You are on page 1of 10

Human Microbiome Journal 10 (2018) 11–20

Contents lists available at ScienceDirect

Human Microbiome Journal


journal homepage: www.elsevier.com/locate/humic

Microbiome and the immune system: From a healthy steady-state to allergy T


associated disruption
Soraya Mezouara, Yannick Chantranb,c, Justin Micheld,e, Alexandre Fabref,g,
Jean-Christophe Dubusa,h, Marc Leonea,i, Youssouf Seremea, Jean-Louis Mègea,

Stéphane Ranquej, Benoît Desnuesa, Pascal Chanezk,l, Joana Vittea,
a
Aix-Marseille Univ, IRD, IHU Méditerranée Infection, MEPHI, Marseille, France
b
Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine, team “Immune System, Neuroinflammation and Neurodegenerative Diseases”,
Hôpital Saint-Antoine, Paris, France
c
Immunology Department, AP-HP Saint-Antoine Hospital, Paris, France
d
Aix Marseille Univ, CNRS, IUSTI, Marseille, France
e
Aix-Marseille Univ, APHM, La Conception Universitary Hospital, Department of ENT and Head and Neck Surgery, Marseille, France
f
Aix-Marseille Univ, APHM, Hopital de la Timone, Service de Pédiatrie Multidisciplinaire, Marseille, France
g
Aix Marseille Univ, INSERM, MMG, Marseille, France
h
Aix-Marseille Univ, APHM Assistance Publique Hôpitaux de Marseille, Hôpital Timone Enfants, Pneumo-pédiatrie, Centre de Ressources et de Compétences en
Mucoviscidose, Marseille, France
i
Aix Marseille Univ, APHM, Hôpital Nord, Service d’anesthésie et de réanimation, Marseille, France
j
Aix-Marseille Univ, APHM, IRD, IHU Méditerranée Infection, VITROME, Marseille, France
k
Aix-Marseille Univ, APHM, Hôpital Nord, Department of Respiratory Diseases, Marseille, France
l
Aix-Marseille Univ, INSERM UMR 1062, INRA UMR 1260, C2VN, Marseille, France

A R T I C LE I N FO A B S T R A C T

Keywords: Microbiome and the immune system are constantly shaping each other, in a mutual aim to thrive, defining the
Microbiome unstable equilibrium of the healthy individual. Microbiome is growingly involved in dysimmune conditions such
Mycobiome as allergy, asthma, autoimmunity, and primary or acquired immune deficiencies. The current epidemics of al-
Immune system lergic diseases and asthma has long been linked to the microbial environment through the hygiene hypothesis.
Dysbiosis
Progress in the understanding of the microbiome-immune system crosstalk has unraveled a tight connection
Allergy
between microbial communities and the development of allergic diseases and asthma. Disruption of the mi-
Asthma
Atopy crobiome affects the immune response of the host and paves the way for disease pathogenesis. Conversely,
Atopic dermatitis disease and therapeutic interventions affect microbial communities. We aimed at providing the reader with a
Dendritic cell view of the state-of-the art of microbiome – immune system crosstalk, with special focus on the loopholes giving
Macrophage potential grip to the pathogenesis of microbiome-related dysimmunity.
Mast cell
Chronic rhinosinusitis

1. Introduction 15,000 articles published since 2011 [1]. However, although much has
been learned this field is still a frontier and even simple facts like the
The microbiome consists of a complex mixture of microbes in- number of bacterial cells in the human body have only recently been
cluding Archaea, bacteria, fungi, protozoa, helminths and viruses. The established [2]. Moreover, nearly 70% of the microbiome studies have
description and the understanding of the human microbiome is one of focused on the gut microbiome [1]. Knowledge of the microbiome al-
the most active fields of the biomedical research, with more than lows better identification of the pathophysiological pathways and opens

Abbreviations: AD, atopic dermatitis; BEC, bronchial epithelial cells; COPD, chronic obstructive pulmonary disease; CRS, chronic rhinosinusitis; DC, dendritic cell;
ICU, intensive care unit; Ig, immunoglobulin; IL, interleukin; IS, immune system; LPS, lipopolysaccharide; LTA, lipoteichoic acid; MALT, mucosal-associated lym-
phoid tissue; MC, mast cell; SA, severe asthma; SCFA, short-chain fatty acid; SDD, selective digestive decontamination; TGF-β, Transforming Growth Factor-beta; Th,
helper T cell; TLR, toll-like receptor; Treg, regulatory T cell

Corresponding author at: IHU Méditerranée Infection, UF Immunologie, 19-21, Boulevard Jean Moulin, 13005 Marseille, France.
E-mail address: jvitte@ap-hm.fr (J. Vitte).

https://doi.org/10.1016/j.humic.2018.10.001

Available online 24 October 2018


2452-2317/ © 2018 Elsevier Ltd. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/BY-NC-ND/4.0/).
S. Mezouar et al. Human Microbiome Journal 10 (2018) 11–20

Fig. 1. The microbiota-immune system in-


teractions shape the mucosal homeostasis.
The microbiota provides direct protection
against pathogens through competition for
nutrients, by affecting the physicochemical
conditions of the local microenvironment
(02, pH…), by the production of bacter-
iocins or by down-modulating the expres-
sion of virulence factors (black arrows).
Commensals can also provide indirect, im-
mune mediated protection by sending “out-
side-in” signals (green arrows) that result in
the proper maturation (both cellular and
organizational) of the mucosal-associated
lymphoid tissue, in the maintain of the
physiological inflammation through Tregs
and Th17 generation and in by favoring the
production of IgA by plasmocytes.
Commensals also induces the expression of
mucins by goblet cells and antimicrobial
molecules, such as RegIIIγ or AMP by
Paneth cells. On the other hand, the mucosal
immune system controls its microbiota
through stratification and compartmentali-
zation. Stratification protect the epithelial
cells by keeping the microbiota at a safe
distance (red arrows) while compartmenta-
lization is provided by the unique structure
of the mucosal associated lymphoid tissue
that restrict DC-B and/or T cell interactions
in the mesenteric lymph nodes and avoid
the systemic immune response.
Abbreviations: PSA, polysaccharide A; SFB,
segmented filamentous bacteria; AMP, anti-
microbial peptides. (For interpretation of
the references to colour in this figure legend,
the reader is referred to the web version of
this article.)

the field of intervention through immune modulation [3]. human lineage, as opposite to great apes [12]. The presence of En-
Two drivers of the microbiome composition have been identified in tamoeba is strongly correlated with microbiome composition and di-
humans: genetic and immunological factors on one side and environ- versity [10]. Thus, microbiome is still being deciphered, its role in
mental, notably diet and environmental biodiversity, on the other side. health is paramount, however numerous forces shape it at different
Their relative importance is variable according to the site of the con- levels.
sidered microbiome. For example, diet and hygiene, a correlate of en- Although the microbiome has become synonymous for the collec-
vironmental biodiversity, seem to shape gut microbiome whereas ge- tion of bacterial microbiome, the mycobiome, or the fungal community,
netic and immunological factors are more prevalent for urogenital tract is an essential part of the microbiome [13]. Strikingly, to date less than
microbiome [4]. From an evolutionary perspective the two main factors 400 fungal species have been isolated from humans [14–16]. The es-
driving microbiome composition in mammals are host phylogeny [dif- timated relative mycobiota abundance is heterogeneous, depending on
ferential organismal lineage through time] and diet, each acting at a the human body site; it ranges from less than 0.1% in the gut to ∼10%
different temporal scale and on different subsets of bacteria [5]. At a on the skin [17]. Yet an average fungal cell is ∼100 times larger than
smaller scale, humans display a substantial loss of gut bacteria in an average bacterium, which puts into perspective this relatively low
comparison to chimpanzees and gorillas. This loss is ancient and partly abundance when considering the total fungal biomass. The fungal
linked to diet modification, e.g. increase of animal fats and proteins and biomass constitutes a significant source of bioactive molecules, which
anatomical modification (reduction of gut size) [6]. This reduction of might significantly influence host physiology [18]. In particular, gut
diversity increased with the Neolithic transition and the industrial mycobiota has been shown to modulate the human innate immune
transition with the loss of bacterial lineages in Western world [4,7]. system (IS) via the Dectin-1 receptor [19]. Fungal gut dysbiosis has
Thus, humans seem to undergo a drastic modification of microbiome been associated with gut inflammation; especially colitis exacerbation,
following these transition steps. The effect of environmental changes is associated with alterations of the fungal gut communities, occurred in
also demonstrated by the fact that non-human primates kept in zoos mice treated with antifungal drugs [20]. Shifts in both bacterial and
display altered microbiomes, reminiscent of the shift observed in hu- fungal microbiome compositions have been evidenced in various
mans moving from non-industrialized to westernized environments [8]. chronic diseases, including inflammatory bowel disease [21–23] or in
Finally, some microbiome components are usually under-evaluated, cystic fibrosis patients’ respiratory tract [24].
like Archaea [9] and either unicellular [10] or multicellular [11] eu- Allergic diseases have evolved into a genuine epidemics over the
karyotes. The Archaea are a minor but important component of the gut last half century [25]. Such a rapid evolution is not attributable to
microbiome and are often not detected with universal primers. Pre- genetics. Environment and lifestyle changes were soon identified as key
liminary results suggest a dramatic decrease of their diversity in the drivers [26] through alterations of human microbiome and related

12
S. Mezouar et al. Human Microbiome Journal 10 (2018) 11–20

immune responses [27,28]. Biodiversity loss and climate change am- responses [43].
plify the environment-driven aberrancies of the microbiome [29]. On the other hand, the IS controls host microbiome (back-outside
Moreover, external exposome players such as air pollutants, in utero signals) through stratification and compartmentalization which sepa-
exposure to tobacco, biocontaminants and even diet exert epigenetic rate mucosal immune responses both geographically and functionally
effects which have been associated with the occurrence of allergic from systemic immunity. Stratification or segregation is provided by the
diseases in the offspring [29]. However, general mechanisms might be mucus and anti-microbial molecules. In the colon, where the bacterial
involved in the microbiota – allergy axis, as suggested by a recent paper density is the highest, microbiome is kept away from the epithelium by
replicating the association of early-life dysbiosis and later occurrence of a dense impermeable inner mucus layer composed of O-glycosylated
atopic wheeze in an Ecuadorian pediatric cohort [30]. In this review, MUC2 mucin [44] enriched in lectin-like protein such as ZG16 and β-
we will first outline the major players and mechanisms of the micro- defensins [45]. This spatial segregation is also observed in the intestine
biome – IS interaction. Then, we will present a state-of-the art summary where the mucus is more heterogeneous, rich in Paneth cell-derived α-
of microbiome disruption in atopic/allergic and asthmatic conditions. defensins [46], RegIIIγ C-type lectin [47] and secretory IgA [48].
Anatomical stratification of commensal bacteria is also dependent on
2. Microbiome – Immune system interactions (Fig. 1) the interleukin (IL)-22 produced by the ILC3 that governs immune ex-
clusion by controlling anti-microbial peptide production [49]. In mice
Host-microbiome mutualism largely exceeds the sole metabolic and lacking T CD4 cells, cytokine responses such as IL-22 and IL-23 persist
nutritional aspects and also involves the interaction between the mi- after weaning, leading to microbiota alterations and subsequent meta-
crobiome and the IS. Mucosal surfaces and more specifically the gas- bolic changes [50]. Compartmentalization of the immune response to-
trointestinal tract represent the major interface between the IS and the wards commensal bacteria is provided by the unique structure of the
microorganisms and the ability to discriminate commensals from pa- MALT which reduces the exposure of commensal bacteria to the sys-
thogens can be considered as the driving force of the evolution of the IS. temic IS. Indeed, commensal bacteria are physiologically taken up by T
The commensal microbiome can confer resistance by direct com- cells or directly by DCs from the sub-follicular zone. DCs interact in the
mensal pathogen – interaction through the preferential consumption of Peyer patch with B and T cells or migrate to the mesenteric lymph
nutrients required for the growth of pathogens. This competition for nodes. In the mesenteric lymph nodes, DCs activate B and T cells, which
nutrients has been exemplified with commensal Escherichia coli com- then leave the mesenteric lymph nodes by the thoracic duct, to reach
peting with enterohemorrhagic strains for amino acids and other or- the systemic circulation and home to mucosal tissues generalizing the
ganic acids [31,32]. In addition, commensal bacteria generate small mucosal response to all the mucosal surfaces of the organism [51].
metabolites such as short chain fatty acids (SCFA) or acetate, thereby In summary, extensive IS – microbiome interactions have a sig-
limiting the growth of pathogens [33] or downmodulating the expres- nificant impact on immune homeostasis that can be linked to numerous
sion of virulence genes [34,35]. The indigenous microbiome may also multifactorial diseases. However, it is still unclear whether those al-
produce bactericidal or bacteriostatic substances, such as bacteriocins. terations are the cause of such diseases or the consequence of IS
For example, Lactobacillus salivarius UCC118 produces a bacteriocin in changes related to those diseases.
vivo that can significantly protect mice against Listeria monocytogenes
[36]. Interestingly, protection is also observed against a naturally 3. Macrophage polarization and microbiome
bacteriocin-resistant foodborne pathogen, Salmonella typhimurium UK1,
suggesting competitive exclusion, or immunomodulatory mechanism Macrophages display a large array of functions from host defense to
[36]. host homeostasis. All these functions are impacted by environment
Indeed, commensal microbiome can confer immune-mediated in- from tissues to mutualistic commensals. This diversity enabled the
direct resistance against pathogens. The role of the microbiome in emergence of macrophage polarization complex. The polarization refers
shaping the IS throughout life (outside-in signals) arise from numerous to the set-up of a specific phenotype of macrophage depending of the
studies comparing axenic (germ-free) and specific-pathogen-free mice. pathogens or the microenvironment [52,53].”Classical” activation of
Axenic mice are characterized by atrophy of Peyer’s patches with few the macrophage resulting in enhanced phagocytic and intracellular lysis
germinal centers, immature mesenteric lymph nodes, few isolated ability and type 1 cytokine secretion is termed M1 polarization, while
lymphoid follicles, decreased levels of antimicrobial peptides and im- “alternative” activation with an anti-inflammatory response corre-
munoglobulin (Ig) A and an overall reduced number of B, T and den- sponds to a M2 phenotype. The success of this classification as a
dritic cells (DCs) [37]. Moreover, villi are longer and narrower, with a working hypothesis is related to the reminiscence of T cell polarization
less complex vascular network while crypts are less deep, with fewer and has generated several debates including our contributions [54,55].
proliferating stem cells. Axenic rats have less goblet cells, and a thinner M1-type macrophages are considered as protective in infectious dis-
mucus layer characterized by increased neutral mucins [37]. In addi- eases and cancer whereas M2-type macrophages are protective in
tion, they display defects in systemic IS, since the structure of the spleen parasitic infections and repair responses. We described the role of
and lymph nodes are affected with fewer germinal centers, diffuse B- macrophage polarization in bacterial infectious diseases and identified
cell and T-cell zones and reduced levels of circulating IgG [38]. Very situations in which polarization was associated with a given clinical
importantly, colonizing axenic mice with commensal bacteria reverses condition, prognosis or pathophysiological process [56].
these defects, allowing the IS of these mice to develop and normalize Macrophages are abundant in mucosae at the host-environment
within weeks [39]. These data emphasize the major role of the com- interface. They interact with commensal organisms that have developed
mensal microbiome in the maturation of the mucosal associated lym- mutualistic relationships with the host. Using axenic animals or anti-
phoid tissue (MALT) as well as the systemic IS. For example, the biotic-treated hosts, it has been established that microbiome affects
polysaccharide A from Bacteroides fragilis restores systemic T cell defi- macrophage functions and likely their polarization status. Most reports
ciencies, lymphoid organogenesis and the differentiation of CD4 T cells have established that the intestinal microbiome maintains a tolerant
into regulatory T cells (Tregs), which in turn favor mucosal im- environment through the induction of M2-like intestinal macrophages.
munomodulation and TGF-β production [40,41]. Segmented fila- Indeed, the macrophages from lamina propria show down-regulated
mentous bacteria induce the maturation of T helper (Th) 17 cells to expression of innate response receptors and inflammatory functions,
coordinate the equilibrium with Tregs [42] and maintain a physiolo- but they retain phagocytosis and bactericidal activities [57]. Com-
gical low-grade inflammation. The role played by Th17 cells may vary mensals shape the polarization status of intestinal macrophages via
as a function of the global context: in a murine experimental model, direct and indirect pathways. B. fragilis and intestinal Clostridia stimu-
microbiota of infants with atopic heredity induced predominant Th-17 late M2 polarization and Tregs [58]. Some endproducts of bacterial

13
S. Mezouar et al. Human Microbiome Journal 10 (2018) 11–20

anaerobic fermentation, such as SCFA (α-butyrate), inhibit the in- of a MCs- microbiome crosstalk. Gut and skin microbiomes have re-
flammatory response of macrophages via a mechanism based on the ceived most of the research focus. Several reports have shown a re-
inhibition of histone deacetylase [59]. Helicobacter hepaticus produces a lationship between MCs, microbiome and allergy. For example, it has
large soluble polysaccharide that induces IL-10-mediated M2 type po- been suggested that gut microbes interact with TLRs expressed by MCs
larization in intestinal macrophages [60]. In contrast, intestinal com- to regulate allergic response to food [76]. In mice models, luminal flora
mensals such as Enterococcus faecalis polarize colon macrophages to a was shown to modulate antigen-specific anaphylactic responses in a
M1 phenotype in a murine model in which macrophages are depleted TLR4-dependent manner. Other studies suggested that commensal
with clodronate [61]. Recruited intestinal Ly6Chigh monocytes release bacteria and products from gut lumen play key roles in regulating MCs
IL-1β in response to commensals in an inflammasome-dependent activation and secretion. For example, gut bacteria induced the acti-
manner, favoring M1 polarization [62]. M1 polarization enables sur- vation of intestinal MCs leading to the increased of gut permeability
vival of oral commensals such as Streptococcus gordoni [63]. [77]. In contrast, other studies showed that intestinal commensal or
The effect of microbiome may explain the therapeutic effect of probiotic bacteria could suppress MCs activation, thus linking gut mi-
probiotics. The probiotic Clostridium butyricum promotes the develop- crobiome and the allergic response [78–80]. Skin microbiome can also
ment of IL-10-producing macrophages that prevent inflammatory colitis influence intestinal MCs. Indeed, recently Wang et al. reported that skin
[64]. The strain G-101 of Lactobacillus brevis inhibits the inflammatory microbiome induces keratinocytes to produce stem cell factor (SCF), a
response of mice treated by trinitrobenzenesulfonic acid. This anti-in- major player in MCs maturation [81]. Using axenic mice, the same team
flammatory property is related to the ability of these bacteria to prevent showed that these mice expressed abnormally low levels of SCF and
the expression of M1 markers and to favor M2 markers, presumably via contained undifferentiated MCs. The reconstitution of the microbiome
the production of IL-10 [65]. These findings are inconstant. For in- or the injection of lipoteichoic acid (LTA, a component of the bacterial
stance, some authors failed to evidence any effect on the polarization of cell wall) rescued the phenotype of MCs in a TLR2-dependent manner
RAW 264.7 murine macrophages as a readout [66], while others re- [82]. These experimental results are reminiscent of findings with in-
ported that probiotics bacteria promote an activation profile of the M1 flammatory diseases such as psoriasis or atopic dermatitis. For example,
type in the human monocytic cell line THP-1 stimulated with lipopo- Nakamura et al. reported that Staphylococcus aureus, found on the skin
lysaccharide (LPS) [67]. It is noteworthy that all these studies are of 90% of atopic dermatitis (AD) patients, secreted δ-toxin involved in
limited to in vitro experiments or animal models, and the extrapolation IgE dependent MCs degranulation [83]. Collectively, these data support
to humans must be careful. a contribution of MCs as key link between microbiome and allergy.
The breach of intestinal homeostasis due to the presence of patho- Better understanding the MCs – microbiome interplay will contribute to
genic bacteria would interfere with the polarization status of intestinal new insights into microbial involvement in allergic disorders and to the
and systemic macrophages. The infection with Salmonella typhi or design of novel therapeutic approaches.
Helicobacter pylori in patients is associated with reprogramming mac-
rophages towards M1 phenotype whereas M2 phenotype is found in 5. Allergy and microbiome – B cells and humoral immunity
convalescent patients, suggesting the role of commensals in main-
taining M2 macrophages as major regulators of tolerance [68]. Im- IgG and IgA might contribute to protection against IgE-mediated
balances in gut microbiome have also been associated with systemic allergic reactions. Allergen-specific IgG4 induction during allergen
diseases such as allergy. Recently, Kim et al. [69] reported the induc- immunotherapy parallels tolerance acquisition [84]. Early-life IgG re-
tion of allergen-induced infiltration of inflammatory cells in mice sponses to environmental and food allergens occur at low levels in non-
treated with antibiotics. This treatment alters macrophage functions but allergic individuals [85–87]. However, the most prominent antibody
reorients alveolar macrophages and circulating monocytes toward a M2 involved in mucosal immunity is IgA. IgA is the most abundant anti-
phenotype. The latter is involved in allergic airway inflammation in- body in humans, and an important share of its production is targeted to
duced by allergens. Antibiotic treatment facilitates fungal overgrowth the gut lumen as secretory IgA [88]. Luminal secretory IgA binding to
that exacerbates airway inflammation. The prostaglandin E2 produced selected bacterial antigens is the first step to multiple mechanisms
by gut fungi is responsible for eosinophil-mediated inflammation and aimed at maintaining mucosal homeostasis: immune exclusion [89],
M2 polarization of macrophages [69]. modulation of bacterial functions such as translation [90] and growth
In conclusion, he diversity of microbiome needs to be addressed [91], and support of gut microbial diversity [88,89,92]. Physiological
specifically with respect to macrophage polarization since divergent or pathological deprivation of mucosal IgA responses may be associated
results have been reported in different experimental settings. with inflammatory diseases. IgA production is scarce in the young in-
Unraveling the finely tuned macrophage polarization will complement fant, and compensated by maternal IgA through breastfeeding [90].
our understanding of allergic diseases and support therapeutic inter- Abnormal gut IgA responses in the first year of life have been associated
vention through probiotics. with allergy and asthma occurrence at the age of 7 years [92]. Ab-
normalities include a lower proportion of IgA bound to fecal bacteria at
4. Mast cells at the crossroads of microbiome and allergy 12 months of age and altered IgA recognition patterns [92]. Moreover,
in undernourished children, IgA-coated strains from gut microbiota
There is increasing evidence of a relationship between allergic dis- were able to convey a diet-dependent enteropathy (disruption of the
order and microbiome. Several studies showed that microbiome mod- small intestinal and colonic epithelial barrier, weight loss, and sepsis),
ulates allergy by an alteration of the function of antigen presenting cells with Enterobacteriaceae playing a major role [89].
or the regulation of the Th1/Th2 balance [70,71]. However, there is At the other end of the lifespan, immunosenescence has been shown
increasing awareness of a direct modulation of mast cells (MCs) re- to affect the composition of gut mucosal IgA. Despite a stable amount of
sponses by microbiome [72,73]. In addition to their well-documented fecal IgA, IgA coating of some bacterial strains, namely Clostridiaceae
role in allergy, MCs possess multifunctional properties. First, they in- and Enterobacteriaceae with potential pathogenicity, was impaired [93].
fluence tissue integrity, function and homeostasis [74]. Second, MCs Many studies have focused on the role of microbiome in shaping the
are involved in innate and adaptive immune responses notably by their Th1/Th2 or the Th/Treg balance of adaptive responses to supposedly
Toll-like receptors (TLRs) and pattern recognition receptors [75]. unrelated non-microbial allergens. Much less attention has been paid to
Emerging data show a relationship between MCs and microbiome, with the direct interaction between B-lineage cells involved in allergy and
microbes altering MCs activation and function. In addition, the position commensal or opportunistic flora. It has been described in mouse and
of MCs as immune sentinels in skin and mucosal surfaces, that contain a human that early-life exposure to T independent antigens, such as
complex composite of non-pathogenic microbes, suggests the existence bacterial carbohydrates, appears critical for the establishment of certain

14
S. Mezouar et al. Human Microbiome Journal 10 (2018) 11–20

B-cell clonotype and imprints lastingly the antibody repertoire [94]. allergen) was associated with a higher proportion of E. coli and Bac-
Natural antibodies against bacterial glycans produced by these B-cell teroides pseudocatenulatum, and less Bacteroides breve, Bacteroides ado-
lineages are highly specific and confer effective humoral protection lescentis, F. prausnitzii, and Akkermansia muciniphila than counterparts
against bacteria bearing those carbohydrate moieties. Interestingly, without food allergy [107]. Enterobacteriaceae and selected Clostridiae
some of these antibodies cross-react between bacterial glycans (e.g. were also enriched in gut microbiome of Japanese food-allergic infants
from Streptococcus or Enterobacter cloacae) and allergens (e.g. Aspergillus [106]. A gut microbiome-wide study of infants aged 3–6 months iden-
fumigatus and house dust mite) bearing similar motifs and appear to be tified seven individual genera with significant inverse associations with
protective against allergic airway disease in mouse models [95]. The food sensitization and/or allergy [108]: Dorea (Lachnospiraceae), Ci-
mechanisms of this protective effect remain elusive, and it is yet unclear trobacter (Enterobacteriaceae), Lactococcus (Strepotococcaceae), Oscillos-
if these results apply in human. Nevertheless, cross-reactivity is a fea- pira (Ruminococcaceae), Dialister (Veillonellaceae), and Dialister (Pas-
ture of IgE known for decades. It may occur between several protein or teurellaceae).
carbohydrate allergens in sensitized individuals, reflecting similar epi-
topes on homologous molecules present in different species [96]. It
must be stressed that inter-allergen cross-reactive IgE are now believed 7. Skin microbiome and atopic dermatitis
to display poor biological activity, and no clinical relevance [97]. If IgE
cross reactivity happens between more or less phylogenetically distant Numerous microorganisms including bacteria, fungi, viruses, and
allergenic species, it should also occur in other species inasmuch as they mites make a living at the surface of human skin. Skin resident bacteria
share homologous determinants. Indeed, cross-reactivity has been are influenced by environmental biodiversity, but most of them belong
shown to happen between protein [98–101] and carbohydrate [102] to Firmicutes, Bacteroidetes, Proteobacteria and Actinobacteria [111].
allergens from insects and parasitic helminths. Similarly to inter-al- Together with keratinocyte-derived antimicrobial peptides, skin com-
lergen cross-reactive IgE, these IgE are not believed to be associated to mensals exert a protective action at the cutaneous level [112,113].
allergic manifestations [102] but could rather explain the negative as- Their disruption paves the way for the growth of pathogenic micro-
sociation between chronic helminth infections and skin prick test re- organisms and facilitates the attack of allergenic molecules. The mi-
activity. Antigenic cross-reactivity can be harnessed by the host’s im- crobiome-host crosstalk at the cutaneous level is established in early life
mune response in order to control microbiome components. For during a well-defined developmental window, with Langerhans cells
instance, flagellin from various bacterial species displays conserved and regulatory T cells (Tregs) setting up a non-allergic, low-level im-
motifs which are recognized through TLR-5, leading to the production mune response to skin commensals [111,114].
of flagellin-specific IgG and IgA that coat and inhibit bacterial AD can appear at any age but is usually one of the earliest mani-
spreading [90]. In another murine model, IgA coating identified in- festations of the atopic march, suggesting a pathophysiologic role for
flammatory commensals associated with inflammatory intestinal dis- abnormal microbiome implementation in neonates and infants. AD is a
eases [103]. frequent disease, with a lifetime prevalence of 10% to 20% [115]. The
However, IgA might play redundant roles with other Ig isotypes, as pathophysiology of AD is multifactorial, including genetic, im-
IgA deficiency does not markedly alter global fecal microbiota [104]. munological, microbial, and environmental factors [112]. Among bac-
In brief, indirect arguments suggest that early-life exposure to mi- teria, S. aureus has been prominently associated with AD [113]. In AD
crobiome determinants would be able to shape later-life B-cell re- skin, colonization with S. aureus is observed in association with a de-
pertoire and antibody response, enhancing production of potentially creased diversity of other bacterial species. Moreover, S. aureus-derived
protective IgE or non-IgE cross-reactive antibodies. δ-toxin induces skin mast cells to degranulate and produce Th2-related
cytokines, which in turn favor the development and persistence of al-
6. Microbiome and food allergy lergic responses [116,117]. Intrinsic skin defects such as filaggrin or
cathelicidin deficiency are also associated with an increased load of S.
Although gut microbiome is both the most abundant and the most aureus in the skin microbiome [112,115]. Finally, increased S. aureus
studied, the link between microbiome and food allergy is still elusive. A numbers in the gut microbiome of infants are predictors of current AD
relatively low prevalence of food allergy in general population, the [118].
multiplicity and the biochemical complexity of allergy-eliciting foods, Skin microbiome displays two prominent features: high site-de-
differences between food allergies occurring during childhood or in pendent variability and richness in fungal components. Among the
adults, the existence of a so-called developmental window for gut mi- latter, the yeast Malassezia is the predominant eukaryotic symbiont of
crobiome implementation and marked interindividual variability in human skin, and a growing body of evidence supports its implication in
clinical presentations of food allergies might explain why research in the development of inflammation and exacerbation associated with AD,
this field is lagging behind. A curious finding is the inconsistent evi- particularly the recognition of IgE binding allergens produced by
dence for decreased gut bacterial diversity in food-allergic patients. Malassezia and the selectivity of immune cells response [119]. Indeed,
Indeed, Azad [105] and Tanaka [106] described low richness of bac- both topical and systemic antifungal treatments reduce the severity of
terial communities in the gut of food-allergic infants, while Fieten skin symptoms in yeast-sensitized AD patients [120] and patients with
[107] and Savage [108] did not find differences in the diversity of fecal AD react more frequently to Malassezia extract or recombinant antigens
bacterial signatures in food-allergic children as compared with non- than healthy controls [121]. Several Malassezia species have been as-
allergic counterparts, and Fazlollahi [109] reported increased bacterial sociated with AD, with conflicting results among published studies.
diversity in gut microbiome of egg-allergic children. However, quali- However, the association of the species M. sympodialis with AD has been
tative bacterial signatures found in food-allergic patients versus non- most frequently pointed out (reviewed in ref. [119]). The major argu-
allergic controls are consistently different. In egg-allergic children, ment supporting this hypothesis is that M. sympodialis sensitization is
Lachnospiraceae (a Firmicutes division) and Streptococcaceae families specific for the clinical manifestations of AD and is absent in patients
were enriched, while Leuconostocaceae were enriched in controls [109]. with other allergic diseases [122]. Interestingly, Malassezia yeast trig-
Leuconostoccaceae have demonstrated protective effects against allergy gers divergent skin conditions where the inflammatory response is ei-
development in a mouse model of ovalbumin-induced food allergy, ther upregulated, such as in AD and seborrheic dermatitis, or down-
through mechanisms resulting in a decrease in IgE production and an regulated, such as in pityriasis versicolor [123]. Complex interactions
increase in interferon-gamma production [110]. A diminished propor- between Malassezia yeasts and other communities of the skin micro-
tion of Leuconostoccaceae has been reported in food-allergic Japanese biome might contribute to the pathogenesis of Malassezia-related skin
infants [106]. In children with AD, concomitant food allergy (any diseases.

15
S. Mezouar et al. Human Microbiome Journal 10 (2018) 11–20

Fig. 2. Microbiome in asthma. The main pathophysiological events involving microbiome are depicted from the prenatal stage of development to the established
pulmonary disease (asthma).

8. Microbiome and allergic diseases of the airways disease but also on the type of medication [132]. Low abundance mi-
crobial pathogens can orchestrate inflammatory disease by remodeling
8.1. Microbiome in asthma (Fig. 2) a normally benign microbiome into a dysbiotic one.
Dysbiosis has also been reported for the respiratory mycobiome of
Airway and gut microbiology may play an important role in the SA patients. A higher density of genera including Rhodosporidium,
pathobiology of chronic airway diseases such as severe asthma (SA) Pneumocystis, Leucosporidium, and Rhodotorula has been show in the
[124]. Airway microbiome is certainly important at the inception of respiratory tract of patients with SA compared with non-asthmatic
asthma as it was found that early in life, the occurrence of bacteria patients, in whom Davidiella, Cryptococcus, and Sterigmatomyces were
species drove the IS towards an asthma susceptible phenotype related to relatively more abundant [133]. In patients with fungal asthma, corti-
the promotion of PDL1 harboring cells [125]. T regulatory cells are costeroid treatment was associated with an increased fungal burden in
finely tuned by an early contact with lung microbiome leading to the the bronchoalveolar lavage, with the Aspergillus fumigatus complex
prevention of susceptibility for asthma [125]. Similarly, early farm dust being the most prevalent [134].
exposure decreases airway susceptibility to asthma through the reg- Overall, the research investigating the relationships between lung
ulation of the ubiquitin A20 which is severely down expressed in microbiome and asthma is paramount to better understand and treat
bronchial epithelial cells (BEC) derived from SA patients [126]. Ex- patients suffering from these devastating diseases. 8.1.
posure to endotoxin in vitro abrogates the proinflammatory response of
BEC from SA exposed to house dust mites [126]. 8.2. Microbiome and asthma in children
At steady state, airway microbiome in SA is different from the
bacterial diversity observed in the normal lung and in lungs of mild-to- Asthma, which has increased in the past few decades, affects one in
moderate asthma patients. Patients with SA were significantly enriched ten children in westernized countries. Albeit pediatric asthma is a
in Klebsiella spp and Actinobacteria [127]. These findings have been multifactorial disease, accumulating evidence suggests that exposure to
reproduced using various sampling methods from bronchoalveolar la- a diverse environmental microbiome in early life has a protective effect
vage to sputum analysis, and from childhood to adulthood [128,129]. against childhood asthma [135,136]. Birth by Caesarean section, for-
Heterogeneity of airway microbiome has been recently related to in- mula-feeding, urban environment, lack of sustained contacts with a
flammatory phenotypes in SA [130]. Non-eosinophilic SA phenotype diversity of humans and animals, and antibiotic intake are clearly as-
was associated with a relative increase in the abundance of γ-Proteo- sociated with an increased risk to develop asthma and allergies later in
bacteria and a reduced load of Streptococcus, Gemella and Porphyr- life. These factors modify the gut fungal and bacterial microbiome
omonas [130]. This is a relevant finding since non-eosinophilic SA is (dysbiosis), particularly during the first 100 days of life which represent
related to Th17 inflammation and poorly responsive to corticosteroid the critical window for acquiring a Th2-lymphocyte profile. Analysis of
therapy. These findings are confirmatory along with the non-response gut microbiota in a US cohort showed that distinct groups, character-
of airway macrophages derived from SA patients to dexamethasone ized by distinct relative risks of atopy and asthma at later stages, could
inhibition, after an exposure to Haemophilus influenzae in vitro [131]. At be identified already during the neonatal period [137]. For instance,
exacerbation, SA microbiome changes and contributes to the severity of the group with the highest risk of multisensitized atopy at age 2 years
the event. Several reports show the major impact of treatments on lung and doctor-diagnosed asthma at age 4 years displayed lower relative
microbiome. It was found that antibiotic use affects the lung micro- abundance of Bifidobacterium, Akkermansia and Faecalibacterium, higher
biome, leading to an increase of alpha diversity and Proteobacteria and relative abundance of fungi such as Candida and Rhodotorula and a pro-
decrease of Firmicutes. A change in microbiome is induced by corti- inflammatory fecal metabolome [137]. Conversely, pre- and post-natal
costeroids alone with an increase, at the genus level, of Haemophilus and exposure to microbiome-loaded farm environment overrides the effect
Moraxella. The microbiome of patients treated with inhaled corticos- of risk alleles in chromosome 17q21 which expose to asthma develop-
teroids and bronchodilators was different from that of untreated pa- ment following rhinovirus respiratory infections. Early colonization of
tients demonstrating that the microbiome is not only dependent on the the airways by Streptococci and Moraxella predispose for recurrent

16
S. Mezouar et al. Human Microbiome Journal 10 (2018) 11–20

Fig. 3. Microbiome-host interplay, main checkpoints and external influences. The host microbiome crosstalk is depicted through its main functional checkpoints
which can be disturbed by external influences or loss of internal homeostasis.

wheezing respiratory episodes and asthma. When asthma is declared, antimicrobial treatment. After therapy, the increase in diversity is ac-
the airway microbiome differs from that of healthy children, with a companied by a decrease in the total abundance of the bacterial po-
usually higher bacterial density. A higher fungal density has also been pulation [145]. However, the role of the sinonasal microbiome in
observed in more severe 9-year-old asthmatic children (increased re- chronic rhinosinusitis (CRS) remains unclear. CRS is a chronic in-
presentation of Bacteroides and Pneumocystis) [133]. Allergy may flammatory disease of the mucosae of the upper airways including the
modify this tendency as the global bacterial species richness and di- nose and paranasal sinuses, often comorbid with asthma and linked to
versity were significantly reduced in 4-year old mites-sensitized asth- increased asthma severity and exacerbation rate [146].
matics in comparison to healthy non-sensitized children, with an A recent study reported that among 111 CRS cases, 46 (41.4%) had
overrepresentation of Moraxella and Leptotrichia [138]. In the future, concurrent physician-diagnosed asthma [147]. Asthmatic patients had
acting on the microbiome, for instance with fecal microbiome trans- higher rates of allergic rhinitis (60.9% asthmatic vs. 26.2% non-asth-
plantation, may be of interest to prevent the development of asthma in matic, p < 0.05). In this study, asthmatic CRS had significantly higher
children or even to manage their asthma. Another axis of research deals relative abundance (RA) of the Streptococcus genus. Moreover, asthma-
with antibiotic use during the perinatal window. At least in murine related exacerbations as reflected by emergency department visits were
models [139], not all antibiotics are associated with an increased risk associated with significant nasal microbiome changes, including higher
on allergic disease later in life. In their model, vancomycine (a Gram- RA of Proteobacteria phylum, notably Burkholderia spp [147].
positive targeting antibiotic) but not streptomycine (targeting both The presence of microbial dysbiosis in CRS is now supported by
Gram-positive and Gram-negative bacteria)-treated mouse pups were many studies but it is still impossible to assess whether this dysbiosis is
prone to experimental asthma. a cause or rather an association of the disease process [144]. Although
probiotic therapies showed early promise, larger studies are required to
establish their real role as a treatment for CRS [144].
8.3. Microbiome and rhinosinusitis

In the field of oto-rhino-laryngology the microbiome has mainly 9. Concluding remarks


been studied in the sinonasal tract. Rhinitis and asthma share key ele-
ments of pathogenesis and have long been noted to co-occur, in support We have aimed at providing the reader with an overview of the
of the unified airway model: microbial, epithelial, and immune con- current knowledge in the field of microbiome – IS crosstalk, with spe-
tinuity from one section of the respiratory tract to the others [140]. In cial emphasis on the unsteady balance associated with health. Dysbiosis
healthy condition rich and diverse bacterial assemblages are present in is associated with dysimmunity, often characterized as a Th2-excessive,
the sinonasal cavity [141]. In CRS, molecular analyses revealed a large Treg-deficient state. Allergy and asthma are typically associated with
amount of interpersonal variation between patients [142]. Analysis of such immune deviations. The intervention of microbiome during the
microbiome in a large cohort revealed that different CRS associated development of allergies and asthma is still incompletely described, and
with asthma or with purulence are characterized by distinct composi- the description of dynamic aspects of microbial communities and their
tions of resident bacterial communities. According to Ramakrishnan shaping of the host IS is still in infancy. The perinatal window of op-
et al. the bacterial diversity and composition are predictors of surgical portunity has gained momentum as a concept, but general-population
outcome [143]. CRS patients show less biodiversity in their microbiome cohorts over long periods of time are lacking in most countries, and
than healthy subjects [144]. Significant differences exist in the diversity studies combining prenatal and postnatal data are hampered by ethical,
of bacteria populations during acute exacerbations of CRS and after material, and social constraints.

17
S. Mezouar et al. Human Microbiome Journal 10 (2018) 11–20

The current understanding of pathophysiologic checkpoints is [18] Underhill DM, Iliev ID. The mycobiota: interactions between commensal fungi and
summarized in Fig. 3. Research in this field is hampered by the multi- the host immune system. Nat Rev Immunol 2014;14:405–16.
[19] Iliev ID, Funari VA, Taylor KD, Nguyen Q, Reyes CN, Strom SP, et al. Interactions
plicity of multifactorial determinants of allergic/atopic diseases and the between commensal fungi and the C-type lectin receptor Dectin-1 influence colitis.
patchy knowledge of mechanistic endotypes underlying clinically de- Science 2012;336:1314–7.
fined phenotypes. Harnessing the microbiome – IS interaction is cur- [20] Wheeler ML, Limon JJ, Bar AS, Leal CA, Gargus M, Tang J, et al. Immunological
consequences of intestinal fungal dysbiosis. Cell Host Microbe 2016;19:865–73.
rently limited to pre/probiotic supplementation and environmental [21] Ott SJ, Kühbacher T, Musfeldt M, Rosenstiel P, Hellmig S, Rehman A, et al. Fungi
interventions. Large-scale future studies on birth and adult cohorts, and inflammatory bowel diseases: Alterations of composition and diversity. Scand
taking advantage of the most recent tools in microbiology, im- J Gastroenterol 2008;43:831–41.
[22] Hoarau G, Mukherjee PK, Gower-Rousseau C, Hager C, Chandra J, Retuerto MA,
munology, allergology, and statistics are highly needed. Novel ap- et al. Bacteriome and mycobiome interactions underscore microbial dysbiosis in
proaches also imply breaking previous categories. For instance, trans- familial Crohn's disease. MBio 2016;7(5)..
kingdom analysis has been proposed in order to address the complexity [23] Sokol H, Leducq V, Aschard H, Pham HP, Jegou S, Landman C, et al. Fungal mi-
crobiota dysbiosis in IBD. Gut 2017;66:1039–48.
of antibiotic-induced alterations [148]. We are at the dawn of a new
[24] Kim SH, Clark ST, Surendra A, Copeland JK, Wang PW, Ammar R, et al. Global
global perspective of analyzing the complex interactions between the analysis of the fungal microbiome in cystic fibrosis patients reveals loss of function
host’s immune defenses and its resident microbiome and between the of the transcriptional repressor Nrg1 as a mechanism of pathogen adaptation. PLoS
communities within the host’s microbiome. This approach should sig- Pathog 2015;11:e1005308.
[25] Platts-Mills TA. The allergy epidemics: 1870–2010. J Allergy Clin Immunol
nificantly enhance our deciphering of their influence on human health 2015;136:3–13.
in general and, particularly, on allergic diseases. [26] Strachan DP. Hay fever, hygiene, and household size. BMJ 1989;299:1259–60.
[27] Stein MM, Hrusch CL, Gozdz J, Igartua C, Pivniouk V, Murray SE, et al. Innate
immunity and asthma risk in Amish and Hutterite farm children. N Engl J Med
Declaration of interest 2016;375:411–21.
[28] Ober C, Sperling AI, von Mutius E, Vercelli D. Immune development and en-
None. vironment: lessons from Amish and Hutterite children. Curr Opin Immunol
2017;48:51–60.
[29] Cecchi L, D'Amato G, Annesi-Maesano I. External exposome and allergic re-
Funding spiratory and skin diseases. J Allergy Clin Immunol 2018;141:846–57.
[30] Arrieta MC, Arévalo A, Stiemsma L, Dimitriu P, Chico ME, Loor S, et al.
Associations between infant fungal and bacterial dysbiosis and childhood atopic
No specific funding was allocated for this paper. wheeze in a nonindustrialized setting. J Allergy Clin Immunol
2018;142(424–34):e10.
Conflict of interest [31] Momose Y, Hirayama K, Itoh K. Effect of organic acids on inhibition of Escherichia
coli O157:H7 colonization in gnotobiotic mice associated with infant intestinal
microbiota. Antonie Van Leeuwenhoek 2008;93:141–9.
None. [32] Momose Y, Hirayama K. Itoh K. Competition for proline between indigenous
Escherichia coli and E. coli O157:H7 in gnotobiotic mice associated with infant
Appendix A. Supplementary data intestinal microbiota and its contribution to the colonization resistance against E.
coli O157:H7. Antonie Van Leeuwenhoek 2008;94:165–71.
[33] Fukuda S, Toh H, Hase K, Oshima K, Nakanishi Y, Yoshimura K, et al.
Supplementary data to this article can be found online at https:// Bifidobacteria can protect from enteropathogenic infection through production of
doi.org/10.1016/j.humic.2018.10.001. acetate. Nature 2011;469:543–7.
[34] Gantois I, Ducatelle R, Pasmans F, Haesebrouck F, Hautefort I, Thompson A, et al.
Butyrate specifically downregulates salmonella pathogenicity island 1 gene ex-
References pression. Appl Environ Microbiol 2006;72:946–9.
[35] Pacheco AR, Curtis MM, Ritchie JM, Munera D, Waldor MK, Moreira CG, et al.
Fucose sensing regulates bacterial intestinal colonization. Nature 2012;492:113–7.
[1] Lloyd-Price J, Abu-Ali G, Huttenhower C. The healthy human microbiome.
[36] Corr SC, Li Y, Riedel CU, O'Toole PW, Hill C, Gahan CG. Bacteriocin production as
Genome Med 2016;8:51.
a mechanism for the anti-infective activity of Lactobacillus salivarius UCC118.
[2] Sender R, Fuchs S, Milo R. Revised estimates for the number of human and bacteria
PNAS 2007;104:7617–21.
cells in the body. PLoS Biol 2016;14(8):e1002533.
[37] Sommer F, Bäckhed F. The gut microbiota–masters of host development and
[3] Geva-Zatorsky N, Sefik E, Kua L, Pasman L, Tan TG, Ortiz-Lopez A, et al. Mining
physiology. Nat Rev Microbiol 2013;11:227–38.
the human gut microbiota for immunomodulatory organisms. Cell
[38] Bauer H, Horowitz RE, Levenson SM, Popper H. The response of the lymphatic
2017;168(928–43):e11.
tissue to the microbial flora. Studies on germfree mice. Am J Pathol
[4] Gupta VK, Paul S, Dutta C. Geography, ethnicity or subsistence-specific variations
1963;42:471–83.
in human microbiome composition and diversity. Front Microbiol 2017;8:1162.
[39] Macpherson AJ, Harris NL. Interactions between commensal intestinal bacteria
[5] Groussin M, Mazel F, Sanders JG, Smillie CS, Lavergne S, Thuiller W, et al.
and the immune system. Nat Rev Immunol 2004;4:478–85.
Unraveling the processes shaping mammalian gut microbiomes over evolutionary
[40] Mazmanian SK, Liu CH, Tzianabos AO, Kasper DL. An immunomodulatory mole-
time. Nat. Commun. 2017;8:14319.
cule of symbiotic bacteria directs maturation of the host immune system. Cell
[6] Moeller AH, Caro-Quintero A, Mjungu D, Georgiev AV, Lonsdorf EV, Muller MN,
2005;122:107–18.
et al. Co-speciation of gut microbiome with hominids. Science 2016;353:380–2.
[41] Round JL, Mazmanian SK. Inducible Foxp3+ regulatory T-cell development by a
[7] Moeller AH. The shrinking human gut microbiome. Curr Opin Microbiol
commensal bacterium of the intestinal microbiota. PNAS 2010;107:12204–9.
2017;38:30–5.
[42] Tanoue T, Umesaki Y, Honda K. Immune responses to gut microbiota-commensals
[8] Clayton JB, Vangay P, Huang H, Ward T, Hillmann BM, Al-Ghalith GA, et al.
and pathogens. Gut Microbes 2010;1:224–33.
Captivity humanizes the primate microbiome. PNAS 2016;113:10376–81.
[43] Petursdottir DH, Nordlander S, Qazi KR, Carvalho-Queiroz C, Ahmed Osman O,
[9] Horz HP. Archaeal lineages within the human microbiome: Absent, Rare or
Hell E, et al. Early-Life Human Microbiota Associated With Childhood Allergy
Elusive? Life (Basel) 2015;5:1333–45.
Promotes the T Helper 17 Axis in Mice. Front Immunol 2017 Dec;1(8):1699.
[10] Morton ER, Lynch J, Froment A, Lafosse S, Heyer E, Przeworski M, et al. Variation
[44] Hansson GC, Johansson ME. The inner of the two Muc2 mucin-dependent mucus
in rural African gut microbiome is strongly correlated with colonization by en-
layers in colon is devoid of bacteria. Gut Microbes 2010;1:51–4.
tamoeba and subsistence. PLoS Genet 2015;11:e1005658.
[45] Bergström JH, Birchenough GM, Katona G, Schroeder BO, Schütte A, Ermund A,
[11] Zaiss MM, Harris NL. Interactions between the intestinal microbiome and helminth
et al. Gram-positive bacteria are held at a distance in the colon mucus by the
parasites. Parasite Immunol 2016;38:5–11.
lectin-like protein ZG16. PNAS 2016;113:13833–8.
[12] Raymann K, Moeller AH, Goodman AL. Ochman H. Unexplored archaeal diversity
[46] Salzman NH, Underwood MA, Bevins CL. Paneth cells, defensins, and the com-
in the great ape gut microbiome. mSphere 2017;2(1).
mensal microbiota: a hypothesis on intimate interplay at the intestinal mucosa.
[13] Seed PC. The human mycobiome. Cold Spring Harb Perspect Med 2014;5:a019810.
Semin Immunol 2007;19:70–83.
[14] Halwachs B, Madhusudhan N, Krause R, Nilsson RH, Moissl-Eichinger C,
[47] Cash HL, Whitham CV, Behrendt CL, Hooper LV. Symbiotic bacteria direct ex-
Högenauer C, et al. Critical issues in mycobiota analysis. Front Microbiol
pression of an intestinal bactericidal lectin. Science 2006;313:1126–30.
2017;8:180.
[48] Macpherson AJ, Uhr T. Induction of protective IgA by intestinal dendritic cells
[15] Oever JT, Netea MG. The bacteriome-mycobiome interaction and antifungal host
carrying commensal bacteria. Science 2004;303:1662–5.
defense. Eur J Immunol 2014;44:3182–91.
[49] Sonnenberg GF, Monticelli LA, Alenghat T, Fung TC, Hutnick NA, Kunisawa J,
[16] Gouba N, Drancourt M. Digestive tract mycobiota: a source of infection. Med Mal
et al. Innate lymphoid cells promote anatomical containment of lymphoid-resident
Infect 2015;45:9–16.
commensal bacteria. Science 2012;336:1321–5.
[17] Belkaid Y, Naik S. Compartmentalized and systemic control of tissue immunity by
[50] Mao K, Baptista AP, Tamoutounour S, Zhuang L, Bouladoux N, Martins AJ, et al.
commensals. Nat Immunol 2013;14:646–53.
Innate and adaptive lymphocytes sequentially shape the gut microbiota and lipid

18
S. Mezouar et al. Human Microbiome Journal 10 (2018) 11–20

metabolism. Nature 2018;554:255–9. 2012;189:1551–8.


[51] Macpherson AJ, Uhr T. Compartmentalization of the mucosal immune responses to [83] Nakamura Y, Oscherwitz J, Cease KB, Chan SM, Munoz-Planillo R, Hasegawa M,
commensal intestinal bacteria. Ann N Y Acad Sci 2004;1029:36–43. et al. Staphylococcus delta-toxin induces allergic skin disease by activating mast
[52] Gordon S, Martinez FO. Alternative activation of macrophages: mechanism and cells. Nature 2013;503:397–401.
functions. Immunity 2010;32:593–604. [84] Akdis M, Akdis CA. Mechanisms of allergen-specific immunotherapy: multiple
[53] Sica A, Mantovani A. Macrophage plasticity and polarization: in vivo veritas. J Clin suppressor factors at work in immune tolerance to allergens. J Allergy Clin
Invest 2012;122:787–95. Immunol 2014;133:621–31.
[54] Ka MB, Daumas A, Textoris J, Mege JL. Phenotypic diversity and emerging new [85] Schwarz A, Panetta V, Cappella A, Hofmaier S, Hatzler L, Rohrbach A, et al. IgG
tools to study macrophage activation in bacterial infectious diseases. Front and IgG4 to 91 allergenic molecules in early childhood by route of exposure and
Immunol 2014;5:500. current and future IgE sensitization: Results from the Multicentre Allergy Study
[55] Murray PJ, Allen JE, Biswas SK, Fisher EA, Gilroy DW, Goerdt S, et al. Macrophage birth cohort. J Allergy Clin Immunol 2016;138:1426–33.
activation and polarization: nomenclature and experimental guidelines. Immunity [86] Hofmaier S, Hatzler L, Rohrbach A, Panetta V, Hakimeh D, Bauer CP, et al.
2014;41:14–20. Default“ versus ”pre-atopic“ IgG responses to foodborne and airborne pathogen-
[56] Benoit M, Desnues B, Mege JL. Macrophage polarization in bacterial infections. J esis-related group 10 protein molecules in birch-sensitized and nonatopic children.
Immunol 2008;181:3733–9. J Allergy Clin Immunol 2015;135. 1367-74.e1-8.
[57] Smith PD, Ochsenbauer-Jambor C, Smythies LE. Intestinal macrophages: unique [87] Aalberse RC, Lupinek C, Siroux V, Nadif R, Just J, Bousquet J, et al. sIgE and sIgG
effector cells of the innate immune system. Immunol Rev 2005;206:149–59. to airborne atopic allergens: coupled rather than inversely related responses.
[58] Ivanov II, Honda K. Intestinal commensal microbes as immune modulators. Cell Allergy 2018 Jul 10. https://doi.org/10.1111/all.13548.
Host Microbe 2012;12:496–508. [88] Corthesy B. Multi-faceted functions of secretory IgA at mucosal surfaces. Front
[59] Chang PV, Hao L, Offermanns S, Medzhitov R. The microbial metabolite butyrate Immunol 2013;4:185.
regulates intestinal macrophage function via histone deacetylase inhibition. PNAS [89] Kau AL, Planer JD, Liu J, Rao S, Yatsunenko T, Trehan I, et al. Functional char-
2014;111:2247–52. acterization of IgA-targeted bacterial taxa from undernourished Malawian children
[60] Danne C, Ryzhakov G, Martínez-López M, Ilott NE, Franchini F, Cuskin F, et al. A that produce diet-dependent enteropathy. Sci Transl Med 2015;7(276). 276ra24.
Large polysaccharide produced by Helicobacter hepaticus induces an anti-in- [90] Cullender TC, Chassaing B, Janzon A, Kumar K, Muller CE, Werner JJ, et al. Innate
flammatory gene signature in macrophages. Cell Host Microbe 2017;22:733–45. and adaptive immunity interact to quench microbiome flagellar motility in the gut.
[61] Yang Y, Wang X, Huycke T, Moore DR, Lightfoot SA, Huycke MM. Colon macro- Cell Host Microbe 2013;14:571–81.
phages polarized by commensal bacteria cause colitis and cancer through the [91] Moor K, Diard M, Sellin ME, Felmy B, Wotzka SY, Toska A, et al. High-avidity IgA
bystander effect. Transl Oncol 2013;6:596–606. protects the intestine by enchaining growing bacteria. Nature 2017;544:498–502.
[62] Seo SU, Kamada N, Muñoz-Planillo R, Kim YG, Kim D, Koizumi Y, et al. Distinct [92] Dzidic M, Abrahamsson TR, Artacho A, Björkstén B, Collado MC, Mira A, et al.
commensals induce interleukin-1β via NLRP3 inflammasome in inflammatory Aberrant IgA responses to the gut microbiota during infancy precede asthma and
monocytes to promote intestinal inflammation in response to injury. Immunity allergy development. J Allergy Clin Immunol 2017;139(3):1017–1025.e14.
2015;42:744–55. [93] Sugahara H, Okai S, Odamaki T, Wong CB, Kato K, Mitsuyama E, et al. Decreased
[63] Croft AJ, Metcalfe S, Honma K, Kay JG. Macrophage polarization alters post- taxon-specific IgA response in relation to the changes of gut microbiota compo-
phagocytosis survivability of the commensal Streptococcus gordonii. Infect Immun sition in the elderly. Front Microbiol 2017(8). 1757.94.
2018;86(3). pii: e00858-17. [94] Kearney JF, Patel P, Stefanov EK, King RG. Natural antibody repertoires: devel-
[64] Hayashi A, Sato T, Kamada N, Mikami Y, Matsuoka K, Hisamatsu T, et al. A single opment and functional role in inhibiting allergic airway disease. Annu Rev
strain of Clostridium butyricum induces intestinal IL-10-producing macrophages to Immunol 2015;33:475–504.
suppress acute experimental colitis in mice. Cell Host Microbe 2013;13:711–22. [95] Patel PS, Kearney JF. Neonatal exposure to pneumococcal phosphorylcholine
[65] Jang SE, Hyam SR, Han MJ, Kim SY, Lee BG, Kim DH. Lactobacillus brevis G-101 modulates the development of house dust mite allergy during adult life. J Immunol
ameliorates colitis in mice by inhibiting NF-κB, MAPK and AKT pathways and by 2015;194:5838–50.
polarizing M1 macrophages to M2-like macrophages. J Appl Microbiol [96] McKenna OE, Asam C, Araujo GR, Roulias A, Goulart LR, Ferreira F. How relevant
2013;115:888–96. is pan allergen sensitization in the development of allergies? Pediatr Allergy
[66] Christoffersen TE, Hult LT, Kuczkowska K, Moe KM, Skeie S, Lea T, et al. In vitro Immunol 2016;27:560–8.
comparison of the effects of probiotic, commensal and pathogenic strains on [97] Mari A, Ooievaar-de Heer P, Scala E, Giani M, Pirrotta L, Zuidmeer L, et al.
macrophage polarization. Probiotics Antimicrob Proteins 2014;6:1–10. Evaluation by double-blind placebo controlled oral challenge of the clinical re-
[67] Habil N, Al-Murrani W, Beal J, Foey AD. Probiotic bacterial strains differentially levance of IgE antibodies against plant glycans. Allergy 2008;63:891–6.
modulate macrophage cytokine production in a strain-dependent and cell subset- [98] Acevedo N, Sanchez J, Erler A, Mercado D, Briza P, Kennedy M, et al. IgE cross-
specific manner. Benef Microbes 2011;2:283–93. reactivity between Ascaris and domestic mite allergens: the role of tropomyosin
[68] Mège JL, Mehraj V, Capo C. Macrophage polarization and bacterial infections. Curr and the nematode polyprotein ABA-1. Allergy 2009;64:1635–43.
Opin Infect Dis 2011;24:230–4. [99] Santiago HC, LeeVan E, Bennuru S, Ribeiro-Gomes F, Mueller E, Wilson M, et al.
[69] Kim YG, Udayanga KG, Totsuka N, Weinberg JB, Núñez G, Shibuya A. Gut dys- Molecular mimicry between cockroach and helminth glutathione S-transferases
biosis promotes M2 macrophage polarization and allergic airway inflammation via promotes cross-reactivity and cross-sensitization. J Allergy Clin Immunol
fungi-induced PGE₂. Cell Host Microbe 2014;15:95–102. 2012;130:248–56.
[70] Swiatczak B, Rescigno M. How the interplay between antigen presenting cells and [100] Santos AB, Rocha GM, Oliver C, Ferriani VP, Lima RC, Palma MS, et al. Cross-
microbiome tunes host immune responses in the gut. Semin Immunol reactive IgE antibody responses to tropomyosin from Ascaris lumbricoides and
2012;24:43–9. cockroach. J Allergy Clin Immunol 2008;121:1040–6.
[71] Wu HJ, Wu E. The role of gut microbiome in immune homeostasis and auto- [101] Valmonte GR, Cauyan GA, Ramos JD. IgE cross-reactivity between house dust mite
immunity. Gut Microbes 2012;3:4–14. allergens and Ascaris lumbricoides antigens. Asia Pac Allergy 2012;2:35–44.
[72] Forsythe P. Microbes taming mast cells: Implications for allergic inflammation and [102] Amoah AS, Obeng BB, Larbi IA, Versteeg SA, Aryeetey Y, Akkerdaas JH, et al.
beyond. Eur J Pharmacol 2016;778(73):169–75. Peanut-specific IgE antibodies in asymptomatic Ghanaian children possibly caused
[73] Hershko AY. Insights into the mast cell-microbiome connection in the skin. J by carbohydrate determinant cross-reactivity. J Allergy Clin Immunol
Allergy Clin Immunol 2017;139:1137–9. 2013;132:639–47.
[74] Galli SJ, Tsai M. Mast cells: Versatile regulators of inflammation, tissue re- [103] Palm NW, de Zoete MR, Cullen TW, Barry NA, Stefanowski J, Hao L, et al.
modeling, host defense and homeostasis. J Dermatol Sci 2008;49:7–19. Immunoglobulin A coating identifies colitogenic bacteria in inflammatory bowel
[75] Vliagoftis H, Befus AD. Rapidly changing perspectives about mast cells at mucosal disease. Cell 2014;158:1000–10.
surfaces. Immunol Rev 2005;206:190–203. [104] Fadlallah J, El Kafsi H, Sterlin D, Juste C, Parizot C, Dorgham K, et al. Microbial
[76] Bashir MEE, Louie HN, Shi HN, Nagler-Anderson C. Toll-like receptor 4 signaling ecology perturbation in human IgA deficiency. Sci Transl Med
by intestinal microbes’ influences susceptibility to food allergy. J Immunol 2018;10(439).;pii:eaan1217.
2004;172:6978–87. [105] Azad MB, Konya T, Guttman DS, Field CJ, Sears MR, HayGlass KT, et al. Infant gut
[77] Sato H, Zhang LS, Martinez K, Chang EB, Yang Q, Wang F, et al. Antibiotics sup- microbiota and food sensitization: associations in the first year of life. Clin Exp
press activation of intestinal mucosal mast cells and reduce dietary lipid absorp- Allergy 2015;45:632–43.
tion in Sprague-Dawley Rats. Gastroenterology 2016;151:923–32. [106] Tanaka M, Korenori Y, Washio M, Kobayashi T, Momoda R, Kiyohara C, et al.
[78] Kasakura K, Takahashi K, Hosono A, Itoh K, Nishiyama C, Kaminogawa S. Signatures in the gut microbiota of Japanese infants who developed food allergies
Commensal bacteria directly suppress in vitro degranulation of mast cells in in early childhood. FEMS Microbiol Ecol 2017;93.
MyD88-independent manner. Biosci Biotechnol Biochem 2014;78:1669–76. [107] Fieten KB, Totté JEE, Levin E, Reyman M, Meijer Y, Knulst A, et al. Fecal micro-
[79] Rodriguez B, Prioult G, Bibiloni R, Nicolis I, Mercenier A, Butel MJ, et al. Germ- biome and food allergy in pediatric atopic dermatitis: A cross-sectional pilot study.
free status and altered caecal subdominant microbiome are associated with a high Int Arch Allergy Immunol 2018;175:77–84.
susceptibility to cow’s milk allergy in mice. FEMS Microbiol Ecol 2011;76:133–44. [108] Savage JH, Lee-Sarwar KA, Sordillo J, Bunyavanich S, Zhou Y, O'Connor G, et al. A
[80] Wang JH, Fan SW, Zhu WY. Development of gut microbiome in a mouse model of prospective microbiome-wide association study of food sensitization and food
ovalbumin-induced allergic diarrhea under sub-barrier system. Asian-Aus J Anim allergy in early childhood. Allergy 2018;73. 145–52.9.
Sci 2013;26:545–51. [109] Fazlollahi M, Chun Y, Grishin A, Wood RA, Burks AW, Dawson P, et al. Early-life
[81] Wang Z, Mascarenhas N, Eckmann L, Miyamoto Y, Sun X, Kawakami T, et al. Skin gut microbiome and egg allergy. Allergy 2018;73:1515–24.
Microbiome promotes mast cell maturation by triggering stem cell factor pro- [110] Kang H, Oh YJ, Ahn KS, Eom HJ, Han N, Kim YB, et al. Leuconostoc citreum HJ-P4
duction in keratinocytes. J Allergy Clin Immunol 2017;139:1205–16. (KACC 91035) regulates immunoglobulin E in an ovalbumin-induced allergy
[82] Wang Z, MacLeod DT, Di Nardo A. Commensal bacteria lipoteichoic acid increases model and induces interleukin-12 through nuclear factor kappa B and p38/c-Jun
skin mast cell antimicrobial activity against vaccinia viruses. J Immunol N-terminal kinases signaling in macrophages. Microbiol Immunol 2009;53:331–9.

19
S. Mezouar et al. Human Microbiome Journal 10 (2018) 11–20

[111] Prescott SL, Larcombe DL, Logan AC, West C, Burks W, Caraballo L, et al. The skin Inflammatory phenotypes in patients with severe asthma are associated with
microbiome: impact of modern environments on skin ecology, barrier integrity, distinct airway microbiology. J Allergy Clin Immunol 2018;141(94–103):e15.
and systemic immune programming. World Allergy Organ J 2017;10:29. [131] Goleva E, Jackson LP, Harris JK, Robertson CE, Sutherland ER, Hall CF, et al. The
[112] Clausen ML, Slotved HC, Krogfelt KA, Andersen PS, Agner T. In vivo expression of effects of airway microbiome on corticosteroid responsiveness in asthma. Am J
antimicrobial peptides in atopic dermatitis. Exp Dermatol 2016;25:3–9. Respir Crit Care Med 2013;188:1193–201.
[113] Knaysi G, Smith AR, Wilson JM, Wisniewski JA. The skin as a route of allergen [132] Durack J, Lynch SV, Nariya S, Bhakta NR, Beigelman A, Castro M, et al. Features of
exposure: Part II. Allergens and role of the microbiome and environmental ex- the bronchial bacterial microbiome associated with atopy, asthma, and respon-
posures. Curr Allergy Asthma Rep 2017;17:7. siveness to inhaled corticosteroid treatment. J Allergy Clin Immunol
[114] Zeeuwen PL, Kleerebezem M, Timmerman HM, Schalkwijk J. Microbiome and skin 2017;140:63–75.
diseases. Curr Opin Allergy Clin Immunol 2013;13:514–20. [133] Goldman DL, Chen Z, Shankar V, Tyberg M, Vicencio A, Burk R. Lower airway
[115] Weidinger S, Novak N. Atopic dermatitis. Lancet 2016;387:1109–22. microbiota and mycobiota in children with severe asthma. J Allergy Clin Immunol
[116] Nakamura Y, Oscherwitz J, Cease KB, Chan SM, Muñoz-Planillo R, Hasegawa M, 2018;141(808–11):e7.
et al. Staphylococcus delta-toxin induces allergic skin disease by activating mast [134] Fraczek MG, Chishimba L, Niven RM, Bromley M, Simpson A, Smyth L, et al.
cells. Nature 2013;503:397–401. Corticosteroid treatment is associated with increased filamentous fungal burden in
[117] Nakatsuji T, Chen TH, Two AM, Chun KA, Narala S, Geha RS, et al. Staphylococcus allergic fungal disease. J Allergy Clin Immunol 2018;142:407–14.
aureus exploits epidermal barrier defects in atopic dermatitis to trigger cytokine [135] Jatzlauk G, Bartel S, Heine H, Schloter M. Krauss-Etschmann. Influences of en-
expression. J, Invest Dermatol 2016;136:2192–200. vironmental bacteria and their metabolites on allergies, asthma, and host mi-
[118] Watanabe S, Narisawa Y, Arase S, Okamatsu H, Ikenaga T, Tajiri Y, et al. crobia. Allergy 2017;72:1859–67.
Differences in fecal microflora between patients with atopic dermatitis and [136] Martinez FD, Guerra S. Early origins of asthma: role of microbial dysbiosis and
healthy control subjects. J Allergy Clin Immunol 2003;111:587–91. metabolic dysfunction. Am J Respir Crit Care Med 2018;197:573–9.
[119] Prohic A, Jovovic Sadikovic T, Krupalija-Fazlic M, Kuskunovic-Vlahovljak S. [137] Fujimura KE, Sitarik AR, Havstad S, Lin DL, Levan S, Fadrosh D, et al. Neonatal gut
Malassezia species in healthy skin and in dermatological conditions. Int J microbiota associates with childhood multisensitized atopy and T cell differ-
Dermatol 2016;55:494–504. entiation. Nat Med 2016;22:1187–91.
[120] Svejgaard EL, Larsen PØ, Deleuran M, Ternowitz T, Roed-Petersen J, Nilsson J. [138] Chiu CY, Chan YL, Tsai YS, Chen SA, Wang CJ, Chen KF, et al. Airway microbial
Treatment of head and neck dermatitis comparing itraconazole 200 mg and 400 diversity is inversely associated with mite-sensitized rhinitis and asthma in early
mg daily for 1 week with placebo. J Eur Acad Dermatol Venereol 2004;18:445–9. childhood. Sci Reports 2017;7:1820.
[121] Sonesson A, Bartosik J, Christiansen J, Roscher I, Nilsson F, Schmidtchen A, et al. [139] Russell SL, Gold MJ, Hartmann M, Willing BP, Thorson L, Wlodarska M, et al.
Sensitization to skin-associated microorganisms in adult patients with atopic Early life antibiotic-driven changes in microbiota enhance susceptibility to allergic
dermatitis is of importance for disease severity. Acta Derm Venereol asthma. EMBO Rep 2012;13:440–7.
2013;93:340–5. [140] Hanshew AS, Jetté ME, Rosen SP, Thibeault SL. Integrating the microbiota of the
[122] Casagrande BF, Flückiger S, Linder MT, Johansson C, Scheynius A, Crameri R, respiratory tract with the unified airway model. Respir Med 2017;126:68–74.
et al. Sensitization to the yeast Malassezia sympodialis is specific for extrinsic and [141] Ramakrishnan VR, Feazel LM, Gitomer SA, Ir D, Robertson CE, Frank DN. The
intrinsic atopic eczema. J Invest Dermatol 2006;126:2414–21. microbiome of the middle meatus in healthy adults. PLoS One 2013;8:e85507.
[123] Gaitanis G, Magiatis P, Hantschke M, Bassukas ID, Velegraki A. The Malassezia [142] Kim RJ, Biswas K, Hoggard M, Taylor MW, Douglas RG. Paired analysis of the
genus in skin and systemic diseases. Clin Microbiol Rev 2012;25:106–41. microbiome of surface mucus and whole-tissue specimens in patients with chronic
[124] Fujimura KE, Lynch SV. Microbiota in allergy and asthma and the emerging re- rhinosinusitis. Int Forum Allergy Rhinol 2015;5:877–83.
lationship with the gut microbiome. Cell Host Microbe 2015;17:592–602. [143] Ramakrishnan VR, Hauser LJ, Feazel LM, Ir D, Robertson CE, Frank DN. Sinus
[125] Gollwitzer ES, Saglani S, Trompette A, Yadava K, Sherburn R, McCoy KD, et al. microbiome varies among chronic rhinosinusitis phenotypes and predicts surgical
Lung microbiome promotes tolerance to allergens in neonates via PD-L1. Nat Med outcome. J Allergy Clin Immunol 2015;136(334–42):e1.
2014;20:642–7. [144] Psaltis AJ, Wormald PJ. Therapy of sinonasal microbiome in CRS: A critical ap-
[126] Schuijs MJ, Willart MA, Vergote K, Gras D, Deswarte K, Ege MJ, et al. Farm dust proach. Curr Allergy Asthma Rep 2017;17:59.
and endotoxin protect against allergy through A20 induction in lung epithelial [145] Merkley MA, Bice TC, Grier A, Strohl AM, Man LX, Gill SR. The effect of antibiotics
cells. Science 2015;349:1106–10. on the microbiome in acute exacerbations of chronic rhinosinusitis. Int Forum
[127] Huang YJ, Boushey HA. The microbiome in asthma. J Allergy Clin Immunol Allergy Rhinol 2015;5:884–93.
2015;135:25–30. [146] Dixon AE, Kaminsky DA, Holbrook JT, Wise RA, Shade DM, Irvin CG. Allergic
[128] Marri PR, Stern DA, Wright AL, Billheimer D, Martinez FD. Asthma-associated rhinitis and sinusitis in asthma: differential effects on symptoms and pulmonary
differences in microbial composition of induced sputum. J Allergy Clin Immunol function. Chest 2006;130:429–35.
2013. 131:346-52.e1-3. [147] Yang HJ, LoSavio PS, Engen PA, Naqib A, Mehta A, Kota R, et al. Association of
[129] Zakharkina T, Heinzel E, Koczulla RA, Greulich T, Rentz K, Pauling JK, et al. nasal microbiome and asthma control in patients with chronic rhinosinusitis. Clin
Analysis of the airway microbiota of healthy individuals and patients with chronic Exp Allergy 2018 Aug 20. https://doi.org/10.1111/cea.
obstructive pulmonary disease by T-RFLP and clone sequencing. PLoS One [148] Morgun A, Dzutsev A, Dong X, Greer RL, Sexton DJ, Ravel J, et al. Uncovering
2013;8:e68302. effects of antibiotics on the host and microbiota using transkingdom gene net-
[130] Taylor SL, Leong LEX, Choo JM, Wesselingh S, Yang IA, Upham JW, et al. works. Gut 2015;64:1732–43.

20

You might also like