Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Nutrients ; 16(16)2024 Aug 18.
Article in English | MEDLINE | ID: mdl-39203888

ABSTRACT

Although gut dysbiosis is associated with cow's milk allergy (CMA), causality remains uncertain. This study aimed to identify specific bacterial signatures that influence the development and outcome of the disease. We also investigated the effect of hypoallergenic formula (HF) consumption on the gut microbiome of milk-allergic children. 16S rRNA amplicon sequencing was applied to characterize the gut microbiome of 32 milk-allergic children aged 5-12 years and 36 age-matched healthy controls. We showed that the gut microbiome of children with CMA differed significantly from that of healthy children, regardless of whether they consumed cow's milk. Compared to that of healthy cow's milk consumers, it was depleted in Bifidobacterium, Coprococcus catus, Monoglobus, and Lachnospiraceae GCA-900066575, while being enriched in Oscillibacter valericigenes, Negativibacillus massiliensis, and three genera of the Ruminococcaceae family. Of these, only the Ruminococcaceae taxa were also enriched in healthy children not consuming cow's milk. Furthermore, the gut microbiome of children who developed tolerance and had received an HF was similar to that of healthy children, whereas that of children who had not received an HF was significantly different. Our results demonstrate that specific gut microbiome signatures are associated with CMA, which differ from those of dietary milk elimination. Moreover, HF consumption affects the gut microbiome of children who develop tolerance.


Subject(s)
Gastrointestinal Microbiome , Milk Hypersensitivity , Milk , RNA, Ribosomal, 16S , Humans , Milk Hypersensitivity/microbiology , Child, Preschool , Child , Female , Male , Animals , RNA, Ribosomal, 16S/genetics , Milk/microbiology , Dysbiosis/microbiology , Bacteria/classification , Bacteria/genetics , Cattle , Case-Control Studies , Feces/microbiology
2.
J Pediatr Gastroenterol Nutr ; 79(4): 841-849, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39175183

ABSTRACT

OBJECTIVES: Food protein-induced enterocolitis syndrome (FPIES) is a severe type of non-IgE (immunoglobulin E)-mediated (NIM) food allergy, with cow's milk (CM) being the most common offending food. The relationship between the gut microbiota and its metabolites with the inflammatory process in infants with CM FPIES is unknown, although evidence suggests a microbial dysbiosis in NIM patients. This study was performed to contribute to the knowledge of the interaction between the gut microbiota and its derived metabolites with the local immune system in feces of infants with CM FPIES at diagnosis. METHODS: Twelve infants with CM FPIES and a matched healthy control group were recruited and the gut microbiota was investigated by 16S amplicon and shotgun sequencing. Fatty acids (FAs) were measured by gas chromatography, while immune factors were determined by enzyme-linked immunosorbent assay and Luminex technology. RESULTS: A specific pattern of microbiota in the gut of CM FPIES patients was found, characterized by a high abundance of enterobacteria. Also, an intense excretion of FAs in the feces of these infants was observed. Furthermore, correlations were found between fecal bifidobacteria and immune factors. CONCLUSION: These fecal determinations may be useful to gain insight into the pathophysiology of this syndrome and should be taken in consideration for future studies of FPIES patients.


Subject(s)
Enterobacteriaceae , Enterocolitis , Feces , Gastrointestinal Microbiome , Milk Hypersensitivity , Humans , Infant , Male , Female , Milk Hypersensitivity/microbiology , Milk Hypersensitivity/immunology , Feces/microbiology , Enterobacteriaceae/isolation & purification , Enterocolitis/microbiology , Animals , Case-Control Studies , Fatty Acids/metabolism , Milk/microbiology , Dysbiosis/microbiology
3.
Ann Allergy Asthma Immunol ; 133(2): 203-210.e6, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38697287

ABSTRACT

BACKGROUND: Immune regulation by gut microbiota is affected by dysbiosis and may precede food allergy onset. Prior studies lacked comparisons stratified by age and clinical phenotype. OBJECTIVE: To assess the microbiome of children with food allergy (<3 years, 3-18 years) compared with similar aged children without food allergy. METHODS: A real-world prospective cross-sectional study performed from 2014 to 2019 recruited children highly likely to have milk, egg, or peanut allergy defined by history and serum IgE or confirmed by food challenge. 16S ribosomal RNA sequencing identified stool microbial DNA. Alpha and beta diversity was compared between groups with food allergy and healthy controls stratified by age. Differential abundance for non a priori taxa was accepted at absolute fold-change greater than 2 and q value less than 0.05. RESULTS: A total of 70 patients were included (56 with food allergy and 14 healthy controls). Groups were not significantly different in age, gender at birth, race, mode of delivery, breastfeeding duration, or antibiotic exposure. Younger children with food allergy had similar alpha diversity compared with controls. Beta diversity was significantly different by age (P = .001). There was differential abundance of several a priori (P < .05) taxa (including Clostridia) only in younger children. Both a priori (including Coprococcus and Clostridia) and non a priori (q < 0.05) Acidobacteria_Gp15, Aestuariispira, Tindallia, and Desulfitispora were significant in older children with food allergy, especially with peanut allergy. CONCLUSION: Dysbiosis associates with food allergy, most prominent in older children with peanut allergy. Younger children with and without food allergy have fewer differences in gut microbiota. This correlates with clinical observations of persistence of peanut allergy and improved efficacy and safety of oral immunotherapy in younger children. Age younger than 3 years should be considered when initiating therapeutic interventions.


Subject(s)
Egg Hypersensitivity , Gastrointestinal Microbiome , Milk Hypersensitivity , Peanut Hypersensitivity , Humans , Female , Child, Preschool , Child , Male , Peanut Hypersensitivity/immunology , Peanut Hypersensitivity/microbiology , Gastrointestinal Microbiome/immunology , Cross-Sectional Studies , Adolescent , Milk Hypersensitivity/immunology , Milk Hypersensitivity/microbiology , Egg Hypersensitivity/immunology , Prospective Studies , Dysbiosis/immunology , Dysbiosis/microbiology , Age Factors , Infant , Immunoglobulin E/blood , Immunoglobulin E/immunology , Feces/microbiology , Allergens/immunology , RNA, Ribosomal, 16S/genetics
5.
Cell Immunol ; 382: 104633, 2022 12.
Article in English | MEDLINE | ID: mdl-36347161

ABSTRACT

Loss of oral tolerance (OT) to food antigens results in food allergies. One component of achieving OT is the symbiotic microorganisms living in the gut (microbiota). The composition of the microbiota can drive either pro-tolerogenic or pro-inflammatory responses against dietary antigens though interactions with the local immune cells within the gut. Products from bacterial fermentation, such as butyrate, are one of the main communication molecules involved in this interaction, however, this is released by a subset of bacterial species. Thus, strategies to specifically expand these bacteria with protolerogenic properties have been explored to complement oral immunotherapy in food allergy. These approaches either provide digestible biomolecules to induce beneficial bacteria species (prebiotics) or the direct administration of live bacteria species (probiotics). While this combined therapy has shown positive outcomes in clinical trials for cow's milk allergy, more research is needed to determine if this therapy can be extended to other food allergens.


Subject(s)
Food Hypersensitivity , Gastrointestinal Microbiome , Microbiota , Milk Hypersensitivity , Probiotics , Cattle , Animals , Female , Food Hypersensitivity/therapy , Milk Hypersensitivity/microbiology , Probiotics/therapeutic use , Bacteria
7.
Nutrients ; 13(11)2021 Oct 26.
Article in English | MEDLINE | ID: mdl-34836050

ABSTRACT

Acute respiratory infections are a common cause of morbidity in infants and young children. This high rate of respiratory infections in early life has a major impact on healthcare resources and antibiotic use, with the associated risk of increasing antibiotic resistance, changes in intestinal microbiota composition and activity and, consequently, on the future health of children. An international group of clinicians and researchers working in infant nutrition and cow's milk allergy (CMA) met to review the available evidence on the prevalence of infections in healthy infants and in those with allergies, particularly CMA; the factors that influence susceptibility to infection in early life; links between infant feeding, CMA and infection risk; and potential strategies to modulate the gut microbiota and infection outcomes. The increased susceptibility of infants with CMA to infections, and the reported potential benefits with prebiotics, probiotics and synbiotics with regard to improving infection outcomes and reducing antibiotic usage in infants with CMA, makes this a clinically important issue that merits further research.


Subject(s)
Milk Hypersensitivity/microbiology , Probiotics/therapeutic use , Respiratory Tract Infections/microbiology , Anti-Bacterial Agents/therapeutic use , Drug Resistance, Microbial , Expert Testimony , Female , Gastrointestinal Microbiome , Humans , Infant , Infant Nutritional Physiological Phenomena , Infant, Newborn , Male , Prevalence , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/epidemiology
8.
Int J Mol Sci ; 22(4)2021 Feb 06.
Article in English | MEDLINE | ID: mdl-33562104

ABSTRACT

Food allergy (FA) and, in particular, IgE-mediated cow's milk allergy is associated with compositional and functional changes of gut microbiota. In this study, we compared the gut microbiota of cow's milk allergic (CMA) infants with that of cow's milk sensitized (CMS) infants and Healthy controls. The effect of the intake of a mixture of Bifidobacterium longum subsp. longum BB536, Bifidobacterium breve M-16V and Bifidobacterium longum subsp. infantis M-63 on gut microbiota modulation of CMA infants and probiotic persistence was also investigated. Gut microbiota of CMA infants resulted to be characterized by a dysbiotic status with a prevalence of some bacteria as Haemophilus, Klebsiella, Prevotella, Actinobacillus and Streptococcus. Among the three strains administered, B.longum subsp. infantis colonized the gastrointestinal tract and persisted in the gut microbiota of infants with CMA for 60 days. This colonization was associated with perturbations of the gut microbiota, specifically with the increase of Akkermansia and Ruminococcus. Multi-strain probiotic formulations can be studied for their persistence in the intestine by monitoring specific bacterial probes persistence and exploiting microbiota profiling modulation before the evaluation of their therapeutic effects.


Subject(s)
Bifidobacterium breve/metabolism , Bifidobacterium longum subspecies infantis/metabolism , Bifidobacterium/metabolism , Gastrointestinal Microbiome/physiology , Milk Hypersensitivity/therapy , Probiotics/therapeutic use , Animals , Breast Feeding , Child, Preschool , Dysbiosis/microbiology , Female , Humans , Immunoglobulin E/immunology , Infant , Male , Milk/immunology , Milk Hypersensitivity/microbiology
9.
Front Immunol ; 11: 1700, 2020.
Article in English | MEDLINE | ID: mdl-33042105

ABSTRACT

Background: Early nutrition may influence the development of food allergies later in life. In the absence of breastfeeding, hydrolysates from cow's milk proteins (CMP) were indicated as a prevention strategy in at risk infants, but their proof of effectiveness in clinical and pre-clinical studies is still insufficient. Thanks to a validated mouse model, we then assessed specific and nonspecific preventive effects of administration of extensive hydrolysates from caseins (eHC) on the development of food allergy to CMP. The additional nonspecific effect of the probiotic Lactobacillus GG (LGG), commonly used in infant formula, was also assessed. Methods: Groups of young BALB/cByJ female mice were pretreated by repeated gavage either with PBS (control mice), or with PBS solution containing non-hydrolyzed milk protein isolate (MPI), eHC or eHC+LGG (eq. of 10 mg of protein/gavage). All mice were then experimentally sensitized to CMP by gavage with whole CM mixed with the Th2 mucosal adjuvant Cholera toxin. All mice were further chronically exposed to cow's milk. A group of mice was kept naïve. Sensitization to both caseins and to the non-related whey protein ß-lactoglobulin (BLG) was evaluated by measuring specific antibodies in plasma and specific ex vivo Th2/Th1/Th17 cytokine secretion. Elicitation of the allergic reaction was assessed by measuring mMCP1 in plasma obtained after oral food challenge (OFC) with CMP. Th/Treg cell frequencies in gut-associated lymphoid tissue and spleen were analyzed by flow cytometry at the end of the protocol. Robust statistical procedure combining non-supervised and supervised multivariate analyses and univariate analyses, was conducted to reveal any effect of the pretreatments. Results: PBS pretreated mice were efficiently sensitized and demonstrated elicitation of allergic reaction after OFC, whereas mice pretreated with MPI were durably protected from allergy to CMP. eHC+/-LGG pretreatments had no protective effect on sensitization to casein (specific) or BLG (non-specific), nor on CMP-induced allergic reactions. Surprisingly, eHC+LGG mice demonstrated significantly enhanced humoral and cellular immune responses after sensitization with CMP. Only some subtle changes were evidenced by flow cytometry. Conclusion: Neither specific nor nonspecific preventive effects of administration of casein-derived peptides on the development of CMP food allergy were evidenced in our experimental setup. Further studies should be conducted to delineate the mechanisms involved in the immunostimulatory potential of LGG and to clarify its significance in clinical use.


Subject(s)
Caseins/administration & dosage , Lacticaseibacillus rhamnosus/physiology , Milk Hypersensitivity/prevention & control , Probiotics/administration & dosage , Animals , Antibodies/blood , Caseins/immunology , Cells, Cultured , Cytokines/blood , Disease Models, Animal , Female , Gastrointestinal Tract/immunology , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Immunity, Cellular , Immunity, Humoral , Lacticaseibacillus rhamnosus/immunology , Mice, Inbred C57BL , Milk Hypersensitivity/blood , Milk Hypersensitivity/immunology , Milk Hypersensitivity/microbiology , Spleen/immunology , Spleen/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
10.
Appl Environ Microbiol ; 86(21)2020 10 15.
Article in English | MEDLINE | ID: mdl-32826221

ABSTRACT

Cow's milk allergy is a worldwide public health issue, especially since there is no effective treatment, apart from milk and dairy product avoidance. The aim of this study was to assess the beneficial role of three probiotic strains previously selected for their prophylactic properties in a mouse model of ß-lactoglobulin allergy. Administration of Lactobacillus rhamnosus LA305, L. salivarius LA307, or Bifidobacterium longum subsp. infantis LA308 for 3 weeks post-sensitization and challenge modified the composition of the gut microbiota, with an increase in the Prevotella NK3B31 group and a decrease in Marvinbryantia, belonging to the Lachnospiraceae family. Although no impact on markers of sensitization was detected, modifications of foxp3, tgfß, and il10 ileal gene expression, as well as plasma metabolomic alterations in the tryptophan pathway, were observed. Moreover, ex vivo studies showed that all probiotic strains induced significant decreases in cytokine production by ß-lactoglobulin-stimulated splenocytes. Taken together, these results suggest that the three probiotic strains tested lead to alterations in immune responses, i.e., induction of a tolerogenic anergy and anti-inflammatory responses. This anergy could be linked to cecal microbiota modifications, although no impact on fecal short-chain fatty acid (SCFA) concentrations was detected. Anergy could also be linked to a direct impact of probiotic strains on dendritic cells, since costimulatory molecule expression was decreased following coincubation of these strains with bone marrow-derived dendritic cells (BMDCs). To conclude, all three candidate probiotic strains induced strain-specific gut microbiota and metabolic changes, which could potentially be beneficial for general health, as well as anergy, which could contribute to oral tolerance acquisition.IMPORTANCE We showed previously that three probiotic strains, i.e., Lactobacillus rhamnosus LA305, L. salivarius LA307, and Bifidobacterium longum subsp. infantis LA308, exerted different preventive effects in a mouse model of cow's milk allergy. In this study, we evaluated their potential benefits in a curative mouse model of cow's milk allergy. When administered for 3 weeks after the sensitization process and a first allergic reaction, none of the strains modified the levels of sensitization and allergic markers. However, all three strains affected gut bacterium communities and modified immune and inflammatory responses, leading to a tolerogenic profile. Interestingly, all three strains exerted a direct effect on dendritic cells, which are known to play a major role in food sensitization through their potentially tolerogenic properties and anergic responses. Taken together, these data indicate a potentially beneficial role of the probiotic strains tested in this model of cow's milk allergy with regard to tolerance acquisition.


Subject(s)
Gastrointestinal Microbiome , Immune Tolerance/immunology , Milk Hypersensitivity/microbiology , Probiotics/administration & dosage , Animals , Bifidobacterium longum subspecies infantis/chemistry , Cattle , Female , Lacticaseibacillus rhamnosus/chemistry , Ligilactobacillus salivarius/chemistry , Mice , Mice, Inbred BALB C , Probiotics/chemistry
11.
Allergol Immunopathol (Madr) ; 48(2): 149-157, 2020.
Article in English | MEDLINE | ID: mdl-31477403

ABSTRACT

OBJECTIVES: ß-lactoglobulin (ß-Lg)-sensitized mice model was employed to investigate the correlation between Lactobacillus acidophilus KLDS 1.0738 (Lap KLDS 1.0738) modulating gut microbiota and inducting Toll-like receptors (TLRs) expression. METHODS: The alterations of mice fecal microbiota were analyzed by 16S rRNA gene sequencing. The serum cytokines production and TLR4/NF-κB mRNA expression in the colon tissues were measured by ELISA kit and quantitative RT-PCR, respectively. RESULTS: The results showed that Lap KLDS 1.0738 pretreatment attenuated ß-Lg-induced hypersensitivity, accompanied with a diminished expression of TLR4/NF-κB signaling. Moreover, oral administration of Lap KLDS 1.0738 improved the richness and diversity of fecal microbiota, which was characterized by fewer Proteobacteria phylum and Helicobacteraceae family, and higher Firmicutes phylum and Lachnospiraceae family than allergic group. Notably, TLR4/NF-κB expression was positively correlated with the family of Helicobacteraceae in allergic group, but negatively correlated with the family of Lachnospiraceae, Ruminococcaceae and anti-inflammatory cytokines level. A significant positive correlation was observed between TLR4/NF-κB expression and the production of histamine, total IgE and pro-inflammatory cytokines. CONCLUSIONS: Intake of Lap KLDS 1.0738 can influence the gut bacterial composition, which might result in recognizing TLRs signaling so as to inhibit allergic response.


Subject(s)
Gastrointestinal Microbiome , Milk Hypersensitivity/immunology , Milk Hypersensitivity/microbiology , Probiotics/pharmacology , Toll-Like Receptor 4/immunology , Animals , Disease Models, Animal , Female , Gastrointestinal Microbiome/immunology , Lactobacillus acidophilus , Lactoglobulins/immunology , Lactoglobulins/toxicity , Mice , Mice, Inbred BALB C
12.
Nat Med ; 25(3): 448-453, 2019 03.
Article in English | MEDLINE | ID: mdl-30643289

ABSTRACT

There has been a striking generational increase in life-threatening food allergies in Westernized societies1,2. One hypothesis to explain this rising prevalence is that twenty-first century lifestyle practices, including misuse of antibiotics, dietary changes, and higher rates of Caesarean birth and formula feeding have altered intestinal bacterial communities; early-life alterations may be particularly detrimental3,4. To better understand how commensal bacteria regulate food allergy in humans, we colonized germ-free mice with feces from healthy or cow's milk allergic (CMA) infants5. We found that germ-free mice colonized with bacteria from healthy, but not CMA, infants were protected against anaphylactic responses to a cow's milk allergen. Differences in bacterial composition separated the healthy and CMA populations in both the human donors and the colonized mice. Healthy and CMA colonized mice also exhibited unique transcriptome signatures in the ileal epithelium. Correlation of ileal bacteria with genes upregulated in the ileum of healthy or CMA colonized mice identified a clostridial species, Anaerostipes caccae, that protected against an allergic response to food. Our findings demonstrate that intestinal bacteria are critical for regulating allergic responses to dietary antigens and suggest that interventions that modulate bacterial communities may be therapeutically relevant for food allergy.


Subject(s)
Anaphylaxis/microbiology , Fecal Microbiota Transplantation , Gastrointestinal Microbiome/genetics , Milk Hypersensitivity/microbiology , Animals , Clostridiales/genetics , Female , Food Hypersensitivity/microbiology , Germ-Free Life , Healthy Volunteers , Humans , Ileum/microbiology , Infant , Male , Mice
13.
Nutrients ; 10(10)2018 Oct 11.
Article in English | MEDLINE | ID: mdl-30314304

ABSTRACT

Cow's milk protein allergy (CMPA) is the most common food allergy in infancy. Non-IgE mediated (NIM) forms are little studied and the responsible mechanisms of tolerance acquisition remain obscure. Our aim was to study the intestinal microbiota and related parameters in the fecal samples of infants with NIM-CMPA, to establish potential links between type of formula substitutes, microbiota, and desensitization. Seventeen infants between one and two years old, diagnosed with NIM-CMPA, were recruited. They were all on an exclusion diet for six months, consuming different therapeutic protein hydrolysates. After this period, stool samples were obtained and tolerance development was evaluated by oral challenges. A control group of 10 age-matched healthy infants on an unrestricted diet were included in the study. Microbiota composition, short-chain fatty acids, calprotectin, and transforming growth factor (TGF)-ß1 levels were determined in fecal samples from both groups. Infants with NIM-CMPA that consumed vegetable protein-based formulas presented microbiota colonization patterns different from those fed with an extensively hydrolyzed formula. Differences in microbiota composition and fecal parameters between NIM-CMPA and healthy infants were observed. Non-allergic infants showed a significantly higher proportion of Bacteroides compared to infants with NIM-CMPA. The type of protein hydrolysate was found to determine gut microbiota colonization and influence food allergy resolution in NIM-CMPA cases.


Subject(s)
Diet/methods , Feces/microbiology , Gastrointestinal Microbiome/immunology , Milk Hypersensitivity/microbiology , Protein Hydrolysates/immunology , Child, Preschool , Female , Humans , Immune Tolerance , Immunoglobulin E/immunology , Infant , Infant Formula/microbiology , Infant, Newborn , Male , Milk Hypersensitivity/immunology , Plant Proteins, Dietary/immunology
14.
Sci Rep ; 8(1): 12500, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30131575

ABSTRACT

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.


Subject(s)
Bacteria/classification , Butyrates/analysis , Dysbiosis/diagnosis , Milk Hypersensitivity/microbiology , Sequence Analysis, DNA/methods , Animals , Bacteria/genetics , Bacteria/isolation & purification , Bacteroides/classification , Bacteroides/genetics , Bacteroides/isolation & purification , Child, Preschool , DNA, Bacterial/genetics , DNA, Ribosomal/genetics , Dysbiosis/etiology , Feces/chemistry , Gastrointestinal Microbiome , Humans , Infant , Longitudinal Studies , Milk Hypersensitivity/metabolism , RNA, Ribosomal, 16S/genetics
15.
Food Res Int ; 109: 416-425, 2018 07.
Article in English | MEDLINE | ID: mdl-29803466

ABSTRACT

The aim of this review paper is to assess the applicability of donkey's milk to infants suffering from Cow Milk Protein Allergy (CMPA) compared to human and other available milk types. The bioactive and immune-supportive character which could be beneficial as a fortifier to the formula-fed infants is described while limitations of this type of milk are also discussed. Studies showed that human and donkey's milk have similar, overall, chemical composition as well as protein homogeneity and antigenic similarities. Several in vitro and in vivo studies showed that donkey's milk has nutraceutical and functional properties that can support immunity, alter metabolism and beneficially modify gut microbiota. Clinical studies illustrated that donkeys' milk is well tolerated (82.6%-88%) by infants. Finally, the effect that processing (i.e. thermal, non-thermal treatments, drying methods) has on donkey milk components is also discussed pointing out the need for minimally processing this type of milk.


Subject(s)
Bottle Feeding , Dietary Supplements , Equidae , Infant Formula/chemistry , Milk Hypersensitivity/diet therapy , Milk/chemistry , Nutritive Value , Animals , Cross Reactions , Dietary Supplements/adverse effects , Gastrointestinal Microbiome , Humans , Infant , Infant Formula/adverse effects , Infant Nutritional Physiological Phenomena , Infant, Newborn , Milk/adverse effects , Milk/immunology , Milk Hypersensitivity/immunology , Milk Hypersensitivity/microbiology , Milk Hypersensitivity/physiopathology , Milk Proteins/immunology , Nutritional Status
16.
Int J Biol Macromol ; 112: 876-881, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29428389

ABSTRACT

Nowadays health benefits of bioactive food constituents, known as probiotic microorganisms, are a growing awareness. Cow's milk is a nutritious food containing probiotic bacteria. However, milk allergenicity is one of the most common food allergies. The milk protein, ß-lactoglobulin (BLG), is in about 80% of all main cases of milk allergies for children and infants. With the aim of screening proteolytic strains of lactic acid bacteria to evaluate their potential for the reduction of allergenicity of the major bovine milk proteins, we isolated new proteolytic strains of cocci lactic acid bacteria from traditional Iranian dairy products. The proteases produced by these strains had strong proteolytic activity against BLG. Proteolysis of BLG, observed after sodium dodecyl sulfate-PAGE, was confirmed by the analysis of the peptide profiles by reversed-phase HPLC. The two isolates were submitted to 16S rDNA sequencing and identified as Lactcoccus lactis subsp. cremoris and Lactcoccus lactis subsp. hordniea. The competitive ELISA experiments confirmed that these isolates, with high proteolytic activity, reduce significantly the allergenicity of BLG. Accordingly, these isolates can reduce the immunoreactivity of bovine milk proteins, which can be helpful for the production of low-allergic dairy products.


Subject(s)
Lactococcus lactis/isolation & purification , Lactoglobulins/adverse effects , Milk Hypersensitivity/microbiology , Milk/adverse effects , Proteolysis , Animals , Cattle , Chromatography, High Pressure Liquid , Chromatography, Reverse-Phase , Enzyme-Linked Immunosorbent Assay , Humans , Hydrolysis , Immunoglobulin E/metabolism , Iran , Serum/metabolism
17.
Nutrients ; 9(6)2017 May 24.
Article in English | MEDLINE | ID: mdl-28538698

ABSTRACT

From an evolutionary standpoint, allergy has only recently emerged as a significant health problem. Various hypotheses were proposed to explain this, but they all indicated the importance of rapid lifestyle changes, which occurred in industrialized countries in the last few decades. In this review, we discuss evidence from epidemiological and experimental studies that indicate changes in dietary habits may have played an important role in this phenomenon. Based on the example of dietary fiber, we discuss molecular mechanisms behind this and point towards the importance of diet-induced changes in the microbiota. Finally, we reason that future studies unraveling mechanisms governing these changes, along with the development of better tools to manipulate microbiota composition in individuals will be crucial for the design of novel strategies to combat numerous inflammatory disorders, including atopic diseases.


Subject(s)
Diet , Gastrointestinal Microbiome , Hypersensitivity/epidemiology , Life Style , Milk Hypersensitivity/epidemiology , Animals , Dietary Fats/administration & dosage , Dietary Fiber/administration & dosage , Disease Models, Animal , Fatty Acids, Volatile/administration & dosage , Humans , Hypersensitivity/microbiology , Lung/metabolism , Lung/microbiology , Milk/chemistry , Milk/immunology , Milk Hypersensitivity/microbiology , Milk, Human/chemistry , Milk, Human/immunology , Observational Studies as Topic
18.
J Allergy Clin Immunol ; 138(4): 1122-1130, 2016 10.
Article in English | MEDLINE | ID: mdl-27292825

ABSTRACT

BACKGROUND: Gut microbiota may play a role in the natural history of cow's milk allergy. OBJECTIVE: We sought to examine the association between early-life gut microbiota and the resolution of cow's milk allergy. METHODS: We studied 226 children with milk allergy who were enrolled at infancy in the Consortium of Food Allergy observational study of food allergy. Fecal samples were collected at age 3 to 16 months, and the children were followed longitudinally with clinical evaluation, milk-specific IgE levels, and milk skin prick test performed at enrollment, 6 months, 12 months, and yearly thereafter up until age 8 years. Gut microbiome was profiled by 16s rRNA sequencing and microbiome analyses performed using Quantitative Insights into Microbial Ecology (QIIME), Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt), and Statistical Analysis of Metagenomic Profiles (STAMP). RESULTS: Milk allergy resolved by age 8 years in 128 (56.6%) of the 226 children. Gut microbiome composition at age 3 to 6 months was associated with milk allergy resolution by age 8 years (PERMANOVA P = .047), with enrichment of Clostridia and Firmicutes in the infant gut microbiome of subjects whose milk allergy resolved. Metagenome functional prediction supported decreased fatty acid metabolism in the gut microbiome of subjects whose milk allergy resolved (η2 = 0.43; ANOVA P = .034). CONCLUSIONS: Early infancy is a window during which gut microbiota may shape food allergy outcomes in childhood. Bacterial taxa within Clostridia and Firmicutes could be studied as probiotic candidates for milk allergy therapy.


Subject(s)
Gastrointestinal Microbiome/physiology , Milk Hypersensitivity/microbiology , Bacteria/classification , Bacteria/genetics , Child , Child, Preschool , Dermatitis, Atopic/physiopathology , Feces/microbiology , Female , Gastrointestinal Microbiome/genetics , Humans , Immunoglobulin E/blood , Infant , Male , Phylogeny , RNA, Ribosomal, 16S/genetics
19.
J Sci Food Agric ; 96(9): 3180-7, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26459934

ABSTRACT

BACKGROUND: Cow milk allergy is the most common food allergy in children. So far, no effective treatment is available to prevent or cure food allergy. This study investigated whether orally administrated probiotics could suppress sensitisation in whey proteins (WP)-induced allergy mouse model. Two types of probiotic Dahi were prepared by co-culturing Dahi bacteria (Lactococcus lactis ssp. cremoris NCDC-86 and Lactococcus lactis ssp. lactis biovar diacetylactis NCDC-60) along with selected strain of Lactobacillus acidophilus LaVK2 and Bifidobacterium bifidum BbVK3. Mice were fed with probiotic Dahi (La-Dahi and LaBb-Dahi) from 7 days before sensitisation with WP, respectively, in addition to milk protein-free basal diet, and control group received no supplements. RESULTS: Feeding of probiotic Dahi suppressed the elevation of whey proteins-specific IgE and IgG response of WP-sensitised mice. In addition, sIgA levels were significantly (P < 0.001) increased in intestinal fluid collected from mice fed with La-Dahi. Production of T helper (Th)-1 cell-specific cytokines, i.e. interferon-γ (IFN-γ), interleukin (IL)-12, and IL-10 increased, while Th2-specific cytokines, i.e. IL-4 decreased in the supernatant of cultured splenocytes collected from mice fed with probiotic Dahi as compared to the other groups. Moreover, the splenic mRNA levels of IFN-γ, interleukin-10 were found to be significantly increased, while that of IL-4 decreased significantly in La-Dahi groups, as compared to control groups. CONCLUSION: Results of the present study indicate that probiotic Dahi skewed Th2-specific immune response towards Th1-specific response and suppressed IgE in serum. Collectively, this study shows the potential use of probiotics intervention in reducing the allergic response to whey proteins in mice. © 2015 Society of Chemical Industry.


Subject(s)
Bifidobacterium bifidum/immunology , Cytokines/biosynthesis , Immunoglobulins/blood , Lactobacillus acidophilus/immunology , Probiotics/pharmacology , Animal Feed/microbiology , Animals , Cell Line , Cytokines/immunology , Dietary Supplements , Disease Models, Animal , Food Hypersensitivity/diet therapy , Food Hypersensitivity/prevention & control , Intestines/immunology , Lactococcus lactis/growth & development , Male , Mice , Milk Hypersensitivity/drug therapy , Milk Hypersensitivity/microbiology , RNA, Messenger/analysis , RNA, Messenger/isolation & purification , Spleen/immunology , Whey Proteins/immunology , Whey Proteins/pharmacology
20.
ISME J ; 10(3): 742-50, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26394008

ABSTRACT

Dietary intervention with extensively hydrolyzed casein formula supplemented with Lactobacillus rhamnosus GG (EHCF+LGG) accelerates tolerance acquisition in infants with cow's milk allergy (CMA). We examined whether this effect is attributable, at least in part, to an influence on the gut microbiota. Fecal samples from healthy controls (n=20) and from CMA infants (n=19) before and after treatment with EHCF with (n=12) and without (n=7) supplementation with LGG were compared by 16S rRNA-based operational taxonomic unit clustering and oligotyping. Differential feature selection and generalized linear model fitting revealed that the CMA infants have a diverse gut microbial community structure dominated by Lachnospiraceae (20.5±9.7%) and Ruminococcaceae (16.2±9.1%). Blautia, Roseburia and Coprococcus were significantly enriched following treatment with EHCF and LGG, but only one genus, Oscillospira, was significantly different between infants that became tolerant and those that remained allergic. However, most tolerant infants showed a significant increase in fecal butyrate levels, and those taxa that were significantly enriched in these samples, Blautia and Roseburia, exhibited specific strain-level demarcations between tolerant and allergic infants. Our data suggest that EHCF+LGG promotes tolerance in infants with CMA, in part, by influencing the strain-level bacterial community structure of the infant gut.


Subject(s)
Butyrates/metabolism , Lacticaseibacillus rhamnosus/metabolism , Milk Hypersensitivity/microbiology , Probiotics/administration & dosage , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Bacteria/metabolism , Caseins/immunology , Cattle , Feces/microbiology , Female , Gastrointestinal Microbiome/drug effects , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Humans , Infant , Lacticaseibacillus rhamnosus/growth & development , Male , Milk Hypersensitivity/drug therapy , Milk Hypersensitivity/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL