Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Invest New Drugs ; 33(5): 1003-11, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26123924

ABSTRACT

PURPOSE: The goal of these studies was to test if local excess of a normal nucleobase substrate prevents the toxicity of protracted 5FU exposure used in human cancer treatment. METHODS: Messenger RNA expression studies were performed of 5FU activating enzymes in human colon cancer cells lines (CaCo-2, HT-29), primary human gingival cells (HEGP), and normal esophageal and gastric clinical tissue samples. Excess nucleobase was then used in vitro to protect cells from 5FU toxicity. RESULTS: Pyrimidine salvage pathways predominate in squamous cells of the gingiva (HEGP) and esophageal tissue. Excess salvage nucleobase uracil but not adenine prevented 5FU toxicity in HEGP cells. Pyrimidine de novo synthesis predominates in columnar Caco-2, HT-29 and gastric tissue. Excess nucleobase adenine but not uracil prevented 5FU toxicity to Caco-2 and HT-29 cells. CONCLUSION: The directed application of the normal nucleobase uracil to the squamous cells of the oral mucosa and palms and soles together with the delivery of the normal nucleobase adenine to the columnar cells of the GI tract may enable the safe delivery of higher 5FU dose intensity. These results also suggest a feature of tissue function where squamous cells grow largely by recycling overlying tissue cell components. Columnar cells use absorbed surface nutrients for de novo growth. A disruption of this tissue function can result in growth derived from an underlying nutrient source. That change would also cause the loss of the region of cell turnover at the tissue surface. Subsequent cell proliferation with limiting nutrient availability could promote oncogenesis in such initiated tissue.


Subject(s)
Adenine/pharmacology , Fluorouracil/toxicity , Protective Agents/pharmacology , Pyrimidines/pharmacology , Uracil/pharmacology , Carcinogenesis , Cell Line, Tumor , DNA Replication , Epithelial Cells/drug effects , Esophagus/cytology , Gastric Mucosa/cytology , Gene Expression/drug effects , Humans , Nucleobase Transport Proteins/drug effects , RNA, Messenger
2.
Blood ; 118(26): 6909-19, 2011 Dec 22.
Article in English | MEDLINE | ID: mdl-21967980

ABSTRACT

Targeting cancer stem cells is of paramount importance in successfully preventing cancer relapse. Recently, in silico screening of public gene-expression datasets identified cyclooxygenase-derived cyclopentenone prostaglandins (CyPGs) as likely agents to target malignant stem cells. We show here that Δ(12)-PGJ(3), a novel and naturally produced CyPG from the dietary fish-oil ω-3 polyunsaturated fatty acid eicosapentaenoic acid (EPA; 20:5) alleviates the development of leukemia in 2 well-studied murine models of leukemia. IP administration of Δ(12)-PGJ(3) to mice infected with Friend erythroleukemia virus or those expressing the chronic myelogenous leukemia oncoprotein BCR-ABL in the hematopoietic stem cell pool completely restored normal hematologic parameters, splenic histology, and enhanced survival. More importantly, Δ(12)-PGJ(3) selectively targeted leukemia stem cells (LSCs) for apoptosis in the spleen and BM. This treatment completely eradicated LSCs in vivo, as demonstrated by the inability of donor cells from treated mice to cause leukemia in secondary transplantations. Given the potency of ω-3 polyunsaturated fatty acid-derived CyPGs and the well-known refractoriness of LSCs to currently used clinical agents, Δ(12)-PGJ(3) may represent a new chemotherapeutic for leukemia that targets LSCs.


Subject(s)
Apoptosis/drug effects , Fatty Acids, Omega-3/pharmacology , Leukemia/drug therapy , Neoplastic Stem Cells/drug effects , Prostaglandins/pharmacology , Animals , Ataxia Telangiectasia Mutated Proteins , Blotting, Western , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cyclopentanes/chemistry , Cyclopentanes/metabolism , Cyclopentanes/pharmacology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Fatty Acids, Omega-3/chemistry , Fatty Acids, Omega-3/metabolism , Leukemia/metabolism , Leukemia/pathology , Leukemia, Erythroblastic, Acute/drug therapy , Leukemia, Erythroblastic, Acute/metabolism , Leukemia, Erythroblastic, Acute/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Structure , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Prostaglandins/chemistry , Prostaglandins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Splenomegaly/pathology , Splenomegaly/prevention & control , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
3.
J Lipid Res ; 53(8): 1679-89, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22556214

ABSTRACT

Omega-3-PUFAs, eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), are associated with prevention of various aspects of metabolic syndrome. In the present studies, the effects of oil rich in EPA on gene expression and activation of nuclear receptors was examined and compared with other ω3-PUFAs. The EPA-rich oil (EO) altered the expression of FA metabolism genes in THP-1 cells, including stearoyl CoA desaturase (SCD) and FA desaturase-1 and -2 (FASDS1 and -2). Other ω3-PUFAs resulted in a similar gene expression response for a subset of genes involved in lipid metabolism and inflammation. In reporter assays, EO activated human peroxisome proliferator-activated receptor α (PPARα) and PPARß/γ with minimal effects on PPARγ, liver X receptor, retinoid X receptor, farnesoid X receptor, and retinoid acid receptor γ (RARγ); these effects were similar to that observed for purified EPA. When serum from a 6 week clinical intervention with dietary supplements containing olive oil (control), DHA, or two levels of EPA were applied to THP-1 cells, the expression of SCD and FADS2 decreased in the cells treated with serum from the ω3-PUFA-supplemented individuals. Taken together, these studies indicate regulation of gene expression by EO that is consistent with treating aspects of dyslipidemia and inflammation.


Subject(s)
Eicosapentaenoic Acid/analysis , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Oils/chemistry , Oils/pharmacology , Cell Line , Dietary Supplements/analysis , Eicosapentaenoic Acid/blood , Eicosapentaenoic Acid/chemistry , Eicosapentaenoic Acid/pharmacology , Humans , Inflammation/genetics , Inflammation/metabolism , Structure-Activity Relationship , Transcription Factors/metabolism , Transcriptome/drug effects
4.
Nutr Cancer ; 64(7): 1078-86, 2012.
Article in English | MEDLINE | ID: mdl-23061909

ABSTRACT

Walnuts contain bioactive molecules that may contribute to their beneficial effects, including alpha-linolenic acid (ALA) and phytosterols. In these studies, extracts of walnut, purified compounds, or postprandial serum were examined for effects on breast cancer cell proliferation and gene expression. Extracts derived from walnut oil decreased proliferation of MCF-7 cells, as did ALA and ß-sitosterol. The gene expression response of ALA in the mouse breast cancer cell line TM2H indicates this molecule has multiple cellular targets with peroxisome proliferator-activated receptor (PPAR) target genes, liver X receptor (LXR), and farnesoid X receptor (FXR) target genes being affected. In transactivation assays, walnut oil extracts increased activity of FXR to a greater extent than the other tested nuclear receptors. When examined separately, walnut components ALA and ß-sitosterol were the most efficacious activators of FXR. When serum from individuals fed walnut components were applied to MCF-7 cells, there was a correlation between body mass index and breast cancer cell proliferation in vitro. Taken together, these data support an effect of walnut and its bioactive constituents on mammary epithelial cells and that multiple molecular targets may be involved.


Subject(s)
Anticarcinogenic Agents/pharmacology , Breast Neoplasms/prevention & control , Nuts/chemistry , Plant Extracts/pharmacology , 3T3 Cells , Animals , Cell Proliferation/drug effects , Chemoprevention , Epithelial Cells/drug effects , Epithelial Cells/pathology , Female , Humans , Juglans , Liver X Receptors , MCF-7 Cells , Mice , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , Peroxisome Proliferator-Activated Receptors/genetics , Peroxisome Proliferator-Activated Receptors/metabolism , Plant Oils/chemistry , Real-Time Polymerase Chain Reaction , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Sitosterols/pharmacology , alpha-Linolenic Acid/pharmacology
5.
Prostaglandins Other Lipid Mediat ; 99(1-2): 30-7, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22583689

ABSTRACT

Conjugated linoleic acids (CLAs) are a group of dietary fatty acids that are widely marketed as weight loss supplements. The isomer responsible for this effect is the trans-10, cis-12 CLA (10E12Z-CLA) isomer. 10E12Z-CLA treatment during differentiation of 3T3-L1 adipocytes induces expression of prostaglandin-endoperoxide synthase-2 (Cyclooxygenase-2; COX-2). This work demonstrates that COX-2 is also induced in fully differentiated 3T3-L1 adipocytes after a single treatment of 10E12Z-CLA at both the mRNA (20-40 fold) and protein level (7 fold). Furthermore, prostaglandin (PG)F(2α), but not PGE(2), is significantly increased 10 fold. In female BALB/c mice fed 0.5% 10E12Z-CLA for 10 days, COX-2 was induced in uterine adipose (2 fold). In vitro, pharmacological COX-2 inhibition did not block the effect of 10E12Z-CLA on adipocyte-specific gene expression although PGF(2α) was dose-dependently decreased. These studies demonstrate that PGF(2α) was not by itself responsible for the reduction in adipocyte character due to 10E12Z-CLA treatment. However, PGF(2α), either exogenously or endogenously in response to 10E12Z-CLA, increased the expression of the potent mitogen and epidermal growth factor (EGF) receptor (EGFR) ligand epiregulin in 3T3-L1 adipocytes. Blocking PGF(2α) signaling with the PGF(2α) receptor (FP) antagonist AL-8810 returned epiregulin mRNA levels back to baseline. Although this pathway is not directly responsible for adipocyte dependent gene expression, these results suggest that this signaling pathway may still have broad effect on the adipocyte and surrounding cells.


Subject(s)
Adipocytes/metabolism , Cyclooxygenase 2/metabolism , Dietary Fats, Unsaturated/pharmacology , Dinoprost/biosynthesis , Epidermal Growth Factor/biosynthesis , Linoleic Acids, Conjugated/pharmacology , 3T3-L1 Cells , Adipocytes/drug effects , Animals , Cell Differentiation/drug effects , Epiregulin , Female , Mice , Mice, Inbred BALB C
6.
Mol Pharmacol ; 80(1): 201-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21511917

ABSTRACT

2-Arachidonyl glycerol (2-AG) is an endogenous arachidonic acid derivative capable of suppressing interleukin (IL)-2 production by activated T cells. 2-AG-mediated IL-2 suppression is dependent on cyclooxygenase-2 (COX-2) metabolism and peroxisome proliferator activated receptor γ (PPARγ) activation. The objective of the present studies was to examine whether 15-deoxy-Δ(12,14)-PGJ(2)-glycerol ester (15d-PGJ(2)-G), a putative metabolite of 2-AG, can mimic the actions of 2-AG on IL-2 regulation through PPARγ activation. 15d-PGJ(2)-G bound PPARγ-ligand binding domain in a PPARγ competitive binding assay. 15d-PGJ(2)-G treatment activated PPARγ in a reporter assay, and PPARγ activation was attenuated when a PPARγ antagonist, 2-chloro-5-nitro-N-4-pyridinylbenzamide (T0070907), was present. 15d-PGJ(2)-G treatment suppressed IL-2 production by activated Jurkat cells, which was partially attenuated when pretreated with T0070907. Moreover, IL-2 suppression was pronounced when 15d-PGJ(2)-G was present 30 min before or after T-cell activation. Concordant with IL-2 suppression, 15d-PGJ(2)-G treatment decreased nuclear factor of activated T cells (NFAT) transcriptional activity in transiently transfected Jurkat cells. It is noteworthy that T0070907 alone markedly increased NFAT reporter activity, suggesting the existence of endogenous PPARγ activation and modulation of NFAT. Because COX-2 metabolism of 2-AG is important for IL-2 suppression, the effect of 2-AG on COX-2 and PPARγ mRNA expression was investigated. 2-AG treatment decreased the up-regulation of COX-2 mRNA after T-cell activation, which suggests negative feedback limiting COX-2-mediated metabolism of 2-AG. PPARγ mRNA expression was increased upon activation, and 2-AG treatment produced a modest decrease in PPARγ mRNA expression. Collectively, our findings suggest that 15d-PGJ(2)-G activates PPARγ to decrease NFAT transcriptional activity and IL-2 expression in activated T cells.


Subject(s)
PPAR gamma/drug effects , Prostaglandin D2/analogs & derivatives , Enzyme-Linked Immunosorbent Assay , Humans , Interleukin-2/antagonists & inhibitors , Jurkat Cells , Prostaglandin D2/metabolism , Prostaglandin D2/pharmacology
7.
J Nutr ; 139(10): 1861-6, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19656856

ABSTRACT

Despite having a similar structure, various conjugated linoleic acid (CLA) isomers have a distinct gene expression pattern in RAW 264.7 (RAW) cells, a mouse macrophage cell line. Among the 5 CLA isomers tested [9cis(Z),11trans(E)-, 9Z,11Z-, 9E,11E-, 10E,12Z-, and 11Z,13E-CLA], only 9E,11E-CLA induced the endogenous antiinflammatory molecule, interleukin (IL)-1 receptor antagonist (IL-1Ra), in RAW cells. In this study, the mechanism and effects of IL-1Ra regulation by 9E,11E-CLA in RAW cells was studied in detail. 9E,11E-CLA induced IL-1Ra in a dose- and time-dependent manner, whereas it decreased lipopolysaccharide (LPS)-induced IL-1alpha, IL-1beta, and IL-6 mRNA levels and protein levels. To determine the importance of IL-1Ra in the antiinflammatory effects of this particular CLA isomer, IL-1Ra protein levels were repressed in RAW cells using small interference RNA inhibitor expression. In the presence of IL-1Ra small interference RNA, the induction of this molecule was ablated, as was the decrease of LPS-induced IL-1alpha and IL-6 mRNA levels by 9E,11E-CLA. The IL-1Ra increase due to this CLA isomer was transcriptionally regulated, although there was no response element(s) affected by 9E,11E-CLA in the first 1.5 kb of the IL-1Ra promoter. The phosphoinositide 3-kinase inhibitors, LY294002 and the mammalian target of rapamycin inhibitor rapamycin, abolished the IL-1Ra induction by 9E,11E-CLA, whereas other kinase inhibitors did not affect this response. Taken together, 9E,11E-CLA exerts unique antiinflammatory effects by increasing an endogenous repressor of IL-1 signaling.


Subject(s)
Gene Expression Regulation/drug effects , Interleukin 1 Receptor Antagonist Protein/metabolism , Linoleic Acids, Conjugated/pharmacology , Macrophages/drug effects , Animals , Cell Line , Interleukin 1 Receptor Antagonist Protein/genetics , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Mice , Sirolimus/pharmacology
8.
Free Radic Biol Med ; 42(8): 1155-64, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17382197

ABSTRACT

Liver insufficiency and damage are major causes of death and disease worldwide and may result from exposure to environmental toxicants, specific combinations or dosages of pharmaceuticals, and microbial metabolites. The generation of reactive intermediates, in particular 4-hydroxynonenal (4-HNE), is a common event in liver damage caused by a variety of hepatotoxic drugs and solvents. The peroxisome proliferator-activated receptors (PPARs) are nuclear receptors that are involved in the transcriptional regulation of lipid metabolism as well as other biological functions. Importantly, we have observed that the PPARbeta/delta-/- mouse is more susceptible to chemically induced hepatotoxicity than its wild-type counterpart, and our objective in this study was to elucidate the mechanism(s) by which PPARbeta/delta confers protection to hepatocytes. We hypothesized that PPARbeta/delta plays a protective role by responding to toxic lipids and altering gene expression accordingly. In support, oxidized-VLDL and constituents including 13-S-hydroxyoctadecadienoic acid (13-S-HODE) and 4-HNE are PPARbeta/delta ligands. A structure-activity relationship was established where 4-HNE and 4-hydroperoxynonenal (4-HpNE) enhanced the activity of the PPARbeta/delta subtype while 4-hyroxyhexenal (4-HHE), 4-oxo-2-Nonenal (4-ONE), and trans-4,5-epoxy-2(E)-decenal did not activate this receptor. Increasing PPARbeta/delta activity with a synthetic agonist decreased sensitivity of hepatocytes to 4-HNE and other toxic agents, whereas inhibition of this receptor had the opposite result. Gene expression microarray analysis identified several important PPARbeta/delta-regulated detoxification enzymes involved in 4-HNE metabolism that are regulated at the transcript level. This research established 4-HNE as an endogenous modulator of PPARbeta/delta activity and raises the possibility that agonists of this nuclear receptor may be utilized to prevent or treat liver disease associated with oxidative damage.


Subject(s)
Aldehydes/pharmacology , Oxidative Stress/physiology , PPAR gamma/genetics , PPAR-beta/genetics , 3T3 Cells , Adipocytes/drug effects , Adipocytes/physiology , Aldehydes/metabolism , Animals , Humans , Linoleic Acids/blood , Linoleic Acids/chemistry , Lipoproteins, VLDL/blood , Mice , Models, Molecular , Oligonucleotide Array Sequence Analysis , Oxidative Stress/drug effects , PPAR gamma/chemistry , PPAR-beta/chemistry , Plasmids , Protein Conformation , RNA, Messenger/genetics , Transcription, Genetic
9.
Food Sci Nutr ; 5(1): 148-159, 2017 01.
Article in English | MEDLINE | ID: mdl-28070326

ABSTRACT

Cranberry contains high levels of nutrients and bioactive molecules that have health-promoting properties. The purpose of the present studies was to determine if cranberry extracts (CEs) contain phytochemicals that exert anti-inflammatory effects. The human monocytic cell line THP-1 was treated with two CEs (CE and 90MX) and subsequently challenged with Lipopolysaccharides (LPS). Tumor necrosis factor α (TNF α) expression was decreased in the CE-treated cells, indicative of an anti-inflammatory effect. Gene expression microarrays identified several immune-related genes that were responsive to CEs including interferon-induced protein with tetratricopeptide repeats 1 and 3 (IFIT 1 and 3), macrophage scavenger receptor 1 (MSR1) and colony-stimulating factor 2 (CSF2). In addition, in the CE-treated cells, metallothionein 1F and other metal-responsive genes were induced. Taken together, this data indicates that CEs contain bioactive components that have anti-inflammatory effects and may protect cells from oxidative damage.

10.
Toxicol Sci ; 92(2): 476-89, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16731579

ABSTRACT

Administration of ammonium salts of perfluorooctanoate (PFOA) to rats results in peroxisome proliferation and benign liver tumors, events associated with activation of the nuclear receptor (NR) peroxisome proliferator-activated receptor-alpha (PPARalpha). Due to its fatty acid structure, PFOA may activate other NRs, such as PPARbeta, PPARgamma, liver X receptor (LXR), or retinoid X receptor (RXR). In this study, the activation of human, mouse, and rat PPARalpha, PPARbeta, PPARgamma, LXRbeta, and RXRalpha by PFOA (including its linear and branched isomers) and perfluorooctane sulfonate (PFOS) was investigated and compared to several structural classes of natural fatty acids and appropriate positive control ligands. An NR ligand-binding domain/Gal4 DNA-binding domain chimeric reporter system was used. Human, mouse, and rat PPARalpha were activated by PFOA isomers and PFOS. PPARbeta was less sensitive to the agents tested, with only PFOA affecting the mouse receptor. PFOA and PFOS also activated human, mouse, and rat PPARgamma, although the maximum induction of PPARgamma was much less than that seen with rosiglitazone, suggesting that PFOA and PFOS are partial agonists of this receptor. Neither LXRbeta nor the common heterodimerization partner RXRalpha was activated by PFOA in any species examined. Taken together, these data show that of the NRs studied, PPARalpha is the most likely target of PFOA and PFOS, although PPARgamma is also activated to some extent. Compared to naturally occurring long-chain fatty acids, e.g. linoleic and alpha-linolenic acids, these perfluorinated fatty acid analogs were more selective and less potent in their activation of the NRs.


Subject(s)
Alkanesulfonic Acids/pharmacology , Fatty Acids/pharmacology , Fluorocarbons/pharmacology , Receptors, Cytoplasmic and Nuclear/drug effects , 3T3-L1 Cells , Animals , DNA-Binding Proteins , Genes, Reporter/genetics , Humans , Luciferases/genetics , Mice , Plasmids , Rats , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/metabolism , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/genetics , Transfection
11.
Biochem Biophys Rep ; 8: 395-402, 2016 Dec.
Article in English | MEDLINE | ID: mdl-28955982

ABSTRACT

Peroxisome proliferator-activated receptor ß/δ (PPARß/δ) is a member of the nuclear receptor superfamily and a ligand-activated transcription factor that is involved in the regulation of the inflammatory response via activation of anti-inflammatory target genes and ligand-induced disassociation with the transcriptional repressor B-cell lymphoma 6 (BCL6). Chronic pancreatitis is considered to be a significant etiological factor for pancreatic cancer development, and a better understanding of the underlying mechanisms of the transition between inflammation and carcinogenesis would help further elucidate chemopreventative options. The aim of this study was to determine the role of PPARß/δ and BCL6 in human pancreatic cancer of ductal origin, as well as the therapeutic potential of PPARß/δ agonist, GW501516. Over-expression of PPARß/δ inhibited basal and TNFα-induced Nfkb luciferase activity. GW501516-activated PPARß/δ suppressed TNFα-induced Nfkb reporter activity. RNAi knockdown of Pparb attenuated the GW501516 effect on Nfkb luciferase, while knockdown of Bcl6 enhanced TNFα-induced Nfkb activity. PPARß/δ activation induced expression of several anti-inflammatory genes in a dose-dependent manner, and GW501516 inhibited Mcp1 promoter-driven luciferase in a BCL6-dependent manner. Several pro-inflammatory genes were suppressed in a BCL6-dependent manner. Conditioned media from GW501516-treated pancreatic cancer cells suppressed pro-inflammatory expression in THP-1 macrophages as well as reduced invasiveness across a basement membrane. These results demonstrate that PPARß/δ and BCL6 regulate anti-inflammatory signaling in human pancreatic cancer cells by inhibiting NFκB and pro-inflammatory gene expression, and via induction of anti-inflammatory target genes. Activation of PPARß/δ may be a useful target in pancreatic cancer therapeutics.

12.
Free Radic Biol Med ; 32(9): 890-7, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11978490

ABSTRACT

Selenium (Se) is an essential micronutrient for all mammalian species and is associated with a variety of physiological functions, notably immune system, in the form of selenoproteins. Inadequate Se nutrition has been linked to various diseases, including rheumatoid arthritis, cardiomyopathy, and cancer. Important to this discussion is that cyclooxygenase-2 (COX-2) is over-expressed in all the aforesaid pathologies; however, a casual relationship between Se status and COX-2 expression remains to be established. The present study is based on the hypothesis that oxidant stress, a consequence of Se deficiency, lowers the activation potential of the redox-sensitive transcription factor, NF-kappaB, and that the activated NF-kappaB is required for the altered expression of COX-2. To test this hypothesis, we have investigated the relationship between Se status and COX-2 expression in response to LPS stimulation in RAW 264.7, a macrophage-like cell line. In Se-deficient cells, the Se-dependent glutathione peroxidase activity (Se-GPx), a measure of Se status, was markedly reduced and the overall oxidative stress was significantly higher than Se-supplemented cells. Upon lipopolysaccharide (LPS) stimulation, we found 2-3-folds higher COX-2 protein expression as well as higher PGE2 levels in Se-deficient cells than Se-supplemented cells. In comparison, COX-1 protein expression was not affected by either LPS stimulation or Se status. Following LPS stimulation, the nuclear localization of NF-kappaB was significantly increased in Se-deficient macrophages, thereby leading to increased expression of COX-2. This is the first report demonstrating an inverse relationship between Se status and the expression of COX-2.


Subject(s)
Isoenzymes/biosynthesis , Macrophage Activation/drug effects , Macrophages/enzymology , NF-kappa B/metabolism , Prostaglandin-Endoperoxide Synthases/biosynthesis , Selenium/deficiency , Animals , Blotting, Western , Cell Line , Cell Nucleus/physiology , Cyclooxygenase 1 , Cyclooxygenase 2 , Dinoprostone/metabolism , Electrophoretic Mobility Shift Assay , Free Radicals/metabolism , Gene Expression Regulation, Enzymologic , Glutathione Peroxidase/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Lipopolysaccharides/pharmacology , Luciferases/metabolism , Membrane Proteins , Mice , Mutagenesis, Site-Directed , NF-kappa B/genetics , Oxidative Stress/physiology , Promoter Regions, Genetic , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandin-Endoperoxide Synthases/metabolism , RNA, Messenger/analysis
13.
Toxicol Sci ; 82(1): 170-82, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15310864

ABSTRACT

Administration of phthalates is known to cause toxicity and liver cancer in rodents through the activation of peroxisome proliferator-activated receptors (PPARs), and the monoesters appear to be the active metabolites that function as ligands of PPARs. There is evidence that PPARs exhibit significant species differences in response to ligand activation. In this study, the activation of mouse and human PPARalpha, PPARbeta, and PPARgamma by a broad class of phthalate monoesters was investigated using a trans-activation assay, functional analysis of PPARalpha target gene expression, and a PPARgamma-mediated differentiation assay. These studies demonstrated a range in the ability of various phthalate monoesters to activate PPARalpha, with the mouse PPARalpha generally being activated at lower concentrations and exhibiting a greater response than human PPARalpha. Similarly, a range in the trans-activation of mouse PPARbeta by phthalate monoesters was also observed, but this effect was not found with human PPARbeta. A number of phthalate monoesters activated both mouse and human PPARgamma, with similar sensitivity being exhibited by both receptors. These studies show that the potency and efficacy of phthalate monoesters for the activation of PPARalpha and PPARgamma increase with increasing side-chain length. These studies also show that mouse PPARalpha and PPARbeta are generally activated at lower concentrations of phthalate monoesters than human PPARalpha and PPARbeta, and that both mouse and human PPARgamma exhibit similar sensitivity to phthalate monoesters. Lastly, there is a good relationship between the relative ability of phthalate monoesters to trans-activate PPARalpha and PPARgamma, and the relative induction of PPARalpha target gene mRNA and PPARgamma-mediated adipocyte differentiation, respectively.


Subject(s)
Environmental Pollutants/toxicity , Peroxisome Proliferator-Activated Receptors/biosynthesis , Phthalic Acids/toxicity , 3T3 Cells , Animals , Carcinoma, Hepatocellular , Cell Differentiation/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Mice , Peroxisome Proliferator-Activated Receptors/classification , Peroxisome Proliferator-Activated Receptors/genetics , RNA, Messenger/metabolism , Rats , Species Specificity , Structure-Activity Relationship , Transcriptional Activation/drug effects
14.
PPAR Res ; 2013: 121956, 2013.
Article in English | MEDLINE | ID: mdl-23737761

ABSTRACT

PPARß/δ is a ligand-activated transcription factor that regulates various cellular functions via induction of target genes directly or in concert with its associated transcriptional repressor, BCL-6. Matrix remodeling proteinases are frequently over-expressed in pancreatic cancer and are involved with metastasis. The present study tested the hypothesis that PPARß/δ is expressed in human pancreatic cancer cells and that its activation could regulate MMP-9, decreasing cancer cells ability to transverse the basement membrane. In human pancreatic cancer tissue there was significantly higher expression of MMP-9 and PPARß/δ, and lower levels of BCL-6 mRNA. PPARß/δ activation reduced the TNF α -induced expression of various genes implicated in metastasis and reduced the invasion through a basement membrane in cell culture models. Through the use of short hairpin RNA inhibitors of PPARß/δ, BCL-6, and MMP-9, it was evident that PPARß/δ was responsible for the ligand-dependent effects whereas BCL-6 dissociation upon GW501516 treatment was ultimately responsible for decreasing MMP-9 expression and hence invasion activity. These results suggest that PPARß/δ plays a role in regulating pancreatic cancer cell invasion through regulation of genes via ligand-dependent release of BCL-6 and that activation of the receptor may provide an alternative therapeutic method for controlling migration and metastasis.

15.
Mol Clin Oncol ; 1(3): 444-452, 2013 May.
Article in English | MEDLINE | ID: mdl-24649190

ABSTRACT

Fish oil contains the marine ω-3 polyunsaturated fatty acids (ω-3 PUFAs) docosahexaenoic (DHA) and eicosapentaenoic acid (EPA). The consumption of diets rich in these fatty acids is associated with a decreased incidence of prostate cancer. However, there is limited knowledge regarding the non-marine ω-3 PUFA α-linolenic acid (ALA). To study which ω-3 PUFAs are more effective in prostate cancer prevention, and whether the mechanisms of action are conserved between them, we investigated the effect of DHA, EPA and ALA on the human prostate cancer cell lines PC-3 and LNCaP. Different trends of inhibition of PC-3 cell proliferation were observed for the three ω-3 PUFA, with DHA having the most pronounced effects on cell proliferation, while ALA had the minimum effects of the three ω-3 PUFAs. All the ω-3 PUFAs decreased fatty acid synthase (FASN) mRNA. Concerning genes involved in inflammation, cell cycle and apoptosis, DHA regulated the most genes in all categories, followed by EPA and then ALA. In addition, DHA and EPA increased the gene expression of the pro-apoptotic protein activating transcription factor 3 mRNA. Moreover, these two fatty acids significantly induced apoptosis. In conclusion, while some mechanisms of cancer cell inhibition are conserved among ω-3 PUFA, the extent, magnitude, and duration of transcriptional changes vary for each individual fatty acid.

16.
Toxicol In Vitro ; 27(7): 2049-60, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23899473

ABSTRACT

The bacteriostat triclosan (2,4,4'-trichloro-2'-hydroxydiphenylether) (TCS) decreases rat serum thyroxine via putative nuclear receptor (NR) interaction(s) and subsequent transcriptional up-regulation of hepatic catabolism and clearance. However, due to the evolutionary divergence of the constitutive androstane and pregnane-X receptors (CAR, PXR), TCS-mediated downstream effects may be species-dependent. To test the hypothesis that TCS activates xenobiotic NRs across species, cell-based NR reporter assays were employed to assess potential activation of rat, mouse, and human PXR, and rat, mouse, and three splice variants of human CAR. TCS activated hPXR, acted as an inverse agonist of hCAR1, and as a weak agonist of hCAR3. TCS failed to activate rPXR in full-length receptor reporter assays, and instead acted as a modest inverse agonist of rCAR. Consistent with the rat data, TCS also failed to activate mPXR and was a modest inverse agonist of mCAR. These data suggest that TCS may interact with multiple NRs, including hPXR, hCAR1, hCAR3, and rCAR in order to potentially affect hepatic catabolism. Overall these data support the conclusion that TCS may interact with NRs to regulate hepatic catabolism and downstream thyroid hormone homeostasis in both rat and human models, though perhaps by divergent mechanisms.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cell Nucleus/drug effects , Hepatocytes/drug effects , Models, Biological , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Steroid/agonists , Triclosan/pharmacology , Animals , Anti-Infective Agents, Local/pharmacology , Cell Nucleus/metabolism , Constitutive Androstane Receptor , Drug Inverse Agonism , Genes, Reporter/drug effects , HEK293 Cells , Hep G2 Cells , Hepatocytes/metabolism , Humans , Kinetics , Mice , Pregnane X Receptor , Protein Isoforms/agonists , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Species Specificity
17.
J Nutr Biochem ; 23(5): 510-8, 2012 May.
Article in English | MEDLINE | ID: mdl-21764284

ABSTRACT

The trans-10, cis-12 (10e12z) conjugated linoleic acid (CLA) isomer of CLA is responsible for loss of lipid storage or adipose tissue in vitro or in vivo. This isomer also induces inflammatory signaling in both mouse and human adipocytes in vitro. However, when these events occur and whether they are significant enough to affect other cell types are unclear. In these experiments, the 3T3-L1 cell line has been used to examine the interaction between inflammatory signaling and decreased differentiation or lipid storage induced by 10e12z CLA. In assays measuring both lipid accumulation and gene expression, differentiating 3T3-L1 cells exhibit concurrent induction of inflammatory signaling, as measured by cyclooxygenase-2 expression, and a decrease in adipocyte marker gene expression. Furthermore, in fully differentiated adipocytes, as identified in microarray assays and confirmed with real-time polymerase chain reaction, 10e12z CLA also significantly affected expression of both matrix metalloprotein-3 (MMP-3), collagen VI α 3 ColVI alpha 3 (VIα3) and the cytokine epiregulin, demonstrating that the effects of 10e12z broadly impact adipocyte function. In agreement with other experimental systems, 10e12z CLA inhibited RAW 264.7 cell proliferation; however, in response to adipocyte-conditioned media, 10e12z-CLA-treated adipocytes induced proliferation of this cell line, suggesting that the effect of 10e12z CLA is context dependent. These results are largely consistent with the known activation of the inflammatory mediator nuclear factor-κB in adipocytes in vitro and in vivo by 10e12z CLA treatment and demonstrate that adipose is an important target tissue of this isomer that impacts other cell types.


Subject(s)
Adipocytes/drug effects , Cell Differentiation , Cytokines/metabolism , Linoleic Acids, Conjugated/pharmacology , Macrophages/drug effects , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Animals , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Macrophages/cytology , Macrophages/metabolism , Mice , Stereoisomerism
18.
J Nutr Biochem ; 23(4): 400-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21658928

ABSTRACT

Increased cholesterol efflux from macrophage-derived foam cells (MDFCs) is an important protective mechanism to decrease lipid load in the atherosclerotic plaque. Dietary alpha-linolenic acid (ALA), an omega-3 polyunsaturated fatty acid (PUFA), decreases circulating cholesterol, but its role in cholesterol efflux has not been extensively studied. Stearoyl CoA desaturase 1 (SCD1) is the rate-limiting enzyme in the synthesis of monounsaturated fatty acids (MUFAs). Endogenous MUFAs are preferentially incorporated into triglycerides, phospholipids and cholesteryl ester, which are abundant in atherosclerotic plaque. This study investigated the mechanisms by which ALA regulated SCD1 and subsequent effect on cholesterol storage and transport in MDFCs. Small interfering RNA (siRNA) also was applied to modify SCD1 expression in foam cells. Alpha-linolenic acid treatment and SCD1 siRNA significantly decreased SCD1 expression in MDFCs. The reduction of SCD1 was accompanied with increased cholesterol efflux and decreased intracellular cholesterol storage within these cells. Alpha-linolenic acid activated the nuclear receptor farnesoid-X-receptor, which in turn increased its target gene small heterodimer partner (SHP) expression, and decreased liver-X-receptor dependent sterol regulatory element binding protein 1c transcription, ultimately resulting in repressed SCD1 expression. In conclusion, repression of SCD1 by ALA favorably increased cholesterol efflux and decreased cholesterol accumulation in foam cells. This may be one mechanism by which dietary omega-3 PUFAs promote atherosclerosis regression.


Subject(s)
Cholesterol/metabolism , Foam Cells/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Stearoyl-CoA Desaturase/genetics , alpha-Linolenic Acid/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Fatty Acids, Monounsaturated/metabolism , Foam Cells/cytology , HEK293 Cells , Humans , Liver X Receptors , Macrophages/metabolism , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , RNA, Small Interfering , Receptors, Cytoplasmic and Nuclear/genetics , Stearoyl-CoA Desaturase/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Triglycerides/metabolism
19.
Nutr Metab (Lond) ; 8: 61, 2011 Aug 26.
Article in English | MEDLINE | ID: mdl-21871057

ABSTRACT

BACKGROUND: Walnuts significantly decrease total and low-density lipoprotein cholesterol in normo- and hypercholesterolemic individuals. No study to date has evaluated the effects of walnuts on cholesterol efflux, the initial step in reverse cholesterol transport, in macrophage-derived foam cells (MDFC). The present study was conducted to investigate the mechanisms by which walnut oil affects cholesterol efflux. METHODS: The extract of English walnuts (walnut oil) was dissolved in DMSO and applied to cultured THP-1 MDFC cells (0.5 mg/mL). THP-1 MDFC also were treated with human sera (10%, v:v) taken from subjects in a walnut feeding study. Cholesterol efflux was examined by liquid scintillation counting. Changes in gene expression were quantified by real time PCR. RESULTS: Walnut oil treatment significantly increased cholesterol efflux through decreasing the expression of the lipogenic enzyme stearoyl CoA desaturase 1 (SCD1) in MDFC. Alpha-linolenic acid (ALA), the major n-3 polyunsaturated fatty acids found in walnuts, recaptured SCD1 reduction in MDFC, a mechanism mediated through activation of nuclear receptor farnesoid-X-receptor (FXR). Postprandial serum treatment also increased cholesterol efflux in MDFC. When categorized by baseline C-reactive protein (CRP; cut point of 2 mg/L), subjects in the lower CRP sub-group benefited more from dietary intervention, including a more increase in cholesterol efflux, a greater reduction in SCD1, and a blunted postprandial lipemia. CONCLUSION: In conclusion, walnut oil contains bioactive molecules that significantly improve cholesterol efflux in MDFC. However, the beneficial effects of walnut intake may be reduced by the presence of a pro-inflammatory state. TRIAL REGISTRATION: ClinicalTrials.gov: NCT00938340.

20.
Mol Nutr Food Res ; 54 Suppl 1: S83-92, 2010 May.
Article in English | MEDLINE | ID: mdl-20013886

ABSTRACT

When incorporated into the diet, pistachios have a beneficial effect on lipid and lipoprotein profiles. However, little is known about potential anti-inflammatory properties. This study was conducted to determine whether pistachio oil and an organic extract from pistachio oil extract (PE) regulated expression of inflammation-related genes. A mouse macrophage cell line (RAW 264.7 cells) was treated with pistachio oil and gene expression microarray analyses were performed. Pistachio oil significantly affected genes involved in immune response, defense response to bacteria, and gene silencing, of which INF-induced protein with tetratricopeptide repeats 2 (Ifit-2) was the most dramatically reduced. PE reduced the LPS-induced Ifit-2 by 78% and the bioactive molecules contained in PE, linoleic acid, and beta-sitosterol recapitulated this inhibition. Promoter analysis identified two adjacent IFN-stimulated response elements, which lie between -110 and -85bp of the 5'-flanking region of the Ifit-2 promoter, as being responsive to LPS activation and inhibition by PE. Our results indicate that pistachio oil and bioactive molecules present therein decrease Ifit-2 expressions, and due to the sensitivity of this effect, this gene is a potential biomarker for monitoring diet-induced changes in inflammation.


Subject(s)
Inflammation/genetics , Interferons/pharmacology , Pistacia , Plant Oils/pharmacology , Proteins/genetics , Animals , Apoptosis Regulatory Proteins , Base Sequence , Biomarkers , Cell Line , Mice , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , RNA/genetics , RNA/isolation & purification , RNA-Binding Proteins , Replication Protein C/genetics , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL