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











Base de dados
Intervalo de ano de publicação
1.
Cancer Drug Resist ; 2(3): 539-549, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-35582586

RESUMO

The drug gefitinib, a specific inhibitor of EGFR tyrosine kinase, has been shown to suppress the activation of EGFR signaling for survival and cell proliferation in non-small cell lung cancer cell lines. For many years, EGFR endocytosis has served as a model for investigating ligand-induced, receptor-mediated endocytosis. On EGF stimulation, EGFR is internalized and transported via clathrin-coated vesicles to early endosomes, and EGFR then recruits and phosphorylates signaling molecules, leading to the activation of downstream signaling such as MAPK/PI3K/AKT pathways-an important mechanism for regulating cell growth. Once delivered to the lysosomes, EGFR is degraded to terminate intracellular EGFR signaling via endocytosis; this process is known as receptor downregulation. Therefore, the endocytosis of EGFR is closely related with attenuation of intracellular EGFR signaling. Alternatively, EGFR is returned to cell surface from early endosomes for the continued signaling. Previous reports revealed that a competent EGF-induced endocytosis of EGFR followed by its rapid downregulation efficiently proceeds in the gefitinib-sensitive NSCLC cell lines. In contrast, gefitinib-resistant cell lines showed that EGFR endocytosis is impaired and the internalized EGFR is aggregated in the early endosomes, which is associated with the overexpressed sorting nexin 1 (SNX1), initially identified as a protein that interacts with EGFR. Thus dysregulated EGFR endocytosis is implicated in gefitinib resistance, as it leads to uncontrolled signal transduction. At present, the therapeutic relevance of EGFR endocytosis with regard to drug resistance in lung cancer has not been clarified. This review focused on the mechanism for EGFR endocytosis associated with SNX1 trafficking in gefitinib-resistant lung cancer cells.

3.
Int J Oncol ; 48(5): 1895-906, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26983447

RESUMO

The MET tyrosine kinase receptor and its ligand, hepatocyte growth factor (HGF), are known to be overexpressed in a variety of malignant tumor cells, and are implicated in the development of gefitinib-resistance in human non-small cell lung cancer (NSCLC) cells. Ephedrae herba was previously reported to prevent HGF-induced cancer cell motility by directly suppressing HGF/MET signaling through the inhibition of MET tyrosine kinase, and treatment with its extract also considerably reduced MET protein levels. To further investigate the mechanism underlying the Ephedrae herba-induced inhibition of MET phosphorylation as well as its degradation and subsequent disappearance, we examined the effect of Ephedrae herba on HGF-stimulated MET endocytosis and downregulation via early/late endocytic pathways in an NSCLC cell line. Using immunofluorescence microscopy, we found that pretreatment of cells with Ephedrae herba extract dramatically changed the intracellular distribution of plasma membrane-associated MET, and that the resultant MET staining was distributed throughout the cytoplasm. Pretreatment of the cells with Ephedrae herba extract also led to the rapid loss of MET and phosphorylated (p)-MET in HGF-stimulated cells. In contrast, inefficient endocytic delivery of MET and p-MET from early to late endosomes was observed in the absence of Ephedrae herba extract, since considerable amounts of the internalized MET accumulated in the early endosomes and were not delivered to lysosomes up to 1 h after HGF-stimulation. Furthermore, large amounts of MET and p-MET that had accumulated in late endosomes of Ephedrae herba-pretreated cells after HGF stimulation were observed along with bafilomycin A1. Therefore, we inferred that degradation of MET occurred in the late endosome/lysosome pathway. Moreover, western blot analysis revealed the accelerated degradation of MET and p-MET proceeds in cells pretreated with Ephedrae herba extract. Collectively, our results suggest that some components of Ephedrae herba have a novel role in promoting HGF-stimulated MET and p-MET endocytosis followed by its downregulation, likely mediated by the early/late endocytic pathways.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Ephedra/química , Fator de Crescimento de Hepatócito/metabolismo , Neoplasias Pulmonares/metabolismo , Extratos Vegetais/farmacologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Antineoplásicos Fitogênicos/farmacologia , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Endocitose/efeitos dos fármacos , Gefitinibe , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Fosforilação/efeitos dos fármacos , Quinazolinas/farmacologia , Transdução de Sinais/efeitos dos fármacos
4.
Int J Oncol ; 48(3): 1251-7, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26782071

RESUMO

Solid tumors are characterized by acidic extracellular pH (pHe). The present study examined the contribution of small GTP-binding proteins to phospholipase D (PLD) activation of acidic pHe-induced matrix metalloproteinase-9 (MMP-9) production. Acidic pHe-induced MMP-9 production was reduced by C3 exoenzyme, which inhibits the Rho family of GTPases; cytochalasin D, which inhibits actin reorganization; and simvastatin, which inhibits geranylgeranylation of Rho. Small interfering RNA (siRNA) against RhoA, but not against Rac1 or Cdc42, significantly inhibited acidic pHe induction of MMP-9. Pull-down assays showed that acidic pHe increased the activated form of RhoA. Forced expression of constitutively active RhoA induced MMP-9 production, even at neutral pHe. RhoA siRNA also reduced acidic pHe induced PLD activity. Specific inhibition of PLD1 and Pld1 gene knockout significantly reduced acidic pHe-induced MMP-9 expression. In contrast, PLD2 inhibition or knockout had no effect on MMP-9 expression. These findings suggested that RhoA-PLD1 signaling is involved in acidic pHe induction of MMP-9.


Assuntos
Metaloproteinase 9 da Matriz/metabolismo , Melanoma Experimental/metabolismo , Fosfolipase D/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Actinas/química , Animais , Citocalasina D/química , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Concentração de Íons de Hidrogênio , Metaloproteinase 9 da Matriz/genética , Camundongos , Metástase Neoplásica , Neuropeptídeos/metabolismo , Regiões Promotoras Genéticas , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Sinvastatina/química , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP
5.
Int J Oncol ; 46(4): 1721-9, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25653196

RESUMO

The receptor tyrosine kinase epidermal growth factor receptor (EGFR) and its ligand epidermal growth factor (EGF) are known to play important roles in malignant tumor cells, and the EGFR signaling pathway is one of the most important targets in various tumors, including non-small cell lung cancer (NSCLC). We reported recently that an aberration in certain steps of EGF-stimulated phosphorylated epidermal growth factor receptor (pEGFR) endocytic trafficking from the early endosomes to the late endosomes occurs in the gefitinib-resistant NSCLC cells, in which large amounts of sorting nexin 1 (SNX1) are colocalized with EGFR in the aggregated early endosomes where the internalized pEGFR is also accumulated of these cells. To further investigate the role of SNX1 in EGF­stimulated pEGFR endocytosis, followed by downstream signaling leading to the activation of phosphatidylinositol 3-kinase (PI3K)--the serine/threonine kinase AKT pathway, we examined the effect of depletion of SNX1 knock-down expression by siRNA and an inhibition of targeting membrane recycling using monensin. Using immunofluorescence, we observed an efficient endocytic transport of pEGFR from early endosomes to late endosomes/lysosomes after EGF-stimulation in the cells transfected with siRNA­SNX1, whereas the delayed endocytic delivery of pEGFR was evident in the siRNA-control-transfected cells. Furthermore, a large amount of endocytosed pEGFR was accumulated in the presence of monensin in the early endosomes of the SNX1 knock-down cells. In western blot analysis, EGF stimulation of both control and cells transfected with siRNA-SNX1 resulted in rapid phosphorylation of EGFR and enhanced AKT phosphorylation. Monensin-dependent inhibition of AKT phosphorylation was stronger in SNX1 knock-down cells than in controls. In contrast, however, monensin had no effect on AKT phosphorylation triggered by activation of the MET receptor tyrosine kinase. Collectively, we suggest that EGF-stimulated recycling of EGFR to the plasma membrane induces downstream signaling leading to AKT phosphorylation. Suppression of EGFR membrane recycling by SNX1 appears to be critical for the activation of EGFR/PI3K/AKT signaling pathway in human lung cancer cells.


Assuntos
Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/metabolismo , Neoplasias Pulmonares/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Nexinas de Classificação/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Resistencia a Medicamentos Antineoplásicos , Endocitose/efeitos dos fármacos , Gefitinibe , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Pulmonares/patologia , Monensin/farmacologia , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Nexinas de Classificação/genética
6.
Int J Oncol ; 44(2): 412-26, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24297483

RESUMO

The receptor tyrosine kinase MET and its ligand HGF are known to be overexpressed in malignant tumor cells, and they have been implicated in gefitinib resistance in lung cancer cells. We recently found that sorting nexin 1 (SNX1), a protein that interacts with EGFR, exhibited negative regulation of EGFR trafficking out of early to late endosomes in gefitinib-resistant NSCLC cell lines. To investigate the role of SNX1 on HGF-stimulated MET endocytosis and its downregulation via the early/late endocytic pathway, we examined the effect of depletion of SNX1 expression by siRNA in NSCLC cells. Using immunofluorescence, we found that the silencing of SNX1 by siRNA caused a dramatic change in the intracellular distribution of plasma membrane-associated MET and that the resultant MET staining was spread throughout the cytoplasm, and it co-localized well with the endocytosed Texas red-labeled transferrin in the siRNA-SNX1-transfected cells. We also found efficient MET phosphorylation and rapid endocytic delivery of phosphorylated MET from early endosomes to late endosomes in the siRNA-SNX1-transfected cells. By contrast, the siRNA-control transfected cells showed inefficient endocytic delivery of phosphorylated MET from early endosomes to late endosomes. Furthermore, large amounts of phosphorylated MET that had accumulated in late endosomes were seen even after 60 min of HGF-stimulation in the presence of bafilomycin A1, indicating that degradation of phosphorylated MET proceeds in a late endosome/lysosome pathway. Western blot analysis revealed that depletion of SNX1 by siRNA induced a maximal and dramatic increase in phosphorylated MET at 60 min, followed by an accelerated degradation of phosphorylated MET after HGF stimulation in the cells. Taken together, we suggest that SNX1 plays a suppressive role in the regulation of HGF-stimulated MET/phosphorylated MET endocytosis and downregulation via the early/late endocytic pathway in the gefitinib-resistant NSCLC cells.


Assuntos
Endocitose/efeitos dos fármacos , Fator de Crescimento de Hepatócito/farmacologia , Neoplasias Pulmonares/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Quinazolinas/farmacologia , RNA Interferente Pequeno/genética , Nexinas de Classificação/fisiologia , Antineoplásicos/farmacologia , Western Blotting , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Resistencia a Medicamentos Antineoplásicos , Endossomos/efeitos dos fármacos , Endossomos/metabolismo , Gefitinibe , Inativação Gênica , Humanos , Neoplasias Pulmonares/genética , Microscopia de Fluorescência , Fosforilação/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-met/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas
7.
Int J Oncol ; 41(4): 1520-30, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22859339

RESUMO

Gefitinib is known to suppress the activation of EGFR signaling, which is required for cell survival and proliferation in non-small cell lung cancer (NSCLC) cell lines. We previously demonstrated that the gefitinib-sensitive NSCLC cell line PC9 shows efficient ligand-induced endocytosis of phosphorylated EGFR (pEGFR). In contrast, the gefitinib-resistant NSCLC cell lines QG56 and A549 showed internalized pEGFR accumulation in the aggregated early endosomes, and this was associated with SNX1, a protein that interacts with and enhances the degradation of EGFR upon EGF stimulation. In the present study, to investigate the role of SNX1 on EGF-stimulated EGFR/pEGFR endocytosis via the endocytic pathway, we examined the effect of depletion of SNX1 expression by siRNA in human NSCLC cell lines. Using immunofluorescence, we demonstrated that transfection of SNX1 siRNA into gefitinib-resistant NSCLC cells resulted in the disappearance of a large amounts of SNX1 staining. In addition, upon 15 min of EGF stimulation, we observed an efficient EGFR phosphorylation and a rapid endocytic delivery of pEGFR from early endosomes to late endosomes. Further, western blot analysis revealed that silencing of SNX1 expression by siRNA in the gefitinib-resistant cells leads to an accelerated degradation of EGFR along with a dramatic increase in the amounts of pEGFR after EGF stimulation. Based on these findings, we suggest that SNX1 is involved in the negative regulation of ligand-induced EGFR phosphorylation and mediates EGFR/pEGFR trafficking out of early endosomes for targeting to late endosomes/lysosomes via the early/late endocytic pathway in human lung cancer cells.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Receptores ErbB/genética , Quinazolinas/administração & dosagem , Nexinas de Classificação/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Endocitose/efeitos dos fármacos , Endossomos/efeitos dos fármacos , Endossomos/metabolismo , Receptores ErbB/biossíntese , Gefitinibe , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Fosforilação/efeitos dos fármacos , RNA Interferente Pequeno/genética , Transdução de Sinais/efeitos dos fármacos , Nexinas de Classificação/antagonistas & inibidores , Nexinas de Classificação/biossíntese
8.
Nature ; 487(7406): 235-8, 2012 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-22722837

RESUMO

It is generally accepted that the direct connection from the motor cortex to spinal motor neurons is responsible for dexterous hand movements in primates. However, the role of the 'phylogenetically older' indirect pathways from the motor cortex to motor neurons, mediated by spinal interneurons, remains elusive. Here we used a novel double-infection technique to interrupt the transmission through the propriospinal neurons (PNs), which act as a relay of the indirect pathway in macaque monkeys (Macaca fuscata and Macaca mulatta). The PNs were double infected by injection of a highly efficient retrograde gene-transfer vector into their target area and subsequent injection of adeno-associated viral vector at the location of cell somata. This method enabled reversible expression of green fluorescent protein (GFP)-tagged tetanus neurotoxin, thereby permitting the selective and temporal blockade of the motor cortex­PN­motor neuron pathway. This treatment impaired reach and grasp movements, revealing a critical role for the PN-mediated pathway in the control of hand dexterity. Anti-GFP immunohistochemistry visualized the cell bodies and axonal trajectories of the blocked PNs, which confirmed their anatomical connection to motor neurons. This pathway-selective and reversible technique for blocking neural transmission does not depend on cell-specific promoters or transgenic techniques, and is a new and powerful tool for functional dissection in system-level neuroscience studies.


Assuntos
Mãos/fisiologia , Neurônios Motores/fisiologia , Neurociências , Animais , Dependovirus/genética , Proteínas de Fluorescência Verde/metabolismo , Macaca , Metaloendopeptidases/metabolismo , Córtex Motor/fisiologia , Transmissão Sináptica/genética , Transmissão Sináptica/fisiologia , Toxina Tetânica/metabolismo
9.
J Mol Histol ; 42(5): 427-42, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21847509

RESUMO

The small GTPase RhoA and its downstream effectors, the Rho-associated kinase (Rho-kinase) family, are known to regulate cell morphology, motility, and tumor progression via the regulation of actin cytoskeleton rearrangement. In the present study, we evaluated the role of Rho-kinase in the intracellular endocytic trafficking of ligand-induced phosphorylated epidermal growth factor receptor (pEGFR). We investigated the time course of the internalization fate of EGF-induced pEGFR via the early/late endocytic pathway in human fibrosarcoma cell line HT1080 cells using Y-27632, a selective Rho-kinase inhibitor. We found, using confocal immunofluorescence microscopy and Western blot analysis, a large accumulation of pEGFR in the nuclei of HT1080 cells. In contrast, we observed decreased amounts of the pEGFR-positive staining in the nuclei along with an accumulation of cytosolic pEGFR staining when the cells were incubated for 15-30 min in the presence of Y-27632, implying that an aberrant endocytic trafficking mechanism of pEGFR occurs in HT1080 cells whereby pEGFR might be selectively translocated into the nucleus. Moreover, we demonstrated that after 15-min of stimulation with Texas Red-EGF, increasing numbers of pEGFR-positive staining that had colocalized with Texas Red-EGF-positive punctate staining were seen in the cytoplasm of HT1080 cells but after 30-min of stimulation, most of this staining had disappeared from the cytoplasm and a large accumulation of pEGFR-positive staining appeared in the nucleus. Thus, nuclear accumulation of pEGFR appears to occur in an EGF-dependent manner. In contrast, such nuclear pEGFR-positive staining was not seen in the Y-27632-treated cells. Furthermore, silencing of RhoA or Rho-kinases I/II by sequence specific siRNAs considerably inhibited the EGF-dependent nuclear accumulation of pEGFR. Collectively, these results provide the first evidence that Rho-kinase signaling pathway plays a suppressive role in the intracellular vesicle trafficking of pEGFR via the endocytic pathway and that an increased Rho-kinase activity leads to the attenuation of the normal endocytic vesicular traffic of pEGFR via the early/late endocytic pathway, instead causing pEGFR to be trafficked out of the endocytic vesicles into the nucleus.


Assuntos
Endocitose , Receptores ErbB/metabolismo , Fibrossarcoma/enzimologia , Fibrossarcoma/patologia , Transdução de Sinais , Quinases Associadas a rho/metabolismo , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/metabolismo , Amidas/farmacologia , Western Blotting , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Endocitose/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , Inativação Gênica/efeitos dos fármacos , Humanos , Ligantes , Fosforilação/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Piridinas/farmacologia , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/antagonistas & inibidores , Proteínas rac1 de Ligação ao GTP/metabolismo , Quinases Associadas a rho/antagonistas & inibidores
10.
J Comp Neurol ; 516(6): 493-506, 2009 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-19672995

RESUMO

To investigate the neural basis for functional recovery of the cerebral cortex following spinal cord injury, we measured the expression of growth-associated protein 43 (GAP-43), which is involved in the process of synaptic sprouting. We determined the GAP-43 mRNA expression levels in the sensorimotor cortical areas of macaque monkeys with a unilateral lesion of the lateral corticospinal tract (l-CST) at the C4/C5 level of the cervical cord and compared them with the levels in the corresponding regions of intact monkeys. Lesioned monkeys recovered finger dexterity during the first months after surgery, and the GAP-43 mRNA levels increased in layers II-III in primary motor areas (M1), bilaterally. Double-labeling analysis of the lesioned monkeys showed that GAP-43 mRNA was expressed strongly in excitatory neurons but only rarely in inhibitory interneurons. Expression also increased in the medium-sized (area, 500-1,000 microm(2)) and large pyramidal cells (area, >1,000 microm(2)) in layer V of the bilateral M1. The increased expression of GAP-43 mRNA in the M1 contralateral to the lesion was more prominent during the early recovery stage than during the late recovery stage. In addition, GAP-43 mRNA increased in layers II-III of both the contralesional ventral premotor area and the primary somatosensory area. These results suggest that GAP-43 is involved in time-dependent and brain region-specific plastic changes after l-CST lesioning. The expression patterns imply that plastic changes occur not only in M1 but also in the broad associative cortical network, including the ventral premotor and primary sensory areas.


Assuntos
Lobo Frontal/metabolismo , Proteína GAP-43/metabolismo , Córtex Motor/metabolismo , Neurônios/metabolismo , Tratos Piramidais/lesões , Córtex Somatossensorial/metabolismo , Animais , Vértebras Cervicais , Feminino , Interneurônios/metabolismo , Macaca , Macaca mulatta , Masculino , Células Piramidais/metabolismo , RNA Mensageiro/metabolismo , Recuperação de Função Fisiológica , Fatores de Tempo
11.
Anim Reprod Sci ; 113(1-4): 114-24, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18614302

RESUMO

Full-grown oocytes arrested at germinal vesicle stage contain many dormant maternal mRNAs, and Aurora A has been reported to play a key role for the translation of these maternal mRNAs in Xenopus oocytes. Although the presence of Aurora A has been reported in mammals, the functions of Aurora A on the protein synthesis and the meiotic resumption have never been elucidated in mammalian oocytes. In the present study, the effects of porcine Aurora A on meiotic resumption of porcine oocytes were examined. At first, we cloned porcine Aurora A from total RNA of immature porcine oocytes by RT-PCR and obtained full-length cDNA that was 77%, 86% and 54% homologous with mouse, human and Xenopus Aurora A, respectively. The Aurora A mRNA and large amounts of protein were present throughout maturation period in porcine oocytes. The overexpression of porcine Aurora A by the mRNA injection into immature porcine oocytes had no effects on Cyclin B synthesis and meiotic resumption. Therefore we constructed a mutated Aurora A (AA-Aurora A), which was replaced the expecting inhibitory phosphorylation sites, serines 283 and 284, to non-phosphorylatable alanines. The oocytes expressed AA-Aurora A were accelerated their Cyclin B synthesis and Rsk phosphorylation, an indicator of Mos synthesis, then their meiotic resumption was promoted significantly. These results suggest for the first time in mammalian oocytes that mammalian Aurora A stimulates the protein synthesis and promotes the meiotic resumption. In addition, we identified the inhibitory phosphorylation sites of porcine Aurora A, and indicate the presence of phosphorylation-dependent regulation mechanisms in mammalian Aurora A.


Assuntos
Ciclina B/biossíntese , Meiose/genética , Oócitos/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas c-mos/biossíntese , Sus scrofa , Sequência de Aminoácidos , Animais , Aurora Quinase A , Aurora Quinases , Sequência de Bases , Clonagem Molecular , Ciclina B/genética , Feminino , Dados de Sequência Molecular , Oócitos/metabolismo , Fosforilação , Biossíntese de Proteínas/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-mos/genética , RNA Mensageiro/metabolismo , Homologia de Sequência de Aminoácidos , Sus scrofa/genética , Sus scrofa/metabolismo , Sus scrofa/fisiologia
12.
Mol Cancer ; 7: 42, 2008 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-18492291

RESUMO

Gefitinib (Iressa)-a specific inhibitor of epidermal growth factor receptor (EGFR) tyrosine kinase-has been shown to suppress the activation of EGFR signaling required for cell survival and proliferation in non-small cell lung cancer (NSCLC) cell lines. We recently provided novel evidence that gefitinib-sensitive PC9 cells show normal endocytosis of EGFR: internalized EGF-EGFR complexes were transported to late endosomes/lysosomes 15 min after EGF stimulation, and then degraded within the lysosomes. However, gefitinib-resistant QG56 cells showed internalized EGFR accumulation in early endosomes after 60 min of internalization, instead of its trafficking to lysosomes, indicating an aberration in some steps of EGF-EGFR trafficking from the early endosomes to late endosomes/lysosomes. Therefore, we postulate that impairment in some steps of EGF-EGFR trafficking from early endosomes to late endosomes/lysosomes might confer gefitinib-resistance in NSCLC cell lines. To further substantiate the detailed internalization mechanism of gefitinib-sensitive and gefitinib-resistant cells, using confocal immunofluorescence microscopy, we examined the endocytic trafficking of phosphorylated EGFR (pEGFR) in the absence or presence of gefitinib. In PC9 and QG56 cells without EGF stimulation, a large number of pEGFR-positive small vesicular structures not colocalized with late endosomes/lysosomes were spread throughout the cytoplasm, and some pEGFR staining was distributed in the nucleus. This implies a novel intracellular trafficking pathway for pEGFR from cytoplasmic vesicles to the nucleus. Furthermore, an aggregated vesicular structure of early endosomes was observed in the perinuclear region of QG56 cells; it was revealed to be associated with SNX1, originally identified as a protein that interacts with EGFR. Therefore, we confirmed our previous data that an aberration in some steps of EGF-EGFR trafficking from the early endosomes to late endosomes/lysosomes occurs in QG56 cells. Furthermore, in PC9 cells, efficient phosphorylation of EGFR and rapid internalization of pEGFR was observed at 3 min after EGF stimulation; these internalized pEGFR-positive vesicles were trafficked to late endosomes at 15 min, indicating rapid trafficking of EGF-pEGFR complexes from early to late endosomes in PC9 cells. Gefitinib treatment strongly reduced the phosphorylation level of EGFR, and subsequent endocytosis of EGFR was significantly suppressed in PC9 cells. In contrast, in QG56 cells, EGFR trafficking via the early endocytic pathway was basically impaired; therefore, gefitinib appeared to slightly suppress the internalization of pEGFR. Collectively, our data provide novel evidence that extensive impairment in pEGFR endocytosis via the early endocytic pathway might confer gefitinib-resistance in QG56 cells.


Assuntos
Endocitose/efeitos dos fármacos , Receptores ErbB/metabolismo , Quinazolinas/farmacologia , Transporte Biológico/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Endossomos/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Gefitinibe , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Lisossomos/metabolismo , Microscopia de Fluorescência , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Nexinas de Classificação , Proteínas de Transporte Vesicular/metabolismo
13.
J Reprod Dev ; 53(6): 1151-7, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17693700

RESUMO

A high cyclic adenosine monophosphate (cAMP) level in fully-grown immature oocytes prevents meiotic resumption. In Xenopus, inhibitory cAMP is synthesized within oocytes depending on a stimulatory alpha-subunit of G-protein (Gsalpha). In the present study, we examined whether ooplasmic Gsalpha is involved in meiotic arrest of porcine oocytes. First, we studied the presence of Gsalpha molecules in porcine oocytes by immunoblotting, and this suggested the presence of reported isoforms (45 and 48 kDa) not only in cumulus cells but also in porcine oocytes. Then we injected an anti-Gsalpha antibody into porcine immature oocytes and found that inhibition of ooplasmic Gsalpha functions significantly promoted germinal vesicle breakdown of the oocytes, whose spontaneous meiotic resumption was prevented by 3-isobutyl-l-methylxanthine (IBMX) treatment. Although cyclin B synthesis and M-phase promoting factor (MPF) activation were largely prevented until 30 h of culture in IBMX-treated oocytes, injection of anti-Gsalpha antibody into these oocytes partially recovered cyclin B synthesis and activated MPF activity at 30 h. These results suggest that meiotic resumption of porcine oocytes is prevented by ooplasmic Gsalpha, which may stimulate cAMP synthesis within porcine oocytes, and that synthesized cAMP prevents meiotic resumption of oocytes through the signaling pathways involved in MPF activation.


Assuntos
Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Meiose/fisiologia , Oócitos/citologia , Oócitos/metabolismo , Animais , Anticorpos/farmacologia , Células Cultivadas , AMP Cíclico/metabolismo , Ciclina B/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/imunologia , Fator Promotor de Maturação/metabolismo , Transdução de Sinais/fisiologia , Sus scrofa
14.
Histochem Cell Biol ; 127(5): 541-53, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17361439

RESUMO

The drug gefitinib (Iressa), which is a specific inhibitor of EGFR tyrosine kinase, has been shown to suppress the activation of EGFR signaling for survival and proliferation in non-small cell lung cancer (NSCLC) cell lines. A recent study demonstrated rapid down-regulation of ligand-induced EGFR in a gefitinib-sensitive cell line and inefficient down-regulation of EGFR in a gefitinib-resistant cell line in the exponential phase of growth; this implies that each cell type employs a different unknown down-regulation mechanism occurs. However, the mechanism of drug sensitivity to gefitinib remains unclear. In this study, to further substantiate the effect of gefitinib on the EGFR down-regulation pathway and to understand the detailed internalization mechanism of gefitinib-sensitive PC9 and gefitinib-resistant QG56 cell lines, we examined the internalization of Texas red-EGF in the absence or presence of gefitinib in both cell lines. The distribution of internalized Texas red-EGF, early endosomes, and late endosomes/lysosomes was then assessed by confocal immunofluorescence microscopy. Here, we provide novel evidence that efficient endocytosis of EGF-EGFR occurs via the endocytic pathway in the PC9 cells, because the internalized Texas red-EGF-positive small punctate vesicles were transported to the late endosomes/lysosomes and then degraded within the lysosomes after 60 min of internalization. Additionally, gefitinib exerted a strong inhibitory effect on the endocytosis of EGFR in PC9 cells, and the internalization rate of EGFR from the plasma membrane via the early endosomes to the late endosomes/lysosomes was considerably delayed. This indicates that gefitinib efficiently suppresses ligand-stimulated endocytosis of EGFR via the early/late endocytic pathway in PC9 cells. In contrast, the internalization rate of ligand-induced EGFR was not significantly changed by gefitinib in QG56 cells because even in the absence of gefitinib, internalized EGFR accumulation was noted in the early and late endosomes after 60 min of internalization instead of its delivery to the lysosomes in QG56 cells. This suggests that the endocytic machinery of EGFR might be basically impaired at the level of the early/late endosomes. Taken together, this is the first report demonstrating that the suppressive effect of gefitinib on the endocytosis of EGFR is much stronger with PC9 cells than QG56 cells. Thus, impairment in some steps of the EGF-EGFR traffic out of early endosomes toward the late endosomes/lysosomes might confer gefitinib-resistance in NSCLC cell lines.


Assuntos
Endocitose/efeitos dos fármacos , Receptores ErbB/metabolismo , Quinazolinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Antineoplásicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Catepsina D/metabolismo , Linhagem Celular Tumoral , Membrana Celular/química , Membrana Celular/metabolismo , Resistencia a Medicamentos Antineoplásicos , Endocitose/fisiologia , Endossomos/química , Endossomos/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/análise , Receptores ErbB/antagonistas & inibidores , Gefitinibe , Humanos , Proteínas de Membrana Lisossomal/análise , Lisossomos/metabolismo , Microscopia de Fluorescência , Transporte Proteico/efeitos dos fármacos , Proteínas Qa-SNARE/análise , Receptores Depuradores/análise , Transferrina/metabolismo
15.
Histochem Cell Biol ; 126(5): 627-38, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16763828

RESUMO

We have previously shown that overexpression of LIM kinase1 (LIMK1) resulted in a marked retardation of the internalization of the receptor-mediated endocytic tracer, Texas red-labeled epidermal growth factor (EGF) in low-invasive human breast cancer cell MCF-7. We thereby postulate that LIMK1 signaling plays an important role in the regulation of ligand-induced endocytosis of EGF receptor (EGFR) in tumor cells by reorganizing and influencing actin-filament dynamics. In the present study, we further assessed the effect of wild-type LIMK1, a kinase-deficient dominant negative mutant of LIMK1 (DN-LIMK1) and an active, unphosphorylatable cofilin mutant (S3A cofilin) on internalization of EGF-EGFR in MDA-MB-231, a highly invasive human breast cancer cell line. We demonstrate here that a marked delay in the receptor-mediated internalization of Texas red-labeled EGF was observed in the wild-type LIMK1 transfectants, and that most of the internalized EGF staining were accumulated within transferrin receptor-positive early endosomes even after 30 min internalization. In contrast, the expression of dominant-negative LIMK1 mutant rescued the efficient endocytosis of Texas red-EGF, and large amounts of Texas red-EGF staining already reached LIMPII-positive late endosomes/lysosomal vacuoles after 15 min internalization. We further analyzed the effect of S3A cofilin mutant on EGFR trafficking, and found an efficient delivery of Texas red-EGF into late endosomes/lysosomes at 15-30 min after internalization. Taken together, our novel findings presented in this paper implicate that LIMK1 signaling indeed plays a pivotal role in the regulation of EGFR trafficking through the endocytic pathway in invasive tumor cells.


Assuntos
Fatores de Despolimerização de Actina/fisiologia , Neoplasias da Mama/metabolismo , Endocitose/fisiologia , Receptores ErbB/metabolismo , Proteínas Quinases/fisiologia , Fatores de Despolimerização de Actina/genética , Catepsina D/metabolismo , Feminino , Humanos , Immunoblotting , Quinases Lim , Proteínas de Membrana Lisossomal/metabolismo , Transporte Proteico , Receptores Depuradores/metabolismo , Transdução de Sinais , Transfecção , Células Tumorais Cultivadas
16.
Eur J Cell Biol ; 83(7): 369-80, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15503860

RESUMO

LIM kinase (LIMK) plays a critical role in stimulus-induced remodeling of the actin cytoskeleton by linking signals from the Rho family GTPases to changes in cofilin activity. Recent studies have shown an important role for LIMK1 signaling in tumor cell invasion through regulating actin dynamics. In this study, we investigate the role of LIMK1 in intracellular vesicle trafficking of lysosomes/endosomes. We analyzed by confocal immunofluorescence microscopy the cellular distribution of lysosomal proteins and the endocytosis of an endocytic tracer, epidermal growth factor (EGF), in LIMK1-transfected cells. We found in these cells an abnormal dispersed translocation of lysosomes stained for LIMPII and cathepsin D throughout the cytoplasm. The small punctate structures that stained for these lysosomal proteins were redistributed to the periphery of the cell. Computational 3D-image analysis of confocal immunofluorescence micrographs further demonstrated that these vesicles did not colocalize with the transferrin receptor, an early endosomal marker. Furthermore, LIMPII-positive lysosomes did not colocalize with early endosomes labeled with endocytosed Texas red-transferrin. These results indicate that there is no mixing between dispersed lysosomes and early endosomes in the LIMK1-transfected cells. Moreover, LIMK1 overexpression resulted in a marked retardation in the receptor-mediated internalization of Texas red-labeled EGF in comparison with mock-transfected cells. At 30 min after internalization, most of the Texas red-EGF staining overlapped with LIMPII-positive late endosomes/lysosomes in mock-transfected cells, whereas in LIMK1 transfectants only a small fraction of internalized EGF colocalized with LIMPII-positive structures in the perinuclear region. Taken together, the findings presented in this paper suggest that LIMK1 has a role in regulating vesicle trafficking of lysosomes and endosomes in invasive tumor cells.


Assuntos
Actinas/metabolismo , Neoplasias da Mama/enzimologia , Citoesqueleto/metabolismo , Endossomos/metabolismo , Lisossomos/metabolismo , Proteínas Quinases/metabolismo , Transporte Biológico Ativo , Neoplasias da Mama/patologia , Catepsina D/metabolismo , Linhagem Celular Tumoral , Endocitose/fisiologia , Endossomos/patologia , Fator de Crescimento Epidérmico/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Quinases Lim , Proteínas de Membrana Lisossomal , Lisossomos/patologia , Proteínas de Membrana/metabolismo , Microscopia Confocal , Microscopia de Fluorescência , Metástase Neoplásica/fisiopatologia , Proteínas/metabolismo , Receptores Depuradores , Sialoglicoproteínas/metabolismo , Transdução de Sinais/efeitos dos fármacos
17.
Anat Rec A Discov Mol Cell Evol Biol ; 274(1): 798-806, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12923890

RESUMO

Previous studies have concentrated on elucidating the subcellular localization of major histocompatibility (MHC) class II molecules mainly in B cells, macrophages, and dendritic cells. Despite very rich cell-surface expression of MHC class II molecules by cortical thymic epithelial cells (cTECs), little is known regarding the expression of these molecules by cTECs at the subcellular level. In the present study we focused on the identification and characterization of MHC class II compartments (MIICs) in cTECs in situ by immunogold electron microscopy (IEM). We found that MHC class II molecules were located exclusively in the cytoplasmic vacuoles, and we identified these MHC class II molecule-containing cytoplasmic vacuoles as MIICs in cTECs. These MIICs were immunopositive for early endosomal, late endosomal, and lysosomal markers. Moreover, in these MIICs, MHC class II molecules were colocalized with cathepsin L, H2-DM, class II-associated invariant chain (Ii), and class II-associated invariant chain peptide (CLIP). Similarly, Ii molecules were colocalized with endosomal and lysosomal markers, cathepsin L, and H2-DM in the vacuoles. Taken together, these results suggest that MIICs in cTECs represent conventional endocytic compartments. The colocalization of MHC class II molecule or Ii with cathepsin L and H2-DM in the MIICs suggests that MIICs in cTECs may be sites of Ii degradation and peptide loading.


Assuntos
Antígenos de Histocompatibilidade Classe II/metabolismo , Timo/metabolismo , Animais , Antígenos de Diferenciação de Linfócitos B/metabolismo , Biomarcadores/análise , Catepsina L , Catepsinas/metabolismo , Compartimento Celular , Cisteína Endopeptidases , Endossomos/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/ultraestrutura , Feminino , Lisossomos/metabolismo , Microscopia Eletrônica , Microscopia Imunoeletrônica , Ratos , Ratos Endogâmicos , Timo/citologia , Timo/ultraestrutura , Distribuição Tecidual
18.
Pathol Oncol Res ; 9(2): 83-95, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12858212

RESUMO

Small GTPase Rho and its downstream effectors, ROCK family of Rho-associated serine-threonine kinases, are thought to participate in cell morphology, motility, and tumor progression through regulating the rearrangement of actin cytoskeleton. Here we present evidence that transfection of human breast cancer cells with cDNA encoding a dominant active mutant of ROCK causes dispersal of lysosomal vesicles throughout the cytoplasm without perturbing the machinery of the endocytic pathway. The intracellular distribution of lysosomes and endocytosed transferrin, an early endosomal marker, were further assessed by confocal immunofluorescence microscopy. In the active ROCK transfected cells the lysosomal proteins, cathepsin D, LIMPII, and LAMP1, were found throughout the cytoplasm in dispersed small vesicles, which were accessible to the endocytosed Texas Red-labeled transferrin. 3D-image analysis of lysosomal distribution in the active ROCK transfectants revealed abundant punctate signals in the peripheral region of the basal plasma membrane. Cells expressing vector alone did not exhibit these alterations. Wortmannin, a phosphatidylinositol 3-kinase inhibitor, induced LIMPII-positive/ transferrin negative large vacuoles in the perinuclear region, and disappearence of the dispersed small vesicular structures. To our knowledge, this is the first evidence that increasing ROCK expression contributes to selective cellular dispersion of lysosomes in invasive breast cancer cells.


Assuntos
Neoplasias da Mama/enzimologia , Membranas Intracelulares/enzimologia , Lisossomos/enzimologia , Proteínas de Membrana , Proteínas Serina-Treonina Quinases/metabolismo , Transporte Proteico , Sialoglicoproteínas , Androstadienos/farmacologia , Antígenos CD/metabolismo , Antígenos CD36/metabolismo , Catepsina D/metabolismo , Endocitose/fisiologia , Endossomos/efeitos dos fármacos , Endossomos/enzimologia , Endossomos/ultraestrutura , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteína 1 de Membrana Associada ao Lisossomo , Proteínas de Membrana Lisossomal , Lisossomos/ultraestrutura , Microscopia Confocal , Microscopia de Fluorescência , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Receptores Depuradores , Receptores da Transferrina/imunologia , Transdução de Sinais , Transfecção , Transferrina/metabolismo , Células Tumorais Cultivadas , Regulação para Cima , Wortmanina , Quinases Associadas a rho
19.
J Electron Microsc (Tokyo) ; 51(3): 173-81, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12113625

RESUMO

To better comprehend the thymic microenvironment, it is necessary to identify the antigenic profile of cortical thymic epithelial cells (cTECs) that are involved in the development of major histocompatibility complex (MHC)-restricted T cells. Ultrastructurally, cTECs can be classified into four morphologically distinct subtypes: subcapsular/perivascular (EC1), pale (EC2), intermediate (EC3) and dark (EC4) cells. Several immunohistochemical studies were done on cTECs at the light and electron microscopic levels, but not with reference to the above subtypes. In the present paper, we analysed the expression of MHC class II antigen and cathepsin L by individual cTEC subtypes at the electron microscopic level. We show that (1) MHC class II antigens are expressed on the cell surfaces except on the basal surface of EC1, both on the cell surface and in intracytoplasmic vacuoles of EC2, and only in the intracytoplasmic vacuoles of EC3 and EC4, and (2) that cathepsin L is expressed strongly and uniformly throughout the cytoplasm of EC2, but weakly and non-uniformly in the cytoplasm of EC1, EC3 and EC4. These results show that MHC class II antigen expression and cathepsin L expression is heterogeneous in cTEC subtypes and suggest that EC2 might play a significant role in the development of CD4+ T cells.


Assuntos
Catepsinas/análise , Células Epiteliais/química , Células Epiteliais/ultraestrutura , Antígenos de Histocompatibilidade Classe II/análise , Microscopia Imunoeletrônica/métodos , Timo/química , Timo/ultraestrutura , Animais , Células Apresentadoras de Antígenos/química , Células Apresentadoras de Antígenos/citologia , Células Apresentadoras de Antígenos/ultraestrutura , Catepsina L , Cisteína Endopeptidases , Células Epiteliais/citologia , Feminino , Imuno-Histoquímica , Ratos , Timo/citologia
20.
Histochem J ; 34(5): 189-213, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12587997

RESUMO

Small GTPase RhoA regulates signal transduction from receptors in the membrane to a variety of cellular events related to cell morphology, motility, cytoskeletal dynamics, cytokinesis, and tumour progression, but it is unclear how RhoA regulates intracellular membrane dynamics of lysosomes. We showed previously by confocal immunofluorescence microscopy that the transfection of dominant active RhoA in MM1 cells causes the dispersal translocation of lysosomes stained for cathepsin D throughout the cytoplasm. Y-27632, a selective inhibitor of p160ROCK, impeded the cellular redistribution of lysosomes and promoted reclustering of lysosomes toward the perinuclear region. Here we have further investigated whether the acidic lysosomal vesicles dispersed throughout the cytoplasm are applied to the early endosomes in the endocytic pathway, and we demonstrate that the dispersed lysosomes were accessible to endocytosed molecule such as dextran, and their acidity was not changed, as determined by increased accumulation of the acidotropic probe LysoTracker Red. Brefeldin A did not induce the tabulation of these dispersed lysosomes, but it caused early endosomes to form an extensive tubular network. The dispersed lysosomes associated with cathepsin D and LIMPII were not colocalized with early endosomes, and these vesicles were not inaccessible to the endocytosed anti-transferrin receptor antibody. Moreover, wortmannin, an inhibitor of phosphatidylinositol 3-kinase, induced a dramatic change in LIMPII-containing structures in which LIMPII-positive swollen large vacuoles were increased and small punctate structures disappeared in the cytoplasm. These swollen vacuoles were not doubly positive for LIMPII and transferrin receptor, and were not inaccessible to the internalized anti-transferrin receptor antibody. Therefore, our novel findings presented in this paper indicate that RhoA activity causes a selective translocation of lysosomes without perturbing the machinery of endocytic pathway.


Assuntos
Carcinoma Hepatocelular/enzimologia , Membranas Intracelulares/enzimologia , Lisossomos/enzimologia , Proteínas rho de Ligação ao GTP/metabolismo , Androstadienos/farmacologia , Brefeldina A/farmacologia , Antígenos CD36/metabolismo , Dextranos/metabolismo , Endocitose/fisiologia , Endossomos/efeitos dos fármacos , Endossomos/enzimologia , Endossomos/ultraestrutura , Inibidores Enzimáticos/farmacologia , Lisossomos/efeitos dos fármacos , Lisossomos/ultraestrutura , Microscopia Confocal , Microscopia de Fluorescência , Receptores da Transferrina/imunologia , Transdução de Sinais , Transfecção , Células Tumorais Cultivadas , Wortmanina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA