Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nutr Cancer ; 69(2): 299-306, 2017.
Article in English | MEDLINE | ID: mdl-28094560

ABSTRACT

INTRODUCTION: Freeze-dried black raspberries (BRBs) elicit chemopreventive effects against colorectal cancer in humans and in rodents. The objective of this study was to investigate potential BRB-caused metabolite changes using wild-type (WT) C57BL/6 mice. METHODS AND RESULTS: WT mice were fed either control diet or control diet supplemented with 5% BRBs for 8 wk. A nontargeted metabolomic analysis was conducted on colonic mucosa, liver, and fecal specimens collected from both diet groups. BRBs significantly changed the levels of 41 colonic mucosa metabolites, 40 liver metabolites, and 34 fecal metabolites compared to control diet-fed mice. BRBs reduced 34 lipid metabolites in colonic mucosa and increased levels of amino acids in liver. One metabolite, 3-[3-(sulfooxy) phenyl] propanoic acid, might be a useful biomarker of BRB consumption. In addition, BRB powder was found to contain 30-fold higher levels of linolenate compared to control diets. Consistently, multiple omega-3 polyunsaturated fatty acids (ω-3 PUFAs), including stearidonate, docosapentaenoate (ω-3 DPA), eicosapentaenoate (EPA), and docosahexaenoate (DHA), were significantly elevated in livers of BRB-fed mice. CONCLUSION: The data from the current study suggest that BRBs produce systemic metabolite changes in multiple tissue matrices, supporting our hypothesis that BRBs may serve as both a chemopreventive agent and a beneficial dietary supplement.


Subject(s)
Amino Acids/metabolism , Anticarcinogenic Agents/pharmacology , Colon/metabolism , Lipid Metabolism , Liver/metabolism , Rubus , Animals , Benzoates/metabolism , Dietary Supplements , Fatty Acids, Omega-3/metabolism , Feces , Intestinal Mucosa/metabolism , Mice, Inbred C57BL
2.
Carcinogenesis ; 36(10): 1245-53, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26246425

ABSTRACT

Freeze-dried black raspberries (BRBs) have demonstrated chemopreventive effects in a dietary intervention trial with human colorectal cancer patients. The aim of this study was to investigate BRB-caused metabolite changes using the Apc(Min/+) mouse as a model of human colorectal cancer. Wild-type (WT) mice were fed control diet, and Apc(Min/+) mice were fed either control diet or control diet supplemented with 5% BRBs for 8 weeks. Colonic and intestinal polyp size and number were measured. A non-targeted metabolomic analysis was conducted on colonic mucosa, liver and fecal specimens. Eight weeks of BRB treatment significantly decreased intestinal and colonic polyp number and size in Apc(Min/+) mice. The apc gene mutation significantly changed 52 metabolites in colonic mucosa associated with increased amino acid and decreased lipid metabolites, as well as 39 liver and 8 fecal metabolites. BRBs significantly reversed 23 apc-regulated metabolites, including 13 colonic mucosa, 8 liver and 2 fecal metabolites that were involved in amino acid, glutathione, lipid and nucleotide metabolism. Of these, changes in eight metabolites were linearly correlated with decreased colonic polyp number and size in BRB-treated Apc(Min/+) mice. Elevated levels of putrescine and linolenate in Apc(Min/+) mice were significantly decreased by BRBs. Ornithine decarboxylase expression, the key enzyme in putrescine generation, was fully suppressed by BRBs. These results suggest that BRBs produced beneficial effects against colonic adenoma development in Apc(Min/+) mice and modulated multiple metabolic pathways. The metabolite changes produced by BRBs might potentially reflect the BRB-mediated chemopreventive effects in colorectal cancer patients.


Subject(s)
Adenoma/diet therapy , Adenomatous Polyposis Coli Protein/genetics , Colorectal Neoplasms/diet therapy , Fruit , Rubus , Adenoma/metabolism , Adenoma/pathology , Animals , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Disease Models, Animal , Gene Expression Regulation, Neoplastic/drug effects , Humans , Intestinal Mucosa/drug effects , Mice , Mice, Transgenic , Putrescine/biosynthesis , alpha-Linolenic Acid/biosynthesis
3.
Mol Carcinog ; 54(10): 1096-109, 2015 Oct.
Article in English | MEDLINE | ID: mdl-24838344

ABSTRACT

Colorectal cancer, a leading cause of cancer death, has been linked to inflammation and obesity. Berberine, an isoquinoline alkaloid, possesses anti-inflammatory, anti-diabetes and anti-tumor properties. In the azoxymethane initiated and dextran sulfate sodium (AOM/DSS) promoted colorectal carcinogenesis mouse model, berberine treated mice showed a 60% reduction in tumor number (P = 0.009), a 48% reduction in tumors <2 mm, (P = 0.05); 94% reduction in tumors 2-4 mm, (P = 0.001), and 100% reduction in tumors >4 mm (P = 0.02) compared to vehicle treated mice. Berberine also decreased AOM/DSS induced Ki-67 and COX-2 expression. In vitro analysis showed that in addition to its anti-proliferation activity, berberine also induced apoptosis in colorectal cancer cell lines. Berberine activated AMP-activated protein kinase (AMPK), a major regulator of metabolic pathways, and inhibited mammalian target of rapamycin (mTOR), a downstream target of AMPK. Furthermore, 4E-binding protein-1 and p70 ribosomal S6 kinases, downstream targets of mTOR, were down regulated by berberine treatment. Berberine did not affect Liver kinase B1 (LKB1) activity or the mitogen-activated protein kinase pathway. Berberine inhibited Nuclear Factor kappa-B (NF-κB) activity, reduced the expression of cyclin D1 and survivin, induced phosphorylation of p53 and increased caspase-3 cleavage in vitro. Berberine inhibition of mTOR activity and p53 phosphorylation was found to be AMPK dependent, while inhibition NF-κB was AMPK independent. In vivo, berberine also activated AMPK, inhibited mTOR and p65 phosphorylation and activated caspase-3 cleavage. Our data suggests that berberine suppresses colon epithelial proliferation and tumorigenesis via AMPK dependent inhibition of mTOR activity and AMPK independent inhibition of NF-κB.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Berberine/pharmacology , Carcinogenesis/drug effects , Colon/drug effects , Colorectal Neoplasms/drug therapy , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Azoxymethane/pharmacology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Caspase 3/metabolism , Cell Line, Tumor , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/metabolism , Cyclin D1/metabolism , Cyclooxygenase 2/metabolism , Female , HCT116 Cells , Humans , Mice , NF-kappa B/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , eIF-2 Kinase/metabolism
4.
Mol Cell Biochem ; 406(1-2): 63-73, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25912550

ABSTRACT

Cryptotanshinone (CPT) is a natural compound extracted from herbal medicine that has been previously shown to possess antitumor properties in various types of human cancer cells. In the present study, we examined the potential role of CPT in the treatment of colorectal cancer. Using SW480, HCT116, and LOVO colorectal cancer cell lines, the effects of CPT on cell viability, apoptosis, and tumorigenicity were evaluated. The results showed that CPT significantly inhibited the growth and viability of SW480, HCT116, and LOVO cell lines by inducing apoptosis and prevented anchorage dependent growth on agar. In addition, CPT inhibited the activation of Signal transducer and activator of transcription 3 (Stat3) pathways in colorectal cancer cells. Stat3 is a transcription factor that mediates the expression of various genes associated with many cellular processes, such as inflammation and cell growth, and has been shown to promote several cancer types, including colorectal cancer. These findings indicate that CPT may be a potential candidate for the treatment and prevention of colorectal cancer in part by inhibiting the activation of Stat3.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Phenanthrenes/pharmacology , STAT3 Transcription Factor/metabolism , Apoptosis , Cell Survival/drug effects , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Drug Screening Assays, Antitumor , HCT116 Cells , Humans , Inhibitor of Apoptosis Proteins/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Survivin
5.
Carcinogenesis ; 35(7): 1469-80, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24413684

ABSTRACT

PDCD4 is a tumor suppressor induced by apoptotic stimuli that regulates both translation and transcription. Previously, we showed that overexpression of PDCD4 leads to decreased anchorage-independent growth in glioblastoma (GBM)-derived cell lines and decreased tumor growth in a GBM xenograft model. In inflammatory cells, PDCD4 stimulates tumor necrosis factor-induced activation of the transcription factor NF-κB, an oncogenic driver in many cancer sites. However, the effect of PDCD4 on NF-κB transcriptional activity in most cancers including GBM is still unknown. We studied the effect of PDCD4 on NF-κB-dependent transcriptional activity in GBM by stably overexpressing PDCD4 in U251 and LN229 cells. Stable PDCD4 expression inhibits NF-κB transcriptional activation measured by a luciferase reporter. The molecular mechanism by which PDCD4 inhibits NF-κB transcriptional activation does not involve inhibited expression of NF-κB p65 or p50 proteins. PDCD4 does not inhibit pathways upstream of NF-κB including the activation of IKKα and IKKß kinases or degradation of IκBα, events needed for nuclear transport of p65 and p50. PDCD4 overexpression does inhibit localization of p65 but not p50 in the nucleus. PDCD4 protein interacts preferentially with p65 protein as shown by co-immunoprecipitation and confocal imaging. PDCD4 overexpression inhibits the mRNA expression of two NF-κB target genes in a p65-dependent manner. These results suggest that PDCD4 can significantly inhibit NF-κB activity in GBM cells by a mechanism that involves direct or indirect protein-protein interaction independent of the expected mRNA-selective translational inhibition. These findings offer novel opportunities for NF-κB-targeted interventions to prevent or treat cancer.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , NF-kappa B p50 Subunit/metabolism , RNA-Binding Proteins/genetics , Transcription Factor RelA/metabolism , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Blotting, Western , Cell Movement , Cell Proliferation , Chromatin Immunoprecipitation , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Immunoprecipitation , NF-kappa B p50 Subunit/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor RelA/genetics , Transcription, Genetic , Tumor Cells, Cultured
6.
Carcinogenesis ; 35(12): 2778-86, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25280562

ABSTRACT

Sporadic and non-hereditary mutations account for the majority of colorectal cancers (CRC). After the loss of adenomatous polyposis coli (APC) function and activation of the ß-catenin/LEF signaling pathway, activating mutations in Kras are major drivers of sporadic CRC. Preventing the outgrowth of cells that develop sporadic mutations will decrease CRC. Resveratrol, a naturally occurring polyphenolic compound has anti-inflammatory, anti-oxidant and anti-cancer activities. We used a genetically engineered mouse model for sporadic CRC where the APC locus is knocked out and Kras is activated specifically in the distal colon to determine the effects of resveratrol on preventing and treating CRC. Feeding mice a diet supplemented with 150 or 300 ppm resveratrol (105 and 210mg daily human equivalent dose, respectively) before tumors were visible by colonoscopy resulted in a 60% inhibition of tumor production. In the 40% of mice that did develop tumors Kras expression was lost in the tumors. In a therapeutic assay where tumors were allowed to develop prior to treatment, feeding tumor bearing mice with resveratrol resulted in a complete remission in 33% of the mice and a 97% decrease in tumor size in the remaining mice. Analysis of miRNA expression in non-tumoral and tumoral colonic tissue of resveratrol treated mice showed an increased expression of miR-96, a miRNA previously shown to regulate Kras translation. These data indicate that resveratrol can prevent the formation and growth of colorectal tumors by downregulating Kras expression.


Subject(s)
Adenomatous Polyposis Coli Protein/physiology , Anticarcinogenic Agents/therapeutic use , Cell Transformation, Neoplastic/drug effects , Colorectal Neoplasms/prevention & control , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Stilbenes/therapeutic use , Animals , Blotting, Western , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Colorectal Neoplasms/etiology , Colorectal Neoplasms/pathology , Disease Models, Animal , Female , Humans , Immunoenzyme Techniques , Male , Mice , Mice, Knockout , Mutation/genetics , Proto-Oncogene Proteins p21(ras)/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Resveratrol , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
7.
Carcinogenesis ; 35(5): 1177-84, 2014 May.
Article in English | MEDLINE | ID: mdl-24503444

ABSTRACT

Sulfiredoxin (Srx), the exclusive enzyme that reduces the hyperoxidized inactive form of peroxiredoxins (Prxs), has been found highly expressed in several types of human skin cancer. To determine whether Srx contributed to skin tumorigenesis in vivo, Srx null mice were generated on an FVB background. Mouse skin tumorigenesis was induced by a 7,12-dimethylbenz[α]anthracene/12-O-tetradecanoylphorbol-13-acetate (DMBA/TPA) protocol. We found that the number, volume and size of papillomas in Srx(-/-) mice were significantly fewer compared with either wild-type (Wt) or heterozygous (Het) siblings. Histopathological analysis revealed more apoptotic cells in tumors from Srx(-/-) mice. Mechanistic studies in cell culture revealed that Srx was stimulated by TPA in a redox-independent manner. This effect was mediated transcriptionally through the activation of mitogen-activated protein kinase and Jun-N-terminal kinase. We also demonstrated that Srx was capable of reducing hyperoxidized Prxs to facilitate cell survival under oxidative stress conditions. These findings suggested that loss of Srx protected mice, at least partially, from DMBA/TPA-induced skin tumorigenesis. Therefore, Srx has an oncogenic role in skin tumorigenesis and targeting Srx may provide novel strategies for skin cancer prevention or treatment.


Subject(s)
Cell Transformation, Neoplastic/genetics , Oxidoreductases Acting on Sulfur Group Donors/genetics , Skin Neoplasms/genetics , Skin/metabolism , Skin/pathology , 9,10-Dimethyl-1,2-benzanthracene/adverse effects , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line , Cell Proliferation , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Oxidation-Reduction , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Skin/drug effects , Skin Neoplasms/chemically induced , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tetradecanoylphorbol Acetate/adverse effects , Transcriptional Activation/drug effects
8.
Stem Cells ; 31(8): 1454-66, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23592398

ABSTRACT

Pancreatic cancer is the fourth leading cause of cancer-related mortality in the world. Pancreatic cancer can be localized, locally advanced, or metastatic. The median 1- and 5-year survival rates are 25% and 6%, respectively. Epigenetic modifications such as DNA methylation play a significant role during both normal human development and cancer progression. To investigate epigenetic regulation of genes in the tumor-initiating population of pancreatic cancer cells, which are also termed cancer stem cells (CSCs), we conducted epigenetic arrays in PANC1 and HPAC pancreatic cancer cell lines and compared the global DNA methylation status of CpG promoters in invasive cells, demonstrated to be CSCs, to their noninvasive counterparts, or non-CSCs. Our results suggested that the NF-κB pathway is one of the most activated pathways in pancreatic CSCs. In agreement with this, we determined that upon treatment with NF-κB pathway inhibitors, the stem cell-like properties of cells are significantly disrupted. Moreover, SOX9, demethylated in CSCs, is shown to play a crucial role in the invasion process. Additionally, we found a potential NF-κB binding site located in the SOX9 promoter and determined that the NF-κB subunit p65 positively regulates SOX9 expression by binding to its promoter directly. This interaction can be efficiently blocked by NF-κB inhibitors. Thus, our work establishes a link between the classic NF-κB signaling transduction pathway and the invasiveness of pancreatic CSCs, which may result in the identification of novel signals and molecules that function at an epigenetic level, and could potentially be targeted for pharmaceutical investigations and clinical trials.


Subject(s)
NF-kappa B/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Animals , Cell Line, Tumor , DNA Methylation , Epigenomics , Female , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Mice , Mice, Inbred NOD , Mice, SCID , NF-kappa B/genetics , Neoplasm Invasiveness , Signal Transduction , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
9.
Cancer Cell Int ; 14: 75, 2014.
Article in English | MEDLINE | ID: mdl-25324692

ABSTRACT

BACKGROUND: The small molecule NSC676914A was previously identified as an NF-κB inhibitor in TPA-stimulated HEK293 cells (Mol Can Ther 8:571-581, 2009). We hypothesized that this effect would also be seen in ovarian cancer cells, and serve as its mechanism of cytotoxicity. OVCAR3 and HEK293 cell lines stably containing a NF-κB luciferase reporter gene were generated. METHODS: Levels of NF-κB activity were assessed by luciferase reporter assays, after stimulation with LPA, LPS, TPA, and TNFα, in the presence or absence of a known NF-κB inhibitor or NSC676914A, and cytotoxicity was measured. RESULTS: NSC676914A was toxic to both OVCAR3 and HEK293 cells. We also investigated the cytotoxicity of NSC676914A on a panel of lymphoma cell lines with well characterized mutations previously shown to determine sensitivity or resistance to NF-κB inhibition. The compound did not show predicted patterns of effects on NF-κB activity in either lymphoma, ovarian or HEK293 cell lines. In HEK293 cells, the small molecule inhibited NF-κB when cells were stimulated, while in OVCAR3 cells it only partially inhibited NF-κB. Interestingly, we observed rescue of cell death with ROS inhibition. CONCLUSIONS: The current study suggests that the effect of NSC676914A on NF-κB depends on cell type and the manner in which the pathway is stimulated. Furthermore, as it is similarly toxic to lymphoma, OVCAR3 and HEK293 cells, NSC676914A shows promising NF-κB-independent anti-cancer activity in ovarian tumor cells.

10.
Proc Natl Acad Sci U S A ; 108(17): 7004-9, 2011 Apr 26.
Article in English | MEDLINE | ID: mdl-21487000

ABSTRACT

Oxidative stress is known to cause tumorigenesis through induction of DNA and lipid damage. It also promotes cancer progression through a largely unknown mechanism. Sulfiredoxin (Srx) is a novel oxidative stress-induced antioxidant protein whose function in tumorigenesis and cancer progression has not been well studied. We report that Srx is highly expressed in human lung cancer. Knockdown of Srx reduces anchorage-independent colony formation, cell migration, and invasion of human lung cancer cells. Srx preferentially interacts with Peroxiredoxin (Prx) IV relative to other Prxs due to its intrinsic higher binding affinity. Knockdown of Prx IV recapitulates the phenotypic changes of depleting Srx. Disruption or enhancement of the Srx-Prx IV axis leads respectively to reduction or acceleration of tumor growth and metastasis formation in vivo. Through identification and validation of the downstream mediators we unraveled the Srx-mediated signaling network that traverses AP-1-activating and other phosphokinase signaling cascades. Our work reveals that the Srx-Prx IV axis is critical for lung cancer maintenance and metastasis, suggesting that targeting the Srx-Prx IV axis may provide unique effective strategies for cancer prevention and treatment.


Subject(s)
Lung Neoplasms/enzymology , Neoplasm Proteins/metabolism , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Peroxiredoxins/metabolism , Phosphotransferases/metabolism , Cell Line, Tumor , Gene Knockdown Techniques , HEK293 Cells , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/genetics , Oxidative Stress/genetics , Oxidoreductases Acting on Sulfur Group Donors/genetics , Peroxiredoxins/genetics , Phosphotransferases/genetics , Signal Transduction , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
11.
Gut ; 62(8): 1179-86, 2013 Aug.
Article in English | MEDLINE | ID: mdl-22698650

ABSTRACT

OBJECTIVES: Screening colonoscopy to monitor for early colitis-associated colon cancer (CAC) is difficult due to the aberrant mucosal patterns associated with long-standing colitis. The aim of this study was to develop a rapid fluorescent detection method for use during colonoscopy for improving the detection of CAC utilising a topically applied enzymatically activatable probe (gGlu-HMRG) which fluoresces in the presence of γ-glutamyltranspeptidase (GGT), an enzyme associated with cancer. METHODS: Expression of GGT in colon cell lines was examined with fluorescence microscopy and flow cytometry. A mouse model (azoxymethane/dextran sulphate sodium) of CAC was used and mice were examined with white light and fluorescence colonoscopy before and after topical gGlu-HMRG administration. RESULTS: Expression of GGT, although variable, was higher in human colon cancer cells than normal human colon cells. Using fluorescence colonoscopy in mice, gGlu-HMRG fluorescent lesions were detected 5 min after topical administration and fluorescence persisted for at least 30 min. Fluorescence guided biopsy revealed all fluorescent lesions that contained cancer or dysplasia (n=16), whereas three out of 12 non-fluorescent lesions contained low grade dysplasia and others did not contain neoplastic histology. Microscopic inflammatory infiltration also had variable fluorescence but in general was much lower (∼10-fold) in signal than cancer. Repeat fluorescence endoscopy allowed individual tumours to be monitored. CONCLUSION: These results suggest that gGlu-HMRG can improve endoscopic detection of CAC with a higher target to background ratio than conventional white light colonoscopy. This could be of benefit to patients with long-standing colitis who must undergo repeated screening colonoscopies.


Subject(s)
Colitis/complications , Colonic Neoplasms/diagnosis , Colonic Neoplasms/etiology , Adenocarcinoma/diagnosis , Adenocarcinoma/enzymology , Adenocarcinoma/etiology , Administration, Topical , Animals , Biomarkers, Tumor/metabolism , Biopsy , Colon/enzymology , Colonic Neoplasms/enzymology , Colonoscopy/methods , Disease Models, Animal , Early Detection of Cancer/methods , Fluorescent Dyes/administration & dosage , Humans , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence/methods , Precancerous Conditions/diagnosis , Precancerous Conditions/enzymology , Precancerous Conditions/etiology , Tumor Cells, Cultured , gamma-Glutamyltransferase/metabolism
12.
Carcinogenesis ; 34(6): 1403-10, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23393226

ABSTRACT

Sulfiredoxin (Srx) is the enzyme that reduces the hyperoxidized inactive form of peroxiredoxins. To study the function of Srx in carcinogenesis in vivo, we tested whether loss of Srx protects mice from cancer development. Srx null mice were generated and colon carcinogenesis was induced by an azoxymethane (AOM) and dextran sulfate sodium (DSS) protocol. Compared with either wild-type (Wt) or heterozygotes, Srx(-/-) mice had significantly reduced rates in both tumor multiplicity and volume. Mechanistic studies reveal that loss of Srx did not alter tumor cell proliferation; however, increased apoptosis and decreased inflammatory cell infiltration were obvious in tumors from Srx null mice compared with those from Wt control. In addition to the AOM/DSS model, examination of Srx expression in human reveals a tissue-specific expression pattern. Srx expression was also demonstrated in tumors from colorectal cancer patients and the levels of expression were associated with patients' clinic stages. These data provide the first in vivo evidence that loss of Srx renders mice resistant to AOM/DSS-induced colon carcinogenesis, suggesting that Srx has a critical oncogenic role in cancer development, and Srx may be used as a marker for human colon cancer pathogenicity.


Subject(s)
Cell Transformation, Neoplastic , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Animals , Apoptosis , Azoxymethane , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/chemically induced , Dextran Sulfate , Genotype , Humans , Lung Neoplasms , Macrophages/immunology , Mice , Mice, Knockout , Oxidoreductases Acting on Sulfur Group Donors/genetics , Peroxiredoxins/metabolism
13.
J Cell Physiol ; 228(7): 1536-50, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23280476

ABSTRACT

Recent studies have suggested that changes in serum phosphate levels influence pathological states associated with aging such as cancer, bone metabolism, and cardiovascular function, even in individuals with normal renal function. The causes are only beginning to be elucidated but are likely a combination of endocrine, paracrine, autocrine, and cell autonomous effects. We have used an integrated quantitative biology approach, combining transcriptomics and proteomics to define a multi-phase, extracellular phosphate-induced, signaling network in pre-osteoblasts as well as primary human and mouse mesenchymal stromal cells. We identified a rapid mitogenic response stimulated by elevated phosphate that results in the induction of immediate early genes including c-fos. The mechanism of activation requires FGF receptor signaling followed by stimulation of N-Ras and activation of AP-1 and serum response elements. A distinct long-term response also requires FGF receptor signaling and results in N-Ras activation and expression of genes and secretion of proteins involved in matrix regulation, calcification, and angiogenesis. The late response is synergistically enhanced by addition of FGF23 peptide. The intermediate phase results in increased oxidative phosphorylation and ATP production and is necessary for the late response providing a functional link between the phases. Collectively, the results define elevated phosphate, as a mitogen and define specific mechanisms by which phosphate stimulates proliferation and matrix regulation. Our approach provides a comprehensive understanding of the cellular response to elevated extracellular phosphate, functionally connecting temporally coordinated signaling, transcriptional, and metabolic events with changes in long-term cell behavior.


Subject(s)
Mesenchymal Stem Cells/metabolism , Phosphates/metabolism , Signal Transduction/physiology , 3T3 Cells , Adenosine Triphosphate/biosynthesis , Animals , Cells, Cultured , Computational Biology , Extracellular Space/metabolism , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/metabolism , GTP-Binding Proteins/metabolism , Gene Expression , Genes, Immediate-Early , Genes, fos , Genes, ras , Humans , Mice , Neovascularization, Physiologic , Osteoblasts/metabolism , Promoter Regions, Genetic , Proteins/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Transcription Factor AP-1/metabolism , Transcription Factors/metabolism
14.
Pancreas ; 50(7): 916-922, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34629446

ABSTRACT

ABSTRACT: The potential of artificial intelligence (AI) applied to clinical data from electronic health records (EHRs) to improve early detection for pancreatic and other cancers remains underexplored. The Kenner Family Research Fund, in collaboration with the Cancer Biomarker Research Group at the National Cancer Institute, organized the workshop entitled: "Early Detection of Pancreatic Cancer: Opportunities and Challenges in Utilizing Electronic Health Records (EHR)" in March 2021. The workshop included a select group of panelists with expertise in pancreatic cancer, EHR data mining, and AI-based modeling. This review article reflects the findings from the workshop and assesses the feasibility of AI-based data extraction and modeling applied to EHRs. It highlights the increasing role of data sharing networks and common data models in improving the secondary use of EHR data. Current efforts using EHR data for AI-based modeling to enhance early detection of pancreatic cancer show promise. Specific challenges (biology, limited data, standards, compatibility, legal, quality, AI chasm, incentives) are identified, with mitigation strategies summarized and next steps identified.


Subject(s)
Artificial Intelligence , Congresses as Topic , Early Detection of Cancer/methods , Electronic Health Records/statistics & numerical data , Pancreatic Neoplasms/diagnosis , Biomedical Research/methods , Biomedical Research/statistics & numerical data , Humans , Information Dissemination/methods
15.
Mol Cancer Ther ; 8(3): 571-81, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19258426

ABSTRACT

NSC 676914 has been identified as a selective nuclear factor-kappaB (NF-kappaB) inhibitor that does not inhibit cell proliferation. This compound was originally identified in a high-throughput cell-based assay for activator protein-1 (AP-1) inhibitors using synthetic compound libraries and the National Cancer Institute natural product repository. NSC 676914 shows activity against NF-kappaB in luciferase reporter assays at concentrations much less than the IC50 for AP-1. A serum response element reporter used as a specificity control and indicator of cell proliferation was relatively insensitive to the compound. Pretreatment with NSC 676914 is here shown to repress 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced IkappaB-alpha phosphorylation and translocation of p65/50 to the nucleus but not the processing of p52 from p100, suggesting the inhibition of NF-kappaB regulator IKKbeta rather than IKKalpha. Inhibition of NF-kappaB activation occurred as a consequence of blocking phosphorylation of IKK. Induction of IkappaB-alpha phosphorylation by TPA was diminished by pretreatment of NSC 676914 even at 1.1 mumol/L. In contrast, kinases c-Jun-NH2-kinase and extracellular signal-regulated kinases 1 and 2, important for AP-1 activation, showed no significant repression by this compound. Furthermore, a Matrigel invasion assay with breast cancer cell lines and a transformation assay in mouse JB6 cells revealed that TPA-induced invasion and transformation responses were completely repressed by this compound. These results suggest that NSC 676914 could be a novel inhibitor having potential therapeutic activity to target NF-kappaB for cancer treatment or prevention.


Subject(s)
Antineoplastic Agents/isolation & purification , Breast Neoplasms/pathology , NF-kappa B/antagonists & inhibitors , Small Molecule Libraries/analysis , Transcription Factor AP-1/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Azo Compounds/pharmacology , Azo Compounds/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , I-kappa B Proteins/metabolism , Models, Biological , NF-kappa B/genetics , Phosphorylation/drug effects , Substrate Specificity , Sulfonic Acids/pharmacology , Sulfonic Acids/therapeutic use , Transcription Factor AP-1/genetics , Transfection
16.
Pancreas ; 49(7): 882-886, 2020 08.
Article in English | MEDLINE | ID: mdl-32675784

ABSTRACT

Pancreatic cancer continues to be one of the deadliest malignancies and is the third leading cause of cancer-related mortality in the United States. Based on several models, it is projected to become the second leading cause of cancer-related deaths by 2030. Although the overall survival rate for patients diagnosed with pancreatic cancer is less than 10%, survival rates are increasing in those whose cancers are detected at an early stage, when intervention is possible. There are, however, no reliable biomarkers or imaging technology that can detect early-stage pancreatic cancer or accurately identify precursors that are likely to progress to malignancy. The Alliance of Pancreatic Cancer Consortia, a virtual consortium of researchers, clinicians, and advocacies focused on early diagnosis of pancreatic cancer, was formed in 2016 to provide a platform and resources to discover and validate biomarkers and imaging methods for early detection. The focus of discussion at the most recent alliance meeting was on imaging methods and the use of artificial intelligence for early detection of pancreatic cancer.


Subject(s)
Artificial Intelligence , Diagnostic Imaging/methods , Early Detection of Cancer/methods , Pancreas/diagnostic imaging , Pancreatic Neoplasms/diagnosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Diagnostic Imaging/trends , Humans , Pancreas/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism
17.
Cancer Res ; 67(6): 2430-8, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17363560

ABSTRACT

Activation of activator protein 1 (AP-1) and nuclear factor kappaB (NFkappaB)-dependent transcription is required for tumor promotion in cell culture models and transgenic mice. Dominant-negative c-Jun (TAM67) blocks AP-1 activation by dimerizing with Jun or Fos family proteins and blocks NFkappaB activation by interacting with NFkappaB p65. Two-stage [7,12-dimethylbenz(a)anthracene (DMBA)/12-O-tetradecanoylphorbol-13-acetate (TPA)] skin carcinogenesis experiments in a model relevant to human cancer risk, transgenic mice expressing human papillomavirus 16 E7 oncogene (K14-HPV16-E7), show E7-enhanced tumor promotion. A cross to K14-TAM67-expressing mice results in dramatic inhibition of tumor promoter-induced AP-1 luciferase reporter activation and papillomagenesis. Epithelial specific TAM67 expression inhibits tumorigenesis without affecting TPA- or E7-induced hyperproliferation of the skin. Thus, the mouse model enriches for TAM67 targets relevant to tumorigenesis rather than to general cell proliferation or hyperplasia, implicating a subset of AP-1- and/or NFkappaB-dependent genes. The aim of the present study was to identify target genes responsible for TAM67 inhibition of DMBA-TPA-induced tumorigenesis. Microarray expression analysis of epidermal tissues revealed small sets of genes in which expression is both up-regulated by tumor promoter and down-regulated by TAM67. Among these, cyclooxygenase-2 (Cox-2/Ptgs2) and osteopontin (Opn/Spp1) are known to be functionally significant in driving carcinogenesis. Results identify both Cox-2 and Opn as transcriptional targets of TAM67 with CRE, but not NFkappaB sites important in the Cox-2 promoter and an AP-1 site important in the Opn promoter.


Subject(s)
Cyclooxygenase 2/genetics , Gene Expression Regulation, Neoplastic/genetics , Osteopontin/genetics , Peptide Fragments/genetics , Proto-Oncogene Proteins c-jun/genetics , Skin Neoplasms/genetics , Transcription Factor AP-1/genetics , 9,10-Dimethyl-1,2-benzanthracene , Animals , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclooxygenase 2/biosynthesis , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/metabolism , Oncogene Proteins, Viral/genetics , Osteopontin/biosynthesis , Papillomavirus E7 Proteins , Peptide Fragments/biosynthesis , Promoter Regions, Genetic , Proto-Oncogene Proteins c-jun/biosynthesis , Skin Neoplasms/chemically induced , Tetradecanoylphorbol Acetate , Transcription, Genetic , Transfection
18.
Pancreas ; 47(2): 135-141, 2018 02.
Article in English | MEDLINE | ID: mdl-29346214

ABSTRACT

Pancreatic cancer is the third leading cause of cancer death in the United States, and the 5-year relative survival for patients diagnosed with pancreatic cancer is less than 10%. Early intervention is the key to a better survival outcome. Currently, there are no biomarkers that can reliably detect pancreatic cancer at an early stage or identify precursors that are destined to progress to malignancy. The National Cancer Institute in partnership with the Kenner Family Research Fund and the Pancreatic Cancer Action Network convened a Data Jamboree on Biomarkers workshop on December 5, 2016, to discuss and evaluate existing or newly developed biomarkers and imaging methods for early detection of pancreatic cancer. The primary goal of this workshop was to determine if there are any promising biomarkers for early detection of pancreatic cancer that are ready for clinical validation. The Alliance of Pancreatic Cancer Consortia for Biomarkers for Early Detection, formed under the auspices of this workshop, will provide the common platform and the resources necessary for validation. Although none of the biomarkers evaluated seemed ready for a large-scale biomarker validation trial, a number of them had sufficiently high sensitivity and specificity to warrant additional research, especially if combined with other biomarkers to form a panel.


Subject(s)
Biomarkers, Tumor/blood , Early Detection of Cancer/methods , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/diagnosis , Biomarkers, Tumor/genetics , Genetic Predisposition to Disease/genetics , Humans , Mutation , Pancreatic Neoplasms/genetics , Sensitivity and Specificity
19.
J Biomol Screen ; 12(1): 133-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17175522

ABSTRACT

The oncogenic transcription factor AP-1 (activator protein-1) is required for tumor promotion and progression. Identification of novel and specific AP-1 inhibitors would be beneficial for cancer prevention and therapy. The authors have developed a high-throughput assay to screen synthetic and natural product libraries for noncytotoxic inhibitors of mitogen-activated AP-1 activity. The cell-based high-throughput screen is conducted in a 384-well format using a fluorescent resonance energy transfer (FRET) substrate to quantify the activity of a beta-lactamase reporter under the control of an AP-1-dependent promoter. The ratiometric FRET readout makes this assay extremely robust and reproducible, particularly for use with natural product extracts. To eliminate false positives due to cell killing, a cytotoxicity assay was incorporated. The AP-1 beta-lactamase reporter was validated with inhibitors of kinases located upstream of AP-1 and with known natural product inhibitors of AP-1 (nordihydroguaiaretic acid and curcumin). The assay was able to identify other known AP-1 inhibitors and protein kinase C modulators, as well as a number of chemically diverse compounds with unknown mechanisms of action from natural products libraries. Application to natural product extracts identified hits from a range of taxonomic groups. Screening of synthetic compounds and natural products should identify novel AP-1 inhibitors that may be useful in the prevention and treatment of cancers.


Subject(s)
Antineoplastic Agents/analysis , Antineoplastic Agents/pharmacology , Biological Assay/methods , Transcription Factor AP-1/antagonists & inhibitors , False Negative Reactions , False Positive Reactions , Humans , Plant Extracts/analysis , Plant Extracts/pharmacology , Reproducibility of Results , beta-Lactamases/metabolism
20.
Mol Cell Biol ; 22(2): 587-98, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11756554

ABSTRACT

Mitogen-activated protein (MAP) kinase, extracellular-signal-regulated kinases (ERKs) play an important role in activating AP-1-dependent transcription. Studies using the JB6 mouse epidermal model and a transgenic mouse model have established a requirement for AP-1-dependent transcription in tumor promotion. Tumor promoters such as 12-O-tetradecanoylphorbol-13-acetate (TPA) and epidermal growth factor induce activator protein 1 (AP-1) activity and neoplastic transformation in JB6 transformation-sensitive (P(+)) cells, but not in transformation-resistant (P(-)) variants. The resistance in one of the P(-) variants can be attributed to the low levels of the MAP kinases, ERKs 1 and 2, and consequent nonresponsiveness to AP-1 activation. The resistant variant is not deficient in c-fos transcription. The purpose of these studies was to define the targets of activated ERK that lead to AP-1 transactivation. The results establish that the transactivation domain of Fra-1 can be activated, that activation of Fra-1 is ERK dependent, and that a putative ERK phosphorylation site must be intact for activation to occur. Fra-1 was activated by TPA in ERK-sufficient P(+) cells but not in ERK-deficient P(-) cells. A similar activation pattern was seen for c-Fos but not for Fra-2. Gel shift analysis identified Fra-1 as distinguishing mitogen-activated (P(+)) from nonactivated (P(-)) AP-1 complexes. A second AP-1-nonresponsive P(-) variant that underexpresses Fra-1 gained AP-1 response upon introduction of a Fra-1 expression construct. These observations suggest that ERK-dependent activation of Fra-1 is required for AP-1 transactivation in JB6 cells.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-fos/genetics , Transcription Factor AP-1/metabolism , Animals , Base Sequence , Binding Sites , Carcinogens/toxicity , Cell Line , Cell Transformation, Neoplastic/genetics , Genes, fos , Genetic Variation , Mice , Plasmids/genetics , Protein Structure, Tertiary , Proto-Oncogene Proteins c-fos/chemistry , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Tetradecanoylphorbol Acetate/toxicity , Transcriptional Activation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL