FMO3) As a Novel Genetic Determinant of Acetaminophen (APAP) Induced Hepatotoxicity Swetha Rudraiah University of Connecticut - Storrs, [email protected]

Total Page:16

File Type:pdf, Size:1020Kb

FMO3) As a Novel Genetic Determinant of Acetaminophen (APAP) Induced Hepatotoxicity Swetha Rudraiah University of Connecticut - Storrs, Swetha.Rudraiah@Uconn.Edu University of Connecticut OpenCommons@UConn Doctoral Dissertations University of Connecticut Graduate School 8-4-2014 Characterization of Flavin-containing Monooxygenase-3 (FMO3) as a Novel Genetic Determinant of Acetaminophen (APAP) Induced Hepatotoxicity Swetha Rudraiah University of Connecticut - Storrs, [email protected] Follow this and additional works at: https://opencommons.uconn.edu/dissertations Recommended Citation Rudraiah, Swetha, "Characterization of Flavin-containing Monooxygenase-3 (FMO3) as a Novel Genetic Determinant of Acetaminophen (APAP) Induced Hepatotoxicity" (2014). Doctoral Dissertations. 506. https://opencommons.uconn.edu/dissertations/506 Characterization of Flavin-containing Monooxygenase-3 (FMO3) as a Novel Genetic Determinant of Acetaminophen (APAP) Induced Hepatotoxicity Swetha Rudraiah, Ph.D. University of Connecticut, 2014 Mice pretreated with a mild toxic dose of acetaminophen (APAP) acquire resistance to a second, higher APAP dose. This phenomenon is termed APAP autoprotection and the exact mechanism by which such resistance develops is not clearly known. Given the prevalence of APAP- hepatotoxicity and the human health impact of this potentially hepatotoxic agent, a further understanding of the mechanism(s) involved in such protection are of considerable significance and could lead to new modalities of treatment of acute drug-induced liver injury. The work presented in this thesis investigates FMO3 gene expression during APAP-induced liver injury as well as the functional significance of FMO3 over-expression during APAP- induced liver injury. Furthermore, FMO3 gene regulation during oxidative stress conditions is also examined. Acetaminophen treatment resulted in up-regulation of liver Fmo3 protein in male mice. Female mice express higher liver Fmo3 than males and are highly resistant to APAP hepatotoxicity. Inhibition of Fmo3, by methimazole, renders female mice susceptible to APAP-induced liver injury. These findings are suggestive of a protective function for Fmo3. In addition to Swetha Rudraiah – University of Connecticut, 2014 APAP, ANIT and BDL also increase Fmo3 gene expression in mice. Because these hepatotoxicants also induce oxidative stress, we also investigated the potential role of the oxidative stress sensor and transcription factor, NRF2, in FMO3 gene regulation. Both in vivo and in vitro results show that transcriptional regulation of FMO3 might not involve the NRF2-KEAP1 regulatory pathway. Human liver can adapt to APAP-induced hepatotoxicity similar to our APAP autoprotection mouse model. The last part of this dissertation examined FMO3 gene induction in a human hepatoma cell line, HepaRG. APAP induced FMO3 gene expression in HepaRG cells, and over-expression of FMO3 protects cells against APAP-induced cytotoxicity. The unexpected observation of a faster differentiation phenotype in HepaRG cells over- expressing FMO3 suggests that FMO3 may play an important role in cellular differentiation. Collectively, data presented in this thesis provide evidence for Fmo3 as a novel genetic determinant of APAP-induced liver injury. Furthermore, this thesis describes a novel protective function for FMO3. Findings from HepaRG cells suggest that FMO3 over-expression in response to APAP may be a driving force for differentiation in regenerating hepatocytes. Characterization of Flavin-containing Monooxygenase-3 (FMO3) as a Novel Genetic Determinant of Acetaminophen (APAP) Induced Hepatotoxicity Swetha Rudraiah B.V.Sc. & A.H., University of Agricultural Sciences, India, 2004 M.V.Sc., Karnataka Veterinary, Animal and Fisheries Sciences University, India, 2006 A Dissertation Submitted in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy at the University of Connecticut 2014 i COPYRIGHT BY Swetha Rudraiah 2014 ii APPROVAL PAGE Doctor of Philosophy Dissertation Characterization of Flavin-containing Monooxygenase-3 (FMO3) as a Novel Genetic Determinant of Acetaminophen (APAP) Induced Hepatotoxicity Presented by Swetha Rudraiah Major Advisor: ___________________________________________ José E. Manautou, Ph.D. Associate Advisor: ________________________________________ Ronald N. Hines, Ph.D. Associate Advisor: ________________________________________ Urs A. Boelsterli, Ph.D. Associate Advisor: ________________________________________ Richard S. Bruno, Ph.D. University of Connecticut 2014 iii DEDICATION To everyone who taught me, guided me, and inspired me. iv ACKNOWLEDGEMENTS I would first and foremost like to thank my major advisor, Dr. José Manautou. Dr. Manautou has been an excellent mentor and has provided me invaluable guidance, continued advice and critical discussions that have allowed me to grow both professionally and personally. I would also like to thank my committee members Drs. Ronald Hines, Urs Boelsterli and Richard Bruno for their scientific inputs during the completion of my graduate degree. My special thanks to Dr. Hines for his willingness to help and answer my questions even before being a part of my thesis committee. Thank you for your guidance throughout. Many thanks to other faculty at the University of Connecticut School of Pharmacy for their assistance and advise during this research. Thanks to our collaborators Drs. Angela Slitt, Lauren Aleksunes, and Sarah Campion for providing samples and for insightful scientific discussions. My special thanks to Dr. Carolina Ghanem for the inspiration and friendship. I am very grateful for the support and friendship of the other members of our lab, both past and present, Xinsheng Gu, Amy Bataille, Ron Scialis, Dan Ferreira, Phil Rohrer, and Meeghan O’Connor. Also, thanks to fellow graduate students and postdocs including Bindu, Igor, Supriya, Joe, Greg, Kyle, Vince, Janine, Ritvik, Chad, Stephanie, Lai and Erica for making my stay here at UCONN memorable. I am grateful for the assistance provided by staff at the Department of Pharmaceutical Sciences especially Mark Armati and Leslie v Lebel, Denise Long at the histology laboratory, and all the hardworking staff in the animal care facility. I would also like to acknowledge the financial support of the National Institutes of Health Grant (DK069557) and the UCONN School of Pharmacy Department of Pharmaceutical Sciences without which none of this would have been possible. Thanks to the Society of Toxicology Mechanisms Specialty Section, for recognizing this work through Carl C. Smith Graduate Student Award. Lastly, I would like to extend my sincere thanks to my parents for instilling in me the values of hard work, perseverance, and good education. Their unconditional love, sacrifices, continued support, encouragement and constant pride in my accomplishments have led me so far. Thanks to my sister Shylaja and brother Omprakash, for always being there for me and for believing in me. Thanks to my extended family for all the motivation. To my husband, Sangamesh, thanks for the daily support and love and for this I will always be grateful. Finally, to my daughter, Gauri, your smile and cuteness have immensely helped me cheer, especially on some of those gloomy days. I am very fortunate to have you all in my life. vi TABLE OF CONTENTS APPROVAL PAGE iii DEDICATION iv ACKNOWLEDGEMENTS v TABLE OF CONTENTS vii LIST OF FIGURES ix LIST OF TABLES xii LIST OF ABBREVIATIONS xiii CHAPTER 1. Review of Literature 1 1.1 Liver Function and Structure 1 1.2 Hepatic Metabolizing Systems 4 1.3 APAP Hepatotoxicity 6 1.4 Liver Injury Models and Mechanisms of Hepatotoxicity 12 1.5 Nrf2 Signaling Pathway 17 1.6 APAP Autoprotection 24 1.7 Flavin-containing Monooxygenase-3 27 1.8 Summary 40 CHAPTER 2. Tolerance to Acetaminophen Hepatotoxicity in the Mouse Model of Autoprotection is Associated With Induction of Flavin-containing Monooxygenase-3 (FMO3) in Hepatocytes 48 2.1 Abstract 48 2.2 Introduction 50 2.3 Materials and Methods 55 2.4 Results 63 2.5 Discussion 98 CHAPTER 3. Differential FMO3 Gene Expression in Various Liver Injury Models Involving Hepatic Oxidative Stress in Mice 107 3.1 Abstract 107 3.2 Introduction 109 3.3 Materials and Methods 114 3.4 Results 118 3.5 Discussion 138 vii CHAPTER 4. Establishment and Characterization of HepaRG Cell Line Overexpressing Human Flavin-containing Monooxygenase-3 (FMO3) 145 4.1 Abstract 145 4.2 Introduction 147 4.3 Materials and Methods 150 4.4 Results 156 4.5 Discussion 186 CHAPTER 5. Human Hepatic Flavin-containing Monooxygenase-3 (FMO3) Gene Expression is not Regulated by Nuclear Factor Erythroid 2-Like 2 (NRF2/NFE2L2) Under Oxidative Stress Conditions 193 5.1 Abstract 193 5.2 Introduction 195 5.3 Materials and Methods 196 5.4 Results 202 5.5 Discussion 212 CHAPTER 6. Summary 215 REFERENCES 223 viii LIST OF FIGURES Page Figure 1.1 Scheme showing structural organization of the liver lobule 42 Figure 1.2 Schematic drawing of various domains of Nrf (Nrf1, Nrf2, Nrf3) and Keap1/INrf2 43 Figure 1.3 A schematic representation of Nrf2-mediated gene transcription 44 Figure 1.4 A schematic representation of catalytic cycle of FMO 45 Figure 2.1 Plasma ALT activity and quantitative RT-PCR analysis of liver Fmo3 transcripts following a single dose APAP treatment 74 Figure 2.2 Analysis of liver Fmo3 protein expression following a single dose APAP treatment by western blotting, enzyme activity assay as well as by immunohistochemistry 76 Figure 2.3 Plasma ALT activity in the mouse model of APAP autoprotection 78 Figure 2.4 Analysis of liver Fmo3 protein expression in the mouse model of APAP autoprotection by western blotting and enzyme activity
Recommended publications
  • Table 2. Functional Classification of Genes Differentially Regulated After HOXB4 Inactivation in HSC/Hpcs
    Table 2. Functional classification of genes differentially regulated after HOXB4 inactivation in HSC/HPCs Symbol Gene description Fold-change (mean ± SD) Signal transduction Adam8 A disintegrin and metalloprotease domain 8 1.91 ± 0.51 Arl4 ADP-ribosylation factor-like 4 - 1.80 ± 0.40 Dusp6 Dual specificity phosphatase 6 (Mkp3) - 2.30 ± 0.46 Ksr1 Kinase suppressor of ras 1 1.92 ± 0.42 Lyst Lysosomal trafficking regulator 1.89 ± 0.34 Mapk1ip1 Mitogen activated protein kinase 1 interacting protein 1 1.84 ± 0.22 Narf* Nuclear prelamin A recognition factor 2.12 ± 0.04 Plekha2 Pleckstrin homology domain-containing. family A. (phosphoinosite 2.15 ± 0.22 binding specific) member 2 Ptp4a2 Protein tyrosine phosphatase 4a2 - 2.04 ± 0.94 Rasa2* RAS p21 activator protein 2 - 2.80 ± 0.13 Rassf4 RAS association (RalGDS/AF-6) domain family 4 3.44 ± 2.56 Rgs18 Regulator of G-protein signaling - 1.93 ± 0.57 Rrad Ras-related associated with diabetes 1.81 ± 0.73 Sh3kbp1 SH3 domain kinase bindings protein 1 - 2.19 ± 0.53 Senp2 SUMO/sentrin specific protease 2 - 1.97 ± 0.49 Socs2 Suppressor of cytokine signaling 2 - 2.82 ± 0.85 Socs5 Suppressor of cytokine signaling 5 2.13 ± 0.08 Socs6 Suppressor of cytokine signaling 6 - 2.18 ± 0.38 Spry1 Sprouty 1 - 2.69 ± 0.19 Sos1 Son of sevenless homolog 1 (Drosophila) 2.16 ± 0.71 Ywhag 3-monooxygenase/tryptophan 5- monooxygenase activation protein. - 2.37 ± 1.42 gamma polypeptide Zfyve21 Zinc finger. FYVE domain containing 21 1.93 ± 0.57 Ligands and receptors Bambi BMP and activin membrane-bound inhibitor - 2.94 ± 0.62
    [Show full text]
  • Carbon Monoxide Prevents TNF-Α-Induced Enos Downregulation by Inhibiting NF-Κb-Responsive Mir-155-5P Biogenesis
    OPEN Experimental & Molecular Medicine (2017) 49, e403; doi:10.1038/emm.2017.193 Official journal of the Korean Society for Biochemistry and Molecular Biology www.nature.com/emm ORIGINAL ARTICLE Carbon monoxide prevents TNF-α-induced eNOS downregulation by inhibiting NF-κB-responsive miR-155-5p biogenesis Seunghwan Choi1, Joohwan Kim1, Ji-Hee Kim1, Dong-Keon Lee1, Wonjin Park1, Minsik Park1, Suji Kim1, Jong Yun Hwang2, Moo-Ho Won3, Yoon Kyung Choi1,4, Sungwoo Ryoo5, Kwon-Soo Ha1, Young-Guen Kwon6 and Young-Myeong Kim1 Heme oxygenase-1-derived carbon monoxide prevents inflammatory vascular disorders. To date, there is no clear evidence that HO-1/CO prevents endothelial dysfunction associated with the downregulation of endothelial NO synthesis in human endothelial cells stimulated with TNF-α. Here, we found that the CO-releasing compound CORM-2 prevented TNF-α-mediated decreases in eNOS expression and NO/cGMP production, without affecting eNOS promoter activity, by maintaining the functional activity of the eNOS mRNA 3′-untranslated region. By contrast, CORM-2 inhibited MIR155HG expression and miR-155-5p biogenesis in TNF-α-stimulated endothelial cells, resulting in recovery of the 3′-UTR activity of eNOS mRNA, a target of miR-155-5p. The beneficial effect of CORM-2 was blocked by an NF-κB inhibitor, a miR-155-5p mimic, a HO-1 inhibitor and siRNA against HO-1, indicating that CO rescues TNF-α-induced eNOS downregulation through NF-κB-responsive miR-155-5p expression via HO-1 induction; similar protective effects of ectopic HO-1 expression and bilirubin were observed in endothelial cells treated with TNF-α.
    [Show full text]
  • Upregulation of Peroxisome Proliferator-Activated Receptor-Α And
    Upregulation of peroxisome proliferator-activated receptor-α and the lipid metabolism pathway promotes carcinogenesis of ampullary cancer Chih-Yang Wang, Ying-Jui Chao, Yi-Ling Chen, Tzu-Wen Wang, Nam Nhut Phan, Hui-Ping Hsu, Yan-Shen Shan, Ming-Derg Lai 1 Supplementary Table 1. Demographics and clinical outcomes of five patients with ampullary cancer Time of Tumor Time to Age Differentia survival/ Sex Staging size Morphology Recurrence recurrence Condition (years) tion expired (cm) (months) (months) T2N0, 51 F 211 Polypoid Unknown No -- Survived 193 stage Ib T2N0, 2.41.5 58 F Mixed Good Yes 14 Expired 17 stage Ib 0.6 T3N0, 4.53.5 68 M Polypoid Good No -- Survived 162 stage IIA 1.2 T3N0, 66 M 110.8 Ulcerative Good Yes 64 Expired 227 stage IIA T3N0, 60 M 21.81 Mixed Moderate Yes 5.6 Expired 16.7 stage IIA 2 Supplementary Table 2. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of an ampullary cancer microarray using the Database for Annotation, Visualization and Integrated Discovery (DAVID). This table contains only pathways with p values that ranged 0.0001~0.05. KEGG Pathway p value Genes Pentose and 1.50E-04 UGT1A6, CRYL1, UGT1A8, AKR1B1, UGT2B11, UGT2A3, glucuronate UGT2B10, UGT2B7, XYLB interconversions Drug metabolism 1.63E-04 CYP3A4, XDH, UGT1A6, CYP3A5, CES2, CYP3A7, UGT1A8, NAT2, UGT2B11, DPYD, UGT2A3, UGT2B10, UGT2B7 Maturity-onset 2.43E-04 HNF1A, HNF4A, SLC2A2, PKLR, NEUROD1, HNF4G, diabetes of the PDX1, NR5A2, NKX2-2 young Starch and sucrose 6.03E-04 GBA3, UGT1A6, G6PC, UGT1A8, ENPP3, MGAM, SI, metabolism
    [Show full text]
  • Table 2. Significant
    Table 2. Significant (Q < 0.05 and |d | > 0.5) transcripts from the meta-analysis Gene Chr Mb Gene Name Affy ProbeSet cDNA_IDs d HAP/LAP d HAP/LAP d d IS Average d Ztest P values Q-value Symbol ID (study #5) 1 2 STS B2m 2 122 beta-2 microglobulin 1452428_a_at AI848245 1.75334941 4 3.2 4 3.2316485 1.07398E-09 5.69E-08 Man2b1 8 84.4 mannosidase 2, alpha B1 1416340_a_at H4049B01 3.75722111 3.87309653 2.1 1.6 2.84852656 5.32443E-07 1.58E-05 1110032A03Rik 9 50.9 RIKEN cDNA 1110032A03 gene 1417211_a_at H4035E05 4 1.66015788 4 1.7 2.82772795 2.94266E-05 0.000527 NA 9 48.5 --- 1456111_at 3.43701477 1.85785922 4 2 2.8237185 9.97969E-08 3.48E-06 Scn4b 9 45.3 Sodium channel, type IV, beta 1434008_at AI844796 3.79536664 1.63774235 3.3 2.3 2.75319499 1.48057E-08 6.21E-07 polypeptide Gadd45gip1 8 84.1 RIKEN cDNA 2310040G17 gene 1417619_at 4 3.38875643 1.4 2 2.69163229 8.84279E-06 0.0001904 BC056474 15 12.1 Mus musculus cDNA clone 1424117_at H3030A06 3.95752801 2.42838452 1.9 2.2 2.62132809 1.3344E-08 5.66E-07 MGC:67360 IMAGE:6823629, complete cds NA 4 153 guanine nucleotide binding protein, 1454696_at -3.46081884 -4 -1.3 -1.6 -2.6026947 8.58458E-05 0.0012617 beta 1 Gnb1 4 153 guanine nucleotide binding protein, 1417432_a_at H3094D02 -3.13334396 -4 -1.6 -1.7 -2.5946297 1.04542E-05 0.0002202 beta 1 Gadd45gip1 8 84.1 RAD23a homolog (S.
    [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]
  • Regulation of Xenobiotic and Bile Acid Metabolism by the Anti-Aging Intervention Calorie Restriction in Mice
    REGULATION OF XENOBIOTIC AND BILE ACID METABOLISM BY THE ANTI-AGING INTERVENTION CALORIE RESTRICTION IN MICE By Zidong Fu Submitted to the Graduate Degree Program in Pharmacology, Toxicology, and Therapeutics and the Graduate Faculty of the University of Kansas in partial fulfillment of the requirements for the degree of Doctor of Philosophy. Dissertation Committee ________________________________ Chairperson: Curtis Klaassen, Ph.D. ________________________________ Udayan Apte, Ph.D. ________________________________ Wen-Xing Ding, Ph.D. ________________________________ Thomas Pazdernik, Ph.D. ________________________________ Hao Zhu, Ph.D. Date Defended: 04-11-2013 The Dissertation Committee for Zidong Fu certifies that this is the approved version of the following dissertation: REGULATION OF XENOBIOTIC AND BILE ACID METABOLISM BY THE ANTI-AGING INTERVENTION CALORIE RESTRICTION IN MICE ________________________________ Chairperson: Curtis Klaassen, Ph.D. Date approved: 04-11-2013 ii ABSTRACT Calorie restriction (CR), defined as reduced calorie intake without causing malnutrition, is the best-known intervention to increase life span and slow aging-related diseases in various species. However, current knowledge on the exact mechanisms of aging and how CR exerts its anti-aging effects is still inadequate. The detoxification theory of aging proposes that the up-regulation of xenobiotic processing genes (XPGs) involved in phase-I and phase-II xenobiotic metabolism as well as transport, which renders a wide spectrum of detoxification, is a longevity mechanism. Interestingly, bile acids (BAs), the metabolites of cholesterol, have recently been connected with longevity. Thus, this dissertation aimed to determine the regulation of xenobiotic and BA metabolism by the well-known anti-aging intervention CR. First, the mRNA expression of XPGs in liver during aging was investigated.
    [Show full text]
  • Flavin-Containing Monooxygenases: Mutations, Disease and Drug Response Phillips, IR; Shephard, EA
    Flavin-containing monooxygenases: mutations, disease and drug response Phillips, IR; Shephard, EA For additional information about this publication click this link. http://qmro.qmul.ac.uk/jspui/handle/123456789/1015 Information about this research object was correct at the time of download; we occasionally make corrections to records, please therefore check the published record when citing. For more information contact [email protected] Flavin-containing monooxygenases: mutations, disease and drug response Ian R. Phillips1 and Elizabeth A. Shephard2 1School of Biological and Chemical Sciences, Queen Mary, University of London, Mile End Road, London E1 4NS, UK 2Department of Biochemistry and Molecular Biology, University College London, Gower Street, London WC1E 6BT, UK Corresponding author: Shephard, E.A. ([email protected]). and, thus, contribute to drug development. This review Flavin-containing monooxygenases (FMOs) metabolize considers the role of FMOs and their genetic variants in numerous foreign chemicals, including drugs, pesticides disease and drug response. and dietary components and, thus, mediate interactions between humans and their chemical environment. We Mechanism and structure describe the mechanism of action of FMOs and insights For catalysis FMOs require flavin adenine dinucleotide gained from the structure of yeast FMO. We then (FAD) as a prosthetic group, NADPH as a cofactor and concentrate on the three FMOs (FMOs 1, 2 and 3) that are molecular oxygen as a cosubstrate [5,6]. In contrast to most important for metabolism of foreign chemicals in CYPs FMOs accept reducing equivalents directly from humans, focusing on the role of the FMOs and their genetic NADPH and, thus, do not require accessory proteins.
    [Show full text]
  • Type of the Paper (Article
    Supplementary Material A Proteomics Study on the Mechanism of Nutmeg-induced Hepatotoxicity Wei Xia 1, †, Zhipeng Cao 1, †, Xiaoyu Zhang 1 and Lina Gao 1,* 1 School of Forensic Medicine, China Medical University, Shenyang 110122, P. R. China; lessen- [email protected] (W.X.); [email protected] (Z.C.); [email protected] (X.Z.) † The authors contributed equally to this work. * Correspondence: [email protected] Figure S1. Table S1. Peptide fraction separation liquid chromatography elution gradient table. Time (min) Flow rate (mL/min) Mobile phase A (%) Mobile phase B (%) 0 1 97 3 10 1 95 5 30 1 80 20 48 1 60 40 50 1 50 50 53 1 30 70 54 1 0 100 1 Table 2. Liquid chromatography elution gradient table. Time (min) Flow rate (nL/min) Mobile phase A (%) Mobile phase B (%) 0 600 94 6 2 600 83 17 82 600 60 40 84 600 50 50 85 600 45 55 90 600 0 100 Table S3. The analysis parameter of Proteome Discoverer 2.2. Item Value Type of Quantification Reporter Quantification (TMT) Enzyme Trypsin Max.Missed Cleavage Sites 2 Precursor Mass Tolerance 10 ppm Fragment Mass Tolerance 0.02 Da Dynamic Modification Oxidation/+15.995 Da (M) and TMT /+229.163 Da (K,Y) N-Terminal Modification Acetyl/+42.011 Da (N-Terminal) and TMT /+229.163 Da (N-Terminal) Static Modification Carbamidomethyl/+57.021 Da (C) 2 Table S4. The DEPs between the low-dose group and the control group. Protein Gene Fold Change P value Trend mRNA H2-K1 0.380 0.010 down Glutamine synthetase 0.426 0.022 down Annexin Anxa6 0.447 0.032 down mRNA H2-D1 0.467 0.002 down Ribokinase Rbks 0.487 0.000
    [Show full text]
  • Targeting Heme Oxygenase-1 in Cardiovascular and Kidney Disease
    antioxidants Review Targeting Heme Oxygenase-1 in Cardiovascular and Kidney Disease Heather A. Drummond 1, Zachary L. Mitchell 1, Nader G. Abraham 2,3 and David E. Stec 1,* 1 Department of Physiology and Biophysics, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MI 39216, USA; [email protected] (H.A.D.); [email protected] (Z.L.M.) 2 Departments of Medicine and Pharmacology, New York Medical College, Vahalla, NY 10595, USA; [email protected] 3 Joan C. Edwards School of Medicine, Marshall University, Huntington, VA 25701, USA * Correspondence: [email protected]; Tel.: +1-601-815-1859; Fax: +1-601-984-1817 Received: 3 June 2019; Accepted: 15 June 2019; Published: 18 June 2019 Abstract: Heme oxygenase (HO) plays an important role in the cardiovascular system. It is involved in many physiological and pathophysiological processes in all organs of the cardiovascular system. From the regulation of blood pressure and blood flow to the adaptive response to end-organ injury, HO plays a critical role in the ability of the cardiovascular system to respond and adapt to changes in homeostasis. There have been great advances in our understanding of the role of HO in the regulation of blood pressure and target organ injury in the last decade. Results from these studies demonstrate that targeting of the HO system could provide novel therapeutic opportunities for the treatment of several cardiovascular and renal diseases. The goal of this review is to highlight the important role of HO in the regulation of cardiovascular and renal function and protection from disease and to highlight areas in which targeting of the HO system needs to be translated to help benefit patient populations.
    [Show full text]
  • Supplementary Materials
    Supplementary Materials COMPARATIVE ANALYSIS OF THE TRANSCRIPTOME, PROTEOME AND miRNA PROFILE OF KUPFFER CELLS AND MONOCYTES Andrey Elchaninov1,3*, Anastasiya Lokhonina1,3, Maria Nikitina2, Polina Vishnyakova1,3, Andrey Makarov1, Irina Arutyunyan1, Anastasiya Poltavets1, Evgeniya Kananykhina2, Sergey Kovalchuk4, Evgeny Karpulevich5,6, Galina Bolshakova2, Gennady Sukhikh1, Timur Fatkhudinov2,3 1 Laboratory of Regenerative Medicine, National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia 2 Laboratory of Growth and Development, Scientific Research Institute of Human Morphology, Moscow, Russia 3 Histology Department, Medical Institute, Peoples' Friendship University of Russia, Moscow, Russia 4 Laboratory of Bioinformatic methods for Combinatorial Chemistry and Biology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia 5 Information Systems Department, Ivannikov Institute for System Programming of the Russian Academy of Sciences, Moscow, Russia 6 Genome Engineering Laboratory, Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia Figure S1. Flow cytometry analysis of unsorted blood sample. Representative forward, side scattering and histogram are shown. The proportions of negative cells were determined in relation to the isotype controls. The percentages of positive cells are indicated. The blue curve corresponds to the isotype control. Figure S2. Flow cytometry analysis of unsorted liver stromal cells. Representative forward, side scattering and histogram are shown. The proportions of negative cells were determined in relation to the isotype controls. The percentages of positive cells are indicated. The blue curve corresponds to the isotype control. Figure S3. MiRNAs expression analysis in monocytes and Kupffer cells. Full-length of heatmaps are presented.
    [Show full text]
  • Supplementary Table S4. FGA Co-Expressed Gene List in LUAD
    Supplementary Table S4. FGA co-expressed gene list in LUAD tumors Symbol R Locus Description FGG 0.919 4q28 fibrinogen gamma chain FGL1 0.635 8p22 fibrinogen-like 1 SLC7A2 0.536 8p22 solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 DUSP4 0.521 8p12-p11 dual specificity phosphatase 4 HAL 0.51 12q22-q24.1histidine ammonia-lyase PDE4D 0.499 5q12 phosphodiesterase 4D, cAMP-specific FURIN 0.497 15q26.1 furin (paired basic amino acid cleaving enzyme) CPS1 0.49 2q35 carbamoyl-phosphate synthase 1, mitochondrial TESC 0.478 12q24.22 tescalcin INHA 0.465 2q35 inhibin, alpha S100P 0.461 4p16 S100 calcium binding protein P VPS37A 0.447 8p22 vacuolar protein sorting 37 homolog A (S. cerevisiae) SLC16A14 0.447 2q36.3 solute carrier family 16, member 14 PPARGC1A 0.443 4p15.1 peroxisome proliferator-activated receptor gamma, coactivator 1 alpha SIK1 0.435 21q22.3 salt-inducible kinase 1 IRS2 0.434 13q34 insulin receptor substrate 2 RND1 0.433 12q12 Rho family GTPase 1 HGD 0.433 3q13.33 homogentisate 1,2-dioxygenase PTP4A1 0.432 6q12 protein tyrosine phosphatase type IVA, member 1 C8orf4 0.428 8p11.2 chromosome 8 open reading frame 4 DDC 0.427 7p12.2 dopa decarboxylase (aromatic L-amino acid decarboxylase) TACC2 0.427 10q26 transforming, acidic coiled-coil containing protein 2 MUC13 0.422 3q21.2 mucin 13, cell surface associated C5 0.412 9q33-q34 complement component 5 NR4A2 0.412 2q22-q23 nuclear receptor subfamily 4, group A, member 2 EYS 0.411 6q12 eyes shut homolog (Drosophila) GPX2 0.406 14q24.1 glutathione peroxidase
    [Show full text]
  • Supplementary Materials
    Supplementary materials Supplementary Table S1: MGNC compound library Ingredien Molecule Caco- Mol ID MW AlogP OB (%) BBB DL FASA- HL t Name Name 2 shengdi MOL012254 campesterol 400.8 7.63 37.58 1.34 0.98 0.7 0.21 20.2 shengdi MOL000519 coniferin 314.4 3.16 31.11 0.42 -0.2 0.3 0.27 74.6 beta- shengdi MOL000359 414.8 8.08 36.91 1.32 0.99 0.8 0.23 20.2 sitosterol pachymic shengdi MOL000289 528.9 6.54 33.63 0.1 -0.6 0.8 0 9.27 acid Poricoic acid shengdi MOL000291 484.7 5.64 30.52 -0.08 -0.9 0.8 0 8.67 B Chrysanthem shengdi MOL004492 585 8.24 38.72 0.51 -1 0.6 0.3 17.5 axanthin 20- shengdi MOL011455 Hexadecano 418.6 1.91 32.7 -0.24 -0.4 0.7 0.29 104 ylingenol huanglian MOL001454 berberine 336.4 3.45 36.86 1.24 0.57 0.8 0.19 6.57 huanglian MOL013352 Obacunone 454.6 2.68 43.29 0.01 -0.4 0.8 0.31 -13 huanglian MOL002894 berberrubine 322.4 3.2 35.74 1.07 0.17 0.7 0.24 6.46 huanglian MOL002897 epiberberine 336.4 3.45 43.09 1.17 0.4 0.8 0.19 6.1 huanglian MOL002903 (R)-Canadine 339.4 3.4 55.37 1.04 0.57 0.8 0.2 6.41 huanglian MOL002904 Berlambine 351.4 2.49 36.68 0.97 0.17 0.8 0.28 7.33 Corchorosid huanglian MOL002907 404.6 1.34 105 -0.91 -1.3 0.8 0.29 6.68 e A_qt Magnogrand huanglian MOL000622 266.4 1.18 63.71 0.02 -0.2 0.2 0.3 3.17 iolide huanglian MOL000762 Palmidin A 510.5 4.52 35.36 -0.38 -1.5 0.7 0.39 33.2 huanglian MOL000785 palmatine 352.4 3.65 64.6 1.33 0.37 0.7 0.13 2.25 huanglian MOL000098 quercetin 302.3 1.5 46.43 0.05 -0.8 0.3 0.38 14.4 huanglian MOL001458 coptisine 320.3 3.25 30.67 1.21 0.32 0.9 0.26 9.33 huanglian MOL002668 Worenine
    [Show full text]