Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
West Afr J Med ; 26(1): 42-7, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17595991

RESUMO

BACKGROUND AND OBJECTIVE: Anti-epidermal growth factor receptor strategies are now established in cancer treatment We have recently described the presence of EGFRvIII (a variant EGFR) in prostatic tumours from UK white men and this is now a target for anti-prostate cancer treatments. However, there has been no report on the expression of this abnormal protein in black men. MATERIALS AND METHODS: We determined EGFRvIII expression in sections of normal, benign hyperplastic (BPH) and carcinomatous (CaP) prostatic archival tissues from Nigerian men and UK white men using streptavidin immunohistochemical techniques. The EGFRvIII immunoreactivity was scored visually using a semi-quantitative method and the results compared statistically. RESULTS: EGFRvIII expression increased with increasing malignancy in both study populations (CaP > BPH > Normal p, <0.0001). Furthermore, EGFRvIII expression was similar in both BPH and CaP tissues in black and white men (p, 0.86 and 0.31 respectively). CONCLUSION: These results demonstrate that EGFRvIII immunoreactivity in prostatic tumours in black men is similar to that in white men. Anti-cancer treatments directed at the EGFRvIII should be equally effective in men from both subpopulations.


Assuntos
População Negra , Receptores ErbB/fisiologia , Hiperplasia Prostática/fisiopatologia , Neoplasias da Próstata/fisiopatologia , População Branca , Estudos de Casos e Controles , Receptores ErbB/genética , Receptores ErbB/imunologia , Humanos , Imuno-Histoquímica , Masculino , Nigéria/epidemiologia , Hiperplasia Prostática/genética , Hiperplasia Prostática/imunologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/imunologia , Estados Unidos/epidemiologia
2.
Afr J Med Med Sci ; 33(3): 245-53, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15819472

RESUMO

Prostatic carcinogenesis has been associated with alterations in the expression of the androgen receptor (AR) and the epidermal growth factor receptor (WT-EGFR), and over-expression of the constitutively active variant epidermal growth factor receptor (EGFRvIII). Changes in the expression of AR, WT-EGFR and EGFRvIII were evaluated in serial sections from 26 normal and 26 benign hyperplastic and 50 prostate cancer tissues using specific immunostaining techniques. The loss of AR expression in peri-epithelial stroma as prostatic tissues de-differentiated correlated strongly with the depletion of WT-EGFR and with increasing expression of the EGFRvIII in the adjacent epithelium. In contrast, changes in epithelial AR immunopositivity in these tissues correlated weakly with the changes in normal and variant EGFR levels. This is the first report correlating the changes in the expression of these three proteins in archival material from the different human prostatic tissue histotypes. The loss of expression of proteins that contribute to the regulation of prostatic homeostasis (AR and WT-EGFR) correlates strongly with the expression of a constitutively active variant EGF receptor (EGFRvIII) in human prostate cancer. These changes occur at an early stage of neoplastic transformation and may contribute to the progression of the disease to hormone independence.


Assuntos
Receptores ErbB/metabolismo , Hiperplasia Prostática/metabolismo , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/metabolismo , Endotélio Vascular/metabolismo , Humanos , Imunoquímica , Masculino
3.
Appl Immunohistochem Mol Morphol ; 9(3): 261-6, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11556755

RESUMO

Storage of unstained paraffin slides may lead to the deterioration of specimens and failure to detect cellular proteins immunohistochemically. Although the implication of age-induced alterations on multicenter immunohistochemical studies would be considerable, they have not been investigated previously. The current study was undertaken to examine the effect of this factor further and to explore new ways of overcoming the resultant shortcomings. The authors now report on the immunodetection of a host of antigens in similarly preserved unstained serial paraffin slides obtained from three centers using a panel of eight antibodies. Staining of recently prepared sections from the authors' centers resulted in similar strong patterns in seven of eight antibodies, with one antibody demonstrating variable immunoreactivity. However, storage of unstained paraffin sections at room temperature resulted in a variable but progressive decrease in expression of several tissue antigens. Although the loss in antigenicity was proportional to the length of storage, the effect was reversible if super antibody concentrations were used. The authors conclude that recently prepared paraffin sections from centers with similar fixation protocols have similar immunoreactivity and are suitable for use in comparative multicenter studies. However, in view of the delays that may attend tissue transportation during these projects, the authors suggest that test systems should be checked for age-induced antigen degradation by incubating sections with higher antibody concentrations.


Assuntos
Inclusão em Parafina , Manejo de Espécimes , Anticorpos/imunologia , Humanos , Imuno-Histoquímica , Estudos Multicêntricos como Assunto
5.
Cancer Res ; 60(11): 3081-7, 2000 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-10850460

RESUMO

Epidermal growth factor receptor vIII (EGFRvIII) is a tumor-specific, ligand-independent, constitutively active variant of the EGFR. Its expression has been detected in gliomas and various other human malignancies. To more fully characterize the function and potential biological role of EGFRvIII in regulating cell proliferation and in tumorigenesis, we transfected EGFRvIII cDNA into a nontumorigenic, interleukin 3 (IL-3)-dependent murine hematopoietic cell line (32D cells). We observed 32D cells expressing high levels of EGFRvIII (32D/EGFRvIII P5) to be capable of abrogating the IL-3-dependent pathway in the absence of ligands. In contrast, the parental cells, 32D/EGFR, 32D/ErbB-4, and 32D/ErbB-2+ErbB-3 cells, all depended on IL-3 or EGF-like ligands for growth. 32D/EGFRvIII P5 cells subjected to long-term culture conditions in the absence of IL-3 revealed further elevation of EGFRvIII expression levels. These results suggested that the IL-3-independent phenotype is mediated by EGFRvIII. The level of expression is a critical driving force for the IL-3-independent phenotype. Dose-response analysis revealed 32D/EGFRvIII cells to require 500-fold higher concentrations (50 ng/ml) of EGF to further stimulate the EGF-mediated proliferation than in the 32D/EGFR cells (100 pg/ml). Similar effects were also observed in beta-cellulin-mediated proliferation. Moreover, 32D cells expressing high levels of EGFRvIII formed large tumors in nude mice, even when no exogenous EGF ligand was administered. In contrast, no tumors grew in mice injected with 32D/EGFR, 32D/ErbB-4, and 32D/ErbB-2+ErbB-3 cells or low-expressing clone 32D/EGFRvIII C2 cells or the parental 32D cells. The changes of the ligand specificity support the notion for an altered conformation of EGFRvIII to reveal an activated ligand-independent oncoprotein with tumorigenic activity analogous to v-erbB. These studies clearly demonstrate that EGFRvIII is capable of transforming a nontumorigenic, IL-3-dependent murine hematopoietic cell line (32D cells) into an IL-3-independent and ligand-independent malignant phenotype in vitro and in vivo. To delineate the biological significance of EGFRvIII in human breast cancer, we expressed EGFRvIII in the MCF-7 human breast cancer cell line. Expression of EGFRvIII in MCF-7 cells produced a constitutively activated EGFRvIII receptor. Expression of EGFRvIII in MCF-7 cells also elevated ErbB-2 phosphorylation, presumably through heterodimerization and cross-talk. These MCF-7/EGFRvIII transfectants exhibited an approximately 3-fold increase in colony formation in 1% serum with no significant effect observed at higher percentages of serum. A similar result was also seen in anchorage-dependent assays. Furthermore, EGFRvIII expression significantly enhanced tumorigenicity of MCF-7 cells in athymic nude mice with P < 0.001. Collectively, these results provide the first evidence that EGFRvIII could play a pivotal role in human breast cancer progression.


Assuntos
Neoplasias da Mama/metabolismo , Receptores ErbB/metabolismo , Animais , Divisão Celular , DNA Complementar/metabolismo , Receptores ErbB/genética , Feminino , Citometria de Fluxo , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Fosforilação , Receptor ErbB-2/metabolismo , Fatores de Tempo , Transfecção , Células Tumorais Cultivadas
6.
Br J Cancer ; 82(1): 186-94, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10638988

RESUMO

Earlier studies have demonstrated an unexplained depletion of the epidermal growth factor receptor (EGFR) protein expression in prostatic cancer. We now attribute this phenomenon to the presence of a variant EGFR (EGFRvIII) that is highly expressed in malignant prostatic neoplasms. In a retrospective study, normal, benign hyperplastic and malignant prostatic tissues were examined at the mRNA and protein levels for the presence of this mutant receptor. The results demonstrated that whilst EGFRvIII was not present in normal prostatic glands, the level of expression of this variant protein increased progressively with the gradual transformation of the tissues to the malignant phenotype. The selective association of high EGFRvIII levels with the cancer phenotype underlines the role that this mutant receptor may maintain in the initiation and progression of malignant prostatic growth, and opens the way for new approaches in the management of this disease including gene therapy.


Assuntos
Receptores ErbB/análise , Proteínas de Neoplasias/análise , Neoplasias da Próstata/química , Análise de Variância , Western Blotting , Resistencia a Medicamentos Antineoplásicos , Humanos , Masculino , Fenótipo , RNA Mensageiro/análise , Estudos Retrospectivos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
J Biol Chem ; 274(44): 31719-26, 1999 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-10531383

RESUMO

Stimulation of the hepatocyte growth factor receptor tyrosine kinase, Met, induces the inherent morphogenic program of epithelial cells. The multisubstrate binding protein Gab1 (Grb2-associated binder-1) is the major phosphorylated protein in epithelial cells following activation of Met. Gab1 contains a pleckstrin homology domain and multiple tyrosine residues that act to couple Met with multiple signaling proteins. Met receptor mutants that are impaired in their association with Gab1 fail to induce a morphogenic program in epithelial cells, which is rescued by overexpression of Gab1. The Gab1 pleckstrin homology domain binds to phosphatidylinositol 3,4, 5-trisphosphate and contains conserved residues, shown from studies of other pleckstrin homology domains to be crucial for phospholipid binding. Mutation of conserved phospholipid binding residues tryptophan 26 and arginine 29, generates Gab1 proteins with decreased phosphatidylinositol 3,4,5-trisphosphate binding, decreased localization at sites of cell-cell contact, and reduced ability to rescue Met-dependent morphogenesis. We conclude that the ability of the Gab1 pleckstrin homology domain to bind phosphatidylinositol 3,4,5-trisphosphate is critical for subcellular localization of Gab1 and for efficient morphogenesis downstream from the Met receptor.


Assuntos
Proteínas Sanguíneas , Sequência Conservada , Células Epiteliais/citologia , Fosfatos de Fosfatidilinositol/metabolismo , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Sequência de Aminoácidos , Arginina/genética , Sítios de Ligação/genética , Morfogênese , Mutagênese Sítio-Dirigida , Fosfoproteínas/genética , Fosforilação , Ligação Proteica , Homologia de Sequência de Aminoácidos , Triptofano/genética
8.
J Biol Chem ; 274(8): 4489-92, 1999 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-9988678

RESUMO

Ectopic expression of decorin induces profound cytostatic effects in transformed cells with diverse histogenetic backgrounds. The mechanism of action has only recently begun to be elucidated. Exogenous decorin activates the epidermal growth factor (EGF) receptor, thereby triggering a signaling cascade that leads to phosphorylation of mitogen-activated protein (MAP) kinase, induction of p21, and growth suppression. In this study we demonstrate a direct interaction of decorin with the EGF receptor. Binding of decorin induces dimerization of the EGF receptor and rapid and sustained phosphorylation of MAP kinase in squamous carcinoma cells. In a cell-free system, decorin induces autophosphorylation of purified EGF receptor by activating the receptor tyrosine kinase and can also act as a substrate for the EGF receptor kinase itself. Using radioligand binding assays we show that both immobilized and soluble decorin bind to the EGF receptor ectodomain or to purified EGF receptor. The binding is mediated by the protein core and has relatively low affinity (Kd approximately 87 nM). Thus, decorin should be considered as a novel biological ligand for the EGF receptor, an interaction that could regulate cell growth during remodeling and cancer growth.


Assuntos
Receptores ErbB/metabolismo , Proteoglicanas/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Decorina , Dimerização , Ativação Enzimática , Proteínas da Matriz Extracelular , Ligantes , Fosforilação , Ligação Proteica , Células Tumorais Cultivadas
9.
J Biol Chem ; 273(1): 200-6, 1998 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-9417065

RESUMO

The most frequently found alteration of the epidermal growth factor receptor (EGFR) in human tumors is a deletion of exons 2-7. This receptor, termed EGFRvIII, can transform NIH 3T3 cells, and the frequent expression of this variant implies that it confers a selective advantage upon tumor cells in vivo. Although EGFRvIII is a constitutively activated tyrosine kinase, there is no increase in Ras.GTP levels and low levels of mitogen-activated protein kinase activity in NIH 3T3 cells expressing this variant. We investigated whether phosphatidylinositol (PI) 3-kinase was an effector in transformation by the EGFRvIII. High levels of PI 3-kinase activity were constitutively present in EGFRvIII-transformed cells and were dependent upon the kinase activity of the receptor. While mitogen-activated protein kinase activity was quickly down-regulated to basal levels after 12 h of continuous EGFR activation, there was a 3-fold increase in PI 3-kinase activity in cells expressing normal EGFR and an 8-fold increase in cells expressing EGFRvIII after 48 h. This increased activity may reflect enhanced binding to EGFRvIII and the presence of novel PI 3-kinase isoforms. Treatment with the PI 3-kinase inhibitors wortmannin and LY294002 blocked both anchorage-independent growth and growth in low serum media and also resulted in morphological reversion of EGFRvIII-transformed cells. These results support an essential role for PI 3-kinase in transformation by this EGFR variant.


Assuntos
Receptores ErbB/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Células 3T3 , Proteínas Adaptadoras de Transdução de Sinal , Animais , Divisão Celular , Linhagem Celular Transformada , Regulação para Baixo , Ativação Enzimática , Receptores ErbB/genética , Camundongos , Mutação , Fosfoproteínas/metabolismo
10.
J Biol Chem ; 273(5): 2817-22, 1998 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-9446590

RESUMO

Epidermal growth factor receptor (EGF) variant type III (EGFRvIII) is a constitutively active, naturally occurring mutation of the EGF receptor that is found in many types of human tumors. When overexpressed in NIH3T3 fibroblasts, EGFRvIII induces transformation by enhancing cell growth and reducing apoptosis. Analysis of downstream signaling pathways has revealed that extracellular signal-regulated kinase activity is down-regulated, raising doubt as to the significance of this pathway in promoting transformation. We investigated whether the c-Jun N-terminal kinase (JNK) pathway was affected by EGFRvIII. NIH3T3 cells expressing EGFRvIII exhibited a high basal level of JNK activity, which was not present in cells overexpressing the normal EGF receptor. Treatment of cells overexpressing EGFRvIII with inhibitors of the EGF receptor or phosphatidylinositol 3-kinase resulted in the down-regulation of JNK activity. Furthermore, the down-regulation of JNK activity was associated with a loss of properties related to transformation, and there was no evidence for JNK activity in the promotion of apoptosis in these cells. These findings implicate constitutive activation of the JNK pathway in transformation by EGFRvIII.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Transformação Celular Neoplásica/genética , Receptores ErbB/genética , Proteínas Quinases Ativadas por Mitógeno , Tirfostinas , Células 3T3 , Animais , Apoptose , Regulação para Baixo , Ativação Enzimática , Receptores ErbB/antagonistas & inibidores , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Camundongos , Mutação , Nitrilas/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Quinazolinas/farmacologia , Transdução de Sinais , Transfecção
11.
J Clin Invest ; 101(2): 406-12, 1998 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-9435313

RESUMO

Decorin, a small leucine-rich proteoglycan, is capable of suppressing the growth of various tumor cell lines when expressed ectopically. In this report, we investigated the biochemical mechanism by which decorin inhibits cell cycle progression. In A431 squamous carcinoma cells, decorin proteoglycan or protein core induced a marked growth suppression, when either exogenously added or endogenously produced by a transgene. Decorin caused rapid phosphorylation of the EGF receptor and a concurrent activation of mitogen-activated protein (MAP) kinase signal pathway. This led to a protracted induction of endogenous p21, a potent inhibitor of cyclin-dependent kinases, and ultimate cell cycle arrest. Biglycan, a related proteoglycan, had no effect. Moreover, decorin activated the EGF receptor/MAP kinase/ p21 axis in cell lines of various histogenetic backgrounds. These results provide the first evidence that EGF and decorin converge functionally to regulate the cell cycle through activation of a common pathway which ultimately leads to growth suppression.


Assuntos
Carcinoma de Células Escamosas/patologia , Receptores ErbB/fisiologia , Proteoglicanas/fisiologia , Tirfostinas , Células 3T3 , Animais , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/biossíntese , Decorina , Ativação Enzimática , Proteínas da Matriz Extracelular , Humanos , Camundongos , Nitrilas/farmacologia , Fosforilação , Quinazolinas/farmacologia , Células Tumorais Cultivadas
12.
Proc Natl Acad Sci U S A ; 94(23): 12419-24, 1997 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-9356464

RESUMO

Nerve growth factor (NGF) prevents apoptosis through stimulation of the TrkA receptor protein tyrosine kinase. The downstream activation of phosphatidylinositol 3-kinase (PI 3-kinase) is essential for the inhibition of apoptosis, although this enzyme does not bind to and is not directly activated by TrkA. We have found that the addition of NGF to PC-12 cells resulted in the phosphorylation of the Grb2-associated binder-1 (Gab1) docking protein and induced the association of several SH2 domain-containing proteins, including PI 3-kinase. A substantial fraction of the total cellular PI 3-kinase activity was associated with Gab1. PC-12 cells that overexpressed Gab1 show a decreased requirement for the amount of NGF necessary to inhibit apoptosis. The expression of a Gab1 mutant that lacked the binding sites for PI 3-kinase enhanced apoptosis and diminished the protective effect of NGF. Hence, Gab1 has a major role in connecting TrkA with PI 3-kinase activation and for the promotion of cell survival by NGF.


Assuntos
Apoptose/efeitos dos fármacos , Fatores de Crescimento Neural/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Sobrevivência Celular/efeitos dos fármacos , Mutação , Células PC12 , Fosfoproteínas/genética , Ratos
13.
Cancer Res ; 57(8): 1419-24, 1997 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-9108438

RESUMO

The type III EGF receptor (EGFRvIII) is the result of an in-frame deletion from nucleotides 275 to 1075 in the EGF receptor cDNA sequence creating a novel epitope at the fusion junction. This spontaneously occurring alteration is found in a high percentage of primary human brain, breast, lung and ovarian tumors. We have explored whether a peptide derived from the fusion junction could serve as the basis for an antitumor vaccine. Preimmunization of mice with this peptide substantially inhibited tumor formation by cells expressing EGFRvIII. Tumor cell inoculation followed by immunization could also enhance the regression of existing tumors. Antibody production was elicited in animals that was highly specific for the novel epitope and also a CTL response that was mediated by CD8+ T lymphocytes. The alteration present in EGFRvIII could serve as the basis for an antitumor vaccine with potentially wide application in humans.


Assuntos
Vacinas Anticâncer/uso terapêutico , DNA Complementar/genética , Epitopos/imunologia , Receptores ErbB/imunologia , Imunoterapia , Neoplasias Experimentais/terapia , Deleção de Sequência , Linfócitos T Citotóxicos/imunologia , Células 3T3 , Animais , Vacinas Anticâncer/imunologia , Receptores ErbB/classificação , Receptores ErbB/genética , Feminino , Humanos , Camundongos , Neoplasias Experimentais/genética , Neoplasias Experimentais/imunologia , Ratos , Ratos Endogâmicos F344 , Transfecção
14.
Biochem Biophys Res Commun ; 232(1): 111-6, 1997 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-9125112

RESUMO

The epidermal growth factor receptor (EGFR) activates formation of the phospholipid signal messenger phosphatidic acid (PA) on ligand binding. We explored the effects of chronic EGF stimulation on cellular PA in NIH3T3 cells expressing intact EGFR a mutant EGFR (EGFRvIII). The presence of EGFRvIII increased PA levels to twice those induced by chronic EGFR activation. Fatty acid methyl ester analysis revealed a marked increase in oleic acid containing PA. No apparent increase in phospholipase D (PLD) activity was detected, and diacylglycerol (DAG) kinase assays demonstrated a marked preference for dioleoyl DAG in the presence of activated EGFR or EGFRvIII. Levels of PA which were lower than would be predicted by DAG kinase activation are explained by increased phosphatidate phosphohydrolase activity. Specific inhibitors of EGFR kinase and DAG kinase suppressed DAG kinase activation and PA production by EGFRvIII. EGFR kinase activation by chronic exposure to ligand or by deletional mutation stimulates formation of a specific form of signalling PA.


Assuntos
Receptores ErbB/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Células 3T3 , Animais , Linhagem Celular , Diacilglicerol Quinase , Ativação Enzimática , Camundongos
15.
J Biol Chem ; 272(7): 4079-86, 1997 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-9020117

RESUMO

Mutation of the autophosphorylation sites of receptor protein-tyrosine kinases alters ligand dependent internalization and down-regulation, indicating a critical role for these sites in receptor processing. Currently, no differences in receptor processing based on an individual autophosphorylation site have been defined. By using a glutathione S-transferase fusion protein containing the src homology 2 domains of phospholipase C-gamma1 to specifically recognize tyrosine 992 on the EGF receptor (Tyr(P)992), we have found differences in this subpopulation of receptors. Following EGF stimulation, the number of Tyr(P)992 receptors increased 2-fold over receptors identified by an antibody that recognizes activated EGF receptors (alpha-Act. EGFR) in A431 cells. Confocal fluorescence microscopy showed that Tyr(P)992 receptors underwent endocytosis at a slower rate and did not rapidly concentrate in juxtanuclear bodies. Tyr(P)992 receptors were associated with more SOS, Ras-GTPase activating protein, phosphatidylinositol 3-kinase, and SHPTP2/syp, but less Grb2, than receptors in the general population, and these receptors were more heavily phosphorylated than the general population of active receptors. These findings suggest that autophosphorylation status is relevant to the endocytosis, degradation, and effector molecule interaction of individual EGF receptors. Further investigations based on phosphorylation status should provide new insights into how receptor protein-tyrosine kinase signaling is regulated.


Assuntos
Endocitose , Receptores ErbB/classificação , Actinina/metabolismo , Transporte Biológico , Western Blotting , Linhagem Celular , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Mutagênese Sítio-Dirigida , Ligação Proteica , Especificidade por Substrato , Fosfolipases Tipo C/metabolismo , Domínios de Homologia de src
16.
Oncogene ; 13(1): 85-96, 1996 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-8700557

RESUMO

An amino-truncated variant form of the epidermal growth factor receptor (EGFRvIII) has been identified in human brain, breast, lung and ovarian tumors. We have found that overexpression of this mutant EGF receptor in NIH3T3 cells results in transformation as a result of the activation of the receptor kinase via ligand-independent dimerization. Transformation was correlated with tyrosine phosphorylation of only a subset of the proteins observed in cells overexpressing the normal EGF receptor. This suggested that further studies on cells expressing the EGFRvIII might provide insights into the pathways most relevant to transformation. In clones expressing high levels of mutant EGF receptor, the levels of both Grb2 and SHC were decreased. Despite this decrease, much of the endogenous Grb2 immunoprecipitated with EGFRvIII. Interestingly, no increase in ras-GTP loading was found in clones expressing the EGFRvIII and MAP kinase assays indicated only a small increase in activity. These results indicate that high-level expression of the EGFRvIII induces down-regulation of the ras-MAP kinase pathway and that other components involved in EGF receptor signal transduction may play a greater role in neoplastic transformation by the EGFRvIII.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Transformação Celular Neoplásica/genética , Receptores ErbB/genética , Proteínas de Neoplasias/genética , Transdução de Sinais/genética , Células 3T3 , Sequência de Aminoácidos , Animais , Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , Receptores ErbB/fisiologia , Feminino , Proteína Adaptadora GRB2 , Guanosina Trifosfato/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Dados de Sequência Molecular , Proteínas de Neoplasias/fisiologia , Transplante de Neoplasias , Fosforilação , Biossíntese de Proteínas , Processamento de Proteína Pós-Traducional , Transfecção
17.
Nature ; 379(6565): 560-4, 1996 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-8596638

RESUMO

The protein Grb2 plays a central role in signalling by receptor protein-tyrosine kinases, where its SH2 domain binds to the receptor and its two SH3 domains link to effectors. One target effector is Sos, so Grb2 links receptor protein-tyrosine kinases with the Ras signalling pathway. The SH3 domains can also couple to other signalling proteins, including Vav, c-Abl and dynamin. We have identified several bands in glial and medulloblastoma tumours that are recognized by Grb2 but these did not correspond to any known protein. Here we use recombinant Grb2 to isolate a complementary DNA called Gab1 (for Grb2-associated binder-1). Gab1 shares amino-acid homology and several structural features with IRS-1 (insulin-receptor substrate-1; refs 6,7), is a substrate of the EGF and insulin receptors, and can act as a docking protein for several SH2-containing proteins. Over-expression of Gab1 enhances cell growth and results in transformation. We conclude that Gab1 is a new protein in EGF and insulin receptor signalling which could integrate signals from different systems.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Receptores ErbB/metabolismo , Fosfoproteínas/metabolismo , Proteínas/metabolismo , Receptor de Insulina/metabolismo , Transdução de Sinais , Células 3T3 , Sequência de Aminoácidos , Animais , Northern Blotting , Western Blotting , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Divisão Celular , Transformação Celular Neoplásica , DNA Complementar , Ativação Enzimática , Proteína Adaptadora GRB2 , Humanos , Camundongos , Dados de Sequência Molecular , Fosfatidilinositol 3-Quinases , Fosfoproteínas/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Células Tumorais Cultivadas , Domínios de Homologia de src/genética
18.
Cancer Res ; 55(23): 5536-9, 1995 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-7585629

RESUMO

The epidermal growth factor receptor has received much interest as a target for various antineoplastic agents, but a complication is that many normal tissues also express this receptor. We have previously identified in human glial tumors an 801-bp in-frame deletion within the epidermal growth factor receptor gene that created a novel epitope at the junction. By using Western blot assays with a mutant-specific antibody as a rapid and sensitive means for detecting this alteration in primary human tumors, it was found that 57% (26 of 46) of high-grade and 86% (6 of 7) of low-grade glial tumors, but not normal brain, express this protein. This altered receptor was also present in 66% (4 of 6) of pediatric gliomas and 86% (6 of 7) of medulloblastomas, 78% (21 of 27) of breast carcinomas, and 73% (24 of 32) of ovarian carcinomas. The fact that this receptor is frequently found in tumors but not in normal tissue makes it an attractive candidate for various antitumor strategies.


Assuntos
Receptores ErbB/genética , Proteínas de Neoplasias/genética , Neoplasias/química , Anticorpos Monoclonais , Sequência de Bases , Western Blotting , Neoplasias Encefálicas/química , Neoplasias da Mama/química , Receptores ErbB/análise , Feminino , Humanos , Dados de Sequência Molecular , Proteínas de Neoplasias/análise , Neoplasias Ovarianas/química , Células Tumorais Cultivadas
19.
J Biol Chem ; 270(51): 30562-6, 1995 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-8530489

RESUMO

A paradigm has been established whereby mutant tyrosine kinase receptors such as the v-erbB and v-fms gene products function as oncoproteins in the absence of ligand. A spontaneously occurring deletional mutant of the human epidermal growth factor receptor (EGFR-vIII) has been isolated from astrocytic neoplasms and transforms NIH3T3 cells in the absence of ligand. The EGFRvIII is constitutively complexed with the majority of cellular GRB2, suggesting a link to the Ras-Mitogen-activated protein (MAP) kinase pathway (D. Moscatello, R. B. Montgomery, P. Sundareshan, H. McDanel, M. Y. Wong, and A. J. Wong, submitted for publication). In this report, we document that expression of EGFRvIII in fibroblasts is associated with downstream activation of mitogen-activated protein (MAP) kinase/extracellular signal-regulated kinase (MEK) and modest activation of p42 and p44 MAP kinases. The presence of EGFRvIII suppresses activation of p42 and p44 MAP kinases by phorbol 12-myristate 13-acetate (PMA) and serum; however, MEK activation by PMA is not suppressed by EGFRvIII. Basal and PMA-stimulated MAP kinase activity in EGFRvIII-transfected cells is augmented by the tyrosine phosphatase inhibitor sodium vanadate. EGFR-vIII is capable of transducing downstream signals through MAP kinase as evidenced by activation of cytoplasmic phospholipase A2 at levels similar to that induced by intact EGFR. Our results suggest that EGFR-vIII constitutively activates downstream signal transduction through MAP kinase, and this chronic stimulation of the MAP kinase pathway may represent one means by which mutant EGFR transduces an oncogenic signal.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Proteínas Quinases/metabolismo , Deleção de Sequência , Células 3T3 , Animais , Ativação Enzimática , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/biossíntese , Glioblastoma , Humanos , Cinética , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Transfecção , Células Tumorais Cultivadas , Vanadatos/farmacologia
20.
Cancer Res ; 55(19): 4375-82, 1995 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-7671250

RESUMO

Amplification and rearrangement of the epidermal growth factor receptor (EGFR) gene are characteristics of many types of tumors. One class of EGFR mutations, EGFRvIII, is characterized by an in-frame deletion resulting in a truncated external domain of the receptor. EGFRvIII was first identified in a subset of gliomas and has since been found in some non-small cell lung carcinomas and breast carcinomas. mAbs specific for this variant form of EGFR but unreactive with the wild-type EGFR have been reported from our laboratory. This study further characterizes three of these antibodies. We determined, via radiolabeling techniques and immunofluorescence microscopy, that, after cell binding in vitro, the anti-EGFRvIII-specific mAbs internalize at 37 degrees C. Furthermore, subsequent to internalization, the antibodies were processed intracellularly, presumably by lysosomal degradation. We also examined the use of an alternative radiolabeling procedure that uses nonmetabolizable radio-iodinated tyramine cellobiose. Our results show that the tyramine cellobiose labeling method allows for greater tumor cell retention of radiolabel in vitro (76% for tyramine cellobiose and 27% for Iodo-Gen after 24 h). Paired-label biodistribution studies in athymic mice indicate that anti-EGFRvIII mAb L8A4 localizes specifically to EGFRvIII-expressing tumor xenografts with a maximum of 34.3 +/- 7.6% injected dose/g when labeled using tyramine cellobiose compared with a maximum of 14.9 +/- 4.3% injected dose/g using Iodo-Gen; similar results were obtained with mAb H10. These results suggest that the anti-EGFRvIII mAbs may serve as potential carriers for radioconjugate- and immunotoxin-based therapies for tumors expressing EGFRvIII.


Assuntos
Anticorpos Monoclonais/farmacocinética , Receptores ErbB/imunologia , Radioisótopos do Iodo/farmacocinética , Marcação por Isótopo/métodos , Neoplasias Experimentais/metabolismo , Animais , Anticorpos Monoclonais/uso terapêutico , Celobiose , Portadores de Fármacos , Receptores ErbB/análise , Camundongos , Microscopia de Fluorescência , Transplante de Neoplasias , Neoplasias Experimentais/terapia , Doses de Radiação , Distribuição Tecidual , Transplante Heterólogo , Tiramina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA