Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
1.
Elife ; 112022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35001873

RESUMEN

Atherosclerosis preferentially occurs in atheroprone vasculature where human umbilical vein endothelial cells are exposed to disturbed flow. Disturbed flow is associated with vascular inflammation and focal distribution. Recent studies have revealed the involvement of epigenetic regulation in atherosclerosis progression. N6-methyladenosine (m6A) is the most prevalent internal modification of eukaryotic mRNA, but its function in endothelial atherogenic progression remains unclear. Here, we show that m6A mediates the epidermal growth factor receptor (EGFR) signaling pathway during EC activation to regulate the atherosclerotic process. Oscillatory stress (OS) reduced the expression of methyltransferase like 3 (METTL3), the primary m6A methyltransferase. Through m6A sequencing and functional studies, we determined that m6A mediates the mRNA decay of the vascular pathophysiology gene EGFR which leads to EC dysfunction. m6A modification of the EGFR 3' untranslated regions (3'UTR) accelerated its mRNA degradation. Double mutation of the EGFR 3'UTR abolished METTL3-induced luciferase activity. Adenovirus-mediated METTL3 overexpression significantly reduced EGFR activation and endothelial dysfunction in the presence of OS. Furthermore, thrombospondin-1 (TSP-1), an EGFR ligand, was specifically expressed in atheroprone regions without being affected by METTL3. Inhibition of the TSP-1/EGFR axis by using shRNA and AG1478 significantly ameliorated atherogenesis. Overall, our study revealed that METTL3 alleviates endothelial atherogenic progression through m6A-dependent stabilization of EGFR mRNA, highlighting the important role of RNA transcriptomics in atherosclerosis regulation.


Asunto(s)
Adenosina/análogos & derivados , Aterosclerosis/fisiopatología , Estabilidad del ARN , ARN Mensajero/metabolismo , Adenosina/genética , Adenosina/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Células Endoteliales/fisiología , Genes erbB-1/genética , Genes erbB-1/fisiología , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Metilación , Metiltransferasas/genética , Metiltransferasas/metabolismo , Ratones , Ratones Endogámicos C57BL
2.
Mol Biol Cell ; 32(8): 788-799, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33566630

RESUMEN

During Caenorhabditis elegans larval development, an inductive signal mediated by the LET-23 EGFR (epidermal growth factor receptor), specifies three of six vulva precursor cells (VPCs) to adopt vulval cell fates. An evolutionarily conserved complex consisting of PDZ domain-containing scaffold proteins LIN-2 (CASK), LIN-7 (Lin7 or Veli), and LIN-10 (APBA1 or Mint1) (LIN-2/7/10) mediates basolateral LET-23 EGFR localization in the VPCs to permit signal transmission and development of the vulva. We recently found that the LIN-2/7/10 complex likely forms at Golgi ministacks; however, the mechanism through which the complex targets the receptor to the basolateral membrane remains unknown. Here we found that overexpression of LIN-10 or LIN-7 can compensate for loss of their complex components by promoting LET-23 EGFR signaling through previously unknown complex-independent and receptor-dependent pathways. In particular, LIN-10 can independently promote basolateral LET-23 EGFR localization, and its complex-independent function uniquely requires its PDZ domains that also regulate its localization to Golgi. These studies point to a novel complex-independent function for LIN-7 and LIN-10 that broadens our understanding of how this complex regulates targeted sorting of membrane proteins.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Receptores ErbB/metabolismo , Proteínas de la Membrana/metabolismo , Vulva/embriología , Animales , Caenorhabditis elegans/embriología , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/fisiología , Proteínas Portadoras/metabolismo , Diferenciación Celular , Membrana Celular/metabolismo , Receptores ErbB/fisiología , Femenino , Genes erbB-1/fisiología , Proteínas del Helminto/metabolismo , Proteínas de la Membrana/fisiología , Transporte de Proteínas , Transducción de Señal , Vulva/metabolismo
3.
J Clin Pharm Ther ; 46(2): 256-266, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33152129

RESUMEN

WHAT IS KNOWN AND OBJECTIVE: Smoking has a notable influence on the efficacy of medications for lung cancer. Previous studies illustrated the correlation between smoking and the efficacy of first-line Epidermal Growth Factor Receptors-Tyrosine Kinase Inhibitors (EGFR-TKIs). The benefit of smokers in immunotherapy was still controversial. Here, we investigated the impact of smoking on clinical outcomes of molecularly targeted therapies or immunotherapy in Non-Small Cell Lung Cancer (NSCLC). METHODS: We performed meta-analysis including trials comparing EGFR-TKIs, Anaplastic Lymphoma Kinase (ALK) inhibitors or Immune Checkpoint Inhibitors (ICIs) against chemotherapy in NSCLC. The Progression-Free Survival (PFS)-Hazard Ratios (HRs) of two groups served as the index and we used random effects to pool outcomes. RESULTS AND DISCUSSION: Twenty randomized trials were selected. Compared with chemotherapy, treatment with EGFR-TKIs had similar benefit in never-smokers (PFS: HR = 0.46, 95% CI 0.30 to 0.69) and smokers (PFS: HR = 0.68, 95% CI 0.50 to 0.91; p = 0.135) while non-smokers (PFS: HR = 0.32, 95% CI 0.23 to 0.44) had better benefit from first-line EGFR-TKIs than smokers (PFS: HR = 0.54, 95% CI 0.41 to 0.71; p = 0.02). Treatment with ALK inhibitors had similar benefits in never-smokers (PFS: HR = 0.43, 95% CI 0.35 to 0.53) and smokers (PFS: HR = 0.56, 95% CI 0.44 to 0.71; p = 0.406). The benefit of ICIs in smokers (PFS: HR = 0.79, 95% CI 0.64 to 0.98) was significantly greater than never-smokers (PFS: HR = 1.81, 95% CI 1.27 to 2.57; p = 0.004). WHAT IS NEW AND CONCLUSION: Smoking status is an important clinical predictor of therapy in NSCLC. Never-smokers and smokers have similar benefit with EGFR-TKIs therapy compared with chemotherapy, while never-smokers have greater benefit after first-line EGFR-TKIs therapy. There was similar benefit in never-smokers and smokers when using ALK inhibitors over chemotherapy. Additionally, ICIs treatment over chemotherapy leads to more favourable PFS in smokers both in first-line and second-line settings.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/epidemiología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/epidemiología , Fumar/epidemiología , Quinasa de Linfoma Anaplásico/antagonistas & inhibidores , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Genes erbB-1/fisiología , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Supervivencia sin Progresión , Inhibidores de Proteínas Quinasas/uso terapéutico , Ensayos Clínicos Controlados Aleatorios como Asunto
4.
Mol Biol Rep ; 47(6): 4553-4568, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32383136

RESUMEN

Cervical cancer is one of the most common disorders in females all around the world. Similar to other types of cancer, several signaling pathways are demonstrated to be involved in the progression of this cancer including ERK/MAPK, PI3K/AKT, apoptotic signaling pathways, Wnt, and epidermal growth factor receptor (EGFR). Various microRNAs (miRNAs) and their target genes involved in cervical cancer have been extracted from the kinds of literature of Scopus, Pubmed and Google scholar databases. Regarding the targets, some of them were found to belong in EGFR signaling pathways. The regulation patterns of these miRNA are different in cervical cancer; however, their main aim is to trigger EGFR signaling to proceed with cancer. Moreover, several predicted miRNAs were found to have some interactions with the differentially expressed genes of cervical cancer which are the members of the EGFR signaling pathway by using miRWalk 3.0 (https://mirwalk.umm.uni-heidelberg.de/) and TargetScan 7.1 (https://www.targetscan.org/vert_71/). Also, the microarray data were obtained from the NCBI-Gene Expression Omnibus (GEO) datasets of cervical cancer. In the present review, we highlight the miRNAs involved in cervical cancer and the role of their targets in the EGFR signaling pathway. Furthermore, some predicted miRNAs were the candidate to target EGFR signaling pathway members differentially expressed in cervical cancer samples compared to normal samples.


Asunto(s)
Receptores ErbB/genética , MicroARNs/genética , Neoplasias del Cuello Uterino/genética , Apoptosis/genética , Biomarcadores de Tumor/genética , Biología Computacional/métodos , Femenino , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Genes erbB-1/genética , Genes erbB-1/fisiología , Humanos , MicroARNs/metabolismo , Transducción de Señal/genética
5.
Mol Biol Rep ; 46(5): 5645-5659, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31280421

RESUMEN

Drug resistance is a major setback for advanced therapeutics in multiple cancers. The increasing prevalence of this resistance is a growing concern and bitter headache for the researchers since a decade. Hence, it is essential to revalidate the existing strategies available for cancer treatment and to look after a novel therapeutic approach for target based killing of cancer cells at the genetic level. This review outlines the different mechanisms enabling resistance including drug efflux, drug target alternation, alternative splicing, the release of the extracellular vesicle, tumor heterogeneity, epithelial-mesenchymal transition, tumor microenvironment, the secondary mutation in the receptor, epigenetic alternation, heterodimerization of receptors, amplification of target and amplification of components rather than the target. Furthermore, existing evidence and the role of various signaling pathways like EGFR, Ras, PI3K/Akt, Wnt, Notch, TGF-ß, Integrin-ECM signaling in drug resistance are explained. Lastly, the prevention of this resistance by a contemporary therapeutic strategy, i.e., a combination of specific signaling pathway inhibitors and the cocktail of a cancer drug is summarized showing the new treatment strategies.


Asunto(s)
Resistencia a Antineoplásicos/genética , Resistencia a Antineoplásicos/fisiología , Neoplasias/tratamiento farmacológico , Antineoplásicos/farmacología , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/efectos de los fármacos , Genes erbB-1/fisiología , Humanos , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/fisiología , Neoplasias/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Microambiente Tumoral/efectos de los fármacos , Vía de Señalización Wnt/fisiología
6.
Medicine (Baltimore) ; 98(12): e14924, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30896652

RESUMEN

BACKGROUND: P-cadherin is a calcium-dependent cell-cell adhesion glycoprotein. It has been implicated in invasiveness and metastasis. However, the clinical prognostic value of overexpression of P-cadherin in patients with breast cancer (BC) remains unsettled. METHODS: A systematic literature search will be performed in all available databases to quantitatively review eligible studies and identify all relevant data, which could be used to detect the relationship between overexpression of P-cadherin and overall survival (OS), disease-free survival (DFS), and clinicopathological parameters. Hazard ratio and 95% confidence intervals (CIs) or P value will be employed as effect measures to estimate the correlation between P-cadherin and the oncologic outcomes including overall survival (OS), disease-free survival (DFS). Odds ratios (ORs) and the 95% CIs will be evaluated for the pooled analysis of the correlation between P-cadherin expression and clinicopathological features. We will use the Review Manager (Revman) 5.3.5 software (Cochrane Community, London, United Kingdom) and STATA 14 software (version 14.0; Stata Corp, College Station, TX) to perform the meta-analysis to calculate the data. RESULTS: The review will provide a high-quality synthesis of current evidence of the prognostic role of P-cadherin in BCs. The results will be published in a peer-reviewed journal. CONCLUSION: We hope that the results of this study will provide significant evidence to assess whether the expression of P-cadherin is associated with poor prognosis in patients with BC. PROSPERO REGISTRATION NUMBER: This meta-analysis protocol has been registered in the PROSPERO network with registration number: CRD42019119880.


Asunto(s)
Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Cadherinas/biosíntesis , Biomarcadores de Tumor , Neoplasias de la Mama/sangre , Femenino , Genes erbB-1/fisiología , Genes erbB-2/fisiología , Humanos , Metástasis Linfática , Invasividad Neoplásica , Estadificación de Neoplasias , Oportunidad Relativa , Pronóstico , Proyectos de Investigación , Análisis de Supervivencia
7.
Biosystems ; 149: 125-138, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27297545

RESUMEN

The model checking method has been long since established as an important tool for modelling and reverse engineering of biological systems. However, due to a high complexity of both the method and the biological systems, this approach often requires a vast amount of computational resources. In this article we show that by reducing the expressivity of the method we can gain performance while still being able to use all biologically relevant data. We utilize this approach to conduct a study of mutations in the EGFR signalling, motivated by a paper from Klinger et al. (2013). Here we aim at constructing approximated models of multiple cell-lines from sizeable sets of experimental data. Due to cancerous mutations in each cell line, there is a high degree of parameter uncertainty and the study would not be practically tractable without the performance optimizations described here.


Asunto(s)
Biología Computacional/métodos , Bases de Datos Factuales , Redes Reguladoras de Genes , Modelos Teóricos , Animales , Redes Reguladoras de Genes/fisiología , Genes erbB-1/fisiología , Humanos
8.
Reproduction ; 151(2): 179-85, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26612427

RESUMEN

Human chorionic gonadotropin (hCG) is a known trigger of ovarian hyperstimulation syndrome (OHSS), a potentially life-threatening complication of assisted reproduction. Administration of hCG results in the release of vascular endothelial growth factor (VEGF) from the ovary. We have previously shown that expression of pigment epithelium-derived factor (PEDF) in granulosa cell line is regulated by hCG, reciprocally to VEGF, and that the PEDF-VEGF balance is impaired in OHSS. Our aim was to explore the signaling network by which hCG downregulates the expression of PEDF mRNA and protein in granulosa cells. We applied specific chemical inhibitors and stimuli to human primary granulosa cells and rat granulosa cell line. We found that PKA and protein kinase C, as well as EGFR, ERK1/2 and PI3K, participate in the signaling network. The finding that hCG-induced PEDF downregulation and VEGF upregulation are mediated by similar signaling cascades emphasizes the delicate regulation of ovarian angiogenesis.


Asunto(s)
Gonadotropina Coriónica/farmacología , Proteínas del Ojo/genética , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/metabolismo , Factores de Crecimiento Nervioso/genética , Serpinas/genética , Adulto , Animales , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Proteínas del Ojo/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Genes erbB-1/fisiología , Humanos , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/genética , Factores de Crecimiento Nervioso/metabolismo , Ovario/irrigación sanguínea , Ovario/efectos de los fármacos , Ratas , Serpinas/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Adulto Joven
9.
Zhonghua Zhong Liu Za Zhi ; 37(5): 342-6, 2015 May.
Artículo en Chino | MEDLINE | ID: mdl-26463023

RESUMEN

OBJECTIVE: To investigate the effect and explore its possible mechanisms of epidermal growth factor receptor(EGFR) expression in macrophages on the anti-cancer effect of berberine (BER) on the growth of colorectal cancer. METHODS: Mice with EGFR gene defects in macrophages (Egfr(fl/fl) LysM-Cre) and with EGFR gene expression in macrophages (LysM-Cre) (control group) were treated with azoxymethane (AOM) to establish colorectal tumor models. These models were treated with or without berberine (BER) intervention. The number of colorectal tumors and the gut length in the two groups were measured. The proliferation of tumor cells was detected by Ki-67 immunohistochemistry and apoptosis was detected by annexin V-FITC fluorescence labeling. Western blot was used to detect the expression of cleaved-caspase-3 protein. RESULTS: After treated with AOM, the colorectal tumor number was 10.26 ± 1.43 in the LysM-Cre group and 7.62 ± 1.05 in the Egfr(fl/fl) LysM-Cre group, showing a significant difference (P = 0.021). The gut length was (6.04 ± 1.06) cm in the LysM-Cre group and (6.39 ± 0.92) cm in the gfrfl/flLysM-Cre group, with a non-significant difference between the two groups (P = 0.075). After treated with AOM plus BER intervention, the colorectal tumor number of the LysM-Cre group was 8.35 ± 1.22 and that in the Egfr(fl/fl) LysM-Cre group was 2.66 ± 0.38, showing a very significant difference between the two groups (P = 0.006). The gut length of the LysM-Cre group was (7.34 ± 1.16) cm and that of the Egfr(fl/fl) LysM-Cre group was (10.01 ± 1.72) cm (P = 0.028). After treated with AOM, the ratio of Ki-67-positive tumor cells in the LysM-Cre group was (78.31 ± 3.43)% and that in the Egfr(fl/fl) LysM-Cre group was (75.85 ± 2.92)% (P = 0.282). After AOM plus BER treatment, the ratio of Ki-67-positive tumor cells in the LysM-Cre group was (42.43 ± 3.09)% and that in the Egfr(fl/fl) LysM-Cre group was significantly lower (29.65 ± 2.47)% (P = 0.018). The ratio of annexin V-positive tumor cells was (0.95 ± 0.13)% in the LysM-Cre group, not significantly different from (1.13 ± 0.16)% in the Egfr(fl/fl) LysM-Cre group (P = 0.175). After AOM plus BER treatment, the ratio of annexin V-positive tumor cells in the LysM-Cre group was (32.10 ± 1.97)%, significantly lower than the (47.08 ± 2.83)% in the Egfr(fl/fl) LysM-Cre group (P = 0.010). The level of cleaved-caspase-3 protein expression was 235.92 ± 19.73 in the Egfr(fl/fl) LysM-Cre group, significantly higher than the 119.71 ± 12.87 in the LysM-Cre group (P = 0.012). CONCLUSIONS: The growth of colorectal cancer cells in mice can be inhibited by BER treatment, and this anti-cancer effect of BER can be further enhanced by EGFR gene knockout in macrophages. The mechanisms may be related to the inhibition of proliferation and promotion of apoptosis in colorectal cancer cells.


Asunto(s)
Antineoplásicos/farmacología , Berberina/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Genes erbB-1/fisiología , Macrófagos/metabolismo , Animales , Antineoplásicos/uso terapéutico , Apoptosis , Berberina/uso terapéutico , Caspasa 3 , Inmunohistoquímica , Ratones
10.
Rev. neurol. (Ed. impr.) ; 61(2): 85-93, 16 jul., 2015. ilus
Artículo en Español | IBECS | ID: ibc-141839

RESUMEN

El glioblastoma es un tumor cerebral primario muy agresivo y resistente al tratamiento convencional con quimio y radioterapia. Dado que el receptor del factor de crecimiento epidérmico (EGFR) se encuentra alterado en el 50% de los glioblastomas, representa actualmente una de las dianas terapéuticas más prometedoras en este tipo de tumores. Sin embargo, los inhibidores de la actividad cinasa del EGFR han generado escasos resultados en ensayos clínicos con pacientes con glioblastoma. En esta revisión se analiza la función del EGFR en el glioblastoma y se describen las aproximaciones terapéuticas dirigidas frente a dicho receptor en este tipo de tumores. Este tipo de análisis podría constituir un punto de partida para mejorar el diseño de futuras terapias para los glioblastomas, basadas en la inhibición de la función del EGFR (AU)


A glioblastoma is a primary brain tumour that is very aggressive and resistant to conventional treatment with chemo- or radiotherapy. Given that epidermic growth factor receptor (EGFR) is altered in 50% of glioblastomas, it is currently one of the most promising therapeutic targets in this kind of tumour. Yet, inhibitors of the kinase activity of EGFR have yielded poor results in clinical trials with patients with glioblastomas. In this review we analyse the function of EGFR in glioblastomas and outline the therapeutic approaches aimed against this receptor in this kind of tumour. This sort of analysis could be a starting point for improving the design of future therapies for glioblastomas, based on inhibiting the EGFR function (AU)


Asunto(s)
Femenino , Humanos , Masculino , Genes erbB-1/genética , Péptidos y Proteínas de Señalización Intercelular/administración & dosificación , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Glioblastoma/complicaciones , Glioblastoma/metabolismo , Carcinogénesis/genética , Mitosis/genética , Ensayos Clínicos como Asunto/instrumentación , Ensayos Clínicos como Asunto/métodos , Tomografía/métodos , Genes erbB-1/fisiología , Péptidos y Proteínas de Señalización Intercelular/análisis , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Glioblastoma/patología , Glioblastoma/secundario , Carcinogénesis/patología , Mitosis/fisiología , Ensayos Clínicos como Asunto/normas , Ensayos Clínicos como Asunto , Tomografía/instrumentación
11.
J Pediatr Endocrinol Metab ; 28(1-2): 7-17, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25503464

RESUMEN

Adamantinomatous craniopharyngiomas (ACPs) are histologically benign but clinically aggressive epithelial tumours of the sellar region that are associated with high morbidity and occasional mortality. Research from the last 3 years has provided important insights into the molecular and cellular pathogenesis of these tumours. It has become established that mutations in CTNNB1 (encoding ß-catenin), leading to the over-activation of the WNT pathway, underlie the molecular aetiology of human ACP. Interestingly, the effect of these mutations is restricted to a small number of tumour cells, mostly forming clusters, which recent research has shown to be critical for tumorigenesis in mice and humans. Several pathways have been found to be activated in these clusters including the epidermal growth factor receptor and the sonic hedgehog pathways, offering potential therapeutic targets. A novel and unexpected role for pituitary stem cells has been proposed, which is fundamentally distinct from the cancer stem cell paradigm. The study of these benign tumours could reveal important insights into general mechanisms underlying the initial steps of tumorigenesis and facilitate novel tools to improve managements of the patients.


Asunto(s)
Craneofaringioma , Modelos Animales de Enfermedad , Ratones , Neoplasias Hipofisarias , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Craneofaringioma/genética , Craneofaringioma/patología , Genes erbB-1/fisiología , Proteínas Hedgehog/fisiología , Humanos , Mutación , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/patología , Vía de Señalización Wnt/genética
13.
J Neurochem ; 130(2): 291-300, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24646423

RESUMEN

Expression of a familial Alzheimer's disease (AD)-linked mutant of amyloid ß precursor protein (APP) or the binding of transforming growth factor ß2 to wild-type (wt)-APP causes neuronal death by activating an intracellular death signal (a APP-mediated intracellular death signal) in the absence of the involvement of amyloid ß (Aß) toxicity in vitro. These neuronal death models may therefore be regarded as Aß-independent neuronal death models related to AD. A recent study has shown that the A673T mutation in the APP isoform APP770 , corresponding to the A598T mutation in the most prevalent neuronal APP isoform APP695 (an AD-protective mutant of APP), is linked to a reduction in the incidence rate of AD. Consistent with this, cells expressing the AD-protective mutant of APP produce less Aß than cells expressing wt-APP. In this study, transforming growth factor ß2 caused death in cultured neuronal cells expressing wt-APP, but not in those expressing the AD-protective mutant of APP. This result suggests that the AD-protective mutation of APP reduces the incidence rate of AD by attenuating the APP-mediated intracellular death signal. In addition, a mutation that causes hereditary cerebral hemorrhage with amyloidosis-Dutch type also attenuated the APP-mediated intracellular death signal. The A598T mutation of amyloid precursor protein APP is linked to a reduction in the incidence rate of Alzheimer's disease (AD). This study shows that TGFß2 causes death in neuronal cells expressing wild-type APP, but not in those expressing the AD-protective mutant of APP, suggesting that the AD-protective mutation of APP reduces the incidence rate of AD by attenuating the APP-mediated intracellular death signal.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/toxicidad , Mutación/fisiología , Síndromes de Neurotoxicidad/genética , Síndromes de Neurotoxicidad/patología , Péptidos beta-Amiloides/farmacología , Amiloidosis/genética , Amiloidosis/patología , Animales , Western Blotting , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/fisiología , Genes erbB-1/genética , Genes erbB-1/fisiología , Humanos , MAP Quinasa Quinasa 4/metabolismo , Ratones , Mutación/genética , Neuronas/efectos de los fármacos , Neuronas/fisiología , Fragmentos de Péptidos/farmacología , Fosforilación
14.
J Nat Prod ; 77(2): 250-7, 2014 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-24456004

RESUMEN

Since the administration of synthetic medicines is associated with drug resistance and undesired side effects, utilization of natural compounds could be an alternative and complementary modality to inhibit or prevent the development of tumors. Epigallocatechin 3-O-gallate (EGCG, 1), the major flavan component of green tea, and genistein (2), a soy isoflavonoid, are known to have chemopreventive and chemotherapeutic effects against cancer. This study demonstrated that both flavonoids inhibit cell proliferation, an effect enhanced under serum-free conditions. Compound 1, but not 2, induced downregulation of ErbB1 and ErbB2 in mammary and epidermoid carcinoma cells, and its inhibitory effect on cell viability was mediated by the 67 kDa laminin receptor (67LR). While 1 was superior in inducing cell death, 2 was more efficient in arresting the tumor cells in the G2/M phase. Furthermore, number and brightness analysis revealed that 1 decreased the homoclustering of a lipid raft marker, glycosylphosphatidylinositol-anchored GFP, and it also reduced the co-localization between lipid rafts and 67LR. The main conclusion made is that the primary target of 1 may be the lipid raft component of the plasma membrane followed by secondary changes in the expression of ErbB proteins. Compound 2, on the other hand, must have other unidentified targets.


Asunto(s)
Anticarcinógenos/química , Anticarcinógenos/farmacología , Catequina/análogos & derivados , Flavonoides/farmacología , Genisteína/farmacología , Receptores de Laminina/efectos de los fármacos , Proteínas Ribosómicas/efectos de los fármacos , Té/química , Algoritmos , Anticarcinógenos/aislamiento & purificación , Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/metabolismo , Catequina/química , Catequina/aislamiento & purificación , Catequina/farmacología , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Femenino , Flavonoides/química , Flavonoides/aislamiento & purificación , Puntos de Control de la Fase G2 del Ciclo Celular/genética , Puntos de Control de la Fase G2 del Ciclo Celular/fisiología , Genes erbB-1/genética , Genes erbB-1/fisiología , Genes erbB-2/genética , Genes erbB-2/fisiología , Genisteína/química , Genisteína/aislamiento & purificación , Humanos , Estructura Molecular , Polifenoles/farmacología , Receptores de Laminina/genética , Receptores de Laminina/metabolismo , Receptores de Laminina/fisiología , Glycine max/química
18.
J Endocrinol ; 214(2): 165-75, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22593082

RESUMEN

Progesterone and progestins have been demonstrated to enhance breast cancer cell migration, although the mechanisms are still not fully understood. The protease-activated receptors (PARs) are a family of membrane receptors that are activated by serine proteases in the blood coagulation cascade. PAR1 (F2R) has been reported to be involved in cancer cell migration and overexpressed in breast cancer. We herein demonstrate that PAR1 mRNA and protein are upregulated by progesterone treatment of the breast cancer cell lines ZR-75 and T47D. This regulation is dependent on the progesterone receptor (PR) but does not require PR phosphorylation at serine 294 or the PR proline-rich region mPRO. The increase in PAR1 mRNA was transient, being present at 3  h and returning to basal levels at 18  h. The addition of a PAR1-activating peptide (aPAR1) to cells treated with progesterone resulted in an increase in focal adhesion (FA) formation as measured by the cellular levels of phosphorylated FA kinase. The combined but not individual treatment of progesterone and aPAR1 also markedly increased stress fiber formation and the migratory capacity of breast cancer cells. In agreement with in vitro findings, data mining from the Oncomine platform revealed that PAR1 expression was significantly upregulated in PR-positive breast tumors. Our observation that PAR1 expression and signal transduction are modulated by progesterone provides new insight into how the progestin component in hormone therapies increases the risk of breast cancer in postmenopausal women.


Asunto(s)
Neoplasias de la Mama/patología , Carcinoma/patología , Movimiento Celular/efectos de los fármacos , Adhesiones Focales/efectos de los fármacos , Progesterona/farmacología , Receptor PAR-1/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Carcinoma/genética , Carcinoma/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Evaluación Preclínica de Medicamentos , Femenino , Adhesiones Focales/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes erbB-1/fisiología , Humanos , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 1/fisiología , Posmenopausia/genética , Posmenopausia/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/fisiología , Receptor PAR-1/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
19.
Rev. esp. patol ; 45(1): 14-28, ene.-mar. 2012.
Artículo en Español | IBECS | ID: ibc-96570

RESUMEN

Actualmente los pacientes con cáncer de pulmón no microcítico (CPNM) avanzado portadores de mutaciones del receptor del factor de crecimiento epidérmico (EGFR), y probablemente en un futuro cercano los pacientes con reordenamientos del gen de la quinasa del linfoma anaplásico (ALK), pueden recibir un tratamiento específico basado en el resultado del análisis de biomarcadores. Esto les proporcionará mayor calidad de vida y supervivencia libre de progresión que la quimioterapia convencional. Este documento de consenso nace como una iniciativa conjunta de la Sociedad Española de Anatomía Patológica (SEAP) y de la Sociedad Española de Oncología Médica (SEOM) y propone recomendaciones diagnósticas y terapéuticas para el paciente con CPNM avanzado basadas en la evidencia científica relacionada con el uso de biomarcadores. Por tanto, supone una oportunidad para mejorar la eficiencia de la actividad asistencial y la utilización de recursos, lo que sin duda redundará en un beneficio para estos pacientes. Aunque este campo está en constante evolución, en la actualidad, con los datos disponibles, este grupo de expertos recomienda que en todos los pacientes con CPNM avanzado de células no escamosas, así como en pacientes no fumadores con independencia del subtipo histológico, se determine el estado mutacional del gen EGFR en un plazo máximo de 7 días a partir del diagnóstico anatomopatológico. Los laboratorios involucrados deben participar en programas de gestión de calidad externos. Por el contrario, los reordenamientos del gen ALK solo se deben analizar en el marco de un ensayo clínico, aunque con los datos tan prometedores que se han obtenido se justificará previsiblemente en un futuro próximo su estudio rutinario en pacientes sin mutaciones de EGFR. Por último, no se considera necesaria la determinación rutinaria de otras anormalidades moleculares en la práctica clínica actual(AU)


ingles(AU)


Asunto(s)
Humanos , Masculino , Femenino , Biomarcadores , Biomarcadores Farmacológicos , Sociedades Médicas/organización & administración , Sociedades Médicas , Neoplasias Pulmonares/patología , Calidad de Vida/legislación & jurisprudencia , Genes erbB-1/fisiología , Mutación/genética , Sociedades Médicas/legislación & jurisprudencia , Sociedades Médicas/normas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...