Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
BMC Gastroenterol ; 16: 31, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26940566

ABSTRACT

BACKGROUND: Intestinal bacteria are thought to play a role in the pathogenesis of human inflammatory bowel disease (IBD). We investigated whether oral inoculation with specific intestinal bacteria increased colon inflammation in the multi-drug resistance 1a-deficient (Mdr1a (-/-) ) mouse model of IBD. METHODS: Five-week-old Mdr1a (-/-) mice (FVB background) and FVB mice were randomly assigned to one of two treatment groups (Control or Inoculation, n = 12 per group). All mice were fed AIN-76A rodent diet, and mice in the Inoculation groups also received a single oral bacterial inoculation consisting of twelve cultured Enterococcus species combined with conventional intestinal flora obtained from the gastrointestinal tract of healthy mice (EF.CIF). Body weight, food intake, and disease activity index (DAI) were assessed throughout the study, and at 21 or 24 weeks of age, inflammation was assessed post-mortem by determining colon length and histological injury score (HIS), and plasma serum amyloid A (SAA). RESULTS: Mdr1a (-/-) mice consumed more food than FVB mice at 13 weeks of age (P < 0.05). There was also a significant effect of genotype on body weight, with Mdr1a (-/-) mice weighing less than FVB mice throughout the study (P < 0.05) regardless of treatment, but there was no effect of inoculation on body weight (P > 0.25). Colon HIS of Mdr1a (-/-) mice was significantly higher than that of FVB mice in the Control (9.3 ± 4.7 (mean ± SD) vs. 0.58 ± 0.51; P < 0.001) and Inoculation (6.7 ± 5.1 vs. 0.92 ± 0.39; P < 0.001) groups. There was no difference in colon HIS of Mdr1a (-/-) mice in the Control group compared with Mdr1a (-/-) mice in the Inoculation group (P = 0.25), nor was there any difference in within-group variation of colon HIS in these two Mdr1a (-/-) groups. DAI was higher in Mdr1a (-/-) mice than in FVB mice, but there was no effect of treatment in either strain, nor were there any differences in colon length or plasma SAA. CONCLUSIONS: Inoculation of Mdr1a (-/-) mice with the EF.CIF inoculum described here does not increase colon inflammation or reduce the observed variability of inflammation.


Subject(s)
Colitis/microbiology , Colon/microbiology , Enterococcus , Inflammatory Bowel Diseases/microbiology , ATP Binding Cassette Transporter, Subfamily B/genetics , Animals , Body Weight , Colitis/immunology , Colitis/pathology , Colon/immunology , Colon/pathology , Diet , Disease Models, Animal , Feeding Behavior , Inflammation , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Mice , Mice, Knockout , Serum Amyloid A Protein/immunology
2.
J Nutr Biochem ; 27: 181-92, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26437580

ABSTRACT

The aim of this study was to provide insight into how curcumin reduces colon inflammation in the Mdr1a(-/-) mouse model of human inflammatory bowel disease using a combined transcriptomics and proteomics approach. Mdr1a(-/-) and FVB control mice were randomly assigned to an AIN-76A (control) diet or AIN-76A+0.2% curcumin. At 21 or 24weeks of age, colonic histological injury score (HIS) was determined, colon mRNA transcript levels were assessed using microarrays and colon protein expression was measured using 2D gel electrophoresis and LCMS protein identification. Colonic HIS of Mdr1a(-/-) mice fed the AIN-76A diet was higher (P<.001) than FVB mice fed the same diet; the curcumin-supplemented diet reduced colonic HIS (P<.05) in Mdr1a(-/-) mice. Microarray and proteomics analyses combined with new data analysis tools, such as the Ingenuity Pathways Analysis regulator effects analysis, showed that curcumin's antiinflammatory activity in Mdr1a(-/-) mouse colon may be mediated by activation of α-catenin, which has not previously been reported. We also show evidence to support curcumin's action via multiple molecular pathways including reduced immune response, increased xenobiotic metabolism, resolution of inflammation through decreased neutrophil migration and increased barrier remodeling. Key transcription factors and other regulatory molecules (ERK, FN1, TNFSF12 and PI3K complex) activated in inflammation were down-regulated by dietary intervention with curcumin.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/genetics , Colitis/prevention & control , Curcumin/administration & dosage , Diet , Disease Models, Animal , Inflammatory Bowel Diseases/pathology , Models, Molecular , Animals , Mice
3.
J Nutr Biochem ; 24(10): 1678-90, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23643524

ABSTRACT

Animal models are an important tool to understand the complex pathogenesis of inflammatory bowel diseases (IBDs). This study tested the anti-inflammatory potential of a green tea extract rich in polyphenols (GrTP) in the colon of the multidrug resistance targeted mutation (Mdr1a(-/-)) mouse model of IBD. Insights into mechanisms responsible for this reduction in inflammation were gained using transcriptome and proteome analyses. Mice were randomly assigned to an AIN-76A (control) or GrTP-enriched diet. At 21 or 24 weeks of age, a colonic histological injury score was determined for each mouse, colon mRNA transcript levels were assessed using microarrays, and colon protein expression was measured using two-dimensional gel electrophoresis and liquid chromatography-mass spectrometry protein identification. Mean colonic histological injury score of GrTP-fed Mdr1a(-/-) mice was significantly lower compared to those fed the control diet. Microarray and proteomics analyses showed reduced abundance of transcripts and proteins associated with immune and inflammatory response and fibrinogenesis pathways, and increased abundance of those associated with xenobiotic metabolism pathways in response to GrTP, suggesting that its anti-inflammatory activity is mediated by multiple molecular pathways. Peroxisome proliferator-activated receptor-α and signal transducer and activator of transcription 1 appear to be two key molecules which regulate these effects. These results support the view that dietary intake of polyphenols derived from green tea can ameliorate intestinal inflammation in the colon of a mouse model of IBD, and are in agreement with studies suggesting that consumption of green tea may reduce IBD symptoms and therefore play a part in an overall IBD treatment regimen.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/deficiency , Colitis/prevention & control , Colon/metabolism , Polyphenols/pharmacology , Animals , Colitis/pathology , Colon/drug effects , Colon/pathology , Inflammatory Bowel Diseases/prevention & control , Male , Mice , Models, Animal , PPAR alpha/physiology , Proteome , STAT1 Transcription Factor/physiology , Tea/chemistry , Transcriptome
4.
Biochim Biophys Acta ; 1807(6): 697-706, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20732296

ABSTRACT

Cancer cells are resistant to apoptosis and show a shift in energy production from mitochondrial oxidative phosphorylation to cytosolic glycolysis. Apoptosis resistance and metabolic reprogramming are linked in many cancer cells and both processes center on mitochondria. Clearly, mutated cancer cells escape surveillance and turn into selfish cells. However, many of the mechanisms that operate cellular metabolic control still function in cancer cells. This review describes the metabolic importance of glucose and glutamine, glycolytic enzymes, oxygen, growth cofactors and mitochondria and focuses on the potential role of bioactive food components, including micronutrients. The role of B- and A-vitamin cofactors in (mitochondrial) metabolism is highlighted and the cancer protective potential of omega-3 fatty acids and several polyphenols is discussed in relation to metabolic reprogramming, including the mechanisms that may be involved. Furthermore, it is shown that cancer cell growth reduction by limiting the growth cofactor folic acid seems to be associated with reversal of metabolic reprogramming. Altogether, reversal of metabolic reprogramming may be an attractive strategy to increase susceptibility to apoptotic surveillance. Food bioactive components that affect various aspects of metabolism may be important tools to reverse glycolytic to oxidative metabolism and enhance sensitivity to apoptosis. The success of such a strategy may depend on several actors, acting in concert. Growth cofactors may be one of these, which call for careful (re)evaluation of their function in normal and in cancer metabolism.


Subject(s)
Cell Proliferation , Food , Glycolysis/physiology , Neoplasms/diet therapy , Neoplasms/metabolism , Animals , Down-Regulation , Energy Metabolism/physiology , Humans , Models, Biological , Neoplasms/pathology , Nutritional Physiological Phenomena/physiology
5.
J Nutrigenet Nutrigenomics ; 2(1): 9-28, 2009.
Article in English | MEDLINE | ID: mdl-19776635

ABSTRACT

BACKGROUND/AIMS: Dietary n-3 polyunsaturated fatty acids can reduce inflammation via a range of mechanisms. This study tested the effect of dietary eicosapentaenoic acid (EPA) on intestinal inflammation using interleukin-10 gene-deficient (Il10(-/-)) mice. METHODS: At 35 days of age, 12 weaned Il10(-/-) and 12 C57 mice were randomly assigned to one of two modified AIN-76A diets, supplemented with 3.7% purified ethyl esters of either EPA (n-3) or oleic acid (OA, control). To identify genes relevant to colon inflammation, transcription profiling (microarrays and qRT-PCR) and bioinformatic analyses were used. RESULTS: In this study, dietary EPA reversed the decrease in colon fatty acid beta-oxidation gene expression observed in OA-fed Il10(-/-) compared to C57 mice. Il10(-/-) mice fed the OA diet showed decreased expression of antioxidant enzyme genes, as well as those involved in detoxification of xenobiotics, compared to C57 mice on the same diet. In contrast, dietary EPA increased the expression of these genes in Il10(-/-) mice. CONCLUSIONS: These data indicate that dietary EPA-induced endogenous lipid oxidation which might have a potential anti-inflammatory effect on colon tissue. This is supported by the activation of the Ppara gene that regulates the expression of pro-inflammatory and immunomodulatory genes and proteins.


Subject(s)
Colitis/chemically induced , Colitis/genetics , Eicosapentaenoic Acid/adverse effects , Interleukin-10/genetics , Oleic Acid/adverse effects , Animals , Body Weight/drug effects , Body Weight/genetics , Body Weight/physiology , Dietary Fats, Unsaturated/adverse effects , Eating/drug effects , Eating/genetics , Eating/physiology , Gene Expression Profiling , Gene Regulatory Networks , Genome-Wide Association Study , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Oligonucleotide Array Sequence Analysis , Serum Amyloid A Protein/analysis
6.
Appl Environ Microbiol ; 75(19): 6198-204, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19684171

ABSTRACT

Probiotics are live microorganisms which, when administered in adequate amounts, confer a health benefit on the host. Therefore, probiotic strains should be able to survive passage through the human gastrointestinal tract. Human gastrointestinal tract survival of probiotics in a low-fat spread matrix has, however, never been tested. The objective of this randomized, double-blind, placebo-controlled human intervention study was to test the human gastrointestinal tract survival of Lactobacillus reuteri DSM 17938 and Lactobacillus rhamnosus GG after daily consumption of a low-fat probiotic spread by using traditional culturing, as well as molecular methods. Forty-two healthy human volunteers were randomly assigned to one of three treatment groups provided with 20 g of placebo spread (n = 13), 20 g of spread with a target dose of 1 x 10(9) CFU of L. reuteri DSM 17938 (n = 13), or 20 g of spread with a target dose of 5 x 10(9) CFU of L. rhamnosus GG (n = 16) daily for 3 weeks. Fecal samples were obtained before and after the intervention period. A significant increase, compared to the baseline, in the recovery of viable probiotic lactobacilli in fecal samples was demonstrated after 3 weeks of daily consumption of the spread containing either L. reuteri DSM 17938 or L. rhamnosus GG by selective enumeration. In the placebo group, no increase was detected. The results of selective enumeration were supported by quantitative PCR, detecting a significant increase in DNA resulting from the probiotics after intervention. Overall, our results indicate for the first time that low-fat spread is a suitable carrier for these probiotic strains.


Subject(s)
Gastrointestinal Tract/microbiology , Lacticaseibacillus rhamnosus/physiology , Limosilactobacillus reuteri/physiology , Microbial Viability , Probiotics/administration & dosage , Probiotics/pharmacology , Administration, Oral , Adolescent , Adult , Colony Count, Microbial , Double-Blind Method , Feces/microbiology , Female , Human Experimentation , Humans , Male , Middle Aged , Placebos/administration & dosage , Young Adult
7.
Br J Nutr ; 101(2): 169-81, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18761777

ABSTRACT

Damage of the intestinal epithelial barrier by xenobiotics or reactive oxygen species and a dysregulated immune response are both factors involved in the pathogenesis of inflammatory bowel diseases (IBD). Curcumin and rutin are polyphenolic compounds known to have antioxidant and anti-inflammatory activities, but their mechanism(s) of action are yet to be fully elucidated. Multidrug resistance gene-deficient (mdr1a-/- ) mice spontaneously develop intestinal inflammation, predominantly in the colon, with pathology similar to IBD, so this mouse model is relevant for studying diet-gene interactions and potential effects of foods on remission or development of IBD. The present study tested whether the addition of curcumin or rutin to the diet would alleviate colonic inflammation in mdr1a-/- mice. Using whole-genome microarrays, the effect of dietary curcumin on gene expression in colon tissue was also investigated. Twelve mice were randomly assigned to each of three diets (control (AIN-76A), control +0.2% curcumin or control +0.1% rutin) and monitored from the age of 7 to 24 weeks. Curcumin, but not rutin, significantly reduced histological signs of colonic inflammation in mdr1a-/- mice. Microarray and pathway analyses suggested that the effect of dietary curcumin on colon inflammation could be via an up-regulation of xenobiotic metabolism and a down-regulation of pro-inflammatory pathways, probably mediated by pregnane X receptor (Pxr) and peroxisome proliferator-activated receptor alpha (Ppara) activation of retinoid X receptor (Rxr). These results indicate the potential of global gene expression and pathway analyses to study and better understand the effect of foods in modulating colonic inflammation.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Curcumin/administration & dosage , Gene Expression Regulation/drug effects , Inflammatory Bowel Diseases/prevention & control , Rutin/administration & dosage , ATP Binding Cassette Transporter, Subfamily B/genetics , Animals , Base Sequence , Colitis/genetics , Colitis/pathology , Colitis/prevention & control , Colon/metabolism , Colon/pathology , Dietary Supplements , Fibrosis , Gene Expression/drug effects , Genome-Wide Association Study/methods , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Liver/pathology , Mice , Mice, Knockout , Models, Animal , Molecular Sequence Data , Random Allocation , Reverse Transcriptase Polymerase Chain Reaction/methods , Staining and Labeling
8.
Free Radic Biol Med ; 39(11): 1489-98, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16274884

ABSTRACT

Glutathione (GSH) plays an important role in cellular defense response in many in vitro and in vivo models. Here we investigated its role in NO()-induced toxicity in cell culture and mouse models. Wild-type (TK6) and p53-null (NH32) human lymphoblastoid cells were treated with NO(.) at a steady-state concentration of 0.6 muM, similar to the level estimated to occur in inflamed tissues. In both cell types, GSH was depleted by this exposure in a dose- and time-dependent manner. Contrary to expectations, prior depletion of GSH by treatment with l-buthionine-SR-sulfoximine did not potentiate NO(.)-induced cell killing or DNA deamination in TK6 cells. In activated RAW264.7 murine macrophages producing NO(.), intracellular GSH content did not change, although gamma-glutamate-cysteine ligase was upregulated. NO(.) overproduction in RcsX lymphoma-bearing SJL mice resulted in significantly elevated GSH levels in various organs. Administration of the NO(.) synthase inhibitor N-methylarginine abolished the increase in GSH in these animals. Collectively, these data indicate a multifaceted and complex involvement of GSH in responses of cells and tissues to toxic levels of NO(.). NO(.) treatment effectively depleted GSH levels in human lymphoblastoid cells, but this alteration was not a critical initiating factor for NO(.)-mediated toxicity. Murine macrophages maintained GSH homeostasis when exposed to endogenously produced NO(.). In RcsX lymphoma-bearing mice, upregulation of de novo synthesis of GSH appeared to be a response to the toxic effects of NO(.).


Subject(s)
Glutathione/physiology , Nitric Oxide/toxicity , Animals , Cell Line , Cell Survival , Humans , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Macrophages , Mice , Tumor Cells, Cultured
9.
Food Chem Toxicol ; 41(12): 1739-47, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14563399

ABSTRACT

Modulating effects of high fat fish oil (HFFO) and high fat corn oil (HFCO) diets on azoxymethane (AOM)-induced colonic aberrant crypt foci (ACF) were studied in male F344 rats following 8 weeks of dietary treatment. The incidence of AOM-induced ACF was significantly lower in the proximal colon of rats fed the HFFO diets compared with rats fed the HFCO diets. No differential effects were found on enzyme activities that are involved in metabolic activation and detoxification of AOM. Activities of hepatic P450 IAI and P450 IIBI and hepatic and feacal levels of lipid peroxidation were increased by feeding the HFFO diet. Hepatic GST activity and plasma levels of PGE(2) were significantly lower in rats fed the HFFO diets compared with those fed the HFCO diets. These observations demonstrate that HFFO diets with high levels of n-3 PUFAs are also protective against preneoplastic lesions in the early stages of chemically induced colon carcinogenesis. It seems unlikely from our results that the inhibitory effect of a HFFO diet can be attributed to an altered metabolic activation and detoxification of AOM. Other mechanisms such as oxidative stress or reduction of PGE(2) levels may play an important role in the anticarcinogenic effects of n-3 PUFAs.


Subject(s)
Azoxymethane/antagonists & inhibitors , Azoxymethane/toxicity , Carcinogens/antagonists & inhibitors , Carcinogens/toxicity , Choristoma/chemically induced , Colonic Diseases/chemically induced , Corn Oil/pharmacology , Diet/adverse effects , Fish Oils/pharmacology , Animals , Body Weight/drug effects , Cecum/drug effects , Cecum/enzymology , Choristoma/pathology , Colonic Diseases/pathology , Dinoprostone/metabolism , Eating/drug effects , Epithelial Cells/pathology , Feces/chemistry , Lipid Peroxidation/drug effects , Liver/drug effects , Liver/enzymology , Male , Organ Size/drug effects , Rats , Rats, Inbred F344
10.
Carcinogenesis ; 24(3): 385-92, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12663496

ABSTRACT

This study was conducted to investigate the role of the enzyme cyclooxygenase (COX) and its prostaglandin product PGE(2) in n-6 and n-3 polyunsaturated fatty acid (PUFA)-mediated effects on cellular proliferation of two human colorectal carcinoma cell lines. The long chain PUFAs eicosapentaenoic acid (EPA; 20:5n-3) and arachidonic acid (AA; 20:4n-6) both inhibited cell proliferation of Caco-2 cells compared with the long chain fatty acids alpha-linolenic acid (ALA; 18:3n-3) and linoleic acid (LA; 18:2n-6). Neither incubation with PGE(2) nor reduction in PGE(2) synthesis by EPA compared with AA led to differential effects on cell proliferation in Caco-2 cells. This suggests that n-6 and n-3 PUFA-mediated cell proliferation in Caco-2 cells is not regulated via PGE(2) levels. AA and EPA had no effect on growth of HT-29 colon cancer cells with a low COX activity. However, stimulation of COX-2 activity by IL-1 beta resulted in a decrease in cell proliferation and an induction of cytotoxicity by AA as well as by EPA. Both inhibition of the COX pathway by indomethacin as well as inhibition of direct lipid peroxidation by antioxidants such as vitamin E and C diminished the anti-proliferative effects of AA as well as EPA. Also, malondialdehyde, a product of lipid peroxidation and COX-activity was decreased by addition of vitamin E and partially decreased by indomethacin. These data support the hypothesis that growth inhibitory and cytotoxic effects of PUFAs with methylene-interrupted double bonds such as AA and EPA are due to peroxidation products that are generated during lipid peroxidation and COX activity.


Subject(s)
Cell Division/drug effects , Colorectal Neoplasms/pathology , Dinoprostone/biosynthesis , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Unsaturated/pharmacology , Prostaglandin-Endoperoxide Synthases/metabolism , Antioxidants/pharmacology , Caco-2 Cells , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/metabolism , Cyclooxygenase Inhibitors/pharmacology , Fatty Acids, Omega-6 , HT29 Cells , Humans , Lipid Peroxidation
11.
Nutr Cancer ; 42(1): 125-30, 2002.
Article in English | MEDLINE | ID: mdl-12235644

ABSTRACT

Gap junctional intercellular communication (GJIC), which modulates cell growth and differentiation, may play an important role in tumor growth. Cancer cells have dysfunctional GJIC, but it is not known whether GJIC is mechanistically involved in the carcinogenic and anti-carcinogenic effects of n-6 and n-3 polyunsaturated fatty acids (PUFAs) on colon tumor cells. Caco-2 cells were used as an in vitro model to study the effects of PUFAs on differentiated as well as undifferentiated human colon cells. The GJIC capacity of this cell line increased during spontaneous differentiation. However, no differential effects between n-6 and n-3 PUFAs on GJIC were observed. Short-term incubation with linoleic acid (18:2n-6), alpha-linolenic acid (18:3n-3), arachidonic acid (AA, 20:4n-6), and eicosapentaenoic acid (EPA, 20:5n-3) did not influence GJIC, while long-term incubation (> 10 days) with linoleic acid and alpha-linolenic acid inhibited GJIC of these colon cells. Long-chain metabolites such as AA and EPA were not formed after incubation with linoleic acid and alpha-linolenic acid, thus excluding the involvement of prostaglandins in the observed effects. Although the exact mechanism of GJIC inhibition is unclear, cytotoxicity probably mediated by lipid peroxidation products seems to be related, because incubation with more PUFAs (AA and EPA) completely abolished GJIC.


Subject(s)
Adenocarcinoma/pathology , Cell Communication/drug effects , Colonic Neoplasms/pathology , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Unsaturated/pharmacology , Gap Junctions/drug effects , Intercellular Junctions/drug effects , Caco-2 Cells , Cell Differentiation/drug effects , Cell Division/drug effects , Fatty Acids, Omega-6 , Gap Junctions/physiology , Humans , Intercellular Junctions/physiology , L-Lactate Dehydrogenase/metabolism , Tumor Cells, Cultured
12.
Environ Toxicol Pharmacol ; 11(3-4): 297-308, 2002 Jul.
Article in English | MEDLINE | ID: mdl-21782613

ABSTRACT

During the past few decades, many studies have been conducted to evaluate the effects of n-6 and n-3 polyunsaturated fatty acids (PUFAs) on colorectal carcinogenesis. This report provides a brief overview of the recent studies that have been performed in cultured colon cells, animal models as well as of the population-based and short-term biomarker studies with humans. No differential effect between n-6 and n-3 PUFAs has been observed in vitro. Results from animal models indicate that n-6 PUFAs have a tumor enhancing effect, predominantly during the post-initiation phase. n-3 PUFAs may protect against colorectal carcinogenesis during both the initiation and post-initiation phase. Population-based human studies show little or no associations between n-6 or n-3 PUFA intake and colorectal cancer. Short-term biomarker studies in humans suggest though that fish oil (FO) supplementation with high amounts of n-3 PUFAs may protect against colorectal carcinogenesis and that n-6 PUFA supplementation may increase the risk.

SELECTION OF CITATIONS
SEARCH DETAIL
...