Supplemental Table 1

Total Page:16

File Type:pdf, Size:1020Kb

Supplemental Table 1 Induced CYP3A4 expression in confluent Huh7 hepatoma cells as a result of decreased cell proliferation and subsequent PXR activation Louise Sivertsson, Irene Edebert, Margareta Porsmyr Palmertz, Magnus Ingelman-Sundberg and Etienne P.A. Neve Molecular Pharmacology Supplemental table 1. Genes that were differentially (p<0.05) up-regulated in confluent cells compared to control cells. Genes with FC > 2 are above the red line ProbeSetID GeneAccession GeneSymbol GeneDescription Mean FC p value 8100734 NM_001077 UGT2B17 UDP glucuronosyltransferase 2 family, polypeptid 436.02 6.42E-03 7995820 NM_005947 MT1B metallothionein 1B 48.48 1.05E-02 7983478 NM_032413 C15orf48 chromosome 15 open reading frame 48 46.24 4.59E-02 8147065 NM_173848 RALYL RALY RNA binding protein-like 44.98 5.30E-05 8100853 NM_000583 GC group-specific component (vitamin D binding prot 44.20 2.69E-02 8101874 NM_000667 ADH1A alcohol dehydrogenase 1A (class I), alpha polypep 43.71 2.17E-06 7940775 NM_004585 RARRES3 retinoic acid receptor responder (tazarotene induce 40.49 2.20E-04 7904429 AF252254 LOC128102 3-beta-hydroxysteroid dehydrogenase, tissue-type 36.97 1.37E-03 8089145 NM_015429 ABI3BP ABI family, member 3 (NESH) binding protein 36.57 2.04E-02 8141328 NM_000777 CYP3A5 cytochrome P450, family 3, subfamily A, polypept31.14 4.48E-03 7970949 NM_005584 MAB21L1 mab-21-like 1 (C. elegans) 30.83 3.03E-05 7904726 NM_006472 TXNIP thioredoxin interacting protein 30.72 8.57E-04 8101893 NM_000669 ADH1C alcohol dehydrogenase 1C (class I), gamma polype 26.88 9.95E-04 7933341 NM_019893 ASAH2 N-acylsphingosine amidohydrolase (non-lysosoma 26.68 1.25E-02 8100737 NM_001076 UGT2B15 UDP glucuronosyltransferase 2 family, polypeptid 26.02 6.24E-03 8100746 NM_001076 UGT2B15 UDP glucuronosyltransferase 2 family, polypeptid 26.02 6.24E-03 8035095 NM_021187 CYP4F11 cytochrome P450, family 4, subfamily F, polypept25.97 9.50E-05 7933597 NM_019893 ASAH2 N-acylsphingosine amidohydrolase (non-lysosoma 24.87 1.05E-02 8100760 NM_024743 UGT2A3 UDP glucuronosyltransferase 2 family, polypeptid 24.31 1.26E-02 8086457 NM_004391 CYP8B1 cytochrome P450, family 8, subfamily B, polypept23.81 3.59E-02 8150920 NM_000780 CYP7A1 cytochrome P450, family 7, subfamily A, polypept22.77 4.31E-05 8112053 NM_152623 CDC20B cell division cycle 20 homolog B (S. cerevisiae) 22.55 5.55E-06 7913896 NM_001004434 SLC30A2 solute carrier family 30 (zinc transporter), member 22.12 4.66E-03 8140650 NM_012431 SEMA3E sema domain, immunoglobulin domain (Ig), short 20.43 1.19E-02 8007429 NM_000151 G6PC glucose-6-phosphatase, catalytic subunit 20.26 1.98E-02 7954356 NM_006446 SLCO1B1 solute carrier organic anion transporter family, me19.97 1.86E-06 8133876 NM_001001548 CD36 CD36 molecule (thrombospondin receptor) 19.88 1.57E-02 8077192 NM_000482 APOA4 apolipoprotein A-IV 18.91 5.65E-04 8171449 NM_021804 ACE2 angiotensin I converting enzyme (peptidyl-dipeptid18.90 3.34E-03 7951859 NM_000482 APOA4 apolipoprotein A-IV 18.79 7.05E-04 8126750 NM_021572 ENPP5 ectonucleotide pyrophosphatase/phosphodiesterase 18.03 9.98E-03 8109629 NM_001040442 FABP6 fatty acid binding protein 6, ileal 17.80 9.16E-03 8159491 NM_000606 C8G complement component 8, gamma polypeptide 16.87 1.81E-03 8056151 NM_007366 PLA2R1 phospholipase A2 receptor 1, 180kDa 16.37 7.68E-03 8072735 NM_145343 APOL1 apolipoprotein L, 1 15.90 1.82E-03 8098654 NM_000892 KLKB1 kallikrein B, plasma (Fletcher factor) 1 15.53 7.27E-03 8095402 NM_024743 UGT2A3 UDP glucuronosyltransferase 2 family, polypeptid 15.52 4.25E-03 8140971 NM_152703 SAMD9L sterile alpha motif domain containing 9-like 15.33 3.96E-03 8009145 NM_030779 KCNH6 potassium voltage-gated channel, subfamily H (eag14.93 2.99E-03 8111864 NM_001115131 C6 complement component 6 14.74 2.67E-02 7981998 NR_003339 SNORD116-25 small nucleolar RNA, C/D box 116-25 14.00 2.27E-02 8117304 NM_005495 SLC17A4 solute carrier family 17 (sodium phosphate), memb 13.87 3.59E-03 7916526 NM_000066 C8B complement component 8, beta polypeptide 13.45 1.80E-02 8090180 NM_033049 MUC13 mucin 13, cell surface associated 13.33 2.59E-04 8083301 NM_004617 TM4SF4 transmembrane 4 L six family member 4 13.08 3.06E-02 7904396 NM_001005783 HAO2 hydroxyacid oxidase 2 (long chain) 12.92 3.03E-03 7964183 NM_013267 GLS2 glutaminase 2 (liver, mitochondrial) 12.92 1.07E-03 8108006 NM_052971 LEAP2 liver expressed antimicrobial peptide 2 12.85 3.92E-03 8108251 NR_002713 NPY6R neuropeptide Y receptor Y6 (pseudogene) 12.78 1.62E-02 7919800 NM_004079 CTSS cathepsin S 12.59 1.14E-03 7940341 NM_206893 MS4A10 membrane-spanning 4-domains, subfamily A, mem12.49 4.48E-03 8049297 NR_003008 SCARNA5 small Cajal body-specific RNA 5 12.30 7.68E-04 8092735 NM_207316 TMEM207 transmembrane protein 207 11.91 2.25E-06 8059525 NM_024795 TM4SF20 transmembrane 4 L six family member 20 11.89 3.91E-02 8018975 NM_005567 LGALS3BP lectin, galactoside-binding, soluble, 3 binding prot 11.84 3.78E-03 8165575 NM_001098537 PNPLA7 patatin-like phospholipase domain containing 7 11.62 1.41E-06 7935169 NM_000770 CYP2C8 cytochrome P450, family 2, subfamily C, polypept11.52 1.59E-02 7911754 NM_003820 TNFRSF14 tumor necrosis factor receptor superfamily, membe 11.48 3.86E-03 8141374 NM_001185 AZGP1 alpha-2-glycoprotein 1, zinc-binding 11.48 3.29E-02 8062927 NM_002638 PI3 peptidase inhibitor 3, skin-derived 11.27 3.58E-02 7942717 NM_000260 MYO7A myosin VIIA 11.17 1.15E-04 8075483 NM_052880 PIK3IP1 phosphoinositide-3-kinase interacting protein 1 11.01 9.39E-04 7904408 NM_000198 HSD3B2 hydroxy-delta-5-steroid dehydrogenase, 3 beta- an 10.97 1.29E-02 8173444 NM_000206 IL2RG interleukin 2 receptor, gamma (severe combined im 10.92 6.57E-03 8112920 NM_130767 ACOT12 acyl-CoA thioesterase 12 10.84 8.23E-04 8020037 BC040542 KIAA0802 KIAA0802 10.80 2.08E-03 8118409 NM_007293 C4A complement component 4A (Rodgers blood group 10.61 4.84E-02 8118455 NM_007293 C4A complement component 4A (Rodgers blood group 10.61 4.84E-02 8179399 NM_007293 C4A complement component 4A (Rodgers blood group 10.61 4.84E-02 8134655 NM_022820 CYP3A43 cytochrome P450, family 3, subfamily A, polypept10.54 3.52E-02 7989951 ENST00000380078LOC100131796LP2570 10.43 4.10E-04 7904421 NM_000862 HSD3B1 hydroxy-delta-5-steroid dehydrogenase, 3 beta- an 10.34 1.03E-04 8027760 NM_005031 FXYD1 FXYD domain containing ion transport regulator 1 10.19 1.37E-02 8045795 NM_002239 KCNJ3 potassium inwardly-rectifying channel, subfamily J 10.17 2.91E-03 7917503 NM_018284 GBP3 guanylate binding protein 3 10.14 5.69E-03 8092169 NM_003810 TNFSF10 tumor necrosis factor (ligand) superfamily, membe 9.84 1.30E-03 8065948 NR_024377 FER1L4 fer-1-like 4 (C. elegans) 9.80 1.08E-02 7904414 NM_000862 HSD3B1 hydroxy-delta-5-steroid dehydrogenase, 3 beta- an 9.80 7.25E-05 8056583 NM_003742 ABCB11 ATP-binding cassette, sub-family B (MDR/TAP), 9.74 7.40E-03 8008900 NM_000717 CA4 carbonic anhydrase IV 9.45 1.17E-02 8137483 AK131514 FLJ16734 hypothetical LOC641928 9.45 5.26E-03 8055314 NM_144586 LYPD1 LY6/PLAUR domain containing 1 9.42 7.39E-05 8136709 NR_003715 LOC93432 maltase-glucoamylase-like pseudogene 9.34 1.57E-02 7945169 NM_138788 TMEM45B transmembrane protein 45B 9.34 1.09E-02 7951853 NM_052968 APOA5 apolipoprotein A-V 9.28 1.32E-02 7988350 NM_014080 DUOX2 dual oxidase 2 9.22 1.97E-02 8080466 NM_002217 ITIH3 inter-alpha (globulin) inhibitor H3 8.98 1.30E-03 8135015 AB038783 MUC3B mucin 3B, cell surface associated 8.87 4.20E-04 8145097 NM_018068 PIWIL2 piwi-like 2 (Drosophila) 8.87 3.49E-04 8065920 NR_024377 FER1L4 fer-1-like 4 (C. elegans) 8.75 1.97E-02 8156770 NM_024642 GALNT12 UDP-N-acetyl-alpha-D-galactosamine:polypeptide 8.67 2.04E-04 8020551 NM_198129 LAMA3 laminin, alpha 3 8.44 7.99E-04 7986350 NM_183376 ARRDC4 arrestin domain containing 4 8.41 4.58E-03 7917516 NM_002053 GBP1 guanylate binding protein 1, interferon-inducible, 6 8.29 1.56E-04 7970793 NM_181785 SLC46A3 solute carrier family 46, member 3 8.23 2.62E-02 8022559 NM_173505 ANKRD29 ankyrin repeat domain 29 8.20 2.04E-02 8141016 NM_006528 TFPI2 tissue factor pathway inhibitor 2 8.07 1.09E-03 8095663 NM_001133 AFM afamin 8.05 1.25E-02 8119782 NM_006672 SLC22A7 solute carrier family 22 (organic anion transporter) 7.99 1.14E-03 8075865 NM_153609 TMPRSS6 transmembrane protease, serine 6 7.96 4.17E-03 8023598 NM_173557 RNF152 ring finger protein 152 7.80 4.29E-03 8107909 NM_003059 SLC22A4 solute carrier family 22 (organic cation/ergothione 7.64 1.60E-03 7937782 NM_139022 TSPAN32 tetraspanin 32 7.57 1.87E-02 8102328 NM_000204 CFI complement factor I 7.49 1.40E-02 7917532 NM_004120 GBP2 guanylate binding protein 2, interferon-inducible 7.48 2.29E-03 8009913 NR_003587 MYO15B myosin XVB pseudogene 7.43 9.34E-04 8077198 BC057848 FLJ43315 similar to Asparagine synthetase [glutamine-hydro7.42 3.62E-02 8070467 NM_001135099 TMPRSS2 transmembrane protease, serine 2 7.35 3.54E-03 7986755 NM_019066 MAGEL2 MAGE-like 2 7.22 2.34E-02 8081925 NM_003889 NR1I2 nuclear receptor subfamily 1, group I, member 2 7.21 1.60E-03 8095390 NM_001075 UGT2B10 UDP glucuronosyltransferase 2 family, polypeptid 7.14 9.13E-04 8081298 NM_032787 GPR128 G protein-coupled receptor 128 7.09 1.75E-03 8050115 AK127578 C2orf46 chromosome 2 open reading frame 46 7.08 1.54E-03 8035083 NM_001082 CYP4F2 cytochrome P450, family 4, subfamily F, polypept 7.04 6.78E-04 8098671 NM_000128 F11 coagulation factor XI 6.84 5.50E-03 8011114 NR_028076 SCARF1 scavenger receptor class F, member 1 6.81 3.15E-04 7917548 NM_207398 GBP7 guanylate binding protein 7 6.79 1.02E-03 7980908 NM_006329 FBLN5 fibulin 5 6.76 3.32E-04 8040960 NM_001486 GCKR glucokinase (hexokinase
Recommended publications
  • Human Cathepsin A/ Lysosomal Carboxypeptidase a Antibody
    Human Cathepsin A/ Lysosomal Carboxypeptidase A Antibody Monoclonal Mouse IgG2A Clone # 179803 Catalog Number: MAB1049 DESCRIPTION Species Reactivity Human Specificity Detects human Cathepsin A/Lysosomal Carboxypeptidase A in direct ELISAs and Western blots. In Western blots, detects the single chain (55 kDa) and heavy chain (32 kDa) forms of recombinant human (rh) Cathepsin A. In Western blots, less than 5% cross­reactivity with rhCathepsin B, C, D, E, L, O, S, X and Z is observed and no cross­reactivity with the light chain (20 kDa) of rhCathepsin A is observed. Source Monoclonal Mouse IgG2A Clone # 179803 Purification Protein A or G purified from hybridoma culture supernatant Immunogen Mouse myeloma cell line NS0­derived recombinant human Cathepsin A/Lysosomal Carboxypeptidase A Ala29­Tyr480 (predicted) Accession # P10619 Formulation Lyophilized from a 0.2 μm filtered solution in PBS with Trehalose. See Certificate of Analysis for details. *Small pack size (­SP) is supplied either lyophilized or as a 0.2 μm filtered solution in PBS. APPLICATIONS Please Note: Optimal dilutions should be determined by each laboratory for each application. General Protocols are available in the Technical Information section on our website. Recommended Sample Concentration Western Blot 1 µg/mL Recombinant Human Cathepsin A/Lysosomal Carboxypeptidase A (Catalog # 1049­SE) Immunoprecipitation 25 µg/mL Conditioned cell culture medium spiked with Recombinant Human Cathepsin A/Lysosomal Carboxypeptidase A (Catalog # 1049­SE), see our available Western blot detection antibodies PREPARATION AND STORAGE Reconstitution Reconstitute at 0.5 mg/mL in sterile PBS. Shipping The product is shipped at ambient temperature. Upon receipt, store it immediately at the temperature recommended below.
    [Show full text]
  • Cloning and Sequence Analysis of Canine Apoptosis-Associated Molecules
    Zurich Open Repository and Archive University of Zurich Main Library Strickhofstrasse 39 CH-8057 Zurich www.zora.uzh.ch Year: 2007 Cloning and sequence analysis of canine apoptosis-associated molecules Schade, Benjamin Abstract: The aim of the study was to clone and sequence the coding sequences of a representative set of proteins, primarily from the intrinsic apoptotic pathway in dogs and to assess their conservation with hu- man and murine orthologues. cDNA for these proteins, including Bcl-2 family members (Bcl-XL, Bcl-w, Mcl-1, Bax, Bak, Bad, Noxa), caspases (Caspase-3, Caspase-8, Caspase-9), Inhibitors of Apoptosis Pro- teins (XIAP, cIAP-1, cIAP-2, Survivin), their mitochondrial inhibitors (Smac/ DIABLO, Omi/HtrA2) and p53, were generated by RT-PCR with RNA extracted from two canine non-neoplastic cell lines. Eleven sequences are novel for the dog. Interspecies comparison revealed strongest similarity between the sequences of human and canine intrinsic apoptosis pathway members. Differences with potential func- tional impact, however, were observed in both dogs and mice. In dogs, these changes involve the putative Inhibitor of Apoptosis Protein binding motif of canine Omi/HtrA2, some caspase substrate recognition motifs and some functionally relevant residues of p53. Canine XIAP yields a caspase-cleavage site reported as unique to humans. In conclusion, the generally high degree of similarity of canine apoptosis-associated proteins as compared to human counterparts is supportive of the use of dogs as a model for human dis- eases. Single interspecies sequence variations with potential functional relevance under physiologic and neoplastic conditions do exist, however, and will require further analysis.
    [Show full text]
  • Implications in Parkinson's Disease
    Journal of Clinical Medicine Review Lysosomal Ceramide Metabolism Disorders: Implications in Parkinson’s Disease Silvia Paciotti 1,2 , Elisabetta Albi 3 , Lucilla Parnetti 1 and Tommaso Beccari 3,* 1 Laboratory of Clinical Neurochemistry, Department of Medicine, University of Perugia, Sant’Andrea delle Fratte, 06132 Perugia, Italy; [email protected] (S.P.); [email protected] (L.P.) 2 Section of Physiology and Biochemistry, Department of Experimental Medicine, University of Perugia, Sant’Andrea delle Fratte, 06132 Perugia, Italy 3 Department of Pharmaceutical Sciences, University of Perugia, Via Fabretti, 06123 Perugia, Italy; [email protected] * Correspondence: [email protected] Received: 29 January 2020; Accepted: 20 February 2020; Published: 21 February 2020 Abstract: Ceramides are a family of bioactive lipids belonging to the class of sphingolipids. Sphingolipidoses are a group of inherited genetic diseases characterized by the unmetabolized sphingolipids and the consequent reduction of ceramide pool in lysosomes. Sphingolipidoses include several disorders as Sandhoff disease, Fabry disease, Gaucher disease, metachromatic leukodystrophy, Krabbe disease, Niemann Pick disease, Farber disease, and GM2 gangliosidosis. In sphingolipidosis, lysosomal lipid storage occurs in both the central nervous system and visceral tissues, and central nervous system pathology is a common hallmark for all of them. Parkinson’s disease, the most common neurodegenerative movement disorder, is characterized by the accumulation and aggregation of misfolded α-synuclein that seem associated to some lysosomal disorders, in particular Gaucher disease. This review provides evidence into the role of ceramide metabolism in the pathophysiology of lysosomes, highlighting the more recent findings on its involvement in Parkinson’s disease. Keywords: ceramide metabolism; Parkinson’s disease; α-synuclein; GBA; GLA; HEX A-B; GALC; ASAH1; SMPD1; ARSA * Correspondence [email protected] 1.
    [Show full text]
  • Supplementary Materials For
    Supplementary Materials for Elucidating cellular population dynamics by molecular density function perturbations 1 2,3, Thanneer Malai Perumal ​ and Rudiyanto Gunawan *​ ​ ​ 1 Sage Bionetworks, Seattle, Washington, USA; [email protected] 2 Institute for Chemical and Bioengineering, ETH Zurich, Zurich, Switzerland; [email protected] 3 Swiss Institute of Bioinformatics, Lausanne, Switzerland * Correspondence: [email protected]; Tel.: +41-44-633-2134 Supplementary Material S1. Probability Distance Metrics Signed Engineering Metric: ∞ 2 ∆ (f A (t, x )||f B (t, x )) = sign(∆μ ) ∫ (x f A (t, x ) − x f B (t, x )) dx (S. 1) E Xi i Xi i Xi i Xi i i Xi i i −∞ Signed Jeffrey Divergence: ∞ f A (t,x ) A B A B Xi i ∆ (f (t, x )||f (t, x )) = sgn(∆μ ) ∫ (f (t, x ) − f (t, x ))ln ( B ) )dx (S. 2) JD Xi i Xi i Xi Xi i Xi i f (t,x ) i −∞ Xi i Signed Kullback-Leibler Distance: ∞ f B (t,x ) A B B Xi i ∆ (f (t, x )||f (t, x )) = sgn(∆μ ) ∫ f (t, x )ln ( A ) dx (S. 3) KLD Xi i Xi i Xi Xi i f (t,x ) i −∞ Xi i Signed Jensen-Shannon Divergence: f + (t,x )+f − (t,x ) Xi i Xi i ∆ f (t, x ) = sgn(∆μ ) (S. 4) JSD Xi i Xi 2 Signed Kolmogorov-Smirnov Metric: ∆ (f A (t, x )||f B (t, x )) = sgn(∆μ )sup|F A (t, x ) − F B (t, x )| (S. 5) KS Xi i Xi i Xi Xi i Xi i Supplementary Material S2.
    [Show full text]
  • Epithelial Delamination Is Protective During Pharmaceutical-Induced Enteropathy
    Epithelial delamination is protective during pharmaceutical-induced enteropathy Scott T. Espenschieda, Mark R. Cronana, Molly A. Mattya, Olaf Muellera, Matthew R. Redinbob,c,d, David M. Tobina,e,f, and John F. Rawlsa,e,1 aDepartment of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710; bDepartment of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599; cDepartment of Biochemistry, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599; dDepartment of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599; eDepartment of Medicine, Duke University School of Medicine, Durham, NC 27710; and fDepartment of Immunology, Duke University School of Medicine, Durham, NC 27710 Edited by Dennis L. Kasper, Harvard Medical School, Boston, MA, and approved July 15, 2019 (received for review February 12, 2019) Intestinal epithelial cell (IEC) shedding is a fundamental response to in mediating intestinal responses to injury remains poorly un- intestinal damage, yet underlying mechanisms and functions have derstood for most xenobiotics. been difficult to define. Here we model chronic intestinal damage in Gastrointestinal pathology is common in people using phar- zebrafish larvae using the nonsteroidal antiinflammatory drug maceuticals, including nonsteroidal antiinflammatory drugs (NSAID) Glafenine. Glafenine induced the unfolded protein response (NSAIDs) (11). While gastric ulceration has historically been a (UPR) and inflammatory pathways in IECs, leading to delamination. defining clinical presentation of NSAID-induced enteropathy, Glafenine-induced inflammation was augmented by microbial colo- small intestinal pathology has also been observed, although the nizationandassociatedwithchanges in intestinal and environmental incidence may be underreported due to diagnostic limitations microbiotas.
    [Show full text]
  • Global Analysis of Protein Folding Thermodynamics for Disease State Characterization
    Global Analysis of Protein Folding Thermodynamics for Disease State Characterization and Biomarker Discovery by Jagat Adhikari Department of Biochemistry Duke University Date:_______________________ Approved: ___________________________ Michael C. Fitzgerald, Supervisor ___________________________ Kenneth Kreuzer ___________________________ Terrence G. Oas ___________________________ Jiyong Hong ___________________________ Seok-Yong Lee Dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Department of Biochemistry in the Graduate School of Duke University 2015 ABSTRACT Global Analysis of Protein Folding Thermodynamics for Disease State Characterization and Biomarker Discovery by Jagat Adhikari Department of Biochemistry Duke University Date:_______________________ Approved: ___________________________ Michael C. Fitzgerald, Supervisor ___________________________ Kenneth Kreuzer ___________________________ Terrence G. Oas ___________________________ Jiyong Hong ___________________________ Seok-Yong Lee An abstract of a dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Department of Biochemistry in the Graduate School of Duke University 2015 Copyright by Jagat Adhikari 2015 Abstract Protein biomarkers can facilitate the diagnosis of many diseases such as cancer and they can be important for the development of effective therapeutic interventions. Current large-scale biomarker discovery and disease state characterization
    [Show full text]
  • Neurodevelopmental Signatures of Narcotic and Neuropsychiatric Risk Factors in 3D Human-Derived Forebrain Organoids
    Molecular Psychiatry www.nature.com/mp ARTICLE OPEN Neurodevelopmental signatures of narcotic and neuropsychiatric risk factors in 3D human-derived forebrain organoids 1 1 1 1 2 2 3 Michael Notaras , Aiman Lodhi , Estibaliz✉ Barrio-Alonso , Careen Foord , Tori Rodrick , Drew Jones , Haoyun Fang , David Greening 3,4 and Dilek Colak 1,5 © The Author(s) 2021 It is widely accepted that narcotic use during pregnancy and specific environmental factors (e.g., maternal immune activation and chronic stress) may increase risk of neuropsychiatric illness in offspring. However, little progress has been made in defining human- specific in utero neurodevelopmental pathology due to ethical and technical challenges associated with accessing human prenatal brain tissue. Here we utilized human induced pluripotent stem cells (hiPSCs) to generate reproducible organoids that recapitulate dorsal forebrain development including early corticogenesis. We systemically exposed organoid samples to chemically defined “enviromimetic” compounds to examine the developmental effects of various narcotic and neuropsychiatric-related risk factors within tissue of human origin. In tandem experiments conducted in parallel, we modeled exposure to opiates (μ-opioid agonist endomorphin), cannabinoids (WIN 55,212-2), alcohol (ethanol), smoking (nicotine), chronic stress (human cortisol), and maternal immune activation (human Interleukin-17a; IL17a). Human-derived dorsal forebrain organoids were consequently analyzed via an array of unbiased and high-throughput analytical approaches, including state-of-the-art TMT-16plex liquid chromatography/mass- spectrometry (LC/MS) proteomics, hybrid MS metabolomics, and flow cytometry panels to determine cell-cycle dynamics and rates of cell death. This pipeline subsequently revealed both common and unique proteome, reactome, and metabolome alterations as a consequence of enviromimetic modeling of narcotic use and neuropsychiatric-related risk factors in tissue of human origin.
    [Show full text]
  • Supplementary Table S1. Upregulated Genes Differentially
    Supplementary Table S1. Upregulated genes differentially expressed in athletes (p < 0.05 and 1.3-fold change) Gene Symbol p Value Fold Change 221051_s_at NMRK2 0.01 2.38 236518_at CCDC183 0.00 2.05 218804_at ANO1 0.00 2.05 234675_x_at 0.01 2.02 207076_s_at ASS1 0.00 1.85 209135_at ASPH 0.02 1.81 228434_at BTNL9 0.03 1.81 229985_at BTNL9 0.01 1.79 215795_at MYH7B 0.01 1.78 217979_at TSPAN13 0.01 1.77 230992_at BTNL9 0.01 1.75 226884_at LRRN1 0.03 1.74 220039_s_at CDKAL1 0.01 1.73 236520_at 0.02 1.72 219895_at TMEM255A 0.04 1.72 201030_x_at LDHB 0.00 1.69 233824_at 0.00 1.69 232257_s_at 0.05 1.67 236359_at SCN4B 0.04 1.64 242868_at 0.00 1.63 1557286_at 0.01 1.63 202780_at OXCT1 0.01 1.63 1556542_a_at 0.04 1.63 209992_at PFKFB2 0.04 1.63 205247_at NOTCH4 0.01 1.62 1554182_at TRIM73///TRIM74 0.00 1.61 232892_at MIR1-1HG 0.02 1.61 204726_at CDH13 0.01 1.6 1561167_at 0.01 1.6 1565821_at 0.01 1.6 210169_at SEC14L5 0.01 1.6 236963_at 0.02 1.6 1552880_at SEC16B 0.02 1.6 235228_at CCDC85A 0.02 1.6 1568623_a_at SLC35E4 0.00 1.59 204844_at ENPEP 0.00 1.59 1552256_a_at SCARB1 0.02 1.59 1557283_a_at ZNF519 0.02 1.59 1557293_at LINC00969 0.03 1.59 231644_at 0.01 1.58 228115_at GAREM1 0.01 1.58 223687_s_at LY6K 0.02 1.58 231779_at IRAK2 0.03 1.58 243332_at LOC105379610 0.04 1.58 232118_at 0.01 1.57 203423_at RBP1 0.02 1.57 AMY1A///AMY1B///AMY1C///AMY2A///AMY2B// 208498_s_at 0.03 1.57 /AMYP1 237154_at LOC101930114 0.00 1.56 1559691_at 0.01 1.56 243481_at RHOJ 0.03 1.56 238834_at MYLK3 0.01 1.55 213438_at NFASC 0.02 1.55 242290_at TACC1 0.04 1.55 ANKRD20A1///ANKRD20A12P///ANKRD20A2///
    [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]
  • Supplementary Table S1. Prioritization of Candidate FPC Susceptibility Genes by Private Heterozygous Ptvs
    Supplementary Table S1. Prioritization of candidate FPC susceptibility genes by private heterozygous PTVs Number of private Number of private Number FPC patient heterozygous PTVs in heterozygous PTVs in tumors with somatic FPC susceptibility Hereditary cancer Hereditary Gene FPC kindred BCCS samples mutation DNA repair gene Cancer driver gene gene gene pancreatitis gene ATM 19 1 - Yes Yes Yes Yes - SSPO 12 8 1 - - - - - DNAH14 10 3 - - - - - - CD36 9 3 - - - - - - TET2 9 1 - - Yes - - - MUC16 8 14 - - - - - - DNHD1 7 4 1 - - - - - DNMT3A 7 1 - - Yes - - - PKHD1L1 7 9 - - - - - - DNAH3 6 5 - - - - - - MYH7B 6 1 - - - - - - PKD1L2 6 6 - - - - - - POLN 6 2 - Yes - - - - POLQ 6 7 - Yes - - - - RP1L1 6 6 - - - - - - TTN 6 5 4 - - - - - WDR87 6 7 - - - - - - ABCA13 5 3 1 - - - - - ASXL1 5 1 - - Yes - - - BBS10 5 0 - - - - - - BRCA2 5 6 1 Yes Yes Yes Yes - CENPJ 5 1 - - - - - - CEP290 5 5 - - - - - - CYP3A5 5 2 - - - - - - DNAH12 5 6 - - - - - - DNAH6 5 1 1 - - - - - EPPK1 5 4 - - - - - - ESYT3 5 1 - - - - - - FRAS1 5 4 - - - - - - HGC6.3 5 0 - - - - - - IGFN1 5 5 - - - - - - KCP 5 4 - - - - - - LRRC43 5 0 - - - - - - MCTP2 5 1 - - - - - - MPO 5 1 - - - - - - MUC4 5 5 - - - - - - OBSCN 5 8 2 - - - - - PALB2 5 0 - Yes - Yes Yes - SLCO1B3 5 2 - - - - - - SYT15 5 3 - - - - - - XIRP2 5 3 1 - - - - - ZNF266 5 2 - - - - - - ZNF530 5 1 - - - - - - ACACB 4 1 1 - - - - - ALS2CL 4 2 - - - - - - AMER3 4 0 2 - - - - - ANKRD35 4 4 - - - - - - ATP10B 4 1 - - - - - - ATP8B3 4 6 - - - - - - C10orf95 4 0 - - - - - - C2orf88 4 0 - - - - - - C5orf42 4 2 - - - -
    [Show full text]
  • Differential Proteomic Analysis of the Pancreas of Diabetic Db/Db Mice Reveals the Proteins Involved in the Development of Complications of Diabetes Mellitus
    Int. J. Mol. Sci. 2014, 15, 9579-9593; doi:10.3390/ijms15069579 OPEN ACCESS International Journal of Molecular Sciences ISSN 1422-0067 www.mdpi.com/journal/ijms Article Differential Proteomic Analysis of the Pancreas of Diabetic db/db Mice Reveals the Proteins Involved in the Development of Complications of Diabetes Mellitus Victoriano Pérez-Vázquez 1,*, Juan M. Guzmán-Flores 1, Daniela Mares-Álvarez 1, Magdalena Hernández-Ortiz 2, Maciste H. Macías-Cervantes 1, Joel Ramírez-Emiliano 1 and Sergio Encarnación-Guevara 2 1 Depto. de Ciencias Médicas, División de Ciencias de la Salud, Campus León, Universidad de Guanajuato, León, Guanajuato 37320, Mexico; E-Mails: [email protected] (J.M.G.-F.); [email protected] (D.M.-A.); [email protected] (M.H.M.-C.); [email protected] (J.R.-E.) 2 Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62210, Mexico; E-Mails: [email protected] (M.H.-O.); [email protected] (S.E.-G.) * Author to whom correspondence should be addressed; E-Mail: [email protected]; Tel.: +52-477-7143-812; Fax: +52-477-7167-623. Received: 4 April 2014; in revised form: 14 May 2014 / Accepted: 19 May 2014 / Published: 30 May 2014 Abstract: Type 2 diabetes mellitus is characterized by hyperglycemia and insulin-resistance. Diabetes results from pancreatic inability to secrete the insulin needed to overcome this resistance. We analyzed the protein profile from the pancreas of ten-week old diabetic db/db and wild type mice through proteomics. Pancreatic proteins were separated in two-dimensional polyacrylamide gel electrophoresis (2D-PAGE) and significant changes in db/db mice respect to wild type mice were observed in 27 proteins.
    [Show full text]
  • Investigation of Candidate Genes and Mechanisms Underlying Obesity
    Prashanth et al. BMC Endocrine Disorders (2021) 21:80 https://doi.org/10.1186/s12902-021-00718-5 RESEARCH ARTICLE Open Access Investigation of candidate genes and mechanisms underlying obesity associated type 2 diabetes mellitus using bioinformatics analysis and screening of small drug molecules G. Prashanth1 , Basavaraj Vastrad2 , Anandkumar Tengli3 , Chanabasayya Vastrad4* and Iranna Kotturshetti5 Abstract Background: Obesity associated type 2 diabetes mellitus is a metabolic disorder ; however, the etiology of obesity associated type 2 diabetes mellitus remains largely unknown. There is an urgent need to further broaden the understanding of the molecular mechanism associated in obesity associated type 2 diabetes mellitus. Methods: To screen the differentially expressed genes (DEGs) that might play essential roles in obesity associated type 2 diabetes mellitus, the publicly available expression profiling by high throughput sequencing data (GSE143319) was downloaded and screened for DEGs. Then, Gene Ontology (GO) and REACTOME pathway enrichment analysis were performed. The protein - protein interaction network, miRNA - target genes regulatory network and TF-target gene regulatory network were constructed and analyzed for identification of hub and target genes. The hub genes were validated by receiver operating characteristic (ROC) curve analysis and RT- PCR analysis. Finally, a molecular docking study was performed on over expressed proteins to predict the target small drug molecules. Results: A total of 820 DEGs were identified between
    [Show full text]