Next Article in Journal
Adolescent Attachment to Parents and Peers and the Use of Instagram: The Mediation Role of Psychopathological Risk
Previous Article in Journal
Analysis of China’s Industrial Green Development Efficiency and Driving Factors: Research Based on MGWR
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Prenatal and Perinatal Environmental Influences Shaping the Neonatal Immune System: A Focus on Asthma and Allergy Origins

by
Azahara María García-Serna
1,2,
Elena Martín-Orozco
1,2,3,
Trinidad Hernández-Caselles
1,2,3 and
Eva Morales
1,4,*
1
Biomedical Research Institute of Murcia (IMIB-Arrixaca), 30120 Murcia, Spain
2
Department of Biochemistry and Molecular Biology B and Immunology, Faculty of Medicine, University of Murcia, 30100 Murcia, Spain
3
Network of Asthma and Adverse and Allergic Reactions (ARADyAL), 28029 Madrid, Spain
4
Department of Public Health Sciences, University of Murcia, 30100 Murcia, Spain
*
Author to whom correspondence should be addressed.
Int. J. Environ. Res. Public Health 2021, 18(8), 3962; https://doi.org/10.3390/ijerph18083962
Submission received: 9 February 2021 / Revised: 5 April 2021 / Accepted: 7 April 2021 / Published: 9 April 2021

Abstract

:
It is suggested that programming of the immune system starts before birth and is shaped by environmental influences acting during critical windows of susceptibility for human development. Prenatal and perinatal exposure to physiological, biological, physical, or chemical factors can trigger permanent, irreversible changes to the developing immune system, which may be reflected in cord blood of neonates. The aim of this narrative review is to summarize the evidence on the role of the prenatal and perinatal environment, including season of birth, mode of delivery, exposure to common allergens, a farming environment, pet ownership, and exposure to tobacco smoking and pollutants, in shaping the immune cell populations and cytokines at birth in humans. We also discuss how reported disruptions in the immune system at birth might contribute to the development of asthma and related allergic manifestations later in life.

1. Introduction

Non-communicable diseases (NCDs) are predominantly chronic diseases that include metabolic and cardiovascular diseases, cancer, autoimmune conditions, neurological disorders, chronic lung disease, asthma and other allergic diseases. Typically, these diseases share common features: early-life exposure to environmental agents, chronic low-grade inflammation, and immune disturbance during development. Growing evidence is showing that environmentally induced disruption of normal immune system development may play a significant role in the current global epidemic of NCDs whose prevalence has dramatically increased worldwide in the last decades [1]. Most of the variability in the immune system is due to environmental factors, rather than genes [2]. Moreover, the enhanced vulnerability of the developing immune system for environmental insults is based on unique immune maturational events that occur during critical windows in early life, which includes the fetus and the first years of life. To this regard, the fetal immune system seems to be shaped and programmed markedly by the in utero and the perinatal environment, which can have related adverse health consequences later in life.
In this review, we summarize the current evidence on the role of the prenatal and perinatal environment in shaping the human immune system at birth, including the influences of physiological, biological, physical, and chemical factors. In particular, we focus on neonatal immune cell populations and cytokines and discuss how reported disruptions at birth might contribute to the development of asthma and related allergic manifestations later in life.

2. Immune System Development and Maturation

The immune system is a complex network of cells, proteins, tissues and organs that defends the host against microbes and molecules that are recognized as foreign, and ultimately protects against disease. In humans, there are two principal subsystems: the innate and the adaptive or acquired immune system, each of which comprises cellular and humoral components to perform their functions.
Leukocytes, the cellular component of the immune system, are divided into myeloid and lymphoid cells. Myeloid leukocytes are the main cellular components of the innate system, which includes granulocyte cells (neutrophils, eosinophils, basophils), monocytes, macrophages, mast cells, and dendritic cells (DC) [3,4]. Innate cells sense, capture and process antigens while they inform nearby cells by presenting antigens to T lymphocytes and secreting cytokines, small proteins involved in signaling. Macrophages are the major cytokine producers, including pro-inflammatory cytokines such as interleukin (IL)-1β, IL-6, IL-12, IL-18 and tumor necrosis factor alpha (TNF-α) [3]. Lymphoid progenitors originate lymphocytes B and T, the main actors in adaptive immune system, and natural killer (NK) cells that are involved in both the innate and the adaptive response. Adaptative immune response produces immunological memory through antibodies released by B cells. T cells can differentiate into T-helper (Th), T-cytotoxic (Tc) or T-regulatory (Treg). Tc and NK cells attack invaders through cytotoxicity; while Th cells promote and control the development of the immune response. Th cells that recognize an antigen can differentiate into different Th types, including Th1, Th2, and Th17 [5]. During fetal development, Th1 response is thought to be inhibited in the maternal-fetal interface and immune response typically switch into a Th2 response to avoid fetal rejection [6,7]. Th2 cells, which are involved in defense against helminths and other extracellular parasites, mainly produce IL-4, IL-5, IL-9 and IL-13, activate mast cells and eosinophils, and stimulate production of IgE [8]. At birth, the bias towards a Th1 response starts in the neonatal system. Th1 cells mainly produce interferon gamma (IFN-γ), activate macrophages (M1 classical activation), NK and Tc cells, and stimulate the production of IgG. Th1-related cells defend the host against intracellular pathogens, including bacteria, parasites, yeast, and viruses [9]. Prenatal and postnatal early life environmental factors can induce an unbalanced Th1/Th2 response that could be responsible for several diseases later in life [6,10]. Thus, a predominant Th1 response is associated with autoimmune diseases [9], while a predominant Th2 response is associated with atopic disease and allergies [8].
Five major maturational periods through immune system development are described as critical windows of vulnerability [11]. Firstly, hematopoietic stem cells (HSCs) first emerge in the aorta–gonad–mesonephros (AGM) region at around 5 weeks of gestational age [4] and, later on, initiation of hematopoiesis occurs in mid first trimester when pluripotent stems cells appear in the yolk sac and in the fetal liver progressively, and myeloid and lymphoid precursors colonize the thymus and the fetal liver [12]. Secondly, between late first trimester and early second trimester of gestation, stem cells migrate and progenitors expand to new and peripheral tissues [12]; lymphocytes precursors seed the thymus where maturation of T cells is rendered [10]. Thirdly, hematopoietic stem cells colonize the bone marrow and the thymus between gestational weeks 15–16 until birth. During the third trimester of gestation, immune cell numbers increase, and DCs initiate their maturation towards a Th1 response that continues during first months of life. Finally, immune competence maturation during the first year of life and the establishment of immunological memory between 1 and 18 years of age constitute the two extrauterine critical windows of immune system development [12].

3. Prenatal Environmental Exposures and Susceptibility to Asthma and Allergies

A complex interplay between environmental exposures acting during critical windows of development in early life and genetic susceptibility likely contribute to occurrence of asthma and allergy [13,14]. Identified environmental exposures include physiological, biological, physical, or chemical factors.
Season of birth may influence the occurrence of asthma and allergic manifestations later in life. Children born in autumn have a higher risk of asthma compared with those born in spring [15,16]. Moreover, children born in autumn or winter have a higher risk of food allergy [17,18,19], allergic rhinitis [20] and atopic dermatitis [21] compared to those born in spring or/and summer.
Mode of delivery has also been identified as an important factor in the occurrence of asthma and allergy. Cesarean section, whose rate has increased in parallel with the prevalence of childhood asthma over the past decades, is related to increased risk of wheeze up to school age [22,23], asthma in childhood [24,25,26,27], and bronchial hyperreactivity in early adulthood [28]. Moreover, cesarean delivery may increase risk of food allergy and atopic dermatitis [29,30,31].
Prenatal and postnatal early life exposure to common allergens [32], mold or/and dampness [33,34], and furry pets’ ownership [32,35,36] are involved in the development of asthma and allergy. Exposure to mold and dampness increases risk of allergic rhinitis, wheezing and atopic eczema in childhood [33,37,38,39]; while a farming environment and ownership of pets could reduce risk of allergic diseases and atopy [40,41,42].
Maternal smoking during gestation is associated with increased risk in the offspring of respiratory infections [43], wheezing [44,45,46], asthma [47,48], and impaired lung function in childhood and later in life [49,50]. Secondhand tobacco smoke during pregnancy could also influence risk of asthma [51]. Indoor environmental pollutants could contribute to development of atopy; performing housing renovation and painting during pregnancy could be implicated in development of asthma [34,38] and atopic eczema [38,52,53] through emissions of volatile organic compounds (VOCs) or dust [54,55].
Prenatal exposure to persistent organic pollutants (POPs) such as organochlorine pesticides has been associated with increased risk of wheeze and asthma later in life [56,57,58]. Furthermore, non-persistent organic pollutants have been associated with increased risk of respiratory infections in infancy [59], impaired lung function performance in children [60], and the occurrence of asthma [61,62] and allergies [63,64]. In addition, some studies have shown a higher risk of atopic diseases [65,66] and impaired lung function [67] in childhood in relation to in utero exposure to toxic metals. Finally, growing evidence has shown prenatal exposure to air pollutants, primarily NO2 and particulate-matter (PM2.5 and PM10), to be associated with the occurrence of wheezing [68], asthma [32,69,70,71], rhinitis [38,68], eczema [38], and impaired lung function [72] during childhood.

4. Prenatal and Perinatal Environmental Influences and Immune System at Birth: Review of the Literature

4.1. Search Strategy and Study Selection

A literature search was conducted by two independent reviewers (AMG-S and EM) in MEDLINE (via PubMed) through September 2020. The search strategy used the following keywords: outcome (“leukocytes” OR “lymphocytes” OR “immune system” OR “Th1” OR “Th2” OR “cytokines” OR “IgE”) combined with “cord blood” and with the next keywords for exposures (“season of birth” OR “mode of delivery” OR “cesarean section” OR “vaginal delivery” OR “farming environment” OR “farming” OR “pets” OR “dog” OR “cat” OR “indoor allergens” OR “dust mite” OR “mold” OR “dampness” OR “tobacco smoking” OR “persistent organic pollutants” OR “organochlorine pesticides” OR “volatile organic compounds” OR “metals” OR “arsenic” OR “cadmium” OR “lead” OR “chromium” OR “mercury “ OR “air pollution” OR “nitrogen dioxide” OR “particulate matter”)”. Limits: Human, English. Identification and first screening of the articles were performed using the information available in the title and the abstract. Potentially relevant studies were retrieved in full text and assessed for eligibility; any discrepancies were resolved by discussion between the two independent researchers or by discussion with a third review author.
The selection criteria were: (a) article written in English; (b) original research article based on an epidemiologic study performed in human individuals (abstracts, case reports, comments, and lab-based studies were excluded); (c) outcome assessment included phenotyping of immune system cells (leukocytes, lymphocytes and cellular subsets) or cytokine profile patterns assessed in cord blood of newborns; and d) assessment of season at birth, mode of delivery, farming exposures, pets, common allergens, tobacco smoking, persistent and non-persistent pollutants, toxic metals and outdoor air pollution as exposures during pregnancy or around birth. After screening of retrieved articles, 78 articles met our inclusion criteria, including 49 cohort studies, 28 cross-sectional studies and 1 retrospective study conducted between 1979 and 2020.

4.2. Season of Birth

Immune cell distributions [73,74] and cytokines [75,76,77,78,79] in cord blood of neonates have been investigated in relation to season of birth (Table 1). Collinson et al. [73] found higher leukocyte and lymphocyte counts at birth in children born in wet season compared to those born in dry seasons, but no differences were found for lymphocyte subsets. Moreover, compared to neonates born in summer those born in winter also showed higher leukocyte counts, including granulocytes, neutrophils and monocytes, higher number of plasmacytoid dendritic cells and activated Th and NK cells [74]. Tc cell counts increased in neonates born in spring compared with those born in autumn [74]. No associations were found between season of birth and cord blood distributions of B and T cells, nor lymphocytes subsets including Th and Treg cells [74].
Some studies have reported that neonates born during spring or summer time had significantly lower production and response of cytokines, including TNF-α, IFN-γ, IL-5 and IL-10 [75,79]. However, other studies have reported highest induced responses of IL-4, IL-5, IL-13 among neonates born during summer [75,77]. Moreover, neonates born in autumn or winter showed increased cockroach-induced IL-5 [76] and pro-inflammatory cytokine concentrations and response to specific stimuli (IFN-γ, IFN- α, IL-8, TNF-α and IL-12p40) [78].

4.3. Mode of Delivery

An increased number of leukocytes in cord blood of neonates is associated with vaginal delivery [80,81,82,83,84,85,86] and longer duration of labor [79] (Table 2). Accordingly, cesarean section is associated with decreased counts of leukocytes [79,87,88,89]. Moreover vaginal delivery is related to increased neutrophils [80,82,83,84,86,90] and monocytes [80,86,91], but decreased lymphocytes at birth [83,84]. In addition, cord blood monocytes of neonates born by cesarean section express lower level of the surface innate antigen receptors toll like receptor 2 (TLR2) and TLR4 [87]. Furthermore, vaginal delivery is associated with increased NK cells [80,84,92,93], a higher expression of CD16 and CD56 surface receptors [92], increased Treg cells [94], and a decreased expression of specific Treg transcription factor FoxP3 [95]. Most of the studies have showed no association between mode of delivery and distributions of B and T cytotoxic cells [80,91,92], or memory and naïve T lymphocytes [91,96] in cord blood. Results on T and Th cells are inconsistent. Although some studies did not find an association between mode of delivery and the distribution of T and Th [80,91] cells; others have reported decreased proportions of T [84,92] and Th cells [92] in neonates born by vaginal delivery.
In addition, mode of delivery may alter cytokine patterns at birth. Compared to those delivered by cesarean section neonates born by vaginal delivery had an enhanced innate response, showing higher IL-1β, IL-6 and IL-8 levels but lower granulocyte colony-stimulating factor (G-CSF) levels [23,86,97,98,99,100]. Results on relationship between TNF-α and mode of delivery are contradictory [98,100]. Moreover, vaginal delivery may induce a Th1-related response (IFN-γ and IL-12) at birth [79,84,86,98,101]. On the other hand, cesarean section was associated with an increased IL-13 response to allergens, PHA and LPS [102], and a decreased Granulocyte-macrophage colony-stimulating factor (GM-CSF) concentrations [89], TLR1-induced pro-inflammatory response (i.e., TNF-α and IL-6) [23], and Th2-related response [78,79] at birth. In addition, IL-12p40 showed decreased response after CpG-stimulation but increased response after viral protein induction [78] in cord blood cells of neonates delivered by cesarean section.

4.4. Prenatal Exposure to a Farming Environment, Pets, and Indoor Allergens

Impact of farming-related exposures, pets and indoor allergens on the immune system at birth has been investigated in diverse studies (Table 3). Decreased number of leukocytes [79], increased number and function of Treg cells [103], and increased B-cell activating factor [104] have been found in neonates of farming parents.
Several studies have investigated cord blood immunoglobulin E (IgE), a key feature in allergic manifestations, in relation to prenatal exposure to common allergens. Total IgE levels decreased in cord blood from newborns whose mothers had been exposed to dogs [105,106] and endotoxins [107] during pregnancy, while increased total IgE levels have been found in cord blood of newborns exposed to cats [106,107] and dust mite [107,108]. Furthermore, specific IgE for seasonal allergens decreased in cord blood of newborns whose mothers were exposed to a farm environment during pregnancy, whereas specific IgE for cow’s milk and food allergens increased in cord blood of newborns whose mothers consumed boiled farm milk during pregnancy [109].
Prenatal farm environment is related to increased proinflammatory cytokines (TFN-α and IL-6) in cord blood of neonates [103,110]. Moreover, ownership of dogs during pregnancy has been associated with decreased TNF-α [111] and ownership of dogs or cats has been related to an increased IL-6 response [112]. Th1-related response through IFN-γ production was increased in newborns exposed to farm animals and among those whose mothers consumed unskimmed farm milk or butter made of farm milk during pregnancy [110]. Although, no associations between prenatal exposure to pets (cats and dogs) and IFN-γ in cord blood of neonates has been reported [113]. Prenatal contact with higher number of animal species was associated with increased dust mite allergen-induced IFN-γ response [110]. However, other studies have reported a decreased IFN-γ response in cord blood of newborns of farmer fathers [75], as well as a decreased LPS-induced IFN-γ response in relation to prenatal exposure to high endotoxin levels [114], and a lower proportions of induced Th lymphocytes producing IFN-γ among newborns prenatally exposed to high house dust mite (Der p 1) [115].
In addition, farm environments could reduce a Th2-related response in newborns. Prenatal exposure to stables was related to a decreased dust mite allergen-induced IL-5 response [103]; and newborns whose fathers were farmers showed a decreased IL-5 and IL-10 induced-response [79]. Moreover, neonates whose mothers consumed yogurt made of farm milk during pregnancy showed decreased IL-5 and IL-10 induced-response in cord blood [110]. However, no associations between prenatal exposure to pets (cats or dogs) and IL-13 were found in cord blood of neonates [113].

4.5. Tobacco Smoking

Exposure to maternal tobacco smoking during pregnancy has been associated with a reduction in neutrophils [116,117,118], monocytes [118], lymphocytes [118], and lymphocyte subpopulations including T helper CD4+ cells [119] and Treg cells [120] in cord blood (Table 4). Moreover, some studies have reported lower Th1 related cytokine responses in cord blood of neonates of mother who smoked during pregnancy, including TNF-α [121] and IFN-γ [122]. Neonatal Th2 related cytokine responses to allergen and different stimuli are inconsistent across studies. Reduced responses of Th2-related cytokines (i.e., IL-5 and IL-10) have been found [79,110,121]; however, increased IL-13 response has been reported as well [123]. In addition, reduced levels and induced response to TLR2 and TLR9 pathways of IL-6 have been found in cord blood of neonates whose mothers smoked during pregnancy [121,124].

4.6. Persistent and Non-persistent Organic Pollutants

Diverse studies have investigated the effects of organic pollutants on prenatal immune system development [125,126,127,128,129,130,131,132,133,134,135] (Table 5). Neonates born in areas contaminated with high levels of POPs (DDE, HCB and PCBs) showed decreased percentages of Th lymphocytes [125], dendritic cells, resting T cells, suppressor inducer T cells and NK cells [129,130], while increased proportions of B and activated B cells, Tc lymphocytes [130], and memory T cells [129]. To date, no study has examined the association between prenatal exposure to non-persistent organic pollutants and leukocyte and lymphocyte distributions in newborns.
Furthermore, prenatal exposure to organic pollutants has been associated with impaired immunoglobulin contents in cord blood plasma of neonates, including decreased IgM and increased IgG levels [125]. Furthermore, sex may play an important role in toxicity since male newborns prenatally exposed to dioxin-like compounds showed decreased IgE concentrations in cord blood [135]. Moreover, a positive correlation between IgE levels and perfluoroalkyl (PFO) analytes PFOA and PFOS in cord serum has been found among males [132].
Several studies have associated exposure to organic compounds during pregnancy with changes in cord blood cytokines patterns [136,137,138,139]. Pro-inflammatory cytokines, such as IL-1β, IL-6, TNF-α and IL-33/TSLP were found decreased in cord blood of newborns exposed to PCBs, DDE, phthalates and organochlorines [126,133,134], BPA [133,137,138] and VOCs [139], whereas newborns exposed to permethrin showed higher IL-1β levels than those less exposed [131]. Nevertheless, maternal urine phthalates concentrations during first trimester were associated with decreased chemotactic cytokines in cord blood [136]. Studies researching Th1 and Th2 cytokines are scarce [128], but results suggest that prenatal exposure to organic compound impacts Th2/Th1 ratios; increased IL-13 levels in cord blood were associated with increased POPs concentrations in placenta [128] and newborns exposed to VOCs during pregnancy showed increased IL-4 concentrations [139]. Finally, effects of prenatal exposure to VOCs on Th1-related cytokines are unclear [139].

4.7. Toxic Metals

Diverse epidemiological studies have examined the effects of prenatal exposure to toxic metals on immune system biomarkers in cord blood of neonates [136,140,141,142,143] (Table 6). Changes in cord blood lymphocytes include a decreased number of activated T naïve cells, decreased percentages of Th, Th memory and activated cells, and increased number of Th2-related T cells in newborns exposed to mercury [125], arsenic [141,142] and cadmium [142]. Moreover, prenatal exposure to arsenic has been associated with an increased T cell proliferation and a decreased Treg suppressor function [141]. Furthermore, Belles-Isles et al. found negative correlations between mercury and cord blood IgM levels but positive correlations between lead and IgG cord blood levels [125].
Effects of prenatal exposure to metals on cytokines patters at birth have also been investigated [134,136,138,140]. Prenatal exposure to metals could impair effector Th response and enhance pro-inflammatory and Th2-related response in newborns. Cord blood levels of pro-inflammatory cytokines (TNFα, IL-8, IL-1β and IFN-γ) showed U-shaped associations in relation to maternal exposure to arsenic in late pregnancy [140]. IL-33/TSLP showed an inverse relationship with maternal exposure to lead in early pregnancy [134], and concentrations of chemotactic cytokines increased in newborns prenatally exposed to several metals [136]. In addition, in utero exposure to lead and chromium has been associated with higher IL-13 levels in cord blood [143].

4.8. Outdoor air Pollutants

Studies examining the relationship between prenatal exposure to outdoor air pollutants and distributions of immune cells in cord blood of neonates [88,144,145,146,147,148] are shown in Table 7. Exposure to higher NO2 concentrations 14 days before delivery have been associated with decreased counts of leukocytes, neutrophils and monocytes [88]. Moreover, higher concentrations of NO2 derived from traffic during the first trimester of pregnancy were associated with decreased counts of leukocytes, lymphocytes, monocytes and basophils [148]. In addition, increased percentage of B cells has been reported in relation to higher levels of outdoor air pollutants during late pregnancy including PAHs and PM2.5 [145,146].
Associations between prenatal exposure to outdoor air pollutants and the distribution of NK cells in cord blood are inconsistent. Some studies have showed reduced NK cells in cord blood associated with exposure to PAHs and PM2.5 during early gestation (first trimester) [146], and to NO2 and PM2.5 exposure in early and late gestation [148]. However, other studies have reported increased NK cells in cord blood of neonates in relation to living in a high PM polluted area [144], exposure to PM10 during the first trimester of pregnancy [147], and exposure in late pregnancy to PAHs, PM2.5, NO2 and O3 [146,147,148].
As for T lymphocytes, results are also inconsistent. Reduced T cells have been found in newborns living in a high PM polluted area [144], among those exposed to higher PAH and PM2.5 in late pregnancy [145,146]. Moreover, several studies have reported a decrease in Th cells in relation to prenatal exposure to outdoor air pollutants, including traffic-related NO2 in early pregnancy [148], PAHs and PM2.5 in late pregnancy [145,146], and PM10 during whole pregnancy [147]. Distribution of Th cell subpopulations in cord blood in relation to prenatal exposure to outdoor air pollutants has been poorly investigated [148]. Garcia-Serna et al. reported increased Th cells in relation to high PM concentration during early pregnancy, increased Th1-related cells in neonates exposed to high traffic-related PM levels during early and late pregnancy, and decreased Th1Th2-related cells in relation to higher levels of PM during pregnancy. Moreover, Treg cells were decreased in relation to high concentrations of traffic-related PM10 and NO2 15-days before delivery [148].
Most studies have reported a reduction in Tc cells in newborns associated with prenatal exposure to higher levels of outdoor air pollutants, including short-term exposure in later pregnancy to PAHs and PM2.5 [145,146], and long-term exposure to PM2.5 derived from traffic [148]. However, Baïz et al. found increased Tc in newborns exposed to high NO2 concentrations during the first and second trimester of pregnancy [147].
The influence of prenatal exposure to air pollutants on cytokines in cord blood of neonates has been poorly examined [124,149]. Latzin et al. reported increased levels of IL-1β in cord blood serum in relation to short-term exposure to higher PM10 concentrations before birth, and decreased levels of IL-10 in relation to higher PM10 concentrations in last trimester of pregnancy [124]. In addition, Ashley-Martin et al. found maternal exposure to higher NO2 levels to be associated with increased IL-33 and thymic stromal lymphopoietin (TSLP) concentrations in cord blood of neonates, and increased IgE levels among female neonates exposed to higher PM2.5 levels during pregnancy [149].

5. Discussion

Season of birth, mode of delivery, prenatal exposure to common allergens and chemicals, including tobacco smoking, organic pollutants, metals and outdoor air pollutants, may impair distributions of immune system cells (i.e., leukocytes and lymphocytes), as well as, alter immunoglobulins and cytokine patterns in cord blood of neonates.
Winter birth is associated with increased leukocytes, NK and activated Th cells [74] and a predominant type 1- and type 17-associated immune response with increased concentrations of pro-inflammatory cytokines [78]; whereas spring or summer birth related to a predominant Th2-related cytokine response [75,77]. Findings suggest activation of antimicrobial pathways that might reflect increased maternal exposure to infectious microbes during or before birth, which may serve as a trigger of neonatal immunity during fall, winter, and spring that might prime the immune system in early life. In addition to infections, other season-fluctuating conditions could play a role, including maternal nutrition [150,151], vitamin D status [152], and exposure to common allergens [153,154] and air pollutants [155].
Cord blood of neonates delivered vaginally shows increased leukocytes and some of their subsets (neutrophils, monocytes, NK and Treg cells) [80,81,82,83,84,85,86,90,92,93,94], as well as increased pro-inflammatory cytokines (IL-1β, IL-6 and IL-8) [84,86,97,98,99,100]. Whereas cesarean section is associated with decreased cord blood leukocytes, lymphocytes, and dendritic cells [79,87,88]. Moreover, vaginal delivery seems to be related to a Th1-related response in the newborn characterized by a higher IFN-γ and IL-2 production in response to stimuli and a higher IL-12 response to microbial exposure [84,98,101]. Risk of immune diseases, including asthma, and severity of respiratory infections [25,27,156,157] due to cesarean section likely results from lack of labor-associated stress factors and immune-dysregulation caused by a disturbed gut microbiota immune programming in the offspring at birth [158]. In this sense, redistribution and activation of immune cells under stress of labor may be an adaptive response to enhance defensive mechanisms of the immune system. Furthermore, neonates born by cesarean section are exposed to skin flora instead of birth canal microbes, which alters diversity of the microbes that colonize the gut and thus the activation of the immune response [102,159].
A prenatal farming environment is related to lower IgE response to seasonal allergens [109], a higher pro-inflammatory cytokine (IL-6 and TNF-α) [103,110] and Th1-related (IFN-γ) responses [103,110], but decreased Th2-related (IL-5) [79,103] and Treg-related (IL-10) [79,110] responses. Moreover, prenatal ownership of cats is associated with a higher IgE response to allergens (pollen and food) [106,107] and ownership of dogs is associated with a lower IgE response to allergens (pollen) [105,106]. A farm environment, and particularly the exposure to elevated number of animal species, resulting in farm-associated microbial exposures has been proposed to have immunoregulatory properties on the developing innate immune system [160]. According to this model, several key effector mechanisms of allergic inflammation are inhibited by the immunoregulatory properties of farm-associated microbial exposures.
Maternal smoking during pregnancy impacts both innate and adaptive immunity of neonates, which could attenuate or exacerbate pathogenic immune responses against infections in early life. Reduced number of leukocytes, lymphocytes, including T helper CD4+, and detrimental IFN-γ response may impair infant defense system against respiratory viral infections in early life, which likely contributes to increased incidence and severity of these infections [160], and thereby to higher asthma susceptibility in childhood [43,47,48]. Moreover, an imbalance of Treg cells is associated with airway inflammation in asthma and allergic rhinitis [161].
Although more evidence is warranted, current evidence suggests that prenatal exposure to POPs might contribute to a failed innate immune responses (i.e., decreased dendritic, T and NK cells), and induce lymphocyte activation (increased B cells and IgG production) and a Th2-related response in neonates [125,129,130]. Moreover, prenatal exposure to both persistent and non-persistent organic pollutants is related to a decreased pro-inflammatory cytokine response (TNF-α, IL-6 and IL-33/TSLP) in neonates [126,133,134,137,138]. Overall, reported disturbances in the immune system of neonates could predispose to a higher susceptibility to respiratory infections during the early postnatal period and thus to increased risk of wheeze and asthma in childhood [56,57,58,59].
Prenatal exposure to metals may impair the innate immune response at birth through changes in T, Treg and Th memory cells [125,141,142], which could contribute to a higher risk of respiratory infections in early life [162,163,164]. In addition, prenatal exposure to metals seem to increase a Th2-related cytokine response in neonates [140,143], which might contribute to development of atopic manifestations later in life [66].
In utero exposure to outdoor air pollutants impairs leukocyte and lymphocyte distributions in neonates; early and late gestation seem to be potential developmental windows of higher susceptibility [88,144,145,146,147,148]. Current evidence suggests that prenatal exposure to outdoor air pollution could disbalance Th subsets in neonates reflecting an inflammatory state, which could predispose to allergic immune responses later in life. Reduced Tc cells at birth in relation to in utero exposure to outdoor air pollutants may result into a higher risk of respiratory tract infections in childhood given the role of Tc cells in the defense against virus infections [165]. Limited evidence on cytokine profiles in neonates suggests a higher production of pro-inflammatory cytokines (IL-1β and IL-33) in relation to exposure to higher concentrations of NO2 and PM [124,149].
This review has some limitations. Studies with small sample size were included in the current review. However, our aim was to summarize all the evidence to date on the impact of prenatal and perinatal environmental influences on immune system at birth. Publication bias cannot be discarded, because studies with no significant findings are less likely to be published. Only articles published in English were included. Finally, heterogeneity between studies in exposure assessment, as well as the small number of studies for any given exposure–outcome relationship, currently make the combination of studies for meta-analysis impossible, but this review summarizes the current evidence and may guide future studies.

6. Conclusions

The prenatal and perinatal periods seem to represent crucial biological windows of opportunity for environmental influences to shape the neonatal immune system. Identified disturbances in immune cell populations and cytokines at birth could lead to a higher susceptibility to respiratory infections, asthma and allergic manifestations later in life. Although some associations and mechanisms warrant further investigation, overall, promoting a proper immune system development during early life should be recognized as a major element in the public health agenda to prevent NCDs, especially asthma and allergic manifestations.

Author Contributions

Conceptualization, A.M.G.-S. and E.M.; writing—original draft preparation, A.M.G.-S. and E.M.; writing—review and editing, A.M.G.-S., E.M.-O., T.H.-C. and E.M. All authors have read and agreed to the published version of the manuscript.

Funding

This review received no external funding. A.M.G.S. was funded by a predoctoral Fellowship (FI17/00086) and E.M. was funded by Miguel Servet Fellowships (MS14/00046 and CPII19/00019) awarded by the Instituto de Salud Carlos III (ISCIII), Spanish Ministry of Science, Innovation and Universities, and Fondos FEDER. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Dietert, R.R. Developmental Immunotoxicity, Perinatal Programming, and Noncommunicable Diseases: Focus on Human Studies. Adv. Med. 2014, 2014, 867805. [Google Scholar] [CrossRef] [Green Version]
  2. Brodin, P.; Jojic, V.; Gao, T.; Bhattacharya, S.; Angel, C.J.; Furman, D.; Shen-Orr, S.; Dekker, C.L.; Swan, G.E.; Butte, A.J.; et al. Variation in the human immune system is largely driven by non-heritable influences. Cell 2015, 160, 37–47. [Google Scholar] [CrossRef] [Green Version]
  3. Wynn, T.A.; Chawla, A.; Pollard, J.W. Macrophage biology in development, homeostasis and disease. Nature 2013, 496, 445–455. [Google Scholar] [CrossRef]
  4. Zhang, X.; Zhivaki, D.; Lo-Man, R. Unique aspects of the perinatal immune system. Nat. Rev. Immunol. 2017, 17, 495–507. [Google Scholar] [CrossRef]
  5. Rautajoki, K.J.; Kylaniemi, M.K.; Raghav, S.K.; Rao, K.; Lahesmaa, R. An insight into molecular mechanisms of human T helper cell differentiation. Ann. Med. 2008, 40, 322–335. [Google Scholar] [CrossRef]
  6. D’Elios, M.; Del Prete, G. Th1/Th2 balance in human disease. Transpl. Proc. 1998, 30, 2373–2377. [Google Scholar] [CrossRef]
  7. Wilczyński, J.R. Th1/Th2 cytokines balance--yin and yang of reproductive immunology. Eur. J. Obstet. Gynecol. Reprod. Biol. 2005, 122, 136–143. [Google Scholar] [CrossRef] [PubMed]
  8. Licona-Limón, P.; Kim, L.K.; Palm, N.W.; Flavell, R.A. TH2, allergy and group 2 innate lymphoid cells. Nat. Immunol. 2013, 14, 536–542. [Google Scholar] [CrossRef]
  9. Kamali, A.N.; Noorbakhsh, S.M.; Hamedifar, H.; Jadidi-Niaragh, F.; Yazdani, R.; Bautista, J.M.; Azizi, G. A role for Th1-like Th17 cells in the pathogenesis of inflammatory and autoimmune disorders. Mol. Immunol. 2019, 105, 107–115. [Google Scholar] [CrossRef]
  10. Dietert, R.R. Developmental immunotoxicology (DIT): Windows of vulnerability, immune dysfunction and safety assessment. J. Immunotoxicol. 2008, 5, 401–412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Dietert, R.R.; Etzel, R.A.; Chen, D.; Halonen, M.; Holladay, S.D.; Jarabek, A.M.; Landreth, K.; Peden, D.B.; Pinkerton, K.; Smialowicz, R.J.; et al. Workshop to identify critical windows of exposure for children’s health: Immune and respiratory systems work group summary. Environ. Health Perspect. 2000, 108 (Suppl. 3), 483–490. [Google Scholar]
  12. Kuper, C.F.; van Bilsen, J.; Cnossen, H.; Houben, G.; Garthoff, J.; Wolterbeek, A. Development of immune organs and functioning in humans and test animals: Implications for immune intervention studies. Reprod. Toxicol. 2016, 64, 180–190. [Google Scholar] [CrossRef]
  13. Kahr, N.; Naeser, V.; Stensballe, L.G.; Kyvik, K.O.; Skytthe, A.; Backer, V.; Bønnelykke, K.; Thomsen, S.F. Gene-environment interaction in atopic diseases: A population-based twin study of early-life exposures. Clin. Respir. J. 2015, 9, 79–86. [Google Scholar] [CrossRef]
  14. Morales, E.; Duffy, D. Genetics and Gene-Environment Interactions in Childhood and Adult Onset Asthma. Front. Pediatr. 2019, 7, 499. [Google Scholar] [CrossRef] [Green Version]
  15. Knudsen, T.B.; Thomsen, S.F.; Ulrik, C.S.; Fenger, M.; Nepper-Christensen, S.; Backer, V. Season of birth and risk of atopic disease among children and adolescents. J. Asthma. 2007, 44, 257–260. [Google Scholar] [CrossRef] [PubMed]
  16. Chang, W.C.; Yang, K.D.; Wu, M.T.; Wen, Y.F.; Hsi, E.; Chang, J.C.; Lin, Y.M.; Kuo, H.C.; Chang, W.P. Close correlation between season of birth and the prevalence of bronchial asthma in a Taiwanese population. PLoS ONE 2013, 8, e80285. [Google Scholar] [CrossRef]
  17. Vassallo, M.F.; Banerji, A.; Rudders, S.A.; Clark, S.; Mullins, R.J.; Camargo, C.A., Jr. Season of birth and food allergy in children. Ann. Allergy Asthma Immunol. 2010, 104, 307–313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Keet, C.A.; Matsui, E.C.; Savage, J.H.; Neuman-Sunshine, D.L.; Skripak, J.; Peng, R.D.; Wood, R.A. Potential mechanisms for the association between fall birth and food allergy. Allergy 2012, 67, 775–782. [Google Scholar] [CrossRef]
  19. Kusunoki, T.; Morimoto, T.; Sakuma, M.; Mukaida, K.; Yasumi, T.; Nishikomori, R.; Heike, T. Effect of eczema on the association between season of birth and food allergy in Japanese children. Pediatr. Int. 2013, 55, 7–10. [Google Scholar] [CrossRef]
  20. Li, Y.; Rui, X.; Ma, B.; Jiang, F.; Chen, J. Early-Life Environmental Factors, IFN-γ Methylation Patterns, and Childhood Allergic Rhinitis. Int. Arch. Allergy Immunol. 2019, 178, 323–332. [Google Scholar] [CrossRef] [PubMed]
  21. Calov, M.; Alinaghi, F.; Hamann, C.R.; Silverberg, J.; Egeberg, A.; Thyssen, J.P. The Association Between Season of Birth and Atopic Dermatitis in the Northern Hemisphere: A Systematic Review and Meta-Analysis. J Allergy Clin Immunol Pract. 2020, 8, 674–680.e5. [Google Scholar] [CrossRef]
  22. van Berkel, A.C.; den Dekker, H.T.; Jaddoe, V.W.; Reiss, I.K.; Gaillard, R.; Hofman, A.; de Jongste, J.C.; Duijts, L. Mode of delivery and childhood fractional exhaled nitric oxide, interrupter resistance and asthma: The Generation R study. Pediatr. Allergy Immunol. 2015, 26, 330–336. [Google Scholar] [CrossRef] [PubMed]
  23. Liao, S.L.; Tsai, M.H.; Yao, T.C.; Hua, M.C.; Yeh, K.W.; Chiu, C.Y.; Su, K.W.; Huang, S.Y.; Kao, C.C.; Lai, S.H.; et al. Caesarean Section is associated with reduced perinatal cytokine response, increased risk of bacterial colonization in the airway, and infantile wheezing. Sci. Rep. 2017, 7, 9053. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Keag, O.E.; Norman, J.E.; Stock, S.J. Long-term risks and benefits associated with cesarean delivery for mother, baby, and subsequent pregnancies: Systematic review and meta-analysis. PLoS Med. 2018, 15, e1002494. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Rusconi, F.; Zugna, D.; Annesi-Maesano, I.; Baïz, N.; Barros, H.; Correia, S.; Duijts, L.; Forastiere, F.; Inskip, H.; Kelleher, C.C.; et al. Mode of Delivery and Asthma at School Age in 9 European Birth Cohorts. Am. J. Epidemiol. 2017, 185, 465–473. [Google Scholar] [CrossRef] [Green Version]
  26. Kristensen, K.; Henriksen, L. Cesarean section and disease associated with immune function. J. Allergy Clin. Immunol. 2016, 137, 587–590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Darabi, B.; Rahmati, S.; HafeziAhmadi, M.R.; Badfar, G.; Azami, M. The association between caesarean section and childhood asthma: An updated systematic review and meta-analysis. Allergy Asthma Clin. Immunol. 2019, 15, 62. [Google Scholar] [CrossRef] [PubMed]
  28. Gerlich, J.; Benecke, N.; Peters-Weist, A.S.; Heinrich, S.; Roller, D.; Genuneit, J.; Weinmayr, G.; Windstetter, D.; Dressel, H.; Range, U.; et al. Pregnancy and perinatal conditions and atopic disease prevalence in childhood and adulthood. Allergy. 2018, 73, 1064–1074. [Google Scholar] [CrossRef] [PubMed]
  29. Mitselou, N.; Hallberg, J.; Stephansson, O.; Almqvist, C.; Melén, E.; Ludvigsson, J.F. Cesarean delivery, preterm birth, and risk of food allergy: Nationwide Swedish cohort study of more than 1 million children. J. Allergy Clin. Immunol. 2018, 142, 1510–1514.e2. [Google Scholar] [CrossRef] [Green Version]
  30. Yu, M.; Han, K.; Kim, D.H.; Nam, G.E. Atopic dermatitis is associated with Caesarean sections in Korean adolescents, but asthma is not. Acta Paediatr. 2015, 104, 1253–1258. [Google Scholar] [CrossRef]
  31. Papathoma, E.; Triga, M.; Fouzas, S.; Dimitriou, G. Cesarean section delivery and development of food allergy and atopic dermatitis in early childhood. Pediatr. Allergy Immunol. 2016, 27, 419–424. [Google Scholar] [CrossRef]
  32. Gaffin, J.M.; Kanchongkittiphon, W.; Phipatanakul, W. Perinatal and early childhood environmental factors influencing allergic asthma immunopathogenesis. Int. Immunopharmacol. 2014, 22, 21–30. [Google Scholar] [CrossRef] [Green Version]
  33. Norbäck, D.; Lu, C.; Wang, J.; Zhang, Y.; Li, B.; Zhao, Z.; Huang, C.; Zhang, X.; Qian, H.; Sun, Y.; et al. Asthma and rhinitis among Chinese children—Indoor and outdoor air pollution and indicators of socioeconomic status (SES). Environ. Int. 2018, 115, 1–8. [Google Scholar] [CrossRef]
  34. Wen, H.J.; Chiang, T.L.; Lin, S.J.; Guo, Y.L. Predicting risk for childhood asthma by pre-pregnancy, perinatal, and postnatal factors. Pediatr. Allergy Immunol. 2015, 26, 272–279. [Google Scholar] [CrossRef] [PubMed]
  35. Sasaki, M.; Yoshida, K.; Adachi, Y.; Furukawa, M.; Itazawa, T.; Odajima, H.; Saito, H.; Akasawa, A. Factors associated with asthma control in children: Findings from a national Web-based survey. Pediatr. Allergy Immunol. 2014, 25, 804–809. [Google Scholar] [CrossRef] [PubMed]
  36. Dong, G.H.; Ma, Y.N.; Ding, H.L.; Jin, J.; Cao, Y.; Zhao, Y.D.; He, Q.C. Pets keeping in home, parental atopy, asthma, and asthma-related symptoms in 12,910 elementary school children from northeast China. Indoor Air. 2009, 19, 166–173. [Google Scholar] [CrossRef]
  37. Kang, X.; Tu, H.; Tian, T.; Huang, Z.; Luo, L.; Shen, L.; Ye, J. Home environment and diseases in early life are associated with allergic rhinitis. Int. J. Pediatr. Otorhinolaryngol. 2019, 118, 47–52. [Google Scholar] [CrossRef] [PubMed]
  38. Deng, Q.; Lu, C.; Ou, C.; Chen, L.; Yuan, H. Preconceptional, prenatal and postnatal exposure to outdoor and indoor environmental factors on allergic diseases/symptoms in preschool children. Chemosphere 2016, 152, 459–467. [Google Scholar] [CrossRef] [PubMed]
  39. North, M.L.; Brook, J.R.; Lee, E.Y.; Omana, V.; Daniel, N.M.; Steacy, L.M.; Evans, G.J.; Diamond, M.L.; Ellis, A.K. The Kingston Allergy Birth Cohort: Exploring parentally reported respiratory outcomes through the lens of the exposome. Ann. Allergy Asthma Immunol. 2017, 118, 465–473. [Google Scholar] [CrossRef] [PubMed]
  40. Lodge, C.J.; Allen, K.J.; Lowe, A.J.; Hill, D.J.; Hosking, C.S.; Abramson, M.J.; Dharmage, S.C. Perinatal cat and dog exposure and the risk of asthma and allergy in the urban environment: A systematic review of longitudinal studies. Clin. Dev. Immunol. 2012, 2012, 176484. [Google Scholar] [CrossRef]
  41. Roduit, C.; Wohlgensinger, J.; Frei, R.; Bitter, S.; Bieli, C.; Loeliger, S.; Büchele, G.; Riedler, J.; Dalphin, J.C.; Remes, S.; et al. Prenatal animal contact and gene expression of innate immunity receptors at birth are associated with atopic dermatitis. J. Allergy Clin. Immunol. 2011, 127, 179–185.e1. [Google Scholar] [CrossRef]
  42. Thorsteinsdottir, S.; Thyssen, J.P.; Stokholm, J.; Vissing, N.H.; Waage, J.; Bisgaard, H. Domestic dog exposure at birth reduces the incidence of atopic dermatitis. Allergy 2016, 71, 1736–1744. [Google Scholar] [CrossRef]
  43. Lanari, M.; Vandini, S.; Adorni, F.; Prinelli, F.; Di Santo, S.; Silvestri, M.; Musicco, M.; Study Group of Italian Society of Neonatology on Risk Factors for RSV Hospitalization. Prenatal tobacco smoke exposure increases hospitalizations for bronchiolitis in infants. Respir. Res. 2015, 16, 152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Goudarzi, H.; Konno, S.; Kimura, H.; Araki, A.; Miyashita, C.; Itoh, S.; Ait Bamai, Y.; Kimura, H.; Shimizu, K.; Suzuki, M.; et al. Contrasting associations of maternal smoking and pre-pregnancy BMI with wheeze and eczema in children. Sci. Total Environ. 2018, 639, 1601–1609. [Google Scholar] [CrossRef] [PubMed]
  45. De Queiroz Andrade, E.; Da Silva Sena, C.R.; Collison, A.; Murphy, V.E.; Gould, G.S.; Bonevski, B.; Mattes, J. Association between active tobacco use during pregnancy and infant respiratory health: A systematic review and meta-analysis. BMJ Open. 2020, 10, e037819. [Google Scholar] [CrossRef] [PubMed]
  46. Vardavas, C.I.; Hohmann, C.; Patelarou, E.; Martinez, D.; Henderson, A.J.; Granell, R.; Sunyer, J.; Torrent, M.; Fantini, M.P.; Gori, D.; et al. The independent role of prenatal and postnatal exposure to active and passive smoking on the development of early wheeze in children. Eur. Respir. J. 2016, 48, 115–124. [Google Scholar] [CrossRef] [PubMed]
  47. Cheng, H.; Montgomery, S.; Treglown, L.; Furnham, A. Associations between childhood biomedical factors, maternal smoking, personality traits, Body and Mass Index and the prevalence of asthma in adulthood. Psychol. Health 2018, 33, 1116–1129. [Google Scholar] [CrossRef]
  48. Thacher, J.D.; Gehring, U.; Gruzieva, O.; Standl, M.; Pershagen, G.; Bauer, C.P.; Berdel, D.; Keller, T.; Koletzko, S.; Koppelman, G.H.; et al. Maternal Smoking during Pregnancy and Early Childhood and Development of Asthma and Rhinoconjunctivitis—A MeDALL Project. Environ. Health Perspect. 2018, 126, 047005. [Google Scholar] [CrossRef] [Green Version]
  49. Thacher, J.D.; Schultz, E.S.; Hallberg, J.; Hellberg, U.; Kull, I.; Thunqvist, P.; Pershagen, G.; Gustafsson, P.M.; Melén, E.; Bergström, A. Tobacco smoke exposure in early life and adolescence in relation to lung function. Eur. Respir. J. 2018, 51, 1702111. [Google Scholar] [CrossRef] [PubMed]
  50. Hu, L.W.; Yang, M.; Chen, S.; Shah, K.; Hailegiorgis, Y.; Burgens, R.; Vaughn, M.; Huang, J.; Xaverius, P.; Paul, G.; et al. Effects of in utero and Postnatal Exposure to Secondhand Smoke on Lung Function by Gender and Asthma Status: The Seven Northeastern Cities (SNEC) Study. Respiration 2017, 93, 189–197. [Google Scholar] [CrossRef]
  51. Harju, M.; Keski-Nisula, L.; Georgiadis, L.; Heinonen, S. Parental smoking and cessation during pregnancy and the risk of childhood asthma. BMC Public Health 2016, 16, 428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Cai, J.; Liu, W.; Hu, Y.; Zou, Z.; Shen, L.; Huang, C. Household environment, lifestyle behaviors, and dietary habits in relation to childhood atopic eczema in Shanghai, China. Int. Arch. Occup. Environ. Health 2017, 90, 141–159. [Google Scholar] [CrossRef]
  53. Xu, F.; Yan, S.; Zheng, Q.; Li, F.; Chai, W.; Wu, M.; Kan, H.; Norback, D.; Xu, J.; Zhao, Z. Residential Risk Factors for Atopic Dermatitis in 3- to 6-Year Old Children: A Cross-Sectional Study in Shanghai, China. Int. J. Environ. Res. Public Health 2016, 13, 537. [Google Scholar] [CrossRef]
  54. Dai, H.; Jing, S.; Wang, H.; Ma, Y.; Li, L.; Song, W.; Kan, H. VOC characteristics and inhalation health risks in newly renovated residences in Shanghai, China. Sci. Total Environ. 2017, 577, 73–83. [Google Scholar] [CrossRef] [PubMed]
  55. Franck, U.; Weller, A.; Röder, S.W.; Herberth, G.; Junge, K.M.; Kohajda, T.; von Bergen, M.; Rolle-Kampczyk, U.; Diez, U.; LiNA Study Group; et al. Prenatal VOC exposure and redecoration are related to wheezing in early infancy. Environ. Int. 2014, 73, 393–401. [Google Scholar] [CrossRef] [PubMed]
  56. Hansen, S.; Strøm, M.; Olsen, S.F.; Maslova, E.; Rantakokko, P.; Kiviranta, H.; Rytter, D.; Bech, B.H.; Hansen, L.V.; Halldorsson, T.I. Maternal concentrations of persistent organochlorine pollutants and the risk of asthma in offspring: Results from a prospective cohort with 20 years of follow-up. Environ. Health Perspect. 2014, 122, 93–99. [Google Scholar] [CrossRef] [Green Version]
  57. Mamane, A.; Raherison, C.; Tessier, J.F.; Baldi, I.; Bouvier, G. Environmental exposure to pesticides and respiratory health. Eur. Respir. Rev. 2015, 24, 462–473. [Google Scholar] [CrossRef] [PubMed]
  58. Parker-Lalomio, M.; McCann, K.; Piorkowski, J.; Freels, S.; Persky, V.W. Prenatal exposure to polychlorinated biphenyls and asthma, eczema/hay fever, and frequent ear infections. J. Asthma 2018, 55, 1105–1115. [Google Scholar] [CrossRef]
  59. Impinen, A.; Longnecker, M.P.; Nygaard, U.C.; London, S.J.; Ferguson, K.K.; Haug, L.S.; Granum, B. Maternal levels of perfluoroalkyl substances (PFASs) during pregnancy and childhood allergy and asthma related outcomes and infections in the Norwegian Mother and Child (MoBa) cohort. Environ. Int. 2019, 124, 462–472. [Google Scholar] [CrossRef]
  60. Manzano-Salgado, C.B.; Granum, B.; Lopez-Espinosa, M.J.; Ballester, F.; Iñiguez, C.; Gascón, M.; Martínez, D.; Guxens, M.; Basterretxea, M.; Zabaleta, C.; et al. Prenatal exposure to perfluoroalkyl substances, immune-related outcomes, and lung function in children from a Spanish birth cohort study. Int. J. Hyg. Environ. Health 2019, 222, 945–954. [Google Scholar] [CrossRef]
  61. Vernet, C.; Pin, I.; Giorgis-Allemand, L.; Philippat, C.; Benmerad, M.; Quentin, J.; Calafat, A.M.; Ye, X.; Annesi-Maesano, I.; Siroux, V.; et al. In Utero Exposure to Select Phenols and Phthalates and Respiratory Health in Five-Year-Old Boys: A Prospective Study. Environ. Health Perspect. 2017, 125, 097006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Berger, K.; Eskenazi, B.; Balmes, J.; Kogut, K.; Holland, N.; Calafat, A.M.; Harley, K.G. Prenatal high molecular weight phthalates and bisphenol A, and childhood respiratory and allergic outcomes. Pediatr. Allergy Immunol. 2019, 30, 36–46. [Google Scholar] [CrossRef] [PubMed]
  63. Zhou, A.; Chang, H.; Huo, W.; Zhang, B.; Hu, J.; Xia, W.; Chen, Z.; Xiong, C.; Zhang, Y.; Wang, Y.; et al. Prenatal exposure to bisphenol A and risk of allergic diseases in early life. Pediatr. Res. 2017, 81, 851–856. [Google Scholar] [CrossRef] [PubMed]
  64. Ait Bamai, Y.; Miyashita, C.; Araki, A.; Nakajima, T.; Sasaki, S.; Kishi, R. Effects of prenatal di(2-ethylhexyl) phthalate exposure on childhood allergies and infectious diseases: The Hokkaido Study on Environment and Children’s Health. Sci. Total Environ. 2018, 618, 1408–1415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Kim, J.H.; Jeong, K.S.; Ha, E.H.; Park, H.; Ha, M.; Hong, Y.C.; Lee, S.J.; Lee, K.Y.; Jeong, J.; Kim, Y. Association between prenatal exposure to cadmium and atopic dermatitis in infancy. J. Korean Med. Sci. 2013, 28, 516–521. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Jedrychowski, W.; Perera, F.; Maugeri, U.; Miller, R.L.; Rembiasz, M.; Flak, E.; Mroz, E.; Majewska, R.; Zembala, M. Intrauterine exposure to lead may enhance sensitization to common inhalant allergens in early childhood: A prospective prebirth cohort study. Environ. Res. 2011, 111, 119–124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Recio-Vega, R.; Gonzalez-Cortes, T.; Olivas-Calderon, E.; Lantz, R.C.; Gandolfi, A.J.; Gonzalez-De Alba, C. In utero and early childhood exposure to arsenic decreases lung function in children. J. Appl. Toxicol. 2015, 35, 358–366. [Google Scholar] [CrossRef] [Green Version]
  68. Norbäck, D.; Lu, C.; Zhang, Y.; Li, B.; Zhao, Z.; Huang, C.; Zhang, X.; Qian, H.; Sun, Y.; Sundell, J.; et al. Onset and remission of childhood wheeze and rhinitis across China—Associations with early life indoor and outdoor air pollution. Environ. Int. 2019, 123, 61–69. [Google Scholar] [CrossRef]
  69. Lavigne, E.; Donelle, J.; Hatzopoulou, M.; Van Ryswyk, K.; van Donkelaar, A.; Martin, R.V.; Chen, H.; Stieb, D.M.; Gasparrini, A.; Crighton, E.; et al. Spatiotemporal Variations in Ambient Ultrafine Particles and the Incidence of Childhood Asthma. Am. J. Respir. Crit. Care Med. 2019, 199, 1487–1495. [Google Scholar] [CrossRef] [Green Version]
  70. Yang, S.I.; Lee, S.Y.; Kim, H.B.; Kim, H.C.; Leem, J.H.; Yang, H.J.; Kwon, H.; Seo, J.H.; Cho, H.J.; Yoon, J.; et al. Prenatal particulate matter affects new asthma via airway hyperresponsiveness in schoolchildren. Allergy 2019, 74, 675–684. [Google Scholar] [CrossRef]
  71. Lee, A.; Leon Hsu, H.H.; Mathilda Chiu, Y.H.; Bose, S.; Rosa, M.J.; Kloog, I.; Wilson, A.; Schwartz, J.; Cohen, S.; Coull, B.A.; et al. Prenatal fine particulate exposure and early childhood asthma: Effect of maternal stress and fetal sex. J. Allergy Clin. Immunol. 2018, 141, 1880–1886. [Google Scholar] [CrossRef] [Green Version]
  72. Morales, E.; Garcia-Esteban, R.; de la Cruz, O.A.; Basterrechea, M.; Lertxundi, A.; de Dicastillo, M.D.; Zabaleta, C.; Sunyer, J. Intrauterine and early postnatal exposure to outdoor air pollution and lung function at preschool age. Thorax 2015, 70, 64–73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Collinson, A.C.; Tamba, P.T.; Moore, S.E.; Morgan, G.; Prentice, A.M. Birth season and environmental influences on blood leucocyte and lymphocyte subpopulations in rural Gambian infants. BMC Immunol. 2008, 9, 18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Thysen, A.H.; Rasmussen, M.A.; Kreiner-Møller, E.; Larsen, J.M.; Følsgaard, N.V.; Bønnelykke, K.; Stokholm, J.; Bisgaard, H.; Brix, S. Season of birth shapes neonatal immune function. J. Allergy Clin. Immunol. 2016, 137, 1238–1246.e13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Lehmann, I.; Thoelke, A.; Weiss, M.; Schlink, U.; Schulz, R.; Diez, U.; Sierig, G.; Emmrich, F.; Jacob, B.; Belcredi, P.; et al. T cell reactivity in neonates from an East and a West German city--results of the LISA study. Allergy 2002, 57, 129–136. [Google Scholar] [CrossRef]
  76. Lendor, C.; Johnson, A.; Perzanowski, M.; Chew, G.L.; Goldstein, I.F.; Kelvin, E.; Perera, F.; Miller, R.L. Effects of winter birth season and prenatal cockroach and mouse allergen exposure on indoor allergen-specific cord blood mononuclear cell proliferation and cytokine production. Ann. Allergy Asthma Immunol. 2008, 101, 193–199. [Google Scholar] [CrossRef]
  77. Sullivan Dillie, K.T.; Tisler, C.J.; Dasilva, D.F.; Pappas, T.E.; Roberg, K.A.; Carlson-Dakes, K.T.; Evans, M.D.; Rosenthal, L.A.; Gangnon, R.E.; Gern, J.E.; et al. The influence of processing factors and non-atopy-related maternal and neonate characteristics on yield and cytokine responses of cord blood mononuclear cells. Clin. Exp. Allergy 2008, 38, 298–304. [Google Scholar] [CrossRef]
  78. Gold, D.R.; Bloomberg, G.R.; Cruikshank, W.W.; Visness, C.M.; Schwarz, J.; Kattan, M.; O’Connor, G.T.; Wood, R.A.; Burger, M.S.; Wright, R.J.; et al. Parental characteristics, somatic fetal growth, and season of birth influence innate and adaptive cord blood cytokine responses. J. Allergy Clin. Immunol. 2009, 124, 1078–1087. [Google Scholar] [CrossRef] [Green Version]
  79. Keski-Nisula, L.; Lappalainen, M.H.; Mustonen, K.; Hirvonen, M.R.; Pfefferle, P.I.; Renz, H.; Pekkanen, J.; Roponen, M. Production of interleukin-5, -10 and interferon-γ in cord blood is strongly associated with the season of birth. Clin. Exp. Allergy 2010, 40, 1658–1668. [Google Scholar] [CrossRef]
  80. Thilaganathan, B.; Meher-Homji, N.; Nicolaides, K.H. Labor: An immunologically beneficial process for the neonate. Am. J. Obstet. Gynecol. 1994, 171, 1271–1272. [Google Scholar] [CrossRef]
  81. Nikischin, W.; Peter, M.; Oldigs, H.D. The influence of mode of delivery on hematologic values in the umbilical vein. Gynecol. Obstet. Invest 1997, 43, 104–107. [Google Scholar] [CrossRef]
  82. Chirico, G.; Gasparoni, A.; Ciardelli, L.; Martinotti, L.; Rondini, G. Leukocyte counts in relation to the method of delivery during the first five days of life. Biol. Neonate. 1999, 75, 294–299. [Google Scholar] [CrossRef]
  83. Grönlund, M.M.; Nuutila, J.; Pelto, L.; Lilius, E.M.; Isolauri, E.; Salminen, S.; Kero, P.; Lehtonen, O.P. Mode of delivery directs the phagocyte functions of infants for the first 6 months of life. Clin. Exp. Immunol. 1999, 116, 521–526. [Google Scholar] [CrossRef]
  84. Almanzar, G.; Schönlaub, J.; Hammerer-Lercher, A.; Koppelstaetter, C.; Bernhard, D.; Prelogk, M. Influence of the delivery modus on subpopulations and replication of lymphocytes in mothers and newborns. Early Hum. Dev. 2015, 91, 663–670. [Google Scholar] [CrossRef]
  85. Birle, A.; Nebe, C.T.; Hill, S.; Hartmann, K.; Poeschl, J.; Koch, L. Neutrophil chemotaxis in cord blood of term and preterm neonates is reduced in preterm neonates and influenced by the mode of delivery and anaesthesia. PLoS ONE 2015, 10, e0120341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Yektaei-Karin, E.; Moshfegh, A.; Lundahl, J.; Berggren, V.; Hansson, L.O.; Marchini, G. The stress of birth enhances in vitro spontaneous and IL-8-induced neutrophil chemotaxis in the human newborn. Pediatr. Allergy Immunol. 2007, 18, 643–651. [Google Scholar] [CrossRef]
  87. Shen, C.M.; Lin, S.C.; Niu, D.M.; Kou, Y.R. Labour increases the surface expression of two Toll-like receptors in the cord blood monocytes of healthy term newborns. Acta Paediatr. 2009, 98, 959–962. [Google Scholar] [CrossRef] [PubMed]
  88. Lurà, M.P.; Gorlanova, O.; Müller, L.; Proietti, E.; Vienneau, D.; Reppucci, D.; Pavlovic, R.; Dahinden, C.; Röösli, M.; Latzin, P.; et al. Response of cord blood cells to environmental, hereditary and perinatal factors: A prospective birth cohort study. PLoS ONE 2018, 13, e0200236. [Google Scholar] [CrossRef]
  89. Werlang, I.C.R.; Mueller, N.T.; Pizoni, A.; Wisintainer, H.; Matte, U.; Costa, S.H.A.M.; Ramos, J.G.L.; Goldani, M.Z.; Dominguez-Bello, M.G.; Goldani, H.A.S. Associations of birth mode with cord blood cytokines, white blood cells, and newborn intestinal bifidobacteria. PLoS ONE 2018, 13, e0205962. [Google Scholar] [CrossRef] [PubMed]
  90. Gessler, P.; Dahinden, C. Increased respiratory burst and increased expression of complement receptor-3 (CD11b/CD18) and of IL-8 receptor-A in neutrophil granulocytes from newborns after vaginal delivery. Biol. Neonate 2003, 83, 107–112. [Google Scholar] [CrossRef]
  91. Kotiranta-Ainamo, A.; Apajasalo, M.; Pohjavuori, M.; Rautonen, N.; Rautonen, J. Mononuclear cell subpopulations in preterm and full-term neonates: Independent effects of gestational age, neonatal infection, maternal pre-eclampsia, maternal betamethason therapy, and mode of delivery. Clin. Exp. Immunol. 1999, 115, 309–314. [Google Scholar] [CrossRef]
  92. Samelson, R.; Larkey, D.M.; Amankwah, K.S.; McConnachie, P. Effect of labor on lymphocyte subsets in full-term neonates. Am. J. Reprod. Immunol. 1992, 28, 71–73. [Google Scholar] [CrossRef] [PubMed]
  93. Bili, H.; Fleva, A.; Pados, G.; Argyriou, T.; Tsolakidis, D.; Pavlitou, A.; Tarlatzis, B.C. Regulatory Τ-cell differentiation between maternal and cord blood samples in pregnancies with spontaneous vaginal delivery and with elective cesarean section. Am. J. Reprod. Immunol. 2011, 65, 173–179. [Google Scholar] [CrossRef] [PubMed]
  94. Yildiran, A.; Yurdakul, E.; Guloglu, D.; Dogu, F.; Arsan, S.; Arikan, M.; Cengiz, L.; Tezcan, S.; Ikinciogullari, A. The effect of mode of delivery on T regulatory (Treg) cells of cord blood. Indian J. Pediatr. 2011, 78, 1234–1238. [Google Scholar] [CrossRef] [PubMed]
  95. Steinborn, A.; Engst, M.; Haensch, G.M.; Mahnke, K.; Schmitt, E.; Meuer, S.; Sohn, C. Small for gestational age (SGA) neonates show reduced suppressive activity of their regulatory T cells. Clin. Immunol. 2010, 134, 188–197. [Google Scholar] [CrossRef] [PubMed]
  96. Thornton, C.A.; Capristo, C.C.; Power, L.L.; Holloway, J.; Popplewell, E.J.; Diaper, N.D.; Warner, J.O. The effect of labor on neonatal T-cell phenotype and function. Pediatr. Res. 2003, 54, 120–124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Steinborn, A.; Sohn, C.; Sayehli, C.; Baudendistel, A.; Hüwelmeier, D.; Solbach, C.; Schmitt, E.; Kaufmann, M. Spontaneous labour at term is associated with fetal monocyte activation. Clin. Exp. Immunol. 1999, 117, 147–152. [Google Scholar] [CrossRef] [PubMed]
  98. Malamitsi-Puchner, A.; Protonotariou, E.; Boutsikou, T.; Makrakis, E.; Sarandakou, A.; Creatsas, G. The influence of the mode of delivery on circulating cytokine concentrations in the perinatal period. Early Hum. Dev. 2005, 81, 387–392. [Google Scholar] [CrossRef] [PubMed]
  99. Treviño-Garza, C.; Villarreal-Martínez, L.; Estrada-Zúñiga, C.M.; Leal-Treviño, M.; Rodríguez-Balderrama, I.; Nieto-Sanjuanero, A.; Cárdenas-Del Castillo, B.; Montes-Tapia, F.F.; de la O-Cavazos, M. Leptin, IL-6 and TNF-α levels in umbilical cord blood of healthy term newborns in relation to mode of delivery. J. Obstet. Gynaecol. 2016, 36, 719–721. [Google Scholar] [CrossRef]
  100. Nandanan, B.; Chua, M.C.; Chiang, W.C.; Goh, A.; Kumar, D.; Knippels, L.; Garssen, J.; Sandalova, E. Influence of mode of delivery on cytokine expression in cord blood. Hum. Immunol. 2019, 80, 533–536. [Google Scholar] [CrossRef]
  101. Brown, M.A.; Rad, P.Y.; Halonen, M.J. Method of birth alters interferon-gamma and interleukin-12 production by cord blood MNC. Pediatr. Allergy Immunol. 2003, 14, 106–111. [Google Scholar] [CrossRef]
  102. Ly, N.P.; Ruiz-Pérez, B.; Onderdonk, A.B.; Tzianabos, A.O.; Litonjua, A.A.; Liang, C.; Laskey, D.; Delaney, M.L.; DuBois, A.M.; Levy, H.; et al. Mode of delivery and cord blood cytokines: A birth cohort study. Clin. Mol. Allergy 2006, 4, 13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Schaub, B.; Liu, J.; Höppler, S.; Schleich, I.; Huehn, J.; Olek, S.; Wieczorek, G.; Illi, S.; von Mutius, E. Maternal farm exposure modulates neonatal immune mechanisms through regulatory T cells. J. Allergy Clin. Immunol. 2009, 123, 774–782.e5. [Google Scholar] [CrossRef] [PubMed]
  104. Lundell, A.C.; Hesselmar, B.; Nordström, I.; Adlerberth, I.; Wold, A.E.; Rudin, A. Higher B-cell activating factor levels at birth are positively associated with maternal dairy farm exposure and negatively related to allergy development. J. Allergy Clin. Immunol. 2015, 136, 1074–1082.e3. [Google Scholar] [CrossRef] [PubMed]
  105. Aichbhaumik, N.; Zoratti, E.M.; Strickler, R.; Wegienka, G.; Ownby, D.R.; Havstad, S.; Johnson, C.C. Prenatal exposure to household pets influences fetal immunoglobulin E production. Clin. Exp. Allergy 2008, 38, 1787–1794. [Google Scholar] [CrossRef] [Green Version]
  106. Sybilski, A.J.; Doboszynska, A.; Samolinski, B. Total and antigen-specific IGE levels in umbilical cord blood. Eur. J. Med. Res. 2009, 14 (Suppl. 4), 233–236. [Google Scholar] [CrossRef] [Green Version]
  107. Heinrich, J.; Bolte, G.; Hölscher, B.; Douwes, J.; Lehmann, I.; Fahlbusch, B.; Bischof, W.; Weiss, M.; Borte, M.; Wichmann, H.E.; et al. Allergens and endotoxin on mothers’ mattresses and total immunoglobulin E in cord blood of neonates. Eur. Respir. J. 2002, 20, 617–623. [Google Scholar] [CrossRef] [PubMed]
  108. Peters, J.; Suglia, S.F.; Platts-Mills, T.A.; Hosen, J.; Gold, D.R.; Wright, R.J. Relationships among prenatal aeroallergen exposure and maternal and cord blood IgE: Project ACCESS. J. Allergy Clin. Immunol. 2009, 123, 1041–1046. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Ege, M.J.; Herzum, I.; Büchele, G.; Krauss-Etschmann, S.; Lauener, R.P.; Roponen, M.; Hyvärinen, A.; Vuitton, D.A.; Riedler, J.; Brunekreef, B.; et al. Prenatal exposure to a farm environment modifies atopic sensitization at birth. J. Allergy Clin. Immunol. 2008, 122, 407–412. [Google Scholar] [CrossRef]
  110. Pfefferle, P.I.; Büchele, G.; Blümer, N.; Roponen, M.; Ege, M.J.; Krauss-Etschmann, S.; Genuneit, J.; Hyvärinen, A.; Hirvonen, M.R.; Lauener, R.; et al. Cord blood cytokines are modulated by maternal farming activities and consumption of farm dairy products during pregnancy: The PASTURE Study. J. Allergy Clin. Immunol. 2010, 125, 108–115. [Google Scholar] [CrossRef]
  111. Lappalainen, M.H.; Huttunen, K.; Roponen, M.; Remes, S.; Hirvonen, M.R.; Pekkanen, J. Exposure to dogs is associated with a decreased tumour necrosis factor-α-producing capacity in early life. Clin. Exp. Allergy 2010, 40, 1498–1506. [Google Scholar] [CrossRef] [PubMed]
  112. Roponen, M.; Hyvärinen, A.; Hirvonen, M.R.; Keski-Nisula, L.; Pekkanen, J. Change in IFN-gamma-producing capacity in early life and exposure to environmental microbes. J. Allergy Clin. Immunol. 2005, 116, 1048–1052. [Google Scholar] [CrossRef] [PubMed]
  113. Uzuner, N.; Babayigit Hocaoglu, A.; Olmez Erge, D.; Asilsoy, S.; Karaman, O.; Altun, Z.; Kumral, A.; Tufekci, O.; Islekel, H. Raised interleukin-13 levels in cord blood increases the risk of allergic sensitization at 5 years of age. Iran. J. Allergy Asthma Immunol. 2013, 12, 107–114. [Google Scholar] [PubMed]
  114. Lappalainen, M.H.; Hyvärinen, A.; Hirvonen, M.R.; Rintala, H.; Roivainen, J.; Renz, H.; Pfefferle, P.I.; Nevalainen, A.; Roponen, M.; Pekkanen, J. High indoor microbial levels are associated with reduced Th1 cytokine secretion capacity in infancy. Int. Arch. Allergy Immunol. 2012, 159, 194–203. [Google Scholar] [CrossRef]
  115. Hagendorens, M.M.; Ebo, D.G.; Bridts, C.H.; Van de Water, L.; De Clerck, L.S.; Stevens, W.J. Prenatal exposure to house dust mite allergen (Der p 1), cord blood T cell phenotype and cytokine production and atopic dermatitis during the first year of life. Pediatr. Allergy Immunol. 2004, 15, 308–315. [Google Scholar] [CrossRef]
  116. Harrison, K.L. The Effect of Maternal Smoking on Neonatal Leucocytes. Aust. N. Zeal. J. Obstet. Gynaecol. 1979, 19, 166–168. [Google Scholar] [CrossRef]
  117. Mercelina-Roumans, P.E.A.M.; Breukers, R.B.G.E.; Ubachs, J.M.H.; Van Wersch, J.W.J. Hematological variables in cord blood of neonates of smoking and nonsmoking mothers. J. Clin. Epidemiol. 1996, 49, 449–454. [Google Scholar] [CrossRef]
  118. Pachlopnik Schmid, J.M.; Kuehni, C.E.; Strippoli, M.P.; Roiha, H.L.; Pavlovic, R.; Latzin, P.; Gallati, S.; Kraemer, R.; Dahinden, C.; Frey, U.; et al. Maternal tobacco smoking and decreased leukocytes, including dendritic cells, in neonates. Pediatr. Res. 2007, 61, 462–466. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Karwowska, W.; Rachtan, J.; Wieczorek, E. The effect of cigarette smoking on IgE levels and circulating lymphocytes in cord blood. Centr. Eur. J. Immunol. 2008, 33, 228–231. [Google Scholar]
  120. Hinz, D.; Bauer, M.; Röder, S.; Olek, S.; Huehn, J.; Sack, U.; Borte, M.; Simon, J.C.; Lehmann, I.; Herberth, G.; et al. Cord blood Tregs with stable FOXP3 expression are influenced by prenatal environment and associated with atopic dermatitis at the age of one year. Allergy 2012, 67, 380–389. [Google Scholar] [CrossRef]
  121. Noakes, P.S.; Hale, J.; Thomas, R.; Lane, C.; Devadason, S.G.; Prescott, S.L. Maternal smoking is associated with impaired neonatal toll-like-receptor-mediated immune responses. Eur. Respir. J. 2006, 28, 721–729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Sevgican, U.; Rothers, J.; Stern, D.A.; Lohman, I.C.; Wright, A.L. Predictors of neonatal production of IFN-γ and relation to later wheeze. J. Allergy Clin. Immunol. 2012, 129, 567–568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Noakes, P.S.; Holt, P.G.; Prescott, S.L. Maternal smoking in pregnancy alters neonatal cytokine responses. Allergy 2003, 58, 1053–1058. [Google Scholar] [CrossRef]
  124. Latzin, P.; Frey, U.; Armann, J.; Kieninger, E.; Fuchs, O.; Röösli, M.; Schaub, B. Exposure to moderate air pollution during late pregnancy and cord blood cytokine secretion in healthy neonates. PLoS ONE 2011, 6, e23130. [Google Scholar]
  125. Belles-Isles, M.; Ayotte, P.; Dewailly, E.; Weber, J.P.; Roy, R. Cord blood lymphocyte functions in newborns from a remote maritime population exposed to organochlorines and methylmercury. J. Toxicol. Environ. Health A 2002, 65, 165–182. [Google Scholar] [CrossRef]
  126. Bilrha, H.; Roy, R.; Moreau, B.; Belles-Isles, M.; Dewailly, E.; Ayotte, P. In vitro activation of cord blood mononuclear cells and cytokine production in a remote coastal population exposed to organochlorines and methyl mercury. Environ. Health Perspect. 2003, 111, 1952–1957. [Google Scholar] [CrossRef]
  127. Noakes, P.S.; Taylor, P.; Wilkinson, S.; Prescott, S.L. The relationship between persistent organic pollutants in maternal and neonatal tissues and immune responses to allergens: A novel exploratory study. Chemosphere 2006, 63, 1304–1311. [Google Scholar] [CrossRef] [PubMed]
  128. Brooks, K.; Hasan, H.; Samineni, S.; Gangur, V.; Karmaus, W. Placental p,p′-dichlorodiphenyldichloroethylene and cord blood immune markers. Pediatr. Allergy Immunol. 2007, 18, 621–624. [Google Scholar] [CrossRef]
  129. Horváthová, M.; Jahnová, E.; Palkovičová, L.; Trnovec, T.; Hertz-Picciotto, I. The kinetics of cell surface receptor expression in children perinatally exposed to polychlorinated biphenyls. J. Immunotoxicol. 2011, 8, 367–380. [Google Scholar] [CrossRef] [PubMed]
  130. Horváthová, M.; Jahnová, E.; Palkovičová, L.; Trnovec, T.; Hertz-Picciotto, I. Dynamics of lymphocyte subsets in children living in an area polluted by polychlorinated biphenyls. J. Immunotoxicol. 2011, 8, 333–345. [Google Scholar] [CrossRef] [PubMed]
  131. Neta, G.; Goldman, L.R.; Barr, D.; Apelberg, B.J.; Witter, F.R.; Halden, R.U. Fetal exposure to chlordane and permethrin mixtures in relation to inflammatory cytokines and birth outcomes. Environ. Sci. Technol. 2011, 45, 1680–1687. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Wang, I.J.; Hsieh, W.S.; Chen, C.Y.; Fletcher, T.; Lien, G.W.; Chiang, H.L.; Chiang, C.F.; Wu, T.N.; Chen, P.C. The effect of prenatal perfluorinated chemicals exposures on pediatric atopy. Environ. Res. 2011, 111, 785–791. [Google Scholar] [CrossRef] [PubMed]
  133. Ashley-Martin, J.; Dodds, L.; Levy, A.R.; Platt, R.W.; Marshall, J.S.; Arbuckle, T.E. Prenatal exposure to phthalates, bisphenol A and perfluoroalkyl substances and cord blood levels of IgE, TSLP and IL-33. Environ. Res. 2015, 140, 360–368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Ashley-Martin, J.; Levy, A.R.; Arbuckle, T.E.; Platt, R.W.; Marshall, J.S.; Dodds, L. Maternal exposure to metals and persistent pollutants and cord blood immune system biomarkers. Environ. Health 2015, 14, 52. [Google Scholar] [CrossRef] [Green Version]
  135. Miyashita, C.; Bamai, Y.A.; Araki, A.; Itoh, S.; Minatoya, M.; Kobayashi, S.; Kajiwara, J.; Hori, T.; Kishi, R. Prenatal exposure to dioxin-like compounds is associated with decreased cord blood IgE and increased risk of wheezing in children aged up to 7 years: The Hokkaido study. Sci. Total Environ. 2018, 610–611, 191–199. [Google Scholar] [CrossRef] [Green Version]
  136. Kelley, A.S.; Banker, M.; Goodrich, J.M.; Dolinoy, D.C.; Burant, C.; Domino, S.E.; Smith, Y.R.; Song, P.X.K.; Padmanabhan, V. Early pregnancy exposure to endocrine disrupting chemical mixtures are associated with inflammatory changes in maternal and neonatal circulation. Sci. Rep. 2019, 9, 5422. [Google Scholar] [CrossRef] [Green Version]
  137. Liao, S.L.; Tsai, M.H.; Lai, S.H.; Yao, T.C.; Hua, M.C.; Yeh, K.W.; Chiang, C.H.; Huang, S.Y.; Huang, J.L. Prenatal exposure to bisphenol-A is associated with Toll-like receptor-induced cytokine suppression in neonates. Pediatr. Res. 2016, 79, 438–444. [Google Scholar] [CrossRef] [Green Version]
  138. Huang, Y.F.; Wang, P.W.; Huang, L.W.; Lai, C.H.; Yang, W.; Wu, K.Y.; Lu, C.A.; Chen, H.C.; Chen, M.L. Prenatal Nonylphenol and Bisphenol A Exposures and Inflammation Are Determinants of Oxidative/Nitrative Stress: A Taiwanese Cohort Study. Environ. Sci. Technol. 2017, 51, 6422–6429. [Google Scholar] [CrossRef]
  139. Lehmann, I.; Thoelke, A.; Rehwagen, M.; Rolle-Kampczyk, U.; Schlink, U.; Schulz, R.; Borte, M.; Diez, U.; Herbarth, O. The influence of maternal exposure to volatile organic compounds on the cytokine secretion profile of neonatal T cells. Environ. Toxicol. 2002, 17, 203–210. [Google Scholar] [CrossRef]
  140. Ahmed, S.; Khoda, S.M.; Rekha, R.S.; Gardner, R.M.; Ameer, S.S.; Moore, S.; Ekström, E.C.; Vahter, M.; Raqib, R. Arsenic-Associated Oxidative Stress, Inflammation, and Immune Disruption in Human Placenta and Cord Blood. Environ. Health Perspect. 2011, 119, 258–264. [Google Scholar] [CrossRef] [Green Version]
  141. Nadeau, K.C.; Li, Z.; Farzan, S.; Koestler, D.; Robbins, D.; Fei, D.L.; Malipatlolla, M.; Maecker, H.; Enelow, R.; Korrick, S.; et al. In utero arsenic exposure and fetal immune repertoire in a US pregnancy cohort. Clin. Immunol. 2014, 155, 188–197. [Google Scholar] [CrossRef] [Green Version]
  142. Nygaard, U.C.; Li, Z.; Palys, T.; Jackson, B.; Subbiah, M.; Malipatlolla, M.; Sampath, V.; Maecker, H.; Karagas, M.R.; Nadeau, K.C. Cord blood T cell subpopulations and associations with maternal cadmium and arsenic exposures. PLoS ONE 2017, 12, e0179606. [Google Scholar] [CrossRef]
  143. Kim, J.; Kim, S.; Woo, S.Y.; Chung, J.Y.; Hong, Y.S.; Oh, S.Y.; Choi, S.J.; Oh, S.Y.; Kim, K.W.; Shin, Y.H.; et al. Prenatal Exposure to Lead and Chromium is Associated with IL-13 Levels in Umbilical Cord Blood and Severity of Atopic Dermatitis: COCOA Study. Immune. Netw. 2019, 19, e42. [Google Scholar] [CrossRef] [PubMed]
  144. Hertz-Picciotto, I.; Dostál, M.; Dejmek, J.; Selevan, S.G.; Wegienka, G.; Gomez-Caminero, A.; Srám, R.J. Air pollution and distributions of lymphocyte immunophenotypes in cord and maternal blood at delivery. Epidemiology 2002, 13, 172–183. [Google Scholar] [CrossRef] [PubMed]
  145. Hertz-Picciotto, I.; Herr, C.E.W.; Yap, P.S.; Dostál, M.; Shumway, R.H.; Ashwood, P.; Lipsett, M.; Joad, J.P.; Pinkerton, K.E.; Srám, R.J. Air pollution and lymphocyte phenotype proportions in cord blood. Environ. Health Perspect. 2005, 113, 1391–1398. [Google Scholar] [CrossRef] [Green Version]
  146. Herr, C.E.W.; Dostal, M.; Ghosh, R.; Ashwood, P.; Lipsett, M.; Pinkerton, K.E.; Sram, R.; Hertz-Picciotto, I. Air pollution exposure during critical time periods in gestation and alterations in cord blood lymphocyte distribution: A cohort of livebirths. Environ. Health. 2010, 9, 46. [Google Scholar] [CrossRef] [Green Version]
  147. Baïz, N.; Slama, R.; Béné, M.C.; Charles, M.A.; Kolopp-Sarda, M.N.; Magnan, A.; Thiebaugeorges, O.; Faure, G.; Annesi-Maesano, I. Maternal exposure to air pollution before and during pregnancy related to changes in newborn’s cord blood lymphocyte subpopulations. The EDEN study cohort. BMC Pregnancy Childbirth 2011, 11, 87. [Google Scholar] [CrossRef] [Green Version]
  148. García-Serna, A.M.; Hernández-Caselles, T.; Jiménez-Guerrero, P.; Martín-Orozco, E.; Pérez-Fernández, V.; Cantero-Cano, E.; Muñoz-García, M.; Ballesteros-Meseguer, C.; Pérez de Los Cobos, I.; García-Marcos, L.; et al. Air pollution from traffic during pregnancy impairs newborn’s cord blood immune cells: The NELA cohort. Environ. Res. 2020, 110468. [Google Scholar] [CrossRef] [PubMed]
  149. Ashley-Martin, J.; Lavigne, E.; Arbuckle, T.E.; Johnson, M.; Hystad, P.; Crouse, D.L.; Marshall, J.S.; Dodds, L. Air pollution during pregnancy and cord blood immune system biomarkers. J. Occup. Environ. Med. 2016, 58, 979–986. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Watson, P.E.; McDonald, B.W. Seasonal variation of nutrient intake in pregnancy: Effects on infant measures and possible influence on diseases related to season of birth. Eur. J. Clin. Nutr. 2007, 61, 1271–1280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Pham, M.N.; Bunyavanich, S. Prenatal Diet and the Development of Childhood Allergic Diseases: Food for Thought. Curr. Allergy Asthma Rep. 2018, 18, 58. [Google Scholar] [CrossRef]
  152. Hornsby, E.; Pfeffer, P.E.; Laranjo, N.; Cruikshank, W.; Tuzova, M.; Litonjua, A.A.; Weiss, S.T.; Carey, V.J.; O’Connor, G.; Hawrylowicz, C. Vitamin D supplementation during pregnancy: Effect on the neonatal immune system in a randomized controlled trial. J. Allergy Clin. Immunol. 2018, 141, 269–278.e1. [Google Scholar] [CrossRef] [Green Version]
  153. Nikolaidis, C.; Katotomichelakis, M.; Nena, E.; Makris, M.; Tsakas, M.; Michopoulos, I.; Constantinidis, T.C.; Danielides, V. Seasonal variations of allergenic pollen in a Mediterranean region—Alexandroupolis, north-east Greece. Ann. Agric. Environ. Med. 2015, 22, 685–689. [Google Scholar] [CrossRef]
  154. Antón, S.F.; de la Cruz, D.R.; Sánchez, A.G.; Dávila, I.; Sánchez, S.J.; Sánchez Reyes, E. Urban atmospheric levels of allergenic pollen: Comparison of two locations in Salamanca, Central-Western Spain. Environ. Monit. Assess. 2020, 192, 414. [Google Scholar] [CrossRef] [PubMed]
  155. Ross, Z.; Ito, K.; Johnson, S.; Yee, M.; Pezeshki, G.; Clougherty, J.E.; Savitz, D.; Matte, T. Spatial and temporal estimation of air pollutants in New York City: Exposure assignment for use in a birth outcomes study. Environ. Health. 2013, 12, 51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Kristensen, K.; Fisker, N.; Haerskjold, A.; Ravn, H.; Simões, E.A.; Stensballe, L. Caesarean section and hospitalization for respiratory syncytial virus infection: A population-based study. Pediatr. Infect. Dis. J. 2015, 34, 145–148. [Google Scholar] [CrossRef]
  157. Douglas, L.C.; Leventer-Roberts, M.; Levinkron, O.; Wilson, K.M. Elective caesarean section and bronchiolitis hospitalization: A retrospective cohort study. Pediatr. Allergy Immunol. 2020, 32, 280–287. [Google Scholar] [CrossRef] [PubMed]
  158. Thysen, A.H.; Larsen, J.M.; Rasmussen, M.A.; Stokholm, J.; Bønnelykke, K.; Bisgaard, H.; Brix, S. Prelabor cesarean section bypasses natural immune cell maturation. J. Allergy Clin. Immunol. 2015, 136, 1123–1125.e6. [Google Scholar] [CrossRef] [Green Version]
  159. Francino, M.P. Birth Mode-Related Differences in Gut Microbiota Colonization and Immune System Development. Ann. Nutr. Metab. 2018, 73 (Suppl. 3), 12–16. [Google Scholar] [CrossRef]
  160. von Mutius, E.; Vercelli, D. Farm living: Effects on childhood asthma and allergy. Nat. Rev. Immunol. 2010, 10, 861–868. [Google Scholar] [CrossRef]
  161. Huang, F.; Yin, J.N.; Wang, H.B.; Liu, S.Y.; Li, Y.N. Association of imbalance of effector T cells and regulatory cells with the severity of asthma and allergic rhinitis in children. Allergy Asthma Proc. 2017, 38, 70–77. [Google Scholar] [CrossRef] [PubMed]
  162. Sanchez, T.R.; Perzanowski, M.; Graziano, J.H. Inorganic arsenic and respiratory health, from early life exposure to sex-specific effects: A systematic review. Environ Res. 2016, 147, 537–555. [Google Scholar] [CrossRef] [Green Version]
  163. Farzan, S.F.; Li, Z.; Korrick, S.A.; Spiegelman, D.; Enelow, R.; Nadeau, K.; Baker, E.; Karagas, M.R. Infant Infections and Respiratory Symptoms in Relation to in Utero Arsenic Exposure in a U.S. Cohort. Environ. Health Perspect. 2016, 124, 840–847. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Ahmed, S.M.; Branscum, A.; Welch, B.M.; Megowan, M.; Bethel, J.W.; Odden, M.C.; Joya, S.A.; Ibn Hasan, M.O.S.; Lin, P.I.; Mostofa, G.; et al. A prospective cohort study of in utero and early childhood arsenic exposure and infectious disease in 4- to 5-year-old Bangladeshi children. Environ. Epidemiol. 2020, 4, e086. [Google Scholar] [CrossRef] [PubMed]
  165. Schmidt, M.E.; Varga, S.M. The CD8 T Cell RespoTablense to Respiratory Virus Infections. Front. Immunol. 2018, 9, 678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Table 1. Season of birth and changes in the immune system of neonates (authors ordered by year and then alphabetically).
Table 1. Season of birth and changes in the immune system of neonates (authors ordered by year and then alphabetically).
Author, Year [Ref.]Location
Study Design
N
Outcomes AssessedSeason of BirthStatistically Significant Main Findings
Immune Cells FrequencyCytokine Patterns/Ig
Lehmann et al. 2002 [75]Germany
Cohort
N = 158
Cytokine responseSummer↓ induced-TNF-α (T cells)
↓ induced-IFN-γ (T cells)
↑ induced-IL-4 (T cells)
Sullivan Dillie et al. 2008 [77]USA
Cohort
N = 272
Cytokine responseSpring or summer↑ PHA-induced IL-5
↑ PMA-induced IL-5 (summer)
↑ Staphylococcus aureus-induced IL-5
↑ PHA-induced IL-13
↑ PMA-induced IL-13
Autumn or winter↑ CBMC proliferation
↓ Staphylococcus aureus-induced IL-13
Collison et al. 2008 [73]Gambia
Cohort
N = 138
Lymphocytes (counts) Wet season↑ leukocytes
↑ lymphocytes
Lendor et al. 2008 [76]USA
Cohort
N = 350
Cytokine response
IgE
Winter↑ cockroach-induced IL-5
Gold et al. 2009 [78]USA
Cohort
N = 558
Cytokine productionAutumn or winter↑ IFN-γ, IL-8 and TNF-α
↑ IL-12p40
↑ LPS-induced IFN-α
↑ PG-induced IFN-α
↑ CpG-induced IL-8, TNF-α, and IL-10
↑ cockroach-induced IFN-γ
↑ dust mite-induced IFN-γ
↑ tetanus toxoid-induced IFN-γ
Spring/ summer↑ CpG-induced IFN-α
Keski-Nisula et al. 2010 [79]Finland
Cohort
N = 423
Leukocytes (counts)Cytokine response Spring↓ P/I-induced IL-5, IL-10 and IFN-γ
Thysen et al. 2016 [74]Denmark
Cohort
N = 84
Leukocytes
Lymphocytes
(counts)
Winter↑ Leukocytes, granulocytes, neutrophils and monocytes
↑ Plasmacytoid dendritic cells
↑ CD56 bright NK cells
↑ Activated Th cells
Spring ↑ Tc cells
CBMC: cord blood mononuclear cell; CD: cluster of differentiation; CpG: Immunostimulatory CpG oligonucleotides; IFN: interferon; IL: interleukin; LPS: Lipopolysaccharide; NK: natural killer; PHA: Phytohemagglutinin; PMA: phorbol 12-myristate 13-acetate; P/I: phorbol ester and ionomycin PG: Peptidoglycan; Tc: T cytotoxic cells; Th: T helper cell; TNF: tumor necrosis factor.
Table 2. Mode of delivery and changes in the immune system of neonates (authors ordered by year and then alphabetically).
Table 2. Mode of delivery and changes in the immune system of neonates (authors ordered by year and then alphabetically).
Author, Year [Ref.]Location
Study Design
N
Outcomes AssessedMode of DeliveryStatistically Significant Main Findings
Immune Cells FrequencyCytokine Patterns/Ig
Samelson et al. 1992 [92]USA
Cross-sectional
N = 12
Lymphocytes (%) Vaginal delivery↓ T and Th cells
↑ NK cells
Thilaganathan et al. 1994 [80]UK
Cross-sectional
N = 40
Leukocytes Lymphocytes
(counts)
Vaginal delivery↑ Leukocytes, neutrophils, monocytes and NK cells
Nikischin et al. 1997 [81]Germany
Cross-sectional
N = 121
Leukocytes (counts) Vaginal delivery↑ Leukocytes
Chirico et al. 1999 [82]Italy
Cross-sectional
N = 203
Leukocytes (counts) Vaginal delivery↑ Leukocytes and neutrophils
Grönlund et al. 1999 [83]Finland
Cross-sectional
N = 64
Leukocytes
(counts and %)
Vaginal delivery↑ Leukocytes and neutrophils
↓ Lymphocytes
Kotiranta-Ainamo et al. 1999 [91]Finland
Cross-sectional
N = 50
Lymphocytes Macrophages (%) Cesarean section ↓ CD14+ cells
Steinborn et al. 1999 [97]Germany
Cross-sectional
N = 84
Cytokine productionVaginal delivery↑ IL-6 in myelomonocytic cord blood cells
Brown et al. 2003 [101]USA
Cross-sectional
N = 16
Cytokine responseVaginal delivery↑ ConA-induced IFN-γ
↑ LPS-induced IFN-γ and IL-12
↑ PHA-induced IFN-γ
Gessler et al. 2003 [90]Switzerland Cross-sectional
N = 30
Neutrophils (counts) Vaginal delivery↑ Neutrophils
↑ E.Coli-induced phagocytic respiratory burst
Thornton et al. 2003 [96]UK
Cross-sectional
N = 27
Leukocytes (%)
Lymphocytes (%)
Cytokine responseCesarean section↑ CD62L+ Th cells
Malamitsi-Puchner et al. 2005 [98]Greece
Cohort
N = 78
Cytokine productionVaginal delivery↑ IFN-γ and IL-1β levels
↑ IL-6 and TNF-α levels
Ly et al. 2006 [102]USA
Cohort
N = 37
Cytokine responseCesarean section↑ IFN-γ baseline secretion
↑ PHA-induced IFN-γ response
↑ cat dander allergen-induced IFN-γ response
↑ induced IL-13 response
Yektaei-Karin et al. 2007 [86]Sweden
Cross-sectional
N = 168
LeukocytesCytokine productionVaginal delivery
(compared to C-section)
↑ IL-8
↑ Leukocytes, neutrophils and monocytes
Assisted delivery (compared to C-section)↑ IFN-γ
↑ IL-8
↑ Leukocytes, neutrophils, lymphocytes and monocytes
Gold et al. 2009 [78]USA
Cohort
N = 609
Cytokine responseCesarean section↓ LPS-induced IL-8 response
↓ PG-induced IL-8 response
↓ CpG-induced IL-12p40 response
↑ Respiratory syncytial virus-induced IL-12p40 response
↓ PHA-induced IL-13 response
Shen et al. 2009
[87]
Taiwan
Cross-sectional
N = 62
Leukocytes (counts) Cesarean section↓ Leukocytes, neutrophils, lymphocytes and monocytes
↓ TLR2 and TLR4 surface expression on monocytes
Keski-Nisula et al. 2010 [79]Finland
Cohort
N = 423
Leukocytes (counts)Cytokine responseCesarean section↓ Leukocytes
↓ induced IL-5 response
Long labour↑ Leukocytes
↑ induced IFN-α and IL-5 response
Prostaglandin induction↓ induced IFN-γ response
Steinborn et al. 2010 [95]Germany
Cross-sectional
N = 96
Lymphocytes Vaginal delivery↓ FoxP3 expression in Tregs cells
Bili et al. 2011 [93]Greece
Cross-sectional
N = 81
Lymphocytes (%) Vaginal delivery↑ NK cells
Yildiran et al. 2011 [94]Turkey
Cross-sectional
N = 39
Lymphocytes (counts) Vaginal delivery↑ Treg cells
Almanzar et al. 2015 [84]Austria
Cohort
N = 120
Lymphocytes (counts and %)Cytokine productionVaginal delivery↑ Leukocytes (counts)
↑ Neutrophils and NK cells (%)
↓ Lymphocytes and T cells (%)
↑ IFN-γ, IL-2 and IL-8
Birle et al. 2015 [85]Germany
Cohort
N = 66
Leukocytes (counts) Vaginal delivery↑ Leukocytes
Treviño-Garza et al. 2016 [99]Mexico
Cross-sectional
N = 125
Cytokine productionVaginal delivery↑ IL-6
Liao et al. 2017 [23]Taiwan
Cohort
N = 487
Cytokine responseCesarean section↓ TLR1-stimulated TNF-α and IL-6
Lurà et al. 2018 [88]Switzerland
Cohort
N = 289
Leukocytes (counts) Cesarean section↓ Leukocytes, basophilic granulocytes
↓ Plasmacytoid dendritic cells
Werlang et al. 2018 [89]Brazil
Cross-sectional
N = 64
Leukocytes (counts)Cytokine productionCesarean section↓ Leukocytes
↓ GM-CSF
Nandanan et al. 2019 [100]Singapore
Cohort
N = 98
Cytokine productionVaginal delivery↑ IL-6 and IL-8
↓ TNF-α and G-CSF levels
CD: cluster of differentiation; ConA: Concanavalin A; G-CSF: Granulocyte-colony stimulating Factor; CpG: Immunostimulatory CpG oligonucleotides; E.Coli: Escherichia coli; GM-CSF: Granulocyte-macrophage colony-stimulating factor; IL: interleukin; IFN: interferon; LPS: Lipopolysaccharide; NK: natural killer; PHA: P Phytohemagglutinin; PG: Peptidoglycan; Th: T helper cell; Treg: T regulatory cells; TLR: Toll-like receptor; TNF: tumor necrosis factor.
Table 3. Prenatal exposure to a farming environment, pets and common allergens and changes in the immune system of neonates (authors ordered by exposure, year and then alphabetically).
Table 3. Prenatal exposure to a farming environment, pets and common allergens and changes in the immune system of neonates (authors ordered by exposure, year and then alphabetically).
Author, Year [Ref]Location
Study Design
N
Outcomes AssessedFactor AssessedStatistically Significant Main Findings
Immune Cells FrequencyCytokine Patterns/Ig
Farming environment
Ege et al. 2008 [109]5 European countries
(rural areas)
Cohort N = 922
Specific IgE levels (food and common inhalants)Maternal farm exposures↓ grass pollen-specific IgE
↓ seasonal allergens -specific IgE
↑ cow’s milk-specific IgE
↑ food allergens -specific IgE
Consumption of boiled farm milk↑ cow’s milk-specific IgE
↑ food allergens-specific IgE
Maternal exposure to animal sheds, contact to cattle, removing dung, cleaning the henhouse, handling silage and hay during pregnancy↓ seasonal allergens-specific IgE
Schaub et al. 2009 [103]Germany
Cohort
N = 82
Treg cells
(counts)
Cytokine responsesFarming mothers↑ Treg number and function
↓ Der p 1 plus PG-induced IL-5
↑ Der p 1-induced IL-6
↑ Der p 1 plus PG-induced IL-6
Cytokine responsesStables↑ PG-induced CD4+CD25+ T cells
↓ Der p 1 plus PG-induced IL-5
Cytokine responsesNumber of animal species ≥2↑ IL-10
↑ Der p 1-induced IFN-γ
↑ PG-induced IFN-γ
↓ Der p 1 plus PG-induced IL-5
Keski-Nisula et al. 2010 [79]Finland
Cohort
N = 423
Leukocytes (counts)Cytokine responsesFather as farmer↓ Leukocytes
↓ induced IFN-γ, IL-5 and IL-10
Pfefferle et al. 2010 [110]5 European countries
(rural areas)
Cohort N = 625
Cytokine responseMaternal farm exposures↑ induced IFN-γ
House spent in barn↑ induced TNF-α
Contact with farm animal species
Consumption of unskimmed farm milk
Consumption of butter made of farm milk
↑ induced TNF-α and IFN-γ
Consumption of yogurt made of farm milk↓ induced IL-5 and IL-10
Consumption of cheese made of farm milk↑ induced IL-5
Lundell et al. 2015 [104]Sweden
Cohort
N = 65
Cytokine productionFarming mothers↑ B-cell activating factor (BAFF)
Indoor allergens
Heinrich et al. 2002 [107]Germany
Cohort
N = 1332
IgEEndotoxinU-shaped association with IgE
↓ IgE (medium exposure to endotoxin)
Dust mite (Der p 1)↑ IgE (medium exposure to mite allergen)
Roponen et al. 2005 [112]Finland
Cross-sectional
N = 29
Cytokine responseEndotoxin (in settled dust)↑ induced-IL-6
Hagendorens et al. 2004 [115]Belgium
Cohort
N = 22
Lymphocytes (%)T cell cytokine productionDust mite (Der p 1) during second trimester↓ IFN-γ producing induced Th lymphocytes
Peters et al. 2009 [108]USA
Cohort
N = 301
IgEDust mite (Der f 1 + Der p 1)↑ Dust mite-specific IgE
Cockroach (Bla g 1 + Bla g 2)↓ cockroach-specific IgE (indirectly)
Lappalainen et al. 2012 [114]Finland
Cohort
N = 228
Cytokine responseStaphylococcal enterotoxin B↓ LPS-induced IFN-γ
Mycobacterium spp.↓ LPS-induced IL-8
Combined chemical markers↑induced TNF-α
Pets
Heinrich et al. 2002 [107]Germany
Cohort
N = 1332
IgECat-allergen exposure↑ IgE
Roponen et al. 2005 [112]Finland
Cross-sectional
N = 29
Cytokine responseCat/Dog↑ induced IL-6
Aichbhaumik et al. 2008 [105]USA
Cohort
N = 1049
IgECats
Dogs
↓ IgE
Sybilski et al. 2009 [106]Poland
Retrospective study
N = 173
IgECats↑ grass and grain pollen-specific IgE
↑ food-specific IgE
Dogs↓ grass and grain pollen-specific IgE
Lappalainen et al. 2010 [111]Finland
Cohort
N = 228
Cytokine responseDogs↓ induced TNF-α
Uzuner et al. 2013 [113]Turkey
Cross-sectional
N = 62
Cytokine productionPetsNS
Der: Dermatophagoides pteronyssinus; IFN: interferon; Ig: Immunoglobulin; IL: interleukin; LPS: lipopolysaccharide; NS: no significant results PG: peptidoglycan; Treg: T regulatory cells; TNF: tumor necrosis factor.
Table 4. Prenatal exposure to tobacco smoking and changes in the immune system of neonates (authors ordered by year and then alphabetically).
Table 4. Prenatal exposure to tobacco smoking and changes in the immune system of neonates (authors ordered by year and then alphabetically).
Author, Year [Ref.]Location
Study Design
N
Outcomes AssessedTobacco SmokingStatistically Significant Main Findings
Immune Cells FrequencyCytokine Patterns/Ig
Harrison 1979 [116]-
Cross-sectional N = 257
Leukocytes Maternal smoking ↓ neutrophils
Mercelina-Roumans et al. 1996 [117]The Netherlands
Cohort
N = 142
WBC
Leukocytes Reticulocytes
Maternal smoking↓ neutrophils
Noakes et al. 2003 [123]Australia
Cross-sectional
N = 57
Cytokine responseMaternal smoking↑ IL-13 response to HDM and OVA
Noakes et al. 2006 [121]Australia
Cohort
N = 122
Cytokine responseMaternal smoking↓ TNF-α responses via TLR2, 3 and 4
↓ IL-6 responses via TLR2 and 9
↓ IL-10 via TLR2
Pachlopnik Schmid et al. 2007 [118]Switzerland
Cohort
N = 97
Leukocytes
Lymphocytes
Maternal smoking↓ neutrophils
↓ lymphocytes
↓ monocytes
↓ myeloid precursor dendritic cells
Karwowska et al. 2008 [119]Poland
Cohort
N = 79
LymphocytesIgEMaternal smoking↓ T helper (CD4+) cells
↑ IgE
Keski-Nisula et al. 2010 [79]Finland
Cohort
N = 423
Cytokine responseMaternal smoking↓ induced IL-5 response
Pfefferle et al. 2010 [110]5 European countries
(rural areas)
Cohort
N = 625
Cytokine responseMaternal smoking↓ induced-IL-5 levels
Latzin et al. 2011 [124]Switzerland
Cohort
N = 265
Cytokine productionMaternal smoking↓ IL-6 levels
Hinz et al. 2012 [120]Germany
Cohort
N = 346
Treg cells (counts) Maternal smoking↓ Treg cells
Sevgican et al. 2012 [122]USA
Cohort
N = 277
Cytokine response ↓ mitogen-stimulated IFN-γ
CD: cluster of differentiation; HDM: house dust mite; Ig: Immunoglobulin; IL: interleukin; OVA: ovalbumin; TNF: tumor necrosis factor; Treg: T regulatory cells; TLR: Toll-like receptor; WBC: white blood cells.
Table 5. Prenatal exposure to environmental organic pollutants and changes in the immune system of neonates (authors ordered by year and then alphabetically).
Table 5. Prenatal exposure to environmental organic pollutants and changes in the immune system of neonates (authors ordered by year and then alphabetically).
Author, Year [Ref.]Location
Study Design
N
Outcomes AssessedPollutantStatistically Significant Main Findings
Immune Cells FrequencyCytokine Patterns/Ig
Belles-Isles et al. 2002 [125]Canada
Cross-sectional
N = 108
Lymphocytes
(%)
IgM
IgG
Fishing family ↓ Th naïve cells
↓ IgM and ↑ IgG
PCBs and p,p′-DDE
(cord blood)
↑ IgG
Lehmann et al. 2002 [139]Germany
Cohort
N = 85
T cell cytokine responseVOCs↓ induced IFN-γ response (tetrachloroethylene)
↑ induced IFN-γ response (ethylbenzene and m,p-xylene)
↑ induced IL-2 response (hexane, dodecanem and tridecane)
↓ induced IL-2 response (trichloroethylene and tetrachloroethylene)
↑induced IL-4 response (methylcyclopentane, m,p-xylene and naphthalene)
↓ TNF-α response (methylcyclopentane¸ cyclohexane and tetrachoroethylene)
Bilrha et al. 2003 [126]Canada
Cross-sectional
N = 112
LymphocytesCytokine response
IgG
Fishing group↓ PHA-induced TNF-α
Chlorinated pesticides
ΣPCBs (cord blood)
Negative correlations between TNF-α and PCBs, p,p′-DDE, and HCB
Noakes et al. 2006 [121]Australia
Cohort
N = 31
Cytokine responseOrganochlorines
PCBs
NS
Brooks et al. 2007 [128]USA
Cohort
N = 19
Cytokine productionPlacental p,p′-DDE↑ IL-13 levels
↑ IL-4/IFN-γ and ↑ IL-13/IFN-γ ratio
Horváthová et al. 2011 [129]Slovakia Cross-sectional
N = 362
Leukocytes (%)
Lymphocytes (%)
Living in a region with high levels of PCB contamination↓ Dendritic-like cells
↓ Myeloid dendritic cells
↑ Memory T cells
↓ Naive/resting T cells
↓ Suppressor inducer T-cells CD4+CD62L+
↓ Truly naive helper/inducer T-cells CD4+CD62L+CD45RA+
↑Terminally differentiated effector memory T-cells CD4+CD62L−CD45RA+
Horváthová et al. 2011 [130]Slovakia
Cross-sectional
N = 362
Leukocytes
Lymphocytes
Living in a region with high levels of PCB contamination↑ B cells (%)
↑ Activated B cells (%)
↑ Tc cells (%)
↓ NK cells (%)
Neta et al. 2011 [131]USA
Cross-sectional
N = 272
Cytokine productionCis- and trans-Permethrin
(Cord serum)
↑ IL-1β levels
↓ Anti-inflammatory response (+IL-10-IL-12p70)
Oxychlordane
(Cord serum)
↓ IL-1β levels
Trans-nonachlor
Piperonyl butoxide
(Cord serum)
NS
Wang et al. 2011 [132]Taiwan
Cohort
N = 244
IgEPFOA and PFOS
(Cord serum)
↑ IgE (boys)
PFNA
(Cord serum)
NS
Ashley-Martin et al. 2015 [133]Canada
Cohort
N = 1258
TSLP
IL-33
IgE
Phthalates in maternal urine at first trimesterInverse non-linear association between maternal urinary MCPP and IL-33/TSLP levels
Inverse non-linear association between maternal urinary MCPP and IgE levels
BPA, maternal urine 1st trimesterInverse non-linear association between maternal urinary BPA and IL-33/TSLP levels
Ashley-Martin et al. 2015 [134]Canada
Cohort N = 1258
TSLP
IL-33
IgE
Organochlorines maternal plasma at 1st trimester↓ IL-33/TSLP (DDE)
Organophosphates in maternal urine at 1st trimester↓ IL-33/TSLP (DEP + DETP)
PCB, maternal plasma 1st trimester↓ IL-33/TSLP (PCB 118)
Liao et al. 2016 [137]Taiwan
Cohort
N = 275
Cytokine responseBPA↓ TLR3- and TLR4-stimulated TNF-α response
↓ TLR7-8-stimulated IL-6 response
Huang et al. 2017 [138]Taiwan
Cohort
N = 241
Cytokine productionBPA↓ TNF-α levels
Miyashita et al. 2018 [135]Japan
Cohort
N = 268
IgEDioxin-like compounds, maternal blood, 2nd and 3rd trimesters↓ IgE (boys)
Kelley et al. 2019 [136]USA
Cohort
N = 56
Cytokine production12 phthalates in maternal urine, 1st trimester↑ MIP-α (MnBP)
↑ MCP-3 (MEHP)
BPA: bisphenol A; DEP: Diethylphosphate; p,p′-DDE: p,p′-Dichlorodiphenyldichloroethylene; DETP: Diethylthiophosphate; HCB: hexachlorobenzene; IFN: interferon; Ig: Immunoglobulin; IL: interleukin; MCPP: Mono-3-carboxyypropylphthalate; MIP: Macrophage Inflammatory Proteins; MCP: monocyte chemotactic protein; MnBP: mono n-butyl phthalate; MEHP: mono (2-ethylhexyl) phthalate; NK: Natural Killer cells; NS: no statistically significant results; PAH: polycyclic aromatic hydrocarbons; PCB: Polychlorinated biphenyls; PFOA: Perfluorooctanoic Acid; PFOS: perfluorooctane sulfonate; PFNA: perfluorononanoic acid; PHA: Phytohemagglutinin; PM: particle-matter; Tc: T cytotoxic cells; Th: T helper cells; TLR: toll-like receptor; TNF: tumor necrosis factor; TSLP: thymic stromal lymphopoietin; VOCs: volatile organic compounds.
Table 6. Prenatal exposure to metals and metalloids and changes in the immune system in cord blood of neonates (authors ordered by year and then alphabetically).
Table 6. Prenatal exposure to metals and metalloids and changes in the immune system in cord blood of neonates (authors ordered by year and then alphabetically).
Author, Year [Ref.]Location
Study Design
N
Outcomes AssessedMetal or Metalloid AssessedStatistically Significant Main Findings
Immune Cells FrequencyCytokine Patterns/Ig
Belles-Isles et al. 2002 [125]Canada
Cross-sectional
N = 108
Lymphocytes (%)IgM
IgG
Hg↓ Th naïve cells
↓ IgM
Pb↑ IgG
Ahmed et al. 2011 [140]Bangladesh
Cohort
N = 130
Cytokine productionAsU-shaped association with proinflammatory cytokines IL-1β, IL-8, IFN-γ, TNF-α
Nadeau et al. 2014 [141]USA
Cohort
N = 116
Lymphocytes (counts) As↓ naïve activated T cells (CD69+)
↑ Th2 cells (CD69− CD294+)
↑ T cell proliferation
↓ Treg suppressor function
Ashley-Martin et al. 2015 [134]Canada
Cohort
N = 1258
TSLP
IL-33
IgE
Pb in maternal blood at 1st trimester↓ IL-33/TSLP ODDS
Nygaard et al. 2017 [142]USA
Cohort
N = 63
T-lymphocytes As in toenails (8 wks. of post-partum)↓ Th memory cells
↓ activated Th memory cells (boys)
Cd in toenails within 8 weeks of post-partum↓ Th memory cells (girls)
↓ activated Th memory cells
Kelley et al. 2019 [136]USA
Cohort
N = 56
Cytokine productionAs, Ba, Be, Cd, Cr, Cu, Hg, Mn, Mo, Ni, Pb, Se, Sn, Tl, U, W, Zn↑ MIP-α (Mo, Cd, Zn)
↑ MCP-3 (Mo, Ni, Zn)
Kim et al. 2019 [143]Korea
Cohort
N = 331
Cytokine productionPb, Cr↑ IL-13
CD: cluster of differentiation; IFN: interferon; Ig: Immunoglobulin; IL: interleukin; MIP: Macrophage Inflammatory Proteins; MCP: monocyte chemotactic protein; Th: T helper cells; Th2: T helper type 2 cells; TNF: tumor necrosis factor; Treg: T regulatory cells; TSLP: thymic stromal lymphopoietin; wks.: weeks.
Table 7. Prenatal exposure to outdoor air pollutants and changes in the immune system in cord blood of neonates (authors ordered by year and then alphabetically).
Table 7. Prenatal exposure to outdoor air pollutants and changes in the immune system in cord blood of neonates (authors ordered by year and then alphabetically).
Author, Year [Ref.]Location
Study Design
N
Outcomes AssessedPollutantStatistically Significant Main Findings
Immune Cells FrequencyCytokine Patterns/Ig
Hertz-Picciotto et al. 2002 [144]Czech Republic Cross-sectional
N = 518
Lymphocytes (%) Living in a high PM polluted area↑ NK cells
↓ T-lymphocytes
Hertz-Picciotto et al. 2005 [145]Czech Republic Cross-sectional
N = 1397
Lymphocytes (%) PAH 14 days before birth↓ T-lymphocytes
↓ Tc and Th cells
↑ B-lymphocytes
PM2.5 14 days before birth↓ T-lymphocytes and Th cells
↑ B-lymphocytes
Herr et al. 2010 [146]Czech Republic Cohort
N = 1397
Lymphocytes (%) PAHs
PM2.5
1st Trimester:
↑ T-lymphocytes and Th cells
↓ NK cells
3rd Trimester:
↑ B-lymphocytes and NK cells
↓ T-lymphocytes and Th cells
↓ Tc cells (9th month)
Baïz et al. 2011 [147]France
Cohort
N = 370
Lymphocytes (%) NO21st and 2nd Trimester:
↑ Tc cells (CD8+)
3rd Trimester:
↑ NK cells
PM101st Trimester:
↓ T cells
↑ NK cells
1st, 2nd and 3rd Trimester:
↓ CD25+ Th-cells
Benzene↓ CD4+CD25+ T cells
Latzin et al. 2011 [124]Switzerland
Cohort
N = 265
Cytokine productionPM10 last 3 days of pregnancy↓ IL-10
PM10 last 3 mo. of pregnancy↑ IL-1ß
Ashley-Martin et al. 2016 [149]Canada
Cohort
N = 1081
IL-33 and TSLP levelsNO2↑ IL-33 and TSLP (girls)
↑ IL-33/TSLP (3rd trimester, girls)
Whole pregnancy and 1st trimester PM2.5 ↑ IgE (girls)
Lurà et al. 2018 [88]Switzerland
Cohort
N = 295
Leukocytes
Lymphocytes (counts)
NO2 14 days before delivery↓ leukocytes, neutrophils, and monocytes
García-Serna et al. 2020 [148]Spain
Cohort
N = 190
Leukocytes
Lymphocytes (counts)
Traffic-related NO21st Trimester:
↓ leukocytes, lymphocytes, monocytes and basophils
↓ NK and T cells
↓ Th, Th2 and Th17 cells
↓ Tc cells
2nd Trimester:
↑ T and Th cells
3rd Trimester:
↓ Th1Th2-related cells
15 days before delivery:
↓ NK and Th1Th2 cells
Whole pregnancy:
↓ NK cells
Traffic-related PM2.51st Trimester:
↑ Th, Th1 and Treg cells
2nd Trimester:
↓ Th1 and Th1Th2 cells
3rd Trimester:
↓ NK cells
↑ Th17 cells
15 days before delivery:
↑ Th1 cells
Whole pregnancy:
↓ Tc and Th1Th2 cells
Traffic-related PM101st Trimester:
↑ T, Th, Th1 and Th1Th2 cells
2nd Trimester:
↓ Th1, Th17, Th2 and Th1Th2
3rd Trimester:
↑ Th1
15 days before delivery:
↓ Treg cells
Whole pregnancy:
↓ Th1Th2 cells
Traffic-related O31st Trimester:
↑ Th1 cells
2nd Trimester:
↓ Th1Th2 cells
15 days before delivery:
↑ NK cells
Whole pregnancy:
↓ Th17 cells
IL: interleukin; NK: natural killer; PAH: polycyclic aromatic hydrocarbons; PM: particulate matter; Tc: T cytotoxic cells; Th: T helper cells; Th1: T helper type 1 cells; Th2: T helper type 2 cells; Th17: T helper type 17 cells; Treg: T regulatory cells; TSLP: thymic stromal lymphopoietin.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

García-Serna, A.M.; Martín-Orozco, E.; Hernández-Caselles, T.; Morales, E. Prenatal and Perinatal Environmental Influences Shaping the Neonatal Immune System: A Focus on Asthma and Allergy Origins. Int. J. Environ. Res. Public Health 2021, 18, 3962. https://doi.org/10.3390/ijerph18083962

AMA Style

García-Serna AM, Martín-Orozco E, Hernández-Caselles T, Morales E. Prenatal and Perinatal Environmental Influences Shaping the Neonatal Immune System: A Focus on Asthma and Allergy Origins. International Journal of Environmental Research and Public Health. 2021; 18(8):3962. https://doi.org/10.3390/ijerph18083962

Chicago/Turabian Style

García-Serna, Azahara María, Elena Martín-Orozco, Trinidad Hernández-Caselles, and Eva Morales. 2021. "Prenatal and Perinatal Environmental Influences Shaping the Neonatal Immune System: A Focus on Asthma and Allergy Origins" International Journal of Environmental Research and Public Health 18, no. 8: 3962. https://doi.org/10.3390/ijerph18083962

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop