Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Gut ; 65(7): 1151-64, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26033966

RESUMEN

OBJECTIVE: Colorectal cancer remains the fourth most common cause of cancer-related mortality worldwide. Here we investigate the role of nuclear factor-κB (NF-κB) co-factor B-cell CLL/lymphoma 3 (BCL-3) in promoting colorectal tumour cell survival. DESIGN: Immunohistochemistry was carried out on 47 tumour samples and normal tissue from resection margins. The role of BCL-3/NF-κB complexes on cell growth was studied in vivo and in vitro using an siRNA approach and exogenous BCL-3 expression in colorectal adenoma and carcinoma cells. The question whether BCL-3 activated the AKT/protein kinase B (PKB) pathway in colorectal tumour cells was addressed by western blotting and confocal microscopy, and the ability of 5-aminosalicylic acid (5-ASA) to suppress BCL-3 expression was also investigated. RESULTS: We report increased BCL-3 expression in human colorectal cancers and demonstrate that BCL-3 expression promotes tumour cell survival in vitro and tumour growth in mouse xenografts in vivo, dependent on interaction with NF-κB p50 or p52 homodimers. We show that BCL-3 promotes cell survival under conditions relevant to the tumour microenvironment, protecting both colorectal adenoma and carcinoma cells from apoptosis via activation of the AKT survival pathway: AKT activation is mediated via both PI3K and mammalian target of rapamycin (mTOR) pathways, leading to phosphorylation of downstream targets GSK-3ß and FoxO1/3a. Treatment with 5-ASA suppressed BCL-3 expression in colorectal cancer cells. CONCLUSIONS: Our study helps to unravel the mechanism by which BCL-3 is linked to poor prognosis in colorectal cancer; we suggest that targeting BCL-3 activity represents an exciting therapeutic opportunity potentially increasing the sensitivity of tumour cells to conventional therapy.


Asunto(s)
Neoplasias Colorrectales/química , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas/análisis , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal , Factores de Transcripción/análisis , Factores de Transcripción/metabolismo , Animales , Antiinflamatorios no Esteroideos/farmacología , Apoptosis , Proteínas del Linfoma 3 de Células B , Proliferación Celular , Supervivencia Celular/efectos de los fármacos , Colon/química , Neoplasias Colorrectales/patología , Células HCT116 , Humanos , Mesalamina/farmacología , Ratones , Ratones Desnudos , FN-kappa B/análisis , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , ARN Interferente Pequeño/farmacología , Recto/química , Serina-Treonina Quinasas TOR/metabolismo , Factores de Transcripción/genética , Carga Tumoral
2.
Nat Cell Biol ; 9(2): 210-7, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17220880

RESUMEN

Aberrant activation of beta-catenin promotes cell proliferation and initiates colorectal tumorigenesis. However, the expansion of tumours and the inadequacy of their local vasculature results in areas of hypoxia where cell growth is typically constrained. Here, we report a novel diversion in beta-catenin signalling triggered by hypoxia. We show that hypoxia inhibits beta-catenin-T-cell factor-4 (TCF-4) complex formation and transcriptional activity, resulting in a G1 arrest that involves the c-Myc-p21 axis. Additionally, we find that hypoxia inducible factor-1alpha (HIF-1alpha) competes with TCF-4 for direct binding to beta-catenin. DNA-protein interaction studies reveal that beta-catenin-HIF-1alpha interaction occurs at the promoter region of HIF-1 target genes. Furthermore, rigorous analyses indicate that beta-catenin can enhance HIF-1-mediated transcription, thereby promoting cell survival and adaptation to hypoxia. These findings demonstrate a dynamic role for beta-catenin in colorectal tumorigenesis, where a functional switch is instigated to meet the ever-changing needs of the tumour. This study highlights the importance of the microenvironment in transcriptional regulation.


Asunto(s)
Neoplasias del Colon/metabolismo , Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/metabolismo , beta Catenina/metabolismo , Adaptación Fisiológica , Línea Celular Tumoral , Transformación Celular Neoplásica/metabolismo , Humanos , Transducción de Señal
3.
Carcinogenesis ; 32(11): 1741-7, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21926111

RESUMEN

Due to poor tumour-associated vasculature, tumour cells are subjected to a fluctuating microenvironment with periods of limited oxygen and glucose availability. Adaptive mechanisms to adverse microenvironments are important for tumour cell survival. The cyclooxygenase (COX)-2/prostaglandin E(2) (PGE(2)) pathway has key roles in colorectal tumorigenesis. Although glucose is important as an energy source and in maintaining endoplasmic reticulum homeostasis, relatively little is known regarding how tumour cells adapt to the microenvironmental stress of reduced glucose availability. Here, we report the novel findings that glucose deprivation of colorectal tumour cells not only increases COX-2 expression but also decreases 15-hydroxyprostaglandin dehydrogenase (15-PGDH) expression, resulting in increased extracellular PGE(2). Furthermore, we have shown that PGE(2) promotes tumour cell survival during glucose deprivation. Glucose deprivation enhances phosphoinositide 3-kinase/Akt activity, which has a role in both the up-regulation of COX-2 and down-regulation of 15-PGDH. Glucose deprivation also activates the unfolded protein response (UPR) resulting in elevated C/EBP-homologous protein (CHOP) expression. Interestingly, inhibiting CHOP expression by small interfering RNA during glucose deprivation attenuates the reduction in 15-PGDH expression. This is the first report linking activation of the UPR with a reduction in expression of tumour-suppressive 15-PGDH and may have implications for tumour cells' ability to survive exposure to therapeutic agents that activate the UPR. Our data suggest that diverse microenvironmental stresses converge to regulate PGE(2) as a common and crucial mediator of cell survival during adaptation to the tumour microenvironment and may lead to novel chemopreventive and therapeutic strategies.


Asunto(s)
Proliferación Celular , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Glucosa/deficiencia , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Western Blotting , Neoplasias del Colon/genética , Ciclooxigenasa 2/genética , Elafina/genética , Elafina/metabolismo , Humanos , Hidroxiprostaglandina Deshidrogenasas/genética , Hipoxia , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo , Microambiente Tumoral , Respuesta de Proteína Desplegada
4.
Proteomics ; 10(2): 194-202, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19899082

RESUMEN

This is the first differential expression proteomics study on a human syngeneic cellular in vitro progression model of the colorectal adenoma-to-carcinoma sequence, the anchorage-dependent non-tumorigenic adenoma derived cell line AA/C1 and the derived anchorage-independent and tumorigenic carcinoma cell line AA/C1/SB10C. The study is based on quantitative 2-DE and is complemented by Western blot validation. Excluding redundancies due to proteolysis and post-translational modified isoforms of over 2000 protein spots, 13 proteins were revealed as regulated with statistical variance being within the 95th confidence level and were identified by peptide mass fingerprinting in MALDI MS. Progression-associated proteins belong to the functional complexes of anaerobic glycolysis/gluconeogenesis, steroid biosynthesis, prostaglandin biosynthesis, the regulation and maintenance of the cytoskeleton, protein biosynthesis and degradation, the regulation of apoptosis or other functions. Partial but significant overlap was revealed with previous proteomics and transcriptomics studies in colorectal carcinoma. Among upregulated proteins we identified 3-HMG-CoA synthase, protein phosphatase 1, prostaglandin E synthase 2, villin 1, annexin A1, triosephosphate isomerase, phosphoserine aminotransferase 1, fumarylacetoacetate hydrolase and pyrroline-5-carboxylate reductase 1 (PYCR1), while glucose-regulated protein 78, cathepsin D, lamin A/C and quinolate phosphoribosyltransferase were downregulated.


Asunto(s)
Adenoma/química , Adenoma/patología , Neoplasias Colorrectales/química , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Proteoma/análisis , Línea Celular Tumoral , Regulación hacia Abajo , Electroforesis en Gel Bidimensional , Humanos , Modelos Biológicos , Proteómica , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Regulación hacia Arriba
5.
Int J Oncol ; 56(5): 1304-1313, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32319612

RESUMEN

First discovered as an oncogene in leukaemia, recent reports highlight an emerging role for the proto­oncogene BCL­3 in solid tumours. Importantly, BCL­3 expression is upregulated in >30% of colorectal cancer cases and is reported to be associated with a poor prognosis. However, the mechanism by which BCL­3 regulates tumorigenesis in the large intestine is yet to be fully elucidated. In the present study, it was shown for the first time that knocking down BCL­3 expression suppressed cyclooxygenase­2 (COX­2)/prostaglandin E2 (PGE2) signalling in colorectal cancer cells, a pathway known to drive several of the hallmarks of cancer. RNAi­mediated suppression of BCL­3 expression decreased COX­2 expression in colorectal cancer cells both at the mRNA and protein level. This reduction in COX­2 expression resulted in a significant and functional reduction (30­50%) in the quantity of pro­tumorigenic PGE2 produced by the cancer cells, as shown by enzyme linked immunoassays and medium exchange experiments. In addition, inhibition of BCL­3 expression also significantly suppressed cytokine­induced (TNF­α or IL­1ß) COX­2 expression. Taken together, the results of the present study identified a novel role for BCL­3 in colorectal cancer and suggested that expression of BCL­3 may be a key determinant in the COX­2­meditated response to inflammatory cytokines in colorectal tumour cells. These results suggest that targeting BCL­3 to suppress PGE2 synthesis may represent an alternative or complementary approach to using non­steroidal anti­inflammatory drugs [(NSAIDs), which inhibit cyclooxygenase activity and suppress the conversion of arachidonic acid to prostaglandin], for prevention and/or recurrence in PGE2­driven tumorigenesis.


Asunto(s)
Proteínas del Linfoma 3 de Células B/metabolismo , Neoplasias Colorrectales/metabolismo , Ciclooxigenasa 2/metabolismo , Regulación hacia Arriba , Proteínas del Linfoma 3 de Células B/genética , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Ciclooxigenasa 2/genética , Dinoprostona/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Interleucina-1beta/farmacología , Transducción de Señal , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba/efectos de los fármacos
6.
Carcinogenesis ; 30(3): 377-86, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19136477

RESUMEN

It is widely accepted that alterations to cyclooxygenase-2 (COX-2) expression and the abundance of its enzymatic product prostaglandin E(2) (PGE(2)) have key roles in influencing the development of colorectal cancer. Deregulation of the COX-2/PGE(2) pathway appears to affect colorectal tumorigenesis via a number of distinct mechanisms: promoting tumour maintenance and progression, encouraging metastatic spread, and perhaps even participating in tumour initiation. Here, we review the role of COX-2/PGE(2) signalling in colorectal tumorigenesis and highlight its ability to influence the hallmarks of cancer--attributes defined by Hanahan and Weinberg as being requisite for tumorigenesis. In addition, we consider components of the COX-prostaglandin pathway emerging as important regulators of tumorigenesis; namely, the prostanoid (EP) receptors, 15-hydroxyprostaglandin dehydrogenase and the prostaglandin transporter. Finally, based on recent findings, we propose a model for the cellular adaptation to the hypoxic tumour microenvironment that encompasses the interplay between COX-2, hypoxia-inducible factor 1 and dynamic switches in beta-catenin function that fine-tune signalling networks to meet the ever-changing demands of a tumour.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Ciclooxigenasa 2/fisiología , Dinoprostona/fisiología , Animales , Hipoxia de la Célula , Movimiento Celular/fisiología , Neoplasias Colorrectales/irrigación sanguínea , Neoplasias Colorrectales/patología , Humanos , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neovascularización Patológica/metabolismo , Transportadores de Anión Orgánico/metabolismo , Receptores de Prostaglandina E/metabolismo , Transducción de Señal/fisiología , beta Catenina/metabolismo
7.
Carcinogenesis ; 30(10): 1796-804, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19638428

RESUMEN

Evidence points towards a pivotal role for cyclooxygenase (COX)-2 in promoting colorectal tumorigenesis through increasing prostaglandin E(2) (PGE(2)) levels. PGE(2) signalling is closely associated with the survival, proliferation and invasion of colorectal cancer cells. Recently, a reduction in PGE(2) inactivation, a process mediated by the nicotinamide adenine dinucleotide (NAD+)-dependent 15-hydroxyprostaglandin dehydrogenase (15-PGDH), has also been shown to promote tumoral PGE(2) accumulation. The hepatocyte growth factor (HGF) receptor, Met, is frequently over-expressed in colorectal tumours and promotes cancer growth, metastasis and resistance to therapy, although the mechanisms for this have not been fully elucidated. Here, we report that HGF/Met signalling can promote PGE(2) biogenesis in colorectal cancer cells via COX-2 up-regulation and 15-PGDH down-regulation at the protein and messenger RNA level. Pharmacological inhibition of MEK and PI3K suggested that both extracellular signal-regulated kinase (ERK) and AKT signalling are required for COX-2 protein up-regulation and 15-PGDH down-regulation downstream of Met. Notably, inhibition of Met with the small molecule inhibitor SU11274 reduced COX-2 expression and increased 15-PGDH expression in high Met-expressing cells. We also show that hypoxia potentiated HGF-driven COX-2 expression and enhanced PGE(2) release. Furthermore, inhibition of COX-2 impeded the growth-promoting effects of HGF, suggesting that the COX-2/PGE(2) pathway is an important mediator of HGF/Met signalling. These data reveal a critical role for HGF/Met signalling in promoting PGE(2) biogenesis in colorectal cancer cells. Targeting the crosstalk between these two important pathways may be useful for therapeutic treatment of colorectal cancer.


Asunto(s)
Adenoma/genética , Neoplasias Colorrectales/genética , Ciclooxigenasa 2/genética , Hidroxiprostaglandina Deshidrogenasas/genética , Proteínas Proto-Oncogénicas/fisiología , Receptores de Factores de Crecimiento/fisiología , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/terapia , Ciclooxigenasa 2/metabolismo , Dinoprostona/biosíntesis , Regulación hacia Abajo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Desnudos , Proteínas Proto-Oncogénicas c-met , Transducción de Señal , Regulación hacia Arriba
8.
Glycobiology ; 19(4): 382-98, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19122213

RESUMEN

Our studies provide direct evidence that O-glycosylation pathways play a role in the regulation of cell growth through apoptosis and proliferation pathways. A series of small molecular weight analogs of the GalNAc-alpha-1-O-serine/threonine structure based on 1-benzyl-2-acetamido-2-deoxy-alpha-O-d-galactopyranoside have been synthesized and tested in the human colorectal cancer cell lines PC/AA/C1/SB10C and HCA7/C29. Three inhibitors, 1-benzyl-2-acetamido-2-deoxy-alpha-O-D-galactopyranoside, and the corresponding 2-azido- and C-glycoside analogs were screened in these colorectal cancer cell lines at 0.5 mM and showed induction of apoptosis and downregulation of proliferation. Treatment of both cell lines with inhibitors led to changes in glycosylation detected with peanut lectin. The inhibition of glycosyltransferase activity in cell homogenates from human colorectal mucosal cells and cultured cell lines could be shown. The competitive action of the inhibitors resulted in the intracellular formation of 28 aryl-glycan products which were identified by MALDI and electrospray mass spectroscopy. The structures showed a differential pattern for each of the inhibitors in both cell lines. Gene array analysis of the glycogenes illustrated a pattern of glycosyltransferases that matched the glycan structures found in glycoproteins and aryl-glycans formed in the PC/AA/C1/SB10C cells; however, there was no action of the three inhibitors on glycogene transcript levels. The inhibitors act at both intermediary metabolic and genomic levels, resulting in altered protein glycosylation and aryl-glycan formation. These events may play a part in growth arrest.


Asunto(s)
Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Galactosa/análogos & derivados , Glicoproteínas/metabolismo , Glicosiltransferasas/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Células CACO-2 , Neoplasias Colorrectales/enzimología , Ensayos de Selección de Medicamentos Antitumorales/métodos , Inhibidores Enzimáticos/química , Galactosa/química , Galactosa/farmacología , Glicosilación/efectos de los fármacos , Humanos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos
9.
Clin Chem ; 55(8): 1559-63, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19541867

RESUMEN

BACKGROUND: Aberrantly methylated genes are promising biomarkers for the detection of colon adenomas and colorectal cancers (CRCs). The optimal assay type and specific methylated genes for these assays remain to be determined. METHODS: We used genomewide microarray-based assays to identify methylated genes as candidate biomarkers for colon neoplasms. The frequency of aberrant methylation of these genes in primary tumors was assessed with methylation-specific PCR (MSP). The limits of detection and specificities for different types of PCR-based assays were then assessed with the most promising genes identified in this screen. Finally, we assessed the best-performing MSP assay as an early-detection marker using fecal DNA samples. RESULTS: ITGA4 [integrin, alpha 4 (antigen CD49D, alpha 4 subunit of VLA-4 receptor)] was identified as a novel gene frequently methylated in CRC. Methylated ITGA4 is present in 75% of colon adenomas (n = 36) and 92% of colon adenocarcinomas (n = 75). Comparison of end point MSP, end point MSP with clamped primers, and quantitative fluorescent MSP (qMSP) approaches revealed that both types of end point MSP assays could routinely detect as little as 70 pg DNA, whereas the qMSP assay could routinely detect as little as 7 pg. A fecal DNA qMSP assay for methylated ITGA4 can detect 69% of individuals with colon adenomas (n = 13) with a diagnostic specificity of 79% (n = 28). CONCLUSIONS: Methylated ITGA4 is a promising marker gene for the early detection of colonic neoplasms. qMSP has the lowest limit of detection of the MSP assay types tested, and a qMSP assay that detects methylated ITGA4 has potential as an early-detection assay for colon neoplasms.


Asunto(s)
Adenocarcinoma/diagnóstico , Adenoma/diagnóstico , Neoplasias Colorrectales/diagnóstico , Metilación de ADN , ADN/análisis , Heces/química , Reacción en Cadena de la Polimerasa/métodos , Adenocarcinoma/genética , Adenoma/genética , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Neoplasias Colorrectales/genética , ADN/genética , Detección Precoz del Cáncer , Humanos , Integrina alfa4/genética , Persona de Mediana Edad , Técnicas de Diagnóstico Molecular/métodos , Sensibilidad y Especificidad
10.
Dis Model Mech ; 12(3)2019 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-30792270

RESUMEN

To decrease bowel cancer incidence and improve survival, we need to understand the mechanisms that drive tumorigenesis. Recently, B-cell lymphoma 3 (BCL-3; a key regulator of NF-κB signalling) has been recognised as an important oncogenic player in solid tumours. Although reported to be overexpressed in a subset of colorectal cancers (CRCs), the role of BCL-3 expression in colorectal tumorigenesis remains poorly understood. Despite evidence in the literature that BCL-3 may interact with ß-catenin, it is perhaps surprising, given the importance of deregulated Wnt/ß-catenin/T-cell factor (TCF) signalling in colorectal carcinogenesis, that the functional significance of this interaction is not known. Here, we show for the first time that BCL-3 acts as a co-activator of ß-catenin/TCF-mediated transcriptional activity in CRC cell lines and that this interaction is important for Wnt-regulated intestinal stem cell gene expression. We demonstrate that targeting BCL-3 expression (using RNA interference) reduced ß-catenin/TCF-dependent transcription and the expression of intestinal stem cell genes LGR5 and ASCL2 In contrast, the expression of canonical Wnt targets Myc and cyclin D1 remained unchanged. Furthermore, we show that BCL-3 increases the functional stem cell phenotype, as shown by colorectal spheroid and tumoursphere formation in 3D culture conditions. We propose that BCL-3 acts as a driver of the stem cell phenotype in CRC cells, potentially promoting tumour cell plasticity and therapeutic resistance. As recent reports highlight the limitations of directly targeting cancer stem cells (CSCs), we believe that identifying and targeting drivers of stem cell plasticity have significant potential as new therapeutic targets.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción/metabolismo , Vía de Señalización Wnt , Proteínas del Linfoma 3 de Células B , Línea Celular Tumoral , Núcleo Celular/metabolismo , Regulación Neoplásica de la Expresión Génica , Genes Reporteros , Humanos , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Fenotipo , Unión Proteica , Transporte de Proteínas , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Factores de Transcripción TCF/metabolismo , Factores de Transcripción/genética , beta Catenina/metabolismo
11.
Carcinogenesis ; 29(4): 849-57, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18204076

RESUMEN

Although expression of the anti-apoptotic protein Bcl-2-associated athanogene-1 (BAG-1) has been reported as up-regulated in a number of malignancies, we show for the first time that BAG-1 is over-expressed in medium/large-sized colorectal adenomas and carcinomas compared with normal epithelium. To investigate whether expression of BAG-1 is important for colorectal tumour cell survival, microarray analysis was carried out on the HCT116 colorectal carcinoma cell line following transfection with BAG-1 small interfering RNA (siRNA). Analysis identified altered expression of a subset of potential nuclear factor-kappaB (NF-kappaB)-regulated genes. Furthermore, knock down of BAG-1 was shown to inhibit NF-kappaB transcriptional activity. Inhibition of NF-kappaB activity using BAG-1 siRNA or the NF-kappaB inhibitor BAY-117082 suppressed HCT116 cell yield and induced apoptosis; combined treatment had no additive effect, suggesting that the decrease in cell yield associated with knock down of BAG-1 expression is mediated via inhibition of NF-kappaB. Of clinical relevance, BAG-1 siRNA sensitized colorectal carcinoma cells to apoptosis induced by potential therapeutic agent TRAIL as well as tumour necrosis factor-alpha, both inducers of NF-kappaB activity. In summary, knock down of BAG-1 leads to inhibition of NF-kappaB, identifying BAG-1 as a novel regulator of NF-kappaB. It is proposed that, by inhibiting NF-kappaB, suppression of BAG-1 could represent a novel strategy to impede colorectal cancer cell survival and as an adjuvant increase sensitivity to current therapeutic regimes.


Asunto(s)
Neoplasias del Colon/genética , Neoplasias Colorrectales/genética , Proteínas de Unión al ADN/genética , FN-kappa B/metabolismo , ARN Interferente Pequeño/genética , Factores de Transcripción/genética , Adenoma/genética , Adenoma/patología , Carcinoma/genética , Carcinoma/patología , Línea Celular Tumoral , Supervivencia Celular/genética , Neoplasias del Colon/patología , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Células Tumorales Cultivadas
12.
Cancer Res ; 66(13): 6683-91, 2006 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16818642

RESUMEN

Cyclooxygenase (COX)-2, the inducible key enzyme for prostanoid biosynthesis, is overexpressed in most colorectal carcinomas and a subset of colorectal adenomas. Genetic, biochemical, and clinical evidence indicates an important role for COX-2 in colorectal tumorigenesis. Although COX-2 can be induced by aberrant growth factor signaling and oncogene activation during colorectal tumorigenesis, the role of microenvironmental factors such as hypoxia in COX-2 regulation remains to be elucidated. For the first time, we report that under hypoxic conditions COX-2 protein levels increase in colorectal adenoma and carcinoma cells. Rigorous analyses reveal that COX-2 up-regulation is transcriptional and is associated with hypoxia-inducible factor (HIF)-1alpha induction. Oligonucleotide pull-down and chromatin immunoprecipitation assays reveal that HIF-1alpha binds a hypoxia-responsive element on the COX-2 promoter. COX-2 up-regulation during hypoxia is accompanied by increased levels of prostaglandin E(2) (PGE(2)), which promote tumor cell survival under hypoxic conditions. In addition, elevated levels of PGE(2) in hypoxic colorectal tumor cells enhance vascular endothelial growth factor expression and HIF-1 transcriptional activity by activating the mitogen-activated protein kinase pathway, showing a potential positive feedback loop that contributes to COX-2 up-regulation during hypoxia. This study identifies COX-2 as a direct target for HIF-1 in colorectal tumor cells. In addition, COX-2 up-regulation represents a pivotal cellular adaptive response to hypoxia with implication for colorectal tumor cell survival and angiogenesis. We propose that using modified COX-2-selective inhibitors, which are only activated under hypoxic conditions, could potentially be a novel more selective strategy for colorectal cancer prevention and treatment.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Ciclooxigenasa 2/biosíntesis , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Secuencia de Bases , Procesos de Crecimiento Celular/fisiología , Hipoxia de la Célula/fisiología , Supervivencia Celular/fisiología , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/metabolismo , Ciclooxigenasa 2/genética , Dinoprostona/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Células HT29 , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Sistema de Señalización de MAP Quinasas , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Transcripción Genética , Regulación hacia Arriba
13.
Cancer Res ; 66(6): 3106-13, 2006 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-16540660

RESUMEN

Cyclooxygenase-2 and prostaglandin E(2) (PGE(2)) levels are increased in colorectal cancers and a subset of adenomas. PGE(2) signaling through the EP4 receptor has previously been associated with colorectal tumorigenesis. However, changes in EP4 expression during adenoma to carcinoma progression have not been investigated, neither has whether levels of EP4 influence important markers of malignant potential, such as anchorage-independent growth or the tumors growth response to PGE(2). We report using immunohistochemistry that in vivo EP4 receptor protein expression was increased in colorectal cancers (100%) as well as adenomas (36%) when compared with normal colonic epithelium. EP4 expression was also higher in colorectal carcinoma compared with adenoma cell lines and increased with in vitro models of tumor progression. Adenoma (PC/AA/C1 and RG/C2) and carcinoma cell lines (HT29) were growth stimulated by PGE(2) up to 0.5 micromol/L. However, although carcinoma and transformed adenoma (PC/AA/C1SB10C, a transformed derivative of PC/AA/C1) cells remain stimulated by higher doses of PGE(2) (10 micromol/L), the adenoma cell lines were inhibited. Interestingly, enforced expression of EP4 in the adenoma cell line, RG/C2, resulted in stimulation of growth by 10 micromol/L PGE(2) and promoted anchorage-independent growth. Both in vivo and in vitro data from this study suggest that increased EP4 receptor expression is important during colorectal carcinogenesis. We propose that high levels of PGE(2) in a tumor microenvironment would select for cells with increased EP4 expression, and that the EP4 receptor may therefore represent an important target for colorectal cancer prevention and treatment.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Receptores de Prostaglandina E/biosíntesis , Adenoma/genética , Adenoma/metabolismo , Adenoma/patología , Carcinoma/genética , Carcinoma/metabolismo , Carcinoma/patología , Adhesión Celular/fisiología , Procesos de Crecimiento Celular/efectos de los fármacos , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Transformación Celular Neoplásica/patología , Neoplasias Colorrectales/genética , Dinoprostona/farmacología , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Células HT29 , Humanos , Receptores de Prostaglandina E/genética , Subtipo EP4 de Receptores de Prostaglandina E , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
14.
Biochim Biophys Acta ; 1766(1): 104-19, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16859832

RESUMEN

Colorectal cancer is a major cause of mortality and whilst up to 80% of sporadic colorectal tumours are considered preventable, trends toward increasing obesity suggest the potential for a further increase in its worldwide incidence. Novel methods of colorectal cancer prevention and therapy are therefore of considerable importance. Non-steroidal anti-inflammatory drugs (NSAIDs) are chemopreventive against colorectal cancer, mainly through their inhibitory effects on the cyclooxygenase isoform COX-2. COX enzymes represent the committed step in prostaglandin biosynthesis and it is predominantly increased COX-2-mediated prostaglandin-E2 (PGE2) production that has a strong association with colorectal neoplasia, by promoting cell survival, cell growth, migration, invasion and angiogenesis. COX-1 and COX-2 inhibition by traditional NSAIDs (for example, aspirin) although chemopreventive have some side effects due to the role of COX-1 in maintaining the integrity of the gastric mucosa. Interestingly, the use of COX-2 selective NSAIDs has also shown promise in the prevention/treatment of colorectal cancer while having a reduced impact on the gastric mucosa. However, the prolonged use of high dose COX-2 selective inhibitors is associated with a risk of cardiovascular side effects. Whilst COX-2 inhibitors may still represent viable adjuvants to current colorectal cancer therapy, there is an urgent need to further our understanding of the downstream mechanisms by which PGE2 promotes tumorigenesis and hence identify safer, more effective strategies for the prevention of colorectal cancer. In particular, PGE2 synthases and E-prostanoid receptors (EP1-4) have recently attracted considerable interest in this area. It is hoped that at the appropriate stage, selective (and possibly combinatorial) inhibition of the synthesis and signalling of those prostaglandins most highly associated with colorectal tumorigenesis, such as PGE2, may have advantages over COX-2 selective inhibition and therefore represent more suitable targets for long-term chemoprevention. Furthermore, as COX-2 is found to be overexpressed in cancers such as breast, gastric, lung and pancreatic, these investigations may also have broad implications for the prevention/treatment of a number of other malignancies.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Neoplasias Colorrectales/prevención & control , Inhibidores de la Ciclooxigenasa/uso terapéutico , Dinoprostona/metabolismo , Transducción de Señal , Quimioprevención , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/terapia , Ciclooxigenasa 2/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Receptores de Prostaglandina E/fisiología
15.
Int J Cancer ; 121(10): 2172-80, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17583570

RESUMEN

Deregulation of cell survival pathways and resistance to apoptosis are widely accepted to be fundamental aspects of tumorigenesis. As in many tumours, the aberrant growth and survival of colorectal tumour cells is dependent upon a small number of highly activated signalling pathways, the inhibition of which elicits potent growth inhibitory or apoptotic responses in tumour cells. Accordingly, there is considerable interest in therapeutics that can modulate survival signalling pathways and target cancer cells for death. There is emerging evidence that cannabinoids, especially Delta(9)-tetrahydrocannabinol (THC), may represent novel anticancer agents, due to their ability to regulate signalling pathways critical for cell growth and survival. Here, we report that CB1 and CB2 cannabinoid receptors are expressed in human colorectal adenoma and carcinoma cells, and show for the first time that THC induces apoptosis in colorectal cancer cells. THC-induced apoptosis was rescued by pharmacological blockade of the CB1, but not CB2, cannabinoid receptor. Importantly, THC treatment resulted in CB1-mediated inhibition of both RAS-MAPK/ERK and PI3K-AKT survival signalling cascades; two key cell survival pathways frequently deregulated in colorectal tumours. The inhibition of ERK and AKT activity by THC was accompanied by activation of the proapoptotic BCL-2 family member BAD. Reduction of BAD protein expression by RNA interference rescued colorectal cancer cells from THC-induced apoptosis. These data suggest an important role for CB1 receptors and BAD in the regulation of apoptosis in colorectal cancer cells. The use of THC, or selective targeting of the CB1 receptor, may represent a novel strategy for colorectal cancer therapy.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Dronabinol/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Quinasas/metabolismo , Proteína Letal Asociada a bcl/metabolismo , Proteínas ras/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Fosforilación/efectos de los fármacos , Interferencia de ARN , ARN Mensajero/genética , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/genética , Receptor Cannabinoide CB1/metabolismo , Receptor Cannabinoide CB2/agonistas , Receptor Cannabinoide CB2/genética , Receptor Cannabinoide CB2/metabolismo , Transducción de Señal/efectos de los fármacos
17.
Cancers (Basel) ; 7(3): 1885-99, 2015 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-26393651

RESUMEN

Colorectal cancer is a major global health problem resulting in over 600,000 deaths world-wide every year with the majority of these due to metastatic disease. Wnt signalling, and more specifically ß-catenin-related transcription, has been shown to drive both tumorigenesis and the metastatic process in colorectal neoplasia, yet its complex interactions with other key signalling pathways, such as hedgehog, remain to be elucidated. We have previously shown that the Hedgehog (HH) signalling pathway is active in cells from colorectal tumours, and that inhibition of the pathway with cyclopamine induces apoptosis. We now show that cyclopamine treatment reduces ß-catenin related transcription in colorectal cancer cell lines, and that this effect can be reversed by addition of Sonic Hedgehog protein. We also show that cyclopamine concomitantly induces expression of the tumour suppressor and prognostic indicator E-cadherin. Consistent with a role for HH in regulating the invasive potential we show that cyclopamine reduces the expression of transcription factors (Slug, Snail and Twist) associated with the epithelial-mesenchymal transition and reduces the invasiveness of colorectal cancer cells in vitro. Taken together, Cancers 2015, 7 1886 these data show that pharmacological inhibition of the hedgehog pathway has therapeutic potential in the treatment of colorectal cancer.

19.
Cancer Res ; 73(21): 6435-47, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23950211

RESUMEN

Although microRNAs (miRNA) have been broadly studied in cancer, comparatively less is understood about their role in progression. Here we report that miR-153 has a dual role during progression of colorectal cancer by enhancing cellular invasiveness and platinum-based chemotherapy resistance. miRNA profiling revealed that miR-153 was highly expressed in a cellular model of advanced stage colorectal cancer. Its upregulation was also noted in primary human colorectal cancer compared with normal colonic epithelium and in more advanced colorectal cancer stages compared with early stage disease. In colorectal cancer patients followed for 50 months, 21 of 30 patients with high levels of miR-153 had disease progression compared with others in this group with low levels of miR-153. Functional studies revealed that miR-153 upregulation increased colorectal cancer invasiveness and resistance to oxaliplatin and cisplatin both in vitro and in vivo. Mechanistic investigations indicated that miR-153 promoted invasiveness indirectly by inducing matrix metalloprotease enzyme 9 production, whereas drug resistance was mediated directly by inhibiting the Forkhead transcription factor Forkhead box O3a (FOXO3a). In support of the latter finding, we found that levels of miR-153 and FOXO3a were inversely correlated in matched human colorectal cancer specimens. Our findings establish key roles for miR-153 overexpression in colorectal cancer progression, rationalizing therapeutic strategies to target expression of this miRNA for colorectal cancer treatment.


Asunto(s)
Cisplatino/farmacología , Neoplasias Colorrectales/patología , Resistencia a Antineoplásicos , Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Hibridación Genómica Comparativa , Cartilla de ADN/química , Progresión de la Enfermedad , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/antagonistas & inhibidores , Factores de Transcripción Forkhead/genética , Perfilación de la Expresión Génica , Humanos , Técnicas para Inmunoenzimas , Ratones , Ratones SCID , Invasividad Neoplásica , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Matrices Tisulares , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Curr Pharm Des ; 18(26): 3874-88, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22632753

RESUMEN

TGF-ß1 is an anti-inflammatory cytokine recognised as a key regulator of immunological homeostasis and inflammatory responses. Furthermore, TGF-ß1 is important for the regulation of cell growth, differentiation and apoptosis in a wide range of tissues including the intestinal epithelium. Reduced TGF-ß1 activity is thought to be responsible for the development of autoimmune disorders in several pathological conditions, including inflammatory bowel disease [IBD]. Although the cause of IBD is not yet known, research has shown that a number of factors may be involved including environment, diet and genetics, as well as cytokine exposure. Importantly, IBD is also associated with an increased lifetime risk of developing colorectal cancer, which remains the fourth most common cancer worldwide, representing a significant therapeutic challenge. As functionally implicated in both maintenance of the immune response and tissue homeostasis in the colon, TGF-ß1 signalling potentially sits at the crossroads between aberrant inflammation and colorectal tumorigenesis. Hence, the purpose of this paper is to review the evidence for cross talk between TGF-ß1 signalling and pathways important for colorectal tissue homeostasis, with the emphasis on understanding how deregulation of TGF-ß1 signalling contributes not only to aberrant inflammatory disease but also to colorectal tumour progression.


Asunto(s)
Neoplasias Colorrectales/patología , Inflamación/fisiopatología , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Apoptosis , Diferenciación Celular , Neoplasias Colorrectales/etiología , Progresión de la Enfermedad , Humanos , Inflamación/complicaciones , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA