Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
1.
Oncogene ; 31(6): 706-15, 2012 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-21725367

RESUMO

Many malignancies show increased expression of the epidermal growth factor (EGF) receptor family member ErbB3 (HER3). ErbB3 binds heregulin ß-1 (HRGß1) and forms a heterodimer with other ErbB family members, such as ErbB2 (HER2) or EGF receptor (EGFR; HER1), enhancing phosphorylation of specific C-terminal tyrosine residues and activation of downstream signaling pathways. ErbB3 contains six YXXM motifs that bind the p85 subunit of phosphoinositide 3 (PI3)-kinase. Previous studies demonstrated that overexpression of ErbB3 in mammary tumor cells can significantly enhance chemotaxis to HRGß1 and overall metastatic potential. We tested the hypothesis that ErbB3-mediated PI3-kinase signaling is critical for heregulin-induced motility, and therefore crucial for ErbB3-mediated invasion, intravasation and metastasis. The tyrosines in the six YXXM motifs on the ErbB3 C-terminus were replaced with phenylalanine. In contrast to overexpression of the wild-type ErbB3, overexpression of the mutant ErbB3 did not enhance chemotaxis towards HRGß1 in vitro or in vivo. We also observed reduced tumor cell motility in the primary tumor by multiphoton microscopy, as well as a dramatically reduced ability of these cells to cross the endothelium and intravasate into the circulation. Moreover, whereas mutation of the ErbB3 C-terminus had no effect on tumor growth, it had a dramatic effect on spontaneous metastatic potential. Treatment with the PI3-kinase inhibitor PIK-75 similarly inhibited motility and invasion in vitro and in vivo. Our results indicate that stimulation of the early metastatic steps of motility and invasion by ErbB3 requires activation of the PI3-kinase pathway by the ErbB3 receptor.


Assuntos
Movimento Celular/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Receptor ErbB-3/metabolismo , Transdução de Sinais/fisiologia , Animais , Sítios de Ligação/genética , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Feminino , Humanos , Hidrazonas/farmacologia , Imunoprecipitação , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos SCID , Microscopia de Fluorescência por Excitação Multifotônica , Mutação , Invasividade Neoplásica , Metástase Neoplásica , Neuregulina-1/metabolismo , Neuregulina-1/farmacologia , Fosfatidilinositol 3-Quinases/genética , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Ligação Proteica , Ratos , Receptor ErbB-3/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Sulfonamidas/farmacologia , Transplante Heterólogo , Tirosina/genética , Tirosina/metabolismo
2.
Oncogene ; 29(11): 1598-610, 2010 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-20010870

RESUMO

ErbB2 amplification and overexpression in breast cancer correlates with aggressive disease and poor prognosis. To find novel ErbB2-interacting proteins, we used stable isotope labeling of amino acids in cell culture followed by peptide affinity pull-downs and identified specific binders using relative quantification by mass spectrometry. Copine-III, a member of a Ca(2+)-dependent phospholipid-binding protein family, was identified as binding to phosphorylated Tyr1248 of ErbB2. In breast cancer cells, Copine-III requires Ca(2+) for binding to the plasma membrane, where it interacts with ErbB2 upon receptor stimulation, an interaction that is dependent on receptor activity. Copine-III also binds receptor of activated C kinase 1 and colocalizes with phosphorylated focal adhesion kinase at the leading edge of migrating cells. Importantly, knockdown of Copine-III in T47D breast cancer cells causes a decrease in Src kinase activation and ErbB2-dependent wound healing. Our data suggest that Copine-III is a novel player in the regulation of ErbB2-dependent cancer cell motility. In primary breast tumors, high CPNE3 RNA levels significantly correlate with ERBB2 amplification. Moreover, in an in situ tissue microarray analysis, we detected differential protein expression of Copine-III in normal versus breast, prostate and ovarian tumors, suggesting a more general role for Copine-III in carcinogenesis.


Assuntos
Neoplasias da Mama/metabolismo , Movimento Celular , Fosfoproteínas/metabolismo , Receptor ErbB-2/metabolismo , Sítios de Ligação/genética , Western Blotting , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Modelos Biológicos , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Fosfoproteínas/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Ligação Proteica , Interferência de RNA , Receptor ErbB-2/genética , Tirosina/genética , Tirosina/metabolismo
3.
Tanzan Health Res Bull ; 7(3): 142-8, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16941939

RESUMO

DNA-based vaccination has emerged as a promising method of immunisation since the first demonstration of this technology. Improving the antibody responses is desirable for the protective efficacy and hence broad application of these vaccines. We examined the immunogenicity of a Plasmodium-based DNA vaccine that was targeted to antigen presenting cells by fusion to CTLA4. Fusion proteins comprising the extra-cellular domain of CTLA4, the hinge, CH2 and CH3 domains of human IgG1 and MSP-1 gene fragments were expressed in COS-7 cells. Three of the secreted proteins containing the mouse homologue of CTLA4 were shown to bind differently to the human B7-1 molecule expressed on THP-1 cells. Competition binding assays for two fusion proteins showed that binding was specific. When C57BL/6 mice were immunized with plasmids encoding the fusion proteins, antibodies against two denatured and one non-denatured MSP-1 gene fragments were successfully induced. The usefulness of this strategy in future studies of immunisaton against human malaria is discussed.


Assuntos
Células Dendríticas/efeitos dos fármacos , Malária/imunologia , Vacinas de DNA/imunologia , Animais , Sequência de Bases , Proteínas de Ligação a DNA , Imunização , Imunoglobulina G , Malária/genética , Camundongos , Modelos Animais
4.
Mol Endocrinol ; 15(12): 2182-96, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11731619

RESUMO

Treatment of HC11 mammary epithelial cells with the lactogenic hormone PRL promotes differentiation and induction of milk protein gene expression via stimulation of the Janus kinase (JAK)/signal transducer and activator of transcription pathway. We have previously shown that autocrine activation of epidermal growth factor (EGF) receptor interferes with normal PRL-induced differentiation. Here we show that PRL activation of JAK2 was dramatically reduced in HC11 cells pretreated with EGF, demonstrating that the target of EGF receptor activation is JAK2 kinase. Using an in-gel protein tyrosine phosphatase (PTP) assay, we observed that the activity of a 125-kDa PTP was up-regulated in HC11 cells in response to EGF. A specific antiserum was used to demonstrate that the 125-kDa PTP was PTP-PEST and to show that EGF treatment of HC11 cells led to an increase in the level of PTP-PEST. In intact HC11 cells, PTP-PEST was constitutively associated with JAK2, and in response to EGF treatment there was an increased level of PTP-PEST in JAK2 complexes. An in vitro phosphatase assay, using PRL-activated JAK2 as the substrate and lysates from HC11 cells as the source of PTP-PEST, revealed that JAK2 could serve as a PTP-PEST substrate. However, in intact cells the regulation of JAK2 by PTP-PEST was complex, since transient overexpression of PTP-PEST had a negligible effect on PRL-induced JAK2 activation. EGF's negative influence on JAK2 activity was blocked by actinomycin D treatment of HC11 cells, suggesting that EGF induced a protein that mediated the effects of PTP-PEST on JAK2. In support of this model, PTP-PEST-containing lysates from EGF-treated HC11 cells dephosphorylated JAK2 to a greater extent than lysates prepared from control cells.


Assuntos
Fator de Crescimento Epidérmico/farmacologia , Regulação da Expressão Gênica , Glândulas Mamárias Animais/fisiologia , Proteínas do Leite , Prolactina/fisiologia , Proteínas Tirosina Fosfatases/fisiologia , Proteínas Proto-Oncogênicas , Animais , Western Blotting , Células Cultivadas , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/fisiologia , Eletroforese em Gel de Poliacrilamida , Feminino , Janus Quinase 2 , Luciferases/análise , Glândulas Mamárias Animais/enzimologia , Camundongos , Testes de Precipitina , Proteína Tirosina Fosfatase não Receptora Tipo 12 , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Proteínas Tirosina Quinases/fisiologia , Fator de Transcrição STAT5 , Transativadores/antagonistas & inibidores , Transativadores/fisiologia
5.
Br J Cancer ; 85(11): 1787-93, 2001 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-11742503

RESUMO

Immunophototherapy of cancer combines the specificity of a monoclonal antibody (MAb) to an overexpressed tumor marker with the phototoxic properties of the conjugated dye. To analyze the potential role of internalisation of the dye on photo-induced cytotoxicity, we compared two target antigens, carcinoembryonic antigen (CEA) that does not internalise and ErbB2 that does. Human ovarian carcinoma SKOv3 cells that express a high level of ErbB2 were transfected with the CEA cDNA. Using FACS analysis, the resulting cell line, SKOv3-CEA-1B9, demonstrated comparable levels of expression of the two target antigens. Aluminium tetrasulfophthalocyanine (AlPcS(4)) was covalently coupled to anti-CEA MAb 35A7, anti-ErbB2 MAb FSP77 and a non-specific MAb PX, via a five-carbon sulfonamide spacer chain (A(1)) at molar ratios ranging from 6 to 9 moles of AlPcS(4) per mole of MAb. The 35A7-(AlPcS(4)A(1))(8) conjugate induced 68% growth inhibition of the SKOv3-CEA-1B9 cell line after a 20 h incubation at 2.50 microg/ml (based on AlPcS(4)A(1) content) following light exposure. However, the FSP77-(AlPcS(4)A(1))(6) conjugate gave a 51% growth inhibition for an AlPcS(4)A(1) concentration as low as 0.04 microg/ml after the same incubation time and exposure to the same light dose. At a 1.25 microg/ml AlPcS(4)A(1) concentration, the FSP77-(AlPcS(4)A(1))(6) conjugate gave a 67% growth inhibition after an incubation time as short as 1 h, reaching a 96% inhibition after an 8 h incubation time. Using an unique cell line that expresses two different target antigens, we demonstrated a clear advantage of an internalising over a non-internalising MAb-dye conjugate in terms of phototoxic efficacy. In vivo evaluation of the photodynamic properties of the conjugates is in progress.


Assuntos
Anticorpos Monoclonais/farmacocinética , Divisão Celular/efeitos dos fármacos , Indóis/farmacocinética , Compostos Organometálicos/farmacocinética , Radiossensibilizantes/farmacocinética , Anticorpos Monoclonais/química , Antígeno Carcinoembrionário/genética , Antígeno Carcinoembrionário/imunologia , Relação Dose-Resposta a Droga , Endocitose , Regulação Neoplásica da Expressão Gênica , Humanos , Indóis/química , Luz , Compostos Organometálicos/química , Plasmídeos/genética , Radiossensibilizantes/química , Receptor ErbB-2/genética , Receptor ErbB-2/imunologia , Fatores de Tempo , Transfecção , Células Tumorais Cultivadas
6.
Oncogene ; 20(39): 5459-65, 2001 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-11571643

RESUMO

The ErbB2 receptor tyrosine kinase (RTK) has been intensely pursued as a cancer therapy target due to its association with breast cancer. In this study we used the HC11 mammary epithelial cell line to develop an orthotopic, ErbB2-driven tumor model for testing efficacy of anti-cancer compounds. HC11 cells were infected with a retrovirus encoding oncogenic NeuT, the rat homolog of ErbB2. Drug-selected populations were introduced into mammary fat pads of Balb/c syngeneic mice cleared of host tissue. The majority of glands injected with HC11-NeuT cells developed mammary tumors which appeared after a 3-4 week latency period and grew rapidly. HC11 cells infected with the control retrovirus showed no tumor growth after injection. Tumor-bearing mice were used to compare the in vivo efficacy of two anti-cancer agents: PKI166, a kinase inhibitor selective for EGF receptor and ErbB2, and Taxol, a microtubule assembly blocker. PKI166 inhibited NeuT-induced mammary tumor growth in a dose-dependent manner and at a dose below the maximum tolerated dose (MTD) was significantly more inhibitory than Taxol at its MTD (57% vs. 25% tumor regression). Importantly, there was a dose-dependent decrease in the phosphotyrosine content of NeuT isolated from PKI166-treated, tumor-bearing mice, providing a mechanistic link between kinase inhibition and its anti-tumor activity. Thus, implantation of genetically manipulated HC11 cells into mammary glands appears to be an excellent model for studying effects of anti-cancer agents in an orthotopic site.


Assuntos
Ensaios de Seleção de Medicamentos Antitumorais/métodos , Genes erbB-2 , Neoplasias Mamárias Experimentais/patologia , Animais , Antineoplásicos/farmacologia , Divisão Celular , Linhagem Celular Transformada , Transformação Celular Neoplásica , Receptores ErbB/antagonistas & inibidores , Feminino , Glândulas Mamárias Animais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Paclitaxel/farmacologia , Pirimidinas/farmacologia , Pirróis/farmacologia , Receptor ErbB-2/antagonistas & inibidores
7.
Endocr Relat Cancer ; 8(3): 151-9, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11566606

RESUMO

ErbB receptor tyrosine kinases (RTKs) and their ligands have important roles in normal development and in human cancer. Among the ErbB receptors only ErbB2 has no direct ligand; however, ErbB2 acts as a co-receptor for the other family members, promoting high affinity ligand binding and enhancement of ligand-induced biological responses. These characteristics demonstrate the central role of ErbB2 in the receptor family, which likely explains why it is involved in the development of many human malignancies, including breast cancer. ErbB RTKs also function as signal integrators, cross-regulating different classes of membrane receptors including receptors of the cytokine family. Cross-regulation of ErbB RTKs and cytokines receptors represents another mechanism for controlling and enhancing tumor cell proliferation.


Assuntos
Neoplasias/metabolismo , Receptor ErbB-2/metabolismo , Transdução de Sinais , Feminino , Humanos , Ligantes , Masculino , Receptores de Citocinas/metabolismo
8.
Ann Oncol ; 12 Suppl 1: S21-2, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11521716

RESUMO

The molecular mechanisms mediating the anti-proliferative effects of the murine anti-HER2 monoclonal antibody (4D5) were investigated in HER2-overexpressing human carcinoma cell lines. Treatment with 4D5 resulted in a dramatic accumulation of BT-474 breast carcinoma cells in G1; concomitant with reduced expression of proteins involved in sequestration of the cyclin E/Cdk2 inhibitor protein p27, increased association of p27 with Cdk2 complexes and Cdk2 inactivation. No equivalent effects were observed in BT-474 cells treated with a control, non-inhibitory HER2 monoclonal antibody (FRP5) or in a HER2-overexpressing cell line insensitive to 4D5 treatment (MKN7 gastric carcinoma cells), confirming the relationship between these molecular changes and 4D5-mediated inhibition of proliferation. Increased p27 expression was also observed in 4D5-treated BT-474 cells; however an antisense approach demonstrated that this increase was not required for Cdk2 inactivation or establishment of the G1 block. These data suggest that 4D5 interferes with HER2 receptor signaling, resulting in downregulation of proteins involved in p27 sequestration. This causes release of p27, allowing binding and inhibition of cyclin E/Cdk2 complexes and inhibition of G1/S progression. This model was confirmed using a second 4D5-sensitive. HER2-overexpressing breast tumor line (SKBR3), and suggests that the dependency of a given tumor cell on elevated HER2-receptor signaling for the maintenance of p27 sequestration proteins may determine the clinical response to treatment with the humanized anti-HER2 monoclonal antibody Herceptin (trastuzumab).


Assuntos
Proteínas de Ciclo Celular/farmacologia , Quinases Ciclina-Dependentes/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Receptor ErbB-2/efeitos dos fármacos , Receptor ErbB-2/metabolismo , Proteínas Supressoras de Tumor/farmacologia , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Inibidor de Quinase Dependente de Ciclina p27 , Humanos , Técnicas In Vitro , Camundongos , Transdução de Sinais/efeitos dos fármacos , Trastuzumab
9.
Ann Oncol ; 12 Suppl 1: S9-13, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11521729

RESUMO

The HER family of receptors has an important role in the network of cell signals controlling cell growth and differentiation. Although the activity of the HER receptor is strictly controlled in normal cells, HER2 receptor overexpression plays a pivotal role in transformation and tumorigenesis. HER2 gene amplification and/or overexpression of the receptor has been detected in subsets of a wide range of human cancers including breast cancer, and is an indicator of poor prognosis. It is proposed that overexpressed HER2 in combination with HER3 causes high activity of cell-signaling networks, thereby resulting in tumor cell proliferation. Thus, the HER2 receptor is an attractive target for new anti-cancer treatments. Monoclonal antibodies directed against the receptor are the most promising of these, and the humanized anti-HER2 monoclonal antibody trastuzumab (Herceptin) has shown significant clinical efficacy in clinical trials. The anti-tumor mechanisms of anti-HER2 monoclonal antibodies are not completely understood. However, some tumor types are not sensitive to trastuzumab, suggesting that the response of a tumor to trastuzumab may not only be dependent on overexpressed HER2, but may also be influenced by other members of the HER receptor family expressed in the tumor cell.


Assuntos
Neoplasias da Mama/metabolismo , Regulação Neoplásica da Expressão Gênica , Genes erbB-2 , Receptor ErbB-2/metabolismo , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Transformação Celular Neoplásica , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Tirosina Quinases/metabolismo , Receptor ErbB-2/efeitos dos fármacos , Receptor ErbB-2/genética , Trastuzumab , Regulação para Cima
10.
J Mammary Gland Biol Neoplasia ; 6(1): 141-50, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11467449

RESUMO

The proto-oncogene c-myc encodes a transcription factor which plays a major role in the regulation of normal cellular proliferation and is aberrantly expressed in many breast tumors. In a normal cell Myc expression levels are tightly regulated being subject to many layers of control. Errantly expressed Myc collaborates with other oncogenes to promote transformation. In this review we will focus on the association between abnormal Myc expression and mammary cancer. In particular, we will discuss the role of Myc as a downstream effector of the ErbB2 receptor tyrosine kinase which is overexpressed and constitutively activate in many mammary tumors. The cooperation between Myc and ErbB2 in transformation will be discussed in relation to clinical studies on Myc in human cancer and with consideration of transgenic models of Myc-induced mammary cancer. Data from our laboratory will be presented showing that deregulated ErbB2 activity strongly stimulates cytoplasmic signaling pathways which in turn impinge on Myc at multiple levels causing its deregulated expression.


Assuntos
Neoplasias Mamárias Animais/metabolismo , Proteínas Proto-Oncogênicas c-myc/fisiologia , Receptor ErbB-2/metabolismo , Animais , Feminino , Humanos , Camundongos , Camundongos Transgênicos , Proto-Oncogene Mas , Transdução de Sinais/fisiologia
11.
Exp Cell Res ; 267(1): 81-7, 2001 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-11412040

RESUMO

ErbB receptor tyrosine kinases are activated by multiple ligands such as epidermal growth factor (EGF) and neuregulins (NRGs), leading to stimulation of intracellular signaling pathways, including the mitogen-activated protein kinase (MAPK) cascade. We show here that Src kinase is essential for rapid EGF- and NRG-induced MAPK activation when the breast carcinoma cell lines T47D and SKBR3 are stimulated with low concentrations of ligand. In the presence of the pharmacological inhibitor CGP77675, which specifically blocks the activity of Src family kinases, ligand-induced MAPK activation was almost completely blocked at 5 min. Although this block was only transient, inactivation of Src suppressed ligand-induced transcription from a MAPK-responsive promoter. At the molecular level, the initial inhibition of MAPK by Src inactivation correlated with impaired ligand-induced Shc phosphorylation. Surprisingly, Src inhibition affected neither association of Shc with ErbB receptors nor phosphorylation of receptor-bound Shc. Thus, ErbB signaling requires the engagement of a novel Src-dependent route to MAPK, to trigger its rapid activation and subsequent efficient stimulation of transcription.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas Adaptadoras de Transporte Vesicular , Genes erbB , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Quinases da Família src/metabolismo , Neoplasias da Mama , Fator de Crescimento Epidérmico/farmacologia , Feminino , Proteína Adaptadora GRB2 , Humanos , Neurregulinas/farmacologia , Fosforilação , Ligação Proteica , Proteínas Adaptadoras da Sinalização Shc , Transdução de Sinais , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Transcrição Gênica , Células Tumorais Cultivadas
12.
Cancer Res ; 61(1): 383-91, 2001 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-11196191

RESUMO

Interleukin (IL)-6, a multifunctional regulator of immune response, hematopoiesis, and acute phase reactions, has also been shown to regulate cancer cell proliferation. We have investigated IL-6 signaling pathways and cellular responses in the T47D breast carcinoma cell line. The IL-6-type cytokines, IL-6 and oncostatin M, simultaneously inhibited cell proliferation and increased cell migration. In T47D cells, IL-6 stimulated the activation of Janus-activated kinase 1 tyrosine kinase and signal transducers and activators of transcription (STAT) 1 and STAT3 transcription factors. Expression of dominant negative STAT3 in the cells strongly reduced IL-6-mediated growth inhibition but did not prevent IL-6-induced cell migration. IL-6 treatment led to activation of the mitogen-activated protein kinase (MAPK) and the phosphatidylinositol 3'-kinase (PI3K) pathways. Inhibition of MAPK or PI3K activity reversed IL-6- and oncostatin M-stimulated migration. Because cross-talk between cytokine receptors and members of the ErbB family of receptor tyrosine kinases has been described previously, we have examined their interaction in T47D cells. Down-regulation of ErbB receptor activity, through the use of specific pharmacological inhibitors or dominant negative receptor constructs, revealed that IL-6-induced MAPK activation was largely dependent on epidermal growth factor (EGF) receptor activity, but not on ErbB-2 activity. Using a monoclonal antibody that interferes with EGF receptor-ligand interaction, we have shown that in T47D cells, IL-6 cooperates with an EGF receptor autocrine activity loop for signaling through the MAPK and PI3K pathways and for cell migration. Both the tyrosine phosphatase SHP-2 and the multisubstrate docking molecule Gab1, which are potential links between IL-6 and the MAPK/PI3K pathways, were constitutively associated with the active EGF receptor. On IL-6 stimulation, SHP-2 and Gab1 were recruited to the gp130 subunit of the IL-6 receptor and tyrosine phosphorylated, allowing downstream signaling to the MAPK and PI3K pathways. Thus, in T47D breast carcinoma cells, IL-6 acts in synergy with EGF receptor autocrine activity to signal through the MAPK/PI3K pathways. Cooperation between IL-6 and the EGF receptor in T47D breast carcinoma cells illustrates how a combination of multiple stimuli, either exogenous or endogenous, may result in synergistic cellular responses.


Assuntos
Neoplasias da Mama/patologia , Receptores ErbB/fisiologia , Interleucina-6/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal , Neoplasias da Mama/enzimologia , Neoplasias da Mama/metabolismo , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Ativação Enzimática , Humanos , Interleucina-6/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Janus Quinase 1 , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/fisiologia , Oncostatina M , Peptídeos/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositol 3-Quinases/fisiologia , Fosfoproteínas/metabolismo , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Tirosina Quinases/fisiologia , Receptor ErbB-2/fisiologia , Fator de Transcrição STAT3 , Transdução de Sinais/fisiologia , Transativadores/metabolismo , Transativadores/fisiologia , Células Tumorais Cultivadas , Tirosina/metabolismo
13.
Oncogene ; 19(31): 3460-9, 2000 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-10918604

RESUMO

The interaction between the erbB tyrosine kinase receptors and their ligands plays an important role in tumor growth via the regulation of autocrine and paracrine loops. We report the effect of heregulin beta1, the ligand for erbB-3 and erbB-4 receptors, on the regulation of vascular endothelial growth factor (VEGF) expression, using a panel of breast and lung cancer cell lines with constitutive erbB-2 overexpression or engineered to stably overexpress the erbB-2 receptor. We demonstrate that heregulin beta1 induces VEGF secretion in most cancer cell lines, while no significant effect was observed in normal human mammary and bronchial primary cells. Overexpression of erbB-2 receptor results in induction of the basal level of VEGF and exposure to heregulin further enhances VEGF secretion. This is associated with increased VEGF mRNA expression. In contrast, VEGF induction is significantly decreased in a T47D cell line where erbB-2 is functionally inactivated. Conditioned media from heregulin-treated cancer cells, but not from normal cells, stimulates endothelial cell proliferation; this paracrine stimulation is inhibited by co-exposure to a specific VEGF neutralizing antibody. Furthermore, heregulin-mediated angiogenesis is observed in the in vivo CAM assay. This study reports the first evidence of VEGF regulation by heregulin in cancer cells. Oncogene (2000) 19, 3460 - 3469


Assuntos
Fatores de Crescimento Endotelial/metabolismo , Receptores ErbB/fisiologia , Linfocinas/metabolismo , Proteínas de Neoplasias/fisiologia , Neovascularização Patológica , Neuregulina-1/fisiologia , Receptor ErbB-2/fisiologia , Receptor ErbB-3/fisiologia , Adenocarcinoma/patologia , Animais , Mama/citologia , Neoplasias da Mama/patologia , Brônquios/citologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Divisão Celular , Células Cultivadas/efeitos dos fármacos , Embrião de Galinha , Meios de Cultivo Condicionados/farmacologia , Fatores de Crescimento Endotelial/antagonistas & inibidores , Fatores de Crescimento Endotelial/biossíntese , Fatores de Crescimento Endotelial/genética , Receptores ErbB/biossíntese , Receptores ErbB/genética , Feminino , Genes erbB-2 , Humanos , Neoplasias Pulmonares/patologia , Linfocinas/antagonistas & inibidores , Linfocinas/biossíntese , Linfocinas/genética , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/metabolismo , Neovascularização Fisiológica , Fosforilação , Processamento de Proteína Pós-Traducional , RNA Mensageiro/biossíntese , RNA Neoplásico/biossíntese , Receptor ErbB-2/biossíntese , Receptor ErbB-3/biossíntese , Receptor ErbB-3/genética , Receptor ErbB-4 , Proteínas Recombinantes de Fusão/fisiologia , Transfecção , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo , Veias Umbilicais/citologia , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
15.
Oncogene ; 19(13): 1647-56, 2000 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-10763821

RESUMO

The ErbB2 receptor tyrosine kinase is overexpressed in a variety of human tumours. In order to understand the mechanism by which ErbB2 mediates tumour proliferation we have functionally inactivated the receptor using an intracellularly expressed, ER-targeted single-chain antibody (scFV-5R). Inducible expression of scFv-5R in the ErbB2-overexpressing SKBr3 breast tumour cell line leads to loss of plasma membrane localized ErbB2. Simultaneously, the activity of ErbB3, MAP kinase and PKB/Akt decreased dramatically, suggesting that active ErbB2/ErbB3 dimers are necessary for sustained activity of these kinases. Loss of functional ErbB2 caused the SKBr3 tumour cells to accumulate in the G1 phase of the cell cycle. This was a result of reduction in CDK2 activity, which was mediated by a re-distribution of p27Kip1 from sequestering complexes to cyclin E/CDK2 complexes. The level of c-Myc and D-cyclins, proteins involved in p27KiP1 sequestration, decreased in the absence of functional ErbB2. Ectopic expression of c-Myc led to an increase in D cyclin levels, CDK2 activity and resulted in a partial G1 rescue. We propose that c-Myc is a primary effector of ErbB2-mediated oncogenicity and functions to prevent normal p27Kip1 control of cyclinE/CDK2.


Assuntos
Neoplasias da Mama/patologia , Quinases relacionadas a CDC2 e CDC28 , Carcinoma/patologia , Proteínas de Ciclo Celular , Retículo Endoplasmático/metabolismo , Fase G1/fisiologia , Fragmentos de Imunoglobulinas/imunologia , Proteínas de Neoplasias/fisiologia , Receptor ErbB-2/fisiologia , Proteínas Supressoras de Tumor , Neoplasias da Mama/metabolismo , Carcinoma/metabolismo , Divisão Celular , Ciclina D , Ciclina E/metabolismo , Quinase 2 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p27 , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/metabolismo , Dimerização , Retículo Endoplasmático/imunologia , Feminino , Humanos , Fragmentos de Imunoglobulinas/biossíntese , Fragmentos de Imunoglobulinas/genética , Sistema de Sinalização das MAP Quinases , Proteínas Associadas aos Microtúbulos/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas de Neoplasias/imunologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Proteínas Proto-Oncogênicas c-myc/metabolismo , Receptor ErbB-2/química , Receptor ErbB-2/efeitos dos fármacos , Proteínas Recombinantes de Fusão/metabolismo , Transfecção , Células Tumorais Cultivadas
16.
Mol Cell Biol ; 20(9): 3210-23, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10757805

RESUMO

Overexpression of the ErbB2 receptor, a major component of the ErbB receptor signaling network, contributes to the development of a number of human cancers. ErbB2 presents itself, therefore, as a target for antibody-mediated therapies. In this respect, anti-ErbB2 monoclonal antibody 4D5 specifically inhibits the growth of tumor cells overexpressing ErbB2. We have analyzed the effect of 4D5-mediated ErbB2 inhibition on the cell cycle of the breast tumor cell line BT474. 4D5 treatment of BT474 cells resulted in a G(1) arrest, preceded by rapid dephosphorylation of ErbB2, inhibition of cytoplasmic signal transduction pathways, accumulation of the cyclin-dependent kinase inhibitor p27(Kip1), and inactivation of cyclin-Cdk2 complexes. Time courses demonstrated that 4D5 treatment redirects p27(Kip1) onto Cdk2 complexes, an event preceding increased p27(Kip1) expression; this correlates with the downregulation of c-Myc and D-type cyclins (proteins involved in p27(Kip1) sequestration) and the loss of p27(Kip1) from Cdk4 complexes. Similar events were observed in ErbB2-overexpressing SKBR3 cells, which exhibited reduced proliferation in response to 4D5 treatment. Here, p27(Kip1) redistribution resulted in partial Cdk2 inactivation, consistent with a G1 accumulation. Moreover, p27(Kip1) protein levels remained constant. Antisense-mediated inhibition of p27(Kip1) expression in 4D5-treated BT474 cells further demonstrated that in the absence of p27(Kip1) accumulation, p27(Kip1) redirection onto Cdk2 complexes is sufficient to inactivate Cdk2 and establish the G(1) block. These data suggest that ErbB2 overexpression leads to potentiation of cyclin E-Cdk2 activity through regulation of p27(Kip1) sequestration proteins, thus deregulating the G(1)/S transition. Moreover, through comparison with an ErbB2-overexpressing cell line insensitive to 4D5 treatment, we demonstrate the specificity of these cell cycle events and show that ErbB2 overexpression alone is insufficient to determine the cellular response to receptor inhibition.


Assuntos
Neoplasias da Mama/metabolismo , Quinases relacionadas a CDC2 e CDC28 , Proteínas de Ciclo Celular , Quinases Ciclina-Dependentes/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptor ErbB-2/fisiologia , Proteínas Supressoras de Tumor , Anticorpos Monoclonais , Ciclo Celular , Divisão Celular , Quinase 2 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p27 , Citoplasma/metabolismo , Citometria de Fluxo , Fase G1 , Humanos , Fosforilação , Proteínas Quinases/metabolismo , Transdução de Sinais , Neoplasias Gástricas/metabolismo , Fatores de Tempo , Células Tumorais Cultivadas
17.
J Biol Chem ; 275(14): 10247-55, 2000 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-10744710

RESUMO

The activity of transcription factors of the Stat family is controlled by phosphorylation of a conserved, carboxyl-terminal tyrosine residue. Tyrosine phosphorylation is essential for Stat dimerization, nuclear translocation, DNA binding, and transcriptional activation. Phosphorylation of Stats on specific serine residues has also been described. We have previously shown that in HC11 mammary epithelial cells Stat5a is phosphorylated on Tyr(694) in a prolactin-sensitive manner, whereas serine phosphorylation is constitutive (Wartmann, M., Cella, N., Hofer, P., Groner, B., Xiuwen, L., Hennighausen, L., and Hynes, N. E. (1996) J. Biol. Chem. 271, 31863-31868). By using mass spectrometry and site-directed mutagenesis, we have now identified Ser(779), located in a unique Stat5a SP motif, as the site of serine phosphorylation. By using phospho-Ser(779)-specific antiserum, we have determined that Ser(779) is constitutively phosphorylated in mammary glands taken from different developmental stages. Stat5a isolated from spleen, heart, brain, and lung was also found to be phosphorylated on Ser(779). Ser(725) in Stat5a has also been identified as a phosphorylation site (Yamashita, H., Xu, J., Erwin, R. A., Farrar, W. L., Kirken, R. A., and Rui, H. (1998) J. Biol. Chem. 273, 30218-30224). Here we show that mutagenesis of Ser(725), Ser(779), or a combination of Ser(725/779) to an Ala had no effect on prolactin-induced transcriptional activation of a beta-casein reporter construct. However, following prolactin induction the Ser(725) mutant displayed sustained DNA binding activity compared with that of wild type Stat5a. The results suggest that Ser(725) phosphorylation has an impact on signal duration.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Glândulas Mamárias Animais/metabolismo , Proteínas do Leite , Prolactina/farmacologia , Serina , Transativadores/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Encéfalo/metabolismo , Células COS , Proteínas de Ligação a DNA/química , Feminino , Pulmão/metabolismo , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Miocárdio/metabolismo , Fragmentos de Peptídeos/química , Mapeamento de Peptídeos , Fosfatos/metabolismo , Fosfopeptídeos/química , Fosfopeptídeos/metabolismo , Fosforilação , Fosfosserina , Receptores da Prolactina/genética , Receptores da Prolactina/fisiologia , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Fator de Transcrição STAT5 , Baço/metabolismo , Transativadores/química , Transfecção
18.
J Cell Biol ; 148(2): 385-97, 2000 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-10648571

RESUMO

The epidermal growth factor (EGF) family of tyrosine kinase receptors (ErbB1, -2, -3, and -4) and their ligands are involved in cell differentiation, proliferation, migration, and carcinogenesis. However, it has proven difficult to link a given ErbB receptor to a specific biological process since most cells express multiple ErbB members that heterodimerize, leading to receptor cross-activation. In this study, we utilize carcinoma cells depleted of ErbB2, but not other ErbB receptor members, to specifically examine the role of ErbB2 in carcinoma cell migration and invasion. Cells stimulated with EGF-related peptides show increased invasion of the extracellular matrix, whereas cells devoid of functional ErbB2 receptors do not. ErbB2 facilitates cell invasion through extracellular regulated kinase (ERK) activation and coupling of the adaptor proteins, p130CAS and c-CrkII, which regulate the actin-myosin cytoskeleton of migratory cells. Overexpression of ErbB2 in cells devoid of other ErbB receptor members is sufficient to promote ERK activation and CAS/Crk coupling, leading to cell migration. Thus, ErbB2 serves as a critical component that couples ErbB receptor tyrosine kinases to the migration/invasion machinery of carcinoma cells.


Assuntos
Adenocarcinoma/fisiopatologia , Neoplasias da Mama/fisiopatologia , Invasividade Neoplásica , Proteínas , Proteínas Proto-Oncogênicas , Receptores Proteína Tirosina Quinases/metabolismo , Receptor ErbB-2/metabolismo , Receptores de Fatores de Crescimento/metabolismo , Movimento Celular , Proteína Substrato Associada a Crk , Ativação Enzimática , Fator de Crescimento Epidérmico/metabolismo , Feminino , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fragmentos de Peptídeos/metabolismo , Fosfoproteínas/metabolismo , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-crk , Receptor ErbB-2/genética , Proteínas Recombinantes/metabolismo , Proteína p130 Retinoblastoma-Like , Transdução de Sinais
19.
J Cell Biochem ; 76(3): 394-403, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10649437

RESUMO

Mammary epithelial cell differentiation depends on lactogenic hormones, growth factors, and cell-cell and cell-substrate interactions, all of which modulate transcription factors essential for milk protein gene expression. The CCAAT/enhancer binding protein (C/EBP) family and the signal transducer and activator of transcription 5 (Stat5) have been implicated in mammary epithelial cell growth and differentiation. We have investigated the effects of extracellular matrix components and lactogenic hormones on C/EBP and Stat5 activity. In the mammary gland, tenascin is expressed mainly during embryogenesis and carcinogenesis and in cell culture tenascin downregulates beta-casein gene expression. In HC11 mammary cells, we found that tenascin, but not laminin or fibronectin, specifically downregulated C/EBPalpha levels but had no effect on Stat5 amount or DNA binding activity. Furthermore, we found that the lactogenic hormones, glucocorticoids, prolactin, and insulin, had no effect on C/EBPalpha and C/EBPbeta protein levels but downregulated the DNA binding activity of the transcriptional repressor C/EBPbetaLIP. Thus, C/EBPalpha and beta are regulated by tenascin and lactogenic hormones in mammary epithelial cells.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/metabolismo , Proteínas do Leite , Proteínas Nucleares/metabolismo , Prolactina/farmacologia , Tenascina/farmacologia , Animais , Sequência de Bases , Sítios de Ligação/genética , Proteínas Estimuladoras de Ligação a CCAAT , Caseínas/genética , Diferenciação Celular , Linhagem Celular , DNA/genética , DNA/metabolismo , Dexametasona/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Matriz Extracelular/metabolismo , Feminino , Expressão Gênica/efeitos dos fármacos , Insulina/farmacologia , Glândulas Mamárias Animais/citologia , Camundongos , Prolactina/metabolismo , Ligação Proteica/efeitos dos fármacos , Isoformas de Proteínas/metabolismo , Fator de Transcrição STAT5 , Transdução de Sinais , Tenascina/metabolismo , Transativadores/metabolismo
20.
Breast Cancer Res ; 2(3): 154-7, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11250704

RESUMO

Cells are continuously exposed to diverse stimuli ranging from soluble endocrine and paracrine factors to signalling molecules on neighbouring cells. Receptors of the tyrosine kinase family play an important role in the integration and interpretation of these external stimuli, allowing a cell to respond appropriately to its environment. The activation of receptor tyrosine kinases (RTKs) is tightly controlled, allowing a normal cell to correctly integrate its external environment with internal signal transduction pathways. In contrast, due to numerous molecular alterations arising during the course of malignancy, a tumour is characterized by an abnormal response to its environment, which allows cancer cells to evade the normal mechanisms controlling cellular proliferation. Alterations in the expression of various RTKs, in their activation, and in the signalling molecules lying downstream of the receptors play important roles in the development of cancer. This topic is the major focus of the thematic review section of this issue of Breast Cancer Research.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas Tirosina Quinases/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais , Neoplasias da Mama/genética , Feminino , Genes erbB/genética , Genes src/genética , Humanos , Fator de Crescimento Insulin-Like I/genética , Proteínas Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/genética , Receptores de Fatores de Crescimento de Fibroblastos/genética , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...