Supplemental Information

Total Page:16

File Type:pdf, Size:1020Kb

Supplemental Information Methylation of HSP70 orchestrates its binding to and stabilization of BCL-2 mRNA and renders pancreatic cancer cells resistant to therapeutics Liang Wang1,3, Zhiliang Jia1, Dacheng Xie1,4, Tiansuo Zhao1, Zhi Tan2,3, Shuxing Zhang2,3, Fanyang Kong1, Daoyan Wei1, Keping Xie1,3,4 Departments of 1Gastroenterology, Hepatology and Nutrition, 2Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas; 3The University of Texas MD Anderson Cancer Center and UTHealth Graduate School of Biomedical Sciences, Houston, Texas; and 4The Precision Institutes of Medicine and Oncology, Houston, Texas Supplemental Information 1. Supplemental Figures and Legends 2. Supplemental Tables 3. Supplemental Materials and Methods 1 1. Supplemental Figures 2 Figure S1. PRMT1 is overexpressed in multiple gastrointestinal tumors and is associated with patient overall survival. (A) PRMT1 protein expression level in two pairs of isogeneic pancreatic cancer cell lines. (B) Survival analysis of The Cancer Genome Atlas (TCGA) data for various tumor types. Patients were divided into low and high groups based on PRMT1 mRNA expression level. Five-year overall survival rates and log-rank P values are shown. (C) Immunohistochemical staining demonstrating markedly higher PRMT1 expression in gastric, colon, and liver tumor tissues than in adjacent normal tissues. 3 4 Figure S2. Genetic alterations and inflammatory factors contribute to increased PRMT1 expression during pancreatic cancer (PDA) progression. (A) Representative immunohistochemical staining of PRMT1 expression in pancreatic tissue sections from mouse pancreatic cancer models. In the lower panels, the red arrow indicates a pancreatic intraepithelial neoplasia (PanIN) lesion and the black arrowhead indicates a PDA lesion. (B) PRMT1 staining scores were higher in mouse PanIN and PDA lesions. Error bars indicate mean ± SEM. (C) Double immunofluorescence staining of PRMT1 and Ki67 in two KPC mouse pancreatic tissue sections. (D) Percentage of the pancreatic area that is stained positive for CK19 in PBS and DB75 treated groups. (E) Copy number change of the PRMT1 gene locus in 105 human PDA specimens. (F) TNF-α dose-dependently increased PRMT1 expression in human and mouse PDA cells. 5 6 Figure S3. Altered expression and activity of PRMT1 impacts pancreatic cancer (PDA) cell migration, invasion, and growth. (A) Transfected cells were photographed at 0 hours and 12 hours after transfection, and wound areas are outlined by dashed lines (upper and middle panels). Boyden chamber invasion assays were performed, and photos of invaded cells stained by hematoxylin and eosin are shown (bottom panels). (B) Quantification of cell migration and invasion assay results, which were conducted in triplicate. (C) PRMT1 was knocked down in PANC-1 cells, and the growth of tumor xenografts in nude mice was slower when compared with mock cells. (D) L3.7 cells were treated with TC-E5003 (Low=6.25M, High=12.5M) for 48 hours and cell apoptosis was observed (left panel). MiaPaCa-2 cells were treated with DB75 (Low=20M, High=50M) for 48 hours and cell apoptosis (black arrows) was observed. (E) MiaPaCa-2 cells were treated with either DB75 or TC-E5003 for 36h. asymmetric dimethyl- arginine (ASYM24) and asymmetric demethylation of histone H4R3 were tested by Western blot. (F) Human pancreatic cancer MDA28 cells were injected subcutaneously into nude mice to grow for 3 weeks. Phosphate-buffered saline (PBS) or low-dose (5 mg/kg) and high-dose (20 mg/kg) DB75 was administrated intraperitoneally twice per week. Mice were euthanized at day 21. Xenograft tumors were photographed and weighed (left panel). Tumors tissues were used to detect PRMT1 protein expression and protein methylation level by Western blot (right panel). (G) Human pancreatic stellate cells were treated with either DB75 or TC-E5003 at indicated concentrations and cell viability is determined. *P < 0.05, ***P < 0.001. All bar plot data are means ± SD. 7 8 Figure S4. PRMT1 and HSP70 proteins exhibit subcellular co-localization and binding motifs of PRMT1-HSP70 interaction are evident. (A) HSP70 (green) and PRMT1 (red) were detected by immunofluorescence microscopy using HSP70- and PRMT1-specific antibodies in mouse embryonic fibroblasts (MEFs) under normal culture conditions and after heat shock treatment (1 hour at 43°C and then recovery for 6 hours at 37°C). White arrows indicate cells undergoing apoptosis after heat shock, exhibiting cell shrinkage and condensed nuclei. Merge I is merged green and red fluorescence and Merge II includes all three fluorescence colors. (B) Deletion mutants of HSP70 used in interaction assays. Blue boxes indicate the nucleotide binding domain. Yellow boxes indicate the substrate binding region. Green boxes indicate the helical lid region. FL, full length. (C) Deletion mutants of human PRMT1 used in interaction assays. Red boxes indicate the variable N terminal helix. Green boxes indicate the S-adenosyl methionine binding domain. Yellow boxes indicate the β-barrel structure. Blue boxes indicate the dimerization arm. (D) HA-tagged full-length (FL) or deletion mutants of human PRMT1 and Myc-tagged HSP70 were co-transfected into 293T cells, and cell lysates were subjected to SDS-PAGE and then blotted with anti-Myc and anti-HA antibodies. WCL, whole cell lysate. 9 10 Figure S5. Arginine methylation sites of HSP70 are identified. (A) PRMT1-overexpressed and knockdown cell lysates were separated by SDS-PAGE and immunoblotted (IB) with ASYM24 antibody (left panel), and Ponceau S staining showed equal loading (right panel). (B) HSP70 protein was immunopurified from PRMT1 overexpression and control cells, and HSP70 methylation was detected by ASYM24 antibody. IP, immunoprecipitation. (C) Myc-HSP70 was immunopurified from 293T cells and the gel was stained with Coomassie brilliant blue. The HSP70 band was excised and subjected to mass spectrometry. (D and E) Protein sequence alignment of the regions surrounding 410-460 binding motif in 13 human HSP70 family members (D) or in paralogous HSP70 genes in various organisms (E). 11 12 Figure S6. Arginine methylation of HSP70 protects cells against various stresses. (A) HSP70 knockout (KO) was confirmed by Western blot analysis and DNA sequencing. Left panel, expression of HSP70 was determined by Western blot analysis in 14 MiaPaCa-2 cell clones, including 6 control clones and 8 knockout clones. Right panel, DNA sequencing result of KO clone 20. The CRISPR target region revealed different indel mutations in two alleles, indicated as dual peaks. (B) HSP70 control (WT) and knockout (KO) MiaPaCa-2 cell clones were treated with gemcitabine at different concentrations for 24 hours. Cell growth inhibition was measured using the CCK-8 method. HSP70 WT group is pooled cells of 6 control clones from (A). (C) HSP70 knockout (KO) MiaPaCa-2 cell clones 20 and 41 and PATU-8902 (8902) cells were transfected with HSP70-WT, single mutant (R416A) or double mutant (Mut) HSP70, and then incubated for 3-4 days. Relative cell growth was measured using the CCK-8 method (expressed as “OD” values). (D) Western blot analysis of HSP70 protein in knockout cells with exogenous HSP70 reconstitution. (E) HSP70 wild-type and knockout MiaPaCa-2 cells were subcutaneously injected into nude mice. Xenograft tumors were photographed and weighed after 2 weeks. **P < 0.01. All bar plot data are means ± SD. 13 14 Figure S7. Arginine methylation of HSP70 promotes its mRNA binding and stabilization activity. (A) HSP70 was knocked down or overexpressed in MDA28 cells, and BCL-2 protein levels were determined by Western blot analysis. Red and black arrows indicate exogenous and endogenous HSP70 protein respectively. (B) Four PDA cell lines were transfected with pcDNA3.0, HSP70 wild-type (WT), and HSP70 methylation-deficient mutant (Mut) vectors, and BCL-2 mRNA levels were determined by quantitative real-time PCR. (C) Upper panel: two pancreatic cancer cell lines were treated with gemcitabine (Gem) for 24 hours and BCL-2 expression was determined by Western blot analysis. Bottom panel: two pancreatic cancer cell lines were treated in the presence or absence of 1 μM venetoclax (VENE); 20 μM gemcitabine (GEM), or both (V+G), and cytotoxicity was determined by CCK-8. CTRL, vehicle control. (D) The human and mouse 3′- untranslated regions (UTR) of BCL-2 mRNA were analyzed. There are three typical AU-rich elements (AREs), two pentamers (black boxes), and one nonamer (red box) in the first 500 base pairs (bp) of the BCL-2 3′-UTR. (E) The ribonucleoprotein immunoprecipitation (RNP-IP) assay was conducted in 293T cells transfected with wild-type or mutant HSP70. mRNAs of BCL-2 (lanes 2 and 6 for primer pair 1 and lanes 3 and 7 for primer pair 2), VEGFA (lanes 4 and 8), and GAPDH (lanes 5 and 9) were determined by reverse-transcriptase PCR. Lane 1, 100 bp DNA ladder. (F) The 557bp 3′-UTR of BCL-2 containing three AREs was cloned into a pMIR-REPORT vector (pMir-557WT), and the ATTTA conserved sequences were mutated to AGGAA to generate pMir- 557Mut vector. (G) HSP70 failed to stabilize mRNA without AREs. The pMir-557 Mut vector was co-transfected with wild-type or mutant HSP70 for 36 hours and luciferase activity was measured. ***P < 0.001, NS non-significant. All bar plot data are means ± SD. 15 16 Figure S8. Arginine methylation of heat shock proteins mediates therapeutic resistance in pancreatic cancer. During PDA development, PRMT1 expression is gradually increased. Two conserved arginine residues of HSP70 are methylated by PRMT1, and methylated HSP70 binds to BCL-2 mRNA through AREs in its 3’-UTR. BCL-2 mRNA stabilization promotes BCL-2 protein expression and increases drug resistance and anti-apoptosis of cancer cells. PRMT1 inhibitors may interrupt this pathway and sensitize cancer cells to chemotherapy. 17 2. Supplemental Tables Table S1. Clinicopathologic parameters and PRMT1 expression in the pancreatic cancer patients Table S2. HA-PRMT1 and HA-ctrl binding proteins identified in mass spectrometry Table S3.
Recommended publications
  • Cyclin D1/Cyclin-Dependent Kinase 4 Interacts with Filamin a and Affects the Migration and Invasion Potential of Breast Cancer Cells
    Published OnlineFirst February 28, 2010; DOI: 10.1158/0008-5472.CAN-08-1108 Tumor and Stem Cell Biology Cancer Research Cyclin D1/Cyclin-Dependent Kinase 4 Interacts with Filamin A and Affects the Migration and Invasion Potential of Breast Cancer Cells Zhijiu Zhong, Wen-Shuz Yeow, Chunhua Zou, Richard Wassell, Chenguang Wang, Richard G. Pestell, Judy N. Quong, and Andrew A. Quong Abstract Cyclin D1 belongs to a family of proteins that regulate progression through the G1-S phase of the cell cycle by binding to cyclin-dependent kinase (cdk)-4 to phosphorylate the retinoblastoma protein and release E2F transcription factors for progression through cell cycle. Several cancers, including breast, colon, and prostate, overexpress the cyclin D1 gene. However, the correlation of cyclin D1 overexpression with E2F target gene regulation or of cdk-dependent cyclin D1 activity with tumor development has not been identified. This suggests that the role of cyclin D1 in oncogenesis may be independent of its function as a cell cycle regulator. One such function is the role of cyclin D1 in cell adhesion and motility. Filamin A (FLNa), a member of the actin-binding filamin protein family, regulates signaling events involved in cell motility and invasion. FLNa has also been associated with a variety of cancers including lung cancer, prostate cancer, melanoma, human bladder cancer, and neuroblastoma. We hypothesized that elevated cyclin D1 facilitates motility in the invasive MDA-MB-231 breast cancer cell line. We show that MDA-MB-231 motility is affected by disturbing cyclin D1 levels or cyclin D1-cdk4/6 kinase activity.
    [Show full text]
  • C8cc08685k1.Pdf
    Electronic Supplementary Material (ESI) for ChemComm. This journal is © The Royal Society of Chemistry 2018 Supporting Information Minimalist Linkers Suitable for Irreversible Inhibitors in Simultaneous Proteome Profiling, Live-Cell Imaging and Drug Screening Cuiping Guo,Yu Chang, Xin Wang, Chengqian Zhang, Piliang Hao*, Ke Ding and Zhengqiu Li* School of Pharmacy, Jinan University, Guangzhou, China 510632 *Corresponding author ([email protected]) 1. General Information All chemicals were purchased from commercial vendors and used without further purification, unless indicated otherwise. All reactions requiring anhydrous conditions were carried out under argon or nitrogen atmosphere using oven-dried glassware. AR-grade solvents were used for all reactions. Reaction progress was monitored by TLC on pre-coated silica plates (Merck 60 F254 nm, 0.25 µm) and spots were visualized by UV, iodine or other suitable stains. Flash column chromatography was carried out using silica gel (Qingdao Ocean). All NMR spectra (1H-NMR, 13C-NMR) were recorded on Bruker 300 MHz/400 MHz NMR spectrometers. Chemical shifts were reported in parts per million (ppm) referenced with respect to appropriate internal standards or residual solvent peaks (CDCl3 = 7.26 ppm, DMSO-d6 = 2.50 ppm). The following abbreviations were used in reporting spectra, br s (broad singlet), s (singlet), d (doublet), t (triplet), q (quartet), m (multiplet), dd (doublet of doublets). Mass spectra were obtained on Agilent LC-ESI-MS system. All analytical HPLC were carried out on Agilent system. Water with 0.1% TFA and acetonitrile with 0.1% TFA were used as eluents and the flow rate was 0.5 mL/min.
    [Show full text]
  • The Rise and Fall of the Bovine Corpus Luteum
    University of Nebraska Medical Center DigitalCommons@UNMC Theses & Dissertations Graduate Studies Spring 5-6-2017 The Rise and Fall of the Bovine Corpus Luteum Heather Talbott University of Nebraska Medical Center Follow this and additional works at: https://digitalcommons.unmc.edu/etd Part of the Biochemistry Commons, Molecular Biology Commons, and the Obstetrics and Gynecology Commons Recommended Citation Talbott, Heather, "The Rise and Fall of the Bovine Corpus Luteum" (2017). Theses & Dissertations. 207. https://digitalcommons.unmc.edu/etd/207 This Dissertation is brought to you for free and open access by the Graduate Studies at DigitalCommons@UNMC. It has been accepted for inclusion in Theses & Dissertations by an authorized administrator of DigitalCommons@UNMC. For more information, please contact [email protected]. THE RISE AND FALL OF THE BOVINE CORPUS LUTEUM by Heather Talbott A DISSERTATION Presented to the Faculty of the University of Nebraska Graduate College in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy Biochemistry and Molecular Biology Graduate Program Under the Supervision of Professor John S. Davis University of Nebraska Medical Center Omaha, Nebraska May, 2017 Supervisory Committee: Carol A. Casey, Ph.D. Andrea S. Cupp, Ph.D. Parmender P. Mehta, Ph.D. Justin L. Mott, Ph.D. i ACKNOWLEDGEMENTS This dissertation was supported by the Agriculture and Food Research Initiative from the USDA National Institute of Food and Agriculture (NIFA) Pre-doctoral award; University of Nebraska Medical Center Graduate Student Assistantship; University of Nebraska Medical Center Exceptional Incoming Graduate Student Award; the VA Nebraska-Western Iowa Health Care System Department of Veterans Affairs; and The Olson Center for Women’s Health, Department of Obstetrics and Gynecology, Nebraska Medical Center.
    [Show full text]
  • Reprogramming of Trna Modifications Controls the Oxidative Stress Response by Codon-Biased Translation of Proteins
    Reprogramming of tRNA modifications controls the oxidative stress response by codon-biased translation of proteins The MIT Faculty has made this article openly available. Please share how this access benefits you. Your story matters. Citation Chan, Clement T.Y. et al. “Reprogramming of tRNA Modifications Controls the Oxidative Stress Response by Codon-biased Translation of Proteins.” Nature Communications 3 (2012): 937. As Published http://dx.doi.org/10.1038/ncomms1938 Publisher Nature Publishing Group Version Author's final manuscript Citable link http://hdl.handle.net/1721.1/76775 Terms of Use Article is made available in accordance with the publisher's policy and may be subject to US copyright law. Please refer to the publisher's site for terms of use. Reprogramming of tRNA modifications controls the oxidative stress response by codon-biased translation of proteins Clement T.Y. Chan,1,2 Yan Ling Joy Pang,1 Wenjun Deng,1 I. Ramesh Babu,1 Madhu Dyavaiah,3 Thomas J. Begley3 and Peter C. Dedon1,4* 1Department of Biological Engineering, 2Department of Chemistry and 4Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139; 3College of Nanoscale Science and Engineering, University at Albany, SUNY, Albany, NY 12203 * Corresponding author: PCD, Department of Biological Engineering, NE47-277, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139; tel 617-253-8017; fax 617-324-7554; email [email protected] 2 ABSTRACT Selective translation of survival proteins is an important facet of cellular stress response. We recently demonstrated that this translational control involves a stress-specific reprogramming of modified ribonucleosides in tRNA.
    [Show full text]
  • Protein Name Accession Number Molecular Weight Myovi-GTD
    MyoVI-GTD MyoVI-GTD MyoVa-MGT MyoVa-MGT Molecular Spectral Unique Spectral Unique Protein Name Accession Number Weight Counts Peptides Counts Peptides Dync1h1 Cytoplasmic dynein 1 heavy chain 1 IPI00119876 532 kDa 310 121 515 182 Spna2 Spectrin alpha 2 IPI00757353 285 kDa 853 170 597 149 Myo5a 215 kDa protein IPI00875222 215 kDa 162 47 874 109 AU042671 hypothetical protein LOC269700 isoform 1 IPI00762814 453 kDa 2 2 231 104 Spnb2 Isoform 1 of Spectrin beta chain, brain 1 IPI00319830 274 kDa 505 122 347 100 Dmxl2 Isoform 1 of DmX-like protein 2 IPI00853932 338 kDa 63 38 251 100 Cltc Clathrin heavy chain 1 IPI00169916 (+1) 192 kDa 1994 138 565 90 Mtap2 12 days embryo spinal cord cDNA, RIKEN full-length enriched library, clone:C530026F16 product:microtubule-associated protein 2, full insert sequenceIPI00894724 199 kDa 229 82 258 74 Mtap1a Isoform 1 of Microtubule-associated protein 1A IPI00408909 (+1) 300 kDa 310 86 214 74 Itpr1 Isoform 4 of Inositol 1,4,5-trisphosphate receptor type 1 IPI00230019 (+3) 311 kDa 37 18 155 73 Huwe1 HECT, UBA and WWE domain containing 1 IPI00463909 (+1) 483 kDa 5 5 91 69 Fasn Fatty acid synthase IPI00113223 272 kDa 24 17 140 68 Usp9x Ubiquitin carboxyl-terminal hydrolase IPI00798468 291 kDa 68 45 98 65 Lrp1 Prolow-density lipoprotein receptor-related protein 1 precursor IPI00119063 505 kDa 92 53 109 62 Myh10 Myosin-10 IPI00515398 (+1) 229 kDa 65 40 98 59 Mical1 NEDD9-interacting protein with calponin homology and LIM domains IPI00116371 117 kDa 2 2 203 57 Plec1 Isoform PLEC-1I of Plectin-1 IPI00229509 (+10)
    [Show full text]
  • Allele-Specific Expression of Ribosomal Protein Genes in Interspecific Hybrid Catfish
    Allele-specific Expression of Ribosomal Protein Genes in Interspecific Hybrid Catfish by Ailu Chen A dissertation submitted to the Graduate Faculty of Auburn University in partial fulfillment of the requirements for the Degree of Doctor of Philosophy Auburn, Alabama August 1, 2015 Keywords: catfish, interspecific hybrids, allele-specific expression, ribosomal protein Copyright 2015 by Ailu Chen Approved by Zhanjiang Liu, Chair, Professor, School of Fisheries, Aquaculture and Aquatic Sciences Nannan Liu, Professor, Entomology and Plant Pathology Eric Peatman, Associate Professor, School of Fisheries, Aquaculture and Aquatic Sciences Aaron M. Rashotte, Associate Professor, Biological Sciences Abstract Interspecific hybridization results in a vast reservoir of allelic variations, which may potentially contribute to phenotypical enhancement in the hybrids. Whether the allelic variations are related to the downstream phenotypic differences of interspecific hybrid is still an open question. The recently developed genome-wide allele-specific approaches that harness high- throughput sequencing technology allow direct quantification of allelic variations and gene expression patterns. In this work, I investigated allele-specific expression (ASE) pattern using RNA-Seq datasets generated from interspecific catfish hybrids. The objective of the study is to determine the ASE genes and pathways in which they are involved. Specifically, my study investigated ASE-SNPs, ASE-genes, parent-of-origins of ASE allele and how ASE would possibly contribute to heterosis. My data showed that ASE was operating in the interspecific catfish system. Of the 66,251 and 177,841 SNPs identified from the datasets of the liver and gill, 5,420 (8.2%) and 13,390 (7.5%) SNPs were identified as significant ASE-SNPs, respectively.
    [Show full text]
  • Figure S1. DMD Module Network. the Network Is Formed by 260 Genes from Disgenet and 1101 Interactions from STRING. Red Nodes Are the Five Seed Candidate Genes
    Figure S1. DMD module network. The network is formed by 260 genes from DisGeNET and 1101 interactions from STRING. Red nodes are the five seed candidate genes. Figure S2. DMD module network is more connected than a random module of the same size. It is shown the distribution of the largest connected component of 10.000 random modules of the same size of the DMD module network. The green line (x=260) represents the DMD largest connected component, obtaining a z-score=8.9. Figure S3. Shared genes between BMD and DMD signature. A) A meta-analysis of three microarray datasets (GSE3307, GSE13608 and GSE109178) was performed for the identification of differentially expressed genes (DEGs) in BMD muscle biopsies as compared to normal muscle biopsies. Briefly, the GSE13608 dataset included 6 samples of skeletal muscle biopsy from healthy people and 5 samples from BMD patients. Biopsies were taken from either biceps brachii, triceps brachii or deltoid. The GSE3307 dataset included 17 samples of skeletal muscle biopsy from healthy people and 10 samples from BMD patients. The GSE109178 dataset included 14 samples of controls and 11 samples from BMD patients. For both GSE3307 and GSE10917 datasets, biopsies were taken at the time of diagnosis and from the vastus lateralis. For the meta-analysis of GSE13608, GSE3307 and GSE109178, a random effects model of effect size measure was used to integrate gene expression patterns from the two datasets. Genes with an adjusted p value (FDR) < 0.05 and an │effect size│>2 were identified as DEGs and selected for further analysis. A significant number of DEGs (p<0.001) were in common with the DMD signature genes (blue nodes), as determined by a hypergeometric test assessing the significance of the overlap between the BMD DEGs and the number of DMD signature genes B) MCODE analysis of the overlapping genes between BMD DEGs and DMD signature genes.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Supplementary 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]
  • Prox1regulates the Subtype-Specific Development of Caudal Ganglionic
    The Journal of Neuroscience, September 16, 2015 • 35(37):12869–12889 • 12869 Development/Plasticity/Repair Prox1 Regulates the Subtype-Specific Development of Caudal Ganglionic Eminence-Derived GABAergic Cortical Interneurons X Goichi Miyoshi,1 Allison Young,1 Timothy Petros,1 Theofanis Karayannis,1 Melissa McKenzie Chang,1 Alfonso Lavado,2 Tomohiko Iwano,3 Miho Nakajima,4 Hiroki Taniguchi,5 Z. Josh Huang,5 XNathaniel Heintz,4 Guillermo Oliver,2 Fumio Matsuzaki,3 Robert P. Machold,1 and Gord Fishell1 1Department of Neuroscience and Physiology, NYU Neuroscience Institute, Smilow Research Center, New York University School of Medicine, New York, New York 10016, 2Department of Genetics & Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, 3Laboratory for Cell Asymmetry, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan, 4Laboratory of Molecular Biology, Howard Hughes Medical Institute, GENSAT Project, The Rockefeller University, New York, New York 10065, and 5Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724 Neurogliaform (RELNϩ) and bipolar (VIPϩ) GABAergic interneurons of the mammalian cerebral cortex provide critical inhibition locally within the superficial layers. While these subtypes are known to originate from the embryonic caudal ganglionic eminence (CGE), the specific genetic programs that direct their positioning, maturation, and integration into the cortical network have not been eluci- dated. Here, we report that in mice expression of the transcription factor Prox1 is selectively maintained in postmitotic CGE-derived cortical interneuron precursors and that loss of Prox1 impairs the integration of these cells into superficial layers. Moreover, Prox1 differentially regulates the postnatal maturation of each specific subtype originating from the CGE (RELN, Calb2/VIP, and VIP).
    [Show full text]
  • Immunoprecipitation and Mass Spectrometry Defines an Extensive
    BRES : 44759 Model7 pp: À 1221ðcol:fig: : NILÞ brain research ] ( ]]]]) ]]]– ]]] Available online at www.sciencedirect.com 121 122 123 124 125 126 www.elsevier.com/locate/brainres 127 128 129 Review 130 131 fi 132 Immunoprecipitation and mass spectrometry de nes 133 – 134 an extensive RBM45 protein protein interaction 135 Q2 136 network 137 138 a a,b a a c 139 Yang Li , Mahlon Collins , Jiyan An , Rachel Geiser , Tony Tegeler , c c c a,b,n 140 Q1 Kristine Tsantilas , Krystine Garcia , Patrick Pirrotte , Robert Bowser 141 aDivisions of Neurology and Neurobiology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 142 Phoenix, AZ 85013, USA 143 bUniversity of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA 144 cCenter for Proteomics, TGen (Translational Genomics Research Institute), Phoenix, AZ 85004, USA 145 146 147 article info abstract 148 149 Article history: The pathological accumulation of RNA-binding proteins (RBPs) within inclusion bodies is a 150 Received 30 January 2016 hallmark of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration 151 Received in revised form (FTLD). RBP aggregation results in both toxic gain and loss of normal function. Determining 152 25 February 2016 the protein binding partners and normal functions of disease-associated RBPs is necessary 153 Accepted 28 February 2016 to fully understand molecular mechanisms of RBPs in disease. Herein, we characterized 154 the protein–protein interactions (PPIs) of RBM45, a RBP that localizes to inclusions in ALS/ 155 – fi Keywords: FTLD. Using immunoprecipitation coupled to mass spectrometry (IP MS), we identi ed 132 156 fi RBM45 proteins that speci cally interact with RBM45 within HEK293 cells.
    [Show full text]
  • Anti-Inflammatory Role of Curcumin in LPS Treated A549 Cells at Global Proteome Level and on Mycobacterial Infection
    Anti-inflammatory Role of Curcumin in LPS Treated A549 cells at Global Proteome level and on Mycobacterial infection. Suchita Singh1,+, Rakesh Arya2,3,+, Rhishikesh R Bargaje1, Mrinal Kumar Das2,4, Subia Akram2, Hossain Md. Faruquee2,5, Rajendra Kumar Behera3, Ranjan Kumar Nanda2,*, Anurag Agrawal1 1Center of Excellence for Translational Research in Asthma and Lung Disease, CSIR- Institute of Genomics and Integrative Biology, New Delhi, 110025, India. 2Translational Health Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, 110067, India. 3School of Life Sciences, Sambalpur University, Jyoti Vihar, Sambalpur, Orissa, 768019, India. 4Department of Respiratory Sciences, #211, Maurice Shock Building, University of Leicester, LE1 9HN 5Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia- 7003, Bangladesh. +Contributed equally for this work. S-1 70 G1 S 60 G2/M 50 40 30 % of cells 20 10 0 CURI LPSI LPSCUR Figure S1: Effect of curcumin and/or LPS treatment on A549 cell viability A549 cells were treated with curcumin (10 µM) and/or LPS or 1 µg/ml for the indicated times and after fixation were stained with propidium iodide and Annexin V-FITC. The DNA contents were determined by flow cytometry to calculate percentage of cells present in each phase of the cell cycle (G1, S and G2/M) using Flowing analysis software. S-2 Figure S2: Total proteins identified in all the three experiments and their distribution betwee curcumin and/or LPS treated conditions. The proteins showing differential expressions (log2 fold change≥2) in these experiments were presented in the venn diagram and certain number of proteins are common in all three experiments.
    [Show full text]