Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-22dnz Total loading time: 0 Render date: 2024-04-29T07:40:54.833Z Has data issue: false hasContentIssue false

10 - HER

from Part 2.1 - Molecular pathways underlying carcinogenesis: signal transduction

Published online by Cambridge University Press:  05 February 2015

Wolfgang J. Köstler
Affiliation:
Department of Biological Regulation, The Weizmann Institute of Science, Rehovot, Israel
Yosef Yarden
Affiliation:
Department of Biological Regulation, The Weizmann Institute of Science, Rehovot, Israel
Edward P. Gelmann
Affiliation:
Columbia University, New York
Charles L. Sawyers
Affiliation:
Memorial Sloan-Kettering Cancer Center, New York
Frank J. Rauscher, III
Affiliation:
The Wistar Institute Cancer Centre, Philadelphia
Get access

Summary

ERBB: The receptor network

ERBB receptors (also called HER receptors) are composed of an extra-cellular domain (comprising subdomains I–IV), a single transmembrane portion, and a large intra-cellular domain comprising a short juxtamembrane portion, a bilobular tyrosine kinase domain and a carboxyl-terminal tail. The principal ERBB receptor activation mechanism involves ligand binding, which activates the kinase domains of receptor homo- and heterodimers. Notably, the ligandless ERBB2 and the kinase-dead ERBB3 are non-autonomous, yet confer potent signaling upon heterodimerization. Activated kinase domains then phosphorylate tyrosine residues located in the cytoplasmic receptor's portion, which serve as docking sites for proteins containing phosphotyrosine-binding or Src homology-2 domains (Figure 10.1). These signaling effectors and adaptor proteins link activated receptors directly or indirectly to canonical intra-cellular pathways, depicted in Figure 10.2, as well as to the endocytic, desensitizing machinery. Although there is considerable overlap amongst the individual ERBB receptors with regards to the recruited signaling effectors and adaptor proteins, the stoichiometry of recruited adaptors varies, and some pathways are unique to individual receptors (1,2). Moreover, many tyrosine residues can bind several adaptors and effectors, which, in turn, can act as molecular scaffolds. For instance, phosphorylated tyrosine residues 1068 and 1086 of the EGFR recruit the adaptor protein Grb2, which can bind both positive (Sos) and negative (e.g. Cbl, Ship, Socs, Sprouty, Ack1) regulators of EGFR signaling. Further fine-tuning of receptor activity and connectivity is achieved by phosphorylation of cytoplasmic ERBB receptor residues by intra-cellular kinases (e.g. Src phosphorylates EGFR on multiple residues, including tyrosine 845, which then serves as a novel docking site for STAT5b (3)). ERBB signaling may both activate and undergo activation by several heterologous receptors (e.g. the HGF-receptor MET) through multiple mechanisms, including formation of signaling-competent receptor heteromers, receptor transmodulation, and by transcriptional induction of heterologous ligands and receptors.

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 85 - 109
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Schulze, WX, Deng, L, Mann, M. Phosphotyrosine interactome of the ERBB-receptor kinase family. Molecular Systems Biology 2005;1:2005.0008.
Jones, RB, Gordus, A, Krall, JA, MacBeath, G. A quantitative protein interaction network for the ERBB receptors using protein microarrays. Nature 2006;439:168–74.CrossRef
Kloth, MT, Laughlin, KK, Biscardi, JS, et al. STAT5b, a mediator of synergism between c-Src and the epidermal growth factor receptor. Journal of Biological Chemistry 2003;278:1671–9.CrossRefGoogle ScholarPubMed
Gschwind, A, Fischer, OM, Ullrich, A. The discovery of receptor tyrosine kinases: targets for cancer therapy. Nature Reviews Cancer 2004;4:361–70.CrossRef
Yarden, Y, Sliwkowski, MX. Untangling the ERBB signalling network. Nature Reviews Molecular and Cellular Biology 2001;2:127–37.CrossRef
Normanno, N, Bianco, C, Strizzi, L, et al. The ERBB receptors and their ligands in cancer: an overview. Current Drug Targets 2005;6:243–57.CrossRef
Engelman, JA, Zejnullahu, K, Mitsudomi, T, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 2007;316:1039–43.CrossRef
Schroeder, JA, Lee, DC. Transgenic mice reveal roles for TGFalpha and EGF receptor in mammary gland development and neoplasia. Journal of Mammary Gland Biology and Neoplasia 1997;2:119–29.CrossRefGoogle ScholarPubMed
Humphreys, RC, Hennighausen, L. Transforming growth factor alpha and mouse models of human breast cancer. Oncogene 2000;19:1085–91.CrossRef
Vassar, R, Fuchs, E. Transgenic mice provide new insights into the role of TGF-alpha during epidermal development and differentiation. Genes and Development 1991;5:714–27.CrossRef
Cook, PW, Piepkorn, M, Clegg, CH, et al. Transgenic expression of the human amphiregulin gene induces a psoriasis-like phenotype. Journal of Clinical Investigation 1997;100:2286–94.CrossRefGoogle ScholarPubMed
Ursini-Siegel, J, Schade, B, Cardiff, RD, Muller, WJ. Insights from transgenic mouse models of ERBB2-induced breast cancer. Nature Reviews Cancer 2007;7:389–97.CrossRef
Slamon, DJ, Clark, GM, Wong, SG, et al. Human breast cancer:correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 1987;235:177–82.CrossRef
Perou, CM, Sorlie, T, Eisen, MB, et al. Molecular portraits of human breast tumours. Nature 2000;406:747–52.CrossRef
Moody, SE, Sarkisian, CJ, Hahn, KT, et al. Conditional activation of Neu in the mammary epithelium of transgenic mice results in reversible pulmonary metastasis. Cancer Cell 2002;2:451–61.CrossRef
Hollywood, DP, Hurst, HC. A novel transcription factor, OB2–1, is required for overexpression of the proto-oncogene c-ERBB-2 in mammary tumour lines. EMBO Journal 1993;12:2369–75.
Scott, GK, Daniel, JC, Xiong, X, et al. Binding of an ETS-related protein within the DNase I hypersensitive site of the HER2/neu promoter in human breast cancer cells. Journal of Biological Chemistry 1994;269:19848–58.Google ScholarPubMed
Bosher, JM, Williams, T, Hurst, HC. The developmentally regulated transcription factor AP-2 is involved in c-erbB-2 overexpression in human mammary carcinoma. Proceedings of the National Academy of Sciences USA 1995;92:744–7.CrossRef
Chen, H, Hung, MC. Involvement of co-activator p300 in the transcriptional regulation of the HER-2/neu gene. Journal of Biological Chemistry 1997;272:6101–4.CrossRefGoogle ScholarPubMed
Grooteclaes, M, Vernimmen, D, Plaza, S, et al. A new cis element is involved in the HER2 gene overexpression in human breast cancer cells. Cancer Research 1999;59:2527–31.
Scott, GK, Chang, CH, Erny, KM, et al. Ets regulation of the erbB2 promoter. Oncogene 2000;19:6490–502.CrossRef
Perissi, V, Menini, N, Cottone, E, et al. AP-2 transcription factors in the regulation of ERBB2 gene transcription by oestrogen. Oncogene 2000;19:280–8.CrossRef
Bosc, DG, Janknecht, R. Regulation of Her2/neu promoter activity by the ETS transcription factor, ER81. Journal of Cell Biochemistry 2002;86:174–83.CrossRefGoogle ScholarPubMed
Vernimmen, D, Begon, D, Salvador, C, et al. Identification of HTF (HER2 transcription factor) as an AP-2 (activator protein-2) transcription factor and contribution of the HTF binding site to ERBB2 gene overexpression. Biochemical Journal 2003;370:323–9.CrossRef
Vernimmen, D, Gueders, M, Pisvin, S, Delvenne, P, Winkler, R. Different mechanisms are implicated in ERBB2 gene overexpression in breast and in other cancers. British Journal of Cancer 2003;89:899–906.CrossRefGoogle ScholarPubMed
Begon, DY, Delacroix, L, Vernimmen, D, Jackers, P, Winkler, R. Yin Yang 1 cooperates with activator protein 2 to stimulate ERBB2 gene expression in mammary cancer cells. Journal of Biological Chemistry 2005;280:24428–34.CrossRefGoogle ScholarPubMed
Delacroix, L, Begon, D, Chatel, G, Jackers, P, Winkler, R. Distal ERBB2 promoter fragment displays specific transcriptional and nuclear binding activities in ERBB2 overexpressing breast cancer cells. DNA and Cell Biology2005;24:582–94.
Wu, J, Lee, C, Yokom, D, et al. Disruption of the Y-box binding protein-1 results in suppression of the epidermal growth factor receptor and HER-2. Cancer Research 2006;66:4872–9.CrossRef
Dillon, RL, Brown, ST, Ling, C, Shioda, T, Muller, WJ. An EGR2/CITED1 transcription factor complex and the 14–3–3sigma tumor suppressor are involved in regulating ERBB2 expression in a transgenic-mouse model of human breast cancer. Molecular and Cellular Biology 2007;27:8648–57.CrossRef
Allouche, A, Nolens, G, Tancredi, A, et al. The combined immunodetection of AP-2 alpha and YY1 transcription factors is associated with ERBB2 gene overexpression in primary breast tumours. Breast Cancer Research 2008;10:R9.
Matin, A, Hung, MC. The retinoblastoma gene product, Rb, represses neu expression through two regions within the neu regulatory sequence. Oncogene 1994;9:1333–9.
Zuo, T, Wang, L, Morrison, C, et al. FOXP3 is an X-linked breast cancer suppressor gene and an important repressor of the HER-2/ERBB2 oncogene. Cell 2007;129:1275–86.CrossRef
Yu, D, Suen, TC, Yan, DH, Chang, LS, Hung, MC. Transcriptional repression of the neu protooncogene by the adenovirus 5 E1A gene products. Proceedings of the National Academy of Sciences USA 1990;87:4499–503.CrossRef
Yu, D, Matin, A, Hung, MC. The retinoblastoma gene product suppresses neu oncogene-induced transformation via transcriptional repression of neu. Journal of Biological Chemistry 1992;267:10 203–6.Google ScholarPubMed
Wang, SC, Hung, MC. Transcriptional targeting of the HER-2/neu oncogene. Drugs Today (Barcelona) 2000;36:835–43.
Chen, H, Yu, D, Chinnadurai, G, Karunagaran, D, Hung, MC. Mapping of adenovirus 5 E1A domains responsible for suppression of neu-mediated transformation via transcriptional repression of neu. Oncogene 1997;14:1965–71.CrossRef
Chang, JY, Xia, W, Shao, R, et al. The tumor suppression activity of E1A in HER-2/neu-overexpressing breast cancer. Oncogene 1997;14:561–8.
Qin, HR, Iliopoulos, D, Nakamura, T, et al. Wwox suppresses prostate cancer cell growth through modulation of ERBB2-mediated androgen receptor signaling. Molecular Cancer Research 2007;5:957–65.CrossRef
Xing, X, Wang, SC, Xia, W, et al. The ets protein PEA3 suppresses HER-2/neu overexpression and inhibits tumorigenesis. Nature Medicine 2000;6:189–95.CrossRef
Menendez, JA, Vellon, L, Colomer, R, Lupu, R. Effect of gamma-linolenic acid on the transcriptional activity of the Her-2/neu (erbB-2) oncogene. Journal of the National Cancer Institute 2005;97:1611–15.CrossRefGoogle ScholarPubMed
Johnson, AC, Murphy, BA, Matelis, CM, et al. Activator protein-1 mediates induced but not basal epidermal growth factor receptor gene expression. Molecular Medicine 2000;6:17–27.
Sakla, MS, Shenouda, NS, Ansell, PJ, Macdonald, RS, Lubahn, DB. Genistein affects HER2 protein concentration, activation, and promoter regulation in BT-474 human breast cancer cells. Endocrine 2007;32:69–78.CrossRef
Zhang, YW, Wang, R, Liu, Q, et al. Presenilin/gamma-secretase-dependent processing of beta-amyloid precursor protein regulates EGF receptor expression. Proceedings of the National Academy of Sciences USA 2007;104:10 613–8.
Rubinstein, YR, Proctor, KN, Bergel, M, Murphy, B, Johnson, AC. Interferon regulatory factor-1 is a major regulator of epidermal growth factor receptor gene expression. FEBS Letters 1998;431:268–72.CrossRef
Prudenziati, M, Sirito, M, van Dam, H, Ravazzolo, R. Adenovirus E1A down-regulates the EGF receptor via repression of its promoter. International Journal of Cancer 2000;88:943–8.3.0.CO;2-F>CrossRefGoogle ScholarPubMed
Nishi, H, Senoo, M, Nishi, KH, et al. p53 Homologue p63 represses epidermal growth factor receptor expression. Journal of Biological Chemistry 2001;276:41 717–24.CrossRefGoogle ScholarPubMed
Ludes-Meyers, JH, Subler, MA, Shivakumar, CV, et al. Transcriptional activation of the human epidermal growth factor receptor promoter by human p53. Molecular and Cellular Biology 1996;16:6009–19.CrossRef
McGaffin, KR, Acktinson, LE, Chrysogelos, SA. Growth and EGFR regulation in breast cancer cells by vitamin D and retinoid compounds. Breast Cancer Research and Treatment 2004;86:55–73.CrossRef
Salvatori, L, Ravenna, L, Felli, MP, et al. Identification of an estrogen-mediated deoxyribonucleic acid-binding independent transactivation pathway on the epidermal growth factor receptor gene promoter. Endocrinology 2000;141:2266–74.CrossRef
Vallian, S, Chin, KV, Chang, KS. The promyelocytic leukemia protein interacts with Sp1 and inhibits its transactivation of the epidermal growth factor receptor promoter. Molecular and Cellular Biology 1998;18:7147–56.CrossRef
Xu, J, Thompson, KL, Shephard, LB, Hudson, LG, Gill, GN. T3 receptor suppression of Sp1-dependent transcription from the epidermal growth factor receptor promoter via overlapping DNA-binding sites. Journal of Biological Chemistry 1993;268:16 065–73.Google ScholarPubMed
Skinner, A, Hurst, HC. Transcriptional regulation of the c-erbB-3 gene in human breast carcinoma cell lines. Oncogene 1993;8:3393–401.
Zhu, CH, Domann, FE. Dominant negative interference of transcription factor AP-2 causes inhibition of ERBB-3 expression and suppresses malignant cell growth. Breast Cancer Research Treatment 2002;71:47–57.CrossRef
Sheikh, MS, Carrier, F, Johnson, AC, Ogdon, SE, Fornace, AJ. Identification of an additional p53-responsive site in the human epidermal growth factor receptor gene promotor. Oncogene 1997;15:1095–101.CrossRef
Deb, SP, Munoz, RM, Brown, DR, Subler, MA, Deb, S. Wild-type human p53 activates the human epidermal growth factor receptor promoter. Oncogene 1994;9:1341–9.
Wolf-Yadlin, A, Hautaniemi, S, Lauffenburger, DA, White, FM. Multiple reaction monitoring for robust quantitative proteomic analysis of cellular signaling networks. Proceedings of the National Academy of Sciences USA 2007;104:5860–5.CrossRef
Kholodenko, BN. Cell-signalling dynamics in time and space. Nature Reviews Molecular and Cellular Biology 2006;7:165–76.CrossRef
Birtwistle, MR, Hatakeyama, M, Yumoto, N, et al. Ligand-dependent responses of the ERBB signaling network:experimental and modeling analyses. Molecular Systems Biology 2007;3:144.
Blagoev, B, Kratchmarova, I, Ong, SE, et al. A proteomics strategy to elucidate functional protein-protein interactions applied to EGF signaling. Nature Biotechnology 2003;21:315–18.CrossRef
Olsen, JV, Blagoev, B, Gnad, F, et al. Global, in vivo, and site-specific phosphorylation dynamics in signaling networks. Cell 2006;127:635–48.CrossRef
Reynolds, AR, Tischer, C, Verveer, PJ, Rocks, O, Bastiaens, PI. EGFR activation coupled to inhibition of tyrosine phosphatases causes lateral signal propagation. Nature Cell Biology 2003;5:447–53.CrossRef
Nagashima, T, Shimodaira, H, Ide, K, et al. Quantitative transcriptional control of ERBB receptor signaling undergoes graded to biphasic response for cell differentiation. Journal of Biological Chemistry 2007;282:4045–56.CrossRefGoogle ScholarPubMed
Moscatello, DK, Holgado-Madruga, M, Emlet, DR, Montgomery, RB, Wong, AJ. Constitutive activation of phosphatidylinositol 3-kinase by a naturally occurring mutant epidermal growth factor receptor. Journal of Biological Chemistry 1998;273:200–6.CrossRefGoogle ScholarPubMed
Huang, PH, Mukasa, A, Bonavia, R, et al. Quantitative analysis of EGFRvIII cellular signaling networks reveals a combinatorial therapeutic strategy for glioblastoma. Proceedings of the National Academy of Sciences USA 2007;104:12 867–72.
Sordella, R, Bell, DW, Haber, DA, Settleman J. Gefitinib-sensitizing EGFR mutations in lung cancer activate anti-apoptotic pathways. Science 2004;305:1163–7.CrossRef
Blobel, CP. ADAMs:key components in EGFR signalling and development. Nature Reviews Molecular and Cellular Biology 2005;6:32–43.CrossRef
Daub, H, Weiss, FU, Wallasch, C, Ullrich, A. Role of transactivation of the EGF receptor in signalling by G-protein-coupled receptors. Nature 1996;379:557–60.CrossRef
Prenzel, N, Zwick, E, Daub, H, et al. EGF receptor transactivation by G-protein-coupled receptors requires metalloproteinase cleavage of proHB-EGF. Nature 1999;402:884–8.CrossRef
Civenni, G, Holbro, T, Hynes, NE. Wnt1 and Wnt5a induce cyclin D1 expression through ERBB1 transactivation in HC11 mammary epithelial cells. EMBO Reports 2003;4:166–71.CrossRef
Fischer, OM, Hart, S, Gschwind, A, Ullrich, A. EGFR signal transactivation in cancer cells. Biochemical Society Transactions 2003;31:1203–8.CrossRef
Gschwind, A, Zwick, E, Prenzel, N, Leserer, M, Ullrich, A. Cell communication networks: epidermal growth factor receptor transactivation as the paradigm for interreceptor signal transmission. Oncogene 2001;20:1594–600.CrossRef
Fan, H, Derynck, R. Ectodomain shedding of TGF-alpha and other transmembrane proteins is induced by receptor tyrosine kinase activation and MAP kinase signaling cascades. EMBO Journal 1999;18:6962–72.CrossRef
Bao, J, Wolpowitz, D, Role, LW, Talmage, DA. Back signaling by the Nrg-1 intracellular domain. Journal of Cell Biology 2003;161:1133–41.CrossRefGoogle ScholarPubMed
Bao, J, Lin, H, Ouyang, Y, et al. Activity-dependent transcription regulation of PSD-95 by neuregulin-1 and Eos. Nature Neuroscience 2004;7:1250–8.CrossRef
Hieda, M, Isokane, M, Koizumi, M, et al. Membrane-anchored growth factor, HB-EGF, on the cell surface targeted to the inner nuclear membrane. Journal of Cell Biology 2008;180:763–9.CrossRefGoogle ScholarPubMed
Higashiyama, S, Iwabuki, H, Morimoto, C, et al. Membrane-anchored growth factors, the epidermal growth factor family:beyond receptor ligands. Cancer Science 2008;99:214–20.CrossRef
Yamauchi, T, Yamauchi, N, Ueki, K, et al. Constitutive tyrosine phosphorylation of ERBB-2 via Jak2 by autocrine secretion of prolactin in human breast cancer. Journal of Biological Chemistry 2000;275:33 937–44.CrossRefGoogle ScholarPubMed
Yamauchi, T, Ueki, K, Tobe, K, et al. Tyrosine phosphorylation of the EGF receptor by the kinase Jak2 is induced by growth hormone. Nature 1997;390:91–6.CrossRef
Knebel, A, Rahmsdorf, HJ, Ullrich, A, Herrlich, P. Dephosphorylation of receptor tyrosine kinases as target of regulation by radiation, oxidants or alkylating agents. EMBO Journal 1996;15:5314–25.
Zwang, Y, Yarden, Y. p38 MAP kinase mediates stress-induced internalization of EGFR: implications for cancer chemotherapy. EMBO Journal 2006;25:4195–206.CrossRef
Bao, J, Alroy, I, Waterman, H, et al. Threonine phosphorylation diverts internalized epidermal growth factor receptors from a degradative pathway to the recycling endosome. Journal of Biological Chemistry 2000;275:26 178–86.CrossRefGoogle ScholarPubMed
Chen, Y, Grall, D, Salcini, AE, et al. Shc adaptor proteins are key transducers of mitogenic signaling mediated by the G protein-coupled thrombin receptor. EMBO Journal 1996;15:1037–44.
Bill, HM, Knudsen, B, Moores, SL, et al. Epidermal growth factor receptor-dependent regulation of integrin-mediated signaling and cell cycle entry in epithelial cells. Molecular and Cellular Biology 2004;24:8586–99.CrossRef
De Luca, A, Carotenuto, A, Rachiglio, A, et al. The role of the EGFR signaling in tumor microenvironment. Journal of Cell Physiology 2008;214:559–67.CrossRefGoogle ScholarPubMed
Keates, S, Sougioultzis, S, Keates, AC, et al. cag+ Helicobacter pylori induce transactivation of the epidermal growth factor receptor in AGS gastric epithelial cells. Journal of Biological Chemistry 2001;276:48 127–34.CrossRefGoogle ScholarPubMed
Frank, SJ. Mechanistic aspects of crosstalk between GH and PRL and ERBB receptor family signaling. Journal of Mammary Gland Biology and Neoplasia 2008;13:119–29.CrossRefGoogle ScholarPubMed
Lemjabbar, H, Li, D, Gallup, M, et al. Tobacco smoke-induced lung cell proliferation mediated by tumor necrosis factor alpha-converting enzyme and amphiregulin. Journal of Biological Chemistry 2003;278:26 202–7.CrossRefGoogle ScholarPubMed
Pai, R, Soreghan, B, Szabo, IL, et al. Prostaglandin E2 transactivates EGF receptor: a novel mechanism for promoting colon cancer growth and gastrointestinal hypertrophy. Nature Medicine 2002;8:289–93.CrossRef
Fischer, OM, Hart, S, Gschwind, A, Prenzel, N, Ullrich, F. Oxidative and osmotic stress signaling in tumor cells is mediated by ADAM proteases and heparin-binding epidermal growth factor. Molecular and Cellular Biology 2004;24:5172–83.CrossRef
Hart, S, Fischer, OM, Prenzel, N, et al. GPCR-induced migration of breast carcinoma cells depends on both EGFR signal transactivation and EGFR-independent pathways. Biological Chemistry 2005;386:845–55.CrossRef
Gschwind, A, Hart, S, Fischer, OM, Ullrich, A. TACE cleavage of proamphiregulin regulates GPCR-induced proliferation and motility of cancer cells. EMBO Journal 2003;22:2411–21.CrossRef
Awwad, R, Humphrey, LE, Periyasamy, B, et al. The EGF/TGFalpha response element within the TGFalpha promoter consists of a multi-complex regulatory element. Oncogene 1999;18:5923–35.CrossRef
Howell, GM, Humphrey, LE, Ziober, BL, et al. Regulation of transforming growth factor alpha expression in a growth factor-independent cell line. Molecular and Cellular Biology 1998;18:303–13.CrossRef
Schulze, A, Lehmann, K, Jefferies, HB, McMahon, M, Downward, J. Analysis of the transcriptional program induced by Raf in epithelial cells. Genes and Development 2001;15:981–94.CrossRef
Amit, I, Citri, A, Shay, T, et al. A module of negative feedback regulators defines growth factor signaling. Nature Genetics 2007;39:503–12.CrossRef
Joslin, EJ, Opresko, LK, Wells, A, Wiley, HS, Lauffenburger, DA. EGF-receptor-mediated mammary epithelial cell migration is driven by sustained ERK signaling from autocrine stimulation. Journal of Cell Science 2007;120:3688–99.CrossRefGoogle ScholarPubMed
Motoyama, AB, Hynes, NE, Lane, HA. The efficacy of ERBB receptor-targeted anticancer therapeutics is influenced by the availability of epidermal growth factor-related peptides. Cancer Research 2002;62:3151–8.
Zhou, BB, Peyton, M, He, B, et al. Targeting ADAM-mediated ligand cleavage to inhibit HER3 and EGFR pathways in non-small cell lung cancer. Cancer Cell 2006;10:39–50.CrossRef
Ogiso, H, Ishitani, R, Nureki, O, et al. Crystal structure of the complex of human epidermal growth factor and receptor extracellular domains. Cell 2002;110:775–87.CrossRef
Garrett, TP, McKern, NM, Lou, M, et al. Crystal structure of a truncated epidermal growth factor receptor extracellular domain bound to transforming growth factor alpha. Cell 2002;110:763–73.CrossRef
Ferguson, KM, Berger, MB, Mendrola, JM, et al. EGF activates its receptor by removing interactions that autoinhibit ectodomain dimerization. Molecular Cell 2003;11:507–17.CrossRef
Burgess, AW, Cho, HS, Eigenbrot, C, et al. An open-and-shut case? Recent insights into the activation of EGF/ERBB receptors. Molecular Cell 2003;12:541–52.CrossRef
Li, S, Schmitz, KR, Jeffrey, PD, et al. Structural basis for inhibition of the epidermal growth factor receptor by cetuximab. Cancer Cell 2005;7:301–11.CrossRef
Cho, HS, Leahy, DJ. Structure of the extracellular region of HER3 reveals an interdomain tether. Science 2002;297:1330–3.CrossRef
Bouyain, S, Longo, PA, Li, S, Ferguson, KM, Leahy, DJ. The extracellular region of ERBB4 adopts a tethered conformation in the absence of ligand. Proceedings of the National Academy of Sciences USA 2005;102:15 024–9.
Landau, M, Ben-Tal, N. Dynamic equilibrium between multiple active and inactive conformations explains regulation and oncogenic mutations in ERBB receptors. Biochimica et Biophysica Acta 2008;1785:12–31.
Garrett, TP, McKern, NM, Lou, M, et al. The crystal structure of a truncated ERBB2 ectodomain reveals an active conformation, poised to interact with other ERBB receptors. Molecular Cell 2003;11:495–505.CrossRef
Cho, HS, Mason, K, Ramyar, KX, et al. Structure of the extracellular region of HER2 alone and in complex with the Herceptin Fab. Nature 2003;421:756–60.CrossRef
Karunagaran, D, Tzahar, E, Beerli, RR, et al. ERBB-2 is a common auxiliary subunit of NDF and EGF receptors: implications for breast cancer. EMBO Journal 1996;15:254–64.
Sliwkowski, MX, Schaefer, G, Akita, RW, et al. Coexpression of erbB2 and erbB3 proteins reconstitutes a high affinity receptor for heregulin. Journal of Biological Chemistry 1994;269:14 661–5.Google ScholarPubMed
Tzahar, E, Waterman, H, Chen, X, et al. A hierarchical network of interreceptor interactions determines signal transduction by Neu differentiation factor/neuregulin and epidermal growth factor. Molecular and Cellular Biology 1996;16:5276–87.CrossRef
Graus-Porta, D, Beerli, RR, Daly, JM, Hynes, NE. ERBB-2, the preferred heterodimerization partner of all ERBB receptors, is a mediator of lateral signaling. EMBO Journal 1997;16:1647–55.CrossRef
Xu, W, Mimnaugh, E, Rosser, MF, et al. Sensitivity of mature Erbb2 to geldanamycin is conferred by its kinase domain and is mediated by the chaperone protein Hsp90. Journal of Biological Chemistry 2001;276:3702–8.CrossRefGoogle ScholarPubMed
Mimnaugh, EG, Chavany, C, Neckers, L. Polyubiquitination and proteasomal degradation of the p185c-erbB-2 receptor protein-tyrosine kinase induced by geldanamycin. Journal of Biological Chemistry 1996;271:22796–801.CrossRefGoogle ScholarPubMed
Citri, A, Gan, J, Mosesson, Y, et al. Hsp90 restrains ERBB-2/HER2 signalling by limiting heterodimer formation. EMBO Reports 2004;5:1165–70.CrossRef
Ozcan, F, Klein, P, Lemmon, MA, Lax, I, Schlessinger, J. On the nature of low- and high-affinity EGF receptors on living cells. Proceedings of the National Academy of Sciences USA 2006;103:5735–40.CrossRef
Mattoon, D, Klein, P, Lemmon, MA, Lax, I, Schlessinger, J. The tethered configuration of the EGF receptor extracellular domain exerts only a limited control of receptor function. Proceedings of the National Academy of Sciences USA 2004;101:923–8.CrossRef
Gadella, TW., Jovin, TM. Oligomerization of epidermal growth factor receptors on A431 cells studied by time-resolved fluorescence imaging microscopy. A stereochemical model for tyrosine kinase receptor activation. Journal of Cell Biology 1995;129:1543–58.CrossRefGoogle ScholarPubMed
Yu, X, Sharma, KD, Takahashi, T, Iwamoto, R, Mekada, E. Ligand-independent dimer formation of epidermal growth factor receptor (EGFR) is a step separable from ligand-induced EGFR signaling. Molecular Biology of the Cell 2002;13:2547–57.CrossRef
Teramura, Y, Ichinose, J, Takagi, H, et al. Single-molecule analysis of epidermal growth factor binding on the surface of living cells. EMBO Journal 2006;25:4215–22.CrossRef
Liu, P, Sudhaharan, T, Koh, RM, et al. Investigation of the dimerization of proteins from the epidermal growth factor receptor family by single wavelength fluorescence cross-correlation spectroscopy. Biophysical Journal 2007;93:684–98.CrossRef
Warren, CM, Landgraf, R. Signaling through ERBB receptors: multiple layers of diversity and control. Cellular Signaling 2006;18:923–33.CrossRef
Li, E, Hristova, K. Role of receptor tyrosine kinase transmembrane domains in cell signaling and human pathologies. Biochemistry 2006;45:6241–51.CrossRef
Fleishman, SJ, Schlessinger, J, Ben-Tal, N. A putative molecular-activation switch in the transmembrane domain of erbB2. Proceedings of the National Academy of Sciences USA 2002;99:15 937–40.
Mendrola, JM, Berger, MB, King, MC, Lemmon, MA. The single transmembrane domains of ERBB receptors self-associate in cell membranes. Journal of Biological Chemistry 2002;277:4704–12.CrossRefGoogle ScholarPubMed
Moriki, T, Maruyama, H, Maruyama, IN. Activation of preformed EGF receptor dimers by ligand-induced rotation of the transmembrane domain. Journal of Molecular Biology 2001;311:1011–26.CrossRefGoogle ScholarPubMed
Batra, SK, Castelino-Prabhu, S, Wikstrand, CJ, et al. Epidermal growth factor ligand-independent, unregulated, cell-transforming potential of a naturally occurring human mutant EGFRvIII gene. Cell Growth and Differentiation 1995;6:1251–9.
Sugawa, N, Ekstrand, AJ, James, CD, Collins, VP. Identical splicing of aberrant epidermal growth factor receptor transcripts from amplified rearranged genes in human glioblastomas. Proceedings of the National Academy of Sciences USA 1990;87:8602–6.CrossRef
Wong, AJ, Ruppert, JM, Bigner, SH, et al. Structural alterations of the epidermal growth factor receptor gene in human gliomas. Proceedings of the National Academy of Sciences USA 1992;89:2965–9.CrossRef
Ekstrand, AJ, Sugawa, N, James, CD, Collins, VP. Amplified and rearranged epidermal growth factor receptor genes in human glioblastomas reveal deletions of sequences encoding portions of the N- and/or C-terminal tails. Proceedings of the National Academy of Sciences USA 1992;89:4309–13.CrossRef
de la Iglesia, N, Konopka, G, Puram, SV, et al. Identification of a PTEN-regulated STAT3 brain tumor suppressor pathway. Genes and Development 2008;22:449–62.CrossRef
Lee, JC, Vivanco, I, Beroukhim, R, et al. Epidermal growth factor receptor activation in glioblastoma through novel missense mutations in the extracellular domain. PLoS Medicine 2006;3:e485.
Christianson, TA, Doherty, JK, Lin, YJ, et al. NH2-terminally truncated HER-2/neu protein: relationship with shedding of the extracellular domain and with prognostic factors in breast cancer. Cancer Research 1998;58:5123–9.
Codony-Servat, J, Albanell, J, Lopez-Talavera, JC, Arribas, J, Baselga, J. Cleavage of the HER2 ectodomain is a pervanadate-activable process that is inhibited by the tissue inhibitor of metalloproteases-1 in breast cancer cells. Cancer Research 1999;59:1196–201.
Liu, PC, Liu, X, Li, Y, et al. Identification of ADAM10 as a major source of HER2 ectodomain sheddase activity in HER2 overexpressing breast cancer cells. Cancer Biology and Therapy 2006;5:657–64.CrossRef
Scott, GK, Robles, R, Park, JW, et al. A truncated intracellular HER2/neu receptor produced by alternative RNA processing affects growth of human carcinoma cells. Molecular and Cellular Biology 1993;13:2247–57.CrossRef
Kwong, KY, Hung, MC. A novel splice variant of HER2 with increased transformation activity. Molecular Carcinogenesis 1998;23:62–8.3.0.CO;2-O>CrossRef
Anido, J, Scaltriti, M, Bech Serra, JJ, et al. Biosynthesis of tumorigenic HER2 C-terminal fragments by alternative initiation of translation. EMBO Journal 2006;25:3234–44.CrossRef
Molina, MA, Codony-Servat, J, Albanell, J, et al. Trastuzumab (herceptin), a humanized anti-Her2 receptor monoclonal antibody, inhibits basal and activated Her2 ectodomain cleavage in breast cancer cells. Cancer Research 2001;61:4744–9.
Molina, MA, Saez, R, Ramsey, EE, et al. NH(2)-terminal truncated HER-2 protein but not full-length receptor is associated with nodal metastasis in human breast cancer. Clinical Cancer Research 2002;8:347–53.
Hudelist, G, Kostler, WJ, Attems, J, et al. Her-2/neu-triggered intracellular tyrosine kinase activation: in vivo relevance of ligand-independent activation mechanisms and impact upon the efficacy of trastuzumab-based treatment. British Journal of Cancer 2003;89:983–91.CrossRefGoogle ScholarPubMed
Scaltriti, M, Rojo, F, Ocana, A, et al. Expression of p95HER2, a truncated form of the HER2 receptor, and response to anti-HER2 therapies in breast cancer. Journal of the National Cancer Institute 2007;99:628–38.CrossRefGoogle ScholarPubMed
Frank, B, Hemminki, K, Wirtenberger, M, et al. The rare ERBB2 variant Ile654Val is associated with an increased familial breast cancer risk. Carcinogenesis 2005;26:643–7.CrossRef
McLaughlin, S, Smith, SO, Hayman, MJ, Murray, D. An electrostatic engine model for autoinhibition and activation of the epidermal growth factor receptor (EGFR/ERBB) family. Journal of General Physiology 2005;126:41–53.CrossRefGoogle ScholarPubMed
Thiel, KW, Carpenter, G. Epidermal growth factor receptor juxtamembrane region regulates allosteric tyrosine kinase activation. Proceedings of the National Academy of Sciences USA 2007;104:19 238–43.
Lynch, TJ, Bell, DW, Sordella, R, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. New England Journal of Medicine 2004;350:2129–39.CrossRefGoogle ScholarPubMed
Paez, JG, Janne, PA, Lee, JC, et al. EGFR mutations in lung cancer:correlation with clinical response to gefitinib therapy. Science 2004;304:1497–500.CrossRef
Pao, W, Miller, V, Zakowski, M, et al. EGF receptor gene mutations are common in lung cancers from “never smokers” and are associated with sensitivity of tumors to gefitinib and erlotinib. Proceedings of the National Academy of Sciences USA 2004;101:13 306–11.
Lee, JW, Soung, YH, Seo, SH, et al. Somatic mutations of ERBB2 kinase domain in gastric, colorectal, and breast carcinomas. Clinical Cancer Research 2006;12:57–61.CrossRef
Soung, YH, Lee, JW, Kim, SY, et al. Somatic mutations of the ERBB4 kinase domain in human cancers. International Journal of Cancer 2006;118:1426–9.CrossRefGoogle ScholarPubMed
Sharma, SV, Bell, DW, Settleman, J, Haber, DA. Epidermal growth factor receptor mutations in lung cancer. Nature Reviews Cancer 2007;7:169–81.CrossRef
Stephens, P, Hunter, C, Bignell, G, et al. Lung cancer:intragenic ERBB2 kinase mutations in tumours. Nature 2004;431:525–6.CrossRef
Kwak, EL, Jankowski, J, Thayer, SP, et al. Epidermal growth factor receptor kinase domain mutations in esophageal and pancreatic adenocarcinomas. Clinical Cancer Research 2006;12:4283–7.CrossRef
Okamoto, I, Araki, J, Suto, R, et al. EGFR mutation in gefitinib-responsive small-cell lung cancer. Annals of Oncology 2006;17:1028–9.CrossRef
Gwak, GY, Yoon, JH, Shin, CM, et al. Detection of response-predicting mutations in the kinase domain of the epidermal growth factor receptor gene in cholangiocarcinomas. Journal of Cancer Research and Clinical Oncology 2005;131:649–52.CrossRefGoogle ScholarPubMed
Schilder, RJ, Sill, MW, Chen, X, et al. Phase II study of gefitinib in patients with relapsed or persistent ovarian or primary peritoneal carcinoma and evaluation of epidermal growth factor receptor mutations and immunohistochemical expression: a Gynecologic Oncology Group Study. Clinical Cancer Research 2005;11:5539–48.CrossRef
Barber, TD, Vogelstein, B, Kinzler, KW, Velculescu, VE. Somatic mutations of EGFR in colorectal cancers and glioblastomas. New England Journal of Medicine 2004;351:2883.CrossRefGoogle ScholarPubMed
Lee, JW, Soung, YH, Kim, SY, et al. Somatic mutations of EGFR gene in squamous cell carcinoma of the head and neck. Clinical Cancer Research 2005;11:2879–82.CrossRef
Cai, CQ, Peng, Y, Buckley, MT, et al. Epidermal growth factor receptor activation in prostate cancer by three novel missense mutations. Oncogene 2008;27:3201–10.CrossRef
Greulich, H, Chen, TH, Feng, W, et al. Oncogenic transformation by inhibitor-sensitive and -resistant EGFR mutants. PLoS Medicine 2005;2:e313.
Yun, CH, Boggon, TJ, Li, Y, et al. Structures of lung cancer-derived EGFR mutants and inhibitor complexes: mechanism of activation and insights into differential inhibitor sensitivity. Cancer Cell 2007;11:217–27.CrossRef
Yun, CH, Mengwasser, KE, Toms, AV, et al. The T790M mutation in EGFR kinase causes drug resistance by increasing the affinity for ATP. Proceedings of the National Academy of Sciences USA 2008; 105:2070–5.CrossRef
Politi, K, Zakowski, MF, Fan, PD, et al. Lung adenocarcinomas induced in mice by mutant EGF receptors found in human lung cancers respond to a tyrosine kinase inhibitor or to down-regulation of the receptors. Genes and Development 2006;20:1496–510.CrossRef
Ji, H, Li, D, Chen, L, et al. The impact of human EGFR kinase domain mutations on lung tumorigenesis and in vivo sensitivity to EGFR-targeted therapies. Cancer Cell 2006;9:485–95.CrossRef
Regales, L, Balak, MN, Gong, Y, et al. Development of new mouse lung tumor models expressing EGFR T790M mutants associated with clinical resistance to kinase inhibitors. PLoS One 2007;2:e810.
Li, D, Shimamura, T, Ji, H, et al. Bronchial and peripheral murine lung carcinomas induced by T790M-L858R mutant EGFR respond to HKI-272 and rapamycin combination therapy. Cancer Cell 2007;12:81–93.CrossRef
Carey, KD, Garton, AJ, Romero, MS, et al. Kinetic analysis of epidermal growth factor receptor somatic mutant proteins shows increased sensitivity to the epidermal growth factor receptor tyrosine kinase inhibitor, erlotinib. Cancer Research 2006;66:8163–71.CrossRef
Jiang, J, Greulich, H, Janne, PA, et al. Epidermal growth factor-independent transformation of Ba/F3 cells with cancer-derived epidermal growth factor receptor mutants induces gefitinib-sensitive cell cycle progression. Cancer Research 2005;65:8968–74.CrossRef
Mukohara, T, Engelman, JA, Hanna, NH, et al. Differential effects of gefitinib and cetuximab on non-small-cell lung cancers bearing epidermal growth factor receptor mutations. Journal of the National Cancer Institute 2005;97:1185–94.CrossRefGoogle ScholarPubMed
Bell, DW, Gore, I, Okimoto, RA, et al. Inherited susceptibility to lung cancer may be associated with the T790M drug resistance mutation in EGFR. Nature Genetics 2005;37:1315–16.CrossRef
Pao, W, Miller, VA, Politi, KA, et al. Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Medicine 2005;2:e73.
Landau, M, Fleishman, SJ, Ben-Tal, N. A putative mechanism for downregulation of the catalytic activity of the EGF receptor via direct contact between its kinase and C-terminal domains. Structure 2004;12:2265–75.CrossRef
Zhang, X, Gureasko, J, Shen, K, Cole, PA, Kuriyan, J. An allosteric mechanism for activation of the kinase domain of epidermal growth factor receptor. Cell 2006;125:1137–49.CrossRef
Lee, NY, Hazlett, TL, Koland, JG. Structure and dynamics of the epidermal growth factor receptor C-terminal phosphorylation domain. Protein Science 2006;15:1142–52.CrossRef
Berger, MB, Mendrola, JM, Lemmon, MA. ERBB3/HER3 does not homodimerize upon neuregulin binding at the cell surface. FEBS Letters 2004;569:332–6.CrossRef
Gillgrass, A, Cardiff, RD, Sharan, N, Kannan, S, Muller, WJ. Epidermal growth factor receptor-dependent activation of Gab1 is involved in ERBB-2-mediated mammary tumor progression. Oncogene 2003;22:9151–5.CrossRef
Neckers, L. Heat shock protein 90: the cancer chaperone. Journal of Bioscience 2007;32:517–30.CrossRefGoogle ScholarPubMed
Shigematsu, H, Takahashi, T, Nomura, M, et al. Somatic mutations of the HER2 kinase domain in lung adenocarcinomas. Cancer Research 2005;65:1642–6.CrossRef
Wang, SE, Narasanna, A, Perez-Torres, M, et al. HER2 kinase domain mutation results in constitutive phosphorylation and activation of HER2 and EGFR and resistance to EGFR tyrosine kinase inhibitors. Cancer Cell 2006;10:25–38.CrossRef
Han, SW, Kim, TY, Jeon, YK, et al. Optimization of patient selection for gefitinib in non-small cell lung cancer by combined analysis of epidermal growth factor receptor mutation, K-ras mutation, and Akt phosphorylation. Clinical Cancer Research 2006;12:2538–44.CrossRef
Offterdinger, M, Schofer, C, Weipoltshammer, K, Grunt, TW. c-erbB-3: a nuclear protein in mammary epithelial cells. Journal of Cell Biology 2002;157:929–39.CrossRefGoogle ScholarPubMed
Xie, Y, Hung, MC. Nuclear localization of p185neu tyrosine kinase and its association with transcriptional transactivation. Biochemical and Biophysical Research Communications 1994;203:1589–98.CrossRef
Lin, SY, Makino, K, Xia, W, et al. Nuclear localization of EGF receptor and its potential new role as a transcription factor. Nature Cell Biology 2001;3:802–8.CrossRef
Ni, CY, Murphy, MP, Golde, TE, Carpenter, G. gamma -Secretase cleavage and nuclear localization of ERBB-4 receptor tyrosine kinase. Science 2001;294:2179–81.CrossRef
Wells, A, Marti, U. Signalling shortcuts: cell-surface receptors in the nucleus? Nature Reviews Molecular and Cellular Biology 2002;3:697–702.
Lo, HW, Hsu, SC, Ali-Seyed, M, et al. Nuclear interaction of EGFR and STAT3 in the activation of the iNOS/NO pathway. Cancer Cell 2005;7:575–89.CrossRef
Wang, SC, Lien, HC, Xia, W, et al. Binding at and transactivation of the COX-2 promoter by nuclear tyrosine kinase receptor ERBB-2. Cancer Cell 2004;6:251–61.CrossRef
Wang, SC, Nakajima, Y, Yu, YL, et al. Tyrosine phosphorylation controls PCNA function through protein stability. Nature Cell Biology 2006;8:1359–68.CrossRef
Bandyopadhyay, D, Mandal, M, Adam, L, et al. Physical interaction between epidermal growth factor receptor and DNA-dependent protein kinase in mammalian cells. Journal of Biological Chemistry 1998;273:1568–73.CrossRefGoogle ScholarPubMed
Rodemann, HP, Dittmann, K, Toulany, M. Radiation-induced EGFR-signaling and control of DNA-damage repair. International Journal of Radiation Biology 2007;83:781–91.CrossRefGoogle ScholarPubMed
Klein, C, Gensburger, C, Freyermuth, S, et al. A 120 kDa nuclear phospholipase Cgamma1 protein fragment is stimulated in vivo by EGF signal phosphorylating nuclear membrane EGFR. Biochemistry 2004;43:15 873–83.
Grasl-Kraupp, B, Schausberger, E, Hufnagl, K, et al. A novel mechanism for mitogenic signaling via pro-transforming growth factor alpha within hepatocyte nuclei. Hepatology 2002;35:1372–80.CrossRef
Raper, SE, Burwen, SJ, Barker, ME, Jones, AL. Translocation of epidermal growth factor to the hepatocyte nucleus during rat liver regeneration. Gastroenterology 1987;92:1243–50.CrossRef
Schausberger, E, Eferl, R, Parzefall, W, et al. Induction of DNA synthesis in primary mouse hepatocytes is associated with nuclear pro-transforming growth factor alpha and erbb-1 and is independent of c-jun. Carcinogenesis 2003;24:835–41.CrossRef
Dittmann, K, Mayer, C, Fehrenbacher, B, et al. Radiation-induced epidermal growth factor receptor nuclear import is linked to activation of DNA-dependent protein kinase. Journal of Biological Chemistry 2005;280:31 182–9.CrossRefGoogle ScholarPubMed
Wanner, G, Mayer, C, Kehlbach, R, Rodemann, HP, Dittmann, K. Activation of protein kinase Cepsilon stimulates DNA-repair via epidermal growth factor receptor nuclear accumulation. Radiotherapy and Oncology 2007.
Giri, DK, Ali-Seyed, M, Li, LY, et al. Endosomal transport of ERBB-2: mechanism for nuclear entry of the cell surface receptor. Molecular and Cellular Biology 2005;25:11 005–18.
Lo, HW, Ali-Seyed, M, Wu, Y, et al. Nuclear-cytoplasmic transport of EGFR involves receptor endocytosis, importin beta1 and CRM1. Journal of Cell Biochemistry 2006;98:1570–83.CrossRefGoogle ScholarPubMed
Tsai, B, Ye, Y, Rapoport, TA. Retro-translocation of proteins from the endoplasmic reticulum into the cytosol. Nature Reviews Molecular and Cellular Biology 2002;3:246–55.CrossRef
Liao, HJ, Carpenter, G. Role of the Sec61 translocon in EGF receptor trafficking to the nucleus and gene expression. Molecular Biology of the Cell 2007;18:1064–72.CrossRef
Hsu, SC, Hung, MC. Characterization of a novel tripartite nuclear localization sequence in the EGFR family. Journal of Biological Chemistry 2007;282:10 432–40.CrossRefGoogle ScholarPubMed
Williams, CC, Allison, JG, Vidal, GA, et al. The ERBB4/HER4 receptor tyrosine kinase regulates gene expression by functioning as a STAT5A nuclear chaperone. Journal of Cell Biology 2004;167:469–78.CrossRefGoogle ScholarPubMed
Lo, HW, Xia, W, Wei, Y, et al. Novel prognostic value of nuclear epidermal growth factor receptor in breast cancer. Cancer Research 2005;65:338–48.
Cao, H, Lei, ZM, Bian, L, Rao, CV. Functional nuclear epidermal growth factor receptors in human choriocarcinoma JEG-3 cells and normal human placenta. Endocrinology 1995;136:3163–72.CrossRef
Marti, U, Ruchti, C, Kampf, J, et al. Nuclear localization of epidermal growth factor and epidermal growth factor receptors in human thyroid tissues. Thyroid 2001;11:137–45.CrossRef
Kamio, T, Shigematsu, K, Sou, H, Kawai, K, Tsuchiyama, H. Immunohistochemical expression of epidermal growth factor receptors in human adrenocortical carcinoma. Human Pathology 1990;21:277–82.CrossRef
Psyrri, A, Yu, Z, Weinberger, PM, et al. Quantitative determination of nuclear and cytoplasmic epidermal growth factor receptor expression in oropharyngeal squamous cell cancer by using automated quantitative analysis. Clinical Cancer Research 2005;11:5856–62.CrossRef
Lipponen, P, Eskelinen, M. Expression of epidermal growth factor receptor in bladder cancer as related to established prognostic factors, oncoprotein (c-erbB-2, p53) expression and long-term prognosis. British Journal of Cancer 1994;69:1120–5.CrossRefGoogle ScholarPubMed
Hanada, N, Lo, HW, Day, CP, et al. Co-regulation of B-Myb expression by E2F1 and EGF receptor. Molecular Carcinogenesis 2006;45:10–17.CrossRef
Edwards, J, Traynor, P, Munro, AF, et al. The role of HER1-HER4 and EGFRvIII in hormone-refractory prostate cancer. Clinical Cancer Research 2006;12:123–30.CrossRef
Bonner, JA, Harari, PM, Giralt, J, et al. Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck. New England Journal of Medicine 2006;354:567–78.CrossRefGoogle ScholarPubMed
Burtness, B, Goldwasser, MA, Flood, W, Mattar, B, Forastiere, AA. Phase III randomized trial of cisplatin plus placebo compared with cisplatin plus cetuximab in metastatic/recurrent head and neck cancer: an Eastern Cooperative Oncology Group study. Journal of Clinical Oncology 2005;23:8646–54.CrossRefGoogle ScholarPubMed
Cordero, JB, Cozzolino, M, Lu, Y, et al. 1,25-Dihydroxyvitamin D down-regulates cell membrane growth- and nuclear growth-promoting signals by the epidermal growth factor receptor. Journal of Biological Chemistry 2002;277:38 965–71.CrossRefGoogle ScholarPubMed
Howe, LR, Chang, SH, Tolle, KC, et al. HER2/neu-induced mammary tumorigenesis and angiogenesis are reduced in cyclooxygenase-2 knockout mice. Cancer Research 2005;65:10 113–9.
Endo, K, Yoon, BI, Pairojkul, C, Demetris, AJ, Sirica, AE. ERBB-2 overexpression and cyclooxygenase-2 up-regulation in human cholangiocarcinoma and risk conditions. Hepatology 2002;36:439–50.CrossRef
Vadlamudi, R, Mandal, M, Adam, L, et al. Regulation of cyclooxygenase-2 pathway by HER2 receptor. Oncogene 1999;18:305–14.CrossRef
Ristimaki, A, Sivula, A, Lundin, J, et al. Prognostic significance of elevated cyclooxygenase-2 expression in breast cancer. Cancer Research 2002;62:632–5.
Komuro, A, Nagai, M, Navin, NE, Sudol, M. WW domain-containing protein YAP associates with ERBB-4 and acts as a co-transcriptional activator for the carboxyl-terminal fragment of ERBB-4 that translocates to the nucleus. Journal of Biological Chemistry 2003;278:33 334–41.CrossRefGoogle ScholarPubMed
Clark, DE, Williams, CC, Duplessis, TT, et al. ERBB4/HER4 potentiates STAT5A transcriptional activity by regulating novel STAT5A serine phosphorylation events. Journal of Biological Chemistry 2005;280:24 175–80.CrossRefGoogle ScholarPubMed
Muraoka-Cook, RS, Sandahl, M, Husted, C, et al. The intracellular domain of ERBB4 induces differentiation of mammary epithelial cells. Molecular Biology of the Cell 2006;17:4118–29.CrossRef
Sundvall, M, Peri, L, Maatta, JA, et al. Differential nuclear localization and kinase activity of alternative ERBB4 intracellular domains. Oncogene 2007;26:6905–14.CrossRef
Arasada, RR, Carpenter, G. Secretase-dependent tyrosine phosphorylation of Mdm2 by the ERBB-4 intracellular domain fragment. Journal of Biological Chemistry 2005;280:30 783–7.CrossRefGoogle ScholarPubMed
Vidal, GA, Naresh, A, Marrero, L, Jones, FE. Presenilin-dependent gamma-secretase processing regulates multiple ERBB4/HER4 activities. Journal of Biological Chemistry 2005;280:19 777–83.CrossRefGoogle ScholarPubMed
Naresh, A, Long, W, Vidal, GA, et al. The ERBB4/HER4 intracellular domain 4ICD is a BH3-only protein promoting apoptosis of breast cancer cells. Cancer Research 2006;66:6412–20.CrossRef
Linggi, B, Carpenter, G. ERBB-4 s80 intracellular domain abrogates ETO2-dependent transcriptional repression. Journal of Biological Chemistry 2006;281:25 373–80.CrossRefGoogle ScholarPubMed
Junttila, TT, Sundvall, M, Lundin, M, et al. Cleavable ERBB4 isoform in estrogen receptor-regulated growth of breast cancer cells. Cancer Research 2005;65:1384–93.CrossRef
Zhu, Y, Sullivan, LL, Nair, SS, et al. Coregulation of estrogen receptor by ERBB4/HER4 establishes a growth-promoting autocrine signal in breast tumor cells. Cancer Research 2006;66:7991–8.CrossRef
Bacus, SS, Chin, D, Yarden, Y, Zelnick, CR, Stern, DF. Type 1 receptor tyrosine kinases are differentially phosphorylated in mammary carcinoma and differentially associated with steroid receptors. American Journal of Pathology 1996;148:549–58.Google ScholarPubMed
Knowlden, JM, Gee, JM, Seery, LT, et al. c-erbB3 and c-erbB4 expression is a feature of the endocrine responsive phenotype in clinical breast cancer. Oncogene 1998;17:1949–57.CrossRef
Witton, CJ, Reeves, JR, Going, JJ, Cooke, TG, Bartlett, JM. Expression of the HER1–4 family of receptor tyrosine kinases in breast cancer. Journal of Pathology 2003;200:290–7.CrossRefGoogle ScholarPubMed
Srinivasan, R, Gillett, CE, Barnes, DM, Gullick, WJ. Nuclear expression of the c-erbB-4/HER-4 growth factor receptor in invasive breast cancers. Cancer Research 2000;60:1483–7.
Aqeilan, RI, Donati, V, Palamarchuk, A, et al. WW domain-containing proteins, WWOX and YAP, compete for interaction with ERBB-4 and modulate its transcriptional function. Cancer Research 2005;65:6764–72.CrossRef
Dikic, I, Giordano, S. Negative receptor signalling. Current Opinion in Cell Biology 2003;15:128–35.CrossRef
Haj, FG, Verveer, PJ, Squire, A, Neel, BG, Bastiaens, PI. Imaging sites of receptor dephosphorylation by PTP1B on the surface of the endoplasmic reticulum. Science 2002;295:1708–11.CrossRef
Berset, TA, Hoier, EF, Hajnal, A. The C. elegans homolog of the mammalian tumor suppressor Dep-1/Scc1 inhibits EGFR signaling to regulate binary cell fate decisions. Genes and Development 2005;19:1328–40.CrossRef
Tonks, NK. Protein tyrosine phosphatases: from genes, to function, to disease. Nature Reviews Molecular and Cellular Biology 2006;7:833–46.CrossRef
Ostman, A, Hellberg, C, Bohmer, FD. Protein-tyrosine phosphatases and cancer. Nature Reviews Cancer 2006;6:307–20.CrossRef
Stuible, M, Doody, KM, Tremblay, ML. PTP1B and TC-PTP: regulators of transformation and tumorigenesis. Cancer Metastasis Reviews 2008;27:215–30.CrossRef
Julien, SG, Dube, N, Read, M, et al. Protein tyrosine phosphatase 1B deficiency or inhibition delays ERBB2-induced mammary tumorigenesis and protects from lung metastasis. Nature Genetics 2007;39:338–46.CrossRef
Zhu, JH, Chen, R, Yi, W, et al. Protein tyrosine phosphatase PTPN13 negatively regulates Her2/ErbB2 malignant signaling. Oncogene 2008;27:2525–31.CrossRef
Gensler, M, Buschbeck, M, Ullrich, A. Negative regulation of HER2 signaling by the PEST-type protein-tyrosine phosphatase BDP1. Journal of Biological Chemistry 2004;279:12 110–6.CrossRefGoogle ScholarPubMed
Stoscheck, CM, Carpenter, G. Down regulation of epidermal growth factor receptors:direct demonstration of receptor degradation in human fibroblasts. Journal of Cell Biology 1984;98:1048–53.CrossRefGoogle ScholarPubMed
Gur, G, Zwang, Y, Yarden, Y. Endocytosis of receptor tyrosine kinases: implications for signal transduction by growth factors. In: Dikic I, editor. Endosomes. Austin, TX: Eurekah, Landes Bioscience;2006.
Schmidt, MH, Dikic, I. The Cbl interactome and its functions. Nature Reviews Molecular and Cellular Biology 2005;6:907–18.CrossRef
Miaczynska, M, Pelkmans, L, Zerial, M. Not just a sink: endosomes in control of signal transduction. Current Opinion in Cell Biology 2004;16:400–6.CrossRef
Sorkin, A, Von Zastrow, M. Signal transduction and endocytosis: close encounters of many kinds. Nature Reviews Molecular and Cellular Biology 2002;3:600–14.CrossRef
Burke, P, Schooler, K, Wiley, HS. Regulation of epidermal growth factor receptor signaling by endocytosis and intracellular trafficking. Molecular Biology of the Cell 2001;12:1897–910.CrossRef
Vieira, AV, Lamaze, C, Schmid, SL. Control of EGF receptor signaling by clathrin-mediated endocytosis. Science 1996;274:2086–9.CrossRef
Wiley, HS. Trafficking of the ERBB receptors and its influence on signaling. Experimental Cell Research 2003;284:78–88.CrossRef
Parton, RG, Simons, K. The multiple faces of caveolae. Nature Reviews Molecular and Cellular Biology 2007;8:185–94.CrossRef
Couet, J, Sargiacomo, M, Lisanti, MP. Interaction of a receptor tyrosine kinase, EGF-R, with caveolins. Caveolin binding negatively regulates tyrosine and serine/threonine kinase activities. Journal of Biological Chemistry 1997;272:30 429–38.CrossRefGoogle ScholarPubMed
Sigismund, S, Woelk, T, Puri, C, et al. Clathrin-independent endocytosis of ubiquitinated cargos. Proceedings of the National Academy of Sciences USA 2005;102:2760–5.CrossRef
Levkowitz, G, Waterman, H, Ettenberg, SA, et al. Ubiquitin ligase activity and tyrosine phosphorylation underlie suppression of growth factor signaling by c-Cbl/Sli-1. Molecular Cell 1999;4:1029–40.CrossRef
Waterman, H, Katz, M, Rubin, C, et al. A mutant EGF-receptor defective in ubiquitylation and endocytosis unveils a role for Grb2 in negative signaling. EMBO Journal 2002;21:303–13.CrossRef
Klapper, LN, Waterman, H, Sela, M, Yarden, Y. Tumor-inhibitory antibodies to HER-2/ERBB-2 may act by recruiting c-Cbl and enhancing ubiquitination of HER-2. Cancer Research 2000;60:3384–8.
Jiang, X, Huang, F, Marusyk, A, Sorkin, A. Grb2 regulates internalization of EGF receptors through clathrin-coated pits. Molecular Biology of the Cell 2003;14:858–70.CrossRef
Mosesson, Y, Shtiegman, K, Katz, M, et al. Endocytosis of receptor tyrosine kinases is driven by monoubiquitylation, not polyubiquitylation. Journal of Biological Chemistry 2003;278:21 323–6.CrossRefGoogle Scholar
Haglund, K, Sigismund, S, Polo, S, et al. Multiple monoubiquitination of RTKs is sufficient for their endocytosis and degradation. Nature Cell Biology 2003;5:461–6.CrossRef
Levkowitz, G, Waterman, H, Zamir, E, et al. c-Cbl/Sli-1 regulates endocytic sorting and ubiquitination of the epidermal growth factor receptor. Genes and Development 1998;12:3663–74.CrossRef
de Melker, AA, van der Horst, G, Calafat, J, Jansen, H, Borst, J. c-Cbl ubiquitinates the EGF receptor at the plasma membrane and remains receptor associated throughout the endocytic route. Journal of Cell Science 2001;114:2167–78.Google ScholarPubMed
Longva, KE, Blystad, FD, Stang, E, et al. Ubiquitination and proteasomal activity is required for transport of the EGF receptor to inner membranes of multivesicular bodies. Journal of Cell Biology 2002;156:843–54.CrossRefGoogle ScholarPubMed
Schmidt, A, Wolde, M, Thiele, C, et al. Endophilin I mediates synaptic vesicle formation by transfer of arachidonate to lysophosphatidic acid. Nature 1999;401:133–41.CrossRef
Petrelli, A, Gilestro, GF, Lanzardo, S, et al. The endophilin-CIN85-Cbl complex mediates ligand-dependent downregulation of c-Met. Nature 2002;416:187–90.CrossRef
Soubeyran, P, Kowanetz, K, Szymkiewicz, I, Langdon, WY, Dikic, I. Cbl-CIN85-endophilin complex mediates ligand-induced downregulation of EGF receptors. Nature 2002;416:183–7.CrossRef
Haglund, K, Shimokawa, N, Szymkiewicz, I, Dikic, I. Cbl-directed monoubiquitination of CIN85 is involved in regulation of ligand-induced degradation of EGF receptors. Proceedings of the National Academy of Sciences USA 2002;99:12 191–6.
Ettenberg, SA, Magnifico, A, Cuello, M, et al. Cbl-b-dependent coordinated degradation of the epidermal growth factor receptor signaling complex. Journal of Biological Chemistry 2001;276:27 677–84.CrossRefGoogle ScholarPubMed
Yokouchi, M, Kondo, T, Sanjay, A, et al. Src-catalyzed phosphorylation of c-Cbl leads to the interdependent ubiquitination of both proteins. Journal of Biological Chemistry 2001;276:35 185–93.CrossRefGoogle ScholarPubMed
Hopper, NA, Lee, J, Sternberg, PW. ARK-1 inhibits EGFR signaling in C. elegans. Molecular Cell 2000;6:65–75.
Teo, M, Tan, L, Lim, L, Manser, E. The tyrosine kinase ACK1 associates with clathrin-coated vesicles through a binding motif shared by arrestin and other adaptors. Journal of Biological Chemistry 2001;276:18 392–8.CrossRefGoogle ScholarPubMed
Galisteo, ML, Yang, Y, Urena, J, Schlessinger, J. Activation of the nonreceptor protein tyrosine kinase Ack by multiple extracellular stimuli. Proceedings of the National Academy of Sciences USA 2006;103:9796–801.CrossRef
Shen, F, Lin, Q, Gu, Y, Childress, C, Yang, W. Activated Cdc42-associated kinase 1 is a component of EGF receptor signaling complex and regulates EGF receptor degradation. Molecular Biology of the Cell 2007;18:732–42.CrossRef
Grovdal, LM, Johannessen, LE, Rodland, MS, Madshus, IH, Stang, E. Dysregulation of Ack1 inhibits down-regulation of the EGF receptor. Experimental Cell Research 2008;314:1292–300.CrossRef
Courbard, JR, Fiore, F, Adelaide, J, et al. Interaction between two ubiquitin-protein isopeptide ligases of different classes, CBLC and AIP4/ITCH. Journal of Biological Chemistry 2002;277:45 267–75.CrossRefGoogle ScholarPubMed
Zerial, M, McBride, H. Rab proteins as membrane organizers. Nature Reviews Molecular and Cellular Biology 2001;2:107–17.CrossRef
Katz, M, Shtiegman, K, Tal-Or, P, et al. Ligand-independent degradation of epidermal growth factor receptor involves receptor ubiquitylation and Hgs, an adaptor whose ubiquitin-interacting motif targets ubiquitylation by Nedd4. Traffic 2002;3:740–51.CrossRef
Polo, S, Sigismund, S, Faretta, M, et al. A single motif responsible for ubiquitin recognition and monoubiquitination in endocytic proteins. Nature 2002;416:451–5.CrossRef
Raiborg, C, Bache, KG, Gillooly, DJ, et al. Hrs sorts ubiquitinated proteins into clathrin-coated microdomains of early endosomes. Nature Cell Biology 2002;4:394–8.CrossRef
Katzmann, DJ, Babst, M, Emr, SD. Ubiquitin-dependent sorting into the multivesicular body pathway requires the function of a conserved endosomal protein sorting complex, ESCRT-I. Cell 2001;106:145–55.CrossRef
Bache, KG, Brech, A, Mehlum, A, Stenmark, H. Hrs regulates multivesicular body formation via ESCRT recruitment to endosomes. Journal of Cell Biology 2003;162:435–42.CrossRefGoogle ScholarPubMed
Bache, KG, Slagsvold, T, Cabezas, A, et al. The growth-regulatory protein HCRP1/hVps37A is a subunit of mammalian ESCRT-I and mediates receptor down-regulation. Molecular Biology of the Cell 2004;15:4337–46.CrossRef
Doyotte, A, Russell, MR, Hopkins, CR, Woodman, PG. Depletion of TSG101 forms a mammalian “Class E” compartment: a multicisternal early endosome with multiple sorting defects. Journal of Cell Science 2005;118:3003–17.CrossRefGoogle ScholarPubMed
Lu, Q, Hope, LW, Brasch, M, Reinhard, C, Cohen, SN. TSG101 interaction with HRS mediates endosomal trafficking and receptor down-regulation. Proceedings of the National Academy of Sciences USA 2003;100:7626–31.CrossRef
Scott, A, Chung, HY, Gonciarz-Swiatek, M, et al. Structural and mechanistic studies of VPS4 proteins. EMBO Journal 2005;24:3658–69.CrossRef
Ebner, R, Derynck, R. Epidermal growth factor and transforming growth factor-alpha: differential intracellular routing and processing of ligand-receptor complexes. Cell Regulation1991;2:599–612.
French, AR, Tadaki, DK, Niyogi, SK, Lauffenburger, DA. Intracellular trafficking of epidermal growth factor family ligands is directly influenced by the pH sensitivity of the receptor/ligand interaction. Journal of Biological Chemistry 1995;270:4334–40.CrossRefGoogle Scholar
Kochupurakkal, BS, Harari, D, Di-Segni, A, et al. Epigen, the last ligand of ERBB receptors, reveals intricate relationships between affinity and mitogenicity. Journal of Biological Chemistry 2005;280:8503–12.CrossRefGoogle ScholarPubMed
Ravid, T, Sweeney, C, Gee, P, Carraway, KL, 3rd, Goldkorn, T. Epidermal growth factor receptor activation under oxidative stress fails to promote c-Cbl mediated down-regulation. Journal of Biological Chemistry 2002;277:31 214–9.CrossRefGoogle ScholarPubMed
Khan, EM, Lanir, R, Danielson, AR, Goldkorn, T. Epidermal growth factor receptor exposed to cigarette smoke is aberrantly activated and undergoes perinuclear trafficking. FASEB Journal 2008;22:910–17.CrossRef
Oksvold, MP, Thien, CB, Widerberg, J, et al. UV-radiation-induced internalization of the epidermal growth factor receptor requires distinct serine and tyrosine residues in the cytoplasmic carboxy-terminal domain. Radiation Research 2004;161:685–91.CrossRef
Oksvold, MP, Huitfeldt, HS, Ostvold, AC, Skarpen, E. UV induces tyrosine kinase-independent internalisation and endosome arrest of the EGF receptor. Journal of Cell Science 2002;115:793–803.Google ScholarPubMed
He, YY, Huang, JL, Gentry, JB, Chignell, CF. Epidermal growth factor receptor down-regulation induced by UVA in human keratinocytes does not require the receptor kinase activity. Journal of Biological Chemistry 2003;278:42 457–65.CrossRefGoogle Scholar
Levkowitz, G, Klapper, LN, Tzahar, E, et al. Coupling of the c-Cbl protooncogene product to ERBB-1/EGF-receptor but not to other ERBB proteins. Oncogene 1996;12:1117–25.
Lenferink, AE, Pinkas-Kramarski, R, van de Poll, ML, et al. Differential endocytic routing of homo- and hetero-dimeric ERBB tyrosine kinases confers signaling superiority to receptor heterodimers. EMBO Journal 1998;17:3385–97.CrossRef
Worthylake, R, Opresko, LK, Wiley, HS. ERBB-2 amplification inhibits down-regulation and induces constitutive activation of both ERBB-2 and epidermal growth factor receptors. Journal of Biological Chemistry 1999;274:8865–74.CrossRefGoogle ScholarPubMed
Muthuswamy, SK, Gilman, M, Brugge, JS. Controlled dimerization of ERBB receptors provides evidence for differential signaling by homo- and heterodimers. Molecular and Cellular Biology 1999;19:6845–57.CrossRef
Ouyang, X, Gulliford, T, Zhang, H, et al. Association of ERBB2 Ser1113 phosphorylation with epidermal growth factor receptor co-expression and poor prognosis in human breast cancer. Molecular and Cell Biochemistry 2001;218:47–54.CrossRef
Baulida, J, Kraus, MH, Alimandi, M, Di Fiore, PP, Carpenter, G. All ERBB receptors other than the epidermal growth factor receptor are endocytosis impaired. Journal of Biological Chemistry 1996;271:5251–7.Google Scholar
Haslekas, C, Breen, K, Pedersen, KW, et al. The inhibitory effect of ERBB2 on epidermal growth factor-induced formation of clathrin-coated pits correlates with retention of epidermal growth factor receptor-ERBB2 oligomeric complexes at the plasma membrane. Molecular Biology of the Cell 2005;16:5832–42.CrossRef
Shelly, M, Pinkas-Kramarski, R, Guarino, BC, et al. Epiregulin is a potent pan-ERBB ligand that preferentially activates heterodimeric receptor complexes. Journal of Biological Chemistry 1998;273:10 496–505.CrossRefGoogle ScholarPubMed
Hendriks, BS, Opresko, LK, Wiley, HS, Lauffenburger, D. Coregulation of epidermal growth factor receptor/human epidermal growth factor receptor 2 (HER2) levels and locations:quantitative analysis of HER2 overexpression effects. Cancer Research 2003;63:1130–7.
Offterdinger, M, Bastiaens, PI. Prolonged EGFR signaling by ERBB2-mediated sequestration at the plasma membrane. Traffic 2008;9:147–55.CrossRef
Waterman, H, Alroy, I, Strano, S, Seger, R, Yarden, Y. The C-terminus of the kinase-defective neuregulin receptor ERBB-3 confers mitogenic superiority and dictates endocytic routing. EMBO Journal 1999;18:3348–58.CrossRef
French, AR, Sudlow, GP, Wiley, HS, Lauffenburger, DA. Postendocytic trafficking of epidermal growth factor-receptor complexes is mediated through saturable and specific endosomal interactions. Journal of Biological Chemistry 1994;269:15 749–55.Google ScholarPubMed
Shtiegman, K, Kochupurakkal, BS, Zwang, Y, et al. Defective ubiquitinylation of EGFR mutants of lung cancer confers prolonged signaling. Oncogene 2007;26:6968–78.CrossRef
Chen, YR, Fu, YN, Lin, CH, et al. Distinctive activation patterns in constitutively active and gefitinib-sensitive EGFR mutants. Oncogene 2006;25:1205–15.CrossRef
Padron, D, Sato, M, Shay, JW, et al. Epidermal growth factor receptors with tyrosine kinase domain mutations exhibit reduced Cbl association, poor ubiquitylation, and down-regulation but are efficiently internalized. Cancer Research 2007;67:7695–702.CrossRef
Yang, S, Qu, S, Perez-Tores, M, et al. Association with HSP90 inhibits Cbl-mediated down-regulation of mutant epidermal growth factor receptors. Cancer Research 2006;66:6990–7.CrossRef
Sawai, A, Chandarlapaty, S, Greulich, H, et al. Inhibition of Hsp90 down-regulates mutant epidermal growth factor receptor (EGFR) expression and sensitizes EGFR mutant tumors to paclitaxel. Cancer Research 2008;68:589–96.CrossRef
Huang, HS, Nagane, M, Klingbeil, CK, et al. The enhanced tumorigenic activity of a mutant epidermal growth factor receptor common in human cancers is mediated by threshold levels of constitutive tyrosine phosphorylation and unattenuated signaling. Journal of Biological Chemistry 1997;272:2927–35.CrossRefGoogle ScholarPubMed
Schmidt, MH, Furnari, FB, Cavenee, WK, Bogler, O. Epidermal growth factor receptor signaling intensity determines intracellular protein interactions, ubiquitination, and internalization. Proceedings of the National Academy of Sciences USA 2003;100:6505–10.CrossRef
Grandal, MV, Zandi, R, Pedersen, MW, et al. EGFRvIII escapes down-regulation due to impaired internalization and sorting to lysosomes. Carcinogenesis 2007;28:1408–17.CrossRef
Davies, GC, Ryan, PE, Rahman, L, Zajac-Kaye, M, Lipkowitz, S. EGFRvIII undergoes activation-dependent downregulation mediated by the Cbl proteins. Oncogene 2006;25:6497–509.CrossRef
Frederick, L, Wang, XY, Eley, G, James, CD. Diversity and frequency of epidermal growth factor receptor mutations in human glioblastomas. Cancer Research 2000;60:1383–7.
Friedman, LM, Rinon, A, Schechter, B, et al. Synergistic down-regulation of receptor tyrosine kinases by combinations of mAbs:implications for cancer immunotherapy. Proceedings of the National Academy of Sciences USA 2005;102:1915–20.CrossRef
Bao, J, Gur, G, Yarden, Y. Src promotes destruction of c-Cbl:implications for oncogenic synergy between Src and growth factor receptors. Proceedings of the National Academy of Sciences USA 2003;100:2438–43.CrossRef
Wu, WJ, Tu, S, Cerione, RA. Activated Cdc42 sequesters c-Cbl and prevents EGF receptor degradation. Cell 2003;114:715–25.CrossRef
Hirsch, DS, Shen, Y, Wu, WJ. Growth and motility inhibition of breast cancer cells by epidermal growth factor receptor degradation is correlated with inactivation of Cdc42. Cancer Research 2006;66:3523–30.CrossRef
Feng, Q, Baird, D, Peng, X, et al. Cool-1 functions as an essential regulatory node for EGF receptor- and Src-mediated cell growth. Nature Cell Biology 2006;8:945–56.CrossRef
Schmidt, MH, Husnjak, K, Szymkiewicz, I, Haglund, K, Dikic, I. Cbl escapes Cdc42-mediated inhibition by downregulation of the adaptor molecule betaPix. Oncogene 2006;25:3071–8.CrossRef
Rao, DS, Chang, JC, Kumar, PD, et al. Huntingtin interacting protein 1 Is a clathrin coat binding protein required for differentiation of late spermatogenic progenitors. Molecular and Cellular Biology 2001;21:7796–806.CrossRef
Rao, DS, Bradley, SV, Kumar, PD, et al. Altered receptor trafficking in Huntingtin Interacting Protein 1-transformed cells. Cancer Cell 2003;3:471–82.CrossRef
Rao, DS, Hyun, TS, Kumar, PD, et al. Huntingtin-interacting protein 1 is overexpressed in prostate and colon cancer and is critical for cellular survival. Journal of Clinical Investigation 2002;110:351–60.CrossRefGoogle ScholarPubMed
Li, L, Cohen, SN. Tsg101: a novel tumor susceptibility gene isolated by controlled homozygous functional knockout of allelic loci in mammalian cells. Cell 1996;85:319–29.CrossRef
Oh, KB, Stanton, MJ, West, WW, Todd, GL, Wagner, KU. Tsg101 is upregulated in a subset of invasive human breast cancers and its targeted overexpression in transgenic mice reveals weak oncogenic properties for mammary cancer initiation. Oncogene 2007;26:5950–9.CrossRef
Amit, I, Yakir, L, Katz, M, et al. Tal, a Tsg101-specific E3 ubiquitin ligase, regulates receptor endocytosis and retrovirus budding. Genes and Development 2004;18:1737–52.CrossRef
Wakioka, T, Sasaki, A, Kato, R, et al. Spred is a Sprouty-related suppressor of Ras signalling. Nature 2001;412:647–51.CrossRef
Brems, H, Chmara, M, Sahbatou, M, et al. Germline loss-of-function mutations in SPRED1 cause a neurofibromatosis 1-like phenotype. Nature Genetics 2007;39:1120–6.CrossRef
Bundschu, K, Walter, U, Schuh, K. Getting a first clue about SPRED functions. Bioessays 2007;29:897–907.CrossRef
Rawlings, JS, Rennebeck, G, Harrison, SM, Xi, R, Harrison, DA. Two Drosophila suppressors of cytokine signaling (SOCS) differentially regulate JAK and EGFR pathway activities. BMC Cell Biology 2004;5:38.CrossRef
Kario, E, Marmor, MD, Adamsky, K, et al. Suppressors of cytokine signaling 4 and 5 regulate epidermal growth factor receptor signaling. Journal of Biological Chemistry 2005;280:7038–48.CrossRefGoogle ScholarPubMed
Nicholson, SE, Metcalf, D, Sprigg, NS, et al. Suppressor of cytokine signaling (SOCS)-5 is a potential negative regulator of epidermal growth factor signaling. Proceedings of the National Academy of Sciences USA 2005;102:2328–33.CrossRef
Bullock, AN, Rodriguez, MC, Debreczeni, JE, Songyang, Z, Knapp, S. Structure of the SOCS4-ElonginB/C complex reveals a distinct SOCS box interface and the molecular basis for SOCS-dependent EGFR degradation. Structure 2007;15:1493–504.CrossRef
Gur, G, Rubin, C, Katz, M, et al. LRIG1 restricts growth factor signaling by enhancing receptor ubiquitylation and degradation. EMBO Journal 2004;23:3270–81.CrossRef
Laederich, MB, Funes-Duran, M, Yen, L, et al. The leucine-rich repeat protein LRIG1 is a negative regulator of ERBB family receptor tyrosine kinases. Journal of Biological Chemistry 2004;279:47 050–6.CrossRefGoogle ScholarPubMed
Zhang, YW, Vande Woude, GF. Mig-6, signal transduction, stress response and cancer. Cell Cycle 2007;6:507–13.CrossRef
Fiorentino, L, Pertica, C, Fiorini, M, et al. Inhibition of ERBB-2 mitogenic and transforming activity by RALT, a mitogen-induced signal transducer which binds to the ERBB-2 kinase domain. Molecular and Cellular Biology 2000;20:7735–50.CrossRef
Makkinje, A, Quinn, DA, Chen, A, et al. Gene 33/Mig-6, a transcriptionally inducible adapter protein that binds GTP-Cdc42 and activates SAPK/JNK. A potential marker transcript for chronic pathologic conditions, such as diabetic nephropathy. Possible role in the response to persistent stress. Journal of Biological Chemistry 2000;275:17 838–47.CrossRefGoogle Scholar
Zhang, X, Pickin, KA, Bose, R, et al. Inhibition of the EGF receptor by binding of MIG6 to an activating kinase domain interface. Nature 2007;450:741–4.CrossRef
Anastasi, S, Baietti, MF, Frosi, Y, Alema, S, Segatto, O. The evolutionarily conserved EBR module of RALT/MIG6 mediates suppression of the EGFR catalytic activity. Oncogene 2007;26:7833–46.CrossRef
Hackel, PO, Gishizky, M, Ullrich, A. Mig-6 is a negative regulator of the epidermal growth factor receptor signal. Biological Chemistry 2001;382:1649–62.
Anastasi, S, Fiorentino, L, Fiorini, M, et al. Feedback inhibition by RALT controls signal output by the ERBB network. Oncogene 2003;22:4221–34.CrossRef
Ichinose, J, Murata, M, Yanagida, T, Sako, Y. EGF signalling amplification induced by dynamic clustering of EGFR. Biochemical and Biophysical Research Communications 2004;324:1143–9.CrossRef
Ozaki, K, Kadomoto, R, Asato, K, et al. ERK pathway positively regulates the expression of Sprouty genes. Biochemical and Biophysical Research Communications 2001;285:1084–8.CrossRef
Gross, I, Bassit, B, Benezra, M, Licht, JD. Mammalian sprouty proteins inhibit cell growth and differentiation by preventing ras activation. Journal of Biological Chemistry 2001;276:46 460–8.CrossRefGoogle ScholarPubMed
Yusoff, P, Lao, DH, Ong, SH, et al. Sprouty2 inhibits the Ras/MAP kinase pathway by inhibiting the activation of Raf. Journal of Biological Chemistry 2002;277:3195–201.CrossRefGoogle ScholarPubMed
Hanafusa, H, Torii, S, Yasunaga, T, Nishida, E. Sprouty1 and Sprouty2 provide a control mechanism for the Ras/MAPK signalling pathway. Nature Cell Biology 2002;4:850–8.CrossRef
Ozaki, K, Miyazaki, S, Tanimura, S, Kohno, M. Efficient suppression of FGF-2-induced ERK activation by the cooperative interaction among mammalian Sprouty isoforms. Journal of Cell Science 2005;118:5861–71.CrossRefGoogle ScholarPubMed
Kim, HJ, Taylor, LJ, Bar-Sagi, D. Spatial regulation of EGFR signaling by Sprouty2. Current Biology 2007;17:455–61.CrossRef
Sasaki, A, Taketomi, T, Kato, R, et al. Mammalian Sprouty4 suppresses Ras-independent ERK activation by binding to Raf1. Nature Cell Biology 2003;5:427–32.CrossRef
Impagnatiello, MA, Weitzer, S, Gannon, G, et al. Mammalian sprouty-1 and -2 are membrane-anchored phosphoprotein inhibitors of growth factor signaling in endothelial cells. Journal of Cell Biology 2001;152:1087–98.CrossRefGoogle ScholarPubMed
Lim, J, Wong, ES, Ong, SH, et al. Sprouty proteins are targeted to membrane ruffles upon growth factor receptor tyrosine kinase activation. Identification of a novel translocation domain. Journal of Biological Chemistry 2000;275:32 837–45.CrossRefGoogle ScholarPubMed
Lim, J, Yusoff, P, Wong, ES, et al. The cysteine-rich sprouty translocation domain targets mitogen-activated protein kinase inhibitory proteins to phosphatidylinositol 4,5-bisphosphate in plasma membranes. Molecular and Cellular Biology 2002;22:7953–66.CrossRef
Rubin, C, Zwang, Y, Vaisman, N, Ron, D, Yarden, Y. Phosphorylation of carboxyl-terminal tyrosines modulates the specificity of Sprouty-2 inhibition of different signaling pathways. Journal of Biological Chemistry 2005;280:9735–44.CrossRefGoogle ScholarPubMed
Mason, JM, Morrison, DJ, Bassit, B, et al. Tyrosine phosphorylation of Sprouty proteins regulates their ability to inhibit growth factor signaling: a dual feedback loop. Molecular Biology of the Cell 2004;15:2176–88.CrossRef
Wong, ES, Lim, J, Low, BC, Chen, Q, Guy, GR. Evidence for direct interaction between Sprouty and Cbl. Journal of Biological Chemistry 2001;276:5866–75.CrossRefGoogle ScholarPubMed
Egan, JE, Hall, AB, Yatsula, BA, Bar-Sagi, D. The bimodal regulation of epidermal growth factor signaling by human Sprouty proteins. Proceedings of the National Academy of Sciences USA 2002;99:6041–6.CrossRef
Wong, ES, Fong, CW, Lim, J, et al. Sprouty2 attenuates epidermal growth factor receptor ubiquitylation and endocytosis, and consequently enhances Ras/ERK signalling. EMBO Journal 2002;21:4796–808.CrossRef
Rubin, C, Litvak, V, Medvedovsky, H, et al. Sprouty fine-tunes EGF signaling through interlinked positive and negative feedback loops. Current Biology 2003;13:297–307.CrossRef
Fong, CW, Leong, HF, Wong, ES, et al. Tyrosine phosphorylation of Sprouty2 enhances its interaction with c-Cbl and is crucial for its function. Journal of Biological Chemistry 2003;278:33 456–64.CrossRefGoogle ScholarPubMed
Hall, AB, Jura, N, DaSilva, J, et al. hSpry2 is targeted to the ubiquitin-dependent proteasome pathway by c-Cbl. Current Biology 2003;13:308–14.CrossRef
Haglund, K, Schmidt, MH, Wong, ES, Guy, GR, Dikic, I. Sprouty2 acts at the Cbl/CIN85 interface to inhibit epidermal growth factor receptor downregulation. EMBO Reports 2005;6:635–41.CrossRef
Edwin, F, Singh, R, Endersby, R, Baker, SJ, Patel, TB. The tumor suppressor PTEN is necessary for human Sprouty 2-mediated inhibition of cell proliferation. Journal of Biological Chemistry 2006;281:4816–22.CrossRefGoogle ScholarPubMed
Jensen, KB, Watt, FM. Single-cell expression profiling of human epidermal stem and transit-amplifying cells: Lrig1 is a regulator of stem cell quiescence. Proceedings of the National Academy of Sciences USA 2006;103:11 958–63.
Suzuki, Y, Miura, H, Tanemura, A, et al. Targeted disruption of LIG-1 gene results in psoriasiform epidermal hyperplasia. FEBS Letters 2002;521:67–71.CrossRef
Ledda F, Bieraugel O, Fard SS, Vilar M, Paratcha G. Lrig1 is an endogenous inhibitor of Ret receptor tyrosine kinase activation, downstream signaling, and biological responses to GDNF. Journal Neuroscience 2008;28:39–49.CrossRef
Shattuck, DL, Miller, JK, Laederich, M, et al. LRIG1 is a novel negative regulator of the Met receptor and opposes Met and Her2 synergy. Molecular and Cellular Biology 2007;27:1934–46.CrossRef
Thomasson, M, Hedman, H, Guo, D, Ljungberg, B, Henriksson, R. LRIG1 and epidermal growth factor receptor in renal cell carcinoma:a quantitative RT–PCR and immunohistochemical analysis. British Journal of Cancer 2003;89:1285–9.CrossRefGoogle ScholarPubMed
Ljuslinder, I, Malmer, B, Golovleva, I, et al. Increased copy number at 3p14 in breast cancer. Breast Cancer Research 2005;7:R719–27.
Lindström, AK, Ekman, K, Stendahl, U, et al. LRIG1 and squamous epithelial uterine cervical cancer: correlation to prognosis, other tumor markers, sex steroid hormones, and smoking. International Journal of Gynecological Cancer 2008;18:312–17.CrossRefGoogle ScholarPubMed
Ljuslinder, I, Golovleva, I, Palmqvist, R, et al. LRIG1 expression in colorectal cancer. Acta Oncologica 2007;46:1118–22.CrossRef
Tanemura, A, Nagasawa, T, Inui, S, Itami, S. LRIG-1 provides a novel prognostic predictor in squamous cell carcinoma of the skin:immunohistochemical analysis for 38 cases. Dermatologic Surgery 2005;31:423–30.CrossRef
Hedman, H, Henriksson, R. LRIG inhibitors of growth factor signalling – double-edged swords in human cancer?European Journal of Cancer 2007;43:676–82.CrossRefGoogle ScholarPubMed
Zhang, YW, Staal, B, Su, Y, et al. Evidence that MIG-6 is a tumor-suppressor gene. Oncogene 2007;26:269–76.CrossRef
Farabegoli, F, Ceccarelli, C, Santini, D, et al. Chromosome 1 aneusomy with 1p36 under-representation is related to histologic grade, DNA aneuploidy, high c-erb B-2 and loss of bcl-2 expression in ductal breast carcinoma. International Journal of Cancer 1996;69:381–5.3.0.CO;2-1>CrossRefGoogle ScholarPubMed
Anastasi, S, Sala, G, Huiping, C, et al. Loss of RALT/MIG-6 expression in ERBB2-amplified breast carcinomas enhances ERBB-2 oncogenic potency and favors resistance to Herceptin. Oncogene 2005;24:4540–8.CrossRef
Ferby, I, Reschke, M, Kudlacek, O, et al. Mig6 is a negative regulator of EGF receptor-mediated skin morphogenesis and tumor formation. Nature Medicine 2006;12:568–73.CrossRef
Amatschek, S, Koenig, U, Auer, H, et al. Tissue-wide expression profiling using cDNA subtraction and microarrays to identify tumor-specific genes. Cancer Research 2004;64:844–56.CrossRef
Jin, N, Gilbert, JL, Broaddus, RR, Demayo, FJ, Jeong, JW. Generation of a Mig-6 conditional null allele. Genesis 2007;45:716–21.CrossRef
Gross, I, Morrison, DJ, Hyink, DP, et al. The receptor tyrosine kinase regulator Sprouty1 is a target of the tumor suppressor WT1 and important for kidney development. Journal of Biological Chemistry 2003;278:41 420–30.CrossRefGoogle ScholarPubMed
Lo, TL, Yusoff, P, Fong, CW, et al. The ras/mitogen-activated protein kinase pathway inhibitor and likely tumor suppressor proteins, sprouty 1 and sprouty 2 are deregulated in breast cancer. Cancer Research 2004;64:6127–36.CrossRef
Kwabi-Addo, B, Wang, J, Erdem, H, et al. The expression of Sprouty1, an inhibitor of fibroblast growth factor signal transduction, is decreased in human prostate cancer. Cancer Research 2004;64:4728–35.CrossRef
Lee, CC, Putnam, AJ, Miranti, CK, et al. Overexpression of sprouty 2 inhibits HGF/SF-mediated cell growth, invasion, migration, and cytokinesis. Oncogene 2004;23:5193–202.CrossRef
Fong, CW, Chua, MS, McKie, AB, et al. Sprouty 2, an inhibitor of mitogen-activated protein kinase signaling, is down-regulated in hepatocellular carcinoma. Cancer Research 2006;66:2048–58.CrossRef
Lee, SA, Ho, C, Roy, R, et al. Integration of genomic analysis and in vivo transfection to identify sprouty 2 as a candidate tumor suppressor in liver cancer. Hepatology 2008;47:1200–10.
Fritzsche, S, Kenzelmann, M, Hoffmann, MJ, et al. Concomitant down-regulation of SPRY1 and SPRY2 in prostate carcinoma. Endocrine-Related Cancer 2006;13:839–49.CrossRef
McKie, AB, Douglas, DA, Olijslagers, S, et al. Epigenetic inactivation of the human sprouty2 (hSPRY2) homologue in prostate cancer. Oncogene 2005;24:2166–74.CrossRef
Wang, J, Thompson, B, Ren, C, Ittmann, M, Kwabi-Addo, B. Sprouty4, a suppressor of tumor cell motility, is down regulated by DNA methylation in human prostate cancer. Prostate 2006;66:613–24.CrossRef
Tsavachidou, D, Coleman, ML, Athanasiadis, G, et al. SPRY2 is an inhibitor of the ras/extracellular signal-regulated kinase pathway in melanocytes and melanoma cells with wild-type BRAF but not with the V599E mutant. Cancer Research 2004;64:5556–9.CrossRef
Bloethner, S, Chen, B, Hemminki, K, et al. Effect of common B-RAF and N-RAS mutations on global gene expression in melanoma cell lines. Carcinogenesis 2005;26:1224–32.CrossRef
Shaw, AT, Meissner, A, Dowdle, JA, et al. Sprouty-2 regulates oncogenic K-ras in lung development and tumorigenesis. Genes and Development 2007;21:694–707.CrossRef
Sutterluty, H, Mayer, CE, Setinek, U, et al. Down-regulation of Sprouty2 in non-small cell lung cancer contributes to tumor malignancy via extracellular signal-regulated kinase pathway-dependent and -independent mechanisms. Molecular Cancer Research 2007;5:509–20.CrossRef
Nielsen, TO, West, RB, Linn, SC, et al. Molecular characterisation of soft tissue tumours:a gene expression study. Lancet 2002;359:1301–7.CrossRef
Frolov, A, Chahwan, S, Ochs, M, et al. Response markers and the molecular mechanisms of action of Gleevec in gastrointestinal stromal tumors. Molecular Cancer Therapeutics 2003;2:699–709.
Weinstein, IB. Cancer. Addiction to oncogenes: the Achilles heal of cancer. Science 2002;297:63–4.CrossRef
Reznik, TE, Sang, Y, Ma, Y, et al. Transcription-dependent epidermal growth factor receptor activation by hepatocyte growth factor. Molecular Cancer Research 2008;6:139–50.CrossRef
Mellinghoff, IK, Wang, MY, Vivanco, I, et al. Molecular determinants of the response of glioblastomas to EGFR kinase inhibitors. New England Journal of Medicine 2005;353:2012–24.CrossRefGoogle ScholarPubMed
Khambata-Ford, S, Garrett, CR, Meropol, NJ, et al. Expression of epiregulin and amphiregulin and K-ras mutation status predict disease control in metastatic colorectal cancer patients treated with cetuximab. Journal of Clinical Oncology 2007;25:3230–7.CrossRefGoogle ScholarPubMed
Nagata, Y, Lan, KH, Zhou, X, et al. PTEN activation contributes to tumor inhibition by trastuzumab, and loss of PTEN predicts trastuzumab resistance in patients. Cancer Cell 2004;6:117–27.CrossRef
Berns, K, Horlings, HM, Hennessy, BT, et al. A functional genetic approach identifies the PI3K pathway as a major determinant of trastuzumab resistance in breast cancer. Cancer Cell 2007;12:395–402.CrossRef
Lu, Y, Zi, X, Zhao, Y, Mascarenhas, D, Pollak, M. Insulin-like growth factor-I receptor signaling and resistance to trastuzumab (Herceptin). Journal of the National Cancer Institute 2001;93:1852–7.CrossRefGoogle Scholar
Xing, D, Orsulic, S. Modeling resistance to pathway-targeted therapy in ovarian cancer. Cell Cycle 2005;4:1004–6.CrossRef
Guo, A, Villen, J, Kornhauser, J, et al. Signaling networks assembled by oncogenic EGFR and c-Met. Proceedings of the National Academy of Sciences USA 2008;105:692–7.CrossRef
Araujo, RP, Liotta, LA, Petricoin, EF. Proteins, drug targets and the mechanisms they control:the simple truth about complex networks. Nature Reviews Drug Discovery 2007;6:871–80.CrossRef
Hubbard, SR, Miller, WT. Receptor tyrosine kinases: mechanisms of activation and signaling. Current Opinion in Cell Biology 2007;19:117–23.CrossRef

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

  • HER
    • By Wolfgang J. Köstler, Department of Biological Regulation, The Weizmann Institute of Science, Rehovot, Israel, Yosef Yarden, Department of Biological Regulation, The Weizmann Institute of Science, Rehovot, Israel
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.011
Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

  • HER
    • By Wolfgang J. Köstler, Department of Biological Regulation, The Weizmann Institute of Science, Rehovot, Israel, Yosef Yarden, Department of Biological Regulation, The Weizmann Institute of Science, Rehovot, Israel
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.011
Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

  • HER
    • By Wolfgang J. Köstler, Department of Biological Regulation, The Weizmann Institute of Science, Rehovot, Israel, Yosef Yarden, Department of Biological Regulation, The Weizmann Institute of Science, Rehovot, Israel
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.011
Available formats
×