Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Methods Mol Biol ; 2524: 281-290, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35821479

RESUMO

Bioluminescence (BL) has been widely used to quantitatively monitor various biological phenomena. Here, we describe a protocol for preparing and using cells expressing exosomes labeled with luciferase. The BL of the culture medium of these cells is proportional to the number of secreted exosome particles obtained by well-established nanoparticle tracking analysis, allowing easy, rapid, and sensitive quantification of exosomes in vitro and in vivo. This method, designated the ExoLuc system, is a powerful tool for analyzing the molecular mechanisms of exosome biosynthesis, secretion, uptake, and biodistribution.


Assuntos
Exossomos , Biomarcadores/metabolismo , Diagnóstico por Imagem , Exossomos/metabolismo , Luciferases/genética , Luciferases/metabolismo , Distribuição Tecidual
2.
Sci Rep ; 12(1): 6674, 2022 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-35461323

RESUMO

Cancer cells secrete aberrantly large amounts of extracellular vesicles (EVs) including exosomes, which originate from multivesicular bodies (MVBs). Because EVs potentially contribute to tumor progression, EV inhibitors are of interest as novel therapeutics. We screened a fungal natural product library. Using cancer cells engineered to secrete luciferase-labeled EVs, we identified asteltoxin, which inhibits mitochondrial ATP synthase, as an EV inhibitor. Low concentrations of asteltoxin inhibited EV secretion without inducing mitochondrial damage. Asteltoxin attenuated cellular ATP levels and induced AMPK-mediated mTORC1 inactivation. Consequently, MiT/TFE transcription factors are translocated into the nucleus, promoting transcription of lysosomal genes and lysosome activation. Electron microscopy analysis revealed that the number of lysosomes increased relative to that of MVBs and the level of EVs decreased after treatment with asteltoxin or rapamycin, an mTORC1 inhibitor. These findings suggest that asteltoxin represents a new type of EV inhibitor that controls MVB fate.


Assuntos
Proteínas Quinases Ativadas por AMP , Vesículas Extracelulares , Lisossomos , Alvo Mecanístico do Complexo 1 de Rapamicina , Pironas , Serina-Treonina Quinases TOR
3.
Cancer Sci ; 113(4): 1264-1276, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35108425

RESUMO

Cancer cells secrete large amounts of extracellular vesicles (EVs) originating from multivesicular bodies (MVBs). Mature MVBs fuse either with the plasma membrane for release as EVs, often referred as to exosomes or with lysosomes for degradation. However, the mechanisms regulating MVB fate remain unknown. Here, we investigated the regulators of MVB fate by analyzing the effects of signaling inhibitors on EV secretion from cancer cells engineered to secrete luciferase-labeled EVs. Inhibition of the oncogenic MEK/ERK pathway suppressed EV release and activated lysosome formation. MEK/ERK-mediated lysosomal inactivation impaired MVB degradation, resulting in increased EV secretion from cancer cells. Moreover, MEK/ERK inhibition prevented c-MYC expression and induced the nuclear translocation of MiT/TFE transcription factors, thereby promoting the activation of lysosome-related genes, including the gene encoding a subunit of vacuolar-type H+ -ATPase, which is responsible for lysosomal acidification and function. Furthermore, c-MYC upregulation was associated with lysosomal gene downregulation in MEK/ERK-activated renal cancer cells/tissues. These findings suggest that the MEK/ERK/c-MYC pathway controls MVB fate and promotes EV production in human cancers by inactivating lysosomal function.


Assuntos
Vesículas Extracelulares , ATPases Vacuolares Próton-Translocadoras , Vesículas Extracelulares/metabolismo , Genes myc , Humanos , Lisossomos/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Oncogenes , ATPases Vacuolares Próton-Translocadoras/metabolismo
4.
Nat Commun ; 13(1): 83, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-35013169

RESUMO

Phosphoinositides are a family of membrane lipids essential for many biological and pathological processes. Due to the existence of multiple phosphoinositide regioisomers and their low intracellular concentrations, profiling these lipids and linking a specific acyl variant to a change in biological state have been difficult. To enable the comprehensive analysis of phosphoinositide phosphorylation status and acyl chain identity, we develop PRMC-MS (Phosphoinositide Regioisomer Measurement by Chiral column chromatography and Mass Spectrometry). Using this method, we reveal a severe skewing in acyl chains in phosphoinositides in Pten-deficient prostate cancer tissues, extracellular mobilization of phosphoinositides upon expression of oncogenic PIK3CA, and a unique profile for exosomal phosphoinositides. Thus, our approach allows characterizing the dynamics of phosphoinositide acyl variants in intracellular and extracellular milieus.


Assuntos
Classe I de Fosfatidilinositol 3-Quinases/genética , Metaboloma , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositóis/metabolismo , Neoplasias da Próstata/metabolismo , Animais , Cromatografia de Afinidade , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Exossomos/química , Exossomos/metabolismo , Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Masculino , Espectrometria de Massas , Camundongos , Células PC-3 , PTEN Fosfo-Hidrolase/deficiência , Fosfatidilinositóis/química , Fosfatidilinositóis/classificação , Fosfatidilinositóis/isolamento & purificação , Próstata/química , Próstata/efeitos dos fármacos , Próstata/patologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Pirimidinas/farmacologia , Quinazolinas/farmacologia , Estereoisomerismo
5.
Front Mol Biosci ; 9: 1049402, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36710884

RESUMO

Cancer-derived small extracellular vesicles (sEVs) are multifunctional particles with a lipid bilayer structure that are involved in cancer progression, such as malignant proliferation, distant metastasis, and cancer immunity evasion. The separation protocol used to isolate sEVs is an important process and thus, several have been developed, including ultracentrifugation (UC), size exclusion chromatography (SEC), and affinity purification using antibodies against sEV surface antigens. However, the effects of different separation methods on sEV components have not been adequately examined. Here, we developed a semi-automated system for collecting sEVs by combining SEC and preparative high-performance liquid chromatography and applied it to metabolome analysis. The developed SEC system could recover sEVs more efficiently and non-destructively than UC, suggesting that it is an appropriate recovery method for metabolic analysis and reflects biological conditions. Furthermore, using the developed SEC system, we performed metabolome analysis of sEVs from isocitrate dehydrogenase 1 (IDH)-mutated human colon HCT116 cells, which produce the oncogenic metabolite, 2-hydroxyglutaric acid (2-HG). IDH1-mutated HCT116 cells released significantly more sEVs than wild-type (WT) cells. The metabolomic profiles of IDH1 mutant and WT cells showed distinct differences between the cells and their sEVs. Notably, in IDH mutant cells, large amounts of 2-HG were detected not only in cells, but also in sEVs. These results indicate that the SEC system we developed has wide potential applications in sEVs research.

6.
Oncogene ; 40(23): 3989-4003, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33994542

RESUMO

The tumor microenvironment is deeply involved in the process of tumor growth and development. In this study, we focused on cancer-associated fibroblasts (CAFs) and their derived exosomes on the lymphoma microenvironment to uncover their clinical significance. CAFs were established from primary lymphoma samples, and exosomes secreted from CAFs were obtained by standard procedures. We then investigated the roles of CAFs and their derived exosomes in the survival and drug resistance of lymphoma cells. CAFs supported the survival of lymphoma cells through increased glycolysis, and the extent differed among CAFs. Exosomes were identified as a major component of the extracellular vesicles from CAFs, and they also supported the survival of lymphoma cells. The suppression of RAB27B, which is involved in the secretion of exosomes, using a specific siRNA resulted in reduced exosome secretion and decreased survival of lymphoma cells. Moreover, anti-pyrimidine drug resistance was induced in the presence of exosomes through the suppression of the pyrimidine transporter, equilibrative nucleoside transporter 2 (ENT2), and the suppression of ENT2 was significant in in vivo experiments and clinical samples. RNA sequencing analysis of miRNAs in exosomes identified miR-4717-5p as one of the most abundant miRNAs in the exosome, which suppressed the expression of ENT2 and induced anti-pyrimidine drug resistance in vitro. Our results suggest that exosomes including miR-4717-5p secreted from CAFs play a pivotal role in the lymphoma microenvironment, indicating that they are a promising therapeutic target.


Assuntos
Fibroblastos Associados a Câncer/metabolismo , Desoxicitidina/análogos & derivados , Exossomos/metabolismo , Linfoma/tratamento farmacológico , Animais , Antimetabólitos Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Citarabina/farmacologia , Desoxicitidina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Transportador Equilibrativo 2 de Nucleosídeo/metabolismo , Humanos , Linfoma/genética , Linfoma/metabolismo , Linfoma/patologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , MicroRNAs/genética , MicroRNAs/metabolismo , Cultura Primária de Células , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina
7.
Dev Cell ; 56(6): 842-859.e8, 2021 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-33756122

RESUMO

Extracellular vesicles (EVs) are classified as large EVs (l-EVs, or microvesicles) and small EVs (s-EVs, or exosomes). S-EVs are thought to be generated from endosomes through a process that mainly depends on the ESCRT protein complex, including ALG-2 interacting protein X (ALIX). However, the mechanisms of l-EV generation from the plasma membrane have not been identified. Membrane curvatures are generated by the bin-amphiphysin-rvs (BAR) family proteins, among which the inverse BAR (I-BAR) proteins are involved in filopodial protrusions. Here, we show that the I-BAR proteins, including missing in metastasis (MIM), generate l-EVs by scission of filopodia. Interestingly, MIM-containing l-EV production was promoted by in vivo equivalent external forces and by the suppression of ALIX, suggesting an alternative mechanism of vesicle formation to s-EVs. The MIM-dependent l-EVs contained lysophospholipids and proteins, including IRS4 and Rac1, which stimulated the migration of recipient cells through lamellipodia formation. Thus, these filopodia-dependent l-EVs, which we named as filopodia-derived vesicles (FDVs), modify cellular behavior.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Movimento Celular , Endocitose , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Exossomos/metabolismo , Vesículas Extracelulares/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas de Neoplasias/metabolismo , Pseudópodes/metabolismo , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ciclo Celular/genética , Membrana Celular/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Células HEK293 , Humanos , Proteínas dos Microfilamentos/genética , Proteínas de Neoplasias/genética
8.
Sci Rep ; 10(1): 16616, 2020 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-33024173

RESUMO

Monitoring of exosome dynamics in living organisms is essential to demonstrate the real functions of cancer-derived exosomes. Currently, these have been elucidated in vitro or under non-physiological conditions in vivo in most cases. To overcome these limitations, we developed an imaging method using Antares2-mediated bioluminescence resonance energy transfer (BRET) for observing long-term accumulation of exosomes in vivo. Ectopic expression of CD63-Antares2 effectively labeled exosomes with Antares2, which emitted intense, long-wavelength luminescence suitable for in vivo monitoring. Transplantation of CD63-Antares2-expressing prostate cancer cells into mice allowed determining the amount of cancer-derived exosomes released from primary tumors into the bloodstream and visualizing the long-term homing behavior of exosomes to their target organs or tissues. Interestingly, secreted exosome was decreased upon administration of low dose of dasatinib, an approved tyrosine-kinase inhibitor. The CD63-Antares2 xenograft mouse model will be useful for elucidating the dynamics of cancer-derived exosomes in vivo and evaluating the therapeutic efficacy and mechanism of exosome production inhibitors.


Assuntos
Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Transferência de Energia , Exossomos/metabolismo , Imagem Molecular/métodos , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Animais , Dasatinibe/farmacologia , Xenoenxertos , Masculino , Camundongos , Transplante de Neoplasias , Inibidores de Proteínas Quinases/farmacologia , Fatores de Tempo
9.
Sci Rep ; 9(1): 3265, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30824759

RESUMO

c-Src is a membrane-associated tyrosine kinase that has key roles in the signaling transduction that controls cell growth, adhesion, and migration. In the early stage of carcinogenesis, c-Src is activated under the plasma membrane and transduces oncogenic signals. Here we show that c-Src localized to the endosomal membrane has unique functions in c-Src-transformed cells. Our results indicate that activated c-Src in the endosomal membrane promoted the secretion of exosomes, in which c-Src was encapsulated. In addition, the ESCRT-interacting molecule, Alix was identified as a c-Src-interacting protein in exosomes. We revealed that the interaction between the SH3 domain of c-Src and the proline-rich region of Alix activates ESCRT-mediated intra-luminal vesicle (ILV) formation, resulting in the upregulation of exosome secretion in c-Src-transformed cells. We observed also a correlation between malignant phenotypes and Alix-dependent aberrant exosome secretion in Src-upregulated cancer cells. Collectively, our findings provide a unique mechanism for the upregulation of exosomes in cancer cells, as well as new insights into the significance of exosome secretion in cancer progression.


Assuntos
Proteína Tirosina Quinase CSK/metabolismo , Exossomos/enzimologia , Membranas Intracelulares/enzimologia , Proteínas de Neoplasias/metabolismo , Neoplasias/enzimologia , Proteína Tirosina Quinase CSK/genética , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Exossomos/genética , Exossomos/patologia , Células HCT116 , Células HT29 , Humanos , Membranas Intracelulares/patologia , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patologia , Domínios de Homologia de src
10.
Sci Rep ; 8(1): 14035, 2018 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-30232365

RESUMO

Exosomes have emerged as important mediators of intercellular communication. Although their modes of action have been elucidated, the molecular mechanisms underlying their secretion, sorting of molecules, uptake into recipient cells, and biological distribution in vivo remain elusive. Here, we present a novel system for quantifying secreted exosomes by introducing ectopic or CRISPR/Cas9-mediated knock-in of luciferase-fusion exosome markers such as CD63. This luciferase-based method makes it possible to measure exosomes secreted into the culture medium with high linearity and wide dynamic range in a high-throughput manner. We demonstrate that data obtained by luminescent quantification are well correlated with data obtained by conventional nanoparticle tracking analysis under multiple conditions. In addition, our system is capable of evaluating the recipient cells or tissues that take up exosomes, as well as visualizing exosomes in vivo. The proposed system represents a powerful tool for understanding the molecular mechanisms underlying exosome production, uptake, and long-term distribution.


Assuntos
Exossomos/genética , Nanotecnologia/métodos , Tetraspanina 30/genética , Células A549 , Animais , Sistemas CRISPR-Cas , Linhagem Celular , Exossomos/química , Células HCT116 , Células HT29 , Humanos , Camundongos , Nanopartículas
11.
BMC Cancer ; 17(1): 89, 2017 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-28143428

RESUMO

BACKGROUND: BK-UM (CRM197) is a mutant form of diphtheria toxin and a specific inhibitor of heparin-binding epidermal growth factor-like growth factor (HB-EGF). We assessed the safety, pharmacokinetics, recommended dose, and efficacy of BK-UM in patients with recurrent ovarian cancer (OC) or peritoneal cancer (PC), and measured HB-EGF levels in serum and abdominal fluid after BK-UM administration. METHODS: Eleven patients with advanced or recurrent OC or PC were enrolled and treated with BK-UM via the intraperitoneal route. The dose was escalated (1.0, 2.0, 3.3, and 5.0 mg/m2) using a 3 + 3 design. RESULTS: Eight of 11 patients completed treatment. No dose-limiting toxicity (DLT) was experienced at dose levels 1 (1.0 mg/m2) and 2 (2.0 mg/m2). Grade 3 transient hypotension as an adverse event (defined as a DLT in the present study) was observed in two of four patients at dose level 3 (3.3 mg/m2). Treatment with BK-UM was associated with decreases in HB-EGF levels in serum and abdominal fluid in seven of 11 patients and five of eight patients, respectively. Clinical outcomes included a partial response in one patient, stable disease in five patients, and progressive disease in five patients. CONCLUSIONS: BK-UM was well tolerated at doses of 1.0 and 2.0 mg/m2, with evidence for clinical efficacy in patients with recurrent OC or PC. A dose of 2.0 mg/m2 BK-UM is recommended for subsequent clinical trials. TRIAL REGISTRATION: This trial was prospectively performed as an investigator-initiated clinical trial. The trial numbers are UMIN000001002 and UMIN000001001, with registration dates of 1/30/2008 and 2/4/2008, respectively. UMIN000001001 was registered as a trial for the continuous administration of BK-UM after UMIN000001002 .


Assuntos
Proteínas de Bactérias/administração & dosagem , Recidiva Local de Neoplasia/tratamento farmacológico , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Peritoneais/tratamento farmacológico , Idoso , Proteínas de Bactérias/farmacocinética , Relação Dose-Resposta a Droga , Feminino , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/metabolismo , Humanos , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Peritoneais/metabolismo
12.
PLoS One ; 11(10): e0165257, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27783694

RESUMO

Sialic acid bound to glycans in glycolipids and glycoproteins is essential for synaptic plasticity and memory. Sialidase (EC 3.2.1.18), which has 4 isozymes including Neu1, Neu2, Neu3 and Neu4, regulates the sialylation level of glycans by removing sialic acid from sialylglycoconjugate. In the present study, we investigated the distribution of sialidase activity in rat hippocampus and the role of sialidase in hippocampal memory processing. We previously developed a highly sensitive histochemical imaging probe for sialidase activity, BTP3-Neu5Ac. BTP3-Neu5Ac was cleaved efficiently by rat Neu2 and Neu4 at pH 7.3 and by Neu1 and Neu3 at pH 4.6. When a rat hippocampal acute slice was stained with BTP3-Neu5Ac at pH 7.3, mossy fiber terminal fields showed relatively intense sialidase activity. Thus, the role of sialidase in the synaptic plasticity was investigated at mossy fiber terminal fields. The long-term potentiation (LTP) at mossy fiber-CA3 pyramidal cell synapses was impaired by 2,3-dehydro-2-deoxy-N-acetylneuraminic acid (DANA), a sialidase inhibitor. DANA also failed to decrease paired-pulse facilitation after LTP induction. We also investigated the role of sialidase in hippocampus-dependent spatial memory by using the Morris water maze. The escape latency time to reach the platform was prolonged by DANA injection into the hippocampal CA3 region or by knockdown of Neu4 without affecting motility. The results show that the regulation of sialyl signaling by Neu4 is involved in hippocampal memory processing.


Assuntos
Hipocampo/metabolismo , Neuraminidase/metabolismo , Sinapses/metabolismo , Animais , Hipocampo/efeitos dos fármacos , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , Isoenzimas/metabolismo , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Ácido N-Acetilneuramínico/análogos & derivados , Ácido N-Acetilneuramínico/farmacologia , Neuraminidase/antagonistas & inibidores , Neuraminidase/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Wistar , Memória Espacial/efeitos dos fármacos , Sinapses/efeitos dos fármacos
13.
Glycobiology ; 25(9): 953-62, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26036195

RESUMO

Chondroitin sulfate E (CSE) is a polysaccharide containing mainly disaccharide units of D-glucuronic acid (GlcA) and 4,6-O-disulfated N-acetyl-D-galactosamine (GalNAc) residues (E-unit) in the amount of ∼ 60%. CSE is involved in many biological and pathological processes. In this study, we established new monoclonal antibodies, termed E-12C and E-18H, by using CSE that contained more than 70% of E-units as an immunogen. These antibodies recognized CSE but not other CSs isomers or dermatan sulfate (DS). We evaluated the reactivities of the antibodies to 6-O-sulfated CSA (6S-CSA) and DS (6S-DS) that possessed ∼ 60% of GalNAc (4S, 6S) moieties in their structures. Neither of the antibodies reacted with 6S-DS. The antibodies strictly distinguished the structural difference of GlcA and L-iduronic acid in the polysaccharide. Binding affinities of the antibodies were determined by a surface plasmon resonance assay using CSE and 6S-CSA. The binding affinities were strongly associated with the molecular weight of CSE and the E-unit content of 6S-CSA. Moreover, we demonstrated that the antibodies are applicable to histochemical analysis. In conclusion, the new anti-CSE monoclonal antibodies specifically recognize the E-unit of CSE. The antibodies will become useful tools for the investigation of the biological and pathological significance of CSE.


Assuntos
Anticorpos Monoclonais/imunologia , Sulfatos de Condroitina/imunologia , Animais , Anticorpos Monoclonais/química , Afinidade de Anticorpos , Especificidade de Anticorpos , Linhagem Celular Tumoral , Humanos , Camundongos , Ratos
14.
Genes Cells ; 15(10): 1051-62, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20825494

RESUMO

The nonreceptor tyrosine kinase c-Src is frequently over-expressed or hyperactivated in various human cancers and contributes to cancer progression in cooperation with up-regulated growth factor receptors. However, Src-selective anticancer drugs are still in clinical trials. To identify more effective inhibitors of c-Src-mediated cancer progression, we developed a new screening platform using Csk-deficient cells that can be transformed by c-Src. We found that purvalanol A, developed as a CDK inhibitor, potently suppressed the anchorage-independent growth of c-Src-transformed cells, indicating that the activation of CDKs contributes to the c-Src transformation. We also found that purvalanol A suppressed the c-Src activity as effectively as the Src-selective inhibitor PP2, and that it reverted the transformed morphology to a nearly normal shape with less cytotoxicity than PP2. Purvalanol A induced a strong G2-M arrest, whereas PP2 weakly acted on the G1-S transition. Furthermore, when compared with PP2, purvalanol A more effectively suppressed the growth of human colon cancer HT29 and SW480 cells, in which Src family kinases and CDKs are activated. These findings demonstrate that the coordinated inhibition of cell cycle progression and tyrosine kinase signaling by the multi-selective purvalanol A is effective in suppressing cancer progression associated with c-Src up-regulation.


Assuntos
Proteína Quinase CDC2/antagonistas & inibidores , Transformação Celular Neoplásica/efeitos dos fármacos , Colo/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Purinas/farmacologia , Antineoplásicos/farmacologia , Proteína Quinase CDC2/metabolismo , Proteína Tirosina Quinase CSK , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Colo/citologia , Neoplasias do Colo , Células HT29/efeitos dos fármacos , Humanos , Proteínas Tirosina Quinases/metabolismo , Proteínas Tirosina Quinases/farmacologia , Quinases da Família src
15.
Mol Cell ; 30(4): 426-36, 2008 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-18498747

RESUMO

The tyrosine kinase c-Src is upregulated in various human cancers irrespective of its negative regulator Csk, but the regulatory mechanisms remain unclear. Here, we show that a lipid raft-anchored Csk adaptor, Cbp/PAG, is directly involved in controlling the oncogenicity of c-Src. Using Csk-deficient cells that can be transformed by c-Src overexpression, we found that Cbp expression is markedly downregulated by c-Src activation and re-expression of Cbp efficiently suppresses c-Src transformation as well as tumorigenesis. Cbp-deficient cells are more susceptible to v-Src transformation than their parental cells. Upon phosphorylation, Cbp specifically binds to activated c-Src and sequesters it in lipid rafts, resulting in an efficient suppression of c-Src function independent of Csk. In some human cancer cells and tumors, Cbp is downregulated and the introduction of Cbp significantly suppresses tumorigenesis. These findings indicate a potential role for Cbp as a suppressor of c-Src-mediated tumor progression.


Assuntos
Microdomínios da Membrana/metabolismo , Proteínas de Membrana/metabolismo , Fosfoproteínas/metabolismo , Proteínas Tirosina Quinases/metabolismo , Animais , Proteína Tirosina Quinase CSK , Fracionamento Celular , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/fisiologia , Técnicas de Transferência de Genes , Humanos , Microdomínios da Membrana/química , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Nus , Neoplasias/metabolismo , Fosfoproteínas/genética , Proteínas Tirosina Quinases/genética , Quinases da Família src
16.
Genes Cells ; 13(1): 1-12, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18173743

RESUMO

The c-src proto-oncogene product, c-Src, is frequently over-expressed and activated in various human malignant cancers, implicating a role for c-Src in cancer progression. To verify the role of c-Src, we analyzed the transforming ability of c-Src in mouse embryonic fibroblasts that lack Csk, a negative regulator of Src family kinases. Although Csk deficiency is not sufficient for cell transformation, c-Src over-expression induced characteristic transformed phenotypes including anchorage-independent growth and tumorigenecity. These phenotypes were dose-dependently inhibited by the re-expression of Csk, indicating that there is a certain threshold for c-Src transformation, which is determined by the c-Src : Csk ratio. In contrast to v-Src, c-Src induced the phosphorylation of a limited number of cellular proteins and elicited a restricted change in gene expression profiles. The activation of some critical targets for v-Src transformation, such as STAT3, was not significantly induced by c-Src transformation. Several genes that are involved in cancer progression, that is, cyclin D1 and HIF-1alpha, were induced by v-Src, but not by c-Src. Furthermore, v-Src tumors exhibited aggressive growth and extensive angiogenesis, while c-Src tumors grew more slowly accompanied by the induction of hematomas. These findings demonstrate that c-Src has the potential to induce cell transformation, but it requires coordination with an additional pathway(s) to promote tumor progression in vivo.


Assuntos
Transformação Celular Neoplásica/metabolismo , Fibroblastos/citologia , Proteína Oncogênica pp60(v-src)/fisiologia , Proteínas Tirosina Quinases/fisiologia , Animais , Proteína Tirosina Quinase CSK , Transformação Celular Neoplásica/patologia , Ciclina D1/metabolismo , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Hematoma/metabolismo , Hematoma/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Camundongos Nus , Neovascularização Patológica , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação , Proteínas Tirosina Quinases/genética , Proto-Oncogene Mas , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Transplante Heterólogo , Quinases da Família src
17.
Biochem Biophys Res Commun ; 366(4): 969-75, 2008 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-18086565

RESUMO

To elucidate the regulatory mechanism of cell transformation induced by c-Src tyrosine kinase, we performed a proteomic analysis of tyrosine phosphorylated proteins that interact with c-Src and/or its negative regulator Csk. The c-Src interacting proteins were affinity-purified from Src transformed cells using the Src SH2 domain as a ligand. LC-MS/MS analysis of the purified proteins identified general Src substrates, such as focal adhesion kinase and paxillin, and ZO-1/2 as a transformation-dependent Src target. The Csk binding proteins were analyzed by a tandem affinity purification method. In addition to the previously identified Csk binding proteins, including Cbp/PAG, paxillin, and caveolin-1, we found that ZO-1/2 could also serve as a major Csk binding protein. ZO-2 was phosphorylated concurrently with Src transformation and specifically bound to Csk in a Csk SH2 dependent manner. These results suggest novel roles for ZO proteins as Src/Csk scaffolds potentially involved in the regulation of Src transformation.


Assuntos
Proteínas de Membrana/metabolismo , Fosfoproteínas/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteômica/métodos , Sequência de Aminoácidos , Animais , Proteína Tirosina Quinase CSK , Linhagem Celular , Galinhas , Humanos , Proteínas de Membrana/química , Camundongos , Dados de Sequência Molecular , Fosfoproteínas/química , Ligação Proteica , Mapeamento de Interação de Proteínas , Proteínas Tirosina Quinases/química , Ratos , Proteína da Zônula de Oclusão-1 , Proteína da Zônula de Oclusão-2 , Domínios de Homologia de src , Quinases da Família src
18.
J Biol Chem ; 282(11): 8276-83, 2007 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-17227766

RESUMO

The junctional adhesion molecule (JAM) family is a key molecule in a process called transendothelial migration or diapedesis. Here, we report implications of JAM-C in cancer metastasis. We first determined the mRNA expression of JAMs in 19 kinds of cancer cell lines. JAM-C was expressed in most of tumors having potent metastatic properties. Especially in murine K-1735 melanoma cell lines, the highly metastatic sublines (M2 and X21) strongly expressed JAM-C when compared with the poorly metastatic ones (C-10 and C23). Next, we investigated the role of JAM-C in cancer metastasis by using human JAM-C (hJAM-C) gene-transfected HT1080 fibrosarcoma cells. In comparison with mock-transfected HT1080 cells, these cells showed a significant increase in the adhesion to various extracellular substrates and the invasion across a Matrigel-coated membrane. The knockdown of hJAM-C using small interfering RNA resulted in the suppression of both the adhesion and the invasion of HT1080 cells, suggesting that endogenous hJAM-C might be involved in tumor metastasis. Finally, we studied the role of hJAM-C in an in vivo experimental metastatic model. The results showed that the overexpression of hJAM-C in HT1080 cells significantly decreased the life spans of the tumorbearing mice. In contrast, the knockdown of hJAM-C in HT1080 cells suppressed the weight gain of the lungs with metastatic colonies. We conclude that the expression of JAM-C promotes metastasis by enhancing both the adhesion of cancer cells to extracellular matrices and the subsequent invasion.


Assuntos
Moléculas de Adesão Celular/fisiologia , Imunoglobulinas/fisiologia , Proteínas de Membrana/fisiologia , Animais , Células CHO , Adesão Celular , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Tumoral , Colágeno/farmacologia , Cricetinae , Cricetulus , Combinação de Medicamentos , Humanos , Imunoglobulinas/metabolismo , Laminina/farmacologia , Neoplasias Pulmonares/secundário , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Invasividade Neoplásica , Metástase Neoplásica , Neoplasias da Próstata/metabolismo , Proteoglicanas/farmacologia
19.
Biochim Biophys Acta ; 1669(1): 69-74, 2005 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15843001

RESUMO

For the improvement of therapeutic efficacy in photodynamic therapy (PDT) by using a photosensitizer, benzoporphyrin derivative monoacid ring A (BPD-MA), we previously prepared polyethylene glycol (PEG)-modified liposomes encapsulating BPD-MA (PEG-Lip BPD-MA). PEGylation of liposomes enhanced the accumulation of BPD-MA in tumor tissue at 3 h after injection of it into Meth-A-sarcoma-bearing mice, but, unexpectedly, decreased the suitability of the drug for PDT when laser irradiation was performed at 3 h after the injection of the liposomal photosensitizer. To improve the bioavailability of PEG-Lip BPD-MA, we endowed the liposomes with active-targeting characteristics by using Ala-Pro-Arg-Pro-Gly (APRPG) pentapeptide, which had earlier been isolated as a peptide specific to angiogenic endothelial cells. APRPG-PEG-modified liposomal BPD-MA (APRPG-PEG-Lip BPD-MA) accumulated in tumor tissue similarly as PEG-Lip BPD-MA and to an approx. 4-fold higher degree than BPD-MA delivered with non-modified liposomes at 3 h after the injection of the drugs into tumor-bearing mice. On the contrary, unlike the treatment with PEG-Lip BPD-MA, APRPG-PEG-Lip BPD-MA treatment strongly suppressed tumor growth after laser irradiation at 3 h after injection. Finally, we observed vasculature damage in the dorsal air sac angiogenesis model by APRPG-PEG-Lip BPD-MA-mediated PDT. The present results suggest that antiangiogenic PDT is an efficient modality for tumor treatment and that tumor neovessel-targeted, long-circulating liposomes are a useful carrier for delivering photosensitizer to angiogenic endothelial cells.


Assuntos
Inibidores da Angiogênese/farmacologia , Lipossomos/uso terapêutico , Neovascularização Patológica/tratamento farmacológico , Peptídeos/farmacologia , Fotoquimioterapia , Inibidores da Angiogênese/administração & dosagem , Animais , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Polietilenoglicóis
20.
Biol Pharm Bull ; 27(3): 443-4, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14993821

RESUMO

For the purpose of the avoidance of reticuloendothelial system (RES)-trapping, liposome entrapped benzoporphyrin derivative monoacid ring A (BPD-MA), which is used for cancer photodynamic therapy (PDT), was modified with polyethylene glycol (PEG-LipBPD-MA). Tumor accumulation of BPD-MA at 3 h after injection with PEG-LipBPD-MA in Meth A-sarcoma-bearing mice was significantly higher than that after injection with non-modified liposomal BPD-MA (Cont-LipBPD-MA) as expected. On the contrary, significant tumor growth suppression after PDT was observed only for Cont-LipBPD-MA but not for PEG-LipBPD-MA. Thus, PEGylation enhances the passive targeting of liposomal BPD-MA in tumor, but decreases the susceptibility of the drug in PDT.


Assuntos
Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/administração & dosagem , Fármacos Fotossensibilizantes/uso terapêutico , Polietilenoglicóis/química , Porfirinas/administração & dosagem , Porfirinas/uso terapêutico , Animais , Lipossomos , Camundongos , Camundongos Endogâmicos BALB C , Sarcoma Experimental/tratamento farmacológico , Tecnologia Farmacêutica/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA