Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Int J Gynecol Cancer ; 22(5): 748-54, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22635027

RESUMEN

OBJECTIVE: The insulin-like growth factor I receptor (IGF-IR) and BRCA1 affect cell growth and apoptosis. Little information is available about BRCA1 activity on the IGF signaling pathway. This study evaluated the effect of BRCA1 on IGF-IR expression. METHODS: BRCA1 and IGF-IR immunohistochemistry on archival tissues (35 uterine serous carcinomas [USCs] and 17 metastases) were performed. USPC1 and USPC2 cell lines were transiently cotransfected with an IGF-IR promoter construct driving a luciferase reporter gene and a BRCA1 expression plasmid. Endogenous IGF-IR levels were evaluated by Western immunoblotting. RESULTS: We found high BRCA1 and IGF-IR protein expression in primary and metastatic USC tumors. All samples were immunostained for BRCA1-71% strongly stained; and 33/35 (94%) were stained positive for IGF-IR-2 (6%) strongly stained. No difference in BRCA1 and IGF-IR staining intensity was noted between BRCA1/2 mutation carriers and noncarriers. Metastatic tumors stained more intensely for BRCA1 than did the primary tumor site (P = 0.041) and with borderline significance for IGF-IR (P = 0.069). BRCA1 and IGF-IR staining did not correlate to survival. BRCA1 expression led to 35% and 54% reduction in IGF-IR promoter activity in the USPC1 and USCP2 cell lines, respectively. Western immunoblotting showed a decline in phosphorylated IGF-IR and phosphorylated AKT in both transiently and stably transfected cells. CONCLUSIONS: BRCA1 and IGF-IR are highly expressed in USC tumors. BRCA1 suppresses IGF-IR gene expression and activity. These findings suggest a possible biological link between the BRCA1 and the IGF-I signaling pathways in USC. The clinical implications of this association need to be explored.


Asunto(s)
Proteína BRCA1/metabolismo , Carcinoma Papilar/metabolismo , Cistadenocarcinoma Seroso/metabolismo , Regulación Neoplásica de la Expresión Génica , Receptor IGF Tipo 1/genética , Neoplasias del Cuello Uterino/metabolismo , Western Blotting , Carcinoma Papilar/secundario , Proliferación Celular , Cistadenocarcinoma Seroso/secundario , Femenino , Humanos , Técnicas para Inmunoenzimas , Luciferasas/metabolismo , Metástasis de la Neoplasia , Fosforilación , Pronóstico , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor IGF Tipo 1/metabolismo , Células Tumorales Cultivadas , Neoplasias del Cuello Uterino/patología
2.
Hum Vaccin Immunother ; 18(6): 2124784, 2022 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-36315970

RESUMEN

Currently available health economic models for varicella infection are designed to inform the cost-effectiveness of universal varicella vaccination (UVV) compared with no vaccination. However, in countries with an existing UVV program, these models cannot be used to evaluate whether to continue with the current varicella vaccine or to switch to an alternative vaccine. We developed a dynamic transmission model that incorporates the historical vaccination program to project the health and economic impact of changing vaccination strategies. We applied the model to Israel, which initiated UVV in 2008 with a quadrivalent vaccine, MMRV-GSK, and switched to MMRV-MSD in 2016. The model was calibrated to pre-vaccination incidence data before projecting the impact of the historical and future alternative vaccination strategies on the clinical burden of varicella. Total costs and QALYs lost due to varicella infections were projected to compare continuing with MMRV-MSD versus switching to MMRV-GSK in 2022. Over a 50-year time horizon, continuing with MMRV-MSD reduced varicella incidence further by 64%, reaching 35 cases per 100,000 population by 2072, versus a 136% increase in incidence with MMRV-GSK. Continuing with MMRV-MSD reduced cumulative hospitalization and outpatient cases by 48% and 58% (vs. increase of 137% and 91% with MMRV-GSK), respectively. Continuing with MMRV-MSD resulted in 139 fewer QALYs lost with total cost savings of 3% compared with switching to MMRV-GSK, from the societal perspective. In Israel, maintaining the UVV strategy with MMRV-MSD versus switching to MMRV-GSK is projected to further reduce the burden of varicella and cost less from the societal perspective.


Asunto(s)
Varicela , Vacuna contra el Sarampión-Parotiditis-Rubéola , Humanos , Lactante , Vacuna contra la Varicela , Varicela/epidemiología , Varicela/prevención & control , Herpesvirus Humano 3 , Vacunas Combinadas
3.
Gynecol Oncol ; 121(2): 383-9, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21295335

RESUMEN

PURPOSE: The role of the insulin-like growth factor (IGF) system in endometrial cancer has been well established. The IGF-I receptor (IGF-IR) emerged as a promising therapeutic target in a number of cancers. NVP-AEW541 (Novartis Pharma) is a pyrrolo(2,3-d)pyrimidine derivative with specific IGF-IR tyrosine kinase inhibitory activity. NVP-AEW541 has been shown to abrogate IGF-I-mediated IGF-IR autophosphorylation and to reduce activation of the IGF-IR signaling pathways. The aim of the present study was to investigate the anti-proliferative activity of NVP-AEW541 in Type I (endometrioid) and Type II (uterine serous papillary endometrial carcinoma, USPC) endometrial cancer cell lines. METHODS: Type I (ECC-1, Ishikawa) and Type II (USPC-1, USPC-2) endometrial cancer cell lines were treated with NVP-AEW541 in the presence of IGF-I, and the following parameters were measured: IGF-IR, AKT and ERK phosphorylation, apoptosis, proliferation, cell cycle progression and IGF-IR internalization. RESULTS: Results obtained showed that NVP-AEW541 abolished the IGF-I stimulated IGF-IR phosphorylation in all of the cell lines investigated, whereas it abolished AKT and ERK phosphorylation preferentially in ECC-1 and USPC-1 cells. Furthermore, the inhibitor prevented from IGF-I from exerting its antiapoptotic effect in ECC-1, USPC-1 and USPC-2 cells. In addition, proliferation assays showed that NVP-AEW541 caused a decrease in proliferation rate in all of the cell lines. NVP-AEW541 had no major effect on the insulin receptor. CONCLUSION: Our results suggest that specific IGF-IR inhibition by NVP-AEW541 might be a promising therapeutic tool in endometrial cancer.


Asunto(s)
Carcinoma Endometrioide/tratamiento farmacológico , Carcinoma Papilar/tratamiento farmacológico , Cistadenocarcinoma Seroso/tratamiento farmacológico , Neoplasias Endometriales/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Pirroles/farmacología , Receptor IGF Tipo 1/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Carcinoma Endometrioide/enzimología , Carcinoma Endometrioide/patología , Carcinoma Papilar/enzimología , Carcinoma Papilar/patología , Ciclo Celular/efectos de los fármacos , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Cistadenocarcinoma Seroso/enzimología , Cistadenocarcinoma Seroso/patología , Neoplasias Endometriales/enzimología , Neoplasias Endometriales/patología , Femenino , Humanos , Terapia Molecular Dirigida/métodos , Fosforilación , Receptor IGF Tipo 1/biosíntesis , Receptor de Insulina/metabolismo , Transducción de Señal
4.
Poult Sci ; 96(8): 2601-2613, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28472506

RESUMEN

The transportation process is one of the most stressful practices in poultry and livestock management. Extensive knowledge is available on the impact of transport on stress and animal welfare; however, little is known on the impact of transport on the physiology of turkey pullets, their welfare and health, and even less on the process of homeostatic recovery in the post-transport new environment. The main focus of this manuscript was to focus on trauma, stress, and recovery following transport of turkey pullets from nurseries to pullet farms. Specifically, we determined the physiological consequences of transport, the temporal restoration of homeostasis and its effects on immune system function. We hypothesized that stress signaling by stress hormones would directly activate circulating turkey blood leukocytes (TBL), thus inducing a pro-inflammatory response directed towards tissue repair and recovery. Extensive blood analyses prior to transit and during the collecting, transit, and post-transit stages revealed extensive stress (elevated heat shock protein 70) and blunt-force trauma (internal bleeding and muscle damage as well as limb fractures). TBL were shown to increase mRNA expression of cortisol and adrenergic receptors during transit, thus indicating a possible direct response to circulating stress hormones. Consequently, TBL were shown to increase mRNA expression of pro-inflammatory cytokines, as well as that of serum inflammatory proteins (lysozyme and transferrin) partaking in reducing oxygen radicals as demonstrated by consumption of these proteins. The flare-up due to transit related stress diminished with time until 10 d post-transit, a time at which most parameters returned to resting levels. Though general and vaccine-specific antibody levels were not altered by transport-related stress, the physical and physiological injury caused during transport may explain the susceptibility of turkey pullets to opportunist pathogens in the immediate post-transit period.


Asunto(s)
Bienestar del Animal , Homeostasis , Inmunidad Innata , Estrés Fisiológico , Transportes , Pavos/fisiología , Animales , Femenino , Leucocitos/inmunología , Masculino , Factores de Tiempo , Pavos/inmunología
5.
Cancer Lett ; 335(1): 153-9, 2013 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-23402816

RESUMEN

This study evaluated the potential ability of MK-0646 to inhibit IGF1-mediated biological actions and cell signaling events in Type 1 and Type 2 endometrial cancer. We found that MK-0646 treatment significantly decreased IGF1R expression. In addition, pretreatment with MK-0646 decreased the IGF1-induced phosphorylation of IGF1R, AKT and ERK. Apoptosis analyses showed that MK-0646 abolished the anti-apoptotic effect of IGF1. Furthermore, MK-0646 treatment abolished the IGF1-stimulatory effect on proliferation and enhanced the cytotoxic effect of cisplatin. These findings indicate that specific inhibition of IGF1R could be a useful therapeutic approach for Type 1 and Type 2 endometrial cancer.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Cisplatino/farmacología , Neoplasias Endometriales/tratamiento farmacológico , Receptor IGF Tipo 1/antagonistas & inhibidores , Anticuerpos Monoclonales Humanizados , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Factor I del Crecimiento Similar a la Insulina/fisiología , Terapia Molecular Dirigida , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Transducción de Señal
6.
PLoS One ; 8(4): e61537, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23620761

RESUMEN

Accumulating epidemiological evidence shows that obesity is associated with an increased risk of several types of adult cancers, including endometrial cancer. Chronic hyperinsulinemia, a typical hallmark of diabetes, is one of the leading factors responsible for the obesity-cancer connection. Numerous cellular and circulating factors are involved in the biochemical chain of events leading from hyperinsulinemia and insulin resistance to increased cancer risk and, eventually, tumor development. Metformin is an oral anti-diabetic drug of the biguanide family used for treatment of type 2 diabetes. Recently, metformin was shown to exhibit anti-proliferative effects in ovarian and Type I endometrial cancer, although the mechanisms responsible for this non-classical metformin action remain unclear. The insulin-like growth factors (IGFs) play a prominent role in cancer biology and their mechanisms of action are tightly interconnected with the insulin signaling pathways. Given the cross-talk between the insulin and IGF signaling pathways, the aim of this study was to examine the hypothesis that the anti-proliferative actions of metformin in uterine serous carcinoma (USC) are potentially mediated via suppression of the IGF-I receptor (IGF-IR) pathway. Our results show that metformin interacts with the IGF pathway, and induces apoptosis and inhibition of proliferation and migration of USC cell lines with both wild type and mutant p53. Taken together, our results suggest that metformin therapy could be a novel and attractive therapeutic approach for human USC, a highly aggressive variant of endometrial cancer.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Cistadenocarcinoma Seroso/patología , Regulación hacia Abajo/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Insulina/metabolismo , Metformina/farmacología , Neoplasias Uterinas/patología , Adenilato Quinasa/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/genética , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Cistadenocarcinoma Seroso/tratamiento farmacológico , Cistadenocarcinoma Seroso/enzimología , Neoplasias Endometriales/patología , Femenino , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Factor I del Crecimiento Similar a la Insulina/farmacología , Metformina/uso terapéutico , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Neoplasias Uterinas/tratamiento farmacológico , Neoplasias Uterinas/enzimología
7.
Eur J Cancer ; 48(10): 1570-80, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22033326

RESUMEN

The role of the insulin-like growth factors (IGF) in endometrial cancer has been well established. The IGF-I receptor (IGF-IR), which mediates the biological actions of IGF-I, is usually overexpressed in endometrial tumours. Uterine serous carcinoma (USC) constitutes a defined histological category among endometrial cancers. Mutation of the p53 gene appears early in the course of the disease and is considered a key event in the initiation of USC. The aim of the present study was to evaluate the potential interactions between p53 and the IGF-IR in USC. In addition, we investigated the role of p53 as a biomarker in IGF-IR targeted therapies. Immunohistochemical analysis in a collection of 35 USC specimens revealed that IGF-IR is highly expressed in primary and metastatic USC. Likewise, p53 was expressed in 85.7% of primary tumours and 100% of metastases. A significant negative correlation between p53 expression and survival was noticed. In addition, using USC-derived cell lines we provide evidence that p53 regulates IGF-IR gene expression via a mechanism that involves repression of the IGF-IR promoter. We show that the mechanism of action of p53 involves interaction with zinc finger protein Sp1, a potent transactivator of the IGF-IR gene. Finally, we demonstrate that USC tumours overexpressing p53 are more likely to benefit from anti-IGF-IR therapies. In summary, we provide evidence that p53 regulates IGF-IR gene expression in USC cells via a mechanism that involves repression of the IGF-IR promoter. The interplay between the p53 and IGF-I signalling pathways is of major basic and translational relevance.


Asunto(s)
Cistadenocarcinoma Seroso/metabolismo , Regulación Neoplásica de la Expresión Génica , Receptor IGF Tipo 1/biosíntesis , Proteína p53 Supresora de Tumor/metabolismo , Neoplasias Uterinas/metabolismo , Anciano , Anciano de 80 o más Años , Biomarcadores/metabolismo , Análisis Mutacional de ADN , Femenino , Humanos , Inmunohistoquímica/métodos , Persona de Mediana Edad , Modelos Biológicos , Estudios Retrospectivos , Transducción de Señal , Factor de Transcripción Sp1/química , Activación Transcripcional , Dedos de Zinc
8.
Eur J Cancer ; 47(11): 1717-26, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21450456

RESUMEN

Specific insulin-like growth factor-I receptor (IGF-IR) targeting emerged in recent years as a promising therapeutic strategy in cancer. Endometrial cancer is the most common gynaecological cancer in the Western world. The aim of this study was to evaluate the potential of cixutumumab (IMC-A12, ImClone Systems), a fully human monoclonal antibody against the IGF-IR, to inhibit IGF-I-mediated biological actions and cell signalling events in four endometrial carcinoma-derived cell lines (ECC-1, Ishikawa, USPC-1 and USPC-2). Our results demonstrate that cixutumumab was able to block the IGF-I-induced autophosphorylation of the IGF-IR. In addition, the PI3K and MAPK downstream signalling pathways were also inactivated by cixutumumab in part of the cell lines. Prolonged (24h and 48h) exposures to cixutumumab reduced IGF-IR expression. Furthermore, confocal microscopy of GFP-tagged receptors shows that cixutumumab treatment led to IGF-IR redistribution from the cell membrane to the cytoplasm. Antiapoptotic effects were evaluated by cleavage of caspase 3 and PARP, and mitogenicity and transformation by proliferation and cell cycle assays. Results obtained showed that cixutumumab abrogated the IGF-I-stimulated increase in proliferation rate, and increased caspase-3 and PARP cleavage, two markers of apoptosis. Of importance, cixutumumab had no effect neither on insulin receptor (IR) expression nor on IGF-I activation of IR. In summary, in a cellular model of endometrial cancer cixutumumab was able to inhibit the IGF-I-induced activation of intracellular cascades, apoptosis and proliferation.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Neoplasias Endometriales/metabolismo , Regulación Neoplásica de la Expresión Génica , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Receptor IGF Tipo 1/metabolismo , Anticuerpos Monoclonales Humanizados , Apoptosis , Ciclo Celular , Proliferación Celular , Femenino , Humanos , Microscopía Fluorescente/métodos , Fosforilación , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA