Science - USA (2022-05-27)

(Maropa) #1

reported in different studies. This is likely be-
cause of challenges in separating low-biomass
bacteria in fetal intestines and amniotic fluid
from environmental contaminants during tis-
sue preparation ( 12 , 13 ). Multiple studies that
have controlled for environmental contami-
nants have failed to find evidence of fetal bac-
terial colonization in the absence of infection
( 14 , 15 ). Conversely, in fetuses in which low-
abundance bacteria have been reported, tran-
scriptional adaptation in epithelial cells ( 13 )
and the presence of memory-phenotype T cells
have been interpretated as evidence in favor of
prenatal microbial colonization ( 16 , 17 ). How-
ever, microbial components and metabolites
transferred directly across the placenta, or in-


directly through maternal IgGs that cross the
placenta, are sufficient to prime fetal intestinal
epithelial cells in mice ( 11 )andcanprobably
also induce memory T cell differentiation.
Fetal T cells are hyperresponsive to foreign and
endogenous antigens ( 5 , 18 , 19 ). Such T cell
hyperresponsiveness itself could be considered
evidence for antigen scarcity in utero. This
is because T cells in general require sub-
threshold stimulation through their T cell
receptors and tune their thresholds for acti-
vation accordingly ( 20 ). An environment devoid
of triggering antigens would be expected to
induce lower thresholds for activation and
T cell hyperresponsiveness, as seen in fetuses.
Similar models have been put forward for
other immune cells, such as natural killer
cells ( 21 ).
An additional role of transplacental trans-
fer of microbial components could be to pro-
vide antigens to fetal T cells such that their
thresholds of activation can be tuned down in
preparation for postnatalmicrobialcoloniza-


tion to avoid immunopathology and permit
microbial colonization at birth and thereafter.
Trained immunity is an additional mechanism
by which fetal innate immune cells can be
prepared for postnatal exposures by epigenetic
adaptation ( 21 ). For example, monocytes of
HIV-exposed but uninfected fetuses adapt
and respond more vigorously to Toll-like re-
ceptor ligands after birth compared with HIV-
unexposed controls ( 22 ). Similar adaptive
changes have been reported in children born
to mothers infected by malaria during preg-
nancy ( 23 ). Through these multiple mecha-
nisms, fetal immune systems influenced by
maternal exposures to be prepared for the
challengingbalancingactofpermittingmicro-

bial colonization yet preventing dangerous in-
fections after birth.

Birth and first encounters
Labor is initiated by hormonal factors that are
triggered by physical pressure from the fetus
onto the cervix, but also by inflammatory sig-
nals and the breakdown of feto-maternal tol-
erance. Inflammatory stimuli such as infections
can trigger such loss of tolerance and initiate
preterm labor ( 24 ), and specific strains of
vaginal bacteria have been implicated ( 25 ).
Moreover, fetal cells triggered against mater-
nal antigens can secrete cytokines that lead to
uterine contractions, illustrating the bidirec-
tional nature of feto-maternal tolerance and its
role in preterm labor ( 26 ). Once membranes
rupture, a baby is exposed to microbes through
the birth canal. In vaginal delivery, fecal and
vaginal microbes from the mother seed the
newbornskinandmouth,whereasbabies
born through Cesarean section are populated
mainly by skin microbes, leading to substan-

tial differences in microbial composition early
after birth ( 27 ). Infants continue to receive
microbes from their parents and other sources,
but microbes seeded from the mother during
vaginal labor and originating in the intestine
are optimally adapted to survive in the infant
gut and will predominate over time, illustrating
the niche selection that occurs after birth ( 28 ).
Breastfeeding promotes specific colonizing
microbes in the newborn intestine, including
Bifidobacteria, which are adapted to metabo-
lize HMOs. Antibiotic exposures reduce over-
all microbial diversity and delay the normal
development of the intestinal microbiome ( 29 ),
and such early-life antibiotic exposure is as-
sociated with increased risks of developing
allergy ( 30 ), asthma ( 31 ), type 1 diabetes ( 32 ),
and obesity ( 33 ) later in childhood. Mecha-
nisms underlying thesealtered developmental
trajectories are discussed below.

Immune reactions to microbiota
In the first postnatal days, a strong systemic
immune response occurs in infants, which is
dominated by innate immune cells such as
monocytes expanding in the blood and the ele-
vation of circulating proinflammatory cytokines,
but also by endogenous regulatory factors such
as IL-1 receptor antagonist ( 34 ). Preterm babies
accumulate many disease-associated environ-
mental exposures, such as antibiotics, infections,
and cesarean delivery. They also have elevated
inflammatory responses in cord blood compared
with term children, likely related to prenatal
conditions triggering preterm delivery in the
first place. After birth, preterm and term in-
fants undergo similar changes in response to
postnatal exposures that are similar to all
newborns, leading to a marked convergence
of immune cell and protein profiles during
the first weeks of life in both preterm and term
infants ( 35 ). Despite these similar adaptive
changes during the first weeks of life, very
preterm babies (<31 weeks of gestation) remain
at much elevated risk (odds ratio ~3.6 versus
term infants) of asthma ( 36 ) and other immune-
mediated diseases.
After birth, cells of the immune system,
particularly those at barrier surfaces, must
recognize colonizing microbes and determine
whethertotolerateorresistthem.Theestab-
lishment of host-microbe mutualism is critical
for health, and several processes work to pro-
mote this. Children lacking Tregs (e.g., those
with immunodysregulation polyendocrinopathy
enteropathy X-linked syndrome) typically pre-
sent early in life with severe inflammation
involving both the intestines and skin after
microbial colonization ( 37 ). In mice, a distinct
population of FoxP3+Tregs are induced in the
thymus during the perinatal period that are
essential for maintaining self-tolerance and
preventing autoimmunity later in life ( 38 ). Ad-
ditional experiments in mice have shown that

Brodin,Science 376 , 945–950 (2022) 27 May 2022 2of


A Fetal immune system and layered hematopoiesis B Transplacental transfer of IgG and
microbial components

16 20 36 Birth Adulthood
Gestational week

Maternal IgG
carrying microbial
components

Passive immunity
Priming of fetal
epithelial cells for
postnatal colonization
Tuning/training of fetal
immune cells?

Feto-maternal
tolerance

Fe t a l
hematopoiesis

Resistance
to pathogens

Adult
hematopoiesis

Variable transition period

IFN-1–triggered
switch (infection?)

Fig. 1. Prenatal preparation.(A) Layered hematopoiesis occurs with different stem cell populations that
give rise to cells poised toward feto-maternal tolerance early during pregnancy but gradually switch over
toward pathogen resistance at the end of gestation in preparation for microbial colonization. The transition
period is gradual in nature ( 7 ), and one trigger for this switch from fetal to adult hematopoiesis is a surge
in IFN-I before birth ( 8 ). ( B) Transplacental transfer of maternal IgG, and possibly microbial antigens,
prepares fetal immune systems and epithelia for microbial colonization at birth and thereafter.


ILLUSTRATION: KELLIE HOLOSKI/

SCIENCE
Free download pdf