Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Lipid Res ; 53(10): 2214-2225, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22822037

RESUMO

The submicroscopic spatial organization of cell surface receptors and plasma membrane signaling molecules is readily characterized by electron microscopy (EM) via immunogold labeling of plasma membrane sheets. Although various signaling molecules have been seen to segregate within plasma membrane microdomains, the biochemical identity of these microdomains and the factors affecting their formation are largely unknown. Lipid rafts are envisioned as submicron membrane subdomains of liquid ordered structure with differing lipid and protein constituents that define their specific varieties. To facilitate EM investigation of inner leaflet lipid rafts and the localization of membrane proteins therein, a unique genetically encoded reporter with the dually acylated raft-targeting motif of the Lck kinase was developed. This reporter, designated Lck-BAP-GFP, incorporates green fluorescent protein (GFP) and biotin acceptor peptide (BAP) modules, with the latter allowing its single-step labeling with streptavidin-gold. Lck-BAP-GFP was metabolically biotinylated in mammalian cells, distributed into low-density detergent-resistant membrane fractions, and was readily detected with avidin-based reagents. In EM images of plasma membrane sheets, the streptavidin-gold-labeled reporter was clustered in 20-50 nm microdomains, presumably representative of inner leaflet lipid rafts. The utility of the reporter was demonstrated in an investigation of the potential lipid raft localization of the epidermal growth factor receptor.


Assuntos
Proteína Tirosina Quinase p56(lck) Linfócito-Específica/genética , Microdomínios da Membrana/química , Animais , Biotinilação , Células COS , Células Cultivadas , Chlorocebus aethiops , Genes Reporter , Proteínas de Fluorescência Verde/genética , Humanos , Lipídeos/química , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/metabolismo , Microdomínios da Membrana/metabolismo , Proteínas de Membrana/metabolismo , Microscopia Eletrônica
2.
Mol Cell Biol ; 27(3): 888-98, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17101784

RESUMO

Hepatocyte growth factor-regulated tyrosine kinase substrate (Hrs) is an endosomal protein essential for the efficient sorting of activated growth factor receptors into the lysosomal degradation pathway. Hrs undergoes ligand-induced tyrosine phosphorylation on residues Y329 and Y334 downstream of epidermal growth factor receptor (EGFR) activation. It has been difficult to investigate the functional roles of phosphoHrs, as only a small proportion of the cellular Hrs pool is detectably phosphorylated. Using an HEK 293 model system, we found that ectopic expression of the protein Cbl enhances Hrs ubiquitination and increases Hrs phosphorylation following cell stimulation with EGF. We exploited Cbl's expansion of the phosphoHrs pool to determine whether Hrs tyrosine phosphorylation controls EGFR fate. In structure-function studies of Cbl and EGFR mutants, the level of Hrs phosphorylation and rapidity of apparent Hrs dephosphorylation correlated directly with EGFR degradation. Differential expression of wild-type versus Y329,334F mutant Hrs in Hrs-depleted cells revealed that one or both tyrosines regulate ligand-dependent Hrs degradation, as well as EGFR degradation. By modulating Hrs ubiquitination, phosphorylation, and protein levels, Cbl may control the composition of the endosomal sorting machinery and its ability to target EGFR for lysosomal degradation.


Assuntos
Receptores ErbB/metabolismo , Fosfoproteínas/metabolismo , Fosfotirosina/metabolismo , Processamento de Proteína Pós-Traducional , Animais , Complexos Endossomais de Distribuição Requeridos para Transporte , Fator de Crescimento Epidérmico/farmacologia , Humanos , Cinética , Camundongos , Peso Molecular , Proteínas Mutantes/metabolismo , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-cbl/química , Relação Estrutura-Atividade , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
3.
Biochem J ; 410(3): 585-94, 2008 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-18045238

RESUMO

EGF-R [EGF (epidermal growth factor) receptor] ligands can promote or inhibit cell growth. The biological outcome of receptor activation is dictated, at least in part, by ligand-specified patterns of endocytic trafficking. EGF-R trafficking downstream of the ligands EGF and TGF-alpha (transforming growth factor-alpha) has been investigated extensively. However, less is known about EGF-R fates induced by the ligands BTC (betacellulin) and AR (amphiregulin). We undertook comparative analyses to identify ligand-specific molecular events that regulate EGF-R trafficking and degradation. EGF (17 nM) and BTC (8.5 nM) induced significant EGF-R degradation, with or without ectopic expression of the ubiquitin ligase Cbl. Human recombinant AR (17 nM) failed to affect receptor degradation in either case. Notably, levels of ligand-induced EGF-R ubiquitination did not correlate strictly with receptor degradation. Dose-response experiments revealed that AR at a saturating concentration was a partial agonist at the EGF-R, with approx. 40% efficacy (relative to EGF) at inducing receptor tyrosine phosphorylation, ubiquitination and association with Cbl. EGF-R down-regulation and degradation also were compromised upon cell stimulation with AR (136 nM). These outcomes correlated with decreased degradation of the Cbl substrate and internalization inhibitor hSprouty2. Downstream of the hSprouty2 checkpoint in AR-stimulated cells, Cbl-free EGF-R was incorporated into endosomes from which Cbl-EGF-R complexes were excluded. Our results suggest that the AR-specific EGF-R fate results from decreased hSprouty2 degradation and reduced Cbl recruitment to underphosphorylated EGF-R, two effects that impair EGF-R trafficking to lysosomes.


Assuntos
Endossomos/metabolismo , Fator de Crescimento Epidérmico/fisiologia , Receptores ErbB/metabolismo , Glicoproteínas/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Anfirregulina , Animais , Sequência de Bases , Células COS , Linhagem Celular , Chlorocebus aethiops , Primers do DNA , Regulação para Baixo , Família de Proteínas EGF , Receptores ErbB/agonistas , Imunofluorescência , Humanos , Mutagênese Sítio-Dirigida , Ubiquitina/metabolismo
4.
Sci Signal ; 5(243): pe41, 2012 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-23012653

RESUMO

Excessive signaling by receptor tyrosine kinases (RTKs) can cause cancer. What molecular mechanisms normally control RTK signaling? Are they defective in tumors? If so, should therapeutics be developed to restore particular regulatory pathways to cancer cells? These questions have been approached through mechanistic studies of a prototypical RTK, the epidermal growth factor receptor (EGFR). EGFR signaling is mediated and regulated by both signaling and trafficking effectors. The amplitude of receptor-proximal signals changes as EGFRs move along the degradative trafficking pathway from the cell surface, to endosomes, and into lysosomes. To optimize therapeutic suppression of receptor oncogenicity, it may be crucial to target EGFRs that are signaling from a specific site in the trafficking pathway. Research suggests that EGFRs at the plasma membrane produce the bulk of the global transcriptional response to EGF. EGFRs localized between the internalization and early endosome fusion stages of the pathway enrich the expression of transcripts associated with cancer. EGFRs at later trafficking checkpoints controlled by the endosomal sorting complex required for transport (ESCRT) complexes II and III do not contribute substantially to the EGFR-mediated transcriptional response. These results suggest that therapeutics targeting the receptors at the earliest stages of degradative trafficking might be most effective.


Assuntos
Membrana Celular/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Endossomos/metabolismo , Receptores ErbB/metabolismo , Regulação da Expressão Gênica/fisiologia , Transdução de Sinais/fisiologia , Humanos , Modelos Biológicos , Transporte Proteico/fisiologia , Proteólise
5.
Sci Signal ; 2(102): ra86, 2009 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-20029031

RESUMO

Amino acid residues 1 to 434 of the E3 ubiquitin ligase Cbl control signaling of the epidermal growth factor receptor (EGFR) by enhancing its ubiquitination, down-regulation, and lysosomal degradation. This region of Cbl comprises a tyrosine kinase-binding domain, a linker region, a really interesting new gene finger (RF), and a subset of the residues of the RF tail. In experiments with full-length alanine substitution mutants, we demonstrated that the RF tail of Cbl regulated biochemically distinct checkpoints in the endocytosis of EGFR. The Cbl- and ubiquitin-dependent degradation of the regulator of internalization hSprouty2 was compromised by the Val(431)--> Ala mutation, whereas the Cbl- and EGFR-dependent dephosphorylation or degradation of the endosomal trafficking regulator Hrs was compromised by the Phe(434)--> Ala mutation. Deregulated phosphorylation of Hrs correlated with inhibition of the fusion of early endosomes and of the degradation of EGFR. This study provides the first evidence that Cbl regulates receptor fate by controlling the fusion of sorting endosomes. We postulate that it does so by modulating the abundance of tyrosine-phosphorylated Hrs.


Assuntos
Regulação para Baixo/fisiologia , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Endossomos/metabolismo , Receptores ErbB/metabolismo , Fusão de Membrana/fisiologia , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Transdução de Sinais/fisiologia , Animais , Western Blotting , Células COS , Chlorocebus aethiops , Humanos , Imunoprecipitação , Microscopia de Fluorescência , Fosforilação
6.
Sci Signal ; 1(38): ra5, 2008 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-18812566

RESUMO

The ubiquitin ligase Nedd4 has been proposed to regulate a number of signaling pathways, but its physiological role in mammals has not been characterized. Here we present an analysis of Nedd4-null mice to show that loss of Nedd4 results in reduced insulin-like growth factor 1 (IGF-1) and insulin signaling, delayed embryonic development, reduced growth and body weight, and neonatal lethality. In mouse embryonic fibroblasts, mitogenic activity was reduced, the abundance of the adaptor protein Grb10 was increased, and the IGF-1 receptor, which is normally present on the plasma membrane, was mislocalized. However, surface expression of IGF-1 receptor was restored in homozygous mutant mouse embryonic fibroblasts after knockdown of Grb10, and Nedd4(-/-) lethality was rescued by maternal inheritance of a disrupted Grb10 allele. Thus, in vivo, Nedd4 appears to positively control IGF-1 and insulin signaling partly through the regulation of Grb10 function.


Assuntos
Fator de Crescimento Insulin-Like I/fisiologia , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais , Ubiquitina-Proteína Ligases/fisiologia , Animais , Membrana Celular/metabolismo , Células Cultivadas , Complexos Endossomais de Distribuição Requeridos para Transporte , Proteína Adaptadora GRB10/metabolismo , Insulina/fisiologia , Camundongos , Camundongos Mutantes , Ubiquitina-Proteína Ligases Nedd4 , Fosforilação
7.
Exp Cell Res ; 311(2): 281-93, 2005 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-16246327

RESUMO

Evolutionarily conserved sequences of the E3/protein-ubiquitin ligase Cbl regulate epidermal growth factor receptor (EGF-R) signaling and degradation. These sequences encompass Cbl's tyrosine kinase-binding domain, linker region, RING finger (RF), and an uncharacterized flank C-terminal to the RF (residues 420-436). The latter domain, designated the RF tail, extends beyond Cbl's ubiquitin-conjugating enzyme (Ubc)-binding domain and has no known function. We report structure-function studies evaluating the impact of Cbl RF tail truncations on EGF-R fate in HEK 293 cells. All of the truncation mutants exhibit greatly reduced binding to activated EGF-R and lack proline-rich sequences that mediate direct Cbl association with SH3 proteins such as Grb2, yet a subset of mutants collectively enhances EGF-R ubiquitination, downregulation, and degradation. Significantly, EGF-R degradation correlates better with RF tail-dependent degradation of the Cbl substrate Sprouty2 than with EGF-R ubiquitination: expression of the RF tail truncation mutant Cbl 1-433 enhanced EGF-R ubiquitination while impeding Sprouty2 degradation, and Cbl 1-433 failed to enhance EGF-R downregulation or degradation. Our results suggest that EGF-R fate is controlled by a checkpoint downstream of receptor ubiquitination whose regulation by the Cbl RF tail may require Sprouty2 degradation.


Assuntos
Receptores ErbB/metabolismo , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-cbl/genética , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Sequência de Aminoácidos , Células Cultivadas , Sequência Conservada , Regulação para Baixo , Receptores ErbB/genética , Evolução Molecular , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana , Dados de Sequência Molecular , Mutação , Fosforilação , Estrutura Terciária de Proteína , Deleção de Sequência , Tirosina/metabolismo , Ubiquitina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA