Identification and Characterization of Specific Inhibitors Of

Total Page:16

File Type:pdf, Size:1020Kb

Identification and Characterization of Specific Inhibitors Of IDENTIFICATION AND CHARACTERIZATION OF SPECIFIC INHIBITORS OF THE EYA2 PHOSPHATASE by AARON B. KRUEGER B.S., North Dakota State University, 2006 A thesis submitted to the Faculty of the Graduate School of the University of Colorado in partial fulfillment of the requirements for the degree of Doctor of Philosophy Structural Biology and Biochemistry 2013 This thesis for the Doctor of Philosophy degree by Aaron B. Krueger has been approved for the Structural Biology and Biochemistry by Robert Hodges, Chair Heide Ford Changwei Liu Steve Nordeen Rui Zhao, Advisor Date ___10/21/13____________ ii Krueger, Aaron B. (Ph.D., Structural Biology and Biochemistry) Identification and characterization of specific inhibitors of the Eya2 phosphatase Thesis directed by Associate Professor Rui Zhao. ABSTRACT Eya proteins are transcriptional co-activators of the homeobox gene Six1 and contain a unique protein tyrosine phosphatase activity. Eya and Six1 are required for normal development and are down-regulated in most adult tissues. However, Six1 and Eya are re-expressed in a large number of breast tumors and play a causal role in the initiation and development of these tumors. Eya’s unique phosphatase activity has been shown to be important for the transformation, migration, invasion, and metastasis of breast cancer cells. Because of its mechanistically unique phosphatase activity and its important role in the establishment and spread of breast cancer, I targeted Eya for anti-breast cancer therapy. To this end, I developed an HTS strategy to identify inhibitors of Eya2’s phosphatase activity. In collaboration with the NIH Chemical Genomics Center, I screened over 330,000 compounds and have identified a series of compounds effective at inhibiting Eya2’s phosphatase activity. Cell-based migration assays using MCF10A breast cells have demonstrated an import role for Eya2’s phosphatase activity and that these identified phosphatase inhibitors can effectively inhibit migration of these cells. Using biochemical, biophysical, and structural approaches, I further characterized these inhibitors as reversible, iii mixed mode inhibitors that likely bind at an allosteric site, with a KD of 2.0 µM. I have demonstrated that these inhibitors are specific towards Eya2 over other mechanistically similar phosphatases including its highly-conserved family member, Eya3. These characterizations are leading us to develop a model of how the inhibitor could specifically interact and inhibit Eya2 which will help us to further improve potency to make a viable anti-breast cancer drug. The form and content of this abstract are approved. I recommend its publication. Approved: Rui Zhao iv DEDICATION I dedicate this work to ‘the’ wife Katie who happily provided strength and support throughout the entire process, to my parents who kept me curious and never said no, and to my friends who kept me sane when I otherwise would not be. v ACKNOWLEDGMENTS I would first like to thank the members of my committee for their many insightful questions and advice through the progression of the project. I would like to thank the prior and past members of the Zhao lab for the fun times, the assistance provided, and support given. I would like to especially thank Dr. Zhao for allowing me the opportunity to join her lab and for the years of teaching me to become a successful scientist. I would like to thank our collaborators whose contributions were instrumental in allowing the project to progress. Specifically, I would like to thank members of the NIH Chemical Genomics Center for their contributions to the high throughput screening, of the University of Colorado BioPhysics CORE for their assistance and guidance, Philip Reigan and lab for assistance in modeling, Elan Eisenmesser and lab for assistance with NMR experiments, and Heide Ford and lab for essential contributions. Without these people, I would be nowhere. Finally, for the years of guidance and support, I would like to thank the fellow students, faculty, and staff of the Department of Biochemistry and Molecular Genetics, and specifically those of the Structural Biology and Biochemistry Program. vi CONTENTS CHAPTER I. INTRODUCTION ............................................................................................... 1 Transcription complexes as therapeutic targets ................................... 1 The Six1/Eya transcriptional complex ................................................... 2 Six1 and Eya in cancer ......................................................................... 6 The transcription activator Eya is a HAD family phosphatase .............. 7 Functions of Eya’s protein tyrosine phosphatase activity ................... 15 Targeting protein tyrosine phosphatases for therapeutic drug design 17 Targeting the Eya protein tyrosine phosphatase activity .................... 21 II. THE IDENTIFICATION OF EYA2 PHOSPHATASE INHIBITORS .................. 23 Abstract .............................................................................................. 23 Introduction ......................................................................................... 24 Results ............................................................................................... 27 Design of assays suitable for HTS ................................................ 27 Known phosphatase inhibitors do not significantly inhibit Eya2’s phosphatase activity ...................................................................... 31 The OMFP-based phosphatase assay is suitable for HTS ............ 32 A large scale primary screen identified a class of structurally related initial hits ....................................................................................... 35 The best compounds from this series are active in a pH2AX-based secondary phosphatase assay ...................................................... 38 The N-arylidenebenzohydrazide compounds do not inhibit other cellular phosphatases ................................................................... 40 Discussion .......................................................................................... 41 Methods .............................................................................................. 45 vii Protein expression and purification ............................................... 45 pNPP-based Eya phosphatase assay ........................................... 45 OMFP-based Eya phosphatase assay .......................................... 46 Miniaturized Eya phosphatase assay for HTS .............................. 46 Compound library and instruments for liquid handling .................. 47 HTS data analysis ......................................................................... 48 pH2AX-based Eya phosphatase assay ......................................... 48 Phosphatase assays of PTP1B, PPM1A, and Scp1 ...................... 49 III. DETERMINING THE MECHANISM OF ACTION OF EYA2 INHIBITORS..... 51 Abstract .............................................................................................. 51 Introduction ......................................................................................... 52 Results ............................................................................................... 55 HTS identified a new chemical series that specifically inhibit Eya2’s phosphatase activity. ..................................................................... 55 Enzyme kinetic analyses suggest that the N- arylidenebenzohydrazide compounds are mixed mode inhibitors. 58 Attempt to determine the binding mechanism using X-ray crystallography. ............................................................................. 60 Attempt to determine the binding mechanism using NMR. ........... 61 Compounds likely do not bind in the active site and do not require Mg2+ for function. ........................................................................... 65 Compounds likely bind to an allosteric site as determined by mutational analysis. ....................................................................... 72 Evaluate the effect of inhibitors on cellular phenotypes. ............... 81 Discussion .......................................................................................... 83 Methods .............................................................................................. 90 Protein expression and purification ............................................... 90 viii Eya2 phosphatase assays ............................................................ 91 Eya2 kinetic experiments .............................................................. 91 Isothermal titration calorimetry ...................................................... 92 Circular dichroism ......................................................................... 93 NMR spectroscopy ........................................................................ 93 UV-Vis spectra analysis of selected hydrazides ............................ 94 Molecular docking ......................................................................... 94 Motility assay................................................................................. 95 Cell lines ....................................................................................... 95 IV. DEVELOPMENT OF ASSAYS TARGETING OTHER ASPECTS OF SIX1 ACTION .............................................................................................................. 96 Abstract .............................................................................................
Recommended publications
  • 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]
  • Circhipk3 Facilitates the G2/M Transition in Prostate Cancer Cells by Sponging Mir-338-3P
    OncoTargets and Therapy Dovepress open access to scientific and medical research Open Access Full Text Article ORIGINAL RESEARCH CircHIPK3 Facilitates the G2/M Transition in Prostate Cancer Cells by Sponging miR-338-3p This article was published in the following Dove Press journal: OncoTargets and Therapy Fengchun Liu1 Background: Circular RNAs (circRNAs) play a crucial role in gene expression regulation. Yanru Fan 1 CircHIPK3 is a circRNA derived from Exon 2 of HIPK3 gene and its role in prostate cancer Liping Ou1 (PCa) is still unclear. fl Ting Li1 Methods: CCK8 assays, ow cytometry and colony formation assays were performed to assess Jiaxin Fan1 the effects of circHIPK3 in PCa cells. Bioinformatics analysis, RNA pull-down assay, RNA immunoprecipitation assay (RIP), and luciferase activity assay were performed to dissect the Limei Duan1 mechanism underlying circHIPK3-mediated G2/M transition in PCa cells. Jinxiao Yang1 1 Results: CircHIPK3 expression was upregulated in PCa cells and prostate cancer tissues. Chunli Luo Overexpression of circHIPK3 or circHIPK3 silencing altered PCa viability, proliferation and 2 Xiaohou Wu apoptosis in vitro. CircHIPK3 could sponge miR-338-3p and inhibit its activity, resulting in 1Department of Laboratory Diagnosis, increased expression of Cdc25B and Cdc2 in vitro. Chongqing Medical University, Yuzhong, Conclusion: CircHIPK3 promotes G2/M transition and induces PCa cell proliferation by Chongqing 408000, People’s Republic of China; 2Department of Urology, The First sponging miR-338-3p and increasing the
    [Show full text]
  • Mechanisms of Action for Small Molecules Revealed by Structural Biology in Drug Discovery
    International Journal of Molecular Sciences Review Mechanisms of Action for Small Molecules Revealed by Structural Biology in Drug Discovery Qingxin Li 1,* and CongBao Kang 2,* 1 Guangdong Provincial Engineering Laboratory of Biomass High Value Utilization, Guangdong Provincial Bioengineering Institute (Guangzhou Sugarcane Industry Research Institute), Guangdong Academy of Sciences, Guangzhou 510316, China 2 Experimental Drug Development Centre (EDDC), Agency for Science, Technology and Research (A*STAR), 10 Biopolis Road, Chromos, #05-01, Singapore 138670, Singapore * Correspondence: [email protected] (Q.L.); [email protected] (C.K.); Tel.: +86-020-84168436 (Q.L.); +65-64070602 (C.K.) Received: 12 June 2020; Accepted: 20 July 2020; Published: 24 July 2020 Abstract: Small-molecule drugs are organic compounds affecting molecular pathways by targeting important proteins. These compounds have a low molecular weight, making them penetrate cells easily. Small-molecule drugs can be developed from leads derived from rational drug design or isolated from natural resources. A target-based drug discovery project usually includes target identification, target validation, hit identification, hit to lead and lead optimization. Understanding molecular interactions between small molecules and their targets is critical in drug discovery. Although many biophysical and biochemical methods are able to elucidate molecular interactions of small molecules with their targets, structural biology is the most powerful tool to determine the mechanisms of action for both targets and the developed compounds. Herein, we reviewed the application of structural biology to investigate binding modes of orthosteric and allosteric inhibitors. It is exemplified that structural biology provides a clear view of the binding modes of protease inhibitors and phosphatase inhibitors.
    [Show full text]
  • Supplementary Table S1. Correlation Between the Mutant P53-Interacting Partners and PTTG3P, PTTG1 and PTTG2, Based on Data from Starbase V3.0 Database
    Supplementary Table S1. Correlation between the mutant p53-interacting partners and PTTG3P, PTTG1 and PTTG2, based on data from StarBase v3.0 database. PTTG3P PTTG1 PTTG2 Gene ID Coefficient-R p-value Coefficient-R p-value Coefficient-R p-value NF-YA ENSG00000001167 −0.077 8.59e-2 −0.210 2.09e-6 −0.122 6.23e-3 NF-YB ENSG00000120837 0.176 7.12e-5 0.227 2.82e-7 0.094 3.59e-2 NF-YC ENSG00000066136 0.124 5.45e-3 0.124 5.40e-3 0.051 2.51e-1 Sp1 ENSG00000185591 −0.014 7.50e-1 −0.201 5.82e-6 −0.072 1.07e-1 Ets-1 ENSG00000134954 −0.096 3.14e-2 −0.257 4.83e-9 0.034 4.46e-1 VDR ENSG00000111424 −0.091 4.10e-2 −0.216 1.03e-6 0.014 7.48e-1 SREBP-2 ENSG00000198911 −0.064 1.53e-1 −0.147 9.27e-4 −0.073 1.01e-1 TopBP1 ENSG00000163781 0.067 1.36e-1 0.051 2.57e-1 −0.020 6.57e-1 Pin1 ENSG00000127445 0.250 1.40e-8 0.571 9.56e-45 0.187 2.52e-5 MRE11 ENSG00000020922 0.063 1.56e-1 −0.007 8.81e-1 −0.024 5.93e-1 PML ENSG00000140464 0.072 1.05e-1 0.217 9.36e-7 0.166 1.85e-4 p63 ENSG00000073282 −0.120 7.04e-3 −0.283 1.08e-10 −0.198 7.71e-6 p73 ENSG00000078900 0.104 2.03e-2 0.258 4.67e-9 0.097 3.02e-2 Supplementary Table S2.
    [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]
  • Identification of Candidate Genes and Pathways Associated with Obesity
    animals Article Identification of Candidate Genes and Pathways Associated with Obesity-Related Traits in Canines via Gene-Set Enrichment and Pathway-Based GWAS Analysis Sunirmal Sheet y, Srikanth Krishnamoorthy y , Jihye Cha, Soyoung Choi and Bong-Hwan Choi * Animal Genome & Bioinformatics, National Institute of Animal Science, RDA, Wanju 55365, Korea; [email protected] (S.S.); [email protected] (S.K.); [email protected] (J.C.); [email protected] (S.C.) * Correspondence: [email protected]; Tel.: +82-10-8143-5164 These authors contributed equally. y Received: 10 October 2020; Accepted: 6 November 2020; Published: 9 November 2020 Simple Summary: Obesity is a serious health issue and is increasing at an alarming rate in several dog breeds, but there is limited information on the genetic mechanism underlying it. Moreover, there have been very few reports on genetic markers associated with canine obesity. These studies were limited to the use of a single breed in the association study. In this study, we have performed a GWAS and supplemented it with gene-set enrichment and pathway-based analyses to identify causative loci and genes associated with canine obesity in 18 different dog breeds. From the GWAS, the significant markers associated with obesity-related traits including body weight (CACNA1B, C22orf39, U6, MYH14, PTPN2, SEH1L) and blood sugar (PRSS55, GRIK2), were identified. Furthermore, the gene-set enrichment and pathway-based analysis (GESA) highlighted five enriched pathways (Wnt signaling pathway, adherens junction, pathways in cancer, axon guidance, and insulin secretion) and seven GO terms (fat cell differentiation, calcium ion binding, cytoplasm, nucleus, phospholipid transport, central nervous system development, and cell surface) which were found to be shared among all the traits.
    [Show full text]
  • DNA Methylation Profiling Identifies the HOXA11 Gene As an Early Diagnostic and Prognostic Molecular Marker in Human Lung Adenocarcinoma
    www.impactjournals.com/oncotarget/ Oncotarget, 2017, Vol. 8, (No. 20), pp: 33100-33109 Research Paper DNA methylation profiling identifies the HOXA11 gene as an early diagnostic and prognostic molecular marker in human lung adenocarcinoma Qun Li1,2,*, Chang Chen3, Xiaohui Ren1, Weihong Sun1,* 1Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200031, China 2The State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China 3Department of Orthodontics, The First Affiliated Hospital of Zhengzhou University, Stomatological College Zhengzhou University, Zhengzhou, 450052, China *These authors contributed equally to this work Correspondence to: Weihong Sun, email: [email protected] Keywords: HOXA11, hypermethylation, lung adenocarcinoma, adenocarcinoma in situ, prognosis Received: October 11, 2016 Accepted: March 14, 2017 Published: March 23, 2017 Copyright: Li et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC-BY), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. ABSTRACT DNA hypermethylation plays important roles in carcinogenesis by silencing key genes. The goal of our study was to identify pivotal genes using MethyLight and assessed their diagnostic and prognostic values in lung adenocarcinoma (AD). In the present study, we detected DNA methylation at sixteen loci promoter regions in twenty one pairs of primary human lung AD tissues and adjacent non-tumor lung (AdjNL) tissues using the real-time PCR (RT-PCR)-based method MethyLight.
    [Show full text]
  • Mirc11 Disrupts Inflammatory but Not Cytotoxic Responses of NK Cells
    Published OnlineFirst September 12, 2019; DOI: 10.1158/2326-6066.CIR-18-0934 Research Article Cancer Immunology Research Mirc11 Disrupts Inflammatory but Not Cytotoxic Responses of NK Cells Arash Nanbakhsh1, Anupallavi Srinivasamani1, Sandra Holzhauer2, Matthew J. Riese2,3,4, Yongwei Zheng5, Demin Wang4,5, Robert Burns6, Michael H. Reimer7,8, Sridhar Rao7,8, Angela Lemke9,10, Shirng-Wern Tsaih9,10, Michael J. Flister9,10, Shunhua Lao1,11, Richard Dahl12, Monica S. Thakar1,11, and Subramaniam Malarkannan1,3,4,9,11 Abstract Natural killer (NK) cells generate proinflammatory cyto- g–dependent clearance of Listeria monocytogenes or B16F10 kines that are required to contain infections and tumor melanoma in vivo by NK cells. These functional changes growth. However, the posttranscriptional mechanisms that resulted from Mirc11 silencing ubiquitin modifiers A20, regulate NK cell functions are not fully understood. Here, we Cbl-b, and Itch, allowing TRAF6-dependent activation of define the role of the microRNA cluster known as Mirc11 NF-kB and AP-1. Lack of Mirc11 caused increased translation (which includes miRNA-23a, miRNA-24a, and miRNA-27a) of A20, Cbl-b, and Itch proteins, resulting in deubiquityla- in NK cell–mediated proinflammatory responses. Absence tion of scaffolding K63 and addition of degradative K48 of Mirc11 did not alter the development or the antitumor moieties on TRAF6. Collectively, our results describe a func- cytotoxicity of NK cells. However, loss of Mirc11 reduced tion of Mirc11 that regulates generation of proinflammatory generation of proinflammatory factors in vitro and interferon- cytokines from effector lymphocytes. Introduction TRAF2 and TRAF6 promote K63-linked polyubiquitination that is required for subcellular localization of the substrates (20), Natural killer (NK) cells generate proinflammatory factors and and subsequent activation of NF-kB (21) and AP-1 (22).
    [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]
  • Phosphatome Rnai Screen Identifies Eya1 As a Positive Regulator of Hedgehog Signal Transduction
    Phosphatome RNAi Screen Identifies Eya1 as a Positive Regulator of Hedgehog Signal Transduction The Harvard community has made this article openly available. Please share how this access benefits you. Your story matters Citation Eisner, Adriana. 2013. Phosphatome RNAi Screen Identifies Eya1 as a Positive Regulator of Hedgehog Signal Transduction. Doctoral dissertation, Harvard University. Citable link http://nrs.harvard.edu/urn-3:HUL.InstRepos:11181213 Terms of Use This article was downloaded from Harvard University’s DASH repository, and is made available under the terms and conditions applicable to Other Posted Material, as set forth at http:// nrs.harvard.edu/urn-3:HUL.InstRepos:dash.current.terms-of- use#LAA Phosphatome RNAi Screen Identifies Eya1 as a Positive Regulator of Hedgehog Signal Transduction A dissertation presented by Adriana Eisner to The Division of Medical Sciences in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the subject of Neurobiology Harvard University Cambridge, Massachusetts June 2013 © 2013 Adriana Eisner All rights reserved. Dissertation Advisor: Dr. Rosalind Segal Adriana Eisner Phosphatome RNAi Screen Identifies Eya1 as a Positive Regulator of Hedgehog Signal Transduction Abstract The Hedgehog (Hh) signaling pathway is vital for vertebrate embryogenesis and aberrant activation of the pathway can cause tumorigenesis in humans. In this study, we used a phosphatome RNAi screen for regulators of Hh signaling to identify a member of the Eyes Absent protein family, Eya1, as a positive regulator of Hh signal transduction. Eya1 is both a phosphatase and transcriptional regulator. Eya family members have been implicated in tumor biology, and Eya1 is highly expressed in a particular subtype of medulloblastoma (MB).
    [Show full text]
  • Eya1-Dependent Homeostasis of Morphogenetic Territories During Collective Cell Migration
    bioRxiv preprint doi: https://doi.org/10.1101/2021.01.27.428404; this version posted January 27, 2021. 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. Eya1-dependent homeostasis of morphogenetic territories during collective cell migration Jerónimo R. Miranda-Rodríguez§, Augusto Borges§, Filipe Pinto-Teixeira#, Indra ∫ Wibowo , Hans-Martin Pogoda¶, Matthias Hammerschmidt¶, Koichi Kawakami‡ and Hernán López-Schier§#* § Unit of Sensory Biology & Organogenesis, Helmholtz Zentrum Munchen, Munich, Germany ¶ Institute for Developmental Biology, University of Cologne, Cologne, Germany ∫ School of Technology and Life Sciences, Bandung, Indonesia # Center for Developmental Biology, Université Paul Sabatier, Toulouse, France ‡ Laboratory of Molecular and Developmental Biology, National Institute of Genetics SOKENDAI, 1111 Yata Mishima, Shizuoka 411-8540, Japan * Corresponding author: [email protected] SUMMARY Tissue remodeling presents an enormous challenge to the stability of intercellular signaling domains. Here we investigate this issue during the development of the posterior lateral line in zebrafish. We find that the transcriptional co-activator and phosphatase Eya1, mutated in the branchio-oto-renal syndrome in humans, is essential for the homeostasis of the Wnt/β- catenin and FGF morphogenetic domains during the collective migration of lateral-line primordial cells. Loss of Eya1 strongly diminishes the expression of Dkk1, expanding Wnt/β- catenin activity in the primordium, which in turn abrogates FGFR1 expression. Deficits in Eya1 also abolishes the expression of the chemokine receptor CXCR7b, disrupting primordium migration.
    [Show full text]
  • Genetic Testing Medical Policy – Genetics
    Genetic Testing Medical Policy – Genetics Please complete all appropriate questions fully. Suggested medical record documentation: • Current History & Physical • Progress Notes • Family Genetic History • Genetic Counseling Evaluation *Failure to include suggested medical record documentation may result in delay or possible denial of request. PATIENT INFORMATION Name: Member ID: Group ID: PROCEDURE INFORMATION Genetic Counseling performed: c Yes c No **Please check the requested analyte(s), identify number of units requested, and provide indication/rationale for testing. 81400 Molecular Pathology Level 1 Units _____ c ACADM (acyl-CoA dehydrogenase, C-4 to C-12 straight chain, MCAD) (e.g., medium chain acyl dehydrogenase deficiency), K304E variant _____ c ACE (angiotensin converting enzyme) (e.g., hereditary blood pressure regulation), insertion/deletion variant _____ c AGTR1 (angiotensin II receptor, type 1) (e.g., essential hypertension), 1166A>C variant _____ c BCKDHA (branched chain keto acid dehydrogenase E1, alpha polypeptide) (e.g., maple syrup urine disease, type 1A), Y438N variant _____ c CCR5 (chemokine C-C motif receptor 5) (e.g., HIV resistance), 32-bp deletion mutation/794 825del32 deletion _____ c CLRN1 (clarin 1) (e.g., Usher syndrome, type 3), N48K variant _____ c DPYD (dihydropyrimidine dehydrogenase) (e.g., 5-fluorouracil/5-FU and capecitabine drug metabolism), IVS14+1G>A variant _____ c F13B (coagulation factor XIII, B polypeptide) (e.g., hereditary hypercoagulability), V34L variant _____ c F2 (coagulation factor 2) (e.g.,
    [Show full text]