Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 14(1): 4329, 2024 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-38383774

RESUMEN

A probiotic-related benefit for the host is inherently linked to metabolic activity and integration in the gut ecosystem. To facilitate these, probiotics are often combined with specific prebiotics in a synbiotic formulation. Here, we propose an approach for improving probiotic metabolic activity and engraftment. By cultivating the probiotic strain in the presence of a specific prebiotic (preconditioning), the bacterial enzymatic machinery is geared towards prebiotic consumption. Today, it is not known if preconditioning constitutes an advantage for the synbiotic concept. Therefore, we assessed the effects galacto-oligosaccharide (GOS) addition and preconditioning on GOS of Limosilactobacillus reuteri DSM 17938 on ex vivo colonic metabolic profiles, microbial community dynamics, and osteoblastogenesis. We show that adding GOS and preconditioning L. reuteri DSM 17938 act on different scales, yet both increase ex vivo short-chain fatty acid (SCFA) production and engraftment within the microbial community. Furthermore, preconditioned supernatants or SCFA cocktails mirroring these profiles decrease the migration speed of MC3T3-E1 osteoblasts, increase several osteogenic differentiation markers, and stimulate bone mineralization. Thus, our results demonstrate that preconditioning of L. reuteri with GOS may represent an incremental advantage for synbiotics by optimizing metabolite production, microbial engraftment, microbiome profile, and increased osteoblastogenesis.


Asunto(s)
Limosilactobacillus reuteri , Microbiota , Probióticos , Osteogénesis , Probióticos/farmacología , Prebióticos , Oligosacáridos/farmacología , Oligosacáridos/metabolismo , Ácidos Grasos Volátiles
2.
Nutrients ; 12(8)2020 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-32751149

RESUMEN

BACKGROUND: This study evaluated the impact of Bifidobacterium animalis ssp. lactis CNCM I-3446, Bovine Milk-derived OligoSaccharides (BMOS) and their combination on infant gut microbiota in vitro. In addition, a novel strategy consisting of preculturing B. lactis with BMOS to further enhance their potential synbiotic effects was assessed. METHOD: Short-term fecal batch fermentations (48 h) were used to assess the microbial composition and activity modulated by BMOS alone, B. lactis grown on BMOS or dextrose alone, or their combinations on different three-month-old infant microbiota. RESULTS: BMOS alone significantly induced acetate and lactate production (leading to pH decrease) and stimulated bifidobacterial growth in 10 donors. A further in-depth study on two different donors proved B. lactis ability to colonize the infant microbiota, regardless of the competitiveness of the environment. BMOS further enhanced this engraftment, suggesting a strong synbiotic effect. This was also observed at the microbiota activity level, especially in a donor containing low initial levels of bifidobacteria. In this donor, preculturing B. lactis with BMOS strengthened further the early modulation of microbiota activity observed after 6 h. CONCLUSION: This study demonstrated the strong synbiotic effect of BMOS and B. lactis on the infant gut microbiota, and suggests a strategy to improve its effectiveness in an otherwise low-Bifidobacterium microbiota.


Asunto(s)
Bifidobacterium animalis , Leche/química , Oligosacáridos/farmacología , Probióticos/farmacología , Simbióticos , Animales , Bovinos , Heces/microbiología , Femenino , Fermentación/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Lactante , Masculino , ARN Ribosómico 16S/análisis
3.
Nutrients ; 11(9)2019 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-31540231

RESUMEN

Food allergies have become a major healthcare concern, hence preventive efforts to ensure oral tolerance induction to newly introduced antigens are particularly relevant. Given that transforming growth factor-ß (TGF-ß) plays a key role in immune tolerance, we tested whether an infant formula enriched with TGF-ß would improve oral tolerance induction. A partially hydrolyzed whey protein-based formula was enriched with cow's-milk-derived TGF-ß (TGF-ß-enriched formula) by adding a specific whey protein isolate (WPI). The manufacturing process was optimized to achieve a concentration of TGF-ß within the range of human breast milk concentrations. Protection from allergic sensitization and immune response was assessed in a mouse model. Adult mice received the TGF-ß-enriched formula, a control non-enriched formula, or water ad libitum for 13 days before sensitization and suboptimal tolerization to ovalbumin (OVA). When compared to non-tolerized mice, suboptimally-tolerized mice supplemented with the TGF-ß-enriched formula showed significantly lower levels of total immunoglobulin-E (IgE) and OVA-specific (IgG1). Mouse mast-cell protease-1 (mMCP-1) and cytokine levels were also significantly decreased in suboptimally-tolerized mice fed the TGF-ß-enriched formula. In conclusion, oral supplementation with cow's-milk-derived TGF-ß decreased allergic responses to newly introduced allergens and thus reduced the risk of developing food allergy.


Asunto(s)
Hipersensibilidad a los Alimentos/prevención & control , Tolerancia Inmunológica/efectos de los fármacos , Fórmulas Infantiles/análisis , Factor de Crecimiento Transformador beta/administración & dosificación , Alérgenos/inmunología , Animales , Bovinos , Quimasas , Citocinas/sangre , Femenino , Humanos , Inmunoglobulina G/sangre , Lactante , Ratones , Ratones Endogámicos C57BL , Leche/química , Ovalbúmina/inmunología , Factor de Crecimiento Transformador beta/análisis , Proteína de Suero de Leche
5.
Int Arch Allergy Immunol ; 164(1): 64-73, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24902890

RESUMEN

BACKGROUND: Egg allergy is one of the most common food allergies in children. The standard therapy for egg allergy is strict avoidance. Yet, there is considerable clinical and scientific interest in primary or secondary prevention. A major drawback of oral tolerance (OT) induction protocols, however, is the possibility of severe side effects; thus, we have formulated a hypoallergenic egg product and demonstrate its in vivo capacity to modulate the immune system in the current study. METHODS: Hydrolyzed egg (HE) was produced using a combination of moderate heat treatment and enzymatic hydrolysis. The capacity of HE to induce OT was tested in experimental models and compared to whole egg (WE). Delayed-type hypersensitivity (DTH) responses, immune markers and potential early markers of OT were analyzed. RESULTS: Allergic responses, assessed by both DTH responses upon OVA challenge and serum OVA-specific IgE and IgG1, were decreased after treatment with HE and WE compared to the control group. Additionally, feeding WE and HE significantly decreased Th2 cytokine induction and cell proliferation, induced the activation of effector CD4+ T cells and increased numbers and percentages of ICOS+CD4+CD25+Foxp3+ cells. Furthermore, DO11.10 mouse experiments showed that HE contains other peptides than the OVA323-339 peptide that are able to induce tolerance to OVA. CONCLUSIONS: Altogether, results showed that HE induces OT in mice in a dose-dependent manner. Due to its low allergenicity compared to WE, it may represent a safer alternative for OT induction in at-risk subjects or oral immunotherapy in allergic patients.


Asunto(s)
Desensibilización Inmunológica/métodos , Hipersensibilidad al Huevo/inmunología , Tolerancia Inmunológica/inmunología , Ovalbúmina/inmunología , Animales , Cromatografía Liquida , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Hidrólisis , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL
6.
J Agric Food Chem ; 60(43): 10858-66, 2012 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-23043375

RESUMEN

The immunomodulatory potential of fragments derived from the cow's milk allergen bovine ß-lactoglobulin (BLG) was assessed in a mouse model of oral tolerance (OT) [Adel-Patient, K.; Wavrin, S.; Bernard, H.; Meziti, N.; Ah-Leung, S.; Wal, J. M. Oral tolerance and Treg cells are induced in BALB/c mice after gavage with bovine ß-lactoglobulin. Allergy 2011, 66 (10), 1312-1321]. Native BLG (nBLG) and chemically denatured BLG (lacking S-S bridges, dBLG), products resulting from their hydrolysis using cyanogen bromide (CNBr) and some synthetic peptides, were produced and precisely characterized. CNBr hydrolysates correspond to pools of peptides of various sizes that are still associated by S-S bridges when derived from nBLG. nBLG, dBLG, and CNBr hydrolysate of nBLG efficiently prevented further sensitization. CNBr hydrolysate of dBLG was less efficient, suggesting that the association by S-S bridges of peptides increased their immunomodulatory potential. Conversely, synthetic peptides were inefficient even if covering 50% of the BLG sequence, demonstrating that the immunomodulatory potential requires the presence of all derived fragments of BLG and further supporting the use of partially hydrolyzed milk proteins to favor OT induction in infants with a risk of atopy.


Asunto(s)
Hipersensibilidad a los Alimentos/inmunología , Tolerancia Inmunológica , Lactoglobulinas/química , Lactoglobulinas/inmunología , Péptidos/inmunología , Animales , Bovinos , Bromuro de Cianógeno/química , Femenino , Hipersensibilidad a los Alimentos/dietoterapia , Humanos , Hidrólisis , Lactoglobulinas/metabolismo , Ratones , Ratones Endogámicos BALB C , Péptidos/síntesis química , Péptidos/metabolismo , Hidrolisados de Proteína/inmunología , Hidrolisados de Proteína/metabolismo
7.
Clin Dev Immunol ; 2012: 485750, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21961022

RESUMEN

OBJECTIVE: Utilizing a food allergy murine model, we have investigated the intrinsic antiallergic potential of the Lactococcus lactis NCC 2287 strain. METHODS: BALB/c mice were sensitized at weekly intervals with ovalbumin (OVA) plus cholera toxin (CT) by the oral route for 7 weeks. In this model, an oral challenge with a high dose of OVA at the end of the sensitization period leads to clinical symptoms. Lactococcus lactis NCC 2287 was given to mice via the drinking water during sensitization (prevention phase) or after sensitization (management phase). RESULTS: Lactococcus lactis NCC 2287 administration to sensitized mice strikingly reduced allergic manifestations in the management phase upon challenge, when compared to control mice. No preventive effect was observed with the strain. Lactococcus lactis NCC 2287 significantly decreased relative expression levels of the Th-2 cytokine, IL-13, and associated chemokines CCL11 (eotaxin-1) and CCL17 (TARC) in the ileum. No effect was observed in the jejunum. CONCLUSION/SIGNIFICANCE: These results taken together designate Lactococcus lactis NCC 2287 as a candidate probiotic strain appropriate in the management of allergic symptoms.


Asunto(s)
Quimiocina CCL11/antagonistas & inhibidores , Quimiocina CCL17/antagonistas & inhibidores , Íleon/inmunología , Interleucina-13/antagonistas & inhibidores , Lactococcus lactis/metabolismo , Probióticos/administración & dosificación , Administración Oral , Animales , Quimiocina CCL11/biosíntesis , Quimiocina CCL17/biosíntesis , Toxina del Cólera/administración & dosificación , Toxina del Cólera/efectos adversos , Ensayo de Inmunoadsorción Enzimática , Femenino , Hipersensibilidad a los Alimentos/inmunología , Hipersensibilidad a los Alimentos/metabolismo , Hipersensibilidad a los Alimentos/terapia , Íleon/efectos de los fármacos , Íleon/metabolismo , Inmunoglobulina E/análisis , Inmunoglobulina G/análisis , Interleucina-13/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ovalbúmina/administración & dosificación , Ovalbúmina/efectos adversos , Probióticos/uso terapéutico
8.
FEMS Microbiol Ecol ; 79(1): 192-202, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22029421

RESUMEN

Faecal microbiota of healthy infant displays a large abundance of Bifidobacterium spp. and Bacteroides spp. Although some studies have reported an association between these two genera and allergy, these findings remain a subject of debate. Using a gnotobiotic mouse model of cow's milk allergy, we investigated the impact of an infant gut microbiota ­ mainly composed of Bifidobacterium and Bacteroides spp. ­ on immune activation and allergic manifestations. The transplanted microbiota failed to restore an ileal T-cell response similar to the one observed in conventional mice. This may be due to the low bacterial translocation into Peyer's patches in gnotobiotic mice. The allergic response was then monitored in germ-free, gnotobiotic, and conventional mice after repeated oral sensitization with whey proteins and cholera toxin. Colonized mice displayed a lower drop of rectal temperature upon oral challenge with b-lactoglobulin, lower plasma mMCP-1, and lower anti-BLG IgG1 than germ-free mice. The foxp3 gene was highly expressed in the ileum of both colonized mice that were protected against allergy. This study is the first demonstration that a transplanted healthy infant microbiota mainly composed of Bifidobacterium and Bacteroides had a protective impact on sensitization and food allergy in mice despite altered T-cell response in the ileum.


Asunto(s)
Íleon/microbiología , Inmunidad Celular , Metagenoma/fisiología , Hipersensibilidad a la Leche/microbiología , Leche/efectos adversos , Animales , Bacteroides/fisiología , Bifidobacterium/fisiología , Modelos Animales de Enfermedad , Heces/microbiología , Tracto Gastrointestinal , Vida Libre de Gérmenes , Humanos , Íleon/inmunología , Inmunoglobulina G/sangre , Lactante , Ratones , Ratones Endogámicos C3H , Hipersensibilidad a la Leche/inmunología , Hipersensibilidad a la Leche/prevención & control , Linfocitos T/inmunología , Linfocitos T Reguladores
9.
Int Arch Allergy Immunol ; 158(1): 35-42, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22205338

RESUMEN

BACKGROUND: Enhancing clinical efficacy remains a major goal in allergen-specific immunotherapy. In this study, we tested three strains of bifidobacteria as candidate adjuvants for sublingual allergy vaccines. METHODS: Probiotic candidates were evaluated in human monocyte-derived dendritic cell (h-DC) maturation and CD4(+) T-cell polarization in vitro models and further tested in murine models of sublingual immunotherapy in BALB/c mice sensitized to either ovalbumin or birch pollen. RESULTS: Bifidobacterium adolescentis, B. bifidum and B. longum induced h-DC maturation and polarized naïve CD4(+) T cells toward interferon-γ and interleukin-10 production. B. bifidum increased CD25(high), Foxp3(+) cells within CD4(+) T lymphocytes and was the most potent inducer of interferon-γ in Th2-skewed peripheral blood mononuclear cells and h-DC T-cell cocultures. It also induced a significant decrease in airway hyperresponsiveness in BALB/c mice sensitized to ovalbumin. Sublingual administration of B. bifidum together with recombinant Bet v 1 enhanced tolerance induction in BALB/c mice sensitized to birch pollen, with a downregulation of both airway hyperresponsiveness, lung inflammation and Bet v 1-specific Th2 responses. CONCLUSIONS: Due to its capacity to reorient established Th2 responses toward Th1/regulatory T-cell profiles, B. bifidum represents a valid candidate adjuvant for specific immunotherapy of type I allergies.


Asunto(s)
Bifidobacterium/inmunología , Desensibilización Inmunológica , Tolerancia Inmunológica , Probióticos , Administración Sublingual , Alérgenos/inmunología , Animales , Antígenos de Plantas/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular/inmunología , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Regulación hacia Abajo , Femenino , Humanos , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Interleucina-10/biosíntesis , Interleucina-10/inmunología , Ratones , Ratones Endogámicos BALB C , Ovalbúmina/inmunología , Polen/inmunología
10.
FEMS Microbiol Ecol ; 76(1): 133-44, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21223329

RESUMEN

Studies suggesting that the development of atopy is linked to gut microbiota composition are inconclusive on whether dysbiosis precedes or arises from allergic symptoms. Using a mouse model of cow's milk allergy, we aimed at investigating the link between the intestinal microbiota, allergic sensitization, and the severity of symptoms. Germ-free and conventional mice were orally sensitized with whey proteins and cholera toxin, and then orally challenged with ß-lactoglobulin (BLG). Allergic responses were monitored with clinical symptoms, plasma markers of sensitization, and the T-helper Th1/Th2/regulatory-T-cell balance. Microbiota compositions were analysed using denaturing gradient gel electrophoresis and culture methods. Germ-free mice were found to be more responsive than conventional mice to sensitization, displaying a greater reduction of rectal temperature upon challenge, higher levels of blood mouse mast cell protease-1 (mMCP-1) and BLG-specific immunoglobulin G1 (IgG1), and a systemic Th2-skewed response. This may be explained by a high susceptibility to release mMCP-1 even in the presence of low levels of IgE. Sensitization did not alter the microbiota composition. However, the absence of or low Staphylococcus colonization in the caecum was associated with high allergic manifestations. This work demonstrates that intestinal colonization protects against oral sensitization and allergic response. This is the first study to show a relationship between alterations within the subdominant microbiota and severity of food allergy.


Asunto(s)
Ciego/microbiología , Metagenoma/inmunología , Hipersensibilidad a la Leche/inmunología , Proteínas de la Leche/inmunología , Animales , Bovinos , Ciego/inmunología , Quimasas/sangre , Quimasas/inmunología , ADN Bacteriano/aislamiento & purificación , Modelos Animales de Enfermedad , Heces/microbiología , Vida Libre de Gérmenes , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Lactoglobulinas/farmacología , Ratones , Ratones Endogámicos C3H , Hipersensibilidad a la Leche/sangre , Bazo/citología , Bazo/inmunología , Proteína de Suero de Leche
11.
Gastroenterology ; 137(4): 1380-90.e1-3, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19596011

RESUMEN

BACKGROUND & AIMS: The commensal microbiota is believed to have an important role in regulating immune responsiveness and preventing intestinal inflammation. Intestinal microbes produce signals that regulate inflammation via Toll-like receptor (TLR) signaling, but the mechanisms of this process are poorly understood. We investigated the role of the anti-inflammatory cytokine interleukin (IL)-10 in this signaling pathway using a mouse model of colitis. METHODS: Clinical, histopathologic, and functional parameters of intestinal inflammation were evaluated in TLR4(-/-), IL-10(-/-), and TLR4(-/-) x IL-10(-/-) mice that were free of specific pathogens and in TLR4(-/-) x IL-10(-/-) mice following eradication and reintroduction of Helicobacter hepaticus. Regulatory T-cell (Treg) function was evaluated by crossing each of the lines with transgenic mice that express green fluorescent protein under control of the endogenous regulatory elements of Foxp3. Apoptotic cells in the colonic lamina propria were detected by a TUNEL assay. RESULTS: TLR4-mediated signals have 2 interrelated roles in promoting inflammation in TLR4(-/-) x IL-10(-/-) mice. In the absence of TLR4-mediated signals, secretion of proinflammatory and immunoregulatory cytokines is dysregulated. Tregs (Foxp3(+)) that secrete interferon-gamma and IL-17 accumulate in the colonic lamina propria of TLR4(-/-) x IL-10(-/-) mice and do not prevent inflammation. Aberrant control of epithelial cell turnover results in the persistence of antigen-presenting cells that contain apoptotic epithelial fragments in the colonic lamina propria of Helicobacter-infected TLR4(-/-) mice. CONCLUSIONS: In mice that lack both IL-10- and TLR4-mediated signals, aberrant regulatory T-cell function and dysregulated control of epithelial homeostasis combine to exacerbate intestinal inflammation.


Asunto(s)
Colitis/inmunología , Células Epiteliales/inmunología , Infecciones por Helicobacter/microbiología , Helicobacter hepaticus/inmunología , Mediadores de Inflamación/metabolismo , Interleucina-10/deficiencia , Linfocitos T Reguladores/inmunología , Receptor Toll-Like 4/metabolismo , Animales , Apoptosis , Colitis/microbiología , Colitis/patología , Colitis/prevención & control , Modelos Animales de Enfermedad , Células Epiteliales/microbiología , Células Epiteliales/patología , Factores de Transcripción Forkhead/genética , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/patología , Interferón gamma/metabolismo , Interleucina-10/genética , Interleucina-17/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Prolapso Rectal/inmunología , Prolapso Rectal/microbiología , Bazo/inmunología , Bazo/microbiología , Linfocitos T Reguladores/microbiología , Células TH1/inmunología , Células TH1/microbiología , Factores de Tiempo , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética
12.
Exp Dermatol ; 18(12): 1022-7, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19555432

RESUMEN

Prevalence of allergies has increased during the last two decades. Alteration of the gut microbiota composition is thought to play a crucial role in development of atopic diseases. Oral administration of probiotics has been reported to treat and/or prevent symptoms of atopic diseases in infants, but the results are still controversial. We investigated the potential efficacy of dietary interventions by a probiotic strain on prevention and treatment of atopic dermatitis (AD) in a human-like AD model, NC/NgaTnd mice by perinatal administration. Pregnant NC/NgaTnd mice were orally treated with the probiotic strain Lactobacillus rhamnosus CGMCC 1.3724 (LPR), which was followed by treatment of pups until 12 weeks of age. LPR-treated mice exhibited significant lower clinical symptoms of dermatitis, reduced scratching frequency, lower levels of plasma total Immunoglobulin E and higher levels of interferon-gamma in skin biopsies, compared with untreated mice. The protective effect was also observed when mice started to be treated at weaning time (5 weeks of age) even with limited supplementation period of 2 weeks. However, treatment of mice with the probiotic starting 1 week after the onset of the disease (8 weeks of age) had limited effects. The usefulness of LPR for primary prevention of AD was supported.


Asunto(s)
Dermatitis Atópica/prevención & control , Interferón gamma/biosíntesis , Lacticaseibacillus rhamnosus , Probióticos/farmacología , Probióticos/uso terapéutico , Animales , Animales Recién Nacidos , Dermatitis Atópica/diagnóstico , Dermatitis Atópica/tratamiento farmacológico , Dermatitis Atópica/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/inmunología , Femenino , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Inmunoglobulina E/sangre , Interferón gamma/genética , Interleucina-10/genética , Ratones , Ratones Endogámicos , Embarazo , Probióticos/administración & dosificación , Piel/efectos de los fármacos , Piel/metabolismo , Piel/patología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
13.
Immunol Rev ; 206: 204-18, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16048551

RESUMEN

Allergic hyperreactivity is defined as an exaggerated immune response [typically immunoglobulin E (IgE) but also non-IgE mediated] toward harmless antigenic stimuli. The prevalence of allergic disease has increased dramatically during the last 20 years, especially in developed countries. Both genetic and environmental factors contribute to susceptibility to allergy. Evidence has emerged supporting the hypothesis that a reduction in antigenic stimulation brought about by widespread vaccination, improvements in standards of hygiene, and extensive use of antibiotics has contributed to the dysregulation of T-helper 2 cell (Th2) type responsiveness that typifies allergy. Regulation of the inherently Th2-biased mucosal immune response is crucial both to the maintenance of homeostasis at this strategic defensive barrier and to the prevention of allergic disease. The ability of Th1 responses to counter-regulate Th2 reactivity is well characterized. More recently, interest has centered on regulatory T cells, which can suppress both Th1 and Th2 cells through the secretion of immunosuppressive cytokines such as interleukin-10 and transforming growth factor-beta. In this review, we discuss the basic cellular mechanisms of allergic diseases at mucosal surfaces, focusing on allergic responses to food, before examining newer work that suggests the induction of allergic hyperreactivity is due to a deficient immunoregulatory network, a lack of microbial stimulation, or both.


Asunto(s)
Hipersensibilidad/inmunología , Hipersensibilidad/microbiología , Inmunidad Mucosa , Animales , Humanos , Hipersensibilidad/tratamiento farmacológico
14.
Clin Diagn Lab Immunol ; 11(2): 266-71, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15013974

RESUMEN

We have previously demonstrated that Lactobacillus paracasei NCC2461 may help to prevent cow's milk allergy in mice by inducing oral tolerance to beta-lactoglobulin (BLG). To investigate the mechanisms involved in this beneficial effect, we examined the possibility that L. paracasei induces tolerance by hydrolyzing BLG-derived peptides and liberating peptides that stimulate interleukin-10 (IL-10) production. L. paracasei peptidases have been shown to hydrolyze tryptic-chymotryptic peptides from BLG, releasing numerous small peptides with immunomodulating properties. We have now shown that acidic tryptic-chymotryptic peptides stimulate splenocyte proliferation and gamma interferon (IFN-gamma) production in vitro. Hydrolysis of these peptides with L. paracasei peptidases repressed the lymphocyte stimulation, up-regulated IL-10 production, and down-regulated IFN-gamma and IL-4 secretion. L. paracasei NCC2461 may therefore induce oral tolerance to BLG in vivo by degrading acidic peptides and releasing immunomodulatory peptides stimulating regulatory T cells, which function as major immunosuppressive agents by secreting IL-10.


Asunto(s)
Tolerancia Inmunológica/inmunología , Interleucina-10/metabolismo , Lactobacillus/enzimología , Lactobacillus/inmunología , Lactoglobulinas/inmunología , Lactoglobulinas/metabolismo , Animales , Proteínas Bacterianas/metabolismo , División Celular/inmunología , Femenino , Hidrólisis , Interferón gamma/metabolismo , Linfocitos/citología , Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Péptido Hidrolasas/metabolismo , Bazo/inmunología
15.
Ann N Y Acad Sci ; 1029: 374-8, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15681785

RESUMEN

Ibuprofen and antibiotics are commonly prescribed during early childhood. When given to mice at the time at which oral tolerance is induced, both treatments affect either the induction or the maintenance of oral tolerance. These results suggest that the coadministration of these and similarly acting drugs should be considered cautiously for infants at risk of allergy.


Asunto(s)
Antibacterianos/farmacología , Ibuprofeno/farmacología , Tolerancia Inmunológica/inmunología , Animales , Preescolar , Dinoprostona/metabolismo , Heces/microbiología , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Inmunoglobulina E/sangre , Ratones , Ratones Endogámicos BALB C
16.
Clin Diagn Lab Immunol ; 10(5): 787-92, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12965905

RESUMEN

In this study, the effect of Lactobacillus paracasei (NCC 2461), Lactobacillus johnsonii (NCC 533) and Bifidobacterium lactis Bb12 (NCC 362) on the induction and maintenance of oral tolerance to bovine beta-lactoglobulin (BLG) was investigated in mice. Germfree mice were monocolonized with one of the three strains before oral administration of whey protein to induce tolerance. Mice were then injected with BLG and sacrificed 28 or 50 days after whey protein feeding for humoral and cellular response measurement. Conventional and germfree mice were used as controls. Both humoral and cellular responses were better suppressed in conventional mice than in germfree and monoassociated mice throughout the experiment and better suppressed in L. paracasei-associated mice than in mice colonized with B. lactis or L. johnsonii. The latter two mono-associations suppressed humoral responses only partially and cellular responses not at all. This study provides evidence that probiotics modulate the oral tolerance response to BLG in mice. The mono-colonization effect is strain-dependant, the best result having been obtained with L. paracasei.


Asunto(s)
Vida Libre de Gérmenes , Tolerancia Inmunológica , Lactoglobulinas/inmunología , Probióticos , Administración Oral , Animales , Bifidobacterium/inmunología , Heces/microbiología , Intestinos/inmunología , Intestinos/microbiología , Lactobacillus/inmunología , Ratones , Proteínas de la Leche/administración & dosificación , Proteínas de la Leche/inmunología , Proteína de Suero de Leche
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...