Role of PTPRT in Obesity and Its Substrate Paxillin Tyrosine-88 in Colorectal Cancer

Total Page:16

File Type:pdf, Size:1020Kb

Role of PTPRT in Obesity and Its Substrate Paxillin Tyrosine-88 in Colorectal Cancer� ������� ��� ��� ��������� �������� ROLE OF PTPRT IN OBESITY AND ITS SUBSTRATE PAXILLIN TYROSINE-88 IN COLORECTAL CANCER by ANTHONY SCOTT Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy Department of Genetics and Genome Sciences CASE WESTERN RESERVE UNIVERSITY January 2015 CASE WESTERN RESERVE UNIVERSITY SCHOOL OF GRADUATE STUDIES We hereby approve the thesis/dissertation of Anthony Scott candidate for the degree of Doctor of Philosophy*. Committee Chair Hua Lou Committee Member (Advisor) Zhenghe John Wang Committee Member Sanford Markowitz Committee Member Clark Distelhorst Committee Member Alex Huang Date of Defense 7/17/2014 *We also certify that written approval has been obtained for any proprietary material contained therein. 2 Table of Contents List of Tables 8 List of Figures 9 Acknowledgements 11 List of Abbreviations 12 Abstract 15 Chapter 1: Background and Significance 17 Colorectal Cancer 18 Etiology, staging, and therapy 18 Molecular basis of colorectal cancer 20 Genomics of colorectal cancer 21 PTPRT: Structure and Function 24 PTPRT’s Domain Structure 25 PTPRT’s role in neurological development 26 PTPRT as a tumor suppressor 26 Other Type IIB RPTPs as tumor suppressors 28 3 PTPRT’s role in cell adhesion 29 PTPRT in cancer signaling pathways 30 Paxillin, a substrate of PTPRT 31 Summary of PTPRT structure and function 34 Obesity 36 Diet and obesity 36 Obesity-related pathology 37 Central nervous system and obesity 38 Summary 39 Chapter 2: Identification of paxillin Y88 as a direct target of Src kinase 41 Abstract 42 Introduction 43 Results 45 Src regulates paxillin Y88 phosphorylation 45 Src directly phosphorylates paxillin Y88 47 PY88 paxillin regulates PI3-Kinase activation 48 4 PY88 paxillin does not correlate with clinical 50 characteristics of CRC Paxillin serves as a predictive factor for dasatinib sensitivity 52 Discussion 53 Materials and Methods 56 Chapter 3: PTPRT regulates high-fat diet-induced 59 obesity and insulin resistance Abstract 60 Introduction 61 Results 63 Ptprt-/- mice are resistant to high-fat diet-induced obesity 63 Ptprt-/- mice have less body fat by percentage than 64 wild type littermates Ptprt-/- reduces food intake 66 Ptprt-/- mice have reduced energy expenditure than WT mice 68 Ptprt-/- mice resist high-fat diet-induced hyperglycemia 69 and insulin resistance 5 Metabolic differences between Ptprt+/+ and Ptprt-/- littermates 72 Phospho-STAT3 increased in the hypothalamus of Ptprt-/- mice 72 Discussion 74 Materials and Methods 76 Chapter 4: Discussion and Future Directions 81 Summary 82 PTPRT and obesity: Future Directions 82 What is the role of PTPRT in glucose and lipid metabolism? 82 How does PTPRT affect the relationship between NPY, 90 stress and obesity? What are other implications of hypothalamic 91 phospho-STAT3? What is the in vivo role of PTPRT? 94 Paxillin and cancer: Future Directions 96 Why is pY88 paxillin important therapeutically? 96 How does pY88 paxillin affect p130cas phosphorylation? 98 6 What are alternative pathways through which 100 pY88 paxillin can act? Bibliography 103 7 List of Tables Chapter 2 Table 2-1. Clinical characteristics of colorectal carcinoma samples. 50 8 List of Figures Chapter 1 Figure 1-1. A multiple-stage colorectal progression model. 21 Figure 1-2. Effect of PTPRT mutations on its function. 24 Figure 1-3. Schematic of neurohormonal/nutrient feedback loop. 39 Chapter 2 Figure 2-1. Src phosphorylates paxillin at Y88 in cell lines. 46 Figure 2-2. Src directly phosphorylates paxillin at Y88. 47 Figure 2-3. PY88 paxillin regulates p130CAS-p85 interaction. 49 Figure 2-4. PY88 paxillin is upregulated in colorectal cancer tissues 51 Figure 2-5. PY88 paxillin levels predict sensitivity to dasatinib. 53 Chapter 3 Figure 3-1. PTPRT KO mice demonstrate slightly lower body weight than wild type littermates on normal chow diet. 63 Figure 3-2. PTPRT KO mice are resistant to high-fat diet- induced body composition changes. 65 Figure 3-3. PTPRT KO mice eat less but do not absorb dietary fats differently. 67 9 Figure 3-4. PTPRT KO mice do not have different circulating levels of leptin. 67 Figure 3-5. PTPRT KO mice have decreased NPY levels before high-fat diet. 67 Figure 3-6. PTPRT KO mice utilize more glucose and expend less energy than wild type mice. 69 Figure 3-7. PTPRT KO mice have less insulin resistance than wild type mice after high-fat diet. 71 Figure 3-8. PTPRT KO mice demonstrate better insulin regulation than wild type mice. 73 Figure 3-9. PTPRT KO mice have different blood chemistry values after high-fat diet. 73 Figure 3-10. PTPRT regulates STAT3 phosphorylation in mouse hypothalamus. 74 Chapter 4 Figure 4-1. Summary of PTPRT KO phenotypes after 14 weeks on a high-fat diet. 83 Figure 4-2. Diagram of hepatic insulin signaling. 85 Figure 4-3. Effect of PTPRT KO on end organs. 95 Figure 4-4. Extended pY88 paxillin signaling model. 101 10 Acknowledgements First, I would like to acknowledge the hard work that my adviser Dr. Zhenghe John Wang put in to train me. Specifically, a key scientific skill I needed to work on when I entered the lab was the organization and presentation of my research. Regardless of whether it was a lab journal club or departmental seminar, Dr. Wang emphasized the importance of presenting research in a succinct and easily digestible manner, poring over my and my labmates' presentations to continually improve them. I attribute my three CWRU Biomedical Graduate Student Symposium poster awards, which are indicative of my progress to these ends, to his diligence in training me. I also would like to thank all of my labmates past and present, especially Dr. Yujun Hao and Dr. Xiujing Feng who were with me for all four years. They were both very gracious in providing me with help in lab and Xiujing particularly with conducting mouse studies for our obesity work. Next, I very much appreciate the time put in by my thesis committee, Dr. Hua Lou (chair), Dr. Sanford Markowitz, Dr. Clark Distelhorst and Dr. Alex Huang in preparing me to produce this body of work. Also, I would like to thank my parents and my girlfriend, Dr. Katie Linder, for their support as well. 11 List of Abbreviations ACTH Adrenocorticotropic Hormone AOM Azoxymethane ATP Adenosine Triphosphate BMI Body Mass Index CRC Colorectal Carcinoma Csk C-terminal Src Kinase DMEM Dulbecco's Modified Essential Medium DNA Deoxyribonucleic Acid EDTA Ethylenediaminetetraacetic acid EE Energy Expenditure FAP Familial Adenomatous Polyposis FBS Fetal Bovine Serum FFA/NEFA Free Fatty Acids/Non-Essential Fatty Acids FN Fibronectin G6P Glucose 6-Phosphate GAPDH Glyceraldehyde 3-phosphate dehydrogenase GTT Glucose Tolerance Test HNPCC Hereditary Non-Polyposis Colorectcal Carcinoma HOMA-IR Homestatic Model Assessment-Insulin Resistance HRP Horseradish Peroxidase Ig Immunoglobulin IPTG Isopropyl β-D-1-thiogalactopyranoside 12 ITT Insulin Tolerance Test MAM Meprin/A5-protein/PTPmu NPY Neuropeptide Y p Phospho- p130cas p130-Crk Associated Substrate PAGE Polyacrylamide Gel Electrophoresis PDK1 Phosphoinositide-Dependent Kinase PI3K Phosphoinositide 3-kinase PIP2 Phosphatidylinositol-4,5-bisphosphate PIP3 Phosphatidylinositol-3,4,5-triphosphate PKC Protein Kinase C POMC Proopiomelanocortin PTK Protein Tyrosine Kinase PTP Protein Tyrosine Phosphatase PTPN14 Protein Tyrosine Phosphatase, Non-receptor type 14 PTPRT Protein Tyrosine Phosphatase, Receptor type, T PTPRT KO Protein Tyrosine Phosphatase, Receptor type, T homozygous knockout mice PXN Paxillin pY Phosphotyrosine pY88 Phosphotyrosine-88 (paxillin) RIPA Radioimuniprecipitation assay RPTP Receptor-type Protein Tyrosine Phosphatase RQ Respiratory Quotient 13 SDS Sodium Dodecyl Sulfate SFK Src Family Kinase SH Src Homology SHP-2 Src Homology-2 domain containing phosphatase-2 shRNA Short Hairpin RNA SNP Single Nucleotide Polymorphism STAT3 Signal Transducer and Activator of Transcription 3 VLDL Very Low Density Lipoprotein Y88F Paxillin Tyrosine-88-to-Phenylalanine knock-in mutation 14 Role of PTPRT in Obesity and its Substrate Paxillin Tyrosine-88 in Colorectal Cancer Abstract By ANTHONY SCOTT Regulation of protein tyrosine phosphorylation is important in maintaining appropriate cellular homeostasis. Accordingly, protein tyrosine phosphatases are frequently mutated in cancer. The most commonly mutated protein tyrosine phosphatase in colorectal cancer is PTPRT. Follow-up studies validated it as a tumor suppressor, especially through its activity on its substrates STAT3 phosphotyrosine-705 and paxillin phosphotyrosine-88. While the latter substrate is well characterized, further study is needed into pY88 paxillin. Since previous studies show that PTPRT is inactivated in colorectal carcinoma, understanding what kinase directly phosphorylates its substrates is an important question to investigate. Here, we show that paxillin Y88 is directly targeted by Src kinase for phosphorylation. Consequently, this finding has implications for cells that express high levels of pY88 paxillin, as they become sensitive to dasatinib treatment. Moreover, although prior work demonstrated that pY88 paxillin impacts Akt signaling, how this signal was transduced was not immediately clear. We show that pY88 paxillin promotes 15 interaction between p130Cas and the p85alpha regulatory subunit of PI3K. Therefore, we shed further light into how PTPRT affects colorectal cancer tumorigenesis. Another important aspect of this and many other cancers is the role of obesity in tumor development. One of PTPRT’s substrates, pY705 STAT3, plays a crucial role in energy
Recommended publications
  • PTPRK Expression Is Downregulated in Drug Resistant Ovarian Cancer Cell Lines, and Especially in ALDH1A1 Positive Cscs-Like Popu
    Article PTPRK Expression Is Downregulated in Drug Resistant Ovarian Cancer Cell Lines, and Especially in ALDH1A1 Positive CSCs‐Like Populations Monika Świerczewska 1,*, Karolina Sterzyńska 1, Karolina Wojtowicz 1, Dominika Kaźmierczak 1, Dariusz Iżycki 2, Michał Nowicki 1, Maciej Zabel 1,3 and Radosław Januchowski 1 1 Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61‐781 Poznań, Poland; [email protected] (K.S.); [email protected] (K.W.); [email protected] (D.K.); [email protected] (M.N.); [email protected] (M.Z.); [email protected] (R.J.) 2 Department of Cancer Immunology, Poznan University of Medical Sciences, Garbary 15 St., 61‐866 Poznań, Poland; [email protected] 3 Department of Anatomy and Histology, University of Zielona Góra, Licealna 9 St., 65‐417 Zielona Góra, Poland * Correspondence: [email protected]; Tel.: +48‐61‐8546428 Received: 26 March 2019; Accepted: 24 April 2019; Published: 25 April 2019 Abstract: Background: Ovarian cancer is the 7th most common cancer and 8th most mortal cancer among woman. The standard treatment includes cytoreduction surgery followed by chemotherapy. Unfortunately, in most cases, after treatment, cancer develops drug resistance. Decreased expression and/or activity of protein phosphatases leads to increased signal transduction and development of drug resistance in cancer cells. Methods: Using sensitive (W1, A2780) and resistant ovarian cancer cell lines, the expression of Protein Tyrosine Phosphatase Receptor Type K (PTPRK) was performed at the mRNA (real‐time PCR analysis) and protein level (Western blot, immunofluorescence analysis). The protein expression in ovarian cancer tissues was determined by immunohistochemistry.
    [Show full text]
  • 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]
  • The Rac Gtpase in Cancer: from Old Concepts to New Paradigms Marcelo G
    Published OnlineFirst August 14, 2017; DOI: 10.1158/0008-5472.CAN-17-1456 Cancer Review Research The Rac GTPase in Cancer: From Old Concepts to New Paradigms Marcelo G. Kazanietz1 and Maria J. Caloca2 Abstract Rho family GTPases are critical regulators of cellular func- mislocalization of Rac signaling components. The unexpected tions that play important roles in cancer progression. Aberrant pro-oncogenic functions of Rac GTPase-activating proteins also activity of Rho small G-proteins, particularly Rac1 and their challenged the dogma that these negative Rac regulators solely regulators, is a hallmark of cancer and contributes to the act as tumor suppressors. The potential contribution of Rac tumorigenic and metastatic phenotypes of cancer cells. This hyperactivation to resistance to anticancer agents, including review examines the multiple mechanisms leading to Rac1 targeted therapies, as well as to the suppression of antitumor hyperactivation, particularly focusing on emerging paradigms immune response, highlights the critical need to develop ther- that involve gain-of-function mutations in Rac and guanine apeutic strategies to target the Rac pathway in a clinical setting. nucleotide exchange factors, defects in Rac1 degradation, and Cancer Res; 77(20); 5445–51. Ó2017 AACR. Introduction directed toward targeting Rho-regulated pathways for battling cancer. Exactly 25 years ago, two seminal papers by Alan Hall and Nearly all Rho GTPases act as molecular switches that cycle colleagues illuminated us with one of the most influential dis- between GDP-bound (inactive) and GTP-bound (active) forms. coveries in cancer signaling: the association of Ras-related small Activation is promoted by guanine nucleotide exchange factors GTPases of the Rho family with actin cytoskeleton reorganization (GEF) responsible for GDP dissociation, a process that normally (1, 2).
    [Show full text]
  • Identification of Chebulinic Acid As a Dual Targeting Inhibitor of Protein
    Bioorganic Chemistry 90 (2019) 103087 Contents lists available at ScienceDirect Bioorganic Chemistry journal homepage: www.elsevier.com/locate/bioorg Short communication Identification of chebulinic acid as a dual targeting inhibitor of protein T tyrosine phosphatases relevant to insulin resistance Sun-Young Yoona,1, Hyo Jin Kangb,1, Dohee Ahna, Ji Young Hwanga, Se Jeong Kwona, ⁎ Sang J. Chunga, a School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea b Department of Chemistry, Dongguk University, Seoul 100-715, Republic of Korea ARTICLE INFO ABSTRACT Keywords: Natural products as antidiabetic agents have been shown to stimulate insulin signaling via the inhibition of the Protein tyrosine phosphatases (PTPs) protein tyrosine phosphatases relevant to insulin resistance. Previously, we have identified PTPN9 and DUSP9 as Chebulinic acid potential antidiabetic targets and a multi-targeting natural product thereof. In this study, knockdown of PTPN11 Type 2 diabetes increased AMPK phosphorylation in differentiated C2C12 muscle cells by 3.8 fold, indicating that PTPN11 could Glucose-uptake be an antidiabetic target. Screening of a library of 658 natural products against PTPN9, DUSP9, or PTPN11 PTPN9 identified chebulinic acid (CA) as a strong allosteric inhibitor with a slow cooperative binding toPTPN9 PTPN11 (IC50 = 34 nM) and PTPN11 (IC50 = 37 nM), suggesting that it would be a potential antidiabetic candidate. Furthermore, CA stimulated glucose uptake and resulted in increased AMP-activated protein kinase (AMPK) phosphorylation. Taken together, we demonstrated that CA increased glucose uptake as a dual inhibitor of PTPN9 and PTPN11 through activation of the AMPK signaling pathway. These results strongly suggest that CA could be used as a potential therapeutic candidate for the treatment of type 2 diabetes.
    [Show full text]
  • A GTP-State Specific Cyclic Peptide Inhibitor of the Gtpase Gαs
    bioRxiv preprint doi: https://doi.org/10.1101/2020.04.25.054080; this version posted April 27, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. A GTP-state specific cyclic peptide inhibitor of the GTPase Gαs Shizhong A. Dai1,2†, Qi Hu1,2†, Rong Gao3†, Andre Lazar1,4†, Ziyang Zhang1,2, Mark von Zastrow1,4, Hiroaki Suga3*, Kevan M. Shokat1,2* 5 1Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, 94158, USA 2Howard Hughes Medical Institute 3Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan 10 4Department of Psychiatry, University of California, San Francisco, San Francisco, CA, 94158, USA *Correspondence to: [email protected], [email protected] †These authors contributed equally. 15 20 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.04.25.054080; this version posted April 27, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Abstract: The G protein-coupled receptor (GPCR) cascade leading to production of the second messenger cAMP is replete with pharmacologically targetable receptors and enzymes with the exception of the stimulatory G protein α subunit, Gαs.
    [Show full text]
  • Clinical Utility of Recently Identified Diagnostic, Prognostic, And
    Modern Pathology (2017) 30, 1338–1366 1338 © 2017 USCAP, Inc All rights reserved 0893-3952/17 $32.00 Clinical utility of recently identified diagnostic, prognostic, and predictive molecular biomarkers in mature B-cell neoplasms Arantza Onaindia1, L Jeffrey Medeiros2 and Keyur P Patel2 1Instituto de Investigacion Marques de Valdecilla (IDIVAL)/Hospital Universitario Marques de Valdecilla, Santander, Spain and 2Department of Hematopathology, MD Anderson Cancer Center, Houston, TX, USA Genomic profiling studies have provided new insights into the pathogenesis of mature B-cell neoplasms and have identified markers with prognostic impact. Recurrent mutations in tumor-suppressor genes (TP53, BIRC3, ATM), and common signaling pathways, such as the B-cell receptor (CD79A, CD79B, CARD11, TCF3, ID3), Toll- like receptor (MYD88), NOTCH (NOTCH1/2), nuclear factor-κB, and mitogen activated kinase signaling, have been identified in B-cell neoplasms. Chronic lymphocytic leukemia/small lymphocytic lymphoma, diffuse large B-cell lymphoma, follicular lymphoma, mantle cell lymphoma, Burkitt lymphoma, Waldenström macroglobulinemia, hairy cell leukemia, and marginal zone lymphomas of splenic, nodal, and extranodal types represent examples of B-cell neoplasms in which novel molecular biomarkers have been discovered in recent years. In addition, ongoing retrospective correlative and prospective outcome studies have resulted in an enhanced understanding of the clinical utility of novel biomarkers. This progress is reflected in the 2016 update of the World Health Organization classification of lymphoid neoplasms, which lists as many as 41 mature B-cell neoplasms (including provisional categories). Consequently, molecular genetic studies are increasingly being applied for the clinical workup of many of these neoplasms. In this review, we focus on the diagnostic, prognostic, and/or therapeutic utility of molecular biomarkers in mature B-cell neoplasms.
    [Show full text]
  • Single-Cell RNA Sequencing Demonstrates the Molecular and Cellular Reprogramming of Metastatic Lung Adenocarcinoma
    ARTICLE https://doi.org/10.1038/s41467-020-16164-1 OPEN Single-cell RNA sequencing demonstrates the molecular and cellular reprogramming of metastatic lung adenocarcinoma Nayoung Kim 1,2,3,13, Hong Kwan Kim4,13, Kyungjong Lee 5,13, Yourae Hong 1,6, Jong Ho Cho4, Jung Won Choi7, Jung-Il Lee7, Yeon-Lim Suh8,BoMiKu9, Hye Hyeon Eum 1,2,3, Soyean Choi 1, Yoon-La Choi6,10,11, Je-Gun Joung1, Woong-Yang Park 1,2,6, Hyun Ae Jung12, Jong-Mu Sun12, Se-Hoon Lee12, ✉ ✉ Jin Seok Ahn12, Keunchil Park12, Myung-Ju Ahn 12 & Hae-Ock Lee 1,2,3,6 1234567890():,; Advanced metastatic cancer poses utmost clinical challenges and may present molecular and cellular features distinct from an early-stage cancer. Herein, we present single-cell tran- scriptome profiling of metastatic lung adenocarcinoma, the most prevalent histological lung cancer type diagnosed at stage IV in over 40% of all cases. From 208,506 cells populating the normal tissues or early to metastatic stage cancer in 44 patients, we identify a cancer cell subtype deviating from the normal differentiation trajectory and dominating the metastatic stage. In all stages, the stromal and immune cell dynamics reveal ontological and functional changes that create a pro-tumoral and immunosuppressive microenvironment. Normal resident myeloid cell populations are gradually replaced with monocyte-derived macrophages and dendritic cells, along with T-cell exhaustion. This extensive single-cell analysis enhances our understanding of molecular and cellular dynamics in metastatic lung cancer and reveals potential diagnostic and therapeutic targets in cancer-microenvironment interactions. 1 Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Korea.
    [Show full text]
  • Supp Table 1.Pdf
    Upregulated genes in Hdac8 null cranial neural crest cells fold change Gene Symbol Gene Title 134.39 Stmn4 stathmin-like 4 46.05 Lhx1 LIM homeobox protein 1 31.45 Lect2 leukocyte cell-derived chemotaxin 2 31.09 Zfp108 zinc finger protein 108 27.74 0710007G10Rik RIKEN cDNA 0710007G10 gene 26.31 1700019O17Rik RIKEN cDNA 1700019O17 gene 25.72 Cyb561 Cytochrome b-561 25.35 Tsc22d1 TSC22 domain family, member 1 25.27 4921513I08Rik RIKEN cDNA 4921513I08 gene 24.58 Ofa oncofetal antigen 24.47 B230112I24Rik RIKEN cDNA B230112I24 gene 23.86 Uty ubiquitously transcribed tetratricopeptide repeat gene, Y chromosome 22.84 D8Ertd268e DNA segment, Chr 8, ERATO Doi 268, expressed 19.78 Dag1 Dystroglycan 1 19.74 Pkn1 protein kinase N1 18.64 Cts8 cathepsin 8 18.23 1500012D20Rik RIKEN cDNA 1500012D20 gene 18.09 Slc43a2 solute carrier family 43, member 2 17.17 Pcm1 Pericentriolar material 1 17.17 Prg2 proteoglycan 2, bone marrow 17.11 LOC671579 hypothetical protein LOC671579 17.11 Slco1a5 solute carrier organic anion transporter family, member 1a5 17.02 Fbxl7 F-box and leucine-rich repeat protein 7 17.02 Kcns2 K+ voltage-gated channel, subfamily S, 2 16.93 AW493845 Expressed sequence AW493845 16.12 1600014K23Rik RIKEN cDNA 1600014K23 gene 15.71 Cst8 cystatin 8 (cystatin-related epididymal spermatogenic) 15.68 4922502D21Rik RIKEN cDNA 4922502D21 gene 15.32 2810011L19Rik RIKEN cDNA 2810011L19 gene 15.08 Btbd9 BTB (POZ) domain containing 9 14.77 Hoxa11os homeo box A11, opposite strand transcript 14.74 Obp1a odorant binding protein Ia 14.72 ORF28 open reading
    [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]
  • CDH12 Cadherin 12, Type 2 N-Cadherin 2 RPL5 Ribosomal
    5 6 6 5 . 4 2 1 1 1 2 4 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 2 2 A A A A A A A A A A A A A A A A A A A A C C C C C C C C C C C C C C C C C C C C R R R R R R R R R R R R R R R R R R R R B , B B B B B B B B B B B B B B B B B B B , 9 , , , , 4 , , 3 0 , , , , , , , , 6 2 , , 5 , 0 8 6 4 , 7 5 7 0 2 8 9 1 3 3 3 1 1 7 5 0 4 1 4 0 7 1 0 2 0 6 7 8 0 2 5 7 8 0 3 8 5 4 9 0 1 0 8 8 3 5 6 7 4 7 9 5 2 1 1 8 2 2 1 7 9 6 2 1 7 1 1 0 4 5 3 5 8 9 1 0 0 4 2 5 0 8 1 4 1 6 9 0 0 6 3 6 9 1 0 9 0 3 8 1 3 5 6 3 6 0 4 2 6 1 0 1 2 1 9 9 7 9 5 7 1 5 8 9 8 8 2 1 9 9 1 1 1 9 6 9 8 9 7 8 4 5 8 8 6 4 8 1 1 2 8 6 2 7 9 8 3 5 4 3 2 1 7 9 5 3 1 3 2 1 2 9 5 1 1 1 1 1 1 5 9 5 3 2 6 3 4 1 3 1 1 4 1 4 1 7 1 3 4 3 2 7 6 4 2 7 2 1 2 1 5 1 6 3 5 6 1 3 6 4 7 1 6 5 1 1 4 1 6 1 7 6 4 7 e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e e l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l l p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m m
    [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]