TBC1D1 Regulates Insulin- and Contraction-Induced Glucose Transport in Mouse Skeletal Muscle Ding An,1 Taro Toyoda,1 Eric B

Total Page:16

File Type:pdf, Size:1020Kb

TBC1D1 Regulates Insulin- and Contraction-Induced Glucose Transport in Mouse Skeletal Muscle Ding An,1 Taro Toyoda,1 Eric B ORIGINAL ARTICLE TBC1D1 Regulates Insulin- and Contraction-Induced Glucose Transport in Mouse Skeletal Muscle Ding An,1 Taro Toyoda,1 Eric B. Taylor,1 Haiyan Yu,1 Nobuharu Fujii,1 Michael F. Hirshman,1 and Laurie J. Goodyear1,2 OBJECTIVE—TBC1D1 is a member of the TBC1 Rab-GTPase through a phosphatidylinositol 3 kinase (PI 3-kinase)– family of proteins and is highly expressed in skeletal muscle. dependent mechanism and the exercise effect may involve Insulin and contraction increase TBC1D1 phosphorylation on multiple molecules including the AMP-activated protein phospho-Akt substrate motifs (PASs), but the function of kinase (AMPK) family of protein kinases, an important TBC1D1 in muscle is not known. Genetic linkage analyses show goal in this field has been to elucidate the mechanisms that a TBC1D1 R125W missense variant confers risk for severe connect the proximal insulin and exercise signals to obesity in humans. The objective of this study was to determine whether TBC1D1 regulates glucose transport in skeletal muscle. GLUT4 translocation. Recent studies have identified two molecules, the Akt substrate of 160 kDa (AS160/TBC1D4) RESEARCH DESIGN AND METHODS—In vivo gene injection and its paralog, TBC1D1, as potential molecular links and electroporation were used to overexpress wild-type and among multiple signaling pathways converging on GLUT4 several mutant TBC1D1 proteins in mouse tibialis anterior mus- translocation in skeletal muscle (5–12). cles, and glucose transport was measured in vivo. AS160 was first shown to modulate GLUT4 trafficking in RESULTS—Expression of the obesity-associated R125W mutant insulin-sensitive 3T3-L1 adipocytes (13). A striking struc- significantly decreased insulin-stimulated glucose transport in tural feature of AS160 is that the molecule harbors a the absence of changes in TBC1D1 PAS phosphorylation. Simul- Rab-GTPase (GAP) domain and its activity controls Rab- taneous expression of an inactive Rab-GTPase (GAP) domain of GTP loading and regulation of Rab function (11). AS160 TBC1D1 in the R125W mutant reversed this decrease in glucose contains at least six amino acids that can be phosphory- transport caused by the R125W mutant. Surprisingly, expression of TBC1D1 mutated to Ala on four conserved Akt and/or AMP- lated in response to insulin by Akt (14), and these sites can activated protein kinase predicted phosphorylation sites (4P) be detected in aggregate with an antibody directed against had no effect on insulin-stimulated glucose transport. In contrast, a phospho-Akt substrate motif (PAS). Insulin, exercise, and expression of the TBC1D1 4P mutant decreased contraction- the AMPK activator 5Ј-aminoimidazole-4-carboxymide-1-␤-D- stimulated glucose transport, an effect prevented by concomitant ribofuranoside (AICAR) all cause phosphorylation of disruption of TBC1D1 Rab-GAP activity. There was no effect of the AS160 on PAS sites in skeletal muscle (5,7,10), and there is R125W mutation on contraction-stimulated glucose transport. good evidence that phosphorylation of these sites inhibits CONCLUSIONS—TBC1D1 regulates both insulin- and contrac- AS160 activity, leading to trafficking of GLUT4 vesicles to tion-stimulated glucose transport, and this occurs via distinct the cell surface and glucose transport (8,15). However, mechanisms. The R125W mutation of TBC1D1 impairs skeletal ablation of AS160 phosphorylation at PAS sites only muscle glucose transport, which could be a mechanism for the partially inhibits insulin- and contraction-stimulated glu- obesity associated with this mutation. Diabetes 59:1358–1365, cose transport (8,15). Furthermore, a preliminary report 2010 suggests that whole-body knockout of AS160 does not result in a significant increase in glucose transport in muscle (16). Taken together, these findings suggest that there may be one or more additional Rab-GAP proteins nsulin and exercise are the most physiologically expressed in skeletal muscle that also function to regulate relevant stimulators of glucose transport in skeletal glucose transport. muscle, and in patients with type 2 diabetes, insulin- TBC1D1 (tre-2/USP6, BUB2, cdc16 domain family mem- Ibut not exercise-stimulated glucose transport can be ber 1) is a member of the TBC1 Rab-GAP family of severely impaired (1–4). GLUT4 translocation from an proteins. TBC1D1 and AS160 are 47% identical overall and intracellular location to the sarcolemma and transverse have several comparable structural features. Both proteins tubules is the major mechanism through which both contain two full or partial NH2-terminal phosphotyrosine insulin and exercise increase glucose transport in muscle binding domains, a splice exon, and a putative calmodulin- (1,2). Although it is well established that insulin works binding domain. The COOH-terminal Rab-GAP domains of TBC1D1 and AS160 are 79% identical and exhibit similar From the 1Research Division, Joslin Diabetes Center, Boston, Massachusetts; Rab specificities in vitro (6). Both proteins are detected at and the 2Department of Medicine, Brigham and Women’s Hospital and a molecular weight of ϳ150–160 kDa upon SDS-PAGE, Harvard Medical School, Boston, Massachusetts. and both can be detected using the PAS antibody. Al- Corresponding author: Laurie J. Goodyear, [email protected]. Received 26 August 2009 and accepted 2 March 2010. Published ahead of print though there are several similarities between TBC1D1 and at http://diabetes.diabetesjournals.org on 18 March 2010. DOI: 10.2337/ AS160, there are also clear differences in tissue distribu- db09-1266. tion and protein phosphorylation. For example, whereas © 2010 by the American Diabetes Association. Readers may use this article as AS160 is expressed in multiple tissues at similar levels, long as the work is properly cited, the use is educational and not for profit, and the work is not altered. See http://creativecommons.org/licenses/by TBC1D1 is several-fold higher in skeletal muscle com- -nc-nd/3.0/ for details. pared with other tissues (12). Another key difference The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked “advertisement” in accordance between TBC1D1 and AS160 relates to the phosphoryla- with 18 U.S.C. Section 1734 solely to indicate this fact. tion status of the proteins. Insulin increases the phosphor- 1358 DIABETES, VOL. 59, JUNE 2010 diabetes.diabetesjournals.org D. AN AND ASSOCIATES ylation of five of six Akt substrate motifs in AS160, TBC1D1 mutated to Lys at Arg941 within its Rab-GAP domain to eliminate whereas only one of these insulin-stimulated sites is TBC1D1 GAP activity (R/K); 4) TBC1D1 double mutant containing both the 4P conserved in TBC1D1 (6,12,13). and R/K mutations; and 5) TBC1D1 double mutant containing both the R125W and R/K mutations. TBC1D1 cDNA constructs were sequenced to confirm A recent report has shown that a mouse genetic model accuracy, using the high-throughput DNA sequencing service at Brigham and resulting in a truncation mutation of TBC1D1 that includes Women’s Hospital. Plasmid DNA was amplified in E. coli Top10 cells the Rab-GAP domain of the protein, as well as knockdown (Invitrogen), extracted using an endotoxin-free Plasmid Mega Kit (Qiagen), of TBC1D1 in C2C12 muscle cells, lead to increased fatty and suspended in saline at 4 ␮g/␮l. TBC1D1 plasmids (100 ␮g) were injected acid oxidation (17). Other studies have identified a genetic into mouse tibialis anterior muscles using our protocol (22), modified to that variation in TBC1D1 that confers risk for severe obesity in originally described by Aihara and Miyazaki (23), and mice were studied 7 Caucasian U.S. and French pedigrees (18,19). This R125W days later. Contraction, glucose, and insulin treatment in the study of PAS missense variant occurs in a phosphotyrosine-binding phosphorylation. Mice were anesthetized with intraperitoneal administra- domain of TBC1D1, but how this mutation leads to meta- tion of pentobarbital sodium (90 mg/kg of body wt). Peroneal nerves from bolic dysfunction and obesity is not known. both legs were surgically exposed for electrode placement. One leg was left Using mass spectrometry, we have identified novel unstimulated (basal/sham control) and the other leg was subjected to electri- phosphorylation sites on TBC1D1 (12), and we and other cal stimulation using a Grass S88 pulse generator for 15 min of contractions laboratories have shown that one or more of these sites (train rate 1/s; train duration 500 ms; pulse rate 100 Hz; duration 0.1 ms at 2–7 can be regulated by insulin, contraction, and/or AICAR in V), as we have previously described (5). To study insulin-activated PAS skeletal muscle (12,14,20,21). The function of these phos- signaling, we injected mice with either a bolus of glucose that resulted in a physiological increase in plasma insulin concentrations (8) or an insulin bolus. phorylation sites is not known. However, given that these For the glucose treatment, mice were anesthetized and administered a 20% stimuli are potent regulators of glucose transport, that glucose bolus (1.0 g of glucose/kg of body wt). For the insulin treatment, mice TBC1D1 is highly expressed in skeletal muscle, and that were anesthetized and injected with insulin (10 mU/g). Immediately after R125W mutation of TBC1D1 is associated with obesity, we contraction, 15 min after glucose administration, or 6 min after insulin hypothesized TBC1D1 regulates glucose transport in skel- stimulation, tibialis anterior muscles were dissected and frozen in liquid etal muscle. We found distinct mutations of TBC1D1 nitrogen. differentially decrease insulin- and contraction-stimulated Protein extraction, immunoprecipitation, and immunoblotting. Frozen muscle tissue was homogenized with a Polytron (Brinkman Instruments) in glucose transport in skeletal muscle, and this occurs in a lysis buffer, as described previously (12). Homogenates were centrifuged at Rab-GAP domain–dependent manner. Our findings dem- 14,000g for 10 min and supernatants were collected. Protein concentrations onstrate that TBC1D1 functions as a Rab-GAP protein, were determined by the Bradford assay.
Recommended publications
  • 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]
  • TBC1D1 (V796) Antibody A
    Revision 1 C 0 2 - t TBC1D1 (V796) Antibody a e r o t S Orders: 877-616-CELL (2355) [email protected] Support: 877-678-TECH (8324) 9 2 Web: [email protected] 6 www.cellsignal.com 4 # 3 Trask Lane Danvers Massachusetts 01923 USA For Research Use Only. Not For Use In Diagnostic Procedures. Applications: Reactivity: Sensitivity: MW (kDa): Source: UniProt ID: Entrez-Gene Id: WB, IP M Endogenous 160 Rabbit Q86TI0 23216 Product Usage Information Application Dilution Western Blotting 1:1000 Immunoprecipitation 1:50 Storage Supplied in 10 mM sodium HEPES (pH 7.5), 150 mM NaCl, 100 µg/ml BSA and 50% glycerol. Store at –20°C. Do not aliquot the antibody. Specificity / Sensitivity TBC1D1 (V796) Antibody detects endogenous levels of total TBC1D1 protein. Species Reactivity: Mouse Source / Purification Polyclonal antibodies are produced by immunizing animals with a synthetic peptide corresponding to the sequence around Val796 of mouse TBC1D1. Antibodies are purified by protein A and peptide affinity chromatography. Background TBC1D1 is a paralog of AS160 (1) and both proteins share about 50% identity (2). TBC1D1 was shown to be a candidate gene for severe obesity (3). It plays a role in Glut4 translocation through its GAP activity (2,4). Studies indicate that TBC1D1 is highly expressed in skeletal muscle (1). Insulin, AICAR, and contraction directly regulate TBC1D1 phosphorylation in this tissue (1). Three AMPK phosphorylation sites (Ser231, Ser660, and Ser700) and one Akt phosphorylation site (Thr590) were identified in skeletal muscle (5). Muscle contraction or AICAR treatment increases phosphorylation on Ser231, Ser660, and Ser700 but not on Thr590; insulin increases phosphorylation on Thr590 only (5).
    [Show full text]
  • 11Β-Hydroxysteroid Dehydrogenase Type 1 Regulates Glucocorticoid-Induced Insulin Resistance in Skeletal Muscle
    Diabetes Publish Ahead of Print, published online August 12, 2009 11β-HSD1 and skeletal muscle 11β-hydroxysteroid dehydrogenase type 1 regulates glucocorticoid-induced insulin resistance in skeletal muscle Stuart A Morgan1, Mark Sherlock1, Laura L Gathercole1, Gareth G Lavery1, Carol Lenaghan3, Iwona J Bujalska1, David Laber3, Alice Yu3, Gemma Convey3, Rachel Mayers3, Krisztina Hegyi2, Jaswinder K Sethi2, Paul M Stewart1, David M Smith3 and Jeremy W Tomlinson1 1. Centre for Endocrinology, Diabetes and Metabolism, Institute of Biomedical Research, School of Clinical & Experimental Medicine, University of Birmingham, Birmingham, UK. B15 2TT 2. Department of Clinical Biochemistry, University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, CB2 0QQ 3. AstraZeneca Diabetes & Obesity Drug Discovery, Mereside, Alderley Park, Macclesfield, Cheshire, UK. SK10 4TG Brief title: 11β-HSD1 and skeletal muscle Address for correspondence: Jeremy W Tomlinson PhD MRCP E-mail. [email protected] Submitted 9 April 2009 and accepted 16 July 2009. This is an uncopyedited electronic version of an article accepted for publication in Diabetes. The American Diabetes Association, publisher of Diabetes, is not responsible for any errors or omissions in this version of the manuscript or any version derived from it by third parties. The definitive publisher-authenticated version will be available in a future issue of Diabetes in print and online at http://diabetes.diabetesjournals.org. Copyright American Diabetes Association, Inc., 2009 11β-HSD1 and skeletal muscle Objective: Glucocorticoid (GC) excess is characterized by increased adiposity, skeletal myopathy and insulin resistance, but the precise molecular mechanisms are unknown. Within skeletal muscle, 11β-hydroxysteroid dehydrogenase type 1 (11βHSD1) converts cortisone (11- dehydrocorticosterone, 11DHC in rodents) to active cortisol (corticosterone in rodents).
    [Show full text]
  • Activating Proteins TBC1D1 and TBC1D4 in Mice Eliminates Insulin- and AICAR-Stimulated Glucose Transport
    746 Diabetes Volume 64, March 2015 Alexandra Chadt,1,2 Anja Immisch,3 Christian de Wendt,1 Christian Springer,1 Zhou Zhou,1 Torben Stermann,1 Geoffrey D. Holman,4 Dominique Loffing-Cueni,5 Johannes Loffing,5 Hans-Georg Joost,2,3 and Hadi Al-Hasani1,2 Deletion of Both Rab-GTPase– Activating Proteins TBC1D1 and TBC1D4 in Mice Eliminates Insulin- and AICAR-Stimulated Glucose Transport Diabetes 2015;64:746–759 | DOI: 10.2337/db14-0368 The Rab-GTPase–activating proteins TBC1D1 and TBC1D4 from intracellular vesicles to the cell surface (1,2). The (AS160) were previously shown to regulate GLUT4 trans- two related Rab-GTPase–activating proteins TBC1D1 and location in response to activation of AKT and AMP- TBC1D4 (AS160) are phosphorylated in response to in- dependent kinase. However, knockout mice lacking sulin, AMPK, and exercise/muscle contraction and have either Tbc1d1 or Tbc1d4 displayed only partially impaired been implicated in important roles in regulating the trans- insulin-stimulated glucose uptake in fat and muscle tis- location of GLUT4 (3–9). By positional cloning, we pre- sue. The aim of this study was to determine the impact of viously identified a naturally occurring loss-of-function Tbc1d1 Tbc1d4 the combined inactivation of and on glu- mutation in Tbc1d1 as an obesity suppressor in the cose metabolism in double-deficient (D1/4KO) mice. METABOLISM lean, diabetes-resistant SJL mouse strain (10). In humans, D1/4KO mice displayed normal fasting glucose concen- mutations in TBC1D4 (R3633) and TBC1D1 (R125W) trations but had reduced tolerance to intraperitoneally have been linked to severe postprandial hyperinsulinemia administered glucose, insulin, and AICAR.
    [Show full text]
  • Autocrine IFN Signaling Inducing Profibrotic Fibroblast Responses By
    Downloaded from http://www.jimmunol.org/ by guest on September 23, 2021 Inducing is online at: average * The Journal of Immunology , 11 of which you can access for free at: 2013; 191:2956-2966; Prepublished online 16 from submission to initial decision 4 weeks from acceptance to publication August 2013; doi: 10.4049/jimmunol.1300376 http://www.jimmunol.org/content/191/6/2956 A Synthetic TLR3 Ligand Mitigates Profibrotic Fibroblast Responses by Autocrine IFN Signaling Feng Fang, Kohtaro Ooka, Xiaoyong Sun, Ruchi Shah, Swati Bhattacharyya, Jun Wei and John Varga J Immunol cites 49 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/2013/08/20/jimmunol.130037 6.DC1 This article http://www.jimmunol.org/content/191/6/2956.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 © 2013 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 23, 2021. The Journal of Immunology A Synthetic TLR3 Ligand Mitigates Profibrotic Fibroblast Responses by Inducing Autocrine IFN Signaling Feng Fang,* Kohtaro Ooka,* Xiaoyong Sun,† Ruchi Shah,* Swati Bhattacharyya,* Jun Wei,* and John Varga* Activation of TLR3 by exogenous microbial ligands or endogenous injury-associated ligands leads to production of type I IFN.
    [Show full text]
  • TBC1D1 (CT) Antibody (Pab)
    21.10.2014TBC1D1 (CT) antibody (pAb) Rabbit Anti -Human TBC1D1 (CT) Instru ction Manual Catalog Number PK-AB718-4231 Synonyms TBC1D1 Antibody: TBC1 domain family member 1, TBC, TBC1 Description TBC1D1 is the founding member of a family of proteins sharing a 180- to 200-amino acid TBC domain and presumed to have a role in regulating cell growth and differentiation. These proteins share significant homology with TRE2/USP6, yeast Bub2, and CDC16. TBC1D1 and TBC1D4 (AS160) have been demonstrated to be Rab GAPs (GTPase-activating proteins) that link upstream to Akt and phosphoinositide 3-kinase and downstream to Rabs involved in trafficking of GLUT4 vesicles. TBC1D1 regulates insulin-mediated GLUT4 translocation through its GAP activity, and is a likely Akt substrate. Mutations in the Tbc1d1 gene lead to some cases of severe human obesity. Quantity 100 µg Source / Host Rabbit Immunogen TBC1D1 antibody was raised in rabbits against a 22 amino acid peptide from near the carboxy terminus of human TBC1D1. Purification Method Affinity chromatography purified via peptide column. Clone / IgG Subtype Polyclonal antibody Species Reactivity Human Specificity Formulation Antibody is supplied in PBS containing 0.02% sodium azide. Reconstitution During shipment, small volumes of antibody will occasionally become entrapped in the seal of the product vial. For products with volumes of 200 μl or less, we recommend gently tapping the vial on a hard surface or briefly centrifuging the vial in a tabletop centrifuge to dislodge any liquid in the container’s cap. Storage & Stability Antibody can be stored at 4ºC for three months and at -20°C for up to one year.
    [Show full text]
  • Knockdown of Hnrnpa0, a Del(5Q) Gene, Alters Myeloid Cell Fate In
    Myelodysplastic Syndromes SUPPLEMENTARY APPENDIX Knockdown of Hnrnpa0 , a del(5q) gene, alters myeloid cell fate in murine cells through regulation of AU-rich transcripts David J. Young, 1 Angela Stoddart, 2 Joy Nakitandwe, 3 Shann-Ching Chen, 3 Zhijian Qian, 4 James R. Downing, 3 and Michelle M. Le Beau 2 1Department of Pediatrics, Division of Oncology, Johns Hopkins University, Baltimora, MD; 2Department of Medicine and the Compre - hensive Cancer Center, University of Chicago, IL; 3St. Jude Children's Research Hospital, Memphis, Tennessee; and 4University of Illi - nois Cancer Center, Chicago, IL, USA DJY and AS equally contributed to this work. ©2014 Ferrata Storti Foundation. This is an open-access paper. doi:10.3324/haematol.2013.098657 Manuscript received on September 25, 2013. Manuscript accepted on February 13, 2014. Correspondence: [email protected] Supplementary Materials for D. Young et al. Purification of hematopoietic populations from mice. Cells from the spleens, thymi, and bone marrow of C57BL/6J mice were harvested as appropriate for each population. For primitive populations including Lin–Sca-1+Kit+ (LSK), common lymphoid (CLP) and myeloid (CMP) progenitors, and granulocyte- monocyte progenitors (GMP), the cells were depleted of mature cells using the Mouse Hematopoietic Progenitor Cell Enrichment Kit (StemCell Technologies). The cells were stained for appropriate lineage markers, as described in Supplementary Figure S1, and sorted using a FACSAria fluorescence activated cell sorter (BD Biosciences). Real-time RT-PCR analysis Total RNA was purified from cells using Stat-60 (Tel-Test), according to the manufacturer’s protocols. First-strand cDNA was synthesized using SuperScript III SuperMix for qRT-PCR (Invitrogen) containing both random hexamers and oligo(dT)20 for priming.
    [Show full text]
  • Table S1. 103 Ferroptosis-Related Genes Retrieved from the Genecards
    Table S1. 103 ferroptosis-related genes retrieved from the GeneCards. Gene Symbol Description Category GPX4 Glutathione Peroxidase 4 Protein Coding AIFM2 Apoptosis Inducing Factor Mitochondria Associated 2 Protein Coding TP53 Tumor Protein P53 Protein Coding ACSL4 Acyl-CoA Synthetase Long Chain Family Member 4 Protein Coding SLC7A11 Solute Carrier Family 7 Member 11 Protein Coding VDAC2 Voltage Dependent Anion Channel 2 Protein Coding VDAC3 Voltage Dependent Anion Channel 3 Protein Coding ATG5 Autophagy Related 5 Protein Coding ATG7 Autophagy Related 7 Protein Coding NCOA4 Nuclear Receptor Coactivator 4 Protein Coding HMOX1 Heme Oxygenase 1 Protein Coding SLC3A2 Solute Carrier Family 3 Member 2 Protein Coding ALOX15 Arachidonate 15-Lipoxygenase Protein Coding BECN1 Beclin 1 Protein Coding PRKAA1 Protein Kinase AMP-Activated Catalytic Subunit Alpha 1 Protein Coding SAT1 Spermidine/Spermine N1-Acetyltransferase 1 Protein Coding NF2 Neurofibromin 2 Protein Coding YAP1 Yes1 Associated Transcriptional Regulator Protein Coding FTH1 Ferritin Heavy Chain 1 Protein Coding TF Transferrin Protein Coding TFRC Transferrin Receptor Protein Coding FTL Ferritin Light Chain Protein Coding CYBB Cytochrome B-245 Beta Chain Protein Coding GSS Glutathione Synthetase Protein Coding CP Ceruloplasmin Protein Coding PRNP Prion Protein Protein Coding SLC11A2 Solute Carrier Family 11 Member 2 Protein Coding SLC40A1 Solute Carrier Family 40 Member 1 Protein Coding STEAP3 STEAP3 Metalloreductase Protein Coding ACSL1 Acyl-CoA Synthetase Long Chain Family Member 1 Protein
    [Show full text]
  • And Diffuse-Type Gastric Cancer
    cancers Article Molecular Network Profiling in Intestinal- and Diffuse-Type Gastric Cancer Shihori Tanabe 1,* , Sabina Quader 2 , Ryuichi Ono 3, Horacio Cabral 4, Kazuhiko Aoyagi 5, Akihiko Hirose 1, Hiroshi Yokozaki 6 and Hiroki Sasaki 7 1 Division of Risk Assessment, Center for Biological Safety and Research, National Institute of Health Sciences, Kawasaki 210-9501, Japan; [email protected] 2 Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki 210-0821, Japan; [email protected] 3 Division of Cellular and Molecular Toxicology, Center for Biological Safety and Research, National Institute of Health Sciences, Kawasaki 210-9501, Japan; [email protected] 4 Department of Bioengineering, Graduate School of Engineering, University of Tokyo, Tokyo 113-0033, Japan; [email protected] 5 Department of Clinical Genomics, National Cancer Center Research Institute, Tokyo 104-0045, Japan; [email protected] 6 Department of Pathology, Kobe University of Graduate School of Medicine, Kobe 650-0017, Japan; [email protected] 7 Department of Translational Oncology, National Cancer Center Research Institute, Tokyo 104-0045, Japan; [email protected] * Correspondence: [email protected]; Tel.: +81-44-270-6686 Received: 24 November 2020; Accepted: 17 December 2020; Published: 18 December 2020 Simple Summary: Cancer has several phenotypic subtypes where the responsiveness towards drugs or capacity of migration or recurrence are different. The molecular networks are dynamically altered in various phenotypes of cancer. To reveal the network pathways in epithelial-mesenchymal transition (EMT), we have profiled gene expression in mesenchymal stem cells and diffuse-type gastric cancer (GC), as well as intestinal-type GC.
    [Show full text]
  • TBC1D1 Antibody
    TBC1D1 Antibody CATALOG NUMBER: 4231 Western blot analysis of TBC1D1 in Daudi cell lysate with TBC1D1 antibody at (A) 1, (B) 2 and (C) 4 ug/mL. Specifications SPECIES REACTIVITY: Human HOMOLOGY: Predicted species reactivity based on immunogen sequence: Mouse: (71%) TESTED APPLICATIONS: ELISA, WB APPLICATIONS: TBC1D1 antibody can be used for detection of TBC1D1 by Western blot at 1 - 4 ug/mL. USER NOTE: Optimal dilutions for each application to be determined by the researcher. POSITIVE CONTROL: 1) Cat. No. 1224 - Daudi Cell Lysate IMMUNOGEN: TBC1D1 antibody was raised against a 22 amino acid synthetic peptide from near the carboxy terminus of human TBC1D1. The immunogen is located within the last 50 amino acids of TBC1D1. HOST SPECIES: Rabbit Properties PURIFICATION: TBC1D1 Antibody is affinity chromatography purified via peptide column. PHYSICAL STATE: Liquid BUFFER: TBC1D1 Antibody is supplied in PBS containing 0.02% sodium azide. CONCENTRATION: 1 mg/mL STORAGE CONDITIONS: TBC1D1 antibody can be stored at 4˚C for three months and -20˚C, stable for up to one year. As with all antibodies care should be taken to avoid repeated freeze thaw cycles. Antibodies should not be exposed to prolonged high temperatures. CLONALITY: Polyclonal ISOTYPE: IgG CONJUGATE: Unconjugated Additional Info ALTERNATE NAMES: TBC1D1 Antibody: TBC, TBC1, KIAA1108, TBC1 domain family member 1 ACCESSION NO.: NP_055988 PROTEIN GI NO.: 50658061 OFFICIAL SYMBOL: TBC1D1 GENE ID: 23216 Background BACKGROUND: TBC1D1 Antibody: TBC1D1 is the founding member of a family of proteins sharing a 180- to 200-amino acid TBC domain and presumed to have a role in regulating cell growth and differentiation.
    [Show full text]
  • Rare Copy Number Variants and Congenital Heart Defects in the 22Q11.2 Deletion Syndrome
    UC Davis UC Davis Previously Published Works Title Rare copy number variants and congenital heart defects in the 22q11.2 deletion syndrome. Permalink https://escholarship.org/uc/item/49g9k7pj Journal Human genetics, 135(3) ISSN 0340-6717 Authors Mlynarski, Elisabeth E Xie, Michael Taylor, Deanne et al. Publication Date 2016-03-01 DOI 10.1007/s00439-015-1623-9 Peer reviewed eScholarship.org Powered by the California Digital Library University of California Hum Genet DOI 10.1007/s00439-015-1623-9 ORIGINAL INVESTIGATION Rare copy number variants and congenital heart defects in the 22q11.2 deletion syndrome Elisabeth E. Mlynarski1 · Michael Xie2 · Deanne Taylor2 · Molly B. Sheridan1 · Tingwei Guo3 · Silvia E. Racedo3 · Donna M. McDonald‑McGinn1,4 · Eva W. C. Chow5 · Jacob Vorstman6 · Ann Swillen7 · Koen Devriendt7 · Jeroen Breckpot7 · Maria Cristina Digilio8 · Bruno Marino9 · Bruno Dallapiccola8 · Nicole Philip10 · Tony J. Simon11 · Amy E. Roberts12 · Małgorzata Piotrowicz13 · Carrie E. Bearden14 · Stephan Eliez15 · Doron Gothelf16,17 · Karlene Coleman18 · Wendy R. Kates19 · Marcella Devoto1,4,20,21 · Elaine Zackai1,4 · Damian Heine‑ Suñer22 · Elizabeth Goldmuntz4,23 · Anne S. Bassett5 · Bernice E. Morrow3 · Beverly S. Emanuel1,4 · The International Chromosome 22q11.2 Consortium Received: 16 October 2015 / Accepted: 8 December 2015 © Springer-Verlag Berlin Heidelberg 2016 Abstract The 22q11.2 deletion syndrome (22q11DS; defect (CHD), mostly of the conotruncal type, and/or velocardiofacial/DiGeorge syndrome; VCFS/DGS; MIM aortic arch defect. The etiology of the cardiac pheno- #192430; 188400) is the most common microdeletion typic variability is not currently known for the major- syndrome. The phenotypic presentation of 22q11DS is ity of patients. We hypothesized that rare copy number highly variable; approximately 60–75 % of 22q11DS variants (CNVs) outside the 22q11.2 deleted region may patients have been reported to have a congenital heart modify the risk of being born with a CHD in this sen- sitized population.
    [Show full text]
  • Isoform-Specific AMPK Association with TBC1D1 Is Reduced by a Mutation Associated with Severe Obesity
    Biochemical Journal (2018) 475 2969–2983 https://doi.org/10.1042/BCJ20180475 Research Article Isoform-specific AMPK association with TBC1D1 is reduced by a mutation associated with severe obesity Elaine C. Thomas1, Sharon C. Hook1, Alexander Gray2, Alexandra Chadt3,4, David Carling5, Hadi Al-Hasani3,4, Kate J. Heesom1, D. Grahame Hardie2 and Jeremy M. Tavaré1 1School of Biochemistry, Biomedical Sciences Building, University of Bristol, Bristol, U.K.; 2Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dundee, U.K.; 3German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich-Heine-University, Medical Faculty, Düsseldorf, Germany; 4German Center for Diabetes Research (DZD), Düsseldorf, Germany; 5Cellular Stress Group, Medical Research Council London Institute of Medical Sciences, Hammersmith Hospital, Imperial College, London, U.K. Correspondence: Elaine C. Thomas ([email protected]) AMP-activated protein kinase (AMPK) is a key regulator of cellular and systemic energy homeostasis which achieves this through the phosphorylation of a myriad of downstream targets. One target is TBC1D1 a Rab-GTPase-activating protein that regulates glucose uptake in muscle cells by integrating insulin signalling with that promoted by muscle con- traction. Ser237 in TBC1D1 is a target for phosphorylation by AMPK, an event which may be important in regulating glucose uptake. Here, we show AMPK heterotrimers containing the α1, but not the α2, isoform of the catalytic subunit form an unusual and stable asso- ciation with TBC1D1, but not its paralogue AS160. The interaction between the two pro- teins is direct, involves a dual interaction mechanism employing both phosphotyrosine- binding (PTB) domains of TBC1D1 and is increased by two different pharmacological activators of AMPK (AICAR and A769962).
    [Show full text]