Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Pediatr Allergy Immunol ; 33(6): e13814, 2022 06.
Article in English | MEDLINE | ID: mdl-35754130

ABSTRACT

BACKGROUND: Extensively hydrolyzed formulas are recommended for the dietary management of infants with cow's milk allergy (CMA). OBJECTIVES: Hypoallergenicity, growth, and gastrointestinal (GI) tolerability of a new extensively hydrolyzed whey-protein formula (eHWF) in CMA children were assessed. METHODS: In this prospective, randomized, international, multi-center study (Trial NL3889), 34 children with confirmed CMA (74% IgE-mediated) underwent a double-blind, placebo-controlled food challenge (DBPCFC) with an eHWF developed with non-porcine enzymes, supplemented with prebiotic short-chain galacto- and long-chain fructo-oligosaccharides (0.8 g/L, ratio 9:1), arachidonic acid (0.35/100 g), and docosahexaenoic acid (0.35/100 g). If tolerant to the eHWF, children participated in a 7-day open food challenge with this eHWF. Anthropometrics and GI tolerability were assessed in an optional 16-weeks follow-up. RESULTS: Of the 34 children who started the DBPCFC with the eHWF, 25 subjects (19 boys, mean age: 61 weeks, 18 with IgE-mediated CMA) completed the DBPCFC and 7-day open challenge without major protocol deviations and tested negative at both challenges. One child experienced a late moderate eczematous allergic reaction in the optional follow-up period, indicating the need for close monitoring of subjects starting new formula. Weight and length gain followed the World Health Organization growth curves. Changes in frequency and consistency of stools upon test formula intake were transient. CONCLUSIONS: The newly developed eHWF is a suitable option in CMA treatment as all subjects tolerated the product. This result is in line with the international criteria for hypoallergenicity (American Academy of Pediatrics) that state that more than 90% of CMA children must tolerate the formula. Use of the formula is also associated with normal growth curves and GI tolerability. TRIAL REGISTRATION: Trial NL3889, https://www.trialregister.nl/trial/3889.


Subject(s)
Milk Hypersensitivity , Milk , Animals , Cattle , Child , Female , Humans , Immunoglobulin E , Infant , Infant Formula , Prospective Studies , Whey , Whey Proteins
2.
Clin Exp Allergy ; 48(10): 1345-1353, 2018 10.
Article in English | MEDLINE | ID: mdl-29974988

ABSTRACT

BACKGROUND: Failure to induce oral tolerance may result in food allergy. Hydrolysed cow's milk-based infant formulas are recommended in subjects with a high risk of developing allergic disease. Presentation of T cell epitopes is a prerequisite to generate regulatory T cells that could contribute to oral tolerance. OBJECTIVE: To investigate whether a specific hydrolysed whey-based infant formula contains peptides that function as T cell epitopes to support the development of oral tolerance to whey. METHODS: First, a novel liquid chromatography-mass spectrometry (LC-MS) method was developed to characterize ß-lactoglobulin-derived peptides present in a specific infant formula with a focus on region AA#13-48 of ß-lactoglobulin, which has previously been described to contain T cell epitopes with tolerogenic potential. Second, the formula was subjected to the ProImmune ProPresent® antigen presentation assay and MHC class II binding algorithm to identify relevant HLA-DRB1-restricted peptides. Third, identified peptides were tested on human cow's milk protein-specific T cell lines to determine T cell recognition. RESULTS: Thirteen peptides of minimal 9AAs long that overlap with AA#13-48 of ß-lactoglobulin were identified. Six of them were found across all batches analysed. It was further confirmed that these peptides were processed and presented by human dendritic cells. The identified HLA-DRB1-restricted peptides were correlated to AA#11-30 and AA#23-39 of ß-lactoglobulin. Importantly, the proliferation assay showed that the synthetic peptides were recognized by cow's milk protein-specific T cell lines and induced T cell proliferation. CONCLUSION AND CLINICAL RELEVANCE: This study demonstrates that the tested hydrolysed infant formula contains functional HLA-DRB1-restricted T cell epitopes, which can potentially support the development of oral tolerance to whey.


Subject(s)
Immune Tolerance , Infant Formula , Peptides/immunology , Whey Proteins , Amino Acid Sequence , Animals , Antigen Presentation/immunology , Cattle , Chromatography, Liquid , Epitope Mapping , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , Food Hypersensitivity/genetics , Food Hypersensitivity/immunology , Food Hypersensitivity/prevention & control , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Humans , Hydrolysis , Infant , Infant Formula/adverse effects , Lymphocyte Activation/immunology , Mass Spectrometry , Milk/immunology , Milk Proteins/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Whey Proteins/chemistry , Whey Proteins/immunology
3.
Carcinogenesis ; 34(7): 1628-35, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23455377

ABSTRACT

Red meat consumption is associated with an increased colon cancer risk. Heme, present in red meat, injures the colon surface epithelium by generating cytotoxic and oxidative stress. Recently, we found that this surface injury is compensated by hyperproliferation and hyperplasia of crypt cells, which was induced by a changed surface to crypt signaling. It is unknown whether this changed signaling is caused by cytotoxic stress and/or oxidative stress, as these processes were never studied separately. The aim of this study was to determine the possible differential effects of dietary heme on these luminal stressors and their impact on the colonic mucosa after 2, 4, 7 and 14 days of heme feeding. Mice received a purified, humanized, control diet or the diet supplemented with 0.2 µmol heme/g. Oxidative and cytotoxic stress were measured in fecal water. Proliferation was determined by Ki67-immunohistochemistry and mucosal responses by whole-genome transcriptomics. After heme ingestion, there was an acute increase in reactive oxygen species (ROS) leading to increased levels of lipid peroxidation products. Mucosal gene expression showed an acute antioxidant response, but no change in cell turnover. After day 4, cytotoxicity of the colonic contents was increased and this coincided with differential signaling and hyperproliferation, indicating that cytotoxicity was the causal factor. Simultaneously, several oncogenes were activated, whereas the tumor suppressor p53 was inhibited. In conclusion, luminal cytotoxicity, but not ROS, caused differential surface to crypt signaling resulting in mucosal hyperproliferation and the differential expression of oncogenes and tumor suppressor genes.


Subject(s)
Cell Proliferation , Colon/drug effects , Dietary Supplements , Gene Expression Regulation, Neoplastic , Heme/administration & dosage , Oxidative Stress , Animals , Colon/chemistry , Colon/pathology , Feces/chemistry , Heme/pharmacology , Immunohistochemistry , Intestinal Mucosa/chemistry , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Lipid Peroxidation , Male , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/chemistry , Time Factors , Transcriptome
4.
Gut ; 61(7): 1041-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21948946

ABSTRACT

OBJECTIVE: Colon cancer is a leading cause of cancer deaths in Western countries and is associated with diets high in red meat. Haem, the iron-porphyrin pigment of red meat, induces cytotoxicity of gut contents and damages the colon surface epithelium. Compensatory hyperproliferation leads to epithelial hyperplasia which increases the risk of colon cancer. The aim of this study was to identify molecules signalling from the surface epithelium to the crypt to initiate hyperproliferation upon stress induced by haem. METHODS: C57Bl6/J mice (n=9/group) received a 'westernised' control diet (40 en% fat) with or without 0.5 µmol/g haem for 14 days. Colon mucosa was used to quantify cell proliferation and for microarray transcriptome analysis. Gene expression profiles of surface and crypt cells were compared using laser capture microdissection. Protein levels of potential signalling molecules were quantified. RESULTS: Haem-fed mice showed epithelial hyperproliferation and decreased apoptosis, resulting in hyperplasia. Microarray analysis of the colon mucosa showed 3710 differentially expressed genes (false discovery rate (q) <0.01), with many involved in the cell cycle. Expression levels of haem- and stress-related genes showed that haem affected surface cells but did not directly affect crypt cells. Injured surface cells should therefore signal to crypt cells to induce compensatory hyperproliferation. Haem downregulated the inhibitors of proliferation, Wnt inhibitory factor 1, Indian Hedgehog and bone morphogenetic protein 2. Interleukin-15 was also downregulated. Haem upregulated amphiregulin, epiregulin and cyclo-oxygenase-2 mRNA in surface cells. Their protein/metabolite levels were, however, not increased as haem induced surface-specific inhibition of translation by increasing 4E-BP1. CONCLUSIONS: Haem induces colonic hyperproliferation and hyperplasia by inhibiting the surface to crypt signalling of feedback inhibitors of proliferation.


Subject(s)
Colon/cytology , Epithelial Cells/drug effects , Heme/pharmacology , Intestinal Mucosa/metabolism , Animals , Cell Proliferation/drug effects , Colon/metabolism , Colonic Neoplasms/etiology , Dietary Supplements , Down-Regulation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Feedback, Physiological , Gene Expression , Gene Expression Profiling , Laser Capture Microdissection , Male , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Signal Transduction , Transcriptome
5.
J Immunol ; 185(1): 653-9, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20505143

ABSTRACT

Traditionally, mast cells were regarded as key cells orchestrating type I hypersensitivity responses. However, it is now recognized that mast cells are widely involved in nonallergic (non-IgE) chronic diseases. Also, in inflammatory bowel disease (IBD), a disease not associated with increased IgE concentrations, clear signs of activation of mast cells have been found. In this study, we investigated if Ig-free L chain-induced hypersensitivity-like responses through activation of mast cells could contribute to the pathophysiology of IBD. As a mast cell-dependent model for IBD, mice were skin-sensitized with dinitrofluorobenzene followed by intrarectal application of the hapten. In this murine IBD model, F991 prevented mast cell activation and also abrogated the development of diarrhea, cellular infiltration, and colonic lymphoid follicle hyperplasia. Furthermore, passive immunization with Ag-specific Ig-free L chains (IgLCs) and subsequent rectal hapten challenge elicited local mast cell activation and increased vascular permeability in the colon of mice. Clinical support is provided by the observation that serum concentrations of IgLCs of patients suffering from Crohn's disease are greatly increased. Moreover, increased presence of IgLCs was evident in tissue specimens from colon and ileum tissue of patients with IBD. Our data suggest that IgLCs may play a role in the pathogenesis of IBD, which provides novel therapeutic means to prevent or ameliorate the adverse gastrointestinal manifestations of IBD.


Subject(s)
Colitis/immunology , Colitis/metabolism , Immunoglobulin kappa-Chains/physiology , Immunoglobulin lambda-Chains/physiology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Mast Cells/immunology , Mast Cells/metabolism , Adult , Animals , Colitis/pathology , Crohn Disease/immunology , Crohn Disease/metabolism , Crohn Disease/pathology , Disease Models, Animal , Female , Humans , Immunization, Passive , Immunoglobulin kappa-Chains/biosynthesis , Immunoglobulin kappa-Chains/blood , Immunoglobulin lambda-Chains/biosynthesis , Immunoglobulin lambda-Chains/blood , Inflammatory Bowel Diseases/pathology , Male , Mast Cells/pathology , Mice , Mice, Inbred BALB C , Middle Aged , Up-Regulation/immunology , Young Adult
6.
Br J Nutr ; 104(12): 1780-6, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20691137

ABSTRACT

An increased intestinal permeability is associated with several diseases. Nutrition can influence gut permeability. Previously, we showed that dietary Ca decreases whereas dietary short-chain fructo-oligosaccharides (scFOS) increase intestinal permeability in rats. However, it is unknown how and where in the gastrointestinal tract Ca and scFOS exert their effects. Rats were fed a Western low-Ca control diet, or a similar diet supplemented with either Ca or scFOS. Lactulose plus mannitol and Cr-EDTA were added to the diets to quantify small and total gastrointestinal permeability, respectively. Additionally, colonic tissue was mounted in Ussing chambers and exposed to faecal water of these rats. Dietary Ca immediately decreased urinary Cr-EDTA excretion by 24 % in Ca-fed rats compared with control rats. Dietary scFOS increased total Cr-EDTA permeability gradually with time, likely reflecting relatively slow gut microbiota adaptations, which finally resulted in a 30 % increase. The lactulose:mannitol ratio was 15 % higher for Ca-fed rats and 16 % lower for scFOS-fed rats compared with control rats. However, no dietary effect was present on individual urinary lactulose and mannitol excretion. The faecal waters did not influence colonic permeability in Ussing chambers. In conclusion, despite effects on the lactulose:mannitol ratio, individual lactulose values did not alter, indicating that diet did not influence small-intestinal permeability. Therefore, both nutrients affect permeability only in the colon: Ca decreases, while scFOS increase colonic permeability. As faecal water did not influence permeability in Ussing chambers, probably modulation of mucins and/or microbiota is important for the in vivo effects of dietary Ca and scFOS.


Subject(s)
Calcium, Dietary/pharmacology , Colon/drug effects , Oligosaccharides/chemistry , Oligosaccharides/pharmacology , Animals , Colon/metabolism , Diet , Feces/chemistry , Male , Permeability/drug effects , Rats , Rats, Wistar , Water/analysis
7.
Nutrients ; 12(7)2020 Jul 13.
Article in English | MEDLINE | ID: mdl-32668666

ABSTRACT

The current study evaluates the safety and tolerance of a partially hydrolyzed whey protein-based infant formula (PHF) versus an in intact cow's milk protein formula (IPF). Breastfed infants were included as a reference group. In a multi-country, multicenter, randomized, double-blinded, controlled clinical trial, infants whose mothers intended to fully formula feed were randomized to PHF (n = 134) or IPF (n = 134) from ≤14 days to 17 weeks of age. The equivalence analysis of weight gain per day within margins of +/-3 g/d (primary outcome), the recorded adverse events, growth and gastro-intestinal tolerance parameters were considered for the safety evaluation. Equivalence of weight gain per day from enrolment until 17 weeks of age was demonstrated in the PHF group compared to the IPF group (difference in means -1.2 g/d; 90% CI (-2.42; 0.02)), with estimated means (SE) of 30.2 (0.5) g/d and 31.4 (0.5) g/d, respectively. No significant differences in growth outcomes, the number, severity or type of (serious) adverse events and tolerance outcomes, were observed between the two formula groups. A partially hydrolyzed whey protein-based infant formula supports adequate infant growth, with a daily weight gain equivalent to a standard intact protein-based formula; it is also safe for use and well-tolerated in healthy term infants.


Subject(s)
Child Development/physiology , Infant Formula , Infant Nutritional Physiological Phenomena/physiology , Protein Hydrolysates/administration & dosage , Whey Proteins/administration & dosage , Animals , Breast Feeding , Double-Blind Method , Female , Healthy Volunteers , Humans , Infant , Infant, Newborn , Male , Milk , Milk Proteins , Safety , Weight Gain
8.
Pharmacol Ther ; 116(2): 207-35, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17719089

ABSTRACT

Mast cells are well known as versatile cells capable of releasing and producing a variety of inflammatory mediators upon activation and are often found in close proximity of neurons. In addition, inflammation leads to local activation of neurons resulting in the release neuropeptides, which also play an important immune modulatory role by stimulation of immune cells. In intestinal disorders like inflammatory bowel disease (IBD) and irritable bowel syndrome (IBS), the number of mast cells is known to be much higher than in the normal intestine. Moreover, both these disorders are also reported to be associated with alterations in neuropeptide content and in neural innervation. Mutual association between mast cells and enteric nerves has been demonstrated to be increased in pathophysiological conditions and contribute to spreading and amplification of the response in IBD and IBS. In this review the focus lies on studies appointed to the direct interaction between mast cells and nerves in IBD, IBS, and animal models for these disorders so far.


Subject(s)
Inflammatory Bowel Diseases/physiopathology , Irritable Bowel Syndrome/physiopathology , Mast Cells/immunology , Animals , Disease Models, Animal , Enteric Nervous System/metabolism , Enteric Nervous System/pathology , Humans , Inflammation Mediators/metabolism , Inflammatory Bowel Diseases/immunology , Irritable Bowel Syndrome/immunology , Mast Cells/metabolism , Neurons/metabolism , Neurons/pathology
9.
Eur J Pharmacol ; 585(2-3): 361-74, 2008 May 13.
Article in English | MEDLINE | ID: mdl-18417115

ABSTRACT

The gastro-intestinal tract is well known for its largest neural network outside the central nervous system and for the most extensive immune system in the body. Research in neurogastroenterology implicates the involvement of both enteric nervous system and immune system in symptoms of inflammatory bowel disease and irritable bowel syndrome. Since both disorders are associated with increased immune cell numbers, nerve growth and activation of both immune cells and nerves, we focus in this review on the involvement of immune cell-nerve interactions in inflammatory bowel disease and irritable bowel syndrome. Firstly, the possible effects of enteric nerves, especially of the nonadrenergic and noncholinergic nerves, on the intestinal immune system and their possible role in the pathogenesis of chronic intestinal inflammatory diseases are described. Secondly, the possible effects of immunological factors, from the innate (chemokines and Toll-like receptors) as well as the adaptive (cytokines and immunoglobulins) immune system, on gastro-intestinal nerves and its potential role in the development of inflammatory bowel disease and irritable bowel syndrome are reviewed. Investigations of receptor-mediated and intracellular signal pathways in neuro-immune interactions might help to develop more effective therapeutic approaches for chronic inflammatory intestinal diseases.


Subject(s)
Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/immunology , Irritable Bowel Syndrome/drug therapy , Irritable Bowel Syndrome/immunology , Animals , Autonomic Nervous System/physiopathology , Chemokines/physiology , Cytokines/physiology , Enteric Nervous System/physiopathology , Humans , Immunoglobulins/physiology , Inflammatory Bowel Diseases/physiopathology , Irritable Bowel Syndrome/physiopathology , Neuroimmunomodulation/physiology , Toll-Like Receptors/drug effects , Toll-Like Receptors/physiology
10.
Eur J Pharmacol ; 548(1-3): 150-7, 2006 Oct 24.
Article in English | MEDLINE | ID: mdl-16949572

ABSTRACT

The gastro-intestinal tract is highly innervated by both intrinsic and extrinsic sensory nerves and this neuronal component is thought to play a role in local inflammatory responses. This in vivo study was designed to determine the function of substance P and the tachykinin NK1 receptor in the pathogenesis of inflammatory bowel disease by the use of the specific antagonist RP 67580. The dinitrofluorobenzene (DNFB)-induced colonic hypersensitivity model is associated with increased levels of substance P in the colon. The tachykinin NK1 receptor antagonist RP 67580 was used to investigate the role of substance P on the development of diarrhea, mast cell infiltration and activation, colonic tissue damage, hypertrophy of colonic lymphoid structures and leukocyte infiltration. The formation of watery diarrhea could completely be abrogated by treatment with RP 67580 in DNFB-sensitized animals 72 h after challenge. Antagonizing the tachykinin NK1 receptor in these animals also resulted in significantly reduced colonic patch hypertrophy, leukocyte recruitment and tissue damage. Total levels of substance P in the colon of DNFB-sensitized mice treated with the inactive enantiomer of the tachykinin NK1 receptor antagonist were significantly higher compared to DNFB-sensitized mice treated with RP 67580 72 h after challenge. Although RP 67580 was capable of reducing the total number of mast cells present in the colon, mast cell activation was not affected by this treatment. In conclusion, in this chemically-induced immunological model for inflammatory bowel disease we demonstrated an important role for tachykinin NK1 receptors, and its ligand substance P, in the development of colitis downstream from mast cell activation.


Subject(s)
Colitis/chemically induced , Indoles/pharmacology , Neurokinin-1 Receptor Antagonists , Substance P/analysis , Animals , Capillary Permeability/drug effects , Colitis/immunology , Colitis/metabolism , Colitis/pathology , Colon/immunology , Colon/metabolism , Colon/pathology , Diarrhea/chemically induced , Dinitrofluorobenzene , Disease Models, Animal , Haptens , Hypersensitivity/immunology , Hypersensitivity/metabolism , Hypersensitivity/pathology , Isoindoles , Male , Mast Cells/immunology , Mice , Mice, Inbred BALB C , Sulfonic Acids
11.
Am J Clin Nutr ; 98(2): 572S-7S, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23824724

ABSTRACT

The WHO refers to human milk as the nutritional gold standard for term infants. Human milk contains many immunomodulatory compounds, including oligosaccharides. Human-milk oligosaccharides can serve as prebiotics because the nondigestible oligosaccharides present in human milk show a clear bifidogenic effect on the gut microbiota. Dietary oligosaccharide structures that have prebiotic effects similar to human-milk oligosaccharides include galacto-oligosaccharides, fructo-oligosaccharides, and pectin-derived acidic oligosaccharides. Both animal studies and human clinical trials showed that dietary intervention with these dietary oligosaccharides in early life could lead to the prevention of atopic dermatitis, food allergy, and allergic asthma. The immune-modulating effects of these oligosaccharides are likely assisted via alteration of the intestinal microbiota or in a microbiota-independent manner by direct interaction on immune cells or both. In this review, an overview of the prebiotic role of dietary oligosaccharides on the microbiota and the microbiota-independent immune modulation by these prebiotics is provided. In addition, recent publications that report on the pathways by which the oligosaccharides might exert their direct immunomodulatory effect are summarized.


Subject(s)
Immunologic Factors/administration & dosage , Oligosaccharides/administration & dosage , Probiotics/administration & dosage , Trisaccharides/administration & dosage , Animals , Asthma/prevention & control , Clinical Trials as Topic , Dermatitis, Atopic/prevention & control , Diet , Food Hypersensitivity/prevention & control , Gastrointestinal Tract/microbiology , Humans , Infant , Metagenome , Milk, Human/chemistry , Models, Animal , Prebiotics/analysis , Prebiotics/microbiology
12.
J Neuroimmunol ; 264(1-2): 14-23, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24051200

ABSTRACT

Plasma B cells secrete immunoglobulinfree light chains (IgLC) which by binding to mast cells can mediate hypersensitivity responses and are involved in several immunological disorders. To investigate the effects of antigen-specific IgLC activation, intracellular recordings were made from cultured murine dorsal root ganglion (DRG) neurons, which can specifically bind IgLC. The neurons were sensitized with IgLC for 90min and subsequently activated by application of the corresponding antigen (DNP-HSA). Antigen application induced a decrease in the rate of rise of the action potentials of non-nociceptive neurons (MANOVA, p=2.10(-6)), without affecting the resting membrane potential or firing threshold. The action potentials of the nociceptive neurons (p=0.57) and the electrical excitability of both types of neurons (p>0.35) were not affected. We conclude that IgLC can mediate antigen-specific responses by reducing the rate of rise of action potentials in non-nociceptive murine DRG neurons. We suggest that antigen-specific activation of IgLC-sensitized non-nociceptive DRG neurons may contribute to immunological hypersensitivity responses and neuroinflammation.


Subject(s)
Action Potentials/drug effects , Ganglia, Spinal/cytology , Immunoglobulin Light Chains/pharmacology , Neurons/drug effects , Analysis of Variance , Animals , Antigens/metabolism , Biophysical Phenomena/drug effects , Biophysics , Cells, Cultured , Electric Stimulation , Male , Mice , Mice, Inbred BALB C , Neurons/classification , Neurons/physiology , Time Factors
13.
Curr Pharm Des ; 18(16): 2369-74, 2012.
Article in English | MEDLINE | ID: mdl-22390700

ABSTRACT

The so-called hygiene hypothesis is, at least in part, accountable for the increase in allergic diseases in the developed countries. Although there is support for one of its primary predictions that host-microbe interactions in early life have longterm effects on the development of disease across populations, the theory has already proven to be imperfect as many more recent increases in certain diseases cannot be explained by the hygiene hypothesis. Nevertheless, many research groups are interested in the host-microbe interactions and are exploring the use of "live micro-organisms which, when administered in adequate amounts, confer a health benefit to the host" (probiotics) and "selectively fermented ingredients that result in specific changes, in the composition and/or activity of the gastrointestinal microbiota, thus conferring benefit(s) upon host health" (prebiotics) to reduce the allergic disease onset or clinical outcomes. As the definitions of pre- and probiotics by itself were already adapted after their original dictation, it is not surprising that producing generalistic conclusions on the effectiveness of pre-, pro and synbiotic intervention in allergic diseases is very challenging as large differences exist in used species, methodologies, prebiotic(s) (mixtures) and probiotic strains. In this review we elucidate on the hurdles in describing prebiotics, probiotics and the combination being synbiotics in allergic manifestations.


Subject(s)
Hypersensitivity/therapy , Probiotics/therapeutic use , Humans
14.
PLoS One ; 7(12): e49868, 2012.
Article in English | MEDLINE | ID: mdl-23239972

ABSTRACT

Colon cancer is a major cause of cancer deaths in Western countries and is associated with diets high in red meat. Heme, the iron-porphyrin pigment of red meat, induces cytotoxicity of gut contents which injures surface cells leading to compensatory hyperproliferation of crypt cells. This hyperproliferation results in epithelial hyperplasia which increases the risk of colon cancer. In humans, a high red-meat diet increases Bacteroides spp in feces. Therefore, we simultaneously investigated the effects of dietary heme on colonic microbiota and on the host mucosa of mice. Whole genome microarrays showed that heme injured the colonic surface epithelium and induced hyperproliferation by changing the surface to crypt signaling. Using 16S rRNA phylogenetic microarrays, we investigated whether bacteria play a role in this changed signaling. Heme increased Bacteroidetes and decreased Firmicutes in colonic contents. This shift was most likely caused by a selective susceptibility of gram-positive bacteria to heme cytotoxic fecal water, which is not observed for gram-negative bacteria, allowing expansion of the gram-negative community. The increased amount of gram-negative bacteria most probably increased LPS exposure to colonocytes, however, there is no appreciable immune response detected in the heme-fed mice. There was no functional change in the sensing of the bacteria by the mucosa, as changes in inflammation pathways and Toll-like receptor signaling were not detected. This unaltered host-microbe cross-talk indicates that the changes in microbiota did not play a causal role in the observed hyperproliferation and hyperplasia.


Subject(s)
Colon/microbiology , Diet , Heme/administration & dosage , Metagenome/genetics , Animals , Bacteroidetes/genetics , Feces/microbiology , Host-Pathogen Interactions , Mice , Mucous Membrane/microbiology , Philology , RNA, Ribosomal, 16S/genetics
15.
Eur J Pharmacol ; 668 Suppl 1: S117-23, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21810418

ABSTRACT

Breast feeding is considered as the best nutrition for growth and development of an infant. Human milk consists of a unique combination of nutritional components each with different characteristics. Oligosaccharides or non-digestible carbohydrates as one of these components, are generally accepted to have a beneficial effect by selectively stimulating the growth and/or activity of one or a limited number of bacterial species. Recently more evidence is rising for direct effects of oligosaccharides on the immune system. Oligosaccharides often used as dietary supplements for their beneficial effects on the host and its immune system, are derived from nutritional sources. In this review we aim to summarize the pharmaceutical properties of these food-borne oligosaccharides early in life.


Subject(s)
Dietary Carbohydrates/pharmacology , Oligosaccharides/pharmacology , Animals , Breast Feeding , Humans , Immune System Diseases/diet therapy , Immune System Diseases/drug therapy , Immune System Diseases/prevention & control , Milk, Human/chemistry , Synbiotics
16.
J Neuroimmunol ; 208(1-2): 80-6, 2009 Mar 31.
Article in English | MEDLINE | ID: mdl-19232443

ABSTRACT

Immunoglobulin-free light chains (IgLC) secreted by B lymphocytes, have been shown to mediate hypersensitivity by inducing antigen-specific mast cell activation. Although both mast cells and sensory neurons contribute to the hypersensitivity response, the role of IgLC in relation to sensory neurons is unknown. We therefore aimed to investigate the effects of IgLC on cultures of murine dorsal root ganglion (DRG) neurons. Immunohistochemistry demonstrated that IgLC and IgE could specifically bind to DRG neurons, on which the presence of FcepsilonRI, the specific receptor for IgE, was demonstrated by western blotting. Further, optical recordings with Fluo-4 showed that application of the corresponding antigen to IgLC- or IgE-sensitized DRG neurons induces a sustained increase in intracellular Ca(2+) in about half of these neurons. These results show that IgLC and IgE can mediate antigen-specific responses in murine neurons. Our findings present a novel way of antigen-specific neuronal activation.


Subject(s)
Epitopes/immunology , Ganglia, Spinal/cytology , Ganglia, Spinal/immunology , Immunoglobulin Light Chains/physiology , Sensory Receptor Cells/immunology , Sensory Receptor Cells/metabolism , Animals , Cells, Cultured , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Hypersensitivity/immunology , Hypersensitivity/metabolism , Hypersensitivity/pathology , Immunoglobulin E/metabolism , Immunoglobulin Light Chains/metabolism , Male , Mice , Mice, Inbred BALB C , Sensory Receptor Cells/pathology
17.
Am J Physiol Gastrointest Liver Physiol ; 291(5): G969-76, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16798721

ABSTRACT

Inflammatory bowel disease (IBD) describes chronic inflammatory conditions of the gastrointestinal tract, and TNF-alpha plays a pivotal role in mediating the response. The proinflammatory cytokine TNF-alpha is rapidly released by mast cells after degranulation. In the present study, we hypothesized TNF-alpha to be an important player in our recently described mast cell-dependent murine model for IBD. The effect of neutralizing anti-TNF-alpha MAb was studied on colonic hypersensitivity in mice induced by a skin application of dinitrofluorobenzene (DNFB) followed by an intrarectal challenge with dinitrobenzene sulfonic acid. Features of the colonic hypersensitivity response included diarrhea, mast cell infiltration and activation, infiltration of inflammatory cells in the colon, colonic patch hypertrophy, and increased mast cell-derived TNF-alpha levels in the colon. Anti-TNF-alpha MAb could effectively abrogate diarrhea in DNFB-sensitized mice 72 h after the challenge. The numbers of colonic patches and total tissue damage scores were reduced by anti-TNF-alpha MAb treatment in DNFB-sensitized mice 72 h after the challenge. Mast cell infiltration and activation remained unaffected by neutralizing anti-TNF-alpha MAb. Treatment with the corticosteroid dexamethasone, a frequently used therapeutic treatment in IBD, resulted in a reduction of diarrhea, cellular infiltration, and total tissue damage scores to the same extent as anti-TNF-alpha MAb. Additionally, dexamethasone treatment could also reduce total TNF-alpha levels in the colon, mast cell numbers, and mast cell activation in both vehicle- and DNFB-sensitized mice 72 h after the challenge. These findings suggest that TNF-alpha can play an instrumental role in causing inflammatory responses in the present murine model for IBD downstream from mast cell activation.


Subject(s)
Colitis/pathology , Mast Cells/pathology , Tumor Necrosis Factor-alpha/physiology , Animals , Anti-Inflammatory Agents/therapeutic use , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Colitis/chemically induced , Colon/drug effects , Colon/metabolism , Dexamethasone/therapeutic use , Dinitrofluorobenzene , Immunohistochemistry , Male , Mast Cells/physiology , Mice , Mice, Inbred BALB C , Neutrophil Infiltration/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
18.
J Immunol ; 176(7): 4375-84, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16547276

ABSTRACT

The immunological mechanisms underlying the role of mast cells in the pathogenesis of inflammatory bowel disease (IBD) are poorly defined. In this study, non-IgE mediated colonic hypersensitivity responses in BALB/c mice induced by skin sensitization with dinitrofluorobenzene (DNFB) followed by an intrarectal challenge with dinitrobenzene sulfonic acid featured as a model to study the role of mast cells in the development of IBD. Vehicle- or DNFB-sensitized mice were monitored for clinical symptoms and inflammation 72 h after dinitrobenzene sulfonic acid challenge. DNFB-sensitized mice developed diarrheic stool, increased colonic vascular permeability, hypertrophy of colonic lymphoid follicles (colonic patches), and showed cellular infiltration at the microscopic level. Increased numbers of mast cells were found in the colon of DNFB-sensitized mice located in and around colonic patches associated with elevated levels of mouse mast cell protease-1 in plasma indicating mast cell activation. Colonic patches of DNFB mice, stimulated in vitro with stem cell factor indicated that an increase in TNF-alpha levels in the colon is mainly mast cell originated. Finally, neutrophil infiltration was observed in the colon of DNFB-sensitized mice. Induction of this model in mast cell-deficient WBB6F(1) W/W(v) mice shows a profound reduction of characteristics of the colonic hypersensitivity reaction. Reconstitution with bone marrow-derived mast cells in WBB6F(1) W/W(v) mice fully restored the inflammatory response. This study demonstrates the importance of mast cells in the development of clinical symptoms and inflammation in the presented murine model for IBD.


Subject(s)
Colitis/chemically induced , Colitis/pathology , Dinitrobenzenes/toxicity , Hypersensitivity/immunology , Hypersensitivity/pathology , Mast Cells/immunology , Animals , Colitis/immunology , Colitis/metabolism , Diarrhea/chemically induced , Hypertrophy , Intestinal Absorption , Lymph Nodes/pathology , Male , Mast Cells/cytology , Mice , Mice, Inbred BALB C , Tumor Necrosis Factor-alpha/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL