PTPN3 Expressed in Activated T Lymphocytes Is a Candidate for a Non-Antibody-Type Immune Checkpoint Inhibitor

Total Page:16

File Type:pdf, Size:1020Kb

PTPN3 Expressed in Activated T Lymphocytes Is a Candidate for a Non-Antibody-Type Immune Checkpoint Inhibitor Cancer Immunology, Immunotherapy (2019) 68:1649–1660 https://doi.org/10.1007/s00262-019-02403-y ORIGINAL ARTICLE PTPN3 expressed in activated T lymphocytes is a candidate for a non‑antibody‑type immune checkpoint inhibitor Akiko Fujimura1 · Kazunori Nakayama1 · Akira Imaizumi1 · Makoto Kawamoto1 · Yasuhiro Oyama1 · Shu Ichimiya1 · Masayo Umebayashi2 · Norihiro Koya2 · Takashi Morisaki2 · Takashi Nakagawa3 · Hideya Onishi1 Received: 20 February 2019 / Accepted: 23 September 2019 / Published online: 27 September 2019 © Springer-Verlag GmbH Germany, part of Springer Nature 2019 Abstract It has been shown that protein tyrosine phosphatase non-receptor type (PTPN) 3 inhibits T-cell activation. However, there is no defnitive conclusion about how the inhibition of PTPN3 in lymphocytes afects immune functions in human lym- phocytes. In the present study, we showed that PTPN3 inhibition signifcantly contributes to the enhanced activation of activated human lymphocytes. The PTPN3 expression of lymphocytes was signifcantly increased through the activation process using IL-2 and anti-CD3 mAb. Interestingly, inhibiting the PTPN3 expression in activated lymphocytes signifcantly augmented the proliferation, migration, and cytotoxicity through the phosphorylation of zeta-chain-associated protein kinase 70 (ZAP-70), lymphocyte-specifc protein tyrosine kinase (LCK), and extracellular signal-regulated kinases (ERK). Lym- phocyte activation by PTPN3 inhibition was observed only in activated CD3+ T cells and not in NK cells or resting T cells. In therapy experiments using autologous tumors and lymphocytes, PTPN3 inhibition signifcantly augmented the number of tumor-infltrated lymphocytes and the cytotoxicity of activated lymphocytes. Our results strongly imply that PTPN3 acts as an immune checkpoint in activated lymphocytes and that PTPN3 inhibitor may be a new non-antibody-type immune checkpoint inhibitor for cancer therapy. Keywords PTPN3 · Activated lymphocyte · Immune checkpoint · Cancer immunology · Immunotherapy Abbreviations PTPN Protein tyrosine phosphatase non-receptor type DALs DC-stimulated activated lymphocytes shRNA Short hairpin RNA ERK Extracellular signal-regulated kinases Zap-70 Zeta-chain-associated protein kinase 70 GAPDH Glyceraldehyde-3-phosphate dehydrogenase LCK Lymphocyte-specifc protein tyrosine kinase MOI Multiplicity of infection Introduction ntTILs Non-tumor-tissue-infltrating-administrated lymphocytes Recently, remarkable progress has been made in immuno- PTP Protein tyrosine phosphatase therapy. Many authors have reported the efectiveness of immune check point inhibitors [1, 2]. However, the response rate is still limited, and many severe adverse efects are Electronic supplementary material The online version of this reported, while the treatment remains costly. Therefore, new article (https ://doi.org/10.1007/s0026 2-019-02403 -y) contains methodologies, including combination therapy with existing supplementary material, which is available to authorized users. antibody agents, are still urgently needed. * Hideya Onishi Many molecules that act as immune checkpoint inhibitors [email protected] have been discovered and investigated, including programmed cell death (PD)-1, programmed cell death ligand-1 (PD-L1), 1 Department of Cancer Therapy and Research, Graduate PD-L2, cytotoxic T-lymphocyte associated antigen (CTLA)-4, School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan T-cell immunoglobulin and mucin domain (TIM)-3, and lym- phocyte activation gene (LAG)-3 [1–4]. However, all of these 2 Fukuoka General Cancer Clinic, Fukuoka, Japan molecules are antibody-type immune checkpoint inhibitors 3 Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan Vol.:(0123456789)1 3 1650 Cancer Immunology, Immunotherapy (2019) 68:1649–1660 and no non-antibody-type immune checkpoint inhibitors are supplemented with 2000 units/ml penicillin, 10 mg/ml available at the present time. streptomycin, 0.5% human serum, and 200 U/ml IL-2. After The actions of protein tyrosine phosphatase non-receptor 30–60 min, the supernatant was discarded, and adherent type (PTPN) 3, which removes phosphorylation from protein cells were cultured for 4–7 days in new medium supple- tyrosyl residues, may be the opposite of those of protein mented with 50 ng/ml IL-4 (Primmune) and 100 ng/ml GM- tyrosine kinase [5]. The role of PTPN3 in lymphocytes is CSF (Primmune). Following this, 200 µg of SUIT-2 tumor still controversial. PTPN3 has been shown to inhibit T-cell lysate was added to the culture. After 4–12 h, TNF-α (500 U/ activation [5]. On the other hand, some authors have dem- ml; Gentaur Molecular Products, Brussels, Belgium) and onstrated that PTPN3 is dispensable for the negative regu- 500 U/ml IFN-α (MSD, Kenilworth, NJ, USA) were added lation of T-cell activation using PTPN3 knockout mice [6]. to the culture. After 1 day of incubation, lymphocytes were However, how the inhibition of PTPN3 afects the immune added to the culture and cultured for 4 days. Non-adherent function against cancer in human activated lymphocytes fractions were collected and cultured in RPMI medium sup- remains unclear. The development of new immunotherapy is plemented with 200 U/ml IL-2 in 2.5 µg/ml OKT3-coated signifcantly important for solving problems of the response culture plates for 5 days. Cells were then collected as DALs. rate, treatment cost, and adverse efects of existing antibody agents. In the present study, we focused on PTPN3 in acti- Cancer cell culture vated lymphocytes as a target for non-antibody-type immune checkpoint inhibitors and investigated the functions of lym- The human pancreatic ductal adenocarcinoma cell line phocytes by inhibiting PTPN3. SUIT-2 and human tongue squamous cell carcinoma cell line SCC-9 were maintained in RPMI 1640 medium sup- Materials and methods plemented with 10% fetal calf serum (FCS; Thermo Fisher Scientifc, Waltham, MA, USA) and antibiotics (2000 units/ Induction of activated lymphocytes ml of penicillin and 10 µg/ml of streptomycin). In vitro and in vivo experiments used peripheral blood RNA interference mononuclear cells (PBMCs) collected from the heparinized peripheral blood of ten healthy volunteers and three cancer Nucleotide sequences for short hairpin RNA (shRNA) tar- patients via HISTOPAQUE-1077 (Merck KGaA, Darm- geting PTPN3 were as follows: shPTPN3#1, CAATCA GAA stadt, Germany) density gradient centrifugation. PBMCs GCA GAA TCC TGC TAT A; and shPTPN3#2, GAC AGC were cultured in RPMI-1640 (Nacalai Tesque, Kyoto, Japan) TAC TTA GTC TTG ATC CGT A. These oligonucleotides supplemented with 0.5% human serum, 2000 units/ml peni- were ligated in the plasmid vector (pcDNA™6.2-GW/Em- cillin (Meijiseika, Tokyo, Japan), 10 µg/ml streptomycin miR,#K4934-00; Thermo Fisher Scientifc). This plasmid (Meijiseika), and 200 U/ml IL-2 (Primmune, San Diego, vector was then co-transfected with the ViraPower™ Pack- CA, USA) in a 2.5 µg/ml anti-CD3 monoclonal antibody aging Mix (included in BLOCK-iT™ Lentiviral Poll l miR (OKT3, JANSSEN PHARMACEUTICAL K.K., Osaka, RNAi Expression Systems; Thermo Fisher Scientifc) into Japan)-coated T-75/T-25 fask or 6-well plate for 7 days. the 293FT cell line to produce the lentiviral stock. PTPN3 Non-adherent fractions were collected as activated lympho- transfection using lentivirus in lymphocytes was performed cytes. In the microarray analysis, activated lymphocytes from culture days 3–6. from two patients with pancreatic cancer and lung cancer were used. In in vivo therapy experiments, tumor cells and DNA microarray autologous-activated lymphocytes from a patient with ovar- ian cancer were used. The methodology of the DNA microarray which we used For CD3+CD4+NKG2D− T cell and CD3+CD8+NKG2D+ has been described previously [8]. Intensity-based Z-scores T-cell selection, a magnetic cell separation system was used and ratios (nonlog-scale fold-change) were calculated from (Mojosort Human CD4 T-cell isolation kit and Mojosort the normalized signal intensities of each probe to compare Human CD8 T-cell isolation kit, respectively; Biolegend, lymphocytes before and after activation. The registration San Diego, CA, USA). numbers are GSE86293 and GSE89328. Induction of dendritic cell (DC)‑stimulated activated Reverse transcription (RT)‑polymerase chain lymphocytes (DALs) reaction (PCR) DC-stimulated activated lymphocytes were induced as Total RNA was extracted using a High Pure RNA Isola- described previously [7]. PBMCs were incubated in RPMI tion kit (Roche, Basel, Switzerland) and quantified by 1 3 Cancer Immunology, Immunotherapy (2019) 68:1649–1660 1651 spectrophotometry (Ultrospec 2100 Pro; Amersham Phar- USA), anti-ERK1/2 (1:1000, No. 9102; Cell Signaling Tech- macia Biotech, Piscataway, NJ, USA). For RT-PCR, 1.0 μg nology, Danvers, MA, USA), anti-pERK (1:1000, No. 9101; of RNA was reverse transcribed to cDNA with the Verso Cell Signaling Technology), anti-Zap (1:2000, No. 3165; cDNA Synthesis Kit (Thermo Fisher Scientifc) according Cell Signaling Technology), anti-pZap (1:1000, No. 2701; to the manufacturer’s protocol. RT-PCR reactions were per- Cell Signaling Technology), anti-Lck (1:1000, No. 2657; formed using MasterMix kit (Qiagen, Valencia, CA, USA). Cell Signaling Technology), anti-pSrc (1:1000, No. 2101; All primer sets used were as follows: PTPN3 forward, Cell Signaling Technology), and anti-α tubulin (1:1000; 5′-TGG TAT CGTG GAA GGA CTCA-3′; reverse, 5′-AGC Sigma Aldrich) overnight
Recommended publications
  • 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]
  • Growth and Molecular Profile of Lung Cancer Cells Expressing Ectopic LKB1: Down-Regulation of the Phosphatidylinositol 3؅-Phosphate Kinase/PTEN Pathway1
    [CANCER RESEARCH 63, 1382–1388, March 15, 2003] Growth and Molecular Profile of Lung Cancer Cells Expressing Ectopic LKB1: Down-Regulation of the Phosphatidylinositol 3؅-Phosphate Kinase/PTEN Pathway1 Ana I. Jimenez, Paloma Fernandez, Orlando Dominguez, Ana Dopazo, and Montserrat Sanchez-Cespedes2 Molecular Pathology Program [A. I. J., P. F., M. S-C.], Genomics Unit [O. D.], and Microarray Analysis Unit [A. D.], Spanish National Cancer Center, 28029 Madrid, Spain ABSTRACT the cell cycle in G1 (8, 9). However, the intrinsic mechanism by which LKB1 activity is regulated in cells and how it leads to the suppression Germ-line mutations in LKB1 gene cause the Peutz-Jeghers syndrome of cell growth is still unknown. It has been proposed that growth (PJS), a genetic disease with increased risk of malignancies. Recently, suppression by LKB1 is mediated through p21 in a p53-dependent LKB1-inactivating mutations have been identified in one-third of sporadic lung adenocarcinomas, indicating that LKB1 gene inactivation is critical in mechanism (7). In addition, it has been observed that LKB1 binds to tumors other than those of the PJS syndrome. However, the in vivo brahma-related gene 1 protein (BRG1) and this interaction is required substrates of LKB1 and its role in cancer development have not been for BRG1-induced growth arrest (10). Similar to what happens in the completely elucidated. Here we show that overexpression of wild-type PJS, Lkb1 heterozygous knockout mice show gastrointestinal hamar- LKB1 protein in A549 lung adenocarcinomas cells leads to cell-growth tomatous polyposis and frequent hepatocellular carcinomas (11, 12). suppression. To examine changes in gene expression profiles subsequent to Interestingly, the hamartomas, but not the malignant tumors, arising in exogenous wild-type LKB1 in A549 cells, we used cDNA microarrays.
    [Show full text]
  • The Expression Patterns and the Prognostic Roles of PTPN Family Members in Digestive Tract Cancers
    Preprint: Please note that this article has not completed peer review. The expression patterns and the prognostic roles of PTPN family members in digestive tract cancers CURRENT STATUS: UNDER REVIEW Jing Chen The First Affiliated Hospital of China Medical University Xu Zhao Liaoning Vocational College of Medicine Yuan Yuan The First Affiliated Hospital of China Medical University Jing-jing Jing The First Affiliated Hospital of China Medical University [email protected] Author ORCiD: https://orcid.org/0000-0002-9807-8089 DOI: 10.21203/rs.3.rs-19689/v1 SUBJECT AREAS Cancer Biology KEYWORDS PTPN family members, digestive tract cancers, expression, prognosis, clinical features 1 Abstract Background Non-receptor protein tyrosine phosphatases (PTPNs) are a set of enzymes involved in the tyrosyl phosphorylation. The present study intended to clarify the associations between the expression patterns of PTPN family members and the prognosis of digestive tract cancers. Method Expression profiling of PTPN family genes in digestive tract cancers were analyzed through ONCOMINE and UALCAN. Gene ontology enrichment analysis was conducted using the DAVID database. The gene–gene interaction network was performed by GeneMANIA and the protein–protein interaction (PPI) network was built using STRING portal couple with Cytoscape. Data from The Cancer Genome Atlas (TCGA) were downloaded for validation and to explore the relationship of the PTPN expression with clinicopathological parameters and survival of digestive tract cancers. Results Most PTPN family members were associated with digestive tract cancers according to Oncomine, Ualcan and TCGA data. For esophageal carcinoma (ESCA), expression of PTPN1, PTPN4 and PTPN12 were upregulated; expression of PTPN20 was associated with poor prognosis.
    [Show full text]
  • The Regulatory Roles of Phosphatases in Cancer
    Oncogene (2014) 33, 939–953 & 2014 Macmillan Publishers Limited All rights reserved 0950-9232/14 www.nature.com/onc REVIEW The regulatory roles of phosphatases in cancer J Stebbing1, LC Lit1, H Zhang, RS Darrington, O Melaiu, B Rudraraju and G Giamas The relevance of potentially reversible post-translational modifications required for controlling cellular processes in cancer is one of the most thriving arenas of cellular and molecular biology. Any alteration in the balanced equilibrium between kinases and phosphatases may result in development and progression of various diseases, including different types of cancer, though phosphatases are relatively under-studied. Loss of phosphatases such as PTEN (phosphatase and tensin homologue deleted on chromosome 10), a known tumour suppressor, across tumour types lends credence to the development of phosphatidylinositol 3--kinase inhibitors alongside the use of phosphatase expression as a biomarker, though phase 3 trial data are lacking. In this review, we give an updated report on phosphatase dysregulation linked to organ-specific malignancies. Oncogene (2014) 33, 939–953; doi:10.1038/onc.2013.80; published online 18 March 2013 Keywords: cancer; phosphatases; solid tumours GASTROINTESTINAL MALIGNANCIES abs in sera were significantly associated with poor survival in Oesophageal cancer advanced ESCC, suggesting that they may have a clinical utility in Loss of PTEN (phosphatase and tensin homologue deleted on ESCC screening and diagnosis.5 chromosome 10) expression in oesophageal cancer is frequent, Cao et al.6 investigated the role of protein tyrosine phosphatase, among other gene alterations characterizing this disease. Zhou non-receptor type 12 (PTPN12) in ESCC and showed that PTPN12 et al.1 found that overexpression of PTEN suppresses growth and protein expression is higher in normal para-cancerous tissues than induces apoptosis in oesophageal cancer cell lines, through in 20 ESCC tissues.
    [Show full text]
  • Protein Tyrosine Phosphatase PTPN3 Inhibits Lung Cancer Cell Proliferation and Migration by Promoting EGFR Endocytic Degradation
    Oncogene (2015) 34, 3791–3803 © 2015 Macmillan Publishers Limited All rights reserved 0950-9232/15 www.nature.com/onc ORIGINAL ARTICLE Protein tyrosine phosphatase PTPN3 inhibits lung cancer cell proliferation and migration by promoting EGFR endocytic degradation M-Y Li1,2, P-L Lai1, Y-T Chou3, A-P Chi1, Y-Z Mi1, K-H Khoo1,2, G-D Chang2, C-W Wu3, T-C Meng1,2 and G-C Chen1,2 Epidermal growth factor receptor (EGFR) regulates multiple signaling cascades essential for cell proliferation, growth and differentiation. Using a genetic approach, we found that Drosophila FERM and PDZ domain-containing protein tyrosine phosphatase, dPtpmeg, negatively regulates border cell migration and inhibits the EGFR/Ras/mitogen-activated protein kinase signaling pathway during wing morphogenesis. We further identified EGFR pathway substrate 15 (Eps15) as a target of dPtpmeg and its human homolog PTPN3. Eps15 is a scaffolding adaptor protein known to be involved in EGFR endocytosis and trafficking. Interestingly, PTPN3-mediated tyrosine dephosphorylation of Eps15 promotes EGFR for lipid raft-mediated endocytosis and lysosomal degradation. PTPN3 and the Eps15 tyrosine phosphorylation-deficient mutant suppress non-small-cell lung cancer cell growth and migration in vitro and reduce lung tumor xenograft growth in vivo. Moreover, depletion of PTPN3 impairs the degradation of EGFR and enhances proliferation and tumorigenicity of lung cancer cells. Taken together, these results indicate that PTPN3 may act as a tumor suppressor in lung cancer through its modulation of EGFR signaling. Oncogene (2015) 34, 3791–3803; doi:10.1038/onc.2014.312; published online 29 September 2014 INTRODUCTION sorting EGFR to multivesicular bodies.15 Recently, Ali et al.16 Reversible tyrosine protein phosphorylation by protein tyrosine showed that the ESCRT accessory protein HD-PTP/PTPN23 kinases and protein tyrosine phosphatases (PTPs) acts as a coordinates with the ubiquitin-specific peptidase UBPY to drive molecular switch that regulates a variety of biological pro- EGFR sorting to the multivesicular bodies.
    [Show full text]
  • RT² Profiler PCR Array (96-Well Format and 384-Well [4 X 96] Format)
    RT² Profiler PCR Array (96-Well Format and 384-Well [4 x 96] Format) Human Protein Phosphatases Cat. no. 330231 PAHS-045ZA For pathway expression analysis Format For use with the following real-time cyclers RT² Profiler PCR Array, Applied Biosystems® models 5700, 7000, 7300, 7500, Format A 7700, 7900HT, ViiA™ 7 (96-well block); Bio-Rad® models iCycler®, iQ™5, MyiQ™, MyiQ2; Bio-Rad/MJ Research Chromo4™; Eppendorf® Mastercycler® ep realplex models 2, 2s, 4, 4s; Stratagene® models Mx3005P®, Mx3000P®; Takara TP-800 RT² Profiler PCR Array, Applied Biosystems models 7500 (Fast block), 7900HT (Fast Format C block), StepOnePlus™, ViiA 7 (Fast block) RT² Profiler PCR Array, Bio-Rad CFX96™; Bio-Rad/MJ Research models DNA Format D Engine Opticon®, DNA Engine Opticon 2; Stratagene Mx4000® RT² Profiler PCR Array, Applied Biosystems models 7900HT (384-well block), ViiA 7 Format E (384-well block); Bio-Rad CFX384™ RT² Profiler PCR Array, Roche® LightCycler® 480 (96-well block) Format F RT² Profiler PCR Array, Roche LightCycler 480 (384-well block) Format G RT² Profiler PCR Array, Fluidigm® BioMark™ Format H Sample & Assay Technologies Description The Human Protein Phosphatases RT² Profiler PCR Array profiles the gene expression of the 84 most important and well-studied phosphatases in the mammalian genome. By reversing the phosphorylation of key regulatory proteins mediated by protein kinases, phosphatases serve as a very important complement to kinases and attenuate activated signal transduction pathways. The gene classes on this array include both receptor and non-receptor tyrosine phosphatases, catalytic subunits of the three major protein phosphatase gene families, the dual specificity phosphatases, as well as cell cycle regulatory and other protein phosphatases.
    [Show full text]
  • Live-Cell Imaging Rnai Screen Identifies PP2A–B55α and Importin-Β1 As Key Mitotic Exit Regulators in Human Cells
    LETTERS Live-cell imaging RNAi screen identifies PP2A–B55α and importin-β1 as key mitotic exit regulators in human cells Michael H. A. Schmitz1,2,3, Michael Held1,2, Veerle Janssens4, James R. A. Hutchins5, Otto Hudecz6, Elitsa Ivanova4, Jozef Goris4, Laura Trinkle-Mulcahy7, Angus I. Lamond8, Ina Poser9, Anthony A. Hyman9, Karl Mechtler5,6, Jan-Michael Peters5 and Daniel W. Gerlich1,2,10 When vertebrate cells exit mitosis various cellular structures can contribute to Cdk1 substrate dephosphorylation during vertebrate are re-organized to build functional interphase cells1. This mitotic exit, whereas Ca2+-triggered mitotic exit in cytostatic-factor- depends on Cdk1 (cyclin dependent kinase 1) inactivation arrested egg extracts depends on calcineurin12,13. Early genetic studies in and subsequent dephosphorylation of its substrates2–4. Drosophila melanogaster 14,15 and Aspergillus nidulans16 reported defects Members of the protein phosphatase 1 and 2A (PP1 and in late mitosis of PP1 and PP2A mutants. However, the assays used in PP2A) families can dephosphorylate Cdk1 substrates in these studies were not specific for mitotic exit because they scored pro- biochemical extracts during mitotic exit5,6, but how this relates metaphase arrest or anaphase chromosome bridges, which can result to postmitotic reassembly of interphase structures in intact from defects in early mitosis. cells is not known. Here, we use a live-cell imaging assay and Intracellular targeting of Ser/Thr phosphatase complexes to specific RNAi knockdown to screen a genome-wide library of protein substrates is mediated by a diverse range of regulatory and targeting phosphatases for mitotic exit functions in human cells. We subunits that associate with a small group of catalytic subunits3,4,17.
    [Show full text]
  • Phosphatases Page 1
    Phosphatases esiRNA ID Gene Name Gene Description Ensembl ID HU-05948-1 ACP1 acid phosphatase 1, soluble ENSG00000143727 HU-01870-1 ACP2 acid phosphatase 2, lysosomal ENSG00000134575 HU-05292-1 ACP5 acid phosphatase 5, tartrate resistant ENSG00000102575 HU-02655-1 ACP6 acid phosphatase 6, lysophosphatidic ENSG00000162836 HU-13465-1 ACPL2 acid phosphatase-like 2 ENSG00000155893 HU-06716-1 ACPP acid phosphatase, prostate ENSG00000014257 HU-15218-1 ACPT acid phosphatase, testicular ENSG00000142513 HU-09496-1 ACYP1 acylphosphatase 1, erythrocyte (common) type ENSG00000119640 HU-04746-1 ALPL alkaline phosphatase, liver ENSG00000162551 HU-14729-1 ALPP alkaline phosphatase, placental ENSG00000163283 HU-14729-1 ALPP alkaline phosphatase, placental ENSG00000163283 HU-14729-1 ALPPL2 alkaline phosphatase, placental-like 2 ENSG00000163286 HU-07767-1 BPGM 2,3-bisphosphoglycerate mutase ENSG00000172331 HU-06476-1 BPNT1 3'(2'), 5'-bisphosphate nucleotidase 1 ENSG00000162813 HU-09086-1 CANT1 calcium activated nucleotidase 1 ENSG00000171302 HU-03115-1 CCDC155 coiled-coil domain containing 155 ENSG00000161609 HU-09022-1 CDC14A CDC14 cell division cycle 14 homolog A (S. cerevisiae) ENSG00000079335 HU-11533-1 CDC14B CDC14 cell division cycle 14 homolog B (S. cerevisiae) ENSG00000081377 HU-06323-1 CDC25A cell division cycle 25 homolog A (S. pombe) ENSG00000164045 HU-07288-1 CDC25B cell division cycle 25 homolog B (S. pombe) ENSG00000101224 HU-06033-1 CDKN3 cyclin-dependent kinase inhibitor 3 ENSG00000100526 HU-02274-1 CTDSP1 CTD (carboxy-terminal domain,
    [Show full text]
  • Printer-Friendly Version of PTP Classification
    Nontransmembrane PTP subtypes (NT) Receptor–like PTP subtypes PTPOST http://ptp.cshl.edu & http://science.novonordisk.com/ptp Andersen et al Mol. Cell. Biol. 2001 Classification of the vertebrate family of PTPs into Nontransmembrane (NT) and Receptor-like subtypes (R) based on PTP domain sequence homology hPTPsigma (PTPRS) Inconsistent gene nomenclature is highlighted in red mPTPsigma (Ptprs) rPTPsigma (Ptprd) Subtypes xPTPsigma PTPsigma 0.1 826 cPTPsigma zPTPsigma xPTPdelta R2A 1000 hPTPdelta (PTPRD) PTPdelta mPTPdelta (Ptprd) xLAR hLAR (PTPRF) mLAR (Ptprf) LAR 834 rLAR (Ptprf) xPTPrho hPTPrho (PTPRT) 565 mPTPrho (Ptprt) PTPrho 668 hPTPmu (PTPRM) mPTPmu (Ptprm) PTPmu 1000 hPTPkappa (PTPRK) 712 mPTPkappa (Ptprk) PTPkappa R2B hPTPlamda (PTPRU) mPTPlamda (Ptprl) PTPlambda Tandem rPTPpsi (Ptpru) cPTPgamma PTP Domain rPTPgamma (Ptprg) hPTPgamma (PTPRG) PTPgamma ‘Supertype’ 1000 mPTPgamma (Ptprg) xPTPzeta cPTPzeta R5 906 hPTPzeta (PTPRZ1) PTPzeta rPTPzeta (Ptprz1) zPTPalpha (ptpra) xPTPalpha cPTPalpha hPTPalpha (PTPRA) PTPalpha 1000 mPTPalpha (Ptpra) rPTPalpha (Ptpra) R4 hPTPepsilon (PTPRE) 716 mPTPepsilon (Ptpre) rPTPepsilon (Ptpre) PTPepsilon sharkCD45 cCD45 hCD45 (PTPRC) 671 1000 mCD45 (Ptprc) R1/R6 rCD45 (Ptprc) CD45 cypcaCD45 fuCD45 hDEP1 (PTPRJ) rDEP1 (Ptprj) 940 mDEP1 (Ptprj) DEP1 hPTPbeta (PTPRB) 326 mPTPbeta (Ptprb) rPTPbeta PTPbeta hGLEPP1 (PTPRO) 486 rabPTPoc 562 rGLEPP1 (Ptpro) GLEPP1 mPTPphi (Ptpro) R3 989 hPTPS31 (PTPGMC1) rPTPGMC1 (Ptprq) PTPS31/GMC1 hSAP1 (PTPRH) mSAP1 SAP1 rPTPBEM2 mPTPESP (Ptprv) 636 rOSTPTP (Esp) OST-PTP
    [Show full text]
  • Transcriptome Profiling Reveals the Complexity of Pirfenidone Effects in IPF
    ERJ Express. Published on August 30, 2018 as doi: 10.1183/13993003.00564-2018 Early View Original article Transcriptome profiling reveals the complexity of pirfenidone effects in IPF Grazyna Kwapiszewska, Anna Gungl, Jochen Wilhelm, Leigh M. Marsh, Helene Thekkekara Puthenparampil, Katharina Sinn, Miroslava Didiasova, Walter Klepetko, Djuro Kosanovic, Ralph T. Schermuly, Lukasz Wujak, Benjamin Weiss, Liliana Schaefer, Marc Schneider, Michael Kreuter, Andrea Olschewski, Werner Seeger, Horst Olschewski, Malgorzata Wygrecka Please cite this article as: Kwapiszewska G, Gungl A, Wilhelm J, et al. Transcriptome profiling reveals the complexity of pirfenidone effects in IPF. Eur Respir J 2018; in press (https://doi.org/10.1183/13993003.00564-2018). This manuscript has recently been accepted for publication in the European Respiratory Journal. It is published here in its accepted form prior to copyediting and typesetting by our production team. After these production processes are complete and the authors have approved the resulting proofs, the article will move to the latest issue of the ERJ online. Copyright ©ERS 2018 Copyright 2018 by the European Respiratory Society. Transcriptome profiling reveals the complexity of pirfenidone effects in IPF Grazyna Kwapiszewska1,2, Anna Gungl2, Jochen Wilhelm3†, Leigh M. Marsh1, Helene Thekkekara Puthenparampil1, Katharina Sinn4, Miroslava Didiasova5, Walter Klepetko4, Djuro Kosanovic3, Ralph T. Schermuly3†, Lukasz Wujak5, Benjamin Weiss6, Liliana Schaefer7, Marc Schneider8†, Michael Kreuter8†, Andrea Olschewski1,
    [Show full text]
  • For Reference Purpose Only! Protein Phosphatase Cdc25 Combo Fluorometric Assay Kit User’S Manual for Research Use Only, Not for Use in Diagnostic Procedures
    Protein Phosphatase Cdc25 Combo Fluorometric Assay Kit User’s Manual For Research Use Only, Not for use in diagnostic procedures Fluorometric Assay Kit for Measuring Cdc25A, B and C Phosphatase Activity CycLex Protein Phosphatase Cdc25 Combo Fluorometric Assay Kit 150 Assays Cat# CY-1355 Intended Use............................................. 1 Storage..................................................... 1 Introduction ............................................. 2 Principle of the Assay.............................. 2 Materials Provided .................................. 3 Materials Required but not Provided....... 3 Precautions and Recommendations......... 3-4 Detailed Protocol..................................... 4-6 Evaluation of Results .............................. 7-11 Troubleshooting ...................................... 12 Reagent Stability ..................................... 12 References................................................ 13 Related Products....................................... 13-14 Intended Use The CycLex Research product Protein Phosphatase Cdc25 Combo Fluorometric Assay Kit is a fluorometric and non-radioactive assay designed to measure the activity of Cdc25A, B and C protein phosphatase. This 96-well assay is useful for screening inhibitors and modulators of Cdc25A, B and C activities in HTS. The kit includes all necessary components, including recombinant, human Cdc25A, B and C (catalytic domain), for use in preinvestigational drug discovery assays. This assay kit is for research use only and not
    [Show full text]
  • Molecular Basis of the Interaction of the Human Tyrosine Phosphatase
    www.nature.com/scientificreports OPEN Molecular basis of the interaction of the human tyrosine phosphatase PTPN3 with the hepatitis B virus core protein Mariano Genera1,2, Barbara Quioc‑Salomon3,4,5, Antonin Nourisson1, Baptiste Colcombet‑Cazenave1,2, Ahmed Haouz6, Ariel Mechaly6, Mariette Matondo7, Magalie Duchateau7, Alexander König8, Marc P. Windisch8, Christine Neuveut3,4,9, Nicolas Wolf1 & Célia Caillet‑Saguy1* Interactions between the hepatitis B virus core protein (HBc) and host cell proteins are poorly understood, although they may be essential for the propagation of the virus and its pathogenicity. HBc has a C‑terminal PDZ (PSD‑95, Dlg1, ZO‑1)‑binding motif (PBM) that is responsible for interactions with host PDZ domain‑containing proteins. In this work, we focused on the human protein tyrosine phosphatase non‑receptor type 3 (PTPN3) and its interaction with HBc. We solved the crystal structure of the PDZ domain of PTPN3 in complex with the PBM of HBc, revealing a network of interactions specifc to class I PDZ domains despite the presence of a C‑terminal cysteine in this atypical PBM. We further showed that PTPN3 binds the HBc protein within capsids or as a homodimer. We demonstrate that overexpression of PTPN3 signifcantly afects HBV infection in HepG2 NTCP cells. Finally, we performed proteomics studies on both sides by pull‑down assays and screening of a human PDZ domain library. We identifed a pool of human PBM‑containing proteins that might interact with PTPN3 in cells and that could be in competition with the HBc PBM during infection, and we also identifed potential cellular partners of HBc through PDZ‑PBM interactions.
    [Show full text]