ERK Regulates Hif1α-Mediated Platinum Resistance by Directly Targeting PHD2 in Ovarian Cancer

Total Page:16

File Type:pdf, Size:1020Kb

ERK Regulates Hif1α-Mediated Platinum Resistance by Directly Targeting PHD2 in Ovarian Cancer Published OnlineFirst July 8, 2019; DOI: 10.1158/1078-0432.CCR-18-4145 Translational Cancer Mechanisms and Therapy Clinical Cancer Research ERK Regulates HIF1a-Mediated Platinum Resistance by Directly Targeting PHD2 in Ovarian Cancer Zhuqing Li1,2, Wei Zhou1,2,3, Yi Zhang1,2, Wei Sun4, Mingo M.H. Yung5, Jing Sun1,2, Jing Li1,2, Chi-Wei Chen1,2, Zongzhu Li1,2, Yunxiao Meng1,2, Jie Chai1,2, Yuan Zhou1,2, Stephanie S. Liu6, Annie N.Y. Cheung6, Hextan Y.S. Ngan5, David W. Chan5, Wei Zheng4, and Wenge Zhu1,2 Abstract Purpose: Up to 80% of patients with ovarian cancer Results: YC-1 and selumetinib resensitized PROC cells to develop platinum resistance over time to platinum-based cisplatin. Next, the prolyl hydroxylase domain-containing chemotherapy. Increased HIF1a level is an important mech- protein 2 (PHD2) was shown to be a direct substrate of anism governing platinum resistance in platinum-resistant ERK. Phosphorylation of PHD2 by ERK prevents its binding ovarian cancer (PROC). However, the mechanism regulat- to HIF1a, thus inhibiting HIF1a hydroxylation and degra- ing HIF1a stability in PROC remains largely unknown. dation—increasing HIF1a stability. Significantly, ERK/ Here, we elucidate the mechanism of HIF1a stability reg- PHD2 signaling in PROC cells is dependent on TGFb1, ulation in PROC and explore therapeutic approaches to promoting platinum resistance by stabilizing HIF1a.Inhi- overcome cisplatin resistance in ovarian cancer. bition of TGFb1 by SB431542, ERK by selumetinib, or Experimental Design: We first used a quantitative high- HIF1a by YC-1 efficiently overcame platinum resistance throughput combinational screen (qHTCS) to identify novel both in vitro and in vivo. The results from clinical samples drugs that could resensitize PROC cells to cisplatin. Next, we confirm activation of the ERK/PHD2/HIF1a axis in patients evaluated the combination efficacy of inhibitors of HIF1a with PROC, correlating highly with poor prognoses for (YC-1), ERK (selumetinib), and TGFb1 (SB431542) with patients. platinum drugs by in vitro and in vivo experiments. Moreover, Conclusions: HIF1a stabilization is regulated by TGFb1/ anovelTGFb1/ERK/PHD2-mediated pathway regulating ERK/PHD2 axis in PROC. Hence, inhibiting TGFb1, ERK, or HIF1a stability in PROC was discovered. HIF1a is potential strategy for treating patients with PROC. Introduction platinum drugs is a big hurdle for the treatment of ovarian cancer. Thus, treatments for platinum-resistant ovarian cancer (PROC) Most patients with ovarian cancer initially respond well to remain an unmet need; research into treatments will improve platinum drug-based chemotherapy, but up to 80% patients survival and quality of life for these patients with PROC (1–4). relapse when becoming resistant to platinum. Resistance to Hypoxia-inducible factor-1a (HIF1a) is a subunit of HIF1, a heterodimeric transcription factor that is considered the master 1Department of Biochemistry and Molecular Medicine, The George Washington transcriptional regulator of multiple cellular pathways including University School of Medicine and Health Sciences, Washington, District of cell proliferation, tumor migration, and angiogenesis (5, 6). Ele- Columbia. 2GW Cancer Center, The George Washington University, Washington, vated levels of HIF1a are associated with tumor metastasis and District of Columbia. 3Department of Colorectal Surgery, Sir Run Shaw Hospital, poor patient prognosis as well as chemoresistance in several School of Medicine, Zhejiang University, Hangzhou, China. 4National Center for 5 tumors including breast cancer, oropharyngeal cancer, and ovar- Advancing Translational Sciences, NIH, Bethesda, Maryland. Department of ian cancer (7, 8). HIF1a contains an oxygen-dependent degra- Obstetrics and Gynecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China. 6Department of Pathology, LKS Faculty of dation domain (ODDD, residues 410-603), which is responsible Medicine, The University of Hong Kong, Hong Kong SAR, China. for its rapid degradation under normoxic conditions (9). In the presence of oxygen, HIF1a protein is hydroxylated by prolyl Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). hydroxylase (PHD) domain enzymes on two key proline residues (P402/P564) located within the ODDD, resulting in HIF1a deg- Z. Li, W. Zhou, and Y. Zhang are co-first authors. radation by an ubiquitin-mediated pathway. Hydroxylation of Corresponding Authors: Wenge Zhu, George Washington University, Washing- HIF1a by PHDs creates a binding site for the von Hippel-Lindau ton, DC 20052. Phone: 202-994-3125; Fax: 2029943125; E-mail: (VHL) E3 ligase, which promotes subsequent ubiquitylation and [email protected]; David W. Chan, [email protected]; and Wei Zheng, [email protected] proteasomal degradation of HIF1a. Therefore, PHDs are key factors regulating HIF1a levels in cells; inhibition of PHDs' Clin Cancer Res 2019;XX:XX–XX activity limits HIF1a degradation (10). Nonetheless, the regula- doi: 10.1158/1078-0432.CCR-18-4145 tory pathway governing hydroxylation of HIF1a by PHDs Ó2019 American Association for Cancer Research. remains largely unknown. www.aacrjournals.org OF1 Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst July 8, 2019; DOI: 10.1158/1078-0432.CCR-18-4145 Li et al. and anti-phospho-PHD2 (Ser 125) antibody (MABC1612, Translational Relevance 1:1,000) were from Sigma-Aldrich. p-ERK (CST, 4370S, Identification of mechanisms regulating platinum resis- 1:1000), ERK (4695S, 1:1000), hydroxy-HIF1a (3434S, tance and new approaches for treatment of platinum- 1:1000), phospho-MAPK/CDK substrates (2325S, 1:1000), TGFb resistant ovarian cancer (PROC) will benefit patients with (3711S, 1:1000), and PHD2 (4835, 1:1000) were from Cell cancer. The amount of HIF1a plays an important role in Signaling Technology. Normal mouse IgG (sc-2025), mouse platinum resistance in ovarian cancer, but the mechanism anti-goat IgG-HRP (sc-2354), and mouse anti-rabbit IgG-HRP regulating HIF1a stability, in this context, remained largely (sc-2357) were from Santa Cruz Biotechnologies. Cis-diamine- unknown. This study demonstrates that PHD2 is a direct platinum(II) dichloride (Cisplatin; Sigma-Aldrich, 479306), substrate of ERK and phosphorylation of PHD2 by increased HIF1a inhibitor YC-1 (Selleck Chemicals, S7958), MEK inhibitor, ERK activity in PROC cells prevents PHD2 binding to HIF1a, selumetinib (Selleck Chemicals, S1008), and TGFb receptor resulting in the inhibition of HIFa hydroxylation and degra- inhibitor, SB431542 (Selleck Chemicals, S1067), were dissolved dation. Further studies indicate that this ERK/PHD2/HIF1a in DMSO and sterile 0.9% saline respectively for the in vitro and in pathway is potentiated in PROC cells or tumors by TGFb1. vivo assay. Lambda Protein Phosphatase (Lambda PP; P0753S) Inhibition of HIF1a by YC-1, ERK by selumetinib, or TGFb1by were purchased from New England Biolabs (NEB). SB431542 overcomes platinum resistance in vitro or in vivo. These findings reveal an unexpected TGFb1/ERK/PHD2/ qHTCS HIF1a pathway regulating platinum resistance in ovarian Compound screen experiments were performed in two rounds cancer. Additionally, this study points to three potential drugs as described previously (16). Cisplatin-resistant IGROV1 CR cells to treat patients with PROC. were used against 6,016 compounds from multiple compound libraries including NPC (NCGC Pharmaceutical Collection), MIPE (Mechanism Interrogation Plate), and LOPAC (The Library of Pharmacologically Active Compounds). Briefly, IGROV CR Extracellular regulated kinase (ERK) is a member of the MAPK cells (1,500 cells/well) were plated in 1,536-well polystyrene family and has been implicated in diverse cellular pathways plates (Greiner Bio-One) with 5-mL DMEM medium (10% FBS) including proliferation, survival, differentiation, and motili- and incubated for 16 hours. In the first screen, drugs were tested at ty (11). Although ERK was shown to control HIF1a transcrip- 11 different concentrations from 0.8 nmol/L to 46 mmol/L by tional activation (12, 13), it is unknown if ERK directly regulates serial dilution (1:3). After incubation for 72 hours, a total of 112 HIF1a protein stability and how ERK participates in platinum compounds that efficiently inhibited the proliferation of IGROV1 resistance in PROC. The TGFb, which can activate the ERK CR cells were identified. To further identify compounds that have pathway, belongs to the TGF superfamily comprising a large synergy with cisplatin, these 112 compounds were screened at number of cytokines secreted by many cell types (14). A growing 11 different concentrations in combination with vehicle, or body of evidence indicates that TGFb signaling is altered in 8.5 mmol/L cisplatin (IC50 of cisplatin). Then 4-mL/well ATP cancer. A dual role for TGFb in tumors, depending on tumor content cell viability assay reagent (Promega, G7570) was added stage, cellular context, and tumor microenvironment has long into each well and incubated for 30 minutes followed by detec- been noted. TGFb exerts tumor-suppressive effects; paradoxical- tion of cell viability. Through the second screen, compounds that ly, TGFb can also act as a tumor activator because of its activity exhibited the increased cytotoxicity of cisplatin in IGROV1 CR in suppressing immune and inflammatory responses (15). cells were identified (Supplementary Table S1). The primary We have established
Recommended publications
  • Independence of Hif1a and Androgen Signaling Pathways in Prostate Cancer
    bioRxiv preprint doi: https://doi.org/10.1101/848424; this version posted November 26, 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. Independence of HIF1a and androgen signaling pathways in prostate cancer Maxine GB Tran1, 2*, Becky AS Bibby3†*, Lingjian Yang3, Franklin Lo1, Anne Warren1, Deepa Shukla1, Michelle Osborne1, James Hadfield1, Thomas Carroll1, Rory Stark1, Helen Scott1, Antonio Ramos-Montoya1, Charlie Massie1, Patrick Maxwell1, Catharine ML West3, 4, Ian G. Mills5,6** and David E. Neal1** 1Uro-oncology Research Group, Cancer Research UK Cambridge Institute, Cambridge, CB02 0RE, United Kingdom 2UCL division of Surgery and Interventional Science, Royal Free Hospital, Pond Street, London NW3 2QG 3Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Christie Hospital NHS Trust, Manchester, M20 4BX, United Kingdom 4Manchester Biomedical Research Centre, University of Manchester, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom. 5Centre for Cancer Research and Cell Biology, Queens University Belfast, Belfast, BT9 7AE, United Kingdom 6Nuffield Department of Surgical Sciences, University of Oxford, OX3 9DU, UK *These authors contributed equally to this work **These authors contributed equally to this work †Corresponding author email: Becky Bibby, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Christie Hospital NHS Trust, Manchester, M20 4BX, United Kingdom, [email protected] 1 bioRxiv preprint doi: https://doi.org/10.1101/848424; this version posted November 26, 2019.
    [Show full text]
  • Harnessing Gene Expression Profiles for the Identification of Ex Vivo Drug
    cancers Article Harnessing Gene Expression Profiles for the Identification of Ex Vivo Drug Response Genes in Pediatric Acute Myeloid Leukemia David G.J. Cucchi 1 , Costa Bachas 1 , Marry M. van den Heuvel-Eibrink 2,3, Susan T.C.J.M. Arentsen-Peters 3, Zinia J. Kwidama 1, Gerrit J. Schuurhuis 1, Yehuda G. Assaraf 4, Valérie de Haas 3 , Gertjan J.L. Kaspers 3,5 and Jacqueline Cloos 1,* 1 Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; [email protected] (D.G.J.C.); [email protected] (C.B.); [email protected] (Z.J.K.); [email protected] (G.J.S.) 2 Department of Pediatric Oncology/Hematology, Erasmus MC–Sophia Children’s Hospital, 3015 CN Rotterdam, The Netherlands; [email protected] 3 Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; [email protected] (S.T.C.J.M.A.-P.); [email protected] (V.d.H.); [email protected] (G.J.L.K.) 4 The Fred Wyszkowski Cancer Research, Laboratory, Department of Biology, Technion-Israel Institute of Technology, 3200003 Haifa, Israel; [email protected] 5 Emma’s Children’s Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, 1081 HV Amsterdam, The Netherlands * Correspondence: [email protected] Received: 21 April 2020; Accepted: 12 May 2020; Published: 15 May 2020 Abstract: Novel treatment strategies are of paramount importance to improve clinical outcomes in pediatric AML. Since chemotherapy is likely to remain the cornerstone of curative treatment of AML, insights in the molecular mechanisms that determine its cytotoxic effects could aid further treatment optimization.
    [Show full text]
  • Ten Commandments for a Good Scientist
    Unravelling the mechanism of differential biological responses induced by food-borne xeno- and phyto-estrogenic compounds Ana María Sotoca Covaleda Wageningen 2010 Thesis committee Thesis supervisors Prof. dr. ir. Ivonne M.C.M. Rietjens Professor of Toxicology Wageningen University Prof. dr. Albertinka J. Murk Personal chair at the sub-department of Toxicology Wageningen University Thesis co-supervisor Dr. ir. Jacques J.M. Vervoort Associate professor at the Laboratory of Biochemistry Wageningen University Other members Prof. dr. Michael R. Muller, Wageningen University Prof. dr. ir. Huub F.J. Savelkoul, Wageningen University Prof. dr. Everardus J. van Zoelen, Radboud University Nijmegen Dr. ir. Toine F.H. Bovee, RIKILT, Wageningen This research was conducted under the auspices of the Graduate School VLAG Unravelling the mechanism of differential biological responses induced by food-borne xeno- and phyto-estrogenic compounds Ana María Sotoca Covaleda Thesis submitted in fulfillment of the requirements for the degree of doctor at Wageningen University by the authority of the Rector Magnificus Prof. dr. M.J. Kropff, in the presence of the Thesis Committee appointed by the Academic Board to be defended in public on Tuesday 14 September 2010 at 4 p.m. in the Aula Unravelling the mechanism of differential biological responses induced by food-borne xeno- and phyto-estrogenic compounds. Ana María Sotoca Covaleda Thesis Wageningen University, Wageningen, The Netherlands, 2010, With references, and with summary in Dutch. ISBN: 978-90-8585-707-5 “Caminante no hay camino, se hace camino al andar. Al andar se hace camino, y al volver la vista atrás se ve la senda que nunca se ha de volver a pisar” - Antonio Machado – A mi madre.
    [Show full text]
  • Human/Mouse/Rat HIF-1 Alpha/HIF1A Antibody
    Human/Mouse/Rat HIF-1 alpha/HIF1A Antibody Monoclonal Mouse IgG1 Clone # 241809 Catalog Number: MAB1536 DESCRIPTION Species Reactivity Human/Mouse/Rat Specificity Detects human, mouse, and rat HIF-1 alpha/HIF1A. Source Monoclonal Mouse IgG1 Clone # 241809 Purification Protein A or G purified from hybridoma culture supernatant Immunogen E. coli-derived recombinant human HIF-1 alpha/HIF1A Arg575-Asn826 Accession # Q16665.1 Formulation Lyophilized from a 0.2 μm filtered solution in PBS with Trehalose. See Certificate of Analysis for details. *Small pack size (-SP) is supplied either lyophilized or as a 0.2 μm filtered solution in PBS. APPLICATIONS Please Note: Optimal dilutions should be determined by each laboratory for each application. General Protocols are available in the Technical Information section on our website. Recommended Sample Concentration Western Blot 1 µg/mL See Below Immunohistochemistry 5-25 µg/mL See Below Immunoprecipitation 1-3 µg/500 µg cell MCF‑7 human breast cancer cell line treated with CoCl2, see our available Western blot lysate detection antibodies Simple Western 10 µg/mL See Below Knockout Validated HIF-1 alpha/HIF1A is specifically detected in HeLa human cervical epithelial carcinoma parental cell line but is not detectable in HIF-1 alpha/HIF1A knockout HeLa cell line. DATA Western Blot Immunohistochemistry Detection of Human, Mouse, HIF-1 alpha/HIF1A in Human Kidney. and Rat HIF-1 alpha/HIF1A by HIF-1 alpha/HIF1A was detected in Western Blot. Western blot immersion fixed paraffin-embedded sections shows lysates of MCF-7 human of human kidney using Mouse Anti- breast cancer cell line, Balb-3T3 Human/Mouse/Rat HIF-1 alpha/HIF1A mouse embryonic fibroblast cell Monoclonal Antibody (Catalog # MAB1536) line, and PC-12 rat adrenal at 5 µg/mL for 1 hour at room temperature pheochromocytoma cell line followed by incubation with the Anti-Mouse untreated (-) or treated (+) with IgG VisUCyte™ HRP Polymer Antibody 150 μM CoCl2 for 8 hours.
    [Show full text]
  • On the Biological Properties of Prostate Cancer Cells by Modulation of Inflammatory and Steroidogenesis Pathway Genes
    International Journal of Molecular Sciences Article The Impact of Ang-(1-9) and Ang-(3-7) on the Biological Properties of Prostate Cancer Cells by Modulation of Inflammatory and Steroidogenesis Pathway Genes Kamila Domi ´nska 1,* , Karolina Kowalska 2 , Kinga Anna Urbanek 2 , Dominika Ewa Habrowska-Górczy ´nska 2 , Tomasz Och˛edalski 1 and Agnieszka Wanda Piastowska Ciesielska 2 1 Department of Comparative Endocrinology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland; [email protected] 2 Department of Cell Cultures and Genomic Analysis, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland; [email protected] (K.K.); [email protected] (K.A.U.); [email protected] (D.E.H.-G.); [email protected] (A.W.P.C.) * Correspondence: [email protected] Received: 12 July 2020; Accepted: 26 August 2020; Published: 28 August 2020 Abstract: The local renin–angiotensin system (RAS) plays an important role in the pathophysiology of the prostate, including cancer development and progression. The Ang-(1-9) and Ang-(3-7) are the less known active peptides of RAS. This study examines the influence of these two peptide hormones on the metabolic activity, proliferation and migration of prostate cancer cells. Significant changes in MTT dye reduction were observed depending on the type of angiotensin and its concentration as well as time of incubation. Ang-(1-9) did not regulate the 2D cell division of either prostate cancer lines however, it reduced the size of LNCaP colonies formed in soft agar, maybe through down-regulation of the HIF1a gene.
    [Show full text]
  • Impact of Hypoxia on Hepatitis B Virus Replication
    i IMPACT OF HYPOXIA ON HEPATITIS B VIRUS REPLICATION NICHOLAS ROSS BAKER FRAMPTON A thesis submitted to the University of Birmingham for the degree of Doctor of Philosophy Institute of Immunology and Immunotherapy College of Medical and Dental Sciences University of Birmingham September 2017 ii University of Birmingham Research Archive e-theses repository This unpublished thesis/dissertation is copyright of the author and/or third parties. The intellectual property rights of the author or third parties in respect of this work are as defined by The Copyright Designs and Patents Act 1988 or as modified by any successor legislation. Any use made of information contained in this thesis/dissertation must be in accordance with that legislation and must be properly acknowledged. Further distribution or reproduction in any format is prohibited without the permission of the copyright holder. ABSTRACT Hepatitis B virus (HBV) is one of the world’s unconquered diseases, with 370 million chronically infected globally. HBV replicates in hepatocytes within the liver that exist under a range of oxygen tensions from 11% in the peri-portal area to 3% in the peri- central lobules. HBV transgenic mice show a zonal pattern of viral antigen with expression in the peri-central areas supporting a hypothesis that low oxygen regulates HBV replication. We investigated this hypothesis using a recently developed in vitro model system that supports HBV replication. We demonstrated that low oxygen significantly increases covalently closed circular viral DNA (cccDNA), viral promoter activity and pre-genomic RNA (pgRNA) levels, consistent with low oxygen boosting viral transcription. Hypoxia inducible factors (HIFs) regulate cellular responses to low oxygen and we investigated a role for HIF-1α or HIF-2α on viral transcription.
    [Show full text]
  • HIF-1Α: a Potential Treatment Target in Chronic Lymphocytic Leukemia
    Editorials HIF-1α: a potential treatment target in chronic lymphocytic leukemia Martina Seiffert Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany E-mail: MARTINA SEIFFERT - [email protected] doi:10.3324/haematol.2019.246330 n 2019, William Kaelin Jr., Peter J. Ratcliffe and Gregg in CLL change the binding of protein substrates including L. Semenza were jointly awarded the Nobel Prize in HIF-1α and are therefore involved in the increased stabili- IPhysiology or Medicine for their work in elucidating ty of HIF-1α in patients with these mutations. how the transcription factor Hypoxia-inducible factor 1 HIF-1α has been suggested to be transcriptionally regu- (HIF-1) senses oxygen availability and adapts cellular lated in CLL cells via signals delivered by the microenvi- response accordingly.1 HIF-1 is a heterodimeric protein ronment. In oxygenated blood, circulating CLL cells were complex that consists of two proteins: a constitutively shown to be primed for hypoxia and to undergo a rapid expressed HIF-1β subunit and an inducible HIF-1α sub- induction of HIF-1α activity when entering lymphoid tis- 6 unit. During hypoxia, HIF-1α hydroxylation is reduced, sues. In these tissues, CLL cells are in constant interaction preventing its proteasomal degradation, and the stabi- with accessory cells which deliver essential signals for sur- lized HIF-1 complex comprised of HIF1α and HIF-1β is vival and proliferation of CLL cells and contribute to transported to the nucleus where it regulates the expres- resistance to drug-induced apoptosis.7 The contact of CLL sion of several hundred genes to counter the lack of oxy- cells with stromal cells induces HIF-1α expression via gen.
    [Show full text]
  • (Lncrna) in the Regulation of Hypoxia-Inducible Factor (HIF) in Cancer
    non-coding RNA Review Long-Noncoding RNA (lncRNA) in the Regulation of Hypoxia-Inducible Factor (HIF) in Cancer Dominik A. Barth 1,2 , Felix Prinz 1, Julia Teppan 1, Katharina Jonas 1 , Christiane Klec 1 and Martin Pichler 1,2,* 1 Research Unit of Non-Coding RNAs and Genome Editing in Cancer, Division of Clinical Oncology, Department of Internal Medicine, Comprehensive Cancer Center Graz; Medical University of Graz, 8036 Graz, Austria; [email protected] (D.A.B.); [email protected] (F.P.); [email protected] (J.T.); [email protected] (K.J.); [email protected] (C.K.) 2 Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA * Correspondence: [email protected] Received: 15 May 2020; Accepted: 3 July 2020; Published: 6 July 2020 Abstract: Hypoxia is dangerous for oxygen-dependent cells, therefore, physiological adaption to cellular hypoxic conditions is essential. The transcription factor hypoxia-inducible factor (HIF) is the main regulator of hypoxic metabolic adaption reducing oxygen consumption and is regulated by gradual von Hippel-Lindau (VHL)-dependent proteasomal degradation. Beyond physiology, hypoxia is frequently encountered within solid tumors and first drugs are in clinical trials to tackle this pathway in cancer. Besides hypoxia, cancer cells may promote HIF expression under normoxic conditions by altering various upstream regulators, cumulating in HIF upregulation and enhanced glycolysis and angiogenesis, altogether promoting tumor proliferation and progression. Therefore, understanding the underlying molecular mechanisms is crucial to discover potential future therapeutic targets to evolve cancer therapy. Long non-coding RNAs (lncRNA) are a class of non-protein coding RNA molecules with a length of over 200 nucleotides.
    [Show full text]
  • Tap63g (P51a) and Dnp63a (P73l), Two Major Isoforms of the P63 Gene, Exert Opposite E€Ects on the Vascular Endothelial Growth Factor (VEGF) Gene Expression
    Oncogene (2002) 21, 2455 ± 2465 ã 2002 Nature Publishing Group All rights reserved 0950 ± 9232/02 $25.00 www.nature.com/onc TAp63g (p51A) and dNp63a (p73L), two major isoforms of the p63 gene, exert opposite eects on the vascular endothelial growth factor (VEGF) gene expression Makoto Senoo1, Yasuko Matsumura1 and Sonoko Habu*,1 1Department of Immunology, Tokai University School of Medicine, Bouseidai, Isehara-city, Kanagawa 259-1193, Japan Tumor suppressor p53 has been shown to repress 1995). The VEGF mRNA is expressed predominantly expression of vascular endothelial growth factor in tumor cells and stromal cells, whereas the mRNA (VEGF), an endothelial cell-speci®c mitogen and a key for the VEGF receptors is expressed and upregulated mediator of tumor angiogenesis. The p63 gene, recently in tumor endothelial cells (Suzuki et al., 1996). This identi®ed as a p53-relative, encodes multiple isoforms expression pro®le hypothesizes the VEGF to be a with structural and functional similarities and dierences primary paracrine regulator of tumor angiogenesis. from p53. In this study, we show the evidence that the Several groups have shown that VEGF expression is two major isoforms of the p63 gene, TAp63g (p51A) and induced by exposing cultured cells to 1% O2, CoCl2 or dNp63a (p73L), represses and upregulates VEGF desferrioxamine, and induction can be blocked by expression, respectively, on transcription and protein treating hypoxic cells with cycloheximide (Gleadle et levels. Transient transfection assays show that a al., 1995; Goldberg and Schneider, 1994; Shweiki et al., hypoxia-inducible factor (HIF) 1 binding site within the 1992). Recent studies have demonstrated that hypoxia VEGF promoter region is responsible for both upregula- increases VEGF transcription in several dierent cell tion and repression by dNp63a and by TAp63g, types (Finkenzeller et al., 1995; Ikeda et al., 1995; Levy respectively, of the VEGF promoter activity.
    [Show full text]
  • VHL Inactivation Without Hypoxia Is Sufficient to Achieve Genome Hypermethylation
    bioRxiv preprint doi: https://doi.org/10.1101/093310; this version posted December 12, 2016. 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. VHL inactivation without hypoxia is sufficient to achieve genome hypermethylation Artem V. Artemov1*, Nadezhda Zhigalova1, Svetlana Zhenilo1, Alexander M. Mazur1 and Egor B. Prokhortchouk1 1 Institute of Bioengineering, Research Center of Biotechnology RAS, Moscow, Russian Federation * [email protected] Abstract VHL inactivation is a key oncogenic event for renal carcinomas. In normoxia, VHL suppresses HIF1a-mediated response to hypoxia. It has previously been shown that hypoxic conditions inhibit TET-dependent hydroxymethylation of cytosines and cause DNA hypermethylation at gene promoters. In this work, we performed VHL inactivation by CRISPR/Cas9 and studied its effects on gene expression and DNA methylation. We showed that even without hypoxia, VHL inactivation leads to hypermethylation of the genome which mainly occurred in AP-1 and TRIM28 binding sites. We also observed promoter hypermethylation of several transcription regulators associated with decreased gene expression. Keywords DNA methylation; VHL; hypoxia; HIF1a; JUN; FOS Introduction Sequencing of cancer genomes was initially aimed to find cancer drivers, or genes, that, once mutated, give a selective advantage to a cancer cell, such as increased proliferation, suppression of apoptosis or the ability to avoid immune response. VHL is a key oncosuppressor gene for kidney cancer. Inactivation of the VHL gene is the most common event in renal carcinomas, accounting for 50{70% of sporadic cases (Scelo et al. 2014; Cancer Genome Atlas Research Network 2013; Thomas et al.
    [Show full text]
  • The LIMD1 Protein Represses the Hypoxic Response Through Bridging an Association Between the Phds and VHL
    The LIMD1 Protein represses the Hypoxic Response through Bridging an Association Between the PHDs and VHL Dr Daniel Foxler Centre for Molecular Oncology www.bci.qmul.ac.uk LIMD1 is a 3p21.3 encoded tumour suppressor gene Normal epithelia 3p21.3 loss LIMD1 Squamous metaplasia x200 Hyperplasia p53 mutations Dysplasia Resp epithelium & adjacent HG dysplasia x400 Kras mutations Micro-invasion Invasive SCC in subepithelial stroma x200 LIMD1 knockout mice are LIMD1 loss correlates with predisposed to develop poor prognosis in breast lung cancer cancer Sharp et al 2008 Spendlove et al 2008 LIMD1 is a member of the Zyxin family and represses cell cycle progression miRNA silencing (RNA storage/degradation) AGO E2F PHD1 HIF1 is the master transcriptional regulator of the hypoxic response • Hypoxia inducible factor 1 (HIF1) is the master transcriptional regulator that responds to decreases in intracellular O2 tension Enhanced Gene Transcription •VEGF •VEGF receptor 1 •PDGF Angiogenesis •Ang-2 •Epo •Transferrin Red blood cell production •Transferrin receptor HIF1α HIF1β HIF1 •Lactate dehydrogenase A •Pyruvate kinase 1 Protein •Aldolase A/C Energy metabolism •Phosphoglycerate kinase •Hexokinase •Glucose transporter 1 & 3 •BNIP3 •Insulin-like growth factor 2 •Adenylate kinase 3 Cellular proliferation •WT1 •Nip3 The PHDs and VHL bind to discrete regions within LIMD1 WT LIMD1 LIM1 LIM2 LIM3 676 aa LIMD1 Δ472-676 PHD2 LIMD1 Δ1-467 LIM1 LIM2VHL LIM3 LIM domains PHD2 Pre-LIM region VHL region kDa kDa 75 75 INPUT 50 INPUT 50 BLOT: Xpress BLOT: Xpress 37 37
    [Show full text]
  • HIF-1Α Is Required for Development of the Sympathetic Nervous System
    HIF-1α is required for development of the sympathetic nervous system Romana Bohuslavovaa,1, Radka Cerychovaa, Frantisek Papousekb, Veronika Olejnickovab,c, Martin Bartosc,d, Agnes Görlache,f, Frantisek Kolarb, David Sedmerab,c, Gregg L. Semenzag,h,1, and Gabriela Pavlinkovaa,1 aLaboratory of Molecular Pathogenetics, Institute of Biotechnology, Czech Academy of Sciences, 252 50 Vestec, Czechia; bInstitute of Physiology, Czech Academy of Sciences, 142 00 Prague, Czechia; cInstitute of Anatomy, First Faculty of Medicine, Charles University, 110 00 Prague, Czechia; dInstitute of Dental Medicine, First Faculty of Medicine, Charles University, 110 00 Prague, Czechia; eGerman Heart Centre Munich, Technical University, 80636 Munich, Germany; fGerman Centre for Cardiovascular Research, Partner Site Munich, Munich Heart Alliance, 80636 Munich, Germany; gMcKusick–Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205; and hInstitute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 Contributed by Gregg L. Semenza, May 1, 2019 (sent for review February 28, 2019; reviewed by Gabriel G. Haddad and Michiko Watanabe) The molecular mechanisms regulating sympathetic innervation of The differentiation of sympathetic neurons is induced by sig- the heart during embryogenesis and its importance for cardiac naling molecules of the bone morphogenetic protein family se- development and function remain to be fully elucidated. We creted by the dorsal aorta, followed by expression of a variety of generated mice in which conditional knockout (CKO) of the Hif1a transcription factors that initiate noradrenergic and neuronal gene encoding the transcription factor hypoxia-inducible factor 1α differentiation, including PHOX2A, PHOX2B, GATA2, GATA3, (HIF-1α) is mediated by an Islet1-Cre transgene expressed in the INSM1, HAND2, ASCL1, SOX4, and SOX11 (5, 8).
    [Show full text]