Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cancer Metab ; 11(1): 18, 2023 Oct 19.
Article in English | MEDLINE | ID: mdl-37858256

ABSTRACT

BACKGROUND: To support proliferation and survival within a challenging microenvironment, cancer cells must reprogramme their metabolism. As such, targeting cancer cell metabolism is a promising therapeutic avenue. However, identifying tractable nodes of metabolic vulnerability in cancer cells is challenging due to their metabolic plasticity. Identification of effective treatment combinations to counter this is an active area of research. Aspirin has a well-established role in cancer prevention, particularly in colorectal cancer (CRC), although the mechanisms are not fully understood. METHODS: We generated a model to investigate the impact of long-term (52 weeks) aspirin exposure on CRC cells, which has allowed us comprehensively characterise the metabolic impact of long-term aspirin exposure (2-4mM for 52 weeks) using proteomics, Seahorse Extracellular Flux Analysis and Stable Isotope Labelling (SIL). Using this information, we were able to identify nodes of metabolic vulnerability for further targeting, investigating the impact of combining aspirin with metabolic inhibitors in vitro and in vivo. RESULTS: We show that aspirin regulates several enzymes and transporters of central carbon metabolism and results in a reduction in glutaminolysis and a concomitant increase in glucose metabolism, demonstrating reprogramming of nutrient utilisation. We show that aspirin causes likely compensatory changes that render the cells sensitive to the glutaminase 1 (GLS1) inhibitor-CB-839. Of note given the clinical interest, treatment with CB-839 alone had little effect on CRC cell growth or survival. However, in combination with aspirin, CB-839 inhibited CRC cell proliferation and induced apoptosis in vitro and, importantly, reduced crypt proliferation in Apcfl/fl mice in vivo. CONCLUSIONS: Together, these results show that aspirin leads to significant metabolic reprogramming in colorectal cancer cells and raises the possibility that aspirin could significantly increase the efficacy of metabolic cancer therapies in CRC.

2.
Explor Target Antitumor Ther ; 4(4): 600-615, 2023.
Article in English | MEDLINE | ID: mdl-37720350

ABSTRACT

Aspirin is a well-known nonsteroidal anti-inflammatory drug (NSAID) that has a recognized role in cancer prevention as well as evidence to support its use as an adjuvant for cancer treatment. Importantly there has been an increasing number of studies contributing to the mechanistic understanding of aspirins' anti-tumour effects and these studies continue to inform the potential clinical use of aspirin for both the prevention and treatment of cancer. This review focuses on the emerging role of aspirin as a regulator of metabolic reprogramming, an essential "hallmark of cancer" required to support the increased demand for biosynthetic intermediates needed for sustained proliferation. Cancer cells frequently undergo metabolic rewiring driven by oncogenic pathways such as hypoxia-inducible factor (HIF), wingless-related integration site (Wnt), mammalian target of rapamycin (mTOR), and nuclear factor kappa light chain enhancer of activated B cells (NF-κB), which supports the increased proliferative rate as tumours develop and progress. Reviewed here, cellular metabolic reprogramming has been identified as a key mechanism of action of aspirin and include the regulation of key metabolic drivers, the regulation of enzymes involved in glycolysis and glutaminolysis, and altered nutrient utilisation upon aspirin exposure. Importantly, as aspirin treatment exposes metabolic vulnerabilities in tumour cells, there is an opportunity for the use of aspirin in combination with specific metabolic inhibitors in particular, glutaminase (GLS) inhibitors currently in clinical trials such as telaglenastat (CB-839) and IACS-6274 for the treatment of colorectal and potentially other cancers. The increasing evidence that aspirin impacts metabolism in cancer cells suggests that aspirin could provide a simple, relatively safe, and cost-effective way to target this important hallmark of cancer. Excitingly, this review highlights a potential new role for aspirin in improving the efficacy of a new generation of metabolic inhibitors currently undergoing clinical investigation.

3.
Biofabrication ; 15(1)2022 Nov 02.
Article in English | MEDLINE | ID: mdl-36321254

ABSTRACT

We describe the development of a high-throughput bioprinted colorectal cancer (CRC) spheroid platform with high levels of automation, information content, and low cell number requirement. This is achieved via the formulation of a hydrogel bioink with a compressive Young's modulus that is commensurate with that of colonic tissue (1-3 kPa), which supports exponential growth of spheroids from a wide range of CRC cell lines. The resulting spheroids display tight cell-cell junctions, bioink matrix-cell interactions and necrotic hypoxic cores. By combining high content light microscopy imaging and processing with rapid multiwell plate bioprinting, dose-response profiles are generated from CRC spheroids challenged with oxaliplatin (OX) and fluorouracil (5FU), as well as radiotherapy. Bioprinted CRC spheroids are shown to exhibit high levels of chemoresistance relative to cell monolayers, and OX was found to be significantly less effective against tumour spheroids than in monolayer culture, when compared to 5FU.


Subject(s)
Bioprinting , Colorectal Neoplasms , Humans , Spheroids, Cellular , Bioprinting/methods , Fluorouracil , Cell Line , Oxaliplatin
4.
DNA Repair (Amst) ; 115: 103331, 2022 07.
Article in English | MEDLINE | ID: mdl-35468497

ABSTRACT

The proto-oncogene BCL-3 is upregulated in a subset of colorectal cancers (CRC), where it has been shown to enhance tumour cell survival. However, although increased expression correlates with poor patient prognosis, the role of BCL-3 in determining therapeutic response remains largely unknown. In this study, we use combined approaches in multiple cell lines and pre-clinical mouse models to investigate the function of BCL-3 in the DNA damage response. We show that suppression of BCL-3 increases γH2AX foci formation and decreases homologous recombination in CRC cells, resulting in reduced RAD51 foci number and increased sensitivity to PARP inhibition. Importantly, a similar phenotype is seen in Bcl3-/- mice, where Bcl3-/- mouse crypts also exhibit sensitivity to DNA damage with increased γH2AX foci compared to wild type mice. Additionally, Apc.Kras-mutant x Bcl3-/- mice are more sensitive to cisplatin chemotherapy compared to wild type mice. Taken together, our results identify BCL-3 as a regulator of the cellular response to DNA damage and suggests that elevated BCL-3 expression, as observed in CRC, could increase resistance of tumour cells to DNA damaging agents including radiotherapy. These findings offer a rationale for targeting BCL-3 in CRC as an adjunct to conventional therapies and suggest that BCL-3 expression in tumours could be a useful biomarker in stratification of rectal cancer patients for neo-adjuvant chemoradiotherapy.


Subject(s)
Colorectal Neoplasms , Animals , Cell Line, Tumor , Cisplatin/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , DNA Damage , Homologous Recombination , Humans , Mice
5.
Carcinogenesis ; 41(3): 249-256, 2020 05 14.
Article in English | MEDLINE | ID: mdl-31930327

ABSTRACT

With its identification as a proto-oncogene in chronic lymphocytic leukaemia and central role in regulating NF-κB signalling, it is perhaps not surprising that there have been an increasing number of studies in recent years investigating the role of BCL-3 (B-Cell Chronic Lymphocytic Leukaemia/Lymphoma-3) in a wide range of human cancers. Importantly, this work has begun to shed light on our mechanistic understanding of the function of BCL-3 in tumour promotion and progression. Here, we summarize the current understanding of BCL-3 function in relation to the characteristics or traits associated with tumourigenesis, termed 'Hallmarks of Cancer'. With the focus on colorectal cancer, a major cause of cancer related mortality in the UK, we describe the evidence that potentially explains why increased BCL-3 expression is associated with poor prognosis in colorectal cancer. As well as promoting tumour cell proliferation, survival, invasion and metastasis, a key emerging function of this proto-oncogene is the regulation of the tumour response to inflammation. We suggest that BCL-3 represents an exciting new route for targeting the Hallmarks of Cancer; in particular by limiting the impact of the enabling hallmarks of tumour promoting inflammation and cell plasticity. As BCL-3 has been reported to promote the stem-like potential of cancer cells, we suggest that targeting BCL-3 could increase the tumour response to conventional treatment, reduce the chance of relapse and hence improve the prognosis for cancer patients.


Subject(s)
B-Cell Lymphoma 3 Protein/genetics , Carcinogenesis/genetics , Colorectal Neoplasms/genetics , Neoplasm Recurrence, Local/genetics , Cell Proliferation/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/genetics , Humans , NF-kappa B/genetics , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Prognosis , Proto-Oncogene Mas , Signal Transduction/genetics
6.
Dis Model Mech ; 12(3)2019 03 04.
Article in English | MEDLINE | ID: mdl-30792270

ABSTRACT

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.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Wnt Signaling Pathway , B-Cell Lymphoma 3 Protein , Cell Line, Tumor , Cell Nucleus/metabolism , Gene Expression Regulation, Neoplastic , Genes, Reporter , Humans , Lymphoid Enhancer-Binding Factor 1/metabolism , Phenotype , Protein Binding , Protein Transport , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , TCF Transcription Factors/metabolism , Transcription Factors/genetics , beta Catenin/metabolism
7.
EMBO Mol Med ; 10(11)2018 11.
Article in English | MEDLINE | ID: mdl-30143543

ABSTRACT

Hypoxia is a hallmark of solid tumours and a key physiological feature distinguishing cancer from normal tissue. However, a major challenge remains in identifying tractable molecular targets that hypoxic cancer cells depend on for survival. Here, we used SILAC-based proteomics to identify the orphan G protein-coupled receptor GPRC5A as a novel hypoxia-induced protein that functions to protect cancer cells from apoptosis during oxygen deprivation. Using genetic approaches in vitro and in vivo, we reveal HIFs as direct activators of GPRC5A transcription. Furthermore, we find that GPRC5A is upregulated in the colonic epithelium of patients with mesenteric ischaemia, and in colorectal cancers high GPRC5A correlates with hypoxia gene signatures and poor clinical outcomes. Mechanistically, we show that GPRC5A enables hypoxic cell survival by activating the Hippo pathway effector YAP and its anti-apoptotic target gene BCL2L1 Importantly, we show that the apoptosis induced by GPRC5A depletion in hypoxia can be rescued by constitutively active YAP. Our study identifies a novel HIF-GPRC5A-YAP axis as a critical mediator of the hypoxia-induced adaptive response and a potential target for cancer therapy.


Subject(s)
Adaptation, Physiological , Adaptor Proteins, Signal Transducing/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplasms/pathology , Phosphoproteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Adaptation, Physiological/drug effects , Animals , Antigens, Neoplasm/metabolism , Carbonic Anhydrase IX/metabolism , Cell Hypoxia/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Doxycycline/pharmacology , Humans , Mice, Inbred C57BL , Models, Biological , Neoplasms/genetics , Signal Transduction/drug effects , Transcription Factors , Transcription, Genetic/drug effects , YAP-Signaling Proteins , Zebrafish
8.
Gut ; 65(7): 1151-64, 2016 07.
Article in English | MEDLINE | ID: mdl-26033966

ABSTRACT

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.


Subject(s)
Colorectal Neoplasms/chemistry , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/metabolism , Signal Transduction , Transcription Factors/analysis , Transcription Factors/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis , B-Cell Lymphoma 3 Protein , Cell Proliferation , Cell Survival/drug effects , Colon/chemistry , Colorectal Neoplasms/pathology , HCT116 Cells , Humans , Mesalamine/pharmacology , Mice , Mice, Nude , NF-kappa B/analysis , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/pharmacology , Rectum/chemistry , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/genetics , Tumor Burden
9.
EMBO J ; 32(13): 1903-16, 2013 Jul 03.
Article in English | MEDLINE | ID: mdl-23736261

ABSTRACT

The Wnt/ß-catenin signalling and autophagy pathways each play important roles during development, adult tissue homeostasis and tumorigenesis. Here we identify the Wnt/ß-catenin signalling pathway as a negative regulator of both basal and stress-induced autophagy. Manipulation of ß-catenin expression levels in vitro and in vivo revealed that ß-catenin suppresses autophagosome formation and directly represses p62/SQSTM1 (encoding the autophagy adaptor p62) via TCF4. Furthermore, we show that during nutrient deprivation ß-catenin is selectively degraded via the formation of a ß-catenin-LC3 complex, attenuating ß-catenin/TCF-driven transcription and proliferation to favour adaptation during metabolic stress. Formation of the ß-catenin-LC3 complex is mediated by a W/YXXI/L motif and LC3-interacting region (LIR) in ß-catenin, which is required for interaction with LC3 and non-proteasomal degradation of ß-catenin. Thus, Wnt/ß-catenin represses autophagy and p62 expression, while ß-catenin is itself targeted for autophagic clearance in autolysosomes upon autophagy induction. These findings reveal a regulatory feedback mechanism that place ß-catenin at a key cellular integration point coordinating proliferation with autophagy, with implications for targeting these pathways for cancer therapy.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Colonic Neoplasms/pathology , Lysosomes/metabolism , Transcription Factors/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Blotting, Western , Chromatin Immunoprecipitation , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Leukocyte Immunoglobulin-like Receptor B1 , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Phagosomes/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequestosome-1 Protein , Transcription Factor 4 , Transcription Factors/genetics , Tumor Cells, Cultured , Wnt Proteins/genetics , beta Catenin/antagonists & inhibitors , beta Catenin/genetics
10.
Carcinogenesis ; 34(5): 1150-7, 2013 May.
Article in English | MEDLINE | ID: mdl-23349017

ABSTRACT

Cyclooxygenase-2 is overexpressed in the majority of colorectal tumours leading to elevated levels of prostaglandin E2 (PGE2), promoting many hallmarks of cancer. Importantly, PGE2 is reported to enhance Wnt/ß-catenin signalling in colorectal carcinoma cells and in normal haematopoietic stem cells where it promotes stem cell function. Although Wnt signalling plays a crucial role in intestinal stem cells, the relationship between PGE2 and intestinal stem cells is unclear. Given that the key intestinal cancer stem cell marker LGR5 (leucine-rich G-protein coupled receptor 5) is a Wnt target and PGE2 enhances Wnt signalling, the focus of this study was to investigate whether PGE2 regulated LGR5 expression in colorectal adenoma cells and whether LGR5 was important for tumour cell survival. PGE2 upregulated LGR5 protein in adenoma (RG/C2) and carcinoma (DLD-1) cell lines. LGR5 knockdown induced cell death in RG/C2 and AA/C1 adenoma cells, suggesting that LGR5 has an important survival-promoting role in adenoma cells. Indeed, we detected LGR5 protein expression in 4 of 4 human adenoma cell lines. Furthermore, LGR5 small interfering RNA inhibited the survival-promoting effects of PGE2 in RG/C2, suggesting that PGE2 promotes adenoma cell survival, at least in part, by increasing LGR5 expression. These studies, therefore, show the first link between PGE2 and LGR5 in human colorectal adenoma and carcinoma cells and demonstrate a survival-promoting role of LGR5. As non-steroidal anti-inflammatory drugs (NSAIDs) cause adenomas to regress in FAP patients, these studies could have important implications for the mechanism by which NSAIDs are chemopreventive, as lowering PGE2 levels could reduce LGR5 expression and survival of LGR5(+) adenoma stem cells.


Subject(s)
Adenoma/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Colorectal Neoplasms/metabolism , Dinoprostone/metabolism , Neoplastic Stem Cells/metabolism , Receptors, G-Protein-Coupled/metabolism , Adenoma/genetics , Adenoma/pathology , Carcinoma/drug therapy , Carcinoma/genetics , Carcinoma/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Dinoprostone/genetics , Female , Humans , Middle Aged , Neoplastic Stem Cells/drug effects , Receptors, G-Protein-Coupled/genetics , Up-Regulation , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics , beta Catenin/genetics , beta Catenin/metabolism
11.
Int J Oncol ; 37(1): 187-93, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20514410

ABSTRACT

Despite recent advances in understanding colorectal tumour biology, there is still a need to improve the 5-year survival rate of patients with colorectal cancer as approximately 40% of patients presenting with advanced disease will remain resistant to therapy. One of the major contributing factors in resistance to therapy is the failure of colorectal tumour cells to undergo apoptosis. Hence there is an urgent need to develop novel therapeutic approaches that can target apoptosis-resistant cells. To this end, we investigated the potential efficacy of the endogenous cannabinoid anandamide to induce cell death in apoptosis-resistant colon cancer cells. Here, for the first time, we show that anandamide can induce cell death in the apoptosis-resistant HCT116 Bax-/- colorectal cell line. Importantly, we provide direct genetic evidence that this induction of cell death is dependent on COX-2 expression. Interestingly, increased COX-2 expression also sensitised the SW480 colorectal cancer cell line (low endogenous COX-2) to anandamide-induced death, whereas COX-2 suppression by RNAi inhibited anandamide-induced cell death in the HCA7 colorectal cancer cell line (high endogenous COX-2 expression). This COX-2-dependent death was independent of cannabinoid receptor engagement (CB1 or CB2), and not a direct consequence of reactive oxygen species (ROS) formation. This study demonstrates a novel utilisation for COX-2 expression, targeting apoptotic defective colorectal cancer cells for destruction by anandamide. As COX-2 is not expressed in the normal colorectal epithelium, but highly expressed in colorectal tumours and apoptosis resistance contributes to treatment failure, these data suggest that anandamide has the potential to be an effective therapeutic in colorectal cancer.


Subject(s)
Apoptosis/genetics , Arachidonic Acids/pharmacology , Carcinoma/pathology , Colonic Neoplasms/pathology , Cyclooxygenase 2/physiology , Polyunsaturated Alkamides/pharmacology , Apoptosis/drug effects , Cannabinoid Receptor Modulators/pharmacology , Carcinoma/genetics , Cell Death/drug effects , Cell Death/genetics , Cell Line, Tumor , Colonic Neoplasms/genetics , Cyclooxygenase 2/genetics , Cyclooxygenase 2 Inhibitors/pharmacology , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm/genetics , Endocannabinoids , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , HT29 Cells , Humans , RNA, Small Interfering/pharmacology , Up-Regulation/drug effects , Up-Regulation/genetics , bcl-2-Associated X Protein/genetics
12.
Carcinogenesis ; 29(4): 849-57, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18204076

ABSTRACT

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.


Subject(s)
Colonic Neoplasms/genetics , Colorectal Neoplasms/genetics , DNA-Binding Proteins/genetics , NF-kappa B/metabolism , RNA, Small Interfering/genetics , Transcription Factors/genetics , Adenoma/genetics , Adenoma/pathology , Carcinoma/genetics , Carcinoma/pathology , Cell Line, Tumor , Cell Survival/genetics , Colonic Neoplasms/pathology , Colorectal Neoplasms/pathology , Disease Progression , Humans , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Cells, Cultured
13.
Int J Oncol ; 29(5): 1279-86, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17016662

ABSTRACT

The insulin-like growth factor binding protein 3 (IGFBP-3) is the major circulating IGF binding protein, its function regulated by proteolytic cleavage. The fragments generated have recently been suggested to have IGF-independent biological activity. We have previously established that IGFBP-3 can potentiate apoptosis in colorectal epithelial cells, although its use as a therapeutic reagent may be limited by the fact that it is cleaved in the circulation. Therefore the aim of these experiments was to determine whether the 16-kDa proteolytic fragment (1-95IGFBP-3) would have IGF-independent pro-apoptotic activity in human colonic carcinoma derived cells. We report that the enforced expression of 1-95IGFBP-3 increased the induction of apoptosis by the naturally occurring short chain fatty acid sodium butyrate (NaBt) in the IGF non-responsive HT29 human colorectal carcinoma cell line. Furthermore, the addition of condition medium containing the secreted 1-95IGFBP-3 was as effective as the intact IGFBP-3 protein at potentiating apoptosis. Although not associated with changes in Bcl-2, Bcl-XL, Bax, Bad or Bak expression levels, we report that the expression of the pro-apoptotic 1-95IGFBP-3 fragment is associated with the inhibition of TNFalpha-induced NF-kappaB activity, similar to that reported for the full length IGFBP-3 protein. These results suggest that the 16-kDa 1-95IGFBP-3 fragment is as effective as an intact recombinant protein when used in combination with apoptosis inducing agents, and due to its relative stability in the circulation, it may be important for use as an adjuvant in the treatment of colorectal cancer.


Subject(s)
Apoptosis , Carcinoma/drug therapy , Colonic Neoplasms/drug therapy , Insulin-Like Growth Factor Binding Protein 3/therapeutic use , Peptide Fragments/therapeutic use , Cell Line, Tumor , Chemotherapy, Adjuvant , Culture Media, Conditioned/pharmacology , Humans , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 3/metabolism , NF-kappa B/antagonists & inhibitors , Peptide Fragments/genetics , Peptide Fragments/metabolism , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sodium Oxybate/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
14.
Int J Cancer ; 106(3): 364-71, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12845674

ABSTRACT

Although the retinoblastoma susceptibility gene RB1 is inactivated in a wide range of human tumours, overexpression in colonic carcinomas has been linked to the antiapoptotic function of the protein. In the current study we show that the Retinoblastoma susceptibility protein (Rb) protein interacts with Bag-1, an apoptotic regulator, in human colonic adenoma- and carcinoma-derived cell lines. Coimmunoprecipitation demonstrated that endogenous Rb and Bag-1 interact in both adenoma- and carcinoma-derived cell lines. The specificity of the interaction was demonstrated by expression of human Papillomavirus E7 oncoprotein, an inhibitor of Rb protein interactions, which disrupted the Rb/Bag-1 complex. We report that Bag-1 is predominantly localised in the nucleus of colorectal adenoma- and carcinoma-derived epithelial cells. Disruption of the Rb/Bag-1 complex through expression of E7 changes the subcellular distribution of Bag-1, decreasing nuclear localised Bag-1. Our work establishes that the Rb protein interacts with the Bag-1 apoptotic regulator protein, and introduces a novel function for Rb, involving modulation of the subcellular localisation of Bag-1 in human colonic epithelial cells.


Subject(s)
Adenocarcinoma/metabolism , Adenoma/metabolism , Carrier Proteins/metabolism , Colorectal Neoplasms/metabolism , Retinoblastoma Protein/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenoma/genetics , Adenoma/pathology , Blotting, Western , Carrier Proteins/genetics , Cell Death , Cell Nucleus/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA-Binding Proteins , Epithelial Cells , Genetic Vectors , Glutathione Transferase/metabolism , Humans , Microscopy, Confocal , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins , Precipitin Tests , Protein Transport , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinoblastoma Protein/genetics , Saccharomyces cerevisiae , Subcellular Fractions , Transcription Factors , Tumor Cells, Cultured , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...