Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 123
Filter
1.
J Environ Manage ; 321: 115975, 2022 Nov 01.
Article in English | MEDLINE | ID: mdl-35988399

ABSTRACT

Raw printing ink wastewater (PIW) was treated with various inorganic coagulants and organic flocculants (anionic and cationic polyacrylamides). These processes were also examined as post treatment step following hydrodynamic cavitation. Treatment effectiveness was assessed through color, chemical oxygen demand (COD) and total suspended solids (TSS) removal. The addition of 4500 mg L-1 polyaluminum chloride coagulant in undiluted PIW (COD: 17000 mg L-1) resulted in 99% color removal, 96% COD and TSS removal, after settling for 2 h. The addition of 10 mg L-1 of anionic polyacrylamides in the sample reduced settling time to only 5 min, with concomitant 96-98% removal efficiency. The addition of a 4 min hydrodynamic cavitation pretreatment step reduced coagulant addition by 33%, for the treatment of undiluted PIW (with 10 mg L-1 anionic polyacrylamide), while removals were ranged between 96 and 98%. Economic analysis for the undiluted PIW showed that costs were reduced by ca. 20% with the hydrodynamic cavitation pretreatment step. Moreover, sludge characterization showed the presence of maghemite, aluminum chloride and potassium aluminum silicate. Finally, toxicity tests revealed a significant attenuation of the toxic potential of undiluted PIW, thus indicating the enhanced efficiency of the proposed combined process (hydrodynamic cavitation and coagulation/flocculation).


Subject(s)
Waste Disposal, Fluid , Water Purification , Anions , Flocculation , Hydrodynamics , Ink , Printing, Three-Dimensional , Waste Disposal, Fluid/methods , Wastewater/chemistry
2.
Br J Cancer ; 112(4): 714-9, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25611300

ABSTRACT

BACKGROUND: LGR5 is an important marker of intestinal stem cells and performs its vital functions at the cell membrane. Despite the importance of LGR5 to both normal and cancer stem cell biology, it is not known how microenvironmental stress affects the expression and subcellular distribution of the protein. METHODS: Nutrient stress was induced through glucose starvation. Glycosylation status was assessed using endoglycosidase or tunicamycin treatment. Flow cytometry and confocal microscopy were used to assess subcellular distribution of LGR5. RESULTS: Glucose deprivation altered the glycosylation status of LGR5 resulting in reduced protein stability and cell surface expression. Furthermore, inhibiting LGR5 glycosylation resulted in depleted surface expression and reduced localisation in the cis-Golgi network. CONCLUSIONS: Nutrient stress within a tumour microenvironment has the capacity to alter LGR5 protein stability and membrane localisation through modulation of LGR5 glycosylation status. As LGR5 surface localisation is required for enhanced Wnt signalling, this is the first report to show a mechanism by which the microenvironment could affect LGR5 function.


Subject(s)
Adenoma/metabolism , Cell Membrane/metabolism , Colorectal Neoplasms/metabolism , Glucose/deficiency , Neoplastic Stem Cells/metabolism , Receptors, G-Protein-Coupled/metabolism , Stress, Physiological/physiology , Adenoma/genetics , Adenoma/therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/therapy , Food , Glycosylation , Humans , Protein Stability , Protein Transport , Receptors, G-Protein-Coupled/genetics , Tumor Cells, Cultured , Tumor Microenvironment
3.
Oncogene ; 32(38): 4490-9, 2013 Sep 19.
Article in English | MEDLINE | ID: mdl-23108401

ABSTRACT

As colorectal cancer remains the second highest cause of cancer-related deaths in much of the industrialised world, identifying novel strategies to prevent colorectal tumour development remains an important challenge. BAG-1 is a multi-functional protein, the expression of which is up-regulated at relatively early stages in colorectal tumorigenesis. Importantly, BAG-1 is thought to enhance colorectal tumour progression through promoting tumour cell survival. Here, we report for the first time a novel role for BAG-1, establishing it as a suppressor of transforming growth factor ß (TGF-ß1) expression in colorectal tumour cells. Microarray analysis first highlighted the possibility that BAG-1 may regulate TGF-ß1 expression, a key cytokine in normal colonic tissue homoeostasis. Q-RT-PCR and ELISA demonstrated TGFB1 mRNA and protein expression to be significantly increased when BAG1 levels were reduced by small interfering RNA; additionally, induction of BAG-1L caused suppression of TGFB1 mRNA in colorectal tumour cells. Using reporter and chromatin immunoprecipitation assays, a direct association of BAG-1 with the TGFB1 gene regulatory region was identified. Immunohistochemistry and Weiser fraction data indicated that the levels of BAG-1 and TGF-ß1 are inversely correlated in the normal colonic epithelium in vivo, consistent with a role for BAG-1-mediated repression of TGF-ß1 production. In vitro studies showed that the change in TGF-ß1 production following manipulation of BAG-1 is functionally relevant; through induction of anchorage-independent growth in TGF-ß1-dependent normal rat kidney fibroblasts and regulation of SMAD2 phosphorylation in TGF-ß1-sensitive adenoma cells. Taken together, this study identifies the anti-apoptotic protein BAG-1 as a suppressor of the inhibitory growth factor TGF-ß1, suggesting that high expression of BAG-1 can impact on a number of the hallmarks of cancer, of potential importance in promoting the early stages of colorectal tumorigenesis. Establishing BAG-1 as a repressor of TGF-ß1 has important biological implications, and highlights a new role for BAG-1 in colorectal tumorigenesis.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Transcription Factors/metabolism , Transforming Growth Factor beta1/genetics , Carcinoma/genetics , Carcinoma/metabolism , Cell Line, Tumor , Epithelial Cells/metabolism , Humans , Models, Biological , Protein Binding , Regulatory Sequences, Nucleic Acid , Transcription, Genetic , Transforming Growth Factor beta1/metabolism
4.
Br J Cancer ; 107(9): 1514-7, 2012 Oct 23.
Article in English | MEDLINE | ID: mdl-23033009

ABSTRACT

BACKGROUND: Levels of the pro-tumorigenic prostaglandin PGE(2) are increased in colorectal cancer, previously attributed to increased synthesis through COX-2 upregulation and, more recently, to decreased catabolism. The functionally linked genes 15-prostaglandin dehydrogenase (15-PGDH) and the prostaglandin transporter PGT co-operate in prostaglandin degradation and are downregulated in colorectal cancer. We previously reported repression of 15-PGDH expression by the Wnt/ß-catenin pathway, commonly deregulated during early colorectal neoplasia. Here we asked whether ß-catenin also regulates PGT expression. METHODS: The effect of ß-catenin deletion in vivo was addressed by PGT immunostaining of ß-catenin(-/lox)-villin-cre-ERT2 mouse tissue. The effect of siRNA-mediated ß-catenin knockdown and dnTCF4 induction in vitro was addressed by semi-quantitative and quantitative real-time RT-PCR and immunoblotting. RESULTS: This study shows for the first time that deletion of ß-catenin in murine intestinal epithelium in vivo upregulates PGT protein, especially in the crypt epithelium. Furthermore, ß-catenin knockdown in vitro increases PGT expression in both colorectal adenoma- and carcinoma-derived cell lines, as does dnTCF4 induction in LS174T cells. CONCLUSIONS: These data suggest that ß-catenin employs a two-pronged approach to inhibiting prostaglandin turnover during colorectal neoplasia by repressing PGT expression in addition to 15-PGDH. Furthermore, our data highlight a potential mechanism that may contribute to the non-selective NSAID aspirin's chemopreventive efficacy.


Subject(s)
Aspirin/pharmacology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/prevention & control , Intestinal Mucosa/metabolism , Organic Anion Transporters/biosynthesis , beta Catenin/metabolism , Animals , Anticarcinogenic Agents/pharmacology , Cell Line, Tumor , HCT116 Cells , HT29 Cells , Humans , Hydroxyprostaglandin Dehydrogenases/genetics , Hydroxyprostaglandin Dehydrogenases/metabolism , Mice , Organic Anion Transporters/genetics , Signal Transduction , beta Catenin/genetics
5.
Cell Death Dis ; 3: e408, 2012 Oct 11.
Article in English | MEDLINE | ID: mdl-23059827

ABSTRACT

Although the retinoblastoma-susceptibility gene RB1 is inactivated in a wide range of human tumours, in colorectal cancer, the retinoblastoma protein (Rb) function is often preserved and the RB locus even amplified. Importantly, we have previously shown that Rb interacts with the anti-apoptotic Bcl-2 associated athanogene 1 (BAG-1) protein, which is highly expressed in colorectal carcinogenesis. Here we show for the first time that Rb expression is critical for BAG-1 anti-apoptotic activity in colorectal tumour cells. We demonstrate that Rb expression not only increases the nuclear localisation of the anti-apoptotic BAG-1 protein, but that expression of Rb is required for inhibition of apoptosis by BAG-1 both in a γ-irradiated Saos-2 osteosarcoma cell line and colorectal adenoma and carcinoma cell lines. Further, consistent with the fact that nuclear BAG-1 has previously been shown to promote cell survival through increasing nuclear factor (NF)-κB activity, we demonstrate that the ability of BAG-1 to promote NF-κB activity is significantly inhibited by repression of Rb expression. Taken together, data presented suggest a novel function for Rb, promoting cell survival through regulating the function of BAG-1. As BAG-1 is highly expressed in the majority of colorectal tumours, targeting the Rb-BAG-1 complex to promote apoptosis has exciting potential for future therapeutic development.


Subject(s)
DNA-Binding Proteins/metabolism , Retinoblastoma Protein/metabolism , Transcription Factors/metabolism , Apoptosis , Cell Nucleus/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Humans , NF-kappa B/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Retinoblastoma Protein/antagonists & inhibitors , Retinoblastoma Protein/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Tumor Cells, Cultured
6.
Oncogene ; 31(40): 4409-19, 2012 Oct 04.
Article in English | MEDLINE | ID: mdl-22249255

ABSTRACT

Loss of tumour suppressor gene function can occur as a result of epigenetic silencing of large chromosomal regions, referred to as long-range epigenetic silencing (LRES), and genome-wide analyses have revealed that LRES is present in many cancer types. Here we utilize Illumina Beadchip methylation array analysis to identify LRES across 800 kb of chromosome 5q31 in colorectal adenomas and carcinomas (n=34) relative to normal colonic epithelial DNA (n=6). This region encompasses 53 individual protocadherin (PCDH) genes divided among three gene clusters. Hypermethylation within these gene clusters is asynchronous; while most PCDH hypermethylation occurs early, and is apparent in adenomas, PCDHGC3 promoter methylation occurs later in the adenoma-carcinoma transition. PCDHGC3 was hypermethylated in 17/28 carcinomas (60.7%) according to methylation array analysis. Quantitative real-time reverse transcription-polymerase chain reaction showed that PCDHGC3 is the highest expressed PCDH in normal colonic epithelium, and that there was a strong reciprocal relationship between PCDHGC3 methylation and expression in carcinomas (R=-0.84). PCDH LRES patterns are reflected in colorectal tumour cell lines; adenoma cell lines are not methylated at PCDHGC3 and show abundant expression at the mRNA and protein level, while the expression is suppressed in hypermethylated carcinoma cell lines (R=-0.73). Short-interfering RNA-mediated reduction of PCDHGC3 led to a decrease of apoptosis in RG/C2 adenoma cells, and overexpression of PCDHGC3 in HCT116 cells resulted in the reduction of colony formation, consistent with tumour suppressor capabilities for PCDHGC3. Further functional analysis showed that PCDHGC3 can suppress Wnt and mammalian target of rapamycin signalling in colorectal cancer cell lines. Taken together, our data suggest that the PCDH LRES is an important tumour suppressor locus in colorectal cancer, and that PCDHGC3 may be a strong marker and driver for the adenoma-carcinoma transition.


Subject(s)
Cadherins/genetics , Chromosomes, Human, Pair 5 , Colorectal Neoplasms/genetics , Epigenesis, Genetic , Gene Silencing , Cadherin Related Proteins , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Signal Transduction/genetics
7.
Oncogene ; 31(22): 2761-72, 2012 May 31.
Article in English | MEDLINE | ID: mdl-21963853

ABSTRACT

Understanding the mechanisms that promote aberrant tumour cell survival is critical for the determination of novel strategies to combat colorectal cancer (CRC). We have recently shown that the anti-apoptotic protein BAG-1, highly expressed in pre-malignant and CRC tissue, can potentiate cell survival through regulating NF-κB transcriptional activity. In this study, we identify a novel complex between BAG-1 and the p50-p50 NF-κB homodimers, implicating BAG-1 as a co-regulator of an atypical NF-κB pathway. Importantly, the BAG-1-p50 complex was detected at gene regulatory sequences including the epidermal growth factor receptor (EGFR) and COX-2 (PTGS2) genes. Suppression of BAG-1 expression using small interfering RNA was shown to increase EGFR and suppress COX-2 expression in CRC cells. Furthermore, mouse embryonic fibroblasts derived from the NF-κB1 (p105/p50) knock-out mouse were used to demonstrate that p50 expression was required for BAG-1 to suppress EGFR expression. This was shown to be functionally relevant as attenuation of BAG-1 expression increased ligand activated phosphorylation of EGFR in CRC cells. In summary, this paper identifies a novel role for BAG-1 in modulating gene expression through interaction with the p50-p50 NF-κB complexes. Data presented led us to propose that BAG-1 can act as a selective regulator of p50-p50 NF-κB responsive genes in colorectal tumour cells, potentially important for the promotion of cell survival in the context of the fluctuating tumour microenvironment. As BAG-1 expression is increased in the developing adenoma through to metastatic lesions, understanding the function of the BAG-1-p50 NF-κB complexes may aid in identifying strategies for both the prevention and treatment of CRC.


Subject(s)
Cell Transformation, Neoplastic/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA-Binding Proteins/metabolism , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , NF-kappa B p50 Subunit/metabolism , NF-kappa B/physiology , Transcription Factors/metabolism , Animals , Apoptosis , Blotting, Western , Cell Proliferation , Cells, Cultured , Chromatin Immunoprecipitation , Colorectal Neoplasms/genetics , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , DNA-Binding Proteins/genetics , Electrophoretic Mobility Shift Assay , Embryo, Mammalian , ErbB Receptors/genetics , Fibroblasts , Humans , Immunoenzyme Techniques , Immunoprecipitation , Luciferases/metabolism , Mice , Mice, Knockout , NF-kappa B/antagonists & inhibitors , NF-kappa B p50 Subunit/genetics , Promoter Regions, Genetic , Protein Multimerization , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transfection
8.
Oncogene ; 29(23): 3398-410, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20348947

ABSTRACT

Overexpression of cyclooxygenase-2 (COX-2) and elevated levels of its enzymatic product prostaglandin E2 (PGE(2)) occur in the majority of colorectal cancers and have important roles in colorectal tumorigenesis. However, despite the established prosurvival role of PGE(2) in cancer, the underlying mechanisms are not fully understood. Here, we have shown that PGE(2) suppresses apoptosis via repression of the proapoptotic BH3-only protein Bim in human colorectal adenoma cells. Repression of Bim expression was dependent upon PGE(2)-mediated activation of the Raf-MEK-ERK1/2 pathway, which promoted Bim phosphorylation and proteasomal degradation. Reduction of Bim expression using RNA interference reduced spontaneous apoptosis in adenoma cells and abrogated PGE(2)-dependent apoptosis suppression. Treatment of COX-2-expressing colorectal carcinoma cells with COX-2-selective NSAIDs-induced Bim expression, suggesting that Bim repression via PGE(2) signalling may be opposed by COX-2 inhibition. Examination of Bim expression in two established in vitro models of the adenoma-carcinoma sequence revealed that downregulation of Bim expression was associated with tumour progression towards an anchorage-independent phenotype. Finally, immunohistochemical analyses revealed that Bim expression is markedly reduced in approximately 40% of human colorectal carcinomas in vivo. These observations highlight the COX-2/PGE(2) pathway as an important negative regulator of Bim expression in colorectal tumours and suggest that Bim repression may be an important step during colorectal cancer tumorigenesis.


Subject(s)
Adenoma/etiology , Apoptosis Regulatory Proteins/physiology , Colorectal Neoplasms/etiology , Cyclooxygenase 2/physiology , Dinoprostone/physiology , Membrane Proteins/physiology , Proto-Oncogene Proteins/physiology , Signal Transduction/physiology , Adenoma/pathology , Apoptosis , Bcl-2-Like Protein 11 , Cell Line, Tumor , Colorectal Neoplasms/pathology , Disease Progression , Down-Regulation , Extracellular Signal-Regulated MAP Kinases/physiology , Humans , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Proteasome Endopeptidase Complex/physiology , Proto-Oncogene Proteins c-akt/physiology , bcl-Associated Death Protein/physiology
9.
Br J Cancer ; 101(7): 1124-9, 2009 Oct 06.
Article in English | MEDLINE | ID: mdl-19738613

ABSTRACT

BACKGROUND: Fascin is overexpressed in many cancers, including colorectal, but its role in the malignant transformation of benign colorectal adenomas is unclear. METHODS: Immunohistochemical analysis of fascin expression was carried out in resected human colorectal adenoma specimens. The effects of forced overexpression of fascin on adenoma cell motility were also analysed. RESULTS: We show fascin overexpression in adenomas increasing with tumour size, histological type, and degree of dysplasia and increased cell motility in adenoma cell lines following fascin transfection. CONCLUSION: These data suggest an important role for fascin in the malignant progression of colorectal tumours.


Subject(s)
Adenoma/pathology , Carrier Proteins/physiology , Colorectal Neoplasms/pathology , Microfilament Proteins/physiology , Adenoma/chemistry , Carrier Proteins/analysis , Cell Line, Tumor , Cell Movement , Colorectal Neoplasms/chemistry , Disease Progression , Humans , Immunohistochemistry , Microfilament Proteins/analysis
10.
Br J Cancer ; 98(8): 1366-79, 2008 Apr 22.
Article in English | MEDLINE | ID: mdl-18349829

ABSTRACT

Bevacizumab, an anti-vascular endothelial growth factor (VEGF-A) antibody, is used in metastatic colorectal carcinoma (CRC) treatment, but responses are unpredictable. Vascular endothelial growth factor is alternatively spliced to form proangiogenic VEGF(165) and antiangiogenic VEGF(165)b. Using isoform-specific enzyme-linked immunosorbent assay and quantitative polymerase chain reaction, we found that over 90% of the VEGF in normal colonic tissue was VEGF(xxx)b, but there was a variable upregulation of VEGF(xxx) and downregulation of VEGF(xxx)b in paired human CRC samples. Furthermore, cultured colonic adenoma cells expressed predominantly VEGF(xxx)b, whereas colonic carcinoma cells expressed predominantly VEGF(xxx). However, adenoma cells exposed to hypoxia switched their expression from predominantly VEGF(xxx)b to predominantly VEGF(xxx). VEGF(165)b overexpression in LS174t colon cancer cells inhibited colon carcinoma growth in mouse xenograft models. Western blotting and surface plasmon resonance showed that VEGF(165)b bound to bevacizumab with similar affinity as VEGF(165). However, although bevacizumab effectively inhibited the rapid growth of colon carcinomas expressing VEGF(165), it did not affect the slower growth of tumours from colonic carcinoma cells expressing VEGF(165)b. Both bevacizumab and anti-VEGF(165)b-specific antibodies were cytotoxic to colonic epithelial cells, but less so to colonic carcinoma cells. These results show that the balance of antiangiogenic to proangiogenic isoforms switches to a variable extent in CRC, regulates tumour growth rates and affects the sensitivity of tumours to bevacizumab by competitive binding. Together with the identification of an autocrine cytoprotective role for VEGF(165)b in colonic epithelial cells, these results indicate that bevacizumab treatment of human CRC may depend upon this balance of VEGF isoforms.


Subject(s)
Angiogenesis Inhibitors/physiology , Antibodies, Monoclonal/therapeutic use , Colorectal Neoplasms/drug therapy , Vascular Endothelial Growth Factor A/physiology , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Humanized , Bevacizumab , Cell Line, Tumor , Cell Proliferation , Colon/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Humans , Mice , Protein Isoforms , RNA Splicing , RNA, Messenger/analysis , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/antagonists & inhibitors
11.
Oncogene ; 27(27): 3880-8, 2008 Jun 19.
Article in English | MEDLINE | ID: mdl-18264139

ABSTRACT

MicroRNAs are small, non-coding RNAs that influence gene regulatory networks by post-transcriptional regulation of specific messenger RNA targets. MicroRNA expression is dysregulated in human malignancies, frequently leading to loss of expression of certain microRNAs. We report that expression of hsa-miR-342, a microRNA encoded in an intron of the gene EVL, is commonly suppressed in human colorectal cancer. The expression of hsa-miR-342 is coordinated with that of EVL and our results indicate that the mechanism of silencing is CpG island methylation upstream of EVL. We found methylation at the EVL/hsa-miR-342 locus in 86% of colorectal adenocarcinomas and in 67% of adenomas, indicating that it is an early event in colorectal carcinogenesis. In addition, we observed a higher frequency of methylation (56%) in histologically normal colorectal mucosa from individuals with concurrent cancer compared to mucosa from individuals without colorectal cancer (12%), suggesting the existence of a 'field defect' involving methylated EVL/hsa-miR-342. Furthermore, reconstitution of hsa-miR-342 in the colorectal cancer cell line HT-29 induced apoptosis, suggesting that this microRNA could function as a proapoptotic tumor suppressor. In aggregate, these results support a novel mechanism for silencing intronic microRNAs in cancer by epigenetic alterations of cognate host genes.


Subject(s)
Cell Adhesion Molecules/genetics , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Gene Silencing , Introns , MicroRNAs/genetics , Apoptosis , Cell Line, Tumor , Colorectal Neoplasms/pathology , DNA Methylation , DNA, Neoplasm/genetics , Humans
12.
Biochem Soc Trans ; 35(Pt 5): 1372-4, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17956354

ABSTRACT

Cell growth pathways are mediated through protein-glycan interactions including O-glycosylation. Investigation of these growth pathways can be carried out using appropriate inhibitors to identify stage-specific events. We have adopted this approach to study a group of benzyl-O-N-acetyl-D-galactosamine analogues in human colorectal cancer cell lines. Exposure to O-glycan inhibitors resulted in the induction of apoptosis, a block in proliferation, accumulation of intracellular aryl-glycans and changes in related genes as detected by gene array. Colorectal cancer cell lines susceptible to the inhibitors showed growth arrest with all compounds. However, a differential action of each inhibitor was detected in the pattern of genes affected and in the structure of aryl-glycans formed.


Subject(s)
Apoptosis , Cell Proliferation , Colorectal Neoplasms/pathology , Polysaccharides/metabolism , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Glycosylation , Humans , Polysaccharides/antagonists & inhibitors , Signal Transduction
13.
Cell Prolif ; 40(5): 768-79, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17877615

ABSTRACT

UNLABELLED: The use of non-steroidal anti-inflammatory drugs has proved of great interest in the prevention and treatment of colorectal cancer, although their precise mechanisms of action remain unclear. Overexpression of cyclooxygenase-2 (COX-2) and subsequent prostaglandin production promote metastasis and have been shown to increase cell motility in vitro. OBJECTIVE: We have aimed to elucidate whether specific inhibition of COX-2 with NS-398 (NS-398 is a selective inhibitor of COX-2) would be able to inhibit motility of colorectal cancer cells and whether this was modulated through epidermal growth factor receptor (EGFR) transactivation. MATERIALS AND METHODS: A transwell filter assay was used to study cell motility. Expression of COX-2, EGFR phosphorylation and prostaglandin E(2) (PGE(2)) receptors were assessed by Western blot analysis and reverse transcriptase-polymerase chain reaction. PGE(2) concentrations after NS-398 treatment were estimated by enzyme immunoassay. RESULTS: Treatment with NS-398 significantly reduced PGE(2) levels and reduced cell migration in the HT29 and HCA7 colorectal carcinoma cell lines and this effect was rescued by addition of PGE(2). Furthermore, specific inhibition of COX-2 with NS-398 reduced EGFR phosphorylation in colorectal cancer cells. Direct inhibition of EGFR activity with AG1478 reduced PGE(2)-stimulated motility, clearly demonstrating that PGE(2 )acts via the EGFR-signalling pathway. The novel combination of NS-398 and AG1478 dramatically reduced migration of colorectal cancer cells. CONCLUSION: The data presented indicate that the use of NS-398 in chemoprevention and adjuvant therapy for colorectal cancer may work in part, through the inhibition of cell motility. Furthermore, our data suggest that the combined use of non-steroidal anti-inflammatory drugs with EGFR antagonists could be explored further for future use in the clinic.


Subject(s)
Colonic Neoplasms/drug therapy , Cyclooxygenase 2 Inhibitors/pharmacology , ErbB Receptors/genetics , Nitrobenzenes/pharmacology , Sulfonamides/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Base Sequence , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cyclooxygenase 2 Inhibitors/administration & dosage , DNA Primers/genetics , Dinoprostone/administration & dosage , Dinoprostone/metabolism , Dinoprostone/pharmacology , ErbB Receptors/metabolism , Humans , Phosphorylation , Transcriptional Activation/drug effects
14.
Water Environ Res ; 79(4): 421-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17489277

ABSTRACT

Olive mill wastewater (OMW) produced from small units scattered in rural areas of Southern Europe is a major source of pollution of surface and subsurface water. In the present work, a treatment scheme based on physical separation methods is presented. The investigation was carried out using a pilot-plant unit equipped with ultrafiltration, nanofiltration, and reverse osmosis membranes. Approximately 80% of the total volume of wastewater treated by the membrane units was sufficiently cleaned to meet the standards for irrigation water. The concentrated fractions collected in the treatment concentrates were characterized by high organic load and high content of phenolic compounds. The concentrates were tested in hydroponic systems to examine their toxicity towards undesired herbs. The calculations of the cost of the overall process showed that fixed and operational costs could be recovered from the exploitation of OMW byproducts as water for irrigation and/or as bioherbicides.


Subject(s)
Filtration/methods , Industrial Waste , Plant Oils/isolation & purification , Waste Disposal, Fluid , Water Pollutants, Chemical/isolation & purification , Water Purification/methods , Bioreactors , Olive Oil , Phenols/analysis , Plant Oils/chemistry
15.
Cell Death Differ ; 14(1): 137-45, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16645643

ABSTRACT

There is growing evidence that the insulin-like growth factor-binding protein 3 (IGFBP-3) can have IGF-independent effects on cell growth. However, despite the fact that IGFBP-3 has been reported to be both antiproliferative and proapoptotic, the molecular mechanisms underlying the action of IGFBP-3 have not been elucidated. We report that although addition of IGFBP-3 (either synthetic or secreted protein) had no effect on cell survival, IGFBP-3 (100 ng/ml) significantly enhanced TNF-related apoptosis-inducing ligand (TRAIL)-induced cell death in colonic carcinoma-derived cell lines (20-30% depending on cell line), whereas it had no effect on the survival of the TRAIL-resistant adenoma-derived cells. Both addition of IGFBP-3 protein to cell cultures or enforced expression of IGFBP-3 in the HT29 carcinoma cell line inhibited nuclear factor kappa B (NF-kappaB) activation in response to the induction of apoptosis by TRAIL. We propose that IGFBP-3 is a non-toxic NF-kappaB inhibitor, which could be used as an adjuvant in the treatment of colon cancer.


Subject(s)
Adenoma/physiopathology , Apoptosis , Carcinoma/physiopathology , Colorectal Neoplasms/physiopathology , Insulin-Like Growth Factor Binding Protein 3/metabolism , NF-kappa B/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Cell Line, Tumor , HT29 Cells , Humans , I-kappa B Proteins , NF-KappaB Inhibitor alpha , Signal Transduction , Tumor Cells, Cultured
16.
Gut ; 54(12): 1741-50, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16099783

ABSTRACT

BACKGROUND AND AIMS: Cyclooxygenase 2 (COX-2) is upregulated in most colorectal cancers and is responsible for metabolism of the endogenous cannabinoid, anandamide, into prostaglandin-ethanolamides (PG-EAs). The aims of this study were to determine whether anandamide and PG-EAs induce cell death in colorectal carcinoma (CRC) cells, and whether high levels of COX-2 in CRC cells could be utilised for their specific targeting for cell death by anandamide. METHODS: We determined the effect of anandamide on human CRC cell growth by measuring cell growth and cell death, whether this was dependent on COX-2 protein expression or enzyme activity, and the potential involvement of PG-EAs in induction of cell death. RESULTS: Anandamide inhibited the growth of CRC cell lines HT29 and HCA7/C29 (moderate and high COX-2 expressors, respectively) but had little effect on the very low COX-2 expressing CRC cell line, SW480. Induction of cell death in HT29 and HCA7/C29 cell lines was partially rescued by the COX-2 selective inhibitor NS398. Cell death induced by anandamide was neither apoptosis nor necrosis. Furthermore, inhibition of fatty acid amide hydrolase potentiated the non-apoptotic cell death, indicating that anandamide induced cell death was mediated via metabolism of anandamide by COX-2, rather than its degradation into arachidonic acid and ethanolamine. Interestingly, both PGE2-EA and PGD2-EA induced classical apoptosis. CONCLUSIONS: These findings suggest anandamide may be a useful chemopreventive/therapeutic agent for colorectal cancer as it targets cells that are high expressors of COX-2, and may also be used in the eradication of tumour cells that have become resistant to apoptosis.


Subject(s)
Arachidonic Acids/pharmacology , Colorectal Neoplasms/pathology , Cyclooxygenase 2/physiology , Amidohydrolases/antagonists & inhibitors , Apoptosis/drug effects , Arachidonic Acids/antagonists & inhibitors , Cell Death/drug effects , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/metabolism , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/analogs & derivatives , Dinoprostone/biosynthesis , Dose-Response Relationship, Drug , Endocannabinoids , Humans , Polyunsaturated Alkamides , Tumor Cells, Cultured
17.
Gut ; 54(9): 1209-11, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16099785

ABSTRACT

For the 500,000 new cases of colorectal cancer in the world each year, identification of patients with a worse prognosis and those who are more likely to respond to treatment is a challenge. There is an increasing body of evidence correlating genetic mutations with outcome in tumours derived from human colorectal cancer cohorts. K-ras, but not p53 or APC, mutations appear to be associated with poorer overall survival in colorectal cancer patients.


Subject(s)
Colorectal Neoplasms/genetics , Genes, ras , Mutation , Aged , Colorectal Neoplasms/mortality , DNA, Neoplasm/analysis , Female , Genes, APC , Genes, p53 , Humans , Male , Middle Aged , Polymorphism, Restriction Fragment Length , Prognosis , Survival Rate
18.
Biochem Soc Trans ; 33(Pt 4): 667-71, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16042570

ABSTRACT

There is strong evidence for an important role for increased COX (cyclo-oxygenase)-2 expression and PG (prostaglandin) E2 production in colorectal tumorigenesis. PGE(2) acts through four E-prostanoid receptors (EP1-4). COX-2 has therefore become a target for the potential chemoprevention and therapy of colorectal cancer. However, any therapeutic/preventive strategy has the potential to have an impact on physiological processes and hence result in side effects. General COX (COX-1 and -2) inhibition by traditional NSAIDs (non-steroidal anti-inflammatory drugs), such as aspirin, although chemopreventive, has some side effects, as do some conventional COX-2-selective NSAIDs. As PGE2 is thought to be the major PG species responsible for promoting colorectal tumorigenesis, research is being directed to a number of protein targets downstream of COX-2 that might allow the selective inhibition of the tumour-promoting activities of PGE2, while minimizing the associated adverse events. The PGE synthases and E-prostanoid receptors (EP1-4) have therefore recently attracted considerable interest as potential novel targets for the prevention/therapy of colorectal cancer. Selective (and possibly combinatorial) inhibition of the synthesis and signalling of those PGs most highly associated with colorectal tumorigenesis may have some advantages over COX-2-selective inhibitors.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Anticarcinogenic Agents/therapeutic use , Colorectal Neoplasms/prevention & control , Cyclooxygenase Inhibitors/therapeutic use , Colorectal Neoplasms/epidemiology , Humans , Incidence , Models, Biological , Receptors, Prostaglandin E/physiology , Receptors, Prostaglandin E, EP1 Subtype
19.
Biochem Soc Trans ; 33(Pt 4): 676-8, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16042572

ABSTRACT

Although the retinoblastoma susceptibility gene RB1 is inactivated in a wide variety of human cancers, the retinoblastoma protein (Rb) has been shown to be overexpressed in colon cancers, which is linked to the anti-apoptotic function of the protein. However, the mechanisms by which Rb regulates apoptosis are yet to be fully elucidated. We have established that Rb interacts with the anti-apoptotic BAG-1 (Bcl-2 associated athanogene-1) protein, and that a decrease in nuclear localization of BAG-1 is detectable when the interaction between Rb and BAG-1 is disrupted by expression of the E7 viral oncoprotein. Interestingly, although reported as deregulated in colorectal cancers, we have found that BAG-1 expression is also altered in small adenomas, where its localization was found to be predominantly nuclear. In addition, we have established that maintenance of high nuclear BAG-1 in vitro increases the resistance of adenoma-derived cells to gamma-radiation-induced apoptosis. Our work suggests a novel function for Rb, involving modulation of the subcellular localization of BAG-1. We have found predominant nuclear BAG-1 localization in small adenomas, and suggest that BAG-1 may promote colorectal tumour cell survival by making colonic epithelial cells less sensitive to DNA damage.


Subject(s)
Carrier Proteins/metabolism , Colorectal Neoplasms/genetics , Retinoblastoma Protein/physiology , Adenoma/genetics , Adenoma/pathology , Apoptosis , Cell Nucleus/pathology , Cell Survival/genetics , Colorectal Neoplasms/pathology , DNA Damage , DNA-Binding Proteins , Humans , Intestinal Mucosa , Retinoblastoma Protein/metabolism , Transcription Factors
20.
Biochem Soc Trans ; 33(Pt 4): 712-4, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16042581

ABSTRACT

Despite extensive research into the biology of CRC (colorectal cancer), and recent advances in surgical techniques and chemotherapy, CRC continues to be a major cause of death throughout the world. Therefore it is important to develop novel chemopreventive/chemotherapeutic agents for CRC. Cannabinoids are a class of compounds that are currently used in the treatment of chemotherapy-induced nausea and vomiting, and in the stimulation of appetite. However, there is accumulating evidence that they could also be useful for the inhibition of tumour cell growth by modulating key survival signalling pathways. The chemotherapeutic potential for plant-derived and endogenous cannabinoids in CRC therapy is reviewed.


Subject(s)
Antineoplastic Agents/therapeutic use , Cannabinoids/therapeutic use , Colorectal Neoplasms/drug therapy , Antiemetics/therapeutic use , Antineoplastic Agents/adverse effects , Humans , Nausea/prevention & control , Vomiting/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL
...