Dual Specificity Phosphatases from Molecular Mechanisms to Biological Function

Total Page:16

File Type:pdf, Size:1020Kb

Dual Specificity Phosphatases from Molecular Mechanisms to Biological Function International Journal of Molecular Sciences Dual Specificity Phosphatases From Molecular Mechanisms to Biological Function Edited by Rafael Pulido and Roland Lang Printed Edition of the Special Issue Published in International Journal of Molecular Sciences www.mdpi.com/journal/ijms Dual Specificity Phosphatases Dual Specificity Phosphatases From Molecular Mechanisms to Biological Function Special Issue Editors Rafael Pulido Roland Lang MDPI • Basel • Beijing • Wuhan • Barcelona • Belgrade Special Issue Editors Rafael Pulido Roland Lang Biocruces Health Research Institute University Hospital Erlangen Spain Germany Editorial Office MDPI St. Alban-Anlage 66 4052 Basel, Switzerland This is a reprint of articles from the Special Issue published online in the open access journal International Journal of Molecular Sciences (ISSN 1422-0067) from 2018 to 2019 (available at: https: //www.mdpi.com/journal/ijms/special issues/DUSPs). For citation purposes, cite each article independently as indicated on the article page online and as indicated below: LastName, A.A.; LastName, B.B.; LastName, C.C. Article Title. Journal Name Year, Article Number, Page Range. ISBN 978-3-03921-688-8 (Pbk) ISBN 978-3-03921-689-5 (PDF) c 2019 by the authors. Articles in this book are Open Access and distributed under the Creative Commons Attribution (CC BY) license, which allows users to download, copy and build upon published articles, as long as the author and publisher are properly credited, which ensures maximum dissemination and a wider impact of our publications. The book as a whole is distributed by MDPI under the terms and conditions of the Creative Commons license CC BY-NC-ND. Contents About the Special Issue Editors ..................................... vii Rafael Pulido and Roland Lang Dual Specificity Phosphatases: From Molecular Mechanisms to Biological Function Reprinted from: Int. J. Mol. Sci. 2019, 20, 4372, doi:10.3390/ijms20184372 .............. 1 Gema Gonz´alez-Rubio, Teresa Fern´andez-Acero, Humberto Mart´ın and Mar´ıa Molina Mitogen-Activated Protein Kinase Phosphatases (MKPs) in Fungal Signaling: Conservation, Function, and Regulation Reprinted from: Int. J. Mol. Sci. 2019, 20, 1709, doi:10.3390/ijms20071709 .............. 5 Roland Lang and Faizal A.M. Raffi Dual-Specificity Phosphatases in Immunity and Infection: An Update Reprinted from: Int. J. Mol. Sci. 2019, 20, 2710, doi:10.3390/ijms20112710 .............. 21 Grace C. A. Manley, Lisa C. Parker and Yongliang Zhang Emerging Regulatory Roles of Dual-Specificity Phosphatases in Inflammatory Airway Disease Reprinted from: Int. J. Mol. Sci. 2019, 20, 678, doi:10.3390/ijms20030678 ............... 47 Yashwanth Subbannayya, Sneha M. Pinto, Korbinian B¨osl, T. S. Keshava Prasad and Richard K. Kandasamy Dynamics of Dual Specificity Phosphatases and Their Interplay with Protein Kinases in Immune Signaling Reprinted from: Int. J. Mol. Sci. 2019, 20, 2086, doi:10.3390/ijms20092086 .............. 70 Hsueh-Fen Chen, Huai-Chia Chuang and Tse-Hua Tan Regulation of Dual-Specificity Phosphatase (DUSP) Ubiquitination and Protein Stability Reprinted from: Int. J. Mol. Sci. 2019, 20, 2668, doi:10.3390/ijms20112668 .............. 92 Marta Jim´enez-Mart´ınez, Konstantinos Stamatakis and Manuel Fresno The Dual-Specificity Phosphatase 10 (DUSP10): Its Role in Cancer, Inflammation, and Immunity Reprinted from: Int. J. Mol. Sci. 2019, 20, 1626, doi:10.3390/ijms20071626 ..............109 Jinhui Li, Xiantao Wang, William E. Ackerman IV, Abel J. Batty, Sean G. Kirk, William M. White, Xianxi Wang, Dimitrios Anastasakis, Lobelia Samavati, Irina Buhimschi, and et al. Dysregulation of Lipid Metabolism in Mkp-1 Deficient Mice during Gram-Negative Sepsis Reprinted from: Int. J. Mol. Sci. 2018, 19, 3904, doi:10.3390/ijms19123904 ..............122 Thikryat Neamatallah, Shilan Jabbar, Rothwelle Tate, Juliane Schroeder, Muhannad Shweash, James Alexander and Robin Plevin Whole Genome Microarray Analysis of DUSP4-Deletion Reveals A Novel Role for MAP Kinase Phosphatase-2 (MKP-2) in Macrophage Gene Expression and Function Reprinted from: Int. J. Mol. Sci. 2019, 20, 3434, doi:10.3390/ijms20143434 ..............143 Fan Wu, Robert D. McCuaig, Christopher R. Sutton, Abel H. Y. Tan, Yoshni Jeelall, Elaine G. Bean, Jin Dai, Thiru Prasanna, Jacob Batham, Laeeq Malik, et al. Nuclear-Biased DUSP6 Expression is Associated with Cancer Spreading Including Brain Metastasis in Triple-Negative Breast Cancer Reprinted from: Int. J. Mol. Sci. 2019, 20, 3080, doi:10.3390/ijms20123080 ..............160 v Yuming Cao, Dallas A. Banks, Andrew M. Mattei, Alexys T. Riddick, Kirstin M. Reed, Ashley M. Zhang, Emily S. Pickering and Shant´a D. Hinton Pseudophosphatase MK-STYX Alters Histone Deacetylase 6 Cytoplasmic Localization, Decreases Its Phosphorylation, and Increases Detyrosination of Tubulin Reprinted from: Int. J. Mol. Sci. 2019, 20, 1455, doi:10.3390/ijms20061455 ..............173 Caroline E. Nunes-Xavier, Laura Zaldumbide, Olaia Aurtenetxe, Ricardo L´opez-Almaraz, Jos´eI.L´opez and Rafael Pulido Dual-Specificity Phosphatases in Neuroblastoma Cell Growth and Differentiation Reprinted from: Int. J. Mol. Sci. 2019, 20, 1170, doi:10.3390/ijms20051170 ..............189 Raquel P´erez-Sen, Mar´ıa Jos´e Queipo, Juan Carlos Gil-Redondo, Felipe Ortega, Rosa G´omez-Villafuertes, Mar´ıa Teresa Miras-Portugal and Esmerilda G. Delicado Dual-Specificity Phosphatase Regulation in Neurons and Glial Cells Reprinted from: Int. J. Mol. Sci. 2019, 20, 1999, doi:10.3390/ijms20081999 ..............208 vi About the Special Issue Editors Rafael Pulido is an Ikerbasque Research Professor at Biocruces Bizkaia Health Research Institute (Barakaldo, Bilbao, Spain). His laboratory investigates the role of protein tyrosine phosphatases in human disease, and studies the involvement of phosphatases and kinases in human cancer and cancer-associated hereditary syndromes, with a focus on the PTEN tumor suppressor. His professional interest is the identification, characterization, and validation of new molecular biomarkers and targets for anti-cancer precision therapies. Roland Lang is an MD specialized in Medical Microbiology and an Associate Professor of Innate Immunity at the Institute of Clinical Microbiology, Immunology and Hygiene at the University Hospital Erlangen, Germany. He studied medicine in Regensburg and at the TU Munich, where he was trained in Medical Microbiology. From 1999–2002 he was a postdoctoral fellow at St. Jude Children’s Research Hospital in Memphis, TN. From 2002–2008 he was an established junior investigator at the Institute of Medical Microbiology at TUM. In 2008 he was recruited to Erlangen. His main research interests are innate immune receptors and pathways of pathogen recognition, how innate immune activation is regulated by cytokines and modulators of intracellular signaling, and the implications of these interactions on inflammation and infection, and during immunization. vii International Journal of Molecular Sciences Editorial Dual Specificity Phosphatases: From Molecular Mechanisms to Biological Function Rafael Pulido 1,2,* and Roland Lang 3,* 1 Biomarkers in Cancer Unit, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain 2 IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain 3 Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany * Correspondence: [email protected] or [email protected] (R.P.); [email protected] (R.L.) Received: 3 September 2019; Accepted: 4 September 2019; Published: 6 September 2019 Dual specificity phosphatases (DUSPs) constitute a heterogeneous group of enzymes, relevant in human disease, which belong to the class I Cys-based group of protein tyrosine phosphatase (PTP) gene superfamily [1–4]. DUSPs possess the capability to dephosphorylate Ser/Thr and Tyr residues from proteins as well as to remove phosphates from other non-proteinaceous substrates, including signaling lipids [5]. Catalytically inactive pseudophosphatase DUSPs also exist which regulate phosphorylation-related cell signaling [6]. DUSPs include, among others, mitogen-activated protein kinase (MAPK) phosphatases (MKPs) and small-size atypical DUSPs. These proteins are non-transmembrane enzymes displaying variable substrate specificity and harboring a single PTP catalytic domain with a HCXXGXXR consensus catalytic motif [7,8]. MKPs are enzymes specialized in regulating the catalytic activity and subcellular location of MAPKs, whereas the functions of small-size atypical DUSPs are more diversified. DUSPs have emerged as key players in the regulation of cell growth, differentiation, stress responses and apoptosis. In physiology, DUSPs regulate essential processes, including immunity, neurobiology and metabolic homeostasis, and have been involved in tumorigenesis, pathological inflammation and metabolic disorders [9–11]. Accordingly, alterations in the expression or function of MKPs and small-size atypical DUSPs have important consequences in human disease, making these enzymes potential biological markers and therapeutic targets. Although major biochemical, structural, functional and physiological properties of many DUSPs are currently known, their developmental stage- and tissue-specific involvement in different human pathologies is only starting to be disclosed. This Special Issue provides original research and review articles focused on the involvement of specific MKPs and small-size atypical DUSPs in human
Recommended publications
  • Deciphering the Functions of Ets2, Pten and P53 in Stromal Fibroblasts in Multiple
    Deciphering the Functions of Ets2, Pten and p53 in Stromal Fibroblasts in Multiple Breast Cancer Models DISSERTATION Presented in Partial Fulfillment of the Requirements for the Degree Doctor of Philosophy in the Graduate School of The Ohio State University By Julie Wallace Graduate Program in Molecular, Cellular and Developmental Biology The Ohio State University 2013 Dissertation Committee: Michael C. Ostrowski, PhD, Advisor Gustavo Leone, PhD Denis Guttridge, PhD Dawn Chandler, PhD Copyright by Julie Wallace 2013 Abstract Breast cancer is the second most common cancer in American women, and is also the second leading cause of cancer death in women. It is estimated that nearly a quarter of a million new cases of invasive breast cancer will be diagnosed in women in the United States this year, and approximately 40,000 of these women will die from breast cancer. Although death rates have been on the decline for the past decade, there is still much we need to learn about this disease to improve prevention, detection and treatment strategies. The majority of early studies have focused on the malignant tumor cells themselves, and much has been learned concerning mutations, amplifications and other genetic and epigenetic alterations of these cells. However more recent work has acknowledged the strong influence of tumor stroma on the initiation, progression and recurrence of cancer. Under normal conditions this stroma has been shown to have protective effects against tumorigenesis, however the transformation of tumor cells manipulates this surrounding environment to actually promote malignancy. Fibroblasts in particular make up a significant portion of this stroma, and have been shown to impact various aspects of tumor cell biology.
    [Show full text]
  • Mir-338-3P Functions As a Tumor Suppressor in Gastric Cancer by Targeting PTP1B
    Sun et al. Cell Death and Disease DOI 10.1038/s41419-018-0611-0 Cell Death & Disease ARTICLE Open Access miR-338-3p functions as a tumor suppressor in gastric cancer by targeting PTP1B Feng Sun1, Mengchao Yu2,JingYu2, Zhijian Liu1,XinyanZhou2,YanqingLiu2, Xiaolong Ge3,HaidongGao2, Mei Li4, Xiaohong Jiang2,SongLiu1,XiChen2 and Wenxian Guan 1 Abstract Gastric cancer (GC) is one of the most common malignant tumors and peritoneal metastasis is the primary cause for advanced GC’s mortality. Protein-tyrosine phosphatase 1B (PTP1B) functions as an oncogene and involves in carcinogenesis and cancer dissemination. However, the function and regulation of PTP1B in GC remain poorly understood. In this study, we found that PTP1B was upregulated in GC tissues and overexpression of PTP1B in vitro promoted cell migration and prevented apoptosis. Then, we predicted that PTP1B was a target of miR-338-3p and we revealed an inverse correlation between miR-338-3p levels and PTP1B protein levels in GC tissues. Next, we verified that PTP1B was inhibited by miR-338-3p via direct targeting to its 3′-untranslated regions. Moreover, overexpression of miR-338-3p in vitro attenuated GC cell migration and promoted apoptosis, and these effects could be partially reversed by reintroduction of PTP1B. Finally, we established an orthotopic xenograft model and a peritoneal dissemination model of GC to demonstrate that miR-338-3p restrained tumor growth and dissemination in vivo by targeting PTP1B. Taken together, our results highlight that PTP1B is an oncogene and is negatively regulated by miR- 1234567890():,; 1234567890():,; 338-3p in GC, which may provide new insights into novel molecular therapeutic targets for GC.
    [Show full text]
  • Supplemental Figure 1. Vimentin
    Double mutant specific genes Transcript gene_assignment Gene Symbol RefSeq FDR Fold- FDR Fold- FDR Fold- ID (single vs. Change (double Change (double Change wt) (single vs. wt) (double vs. single) (double vs. wt) vs. wt) vs. single) 10485013 BC085239 // 1110051M20Rik // RIKEN cDNA 1110051M20 gene // 2 E1 // 228356 /// NM 1110051M20Ri BC085239 0.164013 -1.38517 0.0345128 -2.24228 0.154535 -1.61877 k 10358717 NM_197990 // 1700025G04Rik // RIKEN cDNA 1700025G04 gene // 1 G2 // 69399 /// BC 1700025G04Rik NM_197990 0.142593 -1.37878 0.0212926 -3.13385 0.093068 -2.27291 10358713 NM_197990 // 1700025G04Rik // RIKEN cDNA 1700025G04 gene // 1 G2 // 69399 1700025G04Rik NM_197990 0.0655213 -1.71563 0.0222468 -2.32498 0.166843 -1.35517 10481312 NM_027283 // 1700026L06Rik // RIKEN cDNA 1700026L06 gene // 2 A3 // 69987 /// EN 1700026L06Rik NM_027283 0.0503754 -1.46385 0.0140999 -2.19537 0.0825609 -1.49972 10351465 BC150846 // 1700084C01Rik // RIKEN cDNA 1700084C01 gene // 1 H3 // 78465 /// NM_ 1700084C01Rik BC150846 0.107391 -1.5916 0.0385418 -2.05801 0.295457 -1.29305 10569654 AK007416 // 1810010D01Rik // RIKEN cDNA 1810010D01 gene // 7 F5 // 381935 /// XR 1810010D01Rik AK007416 0.145576 1.69432 0.0476957 2.51662 0.288571 1.48533 10508883 NM_001083916 // 1810019J16Rik // RIKEN cDNA 1810019J16 gene // 4 D2.3 // 69073 / 1810019J16Rik NM_001083916 0.0533206 1.57139 0.0145433 2.56417 0.0836674 1.63179 10585282 ENSMUST00000050829 // 2010007H06Rik // RIKEN cDNA 2010007H06 gene // --- // 6984 2010007H06Rik ENSMUST00000050829 0.129914 -1.71998 0.0434862 -2.51672
    [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]
  • Analysis of BMP4 and BMP7 Signaling in Breast Cancer Cells Unveils Time
    Rodriguez-Martinez et al. BMC Medical Genomics 2011, 4:80 http://www.biomedcentral.com/1755-8794/4/80 RESEARCHARTICLE Open Access Analysis of BMP4 and BMP7 signaling in breast cancer cells unveils time-dependent transcription patterns and highlights a common synexpression group of genes Alejandra Rodriguez-Martinez1†, Emma-Leena Alarmo1†, Lilli Saarinen2, Johanna Ketolainen1, Kari Nousiainen2, Sampsa Hautaniemi2 and Anne Kallioniemi1* Abstract Background: Bone morphogenetic proteins (BMPs) are members of the TGF-beta superfamily of growth factors. They are known for their roles in regulation of osteogenesis and developmental processes and, in recent years, evidence has accumulated of their crucial functions in tumor biology. BMP4 and BMP7, in particular, have been implicated in breast cancer. However, little is known about BMP target genes in the context of tumor. We explored the effects of BMP4 and BMP7 treatment on global gene transcription in seven breast cancer cell lines during a 6- point time series, using a whole-genome oligo microarray. Data analysis included hierarchical clustering of differentially expressed genes, gene ontology enrichment analyses and model based clustering of temporal data. Results: Both ligands had a strong effect on gene expression, although the response to BMP4 treatment was more pronounced. The cellular functions most strongly affected by BMP signaling were regulation of transcription and development. The observed transcriptional response, as well as its functional outcome, followed a temporal sequence, with regulation of gene expression and signal transduction leading to changes in metabolism and cell proliferation. Hierarchical clustering revealed distinct differences in the response of individual cell lines to BMPs, but also highlighted a synexpression group of genes for both ligands.
    [Show full text]
  • PRODUCT SPECIFICATION Anti-DUSP15
    Anti-DUSP15 Product Datasheet Polyclonal Antibody PRODUCT SPECIFICATION Product Name Anti-DUSP15 Product Number HPA076649 Gene Description dual specificity phosphatase 15 Clonality Polyclonal Isotype IgG Host Rabbit Antigen Sequence Recombinant Protein Epitope Signature Tag (PrEST) antigen sequence: ICLCFGEEDPGPTQHPKEQLIMADVQVQLRPGSSSCTLSASTERPDGSST PGNPDGITHLQCSCLHPKRA Purification Method Affinity purified using the PrEST antigen as affinity ligand Verified Species Human Reactivity Recommended IHC (Immunohistochemistry) Applications - Antibody dilution: 1:200 - 1:500 - Retrieval method: HIER pH6 ICC-IF (Immunofluorescence) - Fixation/Permeabilization: PFA/Triton X-100 - Working concentration: 0.25-2 µg/ml Characterization Data Available at atlasantibodies.com/products/HPA076649 Buffer 40% glycerol and PBS (pH 7.2). 0.02% sodium azide is added as preservative. Concentration Lot dependent Storage Store at +4°C for short term storage. Long time storage is recommended at -20°C. Notes Gently mix before use. Optimal concentrations and conditions for each application should be determined by the user. For protocols, additional product information, such as images and references, see atlasantibodies.com. Product of Sweden. For research use only. Not intended for pharmaceutical development, diagnostic, therapeutic or any in vivo use. No products from Atlas Antibodies may be resold, modified for resale or used to manufacture commercial products without prior written approval from Atlas Antibodies AB. Warranty: The products supplied by Atlas Antibodies are warranted to meet stated product specifications and to conform to label descriptions when used and stored properly. Unless otherwise stated, this warranty is limited to one year from date of sales for products used, handled and stored according to Atlas Antibodies AB's instructions. Atlas Antibodies AB's sole liability is limited to replacement of the product or refund of the purchase price.
    [Show full text]
  • Exosome-Mediated MIR211 Modulates Tumor Microenvironment Via the DUSP6-ERK5 Axis and Contributes to BRAFV600E Inhibitor Resistan
    bioRxiv preprint doi: https://doi.org/10.1101/548818; this version posted February 13, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Exosome-mediated MIR211 modulates tumor microenvironment via the DUSP6-ERK5 axis and contributes to BRAFV600E inhibitor resistance in melanoma. Bongyong Lee1,3, Anupama Sahoo3, Junko Sawada1,3, John Marchica1,3, Sanjay Sahoo3, Fabiana I. A. L. Layng4, Darren Finlay4, Joseph Mazar5, Piyush Joshi1, Masanobu Komatsu1,3, Kristiina Vuori4, Garth Powis4, Petrus R. de Jong4, Animesh Ray6,7, and Ranjan J. Perera 1,2,3* 1 Department of Cancer and Blood Disorders Institute, Johns Hopkins All Children’s Hospital, St. Petersburg, FL 33701 USA 2 Department of Oncology, Sydney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287 USA 3 Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827 USA 4 Sanford Burnham Prebys Medical Discovery Institute, NCI-Designated Cancer Center, La Jolla, CA 92037, USA 5 Nemours Children Hospital, Orlando, FL 32827 USA 6 Keck Graduate Institute, Claremont CA 91711 USA, 7 Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125 USA. *Correspondence: Ranjan Perera Tel: 1-727-767-3491 Email: [email protected] 1 bioRxiv preprint doi: https://doi.org/10.1101/548818; this version posted February 13, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. ABSTRACT The microRNA MIR211 is an important regulator of melanoma tumor cell behavior.
    [Show full text]
  • 9. Atypical Dusps: 19 Phosphatases in Search of a Role
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Digital.CSIC Transworld Research Network 37/661 (2), Fort P.O. Trivandrum-695 023 Kerala, India Emerging Signaling Pathways in Tumor Biology, 2010: 185-208 ISBN: 978-81-7895-477-6 Editor: Pedro A. Lazo 9. Atypical DUSPs: 19 phosphatases in search of a role Yolanda Bayón and Andrés Alonso Instituto de Biología y Genética Molecular, CSIC-Universidad de Valladolid c/ Sanz y Forés s/n, 47003 Valladolid, Spain Abstract. Atypical Dual Specificity Phosphatases (A-DUSPs) are a group of 19 phosphatases poorly characterized. They are included among the Class I Cys-based PTPs and contain the active site motif HCXXGXXR conserved in the Class I PTPs. These enzymes present a phosphatase domain similar to MKPs, but lack any substrate targeting domain similar to the CH2 present in this group. Although most of these phosphatases have no more than 250 amino acids, their size ranges from the 150 residues of the smallest A-DUSP, VHZ/DUSP23, to the 1158 residues of the putative PTP DUSP27. The substrates of this family include MAPK, but, in general terms, it does not look that MAPK are the general substrates for the whole group. In fact, other substrates have been described for some of these phosphatases, like the 5’CAP structure of mRNA, glycogen, or STATs and still the substrates of many A-DUSPs have not been identified. In addition to the PTP domain, most of these enzymes present no additional recognizable domains in their sequence, with the exception of CBM-20 in laforin, GTase in HCE1 and a Zn binding domain in DUSP12.
    [Show full text]
  • DUSP10 Regulates Intestinal Epithelial Cell Growth and Colorectal Tumorigenesis
    Oncogene (2016) 35, 206–217 © 2016 Macmillan Publishers Limited All rights reserved 0950-9232/16 www.nature.com/onc ORIGINAL ARTICLE DUSP10 regulates intestinal epithelial cell growth and colorectal tumorigenesis CW Png1,2, M Weerasooriya1,2, J Guo3, SJ James1,2,HMPoh1,2, M Osato3, RA Flavell4, C Dong5, H Yang3 and Y Zhang1,2 Dual specificity phosphatase 10 (DUSP10), also known as MAP kinase phosphatase 5 (MKP5), negatively regulates the activation of MAP kinases. Genetic polymorphisms and aberrant expression of this gene are associated with colorectal cancer (CRC) in humans. However, the role of DUSP10 in intestinal epithelial tumorigenesis is not clear. Here, we showed that DUSP10 knockout (KO) mice had increased intestinal epithelial cell (IEC) proliferation and migration and developed less severe colitis than wild-type (WT) mice in response to dextran sodium sulphate (DSS) treatment, which is associated with increased ERK1/2 activation and Krüppel-like factor 5 (KLF5) expression in IEC. In line with increased IEC proliferation, DUSP10 KO mice developed more colon tumours with increased severity compared with WT mice in response to administration of DSS and azoxymethane (AOM). Furthermore, survival analysis of CRC patients demonstrated that high DUSP10 expression in tumours was associated with significant improvement in survival probability. Overexpression of DUSP10 in Caco-2 and RCM-1 cells inhibited cell proliferation. Our study showed that DUSP10 negatively regulates IEC growth and acts as a suppressor for CRC. Therefore, it could be targeted for the development of therapies for colitis and CRC. Oncogene (2016) 35, 206–217; doi:10.1038/onc.2015.74; published online 16 March 2015 INTRODUCTION development.
    [Show full text]
  • Epidermal Growth Factor Receptor (EGFR) Mutation Analysis, Gene
    Peraldo-Neia et al. BMC Cancer 2011, 11:31 http://www.biomedcentral.com/1471-2407/11/31 RESEARCHARTICLE Open Access Epidermal Growth Factor Receptor (EGFR) mutation analysis, gene expression profiling and EGFR protein expression in primary prostate cancer Caterina Peraldo-Neia1,2*, Giorgia Migliardi1, Maurizia Mello-Grand2, Filippo Montemurro3, Raffaella Segir2, Ymera Pignochino1, Giuliana Cavalloni1, Bruno Torchio4, Luciano Mosso4, Giovanna Chiorino2, Massimo Aglietta1,3 Abstract Background: Activating mutations of the epidermal growth factor receptor (EGFR) confer sensitivity to the tyrosine kinase inhibitors (TKi), gefitinib and erlotinib. We analysed EGFR expression, EGFR mutation status and gene expression profiles of prostate cancer (PC) to supply a rationale for EGFR targeted therapies in this disease. Methods: Mutational analysis of EGFR TK domain (exons from 18 to 21) and immunohistochemistry for EGFR were performed on tumour tissues derived from radical prostatectomy from 100 PC patients. Gene expression profiling using oligo-microarrays was also carried out in 51 of the PC samples. Results: EGFR protein overexpression (EGFRhigh) was found in 36% of the tumour samples, and mutations were found in 13% of samples. Patients with EGFRhigh tumours experienced a significantly increased risk of biochemical relapse (hazard ratio-HR 2.52, p=0.02) compared with patients with tumours expressing low levels of EGFR (EGFRlow). Microarray analysis did not reveal any differences in gene expression between EGFRhigh and EGFRlow tumours. Conversely, in EGFRhigh tumours, we were able to identify a 79 gene signature distinguishing mutated from non-mutated tumours. Additionally, 29 genes were found to be differentially expressed between mutated/ EGFRhigh (n=3) and mutated/EGFRlow tumours (n=5).
    [Show full text]
  • Ponatinib Shows Potent Antitumor Activity in Small Cell Carcinoma of the Ovary Hypercalcemic Type (SCCOHT) Through Multikinase Inhibition Jessica D
    Published OnlineFirst February 9, 2018; DOI: 10.1158/1078-0432.CCR-17-1928 Cancer Therapy: Preclinical Clinical Cancer Research Ponatinib Shows Potent Antitumor Activity in Small Cell Carcinoma of the Ovary Hypercalcemic Type (SCCOHT) through Multikinase Inhibition Jessica D. Lang1,William P.D. Hendricks1, Krystal A. Orlando2, Hongwei Yin1, Jeffrey Kiefer1, Pilar Ramos1, Ritin Sharma3, Patrick Pirrotte3, Elizabeth A. Raupach1,3, Chris Sereduk1, Nanyun Tang1, Winnie S. Liang1, Megan Washington1, Salvatore J. Facista1, Victoria L. Zismann1, Emily M. Cousins4, Michael B. Major4, Yemin Wang5, Anthony N. Karnezis5, Aleksandar Sekulic1,6, Ralf Hass7, Barbara C. Vanderhyden8, Praveen Nair9, Bernard E. Weissman2, David G. Huntsman5,10, and Jeffrey M. Trent1 Abstract Purpose: Small cell carcinoma of the ovary, hypercalcemic type three SWI/SNF wild-type ovarian cancer cell lines. We further (SCCOHT) is a rare, aggressive ovarian cancer in young women identified ponatinib as the most effective clinically approved that is universally driven by loss of the SWI/SNF ATPase subunits RTK inhibitor. Reexpression of SMARCA4 was shown to confer SMARCA4 and SMARCA2. A great need exists for effective targeted a 1.7-fold increase in resistance to ponatinib. Subsequent therapies for SCCOHT. proteomic assessment of ponatinib target modulation in Experimental Design: To identify underlying therapeutic vul- SCCOHT cell models confirmed inhibition of nine known nerabilities in SCCOHT, we conducted high-throughput siRNA ponatinib target kinases alongside 77 noncanonical ponatinib and drug screens. Complementary proteomics approaches pro- targets in SCCOHT. Finally, ponatinib delayed tumor dou- filed kinases inhibited by ponatinib. Ponatinib was tested for bling time 4-fold in SCCOHT-1 xenografts while reducing efficacy in two patient-derived xenograft (PDX) models and one final tumor volumes in SCCOHT PDX models by 58.6% and cell-line xenograft model of SCCOHT.
    [Show full text]
  • Hyperactive and Impulsive Behaviors of LMTK1 Knockout Mice
    www.nature.com/scientificreports OPEN Hyperactive and impulsive behaviors of LMTK1 knockout mice Miyuki Takahashi1,7, Arika Sugiyama1, Ran Wei1, Shizuka Kobayashi2, Kimiko Fukuda3, Hironori Nishino1, Roka Takahashi1, Koji Tsutsumi1,8, Ichiro Kita4, Kanae Ando1, Toshiya Manabe2, Hiroyuki Kamiguchi5, Mineko Tomomura6 & Shin‑ichi Hisanaga1* Lemur tail kinase 1 (LMTK1), previously called Apoptosis‑Associated Tyrosine Kinase (AATYK), remains an uncharacterized Ser/Thr protein kinase that is predominantly expressed in the brain. It is recently reported that LMTK1A, an isoform of LMTK1, binds to recycling endosomes through its palmitoylation and regulates endosomal trafcking by suppressing the activity of Rab11 small GTPase. In neurons, knockdown or knockout of LMTK1 results in longer axons, greater branching of dendrites and increased number of spines, suggesting that LMTK1 plays a role in neuronal circuit formation. However, its in vivo function remained to be investigated. Here, we examined the brain structures and behaviors of LMTK1 knockout (KO) mice. LMTK1 was expressed in most neurons throughout the brain. The overall brain structure appeared to be normal in LMTK1 KO mice, but the numbers of synapses were increased. LMTK1 KO mice had a slight impairment in memory formation and exhibited distinct psychiatric behaviors such as hyperactivity, impulsiveness and high motor coordination without social interaction defcits. Some of these abnormal behaviors represent core features of attention defcit hyperactive disorder (ADHD), suggesting the possible involvement of LMTK1 in the pathogenesis of ADHD. Abbreviations AATYK1 Apoptosis-Associated Tyrosine Kinase ADHD Attention defcit hyperactive disorder Cx Cerebral cortex Cb Cerebellum CS Conditioned stimulus ISH In situ hybridization KO Knockout LMTK Lemur tail kinase PPF Paired-pulse facilitation US Unconditioned stimulus WT Wildtype Neuronal circuit wiring is the structural and functional basis of higher brain activities such as recognition and behaviors.
    [Show full text]