Her2/Neu Induces All-Trans Retinoic Acid (ATRA) Resistance in Breast Cancer Cells

Total Page:16

File Type:pdf, Size:1020Kb

Her2/Neu Induces All-Trans Retinoic Acid (ATRA) Resistance in Breast Cancer Cells Oncogene (2002) 21, 5224 – 5232 ª 2002 Nature Publishing Group All rights reserved 0950 – 9232/02 $25.00 www.nature.com/onc Her2/neu induces all-trans retinoic acid (ATRA) resistance in breast cancer cells Ana M Tari*,1,5, Soo-Jeong Lim1,4,5, Mien-Chie Hung2, Francisco J Esteva3 and Gabriel Lopez-Berestein1 1Department of Bioimmunotherapy, Section of Immunobiology and Drug Carriers, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, TX 77030, USA; 2Department of Molecular and Cellular Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, TX 77030, USA; 3Department of Breast Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, TX 77030, USA We observed that all-trans retinoic acid (ATRA) Introduction inhibited the growth of MCF-7 breast cancer cells, but not those transfected with HER2/NEU or its transacti- All-trans retinoic acid (ATRA) inhibits the growth of vating ligand HEREGULIN. This suggests that Her2/ various cell types. ATRA mediates its growth neu causes breast cancer cells to be resistant to the inhibitory effects via the retinoic acid receptors growth inhibitory effects of ATRA. To confirm this (RARs). Upon binding to ATRA, RARs form observation, MDA-MB-453 and BT-474 cells, which heterodimers with retinoic X receptors, bind to retinoic have high levels of Her2/neu and are resistant to ATRA, acid response elements found in the promoters of were incubated with the trastuzumab (HerceptinTM) target genes, and modulate gene transcription antibody so that we could determine whether inhibition (Mangelsdorf et al., 1994). Many human breast cancer of the expression and function of Her2/neu would cell lines are sensitive to the inhibitory actions of resensitize these cells to ATRA. Indeed, we found that ATRA, but there are breast cancer cell lines that are MDA-MB-453 and BT-474 cells treated with trastuzu- resistant to ATRA (Lotan, 1979; Fraker et al., 1984; mab were growth inhibitory by ATRA. We then Fontana, 1987). ATRA-resistant breast cancer cell lines determined whether Her2/neu uses Grb2 and Akt are generally negative for the expression of estrogen proteins to induce ATRA resistance. Liposome-incorpo- receptor-a (ERa) and have low levels of RARs, rated Grb2 antisense oligonucleotides (L-Grb2) and a particularly RARa (Sheikh et al., 1994; van der Leede dominant negative (DN) AKT mutant were used to et al., 1995; Fitzgerald et al., 1997; Schneider et al., down-regulate Grb2 expression and inhibit Akt activity, 2000). respectively. When incubated with L-Grb2 or transfected Amplification of HER2/NEU (also known as with the DN AKT mutant, ATRA-resistant, Her2/neu- ERBB2) is correlated with poor prognosis and survival overexpressing cells became sensitive to ATRA. Our outcome in patients with breast cancer (Slamon et al., results indicate that Her2/neu utilizes Grb2 and Akt 1987). This is partly because Her2/neu induces breast proteins to induce ATRA resistance in breast cancer cancer cells resistance to chemotherapeutic and cells. ATRA sensitivity was also correlated with RARa biological agents, including tumor necrosis factor-a, protein levels since higher RARa protein levels were tamoxifen, cisplatin, doxorubicin, and paclitaxel observed in cells in which the Her2/neu pathway was (Hudziak et al., 1989; Benz et al., 1992; Pietras et al., inhibited. 1994; Yu et al., 1996; Baselga et al., 1998). While Oncogene (2002) 21, 5224 – 5232. doi:10.1038/sj.onc. comparing the characteristics of ATRA-sensitive and 1205660 ATRA-resistant breast cancer cell lines, we observed that many ATRA-resistant cell lines, with the excep- Keywords: HER2/neu; ATRA; Grb2; Akt tion of SKBr3 cells, have moderate to high expression levels of the Her2/neu protein (Sheikh et al., 1993; Rishi et al., 1996; De Luca et al., 1997). We hypothesized that Her2/neu induces ATRA resistance in breast cancer cells. Here we report that transfection of HER2/NEU in ATRA-sensitive breast cancer cells caused cells to become ATRA-resistant. Inhibition of TM *Correspondence: AM Tari, Department of Bioimmunotherapy, Box Her2/neu by the trastuzumab (Herceptin ) antibody 422, The University of Texas M.D. Anderson Cancer Center, 1515 sensitized Her2/neu-overexpressing cells to ATRA. Holcombe Boulevard, Houston, TX 77030, USA Inhibition of the growth factor receptor bound 4 Current address: Division of Basic Science, National Cancer Center, protein-2 (Grb2) and Akt, two well-known down- 809 Madu 1-Dong, ILsan-gu, Goyang-si, Gyeonggi-do, Korea 5The first two authors contributed equally to this study stream signaling proteins of Her2/neu, also sensitized Received 7 December 2001; revised 19 April 2002; accepted 10 Her2/neu-overexpressing cells to ATRA. Furthermore, May 2002 we could correlate inhibition of the Her2/neu signaling Her2/neu induces ATRA resistance AM Tari et al 5225 pathway with increased ATRA sensitivity and sensitivity of MCF-7 cells to ATRA by heregulin was increased RARa protein levels. observed when cells were incubated with up to 250 nM ATRA (Figure 2). However, heregulin did not affect the sensitivity of MCF-7 cells to ATRA when the cells were Results incubated with higher doses of ATRA. At 1 mM concentration, ATRA reduced the growth of MCF-7 Her2/neu and heregulin induced ATRA resistance in cells by 71% in the absence of heregulin, and 55% in the breast cancer cells presence of heregulin (P40.05). Thus, in the presence of To determine whether Her2/neu could induce ATRA heregulin, MCF-7 cells became less sensitive to low doses resistance in breast cancer cells, the sensitivity of of ATRA. Heregulin also reduced the sensitivity of parental MCF-7 cells to the growth inhibitory effects another breast cancer cell line, T-47D, to ATRA (data of ATRA was compared with those of MCF-7 cells not shown). These data indicate that overexpression or transfected with HER2/NEU or HEREGULIN, since activation of Her2/neu led to ATRA resistance in breast Her2/neu is known to be transactivated by heregulin cancer cells. (Lupu et al., 1992; Peles et al., 1992; Plowman et al., 1993; Sliwkowski et al., 1994). We chose MCF-7 cells Reduced RARa protein levels were found in MCF-7 cells because these cells are sensitive to ATRA, have very transfected with Her2/neu or heregulin low levels of Her2/neu (Figure 1a, inset), and due to their ease of transfection, have been used as a model to Since the sensitivity of breast cancer cells to ATRA has study the function of various genes in breast cancer been correlated with RARa expression (Sheikh et al., cells (Kern et al., 1994). As expected, MCF-7 cells 1994; van der Leede et al., 1995; Fitzgerald et al., 1997; stably transfected with either the HER2/NEU gene Schneider et al., 2000), we examined the expression of (MCF-7/HER2) or the HEREGULIN gene (MCF-7/ RARs in MCF-7 parental cells and its variants. HRG) express very high levels of the respective Compared with MCF-7/WT cells, MCF-7/HRG and transfected proteins (Figure 1a, inset). MCF-7/HER2 cells had lower RARa protein expres- After 5 days of treatment with ATRA, the sion (Figure 3). Densitometric data indicated that proliferation of parental MCF-7 cells (MCF-7/WT) RARa protein expression levels in the MCF-7/V, was inhibited in a dose-dependent manner (Figure 1a). MCF-7/HRG and MCF-7/HER2 cells were approxi- At 1 mM concentration, ATRA inhibited MCF-7/WT mately 90, 41 and 63% of the level for MCF-7/WT cell growth by 63%. The sensitivity of vector- cells. It is not clear why two RARa bands were transfected cells (MCF-7/V) to ATRA was similar to detected in MCF-7/HER2 cells. The two RARa bands that of MCF-7/WT cells (data not shown). However, could be phosphorylated and unphosphorylated forms ATRA did not inhibit the proliferation of MCF-7/ of RARa or two different RARa isoforms. The HER2 or MCF-7/HRG cells (Figure 1a). This is expression levels of RARb and RARg proteins were despite the fact that the expression levels of ERa are not modulated in any of the transfected MCF-7 cells similar among MCF-7/WT, MCF-7/HER2 and MCF- (Figure 3), indicating that Her2/neu, when over- 7/HRG cells (Figure 1a, inset). expressed or activated by heregulin, can decrease Since Her2/neu and EGFR are related family endogenous RARa expression. members, we determined whether EGFR could also induce ATRA resistance. The levels of EGFR in MCF- Trastuzumab increased ATRA sensitivity in Her2- 7 cells transfected with vector control (MCF-7/V) are overexpressing cells almost undetectable, whereas the levels of EGFR in MCF-7 cells stably transfected with the EGFR gene The antibody trastuzumab specifically blocks Her2/neu (MCF-7/EGFR cells) are very high (Figure 1b, inset). function and expression (Carter et al., 1992; Sliwkows- The expression levels of ERa in both cell lines are quite ki et al., 1999). Thus we determined whether similar (Figure 1b, inset). At 1 mM concentration, trastuzumab, by blocking Her2/neu signaling, could ATRA inhibited the growth of MCF-7/V cells by sensitize Her2-overexpressing breast cancer cells, such approximately 51% (Figure 1b). In the presence or as MDA-MB-453 and BT-474, to ATRA. In the absence of EGF, ATRA inhibited the growth of MCF- absence of trastuzumab, ATRA did not inhibit the 7/EGFR cells to a very similar extent, approximately growth of MDA-MB-453 (Figure 4a) or BT-474 39 – 43% (Figure 1b). (Figure 4b) cells. When 1 mM trastuzumab was added To avoid the bias of using only one clone of MCF-7/ to MDA-MB-453 cells, growth was inhibited by 56% HER2 and one clone of MCF-7/HRG cells, we decided (Figure 4a).
Recommended publications
  • A Network Model of Early Events in Epidermal Growth Factor Receptor Signaling That Accounts for Combinatorial Complexity Michael L
    BioSystems 83 (2006) 136–151 A network model of early events in epidermal growth factor receptor signaling that accounts for combinatorial complexity Michael L. Blinov, James R. Faeder, Byron Goldstein, William S. Hlavacek ∗ Theoretical Biology and Biophysics Group, Theoretical Division, Los Alamos National Laboratory, Los Alamos, NM 87545, USA Received 8 January 2005; received in revised form 6 May 2005; accepted 21 June 2005 Abstract We consider a model of early events in signaling by the epidermal growth factor (EGF) receptor (EGFR). The model includes EGF, EGFR, the adapter proteins Grb2 and Shc, and the guanine nucleotide exchange factor Sos, which is activated through EGF- induced formation of EGFR–Grb2–Sos and EGFR–Shc–Grb2–Sos assemblies at the plasma membrane. The protein interactions involved in signaling can potentially generate a diversity of protein complexes and phosphoforms; however, this diversity has been largely ignored in models of EGFR signaling. Here, we develop a model that accounts more fully for potential molecular diversity by specifying rules for protein interactions and then using these rules to generate a reaction network that includes all chemical species and reactions implied by the protein interactions. We obtain a model that predicts the dynamics of 356 molecular species, which are connected through 3749 unidirectional reactions. This network model is compared with a previously developed model that includes only 18 chemical species but incorporates the same scope of protein interactions. The predictions of this model are reproduced by the network model, which also yields new predictions. For example, the network model predicts distinct temporal patterns of autophosphorylation for different tyrosine residues of EGFR.
    [Show full text]
  • HER2 Stabilizes EGFR and Itself by Altering Autophosphorylation Patterns in a Manner That Overcomes Regulatory Mechanisms and Pr
    Oncogene (2013) 32, 4169–4180 & 2013 Macmillan Publishers Limited All rights reserved 0950-9232/13 www.nature.com/onc ORIGINAL ARTICLE HER2 stabilizes EGFR and itself by altering autophosphorylation patterns in a manner that overcomes regulatory mechanisms and promotes proliferative and transformation signaling Z Hartman1, H Zhao1 and YM Agazie1,2 One of the causes of breast cancer is overexpression of the human epidermal growth factor receptor 2 (HER2). Enhanced receptor autophosphorylation and resistance to activation-induced downregulation have been suggested as mechanisms for HER2-induced sustained signaling and cell transformation. However, the molecular mechanisms underlying these possibilities remain incompletely understood. In the current report, we present evidence that show that HER2 overexpression does not lead to receptor hyper-autophosphorylation, but alters patterns in a manner that favors receptor stability and sustained signaling. Specifically, HER2 overexpression blocks epidermal growth factor receptor (EGFR) tyrosine phosphorylation on Y1045 and Y1068, the known docking sites of c-Cbl and Grb2, respectively, whereas promoting phosphorylation on Y1173, the known docking site of the Gab adaptor proteins and phospholipase C gamma. Under these conditions, HER2 itself is phosphorylated on Y1221/1222, with no known role, and on Y1248 that corresponds to Y1173 of EGFR. Interestingly, suppressed EGFR autophosphorylation on the Grb2 and c-Cbl-binding sites correlated with receptor stability and sustained signaling, suggesting that HER2 accomplishes these tasks by altering autophosphorylation patterns. In conformity with these findings, mutation of the Grb2-binding site on EGFR (Y1068F–EGFR) conferred resistance to ligand-induced degradation, which in turn induced sustained signaling, and increased cell proliferation and transformation.
    [Show full text]
  • Paxillin: a Crossroad in Pathological Cell Migration Ana María López-Colomé*, Irene Lee-Rivera, Regina Benavides-Hidalgo and Edith López
    López-Colomé et al. Journal of Hematology & Oncology (2017) 10:50 DOI 10.1186/s13045-017-0418-y REVIEW Open Access Paxillin: a crossroad in pathological cell migration Ana María López-Colomé*, Irene Lee-Rivera, Regina Benavides-Hidalgo and Edith López Abstract Paxilllin is a multifunctional and multidomain focal adhesion adapter protein which serves an important scaffolding role at focal adhesions by recruiting structural and signaling molecules involved in cell movement and migration, when phosphorylated on specific Tyr and Ser residues. Upon integrin engagement with extracellular matrix, paxillin is phosphorylated at Tyr31, Tyr118, Ser188, and Ser190, activating numerous signaling cascades which promote cell migration, indicating that the regulation of adhesion dynamics is under the control of a complex display of signaling mechanisms. Among them, paxillin disassembly from focal adhesions induced by extracellular regulated kinase (ERK)- mediated phosphorylation of serines 106, 231, and 290 as well as the binding of the phosphatase PEST to paxillin have been shown to play a key role in cell migration. Paxillin also coordinates the spatiotemporal activation of signaling molecules, including Cdc42, Rac1, and RhoA GTPases, by recruiting GEFs, GAPs, and GITs to focal adhesions. As a major participant in the regulation of cell movement, paxillin plays distinct roles in specific tissues and developmental stages and is involved in immune response, epithelial morphogenesis, and embryonic development. Importantly, paxillin is also an essential player in pathological conditions including oxidative stress, inflammation, endothelial cell barrier dysfunction, and cancer development and metastasis. Keywords: Cancer, Focal adhesions, Cell migration, Signal transduction Background gene transcription, thus acting as a direct link from the Paxillin is a main component of focal adhesions (FAs) and plasma membrane and the cytoskeleton to the nucleus [5].
    [Show full text]
  • Association of Β1 Integrin with Focal Adhesion Kinase and Paxillin In
    The Journal of Neuroscience, May 15, 2000, 20(10):3776–3784 Association of ␤1 Integrin with Focal Adhesion Kinase and Paxillin in Differentiating Schwann Cells Li-Mei Chen, Debora Bailey, and Cristina Fernandez-Valle Department of Molecular Biology and Microbiology, University of Central Florida, Orlando, Florida 32816-2360, and Orlando Regional Healthcare System/Health Research Institute, Orlando, Florida 32806 Schwann cells (SCs) differentiate into a myelinating cell when grin, FAK, and paxillin molecules reside in the insoluble, simultaneously adhering to an axon destined for myelination F-actin-rich fraction of differentiating cocultures. Cytochalasin and basal lamina. We are interested in defining the signaling D, an actin depolymerizing agent, decreases tyrosine phos- pathway activated by basal lamina. Using SC/sensory neuron phorylation of FAK and paxillin and their association with ␤1 (N) cocultures, we identified ␤1 integrin and F-actin as compo- integrin and causes a dose-dependent increase in the abun- nents of a pathway leading to myelin gene expression and dance of insoluble FAK and paxillin complexes. Collectively, our myelination (Fernandez-Valle et al., 1994, 1997). Here, we show work indicates that ␤1 integrin, FAK, paxillin, and fyn kinase that focal adhesion kinase (FAK) and paxillin are constitutively form an actin-associated complex in SCs adhering to basal expressed by SCs contacting axons in the absence of basal lamina in the presence of axons. This complex may be impor- lamina. Tyrosine phosphorylation of FAK and paxillin increases tant for initiating the process of SC differentiation into a myeli- as SCs form basal lamina and differentiate. FAK and paxillin nating cell.
    [Show full text]
  • CELL SIGNALLING EGF Signalling — It’S All in SHC1’S Timing
    RESEARCH HIGHLIGHTS Nature Reviews Molecular Cell Biology | AOP, published online 17 July 2013; doi:10.1038/nrm3630 GETTY CELL SIGNALLING EGF signalling — it’s all in SHC1’s timing When activated, the Tyr kinase EGFR (epidermal included effectors involved in stimulating growth factor receptor) phosphorylates the mitogenic signalling, cluster 2 included the Tyr scaffold protein SHC1. This enables SHC1 to phosphatase PTPN12 and cluster 3 included recruit SH2 (SRC homology 2) domain-containing proteins involved in cytoskeletal re­organization proteins, such as the adaptor GRB2 (growth factor as well as phosphatases and GTPase-activating receptor-bound 2), to trigger the activation of proteins that downregulate RAS–MAPK downstream pathways. Pawson and colleagues signalling. The timing of these SHC1–protein now uncover the true complexity of this system, interactions suggests that SHC1 initially revealing that SHC1 is phosphoryl­ated on both stimulates mitogenic signalling before switching Tyr and Ser/‌Thr residues at specific time points its signalling output to the control of after EGF stimulation; this allows it to interact cytoskeletal architecture and signal reversal. with three discrete clusters of proteins that But, do all of the downstream effects of change the output of EGF signalling over time. SHC1 depend on its interaction with GRB2? To investigate events downstream of The interaction of SHC1 with cluster 1 proteins, EGFR-mediated SHC1 phosphoryl­ation, the but not cluster 2 and cluster 3 proteins, was authors generated cells stably expressing a abolished in GRB2-depleted cells. Further tagged version of the 52 kDa isoform of SHC1. experiments showed that Ser29 phosphorylation They mapped SHC1 phosphorylation sites and is important for the GRB2-independent the time after EGF stimulation at which inter­action of SHC1 with the cluster 2 protein phosphoryl­ation peaks at each site.
    [Show full text]
  • Synergistic Anti-Leukemic Activity of Imatinib in Combination with a Small Molecule Grb2 SH2 Domain Binding Antagonist
    Letters to the Editor 948 12 Choi Y. A fast computation of pairwise sequence alignment scores between a 14 Massova I, Kotra LP, Mobashery S. Structural insight into the binding motifs for protein and a set of single-locus variants of another protein. In: Proceedings of the the calcium ion and the non-catalytic zinc in matrix metalloproteases. Bioorg Med ACM Conference on Bioinformatics, Computational Biology and Biomedicine. ACM: Chem Lett 1998; 8: 853–858. Orlando, FL, USA, 2012; pp 414–417. 15 Bourquin JP, Subramanian A, Langebrake C, Reinhardt D, Bernard O, Ballerini P et al. 13 Dreszer TR, Karolchik D, Zweig AS, Hinrichs AS, Raney BJ, Kuhn RM et al. The UCSC Identification of distinct molecular phenotypes in acute megakaryoblastic Genome Browser database: extensions and updates 2011. Nucleic Acids Res 2012; leukemia by gene expression profiling. Proc Natl Acad Sci USA 2006; 103: 40: D918–D923. 3339–3344. Supplementary Information accompanies this paper on the Leukemia website (http://www.nature.com/leu) Synergistic anti-leukemic activity of imatinib in combination with a small molecule Grb2 SH2 domain binding antagonist Leukemia (2014) 28, 948–951; doi:10.1038/leu.2013.323 combination of these two agents increased apoptosis by 450% (20% vs 36%; Figure 1d), indicating functional complementation of distinct pro-apoptotic effects. Flow cytometry analysis using CellTrace Violet indicated that vehicle-treated K562 cells divided eight times in 72 h. Treatment with 0.25 or 2.5 mM imatinib alone The mechanisms underlying imatinib resistance in chronic resulted in 0.04% and 0.03% of cells reaching the eighth cell myelogenous leukemia (CML) include BCR-ABL gene rearrange- division, respectively, and treatment with 2 mM TB03 alone resulted ment, overexpression of Bcl-Abl tyrosine kinase fusion protein and in 0.08% of cells reaching the eighth cell division (Figure 1e).
    [Show full text]
  • Tyrosine Phosphorylation of Grb2 by Bcr/Abl and Epidermal Growth Factor Receptor: a Novel Regulatory Mechanism for Tyrosine Kinase Signaling
    The EMBO Journal Vol. 20 No. 23 pp. 6793±6804, 2001 Tyrosine phosphorylation of Grb2 by Bcr/Abl and epidermal growth factor receptor: a novel regulatory mechanism for tyrosine kinase signaling Shaoguang Li1,2, Anthony D.Couvillon3,4, 1996; Kouhara et al., 1997) and on non-receptor tyrosine Bradley B.Brasher5 and Richard A.Van Etten1 kinases such as Bcr/Abl and FAK (Pendergast et al., 1993; Puil et al., 1994; Schlaepfer et al., 1994). The SH3 Center for Blood Research, Department of Genetics, Harvard Medical School, Boston, MA 02115 and 3Division of Signal Transduction, domains of Grb2 bind to proline-rich motifs on the guanine Beth Israel-Deaconess Medical Center, Boston, MA, USA nucleotide releasing factor son-of-sevenless (Sos) (Buday and Downward, 1993; Egan et al., 1993; Li et al., 1993; 2Present address: The Jackson Laboratory, Bar Harbor, ME, USA 4Present address: Department of Molecular Physiology and Biophysics, Rozakis-Adcock et al., 1993), which stimulates GTP Vanderbilt University Medical Center, Nashville, TN, USA binding to Ras, leading to the activation of mitogen- 5Present address: Enanta Pharmaceuticals, Watertown, MA, USA activated protein kinase (MAPK) and other signaling 1Corresponding authors pathways (Baltensperger et al., 1993; Gale et al., 1993; e-mail: [email protected] Skolnik et al., 1993a). The product of the Philadelphia chromosome in human Growth factor receptor-binding protein-2 (Grb2) chronic myeloid leukemia (CML), Bcr/Abl, is an onco- plays a key role in signal transduction initiated by genic tyrosine kinase that can transform ®broblasts and Bcr/Abl oncoproteins and growth factors, functioning hematopoietic cells in vitro (Sawyers et al., 1995) and as an adaptor protein through its Src homology 2 and induce CML-like disease in mice (Li et al., 1999).
    [Show full text]
  • Grb2 Is Necessary for EGF-Stimulated EGFR Uptake 1793
    Research Article 1791 Role of Grb2 in EGF-stimulated EGFR internalization Tetsuo Yamazaki1, Kristien Zaal2, Dale Hailey2, John Presley2, Jennifer Lippincott-Schwartz2 and Lawrence E. Samelson1,* 1Laboratory of Cellular and Molecular Biology, Division of Basic Science, National Cancer Institute, Bethesda, MD 20892-4255, USA 2Cell Biology and Metabolism Branch, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA *Author for correspondence (e-mail: [email protected]) Accepted 28 January 2002 Journal of Cell Science 115, 1791-1802 (2002) © The Company of Biologists Ltd Summary Grb2 is an adaptor molecule that couples membrane transferrin uptake, had no effect on EGF-induced receptors such as the epidermal growth factor receptor internalization of EGFR. YFP-tagged Grb2 containing an (EGFR) to intracellular signaling pathways. To gain insight inhibitory mutation in either N- or C-SH3 domain into the trafficking pathways followed by these molecules redistributed to the plasma membrane upon EGF after activation by EGF, we visualized Grb2 and EGFR stimulation, but the macropinocytic structures containing fused to GFP spectral variants in single live cells. In Grb2-YFP and EGFR-CFP did not translocate inward and nonstimulated cells, Grb2-YFP was primarily localized appeared to remain tethered to the plasma membrane. This diffusely in the cytoplasm, whereas EGFR-CFP was found suggested that the Grb2 SH3 domain was responsible for on the plasma membrane and in endocytic structures coupling the membranes containing EGFR with localized in the perinuclear area. Within 1 minute of EGF downstream effectors involved in internalization of these stimulation, Grb2 redistributed to the plasma membrane membranes.
    [Show full text]
  • Stimulation of Growth Factorreceptor Signal Transduction by Activation Of
    Proc. Ntatl. Acadil. Sci. U.SA Vol. 93, pp. 1113-1118, February 1996 Neurobiology Stimulation of growth factor receptor signal transduction by activation of voltage-sensitive calcium channels (epidermal growth factor receptor/Ras/mitogen-activated protein kinase) LAURA B. ROSEN*t AND MICHAEL E. GREENBERG*t§ *Division of Neuroscience, Department of Neurology, Children's Hospital, and tProgram in Biological and Biomedical Sciences and :Department of Neurobiology, Harvard Medical School, Boston, MA t)2115 Comminicated by Gerald D. Fischbach, Harvard Medical School, Boston, MA, October 2, 1995 ABSTRACT To understand the mechanisms by which elec- number of different signaling molecules that associate with trical activity may generate long-term responses in the nervous activated growth factor receptors (15, 16). SH2 domains bind system, we examined how activation of voltage-sensitive calcium to phosphorylated tyrosine residues and adjacent amino acid channels (VSCCs) can stimulate the Ras/mitogen-activated sequences, which determine the specificity of the interaction. protein kinase (MAPK) signaling pathway. Calcium influx One class of signaling molecule that inducibly binds to through L-type VSCCs leads to tyrosine phosphorylation of the growth factor receptors is the adaptor protein, which lacks adaptor protein Shc and its association with the adaptor protein catalytic moieties but mediates protein-protein interactions Grb2, which is bound to the guanine nucleotide exchange factor via modular domains such as SH2 domains. One of the adaptor Sosi. In response to calcium influx, Shc, Grb2, and Sosl induc- proteins that inducibly associates with tyrosine-phosphory- ibly associate with a 180-kDa tyrosine-phosphorylated protein, lated growth factor and cytokine receptors is the SH2/collagen which was determined to be the epidermal growth factor receptor protein (Shc) (17).
    [Show full text]
  • Coexpression of Grb7 with Epidermal Growth Factor Receptor Or Her2/Erbb2 in Human Advanced Esophageal Carcinoma1
    [CANCER RESEARCH 57. 28-31. January 1. 1997] Advances in Brief Coexpression of Grb7 with Epidermal Growth Factor Receptor or Her2/erbB2 in Human Advanced Esophageal Carcinoma1 Shinji Tanaka,2 Masaki Mori, Tsuyoshi Akiyoshi, Youichi Tanaka, Ken-ichi Mafune, Jack R. Wands, and Keizo Sugimachi Department of Surgery. Medical Institute of Bioregulation, Kyushu University, 4546 Tsurumibaru, Beppu 874. Japan [S. T., M. M., 1'.A.]; Department of Surgery Ii, Faculty of Medicine, Kyushu University, Fukuoka, Japan [K. S.]; Department of Surgery, Saitama Cancer Center, Saitama, Japan (Y. T., K. M.]; and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts (JR. WI. Abstract respectively (5, 6). With respect to their possible role in oncogenesis, we have previously found that cellular transformation induced by Growth factor receptors transmit intracellular signals that may be IRS-l requires an interaction with both Grb2 and Grb9, respectively. important in carcisogenesis. The Grb7 protein was recently identified as Thus, these binding motifs of the IRS-l protein have a functional role a substrate of the epidermal growth factor receptor and related Her2/ erbB2 receptor-linked tyrosine kinase activity. The Grb7 gene has been in producing cellular transformation (7). found to be coamplifled with Her2IerbB2 in breast carcinomas. In this The Grb7 is a newly identified SH2-containing protein that binds study, Grb7 expression was studied in 32 human esophageal cancers. A avidly to the activated EGFR (5). Murine Grb7 protein is composed of human Grb7 cDNA encoding for N-terminal amino acids was isolated and a SH2 domain at the carboxyl (C) terminus and PH domain that has found to be 90% homologous to the murine counterpart.
    [Show full text]
  • Composition of Receptor Tyrosine Kinase-Mediated Lipid Micro
    www.nature.com/scientificreports OPEN Composition of receptor tyrosine kinase‑mediated lipid micro‑domains controlled by adaptor protein interaction Arndt Rohwedder1, Sabine Knipp1,2, Lee D. Roberts3 & John E. Ladbury1* Receptor tyrosine kinases (RTKs) are highly regulated, single pass transmembrane proteins, fundamental to cellular function and survival. Aberrancies in regulation lead to corruption of signal transduction and a range of pathological outcomes. Although control mechanisms associated with the receptors and their ligands are well understood, little is known with respect to the impact of lipid/lipid and lipid/protein interactions in the proximal plasma membrane environment. Given that the transmembrane regions of RTKs change in response to extracellular ligand binding, the lipid interactions have important consequences in infuencing signal transduction. Fibroblast growth factor receptor 2 (FGFR2) is a highly regulated RTK, including under basal conditions. Binding of the adaptor protein, growth factor receptor‑bound protein 2 (GRB2) to FGFR2 prevents full activation and recruitment of downstream signalling efector proteins in the absence of extracellular stimulation. Here we demonstrate that the FGFR2‑GRB2 complex is sustained in a defned lipid environment. Dissociation of GRB2 from this complex due to ligand binding, or reduced GRB2 expression, facilitates the dispersion of FGFR2 into detergent‑resistant membrane (DRM) micro‑domains. This modifcation of the plasma membrane proximal to FGFR2 provides a further regulatory checkpoint which controls receptor degradation, recycling and recruitment of intracellular signalling proteins. Difusion through, and localisation within the plasma membrane is important for the control and regulation of receptor tyrosine kinase (RTK) signal output in both the absence and presence of extracellular stimulation 1–5.
    [Show full text]
  • Role of the JAK/STAT Pathway in Cervical Cancer: Its Relationship with HPV E6/E7 Oncoproteins
    cells Review Role of the JAK/STAT Pathway in Cervical Cancer: Its Relationship with HPV E6/E7 Oncoproteins Adriana Gutiérrez-Hoya 1,2 and Isabel Soto-Cruz 1,* 1 Molecular Oncology Laboratory, Cell Differentiation and Cancer Research Unit, FES Zaragoza, National University of Mexico, Batalla 5 de Mayo s/n Col. Ejército de Oriente, Mexico City 09230, Mexico; [email protected] 2 Cátedra CONACYT, CONACYT, Avenida Insurgentes Sur 1582, Col. Crédito Constructor Del. Benito Juárez, Mexico City 03940, Mexico * Correspondence: [email protected]; Tel.: +52-55-56230796 Received: 31 August 2020; Accepted: 12 October 2020; Published: 15 October 2020 Abstract: The janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathway is associated with the regulation of essential cellular mechanisms, such as proliferation, invasion, survival, inflammation, and immunity. Aberrant JAK/STAT signaling contributes to cancer progression and metastatic development. STAT proteins play an essential role in the development of cervical cancer, and the inhibition of the JAK/STAT pathway may be essential for enhancing tumor cell death. Persistent activation of different STATs is present in a variety of cancers, including cervical cancer, and their overactivation may be associated with a poor prognosis and poor overall survival. The oncoproteins E6 and E7 play a critical role in the progression of cervical cancer and may mediate the activation of the JAK/STAT pathway. Inhibition of STAT proteins appears to show promise for establishing new targets in cancer treatment. The present review summarizes the knowledge about the participation of the different components of the JAK/STAT pathway and the participation of the human papillomavirus (HPV) associated with the process of cellular malignancy.
    [Show full text]