ArticlePDF Available

The maternal immune system during pregnancy and its influence on fetal development

Authors:

Abstract and Figures

Sara S Morelli,1 Mili Mandal,2 Laura T Goldsmith,1 Banafsheh N Kashani,1 Nicholas M Ponzio3,41Department of Obstetrics, Gynecology and Women's Health, New Jersey Medical School, Rutgers University, Newark, 2Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, 3Department of Pathology and Laboratory Medicine, New Jersey Medical School, Rutgers University, Newark, 4Graduate School of Biomedical Sciences, Rutgers University, Newark, NJ, USAAbstract: The maternal immune system plays a critical role in the establishment, maintenance, and completion of a healthy pregnancy. However, the specific mechanisms utilized to achieve these goals are not well understood. Various cells and molecules of the immune system are key players in the development and function of the placenta and the fetus. Effector cells of the immune system act to promote and yet limit placental development. The T helper 1 (Th1)/T helper 2 (Th2) immune shift during pregnancy is well established. A fine balance between proinflammatory and anti-inflammatory influences is required. We herein review the evidence regarding maternal tolerance of fetal tissues and the underlying cell-mediated immune and humoral (hormones and cytokines) mechanisms. We also note the many unanswered questions in our understanding of these mechanisms. In addition, we summarize the clinical manifestations of an altered maternal immune system during pregnancy related to susceptibility to common viral, bacterial, and parasitic infections, as well as to autoimmune diseases.Keywords: maternal–fetal interface, immune system, fetal tolerance, lymphocyte subsets, decidua, pregnancy
Content may be subject to copyright.
© 2015 Morelli et al. This work is published by Dove Medical Press Limited, and licensed under Creative Commons Attribution – Non Commercial (unported, v3.0)
License. The full terms of the License are available at http://creativecommons.org/licenses/by-nc/3.0/. Non-commercial uses of the work are permitted without any further
permission from Dove Medical Press Limited, provided the work is properly attributed. Permissions beyond the scope of the License are administered by Dove Medical Press Limited. Information on
how to request permission may be found at: http://www.dovepress.com/permissions.php
Research and Reports in Biology 2015:6 171–189
Research and Reports in Biology Dovepress
submit your manuscript | www.dovepress.com
Dovepress 171
REVIEW
open access to scientific and medical research
Open Access Full Text Article
http://dx.doi.org/10.2147/RRB.S80652
Video abstract
Point your SmartPhone at the code above. If you have a
QR code reader the video abstract will appear. Or use:
http://youtu.be/JvJoMUHyJlE
The maternal immune system during pregnancy
and its inuence on fetal development
Sara S Morelli1
Mili Mandal2
Laura T Goldsmith1
Banafsheh N Kashani1
Nicholas M Ponzio3,4
1Department of Obstetrics,
Gynecology and Women’s Health,
New Jersey Medical School, Rutgers
University, Newark, 2Department of
Pha rma col ogy and Toxicology, Ern est
Mario School of Pharmacy, Rutgers
University, Piscataway, 3Department of
Pathology and Laboratory Medicine,
New Jersey Medical School, Rutgers
University, Newark, 4Graduate School
of Biomedical Sciences, Rutgers
University, Newark, NJ, USA
Correspondence: Sara S Morelli
Department of Obstetrics, Gynecology
and Women’s Health, New Jersey
Medical School, Rutgers University,
MSB E-506, 185 South Orange
Avenue, Newark, NJ 07103, USA
Tel +1 973 972 4127
Fax +1 973 972 4574
Email morellsa@njms.rutgers.edu
Abstract: The maternal immune system plays a critical role in the establishment, maintenance,
and completion of a healthy pregnancy. However, the specific mechanisms utilized to achieve
these goals are not well understood. Various cells and molecules of the immune system are key
players in the development and function of the placenta and the fetus. Effector cells of the immune
system act to promote and yet limit placental development. The T helper 1 (Th1)/T helper 2 (Th2)
immune shift during pregnancy is well established. A fine balance between proinflammatory
and anti-inflammatory influences is required. We herein review the evidence regarding maternal
tolerance of fetal tissues and the underlying cell-mediated immune and humoral (hormones and
cytokines) mechanisms. We also note the many unanswered questions in our understanding of
these mechanisms. In addition, we summarize the clinical manifestations of an altered mater-
nal immune system during pregnancy related to susceptibility to common viral, bacterial, and
parasitic infections, as well as to autoimmune diseases.
Keywords: maternal–fetal interface, immune system, fetal tolerance, lymphocyte subsets,
decidua, pregnancy
Introduction
The relationship between mother and fetus has fascinated immunologists for decades.
Survival of the semiallogeneic fetus was used by Billingham et al1 in 1953 as an
example of immune tolerance to the fetus by the maternal immune system. Numerous
hypotheses related to placental protection of the fetus, including expression (or lack
of expression) of histocompatibility antigens on fetal tissues, maternal immune toler-
ance to fetal antigens, and inhibition and/or regulation of maternal antifetal immune
responses have been put forth to explain the survival of the “immunogenic” fetus. Yet,
the mechanisms still remain to be totally clarified.
Part of the difficulty in studying these mechanisms is due to the variation among
species in which such investigations are conducted. Mice are used for many of these
investigations because of their short gestational time, relatively lower cost, well-defined
genetics (including mutant, transgenic, and knockout strains), and availability of a
wide spectrum of antibodies and reagents to perform immunologic and molecular
studies. However, differences in the reproductive system in general, and the feto–
materno–placental unit in particular, as well as differences in the development and
function of immune elements, often preclude direct extension of results observed in
mice to humans. In contrast, studies designed to investigate such questions in humans
are unethical, and studies incorporating nonhuman primates for these investigations
raise similar moral issues and are also prohibitively expensive.
Number of times this article has been viewed
This article was published in the following Dove Press journal:
Research and Reports in Biology
1 October 2015
Research and Reports in Biology 2015:6
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
172
Morelli et al
Therefore, our review is not designed to address all the
unanswered questions surrounding the significance of the
maternal immune system during pregnancy and its influence
on fetal development. Rather, our goals are to identify the
gaps in the knowledge and understanding about the topic
from the published literature about various species and to
acknowledge contexts wherein differences preclude a direct
comparison with humans. Notwithstanding these differences
however, investigations conducted in other species, such
as rodents, do serve to identify possible strategies to address
some of these unanswered questions.
Additionally, we take an interdisciplinary approach as
coauthors who bring clinical and basic science perspectives
and expertise in reproductive and immunological disciplines.
Thus, we address topics related to definition of the maternal–
fetal interface, as well as the significance of maternal immune
responses in regulating key early events during both preg-
nancy (eg, implantation, angiogenesis, and vascular remodel-
ing) and in development of the fetal immune system. We then
review current understanding about maternal tolerance of
fetal tissues and the underlying cellular and humoral immune
mechanisms. Finally, we examine clinical manifestations of
an altered maternal immune system during pregnancy related
to susceptibility to certain viral, bacterial, and parasitic infec-
tions, as well as to autoimmune disorders.
Description and denition of the
maternal–fetal interface
Maternal: decidua
In women, invasion by the trophoblast is extensive,
encompassing the endometrium as well as the inner third
of the myometrium.2 To accommodate this, a pronounced
remodeling process must occur, involving multiple cellular
compartments of the uterus in preparation for implantation
and establishment and support of pregnancy. This process,
decidualization, occurs in humans on a cyclic basis beginning
in the midluteal phase of the menstrual cycle, independently
of pregnancy. In contrast, in rodents and most other species,
decidualization requires the presence of a blastocyst. The
term maternal decidua thus refers to the uterine mucosal
layer (endometrium) after it has undergone decidualization,
the requisite and complex differentiation process involving
the multiple cellular compartments of the endometrium in
preparation for embryo implantation.
The parenchymal cellular compartments of the maternal
decidua include the glandular epithelial compartment, the
luminal epithelial compartment, the endothelium of the spiral
arteries, and the decidualized stromal cells, all of which
undergo dramatic transformation in preparation for pregnancy.
The glandular epithelium acquires increased secretory activ-
ity under the influence of maternal progesterone.3 Dramatic
remodeling of spiral arteries occurs during decidualization,
discussed in greater detail later in this review. The endome-
trial stromal fibroblasts that undergo dramatic morphologic
and biochemical differentiation in preparation for implanta-
tion and support of pregnancy become known as decidual
cells, or decidualized stromal cells. Decidualized stromal
cells no longer have the characteristic spindle shape of the
endometrial stromal fibroblast and, instead, have acquired
an epithelioid phenotype, characterized by progressive cell
enlargement, rounding of the nucleus, and expansion of the
rough endoplasmic reticulum and Golgi complex, all con-
sistent with the transformation into a secretory cell.3 Major
secretory products of decidualized stromal cells include
prolactin and insulin-like growth-factor-binding protein-1,
the hallmark proteins widely used as phenotypic markers of
decidualization.4 These cells also secrete a number of cytok-
ines and growth factors (eg, interleukin [IL]-11, epidermal
growth factor [EGF], heparin-binding EGF-like growth
factor), which further regulate the process of decidualization
in an autocrine and/or paracrine manner.5
In addition to the parenchymal cellular compartments
making up the maternal decidua, various populations of
immune cells exist in the human endometrium throughout
the menstrual cycle. In early pregnancy, leukocytes are
abundant, comprising 30%–40% of all human decidual
stromal compartment cells.6 The basalis layer of the human
endometrium contains lymphoid aggregates composed of
T-cells and a small number of B-cells. In the functionalis layer
of the proliferative phase, few uterine natural killer (uNK)
cells, T-cells, and macrophages are scattered throughout the
stromal compartment.7 Although the numbers of T-cells and
macrophages remain largely unchanged throughout the luteal
phase and during the process of decidualization,7 there is a
dramatic increase in the number of uNK cells postovulation,
playing a critical role in preparation of the endometrium for
pregnancy. With regard to decidual immune cell popula-
tions during early pregnancy, studies using flow cytometry
and immunostaining of human tissues demonstrate that the
majority of first-trimester human decidual leukocytes are
uNK cells (70%), followed by macrophages (20%).8 T-cells
make up approximately 10%–20% of decidual leukocytes,
and dendritic cells (DCs) and B-cells are rare.8 As in humans,
uNK cells are the predominant leukocyte population in the
decidua of the rhesus macaque and the mouse, but studies to
determine relative numbers of other leukocyte populations in
Research and Reports in Biology 2015:6 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
173
Maternal immune system and fetal development
murine decidua are lacking. The functions of each immune
cell type at the maternal–fetal interface are discussed in more
detail in this review, with a particular focus on uNK cells.
Fetal: placenta, fetal membranes
(amnion and chorion)
Structurally, the interface between the uterine mucosa
and the extraembryonic tissues is commonly referred
to as the maternal–fetal interface. This is represented in
Figure 1, which depicts the maternal immune cells and the
fetal trophoblast.9
Extraembryonic cells in direct contact with maternal
cells are the trophoblast cells, derived from the trophecto-
derm layer surrounding the blastocyst. In women, invasion
by the trophoblast into maternal spiral arteries substantially
increases uterine blood flow, puts maternal blood in direct
contact with fetal trophoblast cells, and ensures sufficient
delivery of maternal nutrients and oxygen to the placenta.10
However, the maternal and fetal circulations do not mix.
After attachment of the blastocyst to the endometrial luminal
epithelium, trophoblast cells invade the decidua as depicted
in Figure 1. The trophoblast, composed of an inner cell layer
(cytotrophoblast) and outer cell layer (syncytiotrophoblast),
does not give rise to the fetus itself, but rather to the placenta
and fetal membranes (amnion and chorion). As the blastocyst
and surrounding trophoblast invade the decidua, one pole
of the blastocyst remains oriented toward the endometrial
lumen, and the other remains buried in the decidua, which will
develop into the anchoring cytotrophoblasts and villous tro-
phoblasts, contributing to formation of the placenta, chorion,
and amnion. Of note are the species differences in the degree
of invasion by trophoblast cells, which have been documented
in detail elsewhere.11 In distinct contrast to the process in
women, trophoblast invasion is minimal in rodents.11,12
Signicance of maternal immune
responses during pregnancy
Immune cell subtypes and their
functional signicance
Immune cells accumulating in the human endometrium at
the time of decidualization play critical and diverse roles
at the maternal–fetal interface, including functions in
implantation, placental development, and immunity against
infectious diseases. Of all decidual leukocyte populations,
the most abundant are the phenotypically unique uNK
cells. These cells dramatically increase in number in the
human endometrium 3–5 days postovulation, accounting for
25%–40% of endometrial leukocytes prior to implantation
Mother
(decidua)
Fetus
(placenta)
Trophoblast
Luminal epithelium
Macrophage
B-cell
Dendritic cell
Th cell
Tc cell
uNK cell
Decidual
stromal cell
Figure 1 Schematic depiction of the human maternal–fetal interface including maternal immune cells such as uterine natural killer (uNK) cells, macrophages, (the predominant
immune cell types) and T helper (Th) cells, T-cytotoxic (Tc) cells, dendritic cells, as well as invading trophoblast cells.
Notes: Copyright © 2009 by SAGE Publications. Modied from: Weiss G, Goldsmith LT, Taylor RN, Bellet D, Taylor HS. Inammation in reproductive disorders. Reproductive
Sciences. 2009;16(2):216–229; by permission of SAGE Publications.9
Research and Reports in Biology 2015:6
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
174
Morelli et al
and accounting for 70% of decidual leukocytes in the first
trimester.7,8 It is critical to note that uNK cells are both
phenotypically and functionally distinct from peripheral
NK cells. Phenotypically, they are identified by expression
of the NK cell marker CD56, expressed at high concentra-
tions (CD56bright), but they lack expression of CD16, found
on most peripheral NK cells (CD56dimCD16+).7 In terms
of function, peripheral CD56dimCD16+ NK cells are highly
cytotoxic, mediating both natural and antibody-dependent
killing, whereas uNK cells are only weakly cytotoxic and
do not normally kill trophoblast cells.13 In addition, uNK
cells are a potent source of immunoregulatory cytokines,14
matrix metalloproteinases (MMPs),15 and angiogenic fac-
tors.16 These various factors mediate extracellular matrix
remodeling, trophoblast invasion, and angiogenesis, which
are key processes in placentation and establishment of early
pregnancy at the maternal-fetal interface.17
In addition to uNK cells, decidual macrophages are rela-
tively abundant, comprising 20% of the human decidual leu-
kocyte population in the first trimester.8 In normal pregnancy,
most of the macrophages at the maternal–fetal interface are
of the M2 (immunomodulatory) phenotype.18 Present in
decidua prior to the presence of extravillous trophoblast,19
macrophages play a role in early spiral artery remodeling
by producing factors associated with tissue remodeling
(MMP-9) and angiogenesis (vascular endothelial growth fac-
tor [VEGF]).18 Apoptosis is an important event during spiral
artery remodeling and trophoblast invasion, and decidual
macrophages phagocytose apoptotic cells in remodeled vas-
cular wall and apoptotic trophoblast cells, thereby preventing
the release of proinflammatory substances from the apoptotic
cells into the decidua.20 First-trimester decidual macrophages
may also be responsible for inhibition of human uNK cell–
mediated lysis of invasive cytotrophoblast, mediated by
decidual secretion of transforming growth factor-beta-1
(TGF-β1), as demonstrated in human in vitro studies.21 In
distinct contrast to human uNK cells, which peak in number
at 20 weeks gestation and are nearly absent in the decidua at
term,12 decidual macrophages are present throughout preg-
nancy, but the precise role of decidual macrophages at the
end of pregnancy remains unknown.18
T-cells are also fairly abundant in human decidua,
comprising 10%–20% of the human decidual leukocyte
population,22,23 of which 30%–45% are CD4+ T-cells and
45%–75% are CD8+ T-cells.23 The main function of T-cells
in the decidua, particularly of CD4+ T-regulatory (Treg) cells,
is generally thought to be the promotion of tolerance to the
fetus24 (discussed in detail later in this review). However,
because a variety of different T-cell subsets are present, the
complex interactions of T-cells in the decidua have not been
completely defined.25 Human in vitro studies of CD8+ T-cells
isolated from first-trimester decidua demonstrate that these
cells exhibit cytotoxic activity as well as cytokine produc-
tion (predominantly interferon-gamma [IFN-γ] and IL-8).26
Since decidual CD8+ T-cell supernatants increase the in vitro
invasive capacity of extravillous trophoblast cells, secreted
products of CD8+ T-cells may play a role in regulation of
trophoblast invasion, but precise mediators have not yet
been identified.26
DCs, which are antigen-presenting cells that play a critical
role in regulation of the adaptive immune response, make up
a very small portion of human decidual leukocytes. However,
no single specific marker for DCs exists and their phenotypic
definition is therefore controversial, thereby limiting the
existing studies of decidual DCs.27 Using lineage-negative
and human leukocyte antigen-DR-positive (HLA-DR+) status
as a combination marker for DCs, Gardner and Moffett28
demonstrated that decidual DCs comprised 1% of first-
trimester human decidual leukocytes. Due to the rarity of
this cell population, functional studies of human decidual
DCs are scarce. Human in vitro studies have demonstrated
that decidual DCs, isolated from early-pregnancy decidua,
are more likely than peripheral DCs to prime naïve CD4+
T-cells into a Th2 phenotype, suggesting a potential role
for decidual DCs in averting Th1-mediated rejection of
the fetus.29 Decidual DCs also appear to regulate uNK cell
function, since coculture of decidual DCs with uNK cells
stimulated uNK cell proliferation and activation.30 In vivo
functional studies of decidual DCs exist only in mice and
are more definitive. Decidual DC–depleted mice exhibit
severely impaired implantation, impaired decidual prolif-
eration and differentiation, impaired angiogenesis, impaired
differentiation of uNK cells, and resorption of embryos.31,32
Therefore, at least in mice, decidual DCs play an important
role in decidualization and establishment and maintenance
of early pregnancy.
Mechanisms by which immune cells
(focus: uNK cells) regulate key early
events in establishment of pregnancy:
implantation, angiogenesis, and vascular
remodeling
uNK cells regulate trophoblast invasion
Studies performed by Hanna et al33 provided strong evi-
dence that human uNK cells play a role in regulation of
Research and Reports in Biology 2015:6 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
175
Maternal immune system and fetal development
trophoblast invasion. These investigators demonstrated
that uNK cells isolated from first-trimester human decidua
express the chemokines IL-8 and IFN-inducible protein (IP)-
10, and that purified human invasive trophoblasts express
the chemokine receptors for these ligands: CXCR1 (IL-8
receptor) and CXCR3 (IP-10 receptor). The ability of uNK
cells, but not peripheral blood NK cells, to induce trophoblast
migration in an in vitro trophoblast migration assay was
significantly reduced in the presence of neutralizing anti-
bodies to IL-8 and IP-10. These investigators subsequently
performed in vivo studies in which NK cell subsets embed-
ded in Matrigel were injected into the subcutaneous tissues
of nude mice, and human trophoblast cells were injected
around the Matrigel plug. These in vivo experiments further
demonstrated that uterine, but not peripheral, NK cells pro-
moted trophoblast invasion, and that migration of tropho-
blast cells into the Matrigel plug was significantly reduced
in the presence of IL-8- and IP-10-neutralizing antibodies.
Overall, these studies demonstrated the ability of uNK cells
to positively regulate invasion of trophoblast, mediated by
the uNK-derived cytokines IL-8 and IP-10.33 However, tro-
phoblast invasiveness into maternal decidua must be tightly
regulated. The balance of factors involved in regulation of
invasion is not yet precisely determined. Excessive invasion
predisposes to placenta accreta, a potentially life-threatening
obstetrical condition in which the placenta attaches abnor-
mally to the uterine myometrium.34 Interestingly, human uNK
cells also have the ability to inhibit trophoblast invasion, as
demonstrated by Lash et al35 using in vitro Matrigel inva-
sion assays. These investigators demonstrated that human
uNK cells isolated from early human pregnancy decidua
are a source of IFN-γ, which inhibits trophoblast invasion
by increasing apoptosis of extravillous trophoblast cells and
decreasing trophoblast secretion of MMP-2.35 Thus, the fine
balance required to avoid either underinvasion or overinva-
sion of trophoblast in early human pregnancy is regulated, at
least in part, by the various cytokines derived from human
uNK cells present in decidua.
Role of uNK cells in angiogenesis and vascular
remodeling in early pregnancy
In humans, extensive vascular remodeling must occur to
allow for placentation and establishment of early pregnancy,
as well as to support the demands of a growing fetus. The
decidual spiral arteries must be transformed into larger-
diameter vessels with low resistance and high flow, capable of
transporting nutrients and oxygen to the fetus.22 In addition,
the endothelium of these vessels is replaced by extravillous
trophoblast cells that have migrated from the placenta,
allowing for diversion of blood flow into the space surround-
ing the placental villous tree and thereby permitting nutrient
and gas exchange between mother and fetus.36 Not only is
adequate vascular remodeling critical for the establishment
of a normal pregnancy, but abnormalities in these early events
are associated with later complications of pregnancy such as
preeclampsia and intrauterine growth restriction, which can
have a major impact on fetal and neonatal health.34
A critical role for uNK cells in vascular remodeling has
been demonstrated in both murine in vivo and human in
vitro studies. However, it is important to note significant
differences among species in terms of strategies to increase
blood flow to the site of maternal–placental exchange. In
humans, extensive invasion and destruction of preexisting
arteries by trophoblast occurs. In nonhuman primates such
as rhesus macaques, trophoblastic invasion and modification
of uterine arteries occurs, but unlike in humans, invasion of
decidual stroma by trophoblast in the rhesus monkey occurs
only to a minimal extent.12 In mice, the extent to which the
trophoblast invades both the decidual stroma and uterine
arteries is even more limited.12 Rodent models thus have
limited value in advancing our understanding of mechanisms
of vascular remodeling that facilitate human pregnancy.
Nevertheless, there are in vivo studies performed in mice
that cannot be performed in humans, and the availability
of nonhuman primates for such in vivo studies in early
pregnancy is limited. Therefore, much of the existing data
on uNK cell functions in vascular remodeling are derived
from murine studies.
Multiple murine in vivo studies demonstrate that uNK
cells play a critical role in the remodeling of endometrial
spiral arteries both prior to and during pregnancy. The
earliest studies demonstrating a critical role for uNK cells
in vascular remodeling in pregnancy were those conducted
by Guimond et al,37 who demonstrated several reproductive
abnormalities in the TgΕ26 mouse strain, which is deficient
in NK cells. Multiple vascular abnormalities associated
with implantation sites, including thickening of the media
and adventitia, endothelial damage, reduction in placental
size, and onset of fetal loss at Day 10 of gestation, were
demonstrated in NK-cell-deficient mice. Subsequent studies
from the same laboratory38 demonstrated that bone marrow
transplantation from severe combined immunodeficient
mice (which lack T- and B- lymphocytes but not NK cells)
to NK-cell-deficient mice led to restoration of the uNK cell
population in recipients, reduced anomalies in decidual
blood vessels, increased placental size, and restored fetal
Research and Reports in Biology 2015:6
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
176
Morelli et al
viability. Overall, these studies provide strong support
for a critical role of murine uNK cells in decidualiza-
tion, placentation, and the appropriate vascularization of
implantation sites.
The role of murine uNK cells in vascular remodeling
and decidualization appears to be mediated via IFN-γ, since
transgenic mice that lack IFN-γ or its receptor fail to initi-
ate modification of decidual arteries and exhibit necrosis
of decidual cells, and treatment of NK-deficient mice with
recombinant IFN-γ rescues decidual morphology and initiates
decidual vessel modification.39,40 However, whether human
uNK cells regulate decidual vascular remodeling via IFN-γ
is yet to be definitively determined. The data regarding IFN-γ
expression by human uNK cells are conflicting, likely due to
differences in methodology between studies and the status of
cytokine stimulation of the uNK cells being studied. Evidence
for production of IFN-γ in unstimulated human uNK cells
is limited, but after exposure to stimulatory cytokines such
as IL-2, IL-12, or IL-15, human uNK cells isolated from
first-trimester decidua exhibit significantly increased IFN-γ
secretion.41,42 In addition, because IFN-γ is rapidly secreted
once produced, and expression of IFN-γ mRNA and protein
by human uNK cells rapidly decreases after 24–48 hours
in culture,35 conflicting data regarding IFN-γ expression by
human uNK cells may be attributable to length of time in
culture before measurement. In a nonhuman primate model
of early pregnancy, the major population of CD56bright uNK
cells isolated from early-pregnancy rhesus monkey decidua
is not a source of IFN-γ.43 Therefore, while compelling evi-
dence exists to support the role of IFN-γ in decidual vascular
remodeling in rodents, whether uNK cell-derived IFN-γ plays
an equally important role in vascular remodeling in humans
and in nonhuman primates remains unclear.
Rather, the finding that human uNK cells isolated from
first-trimester decidua are a potent source of the angiogenic
factors angiopoietin (Ang)1, Ang2, VEGF, and PLGF16,33 sup-
ports an important role for these cells in the vascular remod-
eling required for successful human pregnancy. Functional
studies by Hanna et al33 demonstrated that human uNK cells
isolated from first-trimester decidua are potent secretors of
angiogenic factors such as VEGF and placental growth factor
(PLGF). Supernatants derived from human uterine (but not
peripheral) NK cells promoted in vitro angiogenesis, as dem-
onstrated by an increased ability of human umbilical vascular
endothelial cells to form network-like structures, a process
inhibited in the presence of VEGF- and PLGF-neutralizing
proteins. In addition, these investigators33 demonstrated the
in vivo ability of human uNK cells to promote angiogenesis
and growth of human trophoblast choriocarcinoma (JEG-3)
tumor cells when injected subcutaneously into nude mice.
In vivo angiogenic properties of uNK cells were inhibited
in the presence of a VEGF- and PLGF-neutralizing protein.
These studies provide strong evidence that the angiogenic
properties of human uNK cells are mediated, at least in part,
by their secretion of VEGF and PLGF.
Inuence of maternal immune
response on development of
the fetal immune system
Compelling clinical data demonstrate that children of mothers
exposed to certain infectious organisms during pregnancy
have significantly higher frequencies of neurological dis-
orders,44–53 including schizophrenia and autism spectrum
disorders. In such scenarios, the etiology of these disorders
has been linked to activation of the maternal inflammatory/
immune responses (reviewed by Jonakait54 and Patterson55).
Rodent studies in which the maternal immune system is acti-
vated during pregnancy replicate these clinical findings and
provide validated mouse models of these disorders.46,47,51,56–66
Thus, maternal immune stimulation during pregnancy acts
as an environmental risk factor that affects development of
the brain and the immune system in the offspring.
The underlying mechanisms of these phenomena have
been studied primarily in prenatal rodent models, in which
pregnant dams are injected with either infectious pathogens
or synthetic agents that mimic viral or bacterial infections
(namely, lipopolysaccharides and polyinosinic:polycytidylic
acid [poly(I:C)]). Offspring of such immunostimulated
pregnant dams exhibit immune dysregulation and behavioral
abnormalities, as well as chemical and structural anomalies of
the brain, which are similar to those seen in individuals with
schizophrenia and autism spectrum disorders.63,67–72
There is a transient increase of cytokines (IL-1, IL-6,
IL-12, tumor necrosis factor-alpha [TNF-α], granulocyte-
macrophage colony stimulating factor) in the blood and
amniotic fluid of immunostimulated pregnant dams,73,74
which appears to influence development of the fetal immune
system, a concept known as “fetal programming”.75–79 Mandal
et al73,74,80 have also shown that offspring of immunostimu-
lated pregnant dams exhibit accelerated development and
heightened responsiveness of Th1, Th17, and cytotoxic effec-
tor T-cell subsets, indicating a proinflammatory phenotype
in these offspring.
We hypothesized that in utero exposure of the fetus to
cytokines elicited by maternal immune stimulation acts as
a “first hit” to influence fetal programming of the immune
Research and Reports in Biology 2015:6 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
177
Maternal immune system and fetal development
system, which persists postnatally and into adulthood.
Such alterations of normal fetal programming results in
development of a “proinflammatory” phenotype, and upon
subsequent postnatal exposure to an immune stimulus (ie,
second hit), the offspring of the immunostimulated pregnant
dams exhibit exacerbated responses in comparison to off-
spring of phosphate-buffered saline (PBS)-injected dams.
Such a scenario is also consistent with the “multiple hit”
concept of mental disorders.81,82 In the context of neurode-
velopmental disorders, this would mean that abnormalities
of behavior and immune dysregulation observed in some
affected children could reflect such altered fetal programming
that is manifested postnatally upon encounter with a second
hit (eg, infection) to their immune system. We tested this
hypothesis in adult offspring of immunostimulated pregnant
dams using well-documented in vivo experimental models
that involve activation of the innate and/or adaptive immune
systems. In each of these models, the adult offspring of
immunostimulated dams mounted a more robust inflamma-
tory response than adult offspring of control dams injected
with PBS.73,83 Thus, offspring from immunostimulated dams
exhibit behavioral anomalies reminiscent of those seen in
individuals with some neurodevelopmental disorders, such
as schizophrenia and autism. In addition to their behavioral
abnormalities, our studies show that as a result of in utero
exposure to products of maternal immune stimulation, these
adult offspring also exhibit a “proinflammatory” phenotype
that confers a vulnerability to develop immune-mediated
pathology after birth and into adulthood.73,74,80
In this regard, the results obtained from our investigations
in mouse models have provided the scientific rationale for an
ongoing translational research project to determine whether
similar molecular pathogenic mechanisms are involved in a
cohort of autistic children who also exhibit diagnostic evi-
dence of immune dysregulation.84 Using DNA obtained from
the Autism Genetic Resource Exchange database, we initiated
a study to determine whether polymorphisms in selected
maternal cytokine genes occurred more frequently in moth-
ers of these autistic children. Our results show that mothers
of autistic children in this cohort have significantly higher
frequencies of proinflammatory cytokine gene polymor-
phisms, thereby conferring the genetic capability to respond
more vigorously to immune stimulation by producing the
types and amounts of cytokines that promote inflammatory
reactions. Moreover, analysis of preliminary data from the
offspring indicates that the autistic children of these mothers
inherit the maternal genotype. Thus, results obtained from
our investigation of the experimental prenatal mouse model
of maternal immune stimulation during pregnancy73 appear
to have biological relevance to humans.
Maternal–fetal tolerance
Billingham et al1 in 1953 were the first to propose the con-
cept of immune tolerance during pregnancy. They hypoth-
esized that the semiallogeneic fetus is able to survive due to
regulation of the immunologic interactions between mother
and fetus. Such regulation can be caused by a lack of fetal
antigen expression and/or functional suppression of maternal
immune response.1
HLAs that are expressed in the fetal membranes are
tolerogenic rather than immunogenic,85 and expression
of major histocompatibility complex (MHC) proteins at
the maternal–fetal interface is tightly regulated during
pregnancy.86 The MHC class I genes are subdivided into
classes Ia and Ib. The MHC class Ia is further subdivided
into HLA-A, B, and C and class Ib is subdivided into
HLA-E, F, and G. HLA class II (HLA-D) genes are not
translated in human trophoblast cells.87 Human trophoblast
cells express one MHC class Ia (HLA-C) and all MHC class
Ib molecules. In human placenta, fetal trophoblast cells do
not express MHC class Ia (HLA-A and B) molecules that
are responsible for the rejection of allografts in humans.88,89
Interactions between HLA-C and decidual NK cells may
also cause infiltration of trophoblast into maternal tissue.
Pregnancies with mismatched fetal HLA-C exhibit a greater
number of activated T-cells and functional Tregs in decidual
tissues compared to HLA-C-matched pregnancies.90 This
suggests that in uncomplicated pregnancies, decidual T-cells
recognize fetal HLA-C at the maternal–fetal interface
but are prevented from inducing a destructive immune
response.91
Regarding pregnancy, one of the most important ques-
tions is how the fetal–placental unit escapes maternal
rejection. Although there is a continuous interaction
between the fetus and maternal cells throughout pregnancy,
the fetus acts as a privileged site that is protected from
immune rejection.91 Expression of MHC molecules on
trophoblast cells is repressed in most of the species as a
strategy to avoid recognition and destruction by the mater-
nal immune cells.92 Peripheral blood lymphocytes from
pregnant mares demonstrate reduced capacity to develop
into effector cytotoxic T lymphocytes.93 This reduction
in T-cell-mediated alloreactivity returns to normal after
termination of pregnancy and is not observed in nonpreg-
nant mares. In addition, extracts from Day 80 placentas
from mares have been shown to inhibit proliferation of
Research and Reports in Biology 2015:6
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
178
Morelli et al
maternal lymphocytes, and coculture of trophoblast cells
with maternal lymphocytes caused reduction in prolifera-
tion and cytokine production.94
Cell-mediated immunity:
mechanisms promoting
maternal–fetal tolerance
The Th1–Th2 shift in pregnancy
Pregnancy is a complex immunological state, wherein the
mother must tolerate the “foreign” fetus, and thus requires
a degree of immunosuppression. On the other hand, the
mother must maintain sufficient immune function to fight off
infection. One mechanism that plays a role in maintenance
of successful pregnancy is a switch from the Th1 cytokine
profile to the Th2 profile. This switch is more prominent
at the maternal–fetal interface. Th2 cells accumulate in
decidua, and uterine DCs can drive naïve T-cells to become
Th2 cells.95,96 Therefore, the switch to a Th2 phenotype is due
to both migration of Th2 cells and induction of Th2 cells at
the maternal–fetal interface, but there is little change in the
systemic immune system.96 The hypothesis of Th2 predomi-
nance and downregulation of Th1 response during pregnancy
was proposed by Wegmann et al,97 which is supported by
both murine and human studies. In mice, the proinflamma-
tory cytokines IFN-γ and TNF-α, or stimulation of toll-like
receptors, induce miscarriage, which can be reversed by
inhibitors of Th1 cytokines or by administration of anti-
inflammatory IL-10 (Th2 cytokine).98 However, IFN-γ also
plays an important role in vascular remodeling in early
murine pregnancy. Therefore, Th1-type immunity appears
to be controlled to avoid overstimulation during pregnancy.
Progesterone, estradiol, prostaglandin D2 (PGD2), and leu-
kemic inhibitory factor generated during pregnancy promote
the Th2 profile and are, in part, responsible for the Th2 bias
associated with normal pregnancy.96 However, transgenic
Th2 cytokine single-knockout mice such as IL-4-/-, IL-10-
/-99 and mice with single, double, triple, and quadruple gene
deletions of IL-4, IL-5, IL-9, and IL-13 have normal preg-
nancies, suggesting that a predominant Th2-type immunity
might not be essential for successful pregnancy.100
An increase of Th2 cytokines IL-4, IL-10, and mono-
cyte-colony stimulating factor in the peripheral blood and
the maternal–fetal interface is associated with successful
pregnancy. Trophoblast, decidua, and amnion contribute to
the Th2 cytokine-biased environment by production of IL-13,
IL-10, IL-4, and IL-6.101–103 Human placental cytotrophoblasts
have been shown to produce the immunosuppressive cytokine
IL-10.101 In addition, macrophages and Tregs present within
decidua during pregnancy also produce IL-10 and are involved
in maintenance of immune tolerance toward allogeneic fetal
antigens.91 The placenta also produces PGD2, which can
act as a chemoattractant for Th2 cells to the maternal–fetal
interface via the Th2 receptor CRTH2 (a chemoattractant
receptor-homologous molecule expressed on Th2 cells).
Women suffering recurrent pregnancy loss have reduced
expression of CRTH2+ cells than women undergoing elective
termination of pregnancy.104 Anti-inflammatory cytokines
IL-4 and IL-10 inhibit Th1 cells and macrophages, which
in turn prevent fetal allograft rejection. In addition, these
cytokines also inhibit TNF-α, cyclooxygenase-2 (COX-2),
and prostaglandin E2 in amnion-derived cells, which prevent
the onset of labor.24,105–107
Labor is often associated with a proinflammatory state
with reversal back to Th1 rather than Th2. Studies indicate
increases in Th1 proinflammatory cytokines and reduction in
Th2 cytokines in women who are in active labor. Fetal mem-
branes, myometrium, amnion, amniotic fluid, and decidua
produce proinflammatory cytokines IL-1β and TNF-α at
term and can induce nuclear factor kappa B. This transcrip-
tion factor regulates the expression of labor-associated genes
such as COX-2, IL-8, and MMP-9 and triggers a cascade of
labor-inducing events. Despite the proinflammatory nature
of Th1 cytokines, they are essential for successful pregnancy,
contributing to timely labor.108–110
Role of Tregs in pregnancy
CD4+CD25+ Tregs are a subpopulation of T-cells respon-
sible for the maintenance of immunological self-tolerance
by suppressing self-reactive lymphocytes in a cell contact-
dependent manner by production of TGF-β and IL-10.111,112
Tregs express transcription factor forkhead box transcrip-
tion factor (FoxP3), which acts as a major regulator in
their development and function.113 There are two main Treg
subsets: naturally occurring or thymic Tregs (tTregs) and
induced or extrathymic/peripheral Tregs (pTregs). tTregs are
CD4+CD25+Foxp3+ and express cytotoxic T lymphocyte-
associated antigen 4. pTregs develop from naïve T-cells after
exposure to antigens in the periphery and exposure to either
IL-10 or TGF-β and can be either Foxp3- or Foxp3+.114,115
Owing to their immunosuppressive function, Tregs also play
a key role during pregnancy by maintaining maternal–fetal
tolerance.
Several studies have confirmed an increase in Tregs dur-
ing pregnancy in blood, lymph nodes, and thymus, followed
by decrease from midgestation onward until they reach
nonpregnant levels at term or shortly thereafter. They play a
critical role in embryo implantation and in the maintenance
Research and Reports in Biology 2015:6 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
179
Maternal immune system and fetal development
of the maternal immune tolerance against semiallogeneic
fetal antigens.116,117 Evidence suggests that Tregs during
pregnancy are specific to paternal alloantigens, which
protects the fetus from rejection by the mother’s immune
system.118 Expansion of Tregs in decidua from normal preg-
nant women suppresses maternal Th1/Th17 activity on the
semiallogeneic fetus.119
Murine experiments have shown increased levels of
Tregs in both syngeneic and allogeneic matings, suggesting
alloantigen-independent Treg expansion.120 Treg expansion
appears to be regulated by estradiol. This is supported by
in vitro studies, which show that physiological levels of
estradiol not only expand Tregs but also stimulate conver-
sion of CD4+CD25- T-cells into CD4+CD25+ T-cells.121
On the other hand, Zhao et al122 observed no increase in
Tregs in ovariectomized mice. Moreover, they detected
higher number of Tregs in pregnant mice from allogeneic
versus syngeneic matings, suggesting an involvement of
paternal antigens in Treg expansion.122 Recently, Robertson
et al123 showed that seminal fluid can drive Treg expan-
sion. Therefore, both antigen-dependent and antigen-
independent mechanisms are likely to be involved in Treg
expansion.
Tregs express various chemokine receptors whose
ligands are expressed at the maternal–fetal interface, which
might contribute to chemokine-mediated migration of
Tregs to the decidua.120 Furthermore, other immune cells
produce large amounts of CCL17, CCL4, and CCL1,124–126
which might attract Tregs specifically expressing CCR4
and CCR8.127,128 Besides chemokine-mediated migration of
Tregs, integrins, similar to CD62L, seem to play an impor-
tant role in Treg migration, as neutralizing CD62L-specific
antibody blocks expansion of Tregs in draining lymph nodes
and results in allograft rejection. Schumacher et al129 have
shown the importance of human chorionic gonadotropin as
one of the main attractants of Tregs to the maternal–fetal
interface.
Aluvihare et al117 first noted that Tregs increased in all
lymphoid organs in allogeneic matings of C57BL/6 female
mice with CBA males. They also adoptively transferred
lymphocytes from BALB/c females, either allopregnant
from C57BL/6 males or synpregnant from BALB/c males,
into T-cell-deficient BALB/c females, which were then
mated with C57BL/6 males. Pregnancy proceeded normally
when whole lymphocyte populations were transferred. In
contrast, lymphocytes depleted of Tregs resulted in fetal
resorptions, and there was a massive infiltration of T-cells
into the implantation sites.117 Zenclussen130 and Zenclussen
et al131 have shown complete prevention of abortion in the
CBA × DBA/2J model of naturally occurring spontaneous
abortions by transferring Tregs from alloimmunized mice,
and they also reported that no abortions occurred in the CBA
× BALB/c and CBA × CBA control matings. Finally, Chen
et al116 demonstrated that stimulation of Tregs, either directly
by low dose of IL-2 or indirectly by Fms-related tyrosine
kinase 3 ligand, led to normal pregnancy rates in CBA ×
DBA/2J abortion-prone mice. The results of these experi-
ments all demonstrate that in allogenic matings, Tregs are nec-
essary for prevention of a maternal immune response against
the fetus.
Clinical manifestations of an altered
immune system in pregnancy
The notion of pregnancy as an altered state of immune
suppression is well documented.132–136 Pregnancy is a time
period that poses a risk of increased susceptibility to infec-
tious diseases, and the maternal immune system is solely
responsible for defending against infectious microorgan-
isms and protecting the fetus because both the fetal and the
placental responses are limited.132,136 The Th1/Th2 immune
shifts in pregnancy are well established and have provided
a platform to further study the immune system.136 This
has led to refining our understanding about the immune
system and the development of a new paradigm regard-
ing pregnancy and immune function. This newer theory
proposes that the immune system during pregnancy is a
functional and active system, wherein not only a maternal
immune response exists but also a fetal–placental immune
response, which in combination is powerful in defending
both the mother and the fetus.133,136 With this notion, the
immune system is not suppressed, but rather in a modulated
state, and therefore, this explains why pregnant women have
differential responses to various pathogens.133 During this
altered response, signals are generated in the placenta, which
modulate the maternal immune system to behave uniquely
to different microorganisms.133 Although these old and new
paradigms surrounding the immunology of pregnancy dif-
fer, it is clear that the immune system’s goal in pregnancy
is to ensure that a pregnancy progresses successfully, while
still providing protection for both mother and fetus from
external pathogens.
Endocrine regulation of immune cells
Hormone concentrations vary with the initiation
of pregnancy, and there are specific fluctuations in
hormone levels throughout each trimester of preg-
nancy. In general, pregnancy hormones are thought
to suppress maternal alloresponses, while promoting
Research and Reports in Biology 2015:6
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
180
Morelli et al
Table 1 Endocrine regulation of immune cells and immune function
Hormone Th1 (proinammatory)
pathway
Th2 (anti-inammatory)
pathway
Effects on immune cells References
Estradiol (E2) Inhibits pathway via TNF-α,
IL-1β, and IL-6
Stimulates pathway via IL-4,
IL-10, TGF-β, and IFN-γ
Modulates lymphocyte
development and function
134,148
E2 Treg proliferation
E2 enhances Treg’s
suppressive function
Progesterone (P4) Inhibits pathway via TNF-α
and IL-6
Stimulates pathway via IL-4,
IL-10 (from T-cells)
P4 Treg proliferation 134,148–151
P4 enhances Treg’s
suppressive function
P4 favors development
of Th2 CD4 T-cells (may
be responsible for Th2
predominance during
pregnancy)
P4 suppresses T-cell activation
Wide distribution of P4
receptors in immune cells
(Dendritic, T, and B-cells)
uNK cells do not express
steroid receptors, P4 actions
likely mediated through
glucocorticoid receptors
Human chorionic
gonadotropin (hCG)
Inhibits pathway via TNF-αStimulates pathway via
TGFβ, IL-8, and IL-10
(from B-cells)
hCG attracts Tregs 134,148,152
hCG induces uNK cell
proliferation through mannose
receptor (uNK cells do not
express LH/CG receptor)
hCG promotes dendritic cell
and monocyte proliferation
and function
Abbreviations: CD, cluster of differentiation; IL, interleukin; IFN, interferon; LH/CG, luteinizing hormone/chorionic gonadotropin; TGF, transforming growth factor; Th, T
helper cell; TNF, tumor necrosis factor; Treg, T-regulatory cell; uNK, uterine natural killer; , decreased; , increased.
pathways of tolerance.137 Hormonal shifts are thought
to reduce the number of DCs and monocytes, decrease
macrophage activity, while blocking NK cells, T-cells, and
B-cells.137 Each of the major pregnancy-associated hor-
mones is thought to directly and indirectly affect the func-
tion of the major immune cells and thus impacts the immune
milieu during pregnancy. These alterations are discussed
in Table 1.
Evidence of altered immune function
in pregnancy: effects of infectious
organisms on pregnancy
The alterations in the immune system during pregnancy are
well established, and subsequently, these changes result in
increased susceptibility to certain viral, bacterial, and para-
sitic infections.132 This increased susceptibility is believed
to result from the suppression of cell-mediated immunity,
as pregnancy promotes a shift away from the Th1 to the
Th2 immune environment.132,134 Additionally, infection with
certain pathogens has been documented to result in severe
symptoms in pregnant patients because of these immune
changes.133,138 However, it is important to note that, in certain
infectious diseases among gravid patients, the morbidity
and mortality vary between developed and nondeveloped
countries. For example, pregnant women with varicella
in the US or Canada fare better than those diagnosed in
underdeveloped countries, where resources are limited.139
Thus, some bias may result when evaluating the severity of
disease states in pregnant women depending on geographi-
cal distribution.
Table 2 summarizes the more commonly recognized and
studied pathogens related to pregnancy. As seen in Table 2,
infectious diseases during pregnancy are associated with
not only maternal risks but fetal risks as well. These fetal
effects result from infections that cross the placenta, which
can cause miscarriage, congenital anomalies, or even fetal
death.133 As a result, the American Congress of Obstetricians
and Gynecologists and the US Centers for Disease Control
and Prevention recommend that all women be vaccinated
for influenza and tetanus, diphtheria, and pertussis (Tdap)
during pregnancy.140–142 Both these vaccines appear to be
safe when administered during pregnancy, with few maternal
and fetal adverse events.142,143 In contrast, live vaccines, such
as measles–mumps–rubella (MMR) and varicella, are not
Research and Reports in Biology 2015:6 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
181
Maternal immune system and fetal development
Table 2 Common infectious organisms in pregnancy
Infectious
organisms
Type Risks Transmission Immune cells Other Fetal effects References
Cytomegalovirus
(CMV)
Viral Incidence in US: 1%–3% of
pregnant women
Transmission: 30% in
the rst trimester; up
to 72% in the third
trimester
uNK cells at level of
placenta
Most common cause of
intrauterine viral infection
Leading cause of congenital
infection (IUGR, seizures,
microcephaly, petechial rash,
hepatosplenomegaly), hearing
and visual impairments, mental
retardation, fetal death
36,153
Low risk for healthy pregnant
women
Infection progresses
from decidua
placenta
uNK and T-cells control
CMV infection and spread
during pregnancy
Inuenza virus Viral Risk of infection increases
with GA
Infection can spread
from maternal
bloodstream to
decidua (preferred
environment for viral
infections), where the
virus replicates, and
then it infects fetal
chorion and amnion
Innate response: involves
PRRs that recognize viral
components
Virus can induce apoptosis
in chorion cells (direct
cytopathic effect)
Maternal inuenza infection,
during any trimester, confers
fourfold increased risk of
bipolar disorder in offspring
145,154–156
Pregnant women seven
times more likely to be
hospitalized, twice as likely
to die from inuenza as
nonpregnant women
Pregnancy decreases
adiponectin levels (an
adipokine) a more pronounced
innate immune response
when infected with H1N1
Cell types: neutrophils,
macrophages and DCs
IAV increases gene
expression of cytokines
IL-1b, IL-6, TNF-α; may
lead to premature rupture
of membranes
Rhesus monkey model:
maternal inuenza affects fetal
neural development with
reduction in gray matter and
decreased white matter in
parietal cortex
In humans, IL-8 and serologic
evidence of maternal inuenza
infection associated with
schizophrenia in offspring
Pregnancy may enhance
systemic inammatory
response to inuenza
( TNF-α, G-CSF;
IFN-γ, MCP-1
Varicella zoster
virus (VZV)
Viral Primary varicella infection
in pregnant patients leads to
more severe disease than
reactivation
Infection may be
primary, or reactivation
in utero
Prodromal symptoms:
headache, fever, malaise
Fetal effects worse if infection
occurs early in pregnancy
132,139,157,158
Incidence: 0.7–3.0/1,000
pregnancies
Vertical transmission
occurs transplacentally
2.5% of varicella
infections in pregnancy
complicated by varicella
pneumonia
Congenital varicella syndrome:
cerebral abnormalities, cataracts,
microphthalmia, chorioretinitis,
mental retardation, skin lesions,
limb hypoplasia, IUGR, and low
birth weight
Varicella pneumonia more
common in pregnant women
than in nonpregnant women,
and presence of pneumonia
confers increased risk of
maternal mortality
Greater risk of fetal
transmission when
infection occurs in
third trimester
Other fetal risks: miscarriage,
stillbirth
(Continued)
Research and Reports in Biology 2015:6
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
182
Morelli et al
Table 2 (Continued)
Infectious
organisms
Type Risks Transmission Immune cells Other Fetal effects References
Listeriosis (Listeria
monocytogenes)
Bacterial Infections more common
during pregnancy (25%–33%
of all cases occur in pregnant
women)
Transmission: mostly
foodborne
Innate immune response:
macrophages produce IL-1,
IL-6, and TNF-α; this sets
stage for adaptive immune
response to Listeria; CD8+
T-cells peak 7–10 days after
infection
Symptoms: may be
asymptomatic or
nonspecic inuenza-like,
fever, back pain, and rarely
gastroenteritis
Fetal effects depend on the
timing of exposure to Listeria,
with most infections in the
third trimester
36,132,159–163
Pregnant women have
20-fold increased incidence
of infection
Listeria crosses mucosal
barrier in stomach,
hematogenous
spread to extravillous
trophoblasts and
chorionic villi to access
placenta (decidua
and placenta become
major reservoirs of the
organism)
In pregnancy, Th17
cells, causing IL-17a
and IL-22, potentiating the
inammatory response
Murine studies: Listeria blunts
maternal Treg suppression
which may lead to immune-
mediated fetal wastage
Infection can lead to miscarriage,
preterm labor, stillbirth, or
neonatal death (due to
granulomatosis infantiseptica,
causing microabscesses and
granulomas)
Guinea pig studies indicate that
fetus may be infected as early
as 2 days postinoculation
Tuberculosis (TB)
(Mycobacterium
tuberculosis)
Bacterial Difcult to diagnose, due to
nonspecic symptoms related
to physiologic response to
pregnancy (eg, fatigue,
shortness of breath, sweating,
cough, and mild fever)
Transmission via
respiratory tract
droplets
T-cells release IFN-γ in
response to specic
antigens presented by
M. tuberculosis
Screening high-risk
patients is imperative
for early diagnosis and
treatment
Without treatment, two
times increased risk of
preterm birth, IUGR, low
birth weight, prematurity,
and six times increased risk
of perinatal death
164–168
Without treatment, four
times increased risk of
maternal morbidity (higher
rates of abortion, postpartum
hemorrhage, labor difculties,
and preeclampsia)
Altered immunity
increases susceptibility
to infection and
reactivation of latent
TB, and risks are higher
in HIV-positive women
Higher risks in patients
coinfected with HIV
Congenital TB: neonates born
to mothers with active TB have
high mortality rate (20%–44%);
can be subclinical or associated
with birth defects
Vertical transmission
can occur via the
placenta and amniotic
uid (congenital TB)
or respiratory droplets
(neonatal TB)
Prevalence of active TB in
pregnant patients: 0.06%–
0.25% in low-prevalence
countries, and 0.07%–0.5%
in high-burden countries
Research and Reports in Biology 2015:6 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
183
Maternal immune system and fetal development
Malaria
(Plasmodium
falciparum)
Parasitic Pregnant women more
susceptible than nonpregnant
women, with higher
susceptibility during rst half
of pregnancy
Pathogenesis:
infected erythrocytes
accumulate in IVS
(higher density than
peripheral circulation)
and maternal
phagocytes, deposition
of hemozoin (malaria
pigment) seen in IVS
Placental parasites express
surface ligands and antigens
that differ from those of
other P. falciparum variants,
facilitating evasion of immune
response
Maternal risks: anemia Pregnancy-associated malaria
causes up to 200,000 infant
deaths/year
46,133,147,169,170
Fetal risks: low birth weight
and IUGR
125 million pregnant women
worldwide are at risk of
malaria each year
Chronic infection associated
with low birth weight and
preterm delivery
Highly susceptible during
rst and second pregnancies
(despite acquired immunity
after years of exposure)
Toxoplasmosis
(Toxoplasma
gondii)
Parasitic Seronegative pregnant
women .2× as likely as
nonpregnant women to
seroconvert
Primary infection: 20%
vertical transmission
rate to fetus, and
highest transmission in
third trimester (32%)
uNK cells play important
role in defending against
T. gondii at maternal–fetal
interface, however, parasite
can evade uNK cells and
continue to replicate,
especially during
early pregnancy
Second most common
cause of fetal intrauterine
infection
Primary infection can result in
miscarriage, stillbirth, preterm
delivery, or fetal malformations
132,170–172
Transmission occurs
via ingestion of raw
meat or exposure to
cat feces
Often asymptomatic
(in mother), but treatment
aimed at preventing
congenital infection
Congenital toxoplasmosis:
hydrocephalus, seizures,
IUGR, mental retardation,
microphthalmia, eye disease,
intracranial calcications
Abbreviations: CD, cluster of differentiation; DC, dendritic cell; GA, gestational age; G-CSF, granulocyte-colony stimulating factor; IAV, inuenza A virus; H1N1, inuenza A virus subtype H1N1; IFN, interferon; IL, interleukin; IUGR,
intrauterine growth restriction; IVS, intervillous space; MCP-1, monocyte chemotactic protein-1; PRR, pattern recognition receptor; Th, T helper cell; TNF, tumor necrosis factor; Treg, T-regulatory cell; uNK, uterine natural killer;
, decreased; , increased.
Research and Reports in Biology 2015:6
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
184
Morelli et al
recommended during pregnancy due to the theoretical risks
to the fetus.141,142
The risk of infection during pregnancy is a serious
matter, not only for concerns of maternal well-being but
also the potential fetal risks, which may have long-term
consequences. Animal studies have elucidated that the pla-
centa may trigger fetal inflammatory response syndrome
(FIRS), which is the diagnosis of a placental infection
without the growth of an organism, from the microbiology
standpoint.133,136 FIRS is serious and results in increased
circulating levels of cytokines, such as IL-1, IL-6, IL-8,
and TNF-α.133 These inflammatory shifts have been
demonstrated to increase the risk of fetal abnormalities, such
as ventriculomegaly or hemorrhages. Furthermore, human
studies have demonstrated an association between FIRS and
the development of autism, schizophrenia, neurosensorial
deficits, and psychosis.133,136 These observations further
validate the experimental mouse models described earlier in
which immunostimulation induces high levels of proinflam-
matory cytokines in blood and amniotic fluid of pregnant
dams, which are likely involved in the etiology of neurode-
velopmental disorders exhibited in their offspring.63,72–74 In
contrast, bacterial infections that reach the decidua trigger
a proinflammatory response that leads to the development
Table 3 Autoimmune disease in pregnancy
Autoimmune
disease
Improvement (remission of
symptoms)
Worsens (exacerbation of
symptoms)
Other References
Multiple sclerosis Mediated via suppression of
cell-mediated immunity
148,173
Reduced relapse rate mostly seen
in second and third trimesters
Flares common postpartum
Myasthenia gravis Symptoms may worsen, improve, or
remain unchanged
148
Symptoms vary among women and
between pregnancies in the same
woman
Psoriasis Improvement correlated with
higher levels of E2
148,174
E2 causes further shift from Th1
to Th2 type of immunity
Rheumatoid
arthritis
Mediated via suppression of
cell-mediated immunity
Flares may occur postpartum
Expansion of Tregs in pregnancy may
account for improvement of symptoms
during pregnancy
148,175
Decrease in Tregs postpartum may
account for postpartum disease ares
Systemic lupus
erythematosus
Increase in Th2-mediated
response worsens this humoral-
mediated autoimmune disease
Fetal effects: congenital heart block,
due to passive transplacental transfer
of anti-Ro (SS-A) and anti-La (SS-B)
Abs from mother to fetus
148,176
Causes irreversible damage to fetal
cardiac conduction system
Abs also cause neonatal lupus (skin
rashes, liver abnormalities, hematologic
cytopenias); effects are transient
(months) and improve once maternal
Abs are cleared from infant’s circulation
Autoimmune
hyperthyroidism
(Grave’s disease)
Autoimmune thyrotoxicosis may
improve because of a degree
of immunosuppression during
pregnancy
Fetal effects: TSH-R Abs can cross
placenta, causing fetal hyperthyroidism
that may lead to fetal tachycardia,
hydrops, fetal goiter, or IUGR
177
Flare may occur postpartum Untreated fetal hyperthyroidism has
15% mortality
Infant will continue to have maternal
TSH-R Abs for 3 months (transient
effects)
Abbreviations: Abs, antibodies; CD, cluster of differentiation; E2, estradiol; IUGR, intrauterine growth restriction; Th, T helper cell; Treg, T-regulatory cell; TSH-R, thyroid
stimulating hormone-receptor.
Research and Reports in Biology 2015:6 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
185
Maternal immune system and fetal development
of intrauterine infections.144 This is through the activation of
pattern recognition receptors (PRRs) and increased secretion
of cytokines, such as IL-1 and TNF-α.145 Combined, these
contribute to poor pregnancy outcomes, disruption in fetal
development, or preterm births with resultant low-birth-
weight infants.146,147 Thus, it is important to recognize that
pregnancy can cause increased disease susceptibility, which
not only affects maternal morbidity but contributes to detri-
mental long-term fetal and neonatal outcomes.
Evidence of altered immune function in
pregnancy: effects of pregnancy on
autoimmune disease
As discussed, pregnancy confers a shift from Th1- to Th2-
mediated immunity, and this shift affects disease status in
women with known autoimmune diseases. In general, the
hormonal milieu induced by pregnancy shifts the cytokine
profile away from cell-mediated immunity (Th1 type of
immunity) and, therefore, improves inflammatory-type
autoimmune diseases.132 In contrast, autoimmune diseases
that are humorally (or antibody) mediated are exacerbated,
as pregnancy favors increased Th2-related activities, as
well as a Th2 cytokine profile.132,148 For details, please view
Table 3.
Conclusion and future outlook
Pregnancy in women is a dynamic state, with different
mechanisms used during different trimesters to enable and
ensure successful establishment, maintenance, and timely
termination of the pregnancy. Mechanisms operative in early
pregnancy to establish the pregnancy may differ from those
needed to maintain the pregnancy and from those required to
ensure successful and timely labor and delivery. Recent data
challenge the notion that pregnancy is simply an immunosup-
pressed state protecting the allogeneic fetus from attack by the
maternal immune system. The evidence suggests that rather,
pregnancy may be a state of upregulated innate immune
response and decreased cell-mediated response. Unique
decidual lymphoid cell populations actively contribute to
placental development and to tolerance of the fetus. Although
substantial progress in the understanding of the function of
immune cells during pregnancy, especially early pregnancy,
has been achieved, many unanswered questions regarding
regulation of their proliferation and function by endocrine
and other factors still remain. The published results from
human studies and animal models clearly indicate that a fine
balance between proinflammatory and anti-inflammatory
influences is critical for successful pregnancy. Thus, the
future challenge for translational research in reproductive
immunology will be to define more completely those factors
that favor optimal immunological environments that promote
fetal health and development at specific stages of pregnancy,
so that evidence-based regulatory therapeutic strategies can
then be designed.
Acknowledgments
The authors thank Yingting Zhang for her assistance with the
manuscript. Mili Mandal is currently affiliated with Oncol-
ogy, R&D, GlaxoSmithKline, Collegeville, PA, US.
Disclosure
The authors report no conflicts of interest in this work.
References
1. Billingham RE, Brent L, Medawar PB. Actively acquired tolerance of
foreign cells. Nature. 1953;172(4379):603–606.
2. Brosens JJ, Pijnenborg R, Brosens IA. The myometrial junctional zone
spiral arteries in normal and abnormal pregnancies: a review of the
literature. Am J Obstet Gynecol. 2002;187(5):1416–1423.
3. Gellersen B, Brosens IA, Brosens JJ. Decidualization of the human
endometrium: mechanisms, functions, and clinical perspectives. Semin
Reprod Med. 2007;25(6):445–453.
4. Gellersen B, Brosens J. Cyclic AMP and progesterone receptor cross-
talk in human endometrium: a decidualizing affair. J Endocrinol.
2003;178(3):357–372.
5. Dimitriadis E, White CA, Jones RL, Salamonsen LA. Cytokines,
chemokines and growth factors in endometrium related to implantation.
Hum Reprod Update. 2005;11(6):613–630.
6. Lash GE, Robson SC, Bulmer JN. Review: functional role of uterine
natural killer (uNK) cells in human early pregnancy decidua. Placenta.
2010;31(Suppl):S87–S92.
7. King A. Uterine leukocytes and decidualization. Hum Reprod Update.
2000;6(1):28–36.
8. Bulmer JN, Morrison L, Longfellow M, Ritson A, Pace D.
Granulated lymphocytes in human endometrium: histochemi-
cal and immunohistochemical studies. Hum Reprod. 1991;6(6):
791–798.
9. Weiss G, Goldsmith LT, Taylor RN, Bellet D, Taylor HS. Inflammation
in reproductive disorders. Reprod Sci. 2009;16(2):216–229.
10. Moffett A, Colucci F. Uterine NK cells: active regulators at the maternal-
fetal interface. J Clin Invest. 2014;124(5):1872–1879.
11. Moffett A, Loke C. Immunology of placentation in eutherian mammals.
Nat Rev Immunol. 2006;6(8):584–594.
12. Moffett-King A. Natural killer cells and pregnancy. Nat Rev Immunol.
2002;2(9):656–663.
13. King A, Birkby C, Loke YW. Early human decidual cells exhibit
NK activity against the K562 cell line but not against first trimester
trophoblast. Cell Immunol. 1989;118(2):337–344.
14. Koopman LA, Kopcow HD, Rybalov B, et al. Human decidual natural
killer cells are a unique NK cell subset with immunomodulatory
potential. J Exp Med. 2003;198(8):1201–1212.
15. Naruse K, Lash GE, Innes BA, et al. Localization of matrix metallo-
proteinase (MMP)-2, MMP-9 and tissue inhibitors for MMPs (TIMPs)
in uterine natural killer cells in early human pregnancy. Hum Reprod.
2009;24(3):553–561.
16. Lash GE, Schiessl B, Kirkley M, et al. Expression of angiogenic growth
factors by uterine natural killer cells during early pregnancy. J Leukoc
Biol. 2006;80(3):572–580.
17. Yagel S. The developmental role of natural killer cells at the fetal-
maternal interface. Am J Obstet Gynecol. 2009;201(4):344–350.
Research and Reports in Biology 2015:6
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
186
Morelli et al
18. Faas MM, Spaans F, De Vos P. Monocytes and macrophages in
pregnancy and pre-eclampsia. Front Immunol. 2014;5:298.
19. Smith SD, Dunk CE, Aplin JD, Harris LK, Jones RL. Evidence for
immune cell involvement in decidual spiral arteriole remodeling in
early human pregnancy. Am J Pathol. 2009;174(5):1959–1971.
20. Abrahams VM, Kim YM, Straszewski SL, Romero R, Mor G.
Macrophages and apoptotic cell clearance during pregnancy. Am J
Reprod Immunol. 2004;51(4):275–282.
21. Co EC, Gormley M, Kapidzic M, et al. Maternal decidual macrophages
inhibit NK cell killing of invasive cytotrophoblasts during human
pregnancy. Biol Reprod. 2013;88(6):155.
22. Wallace AE, Fraser R, Cartwright JE. Extravillous trophoblast and
decidual natural killer cells: a remodelling partnership. Hum Reprod
Update. 2012;18(4):458–471.
23. Nancy P, Erlebacher A. T cell behavior at the maternal-fetal interface.
Int J Dev Biol. 2014;58(2–4):189–198.
24. Piccinni MP. T cell tolerance towards the fetal allograft. J Reprod
Immunol. 2010;85(1):71–75.
25. Bulmer JN, Williams PJ, Lash GE. Immune cells in the placental bed.
Int J Dev Biol. 2010;54(2–3):281–294.
26. Scaife PJ, Bulmer JN, Robson SC, Innes BA, Searle RF. Effector activ-
ity of decidual CD8+ T lymphocytes in early human pregnancy. Biol
Reprod. 2006;75(4):562–567.
27. Hsu P, Nanan RK. Innate and adaptive immune interactions at the
fetal-maternal interface in healthy human pregnancy and pre-eclampsia.
Front Immunol. 2014;5:125.
28. Gardner L, Moffett A. Dendritic cells in the human decidua. Biol
Reprod. 2003;69(4):1438–1446.
29. Miyazaki S, Tsuda H, Sakai M, et al. Predominance of Th2-promoting
dendritic cells in early human pregnancy decidua. J Leukoc Biol.
2003;74(4):514–522.
30. Laskarin G, Redzović A, Rubesa Z, et al. Decidual natural killer cell
tuning by autologous dendritic cells. Am J Reprod Immunol. 2 008;59(5):
433–445.
31. Plaks V, Birnberg T, Berkutzki T, et al. Uterine DCs are crucial for
decidua formation during embryo implantation in mice. J Clin Invest.
2008;118(12):3954–3965.
32. Krey G, Frank P, Shaikly V, et al. In vivo dendritic cell depletion reduces
breeding efficiency, affecting implantation and early placental develop-
ment in mice. J Mol Med (Berl). 2008;86(9):999–1011.
33. Hanna J, Goldman-Wohl D, Hamani Y, et al. Decidual NK cells regulate
key developmental processes at the human fetal-maternal interface.
Nat Med. 2006;12(9):1065–1074.
34. Norwitz ER. Defective implantation and placentation: laying the
blueprint for pregnancy complications. Reprod Biomed Online. 2007;14
Spec No 1:101–109.
35. Lash GE, Otun HA, Innes BA, et al. Interferon-gamma inhibits
extravillous trophoblast cell invasion by a mechanism that involves
both changes in apoptosis and protease levels. FASEB J. 2006;20(14):
2512–2518.
36. Erlebacher A. Immunology of the maternal-fetal interface. Annu Rev
Immunol. 2013;31:387–411.
37. Guimond MJ, Luross JA, Wang B, Terhorst C, Danial S, Croy BA.
Absence of natural killer cells during murine pregnancy is associated
with reproductive compromise in TgE26 mice. Biol Reprod. 1997;56(1):
169–179.
38. Guimond MJ, Wang B, Croy BA. Engraftment of bone marrow from
severe combined immunodeficient (SCID) mice reverses the reproduc-
tive deficits in natural killer cell-deficient tg epsilon 26 mice. J Exp
Med. 1998;187(2):217–223.
39. Ashkar AA, Di Santo JP, Croy BA. Interferon gamma contributes to
initiation of uterine vascular modification, decidual integrity, and uterine
natural killer cell maturation during normal murine pregnancy. J Exp
Med. 2000;192(2):259–270.
40. Monk JM, Leonard S, McBey BA, Croy BA. Induction of murine spiral
artery modification by recombinant human interferon-gamma. Placenta.
2005;26(10):835–838.
41. Manaster I, Gazit R, Goldman-Wohl D, et al. Notch activation enhances
IFNgamma secretion by human peripheral blood and decidual NK cells.
J Reprod Immunol. 2010;84(1):1–7.
42. Vigano P, Gaffuri B, Somigliana E, Infantino M, Vignali M, Di Blasio AM.
Interleukin-10 is produced by human uterine natural killer cells but
does not affect their production of interferon-gamma. Mol Hum Reprod.
2001;7(10):971–977.
43. Dambaeva SV, Durning M, Rozner AE, Golos TG. Immunophenotype
and cytokine profiles of rhesus monkey CD56bright and CD56dim
decidual natural killer cells. Biol Reprod. 2012;86(1):1–10.
44. Croonenberghs J, Bosmans E, Deboutte D, Kenis G, Maes M. Activation
of the inflammatory response system in autism. Neuropsychobiology.
2002;45:1–6.
45. Deykin E, MacMahon B. Viral exposure and autism. Am J Epidemiol.
1979;109:628–638.
46. Hagberg H, Mallard C. Effect of inflammation on central nervous system
development and vulnerability: review. Curr Opin Neurol. 2005;18:
117–123.
47. Hornig M, Weissenbock H, Horscroft N, Lipkin WI. An infection-based
model of neurodevelopmental damage. Proc Natl Acad Sci. 1999;96:
12101–12107.
48. Lipkin W, Hornig M. Microbiology and immunology of autism
spectrum disorders. Novartis Found Symp. 2003;251:129–143.
49. Malek-Ahmadi P. Cytokines and etiopathogenesis of pervasive
developmental disorders. Med Hypotheses. 2001;56(3):321–324.
50. Pardo C, Eberhart C. The neurobiology of autism. Brain Pathol.
2007;17:434–447.
51. Patterson P. Maternal infection: window on neuroimmune interactions
in fetal brain development and mental illness. Curr Opin Neurobiol.
2002;12:115–118.
52. Brown AS. Prenatal infection as a risk factor for schizophrenia.
Schizophr Bull. 2006;32(2):200–202.
53. Brown AS, Derkits EJ. Prenatal infection and schizophrenia:
a review of epidemiologic and translational studies. Am J Psychiatry.
2010;167(3):261–280.
54. Jonakait G. The effects of maternal inflammation on neuronal develop-
ment: possible mechanisms. Int J Dev Neurosci. 2007;25:415–425.
55. Patterson PH. Maternal infection and immune involvement in autism.
Trends Mol Med. 2011;17:389–394.
56. Bell M, Hallenbeck J. Effects of intrauterine inflammation on developing
rat brain. J Neurosci Res. 2002;70:570–579.
57. Carvey P, Chang Q, Lipton J, Ling Z. Prenatal exposure to the bac-
teriotoxin lipopolysaccharide leads to long-term losses of dopamine
neurons in offspring: a potential, new model of Parkinson’s disease.
Front Biosci. 2003;8:s826–s837.
58. Fatemi SH, Earle J, Kanodia R, et al. Prenatal viral infection leads to pyra-
midal cell atrophy and macrocephaly in adulthood: implications for genesis
of autism and schizophrenia. Clin Mol Neurobiol. 2002;22: 25–33.
59. Hornig M, Solbrig M, Horscroft N, Weissenbock H, Lipkin W. Borna
disease virus infection of adult and neonatal rats: models for neurop-
sychiatric disease. Curr Top Microbiol Immunol. 2001;253:157–177.
60. Pletnikov MV, Jones ML, Rubin SA, Moran TH, Carbone KM. Rat
model of autism spectrum disorders. Genetic background effects on
Borna disease virus-induced development brain damage. Ann NY Acad
Sci. 2001;939:318–319.
61. Pletnikov M, Rubin S, Schwartz G, Moran T, Carbone K. Persistent neona-
tal Borna disease virus (BDV) infection of teh brain causes chronic emo-
tional abnormalities in adult rats. Physiol Behav. 1999;66: 823–831.
62. Shi L, Fatemi SH, Sidwell RW, Patterson PH. Maternal influenza infec-
tion causes marked behavioral and pharmacological changes in the
offspring. J Neurosci. 2003;23(1):297–302.
63. Smith SE, Li J, Garbett K, Mirnics K, Patterson PH. Maternal immune
activation alters fetal brain development through interleukin-6.
J Neurosci. 2007;27(40):10695–10702.
64. Weissenbock H, Hornig M, Hickey W, Lipkin W. Microglial activation
and neuronal apoptosis in Bornavirus infected neonatal Lewis rats.
Brain Pathol. 2000;10:260–272.
Research and Reports in Biology 2015:6 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
187
Maternal immune system and fetal development
65. Lancaster K, Dietz D, Moran T, Pletnikov M. Abnormal social behaviors
in young and adult rats neonatally infected with Borna disease virus.
Behav Brain Res. 2007;176:141–148.
66. Rousset CI, Chalon S, Cantagrel S, et al. Maternal exposure to LPS
induces hypomyelination in the internal capsule and programmed
cell death in the deep gray matter in newborn rats. Pediatr Res.
2006;59:428–433.
67. Dammann O, Leviton A. Maternal intrauterine infection, cytokines, and
brain damage in the preterm newborn. Pediatr Res. 1997;42:1–8.
68. Conroy SM, Nguyen V, Quina LA, et al. Interleukin-6 produces
neuronal loss in developing cerebellar granule neuron cultures.
J Neuroimmunol. 2004;155:43–54.
69. Gilmore J, Fredrik J, Vadlamudi S, Lauder J. Prenatal infection and
risk for schizophrenia: IL-1beta, IL-6, and TNFalpha inhibit cortical
neuron dendrite development. Neuropsychopharmacology. 2004;29:
1221–1229.
70. Nawa H, Takei N. Recent progress in animal modeling of immune
inflammatory processes in schizophrenia: implication of specific
cytokines. Neurosci Res. 2006;56:2–13.
71. Samuelsson A, Jennische E, Hansson H, Holmang A. Prenatal exposure to
Interleukin-6 results in inflammatory neurodegeneration in hippocampus
with NMDA/GABA(A) dysregulation and impaired spatial learning. Amer
J Physiol Regul Integr Comp Phyusiol. 2006;290: R1345–R1356.
72. Ponzio NM, Servatius R, Beck K, Marzouk A, Kreider T. Cytokine levels
during pregnancy influence immunological profiles and neurobehavioral
patterns of the offspring. Ann N Y Acad Sci. 2007;1107:118–128.
73. Mandal M, Donnelly R, Elkabes S, et al. Maternal immune stimulation
during pregnancy shapes the immunological phenotype of offspring.
Brain Behav Immun. 2013;33:33–45.
74. Mandal M, Marzouk AC, Donnelly R, Ponzio NM. Maternal immune
stimulation during pregnancy affects adaptive immunity in off-
spring to promote development of TH17 cells. Brain Behav Immun.
2011;25(5):863–871.
75. Barker DJ. Intrauterine programming of adult disease. Mol Med Today.
1995;1(9):418–423.
76. Bilbo SD, Schwarz JM. Early-life programming of later-life brain and
behavior: critical role for the immune system. Front Behav Neurosci.
2009;3:1–14.
77. Barrett EG. Maternal influence in the transmission of asthma
susceptibility. Pulm Pharmacol Ther. 2008;21(3):474–484.
78. Bellinger DL, Lubahn C, Lorton D. Maternal and early life stress effects
on immune function: relevance to immunotoxicology. J Immunotoxicol.
2008;5(4):419–444.
79. Conrad ML, Ferstl R, Teich R, et al. Maternal TLR signaling is
required for prenatal asthma protection by the nonpathogenic microbe
Acinetobacter lwoffii F78. J Exp Med. 2009;206(13):2869–2877.
80. Mandal M, Marzouk AC, Donnelly R, Ponzio NM. Preferential
development of Th17 cells in offspring of immunostimulated pregnant
mice. J Reprod Immunol. 2010;87(1–2):97–100.
81. Keshavan MS. Development, disease and degeneration in schizophrenia:
a unitary pathophysiological model. J Psychiatr Res. 1999;33(6):
513–521.
82. Keshavan MS, Hogarty GE. Brain maturational processes and delayed
onset in schizophrenia. Dev Psychopathol. 1999;11(3):525–543.
83. Ponzio NM, Mandal M, Elkabes S, et al. Pro-inflammatory phenotype
induced by maternal immune stimulation during pregnancy. In: Fitzger-
ald M, editor. Recent Advances in Autism Spectrum Disorders. Vol 1.
Rijeka: In Tech; 2013:113–141.
84. Ramanathan M, Ponzio N, Limson F, Shah S, Fernandes H. Maternal
cytokine regulation in the pathogenesis of autism. In: Program and
abstracts of the 9th Annual International Meeting for Autism Research;
May 20–22, 2010; Philadelphia, PA. Abstract 136.101.
85. Blaschitz A, Hutter H, Dohr GHLA. Class I protein expression in the
human placenta. Early Pregnancy. 2001;5(1):67–69.
86. Braud VM, Allan DS, McMichael AJ. Functions of nonclassical
MHC and non-MHC-encoded class I molecules. Curr Opin Immunol.
1999;11(1):100–108.
87. Hunt JS, Andrews GK, Wood GW. Normal trophoblasts resist induction
of class I HLA. J Immunol. 1987;138(8):2481–2487.
88. Hunt JS, Orr HT. HLA and maternal-fetal recognition. FASEB J.
1992;6(6):2344–2348.
89. Claas FH, Gijbels Y, van der Velden-de Munck J, van Rood JJ.
Induction of B cell unresponsiveness to noninherited maternal HLA
antigens during fetal life. Science. 1988;241(4874):1815–1817.
90. Tilburgs T, Scherjon SA, van der Mast BJ, et al. Fetal-maternal
HLA-C mismatch is associated with decidual T cell activation
and induction of functional T regulatory cells. J Reprod Immunol.
2009;82(2):148–157.
91. Chen SJ, Liu YL, Sytwu HK. Immunologic regulation in pregnancy:
from mechanism to therapeutic strategy for immunomodulation. Clin
Dev Immunol. 2012;2012:258391.
92. Noronha LE, Antczak DF. Maternal immune responses to trophoblast:
the contribution of the horse to pregnancy immunology. Am J Reprod
Immunol. 2010;64(4):231–244.
93. Baker JM, Bamford AI, Antczak DF. Modulation of allospecific CTL
responses during pregnancy in equids: an immunological barrier to
interspecies matings? J Immunol. 1999;162(8):4496–4501.
94. Flaminio MJ, Antczak DF. Inhibition of lymphocyte proliferation
and activation: a mechanism used by equine invasive trophoblast
to escape the maternal immune response. Placenta. 2005;26(2–3):
148–159.
95. Thellin O, Coumans B, Zorzi W, Igout A, Heinen E. Tolerance to the
foeto-placental ‘graft’: ten ways to support a child for nine months.
Curr Opin Immunol. 2000;12(6):731–737.
96. Sykes L, MacIntyre DA, Yap XJ, Teoh TG, Bennett PR. The Th1:th2
dichotomy of pregnancy and preterm labour. Mediators Inflamm.
2012;2012:967629.
97. Wegmann TG, Lin H, Guilbert L, Mosmann TR. Bidirectional cytokine
interactions in the maternal-fetal relationship: is successful pregnancy
a TH2 phenomenon? Immunol Today. 1993;14(7):353–356.
98. Chaouat G, Assal Meliani A, Martal J, et al. IL-10 prevents naturally
occurring fetal loss in the CBA × DBA/2 mating combination, and
local defect in IL-10 production in this abortion-prone combination
is corrected by in vivo injection of IFN-tau. J Immunol. 1995;154(9):
4261–4268.
99. Svensson L, Arvola M, Sallstrom MA, Holmdahl R, Mattsson R.
The Th2 cytokines IL-4 and IL-10 are not crucial for the comple-
tion of allogeneic pregnancy in mice. J Reprod Immunol. 2001;
51(1):3–7.
100. Fallon PG, Jolin HE, Smith P, et al. IL-4 induces characteristic Th2
responses even in the combined absence of IL-5, IL-9, and IL-13.
Immunity. 2002;17(1):7–17.
101. Roth I, Corry DB, Locksley RM, Abrams JS, Litton MJ, Fisher SJ.
Human placental cytotrophoblasts produce the immunosuppressive
cytokine interleukin 10. J Exp Med. 1996;184(2):539–548.
102. Jones CA, Finlay-Jones JJ, Hart PH. Type-1 and type-2 cytokines
in human late-gestation decidual tissue. Biol Reprod. 1997;57(2):
303–311.
103. Bennett WA, Lagoo-Deenadayalan S, Brackin MN, Hale E, Cowan BD.
Cytokine expression by models of human trophoblast as assessed by
a semiquantitative reverse transcription-polymerase chain reaction
technique. Am J Reprod Immunol. 1996;36(5):285–294.
104. Michimata T, Sakai M, Miyazaki S, et al. Decrease of T-helper 2 and
T-cytotoxic 2 cells at implantation sites occurs in unexplained recur-
rent spontaneous abortion with normal chromosomal content. Hum
Reprod. 2003;18(7):1523–1528.
105. Gilmour JS, Hansen WR, Miller HC, Keelan JA, Sato TA, Mitchell MD.
Effects of interleukin-4 on the expression and activity of prostaglandin
endoperoxide H synthase-2 in amnion-derived WISH cells. J Mol
Endocrinol. 1998;21(3):317–325.
106. Keelan JA, Sato TA, Mitchell MD. Comparative studies on the effects
of interleukin-4 and interleukin-13 on cytokine and prostaglandin E2
production by amnion-derived WISH cells. Am J Reprod Immunol.
1998;40(5):332–338.
Research and Reports in Biology 2015:6
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
188
Morelli et al
107. Goodwin VJ, Sato TA, Mitchell MD, Keelan JA. Anti-inflammatory
effects of interleukin-4, interleukin-10, and transforming growth
factor-beta on human placental cells in vitro. Am J Reprod Immunol.
1998;40(5):319–325.
108. Keelan JA, Marvin KW, Sato TA, Coleman M, McCowan LM, Mitchell
MD. Cytokine abundance in placental tissues: evidence of inflam-
matory activation in gestational membranes with term and preterm
parturition. Am J Obstet Gynecol. 1999;181(6):1530–1536.
109. Lindstrom TM, Bennett PR. The role of nuclear factor kappa B in
human labour. Reproduction. 2005;130(5):569–581.
110. Elliott CL, Loudon JA, Brown N, Slater DM, Bennett PR, Sullivan MH.
IL-1beta and IL-8 in human fetal membranes: changes with gestational
age, labor, and culture conditions. Am J Reprod Immunol. 2 001;46(4):
260–267.
111. Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic
self-tolerance maintained by activated T cells expressing IL-2 recep-
tor alpha-chains (CD25). Breakdown of a single mechanism of self-
tolerance causes various autoimmune diseases. J Immunol. 1995;
155(3):1151–1164.
112. Kingsley CI, Karim M, Bushell AR, Wood KJ. CD25+CD4+ regulatory
T cells prevent graft rejection: CTLA-4- and IL-10-dependent immu-
noregulation of alloresponses. J Immunol. 2002;168(3):1080–1086.
113. Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell devel-
opment by the transcription factor Foxp3. Science. 2003;299(5609):
1057–1061.
114. Sakaguchi S. Naturally arising CD4+ regulatory T cells for immuno-
logic self-tolerance and negative control of immune responses. Annu
Rev Immunol. 2004;22:531–562.
115. Apostolou I, Sarukhan A, Klein L, von Boehmer H. Origin of
regulatory T cells with known specificity for antigen. Nat Immunol.
2002;3(8):756–763.
116. Chen T, Darrasse-Jèze G, Bergot AS, et al. Self-specific memory
regulatory T cells protect embryos at implantation in mice. J Immunol.
2013;191(5):2273–2281.
117. Aluvihare VR, Kallikourdis M, Betz AG. Regulatory T cells mediate
maternal tolerance to the fetus. Nat Immunol. 2004;5(3):266–271.
118. Moldenhauer LM, Diener KR, Thring DM, Brown MP, Hayball JD,
Robertson SA. Cross-presentation of male seminal fluid antigens elicits
T cell activation to initiate the female immune response to pregnancy.
J Immunol. 2009;182(12):8080–8093.
119. Saito S, Nakashima A, Shima T, Ito M. Th1/Th2/Th17 and regulatory
T-cell paradigm in pregnancy. Am J Reprod Immunol. 2010;63(6):
601–610.
120. Leber A, Teles A, Zenclussen AC. Regulatory T cells and their role
in pregnancy. Am J Reprod Immunol. 2010;63(6):445–459.
121. Tai P, Wang J, Jin H, et al. Induction of regulatory T cells by physi-
ological level estrogen. J Cell Physiol. 2008;214(2):456–464.
122. Zhao JX, Zeng YY, Liu Y. Fetal alloantigen is responsible for the
expansion of the CD4(+)CD25(+) regulatory T cell pool during
pregnancy. J Reprod Immunol. 2007;75(2):71–81.
123. Robertson SA, Guerin LR, Bromfield JJ, Branson KM, Ahlstrom AC,
Care AS. Seminal fluid drives expansion of the CD4+CD25+ T
regulatory cell pool and induces tolerance to paternal alloantigens in
mice. Biol Reprod. 2009;80(5):1036–1045.
124. Bystry RS, Aluvihare V, Welch KA, Kallikourdis M, Betz AG. B
cells and professional APCs recruit regulatory T cells via CCL4.
Nat Immunol. 2001;2(12):1126–1132.
125. Schaniel C, Pardali E, Sallusto F, et al. Activated murine B lympho-
cytes and dendritic cells produce a novel CC chemokine which acts
selectively on activated T cells. J Exp Med. 1998;188(3):451–463.
126. Tang HL, Cyster JG. Chemokine Up-regulation and activated T cell
attraction by maturing dendritic cells. Science. 1999;284(5415):
819–822.
127. Iellem A, Mariani M, Lang R, et al. Unique chemotactic response
profile and specific expression of chemokine receptors CCR4 and
CCR8 by CD4(+)CD25(+) regulatory T cells. J Exp Med. 2001;194(6):
847–853.
128. Colantonio L, Iellem A, Sinigaglia F, D’Ambrosio D. Skin-homing
CLA+ T cells and regulatory CD25+ T cells represent major subsets
of human peripheral blood memory T cells migrating in response to
CCL1/I-309. Eur J Immunol. 2002;32(12):3506–3514.
129. Schumacher A, Brachwitz N, Sohr S, et al. Human chorionic
gonadotropin attracts regulatory T cells into the fetal-maternal
interface during early human pregnancy. J Immunol. 2009;182(9):
5488–5497.
130. Zenclussen AC. CD4(+)CD25+ T regulatory cells in murine pregnancy.
J Reprod Immunol. 2005;65(2):101–110.
131. Zenclussen AC, Gerlof K, Zenclussen ML, et al. Abnormal T-cell
reactivity against paternal antigens in spontaneous abortion: adop-
tive transfer of pregnancy-induced CD4+CD25+ T regulatory cells
prevents fetal rejection in a murine abortion model. Am J Pathol.
2005;166(3):811–822.
132. Jamieson DJ, Theiler RN, Rasmussen SA. Emerging infections and
pregnancy. Emerg Infect Dis. 2006;12(11):1638–1643.
133. Mor G, Cardenas I. The immune system in pregnancy: a unique
complexity. Am J Reprod Immunol. 2010;63(6):425–433.
134. Muzzio D, Zygmunt M, Jensen F. The role of pregnancy-associated
hormones in the development and function of regulatory B cells. Front
Endocrinol (Lausanne). 2014;5:39.
135. Polese B, Gridelet V, Araklioti E, Martens H, Perrier d’Hauterive S,
Geenen V. The endocrine milieu and CD4 T-lymphocyte polarization
during pregnancy. Front Endocrinol (Lausanne). 2014;5:106.
136. Racicot K, Kwon JY, Aldo P, Silasi M, Mor G. Understanding the
complexity of the immune system during pregnancy. Am J Reprod
Immunol. 2014;72(2):107–116.
137. Schumacher A, Costa SD, Zenclussen AC. Endocrine factors modulat-
ing immune responses in pregnancy. Front Immunol. 2014;5:196.
138. Schminkey DL, Groer M. Imitating a stress response: a new hypothesis
about the innate immune system’s role in pregnancy. Med Hypotheses.
2014;82(6):721–729.
139. Zhang HJ, Patenaude V, Abenhaim HA. Maternal outcomes in preg-
nancies affected by varicella zoster virus infections: population-based
study on 7.7 million pregnancy admissions. J Obstet Gynaecol Res.
2014;41(1):62–68.
140. American College of Obstetricians and Gynecologists. Update
on immunization and pregnancy: tetanus, diptheria, and pertussis
vaccination. ACOG Committee Opinion no 566. Obstet Gynecol.
2013;121(6):1411–1414.
141. American College of Obstetricians and Gynecologists (ACOG) Immu-
nization Expert Work Group. Immunizations and routine obstetric-
gynecologic care. Washington, DC: ACOG; 2013.
142. Centers for Disease Control and Prevention (CDC). Guidelines for
Vaccinating Pregnant Women. 2013. Advisory Committee on Immu-
nization Practices (ACIP) recommended immunization schedule for
persons aged 0 through 18 Years. Atlanta, GA: CDC; 2013.
143. Munoz FM, Weisman LE, Read JS, et al. Assessment of safety in
newborns of mothers participating in clinical trials of vaccines
administered during pregnancy. Clin Infect Dis. 2014;59(Suppl 7):
S415–S427.
144. Kraus TA, Engel SM, Sperling RS, et al. Characterizing the pregnancy
immune phenotype: results of the viral immunity and pregnancy (VIP)
study. J Clin Immunol. 2012;32(2):300–311.
145. Raj RS, Bonney EA, Phillippe M. Influenza, immune system, and
pregnancy. Reprod Sci. 2014;21(12):1434–1451.
146. Brabin BJ, Romagosa C, Abdelgalil S, et al. The sick placenta-the role
of malaria. Placenta. 2004;25(5):359–378.
147. Rogerson SJ, Hviid L, Duffy PE, Leke RFG, Taylor DW. Malaria in
pregnancy: pathogenesis and immunity. Lancet Infect Dis. 2007;7(2):
105–117.
148. Ngo ST, Steyn FJ, McCombe PA. Gender differences in autoimmune
disease. Front Neuroendocrinol. 2014;35(3):347–369.
149. Mjosberg J, Berg G, Jenmalm MC, Ernerudh J. FOXP3+ regulatory
T cells and T helper 1, T helper 2, and T helper 17 cells in human
early pregnancy decidua. Biol Reprod. 2010;82(4):698–705.
Research and Reports in Biology
Publish your work in this journal
Submit your manuscript here: http://www.dovepress.com/research-and-reports-in-biology-journal
Research and Reports in Biology is an international, peer-reviewed,
open access journal publishing original research, reports, editorials,
reviews and commentaries on all areas of biology including ani-
mal biology, biochemical biology, cell biology, ecological studies,
evolutionary biology, molecular biology, plant science and botany. The
manuscript management system is completely online and includes a
very quick and fair peer-review system. Visit http://www.dovepress.
com/testimonials.php to read real quotes from published authors.
Research and Reports in Biology 2015:6 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
Dovepress
189
Maternal immune system and fetal development
150. Lee JH, Ulrich B, Cho J, Park J, Kim CH. Progesterone promotes
differentiation of human cord blood fetal T cells into T regulatory
cells but suppresses their differentiation into Th17 cells. J Immunol.
2011;187(4):1778–1787.
151. Henderson TA, Saunders PT, Moffett-King A, Groome NP,
Critchley HO. Steroid receptor expression in uterine natural killer
cells. J Clin Endocrinol Metab. 2003;88(1):440–449.
152. Kosaka K, Fujiwara H, Tatsumi K, et al. Human chorionic gonado-
tropin (HCG) activates monocytes to produce interleukin-8 via a
different pathway from luteinizing hormone/HCG receptor system.
J Clin Endocrinol Metab. 2002;87(11):5199–5208.
153. Siewiera J, El Costa H, Tabiasco J, et al. Human cytomegalovirus
infection elicits new decidual natural killer cell effector functions.
PLoS Pathog. 2013;9(4):e1003257.
154. Gabriel G, Arck PC. Sex, immunity and influenza. J Infect Dis. 2014;
209(Suppl 3):S93–S99.
155. Parboosing R, Bao Y, Shen L, Schaefer CA, Brown AS. Gestational
influenza and bipolar disorder in adult offspring. JAMA Psychiatry.
2013;70(7):677–685.
156. Brown AS, Begg MD, Gravenstein S, et al. Serologic evidence of
prenatal influenza in the etiology of schizophrenia. JAMA Psychiatry.
2004;61(8):774–780.
157. Lamont RF, Sobel JD, Carrington D, et al. Varicella-zoster virus
(chickenpox) infection in pregnancy. BJOG. 2011;118(10):
1155–1162.
158. Mandelbrot L. Fetal varicella – diagnosis, management, and outcome.
Prenat Diagn. 2012;32(6):511–518.
159. Baud D, Greub G. Intracellular bacteria and adverse pregnancy
outcomes. Clin Microbiol Infect. 2011;17(9):1312–1322.
160. Williams D, Dunn S, Richardson A, Frank JF, Smith MA. Time
course of fetal tissue invasion by Listeria monocytogenes following
an oral inoculation in pregnant guinea pigs. J Food Prot. 2011;74(2):
248–253.
161. Poulsen KP, Czuprynski CJ. Pathogenesis of listeriosis during
pregnancy. Anim Health Res Rev. 2013;14(1):30–39.
162. Lamont RF, Sobel J, Mazaki-Tovi S, et al. Listeriosis in human preg-
nancy: a systematic review. J Perinat Med. 2011;39(3):227–236.
163. Rowe JH, Ertelt JM, Xin L, Way SS. Listeria monocytogenes
cytoplasmic entry induces fetal wastage by disrupting maternal
Foxp3+ regulatory T cell-sustained fetal tolerance. PLoS Pathog.
2012;8(8):e1002873.
164. Nguyen HT, Pandolfini C, Chiodini P, Bonati M. Tuberculosis care for
pregnant women: a systematic review. BMC Infect Dis. 2014;14(617):
1–10.
165. Sugarman J, Colvin C, Moran AC, Oxlade O. Tuberculosis in preg-
nancy: an estimate of the global burden of disease. Lancet Glob Health.
2014;2(12):e710–e716.
166. Bates M, Ahmed Y, Kapata N, Maeurer M, Mwaba P, Zumla A. Perspectives
on tuberculosis in pregnancy. Int J Infect Dis. 2015;32:124–127.
167. Dewan P, Gomber S, Das S. Congenital tuberculosis: a rare manifes-
tation of a common disease. Paediatr Int Child Health. 2014;34(1):
60–62.
168. Molina RL, Diouf K, Nour NM. Tuberculosis and the obstetrician-
gynecologist: a global perspective. Rev Obstet Gynecol. 2013;6(3–4):
174–181.
169. Beeson JG, Duffy PE. The immunology and pathogenesis of
malaria during pregnancy. Curr Top Microbiol Immunol. 2005;297:
187–227.
170. Adams Waldorf KM, McAdams RM. Influence of infection dur-
ing pregnancy on fetal development. Reproduction. 2013;146(5):
R151–R162.
171. Li XL, Wei HX, Zhang H, Peng HJ, Lindsay DS. A meta analysis on
risks of adverse pregnancy outcomes in Toxoplasma gondii infection.
PLoS One. 2014;9(5):e97775.
172. Han M, Jiang Y, Lao K, et al. sHLA-G involved in the apoptosis of
decidual natural killer cells following Toxoplasma gondii infection.
Inflammation. 2014;37(5):1718–1727.
173. Miller DH, Fazekas F, Montalban X, Reingold SC, Trojano M.
Pregnancy, sex and hormonal factors in multiple sclerosis. Mult Scler.
2014;20(5):527–536.
174. Murase JE, Chan KK, Garite TJ, Cooper DM, Weinstein GD. Hormonal
effect on psoriasis in pregnancy and post partum. Arch Dermatol.
2005;141(5):601–606.
175. Amin S, Peterson EJ, Reed AM, Mueller DL. Pregnancy and
rheumatoid arthritis: insights into the immunology of fetal tolerance
and control of autoimmunity. Curr Rheumatol Rep. 2011;13(5):
449–455.
176. Friedman DM, Duncanson LJ, Glickstein J, Buyon JP. A review of
congenital heart block. Images Paediatr Cardiol. 2003;5(3):36–48.
177. Frise CJ, Williamson C. Endocrine disease in pregnancy. Clin Med J
R Coll Phys Lond. 2013;13(2):176–181.
... Якщо ці зміни не відбуваються або відбуваються не повністю, настає неспроможність вагітності на ранніх термінах [6]. Усі ці події характеризуються збільшенням різних факторів росту та цитокінів [2,9]. ...
... ня в І і ІІ триместрах вагітності [5,9]. При цьому суттєве значення має локальна концентрація прогестерону в ділянці імплантації плідного яйця, яка запобігає його відторгненню. ...
Article
Full-text available
Основними причинами несприятливих перинатальних наслідків є невиношування вагітності, плацентарна дисфункція, гестоз, які обумовлені гіперпроліферативними захворюваннями матки (ГПЗМ), перенесеними запальними захворюваннями внутрішніх статевих органів (ЗЗВСО) та стресу. Мета - вивчити ендокринно-імунні фактори у вагітних із ГПЗМ і загрозою переривання вагітності в І триместрі вагітності. Матеріали та методи. Обстежено 64 вагітні жінки репродуктивного віку з ГПЗМ (лейоміомою матки та аденоміозом) на тлі загрози раннього аборту і перенесеними ЗЗВСО до вагітності (основна група). Контрольну групу становили 30 вагітних жінок без ГПЗМ і ЗЗВСО в анамнезі. У сироватці крові вагітних жінок вивчено цитокіновий профіль - TNFα, IFNγ, IL-8, IL-6, IL-4, вміст прогестерону, пролактину та кортизолу. Результати. У І триместрі вагітності середній показник прогестерону в основній групі був на 13% нижчим, ніж у контрольній групі, що свідчило про виражену недостатність функції жовтого тіла вагітності. Також рівень кортизолу та пролактину був вищим майже у 2 рази проти контрольної групи (р<0,05). Співвідношення TNFα/IL-4 було майже в 3 рази вищим, ніж у контрольній групі, а співвідношення IFNγ/IL-4 - майже у 2 рази. IL-8 був підвищеним в основній групі майже вдвічі, а IL-6 - на 25,5% (р<0,05). Висновки. Збільшення прозапальних цитокінів у вагітних із ГПЗМ на тлі загрози раннього аборту може свідчити про зсув цитокінового профілю за цієї патології вагітності за запальним типом і вказує на тривалий перебіг ЗЗВСО до вагітності. Збільшення кортизолу та пролактину може свідчити про напруження стрес-реалізуючої ланки адаптаційних реакцій в умовах воєнного стану в Україні і призводити до раннього аборту. Дослідження виконано відповідно до принципів Гельсінської декларації. Протокол дослідження ухвалено Локальним етичним комітетом зазначеної в роботі установи. На проведення досліджень отримано інформовану згоду пацієнток. Автори заявляють про відсутність конфлікту інтересів.
... Studies suggest that conceptus alloantigens alter maternal immune function both locally at the embryonic-maternal junction and systemically in the peripheral blood circulation to prevent embryonic immune rejection 11,12 . This is achieved through the modulation of maternal immune cells that direct the balance of cytokines towards the Th2 anti-inflammatory pathway 13 . Immunological tolerance displayed by immune cells can be triggered by several molecules such as hormones, cytokines, and enzymes 14 . ...
Article
Full-text available
In beef cattle, estrous synchronization aiming a second artificial insemination (AI) requires a reliable estimation of the pregnancy status 20 days (D20) after the first AI. The hypothesis is that the expression of interferon-stimulated genes (ISGs; ISG15, OAS1, RSAD2, and IFI44) and cytokines (IL1β and IL10) in mononuclear (PBMC) and polymorphonuclear (PMN) cells is regulated by interferon-τ (IFN-τ) and predicts the pregnancy status. PBMC and PMN were isolated from non-pregnant beef cows (N = 9), 10–12 days post-ovulation (D0), and stimulated with 100 ng/mL recombinant ovine (ro) IFN-τ or with pooled uterine flush (UF) from D18 pregnant cows. Both roIFNT and UF stimulated the expression of ISG15, RSAD2, and IFI44 in PBMC and PMN. Expression of IL1β was reduced by UF in both PBMC and PMN. On another experiment, PMN were isolated, and luteal blood perfusion was measured on D20 post-timed-AI in beef females. The accuracy of ISG expression and luteal blood perfusion to predict the pregnancy outcome was determined by ROC curve analysis. All gene combinations were tested, and the best association for increased accuracy (92.7%) and reduction of false-negative results (0.9%, 2/233) was obtained through the combination of the four ISGs (ISG15, OAS1, RSAD2, and IFI44). The criterion was that if the expression levels of at least one of the four genes were greater than the predefined cutoffs, the animal would be considered pregnant. In conclusion, the expression of ISGs and IL1β was upregulated by roIFNT and UF from pregnancy cows. The combined PMN expression of classical (ISG15 and OAS1) and unusual (RSAD2 and IFI44) ISGs provided the greatest predictive accuracy of the pregnancy status on D20 in females with active CL by Doppler and is a potential tool to be used in reproductive programs for beef cattle. Supplementary Information The online version contains supplementary material available at 10.1038/s41598-025-01996-y.
... Саме тому правильний баланс між про-та протизапальними реакціями є основою для нормальної течії вагітності, а цитокіни є сигнальними білками, що керують біологічними процесами під час вагітності, від імплантації до пологів. Так, інтерферон-гамма (IFN-γ) є прозапальним цитокіном, який виділяється імунними клітинами, включає природні кілери (NK) і Th1-клітини, як частину імунної відповіді на молекулярні структури, пов'язані з пошкодженням (DAMP) та патогенно-асоційовані молекулярні структури (PAMP) [15]. IFN-γ є ключовим медіатором відповіді на вірусні збудники та пухлини [16,17]. ...
Article
Full-text available
Preeclampsia remains one of the main causes of obstetric and perinatal complications. The frequency of preeclampsia in the structure of pregnancy complications varies from 6 % to 12 % in healthy pregnant women and from 20 % to 40 % in pregnant women with extragenital pathology. There is growing interest in investigating the exact pathophysiological mechanism of the preeclampsia process as this may identify new potential screening biomarkers. There is evidence that inflammatory mechanisms are a key link between placental ischemia and endothelial dysfunction. It has been hypothesized that impaired regulatory systems are unable to regulate the resolution of inflammatory pathways, contributing to an exaggerated immune response. It is appropriate to call pregnancy a unique immune state that is modulated, but not suppressed. Modulation of the immune system leads to differentiated reactions depending on the stage of pregnancy. Each stage of pregnancy is characterized by a unique inflammatory state. The first and third trimesters are pro-inflammatory, while the second trimester is an anti-inflammatory phase. Proinflammatory mediators, such as IL-6, directly and indirectly through TNF-α, bradykinin, thrombin cause degradation of the endothelial glycocalyx, which leads to an increase in the permeability of the vascular wall with the progression of capillary leakage into the interstitial space. Inflammatory damage to the glycocalyx that occurs during preeclampsia increases intercellular permeability associated with leakage of fluid and albumin into the interstitial space. As a result of damage to the endothelium, there is an increase in the volume of extracellular fluid, which is manifested in women with severe preeclampsia in the form of edema with pathological fluid retention. Further studies are needed to clarify the role of inflammatory mediators in predicting the risk of developing preeclampsia. Further study of the problem will reveal potential targets for treatment, primarily related to inhibition of inflammatory processes and prevention of damage to the endothelial glycocalyx.
... Studies suggest that conceptus alloantigens alter maternal immune function both locally at the embryonic-maternal junction and systemically in the peripheral blood circulation, to prevent embryonic immune rejection [11,12]. This is achieved through the modulation of maternal immune cells that direct the balance of cytokines towards the Th2 anti-in ammatory pathway [13]. Immunological tolerance displayed by immune cells can be triggered by several molecules such as hormones, cytokines, and enzymes [14]. ...
Preprint
Full-text available
In beef cattle, estrous synchronization aiming a second artificial insemination (AI) requires a reliable estimation of the pregnancy status 20 days (D20) after the first AI. The hypothesis is that the expression of interferon-stimulated genes (ISGs; ISG15, OAS1, RSAD2, and IFI44 ) and cytokines ( IL1β and IL10 ) in mononuclear (PBMC) and polymorphonuclear (PMN) cells is regulated by interferon-τ (IFN-τ) and predicts the pregnancy status. PBMC and PMN were isolated from non-pregnant beef cows (N=9), 10-12 days post-ovulation (D0), and stimulated with 100 ng/mL recombinant ovine (ro) IFN-τ or with pooled uterine flush (UF) from D18 pregnant cows. Both roIFNT and UF stimulated the expression of ISG15, RSAD2, and IFI44 in PBMC and PMN. Expression of IL1β was reduced by UF in both PBMC and PMN. On another experiment, PMN were isolated, and luteal blood perfusion was measured on D20 post-timed-AI in beef females. The accuracy of ISG expression and luteal blood perfusion to predict the pregnancy outcome was determined by ROC curve analysis. All gene combinations were tested, and the best association for increased accuracy (92.7%) and reduction of false-negative results (0.9%, 2/233) was obtained through the combination of the four ISGs ( ISG15, OAS1, RSAD2 , and IFI44 ). The criterion was that if the expression levels of at least one of the four genes were greater than the predefined cutoffs, the animal would be considered pregnant. In conclusion, the expression of ISGs and IL1β was upregulated by roIFNT and UF from pregnancy cows. The combined expression of classical ( ISG15 and OAS1) and non-classical (RSAD2 and IFI44 ) ISGs provided the greatest predictive accuracy of the pregnancy status on D20 in females with active CL by Doppler and is a potential tool to be used in reproductive programs for beef cattle.
... Chromosomal abnormalities, Mullerian malformations, maternal illnesses, and other environmental exposures, including smoking and caffeine consumption are among the factors that can lead to a threatened abortion during the first trimester of pregnancy. The disequilibrium of the endogenous microflora triggers a stress response cascade that impedes fetomaternal tolerance and increases the hazard to pregnancy maintenance, according to additional research (Morelli et al., 2015). On the other hand, it has been proposed that a likely cause of threatening abortion is insufficient immunological tolerance between the mother and the fetus (De la Rochebrochard & Thonneau 2002). ...
Article
Full-text available
Progesterone is essential for the maintenance of pregnancy. Threatened abortion and recurrent miscarriage are the common complications of pregnancy. This study conducted to determine the effectiveness of progesterone supplementation in preventing miscarriage in cases of threatened abortion and recurrent miscarriages, aiming to extend pregnancy beyond 24 weeks in these women. The objectives of this study are to identify the number of pregnant women treated with progesterone, to know the choice of progesterone formulation prescribed for threatened abortion and previous history of miscarriage, and to determine the effectiveness of progesterone treatment among patients with threatened abortion and previous history of miscarriage. The results showed that a total of 107 cases were treated with progesterone, Among the majority of the cases were threatened abortion cases 50 (46.42%), followed by previous history of miscarriage cases 26 (24.29%), previous history of preterm birth cases 15 (14%), primi gravida cases 12 (11.21%) and short cervix cases 4 (3.7%). The outcome of this study determines that, out of 50 (46.42%) threatened abortion cases, 46(92%) women successfully continued their pregnancies to proceed beyond 24 weeks, and 4(8%) cases got abortion. Among 26 (24.29%) previous history of miscarriage cases 25 (96.1%) women successfully continued their pregnancies and 1(3.8%) got an abortion. It also demonstrates the rate of abortion was reduced in women treated with progesterone supplementation.
Article
In sheep production, due to the limitations of breeding conditions, the uteri of ewes are often infected with bacteria, resulting in the failure of embryo implantation or loss, causing huge losses to the sheep industry. Therefore, in this study, by using RT-qPCR, Western blot, and immunofluorescence, we investigated the effects of LPS infusion on the immune microenvironment and endometrial receptivity, which play an important role in the process of embryo implantation in ruminants, during the three critical periods of embryo implantation in sheep. The results showed that LPS infusion at day 12, day 16, and day 20 significantly increased the expression of Th1 cytokines (TNF-α, IL-1β, IL-8, IL-6), while significantly decreasing the expression of Th2 cytokines (IL-4 and IL-10) and disrupting the expression of implantation factors, such as ITGB3, ITGB5, VEGF, and LIF, in the endometrial tissues of sheep. Additionally, the protein expression level of TLR4 and the phosphorylation level of ERK were significantly elevated at day 12, day 16, and day 20 after LPS infusion, suggesting that LPS may impair endometrial receptivity through the TLR4/ERK pathway. Validation was conducted in a receptive model of sEECs using TLR4 and ERK phosphorylation inhibitors. Compared with the LPS group, TLR4 and ERK phosphorylation inhibitors significantly reduced the expression of TLR4 and p-ERK, down-regulated Th1 cytokines, up-regulated Th2 cytokines, and alleviated the disruption of genes for attachment. Treatment with 50 μM PTE can significantly alleviate the abnormal expression of implantation genes caused by LPS, and its mechanism may be related to the regulation of the ERK signaling pathway.
Article
Full-text available
This review article investigates the intricate relationship between increased ozone depletion-induced radiation, cancer risk, and pregnancy, with a focus on pregnant women as a vulnerable population. By examining the mechanisms of radiation induced carcinogenesis and the physiological changes during pregnancy that may alter susceptibility, the article emphasizes the need for targeted research and public health strategies. Drawing on current knowledge and epidemiological evidence, the paper explores the impact of radiation exposure on cancer risk in pregnant women, emphasizing the need for proactive health strategies and preventive measures to safeguard maternal and fetal well-being. The implications for future research and practice are discussed, highlighting the importance of longitudinal studies, risk assessment models, intervention strategies, and health education initiatives to advance our understanding and enhance health outcomes in this critical area. By fostering collaboration and innovation, we aim to address the multifaceted challenges posed by radiation exposure and cancer risk in pregnant women, working towards a healthier future for all.
Article
Background: Monitoring biologic drug therapy during pregnancy in women with immune-mediated inflammatory diseases (IMIDs) is crucial to ensure treatments align with evidence-based practices. Research design and methods: A retrospective cohort study based on healthcare claims data from eight Italian regions was conducted, analyzing deliveries between 2009 and 2021. The study included women receiving biologic drugs within nine months before their last menstruation. Exposures to biologics, conventional disease-modifying anti-rheumatic drugs (DMARDs) and symptom-relieving medications were assessed in the trimesters (T) before, during and after pregnancy. Factors influencing biologic treatment persistence during pregnancy were analyzed. Results: A cohort of 1,763 deliveries was considered. Biologic drugs were prescribed for rheumatic (33.6%), dermatological (32.6%), and gastrointestinal diseases (28.4%). Biologic use declined during pregnancy (TI = 37.3%; TII = 17.6%; TIII = 11.3%), increasing again postpartum. During pregnancy, there was increased use of symptom-relieving medications for rheumatic diseases and DMARDs for gastrointestinal diseases. Factors associated with continued biologic treatment included being older than 35 years and the region of delivery. Conclusions: This study found a decrease in biologics drug use during pregnancy and highlights the necessity for personalized therapeutic approaches. Geographic variations in biologic drug use emphasize the need for educational initiatives about the risk-benefit profiles of these therapies during pregnancy.
Article
Recurrent pregnancy loss (RPL) is an obstetric condition estimated to affect 2–4% of childbearing individuals globally. Due to its varied nature, medical societies globally differ in their diagnostic criteria. Its aetiologies are numerous, ranging from anatomic abnormalities to endocrine and immunological factors. Autoimmune factors can attribute to approximately 20% of cases and include dysregulation of immune cells, cytokine production and antiphospholipid syndrome. Treatment pathways vary by aetiology; however, many cases remain unexplained, adding an additional level of complexity to this condition. Due to its recurrent nature, this type of pregnancy loss has profound impacts on mental health during subsequent pregnancies. While some aspects of RPL have been widely investigated, there continues to be a gap in research, such as its impacts on non-birthing parents and specific sociodemographic groups.
Article
Full-text available
Tuberculosis (TB) has been recognized as an important cause of morbidity and mortality in pregnancy for nearly a century, but research and efforts to roll out comprehensive TB screening and treatment in high-risk populations such as those with a high prevalence of HIV or other diseases of poverty, have lagged behind similar efforts to address HIV infection in pregnancy and the prevention of mother-to-child-transmission. Immunological changes during pregnancy make the activation of latent TB infection or de novo infection more likely than among non-pregnant women. TB treatment in pregnancy poses several problems that have been under-researched, such as contraindications to anti-TB and anti-HIV drugs and potential risks to the neonate, which are particularly important with respect to second-line TB treatment. Whilst congenital TB is thought to be rare, data from high HIV burden settings suggest this is not the case. There is a need for more studies screening for TB in neonates and observing outcomes, and testing preventative or curative actions. National tuberculosis control programmes (NTPs) should work with antenatal and national HIV programmes in high-burden populations to provide screening at antenatal clinics, or to establish functioning systems whereby pregnant women at high risk can drop in to routine NTP screening stations. Copyright © 2014 The Authors. Published by Elsevier Ltd.. All rights reserved.
Article
The hypothesis that direct exposure to or clinical illness with measles, rubella, mumps or chickenpox may play a causal role in autism has been evaluated. The study subjects were 163 cases of autism and their 355 unaffected siblings. Medical data relating to prenatal life and early infancy were obtained by interviewing the parents, and from medical records. Medical records were found for only about half the instances In which a pertinent illness or exposure was reported in the home Interview. However, there was no evidence of systematic bias In recall on the part of parents when reporting on an index case. When medical records and parental statements were both available they were relatively consonant. Increased frequency of combined illness and exposure was seen for the cases relative to their sibling controls with regard to prenatal measles, rubella and mumps and to postnatal mumps. Yet the proportion of cases with histories of viral experience during the periods of interest was small, indicating that these four viruses were unlikely to have played a major role In any substantial proportion of the cases. Some associations were noted with parental histories of other infections during gestation or infancy. Again, however, the proportion of cases that could be accounted for by any of these associations, even if causal, is small.
Article
PROBLEM: In hematopoietic cells, interleukin (IL)-13 shares many actions with IL-4. The effects of IL-13 in gestational tissues have yet to be reported, however. We compared the effects of IL-4 and IL-13 on the production of cytokines and prostaglandin E-2 (PGE(2)) in epithelial amnion-derived WISH cells. METHOD OF STUDY: WISH cells were treated with IL-4 or IL-13 (0.08-10 ng/ml) with/without cotreatment with IL-1 beta (0.2 ng/ml), tumor necrosis factor-alpha (10 ng/ml) or epidermal growth factor (5 ng/ml). The production of IL-6, IL-8, and PGE(2) was measured by immunoassay after 16 hr. RESULTS: Both IL-4 and IL-13 inhibited PGE(2) production with indistinguishable concentration-response curves, under basal or stimulated conditions. The maximal inhibition of 1 beta-stimulated PGE(2) production (to 28% +/- 10% of control) was seen at 10 ng/ml of IL-4 or IL-13. Basal IL-6 production was stimulated approximately twofold by IL-4 and IL-13, whereas IL-4 and IL-13 both inhibited cytokine-stimulated (but not basal) IL-8 production by approximately 50%. In the presence of 1 ng/ml of IL-4, IL-13 was unable to further inhibit PGE(2) production. CONCLUSIONS: The inhibition of PGE(2) and IL-8 production by IL-4 in WISH cells is mimicked by IL-13. Both cytokines, probably through binding to a common receptor complex, may share a role in suppressing inflammatory reactions within intrauterine tissues.
Article
Maternal viral infection is known to increase the risk for schizophrenia and autism in the offspring. Using this observation in an animal model, we find that respiratory infection of pregnant mice (both BALB/c and C57BL/6 strains) with the human influenza virus yields offspring that display highly abnormal behavioral responses as adults. As in schizophrenia and autism, these offspring display deficits in prepulse inhibition (PPI) in the acoustic startle response. Compared with control mice, the infected mice also display striking responses to the acute administration of antipsychotic (clozapine and chlorpromazine) and psychomimetic (ketamine) drugs. Moreover, these mice are deficient in exploratory behavior in both open-field and novel-object tests, and they are deficient in social interaction. At least some of these behavioral changes likely are attributable to the maternal immune response itself. That is, maternal injection of the synthetic double-stranded RNA polyinosinic-polycytidylic acid causes a PPI deficit in the offspring in the absence of virus. Therefore, maternal viral infection has a profound effect on the behavior of adult offspring, probably via an effect of the maternal immune response on the fetus.
Article
Effects of maternal immune stimulation on Toll-Like Receptors (TLR): We are also examining the effects of poly(I:C) exposure during pregnancy on the expression and function of TLRs during fetal development and in neonates and adult offspring. TLRs are part of a larger family of membrane bound cell surface and intracellular Pattern Recognition Receptors (PRR). Eleven TLRs have been discovered in humans (TLRs 1-11), and 12 TLRs have been found in mice (TLRs 1-12). First discovered in Drosophila [75], this family of molecules is very heterogenous, complex, and highly conserved among species. Individual TLRs bind to particular microbial products, such as LPS, peptidoglycan, lipoproteins, and flagellin on bacteria, as well as viral fusion protein, unmethylated CpG motifs, double- and single-stranded RNAs [76, 77]. TLR expression by cells of the innate and adaptive immune systems allows these cells to recognize and respond to extracellular and intracellular microbial pathogens. Downstream cell signaling pathways are initiated when ligands bind to TLRs, leading to activation of different transcription factors (e.g., NF-κB and others), which stimulate expression of pro-inflammatory cytokine genes (e.g., IL-1, IL-6, TNF, interferons). Response to auto-antigens: In order determine if components of the adaptive immune system also mount more robust responses following immune stimulation we chose a well-characterized EAE model of an immune-mediated disease, and used adult offspring of pregnant dams with immunological memory. We have previously shown that T cells from offspring of poly(I:C)-injected pregnant dams with immunological memory (i.e., immune) preferentially differentiate to become Th17 cells after in vitro activation [43-45, 60]. In contrast, T cells from offspring of poly(I:C)-injected immunologically naïve pregnant dams (i.e., non-immune) do not show such Th17 cell preferential differentiation. Th17 cells have been shown to be involved in the neuropathology responsible for the clinical symptoms that develop in Experimental Autoimmune Encephalomyelitis (EAE), a mouse model of multiple sclerosis [122-128]. Female offspring of poly(I:C) (vs. PBS) -injected pregnant dams were injected s.c. in each hind flank with an encephalogenic-peptide (MOG35-55) in Complete Freund ’s adjuvant (CFA). I.p. injections of pertussis toxin were given after MOG immunization to enhance the immune response and promote T cell migration into the brain [129]. Typically, 10 – 12 days after injection of MOG and pertussis, 90% of B6 mice develop progressively: weakness and paralysis in their tail, hindlimb paresis and finally hindlimb paralysis. Controls that receive CFA and pertussis toxin, but no MOG, do not develop clinical signs of EAE.
Article
Approximately 10% of peripheral CD4+ cells and less than 1% of CD8+ cells in normal unimmunized adult mice express the IL-2 receptor a-chain (CD25++) molecules. When CD4+ cell suspensions prepared from BALB/c nu/+ mice lymph nodes and spleens were depleted of CD25++ cells by specific mAb and C, and then inoculated into BALB/c athymic nude (nu/nu) mice, all recipients spontaneously developed histologically and serologically evi dent autoimmune diseases (such as thyroiditis, gastritis, insulitis, sialoadenitis, adrenalitis, oophoritis, glomerulo nephritis, and polyarthritis); some mice also developed graft-vs-host-like wasting disease. Reconstitution of CD4+ cells within a limited period after transfer of CD4+ cells prevented these autoimmune de velopments in a dose-dependent fashion, whereas the reconstitution several days later, or inoculation of an equivalent dose of CD8+ cells, was far less efficient for the prevention. When nu/nu mice were transplanted with allogeneic skins or immunized with xenogeneic proteins at the time of CD25++ cell inoculation, they showed significantly heightened immune responses to the skins or proteins, and reconstitution of CD4+ cells normalized the responses. Taken together, these results indicate that CD4+ cells contribute to maintaining self-tolerance by down-regulating immune response to self and non-self Ags in an Ag-nonspecific manner, pre sumably at the T cell activation stage; elimination/reduction of CD4+ cells relieves this general suppres sion, thereby not only enhancing immune responses to non-self Ags, but also eliciting autoimmune responses to certain self-Ags. Abnormality of this T cell-mediated mechanism of peripheral tolerance can be a possible cause of various autoimmune diseases.
Article
The mechanism by which the mammalian mother accepts the implanting fetus as an allograft remains unexplained, but is likely to be the result of a combination of factors. Mononuclear cytotrophoblasts, the specialized fetal cells of the placenta that invade the uterus, play an important role. These cells express HLA-G, an unusual major histocompatibility complex class I-B molecule, and secrete cytokines and pregnancy-specific proteins that can regulate immune function. We investigated whether cytotrophoblasts secrete interleukin 10 (IL-10), a cytokine that potently inhibits alloresponses in mixed lymphocyte reactions. Cytotrophoblasts from all stages of pregnancy produced IL-10 in vitro, but neither placental fibroblasts nor choriocarcinoma (malignant trophoblast) cell lines did so. Spontaneous IL-10 production averaged 650, 853, and 992 pg/10(6) cells in the first, second, and third trimesters of pregnancy, respectively. IL-10 secretion dropped approximately 10-fold after the first 24 h of culture, and was paralleled by a decrease in messenger RNA. IL-10 messenger RNA was detected in biopsies of the placenta and the portion of the uterus that contains invasive cytotrophoblasts, suggesting that this cytokine is also produced in vivo. IL-10 secreted by cytotrophoblasts in vitro is bioactive, as determined by its ability to suppress interferon gamma production in an allogeneic mixed lymphocyte reaction. We conclude that human cytotrophoblast IL-10 may be an important factor that contributes to maternal tolerance of the allogeneic fetus.