Top Banner
The incidence and prevalence of chronic inflammatory diseases have increased markedly since the end of the Second World War, in line with the pattern of increas- ing urbanization and industrialization. This is particu- larly evident for inflammatory disorders that develop in the mucosal tissues of the airways and the gut, such as asthma and chronic inflammatory bowl disease (IBD) 1 . The hygiene hypothesis was introduced to explain the increased incidence of these diseases in affluent socie- ties 2,3 . According to this theory, modern hygiene, and dietary and medical practices affect the composition of the gut microbiota and limit exposure of infants to pathogens. This change in the microbiota, in combi- nation with genetic and epigenetic factors, influences not only the epithelial mucosal barrier but also peri- natal maturation of the immune system, thus leading to disease susceptibility. Mucosal tissue homeostasis results from the peri- natal establishment of mucosally induced immune tolerance, which has been extensively studied in terms of immunological hyporesponsiveness to ingested innocuous antigens (known as oral tolerance) and to components of the indigenous gut microbiota. Similar tolerogenic mechanisms can be induced through the respiratory mucosa. Perinatal defects in the induc- tion of mucosal tolerance are associated with the later development of allergies, autoimmune diseases (such as rheumatoid arthritis, type 1 diabetes and systemic lupus erythematosus) 4–6 and chronic inflammation of the gut and respiratory mucosae. In both the gut and the airways, mucosal tolerance is regulated by a set of signals provided by innate immune cells that shape adaptive immune responses. The mucosal epithelium controls this regulatory immune network through its barrier function, cell contact-mediated signals and the production of cytokines. It is now well recognized that environmental fac- tors, either directly or indirectly, have a decisive role in perinatal maturation of the mucosal immune system. The colonization of mucosal surfaces by commensal microorganisms is of eminent importance 7,8 . Although current research focuses mainly on the intestinal micro- biome, the functional relevance of the respiratory microbiome is also emerging 9,10 . In addition, perinatal exposure to cigarette smoke, environmental microorganisms and dietary constitu- ents has a marked effect on the early programming of the innate and adaptive immune systems through the stimulation of pattern recognition receptors (PRRs) and by modifying cellular interactions; thus, such exposure directly influences disease develop- ment. Indeed, identified susceptibility genes for both respiratory and enteric chronic inflammatory dis- eases are associated with epithelial barrier integrity, innate immune recognition and adaptive immune stimulation 11–13 . 1 Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Philipps University Marburg, Medical Faculty, Baldingerstrasse, 35043 Marburg, Germany. 2 Laboratory for Immunohistochemistry and Immunopathology (LIIPAT), Centre for Immune Regulation (CIR), University of Oslo, and Department of Pathology, Oslo University Hospital, Rikshospitalet, Oslo, Norway. 3 Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany. Correspondence to H.R. e-mail: [email protected] doi:10.1038/nri3112 Published online 9 December 2011 The impact of perinatal immune development on mucosal homeostasis and chronic inflammation Harald Renz 1 , Per Brandtzaeg 2 and Mathias Hornef 3 Abstract | The mucosal surfaces of the gut and airways have important barrier functions and regulate the induction of immunological tolerance. The rapidly increasing incidence of chronic inflammatory disorders of these surfaces, such as inflammatory bowel disease and asthma, indicates that the immune functions of these mucosae are becoming disrupted in humans. Recent data indicate that events in prenatal and neonatal life orchestrate mucosal homeostasis. Several environmental factors promote the perinatal programming of the immune system, including colonization of the gut and airways by commensal microorganisms. These complex microbial–host interactions operate in a delicate temporal and spatial manner and have an important role in the induction of homeostatic mechanisms. REVIEWS NATURE REVIEWS | IMMUNOLOGY VOLUME 12 | JANUARY 2012 | 9 © 2012 Macmillan Publishers Limited. All rights reserved
15

The impact of perinatal immune development on mucosal ...manyan/To print/The impact of perinatal immune... · natal development in mice. During the first two weeks of postnatal life,

Aug 27, 2019

Download

Documents

hoangmien
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: The impact of perinatal immune development on mucosal ...manyan/To print/The impact of perinatal immune... · natal development in mice. During the first two weeks of postnatal life,

The incidence and prevalence of chronic inflammatory diseases have increased markedly since the end of the Second World War, in line with the pattern of increas­ing urbanization and industrialization. This is particu­larly evident for inflammatory disorders that develop in the mucosal tissues of the airways and the gut, such as asthma and chronic inflammatory bowl disease (IBD)1. The hygiene hypothesis was introduced to explain the increased incidence of these diseases in affluent socie­ties2,3. According to this theory, modern hygiene, and dietary and medical practices affect the composition of the gut microbiota and limit exposure of infants to pathogens. This change in the microbiota, in combi­nation with genetic and epigenetic factors, influences not only the epithelial mucosal barrier but also peri­natal maturation of the immune system, thus leading to disease susceptibility.

Mucosal tissue homeostasis results from the peri­natal establishment of mucosally induced immune tolerance, which has been extensively studied in terms of immunological hyporesponsiveness to ingested innocuous antigens (known as oral tolerance) and to components of the indigenous gut microbiota. Similar tolerogenic mechanisms can be induced through the respiratory mucosa. Perinatal defects in the induc­tion of mucosal tolerance are associated with the later development of allergies, autoimmune diseases (such as rheumatoid arthritis, type 1 diabetes and systemic

lupus erythematosus)4–6 and chronic inflammation of the gut and respiratory mucosae. In both the gut and the airways, mucosal tolerance is regulated by a set of signals provided by innate immune cells that shape adaptive immune responses. The mucosal epithelium controls this regulatory immune network through its barrier function, cell contact­mediated signals and the production of cytokines.

It is now well recognized that environmental fac­tors, either directly or indirectly, have a decisive role in perinatal maturation of the mucosal immune system. The colonization of mucosal surfaces by commensal microorganisms is of eminent importance7,8. Although current research focuses mainly on the intestinal micro­biome, the functional relevance of the respiratory microbiome is also emerging9,10.

In addition, perinatal exposure to cigarette smoke, environmental microorganisms and dietary constitu­ents has a marked effect on the early programming of the innate and adaptive immune systems through the stimulation of pattern recognition receptors (PRRs) and by modifying cellular interactions; thus, such exposure directly influences disease develop­ment. Indeed, identified susceptibility genes for both respiratory and enteric chronic inflammatory dis­eases are associated with epithelial barrier integrity, innate immune recognition and adaptive immune stimulation11–13.

1Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Philipps University Marburg, Medical Faculty, Baldingerstrasse, 35043 Marburg, Germany.2Laboratory for Immunohistochemistry and Immunopathology (LIIPAT), Centre for Immune Regulation (CIR), University of Oslo, and Department of Pathology, Oslo University Hospital, Rikshospitalet, Oslo, Norway.3Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany.Correspondence to H.R. e-mail: [email protected]:10.1038/nri3112Published online 9 December 2011

The impact of perinatal immune development on mucosal homeostasis and chronic inflammationHarald Renz1, Per Brandtzaeg2 and Mathias Hornef3

Abstract | The mucosal surfaces of the gut and airways have important barrier functions and regulate the induction of immunological tolerance. The rapidly increasing incidence of chronic inflammatory disorders of these surfaces, such as inflammatory bowel disease and asthma, indicates that the immune functions of these mucosae are becoming disrupted in humans. Recent data indicate that events in prenatal and neonatal life orchestrate mucosal homeostasis. Several environmental factors promote the perinatal programming of the immune system, including colonization of the gut and airways by commensal microorganisms. These complex microbial–host interactions operate in a delicate temporal and spatial manner and have an important role in the induction of homeostatic mechanisms.

REVIEWS

NATURE REVIEWS | IMMUNOLOGY VOLUME 12 | JANUARY 2012 | 9

© 2012 Macmillan Publishers Limited. All rights reserved

Page 2: The impact of perinatal immune development on mucosal ...manyan/To print/The impact of perinatal immune... · natal development in mice. During the first two weeks of postnatal life,

Oral toleranceThis form of tolerance is established through the intestinal mucosa to avoid local and systemic hypersensitivity to innocuous antigens that have breached the epithelial barrier. It is mediated mainly through the induction of regulatory T cells but probably also through other mechanisms, such as T cell anergy and clonal deletion.

Crypt–villus architectureCrypts are glandular invaginations of the intestinal epithelium. Paneth cells localize to the base of crypts in the small intestine and secrete antimicrobial peptides. Intestinal stem cells in crypts divide continuously and provide rapid epithelial cell renewal. In the small intestine, villi are finger-like protrusions into the gut lumen, which increase the absorptive surface of the gut epithelium. Villi mainly consist of mature, absorptive enterocytes, but also contain mucus-secreting goblet cells.

CathelicidinCathelicidins are a large family of antimicrobial peptides in ruminants. In mice and humans, only one cathelicidin — CRAMP or LL37, respectively — is expressed. The genes encoding CRAMP and LL37 are expressed by most epithelial cells, as well as granulocytes, and their expression is regulated at the level of transcription. They encode a highly conserved ‘cathelin’ sequence of 12 kDa at the amino terminus, followed by the mature antimicrobial peptide, which requires enzymatic cleavage to become active.

DefensinsA class of antimicrobial peptides with broad antibacterial, antifungal and, in part, antiviral activity. α-defensins are produced constitutively by small intestinal Paneth cells and, in humans, also by neutrophils, whereas many β-defensins are transcriptionally regulated and expressed by most epithelial cells.

This Review focuses on disorders of the mucosae but does not present a complete picture of the many factors that contribute to inflammatory diseases of the gut and airways. Instead, we highlight perinatal variables that influence mucosal barrier function and host defence and that when dysregulated can contribute to the patho gene­sis of chronic inflammatory disorders of the respiratory and intestinal tracts.

Perinatal development of the gut mucosaThe intestinal mucosal barrier. The level of maturity of the neonatal gut varies between species, depending on the length of the gestation period. The small intestinal mucosa of human newborns has a mature crypt–villus architecture, with continuous stem cell proliferation, and epithelial cell migration and differentiation. In mice, small intestinal crypts only develop 10–12 days after birth and this is accompanied by increased epithelial cell renewal and the transcriptional reprogramming of enterocytes, which involves changes in the expression of genes involved in nutrient metabolism, nutrient transport and cell differentiation14,15. Also, the enteric spectrum of antimicrobial peptides changes significantly during neo­natal development in mice. During the first two weeks of postnatal life, when mature crypt­based Paneth cells are absent16,17, the mouse intestinal epithelium expresses cathelicidin­related antimicrobial peptide (CRAMP)18. CRAMP expression decreases with weaning, at the same time that Paneth cells start to produce defensins (FIG. 1). Whereas defensin secretion by mature Paneth cells does not depend on bacterial colonization of the gut19, expres­sion of another antimicrobial peptide by epithelial cells of the adult intestine — the C­type lectin regenerating islet­derived protein 3γ (REG3γ) — requires bacterial coloni­zation of the intestine and is supported by interleukin­22 (IL­22)­producing RORγt+NKp46+ lymphocytes20,21. Of note, changes in the composition of antimicrobial pep­tides and in antibacterial activity during the early post­natal period have also been noted in the intestinal lumen of human neonates22. Thus, significant alterations of the antimicrobial peptide repertoire of the intestinal mucosa occur during the early postnatal period. As enteric anti­microbial peptide synthesis has been associated with changes in the composition of the microbiota and chronic mucosal inflammation, these processes during the post­natal period might significantly affect susceptibility to inflammatory disease later in life23 (FIG. 1).

Colonization of the gut mucosa by commensals. Birth marks the transition from a sterile fetal environment to one that is rich in microorganisms and nutritional or other exogenous substrates24,25. The first microbial encounter of a neonate seems to determine the early postnatal intestinal microbiota: after natural birth, the microbiota composition resembles that of the maternal vaginal or gut microbiota, whereas after Caesarean sec­tion, the intestinal microbiota of the infant includes a large number of environmental bacteria25. With wean­ing, an increasingly diverse microbiota is established that is highly individual and remains relatively stable throughout life26,27.

Perinatal microbiota–epithelial crosstalk. Exposure of mouse pups during or shortly after natural birth to lipopoly saccharide (LPS) from Gram­negative bacteria induces a transient transcriptional activation of the small intestinal epithelium with upregulated expression of microRNA­146a (miR­146a). Increased levels of miR­146a in intestinal epithelial cells cause translational repres­sion of the Toll­like receptor (TLR) signalling molecule IL­1­associated kinase 1 (IRAK1). This, together with the proteasomal degradation of IRAK1, contributes to innate immune tolerance by inhibiting TLR signalling28. Other mechanisms that help to prevent inappropriate immune stimulation by TLR agonists during postnatal colonization of the neonatal intestine include the downregulation of expression of TLR4 in the intestinal epithelium, expres­sion of which is high in late fetal life of mice but decreases at birth29 (FIG. 1). Conversely, epithelial expression of the nuclear factor­κB inhibitor IκBα steadily increases dur­ing the postnatal period30. The combination of decreasing levels of TLR4 and increasing levels of IκBα expression by gut epithelial cells effectively increases the threshold of immune activation in the gut epithelium (FIG. 1).

Interestingly, decreased IRAK1 protein expression in mouse neonatal epithelium requires continuous TLR signalling. This facilitates prolonged upregulation of miR­146a expression and simultaneously induces sus­tained expression of genes supporting cell maturation, survival and nutrient absorption31. Innate immune sig­nalling by epithelial cells seems to be essential for immune tolerance, as lack of the pro­inflammatory signalling mol­ecule transforming growth factor­β (TGFβ)­activated kinase 1 (TAK1) specifically in the murine intestinal epithelium leads to early inflammation, tissue damage and postnatal mortality32. Thus, although inappropriate stimulation of the neonatal innate immune system by the microbiota must be prevented, controlled innate immune activation significantly contributes to nutrient absorption, angiogenesis, epithelial cell differentiation and barrier fortification33,34.

Despite the decreased sensitivity of epithelial cells to TLR stimulation in murine neonates, other innate immune signalling pathways remain fully functional. For example, rotavirus infection of the intestinal epithelium in neonatal mice is efficiently sensed by the helicases retinoic acid­inducible gene I (RIG­I) and melanoma differentiation­associated gene 5 (MDA5)35,36. So, when confronted with the emergence of the intestinal microbiota, the neonatal gut epithelium seems to adjust its sensitivity to micro­organisms and modify its signalling pathways after initial stimulation while maintaining antiviral host defences.

Perinatal maturation of the intestinal immune system. It is well known that both endogenous and exogenous factors drive the development and maturation of the intestinal immune system (FIG. 1). In mice and humans, the formation of secondary lymphoid structures — such as Peyer’s patches and mesenteric lymph nodes (MLNs) — occurs before birth, but their size and the development of germinal centres depend on the postnatal microbial colonization of the gut37–39. Also, cryptopatches and isolated lymphoid follicles are seen only postnatally in mice40.

R E V I E W S

10 | JANUARY 2012 | VOLUME 12 www.nature.com/reviews/immunol

© 2012 Macmillan Publishers Limited. All rights reserved

Page 3: The impact of perinatal immune development on mucosal ...manyan/To print/The impact of perinatal immune... · natal development in mice. During the first two weeks of postnatal life,

Peyer’s patch

Mesenteric lymph nodeMesenteric

lymph node

Nature Reviews | Immunology

Epithelial cellproliferation

Production ofantimicrobialpeptides

Commensaldensity andcomplexity

Innateimmunesensing

Level of IgA-producingplasma cells

Lymphocytehoming anddifferentiation

No TLR stimulation Tolerance to TLR stimulation Reactivity to TLR stimulation

Paneth cell-derived peptides and REG3γ

TLR4 expression IκBα expression

Prenatal intestine Neonatal intestine

Birth Weaning

Adult intestine

Dendritic cell

T cell

B cell

CRAMP

CRAMP

TReg cell

TReg cell

Commensalbacterium

IgA-producingplasma cell

Peyer’spatch

IEL

LTi

Cryptopatch

Increasedmucussecretion

ILFILF

Defensin

Maternal SIgA

SIgA

Crypt

CCR9+

α4β7+

Villus

Paneth cellmaturation

Paneth cell

Bacterialcolonization

CX3CR1+

macrophage

Increasedepithelial cellproliferation

M cell

CD103+ DC

Figure 1 | Postnatal development and maturation of the intestinal mucosal barrier and immune system. Both developmental and environmental signals drive significant changes of the intestinal epithelium during the postnatal period in mice and accompany the establishment of an increasingly complex and dense gut microbiota. The neonatal intestinal mucosa is characterized by low levels of epithelial cell proliferation, the absence of crypts and crypt-based Paneth cells, and expression of cathelicidin-related antimicrobial peptide (CRAMP); by contrast, the formation of intestinal crypts late during the second week after birth initiates increased proliferation and rapid epithelial cell renewal, the generation of α-defensin-producing Paneth cells and the upregulation of expression of the antibacterial C-type lectin regenerating islet-derived protein 3γ (REG3γ). A decrease in the level of expression of Toll-like receptor 4 (TLR4) by epithelial cells before birth and a steady increase in the intestinal expression level of the nuclear factor-κB inhibitor IκBα during the postnatal period decrease the responsiveness to bacterial lipopolysaccharide and other pro-inflammatory stimuli. Simultaneously, the acquisition of epithelial TLR tolerance creates a neonatal period of decreased innate immune responsiveness. Note that the human small intestinal epithelium at birth has a much more mature phenotype than in mice. The secondary lymphoid structures of Peyer’s patches and lymph nodes are generated before birth in mice and humans but mature during the postnatal period. By contrast, cryptopatches and isolated lymphoid follicles (ILFs) are formed after birth in mice. Specialized epithelial cells, known as M cells, reside above ILFs and Peyer’s patches and facilitate antigen transport from the lumen to the underlying lymphoid cells. Simultaneously, innate lymphocytes (such as lymphoid tissue inducer (LTi) cells) and T cells leave the liver and thymus, respectively, and colonize the enteric mucosal tissue, including the epithelium. Intraepithelial lymphocytes (IELs) reside in close proximity to the epithelium. Also, increasing numbers of CD103+ dendritic cells and CX

3CR1+ macrophages home to the gut mucosa. In contrast to innate

lymphocytes, regulatory T (TReg

) cells populate the intestinal mucosa in response to bacterial colonization. Although B cells are present in gut tissue during early development, plasma cells producing dimeric IgA are only generated after birth to provide secretory IgA (SIgA) to the lumen. Maternal SIgA is provided by breast milk during the early postnatal period.

RORγt+NKp46+ lymphocytesA population of innate lymphoid cells in the intestinal lamina propria that expresses the transcription factor retinoic acid receptor-related orphan receptor-γt (RORγt) and the natural killer cell marker NKp46. RORγt+NKp46+ lymphocytes support the expression of the antimicrobial peptide REG3γ by epithelial cells through secretion of IL-22.

R E V I E W S

NATURE REVIEWS | IMMUNOLOGY VOLUME 12 | JANUARY 2012 | 11

© 2012 Macmillan Publishers Limited. All rights reserved

Page 4: The impact of perinatal immune development on mucosal ...manyan/To print/The impact of perinatal immune... · natal development in mice. During the first two weeks of postnatal life,

Cryptopatches Clusters of KIT+IL-7R+THY1+ T cell progenitors found in the murine intestinal lamina propria. Cryptopatches are absent in germ-free mice.

Isolated lymphoid folliclesSmall lymphoid aggregates located in the lamina propria of the small and large intestines that contain B cells, dendritic cells, stromal cells and some T cells and that might form germinal centres. In mice, isolated lymphoid follicles were shown to develop from cryptopatches and they are absent in germ-free animals.

AnaphylaxisA severe and rapid allergic reaction triggered by the activation of high-affinity Fc receptors for IgE in sensitized individuals. Anaphylactic shock is the most severe type of anaphylaxis and can lead to death in minutes if left untreated. In various mouse models, it has been shown that IgG1 rather than IgE antibodies trigger the anaphylactic reaction.

In parallel with bacterial colonization, the hom­ing of lymphocytes to the gut mucosa seems to follow defined kinetics that indicate that a series of exogenous and endogenous signals regulates postnatal immune maturation in the intestine. Innate cells — such as lym­phoid tissue inducer (LTi) cells, natural killer (NK) cells, NK­like NKp46+ cells and T helper 2 (TH2)­like cells — migrate during the first 4 weeks after birth from the murine fetal liver to the gut mucosa driven by endog­enous signals41. By contrast, the recruitment of forkhead box protein 3 (FOXP3)+ regulatory T (TReg) cells to the gut mucosa and secretion of the anti­inflammatory cytokine IL­10 have been associated with bacterial colonization42,43, whereby TReg cells help to keep pro­inflammatory TH cells under control to preserve the epithelial barrier44.

A large body of evidence indicates that there are cer­tain ‘windows of opportunity’ for the induction of immu­nological tolerance in the gut and airways, particularly during the perinatal period (although reprogramming of the immune system might, in fact, be a life­long process). Oral tolerance has been best studied in mice and seems to be part of the normal immune maturation process, depending mainly on a finely tuned cross­talk between the innate and adaptive immune systems (in terms of antigen­presenting cells and T cells, respectively), as well as on epithelial barrier integrity45.

The role of TReg cells. TReg cells with immunosuppres­sive properties are abundant in human fetal MLNs46, and their homing to the gut mucosa seems to be particularly active in infancy47. Mechanistically, it is thought that CD103+ migratory dendritic cells (DCs) carry antigen from the gut to MLNs where they promote the induc­tion of TReg cells, particularly in the prescence of retinoic acid and TGFβ. In addition, subepithelial non­migratory CD103–CX3CR1+ macrophage­like cells produce IL­10, which supports proliferation of the TReg cells when they have homed to the lamina propria48. The increasing prevalence of allergic disorders in infancy indicates that the underlying immune dysregulation is probably an early event that compromises the function of immature antigen­presenting cells and TReg cells49,50.

Bifidobacterium infantis, a prominent member of the gut microbiota in human infants, was shown in mice to markedly induce FOXP3+ TReg cells51. Notably, neonatal CD4+ T cells in mice are prone to differentiate into TReg cells following stimulation52, as are human cord blood cells, probably as a result of perinatal exposure to mater­nal progesterone53. Later in development, members of the Clostridium cluster IV and XIVa might take over the role of B. infantis in promoting the local induction of TReg cells in the colon42. Also, Bacteroides fragilis seems to have unique TReg cell­inducing and epithelium­ associating properties. Conversely, pro­inflammatory TH17 cells are strongly promoted in the murine gut by segmented filamentous bacteria (SFB)54,55. These bac­teria, which attach to the mucosa particularly in the distal ileum, are mainly seen in mice after weaning56. It is currently debated whether equivalents of mouse SFB are found in the human gut.

The role of humoral immunity. Humoral immunity con­tributes to the establishment of an adequate postnatal epithelial barrier at mucosal surfaces. By reinforcing the epithelial barrier, secretory IgA (SIgA) inhibits inap­propriate immune activation by microorganisms and antigens in the lumen of the intestinal and respiratory tracts. A large proportion of commensal microorganisms in the upper aerodigestive tract57 and gut58 is coated with SIgA59. This bacterial coating might be explained by low­affinity cross­reacting antibodies as well as by the high glycan content of SIgA60. SIgA at the epithelial surface restricts colonization of microorganisms and the penetra­tion of agents that could potentially cause hypersensitivity reactions or infection.

Neonates that are not breast­fed lack reinforcement of the gut barrier by maternal SIgA61. Plasma cells that produce IgA are generally undetectable in human lamina propria before 10 days of age and only traces of endogenously synthesized luminal SIgA and some SIgM are found after birth45. The postnatal prolifera­tion of intestinal IgA+ plasma cells is highly variable, and in affluent societies it can take several years for the size of the IgA­producing plasma cell population to reach that of healthy adults62. By contrast, a rela­tively fast postnatal increase in the production of SIgA can be seen in children living in developing countries with a heavy microbial load63. Similarly, the number of intestinal IgA+ plasma cells normalizes 4 weeks after colonization of germ­free mice with commensal micro­organisms64. Interestingly, pioneering studies in mice showed that the microbiota stimulates a self­limiting SIgA response in the gut65. Such transient SIgA produc­tion is probably necessary to allow access of microbial constituents to gut­associated lymphoid tissue. In this manner, it seems that the intestinal IgA response is continuously adapting to the changing microbiota66, which would be particularly important in the early postnatal period.

Similar to the induction of TReg cells, the establish­ment of SIgA­mediated immunity also depends on environmental conditions. For example, the differentia­tion of IgA+ plasma cells depends on retinoic acid, so an optimal intestinal immune response requires adequate supply of vitamin A67.

The role of intestinal barrier reinforcement. The epithelial glycoprotein polymeric immunoglobulin receptor (pIgR; also known as membrane secretory component) translocates SIgA and SIgM to the lumen and is therefore essential for the barrier function of the intestinal and respiratory mucosae68. Mice deficient for pIgR lack secretory­type antibodies69 and have aberrant mucosal ‘leakiness’ and excessive uptake of food pro­teins, as well as of components of commensal bacteria, from the gut lumen69,70. This results in a hyper­reactive state and predisposition to anaphylaxis after systemic sensitization71. Interestingly, pIgR­deficient mice have increased induction of TReg cells after continuous feed­ing of a soluble dietary antigen; this enhanced estab­lishment of oral tolerance was shown to efficiently control IgG1­ and T cell­dependent hypersensitivity71.

R E V I E W S

12 | JANUARY 2012 | VOLUME 12 www.nature.com/reviews/immunol

© 2012 Macmillan Publishers Limited. All rights reserved

Page 5: The impact of perinatal immune development on mucosal ...manyan/To print/The impact of perinatal immune... · natal development in mice. During the first two weeks of postnatal life,

Tight junctions These are specialized intercellular junctions that seal the apical epithelium, in which two plasma membranes form a sealing gasket around a cell (also known as the zonula occludens). They are formed by several proteins, including occludin and claudin. Tight junctions prevent fluid moving through the intercellular gaps and prevent the lateral movement of membrane proteins between the apical and basolateral cellular domains.

Goblet cells Mucus-producing cells that are found in the epithelial cell lining of the intestines and lungs.

Clara cellsDome-shaped cells with short microvilli that are found in the small airways (bronchioles) of the lungs. These cells can secrete glycosaminoglycans to protect the lining of the bronchioles and are also known as bronchiolar exocrine cells.

In adddition, decreased integrity of the structural bar­rier as a result of mild or transient breaching of the tight junctions between intestinal epithelial cells in mice was shown to induce an anti­inflammatory TReg cell response in the gut72. So, a predisposition to hyper­sensitivity might be compensated for by the enhanced induction of intestinal TReg cells that promote tolerance to dietary antigens.

These studies are relevant for newborn human infants, who have a leaky gut epithelium45. Food allergy is much more prevalent in young children than in adults, but many children with food allergy outgrow their disorder before 3 years of age, particularly those with non­IgE­mediated allergy to cow’s milk45. In these allergic children, and also in those who outgrow their IgE­mediated cow’s milk allergy, the expansion of TReg cell populations with suppressive properties has been observed73,74. This indicates that a leaky neonatal gut epithelium and the concurrent microbial colonization might provide a ‘window of opportunity’ for oral tol­erance induction by continuous intestinal exposure to small amounts of dietary antigen.

Of note, there are significant differences in the ‘tight­ness’ of the epithelial barrier between mice and humans. In general, the gut mucosa shields the systemic immune system from exposure to constituents of commensal bacteria to a greater extent in mice than in humans75. This is, however, not true for the immediate postnatal period. The so called ‘gut closure’ — the establishment of a mature epithelial barrier that prevents excessive influx of macromolecules across the intestinal epithe­lium — develops during the first days after birth in humans but only with weaning in mice. Thus, some of the mechanisms that have been proposed for oral tol­erance induction in mice, such as neonatal Fc receptor (FcRn)­mediated uptake of IgG–antigen complexes from the gut lumen, are probably of little or no importance in humans.

Dietary impact on perinatal gut immunity. Food constituents also contribute to the development of the intestinal immune effector compartments, both in mice and humans76,77. Breast milk reinforces mucosal defences not only by providing SIgA (and SIgM) anti­bodies but also by delivering immune cells, cytokines, growth factors and high concentrations of oligosac­charides that promote the proliferation of lactic acid­ producing bacteria, which are a major beneficial fraction of the neonatal intestinal microbiota61. Indeed, a recent meta­analysis showed that breast­feeding has a protective effect on the development of inflammatory bowel disease later in life78.

Both breast milk and the maternal microbiome are transmitted to the neonate, but it is still unclear how the nutritional status of the mother affects the qual­ity of her milk and the composition of her micobiota. It is known that intestinal microbial communities can be reshaped postnatally in response to changes in diet. Considerable efforts are therefore being made to examine the inter actions between food and food ingredients, the microbiota, the immune system and health79.

Vitamin A is a well­known dietary constituent that supports the mucosal milieu and facilitates the estab­lishment of the neonatal immune system. The vitamin A derivative retinoic acid is required for the expression of gut­homing molecules on T and B cells (such as integ­rin α4β7 and CC­chemokine receptor 9 (CCR9))40, for the induction of TReg cells and for IgA class switching in both mice and humans40. Thus, an adequate supply of vitamin A is crucial for intestinal immune homeostasis.

Taken together, the findings discussed above show that early postnatal events might significantly influ­ence priming of the mucosal immune system and the establishment of a life­long immune homeostasis.

Perinatal development of the airway mucosaThe role of neonatal microbiota in airway immunity. Microbial colonization of the upper respiratory tract, including the nose, mouth and throat, occurs rap­idly after birth. Until recently, the lower airways were thought to remain sterile, but recent findings have shown that microbial colonization also occurs in this compartment9,10 (FIG. 2). As PRRs recognize micro­bial compounds and subsequently regulate immune responses to pathogens and commensals, it is thought that TLR expression is crucial for the functional devel­opment of the respiratory mucosa (FIG. 2). In the fetal mouse lung, TLR2 and TLR4 are first expressed during the last trimester of pregnancy (from the late pseudo­glandular to terminal saccular stages), and expression levels increase further after birth80. In humans, a com­parable expression pattern was detected in the lung between early pseudoglandular and canalicular stages of fetal development (with the highest upregulation of TLR2 expression occurring between days 60 and 113 of gestation)81. These TLRs are expressed by non­immune cells of the lung as well as by immune cells. Indeed, TLR4 expression by parenchymal lung cells is important for the normal development of lung elasticity and for air­space development, as shown in TLR4­deficient mice reconstituted with bone marrow from wild­type mice82.

What triggers prenatal TLR expression in the res­piratory mucosa? It seems possible that microbial com­ponents, circulating in the maternal blood, cross the placental barrier to reach the amniotic fluid. Swallowing of amniotic fluid by the foetus could allow direct contact of such molecules with the developing respiratory sur­face. Although this is an attractive concept, so far there is no direct evidence in favour of this mechanism for the induction of prenatal TLR expression. Alternatively, an intrinsic developmental programme could be responsi­ble for TLR expression. In either case, the presence of basal levels of TLR expression at birth is important to appropriately deal with environmental microorganisms that are inhaled with the first breath of life.

The respiratory mucosal barrier. The airway epithelium is the first barrier layer to encounter immunostimulatory exogenous components at birth. Within the airway epi­thelium, ciliated columnar cells, mucus­secreting goblet cells and surfactant­secreting Clara cells are connected by tight junctions to form an impermeable but dynamic

R E V I E W S

NATURE REVIEWS | IMMUNOLOGY VOLUME 12 | JANUARY 2012 | 13

© 2012 Macmillan Publishers Limited. All rights reserved

Page 6: The impact of perinatal immune development on mucosal ...manyan/To print/The impact of perinatal immune... · natal development in mice. During the first two weeks of postnatal life,

Nature Reviews | Immunology

DCEpithelial cell

Mucus

Bact

eria

lco

loni

zati

onTL

R e

xpre

ssio

nD

C n

umbe

rIL

-4IF

IFN

αIL

-10

Prenatalairways

Neonatalairways

Adultairways

TH2 cell

IL-4

Commensalbacterium

a Homeostasis c

b Chronic allergic inflammation of the respiratory mucosa

TH1 cell TReg cell TH17 cell

IL-10

Mucosaltolerance

TSLPIL-25IL-33

Altered densityand diversity ofcommensal bacteria

Delayed maturationof TH1 cells

Diminishedantiviral response

Disturbed tolerance

iBALT formation

Persistenceof TH2 cells

Homeostasis Chronic allergic inflammation

Altered bacterial colonization

Altered TLRexpression

IncreasedDC number

IFNγ TGFβ IL-17F

IncreasedDC turnoverand activation

TH2 cell

Mast cell

TLR

IL-5

ECP

IL-4IL-13

IL-4IL-5IL-9IL-13

Altered epithelial cell barrier function

Eosinophil

Neutrophil

IgE-producingplasma cell

Birth Weaning

VirusVirus

Virus Virus

Aeroallergens

Figure 2 | Development of the respiratory mucosal immune system. The establishment of airway homeostasis (a) or the development of chronic airway inflammation such as asthma (b) depends on early events that affect the maturation of the local immune system (c). Microbial colonization of the lower airways starts after birth (through inhalation). Maternal microbial exposure during pregnancy has an impact on prenatal immune programming (a and c). The expression of Toll-like receptors (TLRs) by airway epithelial cells starts during prenatal life and is further increased during postnatal life, influenced by exposure to microorganisms (c). TLR-triggered signalling cascades contribute to the development of the local dendritic cell (DC) network, which is not fully developed at birth (a and c). Exposure to high levels of TLR ligands favours the development of normal immune responses. The perinatal period is characterized by relatively high levels of expression of T helper 2 (T

H2) cell-associated

cytokines, such as interleukin-4 (IL-4), which are rapidly down-regulated early in life under homeostatic conditions (c). T

H1 cell responses, as

indicated by interferon-γ (IFNγ) secretion, develop following exposure to environmental factors, such as TLR agonists. In parallel, IL-10 secretion steadily increases after birth, leading to the development of mucosal

tolerance (a and c). Lack of and/or altered prenatal maternal exposure to certain environmental microorganisms, Caesarean section and altered microbial colonization and diversity contribute to the development of asthma (b and c). These events result in altered TLR expression (c). Low-dose exposure to TLR agonists favours the development of T

H2

cell-mediated immune responses (b and c). TH2 cell responses are further

supported by the intrinsic adjuvant-like activities of certain aeroallergens, such as house dust mite proteins, leading to altered barrier function (b). TLR signalling cascades lead to the production of thymic stromal lymphopoietin (TSLP), IL-25 and IL-33 by airway epithelial cells, which support DC maturation and function (b). The chronic inflammatory immune response leads to the formation of induced bronchus-associated lymphoid tissue (iBALT), which participates in the initiation, maintenance and amplification of allergic inflammation in the respiratory mucosal tissue (b). This results in postnatal persistence of the prenatally developed T

H2-like

immune responses. Moreover, TH1 cell responses are markedly suppressed

and/or delayed, and antiviral responses (IFNα) are diminished (c). ECP, eosinophil cationic protein; TGFβ, transforming growth factor-β, T

Reg cell, regulatory T cell.

R E V I E W S

14 | JANUARY 2012 | VOLUME 12 www.nature.com/reviews/immunol

© 2012 Macmillan Publishers Limited. All rights reserved

Page 7: The impact of perinatal immune development on mucosal ...manyan/To print/The impact of perinatal immune... · natal development in mice. During the first two weeks of postnatal life,

barrier. Under inflammatory conditions, the permeabil­ity of the barrier is increased. The epithelial cells provide several nonspecific defence mechanisms, including the secretion of surfactants, complement products, anti­microbial peptides and mucins2. It is now well recognized that the airway epithelium has an active role both in the initiation, maintenance and resolution of inflammatory mucosal responses, and in the repair of the respiratory epithelium, through the production of cytokines such as IL­25, IL­33 and thymic stromal lymphopoietin83,84. In contrast to our more detailed knowledge of the expres­sion of antimicrobial peptides in the gut early in life, we have only little information about their expression pat­tern in the airways. Young infants express β­defensins and the cathelicidin LL37 and, although data are lacking regarding prenatal and early postnatal expression and function of these molecules, it is probable that there are no substantial differences between the mucosal systems of the gut and the airways in this regard85.

Perinatal maturation of the immune system. The organization of mucosa­associated lymphoid tissue has a distinct pattern in the bronchus compared with the gut. Classical bronchus­associated lymphoid tissue (BALT) — as defined by B cell follicles and follicle­associated epithelium, with so­called membrane or microfold (M) cells86,87 — appears during late embry­onic development in some species, but not in humans and mice88,89. By contrast, the evidence indicates that de novo organization of lymphoid tissue is induced in murine and human lungs by the encounter of antigens in the airways during infections and inflammation90,91. It is probable that DCs, at least in part, compensate for the lack of BALT in healthy human and mouse lungs in terms of immunological surveillance of the entire air­way surface. Because the local DC network in the lungs develops with increasing amounts of antigen exposure, it is not surprising that DCs are absent or only present at low numbers before birth and continuously increase in number from birth onwards (FIG. 2). Therefore, at the time of birth in humans and mice, the respiratory mucosal barrier is only partly equipped to deal with environmental antigens, including bacteria, viruses and environmental allergens. Furthermore, indoor and outdoor air­pollutants, such as environmental tobacco smoke (ETS), have a marked effect on the development of perinatal immune functions (see below).

As mentioned earlier, TLR expression is tightly regu­lated in the perinatal respiratory tract, and this affects the maturation of the adaptive mucosal immune system. A high­throughput study in humans of immune responses to a panel of TLR ligands in a cohort that was screened over the first 2 years of life showed that TLR ligation induces distinct T cell responses dependent on age92. The levels of TH1­type cytokines that are induced in response to TLR ligation are low at birth but increase gradually with age. The induction of interferon­α (IFNα), which supports antiviral defences, reaches adult levels by 1 year of age. A similar progressive increase in the induction of IL­1β, tumour necrosis factor (TNF), IL­6 and IL­12 with age was also found4,93. By contrast, IL­10

production steadily decreases from birth onwards, and the levels of IL­6 and IL­23 (which support the devel­opment of TH17 cells) peak around the time of birth. These data indicate that the maturation of effector T cell responses occurs in an age­dependent manner under the control of mucosal TLR­mediated signalling (FIG. 2). This maturation process is particularly important to acquire the optimal level of TH1 and TH17 cells, which have a crucial role in defence against many pathogens. Furthermore, this response is required to suppress TH2 cells, the levels of which are increased at birth.

In conclusion, the regulation of TLR expression and function in the respiratory tract is closely associated with the development of innate and adaptive immune responses. Although microbial exposure clearly con­tributes to the development of normal mucosal immune responses, TLR expression in the respiratory mucosa occurs well before birth.

Perinatal tolerance induction to allergens. It has long been known that exposure to aerosolized allergens (for example, ovalbumin) results in allergen­specific immune tolerance94. Further studies have shown that this toler­ance can be transferred from the mother to her offspring if antigen exposure occurs during pregnancy95, but also if the mother has been tolerized before pregnancy96. The effects of placental transfer of IgG antibodies have been extensively studied, and the results indicate that maternal antibodies can inhibit T cell responses in the offspring in an antigen­specific manner95. Furthermore, placental transfer of allergens and antigens into the amniotic fluid, as well as the migration of maternal innate and adaptive immune cells, further facilitates immune development in the foetus97. As a result of prenatal intra­uterine expo­sure to antigens, antigen­specific T cell responses of the neonate are readily detectable in cord blood. It is not yet clear whether this reflects tolerance development and/or affects the development of allergic sensitization and TH2­type responses in the developing infant98,99.

Moreover, it has been shown in mice that the pre­natal establishment of tolerance that protects from allergic asthma continues into the postnatal period through breast milk­mediated transfer of allergens100. Also, alveolar epithelial cells can present inhaled antigens to T cells and promote the development of FOXP3+ TReg cells101; they can also produce IL­10, which induces the development of IL­10­producing TReg cells. Thus, the development of tolerance against environmen­tal allergens is a continuous process that starts in prenatal life. Tolerance­inducing mechanisms operating in the placenta during fetal life are supplemented from birth onwards by local immunoregulatory events that occur in the respiratory mucosa.

Pathology of the gut mucosaImmunological hypersensitivity to food antigens. Several of the above­mentioned environmental factors influence the development of food allergies through modulating perinatal mucosal immunity. For exam­ple, birth by Caesarean section is associated with an altered intestinal microbiota in the offspring, who do

R E V I E W S

NATURE REVIEWS | IMMUNOLOGY VOLUME 12 | JANUARY 2012 | 15

© 2012 Macmillan Publishers Limited. All rights reserved

Page 8: The impact of perinatal immune development on mucosal ...manyan/To print/The impact of perinatal immune... · natal development in mice. During the first two weeks of postnatal life,

not encounter maternal faecal–vaginal commensal microorganisms during delivery25,102,103. As a result, this form of delivery increases the risk of food allergy and coeliac disease104,105 (BOX 1), probably because the result­ing ‘inadequate’ neonatal gut microbiota abrogates immune regulation. Interestingly, a small but signifi­cant decrease in the development of allergies was noted after perinatal probiotic intervention in children deliv­ered by Caesarean section106. Accordingly, it is possible that improved microbial intervention might be available in the future to rectify inadequate gut colonization, as observed after Caesarean section. However, other efforts to enhance oral tolerance by reinforcing the gut micro­biota through probiotic treatment have had few convinc­ing health benefits, even in children with a hereditary risk of food allergy. For example, no general inhibitory effect on the incidence of allergy was observed by the age of 5 years after perinatal probiotic intervention, which aimed to prevent TH2 cell­driven IgE­mediated allergy106. Thus, better molecular characterization of the tolerogenic components of the intestinal microbi­ota will be required for the advancement of microbial intervention as a preventive strategy.

Interestingly, there might be a link between die­tary lipids, such as the long­chain omega­3 fatty acids and short­chain fatty acids (SCFAs), and susceptibil­ity to food allergies. Consumption of fish oil, which is enriched in polyunsaturated omega­3 fatty acids, might protect against the development of food allergies by decreasing immune cell recruitment and the production of pro­inflammatory cytokines in the gut107. However, a positive correlation between maternal omega­3 fatty acid intake and the infant’s protection from childhood asthma — which often accompanies severe food allergy — has not been shown in every study108,109. In addi­tion, SCFAs, which are produced by gut bacteria as a

byproduct of the fermentation of dietary fibres, have sev­eral health­promoting effects, in particular by strength­ening the intestinal epithelial barrier110. Indeed, food allergy in early childhood is associated with relatively low faecal levels of SCFAs, possibly as a result of delayed maturation of the intestinal microbiota111.

Lack of mucosal homeostasis in IBD. Numerous mouse models of IBD have identified a crucial role for both the gut microbiota and immune cells in its pathogenesis (FIG. 3). Despite the identification of disease­associated polymorphisms in genes that contribute to the intestinal epithelial barrier and immune homeostasis — such as nucleotide­binding oligomerization domain 2 (NOD2), autophagy­related 16­like 1 (ATG16L1) and IL­23 recep­tor (IL23R) — the clinical manifestations of IBD are usually only seen in late infancy or early adulthood. By contrast, patients with IL­10 or IL­10R deficiency suf­fer from early onset colitis112. Thus, except in the case of total deficiency of IL­10­mediated signalling, the infant gut mucosa seems to have a greater capacity to control inappropriate immune responses than the adult mucosa, an ability that might be lost during repeated mucosal challenges later in life.

Moreover, perinatal exogenous factors might contrib­ute to the manifestation of overt disease symptoms later in life. These might be associated with postnatal envi­ronmental and microbial exposures, as children born by Caesarean section have recently been shown to have a slightly increased risk of developing Crohn’s disease113.

Strikingly, certain alterations of the microbiota compo­sition might be sufficient by themselves to cause mucosal inflammation, as the co­housing of animals that are genetically susceptible to colitis with neonatal or adult wild­type mice can lead to intestinal inflammation in the healthy immunocompetent animals114,115. The ‘coli­togenic’ potential of an altered microbiota might be of particular importance in the neonatal and infant gut, where the immature microbiota is unstable and susceptible to colonization by additional bacteria.

Another interesting observation is that mice carry­ing a mutation in the IBD­associated gene Atg16l1 only develop epithelial damage and enteric disease after infec­tion with norovirus116. A specific infection, potentially during the particularly susceptible postnatal period, might therefore alter the gut microbiota and/or lead to histological changes, both of which could trigger disease manifestation later in life in individuals with a genetic susceptibility. Thus, both infections and microbiota composition can contribute directly or indirectly to chronic inflammation25,68,117.

Necrotizing enterocolitis of the newborn. A striking example of the detrimental consequences of dysregu­lated perinatal maturation of the mucosal immune sys­tem is necrotizing enterocolitis (NEC) — a disease that is observed in human pre­term neonates118 and is charac­terized histologically by massive epithelial cell apoptosis. NEC has been associated with postnatal bacterial coloni­zation. In animal models, altered epithelial TLR4 signal­ling has been linked to impaired stem cell proliferation

Box 1 | Immune dysregulation in coeliac disease

In certain parts of the world, coeliac disease is becoming more common, and both genetic and environmental factors are involved in the pathogensis. Central to the disease is T helper 1 (T

H1) cell-driven intestinal hypersensitivity to the

immunodominant gliadin peptides in wheat gluten, or to similar prolamins in other types of cereal grain. The disease usually presents as malabsorption during childhood. The innate immune signals that cause the break in immune tolerance to prolamins and that initiate disease remain undefined170. Without a prolamin-restricted diet, coeliac disease is a life-long disorder. In some patients, clinical symptoms do not occur until late in adulthood; this might reflect the possibility that initiating events, for example caused by repeated infections of the upper gastrointestinal tract, need to accumulate over time to break tolerance to prolamins.

Extensive work has been carried out to identify other genetic susceptibility loci for coeliac disease, in addition to the well-defined HLA-DQ2 (or less often HLA-DQ8) MHC class II alleles, which mediate presentation of the proline-rich prolamin peptide epitopes to T

H1 cells. Several studies have linked hereditary single nucleotide

polymorphisms (SNPs) in genes encoding various immune factors or innate immune sensors both with coeliac disease171 and with other types of hypersensitivity to food or aeroallergens172,173. This implies that aberrant innate immune responses are a crucial factor in the postnatal delay or abrogation of tolerance to food and other environmental allergens. Interestingly, some of the non-HLA risk loci for coeliac disease are shared with autoimmune systemic disorders such as rheumatoid arthritis171, which supports the idea that tolerogenic responses induced in the gut can disseminate beyond the gastrointestinal tract.

R E V I E W S

16 | JANUARY 2012 | VOLUME 12 www.nature.com/reviews/immunol

© 2012 Macmillan Publishers Limited. All rights reserved

Page 9: The impact of perinatal immune development on mucosal ...manyan/To print/The impact of perinatal immune... · natal development in mice. During the first two weeks of postnatal life,

Nature Reviews | Immunology

TReg cellTReg cell

IL-10TGFβ

IL-10TGFβ

TSLPTGFβRA

TSLPTGFβ

IL-6IL-1

IL-6IL-12IL-23

TSLPTGFβRA

TSLPTGFβ

Tolerogenic CD103+ DC

CX3CR1+

macrophageCX3CR1+

macrophage

IgA-producing plasma cell

IgA-producing plasma cell

Neutrophils

REG3γ

REG3γ

Defensins

IL-17A/FIFNγ

NKp46+

RORγt+

lymphocyte

Paneth cell

BAFFAPRILRA

IL-22

TGFβRA

a Intestinal mucosal homeostasis

Commensalbacterium

IAPPGRP2

Intraepitheliallymphocyte

CD4+ T cell

SIgA

Mucus

SIgA

IgG TH1 orTH17 cell

Tissuedamage

Defensins

TH1 orTH17 cell

b Chronic inflammation of the intestinal mucosa

IgG-producing plasma cell

Figure 3 | The cellular network of gut immune homeostasis and the cellular processes driving mucosal inflammation. a | Several mechanisms maintain microbial–host homeostasis in mucosal tissues. The production of dimeric IgA by plasma cells followed by its epithelial transport into the gut lumen as secretory IgA (SIgA) is enhanced by retinoic acid (RA), B cell-activating factor (BAFF) and a proliferation-inducing ligand (APRIL); SIgA limits the epithelial translocation of antigen and bacteria. The mucus layer and constitutive α-defensin production shield the epithelial surface from direct microbial exposure. In addition, interleukin-22 (IL-22) secretion by NKp46+RORγt+ innate lymphocytes, CD4+ T cells and CD103+ dendritic cells (DCs) promotes epithelial production of regenerating islet-derived protein 3γ (REG3γ). Inappropriate immune activation is controlled by the enzymatic degradation of stimulatory microbial constituents by intestinal alkaline phosphatase (IAP) or the amidase peptidoglycan recognition protein 2 (PGRP2). Epithelial cell-derived regulatory factors, such as thymic stromal lymphopoietin (TSLP), transforming growth factor β (TGFβ) and retinoic acid, induce tolerogenic CD103+ DCs that in turn promote regulatory T (T

Reg) cell development. Both

TReg

cells and CX3CR1+ macrophages control pro-inflammatory T helper 1 (T

H1) and T

H17 cells through the production of

TGFβ and IL-10. b | Microbial infection or tissue damage involving disruption of the epithelial barrier stimulates the release of antimicrobial peptides and abrogates the tolerogenic properties of CD103+ DCs by inducing their secretion of IL-6, IL-12 and IL-23. Together with IL-6 and IL-1 secretion by activated CX

3CR1+ macrophages, this promotes the emergence of

interferon-γ (IFNγ)- and IL-17-producing TH1 and T

H17 cells, which in turn stimulate a pro-inflammatory reaction involving

the infiltration of neutrophils and IgG-producing plasma cells, tissue destruction and, potentially, organ dysfunction.

and crypt–villus migration and decreased goblet cell differentiation119. Interestingly, oral administration of Gram­positive probiotic bacteria has been shown to decrease the incidence of NEC120. Although NEC does not lead to chronic inflammation later in life (after the affected gut tissue has been surgically removed), it

illustrates the requirement of a mature, fully differen­tiated mucosal tissue for the establishment of mucosal homeostasis during postnatal microbial colonization. Less pronounced disturbances of mucosal maturation might induce similar processes and contribute to chronic inflammation later in life.

R E V I E W S

NATURE REVIEWS | IMMUNOLOGY VOLUME 12 | JANUARY 2012 | 17

© 2012 Macmillan Publishers Limited. All rights reserved

Page 10: The impact of perinatal immune development on mucosal ...manyan/To print/The impact of perinatal immune... · natal development in mice. During the first two weeks of postnatal life,

Pathology of the respiratory mucosaThe role of commensals in allergy and asthma develop-ment. Neonatal intestinal colonization with an altered microbiota can influence susceptibility to allergy and asthma, and the mechanisms involved are currently under investigation (TABLE 1). Differences in the com­position of the gut microbiota between atopic or aller­gic and non­atopic or non­allergic infants have been described in several models; for example, lower counts of Lactobacillae, Bifidobacteriae and Bacteroides spe­cies, together with higher counts of Clostridium dif-ficile, were found in atopic infants121–125. In keeping with this influence of the gut microbiota, the epide­miological association between the disappearance of Helicobacter pylori from the human stomach and the increasing incidence of allergic asthma in Western soci­eties has recently been verified as causal in an allergen­ induced mouse model126. Of note, the accumulation of TReg cells in the lungs and protection against allergic asthma was most robust in mice infected neonatally with H. pylori. It is important to note that most of these studies used classical culture­based techniques to monitor microbiota composition. It is expected that the use of non­culture­based techniques to quali­tatively and quantitatively assess the gut microbiome will shed new light on the relationship between the gut microbiota and allergy.

The effect of intestinal commensals on asthma devel­opment has been elegantly illustrated in mice: depletion of the gut microbiota by antibiotic treatment exacer­bated experimental asthma that is driven by the TH2 cell­associated cytokine IL­13 (REF. 127). Moreover, a recent study provided a molecular link between diet, gastrointestinal bacterial metabolism and asthma devel­opment128. SCFAs, which are produced through the fermentation of dietary fibres by the intestinal micro­biota (namely, Bifidobacteriae and Bacteroides species), bind to G­protein­coupled receptor 43 (GPR43); in the absence of this interaction — as in Gpr43–/– mice — exacerbation of experimental asthma was observed. This was related to an increased production of pro­inflammatory mediators and increased immune cell recruitment in the airways.

Finally, the (uro)genital microbiome of the mother might be involved in protection against asthma in her offspring. This hypothesis is receiving increased atten­tion owing to the observation of a higher risk of asthma in children delivered by Caesarean section129–131.

Environmental microbial exposure. In addition to the microorganisms that colonize the mucosal surfaces, the inhalation of airborne environmental microorganisms can also trigger mucosal immune maturation (FIG. 2). Early­life exposure to the unique environment of tra­ditional farming was found to significantly decrease the risk of developing respiratory allergies (reviewed in REF. 132). The high microbial load that is present in the farm environment is strongly associated with increased production of IFNγ and TNF at birth, but was found to have no effect on the levels of TH2­type cytokines or on the production of IL­10 and IL­12. The data regard­ing IgE production by these children after birth or dur­ing early childhood are inconsistent133,134; however, an inverse relationship between neonatal IgE production and the level of cord blood IFNγ was noted135, which further supports the concept that early development of a strong TH1­type immune response is an important mechanism to suppress the initial polarization of T cells towards a TH2­type response and to further inhibit the development of TH2 cell­mediated allergies.

Exposure to environmental microorganisms during early life has a large effect on the level of TLR expression, as indicated by increased levels of Tlr5 and Tlr9 mRNA at birth in exposed compared with non­exposed newborn mice (FIG. 2). When microbial exposure continues through early childhood, the effects are even greater, as shown in the case of natural exposure to endotoxin136 derived from mattress dust. Moreover, the higher the number of differ­ent farm animal species that a mother had contact with during pregnancy, the higher are the levels of TLR2, TLR4 and CD14 expression in her offspring at school age137.

Furthermore, it has been shown that mucosal admin­istration of natural or synthetic TLR agonists — such as lipopeptides (which stimulate TLR2)138, peptidoglycan (TLR2 and TLR4)139, LPS (TLR4)140,141, polyinosinic–polycytidylic acid R848 (TLR3 and TLR7)142 and DNA

Table 1 | Microorganisms affect the development of experimental and clinical asthma

Source and route of microbial exposure

Examples of bacteria Effect on asthma

Environmental microorganisms

• Acinetobacter lwoffii• Eurotium spp.• Penicillium spp.

• Transgenerational protection from experimental asthma• Asthma-protected children are exposed to a high level of

bacteria and fungi/spores103,146

Respiratory microbiota • Proteobacteria• Bacteroides spp.

• Altered microbial colonization pattern of the respiratory mucosa in asthmatics and patients with COPD9

Intestinal microbiota • Lactobacillae• Bifidobacteriae• Bacteroides spp.• Clostridium spp.

• Altered quantitative distribution pattern of certain culturable microbial strains in young asthmatic patients121–125

• Depletion of gut microbiota exacerbates experimental asthma127

Urogenital microbiota • Vaginal microorganisms • Indirect evidence from a model of Caesarean section; cause–effect relationship needs to be established129–131

COPD, chronic obstructive pulmonary disease.

R E V I E W S

18 | JANUARY 2012 | VOLUME 12 www.nature.com/reviews/immunol

© 2012 Macmillan Publishers Limited. All rights reserved

Page 11: The impact of perinatal immune development on mucosal ...manyan/To print/The impact of perinatal immune... · natal development in mice. During the first two weeks of postnatal life,

from Bordetella pertussis (TLR9)143 — triggers a cascade of immune responses that results in protection from experimental asthma in several murine models. In most of these studies, the absence of a TH2 cell­mediated immune response correlated with the development of a strong TH1­type immune response or increased levels of IL­12 and IL­10, further supporting the concept that TLR sig­nalling favours the development of a TH1 cell­mediated immune response (FIG. 2). Further studies in mice have provided evidence that a similar asthma­protective effect could be achieved by intranasal application of intact Gram­negative or Gram­positive bacteria144.

When infants are exposed to TLR ligands, their response pattern at the level of adaptive immunity dif­fers markedly between healthy and allergic infants10,15; for example, allergic children who were exposed to TLR ligands at a young age had decreased production of pro­inflammatory cytokines (such as IL­1β, TNF and IL­6)145. Moreover, they had a markedly delayed matura­tion of TH1 cell functions (FIG. 2), which is, at least in part, responsible for the sustained and high TH2 cell response profile of allergic individuals (for example, owing to lack of IFNγ production).

These proof­of­principle experiments carried out in adult animals provide the foundation for the explora­tion of transmaternal asthma­protective effects. Several asthma­protective bacterial strains have been identi­fied, but modulation of the asthmatic phenotype in mice showed that there are strain­dependent qualitative and quantitative differences, with the highest degree of protec­tion so far observed by Acinetobacter lwoffii146. Intranasal administration of A. lwoffii to mice throughout pregnancy results in a strong and long­lasting protection from air­way inflammation, airway hyper­responsiveness and IgE production in their offspring. Moreover, it augmented TLR expression in the maternal lung, resulting in mild local and systemic inflammation, as marked by increased expression of IL­6, TNF and IL­12p40. This maternal mucosal TLR signalling was responsible for the initia­tion and eventual transmission of the asthma­protective effect to the next generation, as TLR­sufficient offspring of mothers who are deficient for TLR2, TLR3, TLR4, TLR7 and TLR9 were not protected from asthma146. Thus, TLR expression and signalling in the maternal airway mucosa are crucially involved in generating the asthma­protective immune responses that operate in the offspring. Although the downstream events could be linked to a low­level inflammatory immune response, the detailed molecu­lar and cellular events involved remain to be elucidated. Future studies must be carried out to determine the con­tribution of individual bacterial strains as opposed to the overall microbial burden and diversity in this regard103.

Perinatal tobacco smoke exposure. Environmental tobacco smoke is a major component of indoor air pollution. There is a strong association between maternal smoking during pregnancy, or postnatal ETS exposure, and a higher risk of asthma147,148. Studies in mice and monkeys show that intra­uterine ETS exposure results in decreased lung devel­opment and an accelerated decline in lung function149 — with increased thickness of the smooth muscle layer and

collagen type III deposition around the airways — of the offspring150,151. Moreover, the expression of genes involved in WNT signalling was induced in the offspring of mice exposed to cigarette smoke during pregnancy152. These data imply that signalling pathways involved in inflam­mation and lung cancer development contribute to the detrimental effects of prenatal ETS exposure.

The development of the mucosal immune system in neonates is strongly affected by ETS exposure during preg­nancy. Cells from neonates born to mothers who smoke have an impaired response to TLR ligands, resulting in the decreased production of pro­inflammatory cytokines by antigen­presenting cells153. This involves decreased produc­tion of TNF in response to TLR2, TLR3 and TLR4 signal­ling, decreased IL­6 production in response to TLR2 and TLR9 stimulation, and decreased IL­10 levels after TLR2 activation. These results are in line with previous reports showing that altered neonatal cytokine responses are asso­ciated with maternal smoking in pregnancy154, and the decreased production of IL­10 by DCs of babies exposed to ETS in the first five months of life155. Moreover, there are some indications that levels of IFNγ might be decreased in the offspring of mothers who smoke during pregnancy153. The absence of IFNγ production might contribute to the higher levels of TH2­type cytokines observed following antigen challenge in neonates154. Indeed, prenatal ETS exposure exacerbates experimental lung inflammation in mice through the induction of a strong TH2 cell response in the lung marked by increased levels of IL­4, IL­5, IL­6 and IL­13 and decreased levels of IL­2 and IFNγ156 (FIG. 2).

In BALB/c mice, ETS­exposed female neonates are significantly more susceptible than ETS­exposed male neonates to allergic responses157 and to ETS­driven pro­duction of TH2­type cytokines. This gender difference has been attributed to the TH2 cell­enhancing activity of pro­gesterone158. In addition, ETS exposure during the early postnatal period has strong immunosuppressive effects, particularly on T cells, resulting in impaired T cell recep­tor signalling159,160. It is probable that this effect dampens the normal development of T cell responses, particularly against environmental stimuli and microorganisms, as indicated by the increased susceptibility of smokers to respiratory infections and asthma exacerbation.

Furthermore, the predisposition to asthma can be increased by intra­uterine ETS exposure combined with some genetic factors (such as polymorphisms in the genes encoding glutathione­S­transferase160, in the functional promoter of TGFB1 (REF. 161) or in IL1R162). These are important examples of gene–environment interactions that result in the development of a clinical phenotype.

In conclusion, environmental factors have a marked effect on the development of perinatal mucosal immune responses in the respiratory tract. Prenatal exposure to dietary components might influence the pathogenesis of respiratory disease (BOX 2). Moreover, perinatal ETS expo­sure is one of the most important risk factors for asthma development in early life. In addition, perinatal exposure to commensal bacteria of the gut, airways, skin or vagina and airborne environmental microorganisms (FIG. 2) is the most prominent exogenous regulator of perinatal respira­tory immunity. Other microorganisms, such as viruses

R E V I E W S

NATURE REVIEWS | IMMUNOLOGY VOLUME 12 | JANUARY 2012 | 19

© 2012 Macmillan Publishers Limited. All rights reserved

Page 12: The impact of perinatal immune development on mucosal ...manyan/To print/The impact of perinatal immune... · natal development in mice. During the first two weeks of postnatal life,

and fungi, might also have an important role in mucosal immune responses but their contribution has not yet been extensively studied. Therefore, further research is needed to fully understand the impact of host–microbial inter­actions at the local mucosal surface on shaping innate and adaptive mucosal immune responses.

Emerging interventional approachesRecent research on epithelial homeostasis, mucosal immunity and the mucosal microbiota has revealed the enormous complexity of the host–microbial and host–environmental relationships. Better understanding of this complexity has paved the way for novel therapeutic or preventive measures. Antigen­specific approaches to re­establish tolerance in allergic patients through administration of the allergen as a ‘tolerogen’ have pro­vided promising results in clinical studies45. Also, anti­gen nonspecific approaches, such as helminth­derived immuno modulators, probiotics and diet regimen, might be important in the future treatment of immune­ mediated pathologies3,163, although the data are not yet conclusive. Experiments have indicated that some bacterial strains can induce DCs, directly or through

epithelial mediators, to exert anti­inflammatory effects. In addition, early intestinal colonization with beneficial probiotic bacteria was shown to enhance SIgA export to the gut lumen164. Prebiotics, such as selected oligo­saccharides, increase the concentration of beneficial bacteria and the levels of SIgA in healthy infants. However, the selection of the right probiotic bacterial strains, or of symbiotic combinations of prebiotics and breast milk, remains a difficult task and requires the consideration of several safety issues, as certain bacterial strains might be pro­inflammatory165,166. A more promis­ing strategy might be to identify the underlying molecu­lar mechanisms of immune tolerance and to administer the specific components or metabolites that enhance perinatal mucosal immune homeostasis167,168. Finally, another potential future approach for the treatment of chronic inflammatory disease is the interference with specific signal transduction pathways or transcription factors to reduce or promote a certain type of immune response169. Nevertheless, we are only just beginning to identify the cellular and molecular mechanisms that facilitate the postnatal establishment of homeostasis and, thus, potential targets for rational manipulation.

Box 2 | The role of dietary and nutritional components in the pathogenesis of allergy and asthma

There is increasing evidence that the pathogenesis of respiratory diseases, including asthma, might be associated with prenatal exposure to dietary components. The effects of vitamins A and D on asthma are being investigated.

Vitamin A has an essential role in cellular and subcellular membrane stability and, thus, influences the repair and development of epithelial cells174. However, it might promote an allergic response, as it has been shown to favour the development of T helper 2 (T

H2) cells175.

Vitamin D is important for lung maturation and surfactant production, which is of particular importance at the end of the intra-uterine phase of lung development, when the alveolar epithelium undergoes abrupt differentiation as part of the preparation for gas exchange at birth176,177. Furthermore, vitamin D and its metabolites have a broad range of immunomodulatory effects on both innate and adaptive immune cells178. Vitamin D induces tolerogenic dendritic cells that are characterized by the upregulation of expression of the inhibitory receptors immunoglobulin-like transcript 3 (ILT3) and ILT4 (REFS 179,180). A large cohort study181 associated vitamin D supplementation during pregnancy with a significant increase in the mRNA levels of ILT3 and ILT4 in cord blood.

However, the clinical implications of vitamin A and vitamin D supplementation during pregnancy still remain controversial182,183. Conflicting results might be related to the differential effects of the isoforms and metabolites of these vitamins. Clearly, more mechanistic studies are needed to explore the precise effects on fetal immune development.

A methyl donor-rich diet, consisting of high amounts of vitamin B12, folic acid, choline and methionine, might also influence the development of perinatal immunity184. Intra-uterine exposure to a high dose of methyl donors increased the rate of methylation at CpG DNA, which is an important mechanism of epigenetic regulation185. Supplementation of pregnant mice with such a methyl donor-rich diet increased the development of experimental asthma in the offspring186. However, the implications of these results for the development of human asthma remain a matter of debate187,188.

1. Bach, J. F. The effect of infections on susceptibility to autoimmune and allergic diseases. N. Engl. J. Med. 347, 911–920 (2002).

2. Holt, P. G., Strickland, D. H., Wikstrom, M. E. & Jahnsen, F. L. Regulation of immunological homeostasis in the respiratory tract. Nature Rev. Immunol. 8, 142–152 (2008).

3. Guarner, F. et al. Mechanisms of disease: the hygiene hypothesis revisited. Nature Clin. Pract. Gastroenterol. Hepatol. 3, 275–284 (2006).

4. Tulic, M. K. et al. Differences in innate immune function between allergic and nonallergic children: new insights into immune ontogeny. J. Allergy Clin. Immunol. 127, 470–478 (2011).

5. Prescott, S. L. Role of dietary immunomodulatory factors in the development of immune tolerance. Nestle Nutr. Workshop Ser. Pediatr. Program. 64, 185–194 (2009).

6. Fasano, A. Zonulin and its regulation of intestinal barrier function: the biological door to inflammation,

autoimmunity, and cancer. Physiol. Rev. 91, 151–175 (2011).

7. Elinav, E., Strowig, T., Henao-Mejia, J. & Flavell, R. A. Regulation of the antimicrobial response by NLR proteins. Immunity 34, 665–679 (2011).

8. Garrett, W. S. et al. Enterobacteriaceae act in concert with the gut microbiota to induce spontaneous and maternally transmitted colitis. Cell Host Microbe 8, 292–300 (2010).

9. Hilty, M. et al. Disordered microbial communities in asthmatic airways. PLoS ONE 5, e8578 (2010).

10. Huang, Y. J. et al. Airway microbiota and bronchial hyperresponsiveness in patients with suboptimally controlled asthma. J. Allergy Clin. Immunol. 127, 372–381 (2011).

11. McGovern, D. P. et al. Genome-wide association identifies multiple ulcerative colitis susceptibility loci. Nature Genet. 42, 332–337 (2010).

12. Franke, A. et al. Genome-wide association study for ulcerative colitis identifies risk loci at 7q22 and

22q13 (IL17REL). Nature Genet. 42, 292–294 (2010).

13. Noguchi, E. et al. Genome-wide association study identifies HLA-DP as a susceptibility gene for pediatric asthma in asian populations. PLoS Genet. 7, e1002170 (2011).

14. Harper, J., Mould, A., Andrews, R. M., Bikoff, E. K. & Robertson, E. J. The transcriptional repressor Blimp1/Prdm1 regulates postnatal reprogramming of intestinal enterocytes. Proc. Natl Acad. Sci. USA 13 Jun 2011 (doi:10.1073/pnas.1105852108).

15. de Santa, B. P., van den Brink, G. R. & Roberts, D. J. Development and differentiation of the intestinal epithelium. Cell. Mol. Life Sci. 60, 1322–1332 (2003).

16. Darmoul, D., Brown, D., Selsted, M. E. & Ouellette, A. J. Cryptdin gene expression in developing mouse small intestine. Am. J. Physiol. 272, G197–G206 (1997).

17. Bry, L. et al. Paneth cell differentiation in the developing intestine of normal and transgenic mice. Proc. Natl Acad. Sci. USA 91, 10335–10339 (1994).

R E V I E W S

20 | JANUARY 2012 | VOLUME 12 www.nature.com/reviews/immunol

© 2012 Macmillan Publishers Limited. All rights reserved

Page 13: The impact of perinatal immune development on mucosal ...manyan/To print/The impact of perinatal immune... · natal development in mice. During the first two weeks of postnatal life,

18. Menard, S. et al. Developmental switch of intestinal antimicrobial peptide expression. J. Exp. Med. 205, 183–193 (2008).

19. Putsep, K. et al. Germ-free and colonized mice generate the same products from enteric prodefensins. J. Biol. Chem. 275, 40478–40482 (2000).

20. Sanos, S. L. et al. RORγt and commensal microflora are required for the differentiation of mucosal interleukin 22-producing NKp46+ cells. Nature Immunol. 10, 83–91 (2009).

21. Cash, H. L., Whitham, C. V., Behrendt, C. L. & Hooper, L. V. Symbiotic bacteria direct expression of an intestinal bactericidal lectin. Science 313, 1126–1130 (2006).

22. Kai-Larsen, Y. et al. Antimicrobial components of the neonatal gut affected upon colonization. Pediatr. Res. 61, 530–536 (2007).

23. Salzman, N. H. et al. Enteric defensins are essential regulators of intestinal microbial ecology. Nature Immunol. 11, 76–83 (2010). This is the first study to detail the functional consequences of decreased or increased antimicrobial peptide production on the enteric microbiota.

24. Palmer, C., Bik, E. M., DiGiulio, D. B., Relman, D. A. & Brown, P. O. Development of the human infant intestinal microbiota. PLoS Biol. 5, e177 (2007).

25. Dominguez-Bello, M. G. et al. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc. Natl Acad. Sci. USA 107, 11971–11975 (2010). An important study showing that the human neonatal intestinal microbiota is derived from the mother but depends on the mode of delivery.

26. Koenig, J. E. et al. Succession of microbial consortia in the developing infant gut microbiome. Proc. Natl Acad. Sci. USA 108 (Suppl. 1), 4578–4585 (2011).

27. Costello, E. K. et al. Bacterial community variation in human body habitats across space and time. Science 326, 1694–1697 (2009).

28. Lotz, M. et al. Postnatal acquisition of endotoxin tolerance in intestinal epithelial cells. J. Exp. Med. 203, 973–984 (2006).

29. Gribar, S. C. et al. Reciprocal expression and signaling of TLR4 and TLR9 in the pathogenesis and treatment of necrotizing enterocolitis. J. Immunol. 182, 636–646 (2009).

30. Claud, E. C. et al. Developmentally regulated IκB expression in intestinal epithelium and susceptibility to flagellin-induced inflammation. Proc. Natl Acad. Sci. USA 101, 7404–7408 (2004).

31. Chassin, C. et al. miR-146a mediates protective innate immune tolerance in the neonate intestine. Cell Host Microbe 8, 358–368 (2010). A detailed analysis of the molecular processes that control intestinal epithelial innate immune tolerance in neonatal mice.

32. Kajino-Sakamoto, R. et al. Enterocyte-derived TAK1 signaling prevents epithelium apoptosis and the development of ileitis and colitis. J. Immunol. 181, 1143–1152 (2008).

33. Hooper, L. V. et al. Molecular analysis of commensal host–microbial relationships in the intestine. Science 291, 881–884 (2001).

34. Bates, J. M. et al. Distinct signals from the microbiota promote different aspects of zebrafish gut differentiation. Dev. Biol. 297, 374–386 (2006).

35. Broquet, A. H., Hirata, Y., McAllister, C. S. & Kagnoff, M. F. RIG-I/MDA5/MAVS are required to signal a protective IFN response in rotavirus-infected intestinal epithelium. J. Immunol. 186, 1618–1626 (2011).

36. Pott, J. et al. IFN-λ determines the intestinal epithelial antiviral host defense. Proc. Natl Acad. Sci. USA 108, 7944–7949 (2011).

37. Weitkamp, J. H. et al. Ontogeny of FOXP3+ regulatory T cells in the postnatal human small intestinal and large intestinal lamina propria. Pediatr. Dev. Pathol. 12, 443–449 (2009).

38. Xiong, N. & Raulet, D. H. Development and selection of γδ T cells. Immunol. Rev. 215, 15–31 (2007).

39. van de Pavert, S. A. & Mebius, R. E. New insights into the development of lymphoid tissues. Nature Rev. Immunol. 10, 664–674 (2010).

40. Brandtzaeg, P. Function of mucosa-associated lymphoid tissue in antibody formation. Immunol. Invest. 39, 303–355 (2010).

41. Sawa, S. et al. Lineage relationship analysis of RORγt+ innate lymphoid cells. Science 330, 665–669 (2010).

42. Atarashi, K. et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science 331, 337–341 (2011).

43. Round, J. L. et al. The Toll-like receptor 2 pathway establishes colonization by a commensal of the human microbiota. Science 332, 974–977 (2011).

44. Geuking, M. B. et al. Intestinal bacterial colonization induces mutualistic regulatory T cell responses. Immunity 34, 794–806 (2011).

45. Brandtzaeg, P. Food allergy: separating the science from the mythology. Nature Rev. Gastroenterol. Hepatol. 7, 380–400 (2010).

46. Michaelsson, J., Mold, J. E., McCune, J. M. & Nixon, D. F. Regulation of T cell responses in the developing human fetus. J. Immunol. 176, 5741–5748 (2006). This study showed that CD4+CD25+ TReg cells are present at high frequency in human fetal lymphoid tissue, including the mesenteric lymph nodes; these TReg cells seem to be important in controlling T cell responses in utero and perhaps also neonatally.

47. Grindebacke, H. et al. Dynamic development of homing receptor expression and memory cell differentiation of infant CD4+CD25high regulatory T cells. J. Immunol. 183, 4360–4370 (2009).

48. Hadis, U. et al. Intestinal tolerance requires gut homing and expansion of FoxP3+ regulatory T cells in the lamina propria. Immunity 34, 237–246 (2011).

49. Haddeland, U. et al. Putative regulatory T cells are impaired in cord blood from neonates with hereditary allergy risk. Pediatr. Allergy Immunol. 16, 104–112 (2005). This study of cord blood cells presents data to indicate that neonates at high risk of atopy (familial allergic disease) might generate decreased numbers of CD4+CD25+ TReg cells after stimulation.

50. Smith, M. et al. Children with egg allergy have evidence of reduced neonatal CD4+CD25+CD127lo/– regulatory T cell function. J. Allergy Clin. Immunol. 121, 1460–1466.e7 (2008).

51. O’Mahony, C. et al. Commensal-induced regulatory T cells mediate protection against pathogen-stimulated NF-κB activation. PLoS Pathog. 4, e1000112 (2008).

52. Wang, G. et al. “Default” generation of neonatal regulatory T cells. J. Immunol. 185, 71–78 (2010). Murine neonatal CD4+ T cells were shown to have an inborn ability to preferentially develop into FOXP3+ TReg cells with suppressive function after stimulation, which was in marked contrast to results obtained with adult CD4+ T cells.

53. Lee, J. H., Ulrich, B., Cho, J., Park, J. & Kim, C. H. Progesterone promotes differentiation of human cord blood fetal T cells into T regulatory cells but suppresses their differentiation into TH17 cells. J. Immunol. 187, 1778–1787 (2011).

54. Ivanov, I. I. et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 139, 485–498 (2009).

55. Gaboriau-Routhiau, V. et al. The key role of segmented filamentous bacteria in the coordinated maturation of gut helper T cell responses. Immunity 31, 677–689 (2009). References 54 and 55 demonstrate the striking effects of one particular gut commensal on mucosal immune development.

56. Jiang, H. Q., Bos, N. A. & Cebra, J. J. Timing, localization, and persistence of colonization by segmented filamentous bacteria in the neonatal mouse gut depend on immune status of mothers and pups. Infect. Immun. 69, 3611–3617 (2001).

57. Brandtzaeg, P., Fjellanger, I. & Gjeruldsen, S. T. Adsorption of immunoglobulin A onto oral bacteria in vivo. J. Bacteriol. 96, 242–249 (1968).

58. van der Waaij, L. A., Limburg, P. C., Mesander, G. & van der Waaij, D. In vivo IgA coating of anaerobic bacteria in human faeces. Gut 38, 348–354 (1996).

59. Peterson, D. A., McNulty, N. P., Guruge, J. L. & Gordon, J. I. IgA response to symbiotic bacteria as a mediator of gut homeostasis. Cell Host Microbe 2, 328–339 (2007). In this study, Bacteroides thetaiotaomicron was introduced into germ-free and immunodeficient mice harbouring IgA-producing hybridoma cells with reactivity against a single immunomodulatory molecule; the antibody decreased intestinal pro-inflammatory signalling and bacterial epitope expression, thereby documenting the role of secretory IgA in a balanced mutualistic host–microbial relationship.

60. Mathias, A. & Corthesy, B. Recognition of gram-positive intestinal bacteria by hybridoma- and colostrum-derived secretory immunoglobulin A is mediated by carbohydrates. J. Biol. Chem. 286, 17239–17247 (2011).

61. Hanson, L. A. Session 1: Feeding and infant development breast-feeding and immune function. Proc. Nutr. Soc. 66, 384–396 (2007).

62. Pabst, R., Russell, M. W. & Brandtzaeg, P. Tissue distribution of lymphocytes and plasma cells and the role of the gut. Trends Immunol. 29, 206–208 (2008).

63. Mellander, L., Carlsson, B., Jalil, F., Soderstrom, T. & Hanson, L. A. Secretory IgA antibody response against Escherichia coli antigens in infants in relation to exposure. J. Pediatr. 107, 430–433 (1985).

64. Crabbe, P. A., Nash, D. R., Bazin, H., Eyssen, H. & Heremans, J. F. Immunohistochemical observations on lymphoid tissues from conventional and germ-free mice. Lab. Invest. 22, 448–457 (1970). This is the first experimental study to show that the development of the intestinal population of IgA-producing plasma cells depends on colonization of the gut with a commensal microbiota.

65. Shroff, K. E., Meslin, K. & Cebra, J. J. Commensal enteric bacteria engender a self-limiting humoral mucosal immune response while permanently colonizing the gut. Infect. Immun. 63, 3904–3913 (1995).

66. Hapfelmeier, S. et al. Reversible microbial colonization of germ-free mice reveals the dynamics of IgA immune responses. Science 328, 1705–1709 (2010).

67. Kaufman, D. R. et al. Vitamin A deficiency impairs vaccine-elicited gastrointestinal immunity. J. Immunol. 187, 1877–1883 (2011).

68. Brandtzaeg, P. Update on mucosal immunoglobulin A in gastrointestinal disease. Curr. Opin. Gastroenterol. 26, 554–563 (2010).

69. Johansen, F. E. et al. Absence of epithelial immunoglobulin A transport, with increased mucosal leakiness, in polymeric immunoglobulin receptor/secretory component-deficient mice. J. Exp. Med. 190, 915–922 (1999). This study describes for the first time the generation of knockout mice deficient for a functional polymeric immunoglobulin receptor (pIgR) in secretory epithelia and the liver. These mice could not therefore export IgA or IgM to the mucosal surfaces and had a ‘leaky’ gut mucosa, which was associated with an increased systemic immune responsiveness to commensal bacteria, but remarkably not to food antigens.

70. Sait, L. C. et al. Secretory antibodies reduce systemic antibody responses against the gastrointestinal commensal flora. Int. Immunol. 19, 257–265 (2007).

71. Karlsson, M. R., Johansen, F. E., Kahu, H., Macpherson, A. & Brandtzaeg, P. Hypersensitivity and oral tolerance in the absence of a secretory immune system. Allergy 65, 561–570 (2010). Mice deficient for pIgR were shown to be hyper-reactive and therefore predisposed to anaphylaxis after systemic antigen sensitization and challenge. However, they also had an increased capacity for the induction of oral tolerance, which after continuous feeding of cognate antigen protected the mice from IgG1-induced anaphylaxis and delayed type hypersensitivity. This study showed for the first time that a leaky gut mucosa not only results in hyper-reactivity but can also enhance mucosally induced tolerance to cognate antigen.

72. Boirivant, M. et al. A transient breach in the epithelial barrier leads to regulatory T-cell generation and resistance to experimental colitis. Gastroenterology 135, 1612–1623 (2008).

73. Karlsson, M. R., Rugtveit, J. & Brandtzaeg, P. Allergen-responsive CD4+CD25+ regulatory T cells in children who have outgrown cow’s milk allergy. J. Exp. Med. 199, 1679–1688 (2004). This study provides the first human data to indicate that oral tolerance to dietary antigens is associated with the development or proliferation of CD4+CD25+ TReg cells.

74. Shreffler, W. G., Wanich, N., Moloney, M., Nowak-Wegrzyn, A. & Sampson, H. A. Association of allergen-specific regulatory T cells with the onset of clinical tolerance to milk protein. J. Allergy Clin. Immunol. 123, 43–52 (2009).

75. Feng, T. & Elson, C. O. Adaptive immunity in the host-microbiota dialog. Mucosal Immunol. 4, 15–21 (2011).

R E V I E W S

NATURE REVIEWS | IMMUNOLOGY VOLUME 12 | JANUARY 2012 | 21

© 2012 Macmillan Publishers Limited. All rights reserved

Page 14: The impact of perinatal immune development on mucosal ...manyan/To print/The impact of perinatal immune... · natal development in mice. During the first two weeks of postnatal life,

76. Menezes, J. S. et al. Stimulation by food proteins plays a critical role in the maturation of the immune system. Int. Immunol. 15, 447–455 (2003).

77. Kang, W. & Kudsk, K. A. Is there evidence that the gut contributes to mucosal immunity in humans? JPEN J. Parenter. Enteral Nutr. 31, 246–258 (2007).

78. Barclay, A. R. et al. Systematic review: the role of breastfeeding in the development of pediatric inflammatory bowel disease. J. Pediatr. 155, 421–426 (2009).

79. Kau, A. L., Ahern, P. P., Griffin, N. W., Goodman, A. L. & Gordon, J. I. Human nutrition, the gut microbiome and the immune system. Nature 474, 327–336 (2011).

80. Harju, K., Glumoff, V. & Hallman, M. Ontogeny of Toll-like receptors TLR2 and TLR4 in mice. Pediatr. Res. 49, 81–83 (2001).

81. Petrikin, J. E., Gaedigk, R., Leeder, J. S. & Truog, W. E. Selective Toll-like receptor expression in human fetal lung. Pediatr. Res. 68, 335–338 (2010).

82. Zhang, X., Shan, P., Jiang, G., Cohn, L. & Lee, P. J. Toll-like receptor 4 deficiency causes pulmonary emphysema. J. Clin. Invest. 116, 3050–3059 (2006).

83. Saenz, S. A., Taylor, B. C. & Artis, D. Welcome to the neighborhood: epithelial cell-derived cytokines license innate and adaptive immune responses at mucosal sites. Immunol. Rev. 226, 172–190 (2008).

84. Barrett, N. A. & Austen, K. F. Innate cells and T helper 2 cell immunity in airway inflammation. Immunity 31, 425–437 (2009).

85. Hiemstra, P. S. The role of epithelial β-defensins and cathelicidins in host defense of the lung. Exp. Lung Res. 33, 537–542 (2007).

86. Bienenstock, J., Johnston, N. & Perey, D. Y. Bronchial lymphoid tissue. I. Morphologic characteristics. Lab. Invest. 28, 686–692 (1973).

87. Racz, P., Tenner-Racz, K., Myrvik, Q. N. & Fainter, L. K. Functional architecture of bronchial associated lymphoid tissue and lymphoepithelium in pulmonary cell-mediated reactions in the rabbit. J. Reticuloendothel. Soc. 22, 59–83 (1977).

88. Finke, D., Baribaud, F., Diggelmann, H. & Acha-Orbea, H. Extrafollicular plasmablast B cells play a key role in carrying retroviral infection to peripheral organs. J. Immunol. 166, 6266–6275 (2001).

89. Delventhal, S., Hensel, A., Petzoldt, K. & Pabst, R. Effects of microbial stimulation on the number, size and activity of bronchus-associated lymphoid tissue (BALT) structures in the pig. Int. J. Exp. Pathol. 73, 351–357 (1992).

90. Tschernig, T. & Pabst, R. Bronchus-associated lymphoid tissue (BALT) is not present in the normal adult lung but in different diseases. Pathobiology 68, 1–8 (2000).

91. Chvatchko, Y., Kosco-Vilbois, M. H., Herren, S., Lefort, J. & Bonnefoy, J. Y. Germinal center formation and local immunoglobulin E (IgE) production in the lung after an airway antigenic challenge. J. Exp. Med. 184, 2353–2360 (1996).

92. Corbett, N. P. et al. Ontogeny of Toll-like receptor mediated cytokine responses of human blood mononuclear cells. PLoS ONE 5, e15041 (2010).

93. Yerkovich, S. T. et al. Postnatal development of monocyte cytokine responses to bacterial lipopolysaccharide. Pediatr. Res. 62, 547–552 (2007).

94. Holt, P. G., Batty, J. E. & Turner, K. J. Inhibition of specific IgE responses in mice by pre-exposure to inhaled antigen. Immunology 42, 409–417 (1981).

95. Uthoff, H. et al. Critical role of preconceptional immunization for protective and nonpathological specific immunity in murine neonates. J. Immunol. 171, 3485–3492 (2003).

96. Polte, T., Hennig, C. & Hansen, G. Allergy prevention starts before conception: maternofetal transfer of tolerance protects against the development of asthma. J. Allergy Clin. Immunol. 122, 1022–1030 (2008).

97. Blumer, N., Pfefferle, P. I. & Renz, H. Development of mucosal immune function in the intrauterine and early postnatal environment. Curr. Opin. Gastroenterol. 23, 655–660 (2007).

98. Holloway, J. A. et al. Detection of house-dust-mite allergen in amniotic fluid and umbilical-cord blood. Lancet 356, 1900–1902 (2000).

99. Vance, G. H. et al. Exposure of the fetus and infant to hens’ egg ovalbumin via the placenta and breast milk in relation to maternal intake of dietary egg. Clin. Exp. Allergy 35, 1318–1326 (2005).

100. Verhasselt, V. et al. Breast milk-mediated transfer of an antigen induces tolerance and protection from allergic asthma. Nature Med. 14, 170–175 (2008). Using an asthma model in mice, these authors showed that allergen-exposed dams could transfer small amounts of allergen from their airways to their breast-fed pups; this resulted in the induction of oral tolerance in the pups, which mediated protection against asthma in the same model.

101. Gereke, M., Jung, S., Buer, J. & Bruder, D. Alveolar type II epithelial cells present antigen to CD4+ T cells and induce Foxp3+ regulatory T cells. Am. J. Respir. Crit. Care Med. 179, 344–355 (2009).

102. Vestermark, V., Hogdall, C. K., Birch, M., Plenov, G. & Toftager-Larsen, K. Influence of the mode of delivery on initiation of breast-feeding. Eur. J. Obstet. Gynecol. Reprod. Biol. 38, 33–38 (1991).

103. Ege, M. J. et al. Exposure to environmental microorganisms and childhood asthma. N. Engl. J. Med. 364, 701–709 (2011).

104. Decker, E. et al. Cesarean delivery is associated with celiac disease but not inflammatory bowel disease in children. Pediatrics 125, e1433–e1440 (2010).

105. Koplin, J. et al. Is caesarean delivery associated with sensitization to food allergens and IgE-mediated food allergy: a systematic review. Pediatr. Allergy Immunol. 19, 682–687 (2008).

106. Kuitunen, M. et al. Probiotics prevent IgE-associated allergy until age 5 years in cesarean-delivered children but not in the total cohort. J. Allergy Clin. Immunol. 123, 335–341 (2009). In this study, mothers with infants at high risk of IgE-mediated allergy received a probiotic mixture during the last month of pregnancy and their infants received the probiotic mixture combined with prebiotics until the age of 6 months. No allergy-preventive effect of the probiotic mixture was observed at 5 years of age except in children who had been delivered by Caesarean section.

107. Furuhjelm, C. et al. Fish oil supplementation in pregnancy and lactation may decrease the risk of infant allergy. Acta Paediatr. 98, 1461–1467 (2009).

108. Newson, R. B. et al. Umbilical cord and maternal blood red cell fatty acids and early childhood wheezing and eczema. J. Allergy Clin. Immunol. 114, 531–537 (2004).

109. Dirix, C. E. et al. Prenatal arachidonic acid exposure and selected immune-related variables in childhood. Br. J. Nutr. 102, 387–397 (2009).

110. Maslowski, K. M. & Mackay, C. R. Diet, gut microbiota and immune responses. Nature Immunol. 12, 5–9 (2011). A timely and interesting review focusing on the relationship between the fields of immunology, microbiology, nutrition and metabolism. It is also pointed out how the diet in various parts of the world influences the composition of the gut microbiome.

111. Sandin, A., Braback, L., Norin, E. & Bjorksten, B. Faecal short chain fatty acid pattern and allergy in early childhood. Acta Paediatr. 98, 823–827 (2009).

112. Glocker, E. O. et al. Inflammatory bowel disease and mutations affecting the interleukin-10 receptor. N. Engl. J. Med. 361, 2033–2045 (2009).

113. Malmborg, P., Bahmanyar, S., Grahnquist, L., Hildebrand, H. & Montgomery, S. Cesarean section and the risk of pediatric Crohn’s disease. Inflamm. Bowel Dis. 27 Apr 2011 (doi:10.1002/ibd.21741).

114. Elinav, E. et al. NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis. Cell 145, 745–757 (2011).

115. Garrett, W. S., Gordon, J. I. & Glimcher, L. H. Homeostasis and inflammation in the intestine. Cell 140, 859–870 (2010). References 114 and 115 demonstrate the transmissibility of an altered ‘colitogenic’ microbiota that can induce disease in genetically wild-type animals.

116. Cadwell, K. et al. Virus-plus-susceptibility gene interaction determines Crohn’s disease gene Atg16L1 phenotypes in intestine. Cell 141, 1135–1145 (2010). Exciting work describing the requirement for viral infection to promote cellular dysfunction and disease in a genetically predisposed animal model of Crohn’s disease.

117. Stecher, B. et al. Salmonella enterica serovar typhimurium exploits inflammation to compete with the intestinal microbiota. PLoS Biol. 5, 2177–2189 (2007).

118. Neu, J. & Walker, W. A. Necrotizing enterocolitis. N. Engl. J. Med. 364, 255–264 (2011).

119. Sodhi, C. P. et al. Toll-like receptor-4 inhibits enterocyte proliferation via impaired β-catenin signaling in necrotizing enterocolitis. Gastroenterology 138, 185–196 (2010).

120. Alfaleh, K. & Bassler, D. Probiotics for prevention of necrotizing enterocolitis in preterm infants. Cochrane Database Syst. Rev. CD005496 (2008).

121. Bottcher, M. F., Nordin, E. K., Sandin, A., Midtvedt, T. & Bjorksten, B. Microflora-associated characteristics in faeces from allergic and nonallergic infants. Clin. Exp. Allergy 30, 1590–1596 (2000).

122. Penders, J. et al. Gut microbiota composition and development of atopic manifestations in infancy: the KOALA Birth Cohort Study. Gut 56, 661–667 (2007).

123. Murray, C. S. et al. Fecal microbiota in sensitized wheezy and non-sensitized non-wheezy children: a nested case–control study. Clin. Exp. Allergy 35, 741–745 (2005).

124. Adlerberth, I. et al. Gut microbiota and development of atopic eczema in 3 European birth cohorts. J. Allergy Clin. Immunol. 120, 343–350 (2007).

125. Wang, M. et al. Reduced diversity in the early fecal microbiota of infants with atopic eczema. J. Allergy Clin. Immunol. 121, 129–134 (2008).

126. Arnold, I. C. et al. Helicobacter pylori infection prevents allergic asthma in mouse models through the induction of regulatory T cells. J. Clin. Invest. 121, 3088–3093 (2011).

127. Noverr, M. C., Falkowski, N. R., McDonald, R. A., McKenzie, A. N. & Huffnagle, G. B. Development of allergic airway disease in mice following antibiotic therapy and fungal microbiota increase: role of host genetics, antigen, and interleukin-13. Infect. Immun. 73, 30–38 (2005).

128. Maslowski, K. M. et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 461, 1282–1286 (2009).

129. Thavagnanam, S., Fleming, J., Bromley, A., Shields, M. D. & Cardwell, C. R. A meta-analysis of the association between Caesarean section and childhood asthma. Clin. Exp. Allergy 38, 629–633 (2008).

130. Bager, P., Wohlfahrt, J. & Westergaard, T. Caesarean delivery and risk of atopy and allergic disease: meta-analyses. Clin. Exp. Allergy 38, 634–642 (2008).

131. Metsala, J. et al. Perinatal factors and the risk of asthma in childhood — a population-based register study in Finland. Am. J. Epidemiol. 168, 170–178 (2008).

132. von Mutius, E. & Vercelli, D. Farm living: effects on childhood asthma and allergy. Nature Rev. Immunol. 10, 861–868 (2010).

133. Ege, M. J. et al. Prenatal exposure to a farm environment modifies atopic sensitization at birth. J. Allergy Clin. Immunol. 122, 407–412.e4 (2008).

134. Heinrich, J. et al. Environmental surveys in the areas of Bitterfeld, Hettstedt and a comparative area in 1992–2000. Gesundheitswesen 64, 675–682 (2002) (in German).

135. Pfefferle, P. I. et al. Cord blood allergen-specific IgE is associated with reduced IFN-γ production by cord blood cells: the Protection against Allergy — Study in Rural Environments (PASTURE) Study. J. Allergy Clin. Immunol. 122, 711–716 (2008).

136. Braun-Fahrlander, C. et al. Environmental exposure to endotoxin and its relation to asthma in school-age children. N. Engl. J. Med. 347, 869–877 (2002).

137. Ege, M. J. et al. Prenatal farm exposure is related to the expression of receptors of the innate immunity and to atopic sensitization in school-age children. J. Allergy Clin. Immunol. 117, 817–823 (2006).

138. Patel, M. et al. TLR2 agonist ameliorates established allergic airway inflammation by promoting Th1 response and not via regulatory T cells. J. Immunol. 174, 7558–7563 (2005).

139. Velasco, G. et al. Toll-like receptor 4 or 2 agonists decrease allergic inflammation. Am. J. Respir. Cell Mol. Biol. 32, 218–224 (2005).

140. Gerhold, K., Bluemchen, K., Franke, A., Stock, P. & Hamelmann, E. Exposure to endotoxin and allergen in early life and its effect on allergen sensitization in mice. J. Allergy Clin. Immunol. 112, 389–396 (2003).

141. Blumer, N., Herz, U., Wegmann, M. & Renz, H. Prenatal lipopolysaccharide exposure prevents allergic sensitization and airway inflammation, but not airway responsiveness in a murine model of experimental asthma. Clin. Exp. Allergy 35, 397–402 (2005).

142. Sel, S. et al. Immunomodulatory effects of viral TLR ligands on experimental asthma depend on the additive effects of IL-12 and IL-10. J. Immunol. 178, 7805–7813 (2007).

R E V I E W S

22 | JANUARY 2012 | VOLUME 12 www.nature.com/reviews/immunol

© 2012 Macmillan Publishers Limited. All rights reserved

Page 15: The impact of perinatal immune development on mucosal ...manyan/To print/The impact of perinatal immune... · natal development in mice. During the first two weeks of postnatal life,

143. Kim, Y. S., Kwon, K. S., Kim, D. K., Choi, I. W. & Lee, H. K. Inhibition of murine allergic airway disease by Bordetella pertussis. Immunology 112, 624–630 (2004).

144. Debarry, J. et al. Acinetobacter lwoffii and Lactococcus lactis strains isolated from farm cowsheds possess strong allergy-protective properties. J. Allergy Clin. Immunol. 119, 1514–1521 (2007).

145. Tulic, M. K. et al. Differences in innate immune function between allergic and nonallergic children: new insights into immune ontogeny. J. Allergy Clin. Immunol. 127, 470–478 (2011).

146. Conrad, M. L. et al. Maternal TLR signaling is required for prenatal asthma protection by the nonpathogenic microbe Acinetobacter lwoffii F78. J. Exp. Med. 206, 2869–2877 (2009).

147. Pattenden, S. et al. Parental smoking and children’s respiratory health: independent effects of prenatal and postnatal exposure. Tob. Control 15, 294–301 (2006).

148. Alati, R., Al Mamun, A., O’Callaghan, M., Najman, J. M. & Williams, G. M. In utero and postnatal maternal smoking and asthma in adolescence. Epidemiology 17, 138–144 (2006).

149. Gilliland, F. D. et al. Maternal smoking during pregnancy, environmental tobacco smoke exposure and childhood lung function. Thorax 55, 271–276 (2000).

150. Blacquiere, M. J. et al. Maternal smoking during pregnancy induces airway remodelling in mice offspring. Eur. Respir. J. 33, 1133–1140 (2009).

151. Nakamura, Y. et al. Cigarette smoke inhibits lung fibroblast proliferation and chemotaxis. Am. J. Respir. Crit. Care Med. 151, 1497–1503 (1995).

152. Blacquiere, M. J. et al. Maternal smoking during pregnancy decreases Wnt signalling in neonatal mice. Thorax 65, 553–554 (2010).

153. Noakes, P. S. et al. Maternal smoking is associated with impaired neonatal Toll-like-receptor-mediated immune responses. Eur. Respir. J. 28, 721–729 (2006).

154. Noakes, P. S., Holt, P. G. & Prescott, S. L. Maternal smoking in pregnancy alters neonatal cytokine responses. Allergy 58, 1053–1058 (2003).

155. Gentile, D. et al. Association between environmental tobacco smoke and diminished dendritic cell interleukin-10 production during infancy. Ann. Allergy Asthma Immunol. 92, 433–437 (2004).

156. Singh, S. P. et al. Maternal exposure to secondhand cigarette smoke primes the lung for induction of phosphodiesterase-4D5 isozyme and exacerbated Th2 responses: rolipram attenuates the airway hyperreactivity and muscarinic receptor expression but not lung inflammation and atopy. J. Immunol. 183, 2115–2121 (2009).

157. Seymour, B. W. et al. Gender differences in the allergic response of mice neonatally exposed to environmental tobacco smoke. Dev. Immunol. 9, 47–54 (2002).

158. Romagnani, S. Regulation of the development of type 2 T-helper cells in allergy. Curr. Opin. Immunol. 6, 838–846 (1994).

159. Singh, S. P. et al. Early postnatal exposure to cigarette smoke impairs the antigen-specific T-cell responses in the spleen. Toxicol. Lett. 167, 231–237 (2006).

160. Gilliland, F. D. et al. Effects of glutathione S-transferase M1, maternal smoking during pregnancy, and environmental tobacco smoke on asthma and wheezing in children. Am. J. Respir. Crit. Care Med. 166, 457–463 (2002).

161. Salam, M. T., Gauderman, W. J., McConnell, R., Lin, P. C. & Gilliland, F. D. Transforming growth

factor-1 C-509T polymorphism, oxidant stress, and early-onset childhood asthma. Am. J. Respir. Crit. Care Med. 176, 1192–1199 (2007).

162. Ramadas, R. A. et al. Interleukin-1R antagonist gene and pre-natal smoke exposure are associated with childhood asthma. Eur. Respir. J. 29, 502–508 (2007).

163. Harnett, W. & Harnett, M. M. Helminth-derived immunomodulators: can understanding the worm produce the pill? Nature Rev. Immunol. 10, 278–284 (2010).

164. Kukkonen, K. et al. High intestinal IgA associates with reduced risk of IgE-associated allergic diseases. Pediatr. Allergy Immunol. 21, 67–73 (2010). In this study, prenatal and postnatal probiotic intervention tended to increase IgA levels in the faeces of children at the age of 6 months; this increase of faecal IgA seemed to be associated with a mild inflammatory response in the gut mucosa and a decreased risk of IgE-mediated allergic diseases.

165. Salminen, S., Collado, M. C., Isolauri, E. & Gueimonde, M. Microbial–host interactions: selecting the right probiotics and prebiotics for infants. Nestle Nutr. Workshop Ser. Pediatr. Program 64, 201–213 (2009).

166. Hormannsperger, G. & Haller, D. Molecular crosstalk of probiotic bacteria with the intestinal immune system: clinical relevance in the context of inflammatory bowel disease. Int. J. Med. Microbiol. 300, 63–73 (2010).

167. Rachmilewitz, D. et al. Toll-like receptor 9 signaling mediates the anti-inflammatory effects of probiotics in murine experimental colitis. Gastroenterology 126, 520–528 (2004).

168. Round, J. L. & Mazmanian, S. K. Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc. Natl Acad. Sci. USA 107, 12204–12209 (2010). This study showed that a single microbial immunomodulatory molecule — polysaccharide A from the commensal bacterium Bacteroides fragilis — can induce TReg cells in the murine gut, which express FOXP3 and secrete anti-inflammatory cytokines such as IL-10.

169. Solt, L. A. et al. Suppression of TH17 differentiation and autoimmunity by a synthetic ROR ligand. Nature 472, 491–494 (2011). This study used a high-affinity synthetic ligand to block specifically the retinoic acid receptor-related orphan receptors (RORs) that are indispensable for the development of pro-inflammatory TH17 cells; the results indicate that such compounds have potential for the future treatment of chronic inflammatory diseases.

170. Uibo, R., Tian, Z. & Gershwin, M. E. Celiac disease: a model disease for gene-environment interaction. Cell. Mol. Immunol. 8, 93–95 (2011).

171. Zhernakova, A. et al. Meta-analysis of genome-wide association studies in celiac disease and rheumatoid arthritis identifies fourteen non-HLA shared loci. PLoS Genet. 7, e1002004 (2011).

172. Eder, W. & von Mutius, E. Genetics in asthma: the solution to a lasting conundrum? Allergy 60, 1482–1484 (2005).

173. Hong, X., Tsai, H. J. & Wang, X. Genetics of food allergy. Curr. Opin. Pediatr. 21, 770–776 (2009).

174. Chabra, S. et al. Vitamin A status in preterm neonates with and without chronic lung disease. J. Paediatr. Child Health 30, 432–435 (1994).

175. Schuster, G. U., Kenyon, N. J. & Stephensen, C. B. Vitamin A deficiency decreases and high dietary

vitamin A increases disease severity in the mouse model of asthma. J. Immunol. 180, 1834–1842 (2008).

176. Marin, L., Dufour, M. E., Nguyen, T. M., Tordet, C. & Garabedian, M. Maturational changes induced by 1α, 25-dihydroxyvitamin D3 in type II cells from fetal rat lung explants. Am. J. Physiol. 265, L45–L52 (1993).

177. Nguyen, M., Guillozo, H., Garabedian, M. & Balsan, S. Lung as a possible additional target organ for vitamin D during fetal life in the rat. Biol. Neonate 52, 232–240 (1987).

178. Litonjua, A. A. Childhood asthma may be a consequence of vitamin D deficiency. Curr. Opin. Allergy Clin. Immunol. 9, 202–207 (2009).

179. Adorini, L. & Penna, G. Dendritic cell tolerogenicity: a key mechanism in immunomodulation by vitamin D receptor agonists. Hum. Immunol. 70, 345–352 (2009).

180. Manavalan, J. S. et al. High expression of ILT3 and ILT4 is a general feature of tolerogenic dendritic cells. Transpl. Immunol. 11, 245–258 (2003).

181. Rochat, M. K. et al. Maternal vitamin D intake during pregnancy increases gene expression of ILT3 and ILT4 in cord blood. Clin. Exp. Allergy 40, 786–794 (2010).

182. Greenough, A., Shaheen, S. O., Shennan, A., Seed, P. T. & Poston, L. Respiratory outcomes in early childhood following antenatal vitamin C and E supplementation. Thorax 65, 998–1003 (2010).

183. Turner, S. W. et al. Associations between fetal size, maternal α-tocopherol and childhood asthma. Thorax 65, 391–397 (2010).

184. Dunstan, J. A. et al. The relationship between maternal folate status in pregnancy, cord blood folate levels, and allergic outcomes in early childhood. Allergy 19 Sep 2011 (doi:10.1111/j.1398-9995.2011.02714.x).

185. Waterland, R. A. & Michels, K. B. Epigenetic epidemiology of the developmental origins hypothesis. Annu. Rev. Nutr. 27, 363–388 (2007).

186. Hollingsworth, J. W. et al. In utero supplementation with methyl donors enhances allergic airway disease in mice. J. Clin. Invest. 118, 3462–3469 (2008).

187. Whitrow, M. J., Moore, V. M., Rumbold, A. R. & Davies, M. J. Effect of supplemental folic acid in pregnancy on childhood asthma: a prospective birth cohort study. Am. J. Epidemiol. 170, 1486–1493 (2009).

188. Yang, Q. H. et al. Race-ethnicity differences in folic acid intake in women of childbearing age in the United States after folic acid fortification: findings from the National Health and Nutrition Examination Survey, 2001–2002. Am. J. Clin. Nutr. 85, 1409–1416 (2007).

AcknowledgementsH.R. is supported by the SFB/TR22 DFG-funded network ‘Allergic Immune Responses of the Lung’ and by the Universities of Giessen and Marburg Lung Centre (UGMLC), a LOEWE excellence initiative of the Hessian Government, Germany. P.B. is supported by the Research Council of Norway, The University of Oslo and Oslo University Hospital, through the joint financing of the Center of Excellence named ‘Centre for Immune Regulation’. M.H. is supported by the German Research Foundation (Ho2236/5-3), the Federal Ministry of Education and Research (DLR 01GU0825 and DLR 01KI1003D) and the Collaborative Research Center grants SFB621 and SFB900.

Competing interests statement The authors declare no competing financial interests.

R E V I E W S

NATURE REVIEWS | IMMUNOLOGY VOLUME 12 | JANUARY 2012 | 23

© 2012 Macmillan Publishers Limited. All rights reserved