Journal List > Immune Netw > v.12(4) > 1033336

Hwang, Im, and Im: Immune Disorders and Its Correlation with Gut Microbiome

Abstract

Allergic disorders such as atopic dermatitis and asthma are common hyper-immune disorders in industrialized countries. Along with genetic association, environmental factors and gut microbiota have been suggested as major triggering factors for the development of atopic dermatitis. Numerous studies support the association of hygiene hypothesis in allergic immune disorders that a lack of early childhood exposure to diverse microorganism increases susceptibility to allergic diseases. Among the symbiotic microorganisms (e.g. gut flora or probiotics), probiotics confer health benefits through multiple action mechanisms including modification of immune response in gut associated lymphoid tissue (GALT). Although many human clinical trials and mouse studies demonstrated the beneficial effects of probiotics in diverse immune disorders, this effect is strain specific and needs to apply specific probiotics for specific allergic diseases. Herein, we briefly review the diverse functions and regulation mechanisms of probiotics in diverse disorders.

Abbreviations

GALT

Gut-Associated Lymphoid Tissue

GI

Gastrointestinal

MLN

Messenteric Lymph Node

AD

Atopic Dermatitis

HMP

Human Microbiome Project

IBD

Inflammatory Bowel Disease

T1D

Type 1 Diabetes

DC

Dendritic Cell

NOD

Nucleotide-binding Oligomerization Domain

TLT

Toll-Like Rceptor

Treg

Regulatory T Cells

iDO

Indoleamine 2,3-dioxygenase

SCORD

SCORing Atopic Dermatitis

INTRODUCTION

"Probiotics" are defined as live microorganisms that, when administered in adequate amounts, confer a health benefit on the host (1). Although the beneficial effect of probiotics administration was mainly known to improve the balance of intestinal microflora, recent studies showed that probiotics can also exert beneficial effects by directly modulating immune system. For example, an oral treatment of certain probiotics or a mixture of them prevented and improved symptoms of experimental inflammatory bowel disease, atopic dermatitis and asthma by down-regulating inflammatory cytokines or inducing immune regulatory mechanisms. However, these beneficial effects are strain-specific (2-4), suggesting the necessity of development of screening system to identify specific probiotics to treat specific disorders.
Atopic dermatitis (AD) is chronic, relapsing inflammatory skin disorder and a complex immune response mediated by genetic, environment factors and skin barrier dysfunction. Recently, the incidence of atopic dermatitis has remarkably increased especially in the industrial cities compared to rural areas and developing countries, which strongly support the "hygiene hypothesis" in the development of allergic immune disorders (5,6). Hygiene hypothesis has received worldwide attention from many scientists as the subject remains questioned for many years. Hygiene hypothesis states that a lack of early childhood exposure to infectious agents, symbiotic microorganisms (e.g. gut flora or probiotics), and parasites increases susceptibility to allergic diseases by suppressing natural development of the immune system (7). Even though treatment with corticosteroid is the most well used method to treat atopic dermatitis, long term usage of steroid can cause many side effects including the atrophoderma and growth retardation (8). So many studies were focused on the development of innovative prevention and treatment strategies for AD to replace the steroid treatment. Interestingly, recent studies also suggest the association of dysregulation of gut microbiota with diverse diseases including atopic dermatitis (9). Therefore manipulation of the intestinal flora with good probiotics will be complementary and alternative treatments to divert immune response from the hyper-immune states. In this review, we summarized the hygiene hypothesis and its correlation with gut microbiome, especially on the aspect of probiotics, in the development and regulation mechanisms of diverse disorders.

Microbiome and its relevance to the susceptibility of diverse diseases

Several hypotheses have been proposed as the cause of development in allergic disease, including atopic dermatitis. Among them, "hygiene hypothesis" has gained the most attentions (7). This hypothesis explains that early childhood infection caused by unhygienic contact with older siblings could prevent the development of allergic disease (10). Later, Th1/Th2 paradigm was added that a lack of early childhood infection results a decreased Th1 immune response, which disturbs the Th1/Th2 balance and leads to an abundant Th2 response, causing allergic disease (11). Later this hypothesis was further revised, which considers changes in the intestinal colonization pattern during infancy and overlay hygienic lifestyle as the most important reasons for the increased allergy prevalence. Especially overlay hygiene can induce a lack of activity of regulatory T cells, causing over-activation of Th1 as well as Th2 responses as the underlying mechanism of allergic disease (12,13).
As explained by hygiene hypothesis, a person's life span is largely affected by the distribution of intestinal bacteria. The term "microbiome" was coined by Joshua Lederberg, who argued that microorganisms inhabiting the human body should be included as part of the human genome, because of their influence on human physiology and the human body contains over 10 times more microbial cells than human cells (14,15). To understand the microbiome (totality of microbes) and the factors that influence the distribution and evolution of the constituent microorganisms, the Human Microbiome Project (HMP) was initiated. HMP has aimed to identify and characterize the microorganisms which are found in association with both healthy and diseased human (16-18). After investigating the feces of forty people living in rural communities and forty people inhibiting in cities by metagenomic analysis, scientists discovered that the distribution of intestinal bacteria of the two groups has a stark contrast. Surprisingly, it was found that those living in rural habitats have about three to five times more Lactobacillus acidophilus (LA). LA metabolizes lactic acid, obtains vitamin K, a cofactor for blood clotting and healthy bone tissue turnover. LA also helps recycle the amino acids in bile, and normalizes the body's cholesterol level compared to people living in cities. Possible explanations to this phenomenon are attributing to the level of stress and the difference in abiotic factors such as air and water and life span or health of people. Most importantly, distribution of helpful microorganisms is mainly derived from the types of foods consumed (19,20). The potential roles of microbiome on diverse disorders are summarized below.

Atopic dermatitis

Studies investigating the composition of the intestinal microflora in humans have shown the pivotal role of commensal bacteria in the development of allergies. Studies comparing the microflora composition of atopic and non-atopic infants also show significant differences. Non-atopic infants have more colonization of Bifidobacteria and Lactobacillus in the intestine while there is much colonization of Clostridia in the intestine of atopic infants. However several reports also suggest that there are no significant differences between the two groups (21,22). The exact role of intestinal flora in the development of atopic disease in the childhood is still not clarified (23). In addition to these quantitative differences in the Bifidobacterium, qualitative differences have also been observed. The Bifidobacteria from atopic infants were found to induce much higher levels of pro-inflammatory cytokines in vitro whereas the Bifidobacteria from non-atopic infants induced more secretion of anti-inflammatory cytokines. In addition to different secretion ability, differential adhesion to intestinal mucosa may also result in a different or reduced stimulation of the immune system through the gut-associated lymphoid tissue (GALT) (24).

Obesity

Obesity is associated with the changes in the relative abundance of the two dominant bacterial divisions, the Bacteroids and the Firmicutes. The relative proportion of Bacteroids is decreased in obese people compared to lean people while the proportion of Firmicutes is increased in obese people. Obese microbiome has an increased capacity to harvest energy from the diet and colonization of germ-free mice with an obese microbiota results in greater increase in total body fat without any increase in food consumption than colonization with a lean microbiota (25-27).

Type 1 diabetes (T1D)

Metagenomic analysis revealed the close relatedness of bacteria with the development of T1D associated autoimmunity in young child. Especially the proportion of Bacteroides ovatus was increased in children with T1D while the proportion of Firmicutes, CO19 was increased in normal healthy children (28).
All these results collectively imply that gut microbiota could be an additional contributing factor to the pathophysiology of obesity, T1D as well as atopic dermatitis, which in turn suggest that bacterial markers can be used for early diagnosis for these disorders. In addition, supplementation of large amount of bacteria that negatively correlated with the disease state may be beneficial for the prevention of these disorders.

Microflora and immune system

The intestinal microflora

Based on the hygiene hypothesis, in addition to environmental factors, the intestinal microflora is another contributor to allergic disease by substantially affecting the mucosal immunity. Allergic responses are thought to arise if there is absence of microbial exposure while the immune system is developing (24,29). The gastrointestinal tract is an important interface between host and environment and has the dual role of facilitating the absorption of nutrients while excluding pathogens. The human intestines are inhabited by at least 400 different bacterial species, with the greatest density in the large intestine, where concentration of 1011-1012 cells/g of luminal contents can be found (30). Because a large part of the intestinal microflora cannot be cultured with current culture techniques, it has been suggested that the number of microflora species in the human intestine may exceed 1,000 species (24). Although some intestinal bacteria are potential pathogens, the relationship between the intestinal microflora and the human host is mostly symbiotic in healthy individuals.

Gut-Associated Lymphoid Tissue (GALT)

The largest mass of lymphoid tissue of the human body can be found in the gastrointestinal tract and is called as the gut-associated lymphoid tissue (GALT) which includes Peyer's patch and mesenteric lymph node (MLN) (31). GALT interact with intestinal bacteria which are sampled by dendritic cells (DCs) and intestinal epithelial cells through the pattern recognition receptors such as toll-like receptor (TLR) and nucleotide-binding oligomerization domain (NOD) (32-34). The intestinal antigens also can be captured by DCs in the Peyer's patch through M cells on the surface of enterocytes. DCs in lamina propria can capture antigens directly by dendrites (35-37). Intestinal microflora seems to be involved in the induction of oral tolerance which is established in the periphery to specific antigen after ingestion of orally delivered protein (antigen) and plays a pivotal role in the induction and maintenance of peripheral tolerance mediated by regulatory T cells (38,39).

Skin microflora

Most studies delineating the role of resident microorganisms in regulation of immune system and inflammation have focused on the gut microbiome, since gut is the main site of exposure and contains a wide diversity of microbes. However, skin was considered to just have a physical barrier function to protect our body from the attack of pathogens. Interestingly recent studies discovered that skin microbiome also have similar function that of gut microbiome (40). Microbial profiling by metagenomic analysis has revealed the presence of highly diverse commensal communities in skin sites (41). Moreover, cutaneous inflammatory disorders, such as psoriasis and atopic dermatitis have been associated with imbalance of the cutaneous microbiota (42,43). Indeed, microbial products from skin commensals are known to exert immunoregulatory effects (44). Yasmine Belkaid group reported that the roles for the skin microbiota in controlling the local inflammatory milieu. They showed that protective immunity to a cutaneous pathogen is critically dependent on the skin but not on gut microbiota (45). Furthermore, skin commensals activate the function of local T cells through the interleukin-1 receptor (IL-1R)-MyD88 signaling pathway (45). These findings indicate the critical role of the skin residual commensal bacteria not only as a distinctive feature of tissue compartmentalization also as a key regulator of the immune system in health and disease.

Action mechanisms of probiotics

General regulation mechanisms in gut

The intestinal commensal bacteria play diverse functions in regulating nutrient metabolism including absorption of indigestible carbohydrate and production of short chain fatty acid, amino acid and vitamin. Microflora provides additional energy by digesting exogenous and endogenous substrates, such as fibers and mucins. Microflora also provide a protective barrier against incoming bacteria (46). This protection is mediated by several different mechanisms including competition for nutrients and binding sites and production of antimicrobial substances (37,47).

Immune regulation mechanism

Probiotics could stimulate the immune system by modulating the composition and/or activity of the intestinal microbiota (48,49). The immunomodulatory role of probiotics has been shown in diverse types of inflammatory immune disorders both, in gastrointestinal diseases and in non-gastrointestinal diseases. The beneficial effect of probiotics is strain-specific and their effector mechanisms are also quite diverse. Some probiotics generate IL-10 producing Tr1 or CD4+Foxp3+ Treg cells (50), while others could enhance immunity by producing a large amount of IL-1β, IL-12 or TNF-α (51). For example Lactobacillus reuteri induces the production of IL-12 and TNF-α from DCs while Lactobacillus casei inhibits the production of pro-inflammatory cytokines by producing anti-inflammatory cytokines such as IL-10. Therefore it is crucial to use specific probiotics or their mixture to target specific immune disorders. Most of the allergic diseases are associated with a shift of the Th1/Th2 balance toward a Th2 response, which induces secretion of Th2 type cytokines such as IL-4, IL-5 and IL-13 leading to higher production of IgE. Recently, we have demonstrated that IRT5 probiotics, a mixture of 5 probiotics, could suppress diverse immune disorders through the generation of CD4+Foxp3+ Tregs (52). Oral administration of IRT5 probiotics generated rDCs (iDOhigh, Cox2high, IL10+, TGFβ+), which can convert effector CD4+Foxp3- T cells into CD4+Foxp3+ regulatory T cells in the mucosal immune system. In addition, oral administration of the IRT5 probiotics suppressed ongoing experimental AD and pathogenesis of experimental arthritis as well (52). Currently we are investigating to identify potent probiotic strains that could induce the production of immune-modulatory cytokines such as IL-10 and TGF-β or enhancing Th1 response to improves the symptoms of allergic disease (37,53).

The effect of probiotics on dendritic cells (DCs)

DCs function as a professional antigen presenting cells (APC) and have the ability to activated T cells and directly regulate Th1 and Th2 cell. Especially DCs lead to the induction of regulatory T cells through the secretion of IL-10 and TGF-β and help to regulate immune response by the production of IL-10 from Tr1 cells and TGF-β from Th3 cells. Once DCs capture antigens in the presence of proper inflammatory stimuli, they migrate to secondary lymphoid organs and simultaneously undergo a maturation process. During this maturation, DCs produce diverse cytokines and express surface molecules such as MHC, CD40, CD83, CD80 (B7.1) and CD81 (B7.2). Recently several reports suggest that probiotics can induce the maturation and cytokine secretion by DCs. Different probiotics exert differential effect on the cytokine profiling by DCs such as IL-12, TNF-α and IL-10 (54-56). Recently we have demonstrated that administration of IRT5 probiotics mixture increases the generation of CD4+Foxp3+ Tregs in the mesenteric lymph nodes (MLN) through the generation of CD11c+ regulatory dendritic cells (rDCs) that highly express high levels of IL-10, TGF-β and indoleamine 2,3-dioxygenase (iDO) (52), which in turn potentiated the generation of CD4+Foxp3+ Tregs from the pool of CD4+CD25- cells. These results suggest that probiotics or a mixture of them that can induce regulatory DCs in the MLN, which in turn induce the production of CD4+Foxp3+ cells, could represent an attractive potential therapy for the treatment of diverse inflammatory immune disorders.

The role of probiotics in atopic dermatitis

Atopic dermatitis (AD) is caused by diverse pathogenic factors including genetic susceptibility, environment trigger, skin barrier dysfunction, bacterial infection and immune dysregulation (5). AD is a complex immune reaction mediated by both Th1 and Th2 immune responses (6). Th2 type cytokines including IL-4, IL-5 and IL-13 play important role in the development of AD by increasing the levels of serum IgE and blood eosinophils in AD patients (57). In the later stage of AD where infection mediated inflammation occurs, Th1 type cytokines such as IFN-γ and IL-12 mediate chronic stage of atopic dermatitis (5,58,59). IFN-γ is also involved in the maintenance of chronic stage of atopic dermatitis by elevating the expression of CCL17 (TARC) and CCL22 (MDC) that are involved in the recruitment of effector T cells to the inflamed site (60). IFN-γ increases the sensitivity of Fas-mediated apoptosis of keratinocytes, which is considered to be a key pathogenic event in eczematous dermatitis (61). Based on many clinical as well as animal studies, therapeutic or preventive effects of probiotics on allergic disease have been intensively investigated as summarized hereinafter.

1. Human clinical trials

(1) Prevention

Several randomized controlled trials (RCT) have investigated the effect of probiotics on the prevention of atopic dermatitis. In these trials probiotics were given to infants with a high risk of developing allergy, starting immediately after birth and mothers also received probiotics during the last week of pregnancy. The first trial reported that a 50% reduction of the incidence of AD in the probiotics group compare to placebo group at the age of 2 year and this effect was maintained until at 7 years (62). A clinical trial reported that administration of probiotics only reduced the incidence of IgE-associated AD without altering other allergic diseases (63). In another report, consumption of Lactobacillus rhamnosus reduced the incidence of AD while Bifidobacterium animalis did not show any reduction, suggesting the strain dependent preventive effect of probiotics (64). In contrast, two prevention studies did not show any effect on AD incidence. Moreover one of the studies reported an increased sensitization rate for food or aeroallergens in the probiotics group (65,66). There are several possible explanations for these inconclusive results. First, different lactobacillus strains were used and this indicates that not all strains are effective for atopic dermatitis. Second, probiotic effects are dose dependent and there was considerable variation in the daily dosage. The third explanation could be a difference in study design such as numbers and atopic risk of participants and supplementation of the mother during breastfeeding. These studies suggest that careful consideration much be established to decide which probiotics or a mixture of them are employed to treat allergic atopic dermatitis.

(2) Treatment

Randomized controlled trials to examine the effect of probiotics in the treatment of atopic dermatitis in child also have inconclusive results. Several studies showed a significant reduction of SCORAD (SCORing Atopic dermatitis) in the probiotics treated group (67). Other reports showed that probiotics significantly decreased SCORAD of IgE-associated not general AD (68,69). These reports suggest that only children who at a young age already have IgE sensitization such as elevated total or specific IgE levels get a benefit from probiotics administration. However there are also several studies that did not show any effect of probiotics on the severity or incidence of atopic dermatitis (70,71). There are considerable variations in the probiotic strains and daily dosage and number, age, severity of AD of participants and treatment period, which may result in conflicting results.

2. Experimental animal model

Beneficial effect of probiotics on AD development was evaluated in the experimental mouse models. We have recently showed that administration of IRT5 probiotics mixture to experimental AD mice reduced serum IgE levels and infiltration of lymphocytes to inflamed sites by recruiting CD4+Foxp3+ regulatory T cells (52). Administration of Lactobacillus rhamnosus GG (LGG) to Nc/Nga mice during pregnancy and breastfeeding also delayed onset or incidence of atopic dermatitis by increasing IL-10 level in GALT. (72). As a similar study, administration of three Lactobacillus strains isolated from Kimchi to Nc/Nga mice improved AD symptoms by significantly decreasing serum IgE levels as well as Th2 type cytokine expression while increasing IFN-γ and IL-10 expression (73,74). However, it is still unclear on the mechanism by which administration of probiotics generates regulatory T cell populations. In addition, further studies are required to identify potent immunoregulatroy probiotics among the hundreds of probioctic strains. Recently we developed ex vivo screening system to identify immunoregulatory probiotics that induce high levels of IL-10 and Foxp3+ Treg while reducing pro-inflammatory cytokines. We are currently under investigation to identify potent immunoregulatory probiotics from the traditional Korean fermented foods using the ex vivo screening system.

The role of probiotics in inflammatory bowel disease (IBD)

Inflammatory bowel disease (IBD) is characterized by a chronic dysregulation of the immune response in the gastrointestinal tract (GI tract). Proinflammatory cytokines such as INF-γ, TNF-α, IL-6 and IL-1β are major pathogenic cytokines but the pathogenesis is still not clear (75). Recently many studies have demonstrated that the GI tracts of patients with IBD are populated with increased levels of adherent and pathogenic bacteria. Fecal numbers of anaerobic bacteria such as Bacteroides species were greater while Lactobacilli and Bifidobacteria were fewer than in healthy control (76,77). Probiotics may be capable of recolonizing the bowel with non-pathogenic strains of bacteria. Probiotics have long been shown to be beneficial in both infectious and non-infectious digestive disorders. Growing evidence indicates that probiotics may be effective in the treatment of specific clinical IBD conditions.
Although it is unclear whether the abnormal composition of the entreric flora contributes to the pathogenesis of IBD, many clinical trials have examined the effect of diverse probiotic strains on the modulation of IBD progression including LGG (78) and E. coli strain Nissle 1917 (79,80). In addition, VSL#3, a mixture of 4 lactobacilli strains (Lactobacillus plantarum, Lactobacillus casei, Lactobacillus acidophilus, Lactobacillus delbrueckii ssp. bulgaricus), 3 bifidobacteria strains (Bifidobacterium infantis, Bifidobacterium breve, Bifidobacterium longum), and 1 strain of Streptococcus (Streptococcus salivarius ssp. thermophilus), has been examined in ulcerative colitis (UC), Crohn's disease (CD) (81,82). The therapeutic effects of several probiotic strains in animal studies and clinical trials were summarized in Table I and Table II, respectively.
Several studies have investigated the influence of probiotic consumption on colitis in animal trials. In particular, IL-10 knockout mouse that develops IBD spontaneously has been largely employed as a IBD model. Interestingly, IL-10 knockout mice spontaneously develop colitis when colonized with a conventional flora but remain disease free when maintained under germ-free conditions (83-87). These studies suggest the role of commensal flora in the development of inflammatory bowel disease. The role of probiotic strains in animal model of IBD was summarized in Table II.
Collectively these studies indicate that although the probiotic administration has potential therapeutic efficacy in IBD, larger controlled trials are necessary before the use of probiotics as a routine medical treatment.

Summary and future perspectives

Gastrointestinal microbes act as an environmental factor that affects the host's physiology, particularly in the context of obesity and its related metabolic disorder as well as allergic disorders (88,89). The significant involvement of the gut microbiota in human health and disease suggests that probiotics could be a novel therapeutic modality to treat diverse diseases by manipulating the composition of commensal microbiota (9). In fact, many reports suggest that enough consumption of probiotics is beneficial to suppress the development of atopic dermatitis by modifying the intestinal microflora and immune responses. Further studies are needed to evaluate the clinical and immunological effects of different strains of probiotics in well-designed clinical trials.

Figures and Tables

Table I
The role of probiotics in atopic dermatitis
in-12-129-i001
Table II
The role of probiotics in inflammatory bowel disease
in-12-129-i002

Abbreviations

GALT

Gut-Associated Lymphoid Tissue

GI

Gastrointestinal

MLN

Messenteric Lymph Node

AD

Atopic Dermatitis

HMP

Human Microbiome Project

IBD

Inflammatory Bowel Disease

T1D

Type 1 Diabetes

DC

Dendritic Cell

NOD

Nucleotide-binding Oligomerization Domain

TLT

Toll-Like Rceptor

Treg

Regulatory T Cells

iDO

Indoleamine 2,3-dioxygenase

SCORD

SCORing Atopic Dermatitis

ACKNOWLEDGEMENTS

This work was supported by grants from Research Program for Agricultural Science & Technology Development (Project No.: PJ907153), National Academy of Agricultural Science, Rural Development Administration, Republic of Korea, and by a grant from Korea Food Research Institute.

Notes

The authors have no financial conflict of interest.

References

1. Sanders ME. Probiotics: definition, sources, selection, and uses. Clin Infect Dis. 2008. 46:Suppl 2. S58–S61.
crossref
2. Feleszko W, Jaworska J, Rha RD, Steinhausen S, Avagyan A, Jaudszus A, Ahrens B, Groneberg DA, Wahn U, Hamelmann E. Probiotic-induced suppression of allergic sensitization and airway inflammation is associated with an increase of T regulatory-dependent mechanisms in a murine model of asthma. Clin Exp Allergy. 2007. 37:498–505.
crossref
3. Miraglia del Giudice M Jr, De Luca MG, Capristo C. Probiotics and atopic dermatitis. A new strategy in atopic dermatitis. Dig Liver Dis. 2002. 34:Suppl 2. S68–S71.
crossref
4. Sheil B, Shanahan F, O'Mahony L. Probiotic effects on inflammatory bowel disease. J Nutr. 2007. 137:3 Suppl 2. 819S–824S.
crossref
5. Leung DY, Boguniewicz M, Howel MD, Nomura I, Hamid QA. New insights into atopic dermatitis. J Clin Invest. 2004. 113:651–657.
crossref
6. Leung DY, Jain N, Leo HL. New concepts in the pathogenesis of atopic dermatitis. Curr Opin Immunol. 2003. 15:634–638.
crossref
7. Okada H, Kuhn C, Feillet H, Bach JF. The 'hygiene hypothesis' for autoimmune and allergic diseases: an update. Clin Exp Immunol. 2010. 160:1–9.
crossref
8. Atherton DJ. Topical corticosteroids in atopic dermatitis. BMJ. 2003. 327:942–943.
crossref
9. Jia W, Li H, Zhao L, Nicholson JK. Gut microbiota: a potential new territory for drug targeting. Nat Rev Drug Discov. 2008. 7:123–129.
crossref
10. Bach JF. The effect of infections on susceptibility to autoimmune and allergic diseases. N Engl J Med. 2002. 347:911–920.
crossref
11. Maggi E. The TH1/TH2 paradigm in allergy. Immunotechnology. 1998. 3:233–244.
crossref
12. Rautava S, Ruuskanen O, Ouwehand A, Salminen S, Isolauri E. The hygiene hypothesis of atopic disease--an extended version. J Pediatr Gastroenterol Nutr. 2004. 38:378–388.
crossref
13. Shreiner A, Huffnagle GB, Noverr MC. The "Microflora Hypothesis" of Allergic Disease. GI Microbiota and Regulation of the Immune System. 2008. Springer: New York, USA;113–134.
crossref
14. Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, Knight R, Gordon JI. The human microbiome project. Nature. 2007. 449:804–810.
crossref
15. Human Microbiome Jumpstart Reference Strains Consortium. Nelson KE, Weinstock GM, Highlander SK, Worley KC, Creasy HH, Wortman JR, Rusch DB, Mitreva M, Sodergren E, Chinwalla AT, Feldgarden M, Gevers D, Haas BJ, Madupu R, Ward DV, Birren BW, Gibbs RA, Methe B, Petrosino JF, Strausberg RL, Sutton GG, White OR, Wilson RK, Durkin S, Giglio MG, Gujja S, Howarth C, Kodira CD, Kyrpides N, Mehta T, Muzny DM, Pearson M, Pepin K, Pati A, Qin X, Yandava C, Zeng Q, Zhang L, Berlin AM, Chen L, Hepburn TA, Johnson J, McCorrison J, Miller J, Minx P, Nusbaum C, Russ C, Sykes SM, Tomlinson CM, Young S, Warren WC, Badger J, Crabtree J, Markowitz VM, Orvis J, Cree A, Ferriera S, Fulton LL, Fulton RS, Gillis M, Hemphill LD, Joshi V, Kovar C, Torralba M, Wetterstrand KA, Abouellleil A, Wollam AM, Buhay CJ, Ding Y, Dugan S, FitzGerald MG, Holder M, Hostetler J, Clifton SW, Allen-Vercoe E, Earl AM, Farmer CN, Liolios K, Surette MG, Xu Q, Pohl C, Wilczek-Boney K, Zhu D. A catalog of reference genomes from the human microbiome. Science. 2010. 328:994–999.
crossref
16. Tlaskalová-Hogenová H, Stěpánková R, Kozáková H, Hudcovic T, Vannucci L, Tučková L, Rossmann P, Hrnčíř T, Kverka M, Zákostelská Z, Klimešová K, Přibylová J, Bártová J, Sanchez D, Fundová P, Borovská D, Srůtková D, Zídek Z, Schwarzer M, Drastich P, Funda DP. The role of gut microbiota (commensal bacteria) and the mucosal barrier in the pathogenesis of inflammatory and autoimmune diseases and cancer: contribution of germ-free and gnotobiotic animal models of human diseases. Cell Mol Immunol. 2011. 8:110–120.
crossref
17. Human Microbiome Project Consortium. A framework for human microbiome research. Nature. 2012. 486:215–221.
18. Cho I, Blaser MJ. The human microbiome: at the interface of health and disease. Nat Rev Genet. 2012. 13:260–270.
crossref
19. Flohr C, Pascoe D, Williams HC. Atopic dermatitis and the 'hygiene hypothesis': too clean to be true? Br J Dermatol. 2005. 152:202–216.
crossref
20. Gibbs S, Surridge H, Adamson R, Cohen B, Bentham G, Reading R. Atopic dermatitis and the hygiene hypothesis: a case-control study. Int J Epidemiol. 2004. 33:199–207.
crossref
21. Kalliomäki 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. 107:129–134.
crossref
22. Penders J, Thijs C, van den Brandt PA, Kummeling I, Snijders B, Stelma F, Adams H, van Ree R, Stobberingh EE. Gut microbiota composition and development of atopic manifestations in infancy: the KOALA Birth Cohort Study. Gut. 2007. 56:661–667.
crossref
23. 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. 17:141–147.
crossref
24. Ouwehand AC. Antiallergic effects of probiotics. J Nutr. 2007. 137:3 Suppl 2. 794S–797S.
crossref
25. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006. 444:1027–1031.
crossref
26. Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: human gut microbes associated with obesity. Nature. 2006. 444:1022–1023.
27. Million M, Maraninchi M, Henry M, Armougom F, Richet H, Carrieri P, Valero R, Raccah D, Vialettes B, Raoult D. Obesity-associated gut microbiota is enriched in Lactobacillus reuteri and depleted in Bifidobacterium animalis and Methanobrevibacter smithii. Int J Obes (Lond). 2012. 36:817–825.
crossref
28. Giongo A, Gano KA, Crabb DB, Mukherjee N, Novelo LL, Casella G, Drew JC, Ilonen J, Knip M, Hyöty H, Veijola R, Simell T, Simell O, Neu J, Wasserfall CH, Schatz D, Atkinson MA, Triplett EW. Toward defining the autoimmune microbiome for type 1 diabetes. ISME J. 2011. 5:82–91.
crossref
29. Ogden NS, Bielory L. Probiotics: a complementary approach in the treatment and prevention of pediatric atopic disease. Curr Opin Allergy Clin Immunol. 2005. 5:179–184.
crossref
30. Guarner F, Malagelada JR. Gut flora in health and disease. Lancet. 2003. 361:512–519.
crossref
31. Artis D. Epithelial-cell recognition ofcommensal bacteria and maintenance of immune homeostasis in the gut. Nat Rev Immunol. 2008. 8:411–420.
crossref
32. Macdonald TT, Monteleone G. Immunity, inflammation, and allergy in the gut. Science. 2005. 307:1920–1925.
crossref
33. Kelly D, Conway S, Aminov R. Commensal gut bacteria: mechanisms of immune modulation. Trends Immunol. 2005. 26:326–333.
crossref
34. Coombes JL, Powrie F. Dendritic cells in intestinal immune regulation. Nat Rev Immunol. 2008. 8:435–446.
crossref
35. Rescigno M, Urbano M, Valzasina B, Francolini M, Rotta G, Bonasio R, Granucci F, Kraehenbuhl JP, Ricciardi-Castagnoli P. Dendritic cells express tight junction proteins and penetrate gut epithelial monolayers to sample bacteria. Nat Immunol. 2001. 2:361–367.
crossref
36. Mowat AM. Anatomical basis of tolerance and immunity to intestinal antigens. Nat Rev Immunol. 2003. 3:331–341.
crossref
37. O'Hara AM, Shanahan F. The gut flora as a forgotten organ. EMBO Rep. 2006. 7:688–693.
38. 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 Immunol. 1997. 159:1739–1745.
39. Ostman S, Rask C, Wold AE, Hultkrantz S, Telemo E. Impaired regulatory T cell function in germ-free mice. Eur J Immunol. 2006. 36:2336–2346.
crossref
40. Kong HH. Skin microbiome: genomics-based insights into the diversity and role of skin microbes. Trends Mol Med. 2011. 17:320–328.
crossref
41. Grice EA, Kong HH, Conlan S, Deming CB, Davis J, Young AC, Bouffard GG, Blakesley RW, Murray PR, Green ED, Turner ML, Segre JA. NISC Comparative Sequencing Program. Topographical and temporal diversity of the human skin microbiome. Science. 2009. 324:1190–1192.
crossref
42. Gallo RL, Nakatsuji T. Microbial symbiosis with the innate immune defense system of the skin. J Invest Dermatol. 2011. 131:1974–1980.
crossref
43. Kong HH, Oh J, Deming C, Conlan S, Grice EA, Beatson MA, Nomicos E, Polley EC, Komarow HD, Murray PR, Turner ML, Segre JA. NISC Comparative Sequence Program. Temporal shifts in the skin microbiome associated with disease flares and treatment in children with atopic dermatitis. Genome Res. 2012. 22:850–859.
crossref
44. Lai Y, Di Nardo A, Nakatsuji T, Leichtle A, Yang Y, Cogen AL, Wu ZR, Hooper LV, Schmidt RR, von Aulock S, Radek KA, Huang CM, Ryan AF, Gallo RL. Commensal bacteria regulate Toll-like receptor 3-dependent inflammation after skin injury. Nat Med. 2009. 15:1377–1382.
crossref
45. Naik S, Bouladoux N, Wilhelm C, Molloy MJ, Salcedo R, Kastenmuller W, Deming C, Quinones M, Koo L, Conlan S, Spencer S, Hall JA, Dzutsev A, Kong H, Campbell DJ, Trinchieri G, Segre JA, Belkaid Y. Compartmentalized control of skin immunity by resident commensals. Science. 2012. 337:1115–1119.
crossref
46. Ouwehand AC, Derrien M, de Vos W, Tiihonen K, Rautonen N. Prebiotics and other microbial substrates for gut functionality. Curr Opin Biotechnol. 2005. 16:212–217.
crossref
47. Adlerberth I, Cerquetti M, Poilane I, Wold A, Collignon A. Mechanisms of Colonisation and Colonisation Resistance of the Digestive Tract Part 1: Bacteria/host Interactions. Microb Ecol Health Dis. 2000. 2:223–239.
48. Fang H, Elina T, Heikki A, Seppo S. Modulation of humoral immune response through probiotic intake. FEMS Immunol Med Microbiol. 2000. 29:47–52.
crossref
49. Gill HS, Rutherfurd KJ, Cross ML, Gopal PK. Enhancement of immunity in the elderly by dietary supplementation with the probiotic Bifidobacterium lactis HN019. Am J Clin Nutr. 2001. 74:833–839.
crossref
50. Smits HH, Engering A, van der Kleij D, de Jong EC, Schipper K, van Capel TM, Zaat BA, Yazdanbakhsh M, Wierenga EA, van Kooyk Y, Kapsenberg ML. Selective probiotic bacteria induce IL-10-producing regulatory T cells in vitro by modulating dendritic cell function through dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin. J Allergy Clin Immunol. 2005. 115:1260–1267.
crossref
51. Ueno N, Fujiya M, Segawa S, Nata T, Moriichi K, Tanabe H, Mizukami Y, Kobayashi N, Ito K, Kohgo Y. Heat-killed body of lactobacillus brevis SBC8803 ameliorates intestinal injury in a murine model of colitis by enhancing the intestinal barrier function. Inflamm Bowel Dis. 2011. 17:2235–2250.
crossref
52. Kwon HK, Lee CG, So JS, Chae CS, Hwang JS, Sahoo A, Nam JH, Rhee JH, Hwang KC, Im SH. Generation of regulatory dendritic cells and CD4+Foxp3+ T cells by probiotics administration suppresses immune disorders. Proc Natl Acad Sci U S A. 2010. 107:2159–2164.
crossref
53. Winkler P, Ghadimi D, Schrezenmeir J, Kraehenbuhl JP. Molecular and cellular basis of microflora-host interactions. J Nutr. 2007. 137:3 Suppl 2. 756S–772S.
crossref
54. Fink LN, Zeuthen LH, Ferlazzo G, Frøkiaer H. Human antigen-presenting cells respond differently to gut-derived probiotic bacteria but mediate similar strain-dependent NK and T cell activation. FEMS Immunol Med Microbiol. 2007. 51:535–546.
crossref
55. Young SL, Simon MA, Baird MA, Tannock GW, Bibiloni R, Spencely K, Lane JM, Fitzharris P, Crane J, Town I, Addo-Yobo E, Murray CS, Woodcock A. Bifidobacterial species differentially affect expression of cell surface markers and cytokines of dendritic cells harvested from cord blood. Clin Diagn Lab Immunol. 2004. 11:686–690.
crossref
56. Zeuthen LH, Christensen HR, Frøkiaer H. Lactic acid bacteria inducing a weak interleukin-12 and tumor necrosis factor alpha response in human dendritic cells inhibit strongly stimulating lactic acid bacteria but act synergistically with gram-negative bacteria. Clin Vaccine Immunol. 2006. 13:365–375.
crossref
57. Homey B, Steinhoff M, Ruzicka T, Leung DY. Cytokines and chemokines orchestrate atopic skin inflammation. J Allergy Clin Immunol. 2006. 118:178–189.
crossref
58. Vestergaard C, Kirstejn N, Gesser B, Mortensen JT, Matsushima K, Larsen CG. IL-10 augments the IFN-gamma and TNF-alpha induced TARC production in HaCaT cells: a possible mechanism in the inflammatory reaction of atopic dermatitis. J Dermatol Sci. 2001. 26:46–54.
crossref
59. Konur A, Schulz U, Eissner G, Andreesen R, Holler E. Interferon (IFN)-gamma is a main mediator of keratinocyte (HaCaT) apoptosis and contributes to autocrine IFN-gamma and tumour necrosis factor-alpha production. Br J Dermatol. 2005. 152:1134–1142.
crossref
60. Pivarcsi A, Homey B. Chemokine networks in atopic dermatitis: traffic signals of disease. Curr Allergy Asthma Rep. 2005. 5:284–290.
crossref
61. Trautmann A, Akdis M, Kleemann D, Altznauer F, Simon HU, Graeve T, Noll M, Bröcker EB, Blaser K, Akdis CA. T cell-mediated Fas-induced keratinocyte apoptosis plays a key pathogenetic role in eczematous dermatitis. J Clin Invest. 2000. 106:25–35.
crossref
62. Kalliomäki M, Salminen S, Arvilommi H, Kero P, Koskinen P, Isolauri E. Probiotics in primary prevention of atopic disease: a randomised placebo-controlled trial. Lancet. 2001. 357:1076–1079.
crossref
63. Abrahamsson TR, Jakobsson T, Böttcher MF, Fredrikson M, Jenmalm MC, Björkstén B, Oldaeus G. Probiotics in prevention of IgE-associated eczema: a double-blind, randomized, placebo-controlled trial. J Allergy Clin Immunol. 2007. 119:1174–1180.
crossref
64. Wickens K, Black PN, Stanley TV, Mitchell E, Fitzharris P, Tannock GW, Purdie G, Crane J. Probiotic Study Group. 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. 122:788–794.
crossref
65. Soh SE, Aw M, Gerez I, Chong YS, Rauff M, Ng YP, Wong HB, Pai N, Lee BW, Shek LP. Probiotic supplementation in the first 6 months of life in at risk Asian infants--effects on eczema and atopic sensitization at the age of 1 year. Clin Exp Allergy. 2009. 39:571–578.
crossref
66. Kopp MV, Hennemuth I, Heinzmann A, Urbanek R. Randomized, double-blind, placebo-controlled trial of probiotics forprimary prevention: no clinical effects of Lactobacillus GG supplementation. Pediatrics. 2008. 121:e850–e856.
67. Weston S, Halbert A, Richmond P, Prescott SL. Effects of probiotics on atopic dermatitis: a randomised controlled trial. Arch Dis Child. 2005. 90:892–897.
crossref
68. Viljanen M, Savilahti E, Haahtela T, Juntunen-Backman K, Korpela R, Poussa T, Tuure T, Kuitunen M. Probiotics in the treatment of atopic eczema/dermatitis syndrome in infants: a double-blind placebo-controlled trial. Allergy. 2005. 60:494–500.
crossref
69. Sistek D, Kelly R, Wickens K, Stanley T, Fitzharris P, Crane J. Is the effect of probiotics on atopic dermatitis confined to food sensitized children? Clin Exp Allergy. 2006. 36:629–633.
crossref
70. Fölster-Holst R, Müller F, Schnopp N, Abeck D, Kreiselmaier I, Lenz T, von Rüden U, Schrezenmeir J, Christophers E, Weichenthal M. Prospective, randomized controlled trial on Lactobacillus rhamnosus in infants with moderate to severe atopic dermatitis. Br J Dermatol. 2006. 155:1256–1261.
crossref
71. Chen YS, Jan RL, Lin YL, Chen HH, Wang JY. Randomized placebo-controlled trial of lactobacillus on asthmatic children with allergic rhinitis. Pediatr Pulmonol. 2010. 45:1111–1120.
crossref
72. Sawada J, Morita H, Tanaka A, Salminen S, He F, Matsuda H. Ingestion of heat-treated Lactobacillus rhamnosus GG prevents development of atopic dermatitis in NC/Nga mice. Clin Exp Allergy. 2007. 37:296–303.
crossref
73. Tanaka A, Jung K, Benyacoub J, Prioult G, Okamoto N, Ohmori K, Blum S, Mercenier A, Matsuda H. Oral supplementation with Lactobacillus rhamnosus CGMCC 1.3724 prevents development of atopic dermatitis in NC/NgaTnd mice possibly by modulating local production of IFN-gamma. Exp Dermatol. 2009. 18:1022–1027.
crossref
74. Won TJ, Kim B, Lim YT, Song DS, Park SY, Park ES, Lee DI, Hwang KW. Oral administration of Lactobacillus strains from Kimchi inhibits atopic dermatitis in NC/Nga mice. J Appl Microbiol. 2011. 110:1195–1202.
crossref
75. Bouma G, Strober W. The immunological and genetic basis of inflammatory bowel disease. Nat Rev Immunol. 2003. 3:521–533.
crossref
76. Swidsinski A, Ladhoff A, Pernthaler A, Swidsinski S, Loening-Baucke V, Ortner M, Weber J, Hoffmann U, Schreiber S, Dietel M, Lochs H. Mucosal flora in inflammatory bowel disease. Gastroenterology. 2002. 122:44–54.
crossref
77. Favier C, Neut C, Mizon C, Cortot A, Colombel JF, Mizon J. Fecal beta-D-galactosidase production and Bifidobacteria are decreased in Crohn's disease. Dig Dis Sci. 1997. 42:817–822.
78. Gupta P, Andrew H, Kirschner BS, Guandalini S. Is lactobacillus GG helpful in children with Crohn's disease? Results of a preliminary, open-label study. J Pediatr Gastroenterol Nutr. 2000. 31:453–457.
crossref
79. Kruis W, Fric P, Stolte MS. Maintenance of remission in ulcerative colitis is equally effective with Escherichia coli Nissle 1917 and with standard mesalamine. Gastroenterology. 2001. 120:5 Suppl 1. A127.
crossref
80. Malchow HA. Crohn's disease and Escherichia coli. A new approach in therapy to maintain remission of colonic Crohn's disease? J Clin Gastroenterol. 1997. 25:653–658.
81. Gionchetti P, Rizzello F, Venturi A, Brigidi P, Matteuzzi D, Bazzocchi G, Poggioli G, Miglioli M, Campieri M. Oral bacteriotherapy as maintenance treatment in patients with chronic pouchitis: a double-blind, placebo-controlled trial. Gastroenterology. 2000. 119:305–309.
crossref
82. Venturi A, Gionchetti P, Rizzello F, Johansson R, Zucconi E, Brigidi P, Matteuzzi D, Campieri M. Impact on the composition of the faecal flora by a new probiotic preparation: preliminary data on maintenance treatment of patients with ulcerative colitis. Aliment Pharmacol Ther. 1999. 13:1103–1108.
crossref
83. O'Mahony L, Feeney M, O'Halloran S, Murphy L, Kiely B, Fitzgibbon J, Lee G, O'Sullivan G, Shanahan F, Collins JK. Probiotic impact on microbial flora, inflammation and tumour development in IL-10 knockout mice. Aliment Pharmacol Ther. 2001. 15:1219–1225.
84. Sheil B, MacSharry J, O'Callaghan L, O'Riordan A, Waters A, Morgan J, Collins JK, O'Mahony L, Shanahan F. Role of interleukin (IL-10) in probiotic-mediated immune modulation: an assessment in wild-type and IL-10 knock-out mice. Clin Exp Immunol. 2006. 144:273–280.
crossref
85. Sheil B, McCarthy J, O'Mahony L, Bennett MW, Ryan P, Fitzgibbon JJ, Kiely B, Collins JK, Shanahan F. Is the mucosal route of administration essential for probiotic function? Subcutaneous administration is associated with attenuation of murine colitis and arthritis. Gut. 2004. 53:694–700.
crossref
86. Madsen KL, Doyle JS, Jewell LD, Tavernini MM, Fedorak RN. Lactobacillus species prevents colitis in interleukin 10 gene-deficient mice. Gastroenterology. 1999. 116:1107–1114.
crossref
87. Schultz M, Veltkamp C, Dieleman LA, Grenther WB, Wyrick PB, Tonkonogy SL, Sartor RB. Lactobacillus plantarum 299V in the treatment and prevention of spontaneous colitis in interleukin-10-deficient mice. Inflamm Bowel Dis. 2002. 8:71–80.
crossref
88. Khoruts A, Sadowsky MJ. Therapeutic transplantation of the distal gut microbiota. Mucosal Immunol. 2011. 4:4–7.
crossref
89. Delzenne NM, Neyrinck AM, Bäckhed F, Cani PD. Targeting gut microbiota in obesity: effects of prebiotics and probiotics. Nat Rev Endocrinol. 2011. 7:639–646.
crossref
TOOLS
Similar articles