• No results found

The effects of a synbiotic in infants with atopic dermatitis - Chapter 1: General introduction and outline of the thesis

N/A
N/A
Protected

Academic year: 2021

Share "The effects of a synbiotic in infants with atopic dermatitis - Chapter 1: General introduction and outline of the thesis"

Copied!
13
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

UvA-DARE is a service provided by the library of the University of Amsterdam (https://dare.uva.nl)

The effects of a synbiotic in infants with atopic dermatitis

van der Aa, L.B.

Publication date

2010

Link to publication

Citation for published version (APA):

van der Aa, L. B. (2010). The effects of a synbiotic in infants with atopic dermatitis.

General rights

It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s)

and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an open

content license (like Creative Commons).

Disclaimer/Complaints regulations

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please

let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material

inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter

to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You

will be contacted as soon as possible.

(2)

General introduction and

Outline of the thesis

L.B. van der Aa

H.S.A. Heymans

W.M.C. van Aalderen

A.B. Sprikkelman

Chapter

1

Pediatric Allergy and Immunology 2010 Mar;21(2 Pt 2):e355-67

Adapted from:

Probiotics and prebiotics in atopic dermatitis:

(3)

Ch

ap

ter

1

increase in the prevalence of Th1-mediated autoimmune diseases in the Western world does not

concur with the Th1/Th2 paradigm.

These inconsistencies have lead to the proposal of a revised hygiene hypothesis, which considers

changes in the intestinal colonisation pattern during infancy, also caused by an overly hygienic

lifestyle, as an important reason for the increased allergy prevalence and proposes a lack of activity

of regulatory T cells (Tregs), causing overactive Th2 as well as Th1 responses, as the underlying

mechanism (18-21).

The intestinal microbiota

The human intestines are inhabited by at least 400 different bacterial species, with the greatest

density in the large intestine, where concentrations of 10

11

-10

12

cells/g of luminal contents can

be found (22). Approximately 55% of faecal mass consists of bacteria (23). After birth, bacteria

originating from the mother and the environment start colonizing the infant gut. Factors

influencing the colonization pattern are mode of delivery (vaginal delivery versus caesarean

section), prematurity, hospitalization or antibiotic use after birth, type of feeding (breastfeeding

versus formula feeding) and exposure to older siblings (24).

Although some intestinal bacteria are potential pathogens, the relation between the intestinal

microbiota and the human host is mostly symbiotic. The microbiota has several important

nutritional functions, such as degradation of indigestible dietary carbohydrates, production of

short chain fatty acids (SCFA), amino acid synthesis and vitamin synthesis (25). In addition, the

intestinal flora appears to be crucial for the development of the mucosal and systemic immune

system.

The intestinal microbiota and the immune system

The largest mass of lymphoid tissue of the body is found in the gastrointestinal tract and is called

the gut-associated lymphoid tissue (GALT). The GALT interacts with intestinal bacteria, which are

sampled by dendritic cells and intestinal epithelial cells, through two classes of pattern recognition

receptors, Toll-like receptors (TLR) and nucleotide-binding oligomerization domain (NOD)

molecules (26;27).

The intestinal microbiota appears to be important for the development of the GALT. Studies have

shown that mice without microbiota, i.e. germ-free mice, have an underdeveloped GALT with low

numbers of ab- intestinal intraepithelial lymphocytes (28), hypoplastic Peyer’s patches containing

few germinal centres and greatly reduced numbers of IgA-producing plasma cells and lamina

propria CD4+ T cells (29).

Furthermore, the intestinal microbiota also seems to be involved in oral tolerance induction.

Oral tolerance is the establishment of peripheral tolerance to a specific antigen after ingestion of

that antigen. A pivotal role in the induction and maintenance of peripheral tolerance is played

by Tregs. The importance of the intestinal microbiota in tolerance induction was demonstrated

by Sudo and coworkers, who showed that germ-free mice do not develop oral tolerance after

ingestion of ovalbumin and maintain a Th2 response with IgE production (30). Inoculation with

Introduction

Atopic dermatitis (AD) is a chronic, itching, inflammatory skin disease that often presents in infancy

(1). The prevalence of AD has risen over the past decades, especially in western societies where it

varies in primary school children between 5% and 20% (2). The disease is caused by a combination of

genetic and environmental factors. Severe AD in children is often associated with food allergy (3;4).

The majority of patients have elevated serum IgE levels and peripheral eosinophilia, although in up

to 57% of infants with AD, IgE-sensitization can not (yet) be detected (5).

The disease has a significant impact on children and parents, mainly due to itching, scratching and

sleep disruption (6). The prognosis is reasonable with a recovery rate of 40% at age two (7) and

65% in adolescence (8). However, AD can be the starting point of the ‘allergic march’, the natural

progression of allergic disorders such as asthma and allergic rhinitis. Children with AD have a

chance of approximately 40% to develop asthma (7).

Besides avoiding irritants and moisturizing the skin with emollients, local anti-inflammatory

treatment with topical corticosteroids is the mainstay treatment for infants with AD. In children

with a proven food allergy this is combined with avoidance of the specific allergen. However, flares

occur despite treatment and, although rare, topical corticosteroids can have local side effects, such

as skin atrophy and telangiectasia, and possibly also systemic side effects, such as growth retardation

or osteoporosis. Parents often fear these side effects and this may lead to non-compliance (9).

Innovative prevention and treatment strategies for AD, aiming to manipulate the intestinal flora

with pro-, pre- or synbiotics are now focus of interest. This review provides an overview of the

theoretical basis for using probiotics and prebiotics in AD and presents the current evidence from

randomized controlled trials regarding prevention and treatment of AD in children with pro-,

pre- and synbiotics.

Hygiene hypothesis

Several hypotheses have been proposed to explain the rise in allergic disease, including atopic

dermatitis. The hygiene hypothesis (10) has gained the most attention. This hypothesis was

based on the finding that the prevalence of allergic rhinitis and eczema is inversely related to the

number of older children in a household, which lead to the general hypothesis that early childhood

infections, caused for example by unhygienic contact with older siblings, could prevent the

development of allergic disease (10;11). Later, the T helper 1/ T helper 2 lymfocyte paradigm was

added to the theory. It was argued that a lack of early childhood infections results in a decreased

Th1 response, which disturbs the Th1/Th2 balance and leads to an abundant Th2 response, causing

allergic diseases (12).

However, several facts are not consistent with the hygiene hypothesis as it was first proposed.

Although the protective effect of multiple siblings, as well as other “unhygienic” circumstances like

exposure to farming or day care (13;14), has been a consistent finding, a number of cross-sectional

and prospective studies showed no protective effect of childhood infections (15;16). Moreover,

Th2-dominated helminthic infections are not associated with allergy (17) and the coinciding

(4)

Ch

ap

ter

1

of bifidobacteria. Recently, a Dutch multicenter trial reported an increased mortality risk in

adults with severe acute pancreatitis who received probiotic prophylaxis (44). However, in children

with AD no adverse events are reported.

Immunomodulatory effects of probiotics

Many different effects of probiotics have been described in animal, human and in vitro studies,

most of which are strain-specific. In general, effects include stabilizing of intestinal barrier function

(45;46), stimulation of intestinal IgA-production (47) and, most importantly, modulation of

cytokine production. In vitro studies show that probiotics are able to down-regulate Th2 cytokine

production by stimulation of Th1 cytokines, such as IL-12 and IFN-g (48) or regulatory cytokines,

such as IL-10. The latter is accomplished by either increasing production of these cytokines by

antigen presenting cells or by driving the development of IL-10 or TGF-b producing regulatory T

cells (49-51).

However, ex vivo immunomodulatory effects of probiotics in children with AD are inconclusive

(table 1). There are studies that show up-regulation of IL-10 (52-54) or IFN-g (55;56), although

often without accompanying down-regulation of Th2 cytokines, but several studies do not show

any effect on cytokine production (57-62). Also, one study showed down regulation of the Th2

cytokine IL-5, combined with down-regulation of IL-10 and TGF-b instead of the expected

up-regulation of these cytokines (59). These conflicting outcomes could be caused by the use of

different probiotic strains and different ways of stimulating cytokine production.

Prebiotics

Prebiotics are nondigestible food ingredients that beneficially affect the host by stimulating the

growth and/or activity of a limited number of bacterial species in the colon (63). A food ingredient

must fulfil three criteria to be considered a prebiotic: it should not be hydrolyzed or absorbed

in the upper part of the gastrointestinal tract, it has to be a selective substrate for beneficial

commensal bacteria in the colon, for example bifidobacteria, and it must be able to alter the

intestinal microbiota towards a healthier composition (64).

Breast milk contains natural prebiotics, human milk oligosaccharides, which could explain the

bifidobacteria-dominated microbiota seen in breast fed infants (65). In formula fed infants a

similar effect can be realized with a prebiotic mixture of 90% galacto-oligosaccharides (GOS) and

10% fructo-oligosaccharides (FOS), which stimulates the growth of bifidobacteria (66;67) and, to

a lesser extent, lactobacilli (68).

Prebiotics are generally considered to be safe, as they are naturally present in several kinds of

food. The main side effects of over-consumption in humans are flatulence, bloating and diarrhoea

(69). Increased intestinal tumour formation in mice fed inulin has been reported (70), but on the

opposite, reduction of colon tumours in mice, has also been described (71).

Immunomodulatory effects of prebiotics

Several animal studies demonstrated effects of prebiotics on the immune system. Elevation of the

Bifidobacterium infantis restored susceptibility to oral tolerance induction, but only when this

was done in the neonatal stage (30). Others have shown that germ-free mice have impaired

regulatory T cell function (31) and that colonization of germ-free rats with Lactobacillus

plantarum increases the number of Tregs (32).

Association between microbiota and atopic disease

Studies investigating the composition of the intestinal microbiota in humans have produced

more evidence for the role of commensal bacteria in the development of allergies. Breast fed

infants have a relatively simple microbiota dominated by bifidobacteria, while formula fed

infants have a more diverse flora with higher counts of other anaerobes, e.g. clostridia, and

aerobes (33). The lower prevalence of AD in breast fed infants (34) is possibly linked to this

difference in composition of intestinal microbiota.

Studies comparing flora composition of atopic and non-atopic infants also show significant

differences, which already exist in the first few weeks of life and therefore precede the

development of atopic disease (35-37). Although there are some inconsistencies, generally

these studies show that atopic children have less bifidobacteria and more clostridia than

non-atopic children. However, in a recent multi-centre birth cohort study no significant relation

between colonization pattern in infancy and atopy was found (38). Also, a study comparing the

composition of the intestinal microbiota between children with IgE (against food

allergens)-associated AD and non-IgE-allergens)-associated AD found no significant differences (39). So up to now,

the role of the intestinal microbiota in the development of atopic disease in childhood is still

not clarified.

Probiotics

If the revised hygiene hypothesis holds true, then it is feasible that atopic disease can be treated

or even prevented by manipulating the microbiota. This can be done with probiotics, prebiotics,

or synbiotics.

The term probiotic is derived from the Greek language and means “for life”. Probiotics are

defined as live micro organisms which, when administered in adequate amounts, confer a health

benefit on the host (40). Probiotic agents are preferably isolated from the human gastrointestinal

tract and should be non-pathogenic. They must reach the human intestine alive and be able to

adhere to the epithelial surface; therefore they have to be resistant to gastric acid digestion and

bile salts (41). The bacterial genera most commonly used as probiotics are Lactobacillus and

Bifidobacterium. In addition, yeasts have also been used as probiotics, especially Saccharomyces

boulardii (42).

Probiotics have been safely used in the fermentation of food products for decades. Therefore, the

United States Food and Drug Administration has designated probiotics as Generally Recognized

as Safe (GRAS). However, there have been reports of sepsis linked to probiotic ingestion, all in

patients with underlying medical conditions (43). Sepsis was caused by Lactobacillus species,

(5)

Ch

ap

ter

1

Table 1. Effects of probiotics on cytokine responses in children with AD

Study Probiotic Dose (cfu) Number of subjects Age at inclusion

(mean) Treatment period Cytokines Immunomodulatory effect

Majamaa

1997 (62) LGG

a ATCC 53103 5 x 108 cfu/g

formula 27 (AD and cow’s milk allergy) 2.5-15.7 months (range) 4 weeks PBMCs

f (stimulus conA and cow’s milk protein): no difference in IL-4,

IFN-g and TNF-α between the probiotic and the placebo group No effect

Pessi 2000 (52) LGG ATCC 53103 2dd 1x1010 9 21 months 4 weeks Serum: IL-10 at 8 weeks, no effect on IL-6, IL-12, IFN-g and TNF-a

PBMCs (stimulus anti-CD3, 4 patients): IL-10 and ¯ IL-4, IL-2 and IFN-g at 4 weeks

Up-regulation regulatory cytokine, down regulation Th1 and Th2 cytokines (no placebo group) Rosenfeldt

2003 (61) L.

b rhamnosus

19070-2 and L. reuteri DSM 12246

2dd 1 x 1010 43 5.2 years 6 weeks PBMCs (stimulus LPS-PHA): no significant changes in IL-2, IL-4, IL-10

and IFN-g No effect

Pohjavuori

2004 (55) LGG ATCC 53103 or Mixturea 2dd 5 x 10

9 62, of which 38

IgE-associated AD 1.4-11.5 months (range) 4 weeks PBMCs (stimulus: anti-CD3/anti-CD28): IFN-g in LGG group only in IgE-associated AD. No effect on IL-4, IL-5 and IL-12 and in mix group Up-regulation Th1 cytokine Viljanen 2005

(53) LGG ATCC 53103 or Mixturec 2dd 5 x 10

9

2dd c 121, of which 69 IgE-associated AD 1.4-11.5 months (range) 4 weeks Plasma: IL-10 in mix group, IL-6 in LGG group only in IgE-associated AD. No effect on IL-4, TGF-b and IFN-g Up-regulation regulatory cytokine and proinflammatory cytokine

Prescott 2005

(56) L. fermentum VRI-003 PCC 2dd 1 x 10

9 53 10.9 months 8 weeks PBMCs stimulated with PHAg and SEBh. IFN-g response, stimulated with

heat-killed bacteria: TNF-a response and with ovalbumin: ¯IL-13 response in probiotic group

No effect on IL-6, IL-10 and TGF-b responses

Up-regulation pro-inflammatory cytokines and down regulation TH2 cytokine

Between-group analyses: only significant difference in TNF-a Brouwer 2006

(60) L. rhamnosus or LGG 3 x 10

8

(cfu/g powder) 23 5.2 months 12 weeks PBMCs (stimulatus conA and anti-CD3/anti-CD28): no effect on IL-4, Il-5 and IFN-g No effect

Taylor 2006

(59) L. acidophilus LAVRI-A1 3 x 10

9 118 infants with

allergic mother Prevention study Infants: 6 months PBMCs stimulated with SEB: ¯IL-5 and ¯TGF-b, tetanus toxoid: ¯IL-10, and house dust mite:

¯ frequent IL-10 and TNF-a responses in probiotic group

Other stimuli (PHA, ovalbumin, b-lactoglobulin, house dust mite): no effect on IL-5, IL-6, IL-10, IL-13, IFN-g, TGF-b and TNF-a response

No significant difference in the proportion of circulating CD4+CD25+CTLA4+ T cells or resting FOXp3 expression

Downregulation of Th2, regulatory and proinflammatory cytokines No increase in thymus derived regulatory

T cells Flinterman

2007 (58) Mixture

d 1 x 109 13 (AD and cow’s milk

allergy) 1.7 years 3 months PBMCs unstimulated and stimulated with peanut extract or anti-CD3: ¯IL-10 and ¯IL-6 and TNF-a in probiotic group, but non-significant compared to placebo (p0.063) Non-significant downregulation of proinflammatory and immunosuppressive cytokines Kopp 2007 (57) LGG ATCC 53103 5 x 10

9 68 infants with atopic

family history Prevention study Mothers: 4-6 weeks before

delivery Infants: 6 monthse

CBMCsi and PBMCs stimulated with PHA, LGG or b-lactoglobulin: no

difference in IL-10, IL-13 and IFN-g between the 2 groups No effect

Marschan

2008 (54) Mixture

c c 98 infants with atopic

family history Prevention study Mothers: 2-4 weeks

before delivery Infants: 6 months

Plasma: IL-10 in synbiotic group

Il-2, IL-4, IL-6, IFN-g and TNF-a values below detection limit in both groups Up-regulation of immunosuppressive cytokine

aLactobacillus rhamnosus GG, bLactobacillus, cLGG ATCC 53103 5x109 cfu, L. rhamnosus LC705 5x109

cfu, Bifidobacterium breve Bbi99 2x108 cfu, Propionibacterium freudenreichii ssp shermanii JS 2x109 cfu,

dL. acidophilus, L. casei, Lactococcus lactis, B. infantis, B. lactis and B. longum, ein case of breast feeding mothers took

the probiotics, fperipheral blood mononuclear cells, gphytohemaglutinin mitogen, hStaphylococcus antigen, icord

(6)

Ch

ap

ter

1

In contrast, two prevention studies did not show a reduced AD incidence (87;88). Moreover,

one of these studies found an increased sensitization rate for at least one out of ten food- or

aeroallergens and an increased rate of skin prick test positive (food and/or inhalant allergens)AD

in the probiotic group (87).

There are several possible explanations for these inconclusive results. First, different Lactobacillus

species were used. Probiotic effects are strain specific, therefore not all probiotic strains might

be good candidates for AD prevention. Second, probiotic effects are probably dose-dependent

and there was considerable variation in the daily dosage that was given in these studies. A third

explanation could be differences in study design, such as number of participants, atopic risk of

participants, maternal supplementation during pregnancy, supplementation directly to infants or

via breastfeeding and the supplementation period.

Probiotics and treatment of atopic dermatitis

RCTs exploring the role of probiotics, mainly lactobacilli, in the treatment of AD in children also

have conflicting results (table 3). Four studies had positive outcomes. The first study showed a

significant reduction of SCORAD (SCORing Atopic Dermatitis (89)) score in the probiotic group,

but unfortunately the study had few participants and no between-group analysis was performed

(62). The second study was also small and the included children had relatively mild AD. Although

there was a significant reduction in SCORAD score after two months of probiotic treatment

compared to placebo, this difference disappeared after 6 months of treatment, as SCORAD

score was 0 by that time in all groups (90). The study of Kirjavainen et al (91) was designed to

assess the effect of heat-inactivated probiotics. Although this study was terminated early due to

gastrointestinal adverse events in the heat-inactivated group and therefore had few participants, it

did show that SCORAD score decreased significantly more in the viable probiotic group than in

the placebo group. The fourth study, of Weston et al, had more participants, with more severe AD

(92). A significant reduction of SCORAD score was found in the probiotic group and not in the

placebo group, but this difference was not significant in between-group analysis.

Three other studies showed no effect on AD in general, but did find a significant, although modest,

positive effect on SCORAD score in children with IgE-associated AD (61;93;94). This suggests

that possibly only children who at a young age already have evidence of IgE sensitization (positive

skin prick test and/or elevated total or specific IgE levels) benefit from probiotics. Children in two

of these studies (61;94) had a mean age at inclusion of 4 to 5 years, which was considerably older

than the age at inclusion in other treatment studies. In the third study (93) children were younger

but were only treated for a period of 4 weeks. These factors possibly reduced the beneficial effect

of probiotics.

Finally, three studies didn’t show any effect of probiotics on the severity of AD (60;95;96). In the

study of Brouwer et al (60) groups were small, since three groups were formed (two probiotic

groups and one placebo group) of the 50 children that were included in total. However, the two

other studies (95;96) were well-designed, with more participants, and did not show any effect on

AD or on IgE-associated AD.

total cell number in Peyer’s patches (72), increased natural killer cells and peritoneal macrophage

phagocytic activity (73), increased production of IgA, IL-10 (74), IFN-

g

(75) and TGF-β (76)

have all been described. A human study in which FOS was given to 10 patients with Crohn’s

disease reported an increase in the number of IL-10 positive dendritic cells (77).

There are at least two possible underlying mechanisms that can elicit the immunological effects of

prebiotics. First, most prebiotics stimulate the growth and/or activity of lactic acid bacteria, such as

bifidobacteria or lactobacilli, which have immunomodulatory qualities as described above. Second,

fermentation of prebiotics by lactic acid bacteria enhances SCFA and lactate production (63).

SCFA’s primarily act as energy substrate for colonocytes and several other cells in the human

body (78). The three main SCFA’s are acetate, propionate and butyrate. In vitro, acetate and

propionate are significantly increased after prebiotic fermentation (79). It has been shown that

these two SCFA’s stimulate IFN-γ and IL-10 production (80). Receptors for SCFA’s have been

identified on leukocytes (81), which could explain their immunological effect but more studies

are needed to elucidate this pathway.

Pro-, pre- or synbiotics for prevention and treatment of atopic dermatitis

To identify all randomized controlled trials (RCTs) regarding prevention or treatment of AD in

children with probiotics, prebiotics and combinations of both, i.e. synbiotics, the databases of

Pubmed, Embase and Cochrane up to February 2008 were searched. The following keywords

were used: (probiotics OR prebiotics OR synbiotics) AND (atopic dermatitis OR atopic

eczema OR eczema OR food allergy). Only randomized controlled trials considering children

were included in this review. Reference lists of the found articles were checked for additional

randomized studies.

Probiotics and prevention of atopic dermatitis

Several RCTs have investigated the effect of probiotics on the prevention of AD (table 2). In five

trials the preventive effect on the development of AD in infants was investigated. In these trials

probiotics (different Lactobacillus species and one Bifidobacterium strain) were given to infants

with a high risk of developing allergy, starting immediately after birth. In four of these trials

mothers also received probiotics during the last weeks of pregnancy.

The first study reported a 50% reduction of the incidence of AD in the probiotic group compared

to the placebo group at the age of 2 years (82). This effect was still evident at the age of 4 and 7

years (83;84). No effect on the incidence of other allergic diseases or sensitization was found. In

the study of Abrahamsson and co-workers no reduction of the incidence of AD was found, but

subgroup analysis revealed a significant reduction in IgE-associated AD (85). Again, no effect

on the incidence of other allergic diseases or sensitization was found. Recently, Wickens et al

showed that Lactobacillus rhamnosus, but not Bifidobacterium animalis, significantly reduced

the incidence of AD with almost 50% compared to placebo, without any effect on sensitization

(86).

(7)

Ch

ap

ter

1

Table 2. Prevention of AD with probiotics, prebiotics and synbiotics

Study Probiotic Dose Study design Number of subjects Treatment

period Follow-up period Level of evidencef Effect

Kalliomaki

2001 (82) LGG

a 1x1010 cfu RDBPCTd 132,

mother, father or sibling with atopic disease

Mothers: 2-4 weeks before delivery Infants: 6 monthse

7 years A2 Reduction of AD incidence at age 2 (23% vs. 46%, p 0.008), reduction

still significant at age 4 and 7.

No reduction other allergic diseases or sensitization. Abrahamsson 2007 (85) L. b reuteri (ATCC 55730) 1x10 8 cfu RDBPCT 188, mother, father or sibling with atopic disease

Mothers: week 36 until delivery Infants: 12 months

2 years A2 No reduction of eczema incidence (probiotics: 36%, placebo: 34%). Less

IgE-associated eczema at age 2 (8% vs. 20%, p 0.02). No reduction other allergic diseases or sensitization.

If allergic mother: significantly less IgE-associated AD and less sensitization at age 6-24 months.

Taylor

2007 (87) L. acidophilus (LAVRI-A1) 3x10

9 cfu RDBPCT 178,

atopic mother Infants: 6 months 1 year A2 No prevention of AD (probiotics: 26%, placebo: 23%)Probiotic group: higher sensitization rate (40% vs.24%, p0.03) and more

IgE-associated AD (26% vs.14%, p0.045) at age 12 months Kopp

2008 (88) LGG (ATCC 53103) 1x10

10 cfu RDBPCT 94,

mother, father or sibling with atopic disease

Mothers: 4-6 weeks before delivery

Infants: 6 monthse)

2 years A2 No prevention of AD (probiotics: 28%, placebo: 27%).

No reduction in sensitization to inhalant allergens (food allergens were not determined)

Wickens

2008 (85) L. rhamnosus (HN001) or Bifidobacterium animalis subsp lactis (HN 019)

6x109 cfu

9x109 cfu

RDBPCT 474, parental atopic

disease Mothers: week 35 until baby was 6 months (in case of BF) Infants: 2 years

2 years A2 Reduction of AD (HRh 0.51, 95% CI 0.30-0.85, p0.01) and

IgE-associated AD (HR 0.51, 95% CI 0.27-0.97, p0.04) incidence in Lactobacillus group compared to placebo. Bifidobacterium group: no effect.

Both groups: no effect on sensitization. Moro

2006 (97) Prebiotics 0.8 g/100 ml formula RDBPCT 206,parental atopic

disease

Infants: 6 months 6 months Bg Reduction of AD incidence at age 6 months (10% vs.23%, p0.01),

reduction still significant at age 2.

Less recurrent wheezing and allergic urticaria at age 2 (7.6% vs.20.6% and 1.5% vs.10.3%, p<0.05) Kukkonen 2007 (101) Mixture c + prebiotics (synbiotics) c RDBPCT 925, parental atopic disease Mothers: 2-4 weeks before delivery Infants: 6 months

2 years A2 Reduction of AD (ORi 0.74, 95% CI 0.55-0.98, p0.035) and

IgE-associated AD (OR 0.66, 95% CI, 0.46-0.95, p0.025) incidence at age 2 No reduction other allergic diseases or sensitization

aLactobacillus rhamnosus GG, bLactobacillus, cLGG 5x109 cfu, L. rhamnosus LC705 5x109 cfu, Bifidobacterium breve

Bbi99 2x108 cfu, Propionibacterium JS 2x109 cfu, drandomized, double-blind, placebo-controlled trial, ein case of

breast feeding mothers took the probiotics, fLevel of evidence according to the criteria of the Dutch Institute for

Healthcare Improvement (CBO) : A2=RCT of good quality, B=RCT of less quality, greason: <80% follow-up, hhazard

(8)

Ch

ap

ter

1

Table 3. Treatment of AD with probiotics and synbiotics

Study Probiotic Dose Study design Number

of subjects Age at inclusion (mean) MeanSCORAD at inclusion

Treatment

period Level of evidencef Effect

Majamaa

1997 (62) LGG

a (ATCC 53103) 5 x 108 cfu/g formula RDBPCTe 27 (AD and

suspected cow’s milk allergy)

2.5-15.7 months

(no mean reported) 23.4 4 weeks B

g Significant reduction of SCORAD in probiotic (-11 points)

and not in placebo group (-2 points). No between-group analyses performed, follow-up after 2 months: no differences between the groups.

Isolauri

2000 (90) LGG (ATCC 53103) orB.lactis Bb-12 b 3 x 10

8 cfu/g

1 x 109 cfu/g RDBPCT 27 4.6 months 16 6 months B

g Significant reduction of SCORAD in both probiotic groups

compared to placebo after 2 months, after 6 months no difference between the 3 groups (median SCORAD 0 in all groups).

Kirjavainen

2003 (90) viable LGG (ATCC 53103) or heat-inactivated LGG 1 x 10

9 cfu/g formula RDBPCT 35 (AD and

suspected cow’s milk allergy) 5.5 months 16 mean: 7.5 weeks (range 0.4-45.3)

Bg Significant reduction of SCORAD in viable LGG group

compared to placebo (p 0.02), study was prematurely terminated due to adverse gastrointestinal symptoms in the heat-inactivated LGG group Rosenveldt 2003 (61) L. rhamnosus (19070-2) c and L. reuteri (DSM 12246) 2dd 1 x 10 10 cfu RDBPC

cross-over study 43 5.2 years 35 6 weeks B

h Reduction of SCORAD only in IgE-associated AD (-2.4

points vs +3.2, p 0.04) Weston

2005 (92) L. fermentum (VRI-003 PCC) 2dd 1 x 10

9 cfu RDBPCT 53 10.9 months 42.4 8 weeks A2 Significant reduction of SCORAD in probiotic (-18) and not

in placebo group (-10) at 8 wks.

No significant difference in between-group analyses. Viljanen

2005 (93) LGG (ATCC 53103) orMixtured 2dd 5 x 10

9 cfu

d RDBPCT 230 (AD and suspected

cow’s milk allergy)

6.4 months 32.6 4 weeks A2 LGG: reduction of SCORAD only in IgE-associated AD

(-26 vs -20, p 0.04) Mixture: no effect Sistek

2006 (94) L. rhamnosus and B. lactis 2 x 10

10 cfu/g RDBPCT 59 (AD and

suspected cow’s milk allergy)

4.1 years 30.6 12 weeks A2 Marginal effect in food sensitized children:

SCORAD ratio 0.73 (95% CI 0.54-1.00, P=0.047) Brouwer

2006 (60) L. rhamnosus orLGG 3 x 10

8 cfu/g powder

(5 x 109 cfu/100 ml formula RDBPCT 50 5.2 months 18.7 12 weeks A2 No effect on SCORAD or sensitization in either probiotic group

Folster-Holst

2006 (95) LGG 2dd 5 x 10

9 cfu RDBPCT 47 18.8 months 42.3 8 weeks A2 No effect on SCORAD in AD or in IgE-associated AD

Grüber

2007 (105) LGG 2dd 5 x 10

9 cfu RDBPCT 102 7.4 months 24.1 12 weeks A2 No effect on SCORAD in AD or in IgE-associated AD or on

sensitization Passeron

2006 (102) synbiotics: L. rhamnosus (Lcr35) and prebiotics (placebogroup: only prebiotics)

3dd 1.2 x 109 cfu RDBT 39 5.9 years 39.7 12 weeks A2 No difference in SCORAD between the 2 groups

aLactobacillus rhamnosus GG, bBifidobacterium, cLactobacillus, d2dd LGG 5x109 cfu, L. rhamnosus LC705 5x109 cfu,

B. Bbi99 2x108 cfu, Propionibacterium freudenreichii ssp. shermanii JS 2x109 cfu, erandomized, double-blind,

placebo-controlled trial, flevel of evidence according to the criteria of the Dutch Institute for Healthcare Improvement (CBO):

(9)

Ch

ap

ter

1

Moreover, the number of participants was small and they were school aged children. It needs to be

considered that manipulating the intestinal microbiota probably has more effect in early infancy,

when immune programming is initiated (103). To further explore the role of synbiotics in the

treatment of AD, it is necessary to conduct larger, well-designed, randomized placebo-controlled

trials in a young age group, preferably infants.

Aim of the thesis

The principal aim of this thesis is to investigate the clinical, microbiological and immunological effects

of synbiotics, a combination of the probiotic strain Bifidobacterium breve M-16V and a prebiotic

mixture of 90% short chain galactooligosaccharides and 10% long chain fructooligosaccharides

(Immunofortis®) in infants with atopic dermatitis.

Outline of the thesis

In Chapter 2 of this thesis we present the results of a randomized controlled multi-centre trial that

we performed to evaluate the effects of an infant formula with an added synbiotic, a combination

of Bifidobacterium breve M-16V and a specific mixture of 90% short chain galactooligosaccharides

and 10% long chain fructooligosaccharides (Immunofortis®), on the severity of atopic dermatitis

in infants. Additionally, we investigated the effect of this synbiotic on topical corticosteroid usage,

serum total and specific IgE (against inhalant- and foodallergens), serum eosinophil count and on

the composition and metabolic activity of the intestinal microbiota.

In Chapter 3 we investigate the immunological effects of this synbiotic on plasma levels of IL-5,

IgG1, IgG4 and the AD-associated chemokines cutaneous T cell-attracting chemokine (CTACK)

and thymus and activation-regulated chemokine (TARC), on ex vivo cytokine responses of

peripheral mononuclear blood cells (PBMCs) to non-specific and allergen-specific stimuli, and on

circulating regulatory T cell percentages in these infants.

In Chapter 4, we investigate the differences in atopic markes (eosinophilic granulocytes, IL-5,

IgG1, IgG4, CTACK and TARC), ex vivo cytokine responses of PBMCs, and circulating regulatory

T cell percentages between infants with IgE-associated atopic dermatitis and non IgE-associated

atopic dermatitis.

In Chapter 5, we present the results of a one-year follow-up study of the infants that participated in

the trial. The aim of this follow-up study was to determine the prevalence of asthma-like symptoms,

use of asthma medication and sensitization against aeroallergens in the infants that had received

the synbiotic and the infants that had received placebo.

In Chapter 6 we respond to the paper ‘The impact of maternal atopy and probiotic supplementation

during pregnancy on infant sensitization’ of Huurre and colleagues, published in Clinical and

Experimental Allergy (104).

There are considerable differences in the probiotic strains and dosages that were used in the

treatment studies. Also the number, age and eczema severity of the participants and the treatment

period differs between studies. These differences could possibly explain the conflicting results.

It should also be mentioned that these trials are complicated by the natural tendency of AD to

improve over time, with adequate advice and topical corticosteroid treatment, which explains the

substantial improvement that is often seen in the placebo group.

Prebiotics in the prevention and treatment of atopic dermatitis

One double-blind RCT has been performed to investigate the preventive effect of prebiotics in the

development of AD (97) (table 2). In this study 259 high risk infants were enrolled. They received

a hydrolysed protein formula with either GOS/ FOS mixture or maltodextrine (placebo) for 6

months. The incidence of AD, diagnosed according to clinical criteria, during the study period

was significantly lower in the intervention group than in the placebo group (9.8 % compared to

23.1%). At age 2, the cumulative incidence of AD was still significantly reduced in the prebiotic

group, as were the cumulative incidence of recurrent wheezing and allergic urticaria (98). Faecal

bifidobacteria counts were significantly higher in the intervention group. Although these results

seem promising, a limitation of the study was the relatively large percentage (more than 20%)

of infants that were lost to follow-up during the intervention period. Up until now there are no

studies that explore the role of prebiotics in the treatment of AD.

Synbiotics

Combinations of pro- and prebiotics are called synbiotics. This term should be used to describe

products in which the prebiotic compound selectively stimulates the probiotic compound, thus

creating a synergetic effect (99). Theoretically, optimal synbiotic preparations can be expected to

obtain better results in AD prevention or treatment than either pro- or prebiotics alone. This was

confirmed by an animal study that shows that severity of AD lesions and total immunoglobulin

E levels were significantly reduced in mice fed Lactobacillus casei subsp. casei together with its

prebiotic, dextran, compared to placebo. This effect was also seen in mice that were fed either the

probiotic or the prebiotic compound alone, but to a lesser extent than when the two compounds

were given together (100).

A large human prevention trial showed that a preparation of 4 probiotic strains combined with

prebiotics significantly reduced the incidence of eczema (26% versus 32%, p 0.04) and

IgE-associated eczema (12% versus 18%, p 0.03) in high risk children compared to placebo (101) (table

2).

Only one clinical trial investigated the efficacy of synbiotics as treatment for AD (102) (table 3).

Thirty-nine children, with a mean age of 6 years, were included and randomized to receive either

synbiotics or prebiotics. A significant improvement of AD was found in both study groups, but

synbiotics did not appear to be superior to prebiotics alone. However, the study design, without a

placebo group, makes it impossible to draw conclusions whether synbiotics and prebiotics improve

AD since the improvement in both groups may also be due to the natural course of the disease.

(10)

Ch

ap

ter

1

Reference List

(1) Kay J, Gawkrodger DJ, Mortimer MJ, Jaron AG. The prevalence of childhood atopic eczema in a general population. J Am Acad Dermatol 1994 Jan;30(1):35-9.

(2) Williams H, Robertson C, Stewart A, et al. Worldwide variations in the prevalence of symptoms of ato-pic eczema in the International Study of Asthma and Allergies in Childhood. J Allergy Clin Immunol 1999 Jan;103(1 Pt 1):125-38.

(3) Eigenmann PA, Calza AM. Diagnosis of IgE-mediated food allergy among Swiss children with atopic dermatitis. Pediatr Allergy Immunol 2000 May;11(2):95-100.

(4) Hill DJ, Hosking CS. Food allergy and atopic dermatitis in infancy: an epidemiologic study. Pediatr Al-lergy Immunol 2004 Oct;15(5):421-7.

(5) Park JH, Choi YL, Namkung JH, et al. Characteristics of extrinsic vs. intrinsic atopic dermatitis in in-fancy: correlations with laboratory variables. Br J Dermatol 2006 Oct;155(4):778-83.

(6) Chamlin SL, Frieden IJ, Williams ML, Chren MM. Effects of atopic dermatitis on young American children and their families. Pediatrics 2004 Sep;114(3):607-11.

(7) Gustafsson D, Sjoberg O, Foucard T. Development of allergies and asthma in infants and young child-ren with atopic dermatitis--a prospective follow-up to 7 years of age. Allergy 2000 Mar;55(3):240-5. (8) Williams HC, Strachan DP. The natural history of childhood eczema: observations from the British

1958 birth cohort study. Br J Dermatol 1998 Nov;139(5):834-9.

(9) Charman CR, Morris AD, Williams HC. Topical corticosteroid phobia in patients with atopic eczema. Br J Dermatol 2000 May;142(5):931-6.

(10) Strachan DP. Hay fever, hygiene, and household size. BMJ 1989 Nov 18;299(6710):1259-60.

(11) Bach JF. The effect of infections on susceptibility to autoimmune and allergic diseases. N Engl J Med 2002 Sep 19;347(12):911-20.

(12) Romagnani S. The Th1/Th2 paradigm and allergic disorders. Allergy 1998;53(46 Suppl):12-5.

(13) Riedler J, Braun-Fahrlander C, Eder W, et al. Exposure to farming in early life and development of asthma and allergy: a cross-sectional survey. Lancet 2001 Oct 6;358(9288):1129-33.

(14) Kramer U, Heinrich J, Wjst M, Wichmann HE. Age of entry to day nursery and allergy in later child-hood. Lancet 1999 Feb 6;353(9151):450-4.

(15) Bodner C, Godden D, Seaton A. Family size, childhood infections and atopic diseases. The Aberdeen WHEASE Group. Thorax 1998 Jan;53(1):28-32.

(16) Benn CS, Melbye M, Wohlfahrt J, Bjorksten B, Aaby P. Cohort study of sibling effect, infectious diseases, and risk of atopic dermatitis during first 18 months of life. BMJ 2004 May 22;328(7450):1223. (17) Yazdanbakhsh M, Kremsner PG, van Ree R. Allergy, parasites, and the hygiene hypothesis. Science

2002 Apr 19;296(5567):490-4.

(18) Wold AE. The hygiene hypothesis revised: is the rising frequency of allergy due to changes in the intes-tinal flora? Allergy 1998;53(46 Suppl):20-5.

(19) Rautava S, Ruuskanen O, Ouwehand A, Salminen S, Isolauri E. The hygiene hypothesis of atopic di-sease--an extended version. J Pediatr Gastroenterol Nutr 2004 Apr;38(4):378-88.

(20) Noverr MC, Huffnagle GB. The ‘microflora hypothesis’ of allergic diseases. Clin Exp Allergy 2005 Dec;35(12):1511-20.

Chapter 7 gives an overview of the current European feeding recommendations regarding

breastfeeding, cow’s milk formula, complimentary foods and probiotics/prebiotics, for allergy

prevention in infants. This review was part of the EuroPrevall project , a European multicentre

research project on the prevalence and causes of food allergy in different countries across Europe,

in which the department of Pediatric Respiratory Medicine and Allergy of the Emma Children’s

Hospital AMC participates.

In Chapter 8 the results presented in this thesis are summarized and discussed against the

background of the international literature. Moreover, concluding remarks and suggestions for

further research are made.

(11)

Ch

ap

ter

1

(39) Kendler M, Uter W, Rueffer A, Shimshoni R, Jecht E. Comparison of fecal microflora in children with atopic eczema/dermatitis syndrome according to IgE sensitization to food. Pediatr Allergy Immunol 2006 Mar;17(2):141-7.

(40) FAO/WHO, www.who.int/foodsafety/fs_management/en/probiotic_guidelines.pdf. 2002.

(41) Saarela M, Mogensen G, Fonden R, Matto J, Mattila-Sandholm T. Probiotic bacteria: safety, functional and technological properties. J Biotechnol 2000 Dec 28;84(3):197-215.

(42) Katz JA. Probiotics for the prevention of antibiotic-associated diarrhea and Clostridium difficile diar-rhea. J Clin Gastroenterol 2006 Mar;40(3):249-55.

(43) Boyle RJ, Robins-Browne RM, Tang ML. Probiotic use in clinical practice: what are the risks? Am J Clin Nutr 2006 Jun;83(6):1256-64.

(44) Besselink MG, van Santvoort HC, Buskens E, et al. Probiotic prophylaxis in predicted severe acute pan-creatitis: a randomised, double-blind, placebo-controlled trial. Lancet 2008 Feb 23;371(9613):651-9. (45) Isolauri E, Majamaa H, Arvola T, Rantala I, Virtanen E, Arvilommi H. Lactobacillus casei strain GG

reverses increased intestinal permeability induced by cow milk in suckling rats. Gastroenterology 1993 Dec;105(6):1643-50.

(46) Rosenfeldt V, Benfeldt E, Valerius NH, Paerregaard A, Michaelsen KF. Effect of probiotics on gastroin-testinal symptoms and small ingastroin-testinal permeability in children with atopic dermatitis. J Pediatr 2004 Nov;145(5):612-6.

(47) Park JH, Um JI, Lee BJ, et al. Encapsulated Bifidobacterium bifidum potentiates intestinal IgA produc-tion. Cell Immunol 2002 Sep;219(1):22-7.

(48) Pochard P, Gosset P, Grangette C, et al. Lactic acid bacteria inhibit TH2 cytokine production by mono-nuclear cells from allergic patients. J Allergy Clin Immunol 2002 Oct;110(4):617-23.

(49) Hart AL, Lammers K, Brigidi P, et al. Modulation of human dendritic cell phenotype and function by probiotic bacteria. Gut 2004 Nov;53(11):1602-9.

(50) Smits HH, Engering A, van der Kleij D, et al. Selective probiotic bacteria induce IL-10-producing regu-latory T cells in vitro by modulating dendritic cell function through dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin. J Allergy Clin Immunol 2005 Jun;115(6):1260-7.

(51) Hoarau C, Lagaraine C, Martin L, Velge-Roussel F, Lebranchu Y. Supernatant of Bifidobacterium breve induces dendritic cell maturation, activation, and survival through a Toll-like receptor 2 pathway. J Al-lergy Clin Immunol 2006 Mar;117(3):696-702.

(52) Pessi T, Sutas Y, Hurme M, Isolauri E. Interleukin-10 generation in atopic children following oral Lac-tobacillus rhamnosus GG. Clin Exp Allergy 2000 Dec;30(12):1804-8.

(53) Viljanen M, Pohjavuori E, Haahtela T, et al. Induction of inflammation as a possible mechanism of pro-biotic effect in atopic eczema-dermatitis syndrome. J Allergy Clin Immunol 2005 Jun;115(6):1254-9. (54) Marschan E, Kuitunen M, Kukkonen K, et al. Probiotics in infancy induce protective immune profiles

that are characteristic for chronic low-grade inflammation. Clin Exp Allergy 2008 Apr;38(4):611-8. (55) Pohjavuori E, Viljanen M, Korpela R, et al. Lactobacillus GG effect in increasing IFN-gamma

produc-tion in infants with cow’s milk allergy. J Allergy Clin Immunol 2004 Jul;114(1):131-6.

(56) Prescott SL, Dunstan JA, Hale J, et al. Clinical effects of probiotics are associated with increased interferon-gamma responses in very young children with atopic dermatitis. Clin Exp Allergy 2005 Dec;35(12):1557-64. (21) Rook GA, Adams V, Hunt J, Palmer R, Martinelli R, Brunet LR. Mycobacteria and other

environmen-tal organisms as immunomodulators for immunoregulatory disorders. Springer Semin Immunopathol 2004 Feb;25(3-4):237-55.

(22) Simon GL, Gorbach SL. Intestinal flora in health and disease. Gastroenterology 1984 Jan;86(1):174-93. (23) Stephen AM, Cummings JH. The microbial contribution to human faecal mass. J Med Microbiol 1980

Feb;13(1):45-56.

(24) Penders J, Thijs C, Vink C, et al. Factors influencing the composition of the intestinal microbiota in early infancy. Pediatrics 2006 Aug;118(2):511-21.

(25) Hooper LV, Midtvedt T, Gordon JI. How host-microbial interactions shape the nutrient environment of the mammalian intestine. Annu Rev Nutr 2002;22:283-307.

(26) Kelly D, Conway S, Aminov R. Commensal gut bacteria: mechanisms of immune modulation. Trends Immunol 2005 Jun;26(6):326-33.

(27) Macdonald TT, Monteleone G. Immunity, inflammation, and allergy in the gut. Science 2005 Mar 25;307(5717):1920-5.

(28) Bandeira A, Mota-Santos T, Itohara S, et al. Localization of gamma/delta T cells to the intestinal epithe-lium is independent of normal microbial colonization. J Exp Med 1990 Jul 1;172(1):239-44.

(29) Macpherson AJ, Hunziker L, McCoy K, Lamarre A. IgA responses in the intestinal mucosa against pathogenic and non-pathogenic microorganisms. Microbes Infect 2001 Oct;3(12):1021-35.

(30) Sudo N, Sawamura S, Tanaka K, Aiba Y, Kubo C, Koga Y. The requirement of intestinal bacterial flora for the development of an IgE production system fully susceptible to oral tolerance induction. J Immu-nol 1997 Aug 15;159(4):1739-45.

(31) Ostman S, Rask C, Wold AE, Hultkrantz S, Telemo E. Impaired regulatory T cell function in germ-free mice. Eur J Immunol 2006 Sep;36(9):2336-46.

(32) Herias MV, Hessle C, Telemo E, Midtvedt T, Hanson LA, Wold AE. Immunomodulatory effects of Lactobacillus plantarum colonizing the intestine of gnotobiotic rats. Clin Exp Immunol 1999 May;116(2):283-90.

(33) Stark PL, Lee A. The microbial ecology of the large bowel of breast-fed and formula-fed infants during the first year of life. J Med Microbiol 1982 May;15(2):189-203.

(34) Gdalevich M, Mimouni D, David M, Mimouni M. Breast-feeding and the onset of atopic dermatitis in childhood: a systematic review and meta-analysis of prospective studies. J Am Acad Dermatol 2001 Oct;45(4):520-7.

(35) Kalliomaki M, Kirjavainen P, Eerola E, Kero P, Salminen S, Isolauri E. Distinct patterns of neonatal gut microflora in infants in whom atopy was and was not developing. J Allergy Clin Immunol 2001 Jan;107(1):129-34.

(36) Bjorksten B, Sepp E, Julge K, Voor T, Mikelsaar M. Allergy development and the intestinal microflora during the first year of life. J Allergy Clin Immunol 2001 Oct;108(4):516-20.

(37) Penders J, Thijs C, van den Brandt PA, et al. Gut microbiota composition and development of atopic manifestations in infancy: the KOALA Birth Cohort Study. Gut 2007 May;56(5):661-7.

(38) Adlerberth I, Strachan DP, Matricardi PM, et al. Gut microbiota and development of atopic eczema in 3 European birth cohorts. J Allergy Clin Immunol 2007 Aug;120(2):343-50.

(12)

Ch

ap

ter

1

(57) Kopp MV, Goldstein M, Dietschek A, Sofke J, Heinzmann A, Urbanek R. Lactobacillus GG has in vitro effects on enhanced interleukin-10 and interferon-gamma release of mononuclear cells but no in vivo effects in supplemented mothers and their neonates. Clin Exp Allergy 2007 Apr;38(4):602-10. (58) Flinterman AE, Knol EF, van Ieperen-van Dijk AG, et al. Probiotics have a different

immunomodula-tory potential in vitro versus ex vivo upon oral administration in children with food allergy. Int Arch Allergy Immunol 2007;143(3):237-44.

(59) Taylor AL, Hale J, Wiltschut J, Lehmann H, Dunstan JA, Prescott SL. Effects of probiotic supplementa-tion for the first 6 months of life on allergen- and vaccine-specific immune responses. Clin Exp Allergy 2006 Oct;36(10):1227-35.

(60) Brouwer ML, Wolt-Plompen SA, Dubois AE, et al. No effects of probiotics on atopic dermatitis in in-fancy: a randomized placebo-controlled trial. Clin Exp Allergy 2006 Jul;36(7):899-906.

(61) Rosenfeldt V, Benfeldt E, Nielsen SD, Michaelsen KF, Jeppesen DL, Valerius NH, et al. Effect of probiot-ic Lactobacillus strains in children with atopprobiot-ic dermatitis. J Allergy Clin Immunol 2003 Feb;111(2):389-95.

(62) Majamaa H, Isolauri E. Probiotics: a novel approach in the management of food allergy. J Allergy Clin Immunol 1997 Feb;99(2):179-85.

(63) Gibson GR, Roberfroid MB. Dietary modulation of the human colonic microbiota: introducing the concept of prebiotics. J Nutr 1995 Jun;125(6):1401-12.

(64) Collins MD, Gibson GR. Probiotics, prebiotics, and synbiotics: approaches for modulating the micro-bial ecology of the gut. Am J Clin Nutr 1999 May;69(5):1052S-7S.

(65) Coppa GV, Bruni S, Morelli L, Soldi S, Gabrielli O. The first prebiotics in humans: human milk oligosac-charides. J Clin Gastroenterol 2004 Jul;38(6 Suppl):S80-S83.

(66) Boehm G, Lidestri M, Casetta P, et al. Supplementation of a bovine milk formula with an oligosaccha-ride mixture increases counts of faecal bifidobacteria in preterm infants. Arch Dis Child Fetal Neonatal Ed 2002 May;86(3):F178-F181.

(67) Moro G, Minoli I, Mosca M, et al. Dosage-related bifidogenic effects of galacto- and fructooligosac-charides in formula-fed term infants. J Pediatr Gastroenterol Nutr 2002 Mar;34(3):291-5.

(68) Haarman M, Knol J. Quantitative real-time PCR analysis of fecal Lactobacillus species in infants receiv-ing a prebiotic infant formula. Appl Environ Microbiol 2006 Apr;72(4):2359-65.

(69) Ouwehand AC, Derrien M, de VW, Tiihonen K, Rautonen N. Prebiotics and other microbial substrates for gut functionality. Curr Opin Biotechnol 2005 Apr;16(2):212-7.

(70) Pajari AM, Rajakangas J, Paivarinta E, Kosma VM, Rafter J, Mutanen M. Promotion of intestinal tumor formation by inulin is associated with an accumulation of cytosolic beta-catenin in Min mice. Int J Cancer 2003 Sep 20;106(5):653-60.

(71) Pierre F, Perrin P, Champ M, Bornet F, Meflah K, Menanteau J. Short-chain fructo-oligosaccharides re-duce the occurrence of colon tumors and develop gut-associated lymphoid tissue in Min mice. Cancer Res 1997 Jan 15;57(2):225-8.

(72) Manhart N, Spittler A, Bergmeister H, Mittlbock M, Roth E. Influence of fructooligosaccharides on Peyer’s patch lymphocyte numbers in healthy and endotoxemic mice. Nutrition 2003 Jul;19(7-8):657-60.

(73) Kelly-Quagliana KA, Nelson PD, Buddington RK. Dietary oligofructose and inulin modulate immune functions in mice. Nutr.Res. 23, 257-267. 2003.

(74) Roller M, Rechkemmer G, Watzl B. Prebiotic inulin enriched with oligofructose in combination with the probiotics Lactobacillus rhamnosus and Bifidobacterium lactis modulates intestinal immune func-tions in rats. J Nutr 2004 Jan;134(1):153-6.

(75) Mizubuchi H, Yajima T, Aoi N, Tomita T, Yoshikai Y. Isomalto-oligosaccharides polarize Th1-like res-ponses in intestinal and systemic immunity in mice. J Nutr 2005 Dec;135(12):2857-61.

(76) Hoentjen F, Welling GW, Harmsen HJ, et al. Reduction of colitis by prebiotics in HLA-B27 trans-genic rats is associated with microflora changes and immunomodulation. Inflamm Bowel Dis 2005 Nov;11(11):977-85.

(77) Lindsay JO, Whelan K, Stagg AJ, et al. Clinical, microbiological, and immunological effects of fructo-oligosaccharide in patients with Crohn’s disease. Gut 2006 Mar;55(3):348-55.

(78) Wong JM, de SR, Kendall CW, Emam A, Jenkins DJ. Colonic health: fermentation and short chain fatty acids. J Clin Gastroenterol 2006 Mar;40(3):235-43.

(79) Rycroft CE, Jones MR, Gibson GR, Rastall RA. A comparative in vitro evaluation of the fermentation properties of prebiotic oligosaccharides. J Appl Microbiol 2001 Nov;91(5):878-87.

(80) Cavaglieri CR, Nishiyama A, Fernandes LC, Curi R, Miles EA, Calder PC. Differential effects of short-chain fatty acids on proliferation and production of pro- and anti-inflammatory cytokines by cultured lymphocytes. Life Sci 2003 Aug 15;73(13):1683-90.

(81) Le PE, Loison C, Struyf S, et al. Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J Biol Chem 2003 Jul 11;278(28):25481-9. (82) Kalliomaki M, Salminen S, Arvilommi H, Kero P, Koskinen P, Isolauri E. Probiotics in primary

preven-tion of atopic disease: a randomised placebo-controlled trial. Lancet 2001 Apr 7;357(9262):1076-9. (83) Kalliomaki M, Salminen S, Poussa T, Arvilommi H, Isolauri E. Probiotics and prevention of atopic disease:

4-year follow-up of a randomised placebo-controlled trial. Lancet 2003 May 31;361(9372):1869-71. (84) Kalliomaki M, Salminen S, Poussa T, Isolauri E. Probiotics during the first 7 years of life: a

cumula-tive risk reduction of eczema in a randomized, placebo-controlled trial. J Allergy Clin Immunol 2007 Apr;119(4):1019-21.

(85) Abrahamsson TR, Jakobsson T, Bottcher MF, et al. Probiotics in prevention of IgE-associated eczema: A double-blind, randomized, placebo-controlled trial. J Allergy Clin Immunol 2007 May;119(5):1174-80. (86) Wickens K, Black PN, Stanley TV, et al. A differential effect of 2 probiotics in the prevention of eczema

and atopy: A double-blind, randomized, placebo-controlled trial. J Allergy Clin Immunol 2008 Aug 31. (87) Taylor AL, Dunstan JA, Prescott SL. Probiotic supplementation for the first 6 months of life fails to

reduce the risk of atopic dermatitis and increases the risk of allergen sensitization in high-risk children: a randomized controlled trial. J Allergy Clin Immunol 2007 Jan;119(1):184-91.

(88) Kopp MV, Hennemuth I, Heinzmann A, Urbanek R. Randomized, double-blind, placebo-controlled trial of probiotics for primary prevention: no clinical effects of Lactobacillus GG supplementation. Pe-diatrics 2008 Apr;121(4):e850-e856.

(89) Severity scoring of atopic dermatitis: the SCORAD index. Consensus Report of the European Task Force on Atopic Dermatitis. Dermatology 1993;186(1):23-31.

(13)

Ch

ap

ter

1

(90) Isolauri E, Arvola T, Sutas Y, Moilanen E, Salminen S. Probiotics in the management of atopic eczema. Clin Exp Allergy 2000 Nov;30(11):1604-10.

(91) Kirjavainen PV, Salminen SJ, Isolauri E. Probiotic bacteria in the management of atopic disease: un-derscoring the importance of viability. J Pediatr Gastroenterol Nutr 2003 Feb;36(2):223-7.

(92) Weston S, Halbert A, Richmond P, Prescott SL. Effects of probiotics on atopic dermatitis: a randomised controlled trial. Arch Dis Child 2005 Sep;90(9):892-7.

(93) Viljanen M, Savilahti E, Haahtela T, et al. Probiotics in the treatment of atopic eczema/dermatitis syn-drome in infants: a double-blind placebo-controlled trial. Allergy 2005 Apr;60(4):494-500.

(94) Sistek D, Kelly R, Wickens K, Stanley T, Fitzharris P, Crane J. Is the effect of probiotics on atopic derma-titis confined to food sensitized children? Clin Exp Allergy 2006 May;36(5):629-33.

(95) Folster-Holst R, Muller F, Schnopp N, et al. Prospective, randomized controlled trial on Lactobacillus rhamnosus in infants with moderate to severe atopic dermatitis. Br J Dermatol 2006 Dec;155(6):1256-61.

(96) Gruber C, Wendt M, Sulser C, et al. Randomized, placebo-controlled trial of Lactobacillus rhamnosus GG as treatment of atopic dermatitis in infancy. Allergy 2007 Nov;62(11):1270-6.

(97) Moro G, Arslanoglu S, Stahl B, Jelinek J, Wahn U, Boehm G. A mixture of prebiotic oligosaccharides reduces the incidence of atopic dermatitis during the first six months of age. Arch Dis Child 2006 Oct;91(10):814-9.

(98) Arslanoglu S, Moro GE, Schmitt J, Tandoi L, Rizzardi S, Boehm G. Early dietary intervention with a mixture of prebiotic oligosaccharides reduces the incidence of allergic manifestations and infections during the first two years of life. J Nutr 2008 Jun;138(6):1091-5.

(99) Schrezenmeir J, de VM. Probiotics, prebiotics, and synbiotics--approaching a definition. Am J Clin Nutr 2001 Feb;73(2 Suppl):361S-4S.

(100) Ogawa T, Hashikawa S, Asai Y, Sakamoto H, Yasuda K, Makimura Y. A new synbiotic, Lactobacillus casei subsp. casei together with dextran, reduces murine and human allergic reaction. FEMS Immunol Med Microbiol 2006 Apr;46(3):400-9.

(101) Kukkonen K, Savilahti E, Haahtela T, , et al. Probiotics and prebiotic galacto-oligosaccharides in the prevention of allergic diseases: a randomized, double-blind, placebo-controlled trial. J Allergy Clin Immunol 2007 Jan;119(1):192-8.

(102) Passeron T, Lacour JP, Fontas E, Ortonne JP. Prebiotics and synbiotics: two promising approaches for the treatment of atopic dermatitis in children above 2 years. Allergy 2006 Apr;61(4):431-7.

(103) Prescott SL, Bjorksten B. Probiotics for the prevention or treatment of allergic diseases. J Allergy Clin Immunol 2007 Aug;120(2):255-62.

(104) Huurre A, Laitinen K, Rautava S, Korkeamaki M, Isolauri E. Impact of maternal atopy and probiotic supplementation during pregnancy on infant sensitization: a double-blind placebo-controlled study. Clin Exp Allergy 2008 Aug;38(8):1342-8.

(105) Bieber T, Vick K, Folster-Holst R, et al. Efficacy and safety of methylprednisolone aceponate ointment 0.1% compared to tacrolimus 0.03% in children and adolescents with an acute flare of severe atopic dermatitis. Allergy 2007 Feb;62(2):184-9.

Referenties

GERELATEERDE DOCUMENTEN

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons.. In case of

Based on these case studies, the dissertation offers a somewhat paradoxical answer to the question of the relationship between rupture-talk and web-native

the problem of embodiment, I use the more general description of ‘the computational metaphor’ to describe this conceptual separation of information: in cybercultural rhetoric such

In the following sections I discuss the key concepts in Mondo’s notion of a subversive, computer- driven revolution in culture - cyberpunk, information politics and virtual

Instead, to understand the decision and its significance one has to look at key contextual factors that made the idea that the web would transform media production

9 Rather, this proceeded in an ad-hoc fashion, with features often designed in response to more mundane problems like spam: a more appropriate metaphor than “open source news,”

The overarching logic of exposure - as conventional practice, as generic expectations and formal features, and as the means by which bloggers and readers are captured

Notions of the web’s displacement of mass and mainstream media encountered in the case studies - HotWired’s new publishing paradigm, the Slashdot vision of an