Open Peer Review F1000 Faculty Reviews are written by members of the prestigious  . They areF1000 Faculty commissioned and are peer reviewed before publication to ensure that the final, published version is comprehensive and accessible. The reviewers who approved the final version are listed with their names and affiliations. Any comments on the article can be found at the end of the article. REVIEW Better understanding of childhood asthma, towards primary prevention – are we there yet? Consideration of pertinent  literature [version 1; peer review: 2 approved] Michal Gur ,   Fahed Hakim , Lea Bentur1,2 Pediatric Pulmonary Institute and CF Center, Rappaport Children's Hospital, Rambam Health Care Campus, Haifa, Israel Rappaport Faculty of Medicine, Technion–Israel Institute of Technology, Haifa, Israel Abstract Asthma is a chronic disease, characterized by reversible airway obstruction, airway inflammation and hyper-reactivity. The prevalence of asthma has risen dramatically over the past decade, affecting around 300,000,000 people. The etiology is multifactorial, with genetic, epigenetic, developmental and environmental factors playing a role. A complex interaction between the intrauterine environment, the developing immune system, the infant's microbiome and infectious organisms may lead to the development of allergic sensitization and asthma. Thus, a large number of studies have investigated the risk factors for childhood asthma, with a meticulous search of modifiable factors that could aid in primary prevention. We present a current literature review from 2014-2017, as well as older classic publications, on the pathogenesis and the potential modifiable factors for primary prevention of asthma. No ideal preventive measure has yet been found. Rather, creating favorable prenatal and postnatal environments, minimal exposure to hostile environmental factors, prevention of infections in early life, allergic desensitization and nutritional modifications could possibly reduce asthma inception. In the era of personalized medicine, identifying individual risk factors and tailoring specific preventive measures is warranted. Keywords Asthma, wheezing, environmental, factors, prevention 1 1,2 1,2 1 2    Reviewer Status   Invited Reviewers  version 1 published 20 Dec 2017  1 2 , University of Manitoba, Winnipeg,Allan Becker Canada 1 , Vanderbilt University School ofTina V Hartert Medicine, Nashville, USA , Vanderbilt University SchoolChristian E Lynch of Medicine, Nashville, USA 2  20 Dec 2017,  (F1000 Faculty Rev):2152 (First published: 6 )https://doi.org/10.12688/f1000research.11601.1  20 Dec 2017,  (F1000 Faculty Rev):2152 (Latest published: 6 )https://doi.org/10.12688/f1000research.11601.1 v1 Page 1 of 12 F1000Research 2017, 6(F1000 Faculty Rev):2152 Last updated: 17 JUL 2019 https://f1000research.com/browse/f1000-faculty-reviews http://f1000.com/prime/thefaculty https://f1000research.com/articles/6-2152/v1 https://f1000research.com/articles/6-2152/v1 https://f1000research.com/articles/6-2152/v1 https://orcid.org/0000-0002-9874-7617 https://f1000research.com/articles/6-2152/v1 https://doi.org/10.12688/f1000research.11601.1 https://doi.org/10.12688/f1000research.11601.1 http://crossmark.crossref.org/dialog/?doi=10.12688/f1000research.11601.1&domain=pdf&date_stamp=2017-12-20    Lea Bentur ( )Corresponding author: l_bentur@rambam.health.gov.il  No competing interests were disclosed.Competing interests:  The author(s) declared that no grants were involved in supporting this work.Grant information:  © 2017 Gur M  . This is an open access article distributed under the terms of the  , whichCopyright: et al Creative Commons Attribution Licence permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.  Gur M, Hakim F and Bentur L. How to cite this article: Better understanding of childhood asthma, towards primary prevention – are we  F1000Research 2017,  (F1000 Faculty Rev):2152 (there yet? Consideration of pertinent literature [version 1; peer review: 2 approved] 6 )https://doi.org/10.12688/f1000research.11601.1  20 Dec 2017,  (F1000 Faculty Rev):2152 ( ) First published: 6 https://doi.org/10.12688/f1000research.11601.1 Page 2 of 12 F1000Research 2017, 6(F1000 Faculty Rev):2152 Last updated: 17 JUL 2019 http://creativecommons.org/licenses/by/4.0/ https://doi.org/10.12688/f1000research.11601.1 https://doi.org/10.12688/f1000research.11601.1 Introduction Asthma is a chronic disease, characterized by episodes of reversible airflow obstruction. The prevalence of allergic diseases and asthma has risen substantially over the past decades. Currently, it is the most common non-communicable disease, affecting around 300,000,000 people, especially in developed countries, leading to enormous public health costs1. In a recent study, the total annual health care expenditure attributable to asthma for school-aged children in the United States was 5.92 billion US dollars2. Currently, no available therapeutic regimens can cure asthma, and the burden of asthma will continue to be driven by the increased but, as yet, poorly explained prevalence3. Population-based birth cohorts on asthma and allergies may provide insights into the development and natural history of the diseases. Over 130 birth cohorts have been initiated in the last 30 years4. These birth cohorts have improved our understanding of asthma inception, progressions and persistency. Thus, they may help in targeting the ambitious and important goal of primary pre- vention of asthma. The Tucson birth cohort, developed in 1995 by Martinez et al. is the “classic” and most utilized model. This cohort proposed three categories of wheezing phenotypes at early age: transient wheezers (wheezing symptoms before 3 years, no wheezing at age 6), late onset wheezers (no wheeze until 3 years, wheezing at age 6), and persistent wheeze (wheezing in the first 3 years, wheezing at 6 years). This latter group of persistent wheezers can be divided into non-atopic and atopic. One third of all children aged 3 or younger had lower respiratory tract illnesses with wheezing; however, by the age of 6 close to 60 percent of these children no longer had wheezing symptoms. Transient early wheezers were found to have lower levels of lung function compared to other groups ofwheezers, possibly reflecting congenitally smaller airways5. Understanding the trajectory of early wheezing may help identify early predictors of later childhood persistent asthma. This is of utmost impor- tance in identifying preventive interventions that could potentially reduce the inception of asthma6. The etiology of asthma is multifactorial; genetic, epigenetic, developmental and environmental factors play a role, as do the interactions between them1,3. Two important risk factors have been identified: the development of allergic sensitization and wheezing respiratory tract illnesses at an early age. Since both allergic sensitization and viral/bacterial illnesses occur in children who do not develop asthma, it is crucial to identify genetic and environmental factors that activate, interfere with and direct the immune system toward the development of asthma7. Moreover, it has been found that environmental factors affecting a critical period in lung development (during pre- and postnatal periods) are associated with the development of allergic diseases and asthma. Epigenetic pathways could mediate the gene- environ- ment interactions8 and may explain the impact of external environ- mental factors on disease development. Taking into account the significant burden of asthma, it is crucially important to find preventive measures. Strategies targeting asthma prevention can be primary (e.g. infants at high- risk for asthma) or secondary, dealing with children who have developed allergic sensitization or the first manifestations of allergic diseases (e.g. eczema or wheezing)9. However, the heterogeneity and complex natural history of the disease serve as a barrier for the use of a single prevention strategy and suggest the importance of individualized risk assessment and multiple prevention measures with personalized primary prevention strategies3. This review discusses research from the past 3 years, focusing on childhood asthma and primary preventions. We conducted a PubMed search of observational studies and clinical trials, including systematic reviews and meta-analyses, from 2014 to 2017. We also included older classic publications; thus, the search included studies dealing with potentially modifiable factors (both prenatal and postnatal) implicated in the inception of asthma. Environmental factors, early life respiratory infections, host factors and nutritional interventions will also be discussed. Environmental factors Favorable environment The “farm effect”. The importance of favorable environmen- tal exposures in the development of asthma was demonstrated by epidemiologic studies showing significant protection from asthma and allergic diseases in children raised on traditional dairy farms. In particular, it was found that children who lived on farms were exposed to a greater variety of environmental microorgan- isms than controls. Endotoxin, a cell wall component of Gram negative bacteria, along with peptidoglycan, is widespread in stables and other farming environments and exposure to such microbial compounds was found to be inversely correlated with the risk of atopy10. Higher diversity of microorganism expo- sure was correlated with a reduced prevalence of asthma and atopy11. Compelling data exist showing that the “farm effect”12 is not only present in childhood but also exerts an effect during pregnancy, influencing the developing fetal immune system. The farming environment has been linked with lower rates of asthma in offspring, although early-life farm exposure was shown to boost this effect13. A recent study with the Amish community using tra- ditional farming and the Hutterite community using industrialized farming evaluated asthma prevalence in children with similar genetic ancestries and lifestyles14. Asthma prevalence and allergic sensitization was 4 and 6 times lower in the Amish community, whereas median endotoxin levels in Amish house dust was 6.8 times higher. The authors used a murine model of experimen- tal allergic asthma to assess the effects of dust extracts found in Amish and Hutterite households on immune and airway responses. Experiments in humans and mice indicated that the traditional farm environment guards against asthma via interaction with and molding of the innate immune response. Hence, it is evident that altering innate immune signaling, as by farm environment, may be the primary target of asthma protection. However, the inter- ventions required to create a farm effect without living on a farm are as yet unknown. Pets. Several prospective birth cohort studies found a decreased prevalence of atopic disease in children having daily contact with pets, in particular cats and dogs, during early infancy10. Exposure to two or more dogs or cats in the first year of life was associated with a significantly lower risk of atopy (adjusted OR 0.23, 95% CI 0.09-0.60)15. Another study found that living with Page 3 of 12 F1000Research 2017, 6(F1000 Faculty Rev):2152 Last updated: 17 JUL 2019 a cat was inversely related both to having a positive skin test to cat (RR, 0.62 [0.47–0.83]) and incidence of physician-diagnosis of asthma (RR, 0.49 [0.28–0.83]). The effect was most pronounced among the children with a family history of asthma. Ownership of a dog resulted in weaker protective trends16. A pooled analysis of individual participant data of 11 prospective European birth cohorts of 22,000 children concluded that pet ownership in early life did not appear to either increase or reduce the risk of asthma17. However, a large nationwide cohort study (1,011,051 children) in Sweden found that dog exposure during the first year of life was associated with a decreased risk of asthma in school-aged children (OR 0.87, 95% CI 0.81-0.93) and in children 3 years or older (HR 0.90, 95% CI 0.83-0.99). Exposure to farm animals was correlated with a lowered risk of asthma in both children of school and preschool age. These results were independent of parental asthma18. However, the effect of exposure to dogs and farm animals was less pronounced in children younger than 3 years of age. These young children may reflect a group of children with transient wheeze and not persistent asthma5. Taken together, most (but not all) studies support the theory of protective effects of having pets on asthma development. Day care attendance. The effect of day care attendance on asthma development is complex. In a study involving 1,035 children fol- lowed since birth as part of the Tucson study, children with older siblings or day care attendance were more likely to have frequent wheezing at the age of 2 years, but less likely to have frequent wheezing from the age of 6 to 13 years19. Different results were obtained in a birth cohort of 762 children20. In this study, day care attendance at 12 months was associated with an increased risk of asthma [OR 1.8, 95% CI 1.1-3.0]. A multivariate logis- tic model showed that day care attendance and number of lower respiratory infections at 12 months were associated with asthma [OR 1.2 (1.1-1.5); OR 1.4 (1.2-1.7), respectively]. Never- theless, day care attendance of greater than 37.5 hours per week was associated with a lower risk of asthma [OR 0.6 (0.4-0.9)]. Hence, day care during infancy can either increase or reduce the risk of asthma. Prevention of allergic sensitization There is a clear association between asthma and allergy21. Most school age children and adolescents with asthma also have allergic sensitization. Early allergic sensitization is a risk factor for asthma development. In children of preschool age, researchers have found a sequential relationship of allergic sensitization followed by virus-induced wheezing. Exposure to allergens may increase asthma severity in susceptible patients (for example, an asthmatic patient exposed to dust mite allergens may develop increased airway hyper-reactivity)22. Therefore, prevention of allergic sensitization is a major path for primary prevention of asthma. A Cochrane review in 2009 found that multifaceted interven- tions (reducing exposure to both inhalant and food allergens) resulted in a significant decrease in asthma compared to usual care (<5 years: OR 0.72, 95% CI 0.54 to 0.96; >5 years: OR 0.52, 95% CI 0.32 to 0.85). Monofaceted interventions (reducing exposure to either inhalant or food allergens) did not produce significant effects23. Another interventional study assessed the effectiveness of a multifaceted intervention program for the primary prevention of asthma in high-risk infants. The interventions were initiated shortly before birth and applied until 1 year of age, and included avoidance of house dust mites, pets, environmental tobacco smoke, encouragement of breast-feeding, and delayed introduc- tion of solids. These measures resulted in a 4-fold reduction in the risk of asthma (adjusted OR, 0.26; 95% CI 0.08-0.88) in the children who did not develop atopy by 1 year of age; however, in children with early, persistent atopy, the risk of asthma was not reduced24. Moreover, attempts to reduce exposure to house dust mites, known to be strongly associated with atopic sensitization and the development of asthma in childhood, were not found to be effective in the primary prevention of disease3. Lynch et al. examined a birth cohort and a nested case-control study to assess the factors linking contact with allergens and bacteria in the first 3 years of life with the inception of recurrent wheeze and atopy. As the authors put forward, increasing allergen expo- sure in the first 3 years correlated with allergic sensitization, and allergic sensitization correlated with recurrent wheeze. However, there was a negative association between contact with cockroach, mouse and cat allergens in the first year and recurrent wheeze (OR 0.60, 0.65, and 0.75, respectively, p≤0.01)25. Overall, the reported nature of the relationship between exposure and sensitization varies widely among studies, from no signifi- cant association to a simple linear dose–response relationship or a ‘bell-shaped’ dose–response model with a protective effect of high allergen exposure. A recent study assessed the potential role of immunotherapy to alter the natural course of allergic march from allergic sensitization to asthma. The study included 812 children (5–12 years), with grass pollen allergic rhinocon- junctivitis and no medical history or signs of asthma. Children were double blinded and randomly allocated to receive immu- notherapy or placebo for three years and were followed for two additional years26. The study showed reduced risk of experienc- ing asthma symptoms and using asthma medication, but did not show an effect on the time to onset of asthma. Hence, the relationship of atopic desensitization and asthma is complex and is likely determined by the type of allergen, the timing, pattern, route of exposure (inhaled, oral, transcutane- ous), dose, as well as other environmental factors and individual genetic predispositions21. Thus, the role and the measures of prevention of sensitization in the development of IgE-mediated sensitization asthma are yet to be determined. Hostile environment Air pollution. Air pollutants, representing a complex exposure to inorganic and organic components, are known to exacerbate asthma symptoms and might play a role in initiation of this disease. Particulate matter (PM) carries both environmental pollutants, such as polycyclic aromatic hydrocarbons (PAHs)— formed during incomplete combustion of fossil fuels and oil products—as well as agents causing immune stimulation—such as pollens, endotoxin and fungal spores. Air pollutants probably Page 4 of 12 F1000Research 2017, 6(F1000 Faculty Rev):2152 Last updated: 17 JUL 2019 cause oxidative injury to the airways, leading to inflammation, remodeling, and an increased risk of sensitization. The idea that air pollution can cause exacerbations of pre-existing asthma is supported by evidence-based studies, but evidence suggests that air pollution might cause new-onset asthma as well, both in chil- dren and in adults27. In a recent epidemiologic review, Schulz et al. concluded that early life and school-age exposures to air pollution has a negative impact on lung function, at least up to adolescence28. In a Canadian birth cohort study, postnatal exposure to traffic-related air pollution increased the risk for the development of atopy to any allergens (adjusted OR 1.16; 95% CI 1.00-1.41)29. Gauderman et al. found an association between improvements in air quality in southern California and measurable improve- ments in lung-function development in children30. Consequences of exposure to pollutants, pre- or postnatal, represent a complex interaction between the environment, the host and epigenetic factors. In children, PAH exposure has been associated with changes in DNA methylation, as well as impaired function of regulatory T cells31. Epigenetic processes translate environmen- tal exposures into regulation of the identity, gene expression profile, and activity of specific cell types that participate in the pathophysiology of the disease. Further discussion of the effects of air pollution is beyond the scope of this review. Tobacco smoke. There is evidence indicating a consistent det- rimental effect of prenatal exposure and postnatal environmen- tal smoking on childhood wheezing illnesses. Environmental tobacco smoke (ETS) during critical periods of lung development (prenatally, i.e. during pregnancy, and during early life) is consid- ered a substantial risk factor for childhood allergic diseases. ETS induces over-expression of Toll-like receptors on the surface of the airway epithelium, increases oxidative stress and activates dendritic and innate lymphoid cells through the production of cytokines, such as IL-1, IL-25 and IL-33. The result is a higher susceptibility to allergen sensitization and a further risk for asthma32. Genetic polymorphism on chromosome 17q21 was found to be predictive of childhood-onset asthma, and the risk was further increased by early-life exposure to environmental tobacco smoke33,34. In a meta-analysis, ETS was associated with an increased risk of elevated specific IgE (OR 1.12; 95% CI 1.00-1.25) and posi- tive skin prick test (OR 1.15; 95% CI 1.04-1.28). The relationship was stronger in young children and in prospective studies35. In another meta-analysis of 79 prospective studies, ETS was found to increase the risk of wheeze by age 2 years by 70%; prenatal smoking was related to a 40% increased risk. The risk of asthma decreased with age (30% at age 3–4 years, 23% at 5–18 years)36. In a cohort study of 27,993 mother–child pairs, children of moth- ers exposed to passive smoking whilst pregnant but no other smoking exposure had an increased tendency to develop wheeze up to 2 years old (OR 1.11; 95% CI 1.03–1.20) in comparison with control pairs. Exposure to passive smoke postnally, in addi- tion to their mothers’ prenatal passive exposure, further increased the risk (OR 1.29; 95% CI 1.19–1.40). The risk was highest with passive neonatal exposure in addition to prenatal active smoking (OR 1.73; 95% CI 1.59–1.88)37. Thus, ETS was found to be an important but avoidable risk factor for the development of allergic disease in children35. Smoking is a modifiable risk factor in asthma. There is definitely a need for robust measures to reduce prenatal and postnatal smoking as a strategy for primary prevention of asthma. Early life respiratory infections Infections Childhood asthma and infant respiratory viral infection are the most frequent chronic and acute illnesses of childhood, respec- tively. Over the years, it has been possible to make links between these diseases and spot common clinical traits. Early life respira- tory syncytial virus (RSV) and human rhinovirus (HRV) lower respiratory tract infections (LRTIs) have been found to be strongly associated with increased asthma risk. During early infancy, RSV is a more common cause of severe LRTI. With advancing age, the situation is reversed, with HRV becoming more common. In spite of continuous research, the role of these respiratory viruses in the inception of asthma is still under debate38. Respiratory syncytial virus (RSV). Severe RSV bronchioli- tis requiring hospitalization is considered a risk factor for future asthma. In the RSV Bronchiolitis in Early Life Study, 50% of patients hospitalized with RSV had a physician diagnosis of asthma at age 7 years39. Another study found that 21% of infants hospital- ized for RSV bronchiolitis had asthma at age 6 years, compared to 5% in controls40. There is a direct relationship between the severity of the initial RSV bronchiolitis and the risk of subsequent asthma, while environmental factors may further augment the risk41. Thus, the association between early life RSV LTRI and later wheezing is consistent across most studies with large effect sizes and severity dose-response relationship. The pathway from early-life infections with RSV to asthma is the result of complex interactions between the specific type of the virus, genetic, and environmental factors. RSV induces persistent airway damage and bronchial hyper-responsiveness42. It has been suggested that RSV infection affects Th1/Th2 balance in early childhood, thereby inducing an atopic state and may, therefore, be involved in the inception of asthma43. There is data that suggest that in utero exposure to RSV is followed by dysregulation of neurotrophic pathways, predisposing to postnatal airway hyperreactivity upon reinfection with the virus44. Palivizumab (monoclonal antibody) prophylaxis in the first year of life reduces recurrent wheezing in children aged 1 to 3 years40. Palivizumab resulted in a relative reduction of 61% (95% CI 56-65) in the total number of wheezing days during the first year of life45. However, the substantial cost, the need to treat a large population early in life (before RSV infection), and the need for parenteral administration limit its widespread use41. While there is evidence that palivizumab prophylaxis reduces wheezing, its long-term impact on the development of atopic asthma remains controversial. In a follow-up study of children aged 2 to 5 years, palivizumab administration resulted in a 68% reduction in wheezing in the families of non-asthmatics and an 80% reduction in wheezing in non-atopic families, but no pro- tection was achieved in atopic families46. Similarly, in a recent Page 5 of 12 F1000Research 2017, 6(F1000 Faculty Rev):2152 Last updated: 17 JUL 2019 prospective multicenter observational cohort study of 444 pre- term infants, palivizumab prophylaxis administration significantly reduced subsequent physician diagnosis of recurrent wheezing up to 6 years, but did not reduce the incidence of atopic asthma, casting doubt on the suggestion that RSV prevention may decrease atopic asthma inception47. Carroll et al. looked at infants at high-risk for severe RSV and whether a link could be found between better adherence to immunoprophylaxis and decreased childhood asthma. Analysis revealed that 70% or greater adher- ence decreased the odds of asthma compared to those with 20% or less adherence (OR 0.62; 95% CI 0.50-0.78)48. Recently, in two prospective birth cohort studies, HRV infection was significantly more common among infants administered with RSV immuno- prophylaxis (OR, 1.65; 95% CI 1.65-2.39)49. Taken together, the impact of palivizumab on the development of atopic asthma after RSV infection is still controversial. A definitive large-scale randomized clinical trial (RCT) measur- ing the effect of the prevention of RSV on childhood asthma is yet to be done. A large study evaluated 607 children aged 12 to 71 months at the beginning of a LRTI. Administration of azithromycin significantly reduced the odds of progres- sion to severe LRTI (HR 0.64; 95% CI 0.41-0.98, p=0.04)50. A relatively small double-blind placebo-controlled trial found a protective effect of azithromycin therapy during RSV bronchi- olitis on subsequent recurrent wheeze51. Currently, a larger study assessing asthma prevention following RSV by azithromycin is being conducted (NCT02911935). Other strategies to prevent RSV are under investigation. There are some efforts to develop a specific vaccination against RSV. Recently, a genetically engineered, live attenuated vaccine was found to be effective in a phase I study52. Another innovative approach includes immunization of pregnant women that potentially will decrease airway hyperreactivity upon postnatal reinfection with the virus53. In conclusion, although there is a clear association between RSV and asthma, the role of strategies to prevent RSV (palivizu- mab, azithromycin, vaccine) in asthma prevention has yet to be confirmed. Human rhinovirus (HRV). HRV has been increasingly recognized as an etiologic factor in preschool wheeze, as well as a significant risk factor in the development of asthma41. Episodes of wheezing during which HRV was found in the upper airways have been found to be a strong predictor of subsequent asthma54. HRV wheezing illnesses were found to increase the risk of asthma by 10-fold at 6 years of age55. Lee et al. analyzed 1,445 samples collected from 209 infants enrolled in the COAST (Childhood Origins of ASThma) cohort study; these infants had an increased risk for developing allergies and asthma. HRV species A and C were about 7 times more likely to cause moder- ate to severe illness. These results firmly indicate that antiviral therapies aiming at lowering HRV-related morbidity in those infants at high risk ought to be directed towards HRV-A and HRV-C species. As with RSV, the mechanism by which severe early-life HRV causes future asthma remains uncertain. Postulated mechanisms are airway epithelial injury and/or creating the appropriate pro- inflammatory allergenic milieu; alternatively, wheezing viral LRTI may serve as a marker for asthma susceptibility. Several studies suggest that the initial wheezing HRV LRTI may serve as a marker for asthma tendency, while early-life severe RSV bronchiolitis may have a causative role in the development of asthma41. Thus, preventive measures are aimed at decreasing the incidence and severity of infections caused by HRV. A recent study56 identified day-care attendance (OR 5.0; 95% CI 2.3-10.6), high eosinophil blood counts (OR 2.6; 95% CI 1.2-5.7) and expo- sure to tobacco smoke (OR 2.5; 95% CI 1.1-15.6) as significant risk factors for HRV LRTI. Hence, restricting children’s exposure to tobacco smoke may limit dissemination of viruses to younger children, counteract severe respiratory diseases, and thus may reduce sequelae. An RCT assessed the efficacy of oral corticosteroid treatment during the first HRV LRTI to reduce the frequency of subsequent wheezing within 12 months. The study failed to show reduction in post HRV wheezing; however, children with a high viral load treated with prednisolone had a longer time to the next wheezing episode compared to placebo57. Strategies for developing an effective vaccine58 or for preventing viral contact and invasion by forming a barrier on the host mucosa59 are being developed; however, currently there is no approved strategy against HRV. Limiting viral spread is the main available protective measure. Host factors The microbiome The whole host-microbe system benefits from the essential ecosystem services provided by microbial communities (micro- biota) and the host environment in which they reside. Such services include making vital resources, nutrient bioconver- sion, and guarding against harmful microbes. Disease can arise from a shortfall in these beneficial functions or the maladaptive functions introduced by pathogenic microbes. With the advent of 16s rRNA sequencing, a strong associa- tion between the host microbiome and asthma has emerged. The microbiome can be modulated by various environmental fac- tors, including diet, prebiotic and probiotic, and early-life microbial exposures. Recently, the nasopharyngeal microbiome of 33 healthy infants was compared to 99 infants with confirmed RSV. The abundance of the dominant genera was significantly different between the groups, suggesting that RSV alters the infant nasopharyngeal microbiome and, thereby, may contribute to asthma development60. Affecting the microbial dysbiosis was suggested as a target for the prevention of asthma61. Herein, we will discuss the potential role of a prebiotic and probiotic diet in the prenatal and postnatal period. Page 6 of 12 F1000Research 2017, 6(F1000 Faculty Rev):2152 Last updated: 17 JUL 2019 Prenatal. Prenatal prebiotic and probiotic supplementation may alter maternal gut bacteria and influence maternal immune function. There is an inconsistent effect on the offspring of mothers supplemented during pregnancy, and a comprehensive 2016 World Allergy Organization review found a lack of evidence to support the use of prebiotics during pregnancy62. Maternal urinary tract infection during pregnancy increased the likelihood of asthma (OR 1.21; 95% CI 1.2-1.220)3. Several studies have reported an association between the use of over- the-counter antipyretics during pregnancy or infancy and an increased risk (OR 1.26; 95% CI 1.02-1.58) of asthma in early childhood, but not mid-childhood63. In a recent review, the maternal use of antibiotics or paracetamol during pregnancy was suggested as a modifiable risk factor for childhood asthma64. Postnatal. Microbial gut colonization typically starts at the time of birth, and is influenced by the bacterial load of maternal microbiota, type of delivery (cesarean section vs. vaginal delivery), feeding (formula vs. breast-feeding), and the use of antibiotics. In a retrospective cohort study including 321,287 births, there was no difference in the risk of asthma following planned com- pared to unscheduled cesarean surgery. However, compared to vagi- nal delivery, planned cesarean delivery resulted in a small increase in the risk of asthma requiring hospital admission (adjusted HR 1.22; 95% CI 1.11-1.34) and salbutamol inhaler prescription at age 5 years (adjusted HR 1.13; 95% CI 1.01-1.26)65. In another study, cesarean section increased the risk of childhood asthma by 34% in univariate analysis and 11% after adjusting for other environmental exposures and covariates66. Infants born via cesarean section have higher rates of Clostridum, Klebsiella, Bacteroides, and several other species at age 1 to 6 months, which predispose them to a higher risk for asthma, atopy and allergic rhinitis67. In the newborn period, the gut microbiota plays a crucial role in maintaining the structure and function of the mucosal immune system. Gut-associated mucosal lymphoid tissue becomes reac- tive to pathogenic bacteria but tolerant to “beneficial” bacteria. T regulatory cells (Treg) are the key players in immunological tolerance; changes in their number or function are associated with the development of allergy67. In a recent systematic review, the early microbiota of children who later developed allergies showed lower bacterial diversity; moreover, a predominance of Firmicutes, a higher prevalence of Escherichia coli, Clostridium difficile, and a lower prevalence of Lactobacillus were found68. In the Canadian Healthy Infant Longitudinal Development (CHILD) Study, Arrieta et al. evaluated the gut microbiota of 319 individuals. They found that those at risk of asthma showed transient gut microbial dysbiosis in their first 100 days of life, with reduced levels of Faecalibacterium, Veillonella, Lachnospira, and Rothia. Moreover, when these four bacterial taxa were introduced to germ-free mice, they showed decreased airway inflammation, suggesting a causal role of these bacteria in asthma development69. Several animal and human studies have examined the role of the postnatal use of prebiotics and probiotics in the prevention of allergic diseases. A meta-analysis reported data from 4,755 children (2,381 in the probiotic group, 2,374 controls). Infants treated with probiotics had a significantly lower risk of eczema (RR 0.78; 95% CI 0.69-0.89). No significant difference in terms of prevention of asthma (RR 0.99; 95% CI 0.77-1.27) or wheezing (RR 1.02; 95% CI 0.89-1.17) was found. Another meta-analysis concluded that controlled studies have not yielded sufficient evidence to date to recommend prebiotics and probiotics for the primary prevention of asthma70. Perinatal and postnatal suggested modifiable behaviors were natural childbirth, breastfeeding, increased outdoor activities, diet and the judicious use of antibiotics and antipyretics. These measures may help restore the neonatal microbiome and may reduce the risk for allergic diseases71. Nutritional prevention Dietary interventions during pregnancy are attractive because they are inexpensive and follow the accepted practice of folic acid supplementation3. Several nutritional supplements have been investigated; we will discuss the main recent findings. Current evidence suggests that maternal diet during pregnancy influences the developing immune system of the fetus. Interest- ingly, maternal weight gain or obesity during pregnancy was found not only to increase maternal asthma exacerbations72 but also the risk for childhood asthma64. Thus, efforts to prevent childhood asthma focus on early prenatal and postnatal interventions. The Mediterranean diet (MD) comprises fruit, vegetables and grains, a moderate intake of dairy products and a low intake of meat. This diet has been suggested to have a potential protective role in asthma. In a recent pilot study involving 30 pregnant women, the introduction of MD was feasible and acceptable, with 93% of participants retaining the diet73. In a meta-analysis, MD was associated with a lower prevalence of “asthma ever” (OR -0.86; 95% CI 0.74–1.01), as well as “current wheeze” (OR -0.85; 95% CI 0.75–0.98) and “current severe wheeze” (OR -0.66; 95% CI 0.48–0.90)74. Thus, although MD has been suggested to be beneficial to general health, its impact on asthma prevention is still not proven and requires a large epidemiological study; assessing the mechanism, the relevant window of exposure and addressing specific components of the diet is warranted. Vitamins Most studies focus on vitamin D supplementation and the results are inconclusive. Vitamin D has the ability to regulate inflam- mation and modulate immune responses and cell growth. Experimental data suggest that vitamin D may affect the devel- oping lung and immune system during the prenatal and postnatal periods. Additionally, observational studies have suggested an association between maternal intake of vitamin D, cord blood vitamin D levels and persistent and recurrent wheezing in early childhood75. In an RCT, 623 pregnant Danish women receiving 400 IU/d of vitamin D during the third trimester of pregnancy were randomized to receive an additional 2400 IU/d or placebo. Follow-up of the children (N=581) was completed when the Page 7 of 12 F1000Research 2017, 6(F1000 Faculty Rev):2152 Last updated: 17 JUL 2019 youngest child reached age 3 years. Persistent wheeze was diag- nosed in similar rates (16% and 20%) of children whose moth- ers received supplemental vitamin D and placebo, respectively (hazard ratio 0.76; 95% CI 0.52-1.12; p=0.16). The results suggest that 2800 IU/d during the third trimester of pregnancy cannot reduce the risk of persistent wheeze in the offspring through age 3 years. In the Vitamin D Antenatal Asthma Reduction Trial (VDAART) RCT, 881 pregnant women at risk of having children with asthma were randomized to 4,000 international units (IU)/d vitamin D or placebo plus 400 IU/d of vitamin D. Supplementation with 4400 IU/d resulted in a 20% reduction of recurrent wheeze or asthma (hazard ratio 0.8; 95% CI 0.6-1) that did not reach statistical significance (p=0.051)76. A secondary analysis of the data revealed that the largest protective effect was found in women with higher initial vitamin D levels who were randomized to the intervention group (adjusted OR 0.13; 95% CI 0.02-0.99)77. This suggests that higher vitamin D levels in early pregnancy may be required for asthma/recurrent wheeze prevention in early life. A meta-analysis reporting data from 2,456 children demon- strated that prenatal supplementation of vitamin D significantly decreased recurrent wheeze (RR 0.812; 95% CI 0.67-0.98). Postnatal vitamin D administration was found to be associated with a reduction in upper respiratory tract infections78; however, its role in primary asthma prevention remains unclear. Thus, a large randomized double-blind controlled study is currently being conducted to assess the role of postnatal vitamin D supplementa- tions on multiple end points, including allergy, atopy and asthma later in life (NCT01723852)79. Vitamin C and E studies have not undergone meta-analysis due to high heterogeneity; these vitamins did not appear to have a signifi- cant preventive effect on recurrent wheeze80. To conclude, currently there is no sufficient evidence regarding any of the vitamins in the inception and prevention of asthma. Larger double-blind studies are required to recommend their routine use. Antioxidants and fish oil Antioxidants reduce reactive oxygen species and there are several reports about the inverse associations between intake of antioxidants and allergic diseases. Total antioxidant capacity (TAC) assesses the combined activity of all the dietary antioxidants. A study in 2,359 Swedish children found that higher dietary TAC was inversely associated with the sensitization to aeroallergens (OR 0.73; 95% CI 0.55-0.97) and the risk of allergic asthma (OR 0.57; 95% CI 0.34-0.94). Interestingly, the relationship was modi- fied by exposure to air pollution. A stronger inverse association between dietary TAC and allergy was observed in children with low exposure to air pollution. The authors postulated that high TAC may not be enough to counteract the high oxidative stress caused by air pollution81. More than a decade ago, Hodge et al. examined the relation between certain food consumption and asthma. They found a significantly reduced risk of current asthma in children who ate fresh, oily fish (OR 0.26; 95% CI 0.09-0.72; p<0.01)82. Later on, observational studies suggested an association between low levels of dietary fish-oil derived fatty acids (FA) and the risk of asthma and wheezing. In an RCT, 736 pregnant women were assigned to receive 2.4 gr of fish oil or placebo (olive oil). In the treatment group, there was a 30.7% reduction in the risk of persistent wheeze or asthma. The effect was most prominent in the women with low FA levels at randomization; moreover, the effect of the intervention remained at age 5 years, suggesting that it is not restricted to the “transient wheezers”83. Taken together, dietary interventions may aid in the primary prevention of asthma. Antioxidants, MD and fish oil seem to have a beneficial effect. Vitamin D is associated with asthma, but evidence for its role in primary prevention is still lacking. Other vitamins studied (such as C and E) failed to show a beneficial effect. Stress Stress has been suggested to modify the normal lung morpho- genesis and maturation during pregnancy and the postnatal period. Stress may affect neuroendocrine, autonomic and immune function programming, thereby leading to increased asthma inception. Prenatal maternal stress was found to alter innate and adaptive immune response in cord blood mononuclear cells, suggesting that prenatal stress may impact the expression of allergic diseases and increase the risk for later childhood wheezing84,85. Postnatally, maternal behavior was found to influence the development of hypothalamic-pituitary-adrenal (HPA) responses to stress in rodents, mediated by changes in glucocorticoid receptor (GR) expression86. Dreger et al. found that exposure to maternal distress restricted to the first year of life resulted in a 40% increase in cortisol levels in children; beyond the postna- tal period, response to stress differed according to the presence of asthma87. A recent study in a high-risk birth cohort found that maternal stress at age 2 and 3 years and maternal depression at any age were positively associated with recurrent wheeze (p<0.05 and p≤0.01, respectively)88. Moreover, both active and passive stressors in asthmatic patients were associated with an increased activation of the sympathetic nervous system89. Taken together, the data suggest that prenatal as well as postnatal maternal distress may contribute to asthma development in children; thus, lowering early life stress may help decrease asthma. Conclusions Asthma is a chronic inflammatory disease, and genetic, infectious, nutritional and environmental factors play a role in its pathogen- esis. In recent years, there has been some advance in the concept of primary prevention of asthma. However, there is no consensus on the relative importance of risk factors associated with asthma inception and none of the primary prevention or intervention strate- gies investigated has provided sufficient evidence to lead to wide- spread implementation in clinical practice. Current findings suggest that the major preventing measures during pregnancy are avoiding of passive and active smoking and the possible modification of maternal microbiome (e.g. lifestyle, diet, nutritional supplements). Avoiding unnecessary caesarean delivery is the main perinatal Page 8 of 12 F1000Research 2017, 6(F1000 Faculty Rev):2152 Last updated: 17 JUL 2019 measure that may affect asthma. Postnatally, the most important measures are preventing severe neonatal respiratory infection, increasing favorable environment and behaviors (e.g. mimicking farm residence, breastfeeding) and decreasing hostile environ- ments (e.g. smoking and air pollution). Unfortunately, gaps in knowledge still exist. The exact immune pathways that predis- pose certain infants (and not others) to asthma following early life viral infections are not fully understood. Additionally, although there is a clear association between allergy and asthma, its role in the primary prevention of asthma is under debate. Moreover, none of the suggested therapies or interventions can serve as a sole solution for the prevention of asthma inception. Instead, the combination of several pre- and postnatal factors, such as creat- ing a favorable environment with minimal exposure to a hostile environment, attempts to beneficially affect the maternal and infant microbiome, with prevention of infections in early life, is expected to be more effective. This should be achieved by extensive educational and public health efforts to reduce tobacco smoking and air pollution, to implement dietary interventions in pregnant women, and to encourage breastfeeding and childhood vaccinations. Future research should focus on the prevention of RSV and HRV, possibly by vaccine development. Moreover, in the era of person- alized medicine, a test that would recognize the specific asthma phenotype and endotype of each patient, as well as his or her own risk factors, would enable tailoring specific preventive measures for the individual patient. Competing interests The authors declared that they have no competing interests. Grant information The author(s) declared that no grants were involved in supporting this work. References F1000 recommended 1. Jackson DJ, Hartert TV, Martinez FD, et al.: Asthma: NHLBI Workshop on the Primary Prevention of Chronic Lung Diseases. Ann Am Thorac Soc. 2014; 11(Suppl 3): S139–45. PubMed Abstract | Publisher Full Text | Free Full Text 2. Sullivan PW, Ghushchyan V, Navaratnam P, et al.: The national cost of asthma among school-aged children in the United States. Ann Allergy Asthma Immunol. 2017; 119(3): 246–252.e1. PubMed Abstract | Publisher Full Text | F1000 Recommendation 3. Beasley R, Semprini A, Mitchell EA: Risk factors for asthma: is prevention possible? Lancet. 2015; 386(9998): 1075–85. PubMed Abstract | Publisher Full Text 4. Bousquet J, Gern JE, Martinez FD, et al.: Birth cohorts in asthma and allergic diseases: report of a NIAID/NHLBI/MeDALL joint workshop. J Allergy Clin Immunol. 2014; 133(6): 1535–46. PubMed Abstract | Publisher Full Text | Free Full Text 5. Martinez FD, Wright AL, Taussig LM, et al.: Asthma and wheezing in the first six years of life. The Group Health Medical Associates. N Engl J Med. 1995; 332(3): 133–8. PubMed Abstract | Publisher Full Text 6. Panico L, Stuart B, Bartley M, et al.: Asthma trajectories in early childhood: identifying modifiable factors. PLoS One. 2014; 9(11): e111922. PubMed Abstract | Publisher Full Text | Free Full Text 7. Jackson DJ, Gern JE, Lemanske RF Jr: The contributions of allergic sensitization and respiratory pathogens to asthma inception. J Allergy Clin Immunol. 2016; 137(3): 659–65;quiz 666. PubMed Abstract | Publisher Full Text | Free Full Text 8. Potaczek DP, Harb H, Michel S, et al.: Epigenetics and allergy: from basic mechanisms to clinical applications. Epigenomics. 2017; 9(4): 539–71. PubMed Abstract | Publisher Full Text | F1000 Recommendation 9. Elenius V, Jartti T: Vaccines: could asthma in young children be a preventable disease? Pediatr Allergy Immunol. 2016; 27(7): 682–6. PubMed Abstract | Publisher Full Text 10. von Mutius E, Schmid S: The PASTURE project: EU support for the improvement of knowledge about risk factors and preventive factors for atopy in Europe. Allergy. 2006; 61(4): 407–13. PubMed Abstract | Publisher Full Text 11. Ege MJ, Mayer M, Normand AC, et al.: Exposure to environmental microorganisms and childhood asthma. N Engl J Med. 2011; 364(8): 701–9. PubMed Abstract | Publisher Full Text | F1000 Recommendation 12. Wlasiuk G, Vercelli D: The farm effect, or: when, what and how a farming environment protects from asthma and allergic disease. Curr Opin Allergy Clin Immunol. 2012; 12(5): 461–6. PubMed Abstract | Publisher Full Text 13. Douwes J, Cheng S, Travier N, et al.: Farm exposure in utero may protect against asthma, hay fever and eczema. Eur Respir J. 2008; 32(3): 603–11. PubMed Abstract | Publisher Full Text 14. Stein MM, Hrusch CL, Gozdz J, et al.: Innate Immunity and Asthma Risk in Amish and Hutterite Farm Children. N Engl J Med. 2016; 375(5): 411–21. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation 15. Ownby DR, Johnson CC, Peterson EL: Exposure to dogs and cats in the first year of life and risk of allergic sensitization at 6 to 7 years of age. JAMA. 2002; 288(8): 963–72. PubMed Abstract | Publisher Full Text | F1000 Recommendation 16. Perzanowski MS, Rönmark E, Platts-Mills TA, et al.: Effect of cat and dog ownership on sensitization and development of asthma among preteenage children. Am J Respir Crit Care Med. 2002; 166(5): 696–702. PubMed Abstract | Publisher Full Text 17. Lødrup Carlsen KC, Roll S, Carlsen KH, et al.: Does pet ownership in infancy lead to asthma or allergy at school age? Pooled analysis of individual participant data from 11 European birth cohorts. PLoS One. 2012; 7(8): e43214. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation 18. Fall T, Lundholm C, Örtqvist AK, et al.: Early Exposure to Dogs and Farm Animals and the Risk of Childhood Asthma. JAMA Pediatr. 2015; 169(11): e153219. PubMed Abstract | Publisher Full Text | F1000 Recommendation 19. Ball TM, Castro-Rodriguez JA, Griffith KA, et al.: Siblings, day-care attendance, and the risk of asthma and wheezing during childhood. N Engl J Med. 2000; 343(8): 538–43. PubMed Abstract | Publisher Full Text 20. Cheng G, Smith AM, Levin L: Duration of day care attendance during infancy predicts asthma at the age of seven: the Cincinnati Childhood Allergy and Air Pollution Study. Clin Exp Allergy. 2014; 44(10): 1274–81. PubMed Abstract | Publisher Full Text 21. Custovic A: To what extent is allergen exposure a risk factor for the development of allergic disease? Clin Exp Allergy. 2015; 45(1): 54–62. PubMed Abstract | Publisher Full Text 22. Jackson DJ, Evans MD, Gangnon RE, et al.: Evidence for a causal relationship between allergic sensitization and rhinovirus wheezing in early life. Am J Respir Crit Care Med. 2012; 185(3): 281–5. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation 23. Maas T, Kaper J, Sheikh A, et al.: Mono and multifaceted inhalant and/or food allergen reduction interventions for preventing asthma in children at high risk of developing asthma. Cochrane Database Syst Rev. 2009; (3): CD006480. PubMed Abstract | Publisher Full Text 24. Chan-Yeung M, Ferguson A, Watson W, et al.: The Canadian Childhood Asthma Page 9 of 12 F1000Research 2017, 6(F1000 Faculty Rev):2152 Last updated: 17 JUL 2019 http://www.ncbi.nlm.nih.gov/pubmed/24754822 http://dx.doi.org/10.1513/AnnalsATS.201312-448LD http://www.ncbi.nlm.nih.gov/pmc/articles/4112503 https://f1000.com/prime/731078457 http://www.ncbi.nlm.nih.gov/pubmed/28890020 http://dx.doi.org/10.1016/j.anai.2017.07.002 https://f1000.com/prime/731078457 http://www.ncbi.nlm.nih.gov/pubmed/26382999 http://dx.doi.org/10.1016/S0140-6736(15)00156-7 http://www.ncbi.nlm.nih.gov/pubmed/24636091 http://dx.doi.org/10.1016/j.jaci.2014.01.018 http://www.ncbi.nlm.nih.gov/pmc/articles/4088262 http://www.ncbi.nlm.nih.gov/pubmed/7800004 http://dx.doi.org/10.1056/NEJM199501193320301 http://www.ncbi.nlm.nih.gov/pubmed/25379671 http://dx.doi.org/10.1371/journal.pone.0111922 http://www.ncbi.nlm.nih.gov/pmc/articles/4224405 http://www.ncbi.nlm.nih.gov/pubmed/26947979 http://dx.doi.org/10.1016/j.jaci.2016.01.002 http://www.ncbi.nlm.nih.gov/pmc/articles/4782609 https://f1000.com/prime/727429855 http://www.ncbi.nlm.nih.gov/pubmed/28322581 http://dx.doi.org/10.2217/epi-2016-0162 https://f1000.com/prime/727429855 http://www.ncbi.nlm.nih.gov/pubmed/27171908 http://dx.doi.org/10.1111/pai.12598 http://www.ncbi.nlm.nih.gov/pubmed/16512801 http://dx.doi.org/10.1111/j.1398-9995.2006.01009.x https://f1000.com/prime/8846957 http://www.ncbi.nlm.nih.gov/pubmed/21345099 http://dx.doi.org/10.1056/NEJMoa1007302 https://f1000.com/prime/8846957 http://www.ncbi.nlm.nih.gov/pubmed/22892709 http://dx.doi.org/10.1097/ACI.0b013e328357a3bc http://www.ncbi.nlm.nih.gov/pubmed/18448493 http://dx.doi.org/10.1183/09031936.00033707 https://f1000.com/prime/726625847 http://www.ncbi.nlm.nih.gov/pubmed/27518660 http://dx.doi.org/10.1056/NEJMoa1508749 http://www.ncbi.nlm.nih.gov/pmc/articles/5137793 https://f1000.com/prime/726625847 https://f1000.com/prime/1009325 http://www.ncbi.nlm.nih.gov/pubmed/12190366 http://dx.doi.org/10.1001/jama.288.8.963 https://f1000.com/prime/1009325 http://www.ncbi.nlm.nih.gov/pubmed/12204868 http://dx.doi.org/10.1164/rccm.2201035 https://f1000.com/prime/717955515 http://www.ncbi.nlm.nih.gov/pubmed/22952649 http://dx.doi.org/10.1371/journal.pone.0043214 http://www.ncbi.nlm.nih.gov/pmc/articles/3430634 https://f1000.com/prime/717955515 https://f1000.com/prime/725902620 http://www.ncbi.nlm.nih.gov/pubmed/26523822 http://dx.doi.org/10.1001/jamapediatrics.2015.3219 https://f1000.com/prime/725902620 http://www.ncbi.nlm.nih.gov/pubmed/10954761 http://dx.doi.org/10.1056/NEJM200008243430803 http://www.ncbi.nlm.nih.gov/pubmed/25179746 http://dx.doi.org/10.1111/cea.12397 http://www.ncbi.nlm.nih.gov/pubmed/25381695 http://dx.doi.org/10.1111/cea.12450 https://f1000.com/prime/13833959 http://www.ncbi.nlm.nih.gov/pubmed/21960534 http://dx.doi.org/10.1164/rccm.201104-0660OC http://www.ncbi.nlm.nih.gov/pmc/articles/3297109 https://f1000.com/prime/13833959 http://www.ncbi.nlm.nih.gov/pubmed/19588394 http://dx.doi.org/10.1002/14651858.CD006480.pub2 Primary Prevention Study: outcomes at 7 years of age. J Allergy Clin Immunol. 2005; 116(1): 49–55. PubMed Abstract | Publisher Full Text 25. Lynch SV, Wood RA, Boushey H, et al.: Effects of early-life exposure to allergens and bacteria on recurrent wheeze and atopy in urban children. J Allergy Clin Immunol. 2014; 134(3): 593–601.e12. PubMed Abstract | Publisher Full Text | Free Full Text 26. Valovirta E, Petersen TH, Piotrowska T, et al.: Results from the 5-year SQ grass sublingual immunotherapy tablet asthma prevention (GAP) trial in children with grass pollen allergy. J Allergy Clin Immunol. 2017; pii: S0091-6749(17)31088-6. PubMed Abstract | Publisher Full Text | F1000 Recommendation 27. Guarnieri M, Balmes JR: Outdoor air pollution and asthma. Lancet. 2014; 383(9928): 1581–92. PubMed Abstract | Publisher Full Text | Free Full Text 28. Schultz ES, Litonjua AA, Melén E: Effects of Long-Term Exposure to Traffic- Related Air Pollution on Lung Function in Children. Curr Allergy Asthma Rep. 2017; 17(6): 41. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation 29. Sbihi H, Allen RW, Becker A, et al.: Perinatal Exposure to Traffic-Related Air Pollution and Atopy at 1 Year of Age in a Multi-Center Canadian Birth Cohort Study. Environ Health Perspect. 2015; 123(9): 902–8. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation 30. Gauderman WJ, Urman R, Avol E, et al.: Association of improved air quality with lung development in children. N Engl J Med. 2015; 372(10): 905–13. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation 31. Yang IV, Lozupone CA, Schwartz DA: The environment, epigenome, and asthma. J Allergy Clin Immunol. 2017; 140(1): 14–23. PubMed Abstract | Publisher Full Text | Free Full Text 32. Yang HJ: Impact of perinatal environmental tobacco smoke on the development of childhood allergic diseases. Korean J Pediatr. 2016; 59(8): 319–27. PubMed Abstract | Publisher Full Text | Free Full Text 33. Bouzigon E, Corda E, Aschard H, et al.: Effect of 17q21 variants and smoking exposure in early-onset asthma. N Engl J Med. 2008; 359(19): 1985–94. PubMed Abstract | Publisher Full Text | F1000 Recommendation 34. Moffatt MF, Gut IG, Demenais F, et al.: A large-scale, consortium-based genomewide association study of asthma. N Engl J Med. 2010; 363(13): 1211–21. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation 35. Feleszko W, Ruszczyński M, Jaworska J, et al.: Environmental tobacco smoke exposure and risk of allergic sensitisation in children: a systematic review and meta-analysis. Arch Dis Child. 2014; 99(11): 985–92. PubMed Abstract | Publisher Full Text 36. Burke H, Leonardi-Bee J, Hashim A, et al.: Prenatal and passive smoke exposure and incidence of asthma and wheeze: systematic review and meta- analysis. Pediatrics. 2012; 129(4): 735–44. PubMed Abstract | Publisher Full Text | F1000 Recommendation 37. Vardavas CI, Hohmann C, Patelarou E, 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(1): 115–24. PubMed Abstract | Publisher Full Text | F1000 Recommendation 38. Feldman AS, He Y, Moore ML, et al.: Toward primary prevention of asthma. Reviewing the evidence for early-life respiratory viral infections as modifiable risk factors to prevent childhood asthma. Am J Respir Crit Care Med. 2015; 191(1): 34–44. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation 39. Bacharier LB, Cohen R, Schweiger T, et al.: Determinants of asthma after severe respiratory syncytial virus bronchiolitis. J Allergy Clin Immunol. 2012; 130(1): 91–100.e3. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation 40. Zomer-Kooijker K, van der Ent CK, Ermers MJ, et al.: Increased risk of wheeze and decreased lung function after respiratory syncytial virus infection. PLoS One. 2014; 9(1): e87162. PubMed Abstract | Publisher Full Text | Free Full Text 41. Beigelman A, Bacharier LB: Early-life respiratory infections and asthma development: role in disease pathogenesis and potential targets for disease prevention. Curr Opin Allergy Clin Immunol. 2016; 16(2): 172–8. PubMed Abstract | Publisher Full Text | Free Full Text 42. Jafri HS, Chavez-Bueno S, Mejias A, et al.: Respiratory syncytial virus induces pneumonia, cytokine response, airway obstruction, and chronic inflammatory infiltrates associated with long-term airway hyperresponsiveness in mice. J Infect Dis. 2004; 189(10): 1856–65. PubMed Abstract | Publisher Full Text 43. Román M, Calhoun WJ, Hinton KL, et al.: Respiratory syncytial virus infection in infants is associated with predominant Th-2-like response. Am J Respir Crit Care Med. 1997; 156(1): 190–5. PubMed Abstract | Publisher Full Text 44. Piedimonte G, Walton C, Samsell L: Vertical transmission of respiratory syncytial virus modulates pre- and postnatal innervation and reactivity of rat airways. PLoS One. 2013; 8(4): e61309. PubMed Abstract | Publisher Full Text | Free Full Text 45. Blanken MO, Rovers MM, Molenaar JM, et al.: Respiratory syncytial virus and recurrent wheeze in healthy preterm infants. N Engl J Med. 2013; 368(19): 1791–9. PubMed Abstract | Publisher Full Text | F1000 Recommendation 46. Simões EA, Carbonell-Estrany X, Rieger CH, et al.: The effect of respiratory syncytial virus on subsequent recurrent wheezing in atopic and nonatopic children. J Allergy Clin Immunol. 2010; 126(2): 256–62. PubMed Abstract | Publisher Full Text 47. Mochizuki H, Kusuda S, Okada K, et al.: Palivizumab Prophylaxis in Preterm Infants and Subsequent Recurrent Wheezing. Six-Year Follow-up Study. Am J Respir Crit Care Med. 2017; 196(1): 29–38. PubMed Abstract | Publisher Full Text | F1000 Recommendation 48. Carroll KN, Gebretsadik T, Escobar GJ, et al.: Respiratory syncytial virus immunoprophylaxis in high-risk infants and development of childhood asthma. J Allergy Clin Immunol. 2017; 139(1): 66–71.e3. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation 49. Achten NB, Wu P, Bont L, et al.: Interference Between Respiratory Syncytial Virus and Human Rhinovirus Infection in Infancy. J Infect Dis. 2017; 215(7): 1102–6. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation 50. Bacharier LB, Guilbert TW, Mauger DT, et al.: Early Administration of Azithromycin and Prevention of Severe Lower Respiratory Tract Illnesses in Preschool Children With a History of Such Illnesses: A Randomized Clinical Trial. JAMA. 2015; 314(19): 2034–44. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation 51. Zhou Y, Bacharier LB, Isaacson-Schmid M, et al.: Azithromycin therapy during respiratory syncytial virus bronchiolitis: Upper airway microbiome alterations and subsequent recurrent wheeze. J Allergy Clin Immunol. 2016; 138(4): 1215–1219.e5. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation 52. Karron RA, Luongo C, Thumar B, et al.: A gene deletion that up-regulates viral gene expression yields an attenuated RSV vaccine with improved antibody responses in children. Sci Transl Med. 2015; 7(312): 312ra175. PubMed Abstract | Publisher Full Text 53. Saso A, Kampmann B: Vaccination against respiratory syncytial virus in pregnancy: a suitable tool to combat global infant morbidity and mortality? Lancet Infect Dis. 2016; 16(8): e153–63. PubMed Abstract | Publisher Full Text 54. Martinez FD: New insights into the natural history of asthma: primary prevention on the horizon. J Allergy Clin Immunol. 2011; 128(5): 939–45. PubMed Abstract | Publisher Full Text | Free Full Text 55. Jackson DJ, Gangnon RE, Evans MD, et al.: Wheezing rhinovirus illnesses in early life predict asthma development in high-risk children. Am J Respir Crit Care Med. 2008; 178(7): 667–72. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation 56. Nicolai A, Frassanito A, Nenna R, et al.: Risk Factors for Virus-induced Acute Respiratory Tract Infections in Children Younger Than 3 Years and Recurrent Wheezing at 36 Months Follow-Up After Discharge. Pediatr Infect Dis J. 2017; 36(2): 179–83. PubMed Abstract | Publisher Full Text | F1000 Recommendation 57. Jartti T, Nieminen R, Vuorinen T, et al.: Short- and long-term efficacy of prednisolone for first acute rhinovirus-induced wheezing episode. J Allergy Clin Immunol. 2015; 135(3): 691–8.e9. PubMed Abstract | Publisher Full Text | F1000 Recommendation 58. Lee S, Nguyen MT, Currier MG, et al.: A polyvalent inactivated rhinovirus vaccine is broadly immunogenic in rhesus macaques. Nat Commun. 2016; 7: 12838. PubMed Abstract | Publisher Full Text | Free Full Text 59. Mukherjee PK, Esper F, Buchheit K, et al.: Randomized, double-blind, placebo-controlled clinical trial to assess the safety and effectiveness of a novel dual-action oral topical formulation against upper respiratory infections. BMC Infect Dis. 2017; 17(1): 74. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation 60. Rosas-Salazar C, Shilts MH, Tovchigrechko A, et al.: Nasopharyngeal Microbiome in Respiratory Syncytial Virus Resembles Profile Associated with Increased Childhood Asthma Risk. Am J Respir Crit Care Med. 2016; 193(10): 1180–3. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation 61. Chung KF: Airway microbial dysbiosis in asthmatic patients: A target for prevention and treatment? J Allergy Clin Immunol. 2017; 139(4): 1071–81. PubMed Abstract | Publisher Full Text 62. Cuello-Garcia CA, Fiocchi A, Pawankar R, et al.: World Allergy Organization- McMaster University Guidelines for Allergic Disease Prevention (GLAD-P): Prebiotics. World Allergy Organ J. 2016; 9: 10. PubMed Abstract | Publisher Full Text | Free Full Text 63. Sordillo JE, Scirica CV, Rifas-Shiman SL, et al.: Prenatal and infant exposure to Page 10 of 12 F1000Research 2017, 6(F1000 Faculty Rev):2152 Last updated: 17 JUL 2019 http://www.ncbi.nlm.nih.gov/pubmed/15990772 http://dx.doi.org/10.1016/j.jaci.2005.03.029 http://www.ncbi.nlm.nih.gov/pubmed/24908147 http://dx.doi.org/10.1016/j.jaci.2014.04.018 http://www.ncbi.nlm.nih.gov/pmc/articles/4151305 https://f1000.com/prime/727795776 http://www.ncbi.nlm.nih.gov/pubmed/28689794 http://dx.doi.org/10.1016/j.jaci.2017.06.014 https://f1000.com/prime/727795776 http://www.ncbi.nlm.nih.gov/pubmed/24792855 http://dx.doi.org/10.1016/S0140-6736(14)60617-6 http://www.ncbi.nlm.nih.gov/pmc/articles/4465283 https://f1000.com/prime/727676234 http://www.ncbi.nlm.nih.gov/pubmed/28551888 http://dx.doi.org/10.1007/s11882-017-0709-y http://www.ncbi.nlm.nih.gov/pmc/articles/5446841 https://f1000.com/prime/727676234 https://f1000.com/prime/725411384 http://www.ncbi.nlm.nih.gov/pubmed/25826816 http://dx.doi.org/10.1289/ehp.1408700 http://www.ncbi.nlm.nih.gov/pmc/articles/4559953 https://f1000.com/prime/725411384 https://f1000.com/prime/725375890 http://www.ncbi.nlm.nih.gov/pubmed/25738666 http://dx.doi.org/10.1056/NEJMoa1414123 http://www.ncbi.nlm.nih.gov/pmc/articles/4430551 https://f1000.com/prime/725375890 http://www.ncbi.nlm.nih.gov/pubmed/28673400 http://dx.doi.org/10.1016/j.jaci.2017.05.011 http://www.ncbi.nlm.nih.gov/pmc/articles/5673130 http://www.ncbi.nlm.nih.gov/pubmed/27610180 http://dx.doi.org/10.3345/kjp.2016.59.8.319 http://www.ncbi.nlm.nih.gov/pmc/articles/5014911 https://f1000.com/prime/1125838 http://www.ncbi.nlm.nih.gov/pubmed/18923164 http://dx.doi.org/10.1056/NEJMoa0806604 https://f1000.com/prime/1125838 https://f1000.com/prime/6546960 http://www.ncbi.nlm.nih.gov/pubmed/20860503 http://dx.doi.org/10.1056/NEJMoa0906312 http://www.ncbi.nlm.nih.gov/pmc/articles/4260321 https://f1000.com/prime/6546960 http://www.ncbi.nlm.nih.gov/pubmed/24958794 http://dx.doi.org/10.1136/archdischild-2013-305444 https://f1000.com/prime/716197976 http://www.ncbi.nlm.nih.gov/pubmed/22430451 http://dx.doi.org/10.1542/peds.2011-2196 https://f1000.com/prime/716197976 https://f1000.com/prime/726211842 http://www.ncbi.nlm.nih.gov/pubmed/26965294 http://dx.doi.org/10.1183/13993003.01016-2015 https://f1000.com/prime/726211842 https://f1000.com/prime/725223247 http://www.ncbi.nlm.nih.gov/pubmed/25369458 http://dx.doi.org/10.1164/rccm.201405-0901PP http://www.ncbi.nlm.nih.gov/pmc/articles/4299628 https://f1000.com/prime/725223247 https://f1000.com/prime/14267041 http://www.ncbi.nlm.nih.gov/pubmed/22444510 http://dx.doi.org/10.1016/j.jaci.2012.02.010 http://www.ncbi.nlm.nih.gov/pmc/articles/3612548 https://f1000.com/prime/14267041 http://www.ncbi.nlm.nih.gov/pubmed/24498037 http://dx.doi.org/10.1371/journal.pone.0087162 http://www.ncbi.nlm.nih.gov/pmc/articles/3909049 http://www.ncbi.nlm.nih.gov/pubmed/26854761 http://dx.doi.org/10.1097/ACI.0000000000000244 http://www.ncbi.nlm.nih.gov/pmc/articles/5089840 http://www.ncbi.nlm.nih.gov/pubmed/15122522 http://dx.doi.org/10.1086/386372 http://www.ncbi.nlm.nih.gov/pubmed/9230746 http://dx.doi.org/10.1164/ajrccm.156.1.9611050 http://www.ncbi.nlm.nih.gov/pubmed/23637810 http://dx.doi.org/10.1371/journal.pone.0061309 http://www.ncbi.nlm.nih.gov/pmc/articles/3630224 https://f1000.com/prime/718009158 http://www.ncbi.nlm.nih.gov/pubmed/23656644 http://dx.doi.org/10.1056/NEJMoa1211917 https://f1000.com/prime/718009158 http://www.ncbi.nlm.nih.gov/pubmed/20624638 http://dx.doi.org/10.1016/j.jaci.2010.05.026 https://f1000.com/prime/727261386 http://www.ncbi.nlm.nih.gov/pubmed/28152315 http://dx.doi.org/10.1164/rccm.201609-1812OC https://f1000.com/prime/727261386 https://f1000.com/prime/726387093 http://www.ncbi.nlm.nih.gov/pubmed/27212083 http://dx.doi.org/10.1016/j.jaci.2016.01.055 http://www.ncbi.nlm.nih.gov/pmc/articles/5074917 https://f1000.com/prime/726387093 https://f1000.com/prime/727471626 http://www.ncbi.nlm.nih.gov/pubmed/28368456 http://dx.doi.org/10.1093/infdis/jix031 http://www.ncbi.nlm.nih.gov/pmc/articles/5426371 https://f1000.com/prime/727471626 https://f1000.com/prime/725940727 http://www.ncbi.nlm.nih.gov/pubmed/26575060 http://dx.doi.org/10.1001/jama.2015.13896 http://www.ncbi.nlm.nih.gov/pmc/articles/4757487 https://f1000.com/prime/725940727 https://f1000.com/prime/726448126 http://www.ncbi.nlm.nih.gov/pubmed/27339392 http://dx.doi.org/10.1016/j.jaci.2016.03.054 http://www.ncbi.nlm.nih.gov/pmc/articles/5056860 https://f1000.com/prime/726448126 http://www.ncbi.nlm.nih.gov/pubmed/26537255 http://dx.doi.org/10.1126/scitranslmed.aac8463 http://www.ncbi.nlm.nih.gov/pubmed/27317449 http://dx.doi.org/10.1016/S1473-3099(16)00119-5 http://www.ncbi.nlm.nih.gov/pubmed/22036094 http://dx.doi.org/10.1016/j.jaci.2011.09.020 http://www.ncbi.nlm.nih.gov/pmc/articles/3963825 https://f1000.com/prime/1122999 http://www.ncbi.nlm.nih.gov/pubmed/18565953 http://dx.doi.org/10.1164/rccm.200802-309OC http://www.ncbi.nlm.nih.gov/pmc/articles/2556448 https://f1000.com/prime/1122999 https://f1000.com/prime/726908542 http://www.ncbi.nlm.nih.gov/pubmed/27798551 http://dx.doi.org/10.1097/INF.0000000000001385 https://f1000.com/prime/726908542 https://f1000.com/prime/718535343 http://www.ncbi.nlm.nih.gov/pubmed/25129681 http://dx.doi.org/10.1016/j.jaci.2014.07.001 https://f1000.com/prime/718535343 http://www.ncbi.nlm.nih.gov/pubmed/27653379 http://dx.doi.org/10.1038/ncomms12838 http://www.ncbi.nlm.nih.gov/pmc/articles/5036149 https://f1000.com/prime/727209514 http://www.ncbi.nlm.nih.gov/pubmed/28088167 http://dx.doi.org/10.1186/s12879-016-2177-8 http://www.ncbi.nlm.nih.gov/pmc/articles/5237564 https://f1000.com/prime/727209514 https://f1000.com/prime/726367509 http://www.ncbi.nlm.nih.gov/pubmed/27174483 http://dx.doi.org/10.1164/rccm.201512-2350LE http://www.ncbi.nlm.nih.gov/pmc/articles/4872668 https://f1000.com/prime/726367509 http://www.ncbi.nlm.nih.gov/pubmed/28390574 http://dx.doi.org/10.1016/j.jaci.2017.02.004 http://www.ncbi.nlm.nih.gov/pubmed/26962387 http://dx.doi.org/10.1186/s40413-016-0102-7 http://www.ncbi.nlm.nih.gov/pmc/articles/4772464 acetaminophen and ibuprofen and the risk for wheeze and asthma in children. J Allergy Clin Immunol. 2015; 135(2): 441–8. PubMed Abstract | Publisher Full Text | Free Full Text 64. Castro-Rodriguez JA, Forno E, Rodriguez-Martinez CE, et al.: Risk and Protective Factors for Childhood Asthma: What Is the Evidence? J Allergy Clin Immunol Pract. 2016; 4(6): 1111–22. PubMed Abstract | Publisher Full Text | Free Full Text 65. Black M, Bhattacharya S, Philip S, et al.: Planned Cesarean Delivery at Term and Adverse Outcomes in Childhood Health. JAMA. 2015; 314(21): 2271–9. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation 66. Wu P, Feldman AS, Rosas-Salazar C, et al.: Relative Importance and Additive Effects of Maternal and Infant Risk Factors on Childhood Asthma. PLoS One. 2016; 11(3): e0151705. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation 67. Hendaus MA, Jomha FA, Ehlayel M: Allergic diseases among children: nutritional prevention and intervention. Ther Clin Risk Manag. 2016; 12: 361–72. PubMed Abstract | Publisher Full Text | Free Full Text 68. Melli LC, do Carmo-Rodrigues MS, Araújo-Filho HB, et al.: Intestinal microbiota and allergic diseases: A systematic review. Allergol Immunopathol (Madr). 2016; 44(2): 177–88. PubMed Abstract | Publisher Full Text | F1000 Recommendation 69. Arrieta MC, Stiemsma LT, Dimitriu PA, et al.: Early infancy microbial and metabolic alterations affect risk of childhood asthma. Sci Transl Med. 2015; 7(307): 307ra152. PubMed Abstract | Publisher Full Text | F1000 Recommendation 70. Azad MB, Coneys JG, Kozyrskyj AL, et al.: Probiotic supplementation during pregnancy or infancy for the prevention of asthma and wheeze: systematic review and meta-analysis. BMJ. 2013; 347: f6471. PubMed Abstract | Publisher Full Text | Free Full Text 71. Bloomfield SF, Rook GA, Scott EA, et al.: Time to abandon the hygiene hypothesis: new perspectives on allergic disease, the human microbiome, infectious disease prevention and the role of targeted hygiene. Perspect Public Health. 2016; 136(4): 213–24. PubMed Abstract | Publisher Full Text | Free Full Text 72. Ali Z, Nilas L, Ulrik CS: Excessive gestational weight gain in first trimester is a risk factor for exacerbation of asthma during pregnancy: A prospective study of 1283 pregnancies. J Allergy Clin Immunol. 2017; pii: S0091-6749(17)30673-5. PubMed Abstract | Publisher Full Text | F1000 Recommendation 73. Sewell DA, Hammersley VS, Robertson A, et al.: A pilot randomised controlled trial investigating a Mediterranean diet intervention in pregnant women for the primary prevention of allergic diseases in infants. J Hum Nutr Diet. 2017; 30(5): 604–14. PubMed Abstract | Publisher Full Text | F1000 Recommendation 74. Garcia-Marcos L, Castro-Rodriguez JA, Weinmayr G, et al.: Influence of Mediterranean diet on asthma in children: a systematic review and meta- analysis. Pediatr Allergy Immunol. 2013; 24(4): 330–8. PubMed Abstract | Publisher Full Text 75. von Mutius E, Martinez FD: Inconclusive Results of Randomized Trials of Prenatal Vitamin D for Asthma Prevention in Offspring: Curbing the Enthusiasm. JAMA. 2016; 315(4): 347–8. PubMed Abstract | Publisher Full Text 76. Litonjua AA, Carey VJ, Laranjo N, et al.: Effect of Prenatal Supplementation With Vitamin D on Asthma or Recurrent Wheezing in Offspring by Age 3 Years: The VDAART Randomized Clinical Trial. JAMA. 2016; 315(4): 362–70. PubMed Abstract | Publisher Full Text | F1000 Recommendation 77. Wolsk HM, Harshfield BJ, Laranjo N, et al.: Vitamin D supplementation in pregnancy, prenatal 25(OH)D levels, race, and subsequent asthma or recurrent wheeze in offspring: Secondary analyses from the Vitamin D Antenatal Asthma Reduction Trial. J Allergy Clin Immunol. 2017; 140(5): 1423–1429.e5. PubMed Abstract | Publisher Full Text | F1000 Recommendation 78. Martineau AR, Jolliffe DA, Hooper RL, et al.: Vitamin D supplementation to prevent acute respiratory tract infections: systematic review and meta- analysis of individual participant data. BMJ. 2017; 356: i6583. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation 79. Helve O, Viljakainen H, Holmlund-Suila E, et al.: Towards evidence-based vitamin D supplementation in infants: vitamin D intervention in infants (VIDI) - study design and methods of a randomised controlled double-blinded intervention study. BMC Pediatr. 2017; 17(1): 91. PubMed Abstract | Publisher Full Text | Free Full Text 80. Vahdaninia M, Mackenzie H, Helps S, et al.: Prenatal Intake of Vitamins and Allergic Outcomes in the Offspring: A Systematic Review and Meta-Analysis. J Allergy Clin Immunol Pract. 2017; 5(3): 771–778.e5. PubMed Abstract | Publisher Full Text | F1000 Recommendation 81. Gref A, Rautiainen S, Gruzieva O, et al.: Dietary total antioxidant capacity in early school age and subsequent allergic disease. Clin Exp Allergy. 2017; 47(6): 751–9. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation 82. Hodge L, Salome CM, Peat JK, et al.: Consumption of oily fish and childhood asthma risk. Med J Aust. 1996; 164(3): 137–40. PubMed Abstract 83. Bisgaard H, Stokholm J, Chawes BL, et al.: Fish Oil-Derived Fatty Acids in Pregnancy and Wheeze and Asthma in Offspring. N Engl J Med. 2016; 375(26): 2530–9. PubMed Abstract | Publisher Full Text | F1000 Recommendation 84. Chiu YH, Coull BA, Cohen S, et al.: Prenatal and postnatal maternal stress and wheeze in urban children: effect of maternal sensitization. Am J Respir Crit Care Med. 2012; 186(2): 147–54. PubMed Abstract | Publisher Full Text | Free Full Text 85. Wright RJ, Visness CM, Calatroni A, et al.: Prenatal maternal stress and cord blood innate and adaptive cytokine responses in an inner-city cohort. Am J Respir Crit Care Med. 2010; 182(1): 25–33. PubMed Abstract | Publisher Full Text | Free Full Text 86. Weaver IC, Szyf M, Meaney MJ: From maternal care to gene expression: DNA methylation and the maternal programming of stress responses. Endocr Res. 2002; 28(4): 699. PubMed Abstract | Publisher Full Text 87. Dreger LC, Kozyrskyj AL, HayGlass KT, et al.: Lower cortisol levels in children with asthma exposed to recurrent maternal distress from birth. J Allergy Clin Immunol. 2010; 125(1): 116–22. PubMed Abstract | Publisher Full Text 88. Ramratnam SK, Visness CM, Jaffee KF, et al.: Relationships among Maternal Stress and Depression, Type 2 Responses, and Recurrent Wheezing at Age 3 Years in Low-Income Urban Families. Am J Respir Crit Care Med. 2017; 195(5): 674–81. PubMed Abstract | Publisher Full Text | Free Full Text | F1000 Recommendation 89. Plourde A, Lavoie KL, Raddatz C, et al.: Effects of acute psychological stress induced in laboratory on physiological responses in asthma populations: A systematic review. Respir Med. 2017; 127: 21–32. PubMed Abstract | Publisher Full Text | F1000 Recommendation Page 11 of 12 F1000Research 2017, 6(F1000 Faculty Rev):2152 Last updated: 17 JUL 2019 http://www.ncbi.nlm.nih.gov/pubmed/25441647 http://dx.doi.org/10.1016/j.jaci.2014.07.065 http://www.ncbi.nlm.nih.gov/pmc/articles/4323723 http://www.ncbi.nlm.nih.gov/pubmed/27286779 http://dx.doi.org/10.1016/j.jaip.2016.05.003 http://www.ncbi.nlm.nih.gov/pmc/articles/5107168 https://f1000.com/prime/725975546 http://www.ncbi.nlm.nih.gov/pubmed/26624826 http://dx.doi.org/10.1001/jama.2015.16176 http://www.ncbi.nlm.nih.gov/pmc/articles/5055095 https://f1000.com/prime/725975546 https://f1000.com/prime/726230265 http://www.ncbi.nlm.nih.gov/pubmed/27002979 http://dx.doi.org/10.1371/journal.pone.0151705 http://www.ncbi.nlm.nih.gov/pmc/articles/4803347 https://f1000.com/prime/726230265 http://www.ncbi.nlm.nih.gov/pubmed/27022267 http://dx.doi.org/10.2147/TCRM.S98100 http://www.ncbi.nlm.nih.gov/pmc/articles/4788360 https://f1000.com/prime/725704737 http://www.ncbi.nlm.nih.gov/pubmed/25985709 http://dx.doi.org/10.1016/j.aller.2015.01.013 https://f1000.com/prime/725704737 https://f1000.com/prime/725823144 http://www.ncbi.nlm.nih.gov/pubmed/26424567 http://dx.doi.org/10.1126/scitranslmed.aab2271 https://f1000.com/prime/725823144 http://www.ncbi.nlm.nih.gov/pubmed/24304677 http://dx.doi.org/10.1136/bmj.f6471 http://www.ncbi.nlm.nih.gov/pmc/articles/3898421 http://www.ncbi.nlm.nih.gov/pubmed/27354505 http://dx.doi.org/10.1177/1757913916650225 http://www.ncbi.nlm.nih.gov/pmc/articles/4966430 https://f1000.com/prime/727663565 http://www.ncbi.nlm.nih.gov/pubmed/28551030 http://dx.doi.org/10.1016/j.jaci.2017.03.040 https://f1000.com/prime/727663565 https://f1000.com/prime/727322383 http://www.ncbi.nlm.nih.gov/pubmed/28211106 http://dx.doi.org/10.1111/jhn.12469 https://f1000.com/prime/727322383 http://www.ncbi.nlm.nih.gov/pubmed/23578354 http://dx.doi.org/10.1111/pai.12071 http://www.ncbi.nlm.nih.gov/pubmed/26813205 http://dx.doi.org/10.1001/jama.2015.18963 https://f1000.com/prime/726105973 http://www.ncbi.nlm.nih.gov/pubmed/26813209 http://dx.doi.org/10.1001/jama.2015.18589 https://f1000.com/prime/726105973 https://f1000.com/prime/727397650 http://www.ncbi.nlm.nih.gov/pubmed/28285844 http://dx.doi.org/10.1016/j.jaci.2017.01.013 https://f1000.com/prime/727397650 https://f1000.com/prime/727312221 http://www.ncbi.nlm.nih.gov/pubmed/28202713 http://dx.doi.org/10.1136/bmj.i6583 http://www.ncbi.nlm.nih.gov/pmc/articles/5310969 https://f1000.com/prime/727312221 http://www.ncbi.nlm.nih.gov/pubmed/28356142 http://dx.doi.org/10.1186/s12887-017-0845-5 http://www.ncbi.nlm.nih.gov/pmc/articles/5372327 https://f1000.com/prime/727027160 http://www.ncbi.nlm.nih.gov/pubmed/27888033 http://dx.doi.org/10.1016/j.jaip.2016.09.024 https://f1000.com/prime/727027160 https://f1000.com/prime/727328305 http://www.ncbi.nlm.nih.gov/pubmed/28222232 http://dx.doi.org/10.1111/cea.12911 http://www.ncbi.nlm.nih.gov/pmc/articles/5485024 https://f1000.com/prime/727328305 http://www.ncbi.nlm.nih.gov/pubmed/8628130 https://f1000.com/prime/727149602 http://www.ncbi.nlm.nih.gov/pubmed/28029926 http://dx.doi.org/10.1056/NEJMoa1503734 https://f1000.com/prime/727149602 http://www.ncbi.nlm.nih.gov/pubmed/22582161 http://dx.doi.org/10.1164/rccm.201201-0162OC http://www.ncbi.nlm.nih.gov/pmc/articles/3406080 http://www.ncbi.nlm.nih.gov/pubmed/20194818 http://dx.doi.org/10.1164/rccm.200904-0637OC http://www.ncbi.nlm.nih.gov/pmc/articles/2902757 http://www.ncbi.nlm.nih.gov/pubmed/12530685 http://dx.doi.org/10.1081/ERC-120016989 http://www.ncbi.nlm.nih.gov/pubmed/19962747 http://dx.doi.org/10.1016/j.jaci.2009.09.051 https://f1000.com/prime/726764724 http://www.ncbi.nlm.nih.gov/pubmed/27654103 http://dx.doi.org/10.1164/rccm.201602-0272OC http://www.ncbi.nlm.nih.gov/pmc/articles/5363974 https://f1000.com/prime/726764724 https://f1000.com/prime/727676426 http://www.ncbi.nlm.nih.gov/pubmed/28502415 http://dx.doi.org/10.1016/j.rmed.2017.03.024 https://f1000.com/prime/727676426   Open Peer Review Current Peer Review Status: Editorial Note on the Review Process  are written by members of the prestigious  . They are commissioned andF1000 Faculty Reviews F1000 Faculty are peer reviewed before publication to ensure that the final, published version is comprehensive and accessible. The reviewers who approved the final version are listed with their names and affiliations. The reviewers who approved this article are: Version 1 The benefits of publishing with F1000Research: Your article is published within days, with no editorial bias You can publish traditional articles, null/negative results, case reports, data notes and more The peer review process is transparent and collaborative Your article is indexed in PubMed after passing peer review Dedicated customer support at every stage For pre-submission enquiries, contact   research@f1000.com  Tina V Hartert Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and Center for Asthma Research, Vanderbilt University School of Medicine, Nashville, Tennessee, USA  Christian E Lynch Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and Center for Asthma Research, Vanderbilt University School of Medicine, Nashville, Tennessee, USA  No competing interests were disclosed.Competing Interests: 1  Allan Becker Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, R3T 2N2, Canada  No competing interests were disclosed.Competing Interests: 2 Page 12 of 12 F1000Research 2017, 6(F1000 Faculty Rev):2152 Last updated: 17 JUL 2019 https://f1000research.com/browse/f1000-faculty-reviews http://f1000.com/prime/thefaculty