24
During the past decade, the microbiome has emerged as a major contributor to human health 1 It has been suggested in current studies that the early-life microbiome is a crucial factor for proper immune development and long-term health 2, 3 Transient microbial dysbiosis during this time period has been associated with the development of immune-mediated, metabolic, and neurodevelopmental disorders 47 In addition, increasing evidence has been used to support a vital role of the maternal and intrauterine microbiomes in childhood health and development 8 Collectively, these findings have been used to support the microbiome as a key participant in the Developmental Origins of Health and Disease (DOHaD) In this review, we will discuss the current research surrounding the maturation of the early-life microbiome and how transient variations in this bacterial consortium can have long-term consequences for human health THE DOHAD: WHERE DOES THE MICROBIOME FIT? The developmental origins hypothesis proposes variations in fetal and infant programming through environmental exposures during a critical window of early life 9 Termed originally as the Barker hypothesis, 10, 11 in which the association between fetal malnutrition and hypertension later in life was a focus, this theory has since expanded to account for many types of early- life exposures and birth outcomes associated with long-term health and development For example, high birth The Role of the Microbiome in the Developmental Origins of Health and Disease Leah T. Stiemsma, PhD, Karin B. Michels, ScD, PhD Although the prominent role of the microbiome in human health has been established, the early-life microbiome is now being recognized as a major influence on long-term human health and development Variations in the composition and functional potential of the early-life microbiome are the result of lifestyle factors, such as mode of birth, breastfeeding, diet, and antibiotic usage In addition, variations in the composition of the early-life microbiome have been associated with specific disease outcomes, such as asthma, obesity, and neurodevelopmental disorders This points toward this bacterial consortium as a mediator between early lifestyle factors and health and disease In addition, variations in the microbial intrauterine environment may predispose neonates to specific health outcomes later in life A role of the microbiome in the Developmental Origins of Health and Disease is supported in this collective research Highlighting the early-life critical window of susceptibility associated with microbiome development, we discuss infant microbial colonization, beginning with the maternal- to-fetal exchange of microbes in utero and up through the influence of breastfeeding in the first year of life In addition, we review the available disease-specific evidence pointing toward the microbiome as a mechanistic mediator in the Developmental Origins of Health and Disease abstract PEDIATRICS Volume 141, number 4, April 2018:e20172437 STATE-OF-THE-ART REVIEW ARTICLE To cite: Stiemsma LT and Michels KB. The Role of the Microbiome in the Developmental Origins of Health and Disease. Pediatrics. 2018;141(4): e20172437 Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, California Dr Stiemsma conceptualized and outlined the review, conducted the literature search, drafted the initial manuscript, and reviewed and revised the manuscript; Dr Michels supervised the project and critically reviewed and edited the manuscript; and all authors approved the final manuscript as submitted and agreed to be accountable for all aspects of the work. DOI: https://doi.org/10.1542/peds.2017-2437 Accepted for publication Nov 29, 2017 Address correspondence to Karin B. Michels, ScD, PhD, Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, 650 Charles E. Young Dr S, Los Angeles, CA 90095. E-mail: [email protected] PEDIATRICS (ISSN Numbers: Print, 0031-4005; Online, 1098-4275). Copyright © 2018 by the American Academy of Pediatrics FINANCIAL DISCLOSURE: The authors have indicated they have no financial relationships relevant to this article to disclose. FUNDING: Dr Stiemsma is supported by T32 training grant 5T32CA009142-37 from the National Cancer Institute (NCI), National Institutes of Health (NIH). Funded by the National Institues of Health (NIH). POTENTIAL CONFLICT OF INTEREST: The authors have indicated they have no potential conflicts of interest to disclose. NI H by guest on August 10, 2020 www.aappublications.org/news Downloaded from

The Role of the Microbiome in the Developmental …...the developmental origins field (Fig 1). earLy-LiFe DeveLOPMeNT OF The MicrObiOMe The human microbiota is a composite organism,

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: The Role of the Microbiome in the Developmental …...the developmental origins field (Fig 1). earLy-LiFe DeveLOPMeNT OF The MicrObiOMe The human microbiota is a composite organism,

During the past decade, the microbiome has emerged as a major contributor to human health.‍1 It has been suggested in current studies that the early-life microbiome is a crucial factor for proper immune development and long-term health.‍2, 3 Transient microbial dysbiosis during this time period has been associated with the development of immune-mediated, metabolic, and neurodevelopmental disorders.‍4 – 7 In addition, increasing evidence has been used to support a vital role of the maternal and intrauterine microbiomes in childhood health and development.‍8 Collectively, these findings have been used to support the microbiome as a key participant in the Developmental Origins of Health and Disease (DOHaD).‍ In this review, we will discuss the current

research surrounding the maturation of the early-life microbiome and how transient variations in this bacterial consortium can have long-term consequences for human health.‍

The DOhaD: Where DOes The MicrObiOMe FiT?

The developmental origins hypothesis proposes variations in fetal and infant programming through environmental exposures during a critical window of early life.‍9 Termed originally as the Barker hypothesis, 10, 11 in which the association between fetal malnutrition and hypertension later in life was a focus, this theory has since expanded to account for many types of early-life exposures and birth outcomes associated with long-term health and development.‍ For example, high birth

The Role of the Microbiome in the Developmental Origins of Health and DiseaseLeah T. Stiemsma, PhD, Karin B. Michels, ScD, PhD

Although the prominent role of the microbiome in human health has been established, the early-life microbiome is now being recognized as a major influence on long-term human health and development.‍ Variations in the composition and functional potential of the early-life microbiome are the result of lifestyle factors, such as mode of birth, breastfeeding, diet, and antibiotic usage.‍ In addition, variations in the composition of the early-life microbiome have been associated with specific disease outcomes, such as asthma, obesity, and neurodevelopmental disorders.‍ This points toward this bacterial consortium as a mediator between early lifestyle factors and health and disease.‍ In addition, variations in the microbial intrauterine environment may predispose neonates to specific health outcomes later in life.‍ A role of the microbiome in the Developmental Origins of Health and Disease is supported in this collective research.‍ Highlighting the early-life critical window of susceptibility associated with microbiome development, we discuss infant microbial colonization, beginning with the maternal-to-fetal exchange of microbes in utero and up through the influence of breastfeeding in the first year of life.‍ In addition, we review the available disease-specific evidence pointing toward the microbiome as a mechanistic mediator in the Developmental Origins of Health and Disease.‍

abstract

PEDIATRICS Volume 141, number 4, April 2018:e20172437 State-of-the-art review article

To cite: Stiemsma LT and Michels KB. The Role of the Microbiome in the Developmental Origins of Health and Disease. Pediatrics. 2018;141(4): e20172437

Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, California

Dr Stiemsma conceptualized and outlined the review, conducted the literature search, drafted the initial manuscript, and reviewed and revised the manuscript; Dr Michels supervised the project and critically reviewed and edited the manuscript; and all authors approved the final manuscript as submitted and agreed to be accountable for all aspects of the work.

DOi: https:// doi. org/ 10. 1542/ peds. 2017- 2437

Accepted for publication Nov 29, 2017

Address correspondence to Karin B. Michels, ScD, PhD, Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, 650 Charles E. Young Dr S, Los Angeles, CA 90095. E-mail: [email protected]

PEDIATRICS (ISSN Numbers: Print, 0031-4005; Online, 1098-4275).

Copyright © 2018 by the American Academy of Pediatrics

FiNaNciaL DiscLOsUre: The authors have indicated they have no financial relationships relevant to this article to disclose.

FUNDiNG: Dr Stiemsma is supported by T32 training grant 5T32CA009142-37 from the National Cancer Institute (NCI), National Institutes of Health (NIH). Funded by the National Institues of Health (NIH).

POTeNTiaL cONFLicT OF iNTeresT: The authors have indicated they have no potential conflicts of interest to disclose.

NIH

by guest on August 10, 2020www.aappublications.org/newsDownloaded from

Page 2: The Role of the Microbiome in the Developmental …...the developmental origins field (Fig 1). earLy-LiFe DeveLOPMeNT OF The MicrObiOMe The human microbiota is a composite organism,

weight is associated with an increase in breast and colon cancer risk, 12, 13 and the underlying mechanism of this association may be related to intrauterine exposure to high levels of growth hormones.‍9

Early-life infections and microbial exposures were not originally associated with DOHaD but were proposed as significant environmental influences on infant immune development in the hygiene hypothesis of allergic disease.‍14 With the advancement of human microbiome research, a modern extension of the hygiene hypothesis has since been proposed, known as the microflora hypothesis.‍15 In the microflora hypothesis, it is suggested that early-life environmental exposures alter the development of the human microbiome.‍15 Shifts in the composition of the microbiome are thought to bias maturation of the immune system toward a hypersensitive and/or hyperinflammatory state.‍15 For example, the innate and adaptive branches of the immune system are both highly involved in promoting an inflammatory response.‍ However, exposure to microbes typically evokes a T-helper 1–mediated response, which suppresses the T-helper 2 activity often associated with immune-mediated and hypersensitivity reactions.‍3 Discussion of the mechanisms regulating the interface between the immune system and the microbiota is beyond the scope of this review; please see Tamburini et al3 for a recent in-depth discussion on this topic.‍

Analogous to DOHaD, an early-life critical window of development has also been proposed for the microbiome.‍ Transient microbial dysbiosis (unhealthy microbial state) during this time frame has been associated with long-term immune and metabolic health issues, 2 meriting its exploration within

the developmental origins field (Fig 1).‍

earLy-LiFe DeveLOPMeNT OF The MicrObiOMe

The human microbiota is a composite organism, composed of 10 trillion to 100 trillion microbial cells (bacteria, archaea, and microbial eukaryotes) and viruses.‍16, 17 To highlight the impressive functional potential of the microbiota, the genomic catalog of this super organism, the microbiome, is composed of ∼3.‍3 million nonredundant genes.‍16 Although the terms “microbiota” and “microbiome” are descriptive of the microbial composition and genomic catalog, respectively, they are used interchangeably within this research field.‍ The following sections are devoted to delineating the initial colonization and establishment of the human bacterial microbiota in infancy, from conception through the first year of life.‍

Maternal-to-Fetal Microbial Transfer

Until recently, the intrauterine environment was perceived as sterile.‍18 However, nonpathogenic bacteria have since been detected by

molecular techniques in the amniotic fluid and placentas of healthy infants, 19, 20 suggesting a maternal-to-fetal exchange of microbes.‍ In addition, through comparisons of the amniotic, placental, and meconium microbiotas, Collado et al21 report that the meconium microbiota of infants delivered via cesarean delivery shares ∼55% of its bacterial taxa with the placenta and amniotic fluid microbiotas.‍ The prenatal maternal microbiome may also modulate the infant immune system.‍ For example, gestation-only colonization with Escherichia coli HA107 was reported to modify the intestinal mucosal innate immune system and transcriptome of the offspring.‍22

In humans, variations in the placental microbiome composition have been associated with maternal pregnancy-related (stress and gestational diabetes) and neonatal health outcomes (birth weight, preterm birth).‍7, 23 – 25 The placental microbiome of infants born preterm was also reported to differ in composition according to gestational weight gain, 26 suggesting that this bacterial consortium may mediate

STIEMSMA and MICHELS2

FiGUre 1The infant microbiome is most vulnerable to environmental influences in early life. Maternal to fetal microbial transfer, mode of birth, antibiotics, and diet can alter the colonization and maturation of the early-life microbiome. These lifestyle-induced variations in microbiome composition and function can have prolonged influences on human health and may lead to the development of disease later in life.

by guest on August 10, 2020www.aappublications.org/newsDownloaded from

Page 3: The Role of the Microbiome in the Developmental …...the developmental origins field (Fig 1). earLy-LiFe DeveLOPMeNT OF The MicrObiOMe The human microbiota is a composite organism,

fetal development depending on the health status of the mother.‍

Isolation of bacteria from the placenta is often associated with a pathophysiological state, which threatens the health of the mother and child.‍ Because of the lack of culture-based analyses of the placental microbiome, there is some controversy surrounding its validity.‍27 In addition, inclusion of appropriate controls to address background contamination is also lacking in many molecular-based studies characterizing the intrauterine microbiome.‍28

It is also possible that the placenta does not harbor any viable bacteria but rather is composed of phagocytosed microbial by-products or cell wall components.‍8, 22, 29 The lack of viable bacteria does not negate the capacity of the placental microbiome to modulate fetal development because interactions with pathogen-associated molecular patterns may still regulate cell differentiation and proliferation.‍8, 22, 29 In addition, upon implementation of stringent validation strategies for placental metagenomic analyses, the placental microbiome might be referenced as a biomarker for maternal and fetal health and disease.‍ Few studies have been conducted to explore the function of the placental microbiome, 23, 25 emphasizing an opportunity for future research in this area.‍ Analysis of microbial metabolites and the application of metatranscriptomic approaches to characterize the functional capacity of the placental microbiome will be key in defining its role in DOHaD.‍

vaginal birth: The First step in Postnatal Microbial colonization

Postnatal bacterial colonization of the infant begins during birth, at which time neonates are exposed to the maternal fecal and vaginal microbiotas.‍30 Within 24 hours of delivery, the microbiotas across various body sites (oral, skin,

meconium, etc) of cesarean delivered infants are initially populated with bacteria residing on the mother’s skin (eg, Staphylococcus spp.‍), whereas vaginally delivered infants are populated with typical vaginal bacteria (eg, Prevotella, Atopobium spp.‍).‍30 In a recent study, Chu et al31 suggest this finding may be specific to the neonatal gut microbiome.‍ In their study of 81 mother and infant dyads, the microbiomes of other body sites (nares, skin, etc) from infants delivered vaginally revealed a bimodal pattern of maternal origin, populated by both the mothers’ vaginal and skin bacteria rather than by one or the other.‍31 However, it is suggested in both studies that the microbiotas of infants are homogeneously distributed across body sites (eg, meconium, skin, nares, etc) immediately after birth.‍30

Profiling of the neonatal intestinal microbiome immediately after birth and up to age 2 years suggests that birth mode can result in prolonged infant gut microbial dysbiosis.‍32 In a study of 43 mother and infant dyads, infants delivered via cesarean delivery exhibited increased phylogenetic diversity immediately after birth.‍32 However, after 1 month of age, the phylogenetic diversity of infants delivered via cesarean delivery declined below that of vaginally delivered infants.‍32 Chu et al31 challenge this finding slightly.‍ In their recent study, birth mode was associated with variations in the microbiomes of the nares, skin, and oral cavity immediately after birth but not with variations in the infant meconium microbiome.‍ Researchers of both studies do support the influence of birth mode on neonatal colonization in general; however, more research is needed to determine the influence of this early-life factor on the microbiomes of specific body sites.‍

Because of the health benefits associated with vaginal birth, 33 Dominguez-bello et al34 conducted

the first study aimed at recolonizing infants delivered via cesarean delivery with vaginal bacteria.‍ After swabbing neonates with maternal vaginal fluids within 2 minutes of birth, the authors report partial restoration of the microbiota of infants delivered via cesarean delivery to that of vaginally delivered infants.‍34 However, the long-term health effects and composition of the infant microbiome are not yet known.‍34 Future analysis of these cesarean delivered infants exposed to the vaginal microbiome will be extremely valuable in determining the benefits of vaginally derived bacteria in long-term human health.‍ In addition, the establishment of prospective human studies and animal models attempting similar colonization with vaginal bacteria among offspring delivered via cesarean delivery will be crucial in elucidating the role of vaginal microbes in the development of disease.‍

breastfeeding Furthers Microbiota Maturation in early Life

As the neonate grows, the homogeneous microbiome populating his or her body diverges into microbe-specific body niches.‍18, 31 The maturation of the total infant microbiome has been studied for the first year of life, but beyond this time point, most researchers focus specifically on the gut microbiome.‍ Driven in large part by breastfeeding and infant diet, the human gut microbiome continues to mature until the child reaches 2 to 3 years of age, after which its composition stabilizes.‍35

Breastfeeding seeds the infant gut microbiome through contact with maternal areolar and breast milk microbes and provides key energy sources for many bacteria (human milk oligosaccharides).‍36 – 39 In a study of 107 mother and infant dyads, infants who were breastfed during the first 30 to 40 days of life

PEDIATRICS Volume 141, number 4, April 2018 3 by guest on August 10, 2020www.aappublications.org/newsDownloaded from

Page 4: The Role of the Microbiome in the Developmental …...the developmental origins field (Fig 1). earLy-LiFe DeveLOPMeNT OF The MicrObiOMe The human microbiota is a composite organism,

received a mean of ∼28% of their bacteria from breast milk and ∼10% from maternal areolar skin.‍38 The authors also report a dose-dependent association between the infant gut microbiome composition and the proportion of daily breastfeeding.‍38

There is a clear compositional distinction between breastfed and formula-fed infants, with breastfed infants being populated with higher proportions of Bifidobacteria and Lactobacillus spp.‍ and formula-fed infants being populated with a greater prevalence of clostridiales and proteobacteria.‍32, 40 In addition, formula-fed infants exhibit decreased diversity and bacterial richness even after the first year of life (12–24 months of age).‍32 In a study of 30 preterm infants, the effect of breastfeeding (versus formula feeding) was also reported to mask the influence of birth weight on the infant microbiome, highlighting breastfeeding as being potentially protective (at least with regard to the infant microbiome) against a traditional DOHaD risk factor.‍41 Epidemiologic evidence provides further support for the beneficial roles of breastfeeding in promoting infant health.‍ Formula feeding has been associated with an increased risk of various hyperinflammatory and immune-mediated diseases.‍42, 43 In addition, researchers of a recent epidemiologic study reported that breastfeeding protects against wheezing during the first year of life among infants born to mothers with asthma.‍43 With the work discussed in this section, we suggest that the microbiome may be a mediator between these associations.‍

Gestation-associated Maternal Diet shapes the Developing infant Microbiome

Recent evidence has been used to support a significant role of gestation-associated maternal diet in shaping the microbiome of infancy.‍ A maternal high-fat diet during

pregnancy and breastfeeding was reported to induce dysbiosis in the offspring microbiome of Japanese macaques.‍44 These maternal diet–induced microbial variations persisted in juvenile macaques.‍44 In addition, a postweaning, low-fat control diet was unable to correct this maternal high-fat diet–induced dysbiosis.‍44 In a prospective cohort of 26 mother and infant dyads, a high-fat maternal gestational diet was associated with distinct variations in the neonatal gut microbial composition (meconium), which persisted to 4 to 6 weeks of age.‍45 In a mouse model, a maternal high-fiber diet was associated with increased short-chain fatty acid (SCFA) production in the offspring.‍46 Emphasizing the immune-modulatory capacity of the maternal diet–driven microbiome during pregnancy, higher frequencies of thymic T-regulatory cells were also found in these pups.‍46 Finally, demonstrating the effect of maternal diet on the functional capacity of the infant microbiome, piglets born of sows that were fed a western diet (high-energy, high-fat, fructose-based diet) during pregnancy displayed decreased SCFA production.‍47 In these studies, the importance of the microbiome as a mediator linking gestation-associated maternal diet to infant health is supported, substantiating the role of the microbiome in DOHaD.‍ Future studies may shed more light on the prolonged health and developmental effects of gestation-associated maternal diet.‍

antibiotics alter infant colonization and Decrease Microbiota Maturation

Perhaps unsurprisingly, pre- and postnatal antibiotic exposure is a major early-life environmental stressor on the infant microbiome.‍ Prenatal maternal antibiotic exposure has been reported to alter the diversity of both the neonatal48 – 50 and maternal51 microbiotas.‍ Intrapartum antibiotic exposure

was also reported to alter the infant oral microbiome composition.‍52 Specifically, the bacterial families streptococcaceae and gemellaceae and the order lactobacillales were decreased, whereas bacterial families in the proteobacteria phylum were enriched among infants whose mothers received intrapartum antibiotics.‍52 In addition, among infants whose mothers received antibiotics, the infant oral microbiome composition was further differentiated depending on whether the mother received a cocktail of antibiotics compared with if she received only 1 antibiotic.‍52

Postnatal antibiotic courses given to the infant in the first 3 to 9 months of life were reported to alter abundances of specific gut bacterial taxa (namely, Ruminococcus and clostridiales).‍32 In addition, antibiotic use in the first 6 to 12 months of life has been associated with decreased maturation of the infant microbiota.‍32 This suggests antibiotics may stunt the development of the microbiota if administered during this time period, which could potentially predispose infants to microbiome-associated diseases later in life.‍32

In epidemiologic studies, it has been suggested that antibiotic-induced dysbiosis in infancy promotes the development of many noncommunicable diseases in later childhood and adulthood (ie, obesity, asthma, inflammatory bowel disease [IBD]).‍53 – 56 However, these epidemiologic analyses do not address whether these diseases are causally related to early-life antibiotic use or whether they are indicative of early-life immune deficiencies or propensity for infection.‍ Nevertheless, there are some promising animal models and prospective human cohort studies that attempt to address this quandary and provide additional support for the microbiome as a key developmental mediator in DOHaD.‍

STIEMSMA and MICHELS4 by guest on August 10, 2020www.aappublications.org/newsDownloaded from

Page 5: The Role of the Microbiome in the Developmental …...the developmental origins field (Fig 1). earLy-LiFe DeveLOPMeNT OF The MicrObiOMe The human microbiota is a composite organism,

DysbiOsis-assOciaTeD NONcOMMUNicabLe Diseases PrOviDe aDDiTiONaL sUPPOrT FOr The MicrObiOMe iN DOhaD

The critical Window in early Life

As mentioned at the beginning of this review, DOHaD emphasizes a critical window of susceptibility from conception to early life in which environmental stressors most profoundly affect long-term human health.‍ A similar critical window has emerged for the development of the microbiome.‍ Scientists have narrowed the microbiome early-life critical window to the time period between conception and the first year of life (Fig 1).‍1 Though more research is needed to confirm this theory, the microbiome’s vulnerability to environmental influences appears to be time varying, more substantial earlier in life and lessening as it matures toward that of an adult (Fig 1).‍ In the following sections, we will discuss disease-specific research (specifically, necrotizing enterocolitis [NEC], asthma and atopic disease, obesity, and neurodevelopmental disorders) that points toward this early-life critical window and further supports the role of the microbiome in DOHaD (summarized in Table 1).‍

Nec

In addition to the health risks associated with preterm birth, preterm infants display marked neonatal microbial dysbiosis.‍ This dysbiosis enhances their susceptibility to disease, particularly NEC, which may be modified by exposure to antibiotics during this critical time period.‍57 Through analysis of existing 16S ribosomal RNA gene sequence data, the authors identified differential abundances of proteobacteria, firmicutes, and bacteroidetes, which preceded the onset of NEC.‍57 Gut dysbiosis characterized by similar variations in bacterial taxa was also reported to precede NEC in a study of 122 extremely

low birth weight neonates.‍58 The increase in proteobacteria along with increased enterocyte Toll-like receptor 4 activity in these neonates with NEC suggest a hyperinflammatory response to a dysbiotic microbiome.‍59 However, a recent study was conducted in which researchers identified uropathogenic E coli colonization as a significant risk marker for NEC.‍59 This suggests an invasive microbial species may be synergistic in driving this dysbiosis-associated disorder.‍ In addition, infants treated with antibiotics for 7–14 days (regardless of NEC status) were enriched for E coli relative to infants treated with antibiotics for 0–6 days.‍59 This supports a role of neonatal antibiotic-induced dysbiosis in NEC, which may enhance the vulnerability of the neonatal gut microbiome to pathogen invasion.‍ Because of the apparent link between microbial dysbiosis and NEC, probiotic supplementation of preterm neonates is becoming a prominent research area for NEC prevention and treatment.‍109, 110 In a systematic review and meta-analysis of 26 probiotic intervention studies for NEC, it is suggested that probiotic intervention does prevent NEC.‍60 However, the particular strains to be used and the effects on high-risk populations (extremely low birth weight infants) requires further study.‍60 Nonetheless, it may be possible in the near future to optimize the neonatal microbiome to combat development of this disease and prevent associated infant mortality.‍

asthma and atopic Disease

NEC studies highlight the dynamic relation between the gut microbiome and the developing immune system.‍ However, although the neonatal immune system is on high alert for pathogenic invaders, researchers who have conducted studies of asthma and atopic diseases (food allergies, atopic dermatitis, etc)

suggest its development is strongly influenced by commensal bacteria.‍

In a recent assessment of the microbiome in asthma development, a more substantial role of the maternal microbiota in fetal development is supported than what was previously thought.‍ In a mouse model of allergic airway inflammation, high-fiber (which has been reported to stimulate production of SCFAs by the microbiome111) or acetate (a SCFA) feeding of pregnant mice modulated the maternal microbiota and protected the subsequent offspring from developing allergic airway disease.‍73 The authors of this study also provided preliminary evidence of this association in humans; high acetate levels in the serum of pregnant individuals correlated with reduced general practitioner visits for coughing and wheezing in the offspring during the first 12 months of life.‍73

Postnatal early-life dysbiosis in both the human gut2, 4, 5, 70 and airway78 microbiomes is also associated with asthma and atopic disease development in children.‍ This dysbiosis is characterized by shifts in the prevalence of specific bacterial4, 5 and fungal taxa, 70 which are transient and most prominent between birth and 3 months of life.‍ The majority of human studies in this research area reveal only correlative evidence to link the early-life dysbiosis with asthma.‍ However, Arrieta et al4 provide preliminary evidence of causality related to early-life transient dysbiosis and immune development in the context of asthma.‍ In this study, inoculation of previously germ-free (GF) mice with 4 bacterial species, which were reduced in infants at high risk of asthma, ameliorated allergic airway inflammation in these mice.‍4

There is also evidence to support a role of the early-life microbiome in food allergies.‍ Azad et al74 report a decrease in microbial diversity at

PEDIATRICS Volume 141, number 4, April 2018 5 by guest on August 10, 2020www.aappublications.org/newsDownloaded from

Page 6: The Role of the Microbiome in the Developmental …...the developmental origins field (Fig 1). earLy-LiFe DeveLOPMeNT OF The MicrObiOMe The human microbiota is a composite organism,

STIEMSMA and MICHELS6

TabL

e 1

Stud

ies

in W

hich

Res

earc

hers

Rel

ate

the

Earl

y-Li

fe M

icro

biom

e W

ith H

ealth

Out

com

es in

Lat

er C

hild

hood

and

Adu

lthoo

d

Rese

arch

Obj

ectiv

eSt

udy

Popu

latio

n or

An

imal

Mod

elEa

rly-

Life

Fac

tors

and

Ti

min

g of

Exp

osur

eTi

min

g of

Mic

robi

ome

Anal

ysis

or

Inte

rven

tion

Heal

th O

utco

me

and

Tim

ing

of A

sses

smen

tSu

mm

ary

of F

indi

ngs

Ref.

No.

NEC

To

ass

ess

mic

robi

al

dysb

iosi

s be

fore

NEC

in

a sy

stem

atic

rev

iew

and

m

eta-

anal

ysis

Syst

emat

ic r

evie

w

and

met

a-an

alys

is

of 1

4 hu

man

feca

l m

icro

biom

e st

udie

s of

NEC

None

Vari

atio

ns in

the

mic

robi

ome

befo

re N

EC

deve

lopm

ent

NEC

at ∼

30 w

k po

stco

ncep

tion

Incr

ease

d pr

oteo

bact

eria

and

dec

reas

ed

firm

icut

es a

nd b

acte

roid

etes

pre

cede

d NE

C on

set.

Antib

iotic

s, d

iet,

and

mod

e of

del

iver

y do

con

trib

ute

to m

icro

bial

dy

sbio

sis

asso

ciat

ed w

ith N

EC. H

owev

er,

caus

ality

rel

ated

to th

ese

fact

ors

cann

ot b

e de

term

ined

57

To

det

erm

ine

if 1

or m

ore

gut b

acte

rial

taxa

diff

er

betw

een

case

s of

NEC

and

co

ntro

ls

Pros

pect

ive

hum

an c

ohor

t an

alys

is (

prim

ary

coho

rt, n

= 1

22;

seco

ndar

y co

hort

s,

n =

44)

None

Vari

atio

ns in

the

mic

robi

ome

befo

re N

EC

deve

lopm

ent

NEC

in v

ery

low

bir

th w

t in

fant

sIn

crea

ses

in g

amm

apro

teob

acte

ria

and

decr

ease

s in

neg

ativ

icut

es a

nd c

lost

ridi

a-ne

gativ

icut

es c

lass

es o

ver

time

prec

eded

NE

C de

velo

pmen

t

58

To

enh

ance

str

ain-

leve

l re

solu

tion

of N

EC-

asso

ciat

ed p

atho

gens

by

usi

ng d

eep

shot

gun

met

agen

omic

s se

quen

cing

Pros

pect

ive

hum

an c

ohor

t an

alys

is (

n =

166)

None

Vari

atio

ns in

the

mic

robi

ome

befo

re N

EC

deve

lopm

ent

NEC

in in

fanc

yVa

riat

ions

in th

e m

icro

biom

e w

ere

dete

cted

be

fore

NEC

dev

elop

men

t. Ho

wev

er, a

t 17–

22

d po

stpa

rtum

, inf

ants

with

hig

h an

tibio

tic

trea

tmen

t wer

e en

rich

ed fo

r E

coli.

The

gr

oup

late

r id

entifi

ed u

ropa

thog

enic

E

coli

as a

maj

or r

isk

fact

or fo

r NE

C an

d as

soci

ated

dea

th

59

To

com

pare

the

effic

acy

and

safe

ty o

f ent

eral

pr

obio

tic a

dmin

istr

atio

n in

pre

vent

ing

NEC

Syst

emat

ic r

evie

w a

nd

met

a-an

alys

is o

f 24

rand

omiz

ed o

r qu

asi-

rand

omiz

ed c

ontr

olle

d tr

ials

in h

uman

s

None

Ente

ral p

robi

otic

su

pple

men

tatio

n be

fore

NE

C de

velo

pmen

t

Stag

e II

and

stag

e III

NEC

in

infa

ncy

Ente

ral p

robi

otic

sup

plem

enta

tion

sign

ifica

ntly

red

uced

the

inci

denc

e of

NEC

an

d m

orta

lity

in in

fant

s

60

To

ass

ess

the

inte

stin

al

mic

robi

ota

com

posi

tion

befo

re N

EC d

evel

opm

ent

in in

fant

s w

ho d

evel

oped

NE

C an

d co

ntro

ls

Pros

pect

ive

hum

an c

ohor

t an

alys

is (

n =

38)

None

Vari

atio

ns in

the

mic

robi

ome

befo

re N

EC

deve

lopm

ent

NEC

in in

fanc

yAn

ave

rage

of 7

sam

ples

wer

e co

llect

ed

per

subj

ect a

nd th

e te

mpo

ral c

hang

es

in m

icro

biom

e co

mpo

sitio

n w

ere

asse

ssed

. Thr

ough

out e

arly

life

, bef

ore

the

deve

lopm

ent o

f NEC

, diff

eren

t mic

robi

al

popu

latio

ns d

omin

ate

the

gut a

nd a

re

asso

ciat

ed w

ith N

EC d

evel

opm

ent.

In

addi

tion,

the

mic

robi

ome

com

posi

tiona

l pr

ogre

ssio

n ap

pear

s to

be

asso

ciat

ed w

ith

the

timin

g of

NEC

ons

et

61

To

iden

tify

mic

robi

al a

nd

met

abol

ic b

iom

arke

rs

of N

EC

Nest

ed c

ase-

cont

rol

desi

gn (

n =

35)

None

Vari

atio

ns in

the

mic

robi

ome

befo

re N

EC

deve

lopm

ent

NEC

in in

fanc

yLo

wer

α d

iver

sity

4–9

d p

ostb

irth

was

as

soci

ated

with

NEC

dev

elop

men

t. M

icro

biom

es o

f sub

ject

s te

nded

to c

lust

er

acco

rdin

g to

NEC

sta

tus.

The

se m

icro

bial

va

riat

ions

wer

e as

soci

ated

with

shi

fts

in

urin

e m

etab

olite

s, n

amel

y al

anin

e an

d hi

stid

ine

62

by guest on August 10, 2020www.aappublications.org/newsDownloaded from

Page 7: The Role of the Microbiome in the Developmental …...the developmental origins field (Fig 1). earLy-LiFe DeveLOPMeNT OF The MicrObiOMe The human microbiota is a composite organism,

PEDIATRICS Volume 141, number 4, April 2018 7

Rese

arch

Obj

ectiv

eSt

udy

Popu

latio

n or

An

imal

Mod

elEa

rly-

Life

Fac

tors

and

Ti

min

g of

Exp

osur

eTi

min

g of

Mic

robi

ome

Anal

ysis

or

Inte

rven

tion

Heal

th O

utco

me

and

Tim

ing

of A

sses

smen

tSu

mm

ary

of F

indi

ngs

Ref.

No.

To

det

erm

ine

if gu

t m

icro

biom

e co

mpo

sitio

n ca

n be

use

d to

pre

dict

NE

C se

veri

ty

Pros

pect

ive

hum

an c

ohor

t an

alys

is (

n =

30)

None

Vari

atio

ns in

the

mic

robi

ome

befo

re N

EC

deve

lopm

ent

NEC

in in

fanc

yVa

riat

ions

in th

e m

icro

biom

e w

ere

not

asso

ciat

ed w

ith N

EC s

ever

ity. T

here

wer

e al

so n

o di

ffere

nces

in th

e m

icro

biom

e po

st-

NEC

com

pare

d w

ith c

ontr

ols

63

To

ass

ess

whe

ther

fe

cal m

icro

biot

a tr

ansp

lant

atio

n is

an

effe

ctiv

e tr

eatm

ent o

f NEC

Wild

-type

and

Grx

1−/−

m

ouse

mod

els

of N

ECNo

neFe

cal m

icro

biot

a tr

ansp

lant

fr

om 1

to 4

d p

ostb

irth

NEC

5 d

post

birt

hFe

cal m

icro

biot

a tr

ansp

lant

from

hea

lthy

6–8-

wk-

old

mic

e to

mou

se p

ups

cond

ition

ed

for

NEC

redu

ced

NEC

inci

denc

e an

d se

veri

ty c

ompa

red

with

con

trol

s. T

his

was

de

pend

ent o

n Gr

x1. T

he m

echa

nism

of

actio

n is

pot

entia

lly th

roug

h TL

R-m

edia

ted

infla

mm

atio

n an

d gu

t per

mea

bilit

y

64

To

det

erm

ine

if pa

tient

s at

ris

k fo

r NE

C ca

n be

iden

tified

by

thei

r m

econ

ium

and

ear

ly

post

nata

l mic

robi

ota

Pros

pect

ive

hum

an c

ohor

t an

alys

is (

n =

33)

Earl

y en

tera

l fee

ding

and

br

east

milk

Vari

atio

ns in

mec

oniu

m

mic

robi

ome

and

neon

atal

m

icro

biom

e be

fore

NEC

NEC

in in

fanc

yCl

ostr

idiu

m p

erfr

inge

ns a

nd B

acte

roid

es

dore

i wer

e in

crea

sed

in th

e m

econ

ium

of

infa

nts

who

dev

elop

ed N

EC. C

per

frin

gens

ab

unda

nce

pers

iste

d in

neo

nata

l sto

ol

sam

ples

. The

am

ount

of b

reas

t milk

be

fore

NEC

and

ear

lier

ente

ral f

eedi

ng

was

neg

ativ

ely

asso

ciat

ed w

ith N

EC

and

asso

ciat

ed w

ith in

crea

sed

lact

ate-

prod

ucin

g ba

cilli

65

To

com

pare

ent

eral

ver

sus

pare

nter

al a

ntib

iotic

s in

pre

vent

ing

form

ula-

indu

ced

NEC

lesi

ons

in

pigs

Pigl

et m

odel

of N

ECAn

tibio

tic-in

duce

dEn

tera

l and

par

ente

ral

antib

iotic

trea

tmen

t (fo

r 5

d po

st b

irth

)

NEC

in p

rete

rm p

igle

tsEn

tera

l ant

ibio

tics

prev

ente

d NE

C le

sion

s,

whe

reas

lesi

ons

in p

igle

ts th

at w

ere

trea

ted

with

par

ente

ral a

ntib

iotic

s w

ere

incr

ease

d. E

nter

al a

ntib

iotic

s de

crea

sed

bact

eria

l loa

d an

d ab

unda

nces

of

Gram

-pos

itive

bac

teri

a in

the

inte

stin

e.

It is

sug

gest

ed th

at d

elay

ed c

olon

izat

ion

(par

ticul

arly

with

Gra

m-p

ositi

ve b

acte

ria)

m

ay p

reve

nt N

EC. H

owev

er, a

lthou

gh

mic

robi

ome

vari

atio

ns c

orre

late

with

NEC

, th

ey d

o no

t nec

essa

rily

pre

cede

NEC

66

To

det

erm

ine

if to

tal

pare

nter

al n

utri

tion,

be

fore

the

star

t of

ente

ral f

eedi

ng, c

an

prev

ent N

EC-a

ssoc

iate

d gu

t dys

func

tion

and

infla

mm

atio

n

Pigl

et m

odel

of N

ECEn

tera

l and

tota

l par

ente

ral

nutr

ition

Vari

atio

ns in

the

mic

robi

ome

afte

r fe

edin

g m

etho

ds

NEC

in p

rete

rm p

igle

tsEn

tera

l fee

ding

incr

ease

d m

icro

bial

div

ersi

ty

and

the

abun

danc

e of

Clo

stri

dium

spe

cies

. De

nsity

of C

per

frin

gens

was

ass

ocia

ted

with

NEC

sev

erity

. Mic

robi

ome

vari

atio

ns

corr

elat

e w

ith N

EC b

ut d

o no

t nec

essa

rily

pr

eced

e NE

C

67

To

iden

tify

mic

robi

al p

rofil

es

befo

re N

EC d

iagn

osis

Pros

pect

ive

hum

an c

ohor

t an

alys

is (

n =

369)

None

Vari

atio

ns in

the

mic

robi

ome

befo

re N

EC

diag

nosi

s

NEC

in in

fanc

yId

entifi

catio

n of

2 fe

cal m

icro

biot

a pr

ofile

s as

soci

ated

with

NEC

dev

elop

men

t (C

perf

ring

ens

type

A d

omin

ant a

nd K

lebs

iella

do

min

ant)

68

TabL

e 1

Cont

inue

d

by guest on August 10, 2020www.aappublications.org/newsDownloaded from

Page 8: The Role of the Microbiome in the Developmental …...the developmental origins field (Fig 1). earLy-LiFe DeveLOPMeNT OF The MicrObiOMe The human microbiota is a composite organism,

STIEMSMA and MICHELS8

Rese

arch

Obj

ectiv

eSt

udy

Popu

latio

n or

An

imal

Mod

elEa

rly-

Life

Fac

tors

and

Ti

min

g of

Exp

osur

eTi

min

g of

Mic

robi

ome

Anal

ysis

or

Inte

rven

tion

Heal

th O

utco

me

and

Tim

ing

of A

sses

smen

tSu

mm

ary

of F

indi

ngs

Ref.

No.

To

cha

ract

eriz

e ep

igen

ome

to m

icro

biom

e cr

osst

alk

at c

ritic

al n

eona

tal s

tage

s of

dev

elop

men

t

Tiss

ue-b

ased

(im

mat

ure

ente

rocy

tes)

and

m

ouse

mod

els

(dex

amet

haso

ne o

r 5-

azac

ytid

ine

to in

duce

ep

igen

etic

cha

nges

)

Pren

atal

dex

amet

haso

ne o

r 5-

azac

ytid

ine

trea

tmen

tM

icro

biom

e to

epi

geno

me

cros

stal

k in

per

inat

al li

feTL

R an

d tig

ht ju

nctio

n-si

gnal

ing

path

way

s in

of

fspr

ing

Pren

atal

dex

amet

haso

ne a

nd a

zacy

tidin

e tr

eatm

ent a

lters

DNA

met

hyla

tion

of ti

ght

junc

tion

and

TLR

gene

s an

d as

soci

ated

in

flam

mat

ory

path

way

s in

fetu

ses

and

guts

of 2

-wk-

old

offs

prin

g. B

oth

pren

atal

ex

posu

res

also

alte

red

the

offs

prin

g m

icro

biom

e. A

zacy

tidin

e tr

eatm

ent i

nduc

es

glob

al d

emet

hyla

tion,

sug

gest

ing

that

the

pre-

and

neo

nata

l epi

geno

me

influ

ence

s ne

onat

al m

icro

bial

col

oniz

atio

n

69

Asth

ma

and

atop

ic d

isea

se

To a

naly

ze th

e m

icro

biom

e of

infa

nts

befo

re a

topi

c di

seas

e de

velo

pmen

t at 1

y

of a

ge

Nest

ed c

ase-

cont

rol

desi

gn (

n =

319)

and

Ov

albu

min

-cha

lleng

ed

mou

se m

odel

s of

as

thm

a

None

Vari

atio

ns in

the

3-m

o-ol

d gu

t mic

robi

ome

Atop

y an

d w

heez

ing

at 1

y

of a

geFo

ur b

acte

rial

taxa

(Fa

ecal

ibac

teri

um,

Lach

nosp

ira,

Vei

llone

lla, a

nd R

othi

a) w

ere

decr

ease

d am

ong

infa

nts

with

ato

py a

nd

whe

ezin

g at

1 y

of a

ge. S

uppl

emen

tatio

n of

as

thm

a-in

duce

d m

ice

with

thes

e 4

bact

eria

am

elio

rate

d ai

rway

infla

mm

atio

n

4

To

ana

lyze

the

mic

robi

ota

of in

fant

s be

fore

ast

hma

deve

lopm

ent b

y 4

y of

age

Nest

ed c

ase-

cont

rol

desi

gn (

n =

319)

None

Vari

atio

ns in

the

3-m

o-ol

d gu

t mic

robi

ome

Asth

ma

by 4

y o

f age

Decr

ease

d La

chno

spir

a/Cl

ostr

idiu

m n

eona

tale

ra

tio a

t 3 m

o of

age

was

ass

ocia

ted

with

as

thm

a by

4 y

of a

ge

5

To

cha

ract

eriz

e th

e ba

cter

ial

and

fung

al m

icro

biom

es

in n

eona

tes

befo

re

asth

ma

deve

lopm

ent a

t 4

y of

age

Pros

pect

ive

hum

an c

ohor

t an

alys

is (

n =

168)

None

Vari

atio

ns in

gut

m

icro

biom

e 35

d

post

birt

h

Asth

ma

at 4

y o

f age

Vari

atio

ns in

bac

teri

al a

nd fu

ngal

taxa

at 3

5 d

post

birt

h as

soci

ated

with

hig

hest

rel

ativ

e ri

sk o

f ast

hma.

Ste

rile

feca

l wat

er fr

om

the

high

est-r

isk

grou

p in

duce

s CD

4+ T-

cell

dysf

unct

ion

70

To

cha

ract

eriz

e th

e ea

rly-

life

criti

cal

win

dow

ass

ocia

ted

with

ex

acer

bate

d al

lerg

ic

airw

ays

resp

onse

s in

m

ice

Oval

bum

in-c

halle

nged

m

ouse

mod

elAn

tibio

tic-in

duce

dPe

rina

tal (

in u

tero

and

th

roug

h w

eani

ng)

Asth

ma

indu

ced

late

r in

life

Peri

nata

l van

com

ycin

exp

osur

e pr

omot

es

expa

nsio

n of

firm

icut

es a

nd e

xace

rbat

es

airw

ay in

flam

mat

ion

in m

ice

71

To

ana

lyze

the

effe

cts

of

peri

nata

l ant

ibio

tic

trea

tmen

t on

deve

lopm

ent

of h

yper

sens

itivi

ty

pneu

mon

itis

Th1/

Th17

-med

iate

d m

ouse

m

odel

Antib

iotic

-indu

ced

Peri

nata

l ant

ibio

tic

expo

sure

Hype

rsen

sitiv

ity

pneu

mon

itis

in

adul

thoo

d

Peri

nata

l str

epto

myc

in p

rom

otes

exp

ansi

on

of b

acte

roid

etes

and

res

ults

in e

xagg

erat

ed

hype

rsen

sitiv

ity p

neum

oniti

s

72

To

cha

ract

eriz

e th

e ce

llula

r m

echa

nism

s as

soci

ated

w

ith d

iet o

r m

icro

biot

a-m

edia

ted

imm

une

regu

latio

n

Wild

-type

, Gpr

43−

/−, a

nd

HDAC

9−/−

hou

se d

ust

mite

mou

se m

odel

s of

AA

D; n

este

d ca

se-

cont

rol d

esig

n (n

= 4

0)

Mat

erna

l ace

tate

or

high

-fib

er d

iet;

mat

erna

l se

rum

leve

ls o

f ace

tate

Pren

atal

ant

ibio

tic

expo

sure

; pre

nata

l ac

etat

e ex

posu

re

AAD

indu

ced

in a

dulth

ood;

co

ughi

ng a

nd w

heez

e by

1

y of

age

High

-fibe

r di

et o

r ac

etat

e fe

edin

g of

dam

s in

pre

gnan

cy p

reve

nts

robu

st A

AD in

ad

ult o

ffspr

ing.

Mat

erna

l ser

um le

vels

of

acet

ate

wer

e in

vers

ely

asso

ciat

ed w

ith

gene

ral p

ract

ition

er v

isits

for

coug

hing

and

w

heez

ing

in th

e fir

st 1

2 m

o of

life

73

TabL

e 1

Cont

inue

d

by guest on August 10, 2020www.aappublications.org/newsDownloaded from

Page 9: The Role of the Microbiome in the Developmental …...the developmental origins field (Fig 1). earLy-LiFe DeveLOPMeNT OF The MicrObiOMe The human microbiota is a composite organism,

PEDIATRICS Volume 141, number 4, April 2018 9

Rese

arch

Obj

ectiv

eSt

udy

Popu

latio

n or

An

imal

Mod

elEa

rly-

Life

Fac

tors

and

Ti

min

g of

Exp

osur

eTi

min

g of

Mic

robi

ome

Anal

ysis

or

Inte

rven

tion

Heal

th O

utco

me

and

Tim

ing

of A

sses

smen

tSu

mm

ary

of F

indi

ngs

Ref.

No.

To

ana

lyze

the

infa

nt

mic

robi

ota

in a

ssoc

iatio

n w

ith fo

od s

ensi

tizat

ion

Nest

ed c

ase-

cont

rol

desi

gn (

n =

166)

None

Vari

atio

ns in

the

gut

mic

robi

ome

at 3

mo

of

age

Food

sen

sitiv

ity a

t 1 y

of

age

Decr

ease

d α

dive

rsity

and

incr

ease

d en

tero

bact

eria

ceae

/bac

tero

idac

eae

ratio

as

soci

ated

with

food

sen

sitiz

atio

n to

at

leas

t 1 fo

od a

llerg

en (

milk

, egg

, pea

nut,

soy)

at 1

y o

f age

.

74

To

ana

lyze

the

earl

y-lif

e m

icro

biot

a in

ass

ocia

tion

with

res

olut

ion

of c

ow’s

m

ilk a

llerg

y

Nest

ed c

ase-

cont

rol

desi

gn (

n =

226

subj

ects

with

milk

al

lerg

y)

None

Vari

atio

ns in

the

gut

mic

robi

ome

at 3

–6 m

o of

age

Milk

alle

rgy

reso

lutio

n by

8

y of

age

Firm

icut

es a

nd c

lost

ridi

a w

ere

enri

ched

in

mic

robi

omes

of s

ubje

cts

who

se m

ilk

alle

rgy

reso

lved

by

8 y

of a

ge. B

acte

roid

etes

an

d En

tero

bact

er w

ere

enri

ched

am

ong

thos

e w

hose

milk

alle

rgy

did

not r

esol

ve b

y 8

y of

age

75

To

inve

stig

ate

age-

depe

nden

t m

icro

bial

mod

ulat

ion

of

iNKT

s in

mou

se m

odel

s of

IB

D an

d as

thm

a

Oval

bum

in-c

halle

nged

m

ouse

mod

elNo

neEx

posu

re to

mat

erna

l gut

m

icro

biom

e at

bir

thAs

thm

a in

duce

d la

ter

in li

feNe

onat

al e

xpos

ure

to a

con

vent

iona

l m

icro

biot

a (c

ompa

red

with

GF

cond

ition

s)

incr

ease

d iN

KT in

the

lung

s an

d pr

otec

ted

agai

nst a

llerg

ic a

sthm

a in

duce

d in

ad

ulth

ood.

In a

dditi

on, h

ypom

ethy

latio

n of

CXC

L16,

dri

ven

by th

e m

icro

biom

e, w

as

asso

ciat

ed w

ith iN

KT in

duct

ion,

impl

icat

ing

the

mic

robi

ome

in g

ene

regu

latio

n

76

To

ana

lyze

the

role

of t

he

earl

y-lif

e lu

ng m

icro

biot

a in

alle

rgen

-indu

ced

airw

ay

infla

mm

atio

n

Mou

se m

odel

of h

ouse

du

st m

ite–i

nduc

ed

airw

ay in

flam

mat

ion

None

2-w

k w

indo

w o

f su

scep

tibili

ty (

lung

m

icro

biom

e)

Asth

ma

indu

ced

late

r in

life

Vari

atio

ns in

the

lung

mic

robi

ome

(shi

ft fr

om

gam

map

rote

obac

teri

a an

d fir

mic

utes

to

bact

eroi

dete

s) a

ssoc

iate

d w

ith d

ecre

ased

ae

roal

lerg

en r

espo

nsiv

enes

s an

d in

crea

sed

Helio

s− T-

regu

lato

ry c

ells

. Thi

s is

med

iate

d by

PD-

L1. B

lock

age

of P

D-L1

in th

e fir

st 2

wk

of li

fe r

esul

ts in

enh

ance

d al

lerg

ic a

irw

ay

infla

mm

atio

n

77

To

ana

lyze

the

naso

phar

ynge

al

mic

robi

ome

in in

fanc

y in

ass

ocia

tion

with

re

spir

ator

y di

seas

e la

ter

in li

fe

Pros

pect

ive

hum

an c

ohor

t st

udy

(n =

234

)No

neVa

riat

ions

in th

e na

soph

aryn

geal

m

icro

biom

e 7–

9 w

k po

stbi

rth

Chro

nic

whe

ezin

g at

5–1

0 y

of a

geCh

ildre

n w

ho d

evel

oped

chr

onic

whe

ezin

g at

5–

7 y

of a

ge a

nd w

ere

atop

ic b

y ag

e 2

wer

e tw

ice

as li

kely

to h

ave

been

col

oniz

ed w

ith

asym

ptom

atic

Str

epto

cocc

us

78

To

det

erm

ine

if va

riat

ions

in

the

skin

mic

robi

ome

in

earl

y lif

e ar

e as

soci

ated

w

ith a

topi

c de

rmat

itis

Nest

ed c

ase-

cont

rol

desi

gn (

n =

20)

None

Vari

atio

ns in

the

skin

(a

ntec

ubita

l fos

sa)

mic

robi

ome

at 2

mo

of

age

Atop

ic d

erm

atiti

s at

1 y

of

age

Colo

niza

tion

of a

ntec

ubita

l fos

sa a

t 2 m

o of

ag

e w

ith S

taph

yloc

occu

s w

as a

ssoc

iate

d w

ith d

ecre

ased

inci

denc

e of

ato

pic

derm

atiti

s at

1 y

of a

ge

79

To

ana

lyze

ass

ocia

tions

be

twee

n ne

onat

al g

ut

Bifid

obac

teri

um s

peci

es

and

ecze

ma

or a

topy

de

velo

pmen

t in

the

first

ye

ar o

f life

Nest

ed c

ase-

cont

rol

desi

gn (

n =

117)

Colo

niza

tion

patt

erns

in

fluen

ced

by h

ouse

hold

pe

ts, n

umbe

r of

sib

lings

, an

d m

ater

nal a

llerg

ic

stat

us

Vari

atio

ns in

Bi

fidob

acte

rium

spe

cies

at

1 w

k an

d 3

mo

of a

ge

Atop

ic d

erm

atiti

s at

1 y

of

age

Vari

atio

ns in

Bifi

doba

cter

ium

spe

cies

at 1

w

k an

d 3

mo

of a

ge w

ere

asso

ciat

ed w

ith

risk

of e

czem

a at

1 y

of a

ge. H

owev

er, t

he

mic

robi

ome

was

not

ana

lyze

d af

ter

3 m

o of

age

80

TabL

e 1

Cont

inue

d

by guest on August 10, 2020www.aappublications.org/newsDownloaded from

Page 10: The Role of the Microbiome in the Developmental …...the developmental origins field (Fig 1). earLy-LiFe DeveLOPMeNT OF The MicrObiOMe The human microbiota is a composite organism,

STIEMSMA and MICHELS10

Rese

arch

Obj

ectiv

eSt

udy

Popu

latio

n or

An

imal

Mod

elEa

rly-

Life

Fac

tors

and

Ti

min

g of

Exp

osur

eTi

min

g of

Mic

robi

ome

Anal

ysis

or

Inte

rven

tion

Heal

th O

utco

me

and

Tim

ing

of A

sses

smen

tSu

mm

ary

of F

indi

ngs

Ref.

No.

To

ana

lyze

ass

ocia

tions

be

twee

n pr

opor

tions

of

IgA

coat

ing

and

bact

eria

bo

und

to Ig

A in

infa

ncy

and

alle

rgy

deve

lopm

ent

late

r in

life

Nest

ed c

ase-

cont

rol

desi

gn (

n =

48)

None

IgA

and

tota

l bac

teri

al lo

ad

mea

sure

d at

1 w

k an

d 1

y of

age

Asth

ma,

alle

rgic

rh

inoc

onju

nctiv

itis,

al

lerg

ic u

rtic

aria

, and

ec

zem

a by

7 y

of a

ge

At 1

2 m

o of

age

, chi

ldre

n w

ith a

llerg

ic d

isea

se

(par

ticul

arly

ast

hma)

dis

play

ed a

low

er

prop

ortio

n of

IgA

boun

d to

feca

l bac

teri

a.

IgA

reco

gniti

on p

atte

rns

for

the

mic

robi

ota

vari

ed b

etw

een

child

ren

with

alle

rgie

s an

d he

alth

y ch

ildre

n at

1 w

k of

age

81

To

ana

lyze

feca

l mic

robi

al

dive

rsity

and

bac

teri

al

abun

danc

es in

the

first

ye

ar o

f life

in a

ssoc

iatio

n w

ith a

sthm

a an

d al

lerg

ies

late

r in

life

Nest

ed c

ase-

cont

rol

desi

gn (

n =

47)

None

Mic

robi

al d

iver

sity

at 1

mo

of a

geAs

thm

a at

7 y

of a

geCh

ildre

n w

ith a

sthm

a di

spla

yed

low

er o

vera

ll m

icro

bial

div

ersi

ty th

an c

hild

ren

with

out

asth

ma

82

To

inve

stig

ate

the

infa

nt

inte

stin

al m

icro

biot

a co

mpo

sitio

n in

as

soci

atio

n w

ith m

ater

nal

pren

atal

str

ess

and

infa

nt

heal

th

Nest

ed c

ase-

cont

rol

desi

gn (

n =

56)

Pren

atal

str

ess

Vari

atio

ns in

the

mic

robi

ome

at p

ostn

atal

da

ys 7

, 14,

28,

80,

and

110

GI s

ympt

oms

and

alle

rgic

re

spon

se b

y 3

mo

of a

geIn

fant

s ex

pose

d to

pre

nata

l str

ess

disp

laye

d m

ore

GI s

ympt

oms

(38%

com

pare

d w

ith 2

2%)

and

alle

rgic

rea

ctio

ns (

43%

co

mpa

red

with

0%

), w

hich

wer

e as

soci

ated

w

ith v

aria

tions

in th

e m

icro

biom

e. T

his

mic

robi

ome

was

cha

ract

eriz

ed b

y le

ss

lact

ic a

cid

bact

eria

and

Akk

erm

ansi

a an

d gr

eate

r Es

cher

ichi

a, E

nter

obac

ter,

and

Se

rrat

ia

83

Obes

ity a

nd m

etab

olis

m

To a

naly

ze if

the

earl

y-lif

e m

icro

biot

a co

mpo

sitio

n is

as

soci

ated

with

chi

ldho

od

BMI a

nd if

ant

ibio

tic u

se

mod

ifies

this

ass

ocia

tion

Nest

ed c

ase-

cont

rol

desi

gn fr

om 2

coh

orts

(B

ibo

coho

rt, n

= 8

7;

Flor

a co

hort

, n =

75)

Antib

iotic

trea

tmen

tVa

riat

ions

in th

e gu

t m

icro

biom

e at

3 m

o of

ag

e

BMI a

t 5–6

yIn

the

3-m

o-ol

d m

icro

biom

e, r

elat

ive

abun

danc

e of

str

epto

cocc

i was

pos

itive

ly

asso

ciat

ed w

ith B

MI a

t 5–6

y o

f age

, and

re

lativ

e ab

unda

nce

of b

ifido

bact

eria

was

ne

gativ

ely

asso

ciat

ed w

ith B

MI a

t 5–6

y

of a

ge. A

mon

g ch

ildre

n w

ith a

his

tory

of

mul

tiple

ant

ibio

tic c

ours

es, t

he fi

rmic

utes

ph

ylum

was

sig

nific

antly

ass

ocia

ted

with

BM

I. Ho

wev

er, m

icro

biom

e co

mpo

sitio

n w

as

not m

easu

red

at a

ny o

ther

tim

e po

int

84

To

ana

lyze

the

impa

ct o

f di

et o

n th

e ea

rly-

life

mic

robi

ome

in a

pri

mat

e m

odel

Prim

ate

mod

el (

high

-fat

vers

us s

tand

ard

diet

)M

ater

nal h

igh-

fat d

iet

duri

ng p

regn

ancy

and

br

east

feed

ing

Vari

atio

ns in

the

offs

prin

g m

icro

biom

e co

mpo

sitio

nSh

ifts

in m

icro

bial

m

etab

olic

pat

hway

sIn

this

stu

dy, a

link

to a

spe

cific

hea

lth

outc

ome

was

not

est

ablis

hed.

How

ever

, a

mat

erna

l hig

h-fa

t die

t did

alte

r th

e m

icro

biom

e co

mpo

sitio

n of

offs

prin

g m

acaq

ues,

whi

ch p

ersi

sted

in ju

veni

le

mac

aque

s. O

ffspr

ing

disp

laye

d al

tere

d m

etab

olic

pat

hway

s on

the

basi

s of

m

ater

nal d

iet.

Addi

tiona

lly, t

hese

func

tiona

l pa

thw

ays

(am

ino

acid

, car

bohy

drat

e,

and

lipid

met

abol

ism

) co

rrel

ated

with

ab

unda

nces

of s

peci

fic g

ut b

acte

ria

44

TabL

e 1

Cont

inue

d

by guest on August 10, 2020www.aappublications.org/newsDownloaded from

Page 11: The Role of the Microbiome in the Developmental …...the developmental origins field (Fig 1). earLy-LiFe DeveLOPMeNT OF The MicrObiOMe The human microbiota is a composite organism,

PEDIATRICS Volume 141, number 4, April 2018 11

Rese

arch

Obj

ectiv

eSt

udy

Popu

latio

n or

An

imal

Mod

elEa

rly-

Life

Fac

tors

and

Ti

min

g of

Exp

osur

eTi

min

g of

Mic

robi

ome

Anal

ysis

or

Inte

rven

tion

Heal

th O

utco

me

and

Tim

ing

of A

sses

smen

tSu

mm

ary

of F

indi

ngs

Ref.

No.

To

det

erm

ine

if th

e pl

acen

tal

mic

robi

ome

vari

es in

as

soci

atio

n w

ith b

irth

wt

Pros

pect

ive

hum

an c

ohor

t an

alys

is

(n =

24)

None

Vari

atio

ns in

the

plac

enta

l m

icro

biom

e co

mpo

sitio

nBi

rth

wt

Low

bir

th w

t inf

ants

dis

play

ed lo

wer

gut

m

icro

biom

e ri

chne

ss a

nd v

aria

tions

in

the

abun

danc

es o

f spe

cific

bac

teri

al ta

xa

com

pare

d w

ith n

orm

al b

irth

wt i

nfan

ts.

Lact

obac

illus

per

cent

age

was

pos

itive

ly

corr

elat

ed w

ith b

irth

wt

24

To

ana

lyze

the

effe

ct o

f ea

rly-

life

mic

robi

al

pert

urba

tion

with

an

tibio

tic tr

eatm

ent o

n ho

st m

etab

olis

m a

nd

adip

osity

Mou

se m

odel

(hi

gh-fa

t ve

rsus

sta

ndar

d di

et)

Antib

iotic

-indu

ced

LDP

expo

sure

from

bir

th

thro

ugh

wea

ning

Body

com

posi

tion

in

adul

thoo

dCo

mpa

red

with

con

trol

s an

d m

ice

expo

sed

to

long

-term

LDP

, mic

e ex

pose

d to

LDP

for

4 or

8

wk

afte

r bi

rth

disp

laye

d el

evat

ed c

alor

ic

inta

ke a

nd fa

ster

tota

l mas

s an

d fa

t mas

s ac

cum

ulat

ion.

The

aut

hors

als

o re

port

th

at th

e pe

nici

llin-

sele

cted

mic

robi

ota

can

indu

ce m

etab

olic

cha

nges

whe

n tr

ansf

erre

d to

GF

mic

e

6

To

bet

ter

unde

rsta

nd h

ow

earl

y-lif

e an

tibio

tic u

se

alte

rs th

e gu

t mic

robi

ome

com

posi

tion

and

met

abol

ic d

evel

opm

ent

Mou

se m

odel

(hi

gh fa

t ve

rsus

sta

ndar

d di

et)

Antib

iotic

-indu

ced

Puls

ed a

ntib

iotic

trea

tmen

t co

mpl

eted

sho

rtly

aft

er

wea

ning

Body

com

posi

tion

from

3–

6 w

kEa

rly-

life

puls

ed a

ntib

iotic

trea

tmen

t ac

cele

rate

s to

tal m

ass

and

bone

gro

wth

. Th

e au

thor

s al

so r

epor

t tha

t res

pons

e to

hig

h-fa

t die

t is

alte

red

depe

ndin

g on

th

e pa

rtic

ular

ant

ibio

tic a

nd n

umbe

r of

co

urse

s us

ed to

per

turb

the

mic

robi

ota

85

To

ana

lyze

the

impa

ct o

f m

ater

nal p

repr

egna

ncy

BMI o

n th

e in

fant

gut

m

icro

biom

e co

mpo

sitio

n an

d fu

nctio

nal p

oten

tial

Nest

ed c

ase-

cont

rol

desi

gn (

n =

39)

Mat

erna

l pre

preg

nanc

y ob

esity

Vari

atio

ns in

infa

nt

mic

robi

ome

com

posi

tion

and

func

tion

Infa

nt m

etab

olis

m a

t 18

mo

of a

geFi

rmic

utes

wer

e re

port

ed e

nric

hed

in c

hild

ren

born

to m

othe

rs a

t a n

orm

al w

t, w

here

as

bact

eroi

dete

s w

ere

enri

ched

in in

fant

s bo

rn to

wom

en w

ho w

ere

obes

e. In

this

st

udy,

a li

nk to

an

infa

nt h

ealth

out

com

e w

as n

ot e

stab

lishe

d, b

ut d

iffer

entia

l m

icro

biom

e m

etab

olic

func

tions

that

wer

e ba

sed

on w

heth

er in

fant

s w

ere

born

to

mot

hers

at a

nor

mal

wt o

r to

wom

en w

ho

wer

e ob

ese

was

iden

tified

86

To

inve

stig

ate

the

effe

ct o

f ca

dmiu

m e

xpos

ure

on th

e ea

rly-

life

gut m

icro

biot

a an

d m

etab

olis

m in

ad

ulth

ood

SPF

mou

se m

odel

Cadm

ium

exp

osur

e 1

wk

befo

re p

aren

tal m

atin

gVa

riat

ions

in th

e m

icro

biom

e co

mpo

sitio

n ob

serv

ed a

t 8 w

k an

d at

ad

ulth

ood

(20

wk)

Body

com

posi

tion

mea

sure

d in

adu

lthoo

dCa

dmiu

m e

xpos

ure

in p

aren

tal m

ice

resu

lted

in in

crea

sed

fat a

ccum

ulat

ion

in m

ale

offs

prin

g. A

ltera

tions

in

mic

robi

ome

com

posi

tion

occu

rred

bef

ore

mea

sure

men

ts o

f bod

y co

mpo

sitio

n. In

ad

ditio

n, th

roug

h m

icro

biot

a tr

ansf

er

expe

rim

ents

, the

gro

up r

epor

ted

that

fat

accu

mul

atio

n w

as d

rive

n by

the

cadm

ium

-ex

pose

d m

icro

biom

e

87

To

det

erm

ine

if th

e ea

rly-

life

gut m

icro

biot

a co

mpo

sitio

n is

ass

ocia

ted

with

wt d

evel

opm

ent i

n ea

rly

child

hood

Nest

ed c

ase-

cont

rol

desi

gn (

n =

49)

None

Vari

atio

ns in

the

mic

robi

ome

com

posi

tion

betw

een

6 an

d 12

mo

of a

ge

BMI m

easu

red

at 7

y o

f age

Grea

ter

abun

danc

e of

Sta

phyl

ococ

cus

aure

us

in c

hild

ren

who

wer

e ob

ese

in in

fanc

y.

Grea

ter

abun

danc

es o

f bifi

doba

cter

ia in

ch

ildre

n at

a n

orm

al w

t in

infa

ncy

88

TabL

e 1

Cont

inue

d

by guest on August 10, 2020www.aappublications.org/newsDownloaded from

Page 12: The Role of the Microbiome in the Developmental …...the developmental origins field (Fig 1). earLy-LiFe DeveLOPMeNT OF The MicrObiOMe The human microbiota is a composite organism,

STIEMSMA and MICHELS12

Rese

arch

Obj

ectiv

eSt

udy

Popu

latio

n or

An

imal

Mod

elEa

rly-

Life

Fac

tors

and

Ti

min

g of

Exp

osur

eTi

min

g of

Mic

robi

ome

Anal

ysis

or

Inte

rven

tion

Heal

th O

utco

me

and

Tim

ing

of A

sses

smen

tSu

mm

ary

of F

indi

ngs

Ref.

No.

To

inve

stig

ate

the

effe

cts

of

earl

y-lif

e fa

ctor

s on

the

traj

ecto

ry o

f gut

mic

robi

al

deve

lopm

ent a

nd

child

hood

adi

posi

ty

Pros

pect

ive

hum

an c

ohor

t an

alys

is (

n =

75)

Gest

atio

nal a

ge a

nd

deliv

ery

mod

eVa

riat

ions

in th

e m

icro

biom

e be

fore

6 m

o of

age

Adip

osity

at 1

8 m

o of

age

Infa

nts

with

hig

h Bi

fidob

acte

rium

and

Co

llins

ella

at a

late

r ag

e di

spla

yed

low

er

adip

osity

at 1

8 m

o of

age

. Inf

ants

who

ac

quir

ed th

ese

taxa

at 6

mo

show

ed th

e lo

wes

t adi

posi

ty a

t 18

mo.

In a

dditi

on,

acqu

isiti

on o

f the

se b

acte

rial

taxa

was

in

fluen

ced

by le

ngth

of g

esta

tion

and

deliv

ery

mod

e

89

To

ana

lyze

the

effe

ct o

f su

bthe

rape

utic

ant

ibio

tic

adm

inis

trat

ion

on th

e gu

t mic

robi

ome

and

host

m

etab

olis

m

Mou

se m

odel

of a

ntib

iotic

-in

duce

d ad

ipos

ityAn

tibio

tic-in

duce

dVa

riat

ions

in th

e m

icro

biom

e m

easu

red

befo

re s

acri

fice

Body

com

posi

tion

mea

sure

d in

adu

lthoo

d (1

6–20

wk)

The

auth

ors

gene

rate

d a

mou

se m

odel

of

adip

osity

by

expo

sing

mic

e in

ear

ly li

fe

to a

ntib

iotic

s. V

aria

tions

in m

icro

bial

co

mpo

sitio

n be

fore

adi

posi

ty m

easu

rem

ent

was

not

ass

esse

d. H

owev

er, t

he a

ntib

iotic

s di

d al

ter

the

mic

robi

ome

com

posi

tion,

SCF

A m

etab

olis

m, a

nd h

epat

ic m

etab

olis

m o

f fa

tty

acid

s an

d lip

ids

90

To

ana

lyze

the

asso

ciat

ion

betw

een

the

earl

y-lif

e gu

t m

icro

biom

e co

mpo

sitio

n an

d BM

I in

child

hood

Pros

pect

ive

hum

an c

ohor

t an

alys

is (

n =

138)

None

Vari

atio

ns in

the

mic

robi

ome

com

posi

tion

with

in th

e fir

st y

ear

of

age

BMI S

D sc

ore

betw

een

1 an

d 3

y of

age

Abun

danc

e of

Bac

tero

ides

frag

ilis

at 3

and

26

wk

of a

ge is

ass

ocia

ted

with

BM

I SD

scor

e be

twee

n 1

and

3 y

of a

ge. A

bund

ance

of

Stap

hylo

cocc

us a

t 3 a

nd 5

2 w

k is

inve

rsel

y as

soci

ated

with

BM

I SD

scor

e be

twee

n 1

and

3 y

91

Neur

odev

elop

men

t

To d

eter

min

e if

limite

d ne

stin

g st

ress

alte

rs

offs

prin

g m

icro

biot

a,

cort

icos

tero

ne le

vels

, and

in

test

inal

per

mea

bilit

y

Rat m

odel

of l

imite

d ne

stin

g st

ress

Lim

ited

nest

ing

stre

ssVa

riat

ions

in th

e gu

t m

icro

biom

e co

mpo

sitio

n 21

d p

ost b

irth

(at

w

eani

ng)

Lim

ited

nest

ing

stre

ss

from

pos

tnat

al d

ays

2–10

Lim

ited-

nest

ing

pups

had

hy

perc

ortic

oste

rone

mia

, enh

ance

d in

test

inal

per

mea

bilit

y, d

ecre

ased

m

icro

bial

div

ersi

ty, a

nd v

aria

tions

in

spec

ific

mic

robi

al ta

xa

92

To

det

erm

ine

if m

ater

nal

high

-fat-d

iet-i

nduc

ed

obes

ity is

ass

ocia

ted

with

so

cial

beh

avio

ral d

efici

ts

and

alte

red

mic

robi

ota

in

the

offs

prin

g

High

-fat d

iet m

ouse

mod

elM

ater

nal h

igh-

fat d

iet

Vari

atio

ns in

the

gut

mic

robi

ome

com

posi

tion

in o

ffspr

ing

Beha

vior

exa

ms

on

7–12

-wk-

old

offs

prin

g m

ice

A m

ater

nal h

igh-

fat d

iet i

nduc

es

com

posi

tiona

l var

iatio

ns in

the

gut

mic

robi

ome

of o

ffspr

ing

mic

e. O

ffspr

ing

mic

e of

mot

hers

on

a hi

gh-fa

t die

t co

hous

ed w

ith m

ice

born

of m

othe

rs r

aise

d on

a r

egul

ar d

iet d

ispl

ayed

nor

mal

soc

ial

beha

vior

. In

addi

tion,

rei

ntro

duct

ion

of

L re

uter

i (la

ckin

g in

offs

prin

g m

ice

of a

m

othe

r on

a h

igh-

fat d

iet)

res

tore

d no

rmal

so

cial

beh

avio

r in

thes

e m

ice

93

TabL

e 1

Cont

inue

d

by guest on August 10, 2020www.aappublications.org/newsDownloaded from

Page 13: The Role of the Microbiome in the Developmental …...the developmental origins field (Fig 1). earLy-LiFe DeveLOPMeNT OF The MicrObiOMe The human microbiota is a composite organism,

PEDIATRICS Volume 141, number 4, April 2018 13

Rese

arch

Obj

ectiv

eSt

udy

Popu

latio

n or

An

imal

Mod

elEa

rly-

Life

Fac

tors

and

Ti

min

g of

Exp

osur

eTi

min

g of

Mic

robi

ome

Anal

ysis

or

Inte

rven

tion

Heal

th O

utco

me

and

Tim

ing

of A

sses

smen

tSu

mm

ary

of F

indi

ngs

Ref.

No.

To

det

erm

ine

if co

gniti

ve

abili

ty is

ass

ocia

ted

with

pa

rtic

ular

infa

nt g

ut

mic

robi

ota

profi

les

Pros

pect

ive

hum

an c

ohor

t an

alys

is (

n =

89)

Grou

p 2

was

mor

e lik

ely

to h

ave

been

bre

astf

ed,

less

like

ly to

hav

e be

en b

orn

by c

esar

ean

deliv

ery,

and

ass

ocia

ted

with

whi

te e

thni

city

. Ha

ving

old

er s

iblin

gs

was

ass

ocia

ted

with

in

crea

sed

α di

vers

ity

3 gr

oups

of s

ubje

cts

wer

e id

entifi

ed o

n th

e ba

sis

of th

eir

1-y

mic

robi

ome

anal

ysis

Cogn

itive

out

com

es a

t 1

and

2 y

of a

geTh

ree

grou

ps o

f sub

ject

s w

ere

iden

tified

on

the

basi

s of

thei

r m

icro

biom

e.

Grou

p 1

disp

laye

d hi

gh a

bund

ance

of

Faec

alib

acte

rium

, gro

up 2

dis

play

ed

high

abu

ndan

ce o

f Bac

tero

ides

, and

gr

oup

3 di

spla

yed

high

abu

ndan

ce o

f ru

min

ococ

cace

ae. I

ndiv

idua

l Mul

len

scal

es

diffe

red

betw

een

grou

ps. α

div

ersi

ty w

as

nega

tivel

y as

soci

ated

with

indi

vidu

al M

ulle

n sc

ales

at 2

y o

f age

(ex

pres

sive

lang

uage

an

d vi

sual

rec

eptio

n)

94

To

det

erm

ine

if m

ater

nal

pren

atal

str

ess

alte

rs th

e m

icro

bial

intr

aute

rine

en

viro

nmen

t and

beh

avio

r in

offs

prin

g

Mou

se m

odel

of p

rena

tal

stre

ssPr

enat

al s

tres

sVa

riat

ions

in p

lace

ntal

m

icro

biom

e an

d fe

cal

mic

robi

ome

of o

ffspr

ing

Anxi

ety-

like

beha

vior

in

offs

prin

gPr

enat

al m

ater

nal s

tres

s w

as a

ssoc

iate

d w

ith v

aria

tions

in th

e m

icro

biom

e of

da

ms,

offs

prin

g, a

nd in

the

plac

enta

. Ad

ditio

nally

, pre

nata

l str

ess

is a

ssoc

iate

d w

ith in

crea

sed

IL-1

β in

the

plac

enta

and

re

duce

d br

ain-

deri

ved

neur

otro

phic

fact

or

in p

lace

nta

and

adul

t offs

prin

g am

ygda

la

7

To

det

erm

ine

if pr

obio

tic

adm

inis

trat

ion

in e

arly

lif

e m

odifi

es m

ater

nal

sepa

ratio

n-in

duce

d gu

t dy

sfun

ctio

n

Rat m

odel

(st

ress

in

duce

d by

mat

erna

l se

para

tion)

Mat

erna

l sep

arat

ion

in

earl

y lif

e (d

ay 4

to d

ay

19)

Vari

atio

ns in

mic

robi

ome

and

gut f

unct

ion

in

offs

prin

g

Hypo

thal

amus

-pitu

itary

-ad

rena

l axi

s ac

tivity

Incr

ease

d co

rtic

oste

rone

leve

ls a

nd a

ltere

d co

loni

c m

ucos

al b

arri

er fu

nctio

n in

m

ater

nally

sep

arat

ed r

at p

ups.

Ear

ly-li

fe

adm

inis

trat

ion

of p

robi

otic

s (c

ompo

sed

of

Lact

obac

illus

rha

mno

sus

and

Lact

obac

illus

he

lvet

icus

str

ains

) to

rat

pup

s am

elio

rate

s th

ese

findi

ngs

and

pers

ists

to a

dulth

ood

95

To

det

erm

ine

if ea

rly-

life

stre

ss a

lters

the

gut-b

rain

ax

is

Rat m

odel

(st

ress

in

duce

d by

mat

erna

l se

para

tion)

Mat

erna

l sep

arat

ion

in

earl

y lif

eVa

riat

ions

in m

icro

biom

e co

mpo

sitio

n in

rat

pup

sSy

mpt

oms

of p

sych

iatr

ic

diso

rder

s an

d ir

rita

ble

bow

el s

yndr

ome

Incr

ease

d pl

asm

a co

rtic

oste

rone

and

in

crea

sed

tum

or n

ecro

sis

fact

or-α

an

d in

terf

eron

-γ. A

lso,

the

mic

robi

ome

com

posi

tion

vari

ed in

the

mat

erna

lly

sepa

rate

d gr

oup

com

pare

d w

ith th

e ra

ts

that

wer

e no

t mat

erna

lly s

epar

ated

96

To

exa

min

e th

e ef

fect

s of

pr

enat

al a

nd e

arly

-life

ex

posu

re to

pro

pion

ic

acid

and

LPS

on

offs

prin

g gu

t mic

robi

al m

etab

olis

m,

loco

mot

or a

ctiv

ity, a

nd

anxi

ety-

like

beha

vior

Rat m

odel

None

Pre-

and

pos

tnat

al L

PS o

r pr

opio

nic

acid

exp

osur

eBe

havi

or tr

aits

in o

ffspr

ing

Pren

atal

pro

pion

ic a

cid

incr

ease

d an

xiet

y-lik

e be

havi

or in

mal

e an

d fe

mal

e ad

oles

cent

of

fspr

ing.

Pos

tnat

al p

ropi

onic

aci

d in

crea

sed

anxi

ety-

like

beha

vior

in fe

mal

e of

fspr

ing

only

. Pre

nata

l pro

pion

ic a

cid

and

LPS

indu

ced

deve

lopm

enta

l del

ays

(inc

ludi

ng d

elay

s in

eye

ope

ning

)

97

TabL

e 1

Cont

inue

d

by guest on August 10, 2020www.aappublications.org/newsDownloaded from

Page 14: The Role of the Microbiome in the Developmental …...the developmental origins field (Fig 1). earLy-LiFe DeveLOPMeNT OF The MicrObiOMe The human microbiota is a composite organism,

STIEMSMA and MICHELS14

Rese

arch

Obj

ectiv

eSt

udy

Popu

latio

n or

An

imal

Mod

elEa

rly-

Life

Fac

tors

and

Ti

min

g of

Exp

osur

eTi

min

g of

Mic

robi

ome

Anal

ysis

or

Inte

rven

tion

Heal

th O

utco

me

and

Tim

ing

of A

sses

smen

tSu

mm

ary

of F

indi

ngs

Ref.

No.

To

det

erm

ine

if co

loni

zatio

n by

gut

mic

robi

ota

in

earl

y lif

e im

pact

s br

ain

deve

lopm

ent a

nd a

dult

beha

vior

GF a

nd S

PF m

ouse

mod

els

None

Offs

prin

g of

pre

viou

sly

GF

mic

e co

loni

zed

with

SPF

m

icro

biot

a

Beha

vior

in a

dulth

ood

Adul

t col

oniz

ed o

ffspr

ing

disp

laye

d si

mila

r be

havi

ors

com

pare

d w

ith S

PF m

ice.

The

se

mic

e sp

ent l

ess

time

expl

orin

g th

e op

en

arm

s in

the

maz

e (l

ess

loco

mot

or a

ctiv

ity).

Addi

tiona

lly, c

olon

ized

mic

e ex

pres

sed

less

sy

napt

ophy

sin

and

PSD-

95 in

the

stri

atum

co

mpa

red

with

GF

mic

e, s

ugge

stin

g th

e m

icro

biom

e is

invo

lved

in p

rogr

amm

ing

brai

n de

velo

pmen

t

98

To

det

erm

ine

if a

mat

erna

l hi

gh-fa

t-die

t-alte

red

mic

robi

ome

can

mod

ify

offs

prin

g be

havi

or

Mou

se m

odel

col

oniz

ed

with

mat

erna

l hig

h-fa

t-di

et-s

hape

d m

icro

biot

a

Mat

erna

l hig

h-fa

t die

tFe

mal

e m

ice

tran

spla

nted

w

ith h

igh-

fat d

iet

mic

robi

ome

Beha

vior

trai

ts in

offs

prin

gFe

mal

e m

ice

wer

e tr

ansp

lant

ed w

ith a

hi

gh-fa

t-die

t- or

con

trol

low

-fat-d

iet-

asso

ciat

ed g

ut m

icro

biom

e. O

ffspr

ing

of

thes

e m

ice

disp

laye

d al

tere

d be

havi

or

in a

sex

-dep

ende

nt m

anne

r. Of

fspr

ing

mic

e di

spla

yed

less

str

ess

afte

r m

ater

nal

sepa

ratio

n. M

ale

offs

prin

g di

spla

yed

decr

ease

d ex

plor

ator

y an

d co

gniti

ve

beha

vior

s, w

hich

is in

dica

tive

of in

crea

sed

anxi

ety.

Fem

ale

mic

e di

spla

yed

incr

ease

s in

ad

ipos

ity a

nd b

ody

wt

99

To

ana

lyze

the

mic

robi

al a

nd

mol

ecul

ar m

echa

nism

s th

at u

nder

lie th

e gu

t-bra

in

axis

GF a

nd c

onve

ntio

nally

ra

ised

mou

se m

odel

sNo

neGF

mic

e co

loni

zed

afte

r w

eani

ngNe

uron

al a

ctiv

ity in

the

amyg

dala

Abse

nce

of a

mic

robi

ota

in e

arly

life

res

ults

in

diff

eren

tial g

ene

expr

essi

on, e

xon

usag

e,

RNA

editi

ng, a

nd u

pstr

eam

gen

e re

gula

tion

in th

e am

ygda

la. T

his

was

sim

ilar

to

mic

e w

ho w

ere

rais

ed G

F fo

r th

eir

entir

e liv

es b

ut v

arie

d w

hen

com

pare

d w

ith

conv

entio

naliz

ed m

ice

100

To

exa

min

e w

heth

er

vari

atio

ns in

the

vagi

nal m

icro

biom

e ar

e as

soci

ated

with

var

ied

offs

prin

g pr

ogra

mm

ing

Mou

se m

odel

of p

rena

tal

stre

ssPr

enat

al s

tres

sVa

riat

ions

in th

e m

ater

nal

vagi

nal a

nd n

eona

tal g

ut

mic

robi

ome

Met

abol

ic a

nd n

euro

logi

c pr

ogra

mm

ing

and

in

offs

prin

g

Lact

obac

illus

abu

ndan

ce is

dec

reas

ed in

the

vagi

nal m

icro

biom

e an

d in

neo

nate

s bo

rn

to d

ams

expo

sed

to e

arly

pre

nata

l str

ess.

Ot

her

bact

eria

l pop

ulat

ion

abun

danc

es

also

var

ied

in o

ffspr

ing

expo

sed

to e

arly

pr

enat

al s

tres

s. E

arly

pre

nata

l str

ess

alte

red

met

abol

ic p

rofil

es a

nd a

min

o ac

id

avai

labi

lity

in th

e br

ain

101

Imm

une-

med

iate

d di

seas

es (

IBD,

T1D

, etc

)

To in

vest

igat

e ag

e-de

pend

ent

mic

robi

al m

odul

atio

n of

iN

KTs

in m

ouse

mod

els

of

IBD

and

asth

ma

Mou

se m

odel

of

oxaz

olon

e-in

duce

d co

litis

None

SPF-

colo

niza

tion

duri

ng

preg

nanc

y le

ad to

SPF

-co

loni

zed

offs

prin

g

Colit

is in

duce

d la

ter

in li

feNe

onat

al e

xpos

ure

to a

con

vent

iona

l m

icro

biot

a co

mpa

red

with

GF

cond

ition

s pr

otec

ted

mic

e fr

om o

xazo

lone

-indu

ced

colit

is

76

TabL

e 1

Cont

inue

d

by guest on August 10, 2020www.aappublications.org/newsDownloaded from

Page 15: The Role of the Microbiome in the Developmental …...the developmental origins field (Fig 1). earLy-LiFe DeveLOPMeNT OF The MicrObiOMe The human microbiota is a composite organism,

PEDIATRICS Volume 141, number 4, April 2018 15

Rese

arch

Obj

ectiv

eSt

udy

Popu

latio

n or

An

imal

Mod

elEa

rly-

Life

Fac

tors

and

Ti

min

g of

Exp

osur

eTi

min

g of

Mic

robi

ome

Anal

ysis

or

Inte

rven

tion

Heal

th O

utco

me

and

Tim

ing

of A

sses

smen

tSu

mm

ary

of F

indi

ngs

Ref.

No.

To

det

erm

ine

if an

d ho

w

earl

y-lif

e ex

posu

re to

an

tibio

tics

chan

ges

susc

eptib

ility

to IB

D

Mou

se m

odel

DSS

-indu

ced

colit

isAn

tibio

tic-in

duce

dLD

P af

ter

wea

ning

Colit

is in

duce

d la

ter

in li

feLD

P-tr

eate

d m

ice

disp

laye

d tr

ansi

ent g

ut

mic

robi

al c

ompo

sitio

nal a

ltera

tions

, in

clud

ing

erad

icat

ion

of s

egm

ente

d fil

amen

tous

bac

teri

a. In

add

ition

, aft

er D

SS-

indu

ced

colit

is, L

DP m

ice

disp

laye

d re

duce

d co

litis

sym

ptom

s, Il

-17

expr

essi

on, a

nd il

eal

Th17

diff

eren

tiatio

n co

mpa

red

with

mic

e ex

pose

d to

met

roni

dazo

le, e

nrofl

oxac

in,

and

cont

rols

. Fin

ally

, the

aut

hors

rep

ort

peni

cilli

n’s

effe

cts

are

depe

nden

t on

erad

iatio

n of

seg

men

ted

filam

ento

us

bact

eria

, im

plic

atin

g th

e m

icro

biom

e as

the

med

iato

r be

twee

n th

is e

arly

life

exp

osur

e an

d co

litis

dev

elop

men

t

102

To

exp

lore

the

influ

ence

of

gut

dys

bios

is in

the

prog

ress

ion

of T

1D

NOD

mou

se m

odel

Antib

iotic

-indu

ced

Antib

iotic

trea

tmen

t in

earl

y lif

e (c

once

ptio

n un

til 4

0 w

k po

stna

tal)

Spon

tane

ous

diab

etes

la

ter

in li

feAn

tibio

tics

wer

e ad

min

iste

red

to N

OD m

ice

from

con

cept

ion

until

40

wk

post

nata

l de

velo

pmen

t. Tr

eatm

ent w

ith a

ntib

iotic

s in

crea

sed

inci

denc

e of

T1D

in m

ale

mic

e.

Antib

iotic

trea

tmen

t als

o re

sulte

d in

nea

r ab

latio

n of

the

gut m

icro

biom

e at

8 w

k of

age

, whi

ch m

ay p

artia

lly e

xpla

in th

e in

crea

sed

T1D

inci

denc

e in

mal

e m

ice

103

To

det

erm

ine

if ex

posu

re to

pr

enat

al a

ntib

iotic

s ca

n pr

otec

t offs

prin

g fr

om

T1D

NOD

mou

se m

odel

Antib

iotic

-indu

ced

Pren

atal

ant

ibio

tic

trea

tmen

t ind

uced

va

riat

ions

in o

ffspr

ing

and

mat

erna

l m

icro

biom

es

Spon

tane

ous

diab

etes

la

ter

in li

fePr

enat

al n

eom

ycin

and

van

com

ycin

trea

tmen

t re

sulte

d in

diff

eren

tial s

hift

s in

the

offs

prin

g an

d m

ater

nal m

icro

biom

es.

Offs

prin

g tr

eate

d pr

enat

ally

with

neo

myc

in

wer

e pr

otec

ted

from

T1D

dev

elop

men

t, w

here

as o

ffspr

ing

trea

ted

pren

atal

ly w

ith

vanc

omyc

in d

ispl

ayed

acc

eler

ated

T1D

de

velo

pmen

t. Th

e an

tibio

tic tr

eatm

ent a

lso

resu

lted

in a

ltere

d im

mun

e pr

ofile

s, s

uch

as in

crea

sed

T-cel

l–m

edia

ted

infla

mm

atio

n in

mic

e tr

eate

d w

ith v

anco

myc

in a

nd

alte

red

antig

en-p

rese

ntin

g ce

ll ph

enot

ypes

in

mic

e tr

eate

d w

ith n

eom

ycin

104

To

det

erm

ine

the

impa

ct o

f ta

rget

ing

Gram

-neg

ativ

e gu

t bac

teri

a at

var

ious

tim

e po

ints

in e

arly

life

on

T1D

deve

lopm

ent

NOD

mou

se m

odel

Antib

iotic

-indu

ced

Pren

atal

ant

ibio

tic

trea

tmen

t ind

uced

va

riat

ions

in o

ffspr

ing

mic

robi

ome

Spon

tane

ous

diab

etes

la

ter

in li

fePr

egna

nt, N

OD m

ice

trea

ted

with

an

antib

iotic

m

ixtu

re (

neom

ycin

, pol

ymyx

in B

, and

st

rept

omyc

in)

wer

e pr

otec

ted

from

T1D

co

mpa

red

with

mic

e tr

eate

d po

stna

tally

. M

icro

biot

a tr

ansf

er fr

om th

ese

mic

e to

un

trea

ted

mic

e re

sulte

d in

pro

tect

ion

from

T1

D

105

TabL

e 1

Cont

inue

d

by guest on August 10, 2020www.aappublications.org/newsDownloaded from

Page 16: The Role of the Microbiome in the Developmental …...the developmental origins field (Fig 1). earLy-LiFe DeveLOPMeNT OF The MicrObiOMe The human microbiota is a composite organism,

STIEMSMA and MICHELS16

Rese

arch

Obj

ectiv

eSt

udy

Popu

latio

n or

An

imal

Mod

elEa

rly-

Life

Fac

tors

and

Ti

min

g of

Exp

osur

eTi

min

g of

Mic

robi

ome

Anal

ysis

or

Inte

rven

tion

Heal

th O

utco

me

and

Tim

ing

of A

sses

smen

tSu

mm

ary

of F

indi

ngs

Ref.

No.

To

com

pare

the

effe

cts

of p

ulse

d th

erap

eutic

an

tibio

tics

or c

ontin

uous

lo

w-d

ose

antib

iotic

s in

ear

ly li

fe o

n T1

D de

velo

pmen

t

NOD

mou

se m

odel

Antib

iotic

-indu

ced

Puls

ed a

ntib

iotic

trea

tmen

t in

duce

d va

riat

ions

in

6-w

k-ol

d m

icro

biom

e

Spon

tane

ous

diab

etes

la

ter

in li

fePu

lsed

pos

tnat

al tr

eatm

ent w

ith ty

losi

n al

tere

d th

e m

ouse

mic

robi

ome

and

acce

lera

ted

T1D

deve

lopm

ent c

ompa

red

with

mic

e tr

eate

d w

ith s

ubth

erap

eutic

pe

nici

llin

from

pre

gnan

cy to

wee

k 12

106

To

ana

lyze

the

asso

ciat

ion

betw

een

the

infa

nt g

ut

mic

robi

ome

and

T1D

deve

lopm

ent

Pros

pect

ive

hum

an c

ohor

t an

alys

is (

n =

33)

None

Vari

atio

ns in

the

mic

robi

ome

befo

re

diag

nosi

s w

ith T

1D

T1D

diag

nosi

s at

∼3

y of

age

T1D

dise

ase

stat

e w

as d

istin

guis

habl

e by

the

gut m

icro

biom

e co

mpo

sitio

n.

Sero

conv

erte

d su

bjec

ts d

iagn

osed

with

T1D

di

spla

yed

a m

arke

d de

crea

se in

α d

iver

sity

be

fore

dia

gnos

is w

hen

com

pare

d w

ith

sero

conv

erte

d su

bjec

ts n

ot d

iagn

osed

with

T1

D an

d no

nser

ocon

vert

ed s

ubje

cts

107

To

ana

lyze

the

effe

ct o

f pe

ripa

rtum

cef

oper

azon

e ad

min

istr

atio

n on

the

mat

erna

l and

offs

prin

g m

icro

biot

a an

d IB

D in

the

offs

prin

g

SPF

IL-1

0 kn

ock-

out m

ouse

m

odel

com

bine

d w

ith

DSS-

indu

ced

colit

is

Antib

iotic

-indu

ced

Peri

part

um a

ntib

iotic

tr

eatm

ent i

nduc

ed g

ut

dysb

iosi

s in

offs

prin

g th

at p

ersi

sts

to a

dulth

ood

Spon

tane

ous

colit

is la

ter

in li

fePe

ripa

rtum

exp

osur

e to

cef

oper

azon

e in

crea

ses

risk

of s

pont

aneo

us c

oliti

s in

offs

prin

g. A

ntib

iotic

s al

so c

ontr

ibut

e to

imm

une

skew

ing

and

prom

ote

gut

dysb

iosi

s th

at p

ersi

sts

to a

dulth

ood.

Ad

ditio

nally

, as

dem

onst

rate

d by

feca

l tr

ansp

lant

to G

F IL

-10

knoc

k-ou

t dam

s,

imm

une

skew

ing

is m

edia

ted

by th

e an

tibio

tic-in

duce

d dy

sbio

sis

108

To

ana

lyze

the

impa

ct o

f a

mat

erna

l hig

h-fib

er

diet

on

T -reg

ulat

ory

cell

diffe

rent

iatio

n in

the

offs

prin

g

SPF

GPR4

1−/−

mou

se

mod

elM

ater

nal h

igh-

fiber

die

t du

ring

pre

gnan

cy a

nd

brea

stfe

edin

g

Incr

ease

d pl

asm

a SC

FAs

Incr

ease

d th

ymic

and

pe

riph

eral

T-re

gula

tory

ce

lls

Com

pare

d w

ith o

ffspr

ing

from

mat

erna

l mic

e fe

d a

norm

al d

iet,

high

-fibe

r di

et d

urin

g pr

egna

ncy

and

brea

stfe

edin

g re

sulte

d in

in

crea

sed

plas

ma

SCFA

s in

the

offs

prin

g.

Thes

e of

fspr

ing

also

dis

play

ed h

ighe

r fr

eque

ncie

s of

thym

ic a

nd p

erip

hera

l T-r

egul

ator

y ce

lls, w

hich

may

be

prom

pted

by

incr

ease

d SC

FA le

vels

46

AAD,

alle

rgic

air

way

s di

seas

e; C

D4+,

clu

ster

of d

iffer

entia

tion

4; C

XCL1

6, c

hem

okin

e lig

and

16; D

SS, d

extr

an s

odiu

m s

ulfa

te; G

I, ga

stro

inte

stin

al; G

PR41

, G p

rote

in–c

oupl

ed r

ecep

tor

41; G

PR43

, G p

rote

in–c

oupl

ed r

ecep

tor

43; G

rx1,

Glu

tare

doxi

n-1;

HD

AC9,

his

tone

dea

cety

lase

9; I

gA, i

mm

unog

lobu

lin A

; IL-

1β, i

nter

leuk

in 1

bet

a; IL

-10,

inte

rleu

kin

10; I

L-17

, int

erle

ukin

17;

iNKT

, inv

aria

nt n

atur

al k

iller

T c

ell;

LPS,

lipo

poly

sacc

hari

de; N

OD, n

onob

ese

diab

etic

; PD-

L1, p

rogr

amm

ed d

eath

liga

nd-1

; PSD

-95,

po

stsy

napt

ic d

ensi

ty p

rote

in 9

5; R

ef.,

refe

renc

e; T

h1, T

-hel

per

1; T

h17,

T-he

lper

17;

TLR

, Tol

l-lik

e re

cept

or; T

1D, t

ype

1 di

abet

es.

TabL

e 1

Cont

inue

d

by guest on August 10, 2020www.aappublications.org/newsDownloaded from

Page 17: The Role of the Microbiome in the Developmental …...the developmental origins field (Fig 1). earLy-LiFe DeveLOPMeNT OF The MicrObiOMe The human microbiota is a composite organism,

3 months of age and an increased enterobacteriaceae/bacteroidaceae ratio at 3 months and 1 year of age, which was associated with increased food sensitization among 1-year-old children.‍ Variations in the gut microbiome at 3 and 6 months of age have also been associated with the resolution of a milk allergy by 8 years of age.‍75 Thus, authors of the current microbiome research for asthma and allergies suggest the time period between birth and 1 year of age as the developmental origins window for this disease.‍ These immune hypersensitivities persist into later childhood and adulthood despite the transient nature of microbial dysbiosis.‍

Obesity

Similar to asthma and atopic disease, prospective studies reveal early-life gut microbiome compositional variations, which precede the development of obesity.‍88, 91 A recent study reveals temporal and compositional microbiome associations with early childhood adiposity.‍89 Specifically, infants with high Streptococcus abundance at 6 months of age displayed increased adiposity at 18 months of age.‍89 This emphasizes the importance of both microbiome composition and timing of microbiome maturation in the development of childhood obesity.‍

Mouse model studies have been used to mechanistically support the potential metabolic effects of early-life microbial dysbiosis and emphasize the obesity-inducing abilities of antibiotics.‍ In a study by Cho et al, 90 antibiotics administered to mice in early life resulted in subsequent increases in adiposity and metabolic hormones.‍ Cox et al6 furthered this research by manipulating the early-life microbiota using low-dose penicillin (LDP).‍ Here, they report LDP exposure during early life increased the effect of a high-fat diet on the microbiota, which could

be transferred to GF mice to induce obesity.‍6 It will be important for future researchers to incorporate both prospective human studies and animal models to determine the microbiome-associated effects of early-life antibiotic exposure on human health.‍

Neurodevelopmental Disorders

A less intuitive role of the gut microbiome in human health and development is the impact of early-life microbial dysbiosis in neurodevelopment (ie, the gut-brain axis).‍ There is increasing evidence to support early-life microbial compositional variations in stress response and anxiety.‍ In animal models, it is suggested that neonatal stress after maternal separation results in long-term compositional changes to the intestinal microbiota, 95, 96 and treating rat pups with probiotics can counter the resulting elevated corticosterone levels.‍95 Prenatal exposure to the SCFA, propionic acid, was also reported to increase anxiety-like behavior in mice.‍97 In a recent study, Gur et al7 report the influence of prenatal maternal stress in mice and its ability to alter both the maternal and neonatal intestinal microbiomes.‍ Adult offspring exposed to prenatal maternal stress also displayed increased anxiety-like behavior.‍7 Additionally, prenatal stress also induced variations in the placental microbiome.‍7 This suggests that the intrauterine microbial environment is a potential mediator of maternal prenatal stress and resulting stress in the offspring.‍

Variations in the gut microbiome have also been associated with many facets of social behavior.‍ Adult offspring of conventionalized dams (previously GF but colonized with the microbiome of specific pathogen–free [SPF] mice) were reported to display decreased motor activity compared with offspring from GF dams.‍98 Buffington et al93

report impaired social behavior (similar to that observed in autism spectrum disorder) in the offspring of dams that were fed a high-fat diet, which is mediated by variations in the offspring microbiome.‍ In addition, using shotgun metagenomic sequencing, this group identified a significant reduction in Lactobacillus reuteri among the maternal high-fat diet–fed offspring.‍93 Supplementation of drinking water with L reuteri resulted in restoration of offspring social deficits.‍93 Notably, the majority of studies with researchers focusing on the early-life critical window in relation to neurodevelopment have been conducted in animal models.‍ However, there is recent evidence to support a role of the infant gut microbiome in cognitive development in humans.‍ Carlson et al94 reported variations in gut microbiome α diversity in 1-year-old children, which correlated with their cognitive abilities measured at age 2.‍ Additional studies in humans will substantiate this evidence.‍ However, it is interesting to note that researchers consistently identify the time period between pregnancy and the first year of life as the most influential in human development.‍

Collectively, this disease-specific research is used to support a role of the early-life microbiome in fetal and childhood development in a variety of contexts.‍ However, prediagnostic variations in the early-life microbiome composition have been associated with other chronic noncommunicable diseases in later childhood and adulthood (ie, IBD and type 1 diabetes, Table 1).‍1 In addition, Harris et al112 identified an association between adolescent diet and breast cancer diagnosed in adulthood, suggesting that the timing of susceptibility for the microbiome may also be disease specific.‍ The future of research surrounding the microbiome in DOHaD is thus ripe with opportunity.‍

PEDIATRICS Volume 141, number 4, April 2018 17 by guest on August 10, 2020www.aappublications.org/newsDownloaded from

Page 18: The Role of the Microbiome in the Developmental …...the developmental origins field (Fig 1). earLy-LiFe DeveLOPMeNT OF The MicrObiOMe The human microbiota is a composite organism,

cONcLUsiONs aND FUTUre DirecTiONs

In this review, we summarize evidence supporting the role of the microbiome as a fundamental part of human physiology and key to our understanding of DOHaD.‍ Pre- and postbirth exposures alter the microbiome composition and functional potential in the neonate.‍ In turn, this dysbiosis results in a number of health and disease outcomes later in life.‍ Collaborative translational efforts using both animal models and human samples will mechanistically define the extent to which the microbiome plays a causal role during this critical window of developmental plasticity.‍ Multiomics approaches, combining epigenetic, transcriptome, and microbiome analyses in large, prospective, human birth cohorts will enhance our current understanding of how the microbiome may interact with host components to drive aspects of infant development.‍ In addition, as it is becoming clearer that the microbiota (both maternal and placental) can influence fetal development, more research surrounding the vaginal and intrauterine microbiomes is needed to paint a fuller picture of the fetal and neonatal origins of disease.‍ We propose revisiting DOHaD and incorporating the microbiome into our current understanding of the many aspects of early human development associated with long-term health.‍

reFereNces

1. Stiemsma LT, Reynolds LA, Turvey SE, Finlay BB. The hygiene hypothesis: current perspectives and future therapies. Immunotargets Ther. 2015;4:143–157

2. Stiemsma LT, Turvey SE. Asthma and the microbiome: defining the critical window in early life. Allergy Asthma Clin Immunol. 2017;13:3

3. Tamburini S, Shen N, Wu HC, Clemente JC. The microbiome in early life: implications for health outcomes. Nat Med. 2016;22(7):713–722

4. Arrieta MC, Stiemsma LT, Dimitriu PA, et al; CHILD Study Investigators. Early infancy microbial and metabolic alterations affect risk of childhood asthma. Sci Transl Med. 2015;7(307):307ra152

5. Stiemsma LT, Arrieta MC, Dimitriu PA, et al; Canadian Healthy Infant Longitudinal Development (CHILD) Study Investigators. Shifts in Lachnospira and Clostridium sp. in the 3-month stool microbiome are associated with preschool age asthma. Clin Sci (Lond). 2016;130(23):2199–2207

6. Cox LM, Yamanishi S, Sohn J, et al. Altering the intestinal microbiota during a critical developmental window has lasting metabolic consequences. Cell. 2014;158(4):705–721

7. Gur TL, Shay L, Palkar AV, et al. Prenatal stress affects placental cytokines and neurotrophins, commensal microbes, and anxiety-like behavior in adult female offspring. Brain Behav Immun. 2017;64:50–58

8. Mueller NT, Bakacs E, Combellick J, Grigoryan Z, Dominguez-Bello MG. The infant microbiome development: mom matters. Trends Mol Med. 2015;21(2):109–117

9. Waterland RA, Michels KB. Epigenetic epidemiology of the developmental origins hypothesis. Annu Rev Nutr. 2007;27:363–388

10. Osmond C, Barker DJ, Winter PD, Fall CH, Simmonds SJ. Early growth and death from cardiovascular disease in women. BMJ. 1993;307(6918):1519–1524

11. Barker DJ, Osmond C. Infant mortality, childhood nutrition, and ischaemic heart disease in England and Wales. Lancet. 1986;1(8489):1077–1081

12. Hjalgrim LL, Westergaard T, Rostgaard K, et al. Birth weight as a risk factor for childhood leukemia: a meta-analysis of 18 epidemiologic studies. Am J Epidemiol. 2003;158(8):724–735

13. Trichopoulos D. Hypothesis: does breast cancer originate in utero? Lancet. 1990;335(8695):939–940

14. Strachan DP. Hay fever, hygiene, and household size. BMJ. 1989;299(6710):1259–1260

15. Shreiner A, Huffnagle GB, Noverr MC. The “Microflora Hypothesis” of allergic disease. Adv Exp Med Biol. 2008;635:113–134

16. Ursell LK, Metcalf JL, Parfrey LW, Knight R. Defining the human microbiome. Nutr Rev. 2012;70(suppl 1):S38–S44

17. Robinson CM, Pfeiffer JK. Viruses and the microbiota. Annu Rev Virol. 2014;1:55–69

18. Arrieta MC, Stiemsma LT, Amenyogbe N, Brown EM, Finlay B. The intestinal microbiome in early life: health and disease. Front Immunol. 2014;5:427

19. Steel JH, Malatos S, Kennea N, et al. Bacteria and inflammatory cells in fetal membranes do not always cause preterm labor. Pediatr Res. 2005;57(3):404–411

20. Satokari R, Grönroos T, Laitinen K, Salminen S, Isolauri E. Bifidobacterium and Lactobacillus DNA in the human placenta. Lett Appl Microbiol. 2009;48(1):8–12

21. Collado MC, Rautava S, Aakko J, Isolauri E, Salminen S. Human gut colonisation may be initiated in utero by distinct microbial communities in the placenta and amniotic fluid. Sci Rep. 2016;6:23129

22. Gomez de Agüero M, Ganal-Vonarburg SC, Fuhrer T, et al. The maternal microbiota drives early postnatal innate immune development. Science. 2016;351(6279):1296–1302

23. Bassols J, Serino M, Carreras-Badosa G, et al. Gestational diabetes is associated with changes in placental microbiota and microbiome. Pediatr Res. 2016;80(6):777–784

STIEMSMA and MICHELS18

abbreviaTiONs

DOHaD:  Developmental Origins of Health and Disease

GF:  germ freeIBD:  inflammatory bowel diseaseLDP:  low-dose penicillinNEC:  necrotizing enterocolitisSCFA:  short-chain fatty acidSPF:  specific pathogen free

by guest on August 10, 2020www.aappublications.org/newsDownloaded from

Page 19: The Role of the Microbiome in the Developmental …...the developmental origins field (Fig 1). earLy-LiFe DeveLOPMeNT OF The MicrObiOMe The human microbiota is a composite organism,

24. Zheng J, Xiao X, Zhang Q, Mao L, Yu M, Xu J. The placental microbiome varies in association with low birth weight in full-term neonates. Nutrients. 2015;7(8):6924–6937

25. Prince AL, Ma J, Kannan PS, et al. The placental membrane microbiome is altered among subjects with spontaneous preterm birth with and without chorioamnionitis. Am J Obstet Gynecol. 2016;214(5):627.e1–627.e16

26. Antony KM, Ma J, Mitchell KB, Racusin DA, Versalovic J, Aagaard K. The preterm placental microbiome varies in association with excess maternal gestational weight gain. Am J Obstet Gynecol. 2015;212(5):653.e1–e16

27. Perez-Muñoz ME, Arrieta MC, Ramer-Tait AE, Walter J. A critical assessment of the “sterile womb” and “in utero colonization” hypotheses: implications for research on the pioneer infant microbiome. Microbiome. 2017;5(1):48

28. Lauder AP, Roche AM, Sherrill-Mix S, et al. Comparison of placenta samples with contamination controls does not provide evidence for a distinct placenta microbiota. Microbiome. 2016;4(1):29

29. Boro P, Kumaresan A, Singh AK, et al. Expression of short chain fatty acid receptors and pro-inflammatory cytokines in utero-placental tissues is altered in cows developing retention of fetal membranes. Placenta. 2014;35(7):455–460

30. Dominguez-Bello MG, Costello EK, Contreras M, et al. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc Natl Acad Sci USA. 2010;107(26):11971–11975

31. Chu DM, Ma J, Prince AL, Antony KM, Seferovic MD, Aagaard KM. Maturation of the infant microbiome community structure and function across multiple body sites and in relation to mode of delivery. Nat Med. 2017;23(3):314–326

32. Bokulich NA, Chung J, Battaglia T, et al. Antibiotics, birth mode, and diet shape microbiome maturation during early life. Sci Transl Med. 2016;8(343):343ra82

33. Sevelsted A, Stokholm J, Bønnelykke K, Bisgaard H. Cesarean section and

chronic immune disorders. Pediatrics. 2015;135(1). Available at: www. pediatrics. org/ cgi/ content/ full/ 135/ 1/ e92

34. Dominguez-Bello MG, De Jesus-Laboy KM, Shen N, et al. Partial restoration of the microbiota of cesarean-born infants via vaginal microbial transfer. Nat Med. 2016;22(3):250–253

35. Yatsunenko T, Rey FE, Manary MJ, et al. Human gut microbiome viewed across age and geography. Nature. 2012;486(7402):222–227

36. Cabrera-Rubio R, Collado MC, Laitinen K, Salminen S, Isolauri E, Mira A. The human milk microbiome changes over lactation and is shaped by maternal weight and mode of delivery. Am J Clin Nutr. 2012;96(3):544–551

37. Harmsen HJ, Wildeboer-Veloo AC, Raangs GC, et al. Analysis of intestinal flora development in breast-fed and formula-fed infants by using molecular identification and detection methods. J Pediatr Gastroenterol Nutr. 2000;30(1):61–67

38. Pannaraj PS, Li F, Cerini C, et al. Association between breast milk bacterial communities and establishment and development of the infant gut microbiome. JAMA Pediatr. 2017;171(7):647–654

39. Marcobal A, Barboza M, Froehlich JW, et al. Consumption of human milk oligosaccharides by gut-related microbes. J Agric Food Chem. 2010;58(9):5334–5340

40. Bezirtzoglou E, Tsiotsias A, Welling GW. Microbiota profile in feces of breast- and formula-fed newborns by using fluorescence in situ hybridization (FISH). Anaerobe. 2011;17(6):478–482

41. Gregory KE, Samuel BS, Houghteling P, et al. Influence of maternal breast milk ingestion on acquisition of the intestinal microbiome in preterm infants. Microbiome. 2016;4(1):68

42. Brugman S, Visser JT, Hillebrands JL, Bos NA, Rozing J. Prolonged exclusive breastfeeding reduces autoimmune diabetes incidence and increases regulatory T-cell frequency in bio-breeding diabetes-prone rats. Diabetes Metab Res Rev. 2009;25(4):380–387

43. Azad MB, Vehling L, Lu Z, et al; CHILD Study Investigators. Breastfeeding,

maternal asthma and wheezing in the first year of life: a longitudinal birth cohort study. Eur Respir J. 2017;49(5):1602019

44. Ma J, Prince AL, Bader D, et al. High-fat maternal diet during pregnancy persistently alters the offspring microbiome in a primate model. Nat Commun. 2014;5:3889

45. Chu DM, Antony KM, Ma J, et al. The early infant gut microbiome varies in association with a maternal high-fat diet. Genome Med. 2016;8(1):77

46. Nakajima A, Kaga N, Nakanishi Y, et al. Maternal high fiber diet during pregnancy and lactation influences regulatory T cell differentiation in offspring in mice. J Immunol. 2017;199(10):3516–3524

47. Val-Laillet D, Besson M, Guérin S, et al. A maternal Western diet during gestation and lactation modifies offspring’s microbiota activity, blood lipid levels, cognitive responses, and hippocampal neurogenesis in Yucatan pigs. FASEB J. 2017;31(5):2037–2049

48. Tormo-Badia N, Håkansson Å, Vasudevan K, Molin G, Ahrné S, Cilio CM. Antibiotic treatment of pregnant non-obese diabetic mice leads to altered gut microbiota and intestinal immunological changes in the offspring. Scand J Immunol. 2014;80(4):250–260

49. Corvaglia L, Tonti G, Martini S, et al. Influence of intrapartum antibiotic prophylaxis for Group B Streptococcus on gut microbiota in the first month of life. J Pediatr Gastroenterol Nutr. 2016;62(2):304–308

50. Keski-Nisula L, Kyynäräinen HR, Kärkkäinen U, Karhukorpi J, Heinonen S, Pekkanen J. Maternal intrapartum antibiotics and decreased vertical transmission of Lactobacillus to neonates during birth. Acta Paediatr. 2013;102(5):480–485

51. Khan I, Azhar EI, Abbas AT, et al. Metagenomic analysis of antibiotic-induced changes in gut microbiota in a pregnant rat model. Front Pharmacol. 2016;7:104

52. Gomez-Arango LF, Barrett HL, McIntyre HD, Callaway LK, Morrison M, Dekker Nitert M. Antibiotic treatment at delivery shapes the initial oral

PEDIATRICS Volume 141, number 4, April 2018 19 by guest on August 10, 2020www.aappublications.org/newsDownloaded from

Page 20: The Role of the Microbiome in the Developmental …...the developmental origins field (Fig 1). earLy-LiFe DeveLOPMeNT OF The MicrObiOMe The human microbiota is a composite organism,

microbiome in neonates. Sci Rep. 2017:7:43481

53. Scott FI, Horton DB, Mamtani R, et al. Administration of antibiotics to children before age 2 years increases risk for childhood obesity. Gastroenterology. 2016;151(1):120–129.e5

54. Hviid A, Svanström H, Frisch M. Antibiotic use and inflammatory bowel diseases in childhood. Gut. 2011;60(1):49–54

55. Shaw SY, Blanchard JF, Bernstein CN. Association between the use of antibiotics in the first year of life and pediatric inflammatory bowel disease. Am J Gastroenterol. 2010;105(12):2687–2692

56. Hoskin-Parr L, Teyhan A, Blocker A, Henderson AJ. Antibiotic exposure in the first two years of life and development of asthma and other allergic diseases by 7.5 yr: a dose-dependent relationship. Pediatr Allergy Immunol. 2013;24(8):762–771

57. Pammi M, Cope J, Tarr PI, et al. Intestinal dysbiosis in preterm infants preceding necrotizing enterocolitis: a systematic review and meta-analysis. Microbiome. 2017;5(1):31

58. Warner BB, Deych E, Zhou Y, et al. Gut bacteria dysbiosis and necrotising enterocolitis in very low birthweight infants: a prospective case-control study. Lancet. 2016;387(10031):1928–1936

59. Ward DV, Scholz M, Zolfo M, et al. Metagenomic sequencing with strain-level resolution implicates uropathogenic E. coli in necrotizing enterocolitis and mortality in preterm infants. Cell Reports. 2016;14(12):2912–2924

60. Aceti A, Gori D, Barone G, et al; Italian Society of Neonatology. Probiotics for prevention of necrotizing enterocolitis in preterm infants: systematic review and meta-analysis. Ital J Pediatr. 2015;41:89

61. Zhou Y, Shan G, Sodergren E, Weinstock G, Walker WA, Gregory KE. Longitudinal analysis of the premature infant intestinal microbiome prior to necrotizing enterocolitis: a case-control study. PLoS One. 2015;10(3):e0118632

62. Morrow AL, Lagomarcino AJ, Schibler KR, et al. Early microbial and metabolomic signatures predict later onset of necrotizing enterocolitis in preterm infants. Microbiome. 2013;1(1):13

63. Barron LK, Warner BB, Tarr PI, Shannon WD, Deych E, Warner BW. Independence of gut bacterial content and neonatal necrotizing enterocolitis severity. J Pediatr Surg. 2017;52(6):993–998

64. Li X, Li X, Shang Q, et al. Fecal microbiota transplantation (FMT) could reverse the severity of experimental necrotizing enterocolitis (NEC) via oxidative stress modulation. Free Radic Biol Med. 2017;108:32–43

65. Heida FH, van Zoonen AGJF, Hulscher JBF, et al. A necrotizing enterocolitis-associated gut microbiota is present in the meconium: results of a prospective study. Clin Infect Dis. 2016;62(7):863–870

66. Birck MM, Nguyen DN, Cilieborg MS, et al. Enteral but not parenteral antibiotics enhance gut function and prevent necrotizing enterocolitis in formula-fed newborn preterm pigs. Am J Physiol Gastrointest Liver Physiol. 2016;310(5):G323–G333

67. Bjornvad CR, Thymann T, Deutz NE, et al. Enteral feeding induces diet-dependent mucosal dysfunction, bacterial proliferation, and necrotizing enterocolitis in preterm pigs on parenteral nutrition. Am J Physiol Gastrointest Liver Physiol. 2008;295(5):G1092–G1103

68. Sim K, Shaw AG, Randell P, et al. Dysbiosis anticipating necrotizing enterocolitis in very premature infants. Clin Infect Dis. 2015;60(3):389–397

69. Cortese R, Lu L, Yu Y, Ruden D, Claud EC. Epigenome-microbiome crosstalk: a potential new paradigm influencing neonatal susceptibility to disease. Epigenetics. 2016;11(3):205–215

70. Fujimura KE, Sitarik AR, Havstad S, et al. Neonatal gut microbiota associates with childhood multisensitized atopy and T cell differentiation. Nat Med. 2016;22(10):1187–1191

71. Russell SL, Gold MJ, Willing BP, Thorson L, McNagny KM, Finlay BB. Perinatal antibiotic treatment affects murine microbiota, immune responses

and allergic asthma. Gut Microbes. 2013;4(2):158–164

72. Russell SL, Gold MJ, Reynolds LA, et al. Perinatal antibiotic-induced shifts in gut microbiota have differential effects on inflammatory lung diseases. J Allergy Clin Immunol. 2015;135(1):100–109

73. Thorburn AN, McKenzie CI, Shen S, et al. Evidence that asthma is a developmental origin disease influenced by maternal diet and bacterial metabolites. Nat Commun. 2015;6:7320

74. Azad MB, Konya T, Guttman DS, et al; CHILD Study Investigators. Infant gut microbiota and food sensitization: associations in the first year of life. Clin Exp Allergy. 2015;45(3):632–643

75. Bunyavanich S, Shen N, Grishin A, et al. Early-life gut microbiome composition and milk allergy resolution. J Allergy Clin Immunol. 2016;138(4):1122–1130

76. Olszak T, An D, Zeissig S, et al. Microbial exposure during early life has persistent effects on natural killer T cell function. Science. 2012;336(6080):489–493

77. Gollwitzer ES, Saglani S, Trompette A, et al. Lung microbiota promotes tolerance to allergens in neonates via PD-L1. Nat Med. 2014;20(6):642–647

78. Teo SM, Mok D, Pham K, et al. The infant nasopharyngeal microbiome impacts severity of lower respiratory infection and risk of asthma development. Cell Host Microbe. 2015;17(5):704–715

79. Kennedy EA, Connolly J, Hourihane JO, et al. Skin microbiome before development of atopic dermatitis: early colonization with commensal staphylococci at 2 months is associated with a lower risk of atopic dermatitis at 1 year. J Allergy Clin Immunol. 2017;139(1):166–172

80. Ismail IH, Boyle RJ, Licciardi PV, et al. Early gut colonization by Bifidobacterium breve and B. catenulatum differentially modulates eczema risk in children at high risk of developing allergic disease. Pediatr Allergy Immunol. 2016;27(8):838–846

81. Dzidic M, Abrahamsson TR, Artacho A, et al. Aberrant IgA responses to the gut microbiota during infancy

STIEMSMA and MICHELS20 by guest on August 10, 2020www.aappublications.org/newsDownloaded from

Page 21: The Role of the Microbiome in the Developmental …...the developmental origins field (Fig 1). earLy-LiFe DeveLOPMeNT OF The MicrObiOMe The human microbiota is a composite organism,

precede asthma and allergy development. J Allergy Clin Immunol. 2017;139(3):1017–1025.e14

82. Abrahamsson TR, Jakobsson HE, Andersson AF, Björkstén B, Engstrand L, Jenmalm MC. Low gut microbiota diversity in early infancy precedes asthma at school age. Clin Exp Allergy. 2014;44(6):842–850

83. Zijlmans MA, Korpela K, Riksen-Walraven JM, de Vos WM, de Weerth C. Maternal prenatal stress is associated with the infant intestinal microbiota. Psychoneuroendocrinology. 2015;53:233–245

84. Korpela K, Zijlmans MA, Kuitunen M, et al. Childhood BMI in relation to microbiota in infancy and lifetime antibiotic use. Microbiome. 2017;5(1):26

85. Nobel YR, Cox LM, Kirigin FF, et al. Metabolic and metagenomic outcomes from early-life pulsed antibiotic treatment. Nat Commun. 2015;6:7486

86. Cerdó T, Ruiz A, Jáuregui R, et al. Maternal obesity is associated with gut microbial metabolic potential in offspring during infancy [published online ahead of print August 17, 2017]. J Physiol Biochem. doi: 10. 1007/ s13105- 017- 0577- x

87. Ba Q, Li M, Chen P, et al. Sex-dependent effects of cadmium exposure in early life on gut microbiota and fat accumulation in mice. Environ Health Perspect. 2017;125(3):437–446

88. Kalliomäki M, Collado MC, Salminen S, Isolauri E. Early differences in fecal microbiota composition in children may predict overweight. Am J Clin Nutr. 2008;87(3):534–538

89. Dogra S, Sakwinska O, Soh SE, et al; GUSTO Study Group. Dynamics of infant gut microbiota are influenced by delivery mode and gestational duration and are associated with subsequent adiposity. MBio. 2015;6(1):e02419–14

90. Cho I, Yamanishi S, Cox L, et al. Antibiotics in early life alter the murine colonic microbiome and adiposity. Nature. 2012;488(7413):621–626

91. Vael C, Verhulst SL, Nelen V, Goossens H, Desager KN. Intestinal microflora and body mass index during the first three years of life: an observational study. Gut Pathog. 2011;3(1):8

92. Moussaoui N, Jacobs JP, Larauche M, et al. Chronic early-life stress in rat pups alters basal corticosterone, intestinal permeability, and fecal microbiota at weaning: influence of sex. J Neurogastroenterol Motil. 2017;23(1):135–143

93. Buffington SA, Di Prisco GV, Auchtung TA, Ajami NJ, Petrosino JF, Costa-Mattioli M. Microbial reconstitution reverses maternal diet-induced social and synaptic deficits in offspring. Cell. 2016;165(7):1762–1775

94. Carlson AL, Xia K, Azcarate-Peril MA, et al. Infant gut microbiome associated with cognitive development. Biol Psychiatry. 2018;83(2):148–159

95. Gareau MG, Jury J, MacQueen G, Sherman PM, Perdue MH. Probiotic treatment of rat pups normalises corticosterone release and ameliorates colonic dysfunction induced by maternal separation. Gut. 2007;56(11):1522–1528

96. O’Mahony SM, Marchesi JR, Scully P, et al. Early life stress alters behavior, immunity, and microbiota in rats: implications for irritable bowel syndrome and psychiatric illnesses. Biol Psychiatry. 2009;65(3):263–267

97. Foley KA, Ossenkopp KP, Kavaliers M, Macfabe DF. Pre- and neonatal exposure to lipopolysaccharide or the enteric metabolite, propionic acid, alters development and behavior in adolescent rats in a sexually dimorphic manner. PLoS One. 2014;9(1):e87072

98. Diaz Heijtz R, Wang S, Anuar F, et al. Normal gut microbiota modulates brain development and behavior. Proc Natl Acad Sci USA. 2011;108(7):3047–3052

99. Bruce-Keller AJ, Fernandez-Kim SO, Townsend RL, et al. Maternal obese-type gut microbiota differentially impact cognition, anxiety and compulsive behavior in male and female offspring in mice. PLoS One. 2017;12(4):e0175577

100. Stilling RM, Ryan FJ, Hoban AE, et al. Microbes & neurodevelopment—absence of microbiota during early life increases activity-related transcriptional pathways in the amygdala. Brain Behav Immun. 2015;50:209–220

101. Jašarević E, Howerton CL, Howard CD, Bale TL. Alterations in the vaginal microbiome by maternal stress are associated with metabolic reprogramming of the offspring gut and brain. Endocrinology. 2015;156(9):3265–3276

102. Jin S, Zhao D, Cai C, et al. Low-dose penicillin exposure in early life decreases Th17 and the susceptibility to DSS colitis in mice through gut microbiota modification. Sci Rep. 2017;7:43662

103. Candon S, Perez-Arroyo A, Marquet C, et al. Antibiotics in early life alter the gut microbiome and increase disease incidence in a spontaneous mouse model of autoimmune insulin-dependent diabetes [published correction appears in PLoS One. 2016;11(1):e0147888]. PLoS One. 2015;10(5):e0125448

104. Hu Y, Jin P, Peng J, Zhang X, Wong FS, Wen L. Different immunological responses to early-life antibiotic exposure affecting autoimmune diabetes development in NOD mice. J Autoimmun. 2016;72:47–56

105. Hu Y, Peng J, Tai N, et al. Maternal antibiotic treatment protects offspring from diabetes development in nonobese diabetic mice by generation of tolerogenic APCs. J Immunol. 2015;195(9):4176–4184

106. Livanos AE, Greiner TU, Vangay P, et al. Antibiotic-mediated gut microbiome perturbation accelerates development of type 1 diabetes in mice. Nat Microbiol. 2016;1(11):16140

107. Kostic AD, Gevers D, Siljander H, et al; DIABIMMUNE Study Group. The dynamics of the human infant gut microbiome in development and in progression toward type 1 diabetes. Cell Host Microbe. 2015;17(2):260–273

108. Miyoshi J, Bobe AM, Miyoshi S, et al. Peripartum antibiotics promote gut dysbiosis, loss of immune tolerance, and inflammatory bowel disease in genetically prone offspring. Cell Reports. 2017;20(2):491–504

109. Millar M, Seale J, Greenland M, et al. The microbiome of infants recruited to a randomised placebo-controlled probiotic trial (PiPS trial). EBioMedicine. 2017;20:255–262

PEDIATRICS Volume 141, number 4, April 2018 21 by guest on August 10, 2020www.aappublications.org/newsDownloaded from

Page 22: The Role of the Microbiome in the Developmental …...the developmental origins field (Fig 1). earLy-LiFe DeveLOPMeNT OF The MicrObiOMe The human microbiota is a composite organism,

110. Strunk T, Hibbert J, Doherty D, et al. Probiotics and antimicrobial protein and peptide levels in preterm infants. Acta Paediatr. 2017;106(11): 1747–1753

111. Trompette A, Gollwitzer ES, Yadava K, et al. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat Med. 2014;20(2):159–166

112. Harris HR, Willett WC, Vaidya RL, Michels KB. An adolescent and early adulthood dietary pattern associated with inflammation and the incidence of breast cancer. Cancer Res. 2017;77(5):1179–1187

STIEMSMA and MICHELS22 by guest on August 10, 2020www.aappublications.org/newsDownloaded from

Page 23: The Role of the Microbiome in the Developmental …...the developmental origins field (Fig 1). earLy-LiFe DeveLOPMeNT OF The MicrObiOMe The human microbiota is a composite organism,

DOI: 10.1542/peds.2017-2437 originally published online March 8, 2018; 2018;141;Pediatrics 

Leah T. Stiemsma and Karin B. MichelsThe Role of the Microbiome in the Developmental Origins of Health and Disease

ServicesUpdated Information &

http://pediatrics.aappublications.org/content/141/4/e20172437including high resolution figures, can be found at:

Referenceshttp://pediatrics.aappublications.org/content/141/4/e20172437#BIBLThis article cites 112 articles, 17 of which you can access for free at:

Subspecialty Collections

ubhttp://www.aappublications.org/cgi/collection/allergy:immunology_sAllergy/Immunologymilestones_subhttp://www.aappublications.org/cgi/collection/growth:development_Growth/Development Milestonesal_issues_subhttp://www.aappublications.org/cgi/collection/development:behaviorDevelopmental/Behavioral Pediatricsfollowing collection(s): This article, along with others on similar topics, appears in the

Permissions & Licensing

http://www.aappublications.org/site/misc/Permissions.xhtmlin its entirety can be found online at: Information about reproducing this article in parts (figures, tables) or

Reprintshttp://www.aappublications.org/site/misc/reprints.xhtmlInformation about ordering reprints can be found online:

by guest on August 10, 2020www.aappublications.org/newsDownloaded from

Page 24: The Role of the Microbiome in the Developmental …...the developmental origins field (Fig 1). earLy-LiFe DeveLOPMeNT OF The MicrObiOMe The human microbiota is a composite organism,

DOI: 10.1542/peds.2017-2437 originally published online March 8, 2018; 2018;141;Pediatrics 

Leah T. Stiemsma and Karin B. MichelsThe Role of the Microbiome in the Developmental Origins of Health and Disease

http://pediatrics.aappublications.org/content/141/4/e20172437located on the World Wide Web at:

The online version of this article, along with updated information and services, is

by the American Academy of Pediatrics. All rights reserved. Print ISSN: 1073-0397. the American Academy of Pediatrics, 345 Park Avenue, Itasca, Illinois, 60143. Copyright © 2018has been published continuously since 1948. Pediatrics is owned, published, and trademarked by Pediatrics is the official journal of the American Academy of Pediatrics. A monthly publication, it

by guest on August 10, 2020www.aappublications.org/newsDownloaded from