Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Carcinogenesis ; 41(9): 1246-1252, 2020 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-32525968

RESUMEN

Castration-resistant prostate cancer (CRPC) is an incurable stage of the disease. A multivariate principal component analysis on CRPC in vitro models identified aspartyl (asparaginyl) ß hydrolase (ASPH) as the most relevant molecule associated with the CRPC phenotype. ASPH is overexpressed in various malignant neoplasms and catalyzes the hydroxylation of aspartyl and asparaginyl residues in the epidermal growth factor (EGF)-like domains of proteins like NOTCH receptors and ligands, enhancing cell motility, invasion and metastatic spread. Bioinformatics analyses of ASPH in prostate cancer (PCa) and CRPC datasets indicate that ASPH gene alterations have prognostic value both in PCa and CRPC patients. In CRPC cells, inhibition of ASPH expression obtained through specific small interfering RNA or culturing cells in hypoxic conditions, reduced cell proliferation, invasion and cyclin D1 expression through modulation of the NOTCH signaling. ASPH and HIF1α crosstalk, within a hydroxylation-regulated signaling pathway, might be transiently driven by the oxidative stress evidenced inside CRPC cells. In addition, increased phosphorylation of GSK3ß by ASPH silencing demonstrates that ASPH regulates GSK3ß activity inhibiting its interactions with upstream kinases. These findings demonstrate the critical involvement of ASPH in CRPC development and may represent an attractive molecular target for therapy.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proteínas de Unión al Calcio/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Oxigenasas de Función Mixta/antagonistas & inhibidores , Proteínas Musculares/antagonistas & inhibidores , Neoplasias de la Próstata Resistentes a la Castración/patología , Receptor Notch1/metabolismo , Apoptosis , Biomarcadores de Tumor/genética , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Proliferación Celular , Glucógeno Sintasa Quinasa 3 beta/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Pronóstico , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , ARN Interferente Pequeño/genética , Receptor Notch1/genética , Tasa de Supervivencia , Células Tumorales Cultivadas
2.
Int J Mol Sci ; 20(23)2019 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-31816863

RESUMEN

BACKGROUND: Prostate cancer (PCa) is a significant health concern throughout the world. Standard therapy for advanced disease consists of anti-androgens, however, almost all prostate tumors become castration resistant (CRPC). Progression from androgen-sensitive PCa to CRPC is promoted by inflammatory signaling through cyclooxygenase-2 (COX-2) expression and ErbB family receptors/AKT activation, compensating androgen receptor inactivity. METHODS: Making use of CRPC cell lines, we investigated the effects of the anti-inflammatory drug celecoxib. Biochemical data obtained using immunoblotting, enzyme-linked immunosorbent assay (ELISA), invasion, and xenografts were further integrated by bioinformatic analyses. RESULTS: Celecoxib reduced cell growth and induced apoptosis through AKT blockade, cleavage of poly (ADP-ribose) polymerase-1 (PARP-1), and proteasomal degradation of the anti-apoptotic protein Mcl-1. Epidermal growth factor receptor (EGFR), ErbB2, and ErbB3 degradation, and heterogeneous nuclear ribonucleoprotein K (hnRNP K) downregulation, further amplified the inhibition of androgen signaling. Celecoxib reduced the invasive phenotype of CRPC cells by modulating NF-κB activity and reduced tumor growth in mice xenografts when administered in association with the anti-EGFR receptor antibody cetuximab. Bioinformatic analyses on human prostate cancer datasets support the relevance of these pathways in PCa progression. CONCLUSIONS: Signaling nodes at the intersection of pathways implicated in PCa progression are simultaneously modulated by celecoxib treatment. In combination therapies with cetuximab, celecoxib could represent a novel therapeutic strategy to curb signal transduction during CRPC progression.


Asunto(s)
Celecoxib/uso terapéutico , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Transducción de Señal , Anfirregulina/metabolismo , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/genética , Celecoxib/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Cetuximab/farmacología , Cetuximab/uso terapéutico , Regulación hacia Abajo/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Factor de Crecimiento Epidérmico/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ribonucleoproteína Heterogénea-Nuclear Grupo K/metabolismo , Humanos , Masculino , Ratones SCID , FN-kappa B/metabolismo , Invasividad Neoplásica , Fosforilación/efectos de los fármacos , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor ErbB-2/metabolismo , Receptores Androgénicos/metabolismo , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
3.
Int J Mol Sci ; 19(7)2018 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-29966326

RESUMEN

The major challenge in castration-resistant prostate cancer (CRPC) remains the ability to predict the clinical responses to improve patient selection for appropriate treatments. The finding that androgen deprivation therapy (ADT) induces alterations in the androgen receptor (AR) transcriptional program by AR coregulators activity in a context-dependent manner, offers the opportunity for identifying signatures discriminating different clinical states of prostate cancer (PCa) progression. Gel electrophoretic analyses combined with western blot showed that, in androgen-dependent PCa and CRPC in vitro models, the subcellular distribution of spliced and serine-phosphorylated heterogeneous nuclear ribonucleoprotein K (hnRNP K) isoforms can be associated with different AR activities. Using mass spectrometry and bioinformatic analyses, we showed that the protein sets of androgen-dependent (LNCaP) and ADT-resistant cell lines (PDB and MDB) co-immunoprecipitated with hnRNP K varied depending on the cell type, unravelling a dynamic relationship between hnRNP K and AR during PCa progression to CRPC. By comparing the interactome of LNCaP, PDB, and MDB cell lines, we identified 51 proteins differentially interacting with hnRNP K, among which KLK3, SORD, SPON2, IMPDH2, ACTN4, ATP1B1, HSPB1, and KHDRBS1 were associated with AR and differentially expressed in normal and tumor human prostate tissues. This hnRNP K⁻AR-related signature, associated with androgen sensitivity and PCa progression, may help clinicians to better manage patients with CRPC.


Asunto(s)
Ribonucleoproteína Heterogénea-Nuclear Grupo K/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Receptores Androgénicos/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Proliferación Celular/fisiología , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo K/deficiencia , Humanos , Inmunoprecipitación , Masculino , Fosforilación/genética , Fosforilación/fisiología , Neoplasias de la Próstata Resistentes a la Castración/genética , Receptores Androgénicos/deficiencia
4.
Cell Commun Signal ; 15(1): 51, 2017 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-29216878

RESUMEN

BACKGROUND: Prostate cancer (PCa), the second most common cancer affecting men worldwide, shows a broad spectrum of biological and clinical behaviour representing the epiphenomenon of an extreme heterogeneity. Androgen deprivation therapy is the mainstay of treatment for advanced forms but after few years the majority of patients progress to castration-resistant prostate cancer (CRPC), a lethal form that poses considerable therapeutic challenges. METHODS: Western blotting, immunocytochemistry, invasion and reporter assays, and in vivo studies were performed to characterize androgen resistant sublines phenotype in comparison to the parental cell line LNCaP. RNA microarray, mass spectrometry, integrative transcriptomic and proteomic differential analysis coupled with GeneOntology and multivariate analyses were applied to identify deregulated genes and proteins involved in CRPC evolution. RESULTS: Treating the androgen-responsive LNCaP cell line for over a year with 10 µM bicalutamide both in the presence and absence of 0.1 nM 5-α-dihydrotestosterone (DHT) we obtained two cell sublines, designated PDB and MDB respectively, presenting several analogies with CRPC. Molecular and functional analyses of PDB and MDB, compared to the parental cell line, showed that both resistant cell lines were PSA low/negative with comparable levels of nuclear androgen receptor devoid of activity due to altered phosphorylation; cell growth and survival were dependent on AKT and p38MAPK activation and PARP-1 overexpression; their malignant phenotype increased both in vitro and in vivo. Performing bioinformatic analyses we highlighted biological processes related to environmental and stress adaptation supporting cell survival and growth. We identified 15 proteins that could direct androgen-resistance acquisition. Eleven out of these 15 proteins were closely related to biological processes involved in PCa progression. CONCLUSIONS: Our models suggest that environmental factors and epigenetic modulation can activate processes of phenotypic adaptation driving drug-resistance. The identified key proteins of these adaptive phenotypes could be eligible targets for innovative therapies as well as molecules of prognostic and predictive value.


Asunto(s)
Adaptación Fisiológica/efectos de los fármacos , Andrógenos/metabolismo , Resistencia a Antineoplásicos , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/fisiopatología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Fosforilación/efectos de los fármacos , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Receptores Androgénicos/metabolismo , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento
5.
Int J Cancer ; 137(7): 1574-86, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25808111

RESUMEN

Prostate cancer (PCa) displays infrequent point mutations, whereas genomic rearrangements are highly prevalent. In eukaryotes, the genome is compartmentalized into chromatin loop domains by the attachment to the nuclear matrix (NM), and it has been demonstrated that several recombination hot spots are situated at the base of loops. Here, we have characterized the binding between NM proteins and matrix attachment regions (MARs) in PCa. Nontumor and 44 PCa tissues were analyzed. More aggressive tumors were characterized by an increase in the complexity of the NM protein patterns that was synchronous with a decrease in the number of proteins binding the MAR sequences. PARP-1 was the protein that showed the most evident changes. The expression of the PARP-1 associated with NM increased and it was dependent on tumor aggressiveness. Immunohistochemical analysis showed that the protein was significantly overexpressed in tumor cells. To explore the role of PARP-1 in PCa progression, PCa cells were treated with the PARP inhibitor, ABT-888. In androgen-independent PC3 cells, PARP inhibition significantly decreased cell viability, migration, invasion, chromatin loop dimensions and histone acetylation. Collectively, our study provides evidence that MAR-binding proteins are involved in the development and progression of PCa. PARP could play a key role in the compartmentalization of chromatin and in the development of the more aggressive phenotype. Thus, PARP can no longer be viewed only as an enzyme involved in DNA repair, but that its role in chromatin modulation could provide the basis for a new therapeutic approach to the treatment of PCa.


Asunto(s)
Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Proteínas Asociadas a Matriz Nuclear/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Bencimidazoles/farmacología , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Progresión de la Enfermedad , Humanos , Masculino , Regiones de Fijación a la Matriz , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Neoplasias Hormono-Dependientes/metabolismo , Neoplasias Hormono-Dependientes/patología , Proteínas Asociadas a Matriz Nuclear/biosíntesis , Poli(ADP-Ribosa) Polimerasa-1 , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Poli(ADP-Ribosa) Polimerasas/biosíntesis , Neoplasias de la Próstata/tratamiento farmacológico
6.
Prostate ; 73(9): 941-51, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23334998

RESUMEN

BACKGROUND: The role of CXCL1 in prostate cancer (PCa) progression has been poorly investigated. A limitation of previous studies is linked to the use of human PCa cell lines PC3 and DU145, producing CXCL8 at levels strongly exceeding CXCL1 levels. Moreover, in mouse models the sharing of CXCR2 receptor by both ligands makes the phenotype induced by CXCL8 and CXCL1 almost indistinguishable. To overcome this problem we used the murine TRAMP-C2 cell line, not expressing CXCL8 and expressing CXCL1 at low levels. METHODS: The effect of CXCL1 overexpression was examined by in vivo subcutaneous tumor studies and in vitro functional assays of invasion and adhesion. Biochemical modifications were evaluated by Western blotting and antibody arrays. RESULTS: Our data show that the overexpression of CXCL1 in TRAMP-C2 cells represses tumor establishment and in situ invasion. In vitro, the main action of CXCL1 expression in TRAMP cells is associated with the perturbation of molecules linked to cell adhesion and migration thus explaining in vivo data. Other in vitro findings also suggest that signaling by CXCL1 might activate a secretory network limiting in vivo tumor growth by reinforcing senescence. Immunohistochemical staining of human PCa, BPH, and normal prostate biopsies strengthen our observations on the mouse model: when expressed, CXCL1 is limited to small areas with faint staining and PCa progression does not rely on CXCL1 expression. CONCLUSION: We could speculate that CXCL1 overexpression acts as a suppressor of malignancy limiting the escape of tumor cells from the primary tumor and reinforcing growth arrest.


Asunto(s)
Quimiocina CXCL1/biosíntesis , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Animales , Procesos de Crecimiento Celular/fisiología , Senescencia Celular/fisiología , Quimiocina CXCL1/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Invasividad Neoplásica , Neoplasias de la Próstata/genética , Análisis de Matrices Tisulares
7.
J Transl Med ; 11: 108, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23634660

RESUMEN

BACKGROUND: CTLA-4 (Cytotoxic T lymphocyte antigen-4) is traditionally known as a negative regulator of T cell activation. The blocking of CTLA-4 using human monoclonal antibodies, such as Ipilimumab, is currently used to relieve CTLA-4-mediated inhibition of anti-tumor immune response in metastatic melanoma. Herein, we have analyzed CTLA-4 expression and Ipilimumab reactivity on melanoma cell lines and tumor tissues from cutaneous melanoma patients. Then, we investigated whether Ipilimumab can trigger innate immunity in terms of antibody dependent cellular cytotoxicity (ADCC) or Tumor Necrosis Factor (TNF)-α release. Finally, a xenograft murine model was set up to determine in vivo the effects of Ipilimumab and NK cells on melanoma. METHODS: CTLA-4 expression and Ipilimumab reactivity were analyzed on 17 melanoma cell lines (14 primary and 3 long-term cell lines) by cytofluorimetry and on 33 melanoma tissues by immunohistochemistry. CTLA-4 transcripts were analyzed by quantitative RT-PCR. Soluble CTLA-4 and TNF-α were tested by ELISA. Peripheral blood mononuclear cells (PBMC), NK and γδT cells were tested in ADCC assay with Ipilimumab and melanoma cell lines. TNF-α release was analyzed in NK-melanoma cell co-cultures in the presence of ipilimumab. In vivo experiments of xenotransplantation were carried out in NOD/SCID mice. Results were analyzed using unpaired Student's t-test. RESULTS: All melanoma cell lines expressed mRNA and cytoplasmic CTLA-4 but surface reactivity with Ipilimumab was quite heterogeneous. Accordingly, about 2/3 of melanoma specimens expressed CTLA-4 at different level of intensity.Ipilimumab triggered, via FcγReceptorIIIA (CD16), ex vivo NK cells as well as PBMC, IL-2 activated NK and γδT cells to ADCC of CTLA-4+ melanoma cells. No ADCC was detected upon interaction with CTLA-4- FO-1 melanoma cell line. TNF-α was released upon interaction of NK cells with CTLA-4+ melanoma cell lines. Remarkably, Ipilimumab neither affected proliferation and viability nor triggered ADCC of CTLA-4+ T lymphocytes. In a chimeric murine xenograft model, the co-engraftment of Ipilimumab-treated melanoma cells with human allogeneic NK cells delayed and significantly reduced tumor growth, as compared to mice receiving control xenografts. CONCLUSIONS: Our studies demonstrate that Ipilimumab triggers effector lymphocytes to cytotoxicity and TNF-α release. These findings suggest that Ipilimumab, besides blocking CTLA-4, can directly activate the elimination of CTLA-4+ melanomas.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antígeno CTLA-4/metabolismo , Regulación Neoplásica de la Expresión Génica , Melanoma/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Anticuerpos Monoclonales/química , Línea Celular Tumoral , Citometría de Flujo , Humanos , Inmunohistoquímica , Ipilimumab , Células Asesinas Naturales/citología , Melanoma/tratamiento farmacológico , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Receptores de IgG/metabolismo , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Int J Cancer ; 131(3): 582-90, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22015967

RESUMEN

Current diagnostic tools cannot predict clinical failure and androgen-independent disease progression for patients with prostate cancer (PC). The survival signaling pathways of prostate cells play a central role in the progression of tumors to a neuroendocrine (NE) phenotype. NE cells demonstrate attributes that suggest that they are an integral part of the signaling cascade leading to castration-resistant PC. In this study, making use of in vitro neuroendocrine differentiation (NED) of human LNCaP and mouse TRAMP-C2 cells after androgen withdrawal, and of the transgenic adenocarcinoma of mouse prostate (TRAMP) model, we characterized a sequence of molecular events leading to NED and identified a number of markers that could be detectable by routine analyses not only in castration resistant PC but also in hormone naïve PC at the time of initial diagnosis. We found that NED associates with AKT activation that in turn regulates heterogeneous nuclear ribonucleoprotein K (hnRNP K), androgen receptor (AR) and ß-catenin levels. Addition of molecules targeting membrane-bound receptors and protein kinases blocks NE differentiation in LNCaP and TRAMP-C2 cells. The extent of AKT phosphorylation and hnRNP K, AR and ß-catenin levels may have a potential value as prognostic indicators discriminating between androgen-responsive and unresponsive cells and could be used as molecular targets to monitor the anti-tumor action of new therapeutic protocols based on antireceptor agents and/or neuroendocrine hormone antagonists.


Asunto(s)
Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Células Neuroendocrinas/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Transducción de Señal , Andrógenos/metabolismo , Animales , Diferenciación Celular , Línea Celular Tumoral , Ribonucleoproteína Heterogénea-Nuclear Grupo K/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Neuroendocrinas/patología , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Androgénicos/metabolismo , beta Catenina/metabolismo
9.
Mol Med ; 18: 1292-302, 2012 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-22952060

RESUMEN

Despite recent advances in understanding the biological basis of prostate cancer, management of the disease, especially in the phase resistant to androgen ablation, remains a significant challenge. The long latency and high incidence of prostate carcinogenesis provides the opportunity to intervene with chemoprevention to prevent or eradicate prostate malignancies. In this study, we have used human hormone-resistant prostate cancer cells, DU145 and PC3, as an in vitro model to assess the efficacy of xanthohumol (XN) against cell growth, motility and invasion. We observed that treatment of prostate cancer cells with low micromolar doses of XN inhibits proliferation and modulates focal adhesion kinase (FAK) and AKT phosphorylation leading to reduced cell migration and invasion. Oxidative stress by increased production of reactive oxygen species (ROS) was associated with these effects. Transgenic adenocarcinoma of the mouse prostate (TRAMP) transgenic mice were used as an in vivo model of prostate adenocarcinoma. Oral gavage of XN, three times per week, beginning at 4 wks of age, induced a decrease in the average weight of the urogenital (UG) tract, delayed advanced tumor progression and inhibited the growth of poorly differentiated prostate carcinoma. The ability of XN to inhibit prostate cancer in vitro and in vivo suggests that XN may be a novel agent for the management of prostate cancer.


Asunto(s)
Progresión de la Enfermedad , Flavonoides/farmacología , Flavonoides/uso terapéutico , Propiofenonas/farmacología , Propiofenonas/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Administración Oral , Andrógenos/farmacología , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/patología , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Flavonoides/administración & dosificación , Fase G1/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Invasividad Neoplásica , Estadificación de Neoplasias , Propiofenonas/administración & dosificación , Especies Reactivas de Oxígeno/metabolismo , Fase de Descanso del Ciclo Celular/efectos de los fármacos
10.
Microvasc Res ; 83(3): 281-9, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22386654

RESUMEN

Anoxia modulates the expression of molecules associated with endothelial dysfunction and vascular diseases. Polyphenols have potent antioxidant properties due to their ability to modulate genes involved in oxidative tissue damage. In this study, we investigated the effect of polyphenol extract from olive pomace (PEOP) and its main constituents, Tyrosol and Oleuropein, on endothelial cells subjected to anoxia by evaluating the expression of molecules critical for endothelial function, proliferation and migration, and the signaling pathway involved. EAhy926 human endothelial cells were exposed to anoxic stress in the presence or absence of PEOP. Anoxia increased the nitric oxide (NO) level and the expression of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2) and tumor necrosis factor-α (TNFα). These effects were prevented by PEOP treatment in a dose-dependent manner. Moreover, PEOP prevented the proliferation and migration associated with anoxia in EAhy926 cells, down-regulated the levels of matrix metalloproteinase (MMP)-2, MMP-9 and membrane type-1 MMP (MT1-MMP) and increased tissue MMP inhibitor-1 (TIMP-1) expression. Purified Oleuropein or Tyrosol restored to a basal level the anoxia-induced expression of MMP-9 and partially of MMP-2. The expression of TNFα was reduced by both polyphenols in a dose-dependent manner, but more efficiently by Tyrosol. Conversely, Oleuropein and Tyrosol had no significant effects on iNOS, COX-2 and TIMP-1 expression when used at the concentration found in PEOP. PEOP induced a time-dependent phosphorylation of p38 MAPK and ERK1/2 and inhibited anoxia-induced NF-κB activation. PEOP treatment restores the endothelial functions that are impaired by anoxia by regulating the expression of genes involved in proteolysis, angiogenesis and inflammation more efficiently than the single purified components. Therefore, the combined use of polyphenols, as in PEOP, could represent a powerful tool for the treatment and chemoprevention of endothelial dysfunction-associated vascular diseases.


Asunto(s)
Endotelio Vascular/patología , Hipoxia/metabolismo , Olea/metabolismo , Polifenoles/química , Relación Dosis-Respuesta a Droga , Humanos , Sistema de Señalización de MAP Quinasas , Metaloproteinasa 14 de la Matriz/biosíntesis , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/biosíntesis , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fosforilación , Prostaglandina-Endoperóxido Sintasas/metabolismo , Factores de Tiempo , Inhibidor Tisular de Metaloproteinasa-1/biosíntesis , Factor de Necrosis Tumoral alfa/metabolismo
11.
Prostate ; 71(13): 1466-79, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21321982

RESUMEN

BACKGROUND: Bicalutamide (BIC) is widely used in prostate cancer therapy. The dose and schedule employed are well tolerated, but about 50% of patients develop gynecomastia. Several studies have shown a significant reduction of the troublesome effects when Tamoxifen is concomitantly administered with BIC. However, the results reported in the literature seem to be preliminary and possible interferences could be present. In order to clarify the molecular mechanisms of the combination of the two drugs, we have investigated whether the expression of the proteins belonging to nuclear matrix (NM), one modulator of hormone action, is altered by BIC and/or 4-hydroxy-tamoxifen (4OHT) in LNCaP cells. We focused above all on heterogeneous nuclear ribonucleoprotein K (hnRNP K) a NM protein with a key role in prostate carcinoma. METHODS: NM proteins were analyzed by two-dimensional gel electrophoresis. Modulation and compartmentalization of the androgen receptor and the hnRNP K were studied by Western blotting, confocal microscopy, and immunoprecipitation. RESULTS: Proteomic analysis revealed that there is a similarity in the changes of the NM proteins elicited by drugs alone but that their combination does not result in a simple additive effect. Moreover, we found that in the nucleoplasm the androgen receptor and the hnRNP K colocalize in a complex that is highly proximal to DNA and that both proteins were synchronously modulated by BIC and/or 4OHT treatment. CONCLUSION: This study confirm the pivotal role of hnRNP K in prostate carcinoma and suggest that this role might be played by the interaction with the androgen receptor.


Asunto(s)
Antagonistas de Andrógenos/farmacología , Anilidas/farmacología , Núcleo Celular/química , Antagonistas de Estrógenos/farmacología , Ribonucleoproteína Heterogénea-Nuclear Grupo K/análisis , Nitrilos/farmacología , Neoplasias de la Próstata/química , Receptores Androgénicos/análisis , Tamoxifeno/análogos & derivados , Compuestos de Tosilo/farmacología , Línea Celular Tumoral , Supervivencia Celular , Ribonucleoproteína Heterogénea-Nuclear Grupo K/fisiología , Humanos , Masculino , Fosforilación , Antígeno Prostático Específico/análisis , Neoplasias de la Próstata/patología , Tamoxifeno/farmacología
12.
Mol Cancer ; 9: 142, 2010 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-20537156

RESUMEN

BACKGROUND: Prostate cancer shows an extremely slow progression, appearing in its metastatic, hormone refractory phenotype mostly in elderly men. The chemopreventive targeting of this tumor could accordingly delay its malignancy over life expectancy. The cancer chemopreventive retinoid N-(4 hydroxyphenyl)retinamide (4HPR) has already been shown to restrain prostate cancer growth in vitro and in vivo, though its mechanisms of action are only partially explained. RESULTS: We found that 4HPR impairs DU145 and PC3 prostate cancer cells migration and invasion by down-regulating FAK and AKT activation and by enhancing beta-catenin degradation, causing the downregulation of target genes like cyclin D1, survivin and VEGF. This non-migratory phenotype was similarly produced in both cell lines by stable silencing of beta-catenin. 4HPR was able to decrease AKT phosphorylation also when powerfully upregulated by IGF-1 and, consequently, to impair IGF-1-stimulated cell motility. Conversely, the expression of constitutively active AKT (myr-AKT) overcame the effects of 4HPR and beta-catenin-silencing on cell migration. In addition, we found that BMP-2, a 4HPR target with antiangiogenic activity, decreased prostate cancer cell proliferation, migration and invasion by down-regulating the pathway described involving AKT phosphorylation, beta-catenin stability and cyclin D1 expression. CONCLUSION: These data point to 4HPR as a negative regulator of AKT phosphorylation, effectively targeting the beta-catenin pathway and inducing a relatively benign phenotype in prostate cancer cells, limiting neoangiogenesis and cell invasion.


Asunto(s)
Anticarcinógenos/farmacología , Fenretinida/farmacología , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias de la Próstata/patología , Western Blotting , Proteína Morfogenética Ósea 2/biosíntesis , Línea Celular Tumoral , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Silenciador del Gen , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Masculino , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
13.
Cancers (Basel) ; 12(4)2020 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-32295249

RESUMEN

Colorectal cancer's (CRC) ability to invade local tissues and lymph nodes and generate distant metastases is the key for TNM classification. Aspartate-ß-hydroxylase (ASPH), a transmembrane protein that catalyzes Notch receptors and ligand activation, is involved in tumor invasion. Because Notch is involved in gut homeostasis, it could be a target for CRC therapy. ASPH mRNA and protein expression, promoter methylation and gene copy numbers were evaluated using the TCGA and CPTAC human CRC datasets. Using digital pathology, ASPH was scored in the luminal area (LM), center tumor (CT) and invasive margin (IM) of 100 human CRCs. The effect of ASPH targeting on invasiveness and viability was tested by siRNA knockdown and small molecule inhibitors (SMI). Bioinformatics analysis showed increased expression of ASPH mRNA and protein in CRC, paired with a decreased methylation profile. ASPH genetic gain or amplification was frequent (56%), while deletion was rare (0.03%). Digital pathology analysis showed that ASPH exerted its pathological activity in the invasive margin of the tumor, affecting invasive front morphology, tumor budding and patients' overall survival. In vitro, ASPH targeting by siRNA or SMI reduced cell invasion and growth and caused Notch-1 downregulation. This study demonstrates that ASPH targeting by specific inhibitors could improve CRC treatment strategies.

15.
Cells ; 9(3)2020 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-32168749

RESUMEN

Observational/retrospective studies indicate that prostaglandin-endoperoxide synthase-2 (PTGS2) inhibitors could positively affect colorectal cancer (CRC) patients' survival after diagnosis. To obtain an acceptable cost/benefit balance, the inclusion of PTGS2 inhibitors in the adjuvant setting needs a selective criterion. We quantified the 72 kDa, CRC-associated, glycosylated form of PTGS2 in 100 frozen CRC specimens and evaluated PTGS2 localization by IHC in the same tumors, scoring tumor epithelial-derived and stroma-derived fractions. We also investigated the involvement of interleukin-1 beta (IL1ß) in PTGS2 induction, both in vitro and in CRC lysates. Finally, we used overall survival (OS) as a criterion for patient selection. Glycosylated PTGS2 can be quantified with high sensibility in tissue lysates, but the expression in both tumor and stromal cells limits its use for predictive purposes. Immunohistochemistry (IHC) analysis indicates that stromal PTGS2 expression could exert a protective role on patient OS. Stromal PTGS2 was prevalently expressed by cancer-associated fibroblasts exerting a barrier function near the gut lumen, and it apparently favored the antitumor M1 macrophage population. IL1ß was directly linked to gPTGS2 expression both in vitro and in tumors, but its activity was apparently prevalent on the stromal cell population. We suggest that stromal PTGS2 could exert a positive effect on patients OS when expressed in the luminal area of the tumor.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Neoplasias del Colon/tratamiento farmacológico , Neoplasias Colorrectales/tratamiento farmacológico , Ciclooxigenasa 2/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/análisis , Neoplasias del Colon/enzimología , Neoplasias Colorrectales/patología , Inhibidores de la Ciclooxigenasa 2/farmacología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos
16.
Int J Cancer ; 124(12): 2989-96, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19319982

RESUMEN

Tumor growth requires a competent vascular supply and angiogenesis is now considered a potential target for cancer treatment. Chemotherapeutic drugs, and docetaxel in particular, chronically administered using a frequent schedule at low dose (metronomic dosing), can cause potent antiangiogenic effects by targeting the endothelial cells of newly growing blood vessels. Because the exposure to cytotoxic drugs could target both endothelial and tumor cells, we investigated the effects of "metronomic docetaxel" on hormone refractory prostate carcinoma cells. In vitro, metronomic therapy lowered tumor cell viability, inducing apoptosis and reducing the invasive potential at 10- to100-fold lower concentrations as compared with the maximum tolerated dose. Metronomic regimens resulted in a significant reduction of vascular endothelial cell growth factor expression and up-regulation of endogenous angiogenesis inhibitors. Our studies suggest that heterogeneous nuclear ribonucleoprotein K is a mediator of the effects we observed. Targeting heterogeneous nuclear ribonucleoprotein K may serve as a specific antimetastasis and antiangiogenic therapy and could be a potential predictive marker to determine the optimal dose and schedule for metronomic chemotherapy regimens. These findings highlight the multiple effects that may characterize antiangiogenic metronomic chemotherapy and suggest that docetaxel might act as antitumor compound by affecting both cancer and endothelial cells at the same drug concentration. Careful optimization of drug scheduling and dosages will be required to maximize antitumor responses with metronomic approaches.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Antineoplásicos/uso terapéutico , Ribonucleoproteína Heterogénea-Nuclear Grupo K/metabolismo , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/tratamiento farmacológico , Taxoides/uso terapéutico , Apoptosis/efectos de los fármacos , Western Blotting , Movimiento Celular , Proliferación Celular/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , Docetaxel , Ribonucleoproteína Heterogénea-Nuclear Grupo K/antagonistas & inhibidores , Ribonucleoproteína Heterogénea-Nuclear Grupo K/genética , Humanos , Técnicas para Inmunoenzimas , Masculino , Invasividad Neoplásica , Neovascularización Patológica , Fenotipo , Neoplasias de la Próstata/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trombospondina 1/genética , Trombospondina 1/metabolismo , Células Tumorales Cultivadas
17.
Recent Results Cancer Res ; 181: 71-6, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19213559

RESUMEN

Several cancer chemopreventive agents have been demonstrated to exert antiangiogenic effects. Blocking tumor angiogenesis, a process critical for tumor mass expansion and metastasis, represents an intriguing approach not only to cancer therapy, but also to cancer chemoprevention. We found that angiogenesis is a common and key target of many chemopreventive molecules, where they most likely suppress the angiogenic switch in premalignant tumors, a concept we termed "angioprevention." In this manuscript we use as an example the synthetic retinoid N-(4-hydroxyphenyl)retinamide (4HPR), a molecule with confirmed clinical applications in breast cancer adjuvant therapy to prevent cancer recurrence and under evaluation in neuroblastoma and glioblastoma treatment.


Asunto(s)
Antineoplásicos/uso terapéutico , Fenretinida/uso terapéutico , Neoplasias/irrigación sanguínea , Neovascularización Patológica/prevención & control , Humanos
18.
Mol Cancer Ther ; 7(9): 2692-702, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18790751

RESUMEN

The oncogenic Bcr-Abl tyrosine kinase activates various signaling pathways including phosphoinositide 3-kinase/Akt and nuclear factor-kappaB that mediate proliferation, transformation, and apoptosis resistance in Bcr-Abl+ myeloid leukemia cells. The hop flavonoid xanthohumol inhibits tumor growth by targeting the nuclear factor-kappaB and Akt pathways and angiogenesis. Here, we show that xanthohumol has in vitro activity against Bcr-Abl+ cells and clinical samples and retained its cytotoxicity when imatinib mesylate-resistant K562 cells were examined. Xanthohumol inhibition of K562 cell viability was associated with induction of apoptosis, increased p21 and p53 expression, and decreased survivin levels. We show that xanthohumol strongly inhibited Bcr-Abl expression at both mRNA and protein levels and show that xanthohumol caused elevation of intracellular reactive oxygen species and that the antioxidant N-acetylcysteine blunted xanthohumol-induced events. Further, we observed that xanthohumol inhibits leukemia cell invasion, metalloprotease production, and adhesion to endothelial cells, potentially preventing in vivo life-threatening complications of leukostasis and tissue infiltration by leukemic cells. As structural mutations and/or gene amplification in Bcr-Abl can circumvent an otherwise potent anticancer drug such as imatinib, targeting Bcr-Abl expression as well as its kinase activity could be a novel additional therapeutic approach for the treatment of Bcr-Abl+ myeloid leukemia.


Asunto(s)
Antineoplásicos/farmacología , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , FN-kappa B/metabolismo , Propiofenonas/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/genética , Benzamidas , Adhesión Celular/efectos de los fármacos , Proliferación Celular , Supervivencia Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Regulación hacia Abajo , Resistencia a Antineoplásicos/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Flavonoides , Proteínas de Fusión bcr-abl/genética , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Humanos , Mesilato de Imatinib , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , FN-kappa B/antagonistas & inhibidores , Invasividad Neoplásica , Neovascularización Patológica/metabolismo , Piperazinas/farmacología , Pirimidinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Células U937 , Factor A de Crecimiento Endotelial Vascular/metabolismo
19.
Front Immunol ; 10: 961, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31130953

RESUMEN

It is well established that natural killer (NK) cells are involved in both innate and adaptive immunity. Indeed, they can recognize molecules induced at the cell surface by stress signals and virus infections. The functions of NK cells in the gut are much more complex. Gut NK cells are not precisely organized in lymphoid aggregates but rather scattered in the epithelium or in the stroma, where they come in contact with a multitude of antigens derived from commensal or pathogenic microorganisms in addition to components of microbiota. Furthermore, NK cells in the bowel interact with several cell types, including epithelial cells, fibroblasts, macrophages, dendritic cells, and T lymphocytes, and contribute to the maintenance of immune homeostasis and development of efficient immune responses. NK cells have a key role in the response to intestinal bacterial infections, primarily through production of IFNγ, which can stimulate recruitment of additional NK cells from peripheral blood leading to amplification of the anti-bacterial immune response. Additionally, NK cells can have a role in the pathogenesis of gut autoimmune inflammatory bowel diseases (IBDs), such as Crohn's Disease and Ulcerative Colitis. These diseases are considered relevant to the generation of gastrointestinal malignancies. Indeed, the role of gut-associated NK cells in the immune response to bowel cancers is known. Thus, in the gut immune system, NK cells play a dual role, participating in both physiological and pathogenic processes. In this review, we will analyze the known functions of NK cells in the gut mucosa both in health and disease, focusing on the cross-talk among bowel microenvironment, epithelial barrier integrity, microbiota, and NK cells.


Asunto(s)
Inmunidad Mucosa , Mucosa Intestinal/inmunología , Células Asesinas Naturales/inmunología , Animales , Neoplasias Colorrectales/inmunología , Microbioma Gastrointestinal , Humanos , Enfermedades Inflamatorias del Intestino/inmunología , Intestino Grueso/inmunología , Intestino Delgado/inmunología
20.
Clin Cancer Res ; 13(15 Pt 1): 4320-5, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17671111

RESUMEN

By analogy to the success of cardiovascular medicine in reducing mortality through preventive measures, cancer chemoprevention has the potential to significantly reduce incidence and mortality due to tumors. Angiogenesis is an event inhibited by most of the promising cancer chemoprevention compounds, a concept we termed "angioprevention." Here, we review the signaling pathways that are targeted by diverse angioprevention compounds in endothelial cells. We highlight diverse mechanisms of action, implying that combination angioprevention approaches could further improve efficacy and be transferred to clinical practice.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Neoplasias/irrigación sanguínea , Neovascularización Patológica/prevención & control , Transducción de Señal , Animales , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA