Discovery and Characterization of Natural Products As Novel Indoleamine 2,3-Dioxygenase 1 Inhibitors Through High-Throughput Screening

Total Page:16

File Type:pdf, Size:1020Kb

Discovery and Characterization of Natural Products As Novel Indoleamine 2,3-Dioxygenase 1 Inhibitors Through High-Throughput Screening www.nature.com/aps ARTICLE Discovery and characterization of natural products as novel indoleamine 2,3-dioxygenase 1 inhibitors through high-throughput screening Wei Guo1,2, Sheng Yao3,4, Pu Sun1,2, Tian-biao Yang4, Chun-ping Tang3,4, Ming-yue Zheng4, Yang Ye3,4,5 and Ling-hua Meng1,2 Indoleamine 2,3-dioxygenase 1 (IDO1) is emerging as a promising therapeutic target for the treatment of malignant tumors characterized by dysregulated tryptophan metabolism. However, the antitumor efficacy of existing small-molecule IDO1 inhibitors is still unsatisfactory, and the underlying mechanism remains largely undefined. To identify novel IDO1 inhibitors, an in-house natural product library of 2000 natural products was screened for inhibitory activity against recombinant human IDO1. High- throughput fluorescence-based screening identified 79 compounds with inhibitory activity > 30% at 20 μM. Nine natural products were further confirmed to inhibit IDO1 activity by > 30% using Ehrlich’s reagent reaction. Compounds 2, 7, and 8 were demonstrated to inhibit IDO1 activity in a cellular context. Compounds 2 and 7 were more potent against IDO1 than TDO2 in the enzymatic assay. The kinetic studies showed that compound 2 exhibited noncompetitive inhibition, whereas compounds 7 and 8 were graphically well matched with uncompetitive inhibition. Compounds 7 and 8 were found to bind to the ferric-IDO1 enzyme. Docking stimulations showed that the naphthalene ring of compound 8 formed “T-shaped” π–π interactions with Phe-163 and that the 6-methyl-naphthalene group formed additional hydrophobic interactions with IDO1. Compound 8 was identified as a derivative of tanshinone, and preliminary SAR analysis indicated that tanshinone derivatives may be promising hits for the development of IDO1 inhibitors. This study provides new clues for the discovery of IDO1/TDO2 inhibitors with novel scaffolds. Keywords: indoleamine 2,3-dioxygenase 1 inhibitor; natural product library; high-throughput fluorescence-based screening Acta Pharmacologica Sinica (2020) 41:423–431; https://doi.org/10.1038/s41401-019-0246-4 INTRODUCTION macrophages, myeloid-derived suppressor cells, and dendritic Indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygen- cells. In contrast, TDO2 is predominantly expressed in the liver, ase 2 (TDO2) are intracellular heme-containing metalloproteins where it has a key role in maintaining the systemic homeostasis of that initiate the first and the rate-limiting steps of tryptophan the tryptophan levels [3]. The expression of IDO1 is greatly breakdown along the kynurenine pathway [1]. IDO is ubiquitously elevated in multiple types of human cancer and is correlated with expressed in many types of cells and tissues, with the highest a poor prognosis [4–6]. Recently, IDO1 has been shown to play an expression occurring in antigen-presenting cells, such as macro- important role in the process of immune evasion by tumors [7]. phages and dendritic cells. Two homologs, IDO1 and IDO2, have The IDO-mediated depletion of local tryptophan levels and been found in the IDO family, and IDO1 is by far the better the production of toxic metabolites results in the suppression of characterized of the two, and it has a central role in mediating T-cell responses and the enhancement of immunosuppression immune privilege and preventing T-cell-driven rejection of mediated by regulatory T cells [8, 9]. allogeneic fetuses during pregnancy [2]. Although the sequence As such, IDO1 has emerged as a promising therapeutic target, similarity between human IDO1 and TDO2 is poor (16%), the high prompting searches for highly active inhibitors. The landmark similarity of their catalytic domains has been revealed by protein competitive inhibitor 1-methyl-L-tryptophan (1-L-MT) was identi- crystallography studies. Although TDO2 is specific for metaboliz- fied in the early 1990s and has a reported Kiof34μM. However, ing tryptophan to kynurenine, IDO1 recognizes a broad range of 1-L-MT is a rather poor IDO1 inhibitor when tested in IDO1- indole-containing substrates, including the neurotransmitter expressing human tumor cells, as 1-L-MT concentrations >200 μM melatonin. IDO1 is ubiquitously expressed in many tissues are required to block IDO1-mediated tryptophan degradation by and cells, including endothelial cells and cells that in part 50% [10]. The X-ray crystal structure of human IDO1 in a complex constitute the tumor microenvironment, such as fibroblasts, with an inhibitor (4-phenylimidazole or cyanide) was revealed in 1Division of Anti-tumor Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; 2University of Chinese Academy of Sciences, Beijing 100049, China; 3Department of Natural Products Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; 4State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China and 5School of Life Science and Technology, ShanghaiTech University, Shanghai 201203, China Correspondence: Yang Ye ([email protected]) or Ling-hua Meng ([email protected]) These authors contributed equally: Wei Guo, Sheng Yao Received: 25 January 2019 Accepted: 5 May 2019 Published online: 13 June 2019 © CPS and SIMM 2019 Discovery of natural products as novel IDO1 inhibitors W Guo et al. 424 2006 and facilitated the discovery of new IDO1 inhibitors [11]. kynurenine was measured at 492 nm using a SpectraMax Plus Recently, several potent IDO1 inhibitors have been identified by 384 microplate reader (Molecular Devices, Sunnyvale, CA). The unbiased screening [12, 13], structure-based modification [14, 15], data were analyzed using the enzyme kinetics module in and in silico drug design [15, 16]. However, only six compounds, SigmaPlot, version 10. 1-methyl-D-tryptophan (1-D-MT), hydroxylamidine INCB024360, The fluorescent enzymatic assay was carried out in 384-well NLG802, BMS-986205, SHR9146, LY3381916, and PF-06840003, are plates as described previously [19] with minor modifications. currently in clinical trials. Therefore, the number and structures The reaction was carried out in the presence of IDO1 holoenzyme, of IDO1 inhibitors in clinical trials are very limited, and new which consumed no more than ~30% of the initial L-Trp inhibitors with diverse structures and improved activities are of over 30 min and was terminated by the addition of piperidine great interest. (200 mM, final concentration). The plate was incubated at 65 °C for Herein, we screened an in-house natural product library for 15 min and left at room temperature for 60 min prior to novel inhibitors of IDO1. The screening led to the discovery that measurement with a SYNERGY H1 microplate reader (Bioteck, compounds 2, 7, and 8 showed inhibitory potencies in the Winooski, VT, USA). The inhibitory rate was calculated using the micromolar range. Detailed kinetic studies were performed and formula (ODcontrol−ODblank)−(ODtreatment−ODblank)/(ODcontrol− revealed that compound 2 exhibited noncompetitive inhibition, ODblank) × 100%. The IC50 values were determined by nonlinear whereas compounds 7 and 8 showed uncompetitive inhibition. regression analysis with Prism 4 software (GraphPad Software Inc., Compounds 7 and 8 were shown to interact with the ferric form of San Diego, CA, USA). The Z’ factor of the assay was calculated IDO1. Preliminary SARs were drawn from the analogs of using the formula [(ODmean control−3SDcontrol)−(ODmean blank + compound 8 and corroborated the putative binding orientation 3SDblank)]/(ODmean control−ODmean blank). suggested by molecular docking. Cell-based assay of IDO1 and TDO2 activities HEK 293 cells were seeded in a six-well plate at a density of 5 × 105 MATERIALS AND METHODS cells/well. The next day, the HEK 293 cells were transfected with Materials pcDNA3.1-hIDO or pcDNA3.1-hTDO using Lipofectamine 2000 Methylene Blue hydrate (#28514), L-tryptophan (#T0254), DMSO according to the manufacturer’s instructions. The cells were (#D2650), and p-dimethylaminobenzaldehyde (#D2004) were seeded in 96-well plates at a density of 2.5 × 104 cells/well 24 h purchased from Sigma (St. Louis, MO, USA). Catalase, bovine after transfection and treated with tested compounds. After an 1234567890();,: hemin, and ascorbic acid were purchased from Sangon (Shanghai, additional 12 h of incubation, 200 μL of the culture medium from China). Piperidine and TCA were obtained from SCRC (Shanghai, each well was transferred to a 96-well plate and mixed with 100 μL China). INCB024360 and Selleck Customized Library–Z100688 of 30% trichloroacetic acid. The plate was incubated at 65 °C for were purchased from Selleck Chemicals (Houston, TX, USA). The 15 min to hydrolyze the N-formylkynurenine produced by the natural product library was constructed by the Department of catalytic reaction of IDO1 or TDO2. The reaction mixture was then Natural Products Chemistry, Shanghai Institute of Materia Medica, centrifuged at 12 000 r/min for 10 min. Then, 100 μL of the Chinese Academy of Sciences. Cell Counting Kit-8(#CK04) was pur- supernatant was transferred to another 96-well plate and mixed chased from Dojindo (Kumamoto, Japan). Recombinant IDO1 and with 100 μL of 2% (w/v) p-dimethylaminobenzaldehyde in acetic TDO2 were expressed and purified in our laboratory as described acid. The yellow color derived from kynurenine was measured at previously [17]. The plasmids pET28a-hIDO1, pET28a-hTDO2, 492 nm using a SpectraMax Plus 384 microplate reader (Molecular pcDNA3.1-hIDO1, and pcDNA3.1-TDO2 were synthesized by Devices, Sunnyvale, CA, USA). Synbio Technologies (Suzhou, China). Determination of the inhibition kinetics of IDO1 inhibitors Cell culture The inhibition kinetics were determined as described previously HEK 293 cell line was obtained from ATCC (Manassas, VA, USA). [20]. In brief, the enzymatic activity of IDO1 was determined with a Cells were grown in Dulbecco’s modified Eagle’s medium serial dilution of the test compounds with L-tryptophan at (Corning, New York, USA) supplemented with 10% fetal bovine concentrations ranging from 7.5 μMto30μM. A function of S/V serum (Gibco, New York, USA) in 5% CO2 at 37 °C.
Recommended publications
  • Bioinorganic Chemistry of Nickel
    inorganics Editorial Bioinorganic Chemistry of Nickel Michael J. Maroney 1,* and Stefano Ciurli 2,* 1 Department of Chemistry and Program in Molecular and Cellular Biology, University of Massachusetts Amherst, 240 Thatcher Rd. Life Sciences, Laboratory Rm N373, Amherst, MA 01003, USA 2 Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology, University of Bologna, Viale G. Fanin 40, I-40127 Bologna, Italy * Correspondence: [email protected] (M.J.M.); [email protected] (S.C.) Received: 11 October 2019; Accepted: 11 October 2019; Published: 30 October 2019 Following the discovery of the first specific and essential role of nickel in biology in 1975 (the dinuclear active site of the enzyme urease) [1], nickel has become a major player in bioinorganic chemistry,particularly in microorganisms, having impacts on both environmental settings and human pathologies. At least nine classes of enzymes are now known to require nickel in their active sites, including catalysis of redox [(Ni,Fe) hydrogenases, carbon monoxide dehydrogenase, methyl coenzyme M reductase, acetyl coenzyme A synthase, superoxide dismutase] and nonredox (glyoxalase I, acireductone dioxygenase, lactate isomerase, urease) chemistries. In addition, the dark side of nickel has been illuminated in regard to its participation in microbial pathogenesis, cancer, and immune responses. Knowledge gleaned from the investigations of inorganic chemists into the coordination and redox chemistry of this element have boosted the understanding of these biological roles of nickel in each context. In this issue, eleven contributions, including four original research articles and seven critical reviews, will update the reader on the broad spectrum of the role of nickel in biology.
    [Show full text]
  • Cysteine Dioxygenase 1 Is a Metabolic Liability for Non-Small Cell Lung Cancer Authors: Yun Pyo Kang1, Laura Torrente1, Min Liu2, John M
    bioRxiv preprint doi: https://doi.org/10.1101/459602; this version posted November 1, 2018. 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. Cysteine dioxygenase 1 is a metabolic liability for non-small cell lung cancer Authors: Yun Pyo Kang1, Laura Torrente1, Min Liu2, John M. Asara3,4, Christian C. Dibble5,6 and Gina M. DeNicola1,* Affiliations: 1 Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA 2 Proteomics and Metabolomics Core Facility, Moffitt Cancer Center and Research Institute, Tampa, FL, USA 3 Division of Signal Transduction, Beth Israel Deaconess Medical Center, Boston, MA, USA 4 Department of Medicine, Harvard Medical School, Boston, MA, USA 5 Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Boston, MA, USA 6 Department of Pathology, Harvard Medical School, Boston, MA, USA *Correspondence to: [email protected]. Keywords: KEAP1, NRF2, cysteine, CDO1, sulfite Summary NRF2 is emerging as a major regulator of cellular metabolism. However, most studies have been performed in cancer cells, where co-occurring mutations and tumor selective pressures complicate the influence of NRF2 on metabolism. Here we use genetically engineered, non-transformed primary cells to isolate the most immediate effects of NRF2 on cellular metabolism. We find that NRF2 promotes the accumulation of intracellular cysteine and engages the cysteine homeostatic control mechanism mediated by cysteine dioxygenase 1 (CDO1), which catalyzes the irreversible metabolism of cysteine to cysteine sulfinic acid (CSA). Notably, CDO1 is preferentially silenced by promoter methylation in non-small cell lung cancers (NSCLC) harboring mutations in KEAP1, the negative regulator of NRF2.
    [Show full text]
  • Indoleamine 2,3-Dioxygenase and Its Therapeutic Inhibition in Cancer George C
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Scholarship, Research, and Creative Work at Bryn Mawr College | Bryn Mawr College... Bryn Mawr College Scholarship, Research, and Creative Work at Bryn Mawr College Chemistry Faculty Research and Scholarship Chemistry 2018 Indoleamine 2,3-Dioxygenase and Its Therapeutic Inhibition in Cancer George C. Prendergast William Paul Malachowski Bryn Mawr College, [email protected] Arpita Mondal Peggy Scherle Alexander J. Muller Let us know how access to this document benefits ouy . Follow this and additional works at: https://repository.brynmawr.edu/chem_pubs Part of the Chemistry Commons Custom Citation George C. Prendergast, William J. Malachowski, Arpita Mondal, Peggy Scherle, and Alexander J. Muller. 2018. "Indoleamine 2,3-Dioxygenase and Its Therapeutic Inhibition in Cancer." International Review of Cell and Molecular Biology 336: 175-203. This paper is posted at Scholarship, Research, and Creative Work at Bryn Mawr College. https://repository.brynmawr.edu/chem_pubs/25 For more information, please contact [email protected]. Indoleamine 2,3-Dioxygenase and Its Therapeutic Inhibition in Cancer George C. Prendergast, William, Malachowski, Arpita Mondal, Peggy Scherle, and Alexander J. Muller International Review of Cell and Molecular Biology 336: 175-203. http://doi.org/10.1016/bs.ircmb.2017.07.004 ABSTRACT The tryptophan catabolic enzyme indoleamine 2,3-dioxygenase-1 (IDO1) has attracted enormous attention in driving cancer immunosuppression, neovascularization, and metastasis. IDO1 suppresses local CD8+ T effector cells and natural killer cells and induces CD4+ T regulatory cells (iTreg) and myeloid-derived suppressor cells (MDSC). The structurally distinct enzyme tryptophan dioxygenase (TDO) also has been implicated recently in immune escape and metastatic progression.
    [Show full text]
  • Muscle Regeneration Controlled by a Designated DNA Dioxygenase
    Wang et al. Cell Death and Disease (2021) 12:535 https://doi.org/10.1038/s41419-021-03817-2 Cell Death & Disease ARTICLE Open Access Muscle regeneration controlled by a designated DNA dioxygenase Hongye Wang1, Yile Huang2,MingYu3,YangYu1, Sheng Li4, Huating Wang2,5,HaoSun2,5,BingLi 3, Guoliang Xu6,7 andPingHu4,8,9 Abstract Tet dioxygenases are responsible for the active DNA demethylation. The functions of Tet proteins in muscle regeneration have not been well characterized. Here we find that Tet2, but not Tet1 and Tet3, is specifically required for muscle regeneration in vivo. Loss of Tet2 leads to severe muscle regeneration defects. Further analysis indicates that Tet2 regulates myoblast differentiation and fusion. Tet2 activates transcription of the key differentiation modulator Myogenin (MyoG) by actively demethylating its enhancer region. Re-expressing of MyoG in Tet2 KO myoblasts rescues the differentiation and fusion defects. Further mechanistic analysis reveals that Tet2 enhances MyoD binding by demethylating the flanking CpG sites of E boxes to facilitate the recruitment of active histone modifications and increase chromatin accessibility and activate its transcription. These findings shed new lights on DNA methylation and pioneer transcription factor activity regulation. Introduction Ten-Eleven Translocation (Tet) family of DNA dioxy- 1234567890():,; 1234567890():,; 1234567890():,; 1234567890():,; Skeletal muscles can regenerate due to the existence of genases catalyze the active DNA demethylation and play muscle stem cells (MuSCs)1,2. The normally quiescent critical roles in embryonic development, neural regen- MuSCs are activated after muscle injury and further dif- eration, oncogenesis, aging, and many other important – ferentiate to support muscle regeneration3,4.
    [Show full text]
  • Relating Metatranscriptomic Profiles to the Micropollutant
    1 Relating Metatranscriptomic Profiles to the 2 Micropollutant Biotransformation Potential of 3 Complex Microbial Communities 4 5 Supporting Information 6 7 Stefan Achermann,1,2 Cresten B. Mansfeldt,1 Marcel Müller,1,3 David R. Johnson,1 Kathrin 8 Fenner*,1,2,4 9 1Eawag, Swiss Federal Institute of Aquatic Science and Technology, 8600 Dübendorf, 10 Switzerland. 2Institute of Biogeochemistry and Pollutant Dynamics, ETH Zürich, 8092 11 Zürich, Switzerland. 3Institute of Atmospheric and Climate Science, ETH Zürich, 8092 12 Zürich, Switzerland. 4Department of Chemistry, University of Zürich, 8057 Zürich, 13 Switzerland. 14 *Corresponding author (email: [email protected] ) 15 S.A and C.B.M contributed equally to this work. 16 17 18 19 20 21 This supporting information (SI) is organized in 4 sections (S1-S4) with a total of 10 pages and 22 comprises 7 figures (Figure S1-S7) and 4 tables (Table S1-S4). 23 24 25 S1 26 S1 Data normalization 27 28 29 30 Figure S1. Relative fractions of gene transcripts originating from eukaryotes and bacteria. 31 32 33 Table S1. Relative standard deviation (RSD) for commonly used reference genes across all 34 samples (n=12). EC number mean fraction bacteria (%) RSD (%) RSD bacteria (%) RSD eukaryotes (%) 2.7.7.6 (RNAP) 80 16 6 nda 5.99.1.2 (DNA topoisomerase) 90 11 9 nda 5.99.1.3 (DNA gyrase) 92 16 10 nda 1.2.1.12 (GAPDH) 37 39 6 32 35 and indicates not determined. 36 37 38 39 S2 40 S2 Nitrile hydration 41 42 43 44 Figure S2: Pearson correlation coefficients r for rate constants of bromoxynil and acetamiprid with 45 gene transcripts of ECs describing nucleophilic reactions of water with nitriles.
    [Show full text]
  • ALOX12 Antibody
    Efficient Professional Protein and Antibody Platforms ALOX12 Antibody Basic information: Catalog No.: UPA60470 Source: Rabbit Size: 50ul/100ul Clonality: polyclonal Concentration: 1mg/ml Isotype: Rabbit IgG Purification: affinity purified by Protein A Useful Information: Applications: WB:1:500-2000 Reactivity: Human, Mouse, Rat, Cow Specificity: This antibody recognizes ALOX12 protein. KLH conjugated synthetic peptide derived from human 12 Lipoxygenase Immunogen: 101-200/663 Non-heme iron-containing dioxygenase that catalyzes the stereo-specific peroxidation of free and esterified polyunsaturated fatty acids generating a spectrum of bioactive lipid mediators. Mainly converts arachidonic acid to (12S)-hydroperoxyeicosatetraenoic acid/(12S)-HPETE but can also metabo- lize linoleic acid. Has a dual activity since it also converts leukotriene A4/LTA4 into both the bioactive lipoxin A4/LXA4 and lipoxin B4/LXB4. Description: Through the production of specific bioactive lipids like (12S)-HPETE it regu- lates different biological processes including platelet activation. It also probably positively regulates angiogenesis through regulation of the expres- sion of the vascular endothelial growth factor. Plays a role in apoptotic pro- cess, promoting the survival of vascular smooth muscle cells for instance. May also play a role in the control of cell migration and proliferation. {ECO:0000269|PubMed:16638750, Uniprot: P18054 Human BiowMW: 76 KDa Buffer: 0.01M TBS(pH7.4) with 1% BSA, 0.03% Proclin300 and 50% Glycerol. Storage: Store at 4°C short term and -20°C long term. Avoid freeze-thaw cycles. Note: For research use only, not for use in diagnostic procedure. Data: Gene Universal Technology Co. Ltd www.universalbiol.com Tel: 0550-3121009 E-mail: [email protected] Efficient Professional Protein and Antibody Platforms Sample: Raw264.7 Cell Lysate at 40 ug A431 Cell Lysate at 40 ug Primary: Anti-ALOX12 at 1/300 dilution Secondary: IRDye800CW Goat An- ti-Rabbit IgG at 1/20000 dilution Predicted band size: 73 kD Observed band size: 73 kD Gene Universal Technology Co.
    [Show full text]
  • Mechanistic Study of Cysteine Dioxygenase, a Non-Heme
    MECHANISTIC STUDY OF CYSTEINE DIOXYGENASE, A NON-HEME MONONUCLEAR IRON ENZYME by WEI LI Presented to the Faculty of the Graduate School of The University of Texas at Arlington in Partial Fulfillment of the Requirements for the Degree of DOCTOR OF PHILOSOPHY THE UNIVERSITY OF TEXAS AT ARLINGTON August 2014 Copyright © by Student Name Wei Li All Rights Reserved Acknowledgements I would like to thank Dr. Pierce for your mentoring, guidance and patience over the five years. I cannot go all the way through this without your help. Your intelligence and determination has been and will always be an example for me. I would like to thank my committee members Dr. Dias, Dr. Heo and Dr. Jonhson- Winters for the directions and invaluable advice. I also would like to thank all my lab mates, Josh, Bishnu ,Andra, Priyanka, Eleanor, you all helped me so I could finish my projects. I would like to thank the Department of Chemistry and Biochemistry for the help with my academic and career. At Last, I would like to thank my lovely wife and beautiful daughter who made my life meaningful and full of joy. July 11, 2014 iii Abstract MECHANISTIC STUDY OF CYSTEINE DIOXYGENASE A NON-HEME MONONUCLEAR IRON ENZYME Wei Li, PhD The University of Texas at Arlington, 2014 Supervising Professor: Brad Pierce Cysteine dioxygenase (CDO) is an non-heme mononuclear iron enzymes that catalyzes the O2-dependent oxidation of L-cysteine (Cys) to produce cysteine sulfinic acid (CSA). CDO controls cysteine levels in cells and is a potential drug target for some diseases such as Parkinson’s and Alzhermer’s.
    [Show full text]
  • Unique Coupling of Mono- and Dioxygenase Chemistries in a Single Active Site Promotes Heme Degradation
    Unique coupling of mono- and dioxygenase chemistries in a single active site promotes heme degradation Toshitaka Matsuia,1, Shusuke Nambua, Celia W. Gouldingb,c, Satoshi Takahashia, Hiroshi Fujiid, and Masao Ikeda-Saitoa,1 aInstitute of Multidisciplinary Research for Advanced Materials, Tohoku University, Katahira, Aoba, Sendai 980-8577, Japan; bDepartment of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697; cDepartment of Pharmaceutical Sciences, University of California, Irvine, CA 92697; and dDepartment of Chemistry, Graduate School of Humanities and Sciences, Nara Women’s University, Kitauoyanishi, Nara 630-8506, Japan Edited by Harry B. Gray, California Institute of Technology, Pasadena, CA, and approved February 26, 2016 (received for review November 26, 2015) − + Bacterial pathogens must acquire host iron for survival and colo- RH + O2 + 2e + 2H → ROH + H2O, [1] nization. Because free iron is restricted in the host, numerous pathogens have evolved to overcome this limitation by using a R + O → RðOÞ . [2] family of monooxygenases that mediate the oxidative cleavage of 2 2 heme into biliverdin, carbon monoxide, and iron. However, the Biological heme degradation proceeds through a unique self- etiological agent of tuberculosis, Mycobacterium tuberculosis, ac- oxidation mechanism where the substrate heme activates O complishes this task without generating carbon monoxide, which 2 potentially induces its latent state. Here we show that this unusual molecules. The canonical enzyme, heme oxygenase (HO), de- heme degradation reaction proceeds through sequential mono- grades heme into ferrous iron, carbon monoxide (CO), and bil- and dioxygenation events within the single active center of MhuD, iverdin by three successive monooxygenation reactions (Fig. 1A) a mechanism unparalleled in enzyme catalysis.
    [Show full text]
  • Original Article Genetic Variants of Aloxs Genes in Polyunsaturated Fatty Acid/Arachidonic Acid Metabolism Associated with Type-2 Diabetes Development
    Int J Clin Exp Med 2018;11(12):13797-13805 www.ijcem.com /ISSN:1940-5901/IJCEM0077987 Original Article Genetic variants of ALOXs genes in polyunsaturated fatty acid/arachidonic acid metabolism associated with type-2 diabetes development Jim Jinn-Chyuan Sheu1,3,4,5*, Ying-Ju Lin1,3*, Cherry Yin-Yi Chang2, Shih-Yin Chen1,3, Wen-Ling Liao1,4, Jai-Sing Yang1, Ming-Tsung Lai6, Chih-Mei Chen1, Chun-Cheng Tseng4, Tritium Hwang4, Ping-Ho Chen7, Fuu-Jen Tsai1,3 1Human Genetic Center, 2Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan; 3School of Chinese Medicine, China Medical University, Taichung, Taiwan; 4Institute of Biomedical Sci- ences, National Sun Yat-sen University, Kaohsiung, Taiwan; 5Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan; 6Department of Pathology, Taichung Hospital, Ministry of Health and Welfare, Taichung, Taiwan; 7School of Dentistry, Kaohsiung Medical University, Kaohsiung, Taiwan. *Equal contributors. Received April 16, 2018; Accepted July 24, 2018; Epub December 15, 2018; Published December 30, 2018 Abstract: Poly-unsaturated fatty acids (PUFAs)/arachidonic acids (AAs) and their derived eicosanoids play potent roles in triggering inflammation during obesity and diabetes development. Recent studies have indicated functional roles of ALOX5, ALOX12, ALOX12B, and ALOX15 in the development of insulin resistance and islet β-cell dysfunc- tion. However, the impact of their genetic variants on type 2 diabetes (T2D) development in Asian patients remains unclear. In this study, 1,682 healthy controls and 788 patients with T2D were enrolled for genotyping those four ALOX genes by the TaqMan method. A total of eight Han Chinese-specific SNPs (two SNps for each gene) were selected for this study.
    [Show full text]
  • Structural Enzymology of Sulfide Oxidation by Persulfide Dioxygenase and Rhodanese
    Structural Enzymology of Sulfide Oxidation by Persulfide Dioxygenase and Rhodanese by Nicole A. Motl A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy (Biological Chemistry) in the University of Michigan 2017 Doctoral Committee Professor Ruma Banerjee, Chair Assistant Professor Uhn-Soo Cho Professor Nicolai Lehnert Professor Stephen W. Ragsdale Professor Janet L. Smith Nicole A. Motl [email protected] ORCID iD: 0000-0001-6009-2988 © Nicole A. Motl 2017 ACKNOWLEDGEMENTS I would like to take this opportunity to acknowledge the many people who have provided me with guidance and support during my doctoral studies. First I would like to express my appreciation and gratitude to my advisor Dr. Ruma Banerjee for the mentorship, guidance, support and encouragement she has provided. I would like to thank my committee members Dr. Uhn-Soo Cho, Dr. Nicolai Lehnert, Dr. Stephen Ragsdale and Dr. Janet Smith for their advice, assistance and support. I would like to thank Dr. Janet Smith and members of Dr. Smith’s lab, especially Meredith Skiba, for sharing their expertise in crystallography. I would like to thank Dr. Omer Kabil for his help, suggestions and discussions in various aspects of my study. I would also like to thank members of Dr. Banerjee’s lab for their suggestions and discussions. Additionally, I would like to thank my friends and family for their support. ii TABLE OF CONTENTS ACKNOWLEDGEMENTS ii LIST OF TABLES viii LIST OF FIGURES ix ABBREVIATIONS xi ABSTRACT xii CHAPTER I. Introduction:
    [Show full text]
  • Functional Characterization of Genetic Enzyme Variations in Human Lipoxygenases
    Redox Biology 1 (2013) 566–577 Contents lists available at ScienceDirect Redox Biology journal homepage: www.elsevier.com/locate/redox Functional characterization of genetic enzyme variations in human lipoxygenases Thomas Horn a,n, Kumar Reddy Kakularam b, Monika Anton a, Constanze Richter c, Pallu Reddanna b,d, Hartmut Kuhn a a Institute of Biochemistry, University Medicine Berlin—Charité, Charitéplatz 1, D-10117 Berlin, Germany b Department of Animal Sciences, School of Life Sciences, University of Hyderabad, Gachibowli, Hyderabad 500046, Andhra Pradesh, India c Institute of Nutrition Technology and Nutrition Chemistry, TU Berlin, Gustav-Meyer-Allee 25, D-13355 Berlin, Germany d National Institute of Animal Biotechnology, Hyderabad 500046, Andhra Pradesh, India article info abstract Article history: Mammalian lipoxygenases play a role in normal cell development and differentiation but they have also been Received 28 October 2013 implicated in the pathogenesis of cardiovascular, hyperproliferative and neurodegenerative diseases. As lipid Accepted 1 November 2013 peroxidizing enzymes they are involved in the regulation of cellular redox homeostasis since they produce lipid hydroperoxides, which serve as an efficient source for free radicals. There are various epidemiological Keywords: correlation studies relating naturally occurring variationsinthesixhumanlipoxygenasegenes(SNPsorrare Eicosanoids mutations) to the frequency for various diseases in these individuals, but for most of the described variations no Leukotrienes functional data are available. Employing a combined bioinformatical and enzymological strategy, which Lipoxygenases included structural modeling and experimental site-directed mutagenesis, we systematically explored the Gene polymorphism structural and functional consequences of non-synonymous genetic variations in four different human SNP lipoxygenase genes (ALOX5, ALOX12, ALOX15, and ALOX15B) that have been identified in the human 1000 genome project.
    [Show full text]
  • Positive Allosteric Modulation of Indoleamine 2,3-Dioxygenase 1
    Positive allosteric modulation of indoleamine 2,3- dioxygenase 1 restrains neuroinflammation Giada Mondanellia, Alice Colettib, Francesco Antonio Grecob, Maria Teresa Pallottaa, Ciriana Orabonaa, Alberta Iaconoa, Maria Laura Belladonnaa, Elisa Albinia,b, Eleonora Panfilia, Francesca Fallarinoa, Marco Gargaroa, Giorgia Mannia, Davide Matinoa, Agostinho Carvalhoc,d, Cristina Cunhac,d, Patricia Macielc,d, Massimiliano Di Filippoe, Lorenzo Gaetanie, Roberta Bianchia, Carmine Vaccaa, Ioana Maria Iamandiia, Elisa Proiettia, Francesca Bosciaf, Lucio Annunziatof,1, Maikel Peppelenboschg, Paolo Puccettia, Paolo Calabresie,2, Antonio Macchiarulob,3, Laura Santambrogioh,i,j,3, Claudia Volpia,3,4, and Ursula Grohmanna,k,3,4 aDepartment of Experimental Medicine, University of Perugia, 06100 Perugia, Italy; bDepartment of Pharmaceutical Sciences, University of Perugia, 06100 Perugia, Italy; cLife and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; dICVS/3B’s–PT Government Associate Laboratory, 4704-553 Braga/Guimarães, Portugal; eDepartment of Medicine, University of Perugia, 06100 Perugia, Italy; fDepartment of Neuroscience, University of Naples Federico II, 80131 Naples, Italy; gDepartment of Gastroenterology and Hepatology, Erasmus Medical Centre–University Medical Centre Rotterdam, 3015 GD Rotterdam, The Netherlands; hEnglander Institute of Precision Medicine, Weill Cornell Medicine, New York, NY 10065; iDepartment of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065; jDepartment of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065; and kDepartment of Pathology, Albert Einstein College of Medicine, New York, NY 10461 Edited by Lawrence Steinman, Stanford University School of Medicine, Stanford, CA, and approved January 16, 2020 (received for review October 21, 2019) L-tryptophan (Trp), an essential amino acid for mammals, is the or necessary to contain the pathology (20).
    [Show full text]