Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cureus ; 11(6): e4928, 2019 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-31431834

RESUMO

Gp78/autocrine motility factor receptor (Gp78/AMFR) is a cancer-associated endoplasmic reticulum-localized E3 ubiquitin ligase and also the cell surface receptor for autocrine motility factor (AMF). The study objective was to determine the association between Gp78/AMFR and AMF endocytosis in thyroid cancer cells. Gp78/AMFR expression and AMF internalization were measured in differentiated thyroid cancer (DTC) and anaplastic thyroid cancer (ATC) cell lines and in freshly resected human papillary thyroid cancers (PTC) relative to benign thyroid tissue. Spheroid-like aggregates generated from explants of cancer, goiter, and collateral thyroid tissue were assessed for expression of cancer stem cell markers, surface Gp78/AMFR and AMF endocytosis. DTC cell lines showed elevated total and surface Gp78/AMFR and AMF internalization relative to ATC lines. Gp78/AMFR, Oct-4 and Sox-2 protein expression, Gp78/AMFR surface expression and AMF internalization were elevated in PTC-derived aggregates relative to fibroblasts. Elevated levels of Gp78/AMFR expression and AMF internalization in PTC were associated with expression of cancer stem cell markers. Gp78/AMFR expression and AMF uptake are more closely associated with DTC compared to benign thyroid lesions or ATC and with PTC-derived cancer stem-like cells.

2.
Am J Surg ; 209(5): 884-9; discussion 889, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25852009

RESUMO

BACKGROUND: Autocrine motility factor receptor (AMFR) has been linked to metastasis and tumorigenicity. The aim of this study was to evaluate expression and prognostic significance of AMFR in colorectal carcinoma. METHODS: AMFR expression was evaluated in 127 colon cancer specimens, 131 rectal cancer specimens, and 47 colonic and 25 rectal corresponding lymph node metastases. Clinicopathological correlates of prognostic significance were established by univariate and multivariate analysis. Spearman's correlation determined the association of expression between cancers and their metastases. RESULTS: AMFR was over-expressed by 22% of colon cancers and 18% of rectal cancers. AMFR over-expression correlated significantly with improved disease-free survival (DFS) (P < .05) in colon cancer and decreased DFS in corresponding nodal metastases. In rectal cancer, AMFR over-expression significantly correlated with decreased overall survival, DFS, and disease-specific survival (P < .001, P = .031, P = .005, respectively) and decreased overall survival in corresponding metastases. CONCLUSION: AMFR may serve as a molecular prognosticator for colon cancer and rectal cancer.


Assuntos
Neoplasias Colorretais/metabolismo , Linfonodos/metabolismo , Estadiamento de Neoplasias , Receptores do Fator Autócrino de Motilidade/biossíntese , Biomarcadores Tumorais/biossíntese , Western Blotting , Linhagem Celular Tumoral , Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/secundário , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Linfonodos/patologia , Metástase Linfática , Prognóstico , Estudos Retrospectivos
3.
J Cell Sci ; 126(Pt 15): 3295-304, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23690547

RESUMO

Gp78 is a cell surface receptor that also functions as an E3 ubiquitin ligase in the endoplasmic reticulum (ER)-associated degradation (ERAD) pathway. The Gp78 ligand, the glycolytic enzyme phosphoglucose isomerase (PGI; also called autocrine motility factor, AMF), functions as a cytokine upon secretion by tumor cells. AMF is internalized through a PI3K- and dynamin-dependent raft endocytic pathway to the smooth ER; however, the relationship between AMF and Gp78 ubiquitin ligase activity remains unclear. AMF uptake to the smooth ER is inhibited by the dynamin inhibitor, dynasore, is reduced in Gp78 knockdown cells and induces the dynamin-dependent downregulation of its cell surface receptor. AMF uptake is Rac1-dependent and is inhibited by expression of dominant-negative Rac1 and the Rac1 inhibitor NSC23766, and is therefore distinct from Cdc42- and RhoA-dependent raft endocytic pathways. AMF stimulates Rac1 activation, but this is reduced by dynasore treatment and is absent in Gp78-knockdown cells; therefore, AMF activities require Gp78-mediated endocytosis. AMF also prevents Gp78-induced degradation of the mitochondrial fusion proteins, mitofusin 1 and 2 in a dynamin-, Rac1- and phosphoinositide 3-kinase (PI3K)-dependent manner. Gp78 induces mitochondrial clustering and fission in a manner dependent on GP78 ubiquitin ligase activity, and this is also reversed by uptake of AMF. The raft-dependent endocytosis of AMF, therefore, promotes Rac1-PI3K signaling that feeds back to promote AMF endocytosis and also inhibits the ability of Gp78 to target the mitofusins for degradation, thereby preventing Gp78-dependent mitochondrial fission. Through regulation of an ER-localized ubiquitin ligase, the raft-dependent endocytosis of AMF represents an extracellular regulator of mitochondrial fusion and dynamics.


Assuntos
Glucose-6-Fosfato Isomerase/metabolismo , Mitocôndrias/metabolismo , Receptores do Fator Autócrino de Motilidade/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Neoplasias da Mama , Linhagem Celular Tumoral , Endocitose , Retículo Endoplasmático/metabolismo , Degradação Associada com o Retículo Endoplasmático , Fibrossarcoma , Glucose-6-Fosfato Isomerase/genética , Humanos , Mitocôndrias/genética , Receptores do Fator Autócrino de Motilidade/genética , Transdução de Sinais , Transfecção , Proteínas rac1 de Ligação ao GTP/genética
4.
FEMS Immunol Med Microbiol ; 61(2): 205-17, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21204997

RESUMO

Previous studies have suggested that internalization of the Escherichia coli STb enterotoxin in human and rat intestinal epithelial cells is involved in STb pathogenesis, but toxin uptake in porcine jejunum epithelium, the in vivo target tissue, still remains elusive. Using flow cytometry, we studied the internalization of fluorescein isothiocyanate-labelled STb in porcine intestinal epithelial IPEC-J2 and murine fibroblast NIH-3T3 cell lines. In contrast to the selective pronase resistance of STb in NIH-3T3 cells at 37 °C, but not at 4 °C, indicative of toxin internalization, most of the toxin was pronase-sensitive at both temperatures in IPEC-J2 cells, indicating reduced uptake, but significant cell surface binding. Actin reorganization is required for STb internalization by NIH-3T3 cells, confirming STb endocytosis in these cells. The toxin receptor, sulfatide, could not explain these internalization differences because both cell lines possessed surface sulfatide and internalized antisulfatide antibodies over time at 37 °C. Inhibition of lipid rafts endocytosis, known to contain sulfatide, with methyl-ß-cyclodextrin or genistein, did not influence toxin uptake by either cell line. STb internalization is therefore differentially regulated depending on the cell type, possibly by factors other than sulfatide. Although a small STb fraction could be internalized by porcine intestinal epithelial cells, our findings suggest the ability of STb to induce, from the cell surface, intracellular signalling leading to fluid secretion in porcine intestinal epithelium.


Assuntos
Toxinas Bacterianas/metabolismo , Endocitose , Enterotoxinas/metabolismo , Células Epiteliais/metabolismo , Animais , Proteínas de Escherichia coli , Fibroblastos/metabolismo , Corantes Fluorescentes , Camundongos , Coloração e Rotulagem/métodos , Suínos
5.
J Cell Mol Med ; 13(9B): 3218-25, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19438805

RESUMO

Ganglioside GM1-bound cholera toxin-B sub-unit (CT-b) enters the cell via clathrin-coated pits and dynamin-independent non-caveolar raft-dependent endocytosis. Caveolin-1 (Cav1), associated with caveolae formation, is a negative regulator of non-caveolar raft-dependent endocytosis. In mammary epithelial tumour cells deficient for Mgat5, Cav1 is stably expressed at levels below the threshold for caveolae formation, forming stable oligomerized Cav1 microdomains or scaffolds that were shown to suppress EGFR signalling and reduce the plasma membrane diffusion rate of both EGFR and CT-b. Below threshold levels of Cav1 also inhibit the dynamin-dependent raft-mediated endocytosis of CT-b to the Golgi indicating that Cav1-negative regulation of raft-dependent endocytosis is caveolae independent. Inhibition of CT-b internalization does not require Cav1 phosphorylation but does require an intact Cav1 scaffolding domain. By flow cytometry, both over-expression of Cav1 and the dynamin K44A mutant block CT-b internalization from the plasma membrane defining a dynamin-dependent raft pathway for CT-b endocytosis in these cells. However, only minimal co-localization between CT-b and Cav1 is observed. These results suggest that Cav1 regulates raft-dependent internalization of CT-b indirectly via a mechanism that requires the Cav1 scaffolding domain and the formation of oligomerized Cav1 microdomains but not caveolae.


Assuntos
Caveolina 1/biossíntese , Toxina da Cólera/metabolismo , Regulação da Expressão Gênica , Microdomínios da Membrana/metabolismo , Animais , Membrana Celular/metabolismo , Clatrina/química , Endocitose , Complexo de Golgi/metabolismo , Ligantes , Microdomínios da Membrana/química , Camundongos , Camundongos Transgênicos , Mutação , Estrutura Terciária de Proteína , Transdução de Sinais
6.
PLoS One ; 3(10): e3597, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18974847

RESUMO

BACKGROUND: Autocrine motility factor/phosphoglucose isomerase (AMF/PGI) is the extracellular ligand for the gp78/AMFR receptor overexpressed in a variety of human cancers. We showed previously that raft-dependent internalization of AMF/PGI is elevated in metastatic MDA-435 cells, but not metastatic, caveolin-1-expressing MDA-231 cells, relative to non-metastatic MCF7 and dysplastic MCF10A cells suggesting that it might represent a tumor cell-specific endocytic pathway. METHODOLOGY/PRINCIPAL FINDINGS: Similarly, using flow cytometry, we demonstrate that raft-dependent endocytosis of AMF/PGI is increased in metastatic HT29 cancer cells expressing low levels of caveolin-1 relative to metastatic, caveolin-1-expressing, HCT116 colon cells and non-metastatic Caco-2 cells. Therefore, we exploited the raft-dependent internalization of AMF/PGI as a potential tumor-cell specific targeting mechanism. We synthesized an AMF/PGI-paclitaxel conjugate and found it to be as efficient as free paclitaxel in inducing cytotoxicity and apoptosis in tumor cells that readily internalize AMF/PGI compared to tumor cells that poorly internalize AMF/PGI. Murine K1735-M1 and B16-F1 melanoma cells internalize FITC-conjugated AMF/PGI and are acutely sensitive to AMF/PGI-paclitaxel mediated cytotoxicity in vitro. Moreover, following in vivo intratumoral injection, FITC-conjugated AMF/PGI is internalized in K1735-M1 tumors. Intratumoral injection of AMF/PGI-paclitaxel induced significantly higher tumor regression compared to free paclitaxel, even in B16-F1 cells, known to be resistant to taxol treatment. Treatment with AMF/PGI-paclitaxel significantly prolonged the median survival time of tumor bearing mice. Free AMF/PGI exhibited a pro-survival role, reducing the cytotoxic effect of both AMF/PGI-paclitaxel and free paclitaxel suggesting that AMF/PGI-paclitaxel targets a pathway associated with resistance to chemotherapeutic agents. AMF/PGI-FITC uptake by normal murine spleen and thymus cells was negligible both in vitro and following intravenous injection in vivo where AMF/PGI-FITC was selectively internalized by subcutaneous B16-F1 tumor cells. CONCLUSIONS/SIGNIFICANCE: The raft-dependent endocytosis of AMF/PGI may therefore represent a tumor cell specific endocytic pathway of potential value for drug delivery to tumor cells.


Assuntos
Antineoplásicos/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Endocitose/fisiologia , Glucose-6-Fosfato Isomerase/metabolismo , Microdomínios da Membrana/fisiologia , Neoplasias/metabolismo , Animais , Células CACO-2 , Carcinoma/metabolismo , Carcinoma/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Células HCT116 , Células HT29 , Humanos , Melanoma/metabolismo , Melanoma/patologia , Microdomínios da Membrana/metabolismo , Camundongos , Neoplasias/tratamento farmacológico , Paclitaxel/administração & dosagem , Paclitaxel/química , Receptores do Fator Autócrino de Motilidade , Receptores de Citocinas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
7.
Cancer Res ; 68(20): 8210-20, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18922892

RESUMO

Rho/ROCK signaling and caveolin-1 (Cav1) are implicated in tumor cell migration and metastasis; however, the underlying molecular mechanisms remain poorly defined. Cav1 was found here to be an independent predictor of decreased survival in breast and rectal cancer and significantly associated with the presence of distant metastasis for colon cancer patients. Rho/ROCK signaling promotes tumor cell migration by regulating focal adhesion (FA) dynamics through tyrosine (Y14) phosphorylation of Cav1. Phosphorylated Cav1 is localized to protrusive domains of tumor cells and Cav1 tyrosine phosphorylation is dependent on Src kinase and Rho/ROCK signaling. Increased levels of phosphorylated Cav1 were associated with elevated GTP-RhoA levels in metastatic tumor cells of various tissue origins. Stable expression and knockdown studies of Cav1 in tumor cells showed that phosphorylated Cav1 expression stimulates Rho activation, stabilizes FAK association with FAs, and promotes cell migration and invasion in a ROCK-dependent and Src-dependent manner. Tyrosine-phosphorylated Cav1, therefore, functions as an effector of Rho/ROCK signaling in the regulation of FA turnover and, thereby, tumor cell migration and invasion. These studies define a feedback loop between Rho/ROCK, Src, and phosphorylated Cav1 in tumor cell protrusions, identifying a novel function for Cav1 in tumor metastasis that may contribute to the poor prognosis of some Cav1-expressing tumors.


Assuntos
Caveolina 1/fisiologia , Adesões Focais , Invasividade Neoplásica , Neoplasias/patologia , Proteínas rho de Ligação ao GTP/fisiologia , Quinases Associadas a rho/fisiologia , Caveolina 1/análise , Linhagem Celular Tumoral , Movimento Celular , Proteína-Tirosina Quinases de Adesão Focal/fisiologia , Humanos , Metástase Neoplásica , Neoplasias/química , Fosforilação , Transdução de Sinais , Análise Serial de Tecidos , Quinases da Família src/fisiologia
8.
J Cell Biol ; 180(6): 1261-75, 2008 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-18347068

RESUMO

Both tyrosine-phosphorylated caveolin-1 (pY14Cav1) and GlcNAc-transferase V (Mgat5) are linked with focal adhesions (FAs); however, their function in this context is unknown. Here, we show that galectin-3 binding to Mgat5-modified N-glycans functions together with pY14Cav1 to stabilize focal adhesion kinase (FAK) within FAs, and thereby promotes FA disassembly and turnover. Expression of the Mgat5/galectin lattice alone induces FAs and cell spreading. However, FAK stabilization in FAs also requires expression of pY14Cav1. In cells lacking the Mgat5/galectin lattice, pY14Cav1 is not sufficient to promote FAK stabilization, FA disassembly, and turnover. In human MDA-435 cancer cells, Cav1 expression, but not mutant Y14FCav1, stabilizes FAK exchange and stimulates de novo FA formation in protrusive cellular regions. Thus, transmembrane crosstalk between the galectin lattice and pY14Cav1 promotes FA turnover by stabilizing FAK within FAs defining previously unknown, interdependent roles for galectin-3 and pY14Cav1 in tumor cell migration.


Assuntos
Caveolina 1/metabolismo , Membrana Celular/metabolismo , Movimento Celular/fisiologia , Adesões Focais/metabolismo , Galectina 3/metabolismo , Invasividade Neoplásica/fisiopatologia , Sequência de Aminoácidos/fisiologia , Animais , Caveolina 1/química , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Membrana Celular/ultraestrutura , Citoesqueleto/metabolismo , Citoesqueleto/ultraestrutura , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Adesões Focais/ultraestrutura , Humanos , Substâncias Macromoleculares/metabolismo , Camundongos , N-Acetilglucosaminiltransferases/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fosforilação , Ligação Proteica/fisiologia , Transporte Proteico/fisiologia , Tirosina/metabolismo
9.
J Biol Chem ; 282(40): 29305-13, 2007 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-17690101

RESUMO

Autocrine motility factor (AMF) is internalized via a receptor-mediated, dynamin-dependent, cholesterol-sensitive raft pathway to the smooth endoplasmic reticulum that is negatively regulated by caveolin-1. Expression of AMF and its receptor (AMFR) is associated with tumor progression and malignancy; however, the extent to which the raft-dependent uptake of AMF is tumor cell-specific has yet to be addressed. By Western blot and cell surface fluorescence-activated cell sorter (FACS) analysis, AMFR expression is increased in tumorigenic MCF7 and metastatic MDA-231 and MDA-435 breast cancer cell lines relative to dysplastic MCF10A mammary epithelial cells. AMF uptake, determined by FACS measurement of protease-insensitive internalized fluorescein-conjugated AMF, was increased in MCF7 and MDA-435 cells relative to MCF-10A and caveolin-1-expressing MDA-231 cells. Uptake of fluorescein-conjugated AMF was dynamin-dependent, methyl-beta-cyclodextrin- and genistein-sensitive, reduced upon overexpression of caveolin-1 in MDA-435 cells, and increased upon short hairpin RNA reduction of caveolin-1 in MDA-231 cells. Tissue microarray analysis of invasive primary human breast carcinomas showed that AMFR expression had no impact on survival but did correlate significantly with expression of phospho-Akt. Phospho-Akt expression was increased in AMF-internalizing MCF7 and MDA-435 breast carcinoma cells. AMF uptake in these cells was reduced by phosphatidylinositol 3-kinase inhibition but not by regulators of macropinocytosis such as amiloride, phorbol ester, or actin cytoskeleton disruption by cytochalasin D. The raft-dependent endocytosis of AMF therefore follows a distinct phosphatidylinositol 3-kinase-dependent pathway that is up-regulated in more aggressive tumor cells.


Assuntos
Neoplasias da Mama/patologia , Mama/metabolismo , Carcinoma/patologia , Regulação Neoplásica da Expressão Gênica , Glucose-6-Fosfato Isomerase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Neoplasias da Mama/metabolismo , Carcinoma/metabolismo , Linhagem Celular Tumoral , Separação Celular , Citocalasina D/farmacologia , Fragmentação do DNA , Endocitose , Citometria de Fluxo , Humanos , Microdomínios da Membrana/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...