Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Allergy ; 76(5): 1398-1415, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33043467

RESUMO

BACKGROUND: Food allergy (FA) is a growing health problem worldwide. Effective strategies are advocated to limit the disease burden. Human milk (HM) could be considered as a protective factor against FA, but its mechanisms remain unclear. Butyrate is a gut microbiota-derived metabolite able to exert several immunomodulatory functions. We aimed to define the butyrate concentration in HM, and to see whether the butyrate concentration detected in HM is able to modulate the mechanisms of immune tolerance. METHODS: HM butyrate concentration from 109 healthy women was assessed by GS-MS. The effect of HM butyrate on tolerogenic mechanisms was assessed in in vivo and in vitro models. RESULTS: The median butyrate concentration in mature HM was 0.75 mM. This butyrate concentration was responsible for the maximum modulatory effects observed in all experimental models evaluated in this study. Data from mouse model show that in basal condition, butyrate up-regulated the expression of several biomarkers of gut barrier integrity, and of tolerogenic cytokines. Pretreatment with butyrate significantly reduced allergic response in three animal models of FA, with a stimulation of tolerogenic cytokines, inhibition of Th2 cytokines production and a modulation of oxidative stress. Data from human cell models show that butyrate stimulated human beta defensin-3, mucus components and tight junctions expression in human enterocytes, and IL-10, IFN-γ and FoxP3 expression through epigenetic mechanisms in PBMCs from FA children. Furthermore, it promoted the precursors of M2 macrophages, DCs and regulatory T cells. CONCLUSION: The study's findings suggest the importance of butyrate as a pivotal HM compound able to protect against FA.


Assuntos
Hipersensibilidade Alimentar , Microbioma Gastrointestinal , Animais , Butiratos , Hipersensibilidade Alimentar/prevenção & controle , Tolerância Imunológica , Leite Humano
2.
J Pediatr ; 213: 137-142.e2, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31327562

RESUMO

OBJECTIVE: To investigate whether the addition of the probiotic Lactobacillus rhamnosus GG (LGG) to the extensively hydrolyzed casein formula (EHCF) for cow's milk allergy (CMA) treatment could reduce the occurrence of functional gastrointestinal disorders (FGIDs). STUDY DESIGN: This cohort study included children with a positive history for CMA in the first year of life who were treated with EHCF alone or in combination with LGG and had evidence of immune tolerance acquisition to cow's milk for at least 12 months. FGID was diagnosed according to the Rome III diagnostic criteria by investigators unaware of previous treatment. A cohort of consecutive healthy children was also evaluated as a control population. RESULTS: A total of 330 subjects were included, 110 per cohort (EHCF, EHCF+LGG, and healthy controls). The rate of subjects with ≥1 FGID was significantly lower in the EHCF+LGG cohort compared with the EHCF cohort (40% vs 16.4%; P < .05). In the EHCF+LGG cohort, a lower incidence was observed for all components of the main study outcome. The prevalence of FGIDs in the healthy cohort was lower than that in the EHCF cohort and similar to that in the EHCF+LGG cohort. The incidence rate ratio of FGIDs for the EHCF+LGG cohort vs the EHCF cohort (0.40; 95% CI, 0.25-0.65; P < .001) was unmodified after correction for age at CMA diagnosis, breastfeeding, weaning time, and presence of a first-degree relative with an FGID. CONCLUSIONS: These results confirm the increased risk for developing FGIDs in children with CMA and suggest that EHCF+LGG could reduce this risk.


Assuntos
Caseínas/química , Alimentos Formulados , Gastroenteropatias/prevenção & controle , Lacticaseibacillus rhamnosus , Hipersensibilidade a Leite/dietoterapia , Probióticos/administração & dosagem , Animais , Bovinos , Criança , Pré-Escolar , Dieta , Feminino , Humanos , Hidrólise , Tolerância Imunológica , Masculino , Leite , Prevalência , Estudos Prospectivos , Risco , Resultado do Tratamento
3.
Adv Exp Med Biol ; 1125: 57-68, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30680644

RESUMO

The dramatic increase in food allergy prevalence and severity globally is demanding effective strategies. Food allergy derives from a defect in immune tolerance mechanisms. Immune tolerance is modulated by gut microbiota composition and function, and gut microbiota dysbiosis has been associated with the development of food allergy. Selected probiotic strains could act on immune tolerance mechanisms. The mechanisms are multiple and still not completely defined. Increasing evidence is providing useful information on the choice of optimal bacterial species/strains, dosage, and timing for intervention. The increased knowledge on the crucial role played by gut microbiota-derived metabolites, such as butyrate, is also opening the way to a post-biotic approach in the stimulation of immune tolerance.


Assuntos
Hipersensibilidade Alimentar/terapia , Microbioma Gastrointestinal , Probióticos/uso terapêutico , Disbiose/terapia , Humanos , Tolerância Imunológica
4.
J Allergy Clin Immunol ; 139(6): 1906-1913.e4, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28043872

RESUMO

BACKGROUND: Children with cow's milk allergy (CMA) have an increased risk of other allergic manifestations (AMs). OBJECTIVE: We performed a parallel-arm randomized controlled trial to test whether administration of an extensively hydrolyzed casein formula (EHCF) containing the probiotic Lactobacillus rhamnosus GG (LGG) can reduce the occurrence of other AMs in children with CMA. METHODS: Children with IgE-mediated CMA were randomly allocated to the EHCF or EHCF+LGG groups and followed for 36 months. The main outcome was occurrence of at least 1 AM (eczema, urticaria, asthma, and rhinoconjunctivitis). The secondary outcome was tolerance acquisition, which was defined as the negativization of a double-blind food challenge results at 12, 24, and 36 months. AMs were diagnosed according to standardized criteria. Tolerance acquisition was evaluated every 12 months. RESULTS: A total of 220 children (147 boys [67%]) with a median age of 5.0 months (interquartile range, 3.0-8.0 months) were randomized; 110 children were placed in the EHCF group, and 110 children were placed in the EHCF+LGG group. In the complete case analysis the absolute risk difference for the occurrence of at least 1 AM over 36 months was -0.23 (95% CI, -0.36 to -0.10; P < .001), and the absolute risk difference for the acquisition of cow's milk tolerance was 0.20 (95% CI, 0.05-0.35; P < .01) at 12 months, 0.24 (95% CI, 0.08-0.41; P < .01) at 24 months, and 0.27 (95% CI, 0.11-0.43; P < .001) at 36 months. In the sensitivity analysis the effect size of the main outcome was virtually unchanged when the occurrence of AMs was assigned to all 27 missing children. CONCLUSIONS: EHCF+LGG reduces the incidence of other AMs and hastens the development of oral tolerance in children with IgE-mediated CMA.


Assuntos
Caseínas/uso terapêutico , Alimentos Formulados , Lacticaseibacillus rhamnosus , Hipersensibilidade a Leite/terapia , Probióticos/uso terapêutico , Asma/imunologia , Asma/terapia , Conjuntivite/imunologia , Conjuntivite/terapia , Método Duplo-Cego , Eczema/imunologia , Eczema/terapia , Feminino , Humanos , Hidrólise , Tolerância Imunológica , Imunoglobulina E/imunologia , Lactente , Masculino , Hipersensibilidade a Leite/imunologia , Rinite Alérgica/imunologia , Rinite Alérgica/terapia , Testes Cutâneos , Urticária/imunologia , Urticária/terapia
5.
J Pediatr Gastroenterol Nutr ; 64(4): 632-638, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27437928

RESUMO

OBJECTIVES: The long-term effects of amino acid-based formula (AAF) in the treatment of cow's milk allergy (CMA) are largely unexplored. The present study comparatively evaluates body growth and protein metabolism in CMA children treated with AAF or with extensively hydrolyzed whey formula (eHWF), and healthy controls. METHODS: A 12-month multicenter randomized control trial was conducted in outpatients with CMA (age 5-12 m) randomized in 2 groups, treated with AAF (group 1) and eHWF (group 2), and compared with healthy controls (group 3) fed with follow-on (if age <12 months) or growing-up formula (if age >12 months). At enrolment (T0), after 3 (T3), 6 (T6), and 12 months (T12) a clinical evaluation was performed. At T0 and T3, in subjects with CMA serum levels of albumin, urea, total protein, retinol-binding protein, and insulin-like growth factor 1 were measured. RESULTS: Twenty-one subjects in group 1 (61.9% boys, age 6.5 ±â€Š1.5 months), 19 in group 2 (57.9% boys, age 7 ±â€Š1.7 months) and 25 subjects in group 3 (48% boys, age 5.5 ±â€Š0.5 months) completed the study. At T0, the weight z score was similar in group 1 (-0.74) and 2 (-0.76), with differences compared to group 3 (-0.17, P < 0.05). At T12, the weight z score value was similar between the 3 groups without significant differences. There were no significant changes in protein metabolism in children in groups 1 and 2. CONCLUSION: Long-term treatment with AAF is safe and allows adequate body growth in children with CMA.


Assuntos
Aminoácidos/uso terapêutico , Fórmulas Infantis , Hipersensibilidade a Leite/dietoterapia , Soro do Leite , Biomarcadores/sangue , Proteínas Sanguíneas/metabolismo , Estatura , Estudos de Casos e Controles , Feminino , Seguimentos , Humanos , Lactente , Análise de Intenção de Tratamento , Masculino , Hipersensibilidade a Leite/sangue , Resultado do Tratamento , Aumento de Peso
7.
JAMA Netw Open ; 5(12): e2244912, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36469320

RESUMO

Importance: The pediatric obesity disease burden imposes the necessity of new effective strategies. Objective: To determine whether oral butyrate supplementation as an adjunct to standard care is effective in the treatment of pediatric obesity. Design, Setting, and Participants: A randomized, quadruple-blind, placebo-controlled trial was performed from November 1, 2020, to December 31, 2021, at the Tertiary Center for Pediatric Nutrition, Department of Translational Medical Science, University of Naples Federico II, Naples, Italy. Participants included children aged 5 to 17 years with body mass index (BMI) greater than the 95th percentile. Interventions: Standard care for pediatric obesity supplemented with oral sodium butyrate, 20 mg/kg body weight per day, or placebo for 6 months was administered. Main Outcomes and Measures: The main outcome was the decrease of at least 0.25 BMI SD scores at 6 months. The secondary outcomes were changes in waist circumference; fasting glucose, insulin, total cholesterol, low-density lipoprotein cholesterol, high-density lipoprotein cholesterol, triglyceride, ghrelin, microRNA-221, and interleukin-6 levels; homeostatic model assessment of insulin resistance (HOMA-IR); dietary and lifestyle habits; and gut microbiome structure. Intention-to-treat analysis was conducted. Results: Fifty-four children with obesity (31 girls [57%], mean [SD] age, 11 [2.91] years) were randomized into the butyrate and placebo groups; 4 were lost to follow-up after receiving the intervention in the butyrate group and 2 in the placebo group. At intention-to-treat analysis (n = 54), children treated with butyrate had a higher rate of BMI decrease greater than or equal to 0.25 SD scores at 6 months (96% vs 56%, absolute benefit increase, 40%; 95% CI, 21% to 61%; P < .01). At per-protocol analysis (n = 48), the butyrate group showed the following changes as compared with the placebo group: waist circumference, -5.07 cm (95% CI, -7.68 to -2.46 cm; P < .001); insulin level, -5.41 µU/mL (95% CI, -10.49 to -0.34 µU/mL; P = .03); HOMA-IR, -1.14 (95% CI, -2.13 to -0.15; P = .02); ghrelin level, -47.89 µg/mL (95% CI, -91.80 to -3.98 µg/mL; P < .001); microRNA221 relative expression, -2.17 (95% CI, -3.35 to -0.99; P < .001); and IL-6 level, -4.81 pg/mL (95% CI, -7.74 to -1.88 pg/mL; P < .001). Similar patterns of adherence to standard care were observed in the 2 groups. Baseline gut microbiome signatures predictable of the therapeutic response were identified. Adverse effects included transient mild nausea and headache reported by 2 patients during the first month of butyrate intervention. Conclusions and Relevance: Oral butyrate supplementation may be effective in the treatment of pediatric obesity. Trial Registration: ClinicalTrials.gov Identifier: NCT04620057.


Assuntos
Butiratos , Obesidade Infantil , Criança , Feminino , Humanos , Butiratos/uso terapêutico , Colesterol , Método Duplo-Cego , Grelina , Insulina , MicroRNAs , Obesidade Infantil/tratamento farmacológico , Masculino , Adolescente
8.
Pediatr Ann ; 50(4): e178-e185, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34039171

RESUMO

Lactose intolerance is a common gastrointestinal condition caused by the inability to digest and absorb dietary lactose. Primary lactose intolerance is the most common type of lactose intolerance. It is one of the most common forms of food intolerance and occurs when lactase activity is reduced in the brush border of the small bowel mucosa. People may be lactose intolerant to varying degrees, depending on the severity of these symptoms. When lactose is not digested, it is fermented by gut microbiota, leading to abdominal pain, bloating, flatulence, and diarrhea with a considerable intraindividual and interindividual variability in the severity of clinical manifestations. These gastrointestinal symptoms are similar to cow's milk allergy and could be wrongly labeled as symptoms of "milk allergy." There are important differences between lactose intolerance and cow's milk allergy. Therefore, a better knowledge of these differences could limit misunderstandings in the diagnostic approach and in the management of these conditions. [Pediatr Ann. 2021;50(4):e178-e185.].


Assuntos
Gastroenteropatias , Intolerância à Lactose , Hipersensibilidade a Leite , Dor Abdominal/etiologia , Animais , Bovinos , Criança , Diarreia/etiologia , Feminino , Gastroenteropatias/diagnóstico , Gastroenteropatias/etiologia , Humanos , Intolerância à Lactose/diagnóstico , Hipersensibilidade a Leite/diagnóstico
9.
Ital J Pediatr ; 47(1): 151, 2021 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-34217356

RESUMO

BACKGROUND: Amino acid-based formula (AAF) is a relevant dietary strategy for paediatric patients affected by cow's milk allergy (CMA). The present study was designed to evaluate the hypoallergenicity of a new AAF in children with immunoglobulin (Ig)E-mediated CMA. METHODS: According to the criteria provided by the American Academy of Pediatrics Subcommittee on Nutrition and Allergic Diseases, we designed a prospective trial in CMA children (aged 1-36 months) aimed to demonstrate the hypoallergenicity of the new AAF in 90% of subjects with 95% confidence during the double-blind, placebo-controlled challenge (DBPCFC). A skin prick test (SPT) with the new AAF was also performed. RESULTS: Twenty-nine children [all Caucasian, 55.2% male, mean age (±SD) 16.9 ± 5.7 months] were enrolled. The SPT and the DBPCFC with the new AAF were negative in all study subjects. CONCLUSIONS: The study results support the hypoallergenicity of the new AAF. This formula could be considered an additional dietary option for non-breastfed children affected by CMA. TRIAL REGISTRATION: The trial was registered in the ClinicalTrials.gov Protocol Registration System (ID number: NCT03909113 ).


Assuntos
Aminoácidos/uso terapêutico , Fórmulas Infantis/química , Hipersensibilidade a Leite/imunologia , Animais , Bovinos , Pré-Escolar , Método Duplo-Cego , Feminino , Humanos , Imunoglobulina E/imunologia , Lactente , Masculino , Estudos Prospectivos , Testes Cutâneos
10.
Front Pediatr ; 8: 440, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32850553

RESUMO

Cow's milk allergy (CMA) is one of the most prevalent food allergies and the most expensive allergic diseases in the pediatric age. There is no cure for CMA, and actual disease management is based on strict avoidance of cow milk protein-containing foods, access to rescue medication, and use of substitutive formulas. Early-life CMA could be one of the first steps of the "allergic march" (AM), leading to the occurrence of other atopic manifestations later in the life, including asthma and oculorhinitis, with subsequent further increase of costs for health care systems and families of affected children. In the last years, diet is emerged as a relevant strategy to prevent allergic diseases through, at least in part, epigenetic modulation of immune system. We provide an overview of studies that investigate the potential role of different dietary strategies in preventing the AM in pediatric patients with CMA.

11.
Artigo em Inglês | MEDLINE | ID: mdl-32861173

RESUMO

The fatty acid (FA) composition of human milk (HM) from N = 9 Italian healthy donors following a free diet exhibited FA-dependent ranges of variability, as assessed by GC-FID. The possible short-term changes in the FA profile were monitored in the milk of lactating mothers (three) collected at five time points over a 6 h period, following an oral load (200 mL) of bovine milk. An array of techniques was exploited, including UHPLC-ESI-MS/MS of intact lipids and MALDI-TOF MS before and after chemical hydrogenation or bromination, in addition to MALDI-TOF MS analysis of FA after saponification, to monitor short-chain and odd-chain FA in HM as markers of bovine milk fat. A single administration of bovine milk did not appreciably modify the lipid pattern, suggesting that the maternal diet could induce not detectable short-term changes on the lipid composition of HM. Diet-induced increase of butyric acid was also excluded by 13C NMR. The functions that HM FA exert in infant physiology appear finely regulated through maternal metabolism.


Assuntos
Dieta , Ácidos Graxos/análise , Leite Humano/química , Leite/metabolismo , Animais , Bovinos , Cromatografia Líquida de Alta Pressão , Humanos , Espectroscopia de Ressonância Magnética , Espectrometria de Massas em Tandem
12.
J Clin Med ; 9(1)2020 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-31947953

RESUMO

Fatty liver disease is a serious complication of childhood obesity. Calorie-restricted regimen (RCR) is one of the effective therapy for this condition. Aim of the study was to evaluate the effect of lycopene-rich tomato sauce with oregano and basil extracts in obese children with fatty liver on RCR. 61 obese children with fatty liver were enrolled, 52 completed the study. A randomized cross over clinical trial was performed. Participants were assigned to RCR alone or with a supplement of lycopene-rich tomato juice for 60 days; subsequently, the groups were switched to the alternative regimen for the next 60 days. Reduction in BMI, HOMA-IR, cholesterol, triglycerides, liver size, and steatosis was more profound in tomato-supplemented group. Leptin decreased in both groups whereas adiponectin raised only after tomato supplementation. RCR is associated with the impaired engagement of T-cells glycolysis and proliferation, tomato-supplementation resulted in glycolytic metabolic activation of T-cells. Tomato juice ameliorates glucose and lipid metabolism in obese children, improve oxidative and inflammatory state and modulates the mitochondrial metabolism of T-cells contributing to a maintenance of a proper immune surveillance in children, impaired by RCR. The addition of tomato to RCR could be considered a protective and preventive support to obese child.

13.
Aliment Pharmacol Ther ; 51(1): 110-120, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31797399

RESUMO

BACKGROUND: The pathogenesis of infant colic is poorly defined. Gut microbiota seems to be involved, supporting the potential therapeutic role of probiotics. AIMS: To assess the rate of infants with a reduction of ≥50% of mean daily crying duration after 28 days of intervention with the probiotic Bifidobacterium animalis subsp. lactis BB-12® (BB-12). Secondary outcomes were daily number of crying episodes, sleeping time, number of bowel movements and stool consistency. METHODS: Randomized controlled trial (RCT) on otherwise healthy exclusively breastfed infants with infant colic randomly allocated to receive BB-12 (1 × 109  CFU/day) or placebo for 28 days. Gut microbiota structure and butyrate, beta-defensin-2 (HBD-2), cathelicidin (LL-37), secretory IgA (sIgA) and faecal calprotectin levels were assessed. RESULTS: Eighty infants were randomised, 40/group. The rate of infants with reduction of ≥50% of mean daily crying duration was higher in infants treated with BB-12, starting from the end of 2nd week. No infant relapsed when treatment was stopped. The mean number of crying episodes decreased in both groups, but with a higher effect in BB-12 group (-4.7 ± 3.4 vs -2.3 ± 2.2, P < 0.05). Mean daily stool frequency decreased in both groups but the effect was significantly higher in the BB-12 group; stool consistency was similar between the two groups. An increase in Bifidobacterium abundance (with significant correlation with crying time reduction), butyrate and HBD-2, LL-37, sIgA levels associated with a decrease in faecal calprotectin level were observed in the BB-12 group. CONCLUSIONS: Supplementation with BB-12 is effective in managing infant colic. The effect could derive from immune and non-immune mechanisms associated with a modulation of gut microbiota structure and function.


Assuntos
Bifidobacterium animalis , Cólica/dietoterapia , Probióticos/uso terapêutico , Aleitamento Materno , Cólica/microbiologia , Choro , Defecação , Método Duplo-Cego , Fezes/microbiologia , Feminino , Microbioma Gastrointestinal/fisiologia , Humanos , Lactente , Cuidado do Lactente/métodos , Masculino , Placebos , Resultado do Tratamento
14.
Front Immunol ; 10: 191, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30828329

RESUMO

The dramatic increase in food allergy prevalence and severity globally requires effective strategies. Food allergy derives from a defect in immune tolerance mechanisms. Immune tolerance is modulated by gut microbiota function and structure, and microbiome alterations (dysbiosis) have a pivotal role in the development of food allergy. Environmental factors, including a low-fiber/high-fat diet, cesarean delivery, antiseptic agents, lack of breastfeeding, and drugs can induce gut microbiome dysbiosis, and have been associated with food allergy. New experimental tools and technologies have provided information regarding the role of metabolites generated from dietary nutrients and selected probiotic strains that could act on immune tolerance mechanisms. The mechanisms are multiple and still not completely defined. Increasing evidence has provided useful information on optimal bacterial species/strains, dosage, and timing for intervention. The increased knowledge of the crucial role played by nutrients and gut microbiota-derived metabolites is opening the way to a post-biotic approach in the stimulation of immune tolerance through epigenetic regulation. This review focused on the potential role of gut microbiome as the target for innovative strategies against food allergy.


Assuntos
Hipersensibilidade Alimentar/imunologia , Hipersensibilidade Alimentar/terapia , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/imunologia , Fatores Etários , Animais , Dieta , Gerenciamento Clínico , Suscetibilidade a Doenças , Disbiose/imunologia , Ácidos Graxos/metabolismo , Hipersensibilidade Alimentar/metabolismo , Humanos , Tolerância Imunológica , Probióticos
15.
Sci Rep ; 9(1): 2828, 2019 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-30808949

RESUMO

Epigenetic mechanisms could drive the disease course of cow's milk allergy (CMA) and formula choice could modulate these pathways. We compared the effect of two different dietary approaches on epigenetic mechanisms in CMA children. Randomized controlled trial on IgE-mediated CMA children receiving a 12-month treatment with extensively hydrolyzed casein formula containing the probiotic L.rhamnosus GG (EHCF + LGG) or with soy formula (SF). At the baseline, after 6 and 12 months of treatment FoxP3 methylation rate and its expression in CD4+ T cells were assessed. At same study points IL-4, IL-5, IL-10, and IFN-γ methylation rate, expression and serum concentration, miRNAs expression were also investigated. 20 children (10/group) were evaluated. Baseline demographic, clinical and epigenetic features were similar in the two study groups. At 6 and 12 months, EHCF + LGG group showed a significant increase in FoxP3 demethylation rate compared to SF group. At the same study points, EHCF + LGG group presented a higher increase in IL-4 and IL-5 and a higher reduction in IL-10 and IFN-γ DNA methylation rate compared to SF group. A different modulation of miR-155, -146a, -128 and -193a expression was observed in EHCF + LGG vs. SF. Dietary intervention could exert a different epigenetic modulation on the immune system in CMA children.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Epigênese Genética , Fatores de Transcrição Forkhead/genética , Sistema Imunitário , Hipersensibilidade a Leite/dietoterapia , Animais , Caseínas/metabolismo , Bovinos , Criança , Metilação de DNA , Feminino , Humanos , Hidrólise , Lacticaseibacillus rhamnosus , Masculino , Hipersensibilidade a Leite/genética , Hipersensibilidade a Leite/imunologia , Probióticos
16.
Sci Rep ; 9(1): 9504, 2019 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-31239464

RESUMO

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

17.
Sci Rep ; 8(1): 12500, 2018 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-30131575

RESUMO

Cow's milk allergy (CMA) is one of the earliest and most common food allergy and can be elicited by both IgE- or non-IgE-mediated mechanism. We previously described dysbiosis in children with IgE-mediated CMA and the effect of dietary treatment with extensively hydrolyzed casein formula (EHCF) alone or in combination with the probiotic Lactobacillus rhamnosus GG (LGG). On the contrary, the gut microbiota in non-IgE-mediated CMA remains uncharacterized. In this study we evaluated gut microbiota composition and fecal butyrate levels in children affected by non-IgE-mediated CMA. We found a gut microbiota dysbiosis in non-IgE-mediated CMA, driven by an enrichment of Bacteroides and Alistipes. Comparing these results with those previously obtained in children with IgE-mediated CMA, we demonstrated overlapping signatures in the gut microbiota dysbiosis of non-IgE-mediated and IgE-mediated CMA children, characterized by a progressive increase in Bacteroides from healthy to IgE-mediated CMA patients. EHCF containg LGG was more strongly associated with an effect on dysbiosis and on butyrate production if compared to what observed in children treated with EHCF alone. If longitudinal cohort studies in children with CMA will confirm these results, gut microbiota dysbiosis could be a relevant target for innovative therapeutic strategies in children with non-IgE-mediated CMA.


Assuntos
Bactérias/classificação , Butiratos/análise , Disbiose/diagnóstico , Hipersensibilidade a Leite/microbiologia , Análise de Sequência de DNA/métodos , Animais , Bactérias/genética , Bactérias/isolamento & purificação , Bacteroides/classificação , Bacteroides/genética , Bacteroides/isolamento & purificação , Pré-Escolar , DNA Bacteriano/genética , DNA Ribossômico/genética , Disbiose/etiologia , Fezes/química , Microbioma Gastrointestinal , Humanos , Lactente , Estudos Longitudinais , Hipersensibilidade a Leite/metabolismo , RNA Ribossômico 16S/genética
18.
Nutrients ; 9(7)2017 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-28657607

RESUMO

The gut microbiota plays a pivotal role in immune system development and function. Modification in the gut microbiota composition (dysbiosis) early in life is a critical factor affecting the development of food allergy. Many environmental factors including caesarean delivery, lack of breast milk, drugs, antiseptic agents, and a low-fiber/high-fat diet can induce gut microbiota dysbiosis, and have been associated with the occurrence of food allergy. New technologies and experimental tools have provided information regarding the importance of select bacteria on immune tolerance mechanisms. Short-chain fatty acids are crucial metabolic products of gut microbiota responsible for many protective effects against food allergy. These compounds are involved in epigenetic regulation of the immune system. These evidences provide a foundation for developing innovative strategies to prevent and treat food allergy. Here, we present an overview on the potential role of gut microbiota as the target of intervention against food allergy.


Assuntos
Hipersensibilidade Alimentar/prevenção & controle , Hipersensibilidade Alimentar/terapia , Microbioma Gastrointestinal , Dieta , Dieta Hiperlipídica , Fibras na Dieta/administração & dosagem , Disbiose/prevenção & controle , Disbiose/terapia , Epigênese Genética , Hipersensibilidade Alimentar/imunologia , Humanos , Sistema Imunitário/microbiologia , Lactente , Metabolômica , Probióticos
19.
Nutrients ; 8(3): 157, 2016 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-26978392

RESUMO

Intolerance to carbohydrates is relatively common in childhood, but still poorly recognized and managed. Over recent years it has come to the forefront because of progresses in our knowledge on the mechanisms and treatment of these conditions. Children with intolerance to carbohydrates often present with unexplained signs and symptoms. Here, we examine the most up-to-date research on these intolerances, discuss controversies relating to the diagnostic approach, including the role of molecular analysis, and provide new insights into modern management in the pediatric age, including the most recent evidence for correct dietary treatment.


Assuntos
Erros Inatos do Metabolismo dos Carboidratos/dietoterapia , Erros Inatos do Metabolismo dos Carboidratos/diagnóstico , Doença Celíaca/dietoterapia , Doença Celíaca/diagnóstico , Dieta com Restrição de Carboidratos , Carboidratos da Dieta/efeitos adversos , Hipersensibilidade Alimentar/dietoterapia , Hipersensibilidade Alimentar/diagnóstico , Fatores Etários , Erros Inatos do Metabolismo dos Carboidratos/epidemiologia , Doença Celíaca/epidemiologia , Criança , Hipersensibilidade Alimentar/epidemiologia , Humanos , Intolerância à Lactose/diagnóstico , Intolerância à Lactose/dietoterapia , Valor Preditivo dos Testes , Fatores de Risco , Resultado do Tratamento
20.
Methods Mol Biol ; 1371: 215-21, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26530804

RESUMO

Childhood food allergy (FA) rates have rapidly increased with significant direct medical costs for the health care system and even larger costs for the families with a food-allergic child. The possible causes of food allergy become the target of intense scrutiny in recent years. Increasing evidence underline the importance in early life of gut microbiome in the development of allergic diseases. There are a range of factors in the modern environment that may be associated with changes to both the gut microbiome and risk of FA, such as mode of delivery, antibiotic exposure, infant feeding practices, farming environment, and country of origin. Knowledge of the relationship between early life gut microbiome and allergic diseases may facilitate development of novel preventive and treatment strategies. Based on our current knowledge, there are no currently available approved therapies for food allergy. More studies are needed to evaluate the safety and efficacy of allergen-specific and allergen-nonspecific approaches, as well as combination approaches.


Assuntos
Hipersensibilidade Alimentar/etiologia , Hipersensibilidade Alimentar/terapia , Adjuvantes Imunológicos/uso terapêutico , Alérgenos/imunologia , Animais , Dessensibilização Imunológica/métodos , Microbioma Gastrointestinal , Humanos , Tolerância Imunológica , Probióticos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA