Protein Tyrosine Phosphatase PTPN3 Inhibits Lung Cancer Cell Proliferation and Migration by Promoting EGFR Endocytic Degradation

Total Page:16

File Type:pdf, Size:1020Kb

Protein Tyrosine Phosphatase PTPN3 Inhibits Lung Cancer Cell Proliferation and Migration by Promoting EGFR Endocytic Degradation Oncogene (2015) 34, 3791–3803 © 2015 Macmillan Publishers Limited All rights reserved 0950-9232/15 www.nature.com/onc ORIGINAL ARTICLE Protein tyrosine phosphatase PTPN3 inhibits lung cancer cell proliferation and migration by promoting EGFR endocytic degradation M-Y Li1,2, P-L Lai1, Y-T Chou3, A-P Chi1, Y-Z Mi1, K-H Khoo1,2, G-D Chang2, C-W Wu3, T-C Meng1,2 and G-C Chen1,2 Epidermal growth factor receptor (EGFR) regulates multiple signaling cascades essential for cell proliferation, growth and differentiation. Using a genetic approach, we found that Drosophila FERM and PDZ domain-containing protein tyrosine phosphatase, dPtpmeg, negatively regulates border cell migration and inhibits the EGFR/Ras/mitogen-activated protein kinase signaling pathway during wing morphogenesis. We further identified EGFR pathway substrate 15 (Eps15) as a target of dPtpmeg and its human homolog PTPN3. Eps15 is a scaffolding adaptor protein known to be involved in EGFR endocytosis and trafficking. Interestingly, PTPN3-mediated tyrosine dephosphorylation of Eps15 promotes EGFR for lipid raft-mediated endocytosis and lysosomal degradation. PTPN3 and the Eps15 tyrosine phosphorylation-deficient mutant suppress non-small-cell lung cancer cell growth and migration in vitro and reduce lung tumor xenograft growth in vivo. Moreover, depletion of PTPN3 impairs the degradation of EGFR and enhances proliferation and tumorigenicity of lung cancer cells. Taken together, these results indicate that PTPN3 may act as a tumor suppressor in lung cancer through its modulation of EGFR signaling. Oncogene (2015) 34, 3791–3803; doi:10.1038/onc.2014.312; published online 29 September 2014 INTRODUCTION sorting EGFR to multivesicular bodies.15 Recently, Ali et al.16 Reversible tyrosine protein phosphorylation by protein tyrosine showed that the ESCRT accessory protein HD-PTP/PTPN23 kinases and protein tyrosine phosphatases (PTPs) acts as a coordinates with the ubiquitin-specific peptidase UBPY to drive molecular switch that regulates a variety of biological pro- EGFR sorting to the multivesicular bodies. A better understanding cesses.1,2 The receptor tyrosine kinase epidermal growth factor of the role of PTPs in regulating EGFR signaling will help to receptor (EGFR), the best characterized member of the ErbB family provide insights into the molecular mechanisms behind EGFR- receptors, acts as a critical regulator of numerous cellular mediated tumorigenesis. processes, including growth, proliferation and differentiation. PTPN3 (PTPH1) and the closely-related PTPN4 (PTPMEG) are Upon activation by its growth factor ligands, EGFR undergoes non-transmembrane PTPs that contain an N-terminal FERM dimerization and activation, leading to tyrosine phosphorylation (Band 4.1, Ezrin, Radixin, Moesin homology) domain followed by of the intracellular region of the receptor as well as many a single PDZ (PSD95, Dlg, ZO1) domain and the C-terminal PTP 3,4 cytoplasmic substrates. The activated EGFR is then internalized domain.1 They have been implicated in the regulation of cell by clathrin-mediated endocytosis and sorted into the endosomal growth and proliferation.17,18 However, their role in receptor compartments, through which it is either recycled back to the protein tyrosine kinase signaling is not clear. The dPtpmeg is the plasma membrane or transported to the lysosome for degrada- 5 Drosophila homolog of mammalian PTPN3 and PTPN4. Phenotypic tion. Because overexpression or constitutive activation of EGFR analyses have revealed that dptpmeg mutants exhibit aberrant has been implicated in the pathogenesis and progression of a mushroom body axon projection patterns in the brain.19 Besides variety of human malignancies,6 it is therefore crucial to under- its role in regulating neuronal wiring, the molecular function of stand how EGFR signaling is regulated. dPtpmeg has remained largely unknown. In this study, we Several PTPs have been implicated in the regulation of EGFR fi signaling. Among them, PTPRK, DEP-1 (PTPRJ), PTP1B (PTPN1), identi ed EGFR pathway substrate 15 (Eps15) as a substrate of SHP-1 (PTPN6), TCPTP (PTPN2), PTPN9 and PTPN12 have been dPtpmeg and PTPN3. Eps15 is known to be an endocytic adaptor fi 20 shown to downregulate EGFR signaling by dephosphorylating involved in the regulation of EGFR traf cking. PTPN3 depho- EGFR.7–11 The receptor-type PTP DEP-1 dephosphorylates EGFR on sphorylated Eps15 and promoted EGFR for lipid raft-mediated the cell surface and inhibits its internalization.7 On the other hand, endocytosis and lysosomal degradation. The ectopic expression of the endoplasmic reticulum-localized PTP1B has been reported to PTPN3 or Eps15-Y850F mutant in the non-small-cell lung cancer regulate EGFR signaling from endosomes.12,13 PTP1B promotes (NSCLC) cells inhibited cell proliferation, migration and tumor the sequestration of EGFR onto internal vesicles of multivesicular growth. Our findings uncover a novel role for PTPN3 in the bodies.14 The ESCRT (endosomal sorting complex required for regulation of EGFR endocytic trafficking, degradation and transport) complexes are known to play an important role in signaling. 1Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan; 2Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei, Taiwan and 3Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan. Correspondence: Dr G-C Chen, Institute of Biological Chemistry, Academia Sinica, 128 Academia Road, Section 2, Taipei 115, Taiwan. E-mail: [email protected] Received 3 April 2014; revised 26 July 2014; accepted 16 August 2014; published online 29 September 2014 PTPN3 negatively regulates EGFR signaling M-Y Li et al 3792 RESULTS the activated form of Drosophila Ras (RasV12) (Figures 1k–m), but Drosophila Ptpmeg is involved in regulating the EGFR signaling not by coexpressing the constitutively active phosphoinositide 3- pathway kinase (Dp110-CAAX) or active Akt (myr-Akt) (Figures 1n–o). EGFR We previously performed genetic analyses to identify non- is known to induce extracellular signal-regulated kinase/MAPK transmembrane PTPs that could modulate border cell migration phosphorylation in future vein regions during wing develop- 21 ment.27 Strikingly, we found that clonal expression of dPtpmeg in during Drosophila oogenesis. Although RNA interference (RNAi)- 28 mediated downregulation of Drosophila non-transmembrane PTPs wing imaginal discs using the flipout/Gal4 system led to a did not have an obvious effect on border cell migration at stage marked reduction of MAPK phosphorylation in GFP-positive 10 egg chambers,21 we found that dptpmeg mutation caused dPtpmeg-expressing cells (Figures 1p, p’). Taken together, these accelerated migration of border cells and the clusters reached data indicate that dPtpmeg acts as a negative regulator of the oocyte prematurely at stage 9 (Figures 1a and b). The receptor EGFR/Ras/MAPK signal pathway. tyrosine kinases, platelet-derived growth factor (PDGF)- and vascular endothelial growth factor (VEGF)-related receptor (PVR) Identification of Eps15 as a substrate of dPtpmeg and EGFR, have been shown to play an important role in guiding 22,23 We have recently established a mass spectrometry (MS)-based migration of the border cells toward the oocyte. As shown in substrate trapping strategy to identify putative substrates of Figures 1c and d, ectopic expression of dPtpmeg with the border 26 fi PTP61F, the smallest non-transmembrane PTP in Drosophila. We cell-speci c Slbo-Gal4 driver impaired border cell motility. More- used the same approach to identify potential substrates of over, clonal analysis revealed elevated phosphotyrosine levels in dPtpmeg. The bacterially expressed wild-type (WT) form and the dptpmeg homozygous mutant border cell clones (marked by the substrate-trapping DA mutant form of dPtpmeg were incubated loss of green fluorescent protein (GFP); Figure 1e, e’), suggesting with Drosophila S2 cell lysates. After immunoprecipitation of that dPtpmeg may antagonize receptor tyrosine kinase-mediated dPtpmeg, the associated proteins were eluted, resolved by sodium tyrosine phosphorylation and border cell migration. dodecyl sulfate–polyacrylamide gel electrophoresis (SDS–PAGE) It has been reported that EGFR signaling regulates a variety of developmental processes in Drosophila, including wing vein and subjected to liquid chromatography tandem mass spectro- formation.24,25 To investigate the role of dPtpmeg in vein metry analysis. Several proteins, including Eps15, Pvr, Cortactin and Ter94 (mammalian VCP/p97), were identified to interact patterning, we used engrailed-Gal4 (en-Gal4) driver to restrict fi dPtpmeg expression in the posterior compartment of the wing; speci cally with the dPtpmeg-DA mutant (Figure 2a). Interestingly, fi therefore, the anterior part served as control. Interestingly, previous work has identi ed VCP/p97 as a substrate of PTPN3 in 17 fi expression of wild-type dPtpmeg, but not phosphatase-deficient mammalian cells. Moreover, the identi cation of Pvr as a fi mutant (dPtpmeg-CS), caused a wing vein missing phenotype putative substrate of dPtpmeg is consistent with the nding that (Figures 1f–h). The dPtpmeg-induced wing vein defects could be dPtpmeg plays a role in regulating border cell migration rescued by coexpressing dPtpmeg-RNAi (Figure 1i). Moreover, (Figures 1a–d). Here we have focused our study on Eps15. Eps15 ectopic expression of PTP61F,26 the Drosophila homolog of PTP1B, is a multidomain adaptor protein that contains EH domains at the in the developing
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]
  • PTPN9 Promotes Cell Proliferation and Invasion in Eca109 Cells and Is Negatively Regulated by Microrna-126
    ONCOLOGY LETTERS 14: 1419-1426, 2017 PTPN9 promotes cell proliferation and invasion in Eca109 cells and is negatively regulated by microRNA-126 JUNWEI ZHU1, HAOMIAO LI1, JUN MA2, HAIBO HUANG1, JIANJUN QIN1 and YIN LI1 1Department of Thoracic Surgery, The Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, Henan 450008; 2Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, P.R. China Received September 11, 2015; Accepted April 13, 2017 DOI: 10.3892/ol.2017.6315 Abstract. Protein tyrosine phosphatase non-receptor type 9 Using RNA interference, the present study demonstrated that (PTPN9), also named PTP-MEG2, is an important member knockdown of PTPN9 significantly suppressed cell prolifera- of the protein tyrosine phosphatase family that is involved tion and invasion in Eca109. Additionally, it was hypothesized in variety of human diseases. However, the role of PTPN9 that miR-126, described as a tumor suppressor in ESCC, in esophageal squamous cell carcinoma (ESCC) remains to may act at least in part via its inhibition of PTPN9 at the be established. The present evaluated the potential effect and post-transcriptional level. To the best of our knowledge, this is underlying mechanism of action of PTPN9 in ESCC. Immu- the first study to demonstrate that PTPN9 is overexpressed in nohistochemistry was performed to detect PTPN9 protein ESCC and associated with poor survival, and may therefore be expression in 84 ESCC tumor specimens and 30 normal important in the pathogenesis of ESCC. esophageal tissues. The association between positive expres- sion of PTPN9 and clinicopathological features and prognosis Introduction was analyzed. The prognostic role of PTPN9 was further inves- tigated using multivariate regression analysis.
    [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]
  • 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]
  • 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]
  • Growth and Molecular Profile of Lung Cancer Cells Expressing Ectopic LKB1: Down-Regulation of the Phosphatidylinositol 3؅-Phosphate Kinase/PTEN Pathway1
    [CANCER RESEARCH 63, 1382–1388, March 15, 2003] Growth and Molecular Profile of Lung Cancer Cells Expressing Ectopic LKB1: Down-Regulation of the Phosphatidylinositol 3؅-Phosphate Kinase/PTEN Pathway1 Ana I. Jimenez, Paloma Fernandez, Orlando Dominguez, Ana Dopazo, and Montserrat Sanchez-Cespedes2 Molecular Pathology Program [A. I. J., P. F., M. S-C.], Genomics Unit [O. D.], and Microarray Analysis Unit [A. D.], Spanish National Cancer Center, 28029 Madrid, Spain ABSTRACT the cell cycle in G1 (8, 9). However, the intrinsic mechanism by which LKB1 activity is regulated in cells and how it leads to the suppression Germ-line mutations in LKB1 gene cause the Peutz-Jeghers syndrome of cell growth is still unknown. It has been proposed that growth (PJS), a genetic disease with increased risk of malignancies. Recently, suppression by LKB1 is mediated through p21 in a p53-dependent LKB1-inactivating mutations have been identified in one-third of sporadic lung adenocarcinomas, indicating that LKB1 gene inactivation is critical in mechanism (7). In addition, it has been observed that LKB1 binds to tumors other than those of the PJS syndrome. However, the in vivo brahma-related gene 1 protein (BRG1) and this interaction is required substrates of LKB1 and its role in cancer development have not been for BRG1-induced growth arrest (10). Similar to what happens in the completely elucidated. Here we show that overexpression of wild-type PJS, Lkb1 heterozygous knockout mice show gastrointestinal hamar- LKB1 protein in A549 lung adenocarcinomas cells leads to cell-growth tomatous polyposis and frequent hepatocellular carcinomas (11, 12). suppression. To examine changes in gene expression profiles subsequent to Interestingly, the hamartomas, but not the malignant tumors, arising in exogenous wild-type LKB1 in A549 cells, we used cDNA microarrays.
    [Show full text]
  • The Proteintyrosine Phosphatase Receptor Type J Is Regulated by The
    Zurich Open Repository and Archive University of Zurich Main Library Strickhofstrasse 39 CH-8057 Zurich www.zora.uzh.ch Year: 2013 The Protein-Tyrosine Phosphatase Receptor Type J is regulated by the pVHL-HIF axis in Clear Cell Renal Cell Carcinoma Casagrande, Silvia ; Ruf, Melanie ; Rechsteiner, Markus ; Morra, Laura ; Brun-Schmid, Sonja ; von Teichman, Adriana ; Krek, Wilhelm ; Schraml, Peter ; Moch, Holger Abstract: Mass spectrometry analysis of renal cancer cell lines recently suggested that the Protein- Tyrosine Phosphatase Receptor Type J (PTPRJ), an important regulator of tyrosine kinase receptors, is tightly linked to the von-Hippel Lindau protein (pVHL). Therefore, we aimed to characterize the biological relevance of PTPRJ for clear cell renal cell carcinoma (ccRCC). In pVHL negative ccRCC cell lines both RNA and protein expression levels of PTPRJ were lower than those in the corresponding pVHL reconstituted cells. Quantitative RT-PCR and Western blot analysis of ccRCC with known VHL mutation status and normal matched tissues as well as RNA in situ hybridization on a Tissue Microarray (TMA) confirmed a decrease of PTPRJ expression in more than 80 % of ccRCCs, but inonly12%of papillary RCCs. ccRCC patients with no or reduced PTPRJ mRNA expression had a less favourable outcome than those with a normal expression status (p = 0.05). Sequence analysis of 32 PTPRJ mRNA negative ccRCC samples showed five known polymorphisms, but no mutations implying other mechanisms leading to PTPRJ’s down-regulation. Selective silencing of HIF-฀ by siRNA and reporter gene assays demonstrated that pVHL inactivation reduces PTPRJ expression through a HIF-dependent mechanism, which is mainly driven by HIF-2฀ stabilization.
    [Show full text]
  • Mir-96-5P Targets PTEN Expression Affecting Radio-Chemosensitivity of HNSCC Cells
    Vahabi et al. Journal of Experimental & Clinical Cancer Research (2019) 38:141 https://doi.org/10.1186/s13046-019-1119-x RESEARCH Open Access miR-96-5p targets PTEN expression affecting radio-chemosensitivity of HNSCC cells Mahrou Vahabi1,2, Claudio Pulito1, Andrea Sacconi1, Sara Donzelli1, Marco D’Andrea3, Valentina Manciocco4, Raul Pellini4, Paola Paci5,6, Giuseppe Sanguineti3, Lidia Strigari7, Giuseppe Spriano8, Paola Muti9, Pier Paolo Pandolfi10, Sabrina Strano1, Shahrokh Safarian2*, Federica Ganci1* and Giovanni Blandino1* Abstract Background: Head and neck squamous cell carcinoma (HNSCC) is the sixth leading cancer worldwide. They are typically characterized by a high incidence of local recurrence, which is the most common cause of death in HNSCC patients. TP53 is the most frequently mutated gene in HNSCC and patients carrying TP53 mutations are associated with a higher probability to develop local recurrence. MiRNAs, which are among the mediators of the oncogenic activity of mt-p53 protein, emerge as an appealing tool for screening, diagnosis and prognosis of cancer. We previously identified a signature of 12 miRNAs whose aberrant expression associated with TP53 mutations and was prognostic for HNSCC. Among them miR-96-5p emerges as an oncogenic miRNAs with prognostic significance in HNSCC. Methods: To evaluate the oncogenic role of miR-96-5p in a tumoral context, we performed colony formation, cell migration and cell viability assays in two HNSCC cell lines transfected for miR-96-5p mimic or inhibitor and treated with or without radio/chemo-therapy. In addition, to identify genes positively and negatively correlated to miR-96- 5p expression in HNSCC, we analyzed the correlation between gene expression and miR-96-5p level in the subset of TCGA HNSCC tumors carrying missense TP53 mutations by Spearman and Pearson correlation.
    [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]
  • (12) Patent Application Publication (10) Pub. No.: US 2003/0082511 A1 Brown Et Al
    US 20030082511A1 (19) United States (12) Patent Application Publication (10) Pub. No.: US 2003/0082511 A1 Brown et al. (43) Pub. Date: May 1, 2003 (54) IDENTIFICATION OF MODULATORY Publication Classification MOLECULES USING INDUCIBLE PROMOTERS (51) Int. Cl." ............................... C12O 1/00; C12O 1/68 (52) U.S. Cl. ..................................................... 435/4; 435/6 (76) Inventors: Steven J. Brown, San Diego, CA (US); Damien J. Dunnington, San Diego, CA (US); Imran Clark, San Diego, CA (57) ABSTRACT (US) Correspondence Address: Methods for identifying an ion channel modulator, a target David B. Waller & Associates membrane receptor modulator molecule, and other modula 5677 Oberlin Drive tory molecules are disclosed, as well as cells and vectors for Suit 214 use in those methods. A polynucleotide encoding target is San Diego, CA 92121 (US) provided in a cell under control of an inducible promoter, and candidate modulatory molecules are contacted with the (21) Appl. No.: 09/965,201 cell after induction of the promoter to ascertain whether a change in a measurable physiological parameter occurs as a (22) Filed: Sep. 25, 2001 result of the candidate modulatory molecule. Patent Application Publication May 1, 2003 Sheet 1 of 8 US 2003/0082511 A1 KCNC1 cDNA F.G. 1 Patent Application Publication May 1, 2003 Sheet 2 of 8 US 2003/0082511 A1 49 - -9 G C EH H EH N t R M h so as se W M M MP N FIG.2 Patent Application Publication May 1, 2003 Sheet 3 of 8 US 2003/0082511 A1 FG. 3 Patent Application Publication May 1, 2003 Sheet 4 of 8 US 2003/0082511 A1 KCNC1 ITREXCHO KC 150 mM KC 2000000 so 100 mM induced Uninduced Steady state O 100 200 300 400 500 600 700 Time (seconds) FIG.
    [Show full text]
  • Genetic Alterations of Protein Tyrosine Phosphatases in Human Cancers
    Oncogene (2015) 34, 3885–3894 © 2015 Macmillan Publishers Limited All rights reserved 0950-9232/15 www.nature.com/onc REVIEW Genetic alterations of protein tyrosine phosphatases in human cancers S Zhao1,2,3, D Sedwick3,4 and Z Wang2,3 Protein tyrosine phosphatases (PTPs) are enzymes that remove phosphate from tyrosine residues in proteins. Recent whole-exome sequencing of human cancer genomes reveals that many PTPs are frequently mutated in a variety of cancers. Among these mutated PTPs, PTP receptor T (PTPRT) appears to be the most frequently mutated PTP in human cancers. Beside PTPN11, which functions as an oncogene in leukemia, genetic and functional studies indicate that most of mutant PTPs are tumor suppressor genes. Identification of the substrates and corresponding kinases of the mutant PTPs may provide novel therapeutic targets for cancers harboring these mutant PTPs. Oncogene (2015) 34, 3885–3894; doi:10.1038/onc.2014.326; published online 29 September 2014 INTRODUCTION tyrosine/threonine-specific phosphatases. (4) Class IV PTPs include Protein tyrosine phosphorylation has a critical role in virtually all four Drosophila Eya homologs (Eya1, Eya2, Eya3 and Eya4), which human cellular processes that are involved in oncogenesis.1 can dephosphorylate both tyrosine and serine residues. Protein tyrosine phosphorylation is coordinately regulated by protein tyrosine kinases (PTKs) and protein tyrosine phosphatases 1 THE THREE-DIMENSIONAL STRUCTURE AND CATALYTIC (PTPs). Although PTKs add phosphate to tyrosine residues in MECHANISM OF PTPS proteins, PTPs remove it. Many PTKs are well-documented oncogenes.1 Recent cancer genomic studies provided compelling The three-dimensional structures of the catalytic domains of evidence that many PTPs function as tumor suppressor genes, classical PTPs (RPTPs and non-RPTPs) are extremely well because a majority of PTP mutations that have been identified in conserved.5 Even the catalytic domain structures of the dual- human cancers are loss-of-function mutations.
    [Show full text]