Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J. physiol. biochem ; 79(1): 193-204, feb. 2023. ilus
Article in English | IBECS | ID: ibc-215724

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is the deadliest of the common cancers. A major hallmark of PDAC is an abundant and dense fibrotic stroma, the result of a disproportionate deposition of extracellular matrix (ECM) proteins. Cancer-associated fibroblasts (CAFs) are the main mediators of PDAC desmoplasia. CAFs represent a heterogenous group of activated fibroblasts with different origins and activation mechanisms. microRNAs (miRNAs) are small non-coding RNAs with critical activity during tumour development and resistance to chemotherapy. Increasing evidence has revealed that miRNAs play a relevant role in the differentiation of normal fibroblasts into CAFs in PDAC. In this review, we discuss recent findings on the role of miRNAs in the activation of CAFs during the progression of PDAC and its response to therapy, as well as the potential role that PDAC-derived exosomal miRNAs may play in the activation of hepatic stellate cells (HSCs) and formation of liver metastasis. Since targeting of CAF activation may be a viable strategy for PDAC therapy, and miRNAs have emerged as potential therapeutic targets, understanding the biology underpinning miRNA-mediated tumour cell-CAF interactions is an important component in guiding rational approaches to treating this deadly disease. (AU)


Subject(s)
Humans , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Carcinoma, Pancreatic Ductal/metabolism , MicroRNAs/metabolism , Pancreatic Neoplasms/metabolism
2.
J. physiol. biochem ; 79(1): 223–234, feb. 2023. graf, ilus
Article in English | IBECS | ID: ibc-215727

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is characterised by a pro-inflammatory stroma and multi-faceted microenvironment that promotes and maintains tumorigenesis. However, the models used to test new and emerging therapies for PDAC have not increased in complexity to keep pace with our understanding of the human disease. Promising therapies that pass pre-clinical testing often fail in pancreatic cancer clinical trials. The objective of this study was to investigate whether changes in the drug-dosing regimen or the addition of cancer-associated fibroblasts (CAFs) to current existing models can impact the efficacy of chemotherapy drugs used in the clinic. Here, we reveal that gemcitabine and paclitaxel markedly reduce the viability of pancreatic cell lines, but not CAFs, when cultured in 2D. Following the use of an in vitro drug pulsing experiment, PDAC cell lines showed sensitivity to gemcitabine and paclitaxel. However, CAFs were less sensitive to pulsing with gemcitabine compared to their response to paclitaxel. We also identify that a 3D co-culture model of MIA PaCa-2 or PANC-1 with CAFs showed an increased chemoresistance to gemcitabine when compared to standard 2D mono-cultures a difference to paclitaxel which showed no measurable difference between the 2D and 3D models, suggesting a complex interaction between the drug in study and the cell type used. Changes to standard 2D mono-culture-based assays and implementation of 3D co-culture assays lend complexity to established models and could provide tools for identifying therapies that will match clinically the success observed with in vitro models, thereby aiding in the discovery of novel therapies. (AU)


Subject(s)
Humans , Cancer-Associated Fibroblasts/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Pancreatic Neoplasms/metabolism , Paclitaxel , Deoxycytidine , Cell Line, Tumor , Drug Evaluation, Preclinical , Tumor Microenvironment , Early Detection of Cancer
3.
J Physiol Biochem ; 79(1): 193-204, 2023 Feb.
Article in English | MEDLINE | ID: mdl-35767180

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is the deadliest of the common cancers. A major hallmark of PDAC is an abundant and dense fibrotic stroma, the result of a disproportionate deposition of extracellular matrix (ECM) proteins. Cancer-associated fibroblasts (CAFs) are the main mediators of PDAC desmoplasia. CAFs represent a heterogenous group of activated fibroblasts with different origins and activation mechanisms. microRNAs (miRNAs) are small non-coding RNAs with critical activity during tumour development and resistance to chemotherapy. Increasing evidence has revealed that miRNAs play a relevant role in the differentiation of normal fibroblasts into CAFs in PDAC. In this review, we discuss recent findings on the role of miRNAs in the activation of CAFs during the progression of PDAC and its response to therapy, as well as the potential role that PDAC-derived exosomal miRNAs may play in the activation of hepatic stellate cells (HSCs) and formation of liver metastasis. Since targeting of CAF activation may be a viable strategy for PDAC therapy, and miRNAs have emerged as potential therapeutic targets, understanding the biology underpinning miRNA-mediated tumour cell-CAF interactions is an important component in guiding rational approaches to treating this deadly disease.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Pancreatic Ductal , MicroRNAs , Pancreatic Neoplasms , Humans , MicroRNAs/metabolism , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Pancreatic Neoplasms/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Pancreatic Neoplasms
4.
J Physiol Biochem ; 79(1): 223-234, 2023 Feb.
Article in English | MEDLINE | ID: mdl-34865180

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is characterised by a pro-inflammatory stroma and multi-faceted microenvironment that promotes and maintains tumorigenesis. However, the models used to test new and emerging therapies for PDAC have not increased in complexity to keep pace with our understanding of the human disease. Promising therapies that pass pre-clinical testing often fail in pancreatic cancer clinical trials. The objective of this study was to investigate whether changes in the drug-dosing regimen or the addition of cancer-associated fibroblasts (CAFs) to current existing models can impact the efficacy of chemotherapy drugs used in the clinic. Here, we reveal that gemcitabine and paclitaxel markedly reduce the viability of pancreatic cell lines, but not CAFs, when cultured in 2D. Following the use of an in vitro drug pulsing experiment, PDAC cell lines showed sensitivity to gemcitabine and paclitaxel. However, CAFs were less sensitive to pulsing with gemcitabine compared to their response to paclitaxel. We also identify that a 3D co-culture model of MIA PaCa-2 or PANC-1 with CAFs showed an increased chemoresistance to gemcitabine when compared to standard 2D mono-cultures a difference to paclitaxel which showed no measurable difference between the 2D and 3D models, suggesting a complex interaction between the drug in study and the cell type used. Changes to standard 2D mono-culture-based assays and implementation of 3D co-culture assays lend complexity to established models and could provide tools for identifying therapies that will match clinically the success observed with in vitro models, thereby aiding in the discovery of novel therapies.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Deoxycytidine/metabolism , Deoxycytidine/therapeutic use , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Drug Evaluation, Preclinical , Cell Line, Tumor , Early Detection of Cancer , Pancreatic Neoplasms/metabolism , Gemcitabine , Carcinoma, Pancreatic Ductal/metabolism , Paclitaxel/metabolism , Paclitaxel/therapeutic use , Tumor Microenvironment , Pancreatic Neoplasms
5.
Cancer Res ; 80(13): 2861-2873, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32393661

ABSTRACT

Although fibrotic stroma forms an integral component of pancreatic diseases, whether fibroblasts programmed by different types of pancreatic diseases are phenotypically distinct remains unknown. Here, we show that fibroblasts isolated from patients with pancreatic ductal adenocarcinoma (PDAC), chronic pancreatitis (CP), periampullary tumors, and adjacent normal (NA) tissue (N = 34) have distinct mRNA and miRNA profiles. Compared with NA fibroblasts, PDAC-associated fibroblasts were generally less sensitive to an antifibrotic stimulus (NPPB) and more responsive to positive regulators of activation such as TGFß1 and WNT. Of the disease-associated fibroblasts examined, PDAC- and CP-derived fibroblasts shared greatest similarity, yet PDAC-associated fibroblasts expressed higher levels of tenascin C (TNC), a finding attributable to miR-137, a novel regulator of TNC. TNC protein and transcript levels were higher in PDAC tissue versus CP tissue and were associated with greater levels of stromal activation, and conditioned media from TNC-depleted PDAC-associated fibroblasts modestly increased both PDAC cell proliferation and PDAC cell migration, indicating that stromal TNC may have inhibitory effects on PDAC cells. Finally, circulating TNC levels were higher in patients with PDAC compared with CP. Our characterization of pancreatic fibroblast programming as disease-specific has consequences for therapeutic targeting and for the manner in which fibroblasts are used in research. SIGNIFICANCE: Primary fibroblasts derived from various types of pancreatic diseases possess and retain distinct molecular and functional characteristics in culture, providing a series of cellular models for treatment development and disease-specific research.


Subject(s)
Adenocarcinoma/pathology , Carcinoma, Pancreatic Ductal/pathology , Fibroblasts/pathology , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Pancreatic Neoplasms/pathology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Movement , Cell Proliferation , Fibroblasts/metabolism , Humans , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Tenascin/genetics , Tenascin/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Tumor Cells, Cultured , Pancreatic Neoplasms
6.
J Pathol ; 238(3): 423-33, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26497117

ABSTRACT

The cellular defence protein Nrf2 is a mediator of oncogenesis in pancreatic ductal adenocarcinoma (PDAC) and other cancers. However, the control of Nrf2 expression and activity in cancer is not fully understood. We previously reported the absence of Keap1, a pivotal regulator of Nrf2, in ∼70% of PDAC cases. Here we describe a novel mechanism whereby the epigenetic regulator UHRF1 suppresses Keap1 protein levels. UHRF1 expression was observed in 20% (5 of 25) of benign pancreatic ducts compared to 86% (114 of 132) of pancreatic tumours, and an inverse relationship between UHRF1 and Keap1 levels in PDAC tumours (n = 124) was apparent (p = 0.002). We also provide evidence that UHRF1-mediated regulation of the Nrf2 pathway contributes to the aggressive behaviour of PDAC. Depletion of UHRF1 from PDAC cells decreased growth and enhanced apoptosis and cell cycle arrest. UHRF1 depletion also led to reduced levels of Nrf2-regulated downstream proteins and was accompanied by heightened oxidative stress, in the form of lower glutathione levels and increased reactive oxygen species. Concomitant depletion of Keap1 and UHRF1 restored Nrf2 levels and reversed cell cycle arrest and the increase in reactive oxygen species. Mechanistically, depletion of UHRF1 reduced global and tumour suppressor promoter methylation in pancreatic cancer cell lines, and KEAP1 gene promoter methylation was reduced in one of three cell lines examined. Thus, methylation of the KEAP1 gene promoter may contribute to the suppression of Keap1 protein levels by UHRF1, although our data suggest that additional mechanisms need to be explored. Finally, we demonstrate that K-Ras drives UHRF1 expression, establishing a novel link between this oncogene and Nrf2-mediated cellular protection. Since UHRF1 over-expression occurs in other cancers, its ability to regulate the Keap1-Nrf2 pathway may be critically important to the malignant behaviour of these cancers.


Subject(s)
CCAAT-Enhancer-Binding Proteins/physiology , Intracellular Signaling Peptides and Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Pancreatic Neoplasms/etiology , CCAAT-Enhancer-Binding Proteins/deficiency , Carcinogenesis , Cell Cycle Checkpoints/physiology , Cell Transformation, Neoplastic/pathology , DNA Methylation/physiology , Humans , Kelch-Like ECH-Associated Protein 1 , Oxidative Stress/physiology , Pancreatic Neoplasms/pathology , Signal Transduction/physiology , Tumor Burden , Tumor Cells, Cultured , Ubiquitin-Protein Ligases
7.
Biochem Soc Trans ; 42(4): 747-51, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25109952

ABSTRACT

Human leukaemia cells have an often unique ability to either undergo apoptotic cell death mechanisms or, at other times, undergo proliferative expansion, sometimes to the same stimulus such as the pluripotent cytokine TNFα (tumour necrosis factor α). This potential for life/death switching helps us to understand the molecular signalling machinery that underlies these cellular processes. Furthermore, looking at the involvement of these switching signalling pathways that may be aberrant in leukaemia informs us of their importance in cancer tumorigenesis and how they may be targeted pharmacologically to treat various types of human leukaemias. Furthermore, these important pathways may play a crucial role in acquired chemotherapy resistance and should be studied further to overcome in the clinic many drug-resistant forms of blood cancers. In the present article, we uncover the relationship that exists in human leukaemia life/death switching between the anti-apoptotic pro-inflammatory transcription factor NF-κB (nuclear factor κB) and the cytoprotective antioxidant-responsive transcription factor Nrf2 (nuclear factor-erythroid 2-related factor 2). We also discuss recent findings that reveal a major role for Btk (Bruton's tyrosine kinase) in both lymphocytic and myeloid forms of human leukaemias and lymphomas.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Agammaglobulinaemia Tyrosine Kinase , Humans , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Protein-Tyrosine Kinases/metabolism , Transcription Factors/metabolism
8.
Cell Signal ; 25(1): 106-12, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22975686

ABSTRACT

Ibrutinib (previously known as PCI-32765) has recently shown encouraging clinical activity in chronic lymphocytic leukaemia (CLL) effecting cell death through inhibition of Bruton's tyrosine kinase (BTK). In this study we report for the first time that ibrutinib is cytotoxic to malignant plasma cells from patients with multiple myeloma (MM) and furthermore that treatment with ibrutinib significantly augments the cytotoxic activity of bortezomib and lenalidomide chemotherapies. We describe that the cytotoxicity of ibrutinib in MM is mediated via an inhibitory effect on the nuclear factor-κB (NF-κB) pathway. Specifically, ibrutinib blocks the phosphorylation of serine-536 of the p65 subunit of NF-κB, preventing its nuclear translocation, resulting in down-regulation of anti-apoptotic proteins Bcl-xL, FLIP(L) and survivin and culminating in caspase-mediated apoptosis within the malignant plasma cells. Taken together these data provide a platform for clinical trials of ibrutinib in myeloma and a rationale for its use in combination therapy, particularly with bortezomib.


Subject(s)
Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Cell Survival/drug effects , NF-kappa B/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrazines/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Thalidomide/analogs & derivatives , Adenine/analogs & derivatives , Agammaglobulinaemia Tyrosine Kinase , Amides/pharmacology , Bortezomib , Caspases/metabolism , Humans , I-kappa B Proteins/metabolism , Lenalidomide , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , NF-KappaB Inhibitor alpha , Nitriles/pharmacology , Phosphorylation , Piperidines , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , RNA, Messenger/metabolism , Signal Transduction , Thalidomide/pharmacology , Tumor Cells, Cultured
9.
Eur J Nutr ; 52(4): 1327-41, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22923034

ABSTRACT

PURPOSE: It is relatively unknown how different dietary components, in partnership, regulate gene expression linked to colon pathology. It has been suggested that the combination of various bioactive components present in a plant-based diet is crucial for their potential anticancer activities. This study employed a combinatorial chemopreventive strategy to investigate the impact of selenium and/or isothiocyanates on DNA methylation processes in colorectal carcinoma cell lines. METHODS: To gain insights into the epigenetic-mediated changes in gene expression in response to these dietary constituents cultured Caco-2 and HCT116 cells were exposed for up to 12 days to different concentrations of selenium methylselenocysteine and selenite (ranging from 0.2 to 5 µM) either alone or in combination with sulforaphane and iberin (ranging from 6 to 8 µM), and changes to gene-specific (p16(INK4A) and ESR1), global (LINE-1) methylation and DNMT expression were quantified using real-time PCR-based assays. RESULTS: No effects on the methylation of CpG islands in ESR1, p16(INK4A) or of LINE-1, a marker of global genomic methylation, were observed after exposure of Caco-2 and HCT116 cells to selenium or isothiocyanates. Only transient changes in DNMT mRNA expression, which occurred mostly in the treatment groups containing isothiocyanates, were observed, and these occurred only for specific DNMT transcripts and did not lead to the modification of the aberrant methylation status present in these cells. CONCLUSION: These data suggest that treatment for colon cancer cells with selenium and/or isothiocyanates, either individually or in combination does not impact abnormal methylation patterns of key genes involved in the complex multistep process of colon carcinogenesis in vitro.


Subject(s)
Antineoplastic Agents, Phytogenic/metabolism , Antioxidants/metabolism , Colorectal Neoplasms/metabolism , DNA Methylation , Epigenesis, Genetic , Isothiocyanates/metabolism , Selenium/metabolism , Anticarcinogenic Agents/metabolism , Caco-2 Cells , Cell Proliferation , Colorectal Neoplasms/prevention & control , CpG Islands , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Long Interspersed Nucleotide Elements , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , RNA, Messenger/metabolism , Sulfoxides , Thiocyanates/metabolism
10.
Biochim Biophys Acta ; 1823(10): 1914-24, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22820176

ABSTRACT

Currently, there is significant interest in the field of diet-gene interactions and the mechanisms by which food compounds regulate gene expression to modify cancer susceptibility. From a nutrition perspective, two key components potentially exert cancer chemopreventive effects: isothiocyanates (ITCs), present in cruciferous vegetables, and selenium (Se) which, as selenocysteine, is an integral part of selenoproteins. However, the role of these compounds in the expression of key selenoenzymes once the cancer process has been initiated still needs elucidation. Therefore, this investigation examined the effect of two forms of selenium, selenium-methylselenocysteine and sodium selenite, both individually and in combination with two ITCs, sulforaphane or iberin, on the expression of the two selenoenzymes, thioredoxin reductase 1 (TrxR1) and gastrointestinal glutathione peroxidase (GPx2), which are targets of ITCs, in Caco-2 cells. Co-treatment with both ITCs and Se induced expression of TrxR1 and GPx2 more than either compound alone. Moreover, pre-treatment of cells with ITC+Se enhanced cytoprotection against H(2)O(2)-induced cell death through a ROS-dependent mechanism. Furthermore, a single and double knockdown of TrxR1 and/or GPx2 suggested that both selenoproteins were responsible for protecting against H(2)O(2)-induced cell death. Together, these data shed new light on the mechanism of interactions between ITC and Se in which translational expression of the enhanced transcripts by the former is dependent on an adequate Se supply, resulting in a cooperative antioxidant protective effect against cell death.


Subject(s)
Cytoprotection/drug effects , Free Radicals/toxicity , Glutathione Peroxidase/biosynthesis , Isothiocyanates/pharmacology , Selenium/pharmacology , Thioredoxin Reductase 1/biosynthesis , Caco-2 Cells , Cell Death/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dietary Supplements , Enzyme Induction/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Glutathione Peroxidase/genetics , Humans , Hydrogen Peroxide/toxicity , Immunoblotting , NF-E2-Related Factor 2/metabolism , Protein Biosynthesis/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Thioredoxin Reductase 1/genetics , Time Factors , Up-Regulation/drug effects , Up-Regulation/genetics
11.
Cell Cycle ; 11(12): 2248-52, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22617388

ABSTRACT

Multiple myeloma (MM) is a progressive malignant disorder characterized by accumulation of plasma cells in the bone marrow. MM remains an incurable disease with a 5-y survival rate of approximately 40%. While clinical response rates to first line chemotherapeutics are high, disease relapse is inevitable, and occurs because a small fraction of the original myeloma cells appear to be resistant to treatment. Heme oxygenase-1 (HO-1) is an Nrf2 transcription factor-regulated gene that is commonly induced following oxidative stress and cellular injury, functioning to decrease oxidative stress and inflammatory responses, protecting against apoptosis and altering the cell cycle. We and others have highlighted the role of HO-1 in providing cellular protection against chemotherapeutic drugs in a number of cancer cells, which we have highlighted here in this Extra View. Furthermore, we explored the expression of HO-1 in multiple myeloma cells in response to the key anti-myeloma drugs bortezomib and lenalidomide. We show here for the first time that bortezomib increases HO-1 expression in a time- and concentration-dependent manner. Moreover, we also observe that HO-1 is increased in lenalidomide-resistant MM cell lines. Altogether, we highlight a possible role for HO-1 in basal and acquired chemoresistance in MM.


Subject(s)
Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Heme Oxygenase-1/metabolism , Pyrazines/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Boronic Acids/therapeutic use , Bortezomib , Drug Resistance, Neoplasm/drug effects , Heme Oxygenase-1/genetics , Humans , Lenalidomide , Multiple Myeloma/drug therapy , Multiple Myeloma/enzymology , Pyrazines/therapeutic use , Recurrence , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use , Tumor Cells, Cultured
12.
Proc Nutr Soc ; 71(2): 237-45, 2012 May.
Article in English | MEDLINE | ID: mdl-22391025

ABSTRACT

There is evidence from epidemiological studies suggesting that increased consumption of cruciferous vegetables may protect against specific cancers more effectively than total fruit and vegetable intake. These beneficial effects are attributed to the glucosinolate breakdown products, isothiocyanates (ITC). Similarly, selenium (Se) consumption has also been inversely associated with cancer risk and as an integral part of many selenoproteins may influence multiple pathways in the development of cancer. This paper will briefly review the current state of knowledge concerning the effect of Se and ITC in cancer development with a particular emphasis on its antioxidant properties, and will also address whether alterations in DNA methylation may be a potential mechanism whereby these dietary constituents protect against the carcinogenic process. Furthermore, we will discuss the advantages of combining ITC and Se to benefit from their complementary mechanisms of action to potentially protect against the alterations leading to neoplasia. Based on this review it may be concluded that an understanding of the impact of ITC and Se on aberrant DNA methylation in relation to factors modulating gene-specific and global methylation patterns, in addition to the effect of these food constituents as modulators of key selenoenzymes, such as gastrointestinal glutathione peroxidase-2 (GPx2) and thioredoxin reductase-1 (TrxR1), may provide insights into the potential synergy among various components of a plant-based diet that may counteract the genetic and epigenetic alterations that initiate and sustain neoplasia.


Subject(s)
Antioxidants/therapeutic use , DNA Methylation/drug effects , Diet , Epigenesis, Genetic , Isothiocyanates/therapeutic use , Neoplasms/prevention & control , Selenium/therapeutic use , Antioxidants/pharmacology , Brassicaceae/chemistry , DNA Methylation/genetics , Humans , Isothiocyanates/pharmacology , Neoplasms/genetics , Neoplasms/metabolism , Phytotherapy , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Selenium/pharmacology , Selenoproteins/genetics , Selenoproteins/metabolism
13.
Food Chem ; 133(2): 300-7, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-25683399

ABSTRACT

Dietary isothiocyanates and selenium are chemopreventive agents and potent inducers of antioxidant enzymes. It has been previously shown that sulforaphane and selenium have a synergistic effect on the upregulation of thioredoxin reductase-1 (TrxR-1) in human hepatoma HepG2 cells. In this paper, further evidence is presented to show that sulforaphane and selenium synergistically induce TrxR-1 expression in immortalised human hepatocytes. Sulforaphane was found to be more toxic toward hepatocytes than HepG2 cells with IC50=25.1 and 56.4 µM, respectively. Sulforaphane can protect against hydrogen peroxide-induced cell death and this protection was enhanced by co-treatment with selenium. Using siRNA to knock down TrxR-1 or Nrf2, sulforaphane (5 µM)-protected cell viability was reduced from 73% to 46% and 34%, respectively, suggesting that TrxR-1 is an important enzyme in protection against hydrogen peroxide-induced cell death. Sulforaphane-induced TrxR-1 expression was positively associated with significant levels of Nrf2 translocation into the nucleus, but co-treatment with selenium showed no significant increase in Nrf2 translocation. Moreover, MAPK (ERK, JNK and p38) and PI3K/Akt signalling pathways were found to play no significant role in sulforaphane-induced Nrf2 translocation into the nucleus. However, blocking ERK and JNK signalling pathways decreased sulforaphane-induced TrxR-1 mRNA by about 20%; whereas blocking p38 and PI3K/AKT increased TrxR-1 transcription. In summary, a combination of sulforaphane and selenium resulted in a synergistic upregulation of TrxR-1 that contributed to the enhanced protection against free radical-mediated oxidative damage in human hepatocytes.


Subject(s)
Hepatocytes/drug effects , Isothiocyanates/pharmacology , Selenium/pharmacology , Thioredoxin-Disulfide Reductase/metabolism , Cell Death/drug effects , Cell Line , Drug Synergism , Hep G2 Cells , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Hydrogen Peroxide/toxicity , Isothiocyanates/metabolism , Sulfoxides , Up-Regulation/drug effects
14.
Am J Cancer Res ; 2(1): 65-74, 2012.
Article in English | MEDLINE | ID: mdl-22206046

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression by binding to complementary sequences in mRNAs encoding downstream target genes. A large variety of cellular processes, including differentiation, development, apoptosis and cell cycle progression, are dependent on miRNA-mediated suppression of gene expression for their regulation. As such, it is unsurprising that these small RNA molecules are associated with signaling networks that are often altered in various diseases, including many blood cancers. One such network is the nuclear factor-κB (NF-κB) pathways that universally stimulate transcription of proteins which generally promote cell survival, inhibit apoptosis, allow cellular growth, induce angiogenesis and generate many pro-inflammatory responses. The NF-κB signalling pathway is often constitutively activated in blood cell cancers including myelodysplastic syndrome (MDS), acute myeloid leukaemia (AML), acute lymphocytic leukaemia (ALL), chronic myeloid leukaemia (CML), chronic lymphocytic leukaemia (CLL), lymphomas and in multiple myeloma (MM). This review focuses on the function of miRNAs that directly target NF-κB signaling cascade. Recent findings that connect this pathway through various miRNA families to human blood cancers are reviewed, and support for using miRNA-based therapy as a novel method to counteract this tumour-promoting signalling event is discussed.

15.
J Mol Diagn ; 13(2): 233-42, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21354059

ABSTRACT

Colon cancers are characterized by aberrant gene expression signatures associated with disease initiation and progression. Identification of aberrant gene expression associated with colon carcinogenesis has increased significantly with application of gene array technologies. Downstream processing of these data has been hindered by the lack of robust multiplexed gene quantitative technologies facilitating study of the identified multiple gene targets. The GenomeLab Genetic Analysis System presents a novel technology platform for quantitative multiplexed gene expression analysis. This report describes the custom design of a GeXP multiplexed assay used to assess expression profiles of 14 inflammatory gene targets in normal, polyp, and tumor tissue. Characteristic normal, polyp, and tumor tissue gene expression profiles were obtained. Statistical analysis confirmed comparable relative quantitation of gene expression using the GeXP, macroarray, and single-plex real-time polymerase chain reaction assays. GeXP assays may be usefully applied in clinical and regulatory studies of multiple gene targets. This system permits custom-design options for relative quantification of multiple gene target expression, simultaneously in a single reaction, using nanogram quantities of total RNA template. The system provides an approach to advance the study of multiple targets identified from gene array analysis with potential for characterizing gene expression signatures in clinical diagnostics.


Subject(s)
Colonic Neoplasms/genetics , Colonic Polyps/genetics , Gene Expression Profiling/methods , Neoplasms/genetics , Oligonucleotide Array Sequence Analysis/methods , Polymerase Chain Reaction/methods , Aged , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Biopsy , Colonic Neoplasms/pathology , Colonic Polyps/pathology , Female , Humans , Inflammation/genetics , Male , Middle Aged , Neoplasms/pathology
16.
Am J Physiol Gastrointest Liver Physiol ; 293(4): G682-91, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17656443

ABSTRACT

Obesity has recently become a focus of research to elucidate diet and lifestyle factors as important risk factors for colon cancer. Altered levels of insulin, leptin, and adiponectin have been identified as potential candidates increasing colon cancer risk within the prevailing obesogenic environment. There has been considerable research to characterize signaling via these hormones in the brain, liver, and adipose tissue; however, very little is known of their emerging role in peripheral signaling, particularly in epithelial tissues. This study profiles insulin, leptin, and adipokine receptors in the rat colon, revealing novel microanatomical location of these receptors and thereby supporting a potential role in regulating colonic tissue. Potential involvement of insulin, leptin, and adiponectin receptors in increased risk of colon cancer was investigated using Sprague-Dawley rats, either resistant or susceptible to diet-induced obesity. Regulation of insulin, leptin, and adiponectin receptors as a consequence of differing levels of adiposity was assessed regionally in the colon in response to treatment with the chemical carcinogen 1,2-dimethylhydrazine (DMH). However, significantly increased fat mass, increased levels of plasma insulin, leptin, and triglycerides, previously associated with an increased risk of colon cancer, were not associated with promotion of precancerous lesions in the experimental rats or deregulation of insulin, leptin, or adiponectin receptors. These findings do not support a direct link between the deregulation of insulin and adipokine levels observed in obese rats and an increased risk of colon carcinogenesis.


Subject(s)
1,2-Dimethylhydrazine/pharmacology , Colon/metabolism , Obesity/physiopathology , Receptor, Insulin/physiology , Receptors, Adiponectin/physiology , Receptors, Leptin/physiology , Adiponectin/metabolism , Adipose Tissue/physiopathology , Animals , Colon/drug effects , Male , Rats , Rats, Sprague-Dawley
17.
Biochim Biophys Acta ; 1762(9): 844-8, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16978845

ABSTRACT

Recently a mucosal pentraxin, Mptx, regulated by heme and calcium was reported in rat gut mucosal scrapings using microarray strategies. Considering the heterogeneity of gut mucosa scrapings and the widespread use of the rat as a model to study colon pathologies this study was undertaken to generate detailed mapping of micro-anatomical locations of Mptx and gain further insight into potential functions of this mucosal pentraxin in rat colon. Differential regulation was also examined in colon from different rat strains and rat models of oxidative stress and in pre-cancerous colon tissue. Different regional patterns of expression and discrete localisation in epithelial cells within transverse and distal colon crypts and an absence of expression in proximal colon were confirmed by regional PCR analysis and in situ hybridisation studies of colon. This study demonstrates that consideration of regional differences in Mptx gene expression and micro-anatomical location is necessary in the interpretation and deciphering of its regulation in colon.


Subject(s)
Acute-Phase Proteins/metabolism , Colon/metabolism , Gene Expression Regulation , Animals , Oxidative Stress , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...