Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Microencapsul ; 34(6): 582-591, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28868953

RESUMEN

Peptide active ingredients show great promise regarding the treatment of various health-endangering diseases. It is reported that L-lysine inhibits the proliferation of several tumour lines in vitro and in vivo. However, proteins and peptide drugs possess certain disadvantages such as in vivo instability and short biological half-life. On the grounds that drug delivery systems can overcome a wide spectrum of bioactive compounds issues, a biopolymeric blend-based microparticulated system capable of delivering ε-polylysine (PLL) was developed. PLL-loaded poly((L)Lactic acid)/poly(D,L-Lactide)-co-poly(ethylene glycol)-based microparticles (PLL-PB-MPs) were prepared and fully characterised exhibiting a narrow size distribution (1.2 ± 0.12 µm), high loading efficiency (81%) and improved thermal stability (Td from 250 °C to 291 °C). The cytotoxicity and antiproliferative effect of PLL-PB-MPs in pancreatic adenocarcinoma cell lines BxPC3 and MIA PaCa-2 were confirmed. Due to their physicochemical and biopharmaceutical properties, PB-MPs constitute a promising carrier to deliver bioactive peptides.


Asunto(s)
Antineoplásicos/administración & dosificación , Sistemas de Liberación de Medicamentos , Neoplasias Pancreáticas/tratamiento farmacológico , Polilisina/química , Línea Celular Tumoral , Humanos , Polímeros/química
2.
J Biol Chem ; 289(36): 25227-40, 2014 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-24990947

RESUMEN

The serine/threonine protein kinase Akt promotes cell survival, growth, and proliferation through phosphorylation of different downstream substrates. A key effector of Akt is the mammalian target of rapamycin (mTOR). Akt is known to stimulate mTORC1 activity through phosphorylation of tuberous sclerosis complex 2 (TSC2) and PRAS40, both negative regulators of mTOR activity. We previously reported that IκB kinase α (IKKα), a component of the kinase complex that leads to NF-κB activation, plays an important role in promoting mTORC1 activity downstream of activated Akt. Here, we demonstrate IKKα-dependent regulation of mTORC1 using multiple PTEN null cancer cell lines and an animal model with deletion of IKKα. Importantly, IKKα is shown to phosphorylate mTOR at serine 1415 in a manner dependent on Akt to promote mTORC1 activity. These results demonstrate that IKKα is an effector of Akt in promoting mTORC1 activity.


Asunto(s)
Quinasa I-kappa B/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Activación Enzimática , Femenino , Células HEK293 , Células HeLa , Humanos , Quinasa I-kappa B/genética , Immunoblotting , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones Noqueados , Complejos Multiproteicos/genética , Mutación , Fosforilación , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/genética , Interferencia de ARN , Proteína Reguladora Asociada a mTOR , Serina/genética , Serina/metabolismo , Serina-Treonina Quinasas TOR/genética , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/metabolismo
3.
EMBO J ; 30(19): 4059-70, 2011 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-21873977

RESUMEN

The transcription factor RUNX1 is essential to establish the haematopoietic gene expression programme; however, the mechanism of how it activates transcription of haematopoietic stem cell (HSC) genes is still elusive. Here, we obtained novel insights into RUNX1 function by studying regulation of the human CD34 gene, which is expressed in HSCs. Using transgenic mice carrying human CD34 PAC constructs, we identified a novel downstream regulatory element (DRE), which is bound by RUNX1 and is necessary for human CD34 expression in long-term (LT)-HSCs. Conditional deletion of Runx1 in mice harbouring human CD34 promoter-DRE constructs abrogates human CD34 expression. We demonstrate by chromosome conformation capture assays in LT-HSCs that the DRE physically interacts with the human CD34 promoter. Targeted mutagenesis of RUNX binding sites leads to perturbation of this interaction and decreased human CD34 expression in LT-HSCs. Overall, our in vivo data provide novel evidence about the role of RUNX1 in mediating interactions between distal and proximal elements of the HSC gene CD34.


Asunto(s)
Antígenos CD34/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Animales , Trasplante de Médula Ósea , Cromatina/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Sangre Fetal/citología , Genotipo , Células HL-60 , Humanos , Ratones , Ratones Transgénicos , Modelos Biológicos , Secuencias Reguladoras de Ácidos Nucleicos/genética
4.
Cancer Res Commun ; 4(3): 919-937, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38546390

RESUMEN

Lung cancer is the leading cause of cancer deaths. Lethal pulmonary adenocarcinomas (ADC) present with frequent mutations in the EGFR. Genetically engineered murine models of lung cancer expedited comprehension of the molecular mechanisms driving tumorigenesis and drug response. Here, we systematically analyzed the evolution of tumor heterogeneity in the context of dynamic interactions occurring with the intermingled tumor microenvironment (TME) by high-resolution transcriptomics. Our effort identified vulnerable tumor-specific epithelial cells, as well as their cross-talk with niche components (endothelial cells, fibroblasts, and tumor-infiltrating immune cells), whose symbiotic interface shapes tumor aggressiveness and is almost completely abolished by treatment with Unesbulin, a tubulin binding agent that reduces B cell-specific Moloney murine leukemia virus integration site 1 (BMI-1) activity. Simultaneous magnetic resonance imaging (MRI) analysis demonstrated decreased tumor growth, setting the stage for future investigations into the potential of novel therapeutic strategies for EGFR-mutant ADCs. SIGNIFICANCE: Targeting the TME is an attractive strategy for treatment of solid tumors. Here we revealed how EGFR-mutant landscapes are affected at the single-cell resolution level during Unesbulin treatment. This novel drug, by targeting cancer cells and their interactions with crucial TME components, could be envisioned for future therapeutic advancements.


Asunto(s)
Antineoplásicos , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Animales , Ratones , Células Endoteliales , Microambiente Tumoral/genética , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Comunicación Celular , Receptores ErbB/genética
5.
J Exp Med ; 203(2): 371-81, 2006 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-16446383

RESUMEN

Mutations constitutively activating FLT3 kinase are detected in approximately 30% of acute myelogenous leukemia (AML) patients and affect downstream pathways such as extracellular signal-regulated kinase (ERK)1/2. We found that activation of FLT3 in human AML inhibits CCAAT/enhancer binding protein alpha (C/EBPalpha) function by ERK1/2-mediated phosphorylation, which may explain the differentiation block of leukemic blasts. In MV4;11 cells, pharmacological inhibition of either FLT3 or MEK1 leads to granulocytic differentiation. Differentiation of MV4;11 cells was also observed when C/EBPalpha mutated at serine 21 to alanine (S21A) was stably expressed. In contrast, there was no effect when serine 21 was mutated to aspartate (S21D), which mimics phosphorylation of C/EBPalpha. Thus, our results suggest that therapies targeting the MEK/ERK cascade or development of protein therapies based on transduction of constitutively active C/EBPalpha may prove effective in treatment of FLT3 mutant leukemias resistant to the FLT3 inhibitor therapies.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Mutación Puntual , Tirosina Quinasa 3 Similar a fms/genética , Anciano , Proteína alfa Potenciadora de Unión a CCAAT/fisiología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Activación Enzimática/genética , Femenino , Granulocitos/patología , Humanos , Células K562 , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 1/metabolismo , Masculino , Células Mieloides/patología , Fosforilación , Piperazinas/farmacología , Quinazolinas/farmacología , Serina/metabolismo , Células Tumorales Cultivadas , Células U937 , Tirosina Quinasa 3 Similar a fms/antagonistas & inhibidores , Tirosina Quinasa 3 Similar a fms/metabolismo
6.
Commun Biol ; 4(1): 370, 2021 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-33854168

RESUMEN

Lung cancer is the leading cause of cancer deaths. Tumor heterogeneity, which hampers development of targeted therapies, was herein deconvoluted via single cell RNA sequencing in aggressive human adenocarcinomas (carrying Kras-mutations) and comparable murine model. We identified a tumor-specific, mutant-KRAS-associated subpopulation which is conserved in both human and murine lung cancer. We previously reported a key role for the oncogene BMI-1 in adenocarcinomas. We therefore investigated the effects of in vivo PTC596 treatment, which affects BMI-1 activity, in our murine model. Post-treatment, MRI analysis showed decreased tumor size, while single cell transcriptomics concomitantly detected near complete ablation of the mutant-KRAS-associated subpopulation, signifying the presence of a pharmacologically targetable, tumor-associated subpopulation. Our findings therefore hold promise for the development of a targeted therapy for KRAS-mutant adenocarcinomas.


Asunto(s)
Bencimidazoles/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Células Epiteliales/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Mutación , Proteínas Proto-Oncogénicas p21(ras)/genética , Pirazinas/farmacología , Células A549 , Animales , Antineoplásicos , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Terapia Molecular Dirigida , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , RNA-Seq , Análisis de la Célula Individual , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Mol Cell Biol ; 26(3): 1109-23, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16428462

RESUMEN

The leucine zipper family transcription factor CCAAT enhancer binding protein alpha (C/EBPalpha) inhibits proliferation and promotes differentiation in various cell types. In this study, we show, using a lung-specific conditional mouse model of C/EBPalpha deletion, that loss of C/EBPalpha in the respiratory epithelium leads to respiratory failure at birth due to an arrest in the type II alveolar cell differentiation program. This differentiation arrest results in the lack of type I alveolar cells and differentiated surfactant-secreting type II alveolar cells. In addition to showing a block in type II cell differentiation, the neonatal lungs display increased numbers of proliferating cells and decreased numbers of apoptotic cells, leading to epithelial expansion and loss of airspace. Consistent with the phenotype observed, genes associated with alveolar maturation, survival, and proliferation were differentially expressed. Taken together, these results identify C/EBPalpha as a master regulator of airway epithelial maturation and suggest that the loss of C/EBPalpha could also be an important event in the multistep process of lung tumorigenesis. Furthermore, this study indicates that exploring the C/EBPalpha pathway might have therapeutic benefits for patients with respiratory distress syndromes.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/deficiencia , Pulmón/crecimiento & desarrollo , Pulmón/patología , Alveolos Pulmonares/patología , Insuficiencia Respiratoria/genética , Insuficiencia Respiratoria/patología , Animales , Apoptosis , Proteína alfa Potenciadora de Unión a CCAAT/genética , Diferenciación Celular , Proliferación Celular , Epitelio/metabolismo , Epitelio/patología , Eliminación de Gen , Perfilación de la Expresión Génica , Pulmón/metabolismo , Masculino , Ratones , Ratones Mutantes , Alveolos Pulmonares/citología , Alveolos Pulmonares/crecimiento & desarrollo , Eliminación de Secuencia
8.
Mater Sci Eng C Mater Biol Appl ; 81: 327-333, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-28887980

RESUMEN

Miltefosine presents antineoplastic activity but high hemolytic potential. Its use in cancer has been limited to treating cutaneous metastasis of breast cancer. To decrease hemolytic potential, we developed a formulation of miltefosine-loaded polymeric micelles (PM) of the copolymer Pluronic-F127. A central composite design was applied and the analysis of variance showed that the optimum level of hydrodynamic diameter and polydispersity index predicted by the model and experimentally confirmed were 29nm and 0.105, respectively. Thermal analyses confirmed that miltefosine was molecularly dispersed within PM. Pluronic-F127 PM with miltefosine 80µM presented a significant reduction of hemolytic effect (80%, p<0.05) in comparison to free drug. In vitro assays against HeLa carcinoma cells demonstrated similar cytotoxicity to free miltefosine and PM. Our results suggest that, by lowering hemolytic potential, miltefosine-loaded Pluronic-F127 PM a promising alternative to broaden this drug use in cancer therapy, as well as of other alkylphosphocholines.


Asunto(s)
Fosforilcolina/análogos & derivados , Línea Celular Tumoral , Humanos , Micelas , Fosforilcolina/química , Poloxámero , Polímeros
9.
Cancer Res ; 64(12): 4137-47, 2004 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15205324

RESUMEN

We showed previously that CCAAT/enhancer binding protein alpha (C/EBP alpha), a tissue-specific transcription factor, is a candidate tumor suppressor in lung cancer. In the present study, we have performed a transcriptional profiling study of C/EBP alpha target genes using an inducible cell line system. This study led to the identification of hepatocyte nuclear factor 3beta (HNF3 beta), a transcription factor known to play a role in airway differentiation, as a downstream target of C/EBP alpha. We found down-regulation of HNF3 beta expression in a large proportion of lung cancer cell lines examined and identified two novel mutants of HNF3 beta, as well as hypermethylation of the HNF3 beta promoter. We also developed a tetracycline-inducible cell line model to study the cellular consequences of HNF3 beta expression. Conditional expression of HNF3 beta led to significant growth reduction, proliferation arrest, apoptosis, and loss of clonogenic ability, suggesting additionally that HNF3 beta is a novel tumor suppressor in lung cancer. This is the first study to show genetic abnormalities of lung-specific differentiation pathways in the development of lung cancer.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas de Unión al ADN/genética , Neoplasias Pulmonares/genética , Proteínas Nucleares/genética , Factores de Transcripción , Apoptosis/genética , División Celular/genética , Metilación de ADN , Proteínas de Unión al ADN/biosíntesis , Regulación hacia Abajo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Factor Nuclear 3-beta del Hepatocito , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Mutación , Proteínas Nucleares/biosíntesis , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , Transcripción Genética
10.
Sci Transl Med ; 8(350): 350ra104, 2016 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-27488898

RESUMEN

Lung cancer is the most common cause of cancer deaths. The expression of the transcription factor C/EBPα (CCAAT/enhancer binding protein α) is frequently lost in non-small cell lung cancer, but the mechanisms by which C/EBPα suppresses tumor formation are not fully understood. In addition, no pharmacological therapy is available to specifically target C/EBPα expression. We discovered a subset of pulmonary adenocarcinoma patients in whom negative/low C/EBPα expression and positive expression of the oncogenic protein BMI1 (B lymphoma Mo-MLV insertion region 1 homolog) have prognostic value. We also generated a lung-specific mouse model of C/EBPα deletion that develops lung adenocarcinomas, which are prevented by Bmi1 haploinsufficiency. BMI1 activity is required for both tumor initiation and maintenance in the C/EBPα-null background, and pharmacological inhibition of BMI1 exhibits antitumor effects in both murine and human adenocarcinoma lines. Overall, we show that C/EBPα is a tumor suppressor in lung cancer and that BMI1 is required for the oncogenic process downstream of C/EBPα loss. Therefore, anti-BMI1 pharmacological inhibition may offer a therapeutic benefit for lung cancer patients with low expression of C/EBPα and high BMI1.


Asunto(s)
Adenocarcinoma/patología , Adenocarcinoma/terapia , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma del Pulmón , Animales , Proteína alfa Potenciadora de Unión a CCAAT/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Noqueados , Mutación/genética , Complejo Represivo Polycomb 1/genética , Proteínas Proto-Oncogénicas/genética
11.
Methods Mol Biol ; 1280: 447-58, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25736766

RESUMEN

RNA interference-based gene silencing has become a widely used technology to evaluate how inhibition of expression of individual proteins affects biological readout. Through the use of this technology, a lot has been learned about how different proteins function in a wide variety of biological contexts, including cancer. In this context, RNA interference-mediated gene silencing has contributed to further our understanding of how different proteins in the NF-κB signaling pathway (including the NF-κB members themselves) contribute to cancer. Here, we describe two RNA interference-based protocols in lung cancer cells targeting upstream activators of NF-κB transcription factor: the catalytic subunits of the IKK complex. The first protocol is designed to evaluate the impact of IKKα or IKKß inhibition on NF-κB transcriptional activity, whereas the second protocol is designed to evaluate how siRNA-mediated IKK inhibition affects lung cancer cell proliferation.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Quinasa I-kappa B/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , FN-kappa B/metabolismo , Interferencia de ARN , Transducción de Señal , Línea Celular Tumoral , Proliferación Celular , Expresión Génica , Técnicas de Silenciamiento del Gen , Genes Reporteros , Humanos , Quinasa I-kappa B/genética , Transfección
12.
Genes Cancer ; 5(1-2): 41-55, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24955217

RESUMEN

Activating mutations in KRAS are prevalent in cancer, but therapies targeted to oncogenic RAS have been ineffective to date. These results argue that targeting downstream effectors of RAS will be an alternative route for blocking RAS-driven oncogenic pathways. We and others have shown that oncogenic RAS activates the NF-κB transcription factor pathway and that KRAS-induced lung tumorigenesis is suppressed by expression of a degradation-resistant form of the IκBα inhibitor or by genetic deletion of IKKß or the RELA/p65 subunit of NF-κB. Here, genetic and pharmacological approaches were utilized to inactivate IKK in human primary lung epithelial cells transformed by KRAS, as well as KRAS mutant lung cancer cell lines. Administration of the highly specific IKKß inhibitor Compound A (CmpdA) led to NF-κB inhibition in different KRAS mutant lung cells and siRNA-mediated knockdown of IKKα or IKKß reduced activity of the NF-κB canonical pathway. Next, we determined that both IKKα and IKKß contribute to oncogenic properties of KRAS mutant lung cells, particularly when p53 activity is disrupted. Based on these results, CmpdA was tested for potential therapeutic intervention in the Kras-induced lung cancer mouse model (LSL-Kras (G12D)) combined with loss of p53 (LSL-Kras (G12D)/p53 (fl/fl)). CmpdA treatment was well tolerated and mice treated with this IKKß inhibitor presented smaller and lower grade tumors than mice treated with placebo. Additionally, IKKß inhibition reduced inflammation and angiogenesis. These results support the concept of targeting IKK as a therapeutic approach for oncogenic RAS-driven tumors with altered p53 activity.

13.
Lung Cancer ; 77(1): 31-7, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22341411

RESUMEN

PURPOSE: We sought to determine the mechanisms of downregulation of the airway transcription factor Foxa2 in lung cancer and the expression status of Foxa2 in non-small-cell lung cancer (NSCLC). METHODS: A series of 25 lung cancer cell lines were evaluated for Foxa2 protein expression, FOXA2 mRNA levels, FOXA2 mutations, FOXA2 copy number changes and for evidence of FOXA2 promoter hypermethylation. In addition, 32 NSCLCs were sequenced for FOXA2 mutations and 173 primary NSCLC tumors evaluated for Foxa2 expression using an immunohistochemical assay. RESULTS: Out of the 25 cell lines, 13 (52%) had undetectable FOXA2 mRNA. The expression of FOXA2 mRNA and Foxa2 protein were congruent in 19/22 cells (p = 0.001). FOXA2 mutations were not identified in primary NSCLCs and were infrequent in cell lines. Focal or broad chromosomal deletions involving FOXA2 were not present. The promoter region of FOXA2 had evidence of hypermethylation, with an inverse correlation between FOXA2 mRNA expression and presence of CpG dinucleotide methylation (p < 0.0001). In primary NSCLC tumor specimens, there was a high frequency of either absence (42/173, 24.2%) or no/low expression (96/173, 55.4%) of Foxa2. In 130 patients with stage I NSCLC there was a trend towards decreased survival in tumors with no/low expression of Foxa2 (HR of 1.6, 95%CI 0.9-3.1; p = 0.122). CONCLUSIONS: Loss of expression of Foxa2 is frequent in lung cancer cell lines and NSCLCs. The main mechanism of downregulation of Foxa2 is epigenetic silencing through promoter hypermethylation. Further elucidation of the involvement of Foxa2 and other airway transcription factors in the pathogenesis of lung cancer may identify novel therapeutic targets.


Asunto(s)
Adenocarcinoma/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Regulación hacia Abajo , Epigénesis Genética , Factor Nuclear 3-beta del Hepatocito/genética , Neoplasias Pulmonares/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/mortalidad , Adenocarcinoma/patología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Metilación de ADN , Análisis Mutacional de ADN , Femenino , Dosificación de Gen , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 3-beta del Hepatocito/metabolismo , Humanos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Regiones Promotoras Genéticas
14.
Cancer Res ; 70(9): 3537-46, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20406971

RESUMEN

K-Ras-induced lung cancer is a very common disease, for which there are currently no effective therapies. Because therapy directly targeting the activity of oncogenic Ras has been unsuccessful, a different approach for novel therapy design is to identify critical Ras downstream oncogenic targets. Given that oncogenic Ras proteins activate the transcription factor NF-kappaB, and the importance of NF-kappaB in oncogenesis, we hypothesized that NF-kappaB would be an important K-Ras target in lung cancer. To address this hypothesis, we generated a NF-kappaB-EGFP reporter mouse model of K-Ras-induced lung cancer and determined that K-Ras activates NF-kappaB in lung tumors in situ. Furthermore, a mouse model was generated where activation of oncogenic K-Ras in lung cells was coupled with inactivation of the NF-kappaB subunit p65/RelA. In this model, deletion of p65/RelA reduces the number of K-Ras-induced lung tumors both in the presence and in the absence of the tumor suppressor p53. Lung tumors with loss of p65/RelA have higher numbers of apoptotic cells, reduced spread, and lower grade. Using lung cell lines expressing oncogenic K-Ras, we show that NF-kappaB is activated in these cells in a K-Ras-dependent manner and that NF-kappaB activation by K-Ras requires inhibitor of kappaB kinase beta (IKKbeta) kinase activity. Taken together, these results show the importance of the NF-kappaB subunit p65/RelA in K-Ras-induced lung transformation and identify IKKbeta as a potential therapeutic target for K-Ras-induced lung cancer.


Asunto(s)
Transformación Celular Neoplásica/genética , Genes ras , Neoplasias Pulmonares/genética , Factor de Transcripción ReIA/genética , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Genes p53 , Humanos , Quinasa I-kappa B/antagonistas & inhibidores , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , FN-kappa B/genética , FN-kappa B/metabolismo , Factor de Transcripción ReIA/deficiencia , Transfección , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética
15.
Haematologica ; 87(3): 246-9, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11869935

RESUMEN

BACKGROUND AND OBJECTIVES: Analysis of DNA polymorphic sites is a powerful tool for detection of gene flow in human evolutionary studies and to trace genetic background associated with abnormal genes. The beta-globin locus contains more than 20 single-base restriction fragment length polymorphism (RFLP) sites spanning over 80 kb on chromosome 11. Far downstream of the expressed genes, there is a hypersensitive site (HS). The function of the 3'-HS remains unknown. As an approach to the understanding of the 3'-HS region in sickle cell anemia we searched for sequence polymorphism in the AT-rich region, using a non-radioactive polymerase chain reaction (PCR)-single strand conformational polymorphism (SSCP) technique. DESIGN AND METHODS: A 460 bp fragment located at the 3' of the b globin gene was amplified from patients (with sickle cell anemia and HbSC disease), and from AS individuals. Standard RFLP-haplotyping was performed and compared with the PCR-SSCP screening strategy. RESULTS: Two distinct band patterns were revealed by SSCP testing, each one in strict linkage disequilibrium with either Benin or Bantu haplotypes. Direct sequencing of the amplified segment revealed a TAA insertion in the AT-rich region, in all 121 beta(S) Benin chromosomes tested, but not in other beta(S) haplotypes from the total of 380 beta(S) chromosomes typed. INTERPRETATION AND CONCLUSIONS: SSCP analysis could easily distinguish sequence variations in the 3'AT-rich region of the beta-globin cluster, and a TAA insertion in this region seems to be specific for the Benin-beta(S) chromosome.


Asunto(s)
Anemia de Células Falciformes/genética , Globinas/genética , Mutagénesis Insercional/genética , Secuencia Rica en At/genética , Secuencia de Bases , Sitios de Unión , Desoxirribonucleasa I/metabolismo , Variación Genética , Humanos , Polimorfismo Genético , Polimorfismo Conformacional Retorcido-Simple
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA