CUX1 ESRRA GDF6 HOXA6 IL17F ADNP BRPF3 CERS1 CX3CL1 ETS1 GIN1 HOXA7 IL18 AEBP1 BUD31 CERS2 CXCL10 ETS2 GLIS3 HOXB1 IL19 AFF4 C17ORF77 CERS4 CXCL11 ETV3 GMEB1 HOXB13 IL1A AHR C1QTNF4 CFL2 CXCL12 ETV7 GPBP1 HOXB5 IL1B AIMP1 C21ORF66 CHIA CXCL13 FAM3B GPER HOXB6 IL1F3 ALS2CR8 CBFA2T2 CIR1 CXCL14 FAM3D GPI HOXB7 IL1F5 ALX1 CBFA2T3 CITED1 CXCL16 FASLG GREM1 HOXB9 IL1F6 ARGFX CBFB CITED2 CXCL3 FBLN1 GREM2 HOXC4 IL1F7
    [Show full text]
  • Transcriptional Control of Tissue-Resident Memory T Cell Generation
    Transcriptional control of tissue-resident memory T cell generation Filip Cvetkovski Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Graduate School of Arts and Sciences COLUMBIA UNIVERSITY 2019 © 2019 Filip Cvetkovski All rights reserved ABSTRACT Transcriptional control of tissue-resident memory T cell generation Filip Cvetkovski Tissue-resident memory T cells (TRM) are a non-circulating subset of memory that are maintained at sites of pathogen entry and mediate optimal protection against reinfection. Lung TRM can be generated in response to respiratory infection or vaccination, however, the molecular pathways involved in CD4+TRM establishment have not been defined. Here, we performed transcriptional profiling of influenza-specific lung CD4+TRM following influenza infection to identify pathways implicated in CD4+TRM generation and homeostasis. Lung CD4+TRM displayed a unique transcriptional profile distinct from spleen memory, including up-regulation of a gene network induced by the transcription factor IRF4, a known regulator of effector T cell differentiation. In addition, the gene expression profile of lung CD4+TRM was enriched in gene sets previously described in tissue-resident regulatory T cells. Up-regulation of immunomodulatory molecules such as CTLA-4, PD-1, and ICOS, suggested a potential regulatory role for CD4+TRM in tissues. Using loss-of-function genetic experiments in mice, we demonstrate that IRF4 is required for the generation of lung-localized pathogen-specific effector CD4+T cells during acute influenza infection. Influenza-specific IRF4−/− T cells failed to fully express CD44, and maintained high levels of CD62L compared to wild type, suggesting a defect in complete differentiation into lung-tropic effector T cells.
    [Show full text]
  • The E–Id Protein Axis Modulates the Activities of the PI3K–AKT–Mtorc1
    Downloaded from genesdev.cshlp.org on October 6, 2021 - Published by Cold Spring Harbor Laboratory Press The E–Id protein axis modulates the activities of the PI3K–AKT–mTORC1– Hif1a and c-myc/p19Arf pathways to suppress innate variant TFH cell development, thymocyte expansion, and lymphomagenesis Masaki Miyazaki,1,8 Kazuko Miyazaki,1,8 Shuwen Chen,1 Vivek Chandra,1 Keisuke Wagatsuma,2 Yasutoshi Agata,2 Hans-Reimer Rodewald,3 Rintaro Saito,4 Aaron N. Chang,5 Nissi Varki,6 Hiroshi Kawamoto,7 and Cornelis Murre1 1Department of Molecular Biology, University of California at San Diego, La Jolla, California 92093, USA; 2Department of Biochemistry and Molecular Biology, Shiga University of Medical School, Shiga 520-2192, Japan; 3Division of Cellular Immunology, German Cancer Research Center, D-69120 Heidelberg, Germany; 4Department of Medicine, University of California at San Diego, La Jolla, California 92093, USA; 5Center for Computational Biology, Institute for Genomic Medicine, University of California at San Diego, La Jolla, California 92093, USA; 6Department of Pathology, University of California at San Diego, La Jolla, California 92093, USA; 7Department of Immunology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto 606-8507, Japan It is now well established that the E and Id protein axis regulates multiple steps in lymphocyte development. However, it remains unknown how E and Id proteins mechanistically enforce and maintain the naı¨ve T-cell fate. Here we show that Id2 and Id3 suppressed the development and expansion of innate variant follicular helper T (TFH) cells. Innate variant TFH cells required major histocompatibility complex (MHC) class I-like signaling and were associated with germinal center B cells.
    [Show full text]
  • Follicular Thyroglobulin Enhances Gene Expression Necessary for Thyroid Hormone Secretion
    Endocrine Journal 2015, 62 (11), 1007-1015 Original Follicular thyroglobulin enhances gene expression necessary for thyroid hormone secretion Yuko Ishido1), 2), 3), Yuqian Luo1), 3), Aya Yoshihara1), 3), Moyuru Hayashi3), Akio Yoshida2), Ichiro Hisatome2) and Koichi Suzuki1), 3) 1) Laboratory of Molecular Diagnostics, Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Disease, Tokyo 189-0002, Japan 2) Division of Regenerative Medicine and Therapeutics, Department of Genetic Medicine and Regenerative Therapeutics, Institute of Regenerative Medicine and Biofunction, Tottori University Graduate School of Medical Science, Yonago, 683-8503, Japan 3) Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Tokyo 173-8605, Japan Abstract. We have previously shown that follicular thyroglobulin (Tg) has an unexpected function as an autocrine negative- feedback regulator of thyroid hormone (TH) biosynthesis. Tg significantly suppressed the expression of genes necessary for iodide transport and TH synthesis by counteracting stimulation by TSH. However, whether follicular Tg also regulates intracellular TH transport and its secretion from thyrocytes is not known. In the present study, we examined the potential effect of follicular Tg on TH transport and secretion by quantifying the expression of two TH transporters: monocarboxylate transporter 8 (MCT8) and μ-crystallin (CRYM). Our results showed that follicular Tg at physiologic concentrations enhanced both the mRNA and protein expression levels of MCT8 and CRYM in a time- and dose-dependent manner in rat thyroid FRTL-5 cells. Although both the sodium/iodide symporter (NIS), an essential transporter of iodide from blood into the thyroid, and MCT8, a transporter of synthesized TH from the gland, were co-localized on the basolateral membrane of rat thyrocytes in vivo, Tg decreased NIS expression and increased the expression of MCT8 by counteracting TSH action.
    [Show full text]
  • Natural Course of Congenital Hypothyroidism by Dual Oxidase 2 Mutations from the Neonatal Period Through Puberty
    Y Maruo and others Clinical features of DUOX2 174:4 453–463 Clinical Study defects Natural course of congenital hypothyroidism by dual oxidase 2 mutations from the neonatal period through puberty Yoshihiro Maruo1, Keisuke Nagasaki, Katsuyuki Matsui, Yu Mimura, Asami Mori, Maki Fukami2 and Yoshihiro Takeuchi Correspondence should be addressed Department of Pediatrics, Shiga University of Medical Science, Tsukinowa, Seta, Otsu, Shiga 520-2192, Japan, to Y Maruo 1Department of Pediatrics, Niigata University, Niigata, Japan and 2Department of Molecular Endocrinology, Email National Research Institute for Child Health and Development, Tokyo, Japan [email protected] Abstract Aim: We previously reported that biallelic mutations in dual oxidase 2 (DUOX2) cause transient hypothyroidism. Since then, many cases with DUOX2 mutations have been reported. However, the clinical features and prognosis of individuals with DUOX2 defects have not been clarified. Objective: We investigated the prognosis of patients with congenital hypothyroidism (CH) due to DUOX2 mutations. Patients: Twenty-five patients were identified by a neonatal screening program and included seven familial cases. Their serum TSH values ranged from 18.9 to 734.6 mU/l. Twenty-two of the patients had low serum free thyroxine (fT4) levels (0.17–1.1 ng/dl). Twenty-four of the patients were treated with L-thyroxine. Methods: We analyzed the DUOX2, thyroid peroxidase, NaC/IK symporter, and dual oxidase maturation factor 2 genes of these 25 patients by PCR-amplified direct sequencing. An additional 11 genes were analyzed in 11 of the 25 patients using next-generation sequencing. Results: All patients had biallelic DUOX2 mutations, and seven novel alleles were detected.
    [Show full text]
  • 140503 IPF Signatures Supplement Withfigs Thorax
    Supplementary material for Heterogeneous gene expression signatures correspond to distinct lung pathologies and biomarkers of disease severity in idiopathic pulmonary fibrosis Daryle J. DePianto1*, Sanjay Chandriani1⌘*, Alexander R. Abbas1, Guiquan Jia1, Elsa N. N’Diaye1, Patrick Caplazi1, Steven E. Kauder1, Sabyasachi Biswas1, Satyajit K. Karnik1#, Connie Ha1, Zora Modrusan1, Michael A. Matthay2, Jasleen Kukreja3, Harold R. Collard2, Jackson G. Egen1, Paul J. Wolters2§, and Joseph R. Arron1§ 1Genentech Research and Early Development, South San Francisco, CA 2Department of Medicine, University of California, San Francisco, CA 3Department of Surgery, University of California, San Francisco, CA ⌘Current address: Novartis Institutes for Biomedical Research, Emeryville, CA. #Current address: Gilead Sciences, Foster City, CA. *DJD and SC contributed equally to this manuscript §PJW and JRA co-directed this project Address correspondence to Paul J. Wolters, MD University of California, San Francisco Department of Medicine Box 0111 San Francisco, CA 94143-0111 [email protected] or Joseph R. Arron, MD, PhD Genentech, Inc. MS 231C 1 DNA Way South San Francisco, CA 94080 [email protected] 1 METHODS Human lung tissue samples Tissues were obtained at UCSF from clinical samples from IPF patients at the time of biopsy or lung transplantation. All patients were seen at UCSF and the diagnosis of IPF was established through multidisciplinary review of clinical, radiological, and pathological data according to criteria established by the consensus classification of the American Thoracic Society (ATS) and European Respiratory Society (ERS), Japanese Respiratory Society (JRS), and the Latin American Thoracic Association (ALAT) (ref. 5 in main text). Non-diseased normal lung tissues were procured from lungs not used by the Northern California Transplant Donor Network.
    [Show full text]
  • Tu Cornellgrad 0058F 10421.Pdf (5.371Mb)
    MOLECULAR FUNCTION OF MAMMALIAN TRANSLOCATOR PROTEIN (TSPO) A Dissertation Presented to the Faculty of the Graduate School of Cornell University In Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy by Lan Ngoc Ly Tu August 2017 © 2017 Lan Ngoc Ly Tu MOLECULAR FUNCTION OF MAMMALIAN TRANSLOCATOR PROTEIN (TSPO) Lan Ngoc Ly Tu, Ph. D. Cornell University 2017 Translocator protein (TSPO), previously known as the peripheral benzodiazepine receptor (PBR), is a mitochondrial outer membrane protein highly conserved from bacteria to humans. TSPO is found to be upregulated in many pathological conditions, making it an attractive target for both diagnostic and therapeutic purposes in human medicine. However, the exact molecular function of TSPO remained unclear. For the past 25 years, TSPO was depicted to transport cholesterol from the cytosol to the inner mitochondrial membrane, the rate-limiting step in steroid hormone biosynthesis. Its critical role in survival and development was reinforced by a report that claimed TSPO knockout (Tspo-/-) mice were embryonic lethal. Therefore, there had been no genetic models to study the function of TSPO and all functional interpretations of TSPO were based on in vitro experiments using pharmacological tools. Our lab generated the first global Tspo-/- mice which surprisingly were healthy with no apparent abnormalities. Deletion of TSPO in different steroidogenic cell lines also did not affect cell viability. Furthermore, steroid hormone production was not affected in Tspo-/- mice or in steroidogenic cells lacking TSPO compared to the controls, indicating that TSPO is not involved in steroidogenesis. The stimulating effect of some TSPO binding chemicals on steroid hormone production that formed the early basis for linking TSPO and steroidogenesis was also found to be inaccurate.
    [Show full text]
  • Pancreatic Cancer Prognosis Is Predicted by a Novel ATAC-Array Technology For
    bioRxiv preprint doi: https://doi.org/10.1101/2021.01.21.427604; this version posted January 22, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Pancreatic cancer prognosis is predicted by a novel ATAC-array technology for assessing chromatin accessibility Authors: Dhara S1, Chhangawala S2,3, Chintalapudi H1, Askan G4, Aveson V4,5, Massa AL1, Zhang L4, Torres D1, Makohon-Moore AP4, Lecomte N4, Melchor JP4, Bermeo J4, Cardenas III A4, Sinha S4, Glassman D4, Nicolle R7, Moffitt R6, Yu KH4, Leppanen S8, Laderman S8, Curry B8, Gui J1, Balachandran VP4, Iacobuzio-Donahue C4, Chandwani R5, Leslie CS3* and Leach SD1* Author affiliations: 1, Dartmouth Geisel School of Medicine and Norris Cotton Cancer Center, Hanover, NH, USA 2, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA. 3, Computational Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA. 4, David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA 5, Weill Cornell Medicine, New York, NY, USA 6, Stony Brook University, Stony Brook, NY, USA 7, Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre Le Cancer, Paris, France 8, Agilent Technologies Inc. Santa Clara, CA, USA (* Co-corresponding author e-mails: [email protected]; [email protected]) (First two authors contributed equally to this work) 1 bioRxiv preprint doi: https://doi.org/10.1101/2021.01.21.427604; this version posted January 22, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder.
    [Show full text]