De-Repression of Pdgfrb Transcription Promotes Acquired

Total Page:16

File Type:pdf, Size:1020Kb

De-Repression of Pdgfrb Transcription Promotes Acquired Published OnlineFirst March 26, 2013; DOI: 10.1158/2159-8290.CD-12-0502 RESEARCH ARTICLE De-Repression of PDGFR b Transcription Promotes Acquired Resistance to EGFR Tyrosine Kinase Inhibitors in Glioblastoma Patients David Akhavan1 , 2 , Alexandra L. Pourzia 3 , Alex A. Nourian 3 , Kevin J. Williams3 , 7 , David Nathanson 2 , Ivan Babic 9 , Genaro R. Villa 1 , 2 , 9 , Kazuhiro Tanaka 2 , Ali Nael 2 , Huijun Yang 9 , Julie Dang 2 , Harry V. Vinters 3 , William H. Yong 3 , Mitchell Flagg3 , Fuyuhiko Tamanoi 5 , Takashi Sasayama 12 , C. David James 8 , Harley I. Kornblum 4 , Tim F. Cloughesy 6 , Webster K. Cavenee 9 , 10 , Steven J. Bensinger 2 , 3 , 7 , and Paul S. Mischel 9 , 10 , 11 Downloaded from cancerdiscovery.aacrjournals.org on September 23, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst March 26, 2013; DOI: 10.1158/2159-8290.CD-12-0502 ABSTRACT Acquired resistance to tyrosine kinase inhibitors (TKI) represents a major chal- lenge for personalized cancer therapy. Multiple genetic mechanisms of acquired TKI resistance have been identifi ed in several types of human cancer. However, the possibility that cancer cells may also evade treatment by co-opting physiologically regulated receptors has not been addressed. Here, we show the fi rst example of this alternate mechanism in brain tumors by show- ing that EGF receptor (EGFR)-mutant glioblastomas (GBMs) evade EGFR TKIs by transcriptionally de-repressing platelet-derived growth factor receptor β (PDGFRβ). Mechanistic studies show that EGFRvIII signaling actively suppresses PDGFR b transcription in an mTORC1- and extracellular signal– regulated kinase-dependent manner. Genetic or pharmacologic inhibition of oncogenic EGFR renders GBMs dependent on the consequently de-repressed PDGFRβ signaling for growth and survival. Impor- tantly, combined inhibition of EGFR and PDGFRβ signaling potently suppresses tumor growth in vivo . These data identify a novel, nongenetic TKI resistance mechanism in brain tumors and provide compel- ling rationale for combination therapy. SIGNIFICANCE: These results provide the fi rst clinical and biologic evidence for receptor tyrosine kinase (RTK) “switching” as a mechanism of resistance to EGFR inhibitors in GBM and provide a molecu- lar explanation of how tumors can become “addicted” to a nonamplifi ed, nonmutated, physiologically regulated RTK to evade targeted treatment. Cancer Discov; 3(5); 1–14. ©2013 AACR. INTRODUCTION PTEN ( 2 ), PIK3CA ( 5 ), or KRAS ( 6 ); and (iii) co-occurrence of other amplifi ed or mutated receptor tyrosine kinases The EGF receptor, EGFR, is commonly amplifi ed and/ (RTK), including C-MET and platelet-derived growth factor or mutated in many types of solid cancer, including a receptor α (PDGFRα; refs. 7, 8 ). In addition to these genetic variety of epithelial cancers and glioblastoma (GBM) ( 1–3 ). resistance-promoting mutations, it is suspected that EGFR- Despite compelling evidence for EGFR addiction in experi- dependent cancers may escape targeted therapy by devel- mental models, the clinical benefi t of most EGFR tyrosine oping dependence on other nonamplifi ed, nonmutated kinase inhibitors (TKI) has been quite limited. Multiple RTKs ( 9 ). However, the mechanisms by which cancers, genetic resistance mechanisms enable cancer cells to main- including GBM, evade EGFR TKI treatment by coopt- tain signal fl ux to critical downstream effector pathways, ing other physiologically regulated receptors has not been and thus evade EGFR-targeted therapy, including: (i) addressed. acquisition and/or selection for secondary EGFR muta- Herein, we integrate studies in cell lines, patient-derived tions conferring EGFR TKI resistance ( 4 ); (ii) additional tumor cultures, xenotransplants, and tumor tissue from activating mutations in downstream effectors, such as patients with GBM in a phase II clinical trial to provide the fi rst demonstration of EGFR TKI resistance mediated by transcrip- Authors’ Affi liations: 1 Medical Scientist Training Program, David Geffen tional de-repression of PDGFRb. We show that the persist- School of Medicine; Departments of 2 Molecular and Medical Pharmacol- ently active EGFR mutation, EGFRvIII, suppresses PDGFRβ ogy, 3 Pathology and Laboratory Medicine, 4 Psychiatry and Biobehavioral expression via mTORC1- and extracellular signal–regulated Sciences, and 5 Microbiology, Immunology & Molecular Genetics; 6 Neuro- 7 kinase (ERK)-dependent mechanisms. We show in cells, mice, Oncology Program; Institute for Molecular Medicine, University of Cali- β, fornia Los Angeles, Los Angeles; 8 Brain Tumor Research Center, University and patients that EGFR TKI treatment de-represses PDGFR of California San Francisco, San Francisco; 9 Ludwig Institute for Cancer rendering GBMs dependent on PDGFRβ signaling for growth. Research; 10 Moores Cancer Center; 11 Department of Pathology, Univer- We further show that combined abrogation of EGFRvIII and sity of California San Diego, La Jolla, California; and 12 Department of PDGFRβ potently prevents GBM growth in vivo . These results Neurosurgery, Kobe University Graduate School of Medicine, Kobe, Hyogo, identify a novel physiologic EGFR TKI resistance mechanism Japan in GBM and suggest a clinically actionable approach to sup- Note: Supplementary data for this article are available at Cancer Discovery Online (http://cancerdiscovery.aacrjournals.org/). press it. Corresponding Authors: Paul S. Mischel, Ludwig Institute for Can- cer Research, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0753. Phone: 858-534-6080; Fax: 858-534-7750; RESULTS E-mail: [email protected] ; and Steven J. Bensinger, Institute for Molecular EGFR Inhibition Promotes PDGFRb Medicine Department of Pathology and Laboratory Medicine, 36-128 Upregulation in Glioma Center for Health Sciences, University of California Los Angeles, Los Angeles, CA 90095. Phone: 310-825-9885; Fax: 310-267-6267; To better understand how malignant glioma acquires resist- E-mail: [email protected] ance to EGFR inhibitors in vivo , U87 glioma cells expressing doi: 10.1158/2159-8290.CD-12-0502 the EGFRvIII gain-of-function mutation (designated U87- © 2013 American Association for Cancer Research. EGFRvIII herein) were placed in the fl anks of severe combined MAY 2013CANCER DISCOVERY | OF2 Downloaded from cancerdiscovery.aacrjournals.org on September 23, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst March 26, 2013; DOI: 10.1158/2159-8290.CD-12-0502 RESEARCH ARTICLE Akhavan et al. ABDays post-injection ) 3 600 Drug Drug Drug 500 20181614121086420 400 EGFRvIII 300 Tumor injection MeasureMeasure Measure 200 EGFRvIII 100 Erlotinib 150 mg/kg/d Tumor volume (mm 0 Day 10 Day 13 Day 17 C D U87vIII pEGFR U87 U87vIIIU87vIIIU87vIII Erlot. K.D. PDGFRβ pPDGFRβ Y751 U87vIII erlotinib pEGFR Y1086 150 mg/kg, 18 d β EGFR pPDGFR PI3K-p85 pEGFR pAXL E F GBM39 GBM6 HK296 pEGFR Y1086 PDGFRβ Erlotinib (5 μmol/L) –+ –+ –+ PDGFRα EGFRvIII Vehicle PDGFRβ pEGFR Y1068 PI3K-p85 pEGFR Y1086 PDGFRβ GBM12 HK242 Erlotinib (5 μmol/L) – + –+ Wild-type EGFR PDGFRα Erlotinib PDGFRβ pEGFR Y1068 PI3K-p85 G GBM39 Vehicle Erlotinib Tumor 1 Tumor 2 Tumor 1 Tumor 2 pPDGFRb Y751 PDGFRβ pEGFR Y1086 PI3K-p85 Figure 1. A reciprocal relationship between EGFR and PDGFRβ in glioma. A, experimental design of a mouse model of EGFR inhibitor resistance. U87- EGFRvIII cells were subcutaneously implanted in the mouse fl ank on day 0. Mice were treated with erlotinib (150 mg/kg) on day 2 and as indicated there- after. B, tumor growth curve of U87-EGFRvIII xenografts in mice treated with erlotinib (150 mg/kg as indicated in A) or vehicle. C, immunoblot of indicated tumor lysates determining total and phospho-PDGFRβ or EGFR from U87, U87-EGFRvIII ± eroltinib treatment and U87-EGFRvIII kinase dead xenografts harvested on day 21. PI3K-p85 is used as a loading control in this and subsequent immunoblots. D, RTK array of 42 RTKs conducted on U87-EGFRvIII xenograft lysates on day 21 from mice treated with erlotinib or vehicle as described in A. E, immunohistochemistry (IHC) of PDGFRβ and phospho-EGFR in vehicle- and erlotinib-treated U87-EGFRvIII orthotopic xenografts. F, immunoblot of PDGFRα, PDGFRβ, and phospho-EGFR (pEGFR) from patient-derived GBM neurospheres expressing EGFRvIII or wild-type EGFR as indicated. Whole-cell lysates were collected after 24 hours of erlotinib or vehicle treatment. G, immunoblot of tumor lysates from EGFRvIII expressing GBM39 xenografts following oral gavage with vehicle or erlotinib for 10 days. immunodefi cient (SCID) mice. EGFRvIII, the most common vehicle control, and tumor growth was assessed over 18 days EGFR mutation in GBM, arises from an in-frame genomic ( Fig. 1A ). As expected, erlotinib treatment slowed U87- deletion of exons 2 to 7, resulting in a persistently active, EGFRvIII tumor growth relative to control; however, tumors highly oncogenic protein ( 10 ). Tumor-bearing mice were retained a substantial growth rate despite continued erlotinib gavaged with the EGFR inhibitor erlotinib (150 mg/kg) or treatment ( Fig. 1B ). Immunoblots of tumor lysates confi rmed OF3 | CANCER DISCOVERYMAY 2013 www.aacrjournals.org Downloaded from cancerdiscovery.aacrjournals.org on September 23, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst March 26, 2013; DOI: 10.1158/2159-8290.CD-12-0502 An RTK Switch in Malignant Glioma RESEARCH ARTICLE that erlotinib treatment of mice signifi cantly reduced EGFR a strong inverse correlation between EGFR (total and phos-
Recommended publications
  • Tyrosine Kinase – Role and Significance in Cancer
    Int. J. Med. Sci. 2004 1(2): 101-115 101 International Journal of Medical Sciences ISSN 1449-1907 www.medsci.org 2004 1(2):101-115 ©2004 Ivyspring International Publisher. All rights reserved Review Tyrosine kinase – Role and significance in Cancer Received: 2004.3.30 Accepted: 2004.5.15 Manash K. Paul and Anup K. Mukhopadhyay Published:2004.6.01 Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S Nagar, Mohali, Punjab, India-160062 Abstract Tyrosine kinases are important mediators of the signaling cascade, determining key roles in diverse biological processes like growth, differentiation, metabolism and apoptosis in response to external and internal stimuli. Recent advances have implicated the role of tyrosine kinases in the pathophysiology of cancer. Though their activity is tightly regulated in normal cells, they may acquire transforming functions due to mutation(s), overexpression and autocrine paracrine stimulation, leading to malignancy. Constitutive oncogenic activation in cancer cells can be blocked by selective tyrosine kinase inhibitors and thus considered as a promising approach for innovative genome based therapeutics. The modes of oncogenic activation and the different approaches for tyrosine kinase inhibition, like small molecule inhibitors, monoclonal antibodies, heat shock proteins, immunoconjugates, antisense and peptide drugs are reviewed in light of the important molecules. As angiogenesis is a major event in cancer growth and proliferation, tyrosine kinase inhibitors as a target for anti-angiogenesis can be aptly applied as a new mode of cancer therapy. The review concludes with a discussion on the application of modern techniques and knowledge of the kinome as means to gear up the tyrosine kinase drug discovery process.
    [Show full text]
  • Platelet-Derived Growth Factor Receptor Expression and Amplification in Choroid Plexus Carcinomas
    Modern Pathology (2008) 21, 265–270 & 2008 USCAP, Inc All rights reserved 0893-3952/08 $30.00 www.modernpathology.org Platelet-derived growth factor receptor expression and amplification in choroid plexus carcinomas Nina N Nupponen1,*, Janna Paulsson2,*, Astrid Jeibmann3, Brigitte Wrede4, Minna Tanner5, Johannes EA Wolff 6, Werner Paulus3, Arne O¨ stman2 and Martin Hasselblatt3 1Molecular Cancer Biology Program, University of Helsinki, Helsinki, Finland; 2Department of Oncology–Pathology, Cancer Centrum Karolinska, Karolinska Institutet, Stockholm, Sweden; 3Institute of Neuropathology, University Hospital Mu¨nster, Mu¨nster, Germany; 4Department of Pediatric Oncology, University of Regensburg, Regensburg, Germany; 5Department of Oncology, Tampere University Hospital, Tampere, Finland and 6Children’s Cancer Hospital, MD Anderson Cancer Center, Houston, TX, USA Platelet-derived growth factor (PDGF) receptor signaling has been implicated in the development of glial tumors, but not yet been examined in choroid plexus carcinomas, pediatric tumors with dismal prognosis for which novel treatment options would be desirable. Therefore, protein expression of PDGF receptors a and b as well as amplification status of the respective genes, PDGFRA and PDGFRB, were examined in a series of 22 patients harboring choroid plexus carcinoma using immunohistochemistry and chromogenic in situ hybridization (CISH). The majority of choroid plexus carcinomas expressed PDGF receptors with 6 cases (27%) displaying high staining scores for PDGF receptor a and 13 cases (59%) showing high staining scores for PDGF receptor b. Correspondingly, copy-number gains of PDGFRA were observed in 8 cases out of 12 cases available for CISH and 1 case displayed amplification (six or more signals per nucleus). The proportion of choroid plexus carcinomas with amplification of PDGFRB was even higher (5/12 cases).
    [Show full text]
  • Ephb3 Suppresses Non-Small-Cell Lung Cancer Metastasis Via a PP2A/RACK1/Akt Signalling Complex
    ARTICLE Received 7 Nov 2011 | Accepted 11 Jan 2012 | Published 7 Feb 2012 DOI: 10.1038/ncomms1675 EphB3 suppresses non-small-cell lung cancer metastasis via a PP2A/RACK1/Akt signalling complex Guo Li1, Xiao-Dan Ji1, Hong Gao1, Jiang-Sha Zhao1, Jun-Feng Xu1, Zhi-Jian Sun1, Yue-Zhen Deng1, Shuo Shi1, Yu-Xiong Feng1, Yin-Qiu Zhu1, Tao Wang2, Jing-Jing Li1 & Dong Xie1 Eph receptors are implicated in regulating the malignant progression of cancer. Here we find that despite overexpression of EphB3 in human non-small-cell lung cancer, as reported previously, the expression of its cognate ligands, either ephrin-B1 or ephrin-B2, is significantly downregulated, leading to reduced tyrosine phosphorylation of EphB3. Forced activation of EphB3 kinase in EphB3-overexpressing non-small-cell lung cancer cells inhibits cell migratory capability in vitro as well as metastatic seeding in vivo. Furthermore, we identify a novel EphB3-binding protein, the receptor for activated C-kinase 1, which mediates the assembly of a ternary signal complex comprising protein phosphatase 2A, Akt and itself in response to EphB3 activation, leading to reduced Akt phosphorylation and subsequent inhibition of cell migration. Our study reveals a novel tumour-suppressive signalling pathway associated with kinase-activated EphB3 in non-small-cell lung cancer, and provides a potential therapeutic strategy by activating EphB3 signalling, thus inhibiting tumour metastasis. 1 Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Graduate School of Chinese Academy of Sciences, Shanghai 200031, China. 2 The Eastern Hepatobiliary Surgery Hospital, the Second Military Medical University, Shanghai 200433, China.
    [Show full text]
  • Original Article ERBB3, IGF1R, and TGFBR2 Expression Correlate With
    Am J Cancer Res 2018;8(5):792-809 www.ajcr.us /ISSN:2156-6976/ajcr0077452 Original Article ERBB3, IGF1R, and TGFBR2 expression correlate with PDGFR expression in glioblastoma and participate in PDGFR inhibitor resistance of glioblastoma cells Kang Song1,2*, Ye Yuan1,2*, Yong Lin1,2, Yan-Xia Wang1,2, Jie Zhou1,2, Qu-Jing Gai1,2, Lin Zhang1,2, Min Mao1,2, Xiao-Xue Yao1,2, Yan Qin1,2, Hui-Min Lu1,2, Xiang Zhang1,2, You-Hong Cui1,2, Xiu-Wu Bian1,2, Xia Zhang1,2, Yan Wang1,2 1Department of Pathology, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China; 2Key Laboratory of Tumor Immunology and Pathology of Ministry of Education, Chongqing 400038, China. *Equal contributors. Received April 6, 2018; Accepted April 9, 2018; Epub May 1, 2018; Published May 15, 2018 Abstract: Glioma, the most prevalent malignancy in brain, is classified into four grades (I, II, III, and IV), and grade IV glioma is also known as glioblastoma multiforme (GBM). Aberrant activation of receptor tyrosine kinases (RTKs), including platelet-derived growth factor receptor (PDGFR), are frequently observed in glioma. Accumulating evi- dence suggests that PDGFR plays critical roles during glioma development and progression and is a promising drug target for GBM therapy. However, PDGFR inhibitor (PDGFRi) has failed in clinical trials, at least partially, due to the activation of other RTKs, which compensates for PDGFR inhibition and renders tumor cells resistance to PDGFRi. Therefore, identifying the RTKs responsible for PDGFRi resistance might provide new therapeutic targets to syner- getically enhance the efficacy of PDGFRi.
    [Show full text]
  • Pdgfrβ Regulates Adipose Tissue Expansion and Glucose
    1008 Diabetes Volume 66, April 2017 Yasuhiro Onogi,1 Tsutomu Wada,1 Chie Kamiya,1 Kento Inata,1 Takatoshi Matsuzawa,1 Yuka Inaba,2,3 Kumi Kimura,2 Hiroshi Inoue,2,3 Seiji Yamamoto,4 Yoko Ishii,4 Daisuke Koya,5 Hiroshi Tsuneki,1 Masakiyo Sasahara,4 and Toshiyasu Sasaoka1 PDGFRb Regulates Adipose Tissue Expansion and Glucose Metabolism via Vascular Remodeling in Diet-Induced Obesity Diabetes 2017;66:1008–1021 | DOI: 10.2337/db16-0881 Platelet-derived growth factor (PDGF) is a key factor in The physiological roles of the vasculature in adipose tissue angiogenesis; however, its role in adult obesity remains have been attracting interest from the viewpoint of adipose unclear. In order to clarify its pathophysiological role, tissue expansion and chronic inflammation (1,2). White we investigated the significance of PDGF receptor b adipose tissue (WAT) such as visceral fat possesses the (PDGFRb) in adipose tissue expansion and glucose unique characteristic of plasticity; its volume may change metabolism. Mature vessels in the epididymal white several fold even after growth depending on nutritional adipose tissue (eWAT) were tightly wrapped with peri- conditions. Enlarged adipose tissue is chronically exposed cytes in normal mice. Pericyte desorption from vessels to hypoxia (3,4), which stimulates the production of angio- and the subsequent proliferation of endothelial cells genic factors for the supplementation of nutrients and were markedly increased in the eWAT of diet-induced oxygen to the newly enlarged tissue area (5). Selective ab- obese mice. Analyses with flow cytometry and adipose lation of the vasculature in WAT by apoptosis-inducible tissue cultures indicated that PDGF-B caused the de- peptides or the systemic administration of angiogenic in- PATHOPHYSIOLOGY tachment of pericytes from vessels in a concentration- hibitors has been shown to reduce WAT volumes and result dependent manner.
    [Show full text]
  • 60+ Genes Tested FDA-Approved Targeted Therapies & Gene Indicators
    ® 60+ genes tested ABL1, ABL2, ALK, AR, ARAF, ATM, ATR, BRAF, BRCA1*, BRCA2*, BTK, CCND1, CCND2, CCND3, CDK4, Somatic mutation detection CDK6, CDKN1A, CDKN1B, CDKN2A, CDKN2B, DDR1, DDR2, EGFR, ERBB2 (HER2), ESR1, FGFR1, FGFR2, for approved cancer therapies FGFR3, FGFR4, FLCN, FLT1, FLT3, FLT4, GNA11, GNAQ, HDAC1, HDAC2, HRAS, JAK1, JAK2, KDR, KIT, in solid tumors KRAS, MAP2K1, MET, MTOR, NF1, NF2, NRAS, PALB2, PARP1, PDGFRA, PDGFRB, PIK3CA, PIK3CD, PTCH1, PTEN, RAF1, RET, ROS1, SMO, SRC, STK11, TNK2, TSC1, TSC2 FDA-approved Targeted Therapies & Gene Indicators Abiraterone AR Necitumumab EGFR Ado-Trastuzumab ERBB2 (HER2) Nilotinib ABL1, ABL2, DDR1, DDR2, KIT, PDGFRA, PDGFRB Emtansine FGFR1, FGFR2, FGFR3, FLT1, FLT4, KDR, PDGFRA, Afatinib EGFR, ERBB2 (HER2) Nintedanib PDGFRB Alectinib ALK Olaparib ATM, ATR, BRCA1*, BRCA2*, PALB2, PARP1 Anastrozole ESR1 Osimertinib EGFR Axitinib FLT1, FLT4, KDR, KIT, PDGFRA, PDGFRB CDK4, CDK6, CCND1, CCND2, CCND3, CDKN1A, Palbociclib Belinostat HDAC1, HDAC2 CDKN1B, CDKN2A, CDKN2B Panitumumab EGFR Bicalutamide AR Panobinostat HDAC1, HDAC2 Bosutinib ABL1, SRC Pazopanib FLT1, FLT4, KDR, KIT, PDGFRA, PDGFRB Cabozantinib FLT1, FLT3, FLT4, KDR, KIT, MET, RET Pertuzumab ERBB2 (HER2) Ceritinib ALK ABL1, FGFR1, FGFR2, FGFR3, FGFR4, FLT1, FLT3, FLT4, Cetuximab EGFR Ponatinib KDR, KIT, PDGFRA, PDGFRB, RET, SRC Cobimetinib MAP2K1 Ramucirumab KDR Crizotinib ALK, MET, ROS1 Regorafenib ARAF, BRAF, FLT1, FLT4, KDR, KIT, PDGFRB, RAF1, RET Dabrafenib BRAF Ruxolitinib JAK1, JAK2 Dasatinib ABL1, ABL2, DDR1, DDR2, SRC, TNK2
    [Show full text]
  • Erbb3 Is Involved in Activation of Phosphatidylinositol 3-Kinase by Epidermal Growth Factor STEPHEN P
    MOLECULAR AND CELLULAR BIOLOGY, June 1994, p. 3550-3558 Vol. 14, No. 6 0270-7306/94/$04.00+0 Copyright C 1994, American Society for Microbiology ErbB3 Is Involved in Activation of Phosphatidylinositol 3-Kinase by Epidermal Growth Factor STEPHEN P. SOLTOFF,l* KERMIT L. CARRAWAY III,1 S. A. PRIGENT,2 W. G. GULLICK,2 AND LEWIS C. CANTLEY' Division of Signal Transduction, Department ofMedicine, Beth Israel Hospital, Boston, Massachusetts 02115,1 and Molecular Oncology Laboratory, ICRF Oncology Group, Hammersmith Hospital, London W12 OHS, United Kingdom2 Received 11 October 1993/Returned for modification 11 November 1993/Accepted 24 February 1994 Conflicting results concerning the ability of the epidermal growth factor (EGF) receptor to associate with and/or activate phosphatidylinositol (Ptdlns) 3-kinase have been published. Despite the ability of EGF to stimulate the production of Ptdlns 3-kinase products and to cause the appearance of PtdIns 3-kinase activity in antiphosphotyrosine immunoprecipitates in several cell lines, we did not detect EGF-stimulated Ptdlns 3-kinase activity in anti-EGF receptor immunoprecipitates. This result is consistent with the lack of a phosphorylated Tyr-X-X-Met motif, the p85 Src homology 2 (SH2) domain recognition sequence, in this receptor sequence. The EGF receptor homolog, ErbB2 protein, also lacks this motif. However, the ErbB3 protein has seven repeats of the Tyr-X-X-Met motif in the carboxy-terminal unique domain. Here we show that in A431 cells, which express both the EGF receptor and ErbB3, Ptdlns 3-kinase coprecipitates with the ErbB3 protein (pl80eR3) in response to EGF. p180B3 is also shown to be tyrosine phosphorylated in response to EGF.
    [Show full text]
  • PSM, a Mediator of PDGF-BB-, IGF-I-, and Insulin-Stimulated Mitogenesis
    Oncogene (2000) 19, 39 ± 50 ã 2000 Macmillan Publishers Ltd All rights reserved 0950 ± 9232/00 $15.00 www.nature.com/onc PSM, a mediator of PDGF-BB-, IGF-I-, and insulin-stimulated mitogenesis Heimo Riedel*,1,2, Nasim Yousaf1, Yuyuan Zhao4, Heping Dai1,3, Youping Deng1 and Jian Wang1 1Department of Biological Sciences, Wayne State University, Detroit, Michigan, MI 48202, USA; 2Member, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan, MI 48201, USA PSM/SH2-B has been described as a cellular partner of Introduction the FceRI receptor, insulin receptor (IR), insulin-like growth factor-I (IGF-I) receptor (IGF-IR), and nerve The platelet-derived growth factor (PDGF) family growth factor receptor (TrkA). A function has been represents key mitogenic and chemotactic factors that proposed in neuronal dierentiation and development but have been exploited by the Simian sarcoma virus v-sis its role in other signaling pathways is still unclear. To oncogene which results in altered PDGF expression further elucidate the physiologic role of PSM we have (Water®eld et al., 1983). Normal cellular PDGF identi®ed additional mitogenic receptor tyrosine kinases appears in three distinct isoforms as any combination as putative PSM partners including platelet-derived of the PDGF A and B chains which act in paracrine growth factor (PDGF) receptor (PDGFR) beta, hepato- and autocrine mechanisms (Heldin, 1993). PDGF cyte growth factor receptor (Met), and ®broblast growth receptor (PDGFR) signal transduction appears to be factor receptor. We have mapped Y740 as a site of a largely membrane delineated event (Hughes et al., PDGFR beta that is involved in the association with 1996).
    [Show full text]
  • The Erbb Receptor Tyrosine Family As Signal Integrators
    Endocrine-Related Cancer (2001) 8 151–159 The ErbB receptor tyrosine family as signal integrators N E Hynes, K Horsch, M A Olayioye and A Badache Friedrich Miescher Institute, PO Box 2543, CH-4002 Basel, Switzerland (Requests for offprints should be addressed to N E Hynes, Friedrich Miescher Institute, R-1066.206, Maulbeerstrasse 66, CH-4058 Basel, Switzerland. Email: [email protected]) (M A Olayioye is now at The Walter and Eliza Hall Institute of Medical Research, PO Royal Melbourne Hospital, Victoria 3050, Australia) Abstract ErbB receptor tyrosine kinases (RTKs) and their ligands have important roles in normal development and in human cancer. Among the ErbB receptors only ErbB2 has no direct ligand; however, ErbB2 acts as a co-receptor for the other family members, promoting high affinity ligand binding and enhancement of ligand-induced biological responses. These characteristics demonstrate the central role of ErbB2 in the receptor family, which likely explains why it is involved in the development of many human malignancies, including breast cancer. ErbB RTKs also function as signal integrators, cross-regulating different classes of membrane receptors including receptors of the cytokine family. Cross-regulation of ErbB RTKs and cytokines receptors represents another mechanism for controlling and enhancing tumor cell proliferation. Endocrine-Related Cancer (2001) 8 151–159 Introduction The EGF-related peptide growth factors The epidermal growth factor (EGF) or ErbB family of type ErbB receptors are activated by ligands, known as the I receptor tyrosine kinases (RTKs) has four members:EGF EGF-related peptide growth factors (reviewed in Peles & receptor, also termed ErbB1/HER1, ErbB2/Neu/HER2, Yarden 1993, Riese & Stern 1998).
    [Show full text]
  • Intratumoral Heterogeneity of Receptor Tyrosine Kinases EGFR and PDGFRA Amplification in Glioblastoma Defines Subpopulations with Distinct Growth Factor Response
    Intratumoral heterogeneity of receptor tyrosine kinases EGFR and PDGFRA amplification in glioblastoma defines subpopulations with distinct growth factor response Nicholas J. Szerlipa, Alicia Pedrazab, Debyani Chakravartyb, Mohammad Azimc, Jeremy McGuirec, Yuqiang Fangd, Tatsuya Ozawae, Eric C. Hollande,f,g,h, Jason T. Hused,h, Suresh Jhanward, Margaret A. Levershac, Tom Mikkelseni, and Cameron W. Brennanb,f,h,1 aDepartment of Neurosurgery, Wayne State University Medical School, Detroit, MI 48201; bHuman Oncology and Pathogenesis Program, cMolecular Cytogenetic Core Laboratory, dDepartment of Pathology, eCancer Biology and Genetics Program, fDepartment of Neurosurgery, gDepartment of Surgery, and hBrain Tumor Center, Memorial Sloan-Kettering Cancer Center, New York, NY 10065; and iDepartments of Neurology and Neurosurgery, Henry Ford Health System, Detroit, MI 48202 Edited by Webster K. Cavenee, Ludwig Institute, University of California at San Diego, La Jolla, CA, and approved December 29, 2011 (received for review August 29, 2011) Glioblastoma (GBM) is distinguished by a high degree of intra- demonstrated in GBM and has been shown to mediate resistance tumoral heterogeneity, which extends to the pattern of expression to single-RTK inhibition through “RTK switching” in cell lines and amplification of receptor tyrosine kinases (RTKs). Although (11). Although such RTK coactivation has been measured at the most GBMs harbor RTK amplifications, clinical trials of small-mole- protein level, its significance in maintaining tumor cell sub- cule inhibitors targeting individual RTKs have been disappointing to populations has not been established. date. Activation of multiple RTKs within individual GBMs provides We have previously reported prominent PDGFR activation by a theoretical mechanism of resistance; however, the spectrum of platelet-derived growth factor beta (PDGFB) ligand in GBMs EGFR MET fi functional RTK dependence among tumor cell subpopulations in with or ampli cation (12).
    [Show full text]
  • Ephrin-A Binding and Epha Receptor Expression Delineate the Matrix Compartment of the Striatum
    The Journal of Neuroscience, June 15, 1999, 19(12):4962–4971 Ephrin-A Binding and EphA Receptor Expression Delineate the Matrix Compartment of the Striatum L. Scott Janis, Robert M. Cassidy, and Lawrence F. Kromer Department of Cell Biology and Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20007 The striatum integrates limbic and neocortical inputs to regulate matrix neurons. In situ hybridization for EphA RTKs reveals that sensorimotor and psychomotor behaviors. This function is de- the two different ligand binding patterns strictly match pendent on the segregation of striatal projection neurons into the mRNA expression patterns of EphA4 and EphA7. anatomical and functional components, such as the striosome Ligand–receptor binding assays indicate that ephrin-A1 and and matrix compartments. In the present study the association ephrin-A4 selectively bind EphA4 but not EphA7 in the lysates of ephrin-A cell surface ligands and EphA receptor tyrosine of striatal tissue. Conversely, ephrin-A2, ephrin-A3, and kinases (RTKs) with the organization of these compartments ephrin-A5 bind EphA7 but not EphA4. These observations im- was determined in postnatal rats. Ephrin-A1 and ephrin-A4 plicate selective interactions between ephrin-A molecules and selectively bind to EphA receptors on neurons restricted to the EphA RTKs as potential mechanisms for regulating the com- matrix compartment. Binding is absent from the striosomes, partmental organization of the striatum. which were identified by m-opioid
    [Show full text]
  • Human Ephb3 Antibody Antigen Affinity-Purified Polyclonal Sheep Igg Catalog Number: AF5667
    Human EphB3 Antibody Antigen Affinity-purified Polyclonal Sheep IgG Catalog Number: AF5667 DESCRIPTION Species Reactivity Human Specificity Detects human EphB3 in direct ELISAs and Western blots. In direct ELISAs, approximately 3% cross­reactivity with recombinant mouse EphB3 is observed, and less than 1% cross­reactivity with recombinant rat EphB1, recombinant human (rh) EphB2 and rhEphB4 is observed. Source Polyclonal Sheep IgG Purification Antigen Affinity­purified Immunogen Mouse myeloma cell line NS0­derived recombinant human EphB3 Leu38­Ala550 Accession # P54753 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 DATA Western Blot Detection of Human EphB3 by Western Blot. Western blot shows lysates of SH­SY5Y human neuroblastoma cell line. PVDF Membrane was probed with 1 µg/mL of Sheep Anti­Human EphB3 Antigen Affinity­ purified Polyclonal Antibody (Catalog # AF5667) followed by HRP­ conjugated Anti­Sheep IgG Secondary Antibody (Catalog # HAF016). A specific band was detected for EphB3 at approximately 110 kDa (as indicated). This experiment was conducted under reducing conditions and using Immunoblot Buffer Group 8. PREPARATION AND STORAGE Reconstitution Sterile PBS to a final concentration of 0.2 mg/mL. Shipping The product is shipped at ambient temperature. Upon receipt, store it immediately at the temperature recommended below.
    [Show full text]