Supplemental Material.Pdf

Total Page:16

File Type:pdf, Size:1020Kb

Supplemental Material.Pdf Supplemental Table 1: SOX2 qPCR Array ID Gene Symbol Description Assay ID 1 CD44 CD44 antigen (homing function and Indian blood group system) Hs00174139_m1 2 AQP4 aquaporin 4 Hs00242341_m1 3 GFAP glial fibrillary acidic protein Hs00157674_m1 4 ELAVL4 HuD Hs00222634_m1 5 CD68 CD68 antigen Hs00154355_m1 6 ASCL1 MASH1 Hs00269932_m1 7 PROM1 CD133 Hs01009257_m1 8 CD24 CD24A Hs00273561_s1 9 HES1 hairy and enhancer of split 1 Hs00172878_m1 10 PAX6 paired box gene 6 Hs01088112_m1 11 MSI1 musashi homolog 1 Hs00159291_m1 12 NES Nestin Hs00707120_s1 13 SOX2 SRY (sex determining region Y)-box 2 Hs01053049_s1 14 OLIG2 oligodendrocyte lineage transcription factor 2 Hs00377820_m1 15 SOX10 DOM; WS4; MGC15649 Hs00366918_m1 16 PDGFRA platelet-derived growth factor receptor, alpha polypeptide Hs00998026_m1 17 CSPG4 NG2 Hs00426981_m1 18 ST8SIA1 A2B5, SIAT8A, G3 synthase Hs00268157_m1 19 SLC1A3 GLAST Hs00188193_m1 20 FABP7 brain fatty acid binding protein 7, BLBP (Brain lipid-binding protein) Hs00361426_m1 21 TERT hTERT, Telomerase reverse transcriptase Hs00162669_m1 22 P2RY5 purinergic receptor P2Y Hs00271758_s1 23 ADORA2B adenosine A2b receptor Hs00386497_m1 24 NOG Noggin Hs00271352_s1 25 BAMBI NMA Hs00180818_m1 26 TTYH1 tweety homolog 1 (Drosophila) Hs00221061_m1 27 CD99 CD99 molecule Hs00242853_m1 28 CD58 CD58 molecule Hs00156385_m1 29 CD109 CD109 molecule Hs00370347_m1 30 CD164 sialomucin Hs00985280_m1 31 Gli3 Zinc finger protein GLI3 Hs00609233_m1 32 CDH6 cadherin 6, type 2, K-cadherin (fetal kidney) Hs00191832_m1 33 VEGF vascular endothelial growth factor Hs00900055_m1 34 LIFR leukemia inhibitory factor receptor; CD118 Hs00158730_m1 35 IL6ST interleukin 6 signal transducer (gp130, oncostatin M receptor); CD130 Hs00174360_m1 36 CEBPB CCAAT/enhancer binding protein (C/EBP), beta, Nuclear factor NF-IL6 Hs00270923_s1 37 NMI N-myc (and STAT) interactor Hs00190768_m1 38 SPP1 osteopontin Hs00167093_m1 39 TNFSF10 tumor necrosis factor (ligand) superfamily, member 10 Hs00234356_m1 40 TNFSF13B tumor necrosis factor (ligand) superfamily, member 13b Hs00198106_m1 41 GDF15 growth differentiation factor 15 Hs00171132_m1 42 SHC1 SHC (Src homology 2 domain containing) transforming protein 1 Hs00427539_m1 43 EDG2 endothelial differentiation, lysophosphatidic acid G-protein-coupled receptor, 2 Hs00173500_m1 44 HES5 hairy and enhancer of split 5 (Drosophila) Hs01387463_g1 45 NOTCH2 Notch homolog 2 (Drosophila) Hs01050720_mH 46 JAG1 jagged1 Hs00164982_m1 47 NOTCH1 Notch homolog 1 Hs01062014_m1 48 NOTCH3 Notch homolog 3 (Drosophila) Hs01128541_m1 49 Hey1 HARP Hs00232618_m1 50 NEUROG1 neurogenin 1, NGN1 Hs01029249_s1 51 NEUROG2 neurogenin 2, NGN2 Hs00702774_s1 52 DLL1 delta-like 1 (Drosophila) Hs00194509_m1 53 NEUROD2 neurogenic differentiation 2 Hs00272055_s1 54 NEUROD1 neurogenic differentiation 1 Hs00159598_m1 55 NUMB numb homolog (Drosophila) Hs01105433_m1 56 HAP1 huntingtin-associated protein 1 (neuroan 1) Hs00171598_m1 57 SOX4 SRY (sex determining region Y)-box 4 Hs00268388_s1 58 ZIC1 Zic family member 1 (odd-paired homolog, Drosophila) Hs00602749_m1 59 WEE1 WEE1 homolog, WEE1A Hs00268721_m1 60 RREB1 ras responsive element binding protein 1 Hs00366111_m1 61 WWTR1 WW domain containing transcription regulator 1 (TAZ) Hs00210007_m1 62 DACH1 dachshund homolog 1 (Drosophila) Hs00189301_m1 63 YAP1 Yes-associated protein 1, 65kDa Hs00371735_m1 64 SALL1 sal-like 1 (Drosophila) Hs00231307_m1 65 ELAVL1 Hu antigen R Hs00171309_m1 66 BTG2 BTG family, member 2; NGF-inducible anti-proliferative protein PC3, Hs00198887_m1 67 LMCD1 LIM and cysteine-rich domains 1 Hs00205871_m1 68 FGF2 basic fibroblast growth factor Hs00266645_m1 69 EGF epidermal growth factor (beta-urogastrone) Hs00153181_m1 70 NPY2R neuropeptide Y receptor Y2 Hs00168567_m1 71 WNT5A wingless-type MMTV integration site family, member 5A Hs00998537_m1 72 DAAM2 dishevelled associated activator of morphogenesis 2 Hs00322497_m1 73 LY96 lymphocyte antigen 96 Hs00209771_m1 74 AGTR1 angiotensin II receptor, type 1 Hs00258937_m1 75 FAS Fas (TNF receptor superfamily, member 6); CD95 Hs00163653_m1 76 MET met proto-oncogene (hepatocyte growth factor receptor) Hs00179845_m1 77 IL13RA2 interleukin 13 receptor, alpha 2 Hs00152924_m1 78 ITGA2 integrin, alpha 2 (CD49B, alpha 2 subunit of VLA-2 receptor) Hs00158127_m1 79 TLR3 toll-like receptor 3; CD283 antigen Hs00152933_m1 80 TLR4 toll-like receptor 4; CD284 Hs00152939_m1 81 TNFRSF10D tumor necrosis factor receptor superfamily, member 10d;CD264, TRAILR4 Hs00388742_m1 82 TNFRSF10A tumor necrosis factor receptor superfamily, member 10a; CD261, TRAILR-1 Hs00269492_m1 83 TNFRSF19 tumor necrosis factor receptor superfamily, member 19; TRADE, TROY Hs00218634_m1 84 EPHA2 ephrin receptor EPH receptor A2 Hs00171656_m1 85 PTPRG protein tyrosine phosphatase, receptor type, G Hs00177193_m1 86 RGS16 regulator of G-protein signalling 16 Hs00892674_m1 87 NGFR nerve growth factor receptor; p75(NTR), CD271 Hs00609976_m1 88 NRP2 neuropilin 2; NPN2 Hs00187290_m1 89 PTPRZ1 RPTPzeta / phosphocan Hs00267839_m1 90 ENPP2 ectonucleotide pyrophosphatase/phosphodiesterase2 (autotaxin) Hs00905125_m1 91 FZD8 frizzled homolog 8 (Drosophila) Hs00259040_s1 92 FZD10 frizzled homolog 10 (Drosophila) Hs00273077_s1 93 FZD6 frizzled homolog 6 (Drosophila) Hs00171574_m1 94 DKK3 dickkopf homolog 3 (Xenopus laevis) Hs00247426_m1 95 DKK1 dickkopf 1 Hs00183740_m1 Type Symbol Description GeneID Ratio P‐value Ligand GPNMB glycoprotein (transmembrane) nmb 10457 24.01 4.40E‐03 S100A6 S100 calcium binding protein A6 6277 19.25 2.42E‐02 AGTR1 angiotensin II receptor, type 1 185 18.37 1.17E‐04 NOG Noggin 9241 16.59 3.75E‐03 ADM adrenomedullin 133 13.08 1.92E‐03 SPP1 osteopontin 6696 11.89 9.21E‐04 TNFSF10 tumor necrosis factor (ligand) superfamily, 8743 11.73 7.93E‐05 member 10 ANXA1 annexin A1 301 11.68 9.37E‐04 EGF epidermal growth factor (beta‐urogastrone) 1950 9.89 1.43E‐04 DKK3 dickkopf 3 27122 9.00 1.34E‐05 TNFSF13B tumor necrosis factor (ligand) superfamily, 10673 8.80 1.96E‐02 member 13b GDF15 growth differentiation factor 15 9518 8.48 5.98E‐03 CLU clusterin 1191 8.26 4.14E‐04 IL33 interleukin 33 90865 8.07 8.17E‐05 LAMA5 laminin, alpha 5 3911 7.24 5.81E‐03 SHC1 SHC (Src homology 2 domain containing) 6464 6.86 8.44E‐04 transforming protein 1 CMTM3 CKLF‐like MARVEL transmembrane domain 123920 6.51 1.07E‐04 containing 3 CD276 CD276 molecule 80381 6.19 4.77E‐04 LRDD leucine‐rich repeats and death domain 55367 5.95 9.99E‐04 containing FST follistatin 10468 5.46 9.63E‐05 TNFSF4 tumor necrosis factor (ligand) superfamily, 7292 5.45 1.12E‐02 member 4 CRH corticotropin releasing hormone 1392 5.34 2.40E‐02 EPHB4 EPH receptor B4 2050 5.33 1.84E‐03 TGFB2 transforming growth factor, beta 2 7042 5.32 5.03E‐03 GRN granulin 2896 5.20 3.90E‐04 PHIP pleckstrin homology domain interacting 55023 5.14 7.34E‐03 protein ITGA5 integrin, alpha 5 (fibronectin receptor, alpha 3678 5.01 2.04E‐03 polypeptide) ANGPTL2 angiopoietin‐like 2 23452 4.74 3.13E‐03 ADAM9 ADAM metallopeptidase domain 9 (meltrin 8754 4.36 1.64E‐03 gamma) APOL2 apolipoprotein L, 2 23780 4.35 2.51E‐03 MSN moesin 4478 4.34 8.69E‐03 TNC tenascin C 3371 4.27 5.52E‐04 SQSTM1 sequestosome 1 8878 4.26 5.71E‐04 SERPINE1 serpin peptidase inhibitor, clade E (nexin, 5054 4.18 1.36E‐03 plasminogen activator inhibitor type 1), member 1 ANGPTL1 angiopoietin‐like 1 9068 4.15 5.62E‐03 Receptor ELTD1 EGF, latrophilin and seven transmembrane 64123 33.92 3.94E‐03 domain containing 1 FZD10 frizzled 10 11211 33.20 9.80E‐06 LY96 lymphocyte antigen 96 23643 24.07 2.17E‐02 CFI complement factor I 3426 20.74 1.54E‐02 F2RL2 coagulation factor II (thrombin) receptor‐like 2151 18.93 1.48E‐03 2 Type Symbol Description GeneID Ratio P‐value TRPM3 transient receptor potential cation channel, 80036 18.92 4.68E‐05 subfamily M, member 3 CD44 CD44 antigen (homing function and Indian 960 16.38 1.77E‐05 blood group system) EDNRB endothelin receptor type B 1910 14.01 1.60E‐06 IL13RA2 interleukin 13 receptor, alpha 2 3598 13.99 1.02E‐03 MET met proto‐oncogene (hepatocyte growth 4233 13.00 2.61E‐03 factor receptor) ITGA2 integrin, alpha 2 (CD49B, alpha 2 subunit of 3673 12.95 2.57E‐04 VLA‐2 receptor) NPY2R neuropeptide Y receptor Y2 4887 12.64 5.37E‐04 TNFRSF10D tumor necrosis factor receptor superfamily, 8793 12.07 1.62E‐03 member 10d, decoy with truncated death domain LRP10 low density lipoprotein receptor‐related 26020 11.92 4.98E‐03 protein 10 GEM GTP binding protein overexpressed in 2669 11.84 5.07E‐05 skeletal muscle FAS tumor necrosis factor receptor superfamily, 355 10.84 7.30E‐06 member 6 TLR4 toll‐like receptor 4 7099 10.40 3.84E‐05 TLR3 toll‐like receptor 3 7098 10.30 1.22E‐04 FBN1 fibrillin 1 2200 9.64 9.73E‐05 WEE1 WEE1 homolog (S. pombe) 7465 9.38 3.63E‐04 DAAM2 dishevelled associated activator of 23500 8.83 2.92E‐04 morphogenesis 2 NOTCH2 Notch homolog 2 (Drosophila) 4853 8.53 4.67E‐03 GNG12 guanine nucleotide binding protein (G 55970 8.13 1.62E‐03 protein), gamma 12 BAMBI NMA 25805 8.06 2.44E‐03 HRH1 histamine receptor H1 3269 8.02 9.92E‐04 LGALS3BP lectin, galactoside‐binding, soluble, 3 binding 3959 7.85 7.49E‐05 protein LTBP1 latent transforming growth factor beta 4052 7.69 1.19E‐03 binding protein 1 HLA‐E major histocompatibility complex, class I, E 3133 7.24 2.86E‐04 TNFRSF19 tumor necrosis factor receptor superfamily, 55504 7.20 3.36E‐06 member 19 EPHA2 ephrin receptor EphA2 1969 7.17 6.05E‐05 CLIC1 chloride intracellular channel 1 1192 7.01 9.97E‐04 LIFR leukemia inhibitory factor receptor 3977 6.74 2.89E‐04 NGFR p75(NTR) 4804 6.42 2.17E‐03 IL6ST interleukin 6 signal transducer
Recommended publications
  • Genetic Susceptibility for Cow's Milk Allergy in Dutch Children
    Henneman et al. Clin Transl Allergy (2016) 6:7 DOI 10.1186/s13601-016-0096-9 Clinical and Translational Allergy RESEARCH Open Access Genetic susceptibility for cow’s milk allergy in Dutch children: the start of the allergic march? Peter Henneman1*†, Nicole C. M. Petrus2†, Andrea Venema1, Femke van Sinderen1, Karin van der Lip1, Raoul C. Hennekam1, Marcel Mannens1† and Aline B. Sprikkelman2† Abstract Background: Cow’s milk allergy (CMA) is the most common allergic disease in infancy. It is not clear, whether infants with CMA have an increased risk of developing other allergic diseases later in life, the so-called “allergic march”. We aimed to detect genetic associations of CMA using reported single nucleotide polymorphisms (SNP) in other allergic diseases and genetic mutations within the filaggrin (FLG) gene. Both to investigate possible causes of CMA, which also suggests an “allergic march”. Methods: Thirty children from the Dutch EuroPrevall birth cohort study with CMA in infancy and twenty-three healthy controls were studied. Six candidate SNPs were selected (minor allele frequency 10–50 % combined with a large effect) based on the literature. Thirteen FLG candidate mutations were selected spread over repeats 1, 3, 4, 5, 6, 7, 9 and 10 respectively. Results: We found two SNP’s, rs17616434 (P 0.002) and rs2069772 (P 0.038), significantly associated with CMA. One is located near the toll like receptor 6 (TLR6)= gene, which functionally= interacts with toll-like receptor 2, and is associated with an increased risk of other allergic diseases. One is located at the Interleukin 2 (IL2) locus. Twelve FLG amplicons were analyzed, but showed no significant enrichment.
    [Show full text]
  • Lysophosphatidic Acid and Its Receptors: Pharmacology and Therapeutic Potential in Atherosclerosis and Vascular Disease
    JPT-107404; No of Pages 13 Pharmacology & Therapeutics xxx (2019) xxx Contents lists available at ScienceDirect Pharmacology & Therapeutics journal homepage: www.elsevier.com/locate/pharmthera Lysophosphatidic acid and its receptors: pharmacology and therapeutic potential in atherosclerosis and vascular disease Ying Zhou a, Peter J. Little a,b, Hang T. Ta a,c, Suowen Xu d, Danielle Kamato a,b,⁎ a School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD 4102, Australia b Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou 510520, China c Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, St Lucia, QLD 4072, Australia d Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA article info abstract Available online xxxx Lysophosphatidic acid (LPA) is a collective name for a set of bioactive lipid species. Via six widely distributed G protein-coupled receptors (GPCRs), LPA elicits a plethora of biological responses, contributing to inflammation, Keywords: thrombosis and atherosclerosis. There have recently been considerable advances in GPCR signaling especially Lysophosphatidic acid recognition of the extended role for GPCR transactivation of tyrosine and serine/threonine kinase growth factor G-protein coupled receptors receptors. This review covers LPA signaling pathways in the light of new information. The use of transgenic and Atherosclerosis gene knockout animals, gene manipulated cells, pharmacological LPA receptor agonists and antagonists have Gproteins fi β-arrestins provided many insights into the biological signi cance of LPA and individual LPA receptors in the progression Transactivation of atherosclerosis and vascular diseases.
    [Show full text]
  • Supplemental Table S1
    Entrez Gene Symbol Gene Name Affymetrix EST Glomchip SAGE Stanford Literature HPA confirmed Gene ID Profiling profiling Profiling Profiling array profiling confirmed 1 2 A2M alpha-2-macroglobulin 0 0 0 1 0 2 10347 ABCA7 ATP-binding cassette, sub-family A (ABC1), member 7 1 0 0 0 0 3 10350 ABCA9 ATP-binding cassette, sub-family A (ABC1), member 9 1 0 0 0 0 4 10057 ABCC5 ATP-binding cassette, sub-family C (CFTR/MRP), member 5 1 0 0 0 0 5 10060 ABCC9 ATP-binding cassette, sub-family C (CFTR/MRP), member 9 1 0 0 0 0 6 79575 ABHD8 abhydrolase domain containing 8 1 0 0 0 0 7 51225 ABI3 ABI gene family, member 3 1 0 1 0 0 8 29 ABR active BCR-related gene 1 0 0 0 0 9 25841 ABTB2 ankyrin repeat and BTB (POZ) domain containing 2 1 0 1 0 0 10 30 ACAA1 acetyl-Coenzyme A acyltransferase 1 (peroxisomal 3-oxoacyl-Coenzyme A thiol 0 1 0 0 0 11 43 ACHE acetylcholinesterase (Yt blood group) 1 0 0 0 0 12 58 ACTA1 actin, alpha 1, skeletal muscle 0 1 0 0 0 13 60 ACTB actin, beta 01000 1 14 71 ACTG1 actin, gamma 1 0 1 0 0 0 15 81 ACTN4 actinin, alpha 4 0 0 1 1 1 10700177 16 10096 ACTR3 ARP3 actin-related protein 3 homolog (yeast) 0 1 0 0 0 17 94 ACVRL1 activin A receptor type II-like 1 1 0 1 0 0 18 8038 ADAM12 ADAM metallopeptidase domain 12 (meltrin alpha) 1 0 0 0 0 19 8751 ADAM15 ADAM metallopeptidase domain 15 (metargidin) 1 0 0 0 0 20 8728 ADAM19 ADAM metallopeptidase domain 19 (meltrin beta) 1 0 0 0 0 21 81792 ADAMTS12 ADAM metallopeptidase with thrombospondin type 1 motif, 12 1 0 0 0 0 22 9507 ADAMTS4 ADAM metallopeptidase with thrombospondin type 1
    [Show full text]
  • Aquaporin Channels in the Heart—Physiology and Pathophysiology
    International Journal of Molecular Sciences Review Aquaporin Channels in the Heart—Physiology and Pathophysiology Arie O. Verkerk 1,2,* , Elisabeth M. Lodder 2 and Ronald Wilders 1 1 Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; [email protected] 2 Department of Experimental Cardiology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; [email protected] * Correspondence: [email protected]; Tel.: +31-20-5664670 Received: 29 March 2019; Accepted: 23 April 2019; Published: 25 April 2019 Abstract: Mammalian aquaporins (AQPs) are transmembrane channels expressed in a large variety of cells and tissues throughout the body. They are known as water channels, but they also facilitate the transport of small solutes, gasses, and monovalent cations. To date, 13 different AQPs, encoded by the genes AQP0–AQP12, have been identified in mammals, which regulate various important biological functions in kidney, brain, lung, digestive system, eye, and skin. Consequently, dysfunction of AQPs is involved in a wide variety of disorders. AQPs are also present in the heart, even with a specific distribution pattern in cardiomyocytes, but whether their presence is essential for proper (electro)physiological cardiac function has not intensively been studied. This review summarizes recent findings and highlights the involvement of AQPs in normal and pathological cardiac function. We conclude that AQPs are at least implicated in proper cardiac water homeostasis and energy balance as well as heart failure and arsenic cardiotoxicity. However, this review also demonstrates that many effects of cardiac AQPs, especially on excitation-contraction coupling processes, are virtually unexplored.
    [Show full text]
  • Harnessing Gene Expression Profiles for the Identification of Ex Vivo Drug
    cancers Article Harnessing Gene Expression Profiles for the Identification of Ex Vivo Drug Response Genes in Pediatric Acute Myeloid Leukemia David G.J. Cucchi 1 , Costa Bachas 1 , Marry M. van den Heuvel-Eibrink 2,3, Susan T.C.J.M. Arentsen-Peters 3, Zinia J. Kwidama 1, Gerrit J. Schuurhuis 1, Yehuda G. Assaraf 4, Valérie de Haas 3 , Gertjan J.L. Kaspers 3,5 and Jacqueline Cloos 1,* 1 Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; [email protected] (D.G.J.C.); [email protected] (C.B.); [email protected] (Z.J.K.); [email protected] (G.J.S.) 2 Department of Pediatric Oncology/Hematology, Erasmus MC–Sophia Children’s Hospital, 3015 CN Rotterdam, The Netherlands; [email protected] 3 Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; [email protected] (S.T.C.J.M.A.-P.); [email protected] (V.d.H.); [email protected] (G.J.L.K.) 4 The Fred Wyszkowski Cancer Research, Laboratory, Department of Biology, Technion-Israel Institute of Technology, 3200003 Haifa, Israel; [email protected] 5 Emma’s Children’s Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, 1081 HV Amsterdam, The Netherlands * Correspondence: [email protected] Received: 21 April 2020; Accepted: 12 May 2020; Published: 15 May 2020 Abstract: Novel treatment strategies are of paramount importance to improve clinical outcomes in pediatric AML. Since chemotherapy is likely to remain the cornerstone of curative treatment of AML, insights in the molecular mechanisms that determine its cytotoxic effects could aid further treatment optimization.
    [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]
  • 1 Metabolic Dysfunction Is Restricted to the Sciatic Nerve in Experimental
    Page 1 of 255 Diabetes Metabolic dysfunction is restricted to the sciatic nerve in experimental diabetic neuropathy Oliver J. Freeman1,2, Richard D. Unwin2,3, Andrew W. Dowsey2,3, Paul Begley2,3, Sumia Ali1, Katherine A. Hollywood2,3, Nitin Rustogi2,3, Rasmus S. Petersen1, Warwick B. Dunn2,3†, Garth J.S. Cooper2,3,4,5* & Natalie J. Gardiner1* 1 Faculty of Life Sciences, University of Manchester, UK 2 Centre for Advanced Discovery and Experimental Therapeutics (CADET), Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, UK 3 Centre for Endocrinology and Diabetes, Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, UK 4 School of Biological Sciences, University of Auckland, New Zealand 5 Department of Pharmacology, Medical Sciences Division, University of Oxford, UK † Present address: School of Biosciences, University of Birmingham, UK *Joint corresponding authors: Natalie J. Gardiner and Garth J.S. Cooper Email: [email protected]; [email protected] Address: University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, United Kingdom Telephone: +44 161 275 5768; +44 161 701 0240 Word count: 4,490 Number of tables: 1, Number of figures: 6 Running title: Metabolic dysfunction in diabetic neuropathy 1 Diabetes Publish Ahead of Print, published online October 15, 2015 Diabetes Page 2 of 255 Abstract High glucose levels in the peripheral nervous system (PNS) have been implicated in the pathogenesis of diabetic neuropathy (DN). However our understanding of the molecular mechanisms which cause the marked distal pathology is incomplete. Here we performed a comprehensive, system-wide analysis of the PNS of a rodent model of DN.
    [Show full text]
  • Download Download
    Supplementary Figure S1. Results of flow cytometry analysis, performed to estimate CD34 positivity, after immunomagnetic separation in two different experiments. As monoclonal antibody for labeling the sample, the fluorescein isothiocyanate (FITC)- conjugated mouse anti-human CD34 MoAb (Mylteni) was used. Briefly, cell samples were incubated in the presence of the indicated MoAbs, at the proper dilution, in PBS containing 5% FCS and 1% Fc receptor (FcR) blocking reagent (Miltenyi) for 30 min at 4 C. Cells were then washed twice, resuspended with PBS and analyzed by a Coulter Epics XL (Coulter Electronics Inc., Hialeah, FL, USA) flow cytometer. only use Non-commercial 1 Supplementary Table S1. Complete list of the datasets used in this study and their sources. GEO Total samples Geo selected GEO accession of used Platform Reference series in series samples samples GSM142565 GSM142566 GSM142567 GSM142568 GSE6146 HG-U133A 14 8 - GSM142569 GSM142571 GSM142572 GSM142574 GSM51391 GSM51392 GSE2666 HG-U133A 36 4 1 GSM51393 GSM51394 only GSM321583 GSE12803 HG-U133A 20 3 GSM321584 2 GSM321585 use Promyelocytes_1 Promyelocytes_2 Promyelocytes_3 Promyelocytes_4 HG-U133A 8 8 3 GSE64282 Promyelocytes_5 Promyelocytes_6 Promyelocytes_7 Promyelocytes_8 Non-commercial 2 Supplementary Table S2. Chromosomal regions up-regulated in CD34+ samples as identified by the LAP procedure with the two-class statistics coded in the PREDA R package and an FDR threshold of 0.5. Functional enrichment analysis has been performed using DAVID (http://david.abcc.ncifcrf.gov/)
    [Show full text]
  • Supplementary Table 1: Adhesion Genes Data Set
    Supplementary Table 1: Adhesion genes data set PROBE Entrez Gene ID Celera Gene ID Gene_Symbol Gene_Name 160832 1 hCG201364.3 A1BG alpha-1-B glycoprotein 223658 1 hCG201364.3 A1BG alpha-1-B glycoprotein 212988 102 hCG40040.3 ADAM10 ADAM metallopeptidase domain 10 133411 4185 hCG28232.2 ADAM11 ADAM metallopeptidase domain 11 110695 8038 hCG40937.4 ADAM12 ADAM metallopeptidase domain 12 (meltrin alpha) 195222 8038 hCG40937.4 ADAM12 ADAM metallopeptidase domain 12 (meltrin alpha) 165344 8751 hCG20021.3 ADAM15 ADAM metallopeptidase domain 15 (metargidin) 189065 6868 null ADAM17 ADAM metallopeptidase domain 17 (tumor necrosis factor, alpha, converting enzyme) 108119 8728 hCG15398.4 ADAM19 ADAM metallopeptidase domain 19 (meltrin beta) 117763 8748 hCG20675.3 ADAM20 ADAM metallopeptidase domain 20 126448 8747 hCG1785634.2 ADAM21 ADAM metallopeptidase domain 21 208981 8747 hCG1785634.2|hCG2042897 ADAM21 ADAM metallopeptidase domain 21 180903 53616 hCG17212.4 ADAM22 ADAM metallopeptidase domain 22 177272 8745 hCG1811623.1 ADAM23 ADAM metallopeptidase domain 23 102384 10863 hCG1818505.1 ADAM28 ADAM metallopeptidase domain 28 119968 11086 hCG1786734.2 ADAM29 ADAM metallopeptidase domain 29 205542 11085 hCG1997196.1 ADAM30 ADAM metallopeptidase domain 30 148417 80332 hCG39255.4 ADAM33 ADAM metallopeptidase domain 33 140492 8756 hCG1789002.2 ADAM7 ADAM metallopeptidase domain 7 122603 101 hCG1816947.1 ADAM8 ADAM metallopeptidase domain 8 183965 8754 hCG1996391 ADAM9 ADAM metallopeptidase domain 9 (meltrin gamma) 129974 27299 hCG15447.3 ADAMDEC1 ADAM-like,
    [Show full text]
  • CD29 Identifies IFN-Γ–Producing Human CD8+ T Cells With
    + CD29 identifies IFN-γ–producing human CD8 T cells with an increased cytotoxic potential Benoît P. Nicoleta,b, Aurélie Guislaina,b, Floris P. J. van Alphenc, Raquel Gomez-Eerlandd, Ton N. M. Schumacherd, Maartje van den Biggelaarc,e, and Monika C. Wolkersa,b,1 aDepartment of Hematopoiesis, Sanquin Research, 1066 CX Amsterdam, The Netherlands; bLandsteiner Laboratory, Oncode Institute, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; cDepartment of Research Facilities, Sanquin Research, 1066 CX Amsterdam, The Netherlands; dDivision of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; and eDepartment of Molecular and Cellular Haemostasis, Sanquin Research, 1066 CX Amsterdam, The Netherlands Edited by Anjana Rao, La Jolla Institute for Allergy and Immunology, La Jolla, CA, and approved February 12, 2020 (received for review August 12, 2019) Cytotoxic CD8+ T cells can effectively kill target cells by producing therefore developed a protocol that allowed for efficient iso- cytokines, chemokines, and granzymes. Expression of these effector lation of RNA and protein from fluorescence-activated cell molecules is however highly divergent, and tools that identify and sorting (FACS)-sorted fixed T cells after intracellular cytokine + preselect CD8 T cells with a cytotoxic expression profile are lacking. staining. With this top-down approach, we performed an un- + Human CD8 T cells can be divided into IFN-γ– and IL-2–producing biased RNA-sequencing (RNA-seq) and mass spectrometry cells. Unbiased transcriptomics and proteomics analysis on cytokine- γ– – + + (MS) analyses on IFN- and IL-2 producing primary human producing fixed CD8 T cells revealed that IL-2 cells produce helper + + + CD8 Tcells.
    [Show full text]
  • Ck1δ Over-Expressing Mice Display ADHD-Like Behaviors, Frontostriatal Neuronal Abnormalities and Altered Expressions of ADHD-Candidate Genes
    Molecular Psychiatry (2020) 25:3322–3336 https://doi.org/10.1038/s41380-018-0233-z ARTICLE CK1δ over-expressing mice display ADHD-like behaviors, frontostriatal neuronal abnormalities and altered expressions of ADHD-candidate genes 1 1 1 2 1 1 1 Mingming Zhou ● Jodi Gresack ● Jia Cheng ● Kunihiro Uryu ● Lars Brichta ● Paul Greengard ● Marc Flajolet Received: 8 November 2017 / Revised: 4 July 2018 / Accepted: 18 July 2018 / Published online: 19 October 2018 © Springer Nature Limited 2018 Abstract The cognitive mechanisms underlying attention-deficit hyperactivity disorder (ADHD), a highly heritable disorder with an array of candidate genes and unclear genetic architecture, remain poorly understood. We previously demonstrated that mice overexpressing CK1δ (CK1δ OE) in the forebrain show hyperactivity and ADHD-like pharmacological responses to D- amphetamine. Here, we demonstrate that CK1δ OE mice exhibit impaired visual attention and a lack of D-amphetamine- induced place preference, indicating a disruption of the dopamine-dependent reward pathway. We also demonstrate the presence of abnormalities in the frontostriatal circuitry, differences in synaptic ultra-structures by electron microscopy, as 1234567890();,: 1234567890();,: well as electrophysiological perturbations of both glutamatergic and GABAergic transmission, as observed by altered frequency and amplitude of mEPSCs and mIPSCs. Furthermore, gene expression profiling by next-generation sequencing alone, or in combination with bacTRAP technology to study specifically Drd1a versus Drd2 medium spiny neurons, revealed that developmental CK1δ OE alters transcriptional homeostasis in the striatum, including specific alterations in Drd1a versus Drd2 neurons. These results led us to perform a fine molecular characterization of targeted gene networks and pathway analysis. Importantly, a large fraction of 92 genes identified by GWAS studies as associated with ADHD in humans are significantly altered in our mouse model.
    [Show full text]
  • The Caenorhabditis Elegans Excretory System: a Model for Tubulogenesis, Cell Fate Specification, and Plasticity
    | WORMBOOK CELL AND ORGANELLE BIOLOGY The Caenorhabditis elegans Excretory System: A Model for Tubulogenesis, Cell Fate Specification, and Plasticity Meera V. Sundaram*,1 and Matthew Buechner† *Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104 and †Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66045 ABSTRACT The excretory system of the nematode Caenorhabditis elegans is a superb model of tubular organogenesis involving a minimum of cells. The system consists of just three unicellular tubes (canal, duct, and pore), a secretory gland, and two associated neurons. Just as in more complex organs, cells of the excretory system must first adopt specific identities and then coordinate diverse processes to form tubes of appropriate topology, shape, connectivity, and physiological function. The unicellular topology of excretory tubes, their varied and sometimes complex shapes, and the dynamic reprogramming of cell identity and remodeling of tube connec- tivity that occur during larval development are particularly fascinating features of this organ. The physiological roles of the excretory system in osmoregulation and other aspects of the animal’s life cycle are only beginning to be explored. The cellular mechanisms and molecular pathways used to build and shape excretory tubes appear similar to those used in both unicellular and multicellular tubes in more complex organs, such as the vertebrate vascular system and kidney, making this simple organ system a
    [Show full text]