Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nutrients ; 16(8)2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38674882

RESUMO

BACKGROUND: Tropomyosins (TM) from vertebrates are generally non-allergenic, while invertebrate homologs are potent pan-allergens. This study aims to compare the risk of sensitization between chicken TM and shrimp TM through affecting the intestinal epithelial barrier integrity and type 2 mucosal immune activation. METHODS: Epithelial activation and/or barrier effects upon exposure to 2-50 µg/mL chicken TM, shrimp TM or ovalbumin (OVA) as a control allergen, were studied using Caco-2, HT-29MTX, or HT-29 intestinal epithelial cells. Monocyte-derived dendritic cells (moDC), cocultured with HT-29 cells or moDC alone, were exposed to 50 µg/mL chicken TM or shrimp TM. Primed moDC were cocultured with naïve Th cells. Intestinal barrier integrity (TEER), gene expression, cytokine secretion and immune cell phenotypes were determined in these human in vitro models. RESULTS: Shrimp TM, but not chicken TM or OVA exposure, profoundly disrupted intestinal barrier integrity and increased alarmin genes expression in Caco-2 cells. Proinflammatory cytokine secretion in HT-29 cells was only enhanced upon shrimp TM or OVA, but not chicken TM, exposure. Shrimp TM enhanced the maturation of moDC and chemokine secretion in the presence or absence of HT-29 cells, while only in the absence of epithelial cells chicken TM activated moDC. Direct exposure of moDC to shrimp TM increased IL13 and TNFα secretion by Th cells cocultured with these primed moDC, while shrimp TM exposure via HT-29 cells cocultured with moDC sequentially increased IL13 expression and IL4 secretion in Th cells. CONCLUSIONS: Shrimp TM, but not chicken TM, disrupted the epithelial barrier while triggering type 2 mucosal immune activation, both of which are key events in allergic sensitization.


Assuntos
Alérgenos , Galinhas , Técnicas de Cocultura , Células Dendríticas , Mucosa Intestinal , Células Th2 , Tropomiosina , Animais , Humanos , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/efeitos dos fármacos , Células CACO-2 , Tropomiosina/imunologia , Alérgenos/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Células HT29 , Células Th2/imunologia , Citocinas/metabolismo , Penaeidae/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Ovalbumina
2.
Nutrients ; 15(12)2023 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-37375664

RESUMO

Low-grade inflammation and barrier disruption are increasingly acknowledged for their association with non-communicable diseases (NCDs). Short chain fatty acids (SCFAs), especially butyrate, could be a potential treatment because of their combined anti-inflammatory and barrier- protective capacities, but more insight into their mechanism of action is needed. In the present study, non-activated, lipopolysaccharide-activated and αCD3/CD28-activated peripheral blood mononuclear cells (PBMCs) with and without intestinal epithelial cells (IEC) Caco-2 were used to study the effect of butyrate on barrier function, cytokine release and immune cell phenotype. A Caco-2 model was used to compare the capacities of butyrate, propionate and acetate and study their mechanism of action, while investigating the contribution of lipoxygenase (LOX), cyclooxygenase (COX) and histone deacetylase (HDAC) inhibition. Butyrate protected against inflammatory-induced barrier disruption while modulating inflammatory cytokine release by activated PBMCs (interleukin-1 beta↑, tumor necrosis factor alpha↓, interleukin-17a↓, interferon gamma↓, interleukin-10↓) and immune cell phenotype (regulatory T-cells↓, T helper 17 cells↓, T helper 1 cells↓) in the PBMC/Caco-2 co-culture model. Similar suppression of immune activation was shown in absence of IEC. Butyrate, propionate and acetate reduced inflammatory cytokine-induced IEC activation and, in particular, butyrate was capable of fully protecting against cytokine-induced epithelial permeability for a prolonged period. Different HDAC inhibitors could mimic this barrier-protective effect, showing HDAC might be involved in the mechanism of action of butyrate, whereas LOX and COX did not show involvement. These results show the importance of sufficient butyrate levels to maintain intestinal homeostasis.


Assuntos
Butiratos , Citocinas , Humanos , Butiratos/farmacologia , Leucócitos Mononucleares , Técnicas de Cocultura , Histona Desacetilases , Células CACO-2 , Propionatos/farmacologia , Interleucinas , Mucosa Intestinal
3.
Int J Mol Sci ; 23(18)2022 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-36142892

RESUMO

Human milk contains bioactive components that provide protection against viral infections in early life. In particular, intestinal epithelial cells (IEC) have key regulatory roles in the prevention of enteric viral infections. Here we established an in vitro model to study the modulation of host responses against enteric viruses mimicked by poly I:C (pIC). The effects of 2'-fucosyllactose (2'FL), abundantly present in human milk, were studied on IEC and/or innate immune cells, and the subsequent functional response of the adaptive immune cells. IEC were pre-incubated with 2'FL and stimulated with naked or Lyovec™-complexed pIC (LV-pIC). Additionally, monocyte-derived dendritic cells (moDC) alone or in co-culture with IEC were stimulated with LV-pIC. Then, conditioned-moDC were co-cultured with naïve CD4+ T helper (Th)-cells. IEC stimulation with naked or LV-pIC promoted pro-inflammatory IL-8, CCL20, GROα and CXCL10 cytokine secretion. However, only exposure to LV-pIC additionally induced IFNß, IFNλ1 and CCL5 secretion. Pre-incubation with 2'FL further increased pIC induced CCL20 secretion and LV-pIC induced CXCL10 secretion. LV-pIC-exposed IEC/moDC and moDC cultures showed increased secretion of IL-8, GROα, IFNλ1 and CXCL10, and in the presence of 2'FL galectin-4 and -9 were increased. The LV-pIC-exposed moDC showed a more pronounced secretion of CCL20, CXCL10 and CCL5. The moDC from IEC/moDC cultures did not drive T-cell development in moDC/T-cell cultures, while moDC directly exposed to LV-pIC secreted Th1 driving IL-12p70 and promoted IFNγ secretion by Th-cells. Hereby, a novel intestinal model was established to study mucosal host-defense upon a viral trigger. IEC may support intestinal homeostasis, regulating local viral defense which may be modulated by 2'FL. These results provide insights regarding the protective capacity of human milk components in early life.


Assuntos
Interleucina-8 , Leite Humano , Células Dendríticas , Células Epiteliais , Galectina 4 , Humanos , Oligossacarídeos/farmacologia , Poli I , Trissacarídeos
4.
Int J Mol Sci ; 23(7)2022 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-35409339

RESUMO

Non-communicable diseases are increasing and have an underlying low-grade inflammation in common, which may affect gut health. To maintain intestinal homeostasis, unwanted epithelial activation needs to be avoided. This study compared the efficacy of butyrate, propionate and acetate to suppress IFN-γ+/-TNF-α induced intestinal epithelial activation in association with their HDAC inhibitory capacity, while studying the canonical and non-canonical STAT1 pathway. HT-29 were activated with IFN-γ+/-TNF-α and treated with short chain fatty acids (SCFAs) or histone deacetylase (HDAC) inhibitors. CXCL10 release and protein and mRNA expression of proteins involved in the STAT1 pathway were determined. All SCFAs dose-dependently inhibited CXCL10 release of the cells after activation with IFN-γ or IFN-γ+TNF-α. Butyrate was the most effective, completely preventing CXCL10 induction. Butyrate did not affect phosphorylated STAT1, nor phosphorylated NFκB p65, but inhibited IRF9 and phosphorylated JAK2 protein expression in activated cells. Additionally, butyrate inhibited CXCL10, SOCS1, JAK2 and IRF9 mRNA in activated cells. The effect of butyrate was mimicked by class I HDAC inhibitors and a general HDAC inhibitor Trichostatin A. Butyrate is the most potent inhibitor of CXCL10 release compared to other SCFAs and acts via HDAC inhibition. This causes downregulation of CXCL10, JAK2 and IRF9 genes, resulting in a decreased IRF9 protein expression which inhibits the non-canonical pathway and CXCL10 transcription.


Assuntos
Butiratos , Histona Desacetilases , Butiratos/metabolismo , Butiratos/farmacologia , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Células Epiteliais/metabolismo , Ácidos Graxos Voláteis/farmacologia , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Humanos , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/metabolismo , RNA Mensageiro/genética , Fator de Necrose Tumoral alfa/metabolismo
5.
Biomolecules ; 12(3)2022 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-35327576

RESUMO

Prebiotic galacto-oligosaccharides (GOS) were shown to support mucosal immune development by enhancing regulatory-type Th1 immune polarization induced by synthetic CpG oligodeoxynucleotides (TLR9 agonist mimicking a bacterial DNA trigger). Epithelial-derived galectin-9 was associated with these immunomodulatory effects. We aimed to identify the most active fractions within GOS based on the degree of polymerization (DP), and to study the immunomodulatory capacities of DP3-sized ß-3'galactosyllactose (ß-3'GL) using a transwell co-culture model of human intestinal epithelial cells (IEC) and activated peripheral blood mononuclear cells (PBMC). IEC were apically exposed to different DP fractions of GOS or ß-3'GL in the presence of CpG, and basolaterally co-cultured with αCD3/CD28-activated PBMC, washed, and incubated in fresh medium for IEC-derived galectin analysis. Only DP3-5 in the presence of CpG enhanced galectin-9 secretion. DP3-sized ß-3'GL promoted a regulatory-type Th1 response by increasing IFNγ and IL-10 or galectin-9 concentrations as compared to CpG alone. In addition, IEC-derived galectin-3, -4, and -9 secretion was increased by ß-3'GL when combined with CpG. Therefore, the GOS DP3-5 and most effectively DP3-sized ß-3'GL supported the immunomodulatory properties induced by CpG by enhancing epithelial-derived galectin secretion, which, in turn, could support mucosal immunity.


Assuntos
Células Epiteliais , Leucócitos Mononucleares , Galectinas/farmacologia , Células HT29 , Humanos , Oligossacarídeos/farmacologia
6.
Front Immunol ; 13: 1053107, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36703973

RESUMO

Cow's milk allergy is a common food allergy among infants. Improved hygiene conditions and loss of microbial diversity are associated with increased risk of allergy development. The intestinal immune system is essential for oral tolerance induction. In this respect, bacterial CpG DNA is known to drive Th1 and regulatory T-cell (Treg) development via Toll-Like-Receptor 9 (TLR-9) signaling, skewing away from the allergic Th2 phenotype. We aimed to induce allergen specific tolerance via oral delivery of poly (lactic-co-glycolic acid) nanoparticles (NP) co-encapsulated with a selected ß-lactoglobulin derived peptide (BLG-Pep) and TLR-9 ligand CpG oligodeoxynucleotide (CpG). In vivo, 3-4-week-old female C3H/HeOuJ mice housed in individually ventilated cages received 6-consecutive-daily gavages of either PBS, whey, BLG-Pep/NP, CpG/NP, a mixture of BLG-Pep/NP plus CpG/NP or co-encapsulated BLG-Pep+CpG/NP, before 5-weekly oral sensitizations with whey plus cholera toxin (CT) or only CT (sham) and were challenged with whey 5 days after the last sensitization. The co-encapsulated BLG-Pep+CpG/NP pretreatment, but not BLG-Pep/NP, CpG/NP or the mixture of BLG-Pep/NP plus CpG/NP, prevented the whey-induced allergic skin reactivity and prevented rise in serum BLG-specific IgE compared to whey-sensitized mice. Importantly, co-encapsulated BLG-Pep+CpG/NP pretreatment reduced dendritic cell (DC) activation and lowered the frequencies of PD-L1+ DC in the mesenteric lymph nodes compared to whey-sensitized mice. By contrast, co-encapsulated BLG-Pep+CpG/NP pretreatment increased the frequency of splenic PD-L1+ DC compared to the BLG-Pep/NP plus CpG/NP recipients, in association with lower Th2 development and increased Treg/Th2 and Th1/Th2 ratios in the spleen. Oral administration of PLGA NP co-encapsulated with BLG-Pep and CpG prevented rise in serum BLG-specific IgE and symptom development while lowering splenic Th2 cell frequency in these mice which were kept under strict hygienic conditions.


Assuntos
Hipersensibilidade a Leite , Nanopartículas , Bovinos , Camundongos , Feminino , Animais , Hipersensibilidade a Leite/prevenção & controle , Lactoglobulinas , Antígeno B7-H1 , Receptor Toll-Like 9 , Camundongos Endogâmicos C3H , Proteínas do Soro do Leite , Camundongos Endogâmicos , Peptídeos , Imunoglobulina E
7.
Front Pharmacol ; 11: 1244, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32973501

RESUMO

The Mediterranean diet, containing valuable nutrients such as n-3 long chain poly-unsaturated fatty acids (LCPUFAs) and other fat-soluble micronutrients, is known for its health promoting and anti-inflammatory effects. Its valuable elements might help in the battle against the rising prevalence of non-communicable diseases (NCD), including the development of allergic diseases and other (chronic) inflammatory diseases. The fat fraction of the Mediterranean diet contains bioactive fatty acids but can also serve as a matrix to dissolve and increase the uptake of fat-soluble vitamins and phytochemicals, such as luteolin, quercetin, resveratrol and lycopene with known immunomodulatory and anti-inflammatory capacities. Especially n-3 LCPUFAs such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) derived from marine oils can target specific receptors or signaling cascades, act as eicosanoid precursors and/or alter membrane fluidity and lipid raft formation, hereby exhibiting anti-inflammatory properties. Beyond n-3 LCPUFAs, fat-soluble vitamins A, D, E, and K1/2 have the potential to affect pro-inflammatory signaling cascades by interacting with receptors or activating/inhibiting signaling proteins or phosphorylation in immune cells (DCs, T-cells, mast cells) involved in allergic sensitization or the elicitation/effector phase of allergic reactions. Moreover, fat-soluble plant-derived phytochemicals can manipulate signaling cascades, mostly by interacting with other receptors or signaling proteins compared to those modified by fat-soluble vitamins, suggesting potential additive or synergistic actions by applying a combination of these nutrients which are all part of the regular Mediterranean diet. Research concerning the effects of phytochemicals such as polyphenols has been hampered due to their poor bio-availability. However, their solubility and uptake are improved by applying them within the dietary fat matrix. Alternatively, they can be prepared for targeted delivery by means of pharmaceutical approaches such as encapsulation within liposomes or even unique nanoparticles. This review illuminates the molecular mechanisms of action and possible immunomodulatory effects of n-3 LCPUFAs and fat-soluble micronutrients from the Mediterranean diet in allergic disease development and allergic inflammation. This will enable us to further appreciate how to make use of the beneficial effects of n-3 LCPUFAs, fat-soluble vitamins and a selection of phytochemicals as active biological components in allergy prevention and/or symptom reduction.

8.
Front Immunol ; 11: 748, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32431702

RESUMO

Allergy is becoming a rapidly increasing problem worldwide, and in vitro models are frequently used to study the mechanisms behind the different types of allergic response. The dendritic cell (DC)-T-cell model can be used to study sensitization. However, lipopolysaccharide (LPS) is often used to maturate the DCs, but it gives rise to a DC1 phenotype, whereas Th2-driven inflammatory diseases such as allergy are characterized by the involvement of the DC2 phenotype. Our aim was to create a DC2-T-cell human model (human moDC2s) to study in vitro sensitization and validate the model using polyunsaturated fatty acids (PUFAs) that were previously shown to have immunomodulatory properties. We found that the generated DC2s expressed OX40L and drove naive T-cells into IL-13 production of CD4+ effector T-cells. In line with in vivo findings, n-3 long-chain (LC)PUFA docosahexaenoic acid (DHA) effectively decreased the DC2's surface expression of OX40L, as well as the IL-12p40 and IL-23 cytokine production by DC2s and subsequently lowered IL-13 production by DC2-induced effector T-cells. Similar cytokine production effects were found with eicosapentaenoic acid (EPA) and arachidonic acid (AA), whereas linoleic acid (LA) increased OX40L surface expression and subsequent T-cell-derived IL-13/IFNγ ratios, suggesting an increased risk of allergy development. Altogether, these data show that human moDC2s are able to induce Th2-type IL-13 secretion by T-cell differentiated in the presence of these DC2s and that this model can be differentially modulated by PUFAs. These results are in line with previous in vivo studies using PUFAs, indicating that this model may be of use to predict in vivo outcomes.


Assuntos
Citocinas/metabolismo , Células Dendríticas/efeitos dos fármacos , Ácidos Graxos Insaturados/farmacologia , Diferenciação Celular , Técnicas de Cocultura , Voluntários Saudáveis , Humanos , Hipersensibilidade/prevenção & controle , Imunomodulação/efeitos dos fármacos , Interferon gama/metabolismo , Interleucina-10/metabolismo , Interleucina-13/metabolismo , Ligante OX40 , Linfócitos T/metabolismo , Células Th2 , Vitaminas/farmacologia
9.
Front Immunol ; 9: 923, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29867934

RESUMO

Background: Non-digestible oligosaccharides promote colonization of beneficial gut bacteria and have direct immunomodulatory effects. Apical exposure of intestinal epithelial cells (IECs) to short-chain galacto-oligosaccharides and long-chain fructo-oligosaccharides (scGOS/lcFOS) in a transwell co-culture model enhanced the CpG-induced (TLR-9 ligand) T helper 1 (Th1) phenotype and regulatory IL-10 response of underlying peripheral mononuclear cells (PBMCs) of healthy donors. scGOS is derived from lactose and may pose risks in severe cow's milk allergic patients, and scFOS/lcFOS may be an alternative. The goal of this study was to determine the immunomodulatory effects of scGOS/lcFOS and scFOS/lcFOS in an allergen-specific transwell co-culture model using PBMCs from peanut-allergic patients. Methods: IECs cultured on transwell filters were apically exposed to CpG, either or not in combination with oligosaccharides. These IECs were co-cultured with basolateral PBMCs of peanut-allergic patients that were either activated with aCD3/28 or peanut extract. Basolateral cytokine production and T-cell polarization were measured and the contribution of galectin-9 and the dectin-1 receptor in immune modulation were assessed. Results: IECs exposed to CpG increased IFN-γ, IL-10, and galectin-9 production by aCD3/28-stimulated PBMCs, whereas IL-13 decreased. Both scGOS/lcFOS and scFOS/lcFOS further enhanced IFN-γ and IL-10, while suppressing IL-13 and TNF-α. In the peanut-specific model, only scFOS/lcFOS further increased IFN-γ and IL-10 production, coinciding with enhanced Th1-frequency. Expression of CRTH2 reduced after CpG exposure, and was further reduced by scFOS/lcFOS. Galectin-9 inhibitor TIM-3-Fc abrogated the additional effect of scFOS/lcFOS on peanut-specific IFN-γ production, while neutralization of the dectin-1 receptor was not effective. Conclusion: Epithelial exposure to scFOS/lcFOS enhanced the CpG-induced Th1 and regulatory IL-10 response in a peanut-specific co-culture model. These effects suggest scFOS/lcFOS as candidate for dietary adjunct in allergen-specific immunotherapy.


Assuntos
Arachis/imunologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Intestinos/imunologia , Oligossacarídeos/farmacologia , Células Th1/imunologia , Adolescente , Adulto , Alérgenos/imunologia , Técnicas de Cocultura , Feminino , Células HT29 , Humanos , Imunomodulação , Interleucina-10/imunologia , Intestinos/citologia , Ativação Linfocitária/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Oligodesoxirribonucleotídeos/farmacologia , Oligossacarídeos/química , Adulto Jovem
10.
Br J Nutr ; 114(4): 577-85, 2015 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-26179875

RESUMO

Increased intake of vegetable oils rich in n-6 PUFA, including soyabean oil, has been associated with an increase in allergic disease. The present study aimed to determine the effect of an increasing dose of dietary vegetable oil on allergic outcomes in mice. To study this, mice received a 7 v. 10 % soyabean oil diet before and during oral sensitisation with whey or whey hyperimmune serum transfer. Another group of mice received partial whey hydrolysate (pWH) while being fed the diets before oral sensitisation. The acute allergic skin response, serum Ig level, mouse mast cell protease-1 (mMCP-1) concentration and/or splenic T-cell percentages were determined upon whey challenge. When the diets were provided before and during oral sensitisation, the acute allergic skin response was increased in mice fed the 10 % soyabean oil diet compared with the 7 % soyabean oil diet. Whey IgE and IgG1 levels remained unaltered, whereas mMCP-1 levels increased in mice fed the 10 % soyabean oil diet. Furthermore, allergic symptoms were increased in naive mice fed the 10 % soyabean oil diet and sensitised with whey hyperimmune serum. In addition to enhancing the mast cell response, the 10 % soyabean oil diet increased the percentage of activated Th1 and Th2 cells as well as increased the ratios of Th2:regulatory T cells and Th2:Th1 when compared with the 7 % soyabean oil diet. Oral tolerance induction by pWH was abrogated in mice fed the 10 % soyabean oil diet compared with those fed the 7 % soyabean oil diet during pretreatment with pWH. In conclusion, increased intake of soyabean oil rich in n-6 PUFA suppresses tolerance induction by pWH and enhances the severity of the allergic effector response in whey-allergic mice. Dietary vegetable oils rich in n-6 PUFA may enhance the susceptibility to develop or sustain food allergy.


Assuntos
Dieta/efeitos adversos , Ácidos Graxos Ômega-6/imunologia , Imunidade/efeitos dos fármacos , Hipersensibilidade a Leite , Proteínas do Leite/imunologia , Óleo de Soja/imunologia , Subpopulações de Linfócitos T/metabolismo , Alérgenos , Animais , Quimases/sangue , Gorduras na Dieta/administração & dosagem , Gorduras na Dieta/efeitos adversos , Gorduras na Dieta/imunologia , Modelos Animais de Doenças , Ácidos Graxos Ômega-6/administração & dosagem , Ácidos Graxos Ômega-6/efeitos adversos , Comportamento Alimentar , Feminino , Imunoglobulinas/sangue , Mastócitos/metabolismo , Camundongos , Hipersensibilidade a Leite/etiologia , Óleo de Soja/administração & dosagem , Óleo de Soja/efeitos adversos , Baço/metabolismo , Linfócitos T Reguladores/metabolismo , Células Th1/metabolismo , Equilíbrio Th1-Th2/efeitos dos fármacos , Células Th2/metabolismo , Proteínas do Soro do Leite
11.
J Nutr ; 145(5): 915-22, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25716554

RESUMO

BACKGROUND: Although never evaluated for efficacy, n-3 (ω-3) long-chain polyunsaturated fatty acids (LCPUFAs) are commercially offered as treatment for irritable bowel syndrome (IBS). OBJECTIVE: This study was designed to investigate, in a mast cell-dependent model for visceral hypersensitivity, whether this pathophysiologic mechanism can be reversed by dietary LCPUFA treatment via peroxisome proliferator-activated receptor γ (PPARG) activation. METHODS: Maternally separated rats were subjected to hypersensitivity-inducing acute stress at adult age. Reversal was attempted by protocols with tuna oil-supplemented diets [4% soy oil (SO) and 3% tuna oil (SO-T3) or 3% SO and 7% tuna oil (SO-T7)] and compared with control SO diets (7% or 10% SO) 4 wk after stress. The PPARG agonist rosiglitazone was evaluated in a 1 wk preventive protocol (30 mg · kg⁻¹ · d⁻¹). Erythrocytes were assessed to confirm LCPUFA uptake and tissue expression of lipoprotein lipase and glycerol kinase as indicators of PPARG activation. Colonic mast cell degranulation was evaluated by toluidine blue staining. In vitro, human mast cell line 1 (HMC-1) cells were pretreated with rosiglitazone, eicosapentaenoic acid, or docosahexaenoic acid, stimulated with phorbol 12-myristate 13-acetate (PMA) and calcium ionophore or compound 48/80 and evaluated for tumor necrosis factor α (TNF-α) and ß-hexosaminidase release. RESULTS: Stress led to visceral hypersensitivity in all groups. Hypersensitivity was not reversed by SO-T3 or control treatment [prestress vs. 24 h poststress vs. posttreatment area under the curve; 76 ± 4 vs. 128 ± 12 (P < 0.05) vs. 115 ± 14 and 82 ± 5 vs. 127 ± 16 (P < 0.01) vs. 113 ± 19, respectively]. Comparison of SO-T7 with its control showed similar results [74 ± 6 vs. 103 ± 13 (P < 0.05) vs. 115 ± 17 and 66 ± 3 vs. 103 ± 10 (P < 0.05) vs. 117 ± 11, respectively]. Erythrocytes showed significant LCPUFA uptake in the absence of colonic PPARG activation. Rosiglitazone induced increased PPARG target gene expression, but did not prevent hypersensitivity. Mast cell degranulation never differed between groups. Rosiglitazone and LCPUFAs significantly reduced PMA/calcium ionophore-induced TNF-α release but not degranulation of HMC-1 cells. CONCLUSION: Dietary LCPUFAs did not reverse stress-induced visceral hypersensitivity in maternally separated rats. Although further research is needed, claims concerning LCPUFAs as a treatment option in IBS cannot be confirmed at this point and should be regarded with caution.


Assuntos
Sistema Nervoso Autônomo/fisiopatologia , Colo/inervação , Suplementos Nutricionais , Modelos Animais de Doenças , Ácidos Graxos Ômega-3/uso terapêutico , Óleos de Peixe/uso terapêutico , Síndrome do Intestino Irritável/dietoterapia , Animais , Animais Recém-Nascidos , Sistema Nervoso Autônomo/efeitos dos fármacos , Sistema Nervoso Autônomo/imunologia , Biomarcadores/sangue , Biomarcadores/metabolismo , Degranulação Celular/efeitos dos fármacos , Linhagem Celular , Colo/efeitos dos fármacos , Colo/imunologia , Colo/metabolismo , Eritrócitos/efeitos dos fármacos , Eritrócitos/enzimologia , Eritrócitos/metabolismo , Ácidos Graxos Ômega-3/administração & dosagem , Ácidos Graxos Ômega-3/metabolismo , Feminino , Óleos de Peixe/administração & dosagem , Óleos de Peixe/metabolismo , Hipoglicemiantes/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Mucosa Intestinal/inervação , Mucosa Intestinal/metabolismo , Síndrome do Intestino Irritável/imunologia , Síndrome do Intestino Irritável/metabolismo , Síndrome do Intestino Irritável/fisiopatologia , Masculino , Mastócitos/efeitos dos fármacos , Mastócitos/imunologia , Mastócitos/fisiologia , Privação Materna , PPAR gama/antagonistas & inibidores , PPAR gama/metabolismo , Ratos Long-Evans , Atum
12.
J Nutr ; 144(12): 1970-6, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25342698

RESUMO

BACKGROUND: Supplementation with long-chain n-3 polyunsaturated fatty acids (LCPUFAs) has been found to reduce the development of allergic disease. OBJECTIVE: The aim of this study was to compare the effectiveness of fish oil diets rich in eicosapentaenoic acid (20:5n-3; EPA) or docosahexaenoic acid (22:6n-3; DHA) in suppressing food allergic symptoms. METHODS: Mice were fed a control diet (10% soybean oil) or fish oil diet rich in EPA (4% soybean oil + 6% EPA oil containing 28.8% EPA and 13.7% DHA) or DHA (4% soybean oil + 6% DHA oil containing 7% EPA and 27.8% DHA), starting 14 d before and for 5 wk during oral sensitization with peanut extract (PE) or whey. Acute allergic skin responses, serum immunoglobulins (Igs), and mucosal mast cell protease-1 (mmcp-1) were assessed. Hyperimmune serum was transferred to naive recipient mice fed the different diets. RESULTS: The DHA diet effectively reduced the acute allergic skin response compared with the control or EPA diet in PE-allergic mice (control, 159 ± 15, or EPA, 129 ± 8, vs. DHA, 78 ± 7 µm; P < 0.0001 or P < 0.05, respectively). In contrast, both the DHA and EPA diets reduced the allergic skin response in whey allergic mice (control, 169 ± 9, vs. DHA, 91 ± 13, or EPA, 106 ± 14 µm; P < 0.001 or P < 0.01, respectively); however, only the DHA diet reduced mmcp-1 and whey-specific IgE and IgG1. The DHA and EPA diets also reduced the acute skin response in passively immunized mice. CONCLUSIONS: The DHA-rich fish oil diet reduced allergic sensitization to whey and allergic symptoms in both PE- and whey-allergic mice. These data suggest that DHA-rich fish oil is useful as an intervention to prevent or treat food allergy symptoms.


Assuntos
Ácidos Docosa-Hexaenoicos/farmacologia , Óleos de Peixe/farmacologia , Hipersensibilidade a Leite/prevenção & controle , Hipersensibilidade a Amendoim/prevenção & controle , Animais , Dieta , Suplementos Nutricionais , Ácido Eicosapentaenoico/farmacologia , Feminino , Imunoglobulinas/sangue , Camundongos , Camundongos Endogâmicos C3H , Alimentos Marinhos , Pele/metabolismo , Pele/fisiopatologia , Atum
13.
J Hypertens ; 32(5): 1050-8; discussion 1058, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24569415

RESUMO

OBJECTIVE: Long-chain n-3 polyunsaturated fatty acids from oily fish reduce blood pressure (BP) in hypertension. Previously, we demonstrated that hypertension is associated with marked alterations in sphingolipid biology and elevated ceramide-induced vasoconstriction. Here we investigated in spontaneously hypertensive rats (SHRs) whether fish oil improves endothelial function including reduced vascular contraction induced via the sphingolipid cascade, resulting in reduced BP. METHODS: Twelve-week-old SHRs were fed a control or fish oil-enriched diet during 12 weeks, and BP was recorded. Plasma sphingolipid levels were quantified by mass spectrometry and the response of isolated carotid arteries towards different stimuli was measured. Furthermore, erythrocyte membrane fatty acid composition, thromboxane A2 formation and cytokine secretion in ex-vivo lipopolysaccharide-stimulated thoracic aorta segments were determined. RESULTS: The fish oil diet reduced the mean arterial BP (P < 0.001) and improved endothelial function, as indicated by a substantially increased relaxation potential towards ex-vivo methacholine exposure of the carotid arteries (P < 0.001). The long-chain n-3 polyunsaturated fatty acid diet resulted in altered levels of specific (glucosyl)ceramide subspecies (P < 0.05), reduced membrane arachidonic acid content (P < 0.001) and decreased thromboxane concentrations in plasma (P < 0.01). Concomitantly, the fish oil diet largely reduced ceramide-induced contractions (P < 0.01), which are predominantly mediated by thromboxane. Furthermore, thromboxane A2 and interleukin-10 were reduced in supernatants of lipopolysaccharide-stimulated thoracic aorta of SHRs fed the fish oil diet while RANTES (regulated on activation, normal T-cell expressed and secreted) was enhanced. This may contribute to reduced vasoconstriction in vivo. CONCLUSIONS: Dietary fish oil lowers BP in SHRs and improves endothelial function in association with suppression of sphingolipid-dependent vascular contraction.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Gorduras Insaturadas na Dieta/farmacologia , Endotélio Vascular/efeitos dos fármacos , Óleos de Peixe/farmacologia , Esfingolipídeos/fisiologia , Animais , Cromatografia Líquida , Espectrometria de Massas , Contração Muscular/efeitos dos fármacos , Ratos , Ratos Endogâmicos SHR , Esfingomielina Fosfodiesterase/metabolismo , Tromboxano B2/sangue
14.
J Innate Immun ; 5(6): 625-38, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23735749

RESUMO

Dietary intervention using nondigestible oligosaccharides, short-chain galacto-oligosaccharides (scGOS)/long-chain fructo-oligosaccharides (lcFOS), in combination with Bifidobacterium breve M-16V prevents allergic disease involving galectin-9. In addition, apical TLR9 signaling contributes to intestinal homeostasis. We studied the contribution of galectin-9 secreted by intestinal epithelial cells (IEC; HT-29 and T84) in Th1 and regulatory T-cell (Treg) polarization in vitro. IEC were grown in transwell filters, cocultured with CD3/CD28-activated human peripheral blood mononuclear cells (PBMC) and apically exposed to genomic DNA derived from B. breve M-16V or synthetic TLR9 ligand in the absence or presence of scGOS/lcFOS. Cytokine production and T-cell phenotype were determined and galectin expression by IEC was assessed. Galectin-9 was neutralized using lactose or a TIM-3-Fc fusion protein. IEC exposed to DNA from B. breve M-16V or TLR9 ligand in the presence of scGOS/lcFOS enhanced IFN-γ secretion by PBMC and increased the percentage of Th1 and Treg cells. Expression and secretion of galectin-9 by IEC was increased and neutralization of galectin-9 prevented the induction of IFN-γ secretion and also suppressed the production of IL-10 by PBMC. Furthermore, we show that galectin-9 induces Treg and Th1 polarization through interaction with antigen-presenting cells. Our findings show that galectin-9 secreted by IEC apically exposed to TLR9 ligand in the presence of scGOS/lcFOS is involved in Th1 and Treg polarization and may be a promising target to prevent or treat allergic disease.


Assuntos
Células Epiteliais/imunologia , Galectinas/imunologia , Imunomodulação/imunologia , Mucosa Intestinal/imunologia , Oligossacarídeos/imunologia , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Bifidobacterium/genética , Bifidobacterium/fisiologia , Linhagem Celular , Células Cultivadas , Técnicas de Cocultura , DNA Bacteriano/imunologia , DNA Bacteriano/farmacologia , Células Epiteliais/metabolismo , Citometria de Fluxo , Galectinas/metabolismo , Células HT29 , Humanos , Imunomodulação/efeitos dos fármacos , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-10/imunologia , Interleucina-10/metabolismo , Mucosa Intestinal/metabolismo , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Oligossacarídeos/farmacologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Células Th1/imunologia , Células Th1/metabolismo , Receptor Toll-Like 9/imunologia , Receptor Toll-Like 9/metabolismo
15.
Toxicol Lett ; 201(3): 264-9, 2011 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-21251954

RESUMO

Hypoallergenic infant formulas are commonly used for genetically predisposed children and infants diagnosed with cow's milk allergy. This study describes both in vitro and in vivo approaches to assess residual allergenicity of partially hydrolysed infant formulas. Electrophoretic patterns indicated that ß-lactoglobulin and other whey proteins were largely degraded. For safety reasons, according to the European commission-guidelines, it is required that the sensitizing capacity of hypoallergenic formulas is tested in an animal model. In contrast to whey sensitization, no elevated levels of whey-specific IgE, anaphylactic reactions or drop in body temperature were observed in sensitized mice exposed to whey hydrolysates. This indicates that the whey hydrolysates lost their putative sensitizing capacity in a mouse model using oral sensitization, which is highly relevant in relation to the human situation. In combination with the lost capacity of hydrolysed infant formulas to cross-link human IgE antibodies on RBL-huFcɛRI in vitro, both the sensitization and the challenge phase of the allergic response were studied. This combination of assays is proposed as a strategy for the screening of new hypoallergenic formulas aimed at preventing sensitization in atopic children and avoiding clinical symptoms in infants suffering from cow's milk allergy.


Assuntos
Alérgenos/toxicidade , Hipersensibilidade Alimentar , Alimentos Infantis/toxicidade , Anafilaxia/patologia , Animais , Basófilos/efeitos dos fármacos , Western Blotting , Temperatura Corporal/efeitos dos fármacos , Degranulação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Eletroforese em Gel de Poliacrilamida , Humanos , Hidrólise , Imunoglobulina E/imunologia , Imunoglobulina G/biossíntese , Lactente , Recém-Nascido , Camundongos , Camundongos Endogâmicos C3H , Proteínas do Leite/química , Proteínas do Leite/imunologia , Hidrolisados de Proteína/toxicidade , Ratos , Testes Cutâneos , Proteínas do Soro do Leite
16.
Immunobiology ; 216(4): 518-27, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20850889

RESUMO

Intestinal epithelial cells (IEC) separate the mucosal immune system from the external milieu. Under inflammatory conditions, Toll-like receptor (TLR) expression by IEC is increased. In a transwell co-culture model immune modulation by IEC upon TLR ligation was studied. Human IEC (HT-29 and T84) grown on filters were apically or basolaterally exposed to TLR4 or TLR9 ligands and co-cultured with CD3/CD28-activated healthy donor PBMC in the basolateral compartment. TLR4 ligation of IEC (HT-29) enhanced the production of TNF-α and IEC-derived MDC and decreased numbers of Foxp3(+) regulatory T cells. Neutralization of TSLP abrogated TLR4-induced TNF-α secretion. In contrast, apical TLR9 ligation of IEC (HT-29 and T84) enhanced IFN-γ and IL-10 secretion and increased the number of activated T(h)1 cells. The increase in IFN-γ secretion depended on the presence of IEC. Furthermore, CD14 expression on monocytes was reduced coinciding with enhanced intracellular IL-10 and decreased TNF-α production. However, basolateral TLR9 ligand exposure of HT-29 cells resulted in enhanced IFN-γ, IL-6 and TNF-α, while IL-10 secretion remained unaltered. TLR4 and TLR9 ligands reduced IL-13 secretion in presence and absence of apically exposed IEC and enhanced IL-12 secretion in presence of IEC. These data suggest that TLR4 ligation of IEC drives an inflammatory, while apical TLR9 ligation drives a regulatory T(h)1 effector immune response in vitro in a polarized manner. IEC may be important modulators of the mucosal effector immune response.


Assuntos
Polaridade Celular/imunologia , Células Epiteliais/imunologia , Intestinos/imunologia , Células Th1/imunologia , Receptores Toll-Like/imunologia , Linhagem Celular Tumoral , Técnicas de Cocultura , Citocinas/metabolismo , Células HT29 , Humanos , Imunofenotipagem , Inflamação/imunologia , Mucosa Intestinal/citologia , Mucosa Intestinal/imunologia , Intestinos/citologia , Leucócitos Mononucleares/imunologia , Receptores de Superfície Celular/imunologia
17.
Int Arch Allergy Immunol ; 152(2): 159-68, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20016198

RESUMO

BACKGROUND: Intestinal bacteria and intestinal epithelial cells (IEC) may modulate the mucosal immune response. In this study, immune modulation by Lactobacillus GG (LGG) and Bifidobacterium breve (Bb1, Bb2) in the presence or absence of IEC was addressed in an in vitro transwell co-culture model. METHODS: UV-killed LGG,Bb1, Bb2 or Toll-like receptor (TLR) 2 or nucleotide oligomerization domain (NOD) 2 ligands were added directly to unstimulated or anti-CD3/CD28-stimulated PBMC, or applied apically to human IEC (HT-29) co-cultured with PBMC. A mixture of live bacteria was used as reference. The effect on T helper 1 (IFN-gamma, IL-12), T helper 2 (IL-13), inflammatory (TNF-alpha) and regulatory (IL-10) cytokine secretion was determined. RESULTS: Both UV-killed LGG and Bb enhanced IL-12, IFN-gamma, TNF-alpha and IL-10, and reduced IL-13 secretion when added directly to stimulated PBMC, similar to live bacteria. IEC reduced IL-13, IFN-gamma and IL-10 secretion by stimulated PBMC. Apically added LGG, TLR2 and NOD2 ligands,but not Bb, enhanced IFN-gamma, IL-12 and/or TNF-alpha secretion. Bacteria did not induce cytokine secretion when added to HT-29/unstimulated PBMC co-cultures, whereas direct incubations with PBMC did. CONCLUSION: UV-killed LGG as well as Bb supported a T helper 1 and/or regulatory phenotype when added directly to activated PBMC, similar to live bacteria. In contrast, LGG, TLR2 or NOD2 ligands - but not Bb - enhanced T helper 1 type cytokine secretion when added to IEC, while IL-10 secretion remained suppressed. Co-cultures combining IEC and PBMC may reveal differences between bacterial strains relevant for the in vivo situation.


Assuntos
Bifidobacterium/imunologia , Células Epiteliais/imunologia , Mucosa Intestinal/citologia , Lactobacillus/imunologia , Subpopulações de Linfócitos T/imunologia , Acetilmuramil-Alanil-Isoglutamina/farmacologia , Adjuvantes Imunológicos/farmacologia , Anticorpos/imunologia , Anticorpos/farmacologia , Antígenos de Diferenciação de Linfócitos T/imunologia , Bifidobacterium/efeitos da radiação , Proliferação de Células/efeitos dos fármacos , Técnicas de Cocultura , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células HT29 , Humanos , Interferon gama/metabolismo , Interleucinas/metabolismo , Mucosa Intestinal/imunologia , Lactobacillus/efeitos da radiação , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Lipopeptídeos/farmacologia , Proteína Adaptadora de Sinalização NOD2/agonistas , Probióticos/farmacologia , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/metabolismo , Receptor 2 Toll-Like/agonistas , Fator de Crescimento Transformador beta1/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Raios Ultravioleta
18.
Eur J Nutr ; 47(4): 183-91, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18497998

RESUMO

BACKGROUND: The intestinal mucosa functions as a barrier against harmful dietary and microbial antigens. An intact gut barrier forms a prerequisite for protection against infection and allergy. Both allergic and inflammatory mediators (e.g. IL-4, IFN-gamma) are known to compromise the epithelial barrier integrity by enhancing permeability. Breast milk provides protection against infection and allergy and contains polyunsaturated fatty acids (PUFA). AIM OF THE STUDY: Although PUFA are commonly used in infant formulas their effect on intestinal barrier is still poorly understood. Therefore the effects of distinct PUFA (n-6: LA, GLA, DGLA, AA; n-3: ALA, EPA, DHA) and a fat blend with PUFA composition similar to that of the human breast milk fat fraction, on barrier integrity were investigated. METHODS: Human intestinal epithelial cells (T84) were pre-incubated with individual PUFA or a lipase treated fat blend, with or without subsequent IL-4 exposure. Barrier integrity was evaluated by measuring transepithelial resistance and permeability. Membrane phospholipid composition was determined by capillary gas chromatography. RESULTS: DGLA, AA, EPA, DHA and to a lesser extend GLA enhanced basal TER and strongly reduced IL-4 mediated permeability, while LA and ALA were ineffective. Furthermore, the lipase treated fat blend effectively supported barrier function. PUFA were incorporated in the membrane phospholipid fraction of T84 cells. CONCLUSIONS: Long chain PUFA DGLA, AA, EPA and DHA were particularly effective in supporting barrier integrity by improving resistance and reducing IL-4 mediated permeability. Fat blends that release specific PUFA upon digestion in the gastrointestinal tract may support natural resistance.


Assuntos
Gorduras Insaturadas na Dieta/farmacologia , Ácidos Graxos Insaturados/farmacologia , Imunidade nas Mucosas/efeitos dos fármacos , Interleucina-4/metabolismo , Mucosa Intestinal/fisiologia , Permeabilidade/efeitos dos fármacos , Células Cultivadas , Cromatografia Gasosa , Gorduras Insaturadas na Dieta/metabolismo , Sinergismo Farmacológico , Células Epiteliais , Ácidos Graxos Ômega-3/farmacologia , Ácidos Graxos Ômega-6/farmacologia , Ácidos Graxos Insaturados/metabolismo , Hipersensibilidade Alimentar/imunologia , Hipersensibilidade Alimentar/prevenção & controle , Humanos , Imunidade nas Mucosas/fisiologia , Interleucina-4/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Leite Humano/química , Leite Humano/imunologia
19.
Br J Nutr ; 95(6): 1143-9, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16768837

RESUMO

Fructo-oligosaccharides (FOS) are widely used in commercial food products. Most studies on FOS concern the health benefits, but some negative effects were recently reported concerning the faecal cytotoxicity and excretion of mucin-type oligosaccharides in combination with a Ca-restricted diet. The present study was performed to investigate whether these effects of FOS are observed in adults consuming a regular diet unrestricted in Ca. The study was a randomised, double-blind, placebo-controlled crossover trial, involving eleven healthy adults, who consumed 25-30 g FOS or maltodextrin (control) in a random order for 2 weeks in addition to their regular diet. Stools were collected for analysis of pH and SCFA (as markers of fermentation), for the assessment of faecal water cytotoxicity, and for the analysis of alkaline phosphatase activity (as a marker of epithelial cell turnover) and O-linked oligosaccharides (to estimate the excretion of mucin-type oligosaccharides). FOS consumption significantly altered bacterial fermentation (increased percentage of acetate, decreased percentage of butyrate) and tended to decrease stool pH. Furthermore, FOS consumption resulted in a significantly higher stool frequency and in significantly more complaints of flatulence. No significant differences between the control and FOS period were observed in the mean cytotoxicity of faecal water (37.5 (SEM 6.9)% v. 18.5 (SEM 6.9)%; P=0.084), in mean alkaline phosphatase activity (27.7 (SEM 2.9) v. 24.6 (SEM 3.2) U/g dry faeces; P=0.496) or in the mean excretion of mucin-type oligosaccharides (49.9 (sem 4.0) v. 53.5 (SEM 4.3) mg/g dry faeces; P=0.553). We conclude that dietary FOS in a dose up to 25-30 g/d altered the bacterial fermentation pattern but did not affect faecal cytotoxicity or the faecal concentration of mucin-type oligosaccharides in human adults consuming a regular diet.


Assuntos
Água Corporal/fisiologia , Dieta , Fezes , Oligossacarídeos/farmacologia , Acetatos/análise , Adolescente , Adulto , Fosfatase Alcalina/análise , Análise de Variância , Bactérias/metabolismo , Biomarcadores/análise , Butiratos/análise , Morte Celular , Estudos Cross-Over , Diarreia/etiologia , Método Duplo-Cego , Células Epiteliais/citologia , Eritrócitos/citologia , Fezes/química , Feminino , Fermentação , Flatulência/etiologia , Humanos , Concentração de Íons de Hidrogênio , Masculino , Mucinas/análise , Polissacarídeos/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA