Supplementary Table Paper

Total Page:16

File Type:pdf, Size:1020Kb

Supplementary Table Paper Supplementary Material Metabolic Pathways and Immunometabolism in Rare Kidney Diseases Grayson et al, 2017 Symbol Description Pathway G6PD glucose-6-phosphate dehydrogenase Pentose Phosphate Pathway PGLS 6-phosphogluconolactonase Pentose Phosphate Pathway PGD phosphogluconate dehydrogenase Pentose Phosphate Pathway RPE ribulose-5-phosphate-3-epimerase Pentose Phosphate Pathway RPIA ribose 5-phosphate isomerase A Pentose Phosphate Pathway TKT transketolase Pentose Phosphate Pathway TALDO1 transaldolase 1 Pentose Phosphate Pathway ALDOA aldolase, fructose-bisphosphate A Glycolysis ALDOB aldolase, fructose-bisphosphate B Glycolysis ALDOC aldolase, fructose-bisphosphate C Glycolysis ENO1 enolase 1 Glycolysis ENO2 enolase 2 Glycolysis ENO3 enolase 3 Glycolysis GAPDH glyceraldehyde-3-phosphate dehydrogenase Glycolysis GCK glucokinase Glycolysis GPI glucose-6-phosphate isomerase Glycolysis HK1 hexokinase 1 Glycolysis HK2 hexokinase 2 Glycolysis HK3 hexokinase 3 Glycolysis PFKFB1 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 1 Glycolysis PFKFB2 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 2 Glycolysis PFKFB3 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 Glycolysis PFKFB4 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 4 Glycolysis PFKP phosphofructokinase, platelet Glycolysis PFKM phosphofructokinase, muscle Glycolysis PGAM2 phosphoglycerate mutase 2 Glycolysis PGK1 phosphoglycerate kinase 1 Glycolysis PGK2 phosphoglycerate kinase 2 Glycolysis PGM1 phosphoglucomutase 1 Glycolysis PGM3 phosphoglucomutase 3 Glycolysis PKLR pyruvate kinase, liver and RBC Glycolysis PKM pyruvate kinase, muscle Glycolysis SOD1 superoxide dismutase 1, soluble Glycolysis SOD2 superoxide dismutase 2, mitochondrial Glycolysis SOD3 superoxide dismutase 3, extracellular Glycolysis TPI1 triosephosphate isomerase 1 Glycolysis SLC2A1 solute carrier family 2 member 1 glucose transporters SLC2A2 solute carrier family 2 member 2 glucose transporters SLC2A3 solute carrier family 2 member 3 glucose transporters SLC2A4 solute carrier family 2 member 4 glucose transporters ACLY ATP citrate lyase TCA cycle ACO1 aconitase 1 TCA cycle ACO2 aconitase 2 TCA cycle DLAT dihydrolipoamide S-acetyltransferase TCA cycle DLD dihydrolipoamide dehydrogenase TCA cycle DLST dihydrolipoamide S-succinyltransferase TCA cycle FH fumarate hydratase TCA cycle IDH1 isocitrate dehydrogenase (NADP(+)) 1, cytosolic TCA cycle IDH2 isocitrate dehydrogenase (NADP(+)) 2, mitochondrial TCA cycle IDH3A isocitrate dehydrogenase 3 (NAD(+)) alpha TCA cycle IDH3B isocitrate dehydrogenase 3 (NAD(+)) beta TCA cycle IDH3G isocitrate dehydrogenase 3 (NAD(+)) gamma TCA cycle MDH1 malate dehydrogenase 1 TCA cycle MDH2 malate dehydrogenase 2 TCA cycle OGDHL oxoglutarate dehydrogenase-like TCA cycle OGDH oxoglutarate dehydrogenase TCA cycle PC pyruvate carboxylase TCA cycle PCK1 phosphoenolpyruvate carboxykinase 1 TCA cycle PCK2 phosphoenolpyruvate carboxykinase 2, mitochondrial TCA cycle PDHA1 pyruvate dehydrogenase (lipoamide) alpha 1 TCA cycle PDHA2 pyruvate dehydrogenase (lipoamide) alpha 2 TCA cycle PDHB pyruvate dehydrogenase (lipoamide) beta TCA cycle PDK1 pyruvate dehydrogenase kinase 1 TCA cycle PDK2 pyruvate dehydrogenase kinase 2 TCA cycle PDK3 pyruvate dehydrogenase kinase 3 TCA cycle PDK4 pyruvate dehydrogenase kinase 4 TCA cycle PDPR pyruvate dehydrogenase phosphatase regulatory subunit TCA cycle SDHAF1 succinate dehydrogenase complex assembly factor 1 TCA cycle SDHAF3 succinate dehydrogenase complex assembly factor 3 TCA cycle SDHB succinate dehydrogenase complex iron sulfur subunit B TCA cycle SDHD succinate dehydrogenase complex subunit D TCA cycle SUCLA2 succinate-CoA ligase ADP-forming beta subunit TCA cycle SUCLG1 succinate-CoA ligase alpha subunit TCA cycle SUCLG2 succinate-CoA ligase GDP-forming beta subunit TCA cycle GLS glutaminase Glutaminolysis GLS2 glutaminase 2 Glutaminolysis GLUD1 glutamate dehydrogenase 1 Glutaminolysis GLUD2 glutamate dehydrogenase 2 Glutaminolysis OGDH oxoglutarate dehydrogenase Glutaminolysis GOT1 glutamic-oxaloacetic transaminase 1 Glutaminolysis ACSL3 acyl-CoA synthetase long-chain family member 3 Fatty Acid Oxidation CPT1A carnitine palmitoyltransferase 1A Fatty Acid Oxidation SLC25A20 solute carrier family 25 member 20 Fatty Acid Oxidation CPT2 carnitine palmitoyltransferase 2 Fatty Acid Oxidation ACADM acyl-CoA dehydrogenase, C-4 to C-12 straight chain Fatty Acid Oxidation ECHS1 enoyl-CoA hydratase, short chain, 1, mitochondrial Fatty Acid Oxidation HADH hydroxyacyl-CoA dehydrogenase Fatty Acid Oxidation HADHB hydroxyacyl-CoA dehydrogenase/3-ketoacyl-CoA thiolase/enoyl-CoAFatty Acid hydratase Oxidation (trifunctional protein), beta subunit CD3D CD3d molecule Immune Subset Markers CD4 CD4 molecule Immune Subset Markers CD8A CD8a molecule Immune Subset Markers CCR5 C-C motif chemokine receptor 5 (gene/pseudogene) Immune Subset Markers CXCR3 C-X-C motif chemokine receptor 3 Immune Subset Markers IFNG interferon, gamma Immune Subset Markers CCR3 C-C motif chemokine receptor 3 Immune Subset Markers IL13 interleukin 13 Immune Subset Markers CCR6 C-C motif chemokine receptor 6 Immune Subset Markers CCR4 C-C motif chemokine receptor 4 Immune Subset Markers IL17B interleukin 17B Immune Subset Markers IL21 interleukin 21 Immune Subset Markers IL23A interleukin 23 subunit alpha Immune Subset Markers FOXP3 forkhead box P3 Immune Subset Markers IL10 interleukin 10 Immune Subset Markers CD19 CD19 molecule Immune Subset Markers SDC1 syndecan 1 Immune Subset Markers CD14 CD14 molecule Immune Subset Markers CD68 CD68 molecule Immune Subset Markers CD163 CD163 molecule Immune Subset Markers IL1B interleukin 1 beta Immune Subset Markers IL1A interleukin 1 alpha Immune Subset Markers TNF tumor necrosis factor Immune Subset Markers IL6 interleukin 6 Immune Subset Markers MPO myeloperoxidase Immune Subset Markers.
Recommended publications
  • Impact of Biopersistent Fibrous Dusts on Glycolysis, Glutaminolysis and Serine Metabolism in A549 Cells
    MOLECULAR MEDICINE REPORTS 16: 9233-9241, 2017 Impact of biopersistent fibrous dusts on glycolysis, glutaminolysis and serine metabolism in A549 cells SYBILLE WACHE1, SIMONE HELMIG2, DIRK WALTER2, JOACHIM SCHNEIDER2 and SYBILLE MAZUREK1 1Institute of Veterinary Physiology and Biochemistry; 2Institute and Outpatient Clinic for Occupational and Social Medicine, Justus Liebig University Giessen, D-35392 Giessen, Germany Received March 29, 2017; Accepted August 15, 2017 DOI: 10.3892/mmr.2017.7729 Abstract. The conversion rates of different metabolic examples of which are glass fibers [man‑made vitreous fibers pathways summarized as a metabolic signature mirror the (MMVF)], ceramic fibers (including refractory ceramic physiological functions and the general physiological status of fibers) and, more recently, carbon nanotubes. Epidemiological a cell. The present study compared the impact of crocidolite studies have confirmed an increased risk of lung carcinoma and chrysotile asbestos, glass fibers and multi‑walled carbon and mesothelioma following exposure to asbestos (2,3). Due nanotubes (MWCN) of two different lengths (1-2 µm and to their geological formations, asbestos fibers vary in chemical 5-15 µm) on the conversion rates in glycolysis, glutaminolysis composition, length and diameter. Inhalable asbestos fibers of and serine metabolism of A549 cells. The concentration tested critical dimensions were defined as World Health Organization was 1 µg/cm2 for all fibers. A concentration of 5 µg/cm2 was (WHO; Geneva, Switzerland) fibers: Length ≥5 µm, diameter additionally used for chrysotile and crocidolite, and 25 µg/cm2 <3 µm, length:diameter ratio >3:1 (4). However, this conven- for glass fibers and MWCN. With respect to the inhibitory tion is not a robust criterion by which to categorize fibers as effect on cell proliferation and the extent of metabolic altera- toxic.
    [Show full text]
  • HK3 Overexpression Associated with Epithelial-Mesenchymal Transition in Colorectal Cancer Elena A
    Pudova et al. BMC Genomics 2018, 19(Suppl 3):113 DOI 10.1186/s12864-018-4477-4 RESEARCH Open Access HK3 overexpression associated with epithelial-mesenchymal transition in colorectal cancer Elena A. Pudova1†, Anna V. Kudryavtseva1,2†, Maria S. Fedorova1, Andrew R. Zaretsky3, Dmitry S. Shcherbo3, Elena N. Lukyanova1,4, Anatoly Y. Popov5, Asiya F. Sadritdinova1, Ivan S. Abramov1, Sergey L. Kharitonov1, George S. Krasnov1, Kseniya M. Klimina4, Nadezhda V. Koroban2, Nadezhda N. Volchenko2, Kirill M. Nyushko2, Nataliya V. Melnikova1, Maria A. Chernichenko2, Dmitry V. Sidorov2, Boris Y. Alekseev2, Marina V. Kiseleva2, Andrey D. Kaprin2, Alexey A. Dmitriev1 and Anastasiya V. Snezhkina1* From Belyaev Conference Novosibirsk, Russia. 07-10 August 2017 Abstract Background: Colorectal cancer (CRC) is a common cancer worldwide. The main cause of death in CRC includes tumor progression and metastasis. At molecular level, these processes may be triggered by epithelial-mesenchymal transition (EMT) and necessitates specific alterations in cell metabolism. Although several EMT-related metabolic changes have been described in CRC, the mechanism is still poorly understood. Results: Using CrossHub software, we analyzed RNA-Seq expression profile data of CRC derived from The Cancer Genome Atlas (TCGA) project. Correlation analysis between the change in the expression of genes involved in glycolysis and EMT was performed. We obtained the set of genes with significant correlation coefficients, which included 21 EMT-related genes and a single glycolytic gene, HK3. The mRNA level of these genes was measured in 78 paired colorectal cancer samples by quantitative polymerase chain reaction (qPCR). Upregulation of HK3 and deregulation of 11 genes (COL1A1, TWIST1, NFATC1, GLIPR2, SFPR1, FLNA, GREM1, SFRP2, ZEB2, SPP1, and RARRES1) involved in EMT were found.
    [Show full text]
  • Effect of Citric Acid Cycle Genetic Variants and Their Interactions With
    cancers Article Effect of Citric Acid Cycle Genetic Variants and Their Interactions with Obesity, Physical Activity and Energy Intake on the Risk of Colorectal Cancer: Results from a Nested Case-Control Study in the UK Biobank Sooyoung Cho 1 , Nan Song 2,3 , Ji-Yeob Choi 2,4,5 and Aesun Shin 1,2,* 1 Department of Preventive Medicine, Seoul National University College of Medicine, Seoul 03080, Korea; [email protected] 2 Cancer Research Institute, Seoul National University, Seoul 03080, Korea; [email protected] (N.S.); [email protected] (J.-Y.C.) 3 Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA 4 Department of Biomedical Sciences, Graduate School of Seoul National University, Seoul 03080, Korea 5 Medical Research Center, Institute of Health Policy and Management, Seoul National University, Seoul 03080, Korea * Correspondence: [email protected]; Tel.: +82-2-740-8331; Fax: +82-2-747-4830 Received: 18 August 2020; Accepted: 9 October 2020; Published: 12 October 2020 Simple Summary: The citric acid cycle has a central role in the cellular energy metabolism and biosynthesis of macromolecules in the mitochondrial matrix. We identified the single nucleotide polymorphisms (SNPs) of the citrate acid cycle with colorectal cancer susceptibility in UK population. Furthermore, we found the significant interaction of SNPs in the citric acid cycle with the contributors to energy balance and SNP-SNP interactions. Our findings provide clues to the etiology in cancer development related to energy metabolism and evidence on identification of the population at high risk of colorectal cancer.
    [Show full text]
  • Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model
    Downloaded from http://www.jimmunol.org/ by guest on September 25, 2021 T + is online at: average * The Journal of Immunology , 34 of which you can access for free at: 2016; 197:1477-1488; Prepublished online 1 July from submission to initial decision 4 weeks from acceptance to publication 2016; doi: 10.4049/jimmunol.1600589 http://www.jimmunol.org/content/197/4/1477 Molecular Profile of Tumor-Specific CD8 Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A. Waugh, Sonia M. Leach, Brandon L. Moore, Tullia C. Bruno, Jonathan D. Buhrman and Jill E. Slansky J Immunol cites 95 articles Submit online. Every submission reviewed by practicing scientists ? is published twice each month by Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts http://jimmunol.org/subscription Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html http://www.jimmunol.org/content/suppl/2016/07/01/jimmunol.160058 9.DCSupplemental This article http://www.jimmunol.org/content/197/4/1477.full#ref-list-1 Information about subscribing to The JI No Triage! Fast Publication! Rapid Reviews! 30 days* Why • • • Material References Permissions Email Alerts Subscription Supplementary The Journal of Immunology The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2016 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. This information is current as of September 25, 2021. The Journal of Immunology Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A.
    [Show full text]
  • Class-I and Class-II Fumarases Are a Paradigm of the Recruitment Of
    bioRxiv preprint doi: https://doi.org/10.1101/2020.08.04.232652; this version posted August 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 1 Class-I and Class-II fumarases are a paradigm of the recruitment of 2 metabolites and metabolic enzymes for signalling of the DNA Damage 3 Response during evolution. 4 5 Yardena Silas 1, 2, Esti Singer 1, Norbert Lehming 2 and Ophry Pines 1, 2* 6 1. Department of Microbiology and Molecular Genetics, IMRIC, Faculty of Medicine, 7 Hebrew University, Jerusalem, Israel 8 2. CREATE‑NUS‑HUJ Program and the Department of Microbiology and Immunology, 9 Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 10 Singapore. 11 12 [email protected] 13 [email protected] 14 [email protected] 15 [email protected] 16 17 18 19 20 21 22 23 24 25 26 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.08.04.232652; this version posted August 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 27 Abstract 28 Class-II fumarase (Fumarate Hydratase, FH) and its metabolic intermediates are essential 29 components in the DNA damage response (DDR) in eukaryotic cells (human and yeast) and 30 in the prokaryote Bacillus subtilis.
    [Show full text]
  • Interplay Between Epigenetics and Metabolism in Oncogenesis: Mechanisms and Therapeutic Approaches
    OPEN Oncogene (2017) 36, 3359–3374 www.nature.com/onc REVIEW Interplay between epigenetics and metabolism in oncogenesis: mechanisms and therapeutic approaches CC Wong1, Y Qian2,3 and J Yu1 Epigenetic and metabolic alterations in cancer cells are highly intertwined. Oncogene-driven metabolic rewiring modifies the epigenetic landscape via modulating the activities of DNA and histone modification enzymes at the metabolite level. Conversely, epigenetic mechanisms regulate the expression of metabolic genes, thereby altering the metabolome. Epigenetic-metabolomic interplay has a critical role in tumourigenesis by coordinately sustaining cell proliferation, metastasis and pluripotency. Understanding the link between epigenetics and metabolism could unravel novel molecular targets, whose intervention may lead to improvements in cancer treatment. In this review, we summarized the recent discoveries linking epigenetics and metabolism and their underlying roles in tumorigenesis; and highlighted the promising molecular targets, with an update on the development of small molecule or biologic inhibitors against these abnormalities in cancer. Oncogene (2017) 36, 3359–3374; doi:10.1038/onc.2016.485; published online 16 January 2017 INTRODUCTION metabolic genes have also been identified as driver genes It has been appreciated since the early days of cancer research mutated in some cancers, such as isocitrate dehydrogenase 1 16 17 that the metabolic profiles of tumor cells differ significantly from and 2 (IDH1/2) in gliomas and acute myeloid leukemia (AML), 18 normal cells. Cancer cells have high metabolic demands and they succinate dehydrogenase (SDH) in paragangliomas and fuma- utilize nutrients with an altered metabolic program to support rate hydratase (FH) in hereditary leiomyomatosis and renal cell 19 their high proliferative rates and adapt to the hostile tumor cancer (HLRCC).
    [Show full text]
  • Supplementary Materials
    1 Supplementary Materials: Supplemental Figure 1. Gene expression profiles of kidneys in the Fcgr2b-/- and Fcgr2b-/-. Stinggt/gt mice. (A) A heat map of microarray data show the genes that significantly changed up to 2 fold compared between Fcgr2b-/- and Fcgr2b-/-. Stinggt/gt mice (N=4 mice per group; p<0.05). Data show in log2 (sample/wild-type). 2 Supplemental Figure 2. Sting signaling is essential for immuno-phenotypes of the Fcgr2b-/-lupus mice. (A-C) Flow cytometry analysis of splenocytes isolated from wild-type, Fcgr2b-/- and Fcgr2b-/-. Stinggt/gt mice at the age of 6-7 months (N= 13-14 per group). Data shown in the percentage of (A) CD4+ ICOS+ cells, (B) B220+ I-Ab+ cells and (C) CD138+ cells. Data show as mean ± SEM (*p < 0.05, **p<0.01 and ***p<0.001). 3 Supplemental Figure 3. Phenotypes of Sting activated dendritic cells. (A) Representative of western blot analysis from immunoprecipitation with Sting of Fcgr2b-/- mice (N= 4). The band was shown in STING protein of activated BMDC with DMXAA at 0, 3 and 6 hr. and phosphorylation of STING at Ser357. (B) Mass spectra of phosphorylation of STING at Ser357 of activated BMDC from Fcgr2b-/- mice after stimulated with DMXAA for 3 hour and followed by immunoprecipitation with STING. (C) Sting-activated BMDC were co-cultured with LYN inhibitor PP2 and analyzed by flow cytometry, which showed the mean fluorescence intensity (MFI) of IAb expressing DC (N = 3 mice per group). 4 Supplemental Table 1. Lists of up and down of regulated proteins Accession No.
    [Show full text]
  • Interaction of 6-Phosphofructo-2-Kinase/Fructose-2,6- Bisphosphatase (PFK-2/Fbpase-2) with Glucokinase Activates Glucose Phospho
    Interaction of 6-Phosphofructo-2-Kinase/Fructose-2,6- Bisphosphatase (PFK-2/FBPase-2) With Glucokinase Activates Glucose Phosphorylation and Glucose Metabolism in Insulin-Producing Cells Laura Massa,1 Simone Baltrusch,1 David A. Okar,2,3 Alex J. Lange,2 Sigurd Lenzen,1 and Markus Tiedge1 The bifunctional enzyme 6-phosphofructo-2-kinase/ fructose-2,6-bisphosphatase (PFK-2/FBPase-2) was re- cently identified as a new intracellular binding partner he enzyme glucokinase (GK) plays a pivotal role for glucokinase (GK). Therefore, we studied the impor- in the recognition of glucose in pancreatic tance of this interaction for the activity status of GK ␤-cells and the regulation of glucose metabolism and glucose metabolism in insulin-producing cells by Tin the liver (1–7). In pancreatic ␤-cells, GK acts overexpression of the rat liver and pancreatic islet as a glucose sensor and catalyzes the rate-limiting step for isoforms of PFK-2/FBPase-2. PFK-2/FBPase-2 overex- initiation of glucose-induced insulin secretion (6). GK is pression in RINm5F-GK cells significantly increased the regulated in a complex manner in pancreatic ␤-cells by GK activity by 78% in cells expressing the islet isoform, posttranslational modifications of the enzyme protein that by 130% in cells expressing the liver isoform, and by mainly depend on the intracellular glucose concentration 116% in cells expressing a cAMP-insensitive liver S32A/ (8–13). These posttranslational mechanisms of GK activity H258A double mutant isoform. Only in cells overex- regulation are comprised of conformational changes pressing the wild-type liver PFK-2/FBPase-2 isoform (14,15), sulfhydryl-group conversions (16–18), and inter- was the increase of GK activity abolished by forskolin, ␤ apparently due to the regulatory site for phosphoryla- actions with -cell matrix proteins (13,19), insulin gran- tion by a cAMP-dependent protein kinase.
    [Show full text]
  • Multi-Omic Analysis of Hibernator Skeletal Muscle and Regulation of Calcium Handling
    Multi-omic Analysis of Hibernator Skeletal Muscle and Regulation of Calcium Handling A Thesis SUBMITTED TO THE FACULTY OF UNIVERSITY OF MINNESOTA BY Kyle J. Anderson IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF MASTER OF SCIENCE Advisor: Matthew T. Andrews May 2016 © Kyle J. Anderson 2016 Acknowledgements I would like to first thank my family and friends for their guidance and support through my graduate career. I would also like to thank my thesis committee Dr. Andrews, Dr. Hampton, and Dr. Liang for their guidance and assistance throughout these projects. None of this work would have been possible without the financial support I received from the Biology department through my GTA appointments. Additional financial and scientific support to complete the proteomics project came from Tim Griffin and everyone at the Center for Mass Spectrometry and Proteomics at the University of Minnesota and was greatly appreciated. This work was funded by the United States Army Medical Research and Materiel Command contract W81XWH-11-0409, the University of Minnesota McKnight Presidential Endowment, and NIH grant 1RC2HL101625-01 to M.T.A. Additional funding came from the NSF grant 1147079 for the Galaxy-P team. i Abstract Mammalian hibernation is a strategy employed by many species to survive fluctuations in resource availability and environmental conditions. Hibernating mammals endure conditions of dramatically depressed heart rate, body temperature, and oxygen consumption; yet do not show the typical pathological responses. Because of the high abundance and metabolic cost of skeletal muscle, not only must it adjust to the constraints of hibernation, but it is also positioned to play a more active role in the initiation and maintenance of the hibernation phenotype.
    [Show full text]
  • Yeast Genome Gazetteer P35-65
    gazetteer Metabolism 35 tRNA modification mitochondrial transport amino-acid metabolism other tRNA-transcription activities vesicular transport (Golgi network, etc.) nitrogen and sulphur metabolism mRNA synthesis peroxisomal transport nucleotide metabolism mRNA processing (splicing) vacuolar transport phosphate metabolism mRNA processing (5’-end, 3’-end processing extracellular transport carbohydrate metabolism and mRNA degradation) cellular import lipid, fatty-acid and sterol metabolism other mRNA-transcription activities other intracellular-transport activities biosynthesis of vitamins, cofactors and RNA transport prosthetic groups other transcription activities Cellular organization and biogenesis 54 ionic homeostasis organization and biogenesis of cell wall and Protein synthesis 48 plasma membrane Energy 40 ribosomal proteins organization and biogenesis of glycolysis translation (initiation,elongation and cytoskeleton gluconeogenesis termination) organization and biogenesis of endoplasmic pentose-phosphate pathway translational control reticulum and Golgi tricarboxylic-acid pathway tRNA synthetases organization and biogenesis of chromosome respiration other protein-synthesis activities structure fermentation mitochondrial organization and biogenesis metabolism of energy reserves (glycogen Protein destination 49 peroxisomal organization and biogenesis and trehalose) protein folding and stabilization endosomal organization and biogenesis other energy-generation activities protein targeting, sorting and translocation vacuolar and lysosomal
    [Show full text]
  • Inhibition of Glycolysis in Pathogenic TH17 Cells Through Targeting a -21Mir −Peli1− C-Rel Pathway Prevents Autoimmunity
    Inhibition of Glycolysis in Pathogenic TH17 Cells through Targeting a miR-21−Peli1− c-Rel Pathway Prevents Autoimmunity This information is current as Rong Qiu, Xiang Yu, Li Wang, Zhijun Han, Chao Yao, of September 26, 2021. Yange Cui, Guojun Hou, Dai Dai, Wenfei Jin and Nan Shen J Immunol published online 15 May 2020 http://www.jimmunol.org/content/early/2020/05/14/jimmun ol.2000060 Downloaded from Supplementary http://www.jimmunol.org/content/suppl/2020/05/14/jimmunol.200006 Material 0.DCSupplemental http://www.jimmunol.org/ Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication by guest on September 26, 2021 *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2020 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. Published May 15, 2020, doi:10.4049/jimmunol.2000060 The Journal of Immunology Inhibition of Glycolysis in Pathogenic TH17 Cells through Targeting a miR-21–Peli1–c-Rel Pathway Prevents Autoimmunity Rong Qiu,*,†,1 Xiang Yu,*,1 Li Wang,* Zhijun Han,‡ Chao Yao,† Yange Cui,† Guojun Hou,* Dai Dai,* Wenfei Jin,‡ and Nan Shen*,x,{,||,# It is well known that some pathogenic cells have enhanced glycolysis; the regulatory network leading to increased glycolysis are not well characterized.
    [Show full text]
  • Peripheral Glycolysis in Neurodegenerative Diseases
    International Journal of Molecular Sciences Review Peripheral Glycolysis in Neurodegenerative Diseases Simon M. Bell * , Toby Burgess, James Lee, Daniel J. Blackburn, Scott P. Allen and Heather Mortiboys Sheffield Institute for Translational Neurosciences, University of Sheffield, Sheffield S10 2HQ, UK; t.burgess@sheffield.ac.uk (T.B.); james.lee@sheffield.ac.uk (J.L.); d.blackburn@sheffield.ac.uk (D.J.B.); s.p.allen@sheffield.ac.uk (S.P.A.); [email protected] (H.M.) * Correspondence: s.m.bell@sheffield.ac.uk; Tel.: +44-(0)-114-2222273 Received: 31 October 2020; Accepted: 21 November 2020; Published: 24 November 2020 Abstract: Neurodegenerative diseases are a group of nervous system conditions characterised pathologically by the abnormal deposition of protein throughout the brain and spinal cord. One common pathophysiological change seen in all neurodegenerative disease is a change to the metabolic function of nervous system and peripheral cells. Glycolysis is the conversion of glucose to pyruvate or lactate which results in the generation of ATP and has been shown to be abnormal in peripheral cells in Alzheimer’s disease, Parkinson’s disease, and Amyotrophic Lateral Sclerosis. Changes to the glycolytic pathway are seen early in neurodegenerative disease and highlight how in multiple neurodegenerative conditions pathology is not always confined to the nervous system. In this paper, we review the abnormalities described in glycolysis in the three most common neurodegenerative diseases. We show that in all three diseases glycolytic changes are seen in fibroblasts, and red blood cells, and that liver, kidney, muscle and white blood cells have abnormal glycolysis in certain diseases.
    [Show full text]