Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
J Cell Physiol ; 233(2): 1266-1277, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28488765

RESUMO

Gastric cancer represents a diffuse and aggressive neoplasm, whose mortality index is among the highest in the world. Predisposing factors are E-cadherin mutations, Helicobacter pylori infection, and a diet rich in salted and smoked food, with a low intake of fresh fruits and vegetables. Here, we analyzed the effect of total lipophilic extracts of two Southern Italy tomato varieties, San Marzano and Corbarino, on an in vitro model of gastric cancer, YCC-1, YCC-2 and YCC-3 cell lines, characterized by different aggressiveness. Our results showed a possible role of these two varieties of tomatoes against typical neoplastic features. The treatment with tomato extracts affected cancer cell ability to grow both in adherence and in semisolid medium, reducing also cell migration ability. No toxic effects were observed on non-tumoral cells. We found, on gastric cancer cell lines, effects on both cell cycle progression and apoptosis modulation. The extent of antineoplastic effects, however, did not seem to correlate with the carotenoid content and antioxidant activity of the two tomato varieties. Our data indicate that San Marzano and Corbarino intake might be further considered as nutritional support not only in cancer prevention, but also for cancer patient diet.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Antioxidantes/farmacologia , Carotenoides/farmacologia , Solanum lycopersicum/química , Neoplasias Gástricas/tratamento farmacológico , Antineoplásicos Fitogênicos/isolamento & purificação , Antioxidantes/isolamento & purificação , Apoptose/efeitos dos fármacos , Carotenoides/isolamento & purificação , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Frutas/química , Humanos , Itália , Invasividade Neoplásica , Fitoterapia , Plantas Medicinais , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Fatores de Tempo
3.
J Cell Biochem ; 117(4): 828-35, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26479787

RESUMO

Proteomics is a recent field of research in molecular biology that can help in the fight against cancer through the search for biomarkers that can detect this disease in the early stages of its development. Proteomic is a speedily growing technology, also thanks to the development of even more sensitive and fast mass spectrometry analysis. Although this technique is the most widespread for the discovery of new cancer biomarkers, it still suffers of a poor sensitivity and insufficient reproducibility, essentially due to the tumor heterogeneity. Common technical shortcomings include limitations in the sensitivity of detecting low abundant biomarkers and possible systematic biases in the observed data. Current research attempts are trying to develop high-resolution proteomic instrumentation for high-throughput monitoring of protein changes that occur in cancer. In this review, we describe the basic features of the proteomic tools which have proven to be useful in cancer research, showing their advantages and disadvantages. The application of these proteomic tools could provide early biomarkers detection in various cancer types and could improve the understanding the mechanisms of tumor growth and dissemination.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Pulmonares/diagnóstico , Proteínas de Neoplasias/genética , Neoplasias Ovarianas/diagnóstico , Neoplasias Pancreáticas/diagnóstico , Proteômica/métodos , Biomarcadores Tumorais/sangue , Feminino , Expressão Gênica , Humanos , Neoplasias Pulmonares/sangue , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Proteínas de Neoplasias/sangue , Neoplasias Ovarianas/sangue , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Neoplasias Pancreáticas/sangue , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Proteômica/instrumentação , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Pesquisa Translacional Biomédica
4.
J Cell Physiol ; 230(4): 802-5, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25205458

RESUMO

pRb2/p130 is a key tumor suppressor, whose oncosuppressive activity has mainly been attributed to its ability to negatively regulate cell cycle by interacting with the E2F4 and E2F5 transcription factors. Indeed, pRb2/p130 has been found altered in various cancer types in which it functions as a valuable prognostic marker. Here, we analyzed pRb2/p130 expression in gastric cancer tissue samples of diffuse histotype, in comparison with their normal counterparts. We found a cytoplasmic localization of pRb2/p130 in cancer tissue samples, whereas, in normal counterparts, we observed the expected nuclear localization. pRb2/p130 cytoplasmic delocalization can lead to cell cycle deregulation, but considering the emerging involvement of pRb2/p130 in other key cellular processes, it could contribute to gastric tumorigenesis also through other mechanisms. Our data support the necessity of further investigations to verify the possibility of using pRb2/p130 as a biomarker or potential therapeutic target for diffuse gastric cancer.


Assuntos
Proteína Substrato Associada a Crk/metabolismo , Citoplasma/metabolismo , Proteínas Salivares Ricas em Prolina/metabolismo , Neoplasias Gástricas/metabolismo , Fatores de Transcrição/metabolismo , Proteínas de Ciclo Celular/metabolismo , Divisão Celular/genética , Divisão Celular/fisiologia , Feminino , Genes Supressores de Tumor/fisiologia , Humanos , Masculino , Fosfoproteínas/fisiologia , Proteína do Retinoblastoma/metabolismo , Proteína p130 Retinoblastoma-Like/metabolismo , Neoplasias Gástricas/genética
5.
World J Gastroenterol ; 20(27): 8986-92, 2014 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-25083072

RESUMO

The Tenth International Gastric Cancer Congress (IGCC) was held in Verona, Italy, from June 19 to 22, 2013. The meeting enclosed various aspects of stomach tumor management, including both tightly clinical approaches, and topics more related to basic research. Moreover, an overview on gastrointestinal stromal tumors was provided too, although here not discussed. Here we will discuss some topics related to molecular biology of gastric cancer (GC), inherent to prognostic, diagnostic and therapeutic tools shown at the conference. Results about well known subjects, such as E-cadherin loss of expression/function, were presented. They revealed that other mutations of the gene were identified, showing a continuous research to improve diagnosis and prognosis of stomach tumor. Simultaneously, new possible molecular markers with an established role for other neoplasms, were discussed, such as mesothelin, stomatin-like protein 2 and Notch-1. Hence, a wide overview including both old and new diagnostic/prognostic tools was offered. Great attention was also dedicated to possible drugs to be used against GC. They included monoclonal antibodies, such as MS57-2.1, drugs used in other pathologies, such as maraviroc, and natural extracts from plants such as biflorin. We would like to contribute to summarize the most impressive studies presented at the IGCC, concerning novel findings about molecular biology of gastric cancer. Although further investigations will be necessary, it can be inferred that more and more tools were developed, so as to better face stomach neoplasms.


Assuntos
Neoplasias Gástricas/terapia , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Caderinas/genética , Caderinas/metabolismo , Humanos , Instabilidade de Microssatélites , Terapia de Alvo Molecular , Seleção de Pacientes , Medicina de Precisão , Valor Preditivo dos Testes , Prognóstico , Fatores de Risco , Transdução de Sinais , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Fatores de Crescimento do Endotélio Vascular/metabolismo
6.
J Cell Biochem ; 114(9): 2114-9, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23553770

RESUMO

Phosphoinositide 3-kinase proteins are composed by a catalytic p110 subunit and a regulatory p85 subunit. There are three classes of PI3K, named class I-III, on the bases of the protein domain constituting and determining their specificity. The first one is the best characterized and includes a number of key elements for the integration of different cellular signals. Regulatory p85 subunit shares with the catalytic p110 subunit, a N-terminal SH3 domain showing homology with the protein domain Rho-GTP-ase. After cell stimulation, all class I PI3Ks are recruited to the inner face of the plasma membrane, where they generate phosphatidylinositol-3,4,5-trisphosphate by direct phosphorylation of phosphatidylinositol-4,5-bisphosphate. All pathways trigger the control of different phenomena such as cell growth, proliferation, apoptosis, adhesion and migration through various downstream effectors. We have previously provided direct evidences that a Serine in position 83, adjacent to the N-terminal SH3 domain of regulatory subunit of PI3K, is a substrate of PKA. The aim of this work is to confirm the role of p85αPI3KSer83 in regulating cell proliferation, migration and invasion in prostate cancer cells LNCaP. To this purpose cells were transfected with mutant forms of p85, where Serine was replaced by Alanine, where phosphorylation is prevented, or Aspartic Acid, to mimic the phosphorylated residue. The findings of this study suggest that identifying a peptide mimicking the sequence adjacent to Ser 83 may be used to produce antibodies against this residue that can be proposed as usefool tool for prognosis by correlating phosphorylation at Ser83 with tumor stage.


Assuntos
Fosfatidilinositol 3-Quinases/química , Fosfatidilinositol 3-Quinases/metabolismo , Neoplasias da Próstata/metabolismo , Subunidades Proteicas/metabolismo , Serina/metabolismo , Apoptose/genética , Apoptose/fisiologia , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Masculino , Microscopia Confocal , Fosfatidilinositol 3-Quinases/genética , Fosforilação , Neoplasias da Próstata/genética , Subunidades Proteicas/química , Subunidades Proteicas/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
7.
World J Clin Oncol ; 3(9): 128-36, 2012 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-23061031

RESUMO

Gastric cancer is a multifactorial neoplastic pathology numbering among its causes both environmental and genetic predisposing factors. It is mainly diffused in South America and South-East Asia, where it shows the highest morbility percentages and it is relatively scarcely diffused in Western countries and North America. Although molecular mechanisms leading to gastric cancer development are only partially known, three main causes are well characterized: Helicobacter pylori (H. pylori) infection, diet rich in salted and/or smoked food and red meat, and epithelial cadherin (E-cadherin) mutations. Unhealthy diet and H. pylori infection are able to induce in stomach cancer cells genotypic and phenotypic transformation, but their effects may be crossed by a diet rich in vegetables and fresh fruits. Various authors have recently focused their attention on the importance of a well balanced diet, suggesting a necessary dietary education starting from childhood. A constant surveillance will be necessary in people carrying E-cadherin mutations, since they are highly prone in developing gastric cancer, also within the inner stomach layers. Above all in the United States, several carriers decided to undergo a gastrectomy, preferring changing their lifestyle than living with the awareness of the development of a possible gastric cancer. This kind of choice is strictly personal, hence a decision cannot be suggested within the clinical management. Here we summarize the key points of gastric cancer prevention analyzing possible strategies referred to the different predisposing factors. We will discuss about the effects of diet, H. pylori infection and E-cadherin mutations and how each of them can be handled.

8.
Oncol Rep ; 24(6): 1411-8, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21042734

RESUMO

Gastric cancer is one of the most diffuse neoplastic pathologies in the world whose environmental and molecular causes, although deeply investigated, have not been completely clarified. Besides some well-established etiological factors, such as Helicobacter pylori and E-cadherin mutations, investigations on other possible causes gave contrasting results. Rb family proteins (including pRb/p105, pRb2/p130 and p107) are involved in cell cycle regulation and their function and/or expression is often lost in various kinds of tumours such as lung, bladder, breast and brain cancer. The consequences of RB inactivation in tumours can be very different depending on the context and the type of cancer. Recent evidence indicates that Rb status correlates with a different therapeutic response according to the tumour type and the therapeutic agent. Studies performed on Rb family proteins in gastrointestinal tract tumours suggest that these proteins have an important role in these cancer types. However, owing to contrasting results, further investigation is required to assess whether the expression of Rb family proteins can potentially be used as a prognostic or predictive factor in gastric cancer.


Assuntos
Carcinoma/genética , Proteína do Retinoblastoma/fisiologia , Neoplasias Gástricas/genética , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/fisiologia , Carcinoma/diagnóstico , Carcinoma/patologia , Carcinoma/terapia , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Humanos , Família Multigênica/fisiologia , Prognóstico , Proteína do Retinoblastoma/genética , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/patologia , Neoplasias Gástricas/terapia
9.
Proc Natl Acad Sci U S A ; 107(32): 14484-9, 2010 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-20660740

RESUMO

Tumor growth requires neoangiogenesis. VEGF is the most potent proangiogenic factor. Dysregulation of hypoxia-inducible factor (HIF) or cytokine stimuli such as those involving the chemokine receptor 4/stromal-derived cell factor 1 (CXCR4/SDF-1) axis are the major cause of ectopic overexpression of VEGF in tumors. Although the CXCR4/SDF-1 pathway is well characterized, the transcription factors executing the effector function of this signaling are poorly understood. The multifunctional Yin Yang 1 (YY1) protein is highly expressed in different types of cancers and may regulate some cancer-related genes. The network involving CXCR4/YY1 and neoangiogenesis could play a major role in cancer progression. In this study we have shown that YY1 forms an active complex with HIF-1alpha at VEGF gene promoters and increases VEGF transcription and expression observed by RT-PCR, ELISA, and Western blot using two different antibodies against VEGFB. Long-term treatment with T22 peptide (a CXCR4/SDF-1 inhibitor) and YY1 silencing can reduce in vivo systemic neoangiogenesis (P < 0.01 and P < 0.05 vs. control, respectively) during metastasis. Moreover, using an in vitro angiogenesis assay, we observed that YY1 silencing led to a 60% reduction in branches (P < 0.01) and tube length (P < 0.02) and a 75% reduction in tube area (P < 0.001) compared with control cells. A similar reduction was observed using T22 peptide. We demonstrated that T22 peptide determines YY1 cytoplasmic accumulation by reducing its phosphorylation via down-regulation of AKT, identifying a crosstalk mechanism involving CXCR4/YY1. Thus, YY1 may represent a crucial molecular target for antiangiogenic therapy during cancer progression.


Assuntos
Neoplasias/irrigação sanguínea , Neovascularização Patológica , Receptores CXCR4/antagonistas & inibidores , Fatores de Crescimento do Endotélio Vascular/genética , Fator de Transcrição YY1/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Transplante de Neoplasias , Neoplasias/metabolismo , Peptídeos/farmacologia , Ratos , Receptor Cross-Talk/fisiologia , Receptores CXCR4/metabolismo , Fatores de Transcrição , Transplante Heterólogo , Fator de Transcrição YY1/fisiologia
10.
J Cell Physiol ; 222(2): 474-80, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19918796

RESUMO

Hematopoietic stem cells derive regulatory information also from parathyroid hormone (PTH). To explore the possibility that PTH may have a role in regulation of other stem cells residing in bone marrow, such as mesenchymal stem cells (MSCs) and endothelial progenitor cells (EPCs) we assessed the effect of this hormone on the in vitro behavior of MSCs and EPCs. We evidenced that MSCs were much more responsive to PTH than EPCs. PTH increased the proliferation rate of MSCs with a diminution of senescence and apoptosis. Taken together, our results may suggest a protective effect of PTH on MSCs that reduces stress phenomena and preserve genome integrity. At the opposite, PTH did not modify the fate of EPCs in culture.


Assuntos
Células da Medula Óssea/metabolismo , Células Endoteliais/metabolismo , Células-Tronco Mesenquimais/metabolismo , Hormônio Paratireóideo/metabolismo , Células-Tronco/metabolismo , Células Estromais/metabolismo , 8-Hidroxi-2'-Desoxiguanosina , Apoptose , Células da Medula Óssea/patologia , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Senescência Celular , Dano ao DNA , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Células Endoteliais/patologia , Regulação da Expressão Gênica , Humanos , Células-Tronco Mesenquimais/patologia , RNA Mensageiro/metabolismo , Receptores de Hormônios Paratireóideos/metabolismo , Proteína do Retinoblastoma/metabolismo , Células-Tronco/patologia , Células Estromais/patologia , Fatores de Tempo , Proteína Supressora de Tumor p53/metabolismo
11.
Clin Cancer Res ; 14(15): 4775-9, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18676747

RESUMO

PURPOSE: pRb2/p130, a member of the Retinoblastoma gene family, has been shown to be a powerful prognostic factor in several malignancies. We sought to evaluate pRb2/p130 protein expression and its clinical effect in patients affected with soft tissue sarcomas (STS). EXPERIMENTAL DESIGN: Expression of pRb2/p130 was evaluated by immunohistochemistry on formalin-fixed, paraffin-embedded sections in 41 STSs. Results obtained were correlated with clinicopathologic variables and disease-free and overall survival (OS) in univariate and multivariate analysis. RESULTS: Expression of pRb2/p130 was diminished in 25 (61%) tumors, whereas the remaining ones (39%) were classified as high expressors. No correlation between pRb2/p130 expression and clinicopathologic variables was observed. However, a direct relationship between pRb2/p130 expression and clinical outcome of the patients was found in the subgroup of nonmetastatic tumors (n = 31). In univariate analysis, reduced pRb2/p130 expression was a negative prognostic factor and correlated with shorter disease-free survival (P = 0.021) and OS (P = 0.017) survival. In multivariate analysis, reduced pRb2/p130 expression was confirmed to be an independent predictor of shorter OS when considered together with tumor stage and grading (risk ratio, 7.893; confidence interval, 1.618-38.509; P = 0.011). CONCLUSIONS: This study shows for the first time the potential prognostic value of pRb2/130 expression evaluated on formalin-fixed, paraffin-embedded sections in STSs patients. pRb2/p130 immunoreactivity can be used to predict OS in patients with nonmetastatic STSs and, therefore, may represent a new prognostic marker.


Assuntos
Regulação Neoplásica da Expressão Gênica , Proteína p130 Retinoblastoma-Like/biossíntese , Proteína p130 Retinoblastoma-Like/fisiologia , Sarcoma/metabolismo , Sarcoma/mortalidade , Neoplasias de Tecidos Moles/metabolismo , Neoplasias de Tecidos Moles/mortalidade , Adulto , Idoso , Idoso de 80 Anos ou mais , Intervalo Livre de Doença , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade , Modelos Biológicos , Análise Multivariada , Prognóstico , Sarcoma/genética , Neoplasias de Tecidos Moles/genética , Resultado do Tratamento
12.
Endocr Relat Cancer ; 14(2): 405-20, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17639054

RESUMO

Loss of expression of the cyclin-dependent kinase inhibitor p27 through enhanced protein degradation frequently occurs in human cancer. Degradation of p27 requires ubiquitination by the S-phase kinase-associated protein 2 (Skp2), a member of the F-box family of Skp1-Cullin-F-box protein ubiquitin ligases. In the present study, we have investigated the role of Skp2 in human thyroid tumours. Immunohistochemistry analysis showed that Skp2 was overexpressed significantly in thyroid carcinomas (26 out of 51) compared with goitres (0 out of 12, P<0.001) or adenomas (1 out of 10, P<0.05), and that high Skp2 expression was detected more often in anaplastic thyroid (ATC; 83%, n=12) than follicular thyroid (FTC; 40%, n=20) or papillary thyroid (PTC; 42%, n=19) carcinomas (P<0.05). Thyroid cancer cell lines and tissues with high levels of Skp2 protein presented high p27 degradation activity and there was an inverse correlation between Skp2 and p27 expression in thyroid cancer tissues (n=68; P<0.05). In most cases, the observed overexpression of Skp2 protein was paralleled by an increase in the levels of Skp2 mRNA, and we observed Skp2 gene amplification at 5p13 in 2 out of 6 cell lines and in 9 out of 23 primary tumours (six out of eight ATCs, two out of nine PTCs and one out of six FTCs) using Q-PCR and/or fluorescence in situ hybridization analysis. Finally, in vitro experiments demonstrated that suppression of Skp2 expression drastically reduced proliferation of thyroid cancer cells and, conversely, forced expression of Skp2 circumvented serum dependency and contact inhibition in Skp2-negative cells by promoting p27 degradation. These findings indicate that Skp2 plays an important role for the development of thyroid cancer.


Assuntos
Carcinoma/metabolismo , Carcinoma/patologia , Proteínas Quinases Associadas a Fase S/metabolismo , Neoplasias da Glândula Tireoide/metabolismo , Neoplasias da Glândula Tireoide/patologia , Carcinoma/química , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p27/análise , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Amplificação de Genes , Humanos , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Proteínas Quinases Associadas a Fase S/análise , Proteínas Quinases Associadas a Fase S/genética , Neoplasias da Glândula Tireoide/química
13.
Breast Cancer Res Treat ; 102(3): 275-82, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17004112

RESUMO

AKT and MAPK signaling are involved in the resistance of breast cancer cells to the EGFR tyrosine kinase inhibitor gefitinib. RAS proteins are upstream mediators that transfer messages from surface receptors to intracellular signal transducers including MAPK and AKT pathways. AZD3409 is a novel prenyl inhibitor that has shown activity against both farnesyl transferase and geranylgeranyl transferase in isolated enzyme studies. We explored the activity of AZD3409 on breast cancer cell lines with high (SK-Br-3), intermediate (MDA-MB-361) or low (MDA-MB-468) sensitivity to gefitinib. We found that AZD3409 inhibits the growth of breast cancer cells in a dose-dependent manner, with the MDA-MB-468 and MDA-MB-361 cell lines showing higher sensitivity as compared with SK-Br-3 cells. Treatment with AZD3409 produced a significant reduction in the levels of activation of AKT in the three cell lines. AZD3409 also induced an increase in the expression of p27kip-1 and of hypophosphorylated forms of pRb2 in MDA-MB-468 cells that was associated with accumulation of cells in G0/G1 and the appearance of a sub-G1 peak suggestive of apoptosis. In contrast, AZD3409 produced a G2 arrest associated with reduced expression of pRb2 in MDA-MB-361 cells. A synergistic anti-tumor effect was observed when MDA-MB-468 or MDA-MB-361 cells were treated with both AZD3409 and gefitinib, whereas this combination was only additive in SK-Br-3 cells. However, treatment of breast cancer cells with AZD3409 and gefitinib did not produce a more significant blockade of AKT signaling as compared with gefitinib alone. These data suggest that AZD3409 might be active in gefitinib-resistant breast carcinoma.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/metabolismo , Proliferação de Células/efeitos dos fármacos , Quinazolinas/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Western Blotting , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Inibidores Enzimáticos/farmacologia , Fator de Crescimento Epidérmico/efeitos dos fármacos , Feminino , Citometria de Fluxo , Gefitinibe , Humanos , Proteínas Tirosina Quinases/antagonistas & inibidores
14.
J Cell Sci ; 119(Pt 16): 3424-34, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16882693

RESUMO

The serine protease urokinase (uPA) binds to the urokinase receptor (uPAR) through its growth-factor domain (GFD, residues 1-49), affecting cell migration, adhesion and growth. Here, we show that uPA can promote cytoskeletal rearrangements and directional cell migration in a GFD-independent manner, through a new and specific interaction between an internal uPA domain coined ;connecting peptide' (residues 132-158) and cell-surface integrin alpha v beta 5. Remarkably, a peptide corresponding to this region (CPp, residues 135-158) retains the ability to bind to alpha v beta 5, eliciting cytoskeletal rearrangements and directing cell migration at a concentration as low as 1-10 pM. These effects are lost in cells not expressing uPAR, indicating that the uPAR is required for CPp-dependent signaling. Furthermore, the CPp-alpha v beta 5-integrin interaction enhances F-actin-enriched protrusions and cell migration induced by the well-established interaction between the uPAR-binding peptide (GFDp, residues 12-32) of uPA and uPAR. These results provide new insight into the function of uPA, which--through individual domains--can engage two different surface receptors (uPAR and alpha v beta 5 integrin), thus initiating and potentiating intracellular signaling and migration.


Assuntos
Movimento Celular , Integrinas/metabolismo , Fragmentos de Peptídeos/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores de Vitronectina/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Actinas/metabolismo , Animais , Células Cultivadas , Quimiotaxia , Humanos , Rim/metabolismo , Camundongos , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Transdução de Sinais , Células U937
15.
Oncogene ; 24(11): 1882-94, 2005 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-15674339

RESUMO

PTEN/MMAC1/TEP1: (hereafter PTEN) is a tumor suppressor gene (located at 10q23) that is frequently mutated or deleted in sporadic human tumors. PTEN encodes a multifunctional phosphatase, which negatively regulates cell growth, migration and survival via the phosphatidylinositol 3'-kinase/AKT signalling pathway. Accordingly, Pten+/- mice develop various types of tumors including teratocarcinomas and teratomas. We have investigated PTEN expression in 60 bioptic specimens of germ cell tumors (32 seminomas, 22 embryonal carcinomas and six teratomas) and 22 intratubular germ cell neoplasias (ITGCN) adjacent to the tumors for PTEN protein and mRNA expression. In total, 10 testicular biopsies were used as controls. In the testis, PTEN was abundantly expressed in germ cells whereas it was virtually absent from 56% of seminomas as well as from 86% of embryonal carcinomas and virtually all teratomas. On the contrary, ITGCN intensely expressed PTEN, indicating that loss of PTEN expression is not an early event in testicular tumor development. The loss of PTEN expression occurs mainly at the RNA level as determined by in situ hybridization of cellular mRNA (17/22) but also it may involve some kind of post-transcriptional mechanisms in the remaining 25% of cases. Analysis of microsatellites D10S551, D10S541 and D10S1765 in GCTs (n=22) showed LOH at the PTEN locus at 10q23 in at least 36% of GCTs (three embryonal carcinoma, three seminoma, two teratoma); one seminoma and one embryonal (9%) carcinoma presented an inactivating mutation in the PTEN gene (2/22). Finally, we demonstrated that the phosphatidylinositol 3'-kinase/AKT pathway, which is regulated by the PTEN phosphatase, is crucial in regulating the proliferation of the NT2/D1 embryonal carcinoma cells, and that the cyclin-dependent kinase inhibitor p27(kip1) is a key downstream target of this pathway.


Assuntos
Transformação Celular Neoplásica , Genes Supressores de Tumor , Germinoma/genética , Monoéster Fosfórico Hidrolases/genética , Neoplasias Testiculares/genética , Proteínas Supressoras de Tumor/genética , Animais , Neoplasias da Mama , Linhagem Celular Tumoral , Cromossomos Humanos Par 10 , Feminino , Citometria de Fluxo , Humanos , Hibridização In Situ , Perda de Heterozigosidade , Masculino , Camundongos , PTEN Fosfo-Hidrolase , RNA Mensageiro/genética , Testículo/citologia , Testículo/embriologia , Testículo/patologia
16.
Cancer Res ; 63(21): 7423-7, 2003 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-14612541

RESUMO

Atypical lipomatous tumors (ALTs)/well-differentiated liposarcomas represent a distinctive subset of mesenchymal neoplasms featuring mature adipocytic proliferation. These tumors are characterized cytogenetically by the presence of supernumerary ring and/or long marker chromosomes that contain several copies of the chromosomal region 12q13-15, in which the HMGA2 gene is located. Deregulation of the HMGA2 gene is a common molecular alteration implicated in the development of a variety of benign tumors, such as lipomas, uterine leiomyomas, and pulmonary chondroid hamartomas. In this study, we observed HMGA2 overexpression in 7 of 12 ALT primary cell cultures examined. Subsequently, we generated an adenovirus containing the HMGA2 gene in the antisense orientation (Ad-A2as) to study the effect of HMGA2 protein suppression in ALT cells. The infection of six ALT cells, three of which were positive for HMGA2 expression, resulted in growth inhibition coupled with a significant increase in apoptosis. In addition, the growth of the ALT cells negative for HMGA2 expression was not affected by the infection with either the Ad-A2as or the control virus. On the basis of these findings, the targeting of the HMGA2 protein expression may represent a promising approach for treating the well-differentiated liposarcomas resistant to conventional therapies.


Assuntos
Proteína HMGA2/antagonistas & inibidores , Lipossarcoma/metabolismo , Lipossarcoma/terapia , Adenovírus Humanos/genética , Apoptose/genética , Divisão Celular/genética , DNA Antissenso/administração & dosagem , DNA Antissenso/genética , Terapia Genética/métodos , Proteína HMGA2/biossíntese , Proteína HMGA2/genética , Humanos , Lipossarcoma/genética , Lipossarcoma/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução Genética , Células Tumorais Cultivadas
17.
Oncogene ; 22(48): 7576-86, 2003 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-14576819

RESUMO

We report that cyclin D3 is rate limiting for G1 progression in thyroid follicular cells and that its constitutive upregulation by chronic stimulation of the TSH/cAMP pathway plays a role in human and experimental hyperproliferative diseases of the thyroid gland. These conclusions are supported by in vitro and in vivo studies. In rat thyrocytes (PC Cl 3 cells), cyclin D3 expression is enhanced in response to activation of the TSH/cAMP pathway. Interference with the expression of G1 cyclins (in particular cyclin D3) by the antisense methodology strongly reduced TSH-dependent proliferation of PC Cl 3 cells, indicating that proper progression through G1 requires cyclin D3. Accordingly, PC Cl 3 cells engineered to overexpress cyclin D3 (PC-D3 cells) show enhanced growth rate and elude hormone-dependence and contact inhibition. Using an animal experimental model of thyroid stimulation, we demonstrate that cyclin D3 is a key mediator of TSH-dependent proliferation of thyroid follicular cells also in vivo. Cyclin D3 protein levels were higher in the thyrocytes from glands of propylthiouracil-treated rats compared with control animals. The increase in cyclin D3 expression occurred after the propylthiouracil-induced increase in TSH levels and preceded the burst of cell proliferation. Finally, we found that cyclin D3 protein is expressed in a fraction of human goiters but it is strongly overexpressed in most follicular adenomas.


Assuntos
Ciclinas/metabolismo , Doenças da Glândula Tireoide/metabolismo , Doenças da Glândula Tireoide/patologia , Glândula Tireoide/citologia , Glândula Tireoide/efeitos dos fármacos , Tireotropina/farmacologia , Animais , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Ciclina D3 , Ciclina E , Quinases Ciclina-Dependentes/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Quinases/metabolismo , Ratos , Glândula Tireoide/metabolismo , Glândula Tireoide/patologia
18.
Biol Chem ; 383(1): 107-13, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11928806

RESUMO

We have previously reported that phosphorylation of human urokinase on Ser138/303 abolishes its catalytic-independent motogen and proadhesive abilities, whereas receptor binding is not affected. Here we show that substitution of the two relevant serines with glutamic acid residues impairs the ability of urokinase to mobilize a variety of human and mouse cell lines as well as human primary T lymphocytes. Accordingly, urokinase receptor-dependent signaling, leading to cytoskeletal rearrangements and paxillin re-distribution, does not occur in MCF-7 breast carcinoma cells exposed to 'phosphorylation-like' urokinase. Unlike the wild-type form, di-substituted urokinase is unable to induce the physical association of urokinase receptor with alphavbeta5 vitronectin receptor, which is required for MCF-7 urokinase-dependent cell migration. Finally, the di-substituted variant fails to activate p55fgr, a member of the Src tyrosine kinase family, which mediates cell migration and adhesion of U937 myelomonocytic cells. In conclusion, the finding that specific amino acid substitutions strongly interfere with the ability of urokinase to stimulate cell migration, and the associated intracellular events uncover a novel way to regulate urokinase receptor-dependent signaling.


Assuntos
Proteínas Proto-Oncogênicas , Receptores de Superfície Celular/fisiologia , Transdução de Sinais , Ativador de Plasminogênio Tipo Uroquinase/fisiologia , Substituição de Aminoácidos , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proteínas do Citoesqueleto/metabolismo , Ativação Enzimática/efeitos dos fármacos , Feminino , Ácido Glutâmico , Humanos , Camundongos , Proteínas Oncogênicas Virais/metabolismo , Paxilina , Fosfoproteínas/metabolismo , Ligação Proteica , Receptores de Superfície Celular/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Receptores de Vitronectina/metabolismo , Serina , Células Tumorais Cultivadas , Ativador de Plasminogênio Tipo Uroquinase/genética , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Quinases da Família src
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...