Inhibition of Dual-Specificity Phosphatase 26 by Ethyl-3,4-Dephostatin: Ethyl-3,4-Dephostatin As a Multiphosphatase Inhibitor

Total Page:16

File Type:pdf, Size:1020Kb

Inhibition of Dual-Specificity Phosphatase 26 by Ethyl-3,4-Dephostatin: Ethyl-3,4-Dephostatin As a Multiphosphatase Inhibitor ORIGINAL ARTICLES College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea Inhibition of dual-specificity phosphatase 26 by ethyl-3,4-dephostatin: Ethyl-3,4-dephostatin as a multiphosphatase inhibitor HUIYUN SEO, SAYEON CHO Received September 30, 2015, accepted November 6, 2015 Sayeon Cho, Ph.D., College of Pharmacy, Chung-Ang University, Seoul 156-756, Republic of Korea [email protected] Pharmazie 71: 196–200 (2016) doi: 10.1691/ph.2016.5803 Protein tyrosine phosphatases (PTPs) regulate protein function by dephosphorylating phosphorylated proteins in many signaling cascades and some of them have been targets for drug development against many human diseases. There have been many reports that some chemical inhibitors could regulate particular phosphatases. However, there was no extensive study on specificity of inhibitors towardss phosphatases. We investigated the effects of ethyl-3,4-dephostatin, a potent inhibitor of five PTPs including PTP-1B and Src homology-2-containing protein tyrosine phosphatase-1 (SHP-1), on thirteen other PTPs using in vitro phosphatase assays. Of them, dual-specificity protein phosphatase 26 (DUSP26), which inhibits mitogen-activated protein kinase (MAPK) and p53 tumor suppressor and is known to be overexpressed in anaplastic thyroid carcinoma, was inhibited by ethyl- 3,4-dephostatin in a concentration-dependent manner. Kinetic studies with ethyl-3,4-dephostatin and DUSP26 revealed competitive inhibition, suggesting that ethyl-3,4-dephostatin binds to the catalytic site of DUSP26 like other substrate PTPs. Moreover, ethyl-3,4-dephostatin protects DUSP26-mediated dephosphorylation of p38, a member of the MAPK family, and p53. Taken together, these results suggest that ethyl-3,4-dephostatin functions as a multiphosphatase inhibitor and is useful as a therapeutic agent for cancers overexpressing DUSP26. 1. Introduction DUSPs are able to control the activation of MAPKs by dephos- Most cellular signal transductions in mammalian cells are regulated phorylating residues located on the activation loop of MAPKs, by several post-translational modifications, including phosphory- thus modifying the cellular signal (Stoker 2005). Modulation of lation, acetylation, methylation, ubiquitination, hydroxylation, and PTP activity is involved in regulating various cellular biological nitration (Hunter 2000). From among these, protein phosphory- functions and disease susceptibility (Fischer et al. 1991). There- lation regulates various essential cellular processes such as gene fore, chemical compounds that regulate the activity of PTPs may expression, cell growth, proliferation, differentiation, cell cycle be useful in the treatment of diseases such as cancer, inflammation, arrest, and apoptosis (Ciesla et al. 2011). Protein phosphorylation and diabetes. However, development of specific therapeutic PTP occurs on serine, threonine, or tyrosine residues and is regulated inhibitors is a critical challenge since over 100 PTPs paly diverse by protein kinases and protein phosphatases (Ciesla et al. 2011; roles by acting on a broad range of protein targets and the overall Franklin and Kraft 1997; Schlessinger 2000). Among the protein active site pockets of PTPs are known to be shallower than those of phosphatases, the protein tyrosine phosphatase (PTP) superfamily kinases. In addition, knowledge of the selectivity of PTP inhibitors is composed of over 100 members divided into four main fami- for their target phosphatases will provide important information lies based on the amino acid sequences of their catalytic domains. for the prediction of the effects of inhibitors in vivo. The four families include the class I cysteine (Cys)-based PTPs, Ethyl-3,4-dephostatin is known to inhibit PTP-1B and Src homol- the class II Cys-based PTPs, the class III Cys-based PTPs, and ogy-2-containing protein tyrosine phosphatase-1 (SHP-1) (Suzuki the aspartic acid-based PTPs (Patterson et al. 2009). Dual-speci- et al. 2001). In previous reports, we showed that DUSP14, ficity phosphatases (DUSPs) are a heterogeneous group of protein DUSP22, and protein tyrosine phosphatase non-receptor type 2 phosphatases that belong to a subclass of the class I Cys-based (PTPN2) were inhibited by ethyl-3,4-dephostatin in a competitive PTPs. DUSPs dephosphorylate both phospho-tyrosine and serine/ manner (Seo and Cho 2011a, b; Seo et al. 2011). We have screened threonine residues. for additional targets of ethyl-3,4-dephostatin by in vitro phospha- Protein phosphorylation and dephosphorylation lead to confor- tase assays with thirteen human PTPs. Dual-specificity phospha- mational changes in target proteins and then regulate their protein tase 26 (DUSP26) was identified as a novel target of ethyl-3,4- activity. The mitogen-activated protein kinase (MAPK) pathway is dephostatin. DUSP26, also known as mitogen-activated protein a protein phosphorylation-mediated signal transduction pathway kinase phophatase-8, is a member of atypical DUSPs, first char- that transmits signals from extracellular stimuli to genes in the acterized using a yeast two-hybrid screen by Mivechi’s research nucleus (Johnson and Lapadat 2002). The major subfamilies of group in 2005 (Hu and Mivechi 2006). DUSP26 overexpression MAPK are c-Jun N-terminal kinase (JNK), extracellular signal-reg- enhances colony formation in anaplastic thyroid carcinoma by ulated kinase (ERK), and p38 (Schaeffer and Weber 1999). Several inhibiting p38 kinase, while knock-down of DUSP26 inhibits cell proliferation and induces apoptosis (Yu et al. 2007). In addition, DUSP26 physically interacts with and functionally inhibits p53, Abbreviations: PTP, protein tyrosine phosphatase; DUSP, which results in reduction of p53 activity and its down-stream dual-specificity protein phosphatase; SHP-1, Src homolo- targets that are induced in response to genotoxic stress (Shang et gy-2-containing protein tyrosine phosphatase-1; MAPK, mito- al. 2010). In this study, we show that ethyl-3,4-dephostatin specifi- gen-activated protein kinase cally inhibits DUSP26 phosphatase activity and thus regulates p53 and p38, substrates of DUSP26. 196 Pharmazie 71 (2016) ORIGINAL ARTICLES Fig. 1: Inhibitory effect of ethyl-3,4-dephostatin on DUSP26 and kinetic analysis of DUSP26 inhibition. (A) Chemical structure of ethyl-3,4-dephostatin. (B) DUSP26 was incubated with various concentrations (0, 5, 10, 15, or 20 μM) of ethyl-3,4-dephostatin at 37 ˚C for 30 min. Fluorescence emission from the product was measured. (C) Lineweaver-Burk plots of DUSP26 were generated from the data. Each experiment was performed in triplicate. 2. Investigations and results Protein tyrosine Classification IC50 (μM) References 2.1. Effect of ethyl-3,4-dephostatin on DUSP26 phosphatase (n = 3) Ethyl-3,4-dephostatin is a stable synthetic analog of dephostatin PTPN2 NRPTPs 6.5 ± 0.54 (Seo and Cho that inhibits PTP-1B and SHP-1 (Fig. 1A) (Suzuki et al. 2001). 2011a) Since PTP-1B and SHP-1 are related to diabetes and cancer LAR Transmembrane >100 (μg/ml) * (Suzuki et al. 2001) (Kundu et al. 2010; Zhang and Lee 2003), respectively, ethyl-3,4- Classical PTPs dephostatin might be useful for the treatment of those diseases. CD45 Transmembrane 28.5 (μg/ml) * (Suzuki et al. 2001) However, the selectivity of ethyl-3,4-dephostatin towardss phos- Classical PTPs phatases should be determined before it is considered as a thera- DUSP3 Atypical DUSPs >200 This study peutic reagent. We previously reported that DUSP14, DUSP22, and PTPN2, which DUSP6 MKPs >200 This study play roles in MAPK signaling (Jeffrey et al. 2007), were regulated DUSP7 MKPs >50 This study by ethyl-3,4-dephostatin (Seo and Cho 2011a, b; Seo et al. 2011). We tested additional thirteen recombinant human PTPs with in vitro DUSP8 MKPs >50 This study phosphatase assays to determine the effect of ethyl-3,4-dephostatin DUSP13B Atypical DUSPs >200 This study on the regulation of PTPs. An inhibition curve was plotted for each DUSP18 Atypical DUSPs >100 This study PTP and IC50 values were calculated. As shown in the Table, we found that DUSP26 was inhibited by ethyl-3,4-dephostatin. DUSP23 Atypical DUSPs >200 This study PTPN6 NRPTPs >50 This study Table: Inhibition of PTPs by ethyl-3,4-dephostatin in vitro PTPN7 NRPTPs >100 This study PTPN20 NRPTPs >50 This study Protein tyrosine Classification IC50 (μM) References phosphatase (n = 3) SSH3 Slinghots >50 This study DUSP26 Atypical DUSPs 6.8 ± 0.41 This study ACP1 Class II Cys-Based >200 This study PTP-1B NRPTPs 0.58 (μg/ml) * (Suzuki et al. 2001) PTPs SHP-1 NRPTPs 0.96 (μg/ml) * (Suzuki et al. 2001) * The values were obtained from the experimental conditions that were different from ours. DUSP14 Atypical DUSPs 9.7 ± 0.02 (Seo and Cho 2011b) Each experiment was performed in triplicate. In vitro PTP assay was processed as described in Experimental. Data are presented DUSP22 Atypical DUSPs 3.06 ± 0.07 (Seo et al. 2011) as mean ± SEM. Pharmazie 71 (2016) 197 ORIGINAL ARTICLES The inhibitory activity of ethyl-3,4-dephostatin on DUSP26 was concentration-dependent as shown in Fig. 1B. Ethyl-3,4-de- phostatin inhibits DUSP26 with an IC50 of 6.8±0.4 μM. The Line- weaver-Burk plot showed that the Ki was 5.9 μM (Fig. 1C). We found that ethyl-3,4-dephostatin acts as a competitive inhibitor of DUSP26, suggesting that ethyl-3,4-dephostatin suppresses the activity of DUSP26 through binding to the catalytic site. 2.2. Effect of ethyl-3,4-dephostatin on DUSP26 ex- pressed in mammalian cells To confirm the inhibitory effect of ethyl-3,4-dephostatin on DUSP26 in mammalian cells, HEK 293 cells transfected with FLAG-DUSP26 were treated with various concentrations of ethyl- 3,4-dephostatin. Phosphatase activity was measured after immu- noprecipitation of DUSP26 from cell lysates with anti-FLAG M2 agarose beads. As with the in vitro phosphatase assay, this result showed that ethyl-3,4-dephostatin inhibits the phosphatase activity of DUSP26 expressed in mammalian cells (Fig. 2). Fig. 3: Ethyl-3,4-dephostatin inhibits the action of DUSP26 on p38 in vitro and in cells.
Recommended publications
  • Clinical and Biological Features of PTPN2-Deleted Adult and Pediatric T-Cell Acute Lymphoblastic Leukemia
    REGULAR ARTICLE Clinical and biological features of PTPN2-deleted adult and pediatric T-cell acute lymphoblastic leukemia Marion Alcantara,1 Mathieu Simonin,1-3 Ludovic Lhermitte,1 Aurore Touzart,1 Marie Emilie Dourthe,1,4 Mehdi Latiri,1 Nathalie Grardel,5 Jean Michel Cayuela,6 Yves Chalandon,7 Carlos Graux,8 Herve´ Dombret,9 Norbert Ifrah,10 Arnaud Petit,2,3 Elizabeth Macintyre,1 Andre´ Baruchel,4 Nicolas Boissel,9 and Vahid Asnafi1 1Universite´ Paris Descartes Sorbonne Cite,´ Institut Necker-Enfants Malades, Institut National de Recherche Medicale´ U1151, Laboratory of Onco-Hematology, Assistance Publique–Hopitauxˆ de Paris, Hopitalˆ Necker Enfants-Malades, Paris, France; 2Department of Pediatric Hematology and Oncology, Assistance Publique–Hopitauxˆ de Paris, Groupe Hospitalier des Hopitauxˆ Universitaires Est Parisien, Armand Trousseau Hospital, Paris, France; 3Sorbonne Universites,´ Universite´ Pierre et Marie Curie Universite´ Paris 06, Unite´ Mixte de Recherche S 938, Centre de Recherche Saint-Antoine, Groupe de Recherche (GRC) Clinique n°07, GRC Myeloprolif´ erations´ aigues¨ et chroniques, Paris, France; 4Department of Pediatric Hematology and Immunology, Assistance Publique–Hopitauxˆ de Paris, Hopitalˆ Universitaire Robert-Debre,´ Paris, France; 5Laboratory of Hematology, Centre Hospitalier Regional´ Universitaire de Lille, Lille, France; 6Laboratory of Hematology, Saint-Louis Hospital, Assistance Publique–Hopitauxˆ de Paris, Paris, France; 7Swiss Group for Clinical Cancer Research, Bern, Hopitalˆ Universitaire, Geneva, Switzerland; 8Universite´ Catholique de Louvain, Centre Hospitalier Universitaire, Namur, Yvoir, Belgium; 9Adolescent and Young Adult Hematology Unit, Saint-Louis Hospital, Assistance Publique–Hopitauxˆ de Paris, Paris, France; and 10Centre Hospitalier Universitaire d’Angers, Angers, France Protein tyrosine phosphatase nonreceptor type 2 (PTPN2) is a phosphatase known to be Key Points a tumor suppressor gene in T-cell acute lymphoblastic leukemia (T-ALL).
    [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]
  • Inflammatory Cytokine Signalling by Protein Tyrosine Phosphatases in Pancreatic Β-Cells
    59 4 W J STANLEY and others PTPN1 and PTPN6 modulate 59: 4 325–337 Research cytokine signalling in β-cells Differential regulation of pro- inflammatory cytokine signalling by protein tyrosine phosphatases in pancreatic β-cells William J Stanley1,2, Prerak M Trivedi1,2, Andrew P Sutherland1, Helen E Thomas1,2 and Esteban N Gurzov1,2,3 Correspondence should be addressed 1 St. Vincent’s Institute of Medical Research, Melbourne, Australia to E N Gurzov 2 Department of Medicine, St. Vincent’s Hospital, The University of Melbourne, Melbourne, Australia Email 3 ULB Center for Diabetes Research, Universite Libre de Bruxelles (ULB), Brussels, Belgium esteban.gurzov@unimelb. edu.au Abstract Type 1 diabetes (T1D) is characterized by the destruction of insulin-producing β-cells Key Words by immune cells in the pancreas. Pro-inflammatory including TNF-α, IFN-γ and IL-1β f pancreatic β-cells are released in the islet during the autoimmune assault and signal in β-cells through f protein tyrosine phosphorylation cascades, resulting in pro-apoptotic gene expression and eventually phosphatases β-cell death. Protein tyrosine phosphatases (PTPs) are a family of enzymes that regulate f PTPN1 phosphorylative signalling and are associated with the development of T1D. Here, we f PTPN6 observed expression of PTPN6 and PTPN1 in human islets and islets from non-obese f cytokines diabetic (NOD) mice. To clarify the role of these PTPs in β-cells/islets, we took advantage f inflammation Journal of Molecular Endocrinology of CRISPR/Cas9 technology and pharmacological approaches to inactivate both proteins. We identify PTPN6 as a negative regulator of TNF-α-induced β-cell death, through JNK- dependent BCL-2 protein degradation.
    [Show full text]
  • SSH3 Antibody Cat
    SSH3 Antibody Cat. No.: 55-994 SSH3 Antibody SSH3 Antibody immunohistochemistry analysis in formalin Flow cytometric analysis of Hela cells (right histogram) fixed and paraffin embedded human breast carcinoma compared to a negative control cell (left histogram).FITC- followed by peroxidase conjugation of the secondary conjugated goat-anti-rabbit secondary antibodies were antibody and DAB staining. used for the analysis. Confocal immunofluorescent analysis of SSH3 Antibody with Hela cell followed by Alexa Fluor 488-conjugated goat anti-rabbit lgG (green). Actin filaments have been labeled with Alexa Fluor555 phalloidin (red). DAPI was used to stain the cell nuclear (blue). Specifications October 1, 2021 1 https://www.prosci-inc.com/ssh3-antibody-55-994.html HOST SPECIES: Rabbit SPECIES REACTIVITY: Human This SSH3 antibody is generated from rabbits immunized with a KLH conjugated synthetic IMMUNOGEN: peptide between 575-602 amino acids from the C-terminal region of human SSH3. TESTED APPLICATIONS: Flow, IF, IHC-P, WB For WB starting dilution is: 1:1000 For IHC-P starting dilution is: 1:50~100 APPLICATIONS: For FACS starting dilution is: 1:10~50 For IF starting dilution is: 1:10~50 PREDICTED MOLECULAR 73 kDa WEIGHT: Properties This antibody is purified through a protein A column, followed by peptide affinity PURIFICATION: purification. CLONALITY: Polyclonal ISOTYPE: Rabbit Ig CONJUGATE: Unconjugated PHYSICAL STATE: Liquid BUFFER: Supplied in PBS with 0.09% (W/V) sodium azide. CONCENTRATION: batch dependent Store at 4˚C for three months and -20˚C, stable for up to one year. As with all antibodies STORAGE CONDITIONS: care should be taken to avoid repeated freeze thaw cycles.
    [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]
  • SSH3 Promotes Malignant Progression of HCC by Activating FGF1-Mediated FGF/FGFR Pathway
    European Review for Medical and Pharmacological Sciences 2020; 24: 11561-11568 SSH3 promotes malignant progression of HCC by activating FGF1-mediated FGF/FGFR pathway Q.-S. SHI, Y.-H. ZHANG, J. LONG, Z.-L. QIAN, C.-X. HU Department of Oncology Minimally Invasive Interventional Radiology, Beijing Youan Hospital, Capital Medical University, Beijing, China Abstract. – OBJECTIVE: To investigate the Introduction impact of silencing SSH3 on the expression of FGF/FGFR pathway-related genes FGF1, FG- As the third largest cancer killer in the world, FR1, and FGFR2 in hepatocellular carcinoma the mortality and morbidity of hepatocellular car- (HCC) cell line, so as to further understand the role of SSH3 in proliferation and apoptosis of cinoma (HCC) are increasing year by year around 1,2 HCC cells. the world . In China, it has also become one of PATIENTS AND METHODS: TWe first de- the most common tumors with the highest degree tected SSH3 expression in 51 pairs of tumor of malignancy, and more than 300,000 patients tissue specimens and adjacent tissues collect- die of this cancer every year, which is related to ed from HCC patients through quantitative Re- its biological characteristics of prone to recur- al Time-Polymerase Chain Reaction (qRT-PCR) 3,4 and analyzed the interplay between SSH3 ex- rence and metastasis . The development of HCC pression and clinical characteristics of HCC is a complex process involving multiple genes in patients. In vitro, after SSH3-silenced human vivo and coordinated by multiple steps5,6. At pres- HCC cell line was constructed by lentiviral trans- ent, surgery is one of the most effective methods fection, Cell Counting Kit-8 (CCK-8), cell cloning for HCC treatment, however, the high incidence assay, and flow apoptosis methods were con- of intrahepatic metastasis and vascular invasion ducted to explore the HCC cell functions.
    [Show full text]
  • Protein Tyrosine Phosphatase Non-Receptor Type 2 Function in Dendritic Cells Is Crucial to Maintain Tissue Tolerance
    ORIGINAL RESEARCH published: 18 August 2020 doi: 10.3389/fimmu.2020.01856 Protein Tyrosine Phosphatase Non-Receptor Type 2 Function in Dendritic Cells Is Crucial to Maintain Tissue Tolerance Larissa Hering 1, Egle Katkeviciute 1, Marlene Schwarzfischer 1, Philipp Busenhart 1, Claudia Gottier 1, Dunja Mrdjen 2, Juliana Komuczki 2†, Marcin Wawrzyniak 1, Silvia Lang 1, Kirstin Atrott 1, Burkhard Becher 2, Gerhard Rogler 1,3, Michael Scharl 1,3* and Marianne R. Spalinger 1 1 Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland, 2 Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland, 3 Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland Edited by: Michele Marie Kosiewicz, University of Louisville, United States Protein tyrosine phosphatase non-receptor type 2 (PTPN2) plays a pivotal role in Reviewed by: immune homeostasis and has been associated with human autoimmune and chronic Cong-Yi Wang, inflammatory diseases. Though PTPN2 is well-characterized in lymphocytes, little is Tongji Medical College, China Andrew L. Mellor, known about its function in innate immune cells. Our findings demonstrate that dendritic Newcastle University, United Kingdom cell (DC)-intrinsic PTPN2 might be the key to explain the central role for PTPN2 in *Correspondence: the immune system to maintain immune tolerance. Partial genetic PTPN2 ablation in Michael Scharl DCs resulted in spontaneous inflammation, particularly in skin, liver, lung and kidney 22 [email protected] weeks post-birth. DC-specific PTPN2 controls steady-state immune cell composition † Present address: and even incomplete PTPN2 deficiency in DCs resulted in enhanced organ infiltration of Juliana Komuczki, Roche Innovation Center Basel, conventional type 2 DCs, accompanied by expansion of IFNγ-producing effector T-cells.
    [Show full text]
  • (12) United States Patent (10) Patent No.: US 7,662,770 B2 Kinch (45) Date of Patent: Feb
    USOO766277OB2 (12) United States Patent (10) Patent No.: US 7,662,770 B2 Kinch (45) Date of Patent: Feb. 16, 2010 (54) LOW MOLECULARWEIGHT PROTEIN WO WOO1? 12172 A1 2, 2001 TYROSINE PHOSPHATASE (LMW-PTP) ASA WO WO 01/12840 A2 2, 2001 DAGNOSTIC AND THERAPEUTIC TARGET WO WOO3,O94859 A2 11/2003 WO WOO3,O993 13 A1 12/2003 75 WO WO 2004/O14292 A2 2, 2004 (75) Inventor: Michael S. Kinch, Laytonville, MD WO WO 2004/O14292 A3 2, 2004 WO WO 2005/051307 A2 6, 2005 (73) Assignee: Purdue Research Foundation, West W W SE A. 3. LaFayette, IN (US) WO WO 2005/055948 A3 6, 2005 - WO WO 2005/056766 A2 6, 2005 (*) Notice: Subject to any disclaimer, the term of this patent is extended or adjusted under 35 OTHER PUBLICATIONS U.S.C. 154(b) by 350 days. Carles-Kinchet al., “Antibody targeting of the EphA2tyrosine kinase inhibits malignant cell behavior.” May 15, 2002 Cancer Research (21) Appl. No.: 10/515,358 62(10):2840-2847. Parket al., “Low-molecular-weight protein tyrosine phosphatase is a (22) PCT Filed: May 22, 2003 positive component of the fibroblast growth factor receptor signaling pathway.” May 15, 2002 Molecular and Cellular Biology (86). PCT No.: PCT/USO3A16269 22(10):3404-3414. Souza et al., “From immune response to cancer: a spot on the low S371 (c)(1), molecular weight protein tyrosine phosphatase.” Apr. 2009 Cellular (2), (4) Date: Aug. 11, 2005 and Molecular Life Sciences 66(6):1140-1153. Available online on Nov. 11, 2008. (87) PCT Pub.
    [Show full text]
  • Development and Validation of a Protein-Based Risk Score for Cardiovascular Outcomes Among Patients with Stable Coronary Heart Disease
    Supplementary Online Content Ganz P, Heidecker B, Hveem K, et al. Development and validation of a protein-based risk score for cardiovascular outcomes among patients with stable coronary heart disease. JAMA. doi: 10.1001/jama.2016.5951 eTable 1. List of 1130 Proteins Measured by Somalogic’s Modified Aptamer-Based Proteomic Assay eTable 2. Coefficients for Weibull Recalibration Model Applied to 9-Protein Model eFigure 1. Median Protein Levels in Derivation and Validation Cohort eTable 3. Coefficients for the Recalibration Model Applied to Refit Framingham eFigure 2. Calibration Plots for the Refit Framingham Model eTable 4. List of 200 Proteins Associated With the Risk of MI, Stroke, Heart Failure, and Death eFigure 3. Hazard Ratios of Lasso Selected Proteins for Primary End Point of MI, Stroke, Heart Failure, and Death eFigure 4. 9-Protein Prognostic Model Hazard Ratios Adjusted for Framingham Variables eFigure 5. 9-Protein Risk Scores by Event Type This supplementary material has been provided by the authors to give readers additional information about their work. Downloaded From: https://jamanetwork.com/ on 10/02/2021 Supplemental Material Table of Contents 1 Study Design and Data Processing ......................................................................................................... 3 2 Table of 1130 Proteins Measured .......................................................................................................... 4 3 Variable Selection and Statistical Modeling ........................................................................................
    [Show full text]
  • Pharmacological Targeting of the Mitochondrial Phosphatase PTPMT1 by Dahlia Doughty Shenton Department of Biochemistry Duke
    Pharmacological Targeting of the Mitochondrial Phosphatase PTPMT1 by Dahlia Doughty Shenton Department of Biochemistry Duke University Date: May 1 st 2009 Approved: ___________________________ Dr. Patrick J. Casey, Supervisor ___________________________ Dr. Perry J. Blackshear ___________________________ Dr. Anthony R. Means ___________________________ Dr. Christopher B. Newgard ___________________________ Dr. John D. York Dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Department of Biochemistry in the Graduate School of Duke University 2009 ABSTRACT Pharmacological Targeting of the Mitochondrial Phosphatase PTPMT1 by Dahlia Doughty Shenton Department of Biochemistry Duke University Date: May 1 st 2009 Approved: ___________________________ Dr. Patrick J. Casey, Supervisor ___________________________ Dr. Perry J. Blackshear ___________________________ Dr. Anthony R. Means ___________________________ Dr. Christopher B. Newgard ___________________________ Dr. John D. York An abstract of a dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Department of Biochemistry in the Graduate School of Duke University 2009 Copyright by Dahlia Doughty Shenton 2009 Abstract The dual specificity protein tyrosine phosphatases comprise the largest and most diverse group of protein tyrosine phosphatases and play integral roles in the regulation of cell signaling events. The dual specificity protein tyrosine phosphatases impact multiple
    [Show full text]
  • Molecular Programming of Tumor-Infiltrating CD8 T Cells and IL15 Resistance
    Published OnlineFirst August 2, 2016; DOI: 10.1158/2326-6066.CIR-15-0178 Research Article Cancer Immunology Research Molecular Programming of Tumor-Infiltrating CD8þ T Cells and IL15 Resistance Andrew L. Doedens1, Mark P. Rubinstein1,2, Emilie T. Gross3, J. Adam Best1, David H. Craig2, Megan K. Baker2, David J. Cole2, Jack D. Bui3, and Ananda W. Goldrath1 Abstract Despite clinical potential and recent advances, durable immu- in the lung and spleen were activated and dramatically expanded. þ notherapeutic ablation of solid tumors is not routinely achieved. Tumor-infiltrating CD8 T cells exhibited cell-extrinsic and cell- IL15expandsnaturalkillercell(NK),naturalkillerTcell(NKT)and intrinsic resistance to IL15. Our data showed that in the case of þ CD8 T-cell numbers and engages the cytotoxic program, and thus persistent viral or tumor antigen, single-agent systemic IL15cx is under evaluation for potentiation of cancer immunotherapy. We treatment primarily expanded antigen-irrelevant or extratumoral þ found that short-term therapy with IL15 bound to soluble IL15 CD8 Tcells.Weidentified exhaustion, tissue-resident memory, þ receptor a–Fc (IL15cx; a form of IL15 with increased half-life and and tumor-specific molecules expressed in tumor-infiltrating CD8 activity) was ineffective in the treatment of autochthonous PyMT T cells, which may allow therapeutic targeting or programming þ murine mammary tumors, despite abundant CD8 T-cell infiltra- of specific subsets to evade loss of function and cytokine resist- tion. Probing of this poor responsiveness revealed that IL15cx ance, and, in turn, increase the efficacy of IL2/15 adjuvant cytokine þ only weakly activated intratumoral CD8 T cells, even though cells therapy.
    [Show full text]