Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Food Funct ; 10(7): 4210-4219, 2019 Jul 17.
Article in English | MEDLINE | ID: mdl-31257391

ABSTRACT

Identifying tolerance responses to ingested foodstuff over life is essential for understanding dysfunction in metabolic diseases. This study presents a comparative structural and functional analysis of serine-type protease inhibitors (STPIs) from Chenopodium quinoa, Salvia hispanica L., Avena sativa and Triticum durum. It also evaluates their influence on an in vivo hepatocarcinoma (HCC) model. STPIs are found in all samples with significant differences in protease inhibitory capacity: C. quinoa = S. hispanica < A. sativa = T. durum. STPIs in C. quinoa and S. hispanica appear as heterologous complexes, while those in A. sativa are present as homologous complexes. T. durum provides different subunits with STPI capacity. HPLC-RP-ESI analyses revealed homology between STPIs in the different samples and the partial resistance of those to simulated gastrointestinal digestion. In vivo, STPIs from S. hispanica showed the most positive effects, increasing F4/80+ cells normalizing the expression (mRNA) of CD36 and the innate immune 'Toll-like' receptor (TLR)-4. Only STPIs from C. quinoa and S. hispanica did not impair the production of inflammatory mediators (granulocyte-monocyte colony stimulating factor, stem cell factor and TNFα), contributing to maintaining the polarization of the antitumoral M1 macrophage phenotype. These structural and functional features of STPIs from C. quinoa and S. hispanica can be used to control HCC aggressiveness.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Immunity, Innate , Liver Neoplasms/drug therapy , Plant Extracts/pharmacology , Protease Inhibitors/pharmacology , Seeds/chemistry , Tumor Microenvironment/drug effects , Animals , Avena/chemistry , CD36 Antigens/metabolism , Chenopodium quinoa/chemistry , Cytokines/metabolism , Digestion , Flour , Gastrointestinal Tract , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Salvia/chemistry , Stem Cell Factor/metabolism , Toll-Like Receptor 4/metabolism , Triticum/chemistry , Tumor Necrosis Factor-alpha/metabolism
2.
J Dairy Sci ; 99(12): 9395-9404, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27771084

ABSTRACT

The aim of this study was to examine immune adaptive changes, the expression of innate biomarkers and variations in intestinal microbiota composition after horse-milk administration in BALB/c mice, which were sensitized intraperitoneally using cow ß-lactoglobulin and α-casein with aluminum adjuvant. We measured serum antibody IgE levels and the expression of MCP-1, IL-4, and TNF-α in duodenal samples. Changes in immune cell populations in peripheral blood were quantified using flow cytometry, and intestinal microbiota composition was assessed using real-time PCR. We found that horse-milk administration decreased serum IgE levels in sensitized mice. The groups that received horse milk showed an increased population of regulatory T cells (CD4+Foxp3+). Horse-milk administration decreased the mRNA levels of IL-4 and resulted in higher transcripts of TLR-4 in all treatment groups; however, the levels of MCP-1, TNF-α, and TLR-2 were unaltered. After horse-milk treatment, we observed a positive effect, with increased numbers of intestinal Bifidobacterium spp. We observed immune-modulating properties of horse milk, but future studies should focus on testing horse-milk processing, such as fermentation and destroying most allergenic epitopes to continue research under clinical conditions.


Subject(s)
Lactoglobulins/immunology , Milk , Animals , Caseins/genetics , Cattle , Female , Immunoglobulin E/blood , Mice , Milk Hypersensitivity/immunology
3.
Food Chem Toxicol ; 95: 89-95, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27377345

ABSTRACT

Cereal proteins are of clinical interest because of their cytotoxic and immunogenic features being associated to allergic processes and intestinal disorders. In addition to gliadins, there has been suggested an important role for non-gluten modulating factors (nGMF) on the onset of intestinal inflammatory processes. In this study, the amino acid sequences generated after a simulated human gastrointestinal (sGI) digestion of a commercial extract of gliadins (GEF) and the nGMF-enriched fraction (nGEF) obtained from them were characterized (nanoESI-qQTOF). These fractions were fed (20 days) to Wistar rats, sensitized with interferon (IFN)-γ (1000 IU/rat) and further treated with indomethacin (2 mg/kg). The production of inflammatory mediators (ELISAs) and the expression (rt-qPCR) of innate biomarkers was monitored in duodenal tissue sections. There were also evaluated changes in defined leukocyte (flow cytometry) populations quantified in peripheral blood samples. Expected nGMF components, CM3 and 0.19, as well as toxic gliadin-derived peptides were generated after sGI digestion. Rats fed with the nGEF showed higher concentrations of IFNγ, as well as expression levels of TLR-4 and the peroxisome proliferator activated receptor-α in duodenal samples than those animals fed with GEF. Rats fed with GEF showed higher expression levels of the endocannabinoid receptor-1 and an increased CD4(+)CD3(+)Foxp3(+) cell population. The data points out significant different cytotoxic and immunogenic potential of the nGEF when administered independently of the GEF to which are commonly associated. However, proinflammatory responses to nGMF are masked when present associated to gliadins.


Subject(s)
Gliadin/pharmacology , Glutens/pharmacology , Indomethacin/pharmacology , Inflammation/pathology , Interferon-gamma/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antiviral Agents/pharmacology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , HEK293 Cells , Humans , Inflammation/drug therapy , Inflammation/metabolism , Lymphocytes/drug effects , Macrophages/drug effects , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction
4.
Food Chem Toxicol ; 84: 29-36, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26140950

ABSTRACT

Recent studies have expanded the appreciation of the roles of oxysterols triggering inflammatory, immune cytotoxic and apoptotic processes, but have not been considered for proteome analysis. A comparative proteomic study in intestinal epithelial cell cultures incubated (60 µM/24 h) with 7keto-cholesterol or 7keto-stigmasterol was performed. The influence of both compounds was studied following the nLC-TripleTOF analysis. Findings were compared to results for control cultures. In the principal component analysis (PCA) of proteome patterns, two components were extracted accounting for 99.8% of the variance in the protein expression. PCA analysis clearly discriminated between the perturbations in the proteome of cell cultures incubated with 7keto-cholesterol and 7keto-stigmasterol. These proteins participate in mitochondrial function, lipid homeostasis, inflammation and immunity and cell proliferation. Remarkable differences between proteome patterns in cell cultures exposed to 7keto-cholesterol and 7keto-stigmasterol affect macrophage migration inhibitory factor, apolipoprotein E, Bcl-2-associated transcription factor and cellular retinoic acid-binding protein. Besides, exposure to 7keto-stigmasterol increased the concentration of ubiquitin-conjugating enzyme E2 and the mitochondrial superoxide dismutase protein. Such findings raise new questions about safety studies and the regulatory potential of oxysterols in the differentiation and function of intestinal and associated immune cells, their response to environmental stimuli and impairment of absorption processes.


Subject(s)
Enterocytes/drug effects , Gene Expression Regulation/drug effects , Ketocholesterols/toxicity , Oxidants/toxicity , Proteome/drug effects , Stigmasterol/analogs & derivatives , Caco-2 Cells , Enterocytes/enzymology , Enterocytes/metabolism , Gene Expression Profiling , Humans , Peptide Mapping , Principal Component Analysis , Proteome/metabolism , RNA, Messenger/metabolism , Stigmasterol/toxicity
5.
Food Chem Toxicol ; 69: 140-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24747512

ABSTRACT

High dietary intakes of cholesterol together with sedentary habits have been identified as major contributors to atherosclerosis. The latter has long been considered a cholesterol storage disease; however, today atherosclerosis is considered a more complex disease in which both innate and adaptive immune-inflammatory mechanisms as well as bacteria play a major role, in addition to interactions between the arterial wall and blood components. This scenario has promoted nutritional recommendations to enrich different type of foods with plant sterols (PS) because of their cholesterol-lowering effects. In addition to cholesterol, PS can also be oxidized during food processing or storage, and the oxidized derivatives, known as phytosterol oxidation products (POPs), can make an important contribution to the negative effects of both cholesterol and cholesterol oxidation oxides (COPs) in relation to inflammatory disease onset and the development of atherosclerosis. Most current research efforts have focused on COPs, and evaluations of the particular role and physiopathological implications of specific POPs have been only inferential. Appreciation of the inflammatory role described for both COPs and POPs derived from foods also provides additional reasons for safety studies after long-term consumption of PS. The balance and relevance for health of all these effects deserves further studies in humans. This review summarizes current knowledge about the presence of sterol oxidation products (SOPs) in foods and their potential role in inflammatory process and cardiovascular disease.


Subject(s)
Cardiovascular Diseases/etiology , Food/adverse effects , Inflammation/etiology , Phytosterols/pharmacology , Atherosclerosis/etiology , Biological Availability , Cardiovascular Diseases/chemically induced , Cholesterol/metabolism , Food Handling , Humans , Inflammation/chemically induced , Inflammation/metabolism , Phytosterols/adverse effects , Phytosterols/metabolism , Phytosterols/pharmacokinetics , Risk Factors
6.
Food Chem Toxicol ; 56: 247-53, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23454145

ABSTRACT

Human diets contain sterol oxidation products that can induce cytotoxic effects, mainly caused by cholesterol oxides. However, phytosterol oxides effects have been less extensively investigated. This study evaluates the production of inflammatory biomarkers (IL-1ß, IL-8, IL-10, TNFα) and the influence of gene expression transporters and enzymes related to cholesterol absorption and metabolism (NPC1L1, ABCG5/8, HMGCoA, ACAT) produced by 7-ketosterols (stigmasterol/cholesterol) in Caco-2 cells. These effects were linked to intracellular signaling pathways by using several inhibitors. Results showed 7-ketostigmasterol to have a greater proinflammatory potential than 7-ketocholesterol. In non-pre-treated cells, only efflux transporters were down-regulated by 7-ketosterols, showing a greater influence upon ABCG5 expression. Cell-pre-incubation with bradykinin induced changes in ABCG expression levels after 7-ketostigmasterol-incubation; however, the energetic metabolism inhibition reduced NPC1L1 expression only in 7-ketocholesterol-incubated cells. In non-pre-treated cells, HMG-CoA was up-regulated by both 7-ketosterols. However, exposure to inhibitors down-regulated the expression levels, mainly in 7-ketocholesterol-incubated cells. While ACAT expression values in non-pre-treated cells were unchanged, exposure to inhibitors caused down-regulation of mRNA levels. These results suggest that internalization and excretion of 7-ketostigmasterol is probably influenced by [Ca]i, which also could mediate HMGCoA activity in POPs metabolism. However, energetic metabolism and reducing equivalents exert different influences upon the 7-ketosterol internalization.


Subject(s)
Biomarkers/metabolism , Inflammation/pathology , Ketocholesterols/pharmacokinetics , Stigmasterol/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Acetyl-CoA C-Acetyltransferase/genetics , Acetyl-CoA C-Acetyltransferase/metabolism , Acyl Coenzyme A/genetics , Acyl Coenzyme A/metabolism , Anticholesteremic Agents/pharmacology , Biological Transport/drug effects , Bradykinin/pharmacology , Caco-2 Cells , Down-Regulation , Humans , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Lipoproteins/genetics , Lipoproteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Transport Proteins , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
7.
Food Chem Toxicol ; 50(9): 3106-13, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22750387

ABSTRACT

The biological implications of cholesterol oxidation products have been investigated, though research on plant sterol oxidation products is scarce and in some cases contradictory. The cytotoxicity of 7keto(k)-stigmasterol versus 7keto(k)-cholesterol at different concentrations (0-120 µM) and incubation times (4-24h), in intestinal epithelial cells (Caco-2 cells) was evaluated. The 3-[4,5-dimethylthiazol-2-yl]-2,3-diphenyl tetrazolium bromide and neutral red uptake tests, mitochondrial membrane potential (ΔΨm), and relative DNA and RNA contents in the cell cycle phases were determined. Possible interaction effects between 7k-derivatives or non-oxidized stigmasterol were monitored. Endo/lysosomal activity was not impaired by either oxide. 7k-cholesterol showed a deleterious effect upon the mitochondrial compartment after 24h of exposure (120 µM), as well as upon ΔΨm when incubated at all concentrations (12/24h). Only cells incubated with 7k-cholesterol (120 µM) exhibited a decrease in RNA proportion in the G1 population. The presence of 7k-stigmasterol or stigmasterol with 7k-cholesterol reduced the deleterious metabolic effects upon mitochondrial functionality and integrity and the distribution of RNA contents in G1 and G2 phases. A decrease in the G1 phase proportion was detected in cells exposed to mixtures, without alterations in RNA content. The results obtained indicate the absence of 7k-stigmasterol cytotoxicity in Caco-2 cells and its capacity to reduce 7k-cholesterol toxicity.


Subject(s)
Cholesterol/toxicity , Intestines/drug effects , Stigmasterol/toxicity , Caco-2 Cells , Drug Evaluation, Preclinical , Humans , Intestines/cytology , Membrane Potential, Mitochondrial/drug effects
8.
Appl Environ Microbiol ; 78(18): 6507-15, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22773639

ABSTRACT

Celiac disease (CD) is associated with intestinal dysbiosis, which can theoretically lead to dysfunctions in host-microbe interactions and contribute to the disease. In the present study, possible differences in Bacteroides spp. and their pathogenic features between CD patients and controls were investigated. Bacteroides clones (n = 274) were isolated, identified, and screened for the presence of the virulence genes (bft and mpII) coding for metalloproteases. The proteolytic activity of selected Bacteroides fragilis strains was evaluated by zymography and, after gastrointestinal digestion of gliadin, by high-pressure liquid chromatography/electrospray ionization/tandem mass spectrometry. The effects of B. fragilis strains on Caco-2 cell culture permeability and inflammatory response to digested gliadin were determined. B. fragilis was more frequently identified in CD patients than in healthy controls, in contrast to Bacteroides ovatus. B. fragilis clones carrying virulence genes coding for metalloproteases were more abundant in CD patients than in controls. B. fragilis strains, representing the isolated clones and carrying metalloprotease genes, showed gelatinase activity and exerted the strongest adverse effects on the integrity of the Caco-2 cell monolayer. All B. fragilis strains also showed gliadin-hydrolyzing activity, and some of them generated immunogenic peptides that preserved or increased inflammatory cytokine production (tumor necrosis factor alpha) and showed increased ability to permeate through Caco-2 cell cultures. These findings suggest that increased abundance of B. fragilis strains with metalloprotease activities could play a role in CD pathogenesis, although further in vivo studies are required to support this hypothesis.


Subject(s)
Bacteroides fragilis/pathogenicity , Celiac Disease/microbiology , Celiac Disease/pathology , Metalloproteases/genetics , Virulence Factors/genetics , Bacteroides fragilis/enzymology , Bacteroides fragilis/genetics , Caco-2 Cells , Cell Membrane Permeability/drug effects , Chromatography, High Pressure Liquid , Cytokines/biosynthesis , Epithelial Cells/drug effects , Gliadin/metabolism , Humans , Metalloproteases/metabolism , Proteolysis , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry , Virulence Factors/metabolism
9.
Nutr Hosp ; 26(3): 614-21, 2011.
Article in English | MEDLINE | ID: mdl-21892583

ABSTRACT

INTRODUCTION: Fruit beverages are commonly supplemented with milk, vitamins and/or minerals in order to improve their healthy effects by providing some bioactive components that can act additively or synergistically against oxidative stress. AIMS: To test whether iron, zinc, and milk added to fruit beverages do not affect the cytoprotective effect against oxidative damage to Caco-2 cells through GSH-related enzymes induction and cell cycle progression preservation, in comparison with non-supplemented fruit beverage. METHODS: Caco-2 cells were incubated 24 h with the bioaccessible fraction (BF) of eight fruit beverages with/without iron and/or zinc, and/or milk, and then challenged with H2O2 (5 mmol L-1 -2 h). Mitochondrial enzyme activities (MTT test), GSH-Rd and GSH-Px enzyme activities, cell cycle progression and caspase-3 activity were measured. RESULTS AND DISCUSSION: Fruit beverages prevented the deleterious effect of H2O2 on cell viability, with almost all samples reaching control basal levels. Only independent iron or zinc supplementation with/without milk exerted positive effects upon GSH-Rd activity. Both minerals with milk, afforded improved preservation of GSH-Px activity. All samples prevented the decrease in the G1 phase of cell cycle induced by H2O2, except iron supplemented samples with/without milk, but none of them avoided the increase in sub-G1 phase. However, this fact was not associated to caspase-3 activity, with a probable positive effect of zinc upon this parameter. CONCLUSION: Mineral and/or milk supplementation of fruit beverages helps in the prevention of oxidative stress in Caco-2 cells based on cell viability maintenance, GSH-related enzymes activation, cell cycle distribution preservation and inhibition of caspase-3 activation.


Subject(s)
Beverages , Fruit/chemistry , Hydrogen Peroxide/antagonists & inhibitors , Hydrogen Peroxide/pharmacology , Milk , Minerals/pharmacology , Oxidative Stress/drug effects , Animals , Caco-2 Cells , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Survival/drug effects , Dietary Supplements , Digestion/physiology , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism , Humans , Mitochondria/drug effects , Mitochondria/enzymology
10.
Int J Biol Macromol ; 47(4): 458-64, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20637226

ABSTRACT

Dietary components may have an important role in maintaining a balanced gut microbiota composition. Celiac disease is an autoimmune enteropathy caused by gliadins, and has been associated with a reduced proportion of Bifidobacterium in gut microbiota. This study evaluates the influence of glycosaminoglycans (GAGs) on bacterial adhesion and their contribution in the gliadins-induced inflammatory response. The adhesion of potential probiotic (Bifidobacterium longum CECT 7347 and Bifidobacterium bifidum CECT 7365), commensal (Escherichia coli and Bacteroides fragilis) and pathogenic (Salmonella enterica CECT 443 and Listeria monocytogenes CECT 935) bacteria to mucin and Caco-2 cell cultures was determined. Gliadins were subjected to in vitro digestion (pepsin/pancreatin-bile), with/out GAGs, and the presence or not of cell suspensions of B. longum (10(8) CFU/ml). B. longum, E. coli, and L. monocytogenes, markedly interact with the high-sulphur-containing fraction of GAGs. The GAGs reduced the gliadins-mediated production of interleukin-1ß, but not tumour necrosis factor-α. The results suggest that GAGs may ameliorate gliadin-induced inflammatory response, though they also slightly interfere with the action of B. longum.


Subject(s)
Bacterial Adhesion/drug effects , Diet , Enterocytes/drug effects , Enterocytes/pathology , Glycosaminoglycans/pharmacology , Inflammation/pathology , Animals , Caco-2 Cells , Chemical Fractionation , Chromatography, High Pressure Liquid , Chromatography, Reverse-Phase , Fishes , Gliadin , Humans , Interleukin-1beta/biosynthesis , Plant Lectins/metabolism , Soybean Proteins/metabolism , Spectroscopy, Fourier Transform Infrared , Tumor Necrosis Factor-alpha/biosynthesis
11.
J Cell Biochem ; 109(4): 801-7, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20052669

ABSTRACT

Celiac disease (CD) is a chronic enteropathy triggered by intake of gliadin, the toxic component of gluten. This study aims at evaluating the capacity of different Bifidobacterium strains to counteract the inflammatory effects of gliadin-derived peptides in intestinal epithelial (Caco-2) cells. A commercial extract of several gliadin (Gld) types (alpha, beta, gamma, [symbol: see text] ) was subjected to in vitro gastrointestinal digestion (pepsin at pH 3, pancreatin-bile at pH 6), inoculated or not with cell suspensions (10(8) colony forming units/ml) of either B. animalis IATA-A2, B. longum IATA-ES1, or B. bifidum IATA-ES2, in a bicameral system. The generated gliadin-derived peptides were identified by reverse phase-HPLC-ESI-MS/MS. Caco-2 cell cultures were exposed to the different gliadin peptide digestions (0.25 mg protein/ml), and the mRNA expression of nuclear factor kappa-B (NF-kappaB), tumor necrosis factor alpha (TNF-alpha), and chemokine CXCR3 receptor were analyzed by semi-quantitative reverse transcriptase-polymerase chain reaction (RT-PCR) in stimulated cells. The production of the pro-inflammatory markers NF-kappaB p50, TNF-alpha, and IL-1beta (interleukine 1beta) by Caco-2 cells was also determined by ELISA. The peptides from gliadin digestions inoculated with bifidobacteria did not exhibit the toxic amino acid sequences identified in those noninoculated (alpha/beta-Gld [158-164] and alpha/beta-Gld [122-141]). The RT-PCR analysis evidenced a down-regulation in mRNA expression of pro-inflammatory biomarkers. Consistent with these results the production of NF-kappaB, TNF-alpha, and IL-1beta was reduced (18.2-22.4%, 28.0-64.8%, and abolished, respectively) in cell cultures exposed to gliadin digestions inoculated with bifidobacteria. Therefore, bifidobacteria change the gliadin-derived peptide pattern and, thereby, attenuate their pro-inflammatory effects on Caco-2 cells.


Subject(s)
Bifidobacterium , Gliadin/pharmacology , Inflammation/prevention & control , Intestinal Mucosa/pathology , Biological Therapy , Biomarkers/analysis , Caco-2 Cells , Celiac Disease , Digestion , Epithelial Cells/pathology , Gliadin/metabolism , Humans , Inflammation/etiology , Intestinal Mucosa/microbiology , Peptide Fragments/metabolism , Peptide Fragments/toxicity
12.
Pharmacol Res ; 61(3): 219-25, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19914380

ABSTRACT

The human gut is populated by an array of bacterial species, which develop important metabolic and immune functions, with a marked effect on the nutritional and health status of the host. Dietary component also play beneficial roles beyond basic nutrition, leading to the development of the functional food concept and nutraceuticals. Prebiotics, polyunsaturated fatty acids (PUFAs) and phytochemicals are the most well characterized dietary bioactive compounds. The beneficial effects of prebiotics mainly relay on their influence on the gut microbiota composition and their ability to generate fermentation products (short-chain fatty acids) with diverse biological roles. PUFAs include the omega-3 and omega-6 fatty acids, whose balance may influence diverse aspects of immunity and metabolism. Moreover, interactions between PUFAs and components of the gut microbiota may also influence their biological roles. Phytochemicals are bioactive non-nutrient plant compounds, which have raised interest because of their potential effects as antioxidants, antiestrogenics, anti-inflammatory, immunomodulatory, and anticarcinogenics. However, the bioavailability and effects of polyphenols greatly depend on their transformation by components of the gut microbiota. Phytochemicals and their metabolic products may also inhibit pathogenic bacteria while stimulate the growth of beneficial bacteria, exerting prebiotic-like effects. Therefore, the intestinal microbiota is both a target for nutritional intervention and a factor influencing the biological activity of other food compounds acquired orally. This review focuses on the reciprocal interactions between the gut microbiota and functional food components, and the consequences of these interactions on human health.


Subject(s)
Dietary Supplements , Functional Food , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Metagenome/physiology , Animals , Humans
13.
Lett Appl Microbiol ; 49(6): 695-701, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19843211

ABSTRACT

AIMS: To evaluate the adhesion ability of intestinal bacteria to different in vitro models of intestinal epithelia, and to estimate the suitability of these models and the type of interactions involved. METHODS AND RESULTS: The adhesion of probiotic (Lactobacillus rhamnosus GG and Bifidobacterium animalis subsp. lactis Bb12), commensal (B. animalis IATA-A2 and B. bifidum IATA-ES2) and potentially pathogenic bacteria (E. coli and L. monocytogenes) was determined. The adhesion models used were polycarbonate-well plates, with or without mucin, and different configurations of Caco-2 and/or HT29-MTX cell cultures. All bacteria adhered to wells without mucin (2.6-27.3%), the values being highly variable depending on the bacterial strain. Adhesion percentages of potentially probiotic bacteria to Caco-2 cultures were remarkably lower (P < 0.05) than those to mucin, and more similar to those of pathogenic strains. The lowest adhesion of different bacterial strains was detected on HT29-MTX (0.5-2.3%) cultures and Caco-2/HT29-MTX (0.6-3.2%) cocultures, while these values were increased in Caco-2 cultures plus mucin. CONCLUSIONS: The results suggested that bacterial strains exhibit different capacities to adhere to cellular components and several types of mucin present in different models, showing preferences for intestinal MUC2. SIGNIFICANCE AND IMPACT OF THE STUDY: The use of Caco-2 cells monolayer plus mucin (type II) better approaches the physiological characteristics of in vivo situation, providing a reliable and suitable in vitro model to evaluate bacterial adhesion.


Subject(s)
Bacterial Adhesion , Escherichia coli/physiology , Gram-Positive Asporogenous Rods/physiology , Intestinal Mucosa/microbiology , Caco-2 Cells , Colony Count, Microbial , HT29 Cells , Humans , Models, Biological , Mucins/metabolism , Probiotics
14.
J Food Sci ; 74(2): H40-6, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19323749

ABSTRACT

Inulin, a prebiotic, may enhance intestinal Fe absorption. Our objective was to assess the effects of supplemental inulin and 2 probiotic bacteria (B. infantis and L. acidophillus) on Fe availability to Caco-2 cells from common white and red beans (Phaseolus vulgaris L.). Cooked beans were mixed or not with supplemental inulin (4%, w/w), and then subjected to simulated gastrointestinal digestion (pepsin, pH 2; pancreatin, pH 7.2). Subsequently, the digests were incubated overnight with and without B. infantis or L. acidophilus. Ferritin formation in Caco-2 cells was used to evaluate Fe uptake. Total soluble phenols (Folin-Ciocalteau) and phytate (HPLC-electrochemical detection) were quantified, and the flavonoids profile (HPLC-PDA/UV detection) was monitored in the digests. Supplemental inulin did not affect Fe uptake from white nor red beans. Incubation with B. infantis increased total soluble phenols (TSP) in the digests and decreased Fe uptake. Incubation with L. acidophilus decreased TSP in the digest and increased Fe uptake. Variations in Fe uptake were not associated with soluble phytate concentrations in the digests. The largest change in flavonoids profile were found in the digests incubated with L. acidophilus, which decreased the soluble concentration of astragalin (kaempferol-3-O-glucoside). These results suggest that certain probiotics could increase Fe uptake from common beans.


Subject(s)
Caco-2 Cells/metabolism , Inulin/pharmacology , Iron/metabolism , Phaseolus/metabolism , Probiotics/pharmacology , Animals , Bacillus/metabolism , Bile/physiology , Bile Acids and Salts/pharmacology , Caco-2 Cells/drug effects , Freeze Drying , Humans , Lactobacillus acidophilus/metabolism , Oligosaccharides/pharmacology , Pancreatin/pharmacology , Pepsin A/pharmacology , Phaseolus/drug effects , Swine , Tissue Extracts/pharmacology
15.
J Food Prot ; 71(5): 979-85, 2008 May.
Article in English | MEDLINE | ID: mdl-18522033

ABSTRACT

There are some concerns about the use of Pseudomonas-based products as biocontrol agents because of the hemolytic activity shown by their metabolites. The effects of Pseudomonas lipodepsipeptides (LDPs) on mammals via ingestion and the LDP degradation during the digestion and intestinal permeability have not been evaluated. In this research, the susceptibility of different LDPs to degradation was assayed with enzymatic gastrointestinal digestion, and intestinal permeability to LDPs was investigated in an in vitro system based on an intestinal cell layer system. Results demonstrated that trypsin and chymotrypsin hydrolyze up to 50% of the various LDPs, and that proteolysis was further increased by pronase E treatment. A decrease in LDP hemolytic activity matched LDP degradation during the various steps of the digestion process. Moreover, it was shown that syringomycin E (SRE), the main known LDP, was not able to cross the intestinal cell layer, suggesting that SRE does not reach the bloodstream in vivo. It was concluded that the Pseudomonas-based biocontrol products do not represent a serious risk for consumer health. In fact, LDPs possibly present on biocontrol-treated agricultural commodities would likely be partially digested by gastrointestinal enzymes and would not be absorbed at the intestinal level.


Subject(s)
Digestion , Intestinal Absorption/drug effects , Peptides, Cyclic/metabolism , Pseudomonas syringae/metabolism , Consumer Product Safety , Humans , In Vitro Techniques , Peptides, Cyclic/adverse effects , Pest Control, Biological/methods , Risk Assessment
16.
Toxicol In Vitro ; 22(2): 444-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18079090

ABSTRACT

Foods and drinking water are the main routes for human exposure to inorganic arsenic, the intestinal epithelium being the first barrier against such exogenous toxicants. The present study evaluates the effect of As(III) (0.5-25 microM) upon Caco-2 cells as an intestinal epithelia model. Cell viability, intracellular formation of reactive oxygen species (ROS), mitochondrial membrane potential (Deltapsim) changes, and cell cycle distribution in exposed cultures were evaluated. The intracellular production of ROS was seen to increase in a non-dose dependent manner at all concentrations tested, with impairment of cell mitochondrial enzyme function secondary to a loss of Deltapsim. Concentrations between 0.5 and 5 microM induce cell cycle transition from phase G1 to phase S, with no significant alteration in the proportion of cells in phase G2. These data suggest that As(III) could induce intestinal oxidative stress-cytotoxicity at mitochondrial functional level, and affect cell cycle progression. The data presented in this work may also suggest the impairment of essential survival processes in Caco-2 cells, induced after exposure to As(III) (1-25 microM). Oxidative stress and alteration of mitochondrial functionality could be early indicators of arsenic-induced cytotoxicity, with the resulting abnormal progression of the cell cycle.


Subject(s)
Cell Cycle/drug effects , Oxidative Stress/drug effects , Oxides/toxicity , Arsenic Trioxide , Arsenicals , Caco-2 Cells , Cell Survival/drug effects , G1 Phase/drug effects , G2 Phase/drug effects , Humans , Membrane Potentials/drug effects , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria/metabolism , Mitochondrial Membranes/drug effects , Reactive Oxygen Species/metabolism , Tetrazolium Salts , Thiazoles
17.
J Agric Food Chem ; 55(14): 5892-7, 2007 Jul 11.
Article in English | MEDLINE | ID: mdl-17567035

ABSTRACT

Organoarsenical standards and raw and cooked seafood (DORM-2, sole, and Greenland halibut) were subjected to in vitro gastrointestinal digestion to estimate arsenic bioaccessibility (maximum soluble concentration in gastrointestinal medium). The in vitro digestion did not modify the chemical form of the organoarsenic species standards. In seafood, bioaccessibility was 67.5-100% for arsenobetaine (AB), 30% for dimethylarsinic acid (DMA), 45% for tetramethylarsonium ion (TETRA), and >50% for trimethylarsine oxide (TMAO). Cooking induced no changes in bioaccessible contents. In addition, transport by Caco-2 cells, an intestinal epithelia model, was evaluated from organoarsenical standards and DORM-2. For standards, transport ranged from 1.7% for AB to 15.5% for TETRA. In DORM-2, transport was observed for only AB (12%), with far higher efficiency than in the case of the standard solution, thus illustrating the interest of using whole foods for studying bioavailability.


Subject(s)
Arsenicals/metabolism , Seafood/analysis , Arsenicals/pharmacokinetics , Biological Availability , Biological Transport , Caco-2 Cells , Gastrointestinal Tract/metabolism , Humans
18.
J Agric Food Chem ; 51(20): 6080-5, 2003 Sep 24.
Article in English | MEDLINE | ID: mdl-13129320

ABSTRACT

The aim of this study was to examine the bioaccessibility (maximum soluble concentration in gastrointestinal medium) of total (AsT) and inorganic (AsI) arsenic contents and the effect on them of cooking edible seaweed, a food of great interest because of its high As content. An in vitro gastrointestinal digestion (pepsin, pH 2, and pancreatin-bile extract, pH 7) was applied to obtain the mineral soluble fraction of three seaweeds (Hizikia fusiforme, Porphyra sp., and Enteromorpha sp.). AsT was determined by dry-ashing flow injection hydride generation atomic absorption spectrometry. AsI was determined by acid digestion, solvent extraction, and flow injection hydride generation atomic absorption spectrometry. The bioaccessibility of AsI increased significantly after cooking, attaining 73% in Porphyra sp. and 88% in H. fusiforme. For cooked H. fusiforme, the AsI attained in the bioaccessible fraction was 26 microg g(-1) seaweed, a concentration that is a warning of the toxicological risk of this food.


Subject(s)
Arsenic/pharmacokinetics , Digestion , Seaweed/chemistry , Bile/metabolism , Hot Temperature , Hydrogen-Ion Concentration , In Vitro Techniques , Pancreatin/metabolism , Pepsin A/metabolism , Seaweed/metabolism , Solubility , Spectrophotometry, Atomic
SELECTION OF CITATIONS
SEARCH DETAIL
...