Expression Profile of Tyrosine Phosphatases in HER2 Breast

Total Page:16

File Type:pdf, Size:1020Kb

Expression Profile of Tyrosine Phosphatases in HER2 Breast Cellular Oncology 32 (2010) 361–372 361 DOI 10.3233/CLO-2010-0520 IOS Press Expression profile of tyrosine phosphatases in HER2 breast cancer cells and tumors Maria Antonietta Lucci a, Rosaria Orlandi b, Tiziana Triulzi b, Elda Tagliabue b, Andrea Balsari c and Emma Villa-Moruzzi a,∗ a Department of Experimental Pathology, University of Pisa, Pisa, Italy b Molecular Biology Unit, Department of Experimental Oncology, Istituto Nazionale Tumori, Milan, Italy c Department of Human Morphology and Biomedical Sciences, University of Milan, Milan, Italy Abstract. Background: HER2-overexpression promotes malignancy by modulating signalling molecules, which include PTPs/DSPs (protein tyrosine and dual-specificity phosphatases). Our aim was to identify PTPs/DSPs displaying HER2-associated expression alterations. Methods: HER2 activity was modulated in MDA-MB-453 cells and PTPs/DSPs expression was analysed with a DNA oligoar- ray, by RT-PCR and immunoblotting. Two public breast tumor datasets were analysed to identify PTPs/DSPs differentially ex- pressed in HER2-positive tumors. Results: In cells (1) HER2-inhibition up-regulated 4 PTPs (PTPRA, PTPRK, PTPN11, PTPN18) and 11 DSPs (7 MKPs [MAP Kinase Phosphatases], 2 PTP4, 2 MTMRs [Myotubularin related phosphatases]) and down-regulated 7 DSPs (2 MKPs, 2 MTMRs, CDKN3, PTEN, CDC25C); (2) HER2-activation with EGF affected 10 DSPs (5 MKPs, 2 MTMRs, PTP4A1, CDKN3, CDC25B) and PTPN13; 8 DSPs were found in both groups. Furthermore, 7 PTPs/DSPs displayed also altered protein level. Analysis of 2 breast cancer datasets identified 6 differentially expressed DSPs: DUSP6, strongly up-regulated in both datasets; DUSP10 and CDC25B, up-regulated; PTP4A2, CDC14A and MTMR11 down-regulated in one dataset. Conclusions: Several DSPs, mainly MKPs and, unexpectedly, MTMRs, were altered following HER2-modulation in cells and 3 DSPs (DUSP6, CDC25B and MTMR11) were altered in both cells and tumors. Among these, DUSP6, strongly up-regulated in HER2-positive tumors, would deserve further investigation as tumor marker or potential therapy target. Keywords: Tyrosine phosphatases, phosphorylation, HER2, breast cancer, array analysis 1. Introduction motility and invasion may be up-regulated [30,39,42]. However, the way HER2 over-expression deregulates About 10% of human breast cancers display over- these multiple signaling pathways is still poorly un- expressed HER2 (human epidermal growth factor re- derstood [19,39]. Within this background, PTPs (pro- ceptor, ERBB2), a member of the ERBB receptor tein tyrosine phosphatases) and DSPs (dual-specificity family [13,17]. Such tumors are highly invasive and phosphatases) might play a role, since members of metastatic, and generally do not respond to anti- these families are signaling molecules in RTK (recep- estrogen treatments [28,29]. Studies in tumors and tor tyrosine kinase) pathways. tumor-derived cell lines indicated that HER2 overex- The human genome sequence revealed that only 38 pression is mainly due to gene amplification. HER2 PTPs exist, namely 21 receptor and 17 soluble PTPs. repression, using tyrosine kinase inhibitors or mono- DSPs are approximately 65 [32] and include members clonal antibodies, is a valuable therapeutic approach of the MKPs, SSH, PTP4A, CDC14, PTEN, Myotubu- [3,20]. HER2 activation evokes stimulation of both larins, CDC25 and EYA families [1,2]. PTPs and DSPs MAPK and PI3K/AKT pathways, leading to prolifer- are positive or negative regulators in a wide range of ation and survival [40]. Also pathways governing cell signaling networks, thus contributing to control cell * proliferation, adhesion and migration, differentiation Corresponding author: Emma Villa-Moruzzi, PhD, Depart- and survival [15,32]. ment of Experimental Pathology, via Roma 55, 56126 Pisa, Italy. Tel.: +39 050 2218 551; Fax: +39 050 2218 557; E-mail: In cancer pathways, PTPs and DSPs generally play [email protected]. an anti-oncogenic role, by terminating the action of 1570-5870/10/$27.50 © 2010 – IOS Press and the authors. All rights reserved 362 M.A. Lucci et al. / Tyrosine phosphatases in HER2 breast cancer cells and tumors PTKs (protein tyrosine kinases). Examples are the Placid, NY, USA) and CDKN3 (BD Biosciences, MKPs (i.e., DSPs that dephosphorylate MAPKs) [14] Franklin, NJ, USA). The anti-CDC25C antibodies and several PTPs [32]. However, PTPs may also trans- were described previously [35]. The anti-PTPRA rab- duce positive signals, thus favoring oncogenesis. Ex- bit polyclonal antibodies were a kind gift from Dr. amples are PTPN11 [23], PTPN1 [33], PTPRE [11] J. den Hertog (Utrecht, The Netherlands). and the cell cycle regulators CDC25C, B and A [6]. In any event, PTPs and DSPs must be regulated to pre- vent havoc in the cell. Little is still known on PTPs 2.2. Cell culture, treatments and extracts and DSPs in breast cancer, particularly in the HER2- dependent type. Several studies focused on PTEN, The MDA-MB-453 cells were grown in RPMI sup- a tumor suppressor which is often mutated in tumors plemented with 10% fetal calf serum [36]. The MDA- [25]. A role in favoring HER2-evoked tumorigenesis MB-453 cells carrying the intracellular expression of was also reported for PTPN11 [21,43] and PTPN1 the p185HER2-specific single chain antibody (5R) and [4]. Other studies indicated a role for PTPN18 [10] in control MDA-MB-453 cells carrying the empty vec- HER2-overexpressing tumor cell lines and for PTPRE tor (LBSN) were grown in DMEM supplemented with in neu mice [11]. However, many more PTPs and DSPs 10% fetal calf serum and 0.5 mg/ml geneticin [8]. might be involved, given the complexity of the ERBB In brief, cDNA encoding the p185HER2-specific single receptors signaling. chain antibody 5R was cloned into the LXSN retroviral PTPs and DSPs alterations in cancer may com- vector and used to infect the packaging cell line gp + prise changes in the expression and protein levels, AM12. Following G418 selection the virus-containing or mutations, as described in the case of colon can- medium was collected and used to infect the MDA- cer [38]. Post-translation modifications may also oc- MB-453 cell line. The infection cycle was repeated cur. The present project was undertaken to iden- three times and cells were tested for p185HER2 surface tify PTPs and DSPs displaying altered expression expression by FACScan analysis. For EGF-treatment, levels in HER2-overexpressing human breast can- sub-confluent cells were serum-starved and exposed cer and cancer-derived cell lines. We analyzed the to 50 ng/ml EGF (diluted in DMSO) or to DMSO PTPs and DSPs in: (1) MDA-MB-453 cells (HER2- alone (time 0). For immunoblotting, the cells were overexpressing human breast cancer cells) carrying washes in cold PBS and lysed in 50 mM TRIS-HCl, pH permanent HER2 inhibition or activated with EGF 7.5, 150 mM NaCl, 5 mM EDTA, 1% Triton X-100, and (2) 2 public datasets of primary invasive HER2- positive breast tumors. The aim was to identify PTPs 7.5 mM 2-mercaptoethanol, added with 1 mM ortho- and DSPs displaying HER2-associated alterations, that vanadate, 50 mM NaF and protease inhibitors (0.02% might sustain tumor biology features, such as survival benzamidine, 0.02% PMSF, 0.02% TPCK, 10 µg/ml and motility, and might represent new tumor markers soybean trypsin inhibitor and 4 µg/ml leupeptine). Cell or potential therapy targets. extracts were then diluted 1:2 in Laemmli buffer and boiled. 2. Materials and methods 2.3. RNA extraction and cDNA/cRNA preparation 2.1. Materials RNA was purified from cells grown on 30 mm plates Tissue culture media and additives, fetal calf serum, using the SV total RNA isolation system (Promega, EGF, the antibodies to α-tubulin, peroxidase-conjugat- Madison, WI, USA). Labeled antisense RNA (cRNA) ed antibodies and protein A-peroxidase were pur- was synthesized using the TrueLabeling-AMP 2.0 kit chased from Sigma Aldrich (Munich, Germany). The (SABiosciences, formerly SuperArray Biotech, Fred- other commercial antibodies were as follows: phospho- erick, MA, USA). In brief, cDNA was prepared by Y1248-HER2, pan-EGFR, DUSP1, DUSP2, DUSP6, reverse transcription of total RNA (2–3 µg) and imme- MTM1 and PTEN (Santa Cruz Biotech, Heidelberg, diately used for cRNA synthesis, labeling and ampli- Germany); pan-HER2 (Cell Signaling Technology, fication in the same tube. All the rest was according Danvers, MA, USA); phospshoY1173-EGFR (Nan- to the kit instructions. Four h incubation yielded be- otools, Teningen, Germany); PTPN11 (Upstate, Lake tween 4 and 12 µg of cRNA. M.A. Lucci et al. / Tyrosine phosphatases in HER2 breast cancer cells and tumors 363 2.4. DNA arrays branes were previously treated with 5 mM phosphate buffer, 2% SDS and 2 mM 2-mercaptoethanol at 60◦C Custom oligo GEArray, displaying 60-mer oligonu- for 15 min. For immunoblot quantification, the PTP cleotide probes, targeting 103 PTPs and DSPs and or DSP protein bands from treated and control cells 5 housekeeping genes (RPS27A, GAPDH, B2M, were analyzed by densitometric scanning and normal- HSP90AB1 and ACTB), were produced by SABio- ized to the α-tubulin band analyzed in the same ex- sciences. cRNA (2 µg) was used for overnight hy- periment (i.e., the PTP or DSP/α-tubulin value ratios bridization, followed by peroxidase-based chemilumi- were calculated). Normalized data from different ex- nescence detection on X-ray films, according to the periments were averaged, as specifically indicated. Re- manufacturer’s instructions. Data extraction, normal- sults are given as percent of averages from control cells ization with the indicated housekeeping genes and (controls were LBSN cells in Fig. 1 and time 0 cells in analysis used the GEArray Expression Analysis Suite Fig. 3). (SABiosciences). Expression levels are given as the ra- tio of treated to control cells and we reported changes greater than 30%, a boundary selected to boost the re- 2.7. Public gene expression data analysis sults reliability.
Recommended publications
  • CDC25B Mediates Rapamycin-Induced Oncogenic Responses in Cancer Cells
    Published OnlineFirst March 10, 2009; DOI: 10.1158/0008-5472.CAN-08-3222 Research Article CDC25B Mediates Rapamycin-Induced Oncogenic Responses in Cancer Cells Run-qiang Chen,1 Qing-kai Yang,1 Bing-wen Lu,2 Wei Yi,1 Greg Cantin,2 Yan-ling Chen,1 Colleen Fearns,3 John R. Yates III,2 and Jiing-Dwan Lee1 Departments of 1Immunology and Microbial Science, 2Chemical Physiology, and 3Chemistry, The Scripps Research Institute, La Jolla, California Abstract expression of PTEN, increased PI3K activity, and increased expression or activation of AKT in advanced prostate cancer Because the mammalian target of rapamycin (mTOR) pathway (8–10). These aberrations also are indicators of a poor prognosis is commonly deregulated in human cancer, mTOR inhibitors, for prostate cancer patients (11, 12). More importantly, long-term rapamycin and its derivatives, are being actively tested in androgen deprivation treatment for prostate cancer patients that cancer clinical trials. Clinical updates indicate that the reinforces the PI3K/AKT pathway also up-regulates mTOR anticancer effect of these drugs is limited, perhaps due to activation in prostate tumor (9, 10). These abovementioned rapamycin-dependent induction of oncogenic cascades by an experimental and clinical data lead to the supposition that mTOR as yet unclear mechanism. As such, we investigated rapamy- inhibitors (rapamycin and its derivatives) should be effective in cin-dependent phosphoproteomics and discovered that 250 treating human cancer. Unfortunately, recent clinical data indicates phosphosites in 161 cellular proteins were sensitive to that rapamycin shows therapeutic potential in only few types of rapamycin. Among these, rapamycin regulated four kinases human cancer: endometrial carcinoma, renal cell carcinoma, and and four phosphatases.
    [Show full text]
  • 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]
  • Analysis of the Stability of 70 Housekeeping Genes During Ips Reprogramming Yulia Panina1,2*, Arno Germond1 & Tomonobu M
    www.nature.com/scientificreports OPEN Analysis of the stability of 70 housekeeping genes during iPS reprogramming Yulia Panina1,2*, Arno Germond1 & Tomonobu M. Watanabe1 Studies on induced pluripotent stem (iPS) cells highly rely on the investigation of their gene expression which requires normalization by housekeeping genes. Whether the housekeeping genes are stable during the iPS reprogramming, a transition of cell state known to be associated with profound changes, has been overlooked. In this study we analyzed the expression patterns of the most comprehensive list to date of housekeeping genes during iPS reprogramming of a mouse neural stem cell line N31. Our results show that housekeeping genes’ expression fuctuates signifcantly during the iPS reprogramming. Clustering analysis shows that ribosomal genes’ expression is rising, while the expression of cell-specifc genes, such as vimentin (Vim) or elastin (Eln), is decreasing. To ensure the robustness of the obtained data, we performed a correlative analysis of the genes. Overall, all 70 genes analyzed changed the expression more than two-fold during the reprogramming. The scale of this analysis, that takes into account 70 previously known and newly suggested genes, allowed us to choose the most stable of all genes. We highlight the fact of fuctuation of housekeeping genes during iPS reprogramming, and propose that, to ensure robustness of qPCR experiments in iPS cells, housekeeping genes should be used together in combination, and with a prior testing in a specifc line used in each study. We suggest that the longest splice variants of Rpl13a, Rplp1 and Rps18 can be used as a starting point for such initial testing as the most stable candidates.
    [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]
  • 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]
  • Review Article PTEN Gene: a Model for Genetic Diseases in Dermatology
    The Scientific World Journal Volume 2012, Article ID 252457, 8 pages The cientificWorldJOURNAL doi:10.1100/2012/252457 Review Article PTEN Gene: A Model for Genetic Diseases in Dermatology Corrado Romano1 and Carmelo Schepis2 1 Unit of Pediatrics and Medical Genetics, I.R.C.C.S. Associazione Oasi Maria Santissima, 94018 Troina, Italy 2 Unit of Dermatology, I.R.C.C.S. Associazione Oasi Maria Santissima, 94018 Troina, Italy Correspondence should be addressed to Carmelo Schepis, [email protected] Received 19 October 2011; Accepted 4 January 2012 Academic Editors: G. Vecchio and H. Zitzelsberger Copyright © 2012 C. Romano and C. Schepis. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. PTEN gene is considered one of the most mutated tumor suppressor genes in human cancer, and it’s likely to become the first one in the near future. Since 1997, its involvement in tumor suppression has smoothly increased, up to the current importance. Germline mutations of PTEN cause the PTEN hamartoma tumor syndrome (PHTS), which include the past-called Cowden, Bannayan- Riley-Ruvalcaba, Proteus, Proteus-like, and Lhermitte-Duclos syndromes. Somatic mutations of PTEN have been observed in glioblastoma, prostate cancer, and brest cancer cell lines, quoting only the first tissues where the involvement has been proven. The negative regulation of cell interactions with the extracellular matrix could be the way PTEN phosphatase acts as a tumor suppressor. PTEN gene plays an essential role in human development. A recent model sees PTEN function as a stepwise gradation, which can be impaired not only by heterozygous mutations and homozygous losses, but also by other molecular mechanisms, such as transcriptional regression, epigenetic silencing, regulation by microRNAs, posttranslational modification, and aberrant localization.
    [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]
  • Efficient Immune Cell Genome Engineering with Improved CRISPR Editing Tools
    bioRxiv preprint doi: https://doi.org/10.1101/2020.02.13.947002; this version posted February 14, 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 4.0 International license. 1 Title: Efficient Immune Cell Genome Engineering with Improved CRISPR Editing Tools 2 3 Authors: 4 Waipan Chan1,*, Rachel A. Gottschalk1,4, Yikun Yao2, Joel L. Pomerantz3 & Ronald N. Germain1,* 5 6 Affiliation: 7 1Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy 8 and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892 9 2Molecular Development of the Immune System Section, Laboratory of Immune System Biology, 10 National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 11 20892 12 3Department of Biological Chemistry and Institute for Cell Engineering, The Johns Hopkins 13 University School of Medicine, Baltimore, MD 21205 14 4Current Address: Department of Immunology, University of Pittsburgh School of Medicine, 15 W1047 Biomedical Science Tower, 200 Lothrop Street, Pittsburgh, PA 15261 16 17 *Corresponding authors: [email protected]; [email protected] 18 19 This work was supported by the Intramural Research Program of NIAID, NIH. 20 21 All animal experiments were done in accordance with the guidelines of the NIAID/NIH 22 Institutional Animal Care and Use Committee. 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.02.13.947002; this version posted February 14, 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.
    [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]
  • Bioinformatics-Based Screening of Key Genes for Transformation of Liver
    Jiang et al. J Transl Med (2020) 18:40 https://doi.org/10.1186/s12967-020-02229-8 Journal of Translational Medicine RESEARCH Open Access Bioinformatics-based screening of key genes for transformation of liver cirrhosis to hepatocellular carcinoma Chen Hao Jiang1,2, Xin Yuan1,2, Jiang Fen Li1,2, Yu Fang Xie1,2, An Zhi Zhang1,2, Xue Li Wang1,2, Lan Yang1,2, Chun Xia Liu1,2, Wei Hua Liang1,2, Li Juan Pang1,2, Hong Zou1,2, Xiao Bin Cui1,2, Xi Hua Shen1,2, Yan Qi1,2, Jin Fang Jiang1,2, Wen Yi Gu4, Feng Li1,2,3 and Jian Ming Hu1,2* Abstract Background: Hepatocellular carcinoma (HCC) is the most common type of liver tumour, and is closely related to liver cirrhosis. Previous studies have focussed on the pathogenesis of liver cirrhosis developing into HCC, but the molecular mechanism remains unclear. The aims of the present study were to identify key genes related to the transformation of cirrhosis into HCC, and explore the associated molecular mechanisms. Methods: GSE89377, GSE17548, GSE63898 and GSE54236 mRNA microarray datasets from Gene Expression Omni- bus (GEO) were analysed to obtain diferentially expressed genes (DEGs) between HCC and liver cirrhosis tissues, and network analysis of protein–protein interactions (PPIs) was carried out. String and Cytoscape were used to analyse modules and identify hub genes, Kaplan–Meier Plotter and Oncomine databases were used to explore relationships between hub genes and disease occurrence, development and prognosis of HCC, and the molecular mechanism of the main hub gene was probed using Kyoto Encyclopedia of Genes and Genomes(KEGG) pathway analysis.
    [Show full text]
  • DUSP10/MKP5 Antibody A
    Revision 1 C 0 2 - t DUSP10/MKP5 Antibody a e r o t S Orders: 877-616-CELL (2355) [email protected] Support: 877-678-TECH (8324) 3 8 Web: [email protected] 4 www.cellsignal.com 3 # 3 Trask Lane Danvers Massachusetts 01923 USA For Research Use Only. Not For Use In Diagnostic Procedures. Applications: Reactivity: Sensitivity: MW (kDa): Source: UniProt ID: Entrez-Gene Id: WB H M R Endogenous 54 Rabbit Q9Y6W6 11221 Product Usage Information 3. Salojin, K. and Oravecz, T. (2007) J Leukoc Biol 81, 860-9. 4. Tanoue, T. et al. (2002) J Biol Chem 277, 22942-9. Application Dilution 5. Dickinson, R.J. and Keyse, S.M. (2006) J Cell Sci 119, 4607-15. 6. Wu, G.S. (2007) Cancer Metastasis Rev 26, 579-85. Western Blotting 1:1000 7. Teng, C.H. et al. (2007) J Biol Chem 282, 28395-407. 8. Zhang, Y. et al. (2004) Nature 430, 793-7. Storage Supplied in 10 mM sodium HEPES (pH 7.5), 150 mM NaCl, 100 µg/ml BSA and 50% glycerol. Store at –20°C. Do not aliquot the antibody. Specificity / Sensitivity DUSP10/MKP5 Antibody detects endogenous levels of total DUSP10 protein. Species Reactivity: Human, Mouse, Rat Source / Purification Polyclonal antibodies are produced by immunizing animals with a synthetic peptide corresponding to human DUSP10. Antibodies are purified by protein A and peptide affinity chromatography. Background MAP kinases are inactivated by dual-specificity protein phosphatases (DUSPs) that differ in their substrate specificity, tissue distribution, inducibility by extracellular stimuli, and cellular localization. DUSPs, also known as MAPK phosphatases (MKP), specifically dephosphorylate both threonine and tyrosine residues in MAPK P-loops and have been shown to play important roles in regulating the function of the MAPK family (1,2).
    [Show full text]