ArticlePDF AvailableLiterature Review

Research Progress in Atopic March

Frontiers
Frontiers in Immunology
Authors:

Abstract and Figures

The incidence of allergic diseases continues to rise. Cross-sectional and longitudinal studies have indicated that allergic diseases occur in a time-based order: from atopic dermatitis and food allergy in infancy to gradual development into allergic asthma and allergic rhinitis in childhood. This phenomenon is defined as the “atopic march”. Some scholars have suggested that the atopic march does not progress completely in a temporal pattern with genetic and environmental factors. Also, the mechanisms underlying the atopic march are incompletely understood. Nevertheless, the concept of the atopic march provides a new perspective for the mechanistic research, prediction, prevention, and treatment of atopic diseases. Here, we review the epidemiology, related diseases, mechanistic studies, and treatment strategies for the atopic march.
This content is subject to copyright.
REVIEW
published: 27 August 2020
doi: 10.3389/fimmu.2020.01907
Frontiers in Immunology | www.frontiersin.org 1August 2020 | Volume 11 | Article 1907
Edited by:
Christiane Hilger,
Luxembourg Institute of
Health, Luxembourg
Reviewed by:
Jarmila Celakovska,
Charles University, Czechia
Simon Blank,
Technical University of
Munich, Germany
*Correspondence:
Yufeng Zhou
yfzhou1@fudan.edu.cn
Specialty section:
This article was submitted to
Immunological Tolerance and
Regulation,
a section of the journal
Frontiers in Immunology
Received: 07 May 2020
Accepted: 15 July 2020
Published: 27 August 2020
Citation:
Yang L, Fu J and Zhou Y (2020)
Research Progress in Atopic March.
Front. Immunol. 11:1907.
doi: 10.3389/fimmu.2020.01907
Research Progress in Atopic March
Lan Yang1, Jinrong Fu 1and Yufeng Zhou 1,2
*
1Institute of Pediatrics, Children’s Hospital of Fudan University, The Shanghai Key Laboratory of Medical Epigenetics,
International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of
Biomedical Sciences, Fudan University, Shanghai, China, 2National Health Commission (NHC) Key Laboratory of Neonatal
Diseases, Fudan University, Shanghai, China
The incidence of allergic diseases continues to rise. Cross-sectional and longitudinal
studies have indicated that allergic diseases occur in a time-based order: from atopic
dermatitis and food allergy in infancy to gradual development into allergic asthma
and allergic rhinitis in childhood. This phenomenon is defined as the “atopic march”.
Some scholars have suggested that the atopic march does not progress completely
in a temporal pattern with genetic and environmental factors. Also, the mechanisms
underlying the atopic march are incompletely understood. Nevertheless, the concept
of the atopic march provides a new perspective for the mechanistic research, prediction,
prevention, and treatment of atopic diseases. Here, we review the epidemiology, related
diseases, mechanistic studies, and treatment strategies for the atopic march.
Keywords: atopic dermatitis, asthma, food allergy, allergic rhinitis, atopic march
INTRODUCTION
In recent decades, the incidence of allergic diseases has continued to increase, affecting 20%
of the worldwide population, especially children (1). Cross-sectional and longitudinal studies
have suggested that allergic diseases occur following a time-based order: from atopic dermatitis
(AD) and food allergy in infancy to gradual development into allergic asthma (AA) and allergic
rhinitis (AR) in childhood. In terms of anatomic structure, it follows the spatial evolution of skin–
gastrointestinal tract–respiratory tract, and this phenomenon is defined as the “atopic march (2).
Among the allergic diseases mentioned above, some resolve gradually to disappear with age,
whereas others continue for many years (3). Some studies have shown that the atopic march does
not progress completely in a temporal pattern with genes and the environment (4). Nevertheless,
the concept of the atopic march provides a new perspective for the mechanistic research, prediction,
prevention, and treatment of allergic diseases.
Here, we review the epidemiology, related diseases, mechanism of action, and treatment
strategies of the atopic march.
EPIDEMIOLOGY OF THE ATOPIC MARCH
AD: The First Manifestation of the Atopic March
AD is a chronic recurrent skin disease. Its clinical manifestations are chronic inflammation of the
skin, itching, and an impaired skin barrier. AD affects 3% of adults and 30% of children, and its
prevalence tends to increase with age (5). AD occurs in the early years of life. Some epidemiology
studies have shown that 45% of affected children had the condition before 6 months of age, 60%
before 1 year of age, and up to 85% before 5 years of age (6,7).
Yang et al. Research Progress in Atopic March
AD etiology is a combination of various factors involving
genes and the environment (8). Once external allergens contact
a damaged skin barrier, keratinocytes are stimulated to secrete
thymic stromal lymphopoietin (TSLP) and other factors in
conjunction with Langerhans cells (LCs) to stimulate T-helper
type 2 (Th2) immune responses. Then, the body is stimulated
to produce non-specific immunoglobulin (Ig)E (if children are
exposed to allergens such as mites for a long time, specific IgE
may appear). Subsequently, T cells, eosinophils, macrophages,
mast cells, and type 2 innate lymphoid cells (ILC2s) infiltrate to
secrete cytokines, resulting in local inflammation of the skin (9).
AD patients can be classified into two types based on whether
the IgE level is increased: intrinsic (normal IgE and non-allergic)
and extrinsic (high IgE level associated with increased disease
severity). Studies have shown that extrinsic AD increases the risk
of developing the atopic march (10,11).
AA and AR: The End Progression of the
Atopic March
AA is a common chronic airway disease characterized by
the inflammation, hyperresponsiveness, and remodeling of
airways (1215). With modernization and industrialization, AA
incidence has increased year by year. This may be because
of lifestyle alterations, changes in environmental factors (e.g.,
increase in indoor dust mites and outdoor pollution), changes in
dietary habits, and many other factors. AR involves inflammation
of the nasal mucosa (16) and diminishes the quality of life of
sufferers (17).
Epidemiologic evidence has revealed a link between AA and
AR. A retrospective follow-up study reported the incidence of
AR to be higher in AA patients than in non-AA persons (18). In
another cohort study, Leynaert et al. demonstrated that 74–81%
of AA patients reported AR. Also, they found that AA occurred
in 2% of non-AR persons, but in 18.8% of AR patients upon
exposure to pollen or animal dander (19).
AR may lead to changes in the function of the lower airways
through three main mechanisms. Firstly, stimulation of the
nasal mucosa contracts bronchial smooth muscle through the
nasal–tracheal reflex. Secondly, various chemical mediators and
cytokines released by antigenic stimulation causing nasal mucosa
allergy are absorbed into blood, are transported to the lung
through circulation, and then act on the trachea and bronchi,
causing smooth muscle spasm. Thirdly, nasal inflammatory
mediators and secretions are discharged through the nasal
passage to the lower airways, resulting in a reduced β-adrenergic
receptor functional response (20).
Epidemiology of the Atopic March: Linking
AD With AA or AR
Dharmage et al. found, in infants who have AD within 2 years
of age, that the incidence of AA and AR increased significantly
during age 6–7 years. In particular, early-onset, persistent, and
IgE-positive AD led to a higher risk of developing AA and AR (9).
A longitudinal study on a Canadian birth cohort (2,311 children)
has shown that AD with sensitization at 1 year of age increased
the prevalence of AA and AR at 3 years of age more than 11- and
7-fold, respectively (21). In a recent report from Thailand, 102
children with AD (diagnosed at 1.5 years of age) were reviewed,
and subsequently, AR and AA were diagnosed in 61.8 and 29.4%,
respectively. Concomitantly, 67% of the AA patients also suffered
AR (22). A prospective cohort study (3,124 children aged 1–2
years) reported that, compared with children with no history
of AD, those once having AD, particularly moderate-to-severe,
early, and persistent AD, were more inclined to develop AA and
AR (23).
The discoveries mentioned above strongly support the natural
process of the atopic march.
Roles of Food Allergy
IgE-positive food allergy commonly coexists with AD in early
childhood as the earliest manifestation of the atopic march. In
2011, Japanese scholars conducted a retrospective questionnaire
survey on freshmen, and they found that AD occurred earlier
in those with accompanying food allergy. Also, having food
allergy was regarded as the biggest risk factor for the atopic
march (24). A family-based cohort study from Chicago revealed
that symptomatic food allergy, especially severe or multiple food
allergies, was closely related to AA in children aged 6 years.
Children with food allergy developed AA earlier than those
without food allergy (25). A survey of 2,222 infants with AD
aged 11.5–25.5 months showed that 64% of children diagnosed
with AD within 3 months of birth exhibited an IgE-mediated
sensitivity to milk, peanuts, or eggs. Also, in infants <12 months
of age, the proportion of infants with sensitivity to eggs, milk,
or peanuts increased with AD severity, but this phenomenon
was not manifested in children with AD after 1 year of age
(26). Among adults with AD, food allergy is relatively rare
(2731). In addition, studies have shown that children sensitive
to milk in infancy subsequently exhibited aggravated airway
inflammation and increased airway responsiveness to histamine
(32,33). Remarkably, food allergy commonly exists together with
AD in infants. Therefore, it is worth exploring whether the link
between food allergy and AA or AR is related to AD or is a direct
consequence of the food allergy itself.
EoE: A New Manifestation of the Atopic
March?
Eosinophilic esophagitis (EoE) is a chronic esophageal
inflammatory disease induced by pollens or food allergens
(34). EoE patients are sensitive to allergen avoidance and
glucocorticoid therapy. Genome-wide association study
(GWAS) data have indicated that EoE shares some susceptible
genetic loci with other manifestations of the atopic march,
including polymorphisms in the signal transducer and activator
of transcription 6 gene (STAT6) and TSLP (35). In addition,
epidemiology studies have demonstrated EoE to be associated
with other allergic diseases. For example, Mohammad et al.
found that, of 449 EoE patients, the prevalence rates of AR, AA,
and AD were 61.9, 39, and 46.1%, respectively, and that up to
21.6% of EoE patients developed these three atopic diseases (36).
Another study involving 35,528 people reported that those with
IgE-positive food allergy were at a higher risk of EoE (37). A birth
Frontiers in Immunology | www.frontiersin.org 2August 2020 | Volume 11 | Article 1907
Yang et al. Research Progress in Atopic March
cohort study involving 130,435 children determined a positive
association between EoE and other allergic manifestations (34).
The studies mentioned above suggest the potential of EoE as
the fifth “member” of the atopic march, but this hypothesis is
controversial. For example, EoE occurs not only in childhood but
also after childhood. In addition, EoE can occur in individuals
without a history of atopy. Therefore, larger cohorts are
needed to study the epidemiologic relationship of EoE with
other manifestations of the atopic march and the mechanisms
involved (38).
POSSIBLE MECHANISMS UNDERLYING
THE ATOPIC MARCH
Dysfunction of the Skin Barrier
The skin is the foremost barrier for defense against external
stimuli, such as pathogens, environmental pollutants, and
ultraviolet light. As a component of the innate immune
system, the skin has several defensive functions, including
microbial, chemical, physical, and immune barrier. These
different functions of the skin barrier coordinate with each other
to resist external stimuli and maintain skin homeostasis.
Allergens can enter the body through damaged skin
to cause sensitization, which is defined as “transcutaneous
sensitization” (39). Transcutaneous sensitization can cause AD
and, subsequently, AA and AR (5). Studies have shown that
epicutaneous disruption induces sensitization after exposure to
peanut and egg allergens (40,41). Spergel et al. demonstrated that
repeated cutaneous exposure to egg allergens induced AD-like
skin inflammation and AA-like bronchial hyper-responsiveness
in a mouse model (41). Emerging studies now suggest that the
skin barrier protein filaggrin and epithelial cell-derived cytokines
such as TSLP, IL-25, and IL-33 might be related to the progression
of the atopic march.
Filaggrin
Filaggrin, a barrier protein, has important roles in the integrity
of the stratum corneum in terms of structure and composition.
Mutations in the filaggrin gene (FLG) can impair the barrier
function of the skin and induce an allergic response (42,43).
Several studies have shown patients with impaired or reduced
levels of filaggrin to be more susceptible to food sensitization
(4446). Moreover, FLG mutations increase the risk of early and
severe AD and of AA in individuals who have had AD (4749).
Thymic Stromal Lymphopoietin
TSLP is an interleukin (IL)-7-like epithelial cell-derived cytokine
which regulates the Th2 response (50). Zhang et al. found
that TSLP overexpression in keratinocytes aggravated AA-like
airway inflammation in mice subjected to ovalbumin (OVA)
sensitization intraperitoneally and OVA challenge intranasally
(51). Another in vivo study demonstrated that keratinocytic TSLP
was essential to induce a Th2 response in the skin and to trigger
aeroallergen-challenged AA phenotypes (52). In addition, Noti
et al. found that the effect of TSLP was enough to develop
experimental EoE-like phenotypes in mice (53). Also, they found
that TSLP in skin facilitated food allergy (54).
Interleukin-33
IL-33 is derived from epithelial cells and acts on macrophages,
ILC2s, Th2 cells, mast cells, and basophils through the
suppression of tumorigenicity 2/IL-1 receptor accessory protein
heterodimer (ST2/IL1RL1) (5560). Several studies have explored
the roles of IL-33 in allergic diseases and found high expressions
of IL-33 in the skin or airway epithelial cells in AD or airway
inflammation (6163). Blockade of ST2 expression can alleviate
food allergy in peanut- and OVA-challenged models (64,65).
Interleukin-25
IL-25 is also an epithelial cell-derived cytokine (6668). Kim
et al. found that IL-25 inhibited filaggrin expression in the skin
and aggravated skin inflammation by coordinating with Th2
cytokines (69). Lee et al. reported OVA/alum-sensitized allergic
diarrhea to be inhibited in mice lacking IL-17RB, the receptor of
IL-25, whereas IL-25 overexpression in the intestine accelerated
the development of allergic diarrhea (70). Kang et al. found that
the mRNA expression of IL-25 was upregulated in rat lungs in a
TiO2-induced model of airway inflammation (71).
In conclusion, allergens (including food and aeroallergens)
enter the skin through the damaged skin barrier. Then, they
stimulate skin epithelial cells to release TSLP, IL-25, and IL-
33. This action activates some immune cells in the dermis [e.g.,
basophils, mast cells, dendritic cells (DCs), eosinophils, ILC2]
to secrete cytokines, and subsequently, Th2 cells are generated
and IgE production in local lymph nodes occurs. Th2 cells can
secrete more type 2 cytokines (e.g., IL-4) to activate more ILC2
and eosinophils, and IgE can act on mast cells and basophils.
This positive feedback causes skin inflammation and AD (72).
Furthermore, IgE, Th2, TSLP, IL-25, and IL-33 might enter
the digestive and respiratory tracts through blood circulation
to facilitate the development of AA, AR, and food allergy if
allergens are re-encountered (73,74) (Figure 1). Therefore, skin
barrier dysfunction might be a potential mechanism underlying
the atopic march.
Microbiome Alteration
Many microorganisms are colonized in the intestine, skin, and
respiratory tract (75) and influence health and disease. Several
studies have suggested that microbiome alteration plays roles in
atopic diseases locally or peripherally.
Kennedy et al. observed the skin microbiome dysbiosis in
early life of AD patients, and they also found that colonization
with commensal Staphylococci at 2 months was related to a lower
risk of AD at 1 year of age (76). Forno et al. and Abrahamsson
et al. reported that children who had AD at 6 months (77) and 2
years (78) of age had decreased intestinal microbial diversity at 1
month of life. A study of the KOALA birth cohort demonstrated
that infants with Clostridium difficile colonization in the gut at
1 month of life were inclined to develop AD and other atopic
diseases (79). Azad et al. demonstrated that infants who had a
positive skin prick test for food sensitization at 1 year had lower
gut microbial richness at 3 months (80). Abrahamsson et al.
reported that infants with low diversity in intestinal flora at 1
month of age were inclined to develop AA at school age (81).
In addition, Teo et al. determined that microbiome alteration
Frontiers in Immunology | www.frontiersin.org 3August 2020 | Volume 11 | Article 1907
Yang et al. Research Progress in Atopic March
FIGURE 1 | A possible model of the contribution of skin barrier dysfunction to the atopic march. Allergens (including food allergens and aeroallergens) enter the skin
through the damaged skin barrier. Then, they stimulate epithelial cells in the skin to release thymic stromal lymphopoietin (TSLP), interleukin (IL)-25, and IL-33. This
action activates some immune cells in the dermis (e.g., basophils, mast cells, DCs, eosinophils, and ILC2) to secrete cytokines, followed by the generation of T-helper
type 2 (Th2) cells and immunoglobulin E (IgE) production in local lymph nodes. Th2 cells can secrete more type 2 cytokines (e.g., IL-4) to activate more ILC2 and
eosinophils, and IgE can act on mast cells and basophils. This positive feedback causes skin inflammation and atopic dermatitis (AD). Furthermore, IgE, Th2 cells,
TSLP, IL-25, and IL-33 might enter the digestive tract and respiratory tract through blood circulation to facilitate the development of allergic asthma (AA), allergic
rhinitis (AR), and food allergy if allergens are re-encountered.
following respiratory infections during infancy might contribute
to the development of AA (82).
Moreover, some studies have shown that microbes regulate
atopic diseases by secreting metabolites. Furusawa et al. reported
that the short-chain fatty acids produced by several intestinal
microorganisms induced the proliferation of colonic regulatory
T cells (Tregs) and further ameliorated colitis and allergic
responses (83). Dysbiosis of Faecalibacterium prausnitzii, as
observed in AD, was found to reduce the production of butyrate
and propionate and further destroyed the intestinal mucosa.
Then, some toxins permeated into the circulation and induced
a Th2 immune response to facilitate skin inflammation and AD
development (84). Johnson et al. found that the polysaccharides
derived from Bacteroides fragilis induced CD4+Foxp3T cell
activation and further prevented AA onset (85).
The studies mentioned above strongly suggest that
microbiome alteration may be involved in the atopic march.
However, further studies are needed to determine whether
microbiome shifts are a cause or a consequence of the
atopic march.
Epigenetic Factors
Epigenetic mechanisms can regulate gene expression and
constitute the cause of diseases. Several epigenome-wide
studies have revealed DNA methylation in blood to be
related to food allergy (86,87) and AA (88). Recently, Peng
et al. undertook DNA methylation analyses on the cohorts
of the Generation R Study (343 at mid-childhood and
839 newborns) in the Netherlands and Project Viva (396
at mid-childhood and 232 newborns) in the USA. Meta-
analyses linked the differential methylation profiles of the
peripheral blood of mid-childhood children with food allergens,
environmental/inhalant allergens, and atopic sensitization.
Multiple methylation site-related genes were enriched to
AA pathways, including eosinophil peroxidase (EPX), IL4,
interleukin 5 receptor A (IL5RA), and proteoglycan 2 (PRG2).
Furthermore, Peng et al. identified several methylation sites of
cord blood to be related to allergic phenotypes in mid-childhood
and that some methylation sites of cord blood were also
present in mid-childhood (89), which suggested a longitudinal
time trend.
Frontiers in Immunology | www.frontiersin.org 4August 2020 | Volume 11 | Article 1907
Yang et al. Research Progress in Atopic March
The findings mentioned above suggest that epigenetics may
have roles in allergic diseases. However, these studies show only a
correlation between epigenetics and the atopic march. Whether
epigenetic change is a cause or a result of the atopic march
warrants large and detailed longitudinal studies.
“Social” Dysfunction of Cells and
Molecules
Allergic reactions occur not only in the regions where allergens
are in contact directly but also in long-distance, non-contact
sites. This may be a systemic reaction of the body, and the
mechanisms are incompletely understood. Through literature
search, Luo et al. proposed a model of “social events” of cells
and molecules to explain the atopic march (90). Epithelial cells,
such as epidermal keratinocytes and airway epithelial cells, are
the first line of defense against allergen exposure and initiate the
inflammatory response by releasing proinflammatory cytokines.
Thus, epithelial cells are considered to be key participants in
allergic diseases. In this model, Luo and colleagues considered
that it is the atopic factors produced by epithelial cells locally,
not in the circulation, that drive allergy at different sites and
that certain allergens are the irritants that trigger the release of
atopic factors at different sites. Zhang et al. reported that TSLP
overexpression in keratinocytes induced AD-like symptoms
and also aggravated OVA-induced AA manifestations in mice.
However, they also found that increased TSLP expression in the
skin and, subsequently, peripheral blood was not sufficient to
induce lung inflammation (51). The atopic reaction in the lung
might be induced by the TSLP derived from the lung epithelia
themselves. Therefore, the atopic reaction in the skin and the
lung might be the consequence of the “social dysfunction” of
homologous epithelia and molecules such as TSLP. Despite its
rationality and interpretability, the theory of social events needs
sufficient evidence from in vivo and in vitro studies.
Interference of Other Predicted Genes
Marenholz et al. performed GWAS on 2,428 cases with AD
in infancy and AA in childhood and on 17,034 controls. They
identified seven susceptible sites associated with the atopic
march: FLG [1q21.3], AP5B1/OVOL1 [11q13.1], IL4/KIF3A
[5q31.1], IKZF3 [17q21], C11orf30/LRRC329,EFHC1 [6p12.3],
and rs99322 [12q21.3] (91).
Bioinformatics analyses by Gupta et al. revealed that the
atopic march involved 16 common pathogenic genes: IL4,IL5,
TSLP,RNASE3,IL13,IL10,IGHG4,IFNG,CCL11,FCER2,
RNASE2,FOXP3,KCNE4,CD4,IL4R, and CCL26 (92). These
genes were predicted through large-scale and high-throughput
bioinformatics analyses, and their roles in the atopic march need
to be determined through further experimentation.
Summarily, Paller et al. have reviewed the multifactorial
etiology of the atopic march, including skin barrier damage,
microbiome alteration, and epigenetic factors (93), and we
consider that “social” dysfunction of cells and molecules, and the
interference of other predicted genes, may also contribute to the
atopic march (Figure 2). However, further studies are required to
detail the relevant mechanisms.
ANIMAL MODELS FOR STUDIES ON THE
ATOPIC MARCH
The modeling process of Leyva-Castillo et al. consisted of two
phases. In the first phase, wild-type (WT) BALB/c mice were
treated with calcipotriol MC903 plus OVA through epicutaneous
sensitization. This led to increased levels of Th2 cytokines,
Th17 cytokines, and OVA-specific IgE and IgG1 in serum. In
the second phase, MC903-treated (epicutaneous) OVA-sensitized
mice underwent intranasal challenge with OVA. These mice
exhibited AA-like symptoms with increased mucus secretion,
eosinophil infiltration, and expression of Th2 cytokines (52).
In a model established by Han et al., WT BALB/c mice
were first treated with OVA plus TSLP via the intradermal
route (four times within 2 weeks). After 9 days, the mice were
challenged by OVA via the intranasal route for four consecutive
days. Consequently, the mice exhibited increased OVA-specific
IgE in serum as well as cellular and eosinophil infiltration in
the bronchoalveolar lavage fluid. Histopathology showed severe
inflammatory infiltrates in mouse lungs. In addition, periodic
acid–Schiff staining showed excessive goblet cell metaplasia and
mucus secretion (94).
Moreover, one study showed that epicutaneous exposure
to Aspergillus fumigates aeroallergens followed by intranasal
challenge with A. fumigates induced an allergic nasal response in
BALB/c mice (95).
In conclusion, the models mentioned above have one
similarity: the skin is used as a sensitization site, consistent
with the feature that AD is the initial manifestation during the
atopic march. These animal models facilitate the studies of the
mechanisms underlying the atopic march.
REFUTATIONS OF THE ATOPIC MARCH
Despite substantial epidemiologic and experimental evidence,
some scholars argued that the prevalence of the atopic march may
be overemphasized (96).
First, the methods of the data collection and disease
identification initiate one main concern. Considering the cost
and time required to make physician diagnoses, allergic disease
identification was often based on “yes” or “no” questions. In
existing epidemiologic surveys, the diagnosis of AD, AA, and
AR simply used “yes” or “no” questionnaires, and some even
lacked further physician identification (9799). In addition,
deviations and over-reporting in questionnaire surveys from
some individuals led to an overestimation of the disease
prevalence (100). Another rebutted criticism of the atopic march
is the failure to consider disease heterogeneity or variations.
Martinez et al. found that AD patients were at a higher risk
of developing transient early AA and persistent AA, not late-
onset AA (97). This indicates that the association between
AD and AA may be restricted to specific AA subpopulations,
not universal. Moreover, some individual-level analyses did
not support the typical temporal pattern. At an individual
level rather than a large-scale population level, Belgrave et al.
demonstrated that only 3.1% of children followed the classical
Frontiers in Immunology | www.frontiersin.org 5August 2020 | Volume 11 | Article 1907
Yang et al. Research Progress in Atopic March
FIGURE 2 | The temporal pattern and the possible mechanisms of the atopic march. The temporal pattern of the atopic march is, in general, from atopic dermatitis
(AD) and food allergy in infancy to gradual development into allergic asthma (AA) and allergic rhinitis (AR) in childhood. Several mechanisms could underlie the atopic
march: skin barrier damage, microbiome alteration, “social” dysfunction of cells and molecules, epigenetic factors, and interference of other predicted genes.
atopic march procession (AD first, followed by AA and then
AR) and more than 90% of children with atopic manifestations
did not (101).
Unusually, a study from Italy found an evidence of a
“reverse” atopic march. The study included 745 children aged
6–9 years with AA only, without a history of food allergy or
AD. After a 9-years follow-up, 20% of the children were found
to have developed AD (102). In addition, the prevalence of the
atopic march differed in distinct countries. Colombian scholars
followed up 326 mother–infant pairs in a birth cohort study,
and they found that AA was the most common manifestation
by 24 months. The prevalence of recurrent AA was 7.1%
at 12 months and reached 14.2% at 24 months. However,
allergic symptoms induced by milk, egg, or other food allergens
were scarce, only 1.8%, and AD was not observed in any
cases (103).
Although these studies refute the concept of atopic march
to a certain extent, we cannot deny the contribution of the
theory of atopic march to the early prevention, diagnosis,
and treatment of allergic diseases. Future research on the
atopic march should improve the current data collection and
disease identification methods, not only relying on “yes” or
“no” questionnaires, take disease subtypes into account, and
perform the study in an individual level rather than only in a
group level.
PREVENTION AND TREATMENT
STRATEGIES FOR THE ATOPIC MARCH
Several measures used to prevent and treat allergic diseases are
expected to interfere with, delay, and block the natural process of
the atopic march.
Food Interventions
In most studies, breastfeeding for >6 months has been
recommended because it reduces not only the incidence of AD
but also of other allergic diseases (104). A 15-years follow-up
study of the German Infant Nutritional Intervention (GINI)
cohort has shown that, if breastfeeding is not possible, compared
with standard cow’s milk formula (CMF), the interventional
use of partial whey hydrolyzate (pHF-W) formula and extensive
casein hydrolyzate (eHF-C) formula in the first 4 months of
life has significant preventive effects on AD, and the eHF-
C formula also reduced the prevalence of AA and AR (105).
However, the mechanisms underlying the preventative effects of
hydrolyzed formulas are unknown. In addition, the assessment of
the effects of hydrolyzed formulas was based on parental reports
of physicians’ diagnosis, not on the clinical examinations. These
are the criticisms against the use of hydrolyzed formulas for the
prevention of allergic conditions.
Moreover, Wickens et al. found that supplementation with
Lactobacillus rhamnosus for the first 2 years of life reduced the
prevalence of AD by about half (106). However, further studies
are required to handle the uncertainties about whether other
probiotics are equally effective and how probiotics exert their
effects on allergic diseases.
Furthermore, the Learning Early About Peanut Allergy
(LEAP) trial demonstrated that, compared with children who
avoided peanut, sustained peanut consumption, beginning in the
first 11 months of life, significantly decreased the prevalence of
peanut allergy at 60 months of age in infants with high atopic
risk (107). In addition, a large-scale population-based prospective
study showed that early introduction of cow’s milk protein
as a supplement to breastfeeding might promote tolerance,
reducing the incidence of IgE-mediated cow’s milk protein
allergy (108).
Frontiers in Immunology | www.frontiersin.org 6August 2020 | Volume 11 | Article 1907
Yang et al. Research Progress in Atopic March
Environmental Prevention
Exposure to several environmental factors is closely related to
having allergic diseases. Studies have shown that smoke in the
environment increases children’s risk of allergic sensitization and
AA (109). Therefore, it is strongly recommended that all parents
should stop smoking tobacco. Dust mites, pollen, cockroaches,
pet fur, and fungi are common allergens, and avoiding exposure
to these allergens can reduce the sensitization of children at
high risk. However, it has also been proposed that there was
no correlation between house dust mite (HDM) exposure and
AA (110), and keeping pets (cats or dogs) in the home in the
first year after birth reduced the risk of sensitization to multiple
allergens during childhood, but it impaired lung function once
cat or dog sensitization has occurred, particularly in children
with a family history of AA (111). These controversial views are
initiating further research to evaluate their relevance.
Medical Treatment
Symptomatic Treatment
Antihistamines are used to relieve itching in AD patients and to
prevent skin damage aggravated by scratching. Ketotifen, an H1
antihistamine, significantly lowered AA risk in infants with AD
or other pre-asthmatic conditions (112,113). A double-blind,
randomized, placebo-controlled trial showed that compared to
placebo, cetirizine significantly reduced the incidence of AA in
AD patients sensitized to grass pollen or to HDM (114). However,
considering the side effects of antihistamines, a large number of
clinical trials are needed to evaluate the security of antihistamines
and the effectiveness of interventions in the natural course of
allergic diseases.
Glucocorticoid is also an effective anti-inflammatory
treatment for allergic diseases, and inhaled glucocorticoids has
now become the first-line treatment for AA (115). Although
the symptoms of AD and AA can be significantly improved
by glucocorticoids, it is prone to relapse after withdrawal. In
addition, there are many side effects. Therefore, glucocorticoids
should be prescribed with caution by the physicians.
Allergen-Specific Immunotherapy
Allergen-specific immunotherapy (ASIT), also known as
“desensitization therapy, can alleviate allergy symptoms for a
long time and change the natural course of allergic diseases (116).
Several ASIT routes have been documented in preclinical studies,
including subcutaneous immunotherapy (SCIT), sublingual
immunotherapy (SLIT), epicutaneous immunotherapy (EPIT),
and oral immunotherapy (OIT). The recognized mechanism of
specific immunotherapy is stimulation of the secretion of IL-10
and transforming growth factor-βfrom Tregs, promotion of
the balance of Th1 cells/Th2 cells, and conversion of IgE to IgG
to block the IgE-mediated immune cascade (117,118). Zhong
et al. found that the clinical symptoms and quality of life of AD
patients with HDM sensitization could be improved after 2 years
of ASIT (119). Besh et al. demonstrated that combining basic
therapy with SCIT acquired significantly better results in AA
patients compared to basic therapy only (120). Karakoc-Aydiner
et al. found that the nasal symptom scores of children with AR
were significantly reduced after receiving dust mite allergen
vaccine through SCIT or SLIT (121). However, the lack of
security greatly limits the development of ASIT. For example,
the adverse reactions of SLIT mainly focus on local reactions,
such as oromucosal pruritus and gastrointestinal reaction
(122). In addition, almost all clinical trials related to OIT are
accompanied by one or several serious adverse reactions, such
as severe gastrointestinal reactions, systemic allergic reactions,
etc. (123). Long-term follow-up of milk OIT patients showed
that the complete immune tolerance rate after OIT treatment
was only 31% (124,125). Therefore, further research on ASIT
should be directed at the improvement of not only its efficacy but
also security.
Targeted Therapy
Omalizumab is a human monoclonal antibody against IgE.
In 2003, it was approved for the treatment of severe AA
in adolescents and adults. Esquivel et al. demonstrated that
omalizumab inhibited rhinovirus infections, illnesses, and
exacerbations of AA through specific binding to IgE (126).
Dupilumab is a human IgG4 monoclonal antibody against IL-4
receptor subunit alpha (IL-4Rα), and it can inhibit IL-4 and IL-13
signaling pathways by interacting with IL-4Rα(127). Dupilumab
has been approved by the US Food and Drug Administration
to treat infants with moderate-to-severe AD with poor results
from conventional treatment (128). Tezepelumab (AMG 157)
is a monoclonal antibody (G2λ) against TSLP. In one clinical
trial, tezepelumab treatment for 5–12 weeks blunted inhaled
allergen-induced AA attacks (129). Of note is that these targeted
therapy medicines are only licensed for use in certain allergic
diseases. Although the off-label uses or adjunct to treatment
for numerous allergic conditions have acquired encouraging
results, their potential efficacy still needs to be evaluated through
clinical trials.
BIOMARKERS OF THE ATOPIC MARCH
Although there are no reliable biomarkers to identify subjects
with high risk of atopic march, Davidson et al. have proposed
some recommendations recently for future research to explore
biomarkers, which would provide some possibilities to examine
the atopic march.
The relevant proposals are as follows: (1) to look at the
protein, RNA, and lipid signatures in infants before and after AD
using multi-omics approaches; (2) to analyze transcriptomics,
proteomics, metabolomics, and the cell types of infant blood
sequentially; (3) to perform sequential immune profiling of the
blood, including serology, cytokine profiles, and the evolution of
specific B and T cells; (4) to investigate the microbiomes in the
skin and gut from birth; and (5) to consider potential maternal
delivery effects for atopy (130).
CONCLUSION
The global increase of atopic diseases greatly lowers the quality
of life. The theory of atopic march facilitates our understandings
of the pathophysiology of atopic diseases and further promotes
the early detection, prevention, and treatment of children at
Frontiers in Immunology | www.frontiersin.org 7August 2020 | Volume 11 | Article 1907
Yang et al. Research Progress in Atopic March
risk of allergic diseases. Future studies on atopic march would
be directed at the following points. Firstly, the methods for
data collection should be improved and disease heterogeneity
or variations should be considered when performing substantial
epidemiologic surveys. Secondly, more detailed and logical
mechanisms, including genetic and environmental aspects,
should be explored to account for the temporal pattern, which
would pave the way for novel approaches for the prevention and
timely early treatment of the clinical manifestations, ultimately
reducing the allergy burden.
AUTHOR CONTRIBUTIONS
LY contributed to collection of references and manuscript
preparation. JF and YZ contributed to manuscript modifications.
All authors contributed to the article and approved the
submitted version.
FUNDING
This work was supported by grants from the National Key
R&D Program of China (2016YFC1305102 to YZ), National
Natural Science Foundation of China (81671561, 81974248 to
YZ), the International Joint Laboratory Program of National
Children’s Medical Center (EK1125180109 to YZ), Program
for Outstanding Medical Academic Leader (2019LJ19 to YZ),
and Shanghai Municipal Planning Commission of Science and
Research Fund (201740065 to YZ). Shanghai Pujiang Program
(16PJ1401600 to JF). Shanghai Committee of Science and
Technology (19ZR1406400 to JF).
REFERENCES
1. Bantz SK, Zhu Z, Zheng T. The atopic march: progression from atopic
dermatitis to allergic rhinitis and asthma. J Clin Cell Immunol. (2014)
5:202. doi: 10.4172/2155-9899.1000202
2. Spergel JM, Paller AS. Atopic dermatitis and the atopic march. J Allergy Clin
Immunol. (2003) 112 (6 Suppl.):S118–27. doi: 10.1016/j.jaci.2003.09.033
3. Spergel JM. Epidemiology of atopic dermatitis and atopic
march in children. Immunol Allergy Clin North Am. (2010)
30:269–80. doi: 10.1016/j.iac.2010.06.003
4. Zheng T, Yu J, Oh MH, Zhu Z. The atopic march: progression from atopic
dermatitis to allergic rhinitis and asthma. Allergy Asthma Immunol Res.
(2011) 3:67–73. doi: 10.4168/aair.2011.3.2.67
5. Weidinger S, Novak N. Atopic dermatitis. Lancet. (2016) 387:1109–
22. doi: 10.1016/S0140-6736(15)00149-X
6. Spergel JM. Atopic march: link to upper airways. Curr Opin Allergy Clin
Immunol. (2005) 5:17–21. doi: 10.1097/00130832-200502000-00005
7. Kay J, Gawkrodger DJ, Mortimer MJ, Jaron AG. The prevalence of childhood
atopic eczema in a general population. J Am Acad Dermatol. (1994) 30:35–
9. doi: 10.1016/S0190-9622(94)70004-4
8. Bieber T. Atopic dermatitis. N Engl J Med. (2008) 358:1483–
94. doi: 10.1056/NEJMra074081
9. Dharmage SC, Lowe AJ, Matheson MC, Burgess JA, Allen KJ, Abramson
MJ. Atopic dermatitis and the atopic march revisited. Allergy. (2014) 69:17–
27. doi: 10.1111/all.12268
10. Novembre E, Cianferoni A, Lombardi E, Bernardini R, Pucci N, Vierucci
A. Natural history of “intrinsic” atopic dermatitis. Allergy. (2001) 56:452–
3. doi: 10.1034/j.1398-9995.2001.056005452.x
11. Wuthrich B, Schmid-Grendelmeier P. Natural course of AEDS. Allergy.
(2002) 57:267–8. doi: 10.1034/j.1398-9995.2002.1n3572.x
12. Redington AE, Howarth PH. Airway wall remodelling in asthma. Thorax.
(1997) 52:310–12. doi: 10.1136/thx.52.4.310
13. Kon OM, Kay AB. T cells and chronic asthma. Int Arch Allergy Immunol.
(1999) 118:133–5. doi: 10.1159/000024049
14. Carroll N, Elliot J, Morton A, James A. The structure of large and small
airways in nonfatal and fatal asthma. Am Rev Respir Dis. (1993) 147:405–
10. doi: 10.1164/ajrccm/147.2.405
15. James A. Airway remodeling in asthma. Curr Opin Pulm Med. (2005)
11:1–6. doi: 10.1097/01.mcp.0000146779.26339.d8
16. Hellings PW, Fokkens WJ, Allergic rhinitis and its
impact on otorhinolaryngology. Allergy. (2006) 61:656–
64. doi: 10.1111/j.1398-9995.2006.01109.x
17. Selnes A, Nystad W, Bolle R, Lund E. Diverging prevalence trends of atopic
disorders in Norwegian children. Results from three cross-sectional studies.
Allergy. (2005) 60:894–9. doi: 10.1111/j.1398-9995.2005.00797.x
18. Ricci G, Patrizi A, Baldi E, Menna G, Tabanelli M, Masi M. Long-term follow-
up of atopic dermatitis: retrospective analysis of related risk factors and
association with concomitant allergic diseases. J Am Acad Dermatol. (2006)
55:765–71. doi: 10.1016/j.jaad.2006.04.064
19. Leynaert B, Neukirch C, Kony S, Guenegou A, Bousquet J, Aubier M, et al.
Association between asthma and rhinitis according to atopic sensitization
in a population-based study. J Allergy Clin Immunol. (2004) 113:86–
93. doi: 10.1016/j.jaci.2003.10.010
20. Corren J, Adinoff AD, Irvin CG. Changes in bronchial responsiveness
following nasal provocation with allergen. J Allergy Clin Immunol. (1992)
89:611–8. doi: 10.1016/0091-6749(92)90329-Z
21. Tran MM, Lefebvre DL, Dharma C, Dai DW, Lou YWY, Subbarao P,
et al. Predicting the atopic march: results from the canadian healthy infant
longitudinal development study. J Allergy Clin Immunol. (2018) 141:601–7
e8. doi: 10.1016/j.jaci.2017.08.024
22. Somanunt S, Chinratanapisit S, Pacharn P, Visitsunthorn N,
Jirapongsananuruk O. The natural history of atopic dermatitis and its
association with Atopic March. Asian Pac J Allergy Immunol. (2017)
35:137–43. doi: 10.1016/j.jaci.2015.12.613
23. von Kobyletzki LB, Bornehag CG, Hasselgren M, Larsson M, Lindstrom
CB, Svensson A. Eczema in early childhood is strongly associated with the
development of asthma and rhinitis in a prospective cohort. BMC Dermatol.
(2012) 12:11. doi: 10.1186/1471-5945-12-11
24. Kijima A, Murota H, Takahashi A, Arase N, Yang L, Nishioka M, et al.
Prevalence and impact of past history of food allergy in atopic dermatitis.
Allergol Int. (2013) 62:105–12. doi: 10.2332/allergolint.12-OA-0468
25. Schroeder A, Kumar R, Pongracic JA, Sullivan CL, Caruso DM, Costello
J, et al. Food allergy is associated with an increased risk of asthma.
Clin Exp Allergy. (2009) 39:261–70. doi: 10.1111/j.1365-2222.2008.0
3160.x
26. Hill DJ, Hosking CS, de Benedictis FM, Oranje AP, Diepgen TL, Bauchau V,
et al. Confirmation of the association between high levels of immunoglobulin
E food sensitization and eczema in infancy: an international study. Clin Exp
Allergy. (2008) 38:161–8. doi: 10.1111/j.1365-2222.2007.02861.x
27. Tse K, Horner AA. Allergen tolerance versus the allergic march: the
hygiene hypothesis revisited. Curr Allergy Asthma Rep. (2008) 8:475–
83. doi: 10.1007/s11882-008-0088-5
28. Gustafsson D, Sjoberg O, Foucard T. Development of allergies
and asthma in infants and young children with atopic dermatitis–
a prospective follow-up to 7 years of age. Allergy. (2000)
55:240–5. doi: 10.1034/j.1398-9995.2000.00391.x
29. Lowe AJ, Carlin JB, Bennett CM, Hosking CS, Abramson MJ, Hill DJ, et al.
Do boys do the atopic march while girls dawdle? J Allergy Clin Immunol.
(2008) 121:1190–5. doi: 10.1016/j.jaci.2008.01.034
30. Burgess JA, Lowe AJ, Matheson MC, Varigos G, Abramson MJ,
Dharmage SC. Does eczema lead to asthma? J Asthma. (2009)
46:429–36. doi: 10.1080/02770900902846356
31. Bousquet J, Khaltaev N, Cruz AA, Denburg J, Fokkens WJ, Togias
A, et al. Allergic rhinitis and its impact on asthma (ARIA) 2008
Frontiers in Immunology | www.frontiersin.org 8August 2020 | Volume 11 | Article 1907
Yang et al. Research Progress in Atopic March
update (in collaboration with the World Health Organization,
GA(2)LEN and AllerGen). Allergy. (2008) 63 (Suppl. 86):8–160.
doi: 10.1111/j.1398-9995.2007.01620.x
32. Malmberg LP, Saarinen KM, Pelkonen AS, Savilahti E, Makela MJ.
Cow’s milk allergy as a predictor of bronchial hyperresponsiveness and
airway inflammation at school age. Clin Exp Allergy. (2010) 40:1491–
7. doi: 10.1111/j.1365-2222.2010.03567.x
33. Saarinen KM, Pelkonen AS, Makela MJ, Savilahti E. Clinical course and
prognosis of cow’s milk allergy are dependent on milk-specific IgE status.
J Allergy Clin Immunol. (2005) 116:869–75. doi: 10.1016/j.jaci.2005.06.018
34. Hill DA, Grundmeier RW, Ramos M, Spergel JM. Eosinophilic esophagitis
is a late manifestation of the allergic march. J Allergy Clin Immunol Pract.
(2018) 6:1528–33. doi: 10.1016/j.jaip.2018.05.010
35. Hirota T, Nakayama T, Sato S, Yanagida N, Matsui T, Sugiura S, et al.
Association study of childhood food allergy with genome-wide association
studies-discovered loci of atopic dermatitis and eosinophilic esophagitis. J
Allergy Clin Immunol. (2017) 140:1713–6. doi: 10.1016/j.jaci.2017.05.034
36. Mohammad AA, Wu SZ, Ibrahim O, Bena J, Rizk M, Piliang M, et al.
Prevalence of atopic comorbidities in eosinophilic esophagitis: a case-
control study of 449 patients. J Am Acad Dermatol. (2017) 76:559–
60. doi: 10.1016/j.jaad.2016.08.068
37. Hill DA, Dudley JW, Spergel JM. The prevalence of eosinophilic esophagitis
in pediatric patients with ige-mediated food allergy. J Allergy Clin Immunol
Pract. (2017) 5:369–75. doi: 10.1016/j.jaip.2016.11.020
38. Hill DA, Spergel JM. Is eosinophilic esophagitis a member
of the atopic march? Ann Allergy Asthma Immunol. (2018)
120:113–4. doi: 10.1016/j.anai.2017.10.003
39. Strugar TL, Kuo A, Seite S, Lin M, Lio P. Connecting the dots: from skin
barrier dysfunction to allergic sensitization, and the role of moisturizers in
repairing the skin barrier. J Drugs Dermatol. (2019) 18:581.
40. Strid J, Hourihane J, Kimber I, Callard R, Strobel S. Disruption of the stratum
corneum allows potent epicutaneous immunization with protein antigens
resulting in a dominant systemic Th2 response. Eur J Immunol. (2004)
34:2100–9. doi: 10.1002/eji.200425196
41. Spergel JM, Mizoguchi E, Brewer JP, Martin TR, Bhan AK, Geha
RS. Epicutaneous sensitization with protein antigen induces localized
allergic dermatitis and hyperresponsiveness to methacholine after single
exposure to aerosolized antigen in mice. J Clin Invest. (1998) 101:1614–
22. doi: 10.1172/JCI1647
42. Kezic S, Kemperman PM, Koster ES, de Jongh CM, Thio HB, Campbell LE,
et al. Loss-of-function mutations in the filaggrin gene lead to reduced level of
natural moisturizing factor in the stratum corneum. J Invest Dermatol. (2008)
128:2117–9. doi: 10.1038/jid.2008.29
43. O’Regan GM, Sandilands A, W.McLean HI, Irvine AD.
Filaggrin in atopic dermatitis. J Allergy Clin Immunol. (2008)
122:689–93. doi: 10.1016/j.jaci.2008.08.002
44. Guttman-Yassky E, Zhou L, Krueger JG. The skin as an immune organ:
tolerance versus effector responses and applications to food allergy
and hypersensitivity reactions. J Allergy Clin Immunol. (2019) 144:362–
74. doi: 10.1016/j.jaci.2019.03.021
45. Irvine AD, McLean WH, Leung DY. Filaggrin mutations associated
with skin and allergic diseases. N Engl J Med. (2011) 365:1315–
27. doi: 10.1056/NEJMra1011040
46. Tan HT, Ellis JA, Koplin JJ, Matheson MC, Gurrin LC, Lowe AJ, et al.
Filaggrin loss-of-function mutations do not predict food allergy over and
above the risk of food sensitization among infants. J Allergy Clin Immunol.
(2012) 130:1211–3 e3. doi: 10.1016/j.jaci.2012.07.022
47. Palmer CN, Irvine AD, Terron-Kwiatkowski A, Zhao Y, Liao H, Lee SP,
et al. Common loss-of-function variants of the epidermal barrier protein
filaggrin are a major predisposing factor for atopic dermatitis. Nat Genet.
(2006) 38:441–6. doi: 10.1038/ng1767
48. O’Regan GM, Sandilands A, McLean WH, Irvine AD. Filaggrin in
atopic dermatitis. J Allergy Clin Immunol. (2009) 124(3 Suppl. 2):R2–
6. doi: 10.1016/j.jaci.2009.07.013
49. Hudson TJ. Skin barrier function and allergic risk. Nat Genet. (2006) 38:399–
400. doi: 10.1038/ng0406-399
50. Ziegler SF, Artis D. Sensing the outside world: TSLP regulates barrier
immunity. Nat Immunol. (2010) 11:289–93. doi: 10.1038/ni.1852
51. Zhang Z, Hener P, Frossard N, Kato S, Metzger D, Li M, et al. Thymic
stromal lymphopoietin overproduced by keratinocytes in mouse skin
aggravates experimental asthma. Proc Natl Acad Sci USA. (2009) 106:1536–
41. doi: 10.1073/pnas.0812668106
52. Leyva-Castillo JM, Hener P, Jiang H, Li M. TSLP produced by
keratinocytes promotes allergen sensitization through skin and thereby
triggers atopic march in mice. J Invest Dermatol. (2013) 133:154–
63. doi: 10.1038/jid.2012.239
53. Noti M, Wojno ED, Kim BS, Siracusa MC, Giacomin PR, Nair MG,
et al. Thymic stromal lymphopoietin-elicited basophil responses promote
eosinophilic esophagitis. Nat Med. (2013) 19:1005–13. doi: 10.1038/nm.3281
54. Noti M, Kim BS, Siracusa MC, Rak GD, Kubo M, Moghaddam AE, et al.
Exposure to food allergens through inflamed skin promotes intestinal food
allergy through the thymic stromal lymphopoietin-basophil axis. J Allergy
Clin Immunol. (2014) 133:1390–9:1399 e1–6. doi: 10.1016/j.jaci.2014.01.021
55. Xu D, Chan WL, Leung BP, Huang F, Wheeler R, Piedrafita D, et al. Selective
expression of a stable cell surface molecule on type 2 but not type 1 helper T
cells. J Exp Med. (1998) 187:787–94. doi: 10.1084/jem.187.5.787
56. Allakhverdi Z, Smith DE, Comeau MR, Delespesse G. Cutting edge: the ST2
ligand IL-33 potently activates and drives maturation of human mast cells. J
Immunol. (2007) 179:2051–4. doi: 10.4049/jimmunol.179.4.2051
57. Kondo Y, Yoshimoto T, Yasuda K, Futatsugi-Yumikura S, Morimoto
M, Hayashi N, et al. Administration of IL-33 induces airway
hyperresponsiveness and goblet cell hyperplasia in the lungs in
the absence of adaptive immune system. Int Immunol. (2008)
20:791–800. doi: 10.1093/intimm/dxn037
58. Salimi M, Barlow JL, Saunders SP, Xue L, Gutowska-Owsiak D, Wang X,
et al. A role for IL-25 and IL-33-driven type-2 innate lymphoid cells in atopic
dermatitis. J Exp Med. (2013) 210:2939–50. doi: 10.1084/jem.20130351
59. Siede J, Frohlich A, Datsi A, Hegazy AN, Varga DV, Holecska V, et al. IL-33
receptor-expressing regulatory T cells are highly activated, Th2 biased and
suppress CD4 T cell proliferation through IL-10 and TGFbeta release. PLoS
ONE. (2016) 11:e0161507. doi: 10.1371/journal.pone.0161507
60. Moro K, Yamada T, Tanabe M, Takeuchi T, Ikawa T, Kawamoto
H, et al. Innate production of T(H)2 cytokines by adipose tissue-
associated c-Kit(+)Sca-1(+) lymphoid cells. Nature. (2010) 463:540–
4. doi: 10.1038/nature08636
61. Molofsky AB, Savage AK, Locksley RM, Interleukin-33 in
Tissue Homeostasis, Injury, and Inflammation. Immunity. (2015)
42:1005-19. doi: 10.1016/j.immuni.2015.06.006
62. Savinko T, Karisola P, Lehtimaki S, Lappetelainen AM, Haapakoski R, Wolff
H, et al. ST2 regulates allergic airway inflammation and T-cell polarization
in epicutaneously sensitized mice. J Invest Dermatol. (2013) 133:2522–
9. doi: 10.1038/jid.2013.195
63. Savinko T, Matikainen S, Saarialho-Kere U, Lehto M, Wang G, Lehtimaki
S, et al. IL-33 and ST2 in atopic dermatitis: expression profiles and
modulation by triggering factors. J Invest Dermatol. (2012) 132:1392–
400. doi: 10.1038/jid.2011.446
64. Tordesillas L, Goswami R, Benede S, Grishina G, Dunkin D, Jarvinen KM,
et al. Skin exposure promotes a Th2-dependent sensitization to peanut
allergens. J Clin Invest. (2014) 124:4965–75. doi: 10.1172/JCI75660
65. Galand C, Leyva-Castillo JM, Yoon J, Han A, Lee MS, McKenzie ANJ,
et al. IL-33 promotes food anaphylaxis in epicutaneously sensitized
mice by targeting mast cells. J Allergy Clin Immunol. (2016) 138:1356–
66. doi: 10.1016/j.jaci.2016.03.056
66. Fort MM, Cheung J, Yen D, Li J, Zurawski SM, Lo S, et al. IL-25 induces IL-
4, IL-5, and IL-13 and Th2-associated pathologies in vivo. Immunity. (2001)
15:985–95. doi: 10.1016/S1074-7613(01)00243-6
67. Angkasekwinai P, Park H, Wang YH, Wang YH, Chang SH, Corry
DB, et al. Interleukin 25 promotes the initiation of proallergic type
2 responses. J Exp Med. (2007) 204:1509–17. doi: 10.1084/jem.200
61675
68. Corrigan CJ, Wang W, Meng Q, Fang C, Eid G, Caballero MR, et al.
Allergen-induced expression of IL-25 and IL-25 receptor in atopic asthmatic
airways and late-phase cutaneous responses. J Allergy Clin Immunol. (2011)
128:116–24. doi: 10.1016/j.jaci.2011.03.043
69. Kim BE, Bin L, Ye YM, Ramamoorthy P, D.Leung YM, IL-25 enhances
HSV-1 replication by inhibiting filaggrin expression, and acts synergistically
Frontiers in Immunology | www.frontiersin.org 9August 2020 | Volume 11 | Article 1907
Yang et al. Research Progress in Atopic March
with Th2 cytokines to enhance HSV-1 replication. J Invest Dermatol. (2013)
133:2678–85. doi: 10.1038/jid.2013.223
70. Lee JB, Chen CY, Liu B, Mugge L, Angkasekwinai P, Facchinetti V, et al.
IL-25 and CD4(+) TH2 cells enhance type 2 innate lymphoid cell-derived
IL-13 production, which promotes IgE-mediated experimental food allergy. J
Allergy Clin Immunol. (2016) 137:1216–25 e5. doi: 10.1016/j.jaci.2015.09.019
71. Kang CM, Jang AS, Ahn MH, Shin JA, Kim JH, Choi YS, et al.
Interleukin-25 and interleukin-13 production by alveolar macrophages
in response to particles. Am J Respir Cell Mol Biol. (2005) 33:290–
6. doi: 10.1165/rcmb.2005-0003OC
72. Han H, Roan F, Ziegler SF, The atopic march: current insights into skin
barrier dysfunction and epithelial cell-derived cytokines. Immunol Rev.
(2017) 278:116–30. doi: 10.1111/imr.12546
73. Hill DA, Spergel JM. The atopic march: critical evidence
and clinical relevance. Ann Allergy Asthma Immunol. (2018)
120:131–7. doi: 10.1016/j.anai.2017.10.037
74. Li M. Current evidence of epidermal barrier dysfunction and thymic
stromal lymphopoietin in the atopic march. Eur Respir Rev. (2014) 23:292–
8. doi: 10.1183/09059180.00004314
75. Saad MJ, Santos A, Prada PO. Linking gut microbiota and inflammation
to obesity and insulin resistance. Physiology (Bethesda). (2016) 31:283–
93. doi: 10.1152/physiol.00041.2015
76. Kennedy EA, Connolly J, Hourihane JO, Fallon PG, McLean WHI, Murray
D, et al. Skin microbiome before development of atopic dermatitis: early
colonization with commensal staphylococci at 2 months is associated with
a lower risk of atopic dermatitis at 1 year. J Allergy Clin Immunol. (2017)
139:166–72. doi: 10.1016/j.jaci.2016.07.029
77. Forno E, Onderdonk AB, McCracken J, Litonjua AA, Laskey D, Delaney
ML, et al. Diversity of the gut microbiota and eczema in early life. Clin Mol
Allergy. (2008) 6:11. doi: 10.1186/1476-7961-6-11
78. Abrahamsson TR, Jakobsson HE, Andersson AF, Bjorksten B, Engstrand
L, Jenmalm MC. Low diversity of the gut microbiota in infants with
atopic eczema. J Allergy Clin Immunol. (2012) 129:434–40, 440 e1–
2. doi: 10.1016/j.jaci.2011.10.025
79. Penders J, Thijs C, van den Brandt PA, Kummeling I, Snijders B,
Stelma F, et al. Gut microbiota composition and development of atopic
manifestations in infancy: the KOALA Birth Cohort Study. Gut. (2007)
56:661–7. doi: 10.1136/gut.2006.100164
80. Azad MB, Konya T, Guttman DS, Field CJ, Sears MR, HayGlass KT, et al.
Infant gut microbiota and food sensitization: associations in the first year of
life. Clin Exp Allergy. (2015) 45:632–43. doi: 10.1111/cea.12487
81. Abrahamsson TR, Jakobsson HE, Andersson AF, Bjorksten B, Engstrand
L, Jenmalm MC. Low gut microbiota diversity in early infancy precedes
asthma at school age. Clin Exp Allergy. (2014) 44:842–50. doi: 10.1111/cea.
12253
82. Teo SM, Mok D, Pham K, Kusel M, Serralha M, Troy N, et al. The
infant nasopharyngeal microbiome impacts severity of lower respiratory
infection and risk of asthma development. Cell Host Microbe. (2015) 17:704–
15. doi: 10.1016/j.chom.2015.03.008
83. Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D,
et al. Commensal microbe-derived butyrate induces the differentiation of
colonic regulatory T cells. Nature. (2013) 504:446–50. doi: 10.1038/nature
12721
84. Song H, Yoo Y, Hwang J, Na YC, Kim HS, Faecalibacterium
prausnitzii subspecies-level dysbiosis in the human gut microbiome
underlying atopic dermatitis. J Allergy Clin Immunol. (2016)
137:852–60. doi: 10.1016/j.jaci.2015.08.021
85. Johnson JL, Jones MB, Cobb BA. Bacterial capsular polysaccharide prevents
the onset of asthma through T-cell activation. Glycobiology. (2015) 25:368–
75. doi: 10.1093/glycob/cwu117
86. Martino D, Joo JE, Sexton-Oates A, Dang T, Allen K, Saffery R, et al.
Epigenome-wide association study reveals longitudinally stable DNA
methylation differences in CD4+T cells from children with IgE-mediated
food allergy. Epigenetics. (2014) 9:998–1006. doi: 10.4161/epi.28945
87. Hong X, Hao K, Ladd-Acosta C, Hansen KD, Tsai HJ, Liu X, et al.
Genome-wide association study identifies peanut allergy-specific loci and
evidence of epigenetic mediation in US children. Nat Commun. (2015)
6:6304. doi: 10.1038/ncomms7304
88. Xu CJ, Soderhall C, Bustamante M, Baiz N, Gruzieva O, Gehring U, et al.
DNA methylation in childhood asthma: an epigenome-wide meta-analysis.
Lancet Respir Med. (2018) 6:379–388. doi: 10.1016/S2213-2600(18)30052-3
89. Peng C, Van Meel ER, Cardenas A, Rifas-Shiman SL, Sonawane AR, Glass
KR, et al. Epigenome-wide association study reveals methylation pathways
associated with childhood allergic sensitization. Epigenetics. (2019) 14:445–
66. doi: 10.1080/15592294.2019.1590085
90. Luo J, Li Y, Gong R, The mechanism of atopic march may be the ’social’
event of cells and molecules (Review). Int J Mol Med. (2010) 26:779–
85. doi: 10.3892/ijmm_00000525
91. Marenholz I, Esparza-Gordillo J, Ruschendorf F, Bauerfeind A, Strachan DP,
Spycher BD, et al. Meta-analysis identifies seven susceptibility loci involved
in the atopic march. Nat Commun. (2015) 6:8804. doi: 10.1038/ncomms9804
92. Gupta J, Johansson E, Bernstein JA, Chakraborty R, Khurana Hershey GK,
Rothenberg ME, et al. Resolving the etiology of atopic disorders by using
genetic analysis of racial ancestry. J Allergy Clin Immunol. (2016) 138:676–
99. doi: 10.1016/j.jaci.2016.02.045
93. Paller AS, Spergel JM, Mina-Osorio P, Irvine AD. The atopic march and
atopic multimorbidity: many trajectories, many pathways. J Allergy Clin
Immunol. (2019) 143:46–55. doi: 10.1016/j.jaci.2018.11.006
94. Han H, Xu W, Headley MB, Jessup HK, Lee KS, Omori M,
et al. Thymic stromal lymphopoietin (TSLP)-mediated dermal
inflammation aggravates experimental asthma. Mucosal Immunol. (2012)
5:342–51. doi: 10.1038/mi.2012.14
95. Akei HS, Brandt EB, Mishra A, Strait RT, Finkelman FD, Warrier MR,
et al. Epicutaneous aeroallergen exposure induces systemic TH2 immunity
that predisposes to allergic nasal responses. J Allergy Clin Immunol. (2006)
118:62–9. doi: 10.1016/j.jaci.2006.04.046
96. Busse WW. The atopic march: fact or folklore? Ann Allergy Asthma
Immunol. (2018) 120:116–18. doi: 10.1016/j.anai.2017.10.029
97. Martinez FD, Wright AL, Taussig LM, Holberg CJ, Halonen M,
Morgan WJ. Asthma and wheezing in the first six years of life. The
Group Health Medical Associates. N Engl J Med. (1995) 332:133–
8. doi: 10.1056/NEJM199501193320301
98. Burgess JA, Dharmage SC, Byrnes GB, Matheson MC, Gurrin LC, Wharton
CL, et al. Childhood eczema and asthma incidence and persistence: a cohort
study from childhood to middle age. J Allergy Clin Immunol. (2008) 122:280–
5. doi: 10.1016/j.jaci.2008.05.018
99. Abo-Zaid G, Sharpe RA, Fleming LE, Depledge M, Osborne NJ. Association
of infant eczema with childhood and adult asthma: analysis of data from
the 1958 birth cohort study. Int J Environ Res Public Health. (2018)
15:1415. doi: 10.3390/ijerph15071415
100. Soller L, Ben-Shoshan M, Harrington DW, Fragapane J, Joseph L, St Pierre
Y, et al. Overall prevalence of self-reported food allergy in Canada. J Allergy
Clin Immunol. (2012) 130:986–8. doi: 10.1016/j.jaci.2012.06.029
101. Belgrave DC, Simpson A, Buchan IE, Custovic A. Atopic dermatitis and
respiratory allergy: what is the link. Curr Dermatol Rep. (2015) 4:221–
7. doi: 10.1007/s13671-015-0121-6
102. Barberio G, Pajno GB, Vita D, Caminiti L, Canonica GW, Passalacqua
G. Does a ’reverse’ atopic march exist? Allergy. (2008) 63:1630–
2. doi: 10.1111/j.1398-9995.2008.01710.x
103. Acevedo N, Sanchez J, Zakzuk J, Bornacelly A, Quiroz C, Alvarez A, et al.
Particular characteristics of allergic symptoms in tropical environments:
follow up to 24 months in the FRAAT birth cohort study. BMC Pulm Med.
(2012) 12:13. doi: 10.1186/1471-2466-12-13
104. Kull I, Bohme M, Wahlgren CF, Nordvall L, Pershagen G, Wickman
M, Breast-feeding reduces the risk for childhood eczema. J Allergy Clin
Immunol. (2005) 116:657–61. doi: 10.1016/j.jaci.2005.04.028
105. von Berg A, Filipiak-Pittroff B, Schulz H, Hoffmann U, Link E, Sussmann M,
et al. Allergic manifestation 15 years after early intervention with hydrolyzed
formulas–the GINI Study. Allergy. (2016) 71:210–9. doi: 10.1111/all.12790
106. Wickens K, Black PN, Stanley TV, Mitchell E, Fitzharris P,Tannock GW, et al.
A differential effect of 2 probiotics in the prevention of eczema and atopy: a
double-blind, randomized, placebo-controlled trial. J Allergy Clin Immunol.
(2008) 122:788–94. doi: 10.1016/j.jaci.2008.07.011
107. Du Toit G, Roberts G, Sayre PH, Bahnson HT, Radulovic S, Santos AF, et al.
Randomized trial of peanut consumption in infants at risk for peanut allergy.
N Engl J Med. (2015) 372:803–13. doi: 10.1056/NEJMoa1414850
Frontiers in Immunology | www.frontiersin.org 10 August 2020 | Volume 11 | Article 1907
Yang et al. Research Progress in Atopic March
108. Katz Y, Rajuan N, Goldberg MR, Eisenberg E, Heyman E, Cohen A, et al.
Early exposure to cow’s milk protein is protective against IgE-mediated
cow’s milk protein allergy. J Allergy Clin Immunol. (2010) 126:77–82
e1. doi: 10.1016/j.jaci.2010.04.020
109. Lannero E, Wickman M, van Hage M, Bergstrom A, Pershagen G, Nordvall
L. Exposure to environmental tobacco smoke and sensitisation in children.
Thorax. (2008) 63:172–6. doi: 10.1136/thx.2007.079053
110. Dick S, Friend A, Dynes K, AlKandari F, Doust E, Cowie H, et al. A
systematic review of associations between environmental exposures and
development of asthma in children aged up to 9 years. BMJ Open. (2014)
4:e006554. doi: 10.1136/bmjopen-2014-006554
111. Portnoy J, Kennedy K, Sublett J, Phipatanakul W, Matsui E, Barnes
C, et al. Environmental assessment and exposure control: a practice
parameter–furry animals. Ann Allergy Asthma Immunol. (2012) 108:223
e1–15. doi: 10.1016/j.anai.2012.02.015
112. Iikura Y, Naspitz CK, Mikawa H, Talaricoficho S, Baba M, Sole D, et al.
Prevention of asthma by ketotifen in infants with atopic dermatitis. Ann
Allergy. (1992) 68:233–6.
113. Bustos GJ, Bustos D, Bustos GJ, Romero O. Prevention of asthma with
ketotifen in preasthmatic children: a three-year follow-up study. Clin Exp
Allergy. (1995) 25:568–73. doi: 10.1111/j.1365-2222.1995.tb01096.x
114. Warner JO, Businco L, Casimir G, Diepgen TL, Kjellman M, Knol K, et al.
Allergic factors associated with the development of asthma the influence of
cetirizine in a double-blind, randomised, placebo-controlled trial: first results
of ETAC. Early Treatment of the Atopic Child. Pediatr Allergy Immunol.
(1998) 9:116–24. doi: 10.1111/j.1399-3038.1998.tb00356.x
115. Barnes PJ. Glucocorticoids. Chem Immunol Allergy. (2014) 100:311–
6. doi: 10.1159/000359984
116. Jacobsen L, Wahn U, Bilo MB. Allergen-specific immunotherapy provides
immediate, long-term and preventive clinical effects in children and adults:
the effects of immunotherapy can be categorised by level of benefit -the
centenary of allergen specific subcutaneous immunotherapy. Clin Transl
Allergy. (2012) 2:8. doi: 10.1186/2045-7022-2-8
117. Kawauchi H, Goda K, Tongu M, Yamada T, Aoi N, Morikura I, et al.
Short review on sublingual immunotherapy for patients with allergic
rhinitis: from bench to bedside. Adv Otorhinolaryngol. (2011) 72:103–
6. doi: 10.1159/000324631
118. Bahceciler NN, Arikan C, Taylor A, Akdis M, Blaser K, Barlan IB, et al.
Impact of sublingual immunotherapy on specific antibody levels in asthmatic
children allergic to house dust mites. Int Arch Allergy Immunol. (2005)
136:287–94. doi: 10.1159/000083956
119. Zhong H, Deng X, Song Z, Darsow U, Chen W, Chen S, et al. Immunological
changes after ASIT in AD allergen-specific immunotherapy and their
potential correlation with clinical response in patients with atopic dermatitis
patients sensitized to house dust mite. J Eur Acad Dermatol Venereol. (2015)
29:1318–24. doi: 10.1111/jdv.12813
120. Besh O, Besh D, Sorkopud O, Kondratiuk M, Slaba O, Zhakun I, et al. ASIT
as the component of bronchial asthma’s therapy can improve the adherence
to the treatment. Wiad Lek. (2018) 71:849–54.
121. Karakoc-Aydiner E, Eifan AO, Baris S, Gunay E, Akturk E, Akkoc T, et al.
Long-term effect of sublingual and subcutaneous immunotherapy in dust
mite-allergic children with asthma/rhinitis: a 3-year prospective randomized
controlled trial. J Investig Allergol Clin Immunol. (2015) 25:33442.
122. Canonica GW, Cox L, Pawankar R, Baena-Cagnani CE, Blaiss
M, Bonini S, et al. Sublingual immunotherapy: World Allergy
Organization position paper 2013 update. World Allergy Organ J. (2014)
7:6. doi: 10.1186/1939-4551-7-6
123. Hofmann AM, Scurlock AM, Jones SM, Palmer KP, Lokhnygina Y, Steele
PH, et al. Safety of a peanut oral immunotherapy protocol in children
with peanut allergy. J Allergy Clin Immunol. (2009) 124:286–91:291 e1–
6. doi: 10.1016/j.jaci.2009.03.045
124. Skripak JM, Nash SD, Rowley H, Brereton NH, Oh S, Hamilton RG,
et al. A randomized, double-blind, placebo-controlled study of milk oral
immunotherapy for cow’s milk allergy. J Allergy Clin Immunol. (2008)
122:1154–60. doi: 10.1016/j.jaci.2008.09.030
125. Keet CA, Frischmeyer-Guerrerio PA, Thyagarajan A, Schroeder JT, Hamilton
RG, Boden S, et al. The safety and efficacy of sublingual and oral
immunotherapy for milk allergy. J Allergy Clin Immunol. (2012) 129:448–
55:455 e1–5. doi: 10.1016/j.jaci.2011.10.023
126. Esquivel A, Busse WW, Calatroni A, Togias AG, Grindle KG, Bochkov
YA, et al. Effects of omalizumab on rhinovirus infections, illnesses, and
exacerbations of asthma. Am J Respir Crit Care Med. (2017) 196:985–
92. doi: 10.1164/rccm.201701-0120OC
127. Gandhi NA, Bennett BL, Graham NM, Pirozzi G, Stahl N, Yancopoulos GD.
Targeting key proximal drivers of type 2 inflammation in disease. Nat Rev
Drug Discov. (2016) 15:35–50. doi: 10.1038/nrd4624
128. Chang HY. Nadeau KC, IL-4Ralpha inhibitor for atopic disease. Cell. (2017)
170:222. doi: 10.1016/j.cell.2017.06.046
129. Gauvreau GM, O’Byrne PM, Boulet LP, Wang Y, Cockcroft D, Bigler
J, et al. Effects of an anti-TSLP antibody on allergen-induced asthmatic
responses. N Engl J Med. (2014) 370:2102–10. doi: 10.1056/NEJMoa14
02895
130. Davidson WF, Leung DYM, Beck LA, Berin CM, Boguniewicz M,
Busse WW, et al. Report from the national institute of allergy and
infectious diseases workshop on “Atopic dermatitis and the atopic march:
mechanisms and interventions”. J Allergy Clin Immunol. (2019) 143:894–
913. doi: 10.1016/j.jaci.2019.01.003
Conflict of Interest: The authors declare that the research was conducted in the
absence of any commercial or financial relationships that could be construed as a
potential conflict of interest.
Copyright © 2020 Yang, Fu and Zhou. This is an open-access article distributed
under the terms of the Creative Commons Attribution License (CC BY). The use,
distribution or reproduction in other forums is permitted, provided the original
author(s) and the copyright owner(s) are credited and that the original publication
in this journal is cited, in accordance with accepted academic practice. No use,
distribution or reproduction is permitted which does not comply with these terms.
Frontiers in Immunology | www.frontiersin.org 11 August 2020 | Volume 11 | Article 1907
... Over the years, the role of nutrition, especially weaning, in the context of allergic diseases has been extensively studied, contributing to new guidelines for health professionals [17,20]. In fact, although increased IgE-mediated reactivity to food proteins has been found in many patients with allergic diseases such as AD [17,[21][22][23][24][25], according to some authors, their relationship should be interpreted in a multimorbidity framework, thus their co-occurrence does not imply any specific relationship between them and certainly not a progressive or causal relationship, thus questioning the legitimacy of the atopic march model [1,17,[21][22][23][24][25]. Indeed, while, traditionally, the allergic march has been described as a sequence beginning with FA, it is nowadays known that allergic diseases are complex, multifactorial, and caused by a variety of distinct mechanisms responsible for multiple heterogeneous clinical phenotypes [1,[21][22][23][24][25]. ...
... Over the years, the role of nutrition, especially weaning, in the context of allergic diseases has been extensively studied, contributing to new guidelines for health professionals [17,20]. In fact, although increased IgE-mediated reactivity to food proteins has been found in many patients with allergic diseases such as AD [17,[21][22][23][24][25], according to some authors, their relationship should be interpreted in a multimorbidity framework, thus their co-occurrence does not imply any specific relationship between them and certainly not a progressive or causal relationship, thus questioning the legitimacy of the atopic march model [1,17,[21][22][23][24][25]. Indeed, while, traditionally, the allergic march has been described as a sequence beginning with FA, it is nowadays known that allergic diseases are complex, multifactorial, and caused by a variety of distinct mechanisms responsible for multiple heterogeneous clinical phenotypes [1,[21][22][23][24][25]. ...
... In fact, although increased IgE-mediated reactivity to food proteins has been found in many patients with allergic diseases such as AD [17,[21][22][23][24][25], according to some authors, their relationship should be interpreted in a multimorbidity framework, thus their co-occurrence does not imply any specific relationship between them and certainly not a progressive or causal relationship, thus questioning the legitimacy of the atopic march model [1,17,[21][22][23][24][25]. Indeed, while, traditionally, the allergic march has been described as a sequence beginning with FA, it is nowadays known that allergic diseases are complex, multifactorial, and caused by a variety of distinct mechanisms responsible for multiple heterogeneous clinical phenotypes [1,[21][22][23][24][25]. However, some authors argue for the existence of some mechanistic explanations for the progression of atopic march in a proportion of affected individuals, specifically those with an altered skin barrier [1,[22][23][24][25]. ...
Article
Full-text available
One of the most frequent triggers of food anaphylaxis in pediatric age but also among the most common, early, and complex causes of childhood food allergy is cow’s milk protein allergy (CMPA). The diagnostic course and management of this allergy is defined in a complex clinical picture due to several factors. First of all, the epidemiological data are not uniform, mainly as a consequence of the diagnostic methodology used in the various studies and the different age ranges covered. In addition, there is the complexity of terminology, since although CMPA traditionally refers to immune-mediated reactions to cow’s milk, it is a term encompassing numerous clinical features with different symptoms and the requirement for specific treatments. Moreover, the differential diagnosis with other very frequent diseases, especially in the first year of life, such as gastro-esophageal reflux disease or colic, is still complex. This can result in misdiagnosis and incorrect treatment, with harmful health consequences and significant economic repercussions. In this context, the combination of several omics sciences together, which have already proved useful in clarifying the allergenicity of cow’s milk proteins with greater precision, could improve the diagnostic tests currently in use through the identification of new, more specific, and precise biomarkers that make it possible to improve diagnostic accuracy and predict the patient’s response to the various available treatments for the recovery of tolerance.
... . Упродовж останніх років зростає кількість алергічних захворювань (АЗ), особливо з-поміж дітей. Приблизно 20% населення всього світу страждає цими захворюваннями [43]. Згідно з даними ВООЗ, у всьому світі 260 мільйонів людей хворіють на бронхіальну астму (БА), і у 2019 році вона спричинила близько 460 000 смертей [32]. ...
... На атопічний дерматит (АД) страждають 17-30% дітей [37,43]. Згідно з даними епідеміологічних досліджень, діти з АД мають прояви проявами даного захворювання у 45-48% до 6 місяців, у 60% -до 1 року та до 85-89% -до 5 років [37,43]. ...
... На атопічний дерматит (АД) страждають 17-30% дітей [37,43]. Згідно з даними епідеміологічних досліджень, діти з АД мають прояви проявами даного захворювання у 45-48% до 6 місяців, у 60% -до 1 року та до 85-89% -до 5 років [37,43]. Сенсибілізація до інгаляційних алергенів частіше виникає в дітей з АД після трьох років [37]. ...
Article
Full-text available
Aim: review of modern data of scientific medical literature regarding the prevalence, comorbidity, risk factors, features of pathogenesis, diagnosis, principles of treatment and prevention of allergic diseases in children. In recent years, the number of allergic diseases (ADs) has been increasing, especially among children. Approximately 20% of the world's population suffers from these diseases. Comorbid ADs are quite common. Thus, in 67–85% of children, allergic rhinitis is combined with bronchial asthma. Children with atopic dermatitis are diagnosed with allergic rhinitis in 61,8% of cases, and bronchial asthma in 29,4% of cases. The risk of developing bronchial asthma or allergic rhinitis is higher in children with food polysensitization, and infants and young children with atopic dermatitis have a high risk of developing both food allergy and asthma and allergic rhinitis. According to modern data, most ADs have common causal mechanisms. In the development and progression of asthma and allergic rhinitis and atopic dermatitis, the hypotheses of mechanistic damage of the epithelial barrier and biodiversity, related to each other, are considered. In ADs, mechanisms related to allergen-specific immunoglobulin E and non-allergic coexisting mechanisms play an important role. In addition, views on the formation of the sequence of the development of ADs ("allergic march") from the perspective of multimorbidity and phenotyping are increasingly expanding. Pathogenetic treatment of ADs is aimed at controlling the chronic T-helper 2, caused by the inflammatory process in target organs. Secondary and tertiary prevention of ADs in the early stages improves the quality of life of patients and improves the course of atopic conditions. Conclusions. The prevalence of ADs in modern conditions is pandemic in nature. The hypothesis of mechanical damage to the epithelial barrier is significant in the development of ADs. ADs, in particular in children, can be combined, creating multi- and comorbid conditions, and aggravate the course of these diseases. Timely diagnosis of ADs is important, and a personalized approach to the treatment and prevention of ADs is considered the most rational.
... respectively) compared to children without eczema. 8 In Korea, a recent nationwide study also demonstrated that the ORs for asthma and allergic rhinitis increased in severe AD compared to mild-to-moderate AD (6.55 vs 2.62 and 11.02 vs. 4.88, respectively). 9 Furthermore, there is growing evidence indicating that severe AD is associated with chronic systemic diseases beyond allergic diseases, including cardiovascular, autoimmune disease, inflammatory bowel disease (IBD), and even malignant tumors. ...
... Our findings would seem to corroborate the link between dysbiosis and the risk of atopic dermatitis and respiratory infections, demonstrating that supplementing infants with peculiar bifidobacterial species could reduce the incidence of their manifestation at least during the first year of life [50,51]. Atopic dermatitis is considered the first step of the so-called 'atopic march' which, due to the concomitant sensitization to foods and aeroallergens in early childhood, predispose children to asthma and allergic rhinitis in late childhood and adulthood [52,53]. ...
Article
Full-text available
Cesarean section is considered a possible trigger of atopy and gut dysbiosis in newborns. Bifidobacteria, and specifically B. bifidum, are thought to play a central role in reducing the risk of atopy and in favoring gut eubiosis in children. Nonetheless, no trial has ever prospectively investigated the role played by this single bacterial species in preventing atopic manifestations in children born by cesarean section, and all the results published so far refer to mixtures of probiotics. We have therefore evaluated the impact of 6 months of supplementation with B. bifidum PRL2010 on the incidence, in the first year of life, of atopy, respiratory tract infections, and dyspeptic syndromes in 164 children born by cesarean (versus 249 untreated controls). The results of our multicenter, randomized, and controlled trial have shown that the probiotic supplementation significantly reduced the incidence of atopic dermatitis, upper and lower respiratory tract infections, and signs and symptoms of dyspeptic syndromes. Concerning the gut microbiota, B. bifidum supplementation significantly increased α-biodiversity and the relative values of the phyla Bacteroidota and Actinomycetota, of the genus Bacteroides, Bifidobacterium and of the species B. bifidum and reduced the relative content of Escherichia/Shigella and Haemophilus. A 6-month supplementation with B. bifidum in children born by cesarean section reduces the risk of gut dysbiosis and has a positive clinical impact that remains observable in the following 6 months of follow-up.
... Глобальный рост числа аллергических заболеваний, который наблюдается в последнее время, значительно снижает качество жизни людей. Мировая статистика свидетельствует о том, что около 20% населения земного шара страдает от той или иной аллергопатологии [1]. Значительный вклад вносит и пыльцевая аллергия. ...
Article
Введение. Во всем мире наблюдается рост числа аллергических заболеваний. Внедрение рекомбинантных технологий в аллергологию позволило определить белковые компоненты, отвечающие за формирование аллергической реакции. Синтез рекомбинантных аллергенов дает возможность целенаправленно получать пептиды с заданными свойствами. Цель. Получить рекомбинантный полипептид Bet v 1.0101 в прокариотических клетках Escherichia coli, штамм BL21 (DE3). Материалы и методы. В качестве матрицы использована тотальная РНК пыльцы березы повислой, собранная на территории Республики Беларусь. Для получения целевого полипептида использована прокариотическая система Escherichia coli, штамм BL21(DE3), и экспрессирующий вектор pJC40. Очистка проведена методом металл-хелатной хроматографии в денатурирующих условиях. Для подтверждения специфичности белка применен разработанный иммуноферментный лиа-тест, который основан на реакции агглютинации («антиген – антитело»), где в качестве антигена выступает сорбированный на нитроцеллюлозной мембране полипептид Bet v 1.0101. Результаты. Получен высокоочищенный рекомбинантный полипептид Веt v 1 (изоформа Bet v 1.0101), не содержащий примесей бактериальных белков, обладающий антигенными свойствами. Показана возможность использования белка для выявления специфических антител в иммуноферментном лиа-тесте с референс-сыворотками. Заключение. Открывается возможность использования полученного рекомбинантного полипептида для диагностики аллергических заболеваний и в аллергенспецифической иммунотерапии. Introduction. Allergic diseases are on the rise worldwide. The introduction of recombinant technologies in allergology has allowed identifying protein components responsible for allergic reaction formation. Recombinant allergens synthesis makes it possible to purposefully obtain peptides with targeted properties. Purpose. To obtain recombinant polypeptide Bet v 1.0101 in prokaryotic cells of Escherichia coli, strain BL21 (DE3). Materials and methods. The total RNA of birch pollen collected on the territory of the Republic of Belarus was used as a matrix. To obtain the target polypeptide, the prokaryotic Escherichia coli system, strain BL21(DE3), and the expression vector pJC40 were used. The purification was carried out by metal-chelate chromatography under denaturing conditions. To confirm the protein specificity, an elaborated enzyme immunoassay was used based on agglutination reaction ("antigen – antibody"), where the polypeptide Bet v 1.0101 sorbed on a nitrocellulose membrane acted as antigen. Results. A highly purified recombinant polypeptide Bet v 1 (isoform Bet v 1.0101), not containing bacterial protein impurities and possessing antigenic properties, was obtained. The feasibility of using the protein for specific antibodies detecting by enzyme immunoassay with reference sera has been shown. Conclusion. The opportunity of using the obtained recombinant polypeptide in allergic conditions diagnostics and in allergen-specific immunotherapy has been discovered.
... Considering these findings, the discovery that the ethanolic extract from C. linum biomass can significantly suppress key inflammatory factors in both macrophage and keratinocyte cells presents numerous possibilities for treating various serious inflammatory conditions, including skin diseases. Two studies demonstrated that allergic diseases follow a specific sequence, beginning with atopic dermatitis and food allergies in infancy and progressing gradually to allergic asthma and rhinitis in childhood [29,30]. However, the mechanisms underlying the so-called atopic march remain incompletely understood, given the complexity and heterogeneity of atopic diseases, which result from a combination of genetic, environmental, and epigenetic factors [29]. ...
Article
Full-text available
Utilizing plant-based resources, particularly their by-products, aligns with sustainability principles and circular bioeconomy, contributing to environmental preservation. The therapeutic potential of plant extracts is garnering increasing interest, and this study aimed to demonstrate promising outcomes from an extract obtained from an underutilized plant waste. Chaetomorpha linum, an invasive macroalga found in the Orbetello Lagoon, thrives in eutrophic conditions, forming persistent mats covering approximately 400 hectares since 2005. The biomass of C. linum undergoes mechanical harvesting and is treated as waste, requiring significant human efforts and economic resources—A critical concern for municipalities. Despite posing challenges to local ecosystems, the study identified C. linum as a natural source of bioactive metabolites. Phytochemical characterization revealed lipids, amino acids, and other compounds with potential anti-inflammatory activity in C. linum extract. In vitro assays with LPS-stimulated RAW 264.7 and TNF-α/IFN-γ-stimulated HaCaT cells showed the extract inhibited reactive oxygen species (ROS), nitric oxide (NO), and prostaglandin E2 (PGE2) productions, and reduced inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) expressions via NF-κB nuclear translocation, in RAW 264.7 cells. It also reduced chemokines (TARC/CCL17, RANTES/CCL5, MCP-1/CCL2, and IL-8) and the cytokine IL-1β production in HaCaT cells, suggesting potential as a therapeutic candidate for chronic diseases like atopic dermatitis. Finally, in silico studies indicated palmitic acid as a significant contributor to the observed effect. This research not only uncovered the untapped potential of C. linum but also laid the foundation for its integration into the circular bioeconomy, promoting sustainable practices, and innovative applications across various industries.
... Долгое время считалось, что АМ характеризуется ранним и последовательным появлением клинических симптомов атопического дерматита (АтД) и пищевой аллергии (ПА) с последующим присоединением к ним аллергических респираторных проявлений [1]. Сейчас выдвигаются и обсуждаются идеи, что далеко не все дети проходят путь АМ одинаково [2]. ...
Article
Recently, the atopic march (AM) is no longer considered a clear and sequential development of successive allergic diseases, such as atopic dermatitis (AD), asthma, and allergic rhinitis (AR). Recent studies have shown that associations between allergic disorders in children are much more complex, and there may be different variants of AM trajectories. Previously, eight classes of AM were described: (1) no allergic disorders, (2) AM, (3) AD and transient wheeze, (4) AD with later-onset rhinitis, (5) transient wheeze and later-onset rhinitis, (6) transient wheeze only, (7) eczema only, and (8) rhinitis only. 48.3% of children exhibit specific types of sequential development of allergic disorders, but only 7% exhibit profiles resembling ‘classic' AM. The aim of this article was to analyze AM trajectories in real clinical practice in children and to discuss the role of component resolved diagnosis (CRD) in the analysis of AM. The article presents anamnestic patient data and CRD results using a multiplex panel. The literature shows that AM classes do not always match the actual clinical presentation of a patient, indicating the presence of previously unreported AM trajectories. Molecular allergy diagnostics can identify true and cross-sensitization in complex clinical cases, allowing for consideration of the causes of established sensitization and conceptualization of clinical symptoms and individual AM trajectory. It is important to ensure that sensitization to true allergens is established and clinically verified. The analysis of anamnestic data can make it difficult to understand the sequence of allergic disease development. However, CRD significantly facilitates diagnostic search by allowing consideration of the order of sensitization and clinical manifestations. This can help predict the possible severity of allergic reactions to established allergens. KEYWORDS: atopic march, atopic dermatitis, allergic rhinitis, asthma, children, component resolved diagnosis, respiratory allergens. FOR CITATION: Lepeshkova T.S., Andronova E.V. Can component resolved diagnosis help establish "atopic march" trajectory? Russian Medical Inquiry. 2024;8(3):163–170 (in Russ.). DOI: 10.32364/2587-6821-2024-8-3-7.
... Cross-sectional and longitudinal studies have speculated that allergic diseases occur following a time-based order: from AD and food allergy in infancy to progressive development into Allergic Asthma (AA) and Allergic Rhinitis (AR) in childhood [6]. ...
Article
Atopic Dermatitis (AD) is a chronic relapsing inflammatory skin disease characterized by recurrent eczematous skin lesions and intense itching. Globally, it affects 10-30% of children and 2-10% of adults, with a two- to three-fold increase in prevalence noted over the last decades
Article
Full-text available
Skin is replete with immunocompetent cells that modulate signaling pathways to maintain a salubrious immunogenic/tolerogenic balance. This fertile immune environment plays a significant role in the development of allergic responses and sensitivities, but the mechanisms underlying these pathways have been underappreciated and underused with respect to developing therapeutics. Among the complex repertoire of cells that promote tolerogenic pathways in the periphery, 2 key classes include dendritic cells and regulatory T (Treg) cells. Immature dendritic cells are the first line of defense, patrolling the periphery, sampling antigens, and secreting cytokines that suppress immune cells and promote the survival of Treg cells. Skin-homing Treg cells also play a critical role in mitigating the reactivity of immune cells, secreting high levels of cytokines that promote tolerance. Therapeutic approaches that capitalize on our knowledge of the rich cellular and molecular environment are emerging and show great promise. We will discuss the advantages and challenges of 5 such strategies and how these therapies might mitigate the atopic march by facilitating tolerance. We conclude that skin is a multifaceted structure that provides a fertile ground for therapeutic discovery. Accordingly, ongoing work in this domain will no doubt continue to deliver exciting progress for improved health outcomes.
Article
Full-text available
Atopic dermatitis (AD) affects up to 20% of children worldwide and is an increasing public health problem, particularly in developed countries. Although AD in infants and young children can resolve, there is a well-recognized increased risk of sequential progression from AD to other atopic diseases, including food allergy (FA), allergic rhinitis, allergic asthma, and allergic rhinoconjunctivitis, a process referred to as the atopic march. The mechanisms underlying the development of AD and subsequent progression to other atopic comorbidities, particularly FA, are incompletely understood and the subject of intense investigation. Other major research objectives are the development of effective strategies to prevent AD and FA, as well as therapeutic interventions to inhibit the atopic march. In 2017, the Division of Allergy, Immunology, and Transplantation of the National Institute of Allergy and Infectious Diseases sponsored a workshop to discuss current understanding and important advances in these research areas and to identify gaps in knowledge and future research directions. International and national experts in the field were joined by representatives from several National Institutes of Health institutes. Summaries of workshop presentations, key conclusions, and recommendations are presented herein.
Article
Full-text available
Objective: The aim: To compare the treatment adherence of patients with Bronchial Asthma (BA) receiving basic treatment and its combination with allergen-specific immunotherapy. Patients and methods: Materials and methods: The study included 104 patients aged from 18 to 50 with BA. All patients were divided into two groups. The main group (MG) consisted of 51 patients receiving basic medical treatment and ASIT. 38 of them received subcutaneous ASIT and 13 patients received sublingual ASIT. The comparative group (CG) consisted of 53 patients who received only basic therapy. The patients' observation duration was for a year. Results: Results: All patients were done the computer spirometry with a bronchodilation test, determination of the total IgE level, questionnaires (quality of life, control of asthma, adherence to treatment). Also the major and minor component of allergy house dust mites and specific IgG4 were determined in patients of main groups. After 12 months of observation in both groups the spirometry rates improved (the main group result was 16.9%, the control group - was 12.8%). The indicators of asthma's control also increased (MG by 28%, in CG - 21%, (p <0,05)). After individual conversations and training the patients improved and adherence to the treatment too. In the beginning of our research the mean level in MG was 3.2 ± 0.3 points, in - CG-3.3 ± 0.2 points (p> 0.05), after 12 months it increased to 6.3 ± 0.2 points in MG vs 5. 8 ± 0.1 points in CG (p <0.05). Conclusion: Conclusion: Patients' combining basic therapy with SIT had significantly better results of the overall BA's controllability compared to the patients' receiving only basic therapy.
Article
Full-text available
The influence of early life exposures on later life disease has for some time provided clues to modifiable risk factors of disease. The “atopic march” is thought to play a role in the progression of allergic diseases and may offer an opportunity to lower asthma’s health and socioeconomic burden, although evidence remains controversial. We aimed to examine the relationship between early life eczema and asthma later in life. Using the National Child Development Study, we examined infant eczema and childhood and adult asthma. Data related to asthma or wheezing bronchitis were available for 13,503 (73%; 95% CI 72⁻74), 11,503 (61%; 95% CI 60⁻61), 12,524 (68%; 95% CI 67⁻69), 11,194 (60%; 95% CI 60⁻60), 9377 (51%; 95% CI 51⁻51), and 9760 (53%; 95% CI 52⁻53) subjects at ages 11, 16, 23, 33, 44, and 50 years, respectively. Logistic regression models were fitted to examine each wave separately before and after adjusting for a range of potential confounders. Generalised estimating equation (GEE) methods were undertaken to examine the associations after pooling all data from questionnaires. The prevalence of self-reported asthma in those that had previously reported infant eczema ranged from 1.0%; 95% CI 0.9⁻1.4 (age 44 years) to 2.2%; 95% CI 2.1⁻2.3 (age 33 years). Participants with infant eczema had a 2⁻3-fold increased risk of reporting asthma in childhood and adulthood; this was 1.6 times at age 44 years when using spirometry measures. Similar effect sizes were observed in the GEE models when considering all participants (OR 2.9; 95% CI 2.6⁻3.2). Childhood and adult asthma were consistently associated with infant eczema both by using the self-reported data and lung measures.
Article
The skin is one of the largest immunologic organs in the body and a continuous target for allergic and immunologic responses. Impairment of the skin barrier increases the likelihood of external antigens and pathogens entering and creating inflammation, which can potentially lead to skin infections, allergies, and chronic inflammatory diseases such as atopic and contact dermatitis. Functionally, the skin barrier can be divided into four different levels. From outermost to innermost, these highly interdependent levels are the microbiome, chemical, physical, and immune levels. The objective of this review is to provide an update on current knowledge about the relationship between skin barrier function and how dysfunction at each level of the skin barrier can lead to allergic sensitization, contact dermatitis, and the atopic march, and examine how to best repair and maintain this barrier through the use of moisturizers. J Drugs Dermatol. 2019;18(6):581-586.
Article
Epigenetic mechanisms integrate both genetic variability and environmental exposures. However, comprehensive epigenome-wide analysis has not been performed across major childhood allergic phenotypes. We examined the association of epigenome-wide DNA methylation in mid-childhood peripheral blood (Illumina HumanMethyl450K) with mid-childhood atopic sensitization, environmental/inhalant and food allergen sensitization in 739 children in two birth cohorts (Project Viva–Boston, and the Generation R Study–Rotterdam). We performed covariate-adjusted epigenome-wide association meta-analysis and employed pathway and regional analyses of results. Seven-hundred and five methylation sites (505 genes) were significantly cross-sectionally associated with mid-childhood atopic sensitization, 1411 (905 genes) for environmental and 45 (36 genes) for food allergen sensitization (FDR
Article
The atopic march recognizes the increased occurrence of asthma, allergic rhinitis, or both after atopic dermatitis (AD) onset. Mechanisms for developing atopic comorbidities after AD onset are poorly understood but can involve the impaired cutaneous barrier, which facilitates cutaneous sensitization. The association can also be driven or amplified in susceptible subjects by a systemic TH2-dominant immune response to cutaneous inflammation. However, these associations might merely involve shared genetic loci and environmental triggers, including microbiome dysregulation, with the temporal sequence reflecting tissue-specific peak time of occurrence of each disease, suggesting more of a clustering of disorders than a march. Prospective longitudinal cohort studies provide an opportunity to explore the relationships between postdermatitis development of atopic disorders and potential predictive phenotypic, genotypic, and environmental factors. Recent investigations implicate disease severity and persistence, age of onset, parental atopic history, filaggrin (FLG) mutations, polysensitization, and the nonrural environment among risk factors for development of multiple atopic comorbidities in young children with AD. Early intervention studies to repair the epidermal barrier or alter exposure to the microbiome or allergens might elucidate the relative roles of barrier defects, genetic locus alterations, and environmental exposures in the risk and sequence of occurrence of TH2 activation disorders.
Article
Background: The allergic march describes the natural history of allergic conditions as they develop during childhood. Eosinophilic esophagitis (EoE) is a chronic allergic inflammatory disease that can be triggered by specific foods. Despite its allergic pathophysiology, the epidemiologic relationship between EoE and established members of the allergic march is unknown. Objective: We sought to determine whether EoE meets epidemiologic criteria for being considered a member of the allergic march. Methods: Using a primary care birth cohort of 130,435 children, we determined the natural histories of atopic dermatitis (AD), IgE-mediated food allergy (IgE-FA), asthma, EoE, and allergic rhinitis (AR) in individual patients. We then performed case-control analyses to establish the extent that existing allergic conditions influence the rate of subsequent EoE diagnosis. Results: A total of 139 children developed EoE during the observation period (prevalence of 0.11%). The peak age of EoE diagnosis was 2.6 years, as compared with 0.3 years, 1 year, 1.1 years, and 2.1 years for AD, IgE-FA, asthma, and AR, respectively. The presence of AD (hazard ratio [HR] 3.2, 95% confidence interval [CI] 2.2-4.6), IgE-FA (HR 9.1, 95% CI 6.5-12.6), and asthma (HR 1.9, 95% CI 1.3-2.7) was independently and cumulatively associated with subsequent EoE diagnosis. The presence of AR was associated with subsequent EoE diagnosis (HR 2.8, 95% CI 2.0-3.9), and the presence of EoE was associated with subsequent AR diagnosis (HR 2.5, 95% CI 1.7-3.5). Conclusions: Allergic comorbidities are positively associated with EoE diagnosis. Together, our findings suggest that EoE is a late manifestation of the allergic march.
Article
Background: DNA methylation profiles associated with childhood asthma might provide novel insights into disease pathogenesis. We did an epigenome-wide association study to assess methylation profiles associated with childhood asthma. Methods: We did a large-scale epigenome-wide association study (EWAS) within the Mechanisms of the Development of ALLergy (MeDALL) project. We examined epigenome-wide methylation using Illumina Infinium Human Methylation450 BeadChips (450K) in whole blood in 207 children with asthma and 610 controls at age 4-5 years, and 185 children with asthma and 546 controls at age 8 years using a cross-sectional case-control design. After identification of differentially methylated CpG sites in the discovery analysis, we did a validation study in children (4-16 years; 247 cases and 2949 controls) from six additional European cohorts and meta-analysed the results. We next investigated whether replicated CpG sites in cord blood predict later asthma in 1316 children. We subsequently investigated cell-type-specific methylation of the identified CpG sites in eosinophils and respiratory epithelial cells and their related gene-expression signatures. We studied cell-type specificity of the asthma association of the replicated CpG sites in 455 respiratory epithelial cell samples, collected by nasal brushing of 16-year-old children as well as in DNA isolated from blood eosinophils (16 with asthma, eight controls [age 2-56 years]) and compared this with whole-blood DNA samples of 74 individuals with asthma and 93 controls (age 1-79 years). Whole-blood transcriptional profiles associated with replicated CpG sites were annotated using RNA-seq data of subsets of peripheral blood mononuclear cells sorted by fluorescence-activated cell sorting. Findings: 27 methylated CpG sites were identified in the discovery analysis. 14 of these CpG sites were replicated and passed genome-wide significance (p<1·14 × 10-7) after meta-analysis. Consistently lower methylation levels were observed at all associated loci across childhood from age 4 to 16 years in participants with asthma, but not in cord blood at birth. All 14 CpG sites were significantly associated with asthma in the second replication study using whole-blood DNA, and were strongly associated with asthma in purified eosinophils. Whole-blood transcriptional signatures associated with these CpG sites indicated increased activation of eosinophils, effector and memory CD8 T cells and natural killer cells, and reduced number of naive T cells. Five of the 14 CpG sites were associated with asthma in respiratory epithelial cells, indicating cross-tissue epigenetic effects. Interpretation: Reduced whole-blood DNA methylation at 14 CpG sites acquired after birth was strongly associated with childhood asthma. These CpG sites and their associated transcriptional profiles indicate activation of eosinophils and cytotoxic T cells in childhood asthma. Our findings merit further investigations of the role of epigenetics in a clinical context. Funding: EU and the Seventh Framework Programme (the MeDALL project).