Bilirubin Oxidation End Products (Boxes) Induce Neuronal Oxidative Stress Involving the Nrf2 Pathway

Total Page:16

File Type:pdf, Size:1020Kb

Bilirubin Oxidation End Products (Boxes) Induce Neuronal Oxidative Stress Involving the Nrf2 Pathway Hindawi Oxidative Medicine and Cellular Longevity Volume 2021, Article ID 8869908, 11 pages https://doi.org/10.1155/2021/8869908 Research Article Bilirubin Oxidation End Products (BOXes) Induce Neuronal Oxidative Stress Involving the Nrf2 Pathway Yinzhong Lu ,1,2 Wenyi Zhang,1 Bing Zhang,1 Stefan H. Heinemann,3 Toshinori Hoshi,4 Shangwei Hou ,1,2 and Guangming Zhang 1 1Department of Anesthesiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China 2Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China 3Center for Molecular Biomedicine, Department of Biophysics, Friedrich Schiller University Jena & Jena University Hospital, Hans- Knöll-Str. 2, D-07745 Jena, Germany 4Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104, USA Correspondence should be addressed to Shangwei Hou; [email protected] and Guangming Zhang; [email protected] Received 9 October 2020; Revised 4 June 2021; Accepted 22 June 2021; Published 31 July 2021 Academic Editor: Cinzia Signorini Copyright © 2021 Yinzhong Lu et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Delayed ischemic neurological deficit (DIND) is a severe complication after subarachnoid hemorrhage (SAH). Previous studies have suggested that bilirubin oxidation end products (BOXes) are probably associated with the DIND after SAH, but there is a lack of direct evidence yet even on cellular levels. In the present study, we aim to explore the potential role of BOXes and the involved mechanisms in neuronal function. We synthesized high-purity (>97%) BOX A and BOX B isomers. The pharmacokinetics showed they are permeable to the blood-brain barrier. Exposure of a moderate concentration (10 or 30 μM) of BOX A or BOX B to isolated primary cortical neurons increased the production of reactive oxygen species. In the human neuroblastoma SH-SY5Y cells, BOX A and BOX B decreased the mitochondrial membrane potential and enhanced nuclear accumulation of the protein Nrf2 implicated in oxidative injury repair. In addition, both chemicals increased the mRNA and protein expression levels of multiple antioxidant response genes including Hmox1, Gsta3, Blvrb, Gclm, and Srxn1, indicating that the antioxidant response element (ARE) transcriptional cascade driven by Nrf2 is activated. In conclusion, we demonstrated that primary cortical neurons and neuroblastoma cells undergo an adaptive response against BOX A- and BOX B-mediated oxidative stress by activation of multiple antioxidant responses, in part through the Nrf2 pathway, which provides in-depth insights into the pathophysiological mechanism of DIND after SAH or other neurological dysfunctions related to cerebral hemorrhage. 1. Introduction initial impact of SAH and have had their aneurysm effectively treated [2]. However, the etiology and pathophysiology of Subarachnoid hemorrhage (SAH) is a serious cerebrovascu- SAH and DNID remain incompletely understood. lar complication with complex underlying mechanisms Accumulating evidence has shown that oxidative stress, inflicting brain perfusion and function. Despite great prog- acute and subsequent consisting immunological response, ress in the understanding of SAH pathophysiology and man- and other factors collectively cause the severe adverse effects agement of ruptured aneurysms, SAH remains a severe and of SAH [3, 4]. Erythrocyte lysis, a major initiator, is the significant health problem [1]. SAH causes early brain injury, release of heme and its subsequent degradation cascade, which may be followed after 7 to 14 days by delayed ischemic which collectively exert a strong effect on the affected neu- neurological deficit (DIND). DIND is the leading cause of rons and their electrical excitability [5–7]. Various heme morbidity and mortality in the patients who survive the and heme degradation products (HHDPs) were identified 2 Oxidative Medicine and Cellular Longevity in the cerebrospinal fluid (CSF) from the patients after SAH mm column with gradient elution using mobile phase A con- or other types of brain hemorrhage [8–11] and serum from taining 0.1% formic acid in water and mobile phase B (0.1% hepatic failure patients [12]. formic acid in acetonitrile) at a flow rate of 500 μL/min. Notably, an array of bilirubin oxidation end products The gradient run was as follows: 10% solvent B (0.01–0.30 (BOXes) with small molecular weight were identified [11, min), from 10% to 98% B (0.30–1.30 min), 98% B (1.30– 13, 14] and detected in the CSF of a patient with DIND after 1.80 min), from 98%B to 10% B (1.80–1.90 min), and 10%B SAH [8, 9, 15]. Several studies using crude extraction or (1.90–2.40 min). Detection was performed by MS/MS using synthesized BOXes have shown that BOXes constrict mouse an API4000 electrospray ionization (ESI) mass spectropho- cortical blood vessels and damage the normal contractile of tometer (Applied Biosystems, API 4000 triple quadrupole vascular smooth-muscle cells [16, 17], suggesting that BOXes mass spectrometer, AB SCIEX, Concord, Ontario, Canada). are involved in the delayed cerebral vasospasm, a potential The masses for tolbutamide were precursor ion m/z 271.2 cause of DIND. Our previous study showed that a crude and product ion m/z 155.1, for BOX A, precursor ion m/z BOXes mixture regulates the blood tone by modulation of 179.2 and product ion m/z 162.2, and for BOX B, precursor 2+ + Ca - and voltage-gated K channels (Slo BKCa) [18]. How- ion m/z 179.2 and product ion m/z 162.3. ever, the direct impact of BOXes on neuronal cells remains For pharmacological kinetics study, a single dose of 12 to be examined. mg/kg body weight BOX A or BOX B (powder dissolved in In this study, we hypothesized that BOX A or BOX B are 5% DMSO : 95% saline (pH = 11) [20, 21] with final concen- important functional regulators of neurons during the pro- tration 2.4 mg/mL) was injected in vein (i.v.) with 5 mL/kg gression of DIND after SAH. Therefore, we synthesized body weight in male ICR mice (n =3) obtained from Beijing BOX A and BOX B following the reported protocol [14, 19] Vital River Laboratory Animal Technology Co., Ltd. (Beijing, and examined their biological effects on cultured primary China). After 30 min, the plasma concentrations (ng/mL) cortical neurons and neuronal cell lines to elucidate the and the brain concentrations (ng/g) were measured. For underlying molecular mechanisms. preparations of mouse plasma and brain samples, 100 μL cal- ibration curve samples in single, QC samples in duplicate, 2. Materials and Methods and mouse plasma samples were mixed with 300 μL acetoni- trile containing Internal standard (50 ng/mL of propranolol, 2.1. Reagent and Antibodies. 2′,7′-Dichlorofluorescein diace- 200 ng/mL of tolbutamide, and 50 ng/mL of diclofenac) in tate (DCF-DA) and dimethyl sulfoxide (DMSO) were Eppendorf tubes. After the mixture was vortexed for 1 min, purchased from Sigma-Aldrich, the antibody raised against then centrifuged for 10 min, transfer 50 μL supernatant to a HO-1 (1 : 500), BLVRB (1 : 500), and Gclm (1 : 1000) from Pro- 96- well plate with 100 μL pure water, shaking for 10 min, teintech Group, Inc. (Chicago, IL, USA); Nrf2 (WB, 1 : 1000, IF, and finally inject 10 μL into LC-MS/MS system. For brain 1 : 200) and Gsta3 (1 : 500) from Abcam (Cambridge, MA, samples, brain samples were added with 5 folds (w/v) phos- USA); SRXN1 (1 : 500) from Bioss (Beijing, China); Caspase 3 phate buffer (100 mM, pH 7.4) in terms of the weight of (WB, 1 : 1000) from Abmart (Shanghai, China); and β-actin samples, then homogenated. Subsequent procedures were (WB, 1 : 2000) from OriGene Technologies, Inc. (Rockville, performed as described for plasma. The brain concentration MD, USA). Alexa Fluor 546 anti-mouse secondary Ab (IF:1 : to the plasma concentration was calculated as B/P value. The 2000, A-11030), goat anti-mouse IgG (H + L) peroxidase animal study including the use of embryos for primary neu- conjugated (WB: 1 : 20000, 31430), and goat anti-rabbit IgG ron cultures was carried out in accordance with regulations (H + L) peroxidase conjugated (WB: 1 : 20000, 31460) were pur- for animal experimentation and were approved by the chased from Thermo Fisher Scientific(Waltham,MA,USA). Animal Committee of Tongren Hospital affiliated to Shang- hai Jiao Tong University School of Medicine. 2.2. BOXes Synthesis and B/P Study. BOX A and BOX B were For other assays in this study, the ready-for-use BOX A synthesized as shown in Figure 1(a). Compound 1 (bromoci- or BOX B solution was prepared as 1,000 times in DMSO traconic anhydride) reacts with triphenylphosphoylide in as the final concentration in administered cells and stored toluene with reflux. Because the two carbonyl reaction sites at -20°C in the dark. Therefore, the final concentration of are different, two products can be obtained: 3 is the primary DMSO in all treatments was 0.1%. product, 2 is the by-product, and two of which can be sepa- rated by silica gel column chromatography. Compounds 2 2.3. Primary Cortical Neurons and Neuroblastoma Cell Lines. and 3 have the reactivity of palladium-catalyzed coupling Mouse primary cortical neurons (PCNs) were cultured as reaction and amine acetate substitution reaction (the reac- previously described [22] with minor modifications. Briefly, tion order is not required). Then, compounds 6 and 7 were E17.5 ~ E18.5 embryos of either sex were dissected from hydrolyzed into carboxylic acid intermediates by alkali pregnant C57BL/6J mice for cortex dissection in cold hydrolysis, and then the target products BOX A and BOX B Dulbecco’s Modified Eagle Medium (DMEM).
Recommended publications
  • Nrf2 Modulates Host Defense During Streptococcus Pneumoniae Pneumonia in Mice
    The Journal of Immunology Nrf2 Modulates Host Defense during Streptococcus pneumoniae Pneumonia in Mice John C. Gomez,*,† Hong Dang,†,‡ Jessica R. Martin,*,† and Claire M. Doerschuk*,†,x Nrf2 regulates the transcriptional response to oxidative stress. These studies tested the role of Nrf2 during Streptococcus pneumoniae pneumonia and identified Nrf2-dependent genes and pathways in lung tissue and in recruited neutrophils. Nrf2 null and wild type (WT) mice were studied at 6 and 24 h after instillation of S. pneumoniae or PBS. At 6 h, fewer neutrophils were recruited and the number of bacteria remaining in the lungs tended to be less (p = 0.06) in the Nrf2 null compared with WT mice. In uninfected lungs, 53 genes were already differentially expressed in Nrf2 null compared with WT mouse lungs, and gene sets involved in phagocytosis, Fc receptor function, complement, and Ig regulation are enhanced in PBS-treated Nrf2 null gene profiles compared with those of WT mice. These results suggest that initial host defense is enhanced in Nrf2 null mice, resulting in less recruitment of neutrophils. At 24 h, neutrophil recruitment was greater. The percentages of early apoptotic and late apoptotic/necrotic neutrophils were similar. At increasing inoculum numbers, mortality rates strikingly increased from 15 to 31 and 100% in Nrf2 null mice, whereas all WT mice survived, and Nrf2 null mice had a defect in clearance, particularly at the intermediate dose. The mortality was due to enhanced lung injury and greater systemic response. Gene profiling identified differentially regulated genes and pathways in neutrophils and lung tissue, including those involved in redox stress response, metabolism, inflammation, immunoregulatory pathways, and tissue repair, providing insight into the mechanisms for the greater tissue damage and increased neutrophil accumulation.
    [Show full text]
  • Cellular and Molecular Signatures in the Disease Tissue of Early
    Cellular and Molecular Signatures in the Disease Tissue of Early Rheumatoid Arthritis Stratify Clinical Response to csDMARD-Therapy and Predict Radiographic Progression Frances Humby1,* Myles Lewis1,* Nandhini Ramamoorthi2, Jason Hackney3, Michael Barnes1, Michele Bombardieri1, Francesca Setiadi2, Stephen Kelly1, Fabiola Bene1, Maria di Cicco1, Sudeh Riahi1, Vidalba Rocher-Ros1, Nora Ng1, Ilias Lazorou1, Rebecca E. Hands1, Desiree van der Heijde4, Robert Landewé5, Annette van der Helm-van Mil4, Alberto Cauli6, Iain B. McInnes7, Christopher D. Buckley8, Ernest Choy9, Peter Taylor10, Michael J. Townsend2 & Costantino Pitzalis1 1Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK. Departments of 2Biomarker Discovery OMNI, 3Bioinformatics and Computational Biology, Genentech Research and Early Development, South San Francisco, California 94080 USA 4Department of Rheumatology, Leiden University Medical Center, The Netherlands 5Department of Clinical Immunology & Rheumatology, Amsterdam Rheumatology & Immunology Center, Amsterdam, The Netherlands 6Rheumatology Unit, Department of Medical Sciences, Policlinico of the University of Cagliari, Cagliari, Italy 7Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK 8Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Birmingham B15 2WB, UK 9Institute of
    [Show full text]
  • Gene Section Short Communication
    Atlas of Genetics and Cytogenetics in Oncology and Haematology INIST -CNRS OPEN ACCESS JOURNAL Gene Section Short Communication SRXN1 (sulfiredoxin 1) Hedy A Chawsheen, Hong Jiang, Qiou Wei Graduate Center for Toxicology, College of Medicine, University of Kentucky, Lexington, Kentucky 40513, USA (HAC, HJ, QW) Published in Atlas Database: November 2012 Online updated version : http://AtlasGeneticsOncology.org/Genes/SRXN1ID52295ch20p13.html DOI: 10.4267/2042/48870 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2013 Atlas of Genetics and Cytogenetics in Oncology and Haematology Identity Expression In adult, Srx protein was found in internal organs such Other names: C20orf139, Npn3, SRX1, YKL086W, as mouse liver and kidney. Expression pattern of Srx in dJ850E9.2 embryonic development is not clear. Transcriptional HGNC (Hugo): SRXN1 regulation of Srx expression is mainly mediated Location: 20p13 through AP-1 and/or Nrf-2 activation (Jeong et al., 2012). In yeast, it may also be negatively regulated at DNA/RNA the translational level through Ras-PKA pathway (Molin et al., 2011). Note Localisation Human Srx is located on chromosome 20 in the region of p13. Srx is mainly localized in the cytosol. In the presence of severe oxidative stress, it may also translocate to Description mitochondria (Noh et al., 2009). Human Srx gene is 6632 bp in length, composed of 2 Function exons and located at chromosome 20p13. Srx was first identified as a gene preferentially Transcription expressed in transformed JB6 cells (Sun et al., 1994). The size of Srx mRNA is 2580 bp. Srx transcript The primary biochemical function of Srx is to reduce contains two exons.
    [Show full text]
  • In Vivo Measurements of Interindividual Differences in DNA
    In vivo measurements of interindividual differences in PNAS PLUS DNA glycosylases and APE1 activities Isaac A. Chaima,b, Zachary D. Nagela,b, Jennifer J. Jordana,b, Patrizia Mazzucatoa,b, Le P. Ngoa,b, and Leona D. Samsona,b,c,d,1 aDepartment of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139; bCenter for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139; cDepartment of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139; and dThe David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139 Edited by Paul Modrich, Howard Hughes Medical Institute and Duke University Medical Center, Durham, NC, and approved October 20, 2017 (received for review July 6, 2017) The integrity of our DNA is challenged with at least 100,000 lesions with increased cancer risk and other diseases (8–10). However, per cell on a daily basis. Failure to repair DNA damage efficiently the lack of high-throughput assays that can reliably measure in- can lead to cancer, immunodeficiency, and neurodegenerative dis- terindividual differences in BER capacity have limited epidemio- ease. Base excision repair (BER) recognizes and repairs minimally logical studies linking BER capacity to disease. Moreover, BER helix-distorting DNA base lesions induced by both endogenous repairs DNA lesions induced by radiation and chemotherapy (3, 11), and exogenous DNA damaging agents. Levels of BER-initiating raising the possibility of personalized treatment strategies using DNA glycosylases can vary between individuals, suggesting that BER capacity in tumor tissue to predict which therapies are most quantitating and understanding interindividual differences in DNA likely to be effective, and using BER capacity in healthy tissue to repair capacity (DRC) may enable us to predict and prevent disease determine the dose individual patients can tolerate.
    [Show full text]
  • Network Inference Algorithms Elucidate Nrf2 Regulation of Mouse Lung Oxidative Stress
    Network Inference Algorithms Elucidate Nrf2 Regulation of Mouse Lung Oxidative Stress Ronald C. Taylor1.*, George Acquaah-Mensah2., Mudita Singhal1, Deepti Malhotra3, Shyam Biswal3 1 Computational Biology and Bioinformatics Group, Pacific Northwest National Laboratory, U.S. Department of Energy, Richland, Washington, United States of America, 2 Department of Pharmaceutical Sciences, Massachusetts College of Pharmacy and Health Sciences, Worcester, Massachusetts, United States of America, 3 Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America Abstract A variety of cardiovascular, neurological, and neoplastic conditions have been associated with oxidative stress, i.e., conditions under which levels of reactive oxygen species (ROS) are elevated over significant periods. Nuclear factor erythroid 2-related factor (Nrf2) regulates the transcription of several gene products involved in the protective response to oxidative stress. The transcriptional regulatory and signaling relationships linking gene products involved in the response to oxidative stress are, currently, only partially resolved. Microarray data constitute RNA abundance measures representing gene expression patterns. In some cases, these patterns can identify the molecular interactions of gene products. They can be, in effect, proxies for protein–protein and protein–DNA interactions. Traditional techniques used for clustering coregulated genes on high-throughput gene arrays are rarely
    [Show full text]
  • Mechanistic Analysis of an Extracellular Signal-Regulated
    Supplemental material to this article can be found at: http://jpet.aspetjournals.org/content/suppl/2020/10/26/jpet.120.000266.DC1 1521-0103/376/1/84–97$35.00 https://doi.org/10.1124/jpet.120.000266 THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS J Pharmacol Exp Ther 376:84–97, January 2021 Copyright ª 2020 by The Author(s) This is an open access article distributed under the CC BY-NC Attribution 4.0 International license. Mechanistic Analysis of an Extracellular Signal–Regulated Kinase 2–Interacting Compound that Inhibits Mutant BRAF-Expressing Melanoma Cells by Inducing Oxidative Stress s Ramon Martinez, III,1 Weiliang Huang,1 Ramin Samadani, Bryan Mackowiak, Garrick Centola, Lijia Chen, Ivie L. Conlon, Kellie Hom, Maureen A. Kane, Steven Fletcher, and Paul Shapiro Department of Pharmaceutical Sciences, University of Maryland, Baltimore- School of Pharmacy, Baltimore, Maryland Received August 3, 2020; accepted October 6, 2020 Downloaded from ABSTRACT Constitutively active extracellular signal–regulated kinase (ERK) 1/2 (MEK1/2) or ERK1/2. Like other ERK1/2 pathway inhibitors, 1/2 signaling promotes cancer cell proliferation and survival. We SF-3-030 induced reactive oxygen species (ROS) and genes previously described a class of compounds containing a 1,1- associated with oxidative stress, including nuclear factor ery- dioxido-2,5-dihydrothiophen-3-yl 4-benzenesulfonate scaffold throid 2–related factor 2 (NRF2). Whereas the addition of the jpet.aspetjournals.org that targeted ERK2 substrate docking sites and selectively ROS inhibitor N-acetyl cysteine reversed SF-3-030–induced inhibited ERK1/2-dependent functions, including activator ROS and inhibition of A375 cell proliferation, the addition of protein-1–mediated transcription and growth of cancer cells NRF2 inhibitors has little effect on cell proliferation.
    [Show full text]
  • Mouse Srxn1 Conditional Knockout Project (CRISPR/Cas9)
    https://www.alphaknockout.com Mouse Srxn1 Conditional Knockout Project (CRISPR/Cas9) Objective: To create a Srxn1 conditional knockout Mouse model (C57BL/6J) by CRISPR/Cas-mediated genome engineering. Strategy summary: The Srxn1 gene (NCBI Reference Sequence: NM_029688 ; Ensembl: ENSMUSG00000032802 ) is located on Mouse chromosome 2. 2 exons are identified, with the ATG start codon in exon 1 and the TAG stop codon in exon 2 (Transcript: ENSMUST00000041500). Exon 1 will be selected as conditional knockout region (cKO region). Deletion of this region should result in the loss of function of the Mouse Srxn1 gene. To engineer the targeting vector, homologous arms and cKO region will be generated by PCR using BAC clone RP23-214I20 as template. Cas9, gRNA and targeting vector will be co-injected into fertilized eggs for cKO Mouse production. The pups will be genotyped by PCR followed by sequencing analysis. Note: Mice homozygous for a knock-out allele exhibit increased sensitivity to LPS-induced shock. Exon 1 covers 56.77% of the coding region. Start codon is in exon 1, and stop codon is in exon 2. The size of intron 1 for 3'-loxP site insertion: 3041 bp. The size of effective cKO region: ~524 bp. The cKO region does not have any other known gene. Page 1 of 7 https://www.alphaknockout.com Overview of the Targeting Strategy gRNA region Wildtype allele A gRNA region T 5' G 3' 1 2 Targeting vector A T G Targeted allele A T G Constitutive KO allele (After Cre recombination) Legends Homology arm Exon of mouse Srxn1 cKO region loxP site Page 2 of 7 https://www.alphaknockout.com Overview of the Dot Plot Window size: 10 bp Forward Reverse Complement Sequence 12 Note: The sequence of homologous arms and cKO region is aligned with itself to determine if there are tandem repeats.
    [Show full text]
  • UC San Diego Electronic Theses and Dissertations
    UC San Diego UC San Diego Electronic Theses and Dissertations Title Cardiac Stretch-Induced Transcriptomic Changes are Axis-Dependent Permalink https://escholarship.org/uc/item/7m04f0b0 Author Buchholz, Kyle Stephen Publication Date 2016 Peer reviewed|Thesis/dissertation eScholarship.org Powered by the California Digital Library University of California UNIVERSITY OF CALIFORNIA, SAN DIEGO Cardiac Stretch-Induced Transcriptomic Changes are Axis-Dependent A dissertation submitted in partial satisfaction of the requirements for the degree Doctor of Philosophy in Bioengineering by Kyle Stephen Buchholz Committee in Charge: Professor Jeffrey Omens, Chair Professor Andrew McCulloch, Co-Chair Professor Ju Chen Professor Karen Christman Professor Robert Ross Professor Alexander Zambon 2016 Copyright Kyle Stephen Buchholz, 2016 All rights reserved Signature Page The Dissertation of Kyle Stephen Buchholz is approved and it is acceptable in quality and form for publication on microfilm and electronically: Co-Chair Chair University of California, San Diego 2016 iii Dedication To my beautiful wife, Rhia. iv Table of Contents Signature Page ................................................................................................................... iii Dedication .......................................................................................................................... iv Table of Contents ................................................................................................................ v List of Figures ...................................................................................................................
    [Show full text]
  • Activation of the Nrf2 Response by Intrinsic Hepatotoxic Drugs Correlates with Suppression of NF‑Κb Activation and Sensitizes Toward Tnfα‑Induced Cytotoxicity
    Arch Toxicol (2016) 90:1163–1179 DOI 10.1007/s00204-015-1536-3 ORGAN TOXICITY AND MECHANISMS Activation of the Nrf2 response by intrinsic hepatotoxic drugs correlates with suppression of NF‑κB activation and sensitizes toward TNFα‑induced cytotoxicity Bram Herpers1 · Steven Wink1 · Lisa Fredriksson1 · Zi Di1 · Giel Hendriks2 · Harry Vrieling2 · Hans de Bont1 · Bob van de Water1 Received: 27 January 2015 / Accepted: 12 May 2015 / Published online: 31 May 2015 © The Author(s) 2015. This article is published with open access at Springerlink.com Abstract Drug-induced liver injury (DILI) is an impor- toward TNFα-mediated cytotoxicity. This was related to an tant problem both in the clinic and in the development of adaptive primary protective response of Nrf2, since loss of new safer medicines. Two pivotal adaptation and survival Nrf2 enhanced this cytotoxic synergy with TNFα, while responses to adverse drug reactions are oxidative stress and KEAP1 downregulation was cytoprotective. These data indi- cytokine signaling based on the activation of the transcrip- cate that both Nrf2 and NF-κB signaling may be pivotal in tion factors Nrf2 and NF-κB, respectively. Here, we system- the regulation of DILI. We propose that the NF-κB-inhibiting atically investigated Nrf2 and NF-κB signaling upon DILI- effects that coincide with a strong Nrf2 stress response likely related drug exposure. Transcriptomics analyses of 90 DILI sensitize liver cells to pro-apoptotic signaling cascades compounds in primary human hepatocytes revealed that a induced by intrinsic cytotoxic pro-inflammatory cytokines. strong Nrf2 activation is associated with a suppression of endogenous NF-κB activity.
    [Show full text]
  • PP1-Associated Signaling and − B/AP-1 Κ Inhibition of NF- Tolerance
    Downloaded from http://www.jimmunol.org/ by guest on October 3, 2021 is online at: average * and − B/AP-1 κ The Journal of Immunology published online 26 February 2014 from submission to initial decision 4 weeks from acceptance to publication http://www.jimmunol.org/content/early/2014/02/26/jimmun ol.1301610 Identification of Two Forms of TNF Tolerance in Human Monocytes: Differential Inhibition of NF- PP1-Associated Signaling Johannes Günther, Nico Vogt, Katharina Hampel, Rolf Bikker, Sharon Page, Benjamin Müller, Judith Kandemir, Michael Kracht, Oliver Dittrich-Breiholz, René Huber and Korbinian Brand J Immunol 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/2014/02/26/jimmunol.130161 0.DCSupplemental Information about subscribing to The JI No Triage! Fast Publication! Rapid Reviews! 30 days* Why • • • Material Permissions Email Alerts Subscription Supplementary The Journal of Immunology The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2014 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. This information is current as of October 3, 2021. Published February 26, 2014, doi:10.4049/jimmunol.1301610 The Journal of Immunology Identification of Two Forms of TNF Tolerance in Human Monocytes: Differential Inhibition of NF-kB/AP-1– and PP1-Associated Signaling Johannes Gunther,*€ ,1 Nico Vogt,*,1 Katharina Hampel,*,1 Rolf Bikker,* Sharon Page,* Benjamin Muller,*€ Judith Kandemir,* Michael Kracht,† Oliver Dittrich-Breiholz,‡ Rene´ Huber,* and Korbinian Brand* The molecular basis of TNF tolerance is poorly understood.
    [Show full text]
  • Identification of Novel Regulatory Genes in Acetaminophen
    IDENTIFICATION OF NOVEL REGULATORY GENES IN ACETAMINOPHEN INDUCED HEPATOCYTE TOXICITY BY A GENOME-WIDE CRISPR/CAS9 SCREEN A THESIS IN Cell Biology and Biophysics and Bioinformatics Presented to the Faculty of the University of Missouri-Kansas City in partial fulfillment of the requirements for the degree DOCTOR OF PHILOSOPHY By KATHERINE ANNE SHORTT B.S, Indiana University, Bloomington, 2011 M.S, University of Missouri, Kansas City, 2014 Kansas City, Missouri 2018 © 2018 Katherine Shortt All Rights Reserved IDENTIFICATION OF NOVEL REGULATORY GENES IN ACETAMINOPHEN INDUCED HEPATOCYTE TOXICITY BY A GENOME-WIDE CRISPR/CAS9 SCREEN Katherine Anne Shortt, Candidate for the Doctor of Philosophy degree, University of Missouri-Kansas City, 2018 ABSTRACT Acetaminophen (APAP) is a commonly used analgesic responsible for over 56,000 overdose-related emergency room visits annually. A long asymptomatic period and limited treatment options result in a high rate of liver failure, generally resulting in either organ transplant or mortality. The underlying molecular mechanisms of injury are not well understood and effective therapy is limited. Identification of previously unknown genetic risk factors would provide new mechanistic insights and new therapeutic targets for APAP induced hepatocyte toxicity or liver injury. This study used a genome-wide CRISPR/Cas9 screen to evaluate genes that are protective against or cause susceptibility to APAP-induced liver injury. HuH7 human hepatocellular carcinoma cells containing CRISPR/Cas9 gene knockouts were treated with 15mM APAP for 30 minutes to 4 days. A gene expression profile was developed based on the 1) top screening hits, 2) overlap with gene expression data of APAP overdosed human patients, and 3) biological interpretation including assessment of known and suspected iii APAP-associated genes and their therapeutic potential, predicted affected biological pathways, and functionally validated candidate genes.
    [Show full text]
  • Table S1. 103 Ferroptosis-Related Genes Retrieved from the Genecards
    Table S1. 103 ferroptosis-related genes retrieved from the GeneCards. Gene Symbol Description Category GPX4 Glutathione Peroxidase 4 Protein Coding AIFM2 Apoptosis Inducing Factor Mitochondria Associated 2 Protein Coding TP53 Tumor Protein P53 Protein Coding ACSL4 Acyl-CoA Synthetase Long Chain Family Member 4 Protein Coding SLC7A11 Solute Carrier Family 7 Member 11 Protein Coding VDAC2 Voltage Dependent Anion Channel 2 Protein Coding VDAC3 Voltage Dependent Anion Channel 3 Protein Coding ATG5 Autophagy Related 5 Protein Coding ATG7 Autophagy Related 7 Protein Coding NCOA4 Nuclear Receptor Coactivator 4 Protein Coding HMOX1 Heme Oxygenase 1 Protein Coding SLC3A2 Solute Carrier Family 3 Member 2 Protein Coding ALOX15 Arachidonate 15-Lipoxygenase Protein Coding BECN1 Beclin 1 Protein Coding PRKAA1 Protein Kinase AMP-Activated Catalytic Subunit Alpha 1 Protein Coding SAT1 Spermidine/Spermine N1-Acetyltransferase 1 Protein Coding NF2 Neurofibromin 2 Protein Coding YAP1 Yes1 Associated Transcriptional Regulator Protein Coding FTH1 Ferritin Heavy Chain 1 Protein Coding TF Transferrin Protein Coding TFRC Transferrin Receptor Protein Coding FTL Ferritin Light Chain Protein Coding CYBB Cytochrome B-245 Beta Chain Protein Coding GSS Glutathione Synthetase Protein Coding CP Ceruloplasmin Protein Coding PRNP Prion Protein Protein Coding SLC11A2 Solute Carrier Family 11 Member 2 Protein Coding SLC40A1 Solute Carrier Family 40 Member 1 Protein Coding STEAP3 STEAP3 Metalloreductase Protein Coding ACSL1 Acyl-CoA Synthetase Long Chain Family Member 1 Protein
    [Show full text]