Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Oncotarget ; 5(4): 978-92, 2014 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-24657851

RESUMEN

The key role of the Wnt/ß-catenin signaling in colorectal cancer (CRC) insurgence and progression is now recognized and several therapeutic strategies targeting this pathway are currently in developing. Wnt/ß-catenin signaling not only dominates the early stages of sporadic colorectal cancer (SCC), but could also represent the connection between inflammatory bowel diseases (IBD) and increased risk of developing SCC. The knowledge on the sequential molecular events of Wnt-signaling cascade in IBD and during colorectal carcinogenesis, might provide new diagnostic/prognostic markers and could be helpful for optimizing the treatment protocols, thus improving the efficacy of Wnt-targeting therapies. We performed a comparative evaluation of the expression of some crucial molecules participating to Wnt signaling in an animal model of chemically-induced CRC and in human tissues obtained from patients suffering from IBD or at sequential stages of SCC. Specifically, we analyzed upstream events of Wnt signaling including ß-catenin nuclear translocation and loss of E-cadherin and APC functions, and downstream events including c-Myc and Cyclin-D1 expression. We demonstrated that these crucial components of the Wnt/ß-catenin pathway, when evaluated by immunohistochemistry using a multiparametric approach that includes the analyses of both expression and localization, could be potent markers for diagnosis, prevention and therapy in IBD and SCC, also possessing a predictive value for responsiveness to Wnt-targeting therapies. Furthermore, we showed that the animal model of chemically-induced CRC mimics the molecular events of Wnt signaling during IBD and SCC development in humans and may therefore be suitable for testing chemopreventive or therapeutic drugs targeting this pathway.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Animales , Cadherinas/metabolismo , Carcinogénesis/metabolismo , Carcinogénesis/patología , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/terapia , Ciclina D1/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Enfermedades Inflamatorias del Intestino/diagnóstico , Enfermedades Inflamatorias del Intestino/patología , Enfermedades Inflamatorias del Intestino/terapia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Masculino , Adhesión en Parafina , Proteínas Proto-Oncogénicas c-myc/metabolismo , Ratas , Transducción de Señal
2.
J Innate Immun ; 6(1): 72-88, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-23797159

RESUMEN

Thymosin α1 (Tα1) is a naturally occurring thymic peptide used worldwide in clinical trials for the treatment of infectious diseases and cancer. The immunomodulatory activity of Tα1 on innate immunity effector cells has been extensively described, but its mechanism of action is not completely understood. We report that Tα1-exposed human monocyte-derived macrophages (MDMs) assume the typical activated morphology also exhibited by lipopolysaccharide-activated MDMs, but show a comparatively higher ability of internalizing fluorescent beads and zymosan particles. Tα1 exposure also promptly and dramatically stimulates MDM phagocytosis and killing of Aspergillus niger conidia starting as soon as 30 min after challenge. The effect is dose dependent and early coupled to low transcription of the proinflammatory cytokines tumor necrosis factor α and interleukin-6 and unmodified Toll-like receptor expression. The Tα1-stimulated phagocytosis is strictly dependent on the integrity of the microtubule network and protein kinase C activity and occurs by a variation in the classic zipper model, with recruitment of vinculin and actin at the phagosome exhibiting a punctate distribution. These findings indicate that, in human mature MDMs, Tα1 implements pathogen internalization and killing via the stimulation of the complement receptor-mediated phagocytosis. Our observations document that Tα1 is an early and potent activator of innate immunity and reinforce the concept of its pleiotropy.


Asunto(s)
Aspergillus niger/inmunología , Proteínas del Sistema Complemento/metabolismo , Macrófagos/inmunología , Fagocitosis , Timosina/análogos & derivados , Actinas/metabolismo , Células Cultivadas , Activación de Complemento , Citotoxicidad Inmunológica , Regulación hacia Abajo , Humanos , Inmunidad Innata , Mediadores de Inflamación/metabolismo , Interleucina-6/metabolismo , Lipopolisacáridos/inmunología , Fagocitosis/inmunología , Timalfasina , Timosina/inmunología , Timosina/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Vinculina/metabolismo , Zimosan/metabolismo
3.
Ann N Y Acad Sci ; 1270: 13-20, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23050812

RESUMEN

The innate immune response and its cellular effectors-peripheral blood mononuclear cells and differentiated macrophages-play a crucial role in detection and elimination of pathogenic microorganisms. Chemotherapy and some immunosuppressive drugs used after organ transplantation and for treatment of autoimmune diseases have, as main side effect, bone marrow suppression, which can lead to a reduced response of the innate immune system. Hence, many immune-depressed patients have a higher risk of developing bacterial and invasive fungal infections compared with immune-competent individuals. Thymosin α1 (Tα1) immunomodulatory activity on effector cells of the innate immunity has been extensively described, even if its mechanism of action is not completely understood. Here, we report some of the main knowledge on this topic, focusing on our in vitro and in vivo work in progress that reinforce the validity of Tα1 as a stimulatory agent for detection and elimination of pathogens by differentiated macrophages and for restoring immune parameters after chemotherapy-induced myelosuppression.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Inmunidad Celular/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Timosina/análogos & derivados , Adyuvantes Inmunológicos/uso terapéutico , Animales , Humanos , Macrófagos/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Ratas , Timalfasina , Timosina/farmacología , Timosina/uso terapéutico
4.
Biochim Biophys Acta ; 1822(6): 1004-18, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22387884

RESUMEN

Acidic tumor microenvironment and Wnt/ß-catenin pathway activation have been recognized as two crucial events associated with the initiation and progression of cancer. The aim of this study was to clarify the molecular mechanisms underlying the anti-proliferative effects of atrial natriuretic peptide (ANP) as well as to investigate the relationship between the cellular pH and the Wnt/ß-catenin signaling in cancer cells.To pursue our aims, we conducted investigations in DHD/K12/Trb rat colon adenocarcinoma cells. Intracellular pH was measured by Confocal Laser Scanning Microscopy (CLSM) using the lysosensor Green DND-189 probe. Expression of crucial molecules in the Wnt/ß-catenin signaling pathway was analyzed by CLSM, western blot, and real time PCR. Measurements of activation (phosphorylation state) of Akt, ERK1/2, and p38MAPKinase were performed by Reverse-Phase Protein Microarray Analysis (RPMA).We showed that ANP triggered a NHE-1-mediated increase of the intracellular acidity, inhibiting the Wnt/ß-catenin signaling simultaneously. Moreover, we observed that the Wnt1a, a Wnt signaling activator, affected the intracellular pH in an opposite fashion. Results from the comparative analysis of ANP and EIPA (a NHE-1 specific inhibitor) showed that these two molecules affect both the intracellular acidification and the Wnt/ß-catenin signaling cascade. Specifically, ANP acts on the upstream of the cascade, through a Frizzled-mediated activation, while EIPA does on the downstream.We show for the first time that the Akt activity might be a relevant molecular event linking the NHE-1-regulated intracellular pH and the Wnt/ß-catenin signaling. This provides evidence for a cross-talk between the intracellular alkalinization and the Wnt signaling in tumor cells.


Asunto(s)
Factor Natriurético Atrial/farmacología , Neoplasias Colorrectales/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Vía de Señalización Wnt , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Amilorida/análogos & derivados , Amilorida/farmacología , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/fisiopatología , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/fisiopatología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Concentración de Iones de Hidrógeno , Sistema de Señalización de MAP Quinasas , Fosforilación , Ratas , Intercambiador 1 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/antagonistas & inhibidores , beta Catenina/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
5.
Methods Mol Biol ; 823: 215-35, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22081348

RESUMEN

Cancer is the consequence of intra- and extracellular signaling network deregulation that derives from alteration of genetic and proteomic cellular homeostasis. Mapping the individual molecular circuitry of a patient's tumor cells is the starting point for rational personalized therapy.While genes and RNA encode information about cellular status, proteins are considered the engine of the cellular machine, as they are the effective elements that drive cellular functions, such as proliferation, migration, differentiation, and apoptosis. Consequently, investigations of the cellular protein network are considered a fundamental tool to understand cellular functions. In the last decades, increasing interest has been focused on the improvement of new technologies for proteomic analysis. In this context, reverse-phase protein microarrays (RPMAs) have been developed to study and analyze posttranslational modifications that are responsible for principal cell functions and activities. This innovative technology allows the investigation of protein activation as a consequence of protein-protein interaction or biochemical reactions, such as phosphorylation, glycosylation, ubiquitination, protein cleavage, and conformational alterations.Intracellular balance is carefully conserved by constant rearrangements of proteins through the activity of a series of kinases and phosphatases. Therefore, knowledge of the key cellular signaling cascades reveal information regarding the cellular processes driving a tumor's growth (such as cellular survival, proliferation, invasion, and cell death) and response to treatment.Alteration to cellular homeostasis, driven by elaborate intra- and extracellular interactions, has become one of the most studied fields in the era of personalized medicine and targeted therapy. RPMA technology is a valid tool that can be applied to protein analysis of several diseases for the potential to generate protein interaction and activation maps that lead to the identification of critical nodes for individualized or combinatorial target therapy.


Asunto(s)
Análisis por Matrices de Proteínas/métodos , Proteómica/métodos , Animales , Biomarcadores/metabolismo , Biología Computacional , Humanos , Proteínas/análisis , Proteínas/metabolismo , Coloración y Etiquetado
6.
Anticancer Drugs ; 23(1): 32-42, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21849887

RESUMEN

To evaluate the in-vivo preclinical antitumor activity of sanguinarine in a rat syngeneic model of colorectal cancer. The effects of sanguinarine on DHD/K12/TRb colorectal adenocarcinoma cells were first evaluated in vitro by means of ³H-thymidine incorporation, 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) (MTS) assay, and terminal transferase dUTP nick end labeling (TUNEL) microscopy. For the in-vivo studies, DHD/K12/TRb cells (1.5 × 106 cells/0.3 ml of sterile saline/animal) were injected subcutaneously in syngeneic BDIX rats, which were chronically treated with sanguinarine (5 mg/kg/day per os) or control diluent. Tumor growth, body weight, hematologic, and clinical chemistry measurements were monitored in individual animals at defined time intervals. After killing, subcutaneous tumors were explanted from experimental animals for histopathological examination. In vitro, micromolar concentrations of sanguinarine inhibited dose-dependently DHD/K12/TRb cell proliferation and metabolism and induced cell death by apoptosis. In vivo, oral administration of sanguinarine induced a significant inhibition of tumor growth (P<0.01 vs. untreated controls), in the absence of any toxic or side effects. Marked apoptosis and reduced peritumoral vascularization were observed in tumors from sanguinarine-treated rats as compared with the controls. Additional basic studies are needed to fully characterize the mechanism/s underlying the inhibitory effects of sanguinarine on angiogenesis and tumor growth as well as the pharmacological and safety profile of this drug in experimental tumor models. Overall, findings from this study suggest that sanguinarine is a likely candidate for further evaluation in cancer therapy.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos Fitogénicos/farmacología , Benzofenantridinas/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Isoquinolinas/farmacología , Administración Oral , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Etiquetado Corte-Fin in Situ , Masculino , Neovascularización Patológica , Ratas , Ratas Endogámicas , Células Tumorales Cultivadas
7.
Biochem J ; 440(2): 175-83, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21834791

RESUMEN

Acquired drug resistance was found to be suppressed in the doxorubicin-resistant breast cancer cell line MCF7/Dx after pre-treatment with GSNO (nitrosoglutathione). The effect was accompanied by enhanced protein glutathionylation and accumulation of doxorubicin in the nucleus. Among the glutathionylated proteins, we identified three members of the histone family; this is, to our knowledge, the first time that histone glutathionylation has been reported. Formation of the potential NO donor dinitrosyl-diglutathionyl-iron complex, bound to GSTP1-1 (glutathione transferase P1-1), was observed in both MCF7/Dx cells and drug-sensitive MCF7 cells to a similar extent. In contrast, histone glutathionylation was found to be markedly increased in the resistant MCF7/Dx cells, which also showed a 14-fold higher amount of GSTP1-1 and increased glutathione concentration compared with MCF7 cells. These results suggest that the increased cytotoxic effect of combined doxorubicin and GSNO treatment involves the glutathionylation of histones through a mechanism that requires high glutathione levels and increased expression of GSTP1-1. Owing to the critical role of histones in the regulation of gene expression, the implication of this finding may go beyond the phenomenon of doxorubicin resistance.


Asunto(s)
Doxorrubicina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Glutatión/metabolismo , Histonas/metabolismo , Óxido Nítrico/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Complejos de Coordinación/metabolismo , Femenino , Gutatión-S-Transferasa pi/metabolismo , Humanos , S-Nitrosoglutatión/farmacología
8.
Curr Cancer Drug Targets ; 11(5): 572-85, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21486216

RESUMEN

An innovative approach for cancer therapy implies the use of drugs covalently conjugated to macromolecular carriers that specifically target molecules over-expressed on tumor cells. This drug delivery strategy may allow a controlled release of the drug and a high targeting selectivity on tumor cells, increasing drug cytotoxicity and decreasing its undesirable side effects. We provide in vitro and in vivo preclinical data on the antitumor efficacy of ONCOFID™-S, a new bioconjugate of hyaluronic acid (HA) with SN-38 (the CPT11 active metabolite), that support the validity of the drug delivery strategy implying the use of HA as macromolecular carrier of antineoplastic drugs, an approach based on the over-expression of its target CD44 (the receptor for HA-mediated motility) in a wide variety of cancers. We show that ONCOFID™-S exerts a strong in vitro anti-proliferative activity on CD44 over-expressing rat DHD/K12/trb colon adenocarcinoma cells, as well as on gastric, breast, oesophageal, ovarian and lung human cancer cells, higher than that exerted by unconjugated SN-38. We also demonstrated the in vivo anti-tumor efficacy of locoregional treatment with ONCOFID™-S on two pre-clinical models of colorectal cancer (CRC) in BDIX rats: a) syngeneic model of subcutaneous tumor; b) syngeneic model of metastatic tumor induced by injection of cells into the peritoneal cavity, mimicking the clinical situation of peritoneal carcinomatosis. Specifically, in the latter model ONCOFID™-S is able to dramatically reduce all parameters indicative of a poor prognosis in peritoneal metastatization of CRC without any myelotoxicity or mesothelial inflammation. We propose this CD44-targeted therapeutic strategy for locoregional treatment of peritoneal carcinomatosis from CRC, against which systemic chemotherapy results almost inefficient.


Asunto(s)
Camptotecina/análogos & derivados , Carcinoma/tratamiento farmacológico , Portadores de Fármacos/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/análogos & derivados , Neoplasias Peritoneales/tratamiento farmacológico , Pared Abdominal/patología , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/efectos adversos , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/uso terapéutico , Camptotecina/administración & dosificación , Camptotecina/efectos adversos , Camptotecina/química , Camptotecina/uso terapéutico , Carcinoma/metabolismo , Carcinoma/patología , Carcinoma/secundario , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Portadores de Fármacos/efectos adversos , Sistemas de Liberación de Medicamentos/efectos adversos , Evaluación Preclínica de Medicamentos , Humanos , Ácido Hialurónico/efectos adversos , Ácido Hialurónico/uso terapéutico , Concentración 50 Inhibidora , Inyecciones Intraperitoneales , Irinotecán , Masculino , Trasplante de Neoplasias , Neoplasias Peritoneales/metabolismo , Neoplasias Peritoneales/patología , Neoplasias Peritoneales/secundario , Distribución Aleatoria , Ratas , Ratas Endogámicas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...