Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Membranes (Basel) ; 13(8)2023 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-37623781

RESUMO

ADAM17, a prominent member of the "Disintegrin and Metalloproteinase" (ADAM) family, controls vital cellular functions through the cleavage of transmembrane substrates, including epidermal growth factor receptor (EGFR) ligands such as transforming growth factor (TGF)-alpha and Epiregulin (EREG). Several ADAM17 substrates are relevant to oncogenesis and tumor growth. We have presented evidence that surface exposure of phosphatidylserine (PS) is pivotal for ADAM17 to exert sheddase activity. The scramblase Xkr8 is instrumental for calcium-independent exposure of PS in apoptotic cells. Xkr8 can be dually activated by caspase-3 and by kinases. In this investigation, we examined whether Xkr8 would modulate ADAM17 activity under apoptotic and non-apoptotic conditions. Overexpression of Xkr8 in HEK293T cells led to significantly increased caspase-dependent as well as PMA-induced release of EREG and TGF-alpha. Conversely, siRNA-mediated downregulation of Xkr8 in colorectal Caco-2 cancer cells led to decreased PS externalization upon induction of apoptosis, which was accompanied by reduced shedding of endogenously expressed EREG and reduced cell survival. We conclude that Xkr8 shares with conventional scramblases the propensity to upmodulate the ADAM-sheddase function. Liberation of growth factors could serve a rescue function in cells on the pathway to apoptotic death.

2.
Mol Cell Endocrinol ; 520: 111081, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33181234

RESUMO

During puberty, the mammary gland undergoes an intense growth, dependent on the interplay between the Epidermal Growth Factor Receptor (EGFR) in the stroma and different mammary epithelial receptors. We hypothesize that EGFR expressed in the mammary epithelium also has a role in puberty and the epithelial cells can self-sustain by EGFR-mediated autocrine signaling. We adopted mammary cell lines from different species, as in vitro model for the epithelium, and we observed that EGFR-signaling positively affects their survival and proliferation. Once deprived of external growth factors, mammary cells still showed strong Erk 1/2 phosphorylation, abolished upon EGFR inhibition, coupled with a further reduction in survival and proliferation. Based on gene expression analysis, three EGFR-ligands (AREG, EREG and HBEGF) are likely to mediate this autocrine signaling. In conclusion, internal EGFR-activating signals sustain mammary epithelial cell proliferation and survival in vitro.


Assuntos
Células Epiteliais/citologia , Células Epiteliais/metabolismo , Receptores ErbB/metabolismo , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Humanas/citologia , Transdução de Sinais , Animais , Comunicação Autócrina , Bovinos , Ciclo Celular , Linhagem Celular , Proliferação de Células , Sobrevivência Celular , Receptores ErbB/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Queratina-14/metabolismo , Queratina-18/metabolismo , Ligantes , Camundongos , Receptor ErbB-2/metabolismo , Especificidade da Espécie
3.
Growth Factors ; 37(5-6): 209-225, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31878812

RESUMO

The discovery of epidermal growth factor (EGF) and its receptor (EGFR) revealed the connection between EGF-like ligands, signaling from the EGFR family members and cancer. Over the next fifty years, analysis of EGFR expression and mutation led to the use of monoclonal antibodies to target EGFR in the treatment of metastatic colorectal cancer (mCRC) and this treatment has improved outcomes for patients. The use of the RAS oncogene mutational status has helped to refine patient selection for EGFR antibody therapy, but an effective molecular predictor of likely responders is lacking. This review analyzes the potential utility of measuring the expression, levels and activation of EGF-like ligands and associated processes as prognostic or predictive markers for the identification of patient risk and more effective mCRC therapies.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Família de Proteínas EGF/metabolismo , Mutação , Metástase Neoplásica/tratamento farmacológico , Anticorpos Monoclonais/metabolismo , Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/metabolismo , Receptores ErbB/metabolismo , Genes ras , Humanos , Ligantes , Seleção de Pacientes , Medicina de Precisão , Risco , Transdução de Sinais , Resultado do Tratamento
4.
Front Immunol ; 10: 2990, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31921216

RESUMO

Adoptive immunotherapy with ex vivo-expanded tumor-infiltrating lymphocytes (TILs) has achieved objective clinical responses in a significant number of patients with cancer. The failure of many patients to develop long-term tumor control may be, in part, due to exhaustion of transferred T cells in the presence of a hostile tumor microenvironment. In several tumor types, growth and survival of carcinoma cells appear to be sustained by a network of receptors/ligands of the ErbB family. We speculated that if transferred T cells could benefit from EGFR ligands produced by the tumor, they might proliferate better and exert their anti-tumor activities more efficiently. We found that CD8+ T cells transduced with a retrovirus to express EGFR responded to EGFR ligands activating the EGFR signaling pathway. These EGFR-expressing effector T cells proliferated better and produced more IFN-γ and TNF-α in the presence of EGFR ligands produced by tumor cells in vitro. EGFR-expressing CD8 T cells from OT-1 mice were more efficient killing B16-OVA cells than control OT-1 CD8 T cells. Importantly, EGFR-expressing OT-1 T cells injected into B16-OVA tumor bearing mice were recruited into the tumor, expressed lower levels of the exhaustion markers PD1, TIGIT, and LAG3, and were more efficient in delaying tumor growth. Our results suggest that genetic modification of CD8+ T cells to express EGFR might be considered in immunotherapeutic strategies based on adoptive transfer of anti-tumor T cells against cancers expressing EGFR ligands.


Assuntos
Transferência Adotiva , Linfócitos T CD8-Positivos , Receptores ErbB , Vetores Genéticos , Neoplasias , Retroviridae , Transdução Genética , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Linfócitos T CD8-Positivos/transplante , Linhagem Celular Tumoral , Receptores ErbB/genética , Receptores ErbB/imunologia , Feminino , Camundongos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/imunologia , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/terapia
5.
J Immunol Methods ; 459: 63-69, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29803775

RESUMO

BACKGROUND: Prior to large studies in breast cancer patients and healthy individuals we established a sensitive three-plex immunoassay to measure the EGFR ligands amphiregulin (AR), betacellulin (BTC) and transforming growth factor α (TGF-α) simultaneously in human serum samples. METHOD: The three-plex immunoassay was developed using single molecule array (Simoa) technology and requires only 20 µL of serum. RESULTS: AR, BTC and TGF-α were first established as three single-plex assays. Multiplexing the three single-plex assays showed no significant cross reactivity between the reagents. The concentrations of the ligands in serum samples showed correlations r2 ≥ 0.84 between the single-plex and three-plex methods. The three-plex assay demonstrated limit of detection levels at 0.16 ng/L for AR, 0.23 ng/L for BTC and 0.22 ng/L for TGF-α. Total coefficients of variations were 8.5%-31% for AR, 11%-21.8% for BTC and 12.4%-16.2% for TGF-α. Spiking experiments showed a mean recovery of 97% for AR, 86% for BTC and 81% for TGF-α. The concentrations of the EGFR ligands did not change significantly after series of freeze thaw cycles or incubation at 22 °C for up to 24 h. CONCLUSION: This robust three-plex assay with up to 40-fold increase in sensitivity relative to conventional ELISA is the first published method that has the required sensitivity to measure AR, BTC and TGF-α simultaneously in human blood samples.


Assuntos
Anfirregulina/sangue , Betacelulina/sangue , Imunoensaio/métodos , Imagem Individual de Molécula/métodos , Fator de Crescimento Transformador alfa/sangue , Ensaio de Imunoadsorção Enzimática , Humanos , Ligantes , Análise Serial de Proteínas
6.
Methods Mol Biol ; 1731: 57-64, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29318543

RESUMO

Rhomboids are intramembrane serine proteases that cleave their substrates within or immediately adjacent to their transmembrane domains, a process known as regulated intramembrane proteolysis. In eukaryotes, two main types of rhomboid proteases can be distinguished based on their subcellular localization: mitochondrial rhomboids and secretase-type rhomboids that target the secretory pathway. The latter class can cleave and release the extracellular domain of all epidermal growth factor-like proteins in Drosophila and can liberate epidermal growth factor (EGF) in mammals, in a process known as ectodomain shedding. These released EGFs can then activate the EGF receptor (EGFR). EGFR signaling is crucial for mammalian development and is often deregulated in human cancer. Here we describe a cell-based protocol for detecting the ability of rhomboid proteases to release EGFR ligands into the medium. First, cells are transfected with the corresponding protease- and substrate-expressing vectors; second, cells condition the medium and accumulate shed protein. After this, protein lysates from cells and media are prepared and Western blotting is performed to detect the EGFR ligands that have been released into the medium.


Assuntos
Receptores ErbB/metabolismo , Proteínas de Membrana/metabolismo , Serina Endopeptidases/metabolismo , Animais , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Fator de Crescimento Epidérmico/metabolismo , Ligantes , Mitocôndrias/metabolismo , Domínios Proteicos , Especificidade por Substrato
7.
Oncotarget ; 8(19): 31270-31287, 2017 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-28415726

RESUMO

Prostaglandin E2 (PGE2) interacts with tyrosine kinases receptor signaling in both tumor and stromal cells supporting tumor progression. Here we demonstrate that in non-small cell lung carcinoma (NSCLC) cells, A549 and GLC82, PGE2 promotes nuclear translocation of epidermal growth factor receptor (nEGFR), affects gene expression and induces cell growth. Indeed, cyclin D1, COX-2, iNOS and c-Myc mRNA levels are upregulated following PGE2 treatment. The nuclear localization sequence (NLS) of EGFR as well as its tyrosine kinase activity are required for the effect of PGE2 on nEGFR and downstream signaling activities. PGE2 binds its bona fide receptor EP3 which by activating SRC family kinases, induces ADAMs activation which, in turn, releases EGFR-ligands from the cell membrane and promotes nEGFR. Amphiregulin (AREG) and Epiregulin (EREG) appear to be involved in nEGFR promoted by the PGE2/EP3-SRC axis. Pharmacological inhibition or silencing of the PGE2/EP3/SRC-ADAMs signaling axis or EGFR ligands i.e. AREG and EREG expression abolishes nEGFR induced by PGE2. In conclusion, PGE2 induces NSCLC cell proliferation by EP3 receptor, SRC-ADAMs activation, EGFR ligands shedding and finally, phosphorylation and nEGFR. Since nuclear EGFR is a hallmark of cancer aggressiveness, our findings reveal a novel mechanism for the contribution of PGE2 to tumor progression.


Assuntos
Adenocarcinoma/metabolismo , Dinoprostona/metabolismo , Receptores ErbB/metabolismo , Neoplasias Pulmonares/metabolismo , Receptores de Prostaglandina E Subtipo EP3/metabolismo , Transdução de Sinais , Quinases da Família src/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Linhagem Celular Tumoral , Proliferação de Células , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Receptores ErbB/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Ligantes , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Ligação Proteica , Proteína Quinase C/metabolismo , Transporte Proteico , Proteínas Proto-Oncogênicas c-akt/metabolismo
8.
Biochem Biophys Res Commun ; 478(1): 39-45, 2016 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-27462018

RESUMO

The epidermal growth factor receptor (EGFR) is activated through binding to specific ligands and generates signals for proliferation, differentiation, migration, and cell survival. Recent data show the role of nuclear EGFR in tumors. Although many EGFR ligands are upregulated in cancers, little is known about their effects on EGFR nuclear translocation. We have compared the effects of six EGFR ligands (EGF, HB-EGF, TGF-α, ß-Cellulin, amphiregulin, and epiregulin) on nuclear translocation of EGFR, receptor phosphorylation, migration, and proliferation. Cell fractionation and confocal immunofluorescence detected EGFR in the nucleus after EGF, HB-EGF, TGF-α and ß-Cellulin stimulation in a dose-dependent manner. In contrast, amphiregulin and epiregulin did not generate nuclear translocation of EGFR. EGF, HB-EGF, TGF-α and ß-Cellulin showed correlations between a higher rate of wound closure and increased phosphorylation of residues in the carboxy-terminus of EGFR, compared to amphiregulin and epiregulin. The data indicate that EGFR is translocated to the nucleus after stimulation with EGF, HB-EGF, TGF-α and ß-Cellulin, and that these ligands are related to increased phosphorylation of EGFR tyrosine residues, inducing migration of SkHep-1 cells.


Assuntos
Núcleo Celular/metabolismo , Receptores ErbB/metabolismo , Transporte Ativo do Núcleo Celular , Betacelulina/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Fator de Crescimento Epidérmico/metabolismo , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/metabolismo , Humanos , Neoplasias/metabolismo , Fosforilação , Fator de Crescimento Transformador alfa/metabolismo
10.
Int J Biochem Cell Biol ; 51: 23-8, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24681003

RESUMO

Epidermal growth factor (EGF) receptor (EGFR) signalling regulates diverse cellular functions, promoting cell proliferation, differentiation, migration, cell growth and survival. EGFR signalling is critical during embryogenesis, in particular in epithelial development, and disruption of the EGFR gene results in epithelial immaturity and perinatal death. EGFR signalling also functions during wound healing responses through accelerating wound re-epithelialisation, inducing cell migration, proliferation and angiogenesis. Upregulation of EGFR signalling is often observed in carcinomas and has been shown to promote uncontrolled cell proliferation and metastasis. Therefore aberrant EGFR signalling is a common target for anticancer therapies. Various reports indicate that EGFR signalling primarily occurs at the plasma membrane and EGFR degradation following endocytosis greatly attenuates signalling. Other studies argue that EGFR internalisation is essential for complete activation of downstream signalling cascades and that endosomes can serve as signalling platforms. The aim of this review is to discuss current understanding of intersection between EGFR signalling and trafficking.


Assuntos
Receptores ErbB/metabolismo , Animais , Movimento Celular/fisiologia , Endocitose/fisiologia , Humanos , Ligantes , Camundongos , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...