Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Clin Exp Allergy ; 52(1): 137-148, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34145667

RESUMO

BACKGROUND: Two to four percentage of infants are affected by cow's milk allergy (CMA), which persists in 20% of cases. Intervention approaches using early oral exposure to cow's milk protein or hydrolysed cow's milk formula are being studied for CMA prevention. Yet, concerns regarding safety and/or efficacy remain to be tackled in particular for high-risk non-exclusively breastfed infants. Therefore, safe and effective strategies to improve early life oral tolerance induction may be considered. OBJECTIVE: We aim to investigate the efficacy of CMA prevention using oral pre-exposure of two selected 18-AA ß-lactoglobulin-derived peptides loaded poly (lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) in a whey-protein induced CMA murine model. METHODS: The peptides were loaded in PLGA NPs via a double emulsion solvent evaporation technique. In vivo, 3-week-old female C3H/HeOuJ mice received 6 daily gavages with PBS, whey, Peptide-mix, a high- or low-dose Peptide-NPs or empty-NP plus Peptide-mix, prior to 5 weekly oral sensitizations with cholera toxin plus whey or PBS (sham). One week after the last sensitization, the challenge induced acute allergic skin response, anaphylactic shock score, allergen-specific serum immunoglobulins and ex vivo whey-stimulated cytokine release by splenocytes was measured. RESULTS: Mice pre-treated with high-dose Peptide-NPs but not low-dose or empty-NP plus Peptide-mix, were protected from anaphylaxis and showed a significantly lower acute allergic skin response upon intradermal whey challenge compared to whey-sensitized mice. Compared with the Peptide-mix or empty-NP plus Peptide-mix pre-treatment, the high-dose Peptide-NPs-pre-treatment inhibited ex vivo whey-stimulated pro-inflammatory cytokine TNF-α release by splenocytes. CONCLUSION & CLINICAL RELEVANCE: Oral pre-exposure of mice to two ß-lactoglobulin-derived peptides loaded PLGA NPs induced a dose-related partial prevention of CMA symptoms upon challenge to whole whey protein and silenced whey-specific systemic immune response. These findings encourage further development of the concept of peptide-loaded PLGA NPs for CMA prevention towards clinical application.


Assuntos
Hipersensibilidade a Leite , Nanopartículas , Animais , Bovinos , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Lactoglobulinas , Camundongos , Camundongos Endogâmicos C3H , Proteínas do Leite , Soro do Leite/metabolismo , Proteínas do Soro do Leite
2.
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
3.
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
4.
Int Arch Allergy Immunol ; 181(12): 908-918, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32814335

RESUMO

INTRODUCTION: Cow's milk allergy (CMA) is one of the most common food allergies especially early in life. A mixture of nondigestible short-chain galacto-oligosaccharides, long-chain fructo-oligosaccharides, and pectin-derived acidic-oligosaccharides (GFA) may reduce allergy development and allergic symptoms in murine CMA. Recently, vitamin D (VitD) has been suggested to have beneficial effects in reducing allergy as well. OBJECTIVE: In this study, the immune modulatory effect on allergy prevention using the combination of GFA and VitD was investigated. METHODS: Female C3H/HeOuJ mice were fed a control or GFA-containing diet with depleted, standard (1,000 IU/kg), or supplemented (5,000 IU/kg) VitD content for 2 weeks before and during whey sensitization (n = 10-15). Mice were sensitized 5 times intragastrically with PBS as a control, whey as cow's milk allergen, and/or cholera toxin as adjuvant on a weekly interval. One week after the last sensitization, mice were intradermally challenged in both ear pinnae and orally with whey, subsequently the acute allergic skin response and shock symptoms were measured. After 18 h, terminal blood samples, mesenteric lymph nodes, and spleens were collected. Whey-specific immunoglobulin (Ig) E and IgG1 levels were measured by means of ELISA. T cell subsets and dendritic cells (DCs) were studied using flow cytometry. RESULTS: Additional VitD supplementation did not lower the allergic symptoms compared to the standard VitD diet. CMA mice fed the GFA diet supplemented with VitD (GFA VitD+) significantly decreased the acute allergic skin response of whey sensitized mice when compared to the CMA mice fed VitD (VitD+) group (p < 0.05). The effect of GFA was not improved by extra VitD supplementation even though the CMA mice fed the GFA VitD+ diet had a significantly increased percentage of CD103+ DCs compared to the VitD+ group (p < 0.05). The VitD-deprived mice showed a high percentage of severe shock and many reached the humane endpoint; therefore, these groups were not further analyzed. CONCLUSIONS: High-dose VitD supplementation in mice does not protect against CMA development in the presence or absence of GFA.


Assuntos
Células Dendríticas/imunologia , Hipersensibilidade a Leite/dietoterapia , Pele/patologia , Linfócitos T Reguladores/imunologia , Vitamina D/uso terapêutico , Alérgenos/imunologia , Animais , Bovinos , Dieta , Suplementos Nutricionais , Modelos Animais de Doenças , Feminino , Humanos , Imunoglobulina E/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Leite/imunologia , Oligossacarídeos/uso terapêutico
5.
Mediators Inflamm ; 2019: 8456829, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31427886

RESUMO

BACKGROUND: Short-chain galacto- and long-chain fructo-oligosaccharides (scGOS/lcFOS) and CpG-ODN affect intestinal epithelial cells (IEC). Epithelial IL1α may contribute to allergic sensitization via autocrine mediator release affecting dendritic cells (DC). We studied whether IL1α contributes to Th2-associated mediator release by activated IEC and IEC/DC cocultures and possible modulation by scGOS/lcFOS±CpG-ODN. METHODS: Solid phase or transwell cultured IEC were preincubated with IL1α and/or IFNγ/TNFα for 6 h. The transwell IEC were also apically exposed to scGOS/lcFOS±CpG-ODN for 6 h, washed, and re-exposed, while cocultured with immature moDC (ccDC) for 48 h. These ccDC were subsequently added to allogeneic naïve T cells (MLR). IEC- and/or DC-derived mediators and T cell cytokines were measured. RESULTS: IL1α tended to enhance IL25 and enhanced IL33 and CCL20 release by IEC, while IL1α or TNFα or IFNγ enhanced CCL22. These were all further increased upon combined exposure of IFNγ/TNFα±IL1α coinciding with increased IL33 secretion in the solid phase culture. In the transwell, IL25 and IL33 remained under detection, while CCL20 and CCL22 were induced by IL1α or IFNγ/TNFα, respectively, and a synergistic increase was observed upon combined exposure of IFNγ/TNFα and IL1α. Furthermore, IFNγ was found to enhance galectin-9 secretion, which was more pronounced in IFNγ/TNFα±IL1α-exposed IEC and coincided with TGFß increase. Epithelial CpG-ODN exposure further increased CCL20, while reducing CCL22 release by IFNγ/TNFα/IL1α-activated IEC; however, scGOS/lcFOS suppressed both. Combined scGOS/lcFOS and CpG-ODN reduced CCL22, while CCL20 and regulatory galectin-9 and TGFß remained high in the supernatant of IFNγ/TNFα/IL1α-activated IEC and the following IEC/DC coculture. ccDC of scGOS/lcFOS- and CpG-ODN-exposed IFNγ/TNFα/IL1α-activated IEC increased IFNγ, IL10, TGFß, and galectin-9 secretion in the MLR compared to ccDC exposed to control-activated IEC. CONCLUSION: IL1α enhanced CCL20 and Th2-associated CCL22 release by IFNγ/TNFα-activated IEC. Combined scGOS/lcFOS and CpG-ODN exposure suppressed CCL22, while maintaining high CCL20, TGFß, and galectin-9 concentrations. In addition, ccDC derived from this IEC/DC coculture enhanced Th1 and regulatory mediator secretion mimicking known in vivo effects.


Assuntos
Galectinas/metabolismo , Oligossacarídeos/farmacologia , Células Th1/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/metabolismo , Células Cultivadas , Células HT29 , Humanos , Teste de Cultura Mista de Linfócitos , Células Th1/efeitos dos fármacos
6.
J Nutr ; 148(8): 1372-1379, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29986071

RESUMO

Background: Dietary nondigestible, short-chain galacto-, long-chain fructo-, and pectin-derived acidic oligosaccharides (GFAs) lower the effector response in cow-milk-allergic (CMA) mice; and forkhead box P3 (Foxp3)-positive regulatory T cells (Tregs) were shown to contribute to this. Objective: The aim of this study was to assess the contribution of interleukin 10 (IL-10) and transforming growth factor ß (TGF-ß) to the protective effect of the GFA diet in CMA mice. Methods: Female C3H/HeOuJ mice, 3-4 wk old, were orally sensitized with cholera toxin (Sham) or whey and cholera toxin (Whey) 1 time/wk for 5 consecutive weeks and challenged with whey 1 wk later. The mice were fed a control or 1% GFA (9:2:1) (Whey+GFA) diet starting 2 wk before the first sensitization. In a second experiment, the mice were also injected with αIL-10 receptor (αIL-10r), αTGF-ß, or isotype control antibodies 24 h before each sensitization. The acute allergic skin response, anaphylaxis score, whey-specific IgE, mucosal mast cell protease 1 (mMCP-1), and Treg frequency in the mesenteric lymph nodes (MLNs) and intestinal Foxp3, Il10, and Tgfb mRNA expression were determined. Results: In Whey+GFA mice, intestinal Il10, Tgfb, or Foxp3 mRNA expression was 2-10 times higher (P < 0.05) and the MLN Treg frequency was 25% higher compared with Whey mice (P < 0.05). The acute allergic skin response was 50% lower in Whey+GFA mice compared with Whey mice (P < 0.01), and IL-10 receptor (IL-10r) or TGF-ß neutralizing antibodies prevented this protective effect (P < 0.001). The Whey mice had higher serum mMCP-1 concentrations and whey-immunoglobulin E (-IgE) levels than Sham mice (P < 0.01), whereas these were not higher in Whey+GFA mice, and neutralizing antibodies partially interfered with these responses. Conclusions: Dietary GFAs enhance the Treg frequency in the MLNs and mucosal IL-10 and TGF-ß transcription while suppressing the allergic effector response. Neutralizing antibodies showed that the allergy-protective effect of the GFA diet was mediated by IL-10 and TGF-ß in CMA mice.


Assuntos
Interleucina-10/metabolismo , Hipersensibilidade a Leite/prevenção & controle , Leite/imunologia , Oligossacarídeos/uso terapêutico , Receptores de Interleucina-10/metabolismo , Linfócitos T Reguladores/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Bovinos , Quimases/sangue , Dieta , Carboidratos da Dieta/farmacologia , Carboidratos da Dieta/uso terapêutico , Feminino , Fatores de Transcrição Forkhead/metabolismo , Imunoglobulina E/sangue , Intestinos , Linfonodos/metabolismo , Mastócitos/metabolismo , Mesentério , Camundongos Endogâmicos C3H , Hipersensibilidade a Leite/sangue , Hipersensibilidade a Leite/metabolismo , Mucosa/metabolismo , Oligossacarídeos/farmacologia , RNA Mensageiro/metabolismo , Pele/imunologia , Soro do Leite/imunologia
7.
Respir Res ; 16: 17, 2015 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-25849971

RESUMO

BACKGROUND: Allergic asthma is strongly associated with the exposure to house dust mite (HDM) and is characterized by eosinophilic pulmonary inflammation and airway hyperresponsiveness (AHR). Recently, there is an increased interest in using dietary oligosaccharides, also known as prebiotics, as a novel strategy to prevent the development of, or reduce, symptoms of allergy. AIM: We investigated the preventive capacity of dietary galacto-oligosaccharides (GOS) compared to an intra-airway therapeutic treatment with budesonide on the development of HDM-induced allergic asthma in mice. METHODS: BALB/c mice were intranasally sensitized with 1 µg HDM on day 0 followed by daily intranasal challenge with PBS or 10 µg HDM on days 7 to 11. Two weeks prior to the first sensitization and throughout the experiment mice were fed a control diet or a diet containing 1% GOS. Reference mice were oropharyngeally instilled with budesonide (500 µg/kg) on days 7, 9, 11, and 13, while being fed the control diet. On day 14, AHR was measured by nebulizing increasing doses of methacholine into the airways. At the end of the experiment, bronchoalveolar lavage fluid (BALF) and lungs were collected. RESULTS: Sensitization and challenge with HDM resulted in AHR. In contrast to budesonide, dietary intervention with 1% GOS prevented the development of AHR. HDM sensitization and challenge resulted in a significant increase in BALF leukocytes numbers, which was suppressed by budesonide treatment and dietary intervention with 1% GOS. Moreover, HDM sensitization and challenge resulted in significantly enhanced concentrations of IL-6, CCL17, IL-33, CCL5 and IL-13 in lung tissue. Both dietary intervention with 1% GOS or budesonide treatment significantly decreased the HDM-induced increased concentrations of CCL5 and IL-13 in lung tissue, while budesonide also reduced the HDM-enhanced concentrations of IL-6 and CCL17 in lung tissue. CONCLUSION: Not only did dietary intervention with 1% GOS during sensitization and challenge prevent the induction of airway eosinophilia and Th2-related cytokine and chemokine concentrations in the lung equally effective as budesonide treatment, it also prevented AHR development in HDM-allergic mice. GOS might be useful for the prevention and/or treatment of symptoms in asthmatic disease.


Assuntos
Asma/dietoterapia , Hiper-Reatividade Brônquica/prevenção & controle , Broncoconstrição , Carboidratos da Dieta/administração & dosagem , Galactosídeos/administração & dosagem , Pulmão , Oligossacarídeos/administração & dosagem , Prebióticos/administração & dosagem , Eosinofilia Pulmonar/prevenção & controle , Pyroglyphidae , Animais , Asma/imunologia , Asma/metabolismo , Asma/fisiopatologia , Hiper-Reatividade Brônquica/imunologia , Hiper-Reatividade Brônquica/metabolismo , Hiper-Reatividade Brônquica/fisiopatologia , Líquido da Lavagem Broncoalveolar/imunologia , Broncoconstrição/efeitos dos fármacos , Broncodilatadores/farmacologia , Budesonida/farmacologia , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/fisiopatologia , Masculino , Camundongos Endogâmicos BALB C , Eosinofilia Pulmonar/imunologia , Eosinofilia Pulmonar/metabolismo , Eosinofilia Pulmonar/fisiopatologia , Células Th2/imunologia , Células Th2/metabolismo
8.
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
9.
Int Arch Allergy Immunol ; 167(2): 127-36, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26304032

RESUMO

BACKGROUND: The alarmin interleukin 33 (IL-33) and its receptor ST2 play an important role in mucosal barrier tissues, and seem to be crucial for Th2-cell mediated host defense. Galacto-oligosaccharides (GOS), used in infant formulas, exhibit gut and immune modulatory effects. To enhance our understanding of the immunomodulatory capacity of GOS, this study investigated the impact of dietary GOS intervention on IL-33 and ST2 expression related to intestinal barrier dysfunction and asthma. METHODS: B6C3F1 and BALB/c mice were fed a control diet with or without 1% GOS. To simulate intestinal barrier dysfunction, B6C3F1 mice received a gavage with the mycotoxin deoxynivalenol (DON). To mimic asthma-like inflammatory airway responses, BALB/c mice were sensitized on day 0 and challenged on days 7-11 with house-dust mite (HDM) allergen. Samples from the intestines and lungs were collected for IL-33 and ST2 analysis by qRT-PCR, immunoblotting and immunohistochemistry. RESULTS: Dietary GOS counteracted the DON-induced IL-33 mRNA expression and changed the IL-33 distribution pattern in the mouse small intestine. The IL-33 mRNA expression was positively correlated to the intestinal permeability. A strong positive correlation was also observed between IL-33 mRNA expression in the lung and the number of bronchoalveolar fluid cells. Reduced levels of IL-33 protein, altered IL-33 distribution and reduced ST2 mRNA expression were observed in the lungs of HDM-allergic mice after GOS intervention. CONCLUSIONS: Dietary GOS mitigated IL-33 at the mucosal surfaces in a murine model for intestinal barrier dysfunction and HDM-induced asthma. This promising effect may open up new avenues to use GOS not only as a prebiotic in infant nutrition, but also as a functional ingredient that targets inflammatory processes and allergies associated with IL-33 expression.


Assuntos
Inflamação/prevenção & controle , Interleucina-33/genética , Interleucina-33/metabolismo , Oligossacarídeos/administração & dosagem , Oligossacarídeos/imunologia , Alarminas , Animais , Antígenos de Dermatophagoides/administração & dosagem , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Dieta , Galactosídeos/administração & dosagem , Galactosídeos/imunologia , Imunidade nas Mucosas , Imunossupressores/administração & dosagem , Inflamação/genética , Inflamação/imunologia , Proteína 1 Semelhante a Receptor de Interleucina-1 , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Interleucina/química , Receptores de Interleucina/genética , Receptores de Interleucina/metabolismo , Tricotecenos/toxicidade
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 ; 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
12.
Trends Biotechnol ; 42(1): 119-134, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37580191

RESUMO

The global population is growing, rapidly increasing the demand for sustainable, novel, and safe food proteins with minimal risks of food allergy. In vitro testing of allergy-sensitizing capacity is predominantly based on 2D assays. However, these lack the 3D environment and crosstalk between the gut, skin, and immune cells essential for allergy prediction. Organ-on-a-chip (OoC) technologies are promising to study type 2 immune activation required for sensitization, initiated in the small intestine or skin, in interlinked systems. Increasing the mechanistic understanding and, moreover, finding new strategies to study interorgan communication is of importance to recapitulate food allergen sensitization in vitro. Here, we outline recently developed OoC platforms and discuss the features needed for reliable prediction of sensitizing allergenicity of proteins.


Assuntos
Hipersensibilidade Alimentar , Imunoglobulina E , Humanos , Pele , Alérgenos , Dispositivos Lab-On-A-Chip
13.
Adv Healthc Mater ; : e2304569, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38625078

RESUMO

Ever since the implementation of microfluidics in the biomedical field, in vitro models have experienced unprecedented progress that has led to a new generation of highly complex miniaturized cell culture platforms, known as Organs-on-a-Chip (OoC). These devices aim to emulate biologically relevant environments, encompassing perfusion and other mechanical and/or biochemical stimuli, to recapitulate key physiological events. While OoCs excel in simulating diverse organ functions, the integration of the immune organs and immune cells, though recent and challenging, is pivotal for a more comprehensive representation of human physiology. This comprehensive review covers the state of the art in the intricate landscape of immune OoC models, shedding light on the pivotal role of biofabrication technologies in bridging the gap between conceptual design and physiological relevance. The multifaceted aspects of immune cell behavior, crosstalk, and immune responses that are aimed to be replicated within microfluidic environments, emphasizing the need for precise biomimicry are explored. Furthermore, the latest breakthroughs and challenges of biofabrication technologies in immune OoC platforms are described, guiding researchers toward a deeper understanding of immune physiology and the development of more accurate and human predictive models for a.o., immune-related disorders, immune development, immune programming, and immune regulation.

14.
Food Funct ; 15(12): 6488-6501, 2024 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-38804660

RESUMO

Mustard seeds belong to the food category of mandatory labelling due to the severe reactions they can trigger in allergic patients. However, the mechanisms underlying allergic sensitization to mustard seeds are poorly understood. The aim of this work is to study type 2 immune activation induced by the mustard seed major allergen Sin a1 via the intestinal mucosa, employing an in vitro model mimicking allergen exposure via the intestinal epithelial cells (IECs). Sin a1 was isolated from the total protein extract and exposed to IEC, monocyte derived dendritic cells (DCs) or IEC/DC co-cultures. A system of consecutive co-cultures was employed to study the generic capacity of Sin a1 to induce type 2 activation leading to sensitization: IEC/DC, DC/T-cell, T/B-cell and stem cell derived mast cells (MCs) derived from healthy donors. Immune profiles were determined by ELISA and flow cytometry. Sin a1 activated IEC and induced type-2 cytokine secretion in IEC/DC co-culture or DC alone (IL-15, IL-25 and TSLP), and primed DC induced type 2 T-cell skewing. IgG secretion in the T-cell/B-cell phase was enhanced in the presence of Sin a1 in the first stages of the co-culture. Anti-IgE did not induce degranulation but promoted IL-13 and IL-4 release by MC primed with the supernatant from B-cells co-cultured with Sin a1-IEC/DC or -DC primed T-cells. Sin a1 enhanced the release of type-2 inflammatory mediators by epithelial and dendritic cells; the latter instructed generic type-2 responses in T-cells that resulted in B-cell activation, and finally MC activation upon anti-IgE exposure. This indicates that via activation of IEC and/or DC, mustard seed allergen Sin a1 is capable of driving type 2 immunity which may lead to allergic sensitization.


Assuntos
Alérgenos , Células Dendríticas , Células Epiteliais , Mostardeira , Sementes , Células Dendríticas/imunologia , Células Dendríticas/efeitos dos fármacos , Humanos , Sementes/química , Alérgenos/imunologia , Células Epiteliais/imunologia , Células Epiteliais/efeitos dos fármacos , Mucosa Intestinal/imunologia , Técnicas de Cocultura , Antígenos de Plantas/imunologia , Mastócitos/imunologia , Mastócitos/efeitos dos fármacos , Imunoglobulina E/imunologia , Citocinas/metabolismo , Proteínas de Plantas/imunologia , Proteínas de Plantas/farmacologia
15.
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
16.
Pediatr Allergy Immunol ; 24(7): 633-44, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24112425

RESUMO

Food allergy is a growing health concern in the westernized world with approx. 6% of children suffering from it. A lack of approved treatment has led to strict avoidance of the culprit food proteins being the only standard of care. Nowadays in-depth research is conducted to evaluate the possible use of allergen-specific immunotherapy (SIT) as an active therapeutic option for food allergy. Various routes of administration for the immunotherapy are investigated, including subcutaneous, oral, sublingual, and epicutaneous, and some appear to be successful in inducing a temporary tolerant state. Most research has been conducted with oral immunotherapy due to its efficacious and relatively safe profile. Increasing interest is dedicated to safer and more convenient approaches, such as sublingual and epicutaneous SIT; however, doubts exist about their possible capacity to induce temporary tolerant state and permanent oral tolerance. The high frequency of allergic adverse reactions of the various approaches and the inability to achieve permanent oral tolerance have highlighted the need of refinements in the strategies. A promising strategy for preventing IgE cross-linking and thus enhancing safety of SIT, while still activating T cells, is the use of tolerogenic peptides. The implementation of such an immunotherapy approach has the potential of not only increasing the chance of achieving a permanent state of tolerance, but also improving the safety and tolerability of the therapy. Immunotherapy for food allergy is still not ready for the clinic, but current and upcoming studies are dedicated to collect enough evidence for the possible implementation of allergen-SIT as a standard treatment for food allergy.


Assuntos
Alérgenos/uso terapêutico , Dessensibilização Imunológica/métodos , Hipersensibilidade Alimentar/terapia , Fragmentos de Peptídeos/uso terapêutico , Alérgenos/imunologia , Animais , Protocolos Clínicos , Dessensibilização Imunológica/efeitos adversos , Hipersensibilidade Alimentar/imunologia , Humanos , Tolerância Imunológica , Medição de Risco , Resultado do Tratamento
17.
Pediatr Allergy Immunol ; 24(7): 656-64, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24028387

RESUMO

BACKGROUND: Prior exposure to partial whey hydrolysates has been shown to reduce the allergic response to whey in mice. This effect was more pronounced in combination with a diet containing non-digestible oligosaccharides (scGOS/lcFOS/pAOS). It is unknown which fractions/epitopes are responsible for this effect. Therefore, the prophylactic ability of synthetic peptides of ß-lactoglobulin with/without a scGOS/lcFOS/pAOS-containing diet to reduce the allergic response in a mouse model for cow's milk allergy was investigated. METHODS: Of 31 peptides, nine peptides were selected based on human T cell data. Mice were pre-treated orally with three peptide mixtures or single peptides for six consecutive days. During this period, they received a control or scGOS/lcFOS/pAOS-containing diet. Subsequently, mice were orally sensitized to whey and received an intradermal and oral challenge. After sacrifice, serum and mesenteric lymph nodes (MLN) were collected for further analysis. RESULTS: Prior exposure to peptide mixtures 1 and 3 significantly reduced the acute allergic skin response to whey. Mixture 2 showed no effect. An additive effect of the scGOS/lcFOS/pAOS-containing diet was only observed for mixture 1. Of the peptides in mixture 1, one peptide (LLDAQSAPLRVYVEELKP) showed the strongest effect on the acute allergic skin response. This peptide also tended to decrease whey-specific antibody levels and to increase the percentages of CD11b+CD103+ dendritic cells and CD25+Foxp3+ T cells in the MLN. CONCLUSIONS: Prior exposure to specific peptides of ß-lactoglobulin reduces the allergic response to whey, which may involve regulatory dendritic and T cells. Combining peptides with a sGOS/lcFOS/pAOS-containing diet enhances this effect.


Assuntos
Alérgenos/administração & dosagem , Lactoglobulinas/administração & dosagem , Hipersensibilidade a Leite/terapia , Oligossacarídeos/administração & dosagem , Fragmentos de Peptídeos/administração & dosagem , Linfócitos T/imunologia , Administração Oral , Alérgenos/imunologia , Sequência de Aminoácidos , Animais , Bovinos , Linhagem Celular , Proliferação de Células , Criança , Modelos Animais de Doenças , Feminino , Humanos , Lactoglobulinas/imunologia , Camundongos , Camundongos Endogâmicos C3H , Hipersensibilidade a Leite/imunologia , Dados de Sequência Molecular , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/imunologia
18.
Br J Nutr ; 109(10): 1821-31, 2013 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-23021516

RESUMO

Increased n-6 and reduced n-3 long-chain PUFA (LC-PUFA) intake in Western diets may contribute to the increased prevalence of allergic diseases. Key effector cells in allergy are mast cells (MC). The aim of the present study was to investigate the effects of n-6 v. n-3 LC-PUFA on MC phenotype. Human MC lines (LAD2 and HMC-1) were incubated for 24 h with either arachidonic acid (AA, n-6 LC-PUFA) or the n-3 LC-PUFA EPA or DHA. The effects of these three LC-PUFA on degranulation, mediator secretion and reactive oxygen species (ROS) generation were assessed. ROS, mitogen-activated protein kinase (MAPK) or NF-κB inhibitors were used to unravel signalling pathways involved in cytokine secretion. AA, EPA or DHA did not reduce IgE-mediated degranulation by LAD2 cells. However, AA increased PGD2 and TNF-α secretion by ionomycin/phorbol 12-myristate 13-acetate-stimulated HMC-1, whereas EPA and DHA more prominently inhibited IL-4 and IL-13 secretion. Suppression of IL-4 and IL-13 release by LC-PUFA correlated with reduced ROS generation. IL-4 and IL-13 release by activated HMC-1 was abrogated using ROS inhibitors. Inhibition of MAPK signalling, but not NF-κB, downstream of ROS reduced IL-13 secretion by activated HMC-1. Combined incubation of EPA or DHA with MAPK inhibitors further suppressed IL-13 secretion. In conclusion, the n-6 LC-PUFA AA enhanced pro-inflammatory mediator production by MC, while the n-3 LC-PUFA EPA as well as DHA more effectively suppressed ROS generation and IL-4 and IL-13 release. This suggests that dietary supplementation with EPA and/or DHA may alter the MC phenotype, contributing to a reduced susceptibility to develop and sustain allergic disease.


Assuntos
Ácido Araquidônico/efeitos adversos , Gorduras na Dieta/uso terapêutico , Ácidos Graxos Ômega-3/uso terapêutico , Hipersensibilidade/tratamento farmacológico , Inflamação/prevenção & controle , Mastócitos/efeitos dos fármacos , Espécies Reativas de Oxigênio/antagonistas & inibidores , Linhagem Celular , Gorduras na Dieta/farmacologia , Ácidos Docosa-Hexaenoicos/farmacologia , Ácidos Docosa-Hexaenoicos/uso terapêutico , Ácido Eicosapentaenoico/farmacologia , Ácido Eicosapentaenoico/uso terapêutico , Ácidos Graxos Ômega-3/farmacologia , Humanos , Hipersensibilidade/imunologia , Hipersensibilidade/metabolismo , Inflamação/induzido quimicamente , Inflamação/metabolismo , Interleucinas/metabolismo , Mastócitos/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Transdução de Sinais
19.
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
20.
Biomolecules ; 13(2)2023 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-36830632

RESUMO

Proper early life immune development creates a basis for a healthy and resilient immune system, which balances immune tolerance and activation. Deviations in neonatal immune maturation can have life-long effects, such as development of allergic diseases. Evidence suggests that human milk oligosaccharides (HMOS) possess immunomodulatory properties essential for neonatal immune maturation. To understand the immunomodulatory properties of enzymatic or bacterial produced HMOS, the effects of five HMOS (2'FL, 3FL, 3'SL, 6'SL and LNnT), present in human milk have been studied. A PBMC immune model, the IEC barrier model and IEC/PBMC transwell coculture models were used, representing critical steps in mucosal immune development. HMOS were applied to IEC cocultured with activated PBMC. In the presence of CpG, 2'FL and 3FL enhanced IFNγ (p < 0.01), IL10 (p < 0.0001) and galectin-9 (p < 0.001) secretion when added to IEC; 2'FL and 3FL decreased Th2 cell development while 3FL enhanced Treg polarization (p < 0.05). IEC were required for this 3FL mediated Treg polarization, which was not explained by epithelial-derived galectin-9, TGFß nor retinoic acid secretion. The most pronounced immunomodulatory effects, linking to enhanced type 1 and regulatory mediator secretion, were observed for 2'FL and 3FL. Future studies are needed to further understand the complex interplay between HMO and early life mucosal immune development.


Assuntos
Leucócitos Mononucleares , Leite Humano , Recém-Nascido , Humanos , Leite Humano/metabolismo , Técnicas de Cocultura , Leucócitos Mononucleares/metabolismo , Oligossacarídeos/farmacologia , Galectinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA