Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

Chapter 115

Amniotic Fluid and the Fetal Mucosal


Immune System
Stella Nowicki
Department of Microbiology and Immunology, Meharry Medical College, Nashville, TN, USA

Randall M. Goldblum
Pediatric Child Health Research Center, University of Texas Medical Branch, Galveston, TX, USA

Chapter Outline
Introduction2251 Nonspecific Antimicrobial Factors 2260
Development of the Amniotic and Chorionic Humoral Immunity 2261
Cavities and the Placenta 2252 Immunoglobulins and Immunoglobulin
Production of Chorionic and Amniotic Fluids 2254 Transporters2261
Metabolism of Amniotic Fluid 2256 Increased Activity of the Immune System in
Immunological Factors in Amniotic Fluid 2256 African Americans 2263
Innate Immunity 2257 Pathological Conditions Involving Amniotic Fluid 2264
Complement and Complement Inhibitors 2257 Chorioamnionitis2264
Cytokines, Chemokines, and Other Modulatory and Fetal Growth Restriction 2265
Growth Factors 2258 Antiretroviral Drugs and Amniotic Fluid 2265
Cytokines2259 References2265

INTRODUCTION
responses at these sites. In this respect, amniotic fluid might
Amniotic fluid, the solution contained within the amniotic be considered a precursor of human milk feedings as an
membranes, surrounds the developing fetus from about exogenous source of immunological factors that protect the
the sixth week of gestation until immediately before birth, infant directly and shape the responses of the infant’s own
when the fluid is released. A major function of the amniotic immune system. Conversely, since some of the volume and
fluid is to protect the infant from physical trauma due to perhaps the solute content of amniotic fluid are produced
forces that might be applied to the mother’s abdomen. A by the mucosal surfaces of the fetus, it can also be consid-
less conspicuous but important function of amniotic fluid is ered an early product of the fetus’s mucosal immune sys-
its role in protecting the fetus from microbial colonization. tem. This chapter is largely limited to human amniotic fluid,
This chapter reviews the development of the chorionic and since there are major differences between the structure and
amniotic cavities and the synthesis, composition, and fate the function of hemochorial placentas of primates and those
of amniotic fluid. Evidence concerning the role of amni- of other mammals and since there are very few studies on
otic fluid in protecting the infant from immunological or amniotic fluid in nonhuman primates.
infectious injury will then be addressed. Furthermore, since Among the body fluids, amniotic fluid is perhaps one
amniotic fluid is produced by or at least transferred across of the least well studied. This may be due in part to the
epithelial surfaces and bathes many of the mucosal surfaces limited opportunities to access this fluid. Amniotic fluid is
of the fetus, it should be considered part of the mucosal frequently sampled by amniocentesis for genetic analysis
immune system. This important concept is also explored. of the fetus between 10 weeks and the end of the first half
Early exposure of the fetal respiratory and gastrointestinal of pregnancy. Fluid samples are also removed for studies on
tracts to amniotic fluid might help shape mucosal immune fetal maturity, usually during the last 10 weeks of normal
Mucosal Immunology. http://dx.doi.org/10.1016/B978-0-12-415847-4.00115-4
Copyright © 2015 Elsevier Inc. All rights reserved. 2251
2252  SECTION | I  Genitourinary Tract and Mammary Gland

gestation. Finally, large amounts of fluid are available near cleft lined on one side by the ectodermal plate and on the
the time of delivery. However, the later samples have the other side by cells (amnioblasts) that grow from the edge of
potential to be contaminated with maternal cervical and the ectodermal layer and migrate along the overlying cyto-
vaginal fluid unless they are collected during cesarean sec- trophoblast layer. A similar process occurring on the ventral
tion or after insertion of monitoring devices into the amni- or endodermal surface of the blastocyst results in formation
otic cavity. Thus, studies on the development of various of the primitive yolk sac (Figure 1(a)). The chorionic cav-
constituents in the amniotic fluid often have temporal gaps ity, which develops shortly thereafter from extraembryonic
because of the unavailability of samples during some stages mesoderm, surrounds the developing embryo, the amniotic
of gestation. cavity, and the secondary yolk sac by 13 days of gestation
The diagnostic usefulness of amniotic fluid became (Figure 1(b)).
broadly recognized when intrauterine treatments were As the growing embryo causes the surface of the decidua
developed for isoimmune sensitization against the infant’s (modified uterine endometrium) to protrude into the uter-
rhesus antigens. Amniotic fluid has also been used as a win- ine cavity during the second month of gestation, the cho-
dow into the physiological events that lead up to both nor- rionic and amniotic cavities are pushed closer together as
mal and premature parturition. The latter studies have often they approach from the opposite side of the uterine cavity
focused on cytokines and prostaglandins in the amniotic (Figure 2(a)). By the end of the third month of gestation,
fluid that might also impact the development of the immune the chorionic and amniotic membranes fuse with each other
system of the fetus. (Figure 2(b)). The outer surface of the expanding chorionic
membrane also fuses to the surrounding decidua parietalis,
DEVELOPMENT OF THE AMNIOTIC AND forming a single membrane which surrounds the growing
fetus. From the end of the third month to the end of ges-
CHORIONIC CAVITIES AND THE PLACENTA tation, this fluid-filled chorioamniotic sac containing the
The amniotic cavity begins its development during the fetus fills the entire uterine cavity not occupied by the pla-
bilaminar germ disk stage, in the second week of gestation, centa. At the cervical os, this membrane is modified in that
as a cleft within the ectoderm of the inner mass of embry- the chorioamniotic membrane is not fused to the decidua.
onic cells (Langman, 1981). At first, the cavity is a simple The fetal surface of the placenta and the outer surface

(a) Trophoblastic lacunae Maternal sinusoids (b) Prochordal


plate
Primary stem
villi
Trophoblastic
lacunae

Maternal
sinusoid
Connecting
stalk
Amniotic Amniotic
Endoderm
cavity cavity
cells

Secondary
yolk sac

Extra-embryonic
Extra-embryonic somatopleuric
coelom mesoderm
(chorionic plate)

Extra-embryonic
Extra-embryonic coelom
splanchnopleuric (chorionic cavity
mesoderm

Extra-embroyonic
Exocoelomic cavity somatopleuric mesoderm
Exocoelomic
(primitive yolk sac)
membrane Exocoelomic cyst

FIGURE 1  Embryologic development of the chorionic and amniotic cavities. (a) Human blastocyst at about 12 days of gestation. The amniotic cav-
ity is developing between the columnar ectodermal plate and the small ectodermal cells migrating from the edges of the plate. (b) Human blastocyst at
about 13 days. The extraembryonic coelom has developed into the chorionic cavity, and the amniotic cavity has enlarged. Modified with permission from
Langman (1981).
Amniotic Fluid and the Fetal Mucosal Immune System Chapter | 115  2253

of the umbilical cord are also covered by the amniotic mem- the cells closest to the fetus are less mature but more meta-
brane. Thus, the chorioamniotic membrane, derived from bolically active than those that have migrated farther from
fetal mesoderm and ectoderm, forms a large area of inter- the fetus. The potential ability of amniotic epithelial cells to
face with maternal uterine decidua. secrete and/or transport water and immunologically active
Detailed histological examination of the chorioamniotic agents will be considered later. In addition, the more cuboi-
membranes at term (Danforth and Hull, 1958) revealed that dal amniotic epithelium of the reflected membranes may be
the amnion consisted of a single layer of epithelial cells, important in the more passive transport of fluid and solutes
superimposed and tightly attached to an underlying layer of to and from the maternal decidua through the chorionic
fibrous connective tissue about twice as thick as the cellular membrane.
layer. There is no organized basement membrane separating The placenta is formed from the outer cell mass of the
these two layers of the amnion. The amniotic cells seem to embryo, which differentiates into individual cytotropho-
have a unique morphology that differs for those lining the blasts and a multinucleate mass of syncytiotrophoblasts,
inner surface of the reflected membrane and those overly- which invade the endometrial stroma after implantation. By
ing the fetal surface of the placenta. The amniotic epithelial the ninth day of gestation, the syncytiotrophoblasts begin
cells covering the placenta are more complex in appearance, to form intercommunicating clefts or trophoblastic lacunae.
being columnar in shape and containing a canalicular appa- The syncytiotrophoblasts go on to invade the uterine vessels,
ratus in the Golgi zone, secretory granules, and brush-type allowing maternal blood to course through the lacunae and
borders at the basal surface (Danforth and Hull, 1958). The establish the uteroplacental circulation. The cytotrophoblasts
latter are tightly applied to the underlying fibrous connec- then begin to form villi that protrude into these trophoblastic
tive tissue. The epithelial cells of the reflected amniotic lacunae. These villi are themselves invaded centrally by fetal
membrane are cuboid, having fewer cellular elements and mesoderm, which differentiates into blood vessels by the
a more mature appearance. Given the ectodermal origin, third week of gestation. Thus, the mature villi, or fingerlike
antigenic similarity, and continuity between fetal skin and projections, which interface with maternal blood are formed
amniotic epithelial cells, it is interesting to speculate that by three cellular layers: a syncytiotrophoblastic outer layer,

(a) (b)
Fused decidua parietalis, Placenta
chorion laeve and amnion
Decidua basalis

Chorion
Decidua frondosum
parietalis

Chorionic
cavity Amniotic
cavity

Yolk sac

Decidua
capsularis
Uterine
cavity

Chorion laeve
Amniotic cavity

FIGURE 2  Embryologic development of the fetus, placenta, and chorionic and amniotic cavities. (a) Human fetus at the end of the second month.
Note that the amniotic cavity is still separate, though within the chorionic cavity. (b) Human fetus at the end of the third month. Note that the chorionic
and amniotic cavities have now fused and that the uterine cavity is essentially obliterated by the fusion of the chorion laeve and decidua parietalis. The
small cavity that remains near the cervical os is a potential site for transport of maternal cervicovaginal secretions into the chorioamniotic sac. Modified
with permission from Langman (1981).
2254  SECTION | I  Genitourinary Tract and Mammary Gland

a cytotrophoblastic middle layer, and a mesodermal inner during the first 12 and 14 weeks of gestation, respectively.
or vascular layer. As the villi develop, they lengthen and These results also indicated a marked gradient of total
branch. The ends of these branches are the terminal villi. It protein concentration between maternal blood and cho-
is these terminal chorionic villi, which protrude into the tro- rionic fluid (18 times) and chorionic and amniotic fluids
phoblastic lacunae filled with maternal blood, that form the (54 times), suggesting that the amniotic membrane is rela-
major anatomical site for the exchange of gases and nutri- tively impermeable to proteins during the first trimester of
ents between the fetus and the mother. This is also a site gestation. They concluded that the major sources of the low
for active transport of immunological factors, such as IgG, concentration of proteins in the amniotic cavity during the
directly from the maternal to the fetal circulation without first half of gestation are the embryo and its yolk sac and,
transiting the amniotic cavity. subsequently, the fetus. However, the concentration of pro-
teins in the amniotic cavity increases dramatically at about
PRODUCTION OF CHORIONIC AND 11 weeks of gestation. This is about the time of fusion of
the chorionic and amniotic membranes, which allows much
AMNIOTIC FLUIDS more rapid diffusion of solutes from the maternal decidua
The sources of the numerous constituents of chorionic and across the fused fetal membranes (Jauniaux et al., 1994).
amniotic fluids throughout gestation are not known in detail. Jauniaux et al. (1995) subsequently measured IgG, IgA,
Since the fluids in these compartments are in contact only IgM, C3, C4, and specific IgG antibodies to potential intra-
with fetal surfaces, the proximal sources are predominantly uterine pathogens in the chorionic and amniotic fluids and in
fetal. However, maternal contributions can also reach these the mother’s serum during the same early (6–12 weeks) ges-
spaces, either indirectly via the maternofetal circulation or tation period. These are all proteins that are either absent or
more directly by diffusion or active transport by the fetal present in extremely low concentrations, in the early fetus.
membranes of constituents that diffuse from the decidua or Their assays for C3 and C4 were sensitive enough to detect
intervillus spaces and across the chorionic plate of the pla- only concentrations greater than 10% of serum levels, elimi-
centa. The potential sources of chorionic and amniotic fluids nating their usefulness in assessing the transport of these
thus include fetally derived constituents from the external proteins into the fetal cavities. However, data from Gitlin
and mucosal surfaces of the fetus, including the definitive et al. (1972) suggest that IgG and IgA, in amounts adequate
yolk sac, the umbilical cord, and the fetal circulation to the to achieve μg/mL concentrations, reach the chorionic sac
chorionic and amniotic membranes; maternally derived during early gestation by either size-dependent, passive dif-
constituents from the decidua that diffuse or are transported fusion or active transport of selected molecules from the
across one or more fetal membranes into the cavities; and maternal decidua parietalis or intervillus spaces of the pla-
the maternal blood in the intervillus spaces of the placenta, centa. The difference between the maternal serum to chori-
across the overlying chorioamniotic membranes. onic fluid gradients for IgG (28-fold) and monomeric IgA
In the early stages of development, the amniotic cleft (128-fold) suggests some selectivity of transport for these
must be filled with fluid produced by the fetal ectoder- proteins of similar size and structure. However, the concen-
mal cells surrounding it (Figure 1(a)). However, as the tration of both of these immunoglobulin classes in chorionic
cleft enlarges into a cavity and the lining ectoderm thins, fluid correlated with increasing gestational age and with
it becomes possible for water or specific solutes to be each other. No higher-molecular-weight IgM was detectable
transported from the surrounding chorionic cavity into the in the chorionic fluid. Significantly, of the proteins quanti-
amniotic cavity (Figure 1(b)). Studies of the transport of fied, only IgG (the analyte of highest concentration in the
fluid and various solutes into and out of the chorionic and chorionic fluid) was detected in the amniotic fluid, and in
amniotic cavities of humans are predominantly restricted to only two of 34 fetuses. This is most consistent with limited
the later period of gestation, when the single chorioamni- diffusion of these proteins from the chorionic fluid across the
otic sac becomes accessible by amniocentesis. Studies of amniotic membrane. Given the sensitivity of the assays for
chronically cannulated fetuses in experimental animals, IgG and IgA, a gradient of greater than 30-fold and 10-fold,
predominantly sheep, have also provided data concerning respectively, across the amniotic membrane would have pre-
the production and metabolism of fetal fluids. cluded detection of these proteins in amniotic fluid at about
However, Jauniaux et al. (1994) selectively sampled the 11 weeks of gestation. The sources and potential function of
chorionic and amniotic sacs of human fetuses under ultra- the immunoglobulins and antibodies in the chorionic and
sound guidance before elective abortions at 6–12 weeks of amniotic cavities are considered further later.
gestation. They determined the total protein and albumin in The volume of amniotic fluid increases steadily until
maternal serum, chorionic fluid, and amniotic fluid, as well approximately 30 weeks, when it peaks at about 900 mL.
as the concentration of α-fetoprotein in maternal serum and After that it begins to decrease, at first slowly but then more
chorionic fluid. The total protein increased linearly in the rapidly after 35 weeks of gestation (Lotgering and ­Wallenburg,
chorionic cavity and exponentially in the amniotic cavity 1986). The rate at which water and various solutes flow
Amniotic Fluid and the Fetal Mucosal Immune System Chapter | 115  2255

into and out of the amniotic fluid late in gestation has been infants born after 28–30 weeks of gestation have transepi-
studied with radiolabeled and stable isotope clearance tech- thelial water losses during the first day of life that are up
niques. Water and sodium are replaced quite rapidly: 34% to 15 times higher than those of infants born at term (Sedin
and 7% every hour, respectively (Vosburge et al., 1948). et al., 1981). Although the fetal skin has a high density of
Gitlin et al. (1972) injected several isolated radiolabeled sweat glands, their contribution to amniotic fluid volume
serum proteins into the amniotic cavity via amniocentesis. and content is not well established.
They then measured the volume of amniotic fluid by iso- The fetal kidney is known to produce urine during the
tope dilution and the rate of clearance of the proteins by second half of gestation (Langman, 1981), and urine has
obtaining fluid samples by repeat amniocentesis and at the been detected in the fetal bladder as early as 11 weeks of
time of cesarean section. Their results indicated that near gestation (Wladimiroff and Campbell, 1974). The volume
term, the amniotic fluid volume was about 600 mL ±10%. of urine produced by the human fetus increases in propor-
Approximately 2.6% of all the labeled proteins tested were tion to fetal size, from about 110 mL/kg/day at 25 weeks
cleared per hour in the absence of labor. In the presence of to 190 mL/kg/day at 39 weeks of gestation. Since the vol-
labor the apparent hourly clearance of proteins increased ume of amniotic fluid is stable or decreasing during this
to 3.7%. The role of the infant in protein clearance was period, the increasing output of urine must be matched by
investigated by comparison with two pregnancies with dead an increase in the fluid clearance rate. One effect of the
fetuses. The rate was reduced to 12% per day, or approxi- increasing urine output from the fetus is a decrease in the
mately 0.5% per hour. Gitlin’s results indicate that 60–90% osmolarity and sodium concentration, from about 98% to
of amniotic fluid proteins are replenished each day. The 91% of that in maternal or fetal plasma. This is accompa-
volume of maternal serum required to maintain the protein nied by an increase in urea, from 100% to 168% of that
levels in the fluid varied from 12 to 18 mL for albumin and in maternal plasma. However, measurement of albumin in
transferrin, respectively, to as little as 6.3 and 1.4 mL for the first urine after birth indicated that fetal urine could
IgG and IgA, respectively. Whereas the differences between account for only a small proportion of the production of
the transfer of immunoglobulins and smaller serum proteins amniotic fluid proteins at term (Gitlin et al., 1972). The
might be explained by their concentrations in the decidua, urinary contribution to amniotic fluid IgA and transferrin
the differences might also suggest that the immunoglobu- was also small. However, according to Gitlin’s calculations,
lins are actively transported into the chorioamniotic cavity IgG in amniotic fluid could be derived largely from fetal
late in gestation. urine. Nonetheless, even for IgG, the majority of this IgG
Under the conditions of mass removal of amniotic fluid is probably produced by the mother and transferred via the
(indicated by Gitlin et al., 1972), the rates of clearance of placenta to the infant’s circulation prior to excretion in the
all amniotic fluid proteins—and thus their rate of formation fetal urine. The amount of water and individual proteins
(when the volumes and concentrations are stable)—can be released from other mucosal surfaces of the fetus cannot be
ascertained from the rate of disappearance of any labeled ascertained by this type of study. By examining the levels of
protein and the endogenous concentration of the constitu- specific proteins in the blood of the newborn infant, inves-
ent of interest. Total protein accretion was found to be 0.79 tigators suggested that little or none of the IgA could come
and 1.06 g/day for patients prior to and during labor, respec- from the fetus (Gitlin et al., 1972). However, this conclu-
tively (Gitlin et al., 1972). They also found that approxi- sion ignores the possibility that the IgA is produced locally
mately one-half of the protein in amniotic fluid is albumin. and excreted from the mucosal surfaces (other than urinary
The number of potential sites for secretion of various tract) of the fetus.
constituents into the amniotic fluid is expanded after organ- The concentrations of a product of secretory epithelium,
ogenesis. Each of the fetal and maternal sites is considered free secretory component (fSC), in the amniotic fluid and
in the following discussion. During the first half of gestation first urine samples voided by infants have been investi-
the fetal skin is very thin, consisting of only a few cell lay- gated (Cleveland et al., 1991b). The mean concentrations
ers, and lacks the keratinified epithelium that prevents fluid (7.5 and 6.8 μg/mL) were similar in term amniotic fluid and
loss later in gestation and in postnatal life. The superficial first voided urine. However, with the turnover rates for pro-
layer or periderm cells contain structures suggesting that teins noted by Gitlin et al. (1972), the full-term fetus would
these cells are metabolically active (Brace, 1986). In vitro have to have voided from 3.5 to 48 L of urine to account
and in vivo studies indicate the permeability of the periderm for all the fSC in the amniotic fluid, an unlikely event for a
to water and sodium and transport of urea and electrolytes 2 kg fetus. Thus, some other secretory epithelia of the fetus
(Lind et al., 1972). Thus, early in gestation, the amniotic or the chorioamniotic membranes must account for at least
fluid water and electrolytes are believed to pass relatively 90% of the fSC in the amniotic fluid of term infants. This
freely from the infant’s skin to the amniotic fluid. That proposition is consistent with the observation of Tourville
this exchange process may continue, to some extent, later et al. (1969) that SC can be detected in amniotic epithelial
in gestation is suggested by observations that premature cells by immunofluorescence.
2256  SECTION | I  Genitourinary Tract and Mammary Gland

The fetal respiratory tract is another site of fluid produc- the advancing gestational age, providing excellent metabo-
tion. In fetal sheep, 30–50 mL/kg/day of fluid flows from lomic evidence of fetal maturation and having the potential
the trachea. However, it is not clear how much of this fluid to improve the evaluation of fetal health and development.
is immediately swallowed by the fetus. The observation that The detection and quantification of creatinine may provide
surface-active phospholipids (surfactants) appear in human new biomarkers for assessing the renal status of the fetus.
amniotic fluid during the third trimester of gestation sug- Several findings described in the preceding section sug-
gests that some respiratory secretions reach the amniotic gest that the process by which amniotic fluid proteins are
cavity. cleared late in gestation involves coordinated (mass) trans-
Saliva and nasal secretions derived from the salivary port of water and proteins (Gitlin et al., 1972). However, the
glands and nasal mucosa, respectively, may also contribute data from Gitlin et al. indicate that very little of the radio-
to the volume and content of the amniotic fluid. In the fetal labeled protein injected into the chorioamniotic sac reaches
lamb, this is estimated to represent only about 30 mL/day, the maternal circulation. This may be due, at least in part, to
but this excludes the portions of the secretions that are swal- the rapidity of amniotic fluid removal by the fetus (see later
lowed (Brace, 1986). These secretions may contribute to the discussion), but other pressure or concentration gradients
mucoid nature of the amniotic fluid at term. might also be involved.
It is important to point out that each of these fetal sources As described previously, near the end of gestation, the
of amniotic fluid also contains components of the muco- water and soluble proteins in amniotic fluid are rapidly
sal immune system. Thus, amniotic fluid may represent, to cleared. That much of this fluid is removed by the fetus is
some extent, a mixture of the fetal mucosal secretions. indicated by the relatively low rate of clearance when the
There are also several potential avenues by which mater- fetus was dead: 10–15% per day versus 90% for a live fetus
nally derived fluid and specific solutes can reach the amni- during labor (Gitlin et al., 1972). The low recovery of radio-
otic fluid. These include the fetal surface of the placenta labeled proteins in either the maternal or the fetal serum at a
and the uterine decidua. The contribution of the latter is time when a large proportion of the label had been removed
thought to be limited by the limited maternal vascularity from the chorioamniotic sac seems to eliminate absorption
of the decidua in proximity to the chorion laeve and the of the proteins through the fetal respiratory tract, umbilical
mature appearance of the amniotic epithelium at that site cord, or skin, as well as the maternal decidua. This leaves
(­Lotgering and Wallenburg, 1986). By contrast, the amniotic the fetal gastrointestinal tract as the most likely site for
epithelial cells overlying the chorionic plate have a unique, extensive clearance of amniotic fluid. This proposition is
active-appearing morphology, as described previously. In in keeping with the observation that atresia at several sites
addition, the maternal circulation in the intervillus spaces in the infant’s upper gastrointestinal tract can result in an
below is rich. Two forms of water exchange across the cho- accumulation of excessive amniotic fluid, known as poly-
rion and amnion have been described: fast nondiffusional hydramnios. Deglutition begins near the end of the first tri-
bulk flow through intercellular channels and an inherently mester of gestation. Gitlin et al. (1972) concluded that most
slower diffusion mechanism (Lotgering and Wallenburg, of the proteins in the swallowed amniotic fluid are hydro-
1986). While these mechanisms are highly permeable to lyzed and absorbed into the infant’s circulation since a large
water, small solutes cross by simple diffusion; larger com- fraction of the radiolabel was recovered in the mother’s
pounds (1000 mol wt) do not cross these membranes well, if urine, disassociated from the proteins, 1–2 days after the
at all. The driving forces for this transport are differences in amniotic injection.
hydraulic and osmotic pressures. The water channel, aquaporin-8, is expressed in human
amnion, chorion, and placenta. Aquaporin-8 gene expres-
sion was demonstrated in epithelial cells of chorion and
METABOLISM OF AMNIOTIC FLUID amnion and of the syncytiotrophoblasts and outer layer
The composition of AF changes with gestational age. In trophoblasts of placenta. These findings suggest that aqua-
the second half of pregnancy, there is a decrease in sodium porin-8 may mediate amniotic fluid resorption through the
and chloride concentrations, an increase in urea and creati- intramembranous pathway (Wang et al., 2001). This path-
nine concentrations, and an overall decrease in AF osmo- way of amniotic fluid absorption could be important in
larity. Many studies suggest that AF composition is more regulating amniotic fluid volume homeostasis.
highly regulated than AF volume. Metabolite profiles for
second and third trimester amniotic fluid samples, using IMMUNOLOGICAL FACTORS IN AMNIOTIC
high-resolution 1H NMR spectroscopy, showed signifi-
cant differences with gestational age (Groenen et al., 2004;
FLUID
Cohn et al., 2009; Graca et al., 2010). The concentrations A number of immune factors have been identified and in
of creatinine and betaine increased while concentrations of some cases monitored in the amniotic fluid during the prog-
creatine, glucose, and various amino acids decreased with ress of pregnancy. The precise functions of most of these
Amniotic Fluid and the Fetal Mucosal Immune System Chapter | 115  2257

factors in amniotic fluid are not known. However, on the (Izban at al. 2012). DAF is a key complement regulator at
basis of functions of the same factors at other sites, it is the C3 level and is thought to be unique in its responsive-
likely that immune-related factors in amniotic fluid play a ness to progesterone (Kaul et al., 1994). One hypothesis is
role in the defense of the fetal and uterine structures against that a decrease in the DAF/complement ratio could release
infection, signaling the uterus that the fetus has sustained the complement cascade. Under those circumstances, even
an infection, initiating parturition, and/or modulating the minor activation of complement in the fetomaternal com-
maternal immune response to prevent rejection of the fetus. partment could result in proinflammatory responses, acti-
In the following section we summarize immune factors that vation of the prostaglandin cascade, and precipitation of
participate in the innate immune system, soluble immune labor (Nowicki et al. 2013).
modulators and growth factors, and immunoglobulins and Another complement-modulating factor is CD59, the
their receptors/transporters. plasma membrane attack complex inhibitor protein that
has been demonstrated on the apical surface of the syn-
cytiotrophoblast, on extravillous cytotrophoblast, and in
INNATE IMMUNITY amniotic epithelium (Vanderpuye et al., 1993). During
pregnancy the level of CD59 also increases by 50% in
Complement and Complement Inhibitors maternal plasma. CD59 could help protect extraembry-
A fetus is semiallogeneic tissue, resembling a transplanted onic epithelia from damage by complement in maternal
allograft that must be protected from maternal complement blood and in amniotic fluid. The twofold to sixfold higher
attack. Amniotic fluid contains a potentially functional expression in the syncytiotrophoblast than in erythrocytes
complement system, which gradually increases in concen- and the apical location of CD59 may reflect the immu-
tration during gestation. At term, hemolytic complement nological roles and functional polarization of plasma
activity (CH50) exceeds that of maternal serum by 60% membranes in the syncytiotrophoblast (Vanderpuye et al.,
(Huffaker et al., 1989). While the function of complement 1993). CD59 is secreted by human amniotic epithelial
in amniotic fluid is presumably to provide protection from cells into amniotic fluid and could be incorporated into
infection, nonlethal doses of the complement membrane target cells. This mechanism may protect fetal cells from
attack complex can stimulate prostaglandin E2 produc- lysis by maternal complement.
tion in a number of cell types (Daniels et al., 1990; Hansch Amniotic fluid complement factor C3 can be used in
et al., 1988; Rooney and Morgan, 1990a), and an increase the diagnosis of intraamniotic infection in preterm labor
in prostaglandins has been implicated in the onset of both with intact membranes. Assays for C3 are readily avail-
term and preterm labor. Therefore, activation of amniotic able, and their results compare favorably with other rapid
fluid complement could potentially participate in the nor- markers such as white blood cells, lactate dehydrogenase,
mal process of parturition or precipitate the onset of pre- and glucose as an indicator of amniotic fluid infection. This
mature labor. finding supports the general concept of fetal inflammatory
Cytotoxic alloantibodies are present in the plasma of responses to microbial invasion of amniotic fluid (Elimian
some pregnant women, and IgG and IgM are present in et al., 1998).
amniotic fluid (Quan et al., 1999). Therefore, the poten- Mannan-binding protein (MBP), as well as other mem-
tial exists for activation of amniotic fluid complement bers of the collectin family, including complement protein
by the classical pathway. The alternative pathway could C1q, lung surfactant protein A, CL-43, and conglutinin,
be activated by intrauterine infection and might be caus- has been shown to interact with a single C1q receptor, also
ally associated with premature labor. However, human termed collectin receptor (Stuart et al., 1997) This recep-
amniotic epithelial cells express decay accelerating fac- tor, which is widely distributed in different types of cells,
tor (DAF), which can protect against complement toxicity together with collectins plays an important role in innate
(Rooney and Morgan, 1990b; Pacheco et al. 2011). DAF immunity.
is a complement-modulating factor that is also expressed MBP is a serum protein that can activate the classi-
in the fetomaternal interface and has been implicated in cal complement pathway. MBP has also been shown to
protection of the fetus from rejection (Rooney and Morgan opsonize bacteria and enhance clearance of bacteria by
1990b). The expression of DAF on human trophoblast phagocytosis. MBP is present in amniotic fluid, and its con-
membranes may protect them from maternal comple- centration increases sharply from about 32 weeks of ges-
ment-mediated damage in response to microbial infection tation. MBP may play a role in the antibody-independent
(Nishikori et al., 1993; Jensen et al., 1995; Fenichel et al., recognition and clearance of pathogens in the amniotic cav-
1995). Our recent results support the notion that there are ity toward term (Malhotra et al., 1994). It is interesting that
inherent differences in immunological responsiveness at 32 weeks the CH50 of amniotic fluids also increases and
between individuals and that the vitamin D3 system may exceeds significantly that of cord serum but is lower than
be important in sustaining LPS-induced CD55 expression the CH50 of maternal sera.
2258  SECTION | I  Genitourinary Tract and Mammary Gland

CYTOKINES, CHEMOKINES, AND OTHER these factors can often be detected in fluids near the site
MODULATORY AND GROWTH FACTORS of production or can reach distant sites via the circula-
tion. Those mediators that have been detected in amni-
These small-molecular-weight peptides are usually pro- otic fluid are shown in Table 1. Unfortunately, the results
duced by one or more cell types in various tissues and act of such assays on amniotic fluid do not indicate which
as messengers to induce various activities in nearby cells cells are responsible for their production. These might
or the producing cell itself. By sensitive immunoassay, include cells of the fetus, fetal membranes, or placenta

TABLE 1  Cytokines, Chemokines, Growth Factors, and Prostaglandins in Amniotic Fluid

Gestational Age, Concentration, pg/mL


Weeks (Sample (Proportion Detectable,
Name Number) or Range) Alteration with References
IL-1β/IL-1RA 13–19 (4) 90.9 ± 26.8 – Srivastava et al. (1996)
IL-1RA 13–19 (11) 2835 ± 1775 – Srivastava et al. (1996)
IL-2 13–19 (13) 0.9 ± 3 (5/13) – Srivastava et al. (1996)
S IL-2R 13–19 (11) 609 ± 391 (8/11) – Srivastava et al. (1996)
IL-4 13–19 (14) 21 ± 47 (8/14) – Srivastava et al. (1996)
IL-6 13–19 (14) 3407 ± 236 – Srivastava et al. (1996)
Midtrimester to term 703 ± 401 No labor Santhanam et al. (1991)
2923 ± 5638 Labor
16,904 ± 9343 Labor plus infection
15–21 (22) 560 ± 1071 Weimann et al. (1995)
IL-8 13–19 (14) 2825 ± 690 Srivastava et al. (1996)
14–41 (80) <1800 Intraamniotic infection, Puchner et al. (1993)
>10,000 (9/12)
IL-6 and IL-8 Increase of IL-6 and Fetal chromosomal Vesce et al. (2002)
decrease of IL-8 abnormality
IL-10 13–19 (14) 24 ± 40 (8/14) Srivastava et al. (1996)
Midtrimester (66) <40 (<40–476) SGA > 40, 78 (<40–832) Heyborne et al. (1994)
Term (119) 20 Intraamniotic infection in Greig et al. (1995)
labor, 54 (0–759)
TNFα 13–19 (14) 183 ± 205 – Srivastava et al. (1996)
TNFα-active 14–20 (59) 4 Small for gestational Heyborne et al. (1992)
age, 10.5
INFα 13–19 (14) 11 ± 18 (4/14) – Srivastava et al. (1996)
GM-CSF 13–19 (8) 9 ± 14 (5/7) – Srivastava et al. (1996)
15–21 (22) <10 – Weimann et al. (1995)
G-CSF 15–21 (22) 1396 ± 2073 – Weimann et al. (1995)
HB-EGF Late pregnancy Rapid increase Not altered by infection Michalsky et al. (2002)
TGF-β1
Active 13–19 (11) 2.8 ± 2.9 – Srivastava et al. (1996)
Latent 13–19 (13) 167 ± 106 – Srivastava et al. (1996)

Continued
Amniotic Fluid and the Fetal Mucosal Immune System Chapter | 115  2259

TABLE 1  Cytokines, Chemokines, Growth Factors, and Prostaglandins in Amniotic Fluid—cont’d

Gestational Age, Concentration, pg/mL


Weeks (Sample (Proportion Detectable,
Name Number) or Range) Alteration with References
TGF-β1
Active 14–16 (12) 540 ± 60 – Lang and Searle (1994)
TGF-β2
Active 13–19 (14) 10 ± 11 – Srivastava et al. (1996)
Latent 13–19 (13) 1178 ± 469 – Srivastava et al. (1996)
TGF-β2
Active 14–16 (12) 2300 ± 400 – Lang and Searle (1994)
MIP1-α 13–19 (14) 241 ± 402 – Srivastava et al. (1996)
GRO-α Midtrimester (18) 1100 (400–5000) Intraamniotic infection Cohen et al. (1996)
Term (20) 1900 (1200–4200) 2700 (1400–12,700) Cohen et al. (1996)
Preterm (20) 2300 (500–10,000) Intraamniotic infection Cohen et al. (1996)
5000 (600–47,900) Cohen et al. (1996)
MMPase-9 Intraamniotic infection Harirah et al. (2002)
Stem cell factor 13–9 (14) 1047 ± 383 (13.6–541.9 ng/mL) Srivastava et al. (1996)
SLPI Second trimester (15) 106 ± 15 pg/mL Zhang et al. (2001)
Third trimester (12) 802 ± 138 pg/mL
Prostaglandin 444 ± 78 pg/mL Intraamniotic infection Hillier et al. (1993);
(2.238 ± 434 pg/mL)

SGA, small for gestational age; HB-EGF, heparin-binding EGF-like growth factor; MMPase-9, matrix methalloproteinase-9; SLPI, secretory leukocyte protease
inhibitor.

or cells from maternal tissues that transit across the fetal milk, which also contains numerous cytokines, chemokines,
tissues. and growth factors.
Interest in cytokines, chemokines, growth factors, and The possibility that these milk constituents may alter
prostaglandins in amniotic fluid has been stimulated by the the development or immune function of the infant is dis-
possibility that they are involved in the control of parturi- cussed elsewhere in this book (see Chapter 72). The idea
tion. Enhanced production of cytokines and chemokines that some of these factors are produced at the mucosal sur-
can be induced by various stimuli, particularly infections. faces of the infant and provide important signals to other
Thus, increased mediator levels have been investigated as fetal tissues or to the uterus or placenta should also be con-
a potential explanation for premature onset of labor due sidered.
to intraamniotic infections (Romero et al., 1989). More
recently, the potential effects of potent mediators on the
Cytokines
growth and welfare of the fetus and unborn infant have
begun to be evaluated (Heyborne et al., 1992, 1994). Some The function of cytokines in normal pregnancy and deliv-
of the amniotic fluid cytokines, their concentrations, and ery is not fully established. The cytokines IL-1, IL-6, and
alterations in intraamniotic infection and growth retardation TNFα, as well as epidermal growth factor (EGF), have
are shown in Table 1. been detected in human amniotic fluid before and dur-
To date, there has been little discussion about the specific ing labor (Table 1). Amniotic fluid mean values of IL-1β,
effects of the various cytokines, chemokines, and growth IL-6, TNFα, IFNγ, and EGF were approximately 4, 140,
factors demonstrated in amniotic fluid on the development 4, and 3 times those of maternal serum values, respec-
of the mucosal immune system of the infant. However, one tively. Thus, it seems likely that the fetus and/or placenta
might draw an analogy between amniotic fluid and maternal contributes to the production of the cytokines in amniotic
2260  SECTION | I  Genitourinary Tract and Mammary Gland

fluid. This supposition is in agreement with Menon et al. fact, the LBP and sCD14 concentrations in amniotic fluid of
(2001), who reported that amniochorionic membranes are women in preterm labor are linearly associated with amni-
a site of inflammatory cytokine production. Increases in the otic fluid IL-6 concentrations (Gardella et al., 2001).
concentration of cytokines in amniotic fluid may represent Adrenomedullin, a novel hypotensive peptide with pos-
enhanced macrophage activity for immunosurveillance of sible immunological function, was discovered in human
the fetus. In addition, the elevation of amniotic fluid cyto- pheochromocytoma (Kitamura et al., 1993). In humans,
kines may be a preparatory step in the initiation of labor, adrenomedullin was detected in amniotic fluid and plasma
since cytokines play an important role in the stimulation of as well as in many tissues, such as adrenal medulla, aorta,
prostaglandin synthesis. heart, lung, brain, and kidney (Macri et al., 1996). The
An increase of IL-6 (P = 0.034) and a decrease of IL-8 physiological function of increased adrenomedullin during
(P ≤ 0.0001) in amniotic fluid, as well as a decrease of IL-6 pregnancy is still not established, although it was suggested
in the maternal plasma (P = 0.026), were associated with that adrenomedullin plays an important role in the adapta-
pregnancies with fetal chromosomal abnormalities (Vesce tion of the vascular system to pregnancy needs and may
et al., 2002). regulate placental vascular tone.
Heparin-binding EGF-like growth factor (HB-EGF) is Plasma adrenomedullin concentrations increase with
a member of the EGF family that has been implicated in advancing gestation and are highest in the second trimester
the healing of various organ injuries. HB-EGF production of pregnancy. Adrenomedullin concentrations in amniotic
is upregulated in response to injury to the kidney, liver, fluid are significantly higher than in the maternal plasma
brain, skin, and intestine. Recently HB-EGF was detected throughout gestation and also increase with advancing
in human amniotic fluid and breast milk. The HB-EGF in gestation. There is a significant linear correlation between
amniotic fluid may play a role in the development of the amniotic fluid and maternal plasma adrenomedullin during
gastrointestinal tract in utero, whereas that in breast milk pregnancy. Adrenomedullin mRNA was detected not only
may protect gut mucosa against postnatal injury (Michalsky in the amniotic membranes and intermediate trophoblast
et al., 2002). of third trimester pregnancy but also within the endothelial
Amniotic fluid matrix metalloproteinase-9 and IL-6 layers of villous blood vessels (Kanenishi et al., 2001).
are significantly elevated in women with intraamniotic
infection (Harirah et al., 2002), and the levels of matrix
NONSPECIFIC ANTIMICROBIAL FACTORS
metalloproteinase-9 are correlated with IL-6 (r = 0.813,
P < 0.001); both are positively correlated with amniotic The inability of amniotic fluid to support the growth of dif-
fluid leukocytes and inversely correlated with amniotic ferent bacteria was noted a long time ago. It was not clear
fluid glucose (­Harirah et al., 2002). Therefore, amniotic whether this was due to an antimicrobial activity or to a
fluid matrix metalloproteinase-9 has been proposed as an deficiency of one or more nutrients. Amniotic fluid sup-
accurate biochemical marker of intraamniotic infection plemented by bacterial growth media was shown to allow
with better sensitivity, specificity, and positive and negative growth of bacteria. However, when the constituents of the
predictive values than IL-6 (Harirah et al., 2002). Elevated growth media were added individually, only phosphate was
matrix metalloproteinase-8 (>23 ng/mL) in amniotic fluid required (Larsen et al., 1974). Subsequently, Schlievert
is also a powerful predictor of spontaneous preterm deliv- et al. (1976) found that phosphate inhibits an antimicrobial
ery (<32 weeks), with an odds ratio of 68.4. Thus, amniotic property of amniotic fluid that includes a hexapeptide and
fluid studies can be used to improve the risk assessment zinc. The activity of the hexapeptide was inhibited by phos-
for preterm delivery in women who undergo midtrimester phate. Antimicrobial activity against Escherichia coli was
amniocentesis for genetic indications (Yoon et al., 2001). totally lost after addition of phosphate, suggesting that the
Secretory leukocyte protease inhibitor (SLPI), a potent hexapeptide was a major antimicrobial agent against this
inhibitor of human leukocyte elastase, is a protein found bacterium.
in various human fluids, including amniotic fluid, cervical A number of the antimicrobial substances found in
mucus, seminal plasma, and ascites. SLPI is produced by amniotic fluid are also present in other mucosal secretions,
amniotic membranes in response to cytokines (Zhang et al., including milk (see also Chapter 15). Several of these com-
2001). The SLPI in the amniotic fluid may contribute to ponents are presumably produced by epithelial cells of the
immune mechanisms during pregnancy (Zhang et al., 2001). fetus or by amniotic epithelial cells. We therefore examined
The binding of lipopolysaccharide (LPS) with LPS bind- the relationship among the concentrations of fSC, lactofer-
ing protein (LBP) and sCD14 activates macrophages at LPS rin, and lysozyme (Cleveland et al., 1991a). fSC is pro-
concentrations up to 1000 times lower than those required duced by secretory epithelial cells and serves as a marker
with LPS alone. The presence of LBP and sCD14 in amni- for sites of polymeric Ig transport (see also Chapter 20). As
otic fluid might explain the high concentrations of cytokines in the case of fSC, lactoferrin and lysozyme concentrations
present in the amniotic fluid of culture-negative women. In increase in the amniotic fluid with increasing gestational
Amniotic Fluid and the Fetal Mucosal Immune System Chapter | 115  2261

age. We also found that the concentrations of both lactofer- due to its limited concentration in the interstitium of the
rin and lysozyme are correlated with fSC concentrations in decidua or to lack of transport across the chorionic mem-
amniotic fluid and that the concentrations of lactoferrin and brane. C3 has a size and serum concentration in the same
lysozyme are correlated with each other (Cleveland et al., range as that of monomeric IgA. However, the inability of
1991a). This suggests that both these factors are produced Jauniaux et al. (1995) to detect similar amounts of C3 as IgA
by the same cells or that the cells that produce them are in chorionic fluid could have been due to insensitivity of the
under similar regulatory influences. C3 assay used or selective transport of IgA. Specific trans-
Defensins are nonspecific antimicrobial factors that porters for IgG and IgA are known to be produced by the
may be important in the early defense against infection (see fetus. One of them, the polymeric immunoglobulin receptor
also Chapter 16). Defensins are neutrophil peptides in the (pIgR), has been identified by immunofluorescence in the
azurophilic granules released from activated neutrophils fetal membranes (Tourville et al., 1969). The neonatal Fcγ
­(Faurschou et al., 2002). Defensins can eliminate microor- receptor (FcRn) is expressed in the syncytiotrophoblasts of
ganisms by forming pores in their plasma membrane (Bals, the placenta (Leach et al., 1996), but to our knowledge its
2000). Amniotic fluid defensin levels measured by enzyme- presence in the chorioamniotic membranes has not been
linked immunoabsorbent assay were fourfold to 24-fold reported. However, Bright and Ockleford (1995) did not
higher in patients with intrauterine infection than in normal find evidence of endogenous IgG staining within either the
preterm and term controls. An amniotic fluid defensin level of cytotrophoblast or the viable amniotic epithelial cells of the
400–2500 ng/mL identified over 80% of patients with a pos- chorioamniotic membrane. IgG was detected throughout
itive amniotic fluid culture. Amniotic fluid defensin levels the interstitium of the decidua, chorion, and amnion and in
also identify accurately patients with subclinical intrauter- dying amniotic epithelial cells. They concluded that, at least
ine infection, as defined by placental histologic findings and late in gestation, the entry of IgG into the amniotic cavity is
positive amniotic fluid culture. It was also suggested that by passive diffusion.
the defensin level may contribute to the increased risk of We have quantified albumin and each of the major Ig
spastic cerebral palsy and other fetal morbidity in newborns classes in amniotic fluids collected by amniocentesis for
from patients with intrauterine infection. It is interesting genetic analysis and determination of fetal maturity (Hilton,
to note that labor alone was not associated with elevation 1993). Figure 3 demonstrates the albumin concentration in
of the defensin level. Therefore, it was also suggested that amniotic fluid as a function of gestational age. The progres-
amniotic fluid defensin levels could be another marker of sive fall during gestation is similar to that described by oth-
subclinical intrauterine infection (Heine et al., 1998; Balu ers for total protein concentration. In contrast to the pattern
et al., 2002). for albumin, the concentrations of each class of Ig in amni-
otic fluid did not seem to vary significantly with gestational
age, as shown in Figure 4. When concentrations of amniotic
HUMORAL IMMUNITY fluid proteins are considered in relation to the concentrations
Immunoglobulins and Immunoglobulin
Transporters 4000
During pregnancy, the maternal immune response is gen- y = 3324.6 – 49.562x R2 = 0.363
erally modulated toward humoral immunity (Stirrat, 1994;
Jensen et al., 1995). Activated lymphocytes during murine 3000
pregnancy produce cytokines that preferentially stimulate
Albumin, µg/ml

antibody production versus T cell cytotoxic responses. This


effect is most prominent within the uterine decidua but also 2000
affects systemic immunity. The discussion below consid-
ers the maternal and fetal adaptive immune responses, with
emphasis on humoral immune factors and their receptors in
1000
amniotic fluid.
The sources of the immunoglobulins in the chorionic
and amniotic fluids are known in some detail, but the selec-
tivity and mechanisms of their transport are not yet clear. 0
10 20 30 40 50
As described previously, Jauniaux et al. (1995) found that Gestational age, weeks
the gradient between maternal serum and chorionic fluid
FIGURE 3  Albumin concentration in amniotic fluid versus gesta-
was greater for IgA than for IgG and that neither IgM nor tional age. The concentration of albumin was measured by nephelometry
C3 were detected in this fluid. The absence of detectable in 31 amniotic fluid samples. Gestational age was calculated from the last
amounts of the larger IgM molecules in this fluid might be menstrual period. Reprinted with permission from Hilton (1993).
2262  SECTION | I  Genitourinary Tract and Mammary Gland

D 600 100
y = 2.2063e-2 x 10 (61783e-2x) Rz = 0.658

500

10
400
IgG, Pg/mL

fSC, µg/mL
300

1
200

100
10 20 30 40 50
.1
Gestational age, weeks 10 20 30 40 50
Gestational age, weeks
FIGURE 5  fSC concentration in amniotic fluid versus gestational
40
E age. fSC in amniotic fluid was quantified by ELISA. fSC concentration
was significantly correlated with gestational age.

30
of these same proteins in the maternal serum, albumin and
IgG were of the same magnitude (approximately 1/10–1/30
IgA, Pg/mL

20
of maternal serum concentration). However, the ratios of
amniotic IgA and IgM to maternal serum concentrations
were much lower (1/100 and 1/1000, respectively). The lat-
10 ter finding is consistent with later reports on chorionic fluid
(Jauniaux et al., 1995).
Cederqvist et al. (1978) have stated that the level of IgA
0 in amniotic fluid increases until midgestation and decreases
10 20 30 40 50 thereafter. Because there were so few samples available
Gestational age, weeks between weeks 20 and 30 of gestation, such a pattern is dif-
ficult to discern from their data or our own (Figure 4). Given
the decreasing concentration of albumin during gestation
F 10 and the data of Gitlin et al. (1972) that all radiolabeled pro-
teins are cleared from the amniotic fluid at a similar rate,
it seems likely that there is more than one source of IgA in
the amniotic fluid. The extent to which nonspecific diffu-
1 sion and receptor-mediated transport of maternal IgA and
IgM, Pg/mL

the fetal mucosa contribute to amniotic IgA concentration


may vary with gestational age. This issue will be considered
further in the context of the structure of the amniotic IgA.
.1
fSC, the cleaved extracellular portion of the pIgR
expressed in secretory epithelium, has also been quanti-
fied in a set of amniotic fluid samples (Cleveland et al.,
1991a,b; Hilton, 1993). Figure 5 shows the relationship
.01
10 20 30 40 50 between fSC concentration and gestational age (Hilton,
Gestational age, weeks 1993). There is a strong positive correlation between fSC
FIGURE 4  Ig concentration in amniotic fluid versus gestational age. and gestational age from about 15 or 20–40 weeks of gesta-
(a) The concentration of IgG was measured by nephelometry. (b) The con- tion. As discussed earlier, the anatomical site of production
centration of IgA was measured by ELISA. (c) The concentration of IgM of most of the fSC in amniotic fluid is not known. In most
was determined by ELISA. The number of samples was 31 for each of the biological solutions, some of the SC is found complexed
Ig classes. There was no significant correlation between the concentration
with its ligands, polymeric immunoglobulins (IgA and
of any Ig class and gestational age. Reprinted with permission from Hilton
(1993). IgM), which have been transported across these epithelial
cells. After proteolytic cleavage of the pIgR from the api-
cal surface of the epithelial cell, the complexes (Ig-SC) are
Amniotic Fluid and the Fetal Mucosal Immune System Chapter | 115  2263

called secretory immunoglobulins (e.g., S-IgA). Approxi- The ability of Igs in the amniotic fluid to protect the
mately 80% of the amniotic fluids that we have examined developing fetus from infection is difficult to ascertain.
contained small amounts of S-IgA of typical size (390 kDa), Jauniaux et al. (1995) detected specific antibodies against
as indicated by Western blots stained with anti-α chain and Toxoplasma, cytomegalovirus, and rubella virus in cho-
anti-SC antibodies (Cleveland et al., 1991b). This finding rionic fluids as early as 9 weeks of gestation. These anti-
suggests that this portion of the amniotic IgA is derived bodies were present only when the maternal serum had
from epithelial cell transport of dimeric IgA molecules. No antibodies to the same agents. The ratios of maternal serum/
S-IgA molecules of this size were detectable in unconcen- chorionic fluid titers varied from 5:1 to 32:1, consistent
trated urine collected from the first postnatal void. How- with their mean ratio of 28:1 for total IgG concentration.
ever, high-­molecular-weight bands consistent with 390-kDa Mellander et al. (1986) measured antibodies in amniotic
S-IgA were recovered from lysates of amniotic membranes fluid and found that a minority of samples contained IgA
and from the supernatants of short-term cultures of washed antibodies against E. coli surface antigen and poliovirus.
amniotic membranes (Cleveland et al., 1991b). This sug- IgG and IgA antibodies to Candida have been detected
gests that, at least near the end of gestation, some—presum- in the majority of amniotic fluids as early as 15–18 weeks
ably maternal—IgA is actively transported into the amniotic of gestation by two different groups (Auger et al., 1980;
fluid by amniotic epithelial cells. This maternal IgA may Mathai et al., 1991). Interestingly, in contrast to Candida
be produced locally in the decidua, since Cederqvist antibodies in serum, where IgG predominates, IgA antibod-
et al. (1978) reported that up to one-half of the IgA in amni- ies predominated in the amniotic fluid. This suggested that
otic fluid from 11 to 40 weeks of gestation was of the IgA2 these antibodies were not derived from maternal serum. It
isotype. Normally, only about 10% of serum IgA is IgA2, is interesting to speculate that these antibodies might have
whereas IgA produced at mucosal sites contains a larger been derived from maternal secretions (cervical or vaginal)
proportion of IgA2, depending on the site. and transported into the amniotic fluid.
Western blotting also demonstrated the presence in all Blanco et al. (1983) found that amniotic fluid from 46
amniotic fluids examined of bands in the 200-kDa range, patients soon after the clinical onset of bacterial intraamni-
which stained with polyclonal and monoclonal antibodies otic infection had slightly higher IgG concentrations than
against α chain and SC (Cleveland et al., 1991a). Similar- those from 46 matched controls. However, they did not
sized bands were also seen in the first postnatal urine sam- determine whether there was a selective increase in spe-
ples and in lysates and supernatants from cultured amniotic cific antibodies against the infecting organisms. Thus, the
membranes. A preliminary characterization of these unusual increased IgG noted may have been due to increased trans-
IgA molecules indicated that they contain full-length α and port of total IgG into the amniotic fluid. However, another
light chains, suggesting that they are formed from the 390- group has reported that bacteria from amniotic fluid of
kDa molecules by dissociation of interchain disulfide bonds. women with chorioamnionitis were frequently coated with
Subsequent studies (Hilton, 1993) examined various meth- antibodies (Moller et al., 1997).
ods for isolating and analyzing the small molecular form
of IgA from amniotic fluid. After examination by several INCREASED ACTIVITY OF THE IMMUNE
different techniques, it seemed clear that these molecules
did not contain substantial amounts of the J chain present
SYSTEM IN AFRICAN AMERICANS
in typical 390-kDa S-IgA. It was also clear that not all the Although there are currently no known markers for identi-
IgA molecules of this size contained SC. When the approxi- fying differences in immunity and inflammation in amni-
mately equal amounts of 390- and 200-kDa forms were otic fluid in African Americans and Caucasians, these could
compared by several analytical techniques, SC could be be important since the incidence of preterm labor is much
detected in only the 390-kDa forms. However, the amount higher among African Americans. This may be correlated
and purity of the 200-kDa forms available for analysis were with the higher serum Ig levels, particularly IgG, in African
not adequate to allow detection of SC if it was present on Americans. This difference presumably reflects increased
only a subset of these molecules. It is likely that the prelimi- B-cell activation (Tollerud et al., 1995). Serum IgG was
nary observations of Cleveland et al. (1991b) were based on directly related to human leukocyte antigen (HLA)-DR
a population of 200-kDa IgA molecules that were enriched and the level of soluble IL-2 receptors (sIL-2R) and was
in their SC content by isolation with the use of monoclo- inversely related to the proportion of CD4 cells (Tollerud
nal anti-S-IgA affinity methods. Despite the presence of et al., 1995). African Americans exhibit increased expres-
this IgA form in the urine of newborns, the low concentra- sion of the costimulatory molecules CD80 (B7-1) and CD86
tion of urinary IgA at this time (<0.2 μg/mL) (­Gitlin et al., (B7-2) (Hutchings et al., 1999). Costimulatory interactions
1972) and the rapid clearance rate of amniotic fluid make it have been shown to be important for the production of the
unlikely that fetal urine is a major source of even the small proinflammatory cytokines TNFα and IL-1β (May et al.,
amount of IgA found in late-gestation amniotic fluid. 2000). Individuals with mutations at the TNF promoter
2264  SECTION | I  Genitourinary Tract and Mammary Gland

TNF(-238A) and TNF(-376A/-238A) consistently have E. coli bearing class III P adhesin—but not the related class
lower plasma levels of IL-10 than TNF. In children with I or II adhesins—was associated with a twofold increase in
severe malaria, TNF promoter variants influence the bal- the risk of preterm delivery. Clinical symptoms of amni-
ance of IL-10 to TNF in the plasma, which in turn affects otic fluid infection at term occur 1.5 times more often in
the outcome of clinical complications (May et al., 2000). patients with bacterial vaginosis than in those without (Sil-
Altogether, African Americans appear to have inherently ver et al., 1989). A recent clinical study provides evidence
higher levels of selected components of humoral immunity that treatment of bacterial vaginosis, or even asymptomatic
and, therefore, an increased potential for complement and abnormal vaginal colonization, with oral clindamycin early
anti-inflammatory responses in plasma. We know of no in the second trimester significantly reduces the incidence
studies comparing humoral immune factors in the amniotic of preterm deliveries (Ugwumadu et al., 2003).
fluid of African Americans with other groups. Amniotic fluid infection is strongly associated with
the presence of a cytokine response in the amniotic fluid
(Romero et al., 1989; Hillier et al., 1993). Cytokine pro-
PATHOLOGICAL CONDITIONS duction by macrophages is an immunological response
INVOLVING AMNIOTIC FLUID to pathogens that invade the amniotic fluid. The cytokine
concentration in the amniotic fluid of infected patients can
Chorioamnionitis
be even higher than the concentration of cytokines in the
Chorioamnionitis (amniotic fluid infection) is usually asso- cerebrospinal fluid of children during meningitis or in the
ciated with fever, purulent amniotic fluid (with bacterial peritoneal fluid of patients during peritonitis (Mustafa et al.,
contamination) and tachycardia (maternal or fetal), and/or 1989; Zeni et al., 1993; Hillier et al., 1993; Romero et al.,
increased maternal peripheral leukocyte counts. The fact that 1993; Greig, 1993). Prostaglandin E2 has also been associ-
bacterial cultures of amniotic fluid were positive for 75% of ated with amniotic fluid infection. Although it is not clear
women whose newborns weighed less than 1500 g provides whether the macrophages that produce cytokines are of
evidence that amniotic fluid infection is a major risk factor maternal or fetal origin, it is known that cytokine levels in
for preterm delivery (labor) (Hitchcock et al., 1999). amniotic fluid are correlated with levels of prostaglandins.
Bacterial vaginosis is a risk factor for amniotic fluid Granulocyte colony stimulating factor (G-CSF) can
infection. The presence of several vaginal organisms in the be detected in amniotic fluid. Amniotic fluid G-CSF con-
second trimester of pregnancy has been linked to the devel- centrations are similar in preterm and term labor and are
opment of amniotic fluid infection. Women vaginally colo- not significantly influenced by labor, although intraamni-
nized by Gardnerella vaginalis, Bacteroides species, and otic infection is associated with increased concentrations
Mycoplasma hominis have more than a twofold increased of G-CSF in amniotic fluid, neonatal urine, and maternal
risk for amniotic fluid infection, compared with women not serum (Calhoun et al., 2001).
colonized by any of these organisms (Krohn et al., 1991). The S100 proteins are a family of low-molecular-weight
High vaginal IL-8 concentration, high neutrophil count, (10–14 kDa) calcium-binding multifunctional proteins with
abnormal vaginal gram stain, absence of hydrogen perox- α and β subunits. They are present in the cytosol, intracel-
ide-producing Lactobacillus, and anaerobic vaginal flora lularly and extracellularly, and via binding to a target pro-
are strongly associated with amniotic fluid infection among tein may trigger processes activated by the calcium signal
women in preterm labor (Hitti et al., 2001). transduction pathways important in cellular differentiation,
Most pathogens, including E. coli, develop unique viru- progression, and inflammation. Some pathological condi-
lence mechanisms that allow them to colonize and invade tions during pregnancy related to hypoxic stress can affect
the urogenital tract (Goluszko et al., 2001; Das et al. 2005; the elevation of S100B concentration in the amnion. There
Wroblewska-Seniuk et al. 2005; Rice et al. 2006; Nowicki have been several studies investigating the correlations
et al. 2010; Sledzinska et al. 2010). Bacterial adhesins inter- among the S100B concentrations in the amniotic fluid, cord
act with tissue receptors (Samet et al. 2013a; Samet et al. blood, and maternal serum during pregnancy and pathologi-
2013b; Banadakoppa et al. 2014; Rana et al. 2014a; Rana cal conditions related to fetuses have been suggested in pre-
et al. 2014b), allowing ascending infection that triggers term delivery (Gazzolo et al., 2000), intrauterine fetal death
TNF, IL-6, IL-8, and NO responses (Hedlund et al., 2001; (Florio et al., 2004), pregnancy-related hypertensive disor-
Frendeus et al., 2001; Fang et al. 2004). Lack of bacterial ders (Schmidt et al., 2004), intrauteral growth restriction
attachment or prevention of colonization blocks signaling (IUGR) cases, and preeclampsia (Tskitishvili et al., 2006).
and the host cytokine response. Because only certain genet- The amniotic fluid S100B protein concentration of the
ically related pathogens (not all bacteria) cause infection, preeclampsia and normotensive IUGR cases was signifi-
the identification of genetic and virulence factors could pre- cantly higher than that of the control. This study also shows
dict an association with preterm delivery (Hart et al., 2001). that amnion could be a source responsible for the increased
For example, we found that vaginal colonization with concentration of S100B in amniotic fluid.
Amniotic Fluid and the Fetal Mucosal Immune System Chapter | 115  2265

Fetal Growth Restriction HIV-infected pregnant women. The lamivudine concentra-


tion in the amniotic fluid of pregnant patients who received
EGF is present in large amounts in the amniotic membranes
antiretroviral therapy was higher than that in maternal and
and fetal urine. EGF is a 53-amino acid polypeptide mito-
fetal plasma. It was postulated that lamivudine is able to
gen that stimulates tissue growth (Laborda et al., 2000).
cross the placenta by simple diffusion and is concentrated in
Receptors or binding sites for EGF have been identified in
the amniotic fluid (Mandelbrot et al., 2001). The pharmaco-
placenta and fetal membranes. Amniotic fluid EGF levels
kinetics and safety of nelfinavir when used in combination
increase rapidly in late pregnancy but are not altered by
with zidovudine and lamivudine in HIV-infected pregnant
chorioamnionitis or by term or preterm labor. Although
women have been studied (Bryson et al., 2008).
intrauterine growth restriction is associated with lower EGF
Valacyclovir has been recommended for prevention of
levels in amniotic fluid, EGF is not altered in maternal or
and therapy for herpesvirus infection and 10 years ago was
fetal plasma. EGF increases near term and decreases in
a new candidate for treatment of symptomatic herpes infec-
pregnancies complicated by intrauterine growth restriction
tion during pregnancy (Kleymann, 2003).
(Varner et al., 1996).
Studies have shown that steady-state nevirapine plasma
EGF may also influence the level of parathyroid hor-
concentrations are influenced by pregnancy (Nellen et al.,
mone-related protein (PTHrP). PTHrP may regulate pla-
2008). Maternal-fetal transfer and amniotic fluid accumula-
cental calcium flux, fetal tissue development, and uterine
tion of nucleoside analog reverse transcriptase inhibitors in
contractions. Investigations have shown that EGF (10 μg/L)
HIV-infected pregnant women provide evidence that amni-
increased PTHrP secretion by over 100% and that PTHrP
otic fluid concentrations of nucleoside reverse transcrip-
secretion by EGF was both dose and time dependent; in
tase inhibitors, such as lamivudine (3TC) and zidovudine
contrast, estradiol, progesterone, and human chorionic
(ZDV), were up to14-fold higher than blood plasma drug
gonadotropin had no effect on PTHrP secretion (Dvir et al.
concentrations (Chappuy et al., 2004).
1995). Intrauterine PTHrP concentrations are reduced in
association with growth restriction in the spontaneously
hypertensive rat (SHR) in comparison with those of its nor- REFERENCES
motensive control, the Wistar Kyoto rat, implicating PTHrP Auger, P., Marquis, G., Dallaire, L., Montplaisir, S., Ghoubril, S., 1980.
as a pivotal fetal growth factor (Wlodek et al., 2001). The Natural occurrence of a humoral response to Candida in human amni-
SHR fetus is growth-restricted and has suppressed amniotic otic fluid. Am. J. Obstet. Gynecol. 136, 1075–1079.
fluid PTHrP, which is largely determined by the fetus or Bals, R., 2000. Antimicrobial peptides and peptide antibiotics. Med. Klin.
gestational tissues and is independent of maternal hyper- 95 (9), 496–502.
Balu, R.B., Savitz, D.A., Ananth, C.V., Hartmann, K.E., Miller, W.C., Thorp,
tension or maternal PTHrP (Wlodek et al., 2001). The low
J.M., Heine, R.P., 2002. Bacterial vaginosis and vaginal fluid defensins
SHR amniotic fluid PTHrP may play a role in the growth
during pregnancy. Am. J. Obstet. Gynecol. 187 (5), 1267–1271.
restriction of the SHR fetus (Wlodek et al., 2001). Banadakoppa, M., Goluszko, P., Liebenthal, D., Nowicki, B.J., Nowicki,
Circulating S100B protein is increased in intrauterine S., Yallampalli, C., 2014. PI3K/Akt pathway restricts epithelial adhe-
growth-retarded fetuses (Gazzolo et al., 2002). Insulin-like sion of Dr+ Escherichia coli by down-regulating the expression of
growth factor binding protein-1 (IGFBP-1) is present in decay accelerating factor. Exp. Biol. Med. (239), 581–594.
higher concentrations in amniotic fluid than in serum, cer- Blanco, J.D., Gibbs, R.S., Krebs, L.F., 1983. A controlled study of amni-
vical mucus, urine, or seminal plasma (Darj and ­Lyrenas, otic fluid immunoglobulin levels in intraamniotic infection. Obstet.
1998). Therefore, detection of IGFBP-1 by the dipstick Gynecol. 61, 450–453.
test (Darj and Lyrenas, 1998) with monoclonal antibodies Brace, R.A., 1986. Amniotic fluid volume and its relationship to fetal fluid
to IGFBP-1 could be useful in the diagnosis of premature balance: Review of experimental data. Semin. Perinatol. 10, 103–112.
Bright, N.A., Ockleford, C.D., 1995. Ultrastructural distribution of endog-
rupture of membranes.
enous immunoglobulin-G in human term amniochorion. Cell Tiss.
Res. 281, 367–374.
Bryson, Y.J., Mirochnick, M., Stek, A., PACTG 353 Team, et al., 2008.
ANTIRETROVIRAL DRUGS AND AMNIOTIC
Pharmacokinetics and safety of nelfinavir when used in combination
FLUID with zidovudine and lamivudine in HIV-infected pregnant women:
The timing of mother-to-child transmission of HIV-1 and Pediatric AIDS Clinical Trials Group (PACTG) Protocol 353. HIV
Clin. Trials 9, 115–125.
implications for prevention has been studied (Kourtis et al.,
Calhoun, D.A., Chegini, N., Polliotti, B.M., Gersting, J.A., Miller, R.K.,
2006). Lamivudine is an antiretroviral drug that has been
Christensen, R.D., 2001. Granulocyte colony-stimulating factor in
widely used during pregnancy, and has been reported to preterm and term pregnancy, parturition, and intra-amniotic infection.
reduce significantly HIV-1, including zidovudine-resistant Obstet. Gynecol. 97 (2), 229–234.
strains, as well as hepatitis B virus in patients with chronic Cederqvist, L.L., Ewool, L.C., Bonsnes, R.W., Litwin, S.D., 1978. Detect-
hepatitis B infection. Therefore, lamivudine placental trans- ability and pattern of immunoglobulins in normal amniotic fluid
fer and amniotic fluid concentrations were investigated in throughout gestation. Am. J. Obstet. Gynecol. 130, 220–224.
2266  SECTION | I  Genitourinary Tract and Mammary Gland

Chappuy, H., Treluyer, J.M., Jullien, V., Dimet, J., Rey, E., Fouche, M., Gazzolo, D., Vinesi, P., Marinoni, E., Di Iorio, R., Marras, M., Lituania,
Firtion, G., Pons, G., Mandelbrot, L., 2004. Maternal-fetal transfer M., Bruschettini, P., Michetti, F., 2000. S100B protein concentrations
and amniotic fluid accumulation of nucleoside analogue reverse tran- in cord blood: correlations with gestational age in term and preterm
scriptase inhibitors in human immunodeficiency virus-infected preg- deliveries. Clin. Chem. 46 (7), 998–1000.
nant women. Antimicrob. Agents Chemother. 48, 4332–4336. Gazzolo, D., Marinoni, E., Di Iorio, R., Lituania, M., Bruschettini, P.,
Cleveland, M.G., Bakos, M.A., Hilton, S.M., Goldblum, R.M., 1991a. Michetti, F., 2002. Circulating S100B protein is increased in intrauter-
Amniotic fluid: the first feeding of mucosal immune factors. In: ine growth retarded fetuses. Pediatr. Res. 51 (2), 215–219.
­Mestecky, J., et al. (Ed.), Advances in Experimental Medicine and Gitlin, D., Kumate, J., Morales, C., Noriega, L., Arevalo, N., 1972. The
Biology: Immunology of Milk and the Neonate. Plenum Press, New turnover of amniotic fluid protein in the human conceptus. Am. J.
York, pp. 41–49. Obstet. Gynecol. 113, 632–645.
Cleveland, M.G., Bakos, M.A., Pyron, D.L., Rajaraman, S., Goldblum, Goluszko, P., Niesel, D., Nowicki, B., Selvarangan, R., Nowicki, S., Hart,
R.M., 1991b. Characterization of secretory component in amniotic A., Pawelczyk, E., Das, M., Urvil, P., Hasan, R., 2001. Invasiveness
fluid: Identification of new forms of secretory IgA. J. Immunol. 147, of Escherichia coli from pregnant patients with pyelonephritis. Infect.
181–188. Immun. 69 (7), 4678–4680.
Cohen, J., Ghezzi, F., Romero, R., Ghidini, A., Mazor, M., Tolosa, J.E., Graca, G., Duarte, I.F., Barros, A.S., Goodfellow, B.J., Diaz, S.O., Pinto,
Goncalves, L.F., Gomez, R., 1996. GRO alpha in the fetomaternal and J., Carreira, I.M., Galhano, E., Pita, C., Gil, A.M., 2010. The impact
amniotic fluid compartments during pregnancy and parturition. Am. J. of prenatal disorders on the metabolic profile of 2nd trimester amni-
Reprod. Immunol. 35, 23–29. otic fluid: a nuclear magnetic resonance (NMR) metabonomic study.
Cohn, B.R., Joe, B.N., Zhao, S., Kornak, J., Zhang, V.Y., Iman, R., Kurhane- J. Proteome. Res. 9, 6016–6024.
wicz, J., Vahidi, K., Yu, J., Caughey, A.B., Swanson, M.G., 2009. Greig, P.C., Herbert, W.N., Robinette, B.L., Teot, L.A., 1995. Amniotic
Quantitative metabolic profiles of 2nd and 3rd trimester human amni- fluid interleukin-10 concentrations increase through pregnancy and
otic fluid using (1)H HR-MAS spectroscopy. MAGMA 22, 343–352. are elevated in patients with preterm labor associated with intrauterine
Danforth, D.N., Hull, R.W., 1958. The microscopic anatomy of the fetal infection. Am. J. Obstet. Gynecol. 173, 1223–1227.
membranes with particular reference to the detailed structure of the Groenen, P.M., Engelke, U.F., Wevers, R.A., Hendriks, J.C., Eskes, T.K.,
amnion. Am. J. Obstet. Gynecol. 75, 536–547. Merkus, H.M., Steegers-Theunissen, R.P., 2004. High-­resolution 1H
Daniels, R.H., Houston, W.A., Petersen, M.M., Williams, J.D., Williams, B.D., NMR spectroscopy of amniotic fluids from spina bifida fetuses and
Morgan, B.P., 1990. Stimulation of human rheumatoid synovial cells by controls. Eur. J. Obstet. Gynecol. Reprod. Biol. 112 (1), 16–23.
non-lethal complement membrane attack. Immunology 69, 237–242. Hansch, G.M., Betz, M., Gunther, J., Rother, K.O., Sterzel, B., 1988. The
Darj, E., Lyrenas, S., 1998. Insulin-like growth factor binding protein-1, a complement membrane attack complex stimulates the prostaglandin
quick way to detect amniotic fluid. Acta Obstet. Gynecol. Scand. 77 production of cultured glomerular epithelial cells. Int. Arch. Allergy.
(3), 295–297. Appl. Immunol. 85, 87–93.
Das, M., Hart-Van Tassell, A., Urvil, P.T., Lea, S., Pettigrew, D., Anderson, Harirah, H., Donia, S.E., Hsu, C.D., 2002. Amniotic fluid matrix metal-
K.L., Samet, A., Kur, J., Matthews, S., Nowicki, S., Popov, V., Goluszko, loproteinase-9 and interleukin-6 in predicting intra-amniotic infection.
P., Nowicki, B.J., 2005. Hydrophilic Domain II of Escherichia coli Dr Obstet. Gynecol. 99 (1), 80–84.
Fimbriae Facilitates Cell Invasion. Infect. Immun. (73), 6119–6126. Hart, A., Pham, T., Nowicki, S., Whorton Jr., E.B., Martens, M.G.,
Dvir, R., Golander, A., Jaccard, N., Yedwab, G., Otremski, I., Spirer, Z., Anderson, G.D., Nowicki, B.J., 2001. Gestational pyelonephritis-
­
Weisman, Y., 1995. Amniotic fluid and plasma levels of parathyroid associated Escherichia coli isolates represent nonrandom closely
hormone-related protein and hormonal modulation of its secretion by related population. Am. J. Obstet. Gynecol. 174, 983–989.
amniotic fluid cells. Eur. J. Endocrinol. 133 (3), 277–282. Hedlund, M., Duan, R.D., Nilsson, A.A., Svensson, M., Karpman, D.,
Elimian, A., Figueroa, R., Canterino, J., Verma, U., Aguero-Rosenfeld, M., Svanborg, C., 2001. Fimbriae, transmembrane signaling, and cell acti-
Tejani, N., 1998. Amniotic fluid complement C3 as a marker of intra- vation. J. Infect. Dis. 183 (Suppl. 1), S47–S50.
amniotic infection. Obstet. Gynecol. 92 (1), 72–76. Heine, R.P., Wiesenfeld, H., Mortimer, L., Greig, P.C., 1998. Amniotic
Faurschou, M., Sorensen, O.E., Johnsen, A.H., Askaa, J., Borregaard, N., fluid defensins: Potential markers of subclinical intrauterine infection.
2002. Defensin-rich granules of human neutrophils: characterization Clin. Infect. Dis. 27, 513–518.
of secretory properties. Biochim. Biophys. Acta 1591 (1-3), 29–35. Heyborne, K.D., McGregor, J.A., Henry, G., Witkin, S.S., Abrams, J.S.,
Fenichel, P., Donzeau, M., Cervoni, F., Menezo, Y., His, B.L., 1995. 1994. Interleukin-10 in amniotic fluid at midtrimester: Immune activa-
Expression of complement regulatory proteins on human eggs and tion and suppression in relation to fetal growth. Am. J. Obstet. Gyne-
preimplantation embryos. Am. J. Reprod. Immunol. 33, 155–164. col. 171, 55–59.
Florio, P., Michetti, F., Bruschettini, M., Lituania, M., Bruschettini, P., Heyborne, K.D., Witkin, S.S., McGregor, J.A., 1992. Tumor necrosis
Severi, F.M., Petraglia, F., Gazzolo, D., 2004. Amniotic fluid S100B factor-alpha in midtrimester amniotic fluid is associated with impaired
protein in midgestation and intrauterine fetal death. Lancet 364 intrauterine fetal growth. Am. J. Obstet. Gynecol. 167, 920–925.
(9430), 270–272. Hillier, S.L., Witkin, S.S., Krohn, M.A., Watts, D.H., Kiviat, N.B.,
Frendeus, B., Godaly, G., Hang, L., Karpman, D., Svanborg, C., 2001. ­Eschenbach, D.A., 1993. The relationship of amniotic fluid cytokines
Interleukin-8 receptor deficiency confers susceptibility to acute pyelo- and preterm delivery, amniotic fluid infection, histologic chorioamnio-
nephritis. J. Infect. Dis. 183 (Suppl. 1), S56–S60. nitis, and chorioamnion infection. Obstet. Gynecol. 81 (6), 941–948.
Gardella, C., Hitti, J., Martin, T.R., Ruzinski, J.T., Eschenbach, D., 2001. Hilton, S.M., 1993. Quantitation of Immunoglobulins in Amniotic Fluid
Amniotic fluid lipopolysaccharide-binding protein and soluble CD14 throughout Gestation: Purification and Partial Characterization of a
as mediators of the inflammatory response in preterm labor. Am. J. Novel Form of Immunoglobulin A (Ph.D. thesis). University of Texas
Obstet. Gynecol. 184 (6), 1241–1248. Medical Branch, Galveston, Texas.
Amniotic Fluid and the Fetal Mucosal Immune System Chapter | 115  2267

Hitchcock, P.J., MacKay, H.T., Wasserheit, J.N., 1999. Sexually Trans- Leach, J.L., Sedmak, D.D., Osborne, J.M., Rahill, B., Lairmore, M.D.,
mitted Diseases and Adverse Outcomes of Pregnancy. ASM Press, Anderson, C.L., 1996. Isolation from human placenta of the IgG trans-
Washington, DC. porter, FcRn, and localization to the syncytiotrophoblast: Implications
Hitti, J., Hillier, S.L., Agnew, K.J., Krohn, M.A., Reisner, D.P., E­ schenbach, for maternal-fetal antibody transport. J. Immunol. 157, 3317–3322.
D.A., 2001. Vaginal indicators of amniotic fluid infection in preterm Lind, T., Kendall, A., Hytten, F.E., 1972. The role of the fetus in the forma-
labor. Obstet. Gynecol. 97 (2), 211–219. tion of amniotic fluid. Br. J. Obstet. Gynaecol. 79, 289–298.
Huffaker, J., Witkin, S.S., Cutler, L., Druzin, M.L., Ledger, W.J., 1989. Lotgering, F.K., Wallenburg, H.C., 1986. Mechanisms of production and
Total complement activity in maternal sera, amniotic fluids and cord clearance of amniotic fluid. Semin. Perinatol. 10, 94–102.
sera in women with premature labor, premature rupture of membranes Macri, C.J., Martinez, A., Moody, T.W., Gray, K.D., Miller, M.J.,
or chorioamnionitis. Surg. Gynecol. Obstet. 168, 397–401. ­Gallagher, M., Cuttitta, F., 1996. Detection of adrenomedullin, a hypo-
Hutchings, A., Purcell, W.M., Benfield, M.R., 1999. Peripheral blood tensive peptide, in amniotic fluid and fetal membranes. Am. J. Obstet.
­antigen-presenting cells from African-Americans exhibit increased Gynecol. 175 (4 Pt 1), 906–911.
CD80 and CD86 expression. Clin. Exp. Immunol. 118, 247–252. Malhotra, R., Willis, A.C., Lopez Bernal, A., Thiel, S., Sim, R.B., 1994.
Izban, M.G., Nowicki, B.J., Nowicki, S., 2012. 1,25-Dihydroxyvitamin Mannan-binding protein levels in human amniotic fluid during ges-
D3 Promotes a Sustained LPS-induced NF-κB-dependent Expres- tation and its interaction with collectin receptor from amnion cells.
sion of CD55 in Human Monocytic THP-1 cells. PLoS ONE (7), Immunology 82 (3), 439–444.
49318. Mandelbrot, L., Peytavin, G., Firtion, G., Farinotti, R., 2001. Maternal-
Jauniaux, E., Gulbis, B., Jurkovic, D., Campbell, S., Collins, W.P., Ooms, fetal transfer and amniotic fluid accumulation of lamivudine in human
H.A., 1994. Relationship between protein concentrations in embry- immunodeficiency virus-infected pregnant women. Am. J. Obstet.
ological fluids and maternal serum and yolk sac size during human Gynecol. 184 (2), 153–158.
early pregnancy. Hum. Reprod. 9, 161–166. Mathai, M., Mathai, E., Jairaj, P., 1991. Candida antibodies in amniotic
Jauniaux, E., Jurkovic, D., Gulbis, B., Liesnard, C., Lees, C., Campbell, fluid. Indian J. Med. Res. 93, 353–355.
S., 1995. Materno-fetal immunoglobulin transfer and passive immu- May, J., Lell, B., Luty, A.J., Meyer, C.G., Kremsner, P.G., 2000. Plasma
nity during the first trimester of human pregnancy. Hum. Reprod. 10, interleukin-10 colon; Tumor necrosis factor (TNF)-alpha ratio is asso-
3297–3300. ciated with TNF promoter variants and predicts malarial complica-
Jensen, T.S., Bjorge, L., Wollen, A.L., Ulstein, M., 1995. Identification tions. J. Infect. Dis. 182, 1570–1573.
of the complement regulatory proteins CD46, CD55, and CD59 in Mellander, L., Carlsson, B., Hanson, L.A., 1986. Secretory IgA and IgM
human fallopian tube, endometrium, and cervical mucosa and secre- antibodies to E. coli O and poliovirus type I antigens occur in amni-
tion. Am. J. Reprod. Immunol. 34, 1–9. otic fluid, meconium and saliva from newborns: A neonatal immune
Kanenishi, K., Kuwabara, H., Ueno, M., Sato, C., Sakamoto, H., Hata, response without antigenic exposure—a result of anti-idiotypic induc-
T., 2001. Change of adrenomedullin concentrations in plasma and tion? Clin. Exp. Immunol. 63, 555–561.
amniotic fluid, and human placental adrenomedullin expression with Menon, R., Lombardi, S.J., Fortunato, S.J., 2001. IL-18, a product of cho-
advancing gestation. Placenta 22 (2–3), 244–250. riodecidual cells, increases during premature rupture of membranes
Kaul, A.K., Nowicki, B.J., Martens, M.G., Goluszko, P., Hart, A.R., but fails to turn on the Fas-FasL-mediated apoptosis pathway. J.
­Nagamani, M., Kumar, D., Pham, T., Nowicki, S., 1994. Decay accel- Assist. Reprod. Genet. 18, 276–284.
erating factor (DAF) is expressed in the human endometrium and Michalsky, M.P., Lara-Marquez, M., Chun, L., Besner, G.E., 2002.
may serve as the natural ligand for Dr pili of Escherichia coli. Am. J. ­Heparin-binding EGF-like growth factor is present in human amniotic
Reprod. Immunol. 32, 194–199. fluid and breast milk. J. Pediatr. Surg. 37 (1), 1–6.
Kitamura, K., Kangawa, K., Kawamoto, M., Ichiki, Y., Nakamura, S., Moller, M., Sorensen, J., Jepsen, F.L., Thomsen, A.C., 1997. Diagnosis of
­Matsuo, H., Eto, T., 1993. Adrenomedullin: a novel hypotensive pep- intrauterine infection by demonstration of antibody-coated bacteria in
tide isolated from human pheochromocytoma. Biochem. Biophys. the amniotic fluid. Br. J. Obstet. Gynaecol. 93, 240–244.
Res. Commun. 192 (2), 553–560. Mustafa, M.M., Lebel, M.H., Ramilo, O., Olsen, K.D., Reisch, J.S.,
Kleymann, G., 2003. Novel agents and strategies to treat herpes simplex ­Beutler, B., McCracken Jr, G.H., 1989. Correlation of interleukin-1
virus infections. Expert Opin. Investig. Drugs 12 (2), 165–183. beta and cachectin concentrations in cerebrospinal fluid and outcome
Kourtis, A.P., Lee, F.K., Abrams, E.J., Jamieson, D.J., Bulterys, M., 2006. from bacterial meningitis. J. Pediatr. 115 (2), 208–213.
Mother-to-child transmission of HIV-1: timing and implications for Nellen, J.F., Damming, M., Godfried, M.H., et al., 2008. Steady-state nevi-
prevention. Lancet Infect. Dis. 6, 726–732. rapine plasma concentrations are influenced by pregnancy. HIV Med.
Krohn, M.A., Hillier, S.L., Lee, M.L., Rabe, L.K., Eschenbach, D.A., 9, 234–238.
1991. Vaginal Bacteroides species are associated with an increased Nishikori, K., Noma, J., Hirakawa, S., Amano, T., Kudo, T., 1993. The
rate of preterm delivery among women in preterm labor. J. Infect. Dis. change of membrane complement regulatory protein in chorion of
164 (1), 88–93. early pregnancy. Clin. Immunol. 69, 167–174.
Lang, A.K., Searle, R.F., 1994. The immunomodulatory activity of human Nowicki, B., Sledzinska, A., Samet, A., Nowicki, S., 2011. Pathogenesis
amniotic fluid can be correlated with transforming growth factor-betal of gestational UTI: urinary obstruction vs. immune adaptation and
(TGF-β1) and β2 activity. Clin. Exp. Immunol. 97, 158–163. microbial virulence. BJOG (118), 109–112.
Langman, J., 1981. Medical Embryology. Williams & Wilkins, Baltimore. Puchner, T., Egarter, C., Wimmer, C., Lederhilger, F., Weichselbraun, I.,
Larsen, B., Snyder, I.S., Galask, R.P., 1974. Bacterial growth inhibition by 1993. Amniotic fluid interleukin-8 as a marker for intraamniotic infec-
amniotic fluid. 2, reversal of amniotic fluid bacterial growth inhibition tion. Arch. Gynecol. Obstet. 253, 9–14.
by addition of a chemically defined medium. Am. J. Obstet. Gynecol. Quan, C.P., Forestier, F., Bouvet, J.P., 1999. Immunoglobulins of the
119, 497–501. human amniotic fluid. Am. J. Reprod. Immunol. 42, 219–225.
2268  SECTION | I  Genitourinary Tract and Mammary Gland

Rana, T., Udoko, A.N., Johnson, C.A., Singh, R., Rachakonda, G., Lima, Srivastava, M.D., Lippes, J., Srivastava, B.I., 1996. Cytokines of the human
M.F., Villalta, F., Mandape, S.N., Lawler, J., Pratap, S., Nowicki, S., reproductive tract. Am. J. Reprod. Immunol. 36, 157–166.
Nowicki, B., Nde, P.N., 2014a. Thrombospondin-1 Carries CD55 like Stirrat, G.M., 1994. Pregnancy and immunity. BMJ 308, 1384.
Domains: A Novel Target for E. coli Invasion into Human Heart Cells. Tollerud, D.J., Brown, L.M., Blattner, W.A., Weiss, S.T., Maloney, E.M.,
Circulation Res in press. Kurman, C.C., Nelson, D.L., Hoover, R.N., 1995. Racial differences
Rana, T., Hasan, R.J., Nowicki, S., Venkatarajan, M.S., Singh, R., Urvil, in serum immunoglobulin levels: Relationship to cigarette smoking,
P.T., Popov, V., Braun, W.A., Popik, W., Goodwin, J.S., Nowicki, T-cell subsets, and soluble interleukin-2 receptors. J. Clin. Lab. Anal.
B.J., 2014b. Complement Protective Epitopes and CD55-Microtubule 9, 37–41.
Complexes Facilitate the Invasion and Intracellular Persistence of Tourville, D.R., Lippes, J., Ogra, S., Tomasi, T.B., 1969. Secretory com-
Uropathogenic Escherichia coli. J. Infect. Dis. 209, 1066–1076. ponents in human placental tissues and fetal membranes. In: ­Dayton,
Rice, J.C., Peng, T., Kuo, Y.F., Pendyala, S., Simmons, L., Boughton, J., D.H., et al. (Ed.), The Secretory Immunologic System. National
Ishihara, K., Nowicki, S., Nowicki, B.J., 2006. Renal Allograft Injury Institute of Child Health and Human Development, Bethesda, MD,
Is Associated with Urinary Tract Infection Caused by Escherichia coli pp. 403–411.
Bearing Adherence Factors. Am. J. Transplant (6), 2375–2383. Ugwumadu, A., Manyonda, I., Reid, F., Hay, P., 2003. Effect of early oral
Romero, R., Brody, D.T., Oyarzun, E., Mazor, M., Wu, Y.K., Hobbins, clindamycin on late miscarriage and preterm delivery in asymptomatic
J.C., Durum, S.K., 1989. Infection and labor. III. Interleukin-1: a sig- women with abnormal vaginal flora and bacterial vaginosis: a random-
nal for the onset of parturition. Am. J. Obstet. Gynecol. 160 (5 Pt 1), ized controlled trial. Lancet 361, 983–988.
1117–1123. Vanderpuye, O.A., Labarrere, C.A., McIntyre, J.A., 1993. Expres-
Rooney, I.A., Morgan, B.P., 1990a. Non-lethal doses of antibody and com- sion of CD59, a human complement system regulatory protein, in
plement stimulate release of prostaglandin E2 from human amniotic extraembryonic membranes. Int. Arch. Allergy Immunol. 101 (4),
cells in vitro. Biochem. Soc. Trans. 18, 617. 376–384.
Rooney, I.A., Morgan, B.P., 1990b. Protection of human amniotic epithe- Vesce, F., Scapoli, C., Giovannini, G., Tralli, L., Gotti, G., Valerio, A.,
lial cells (HAEC) from complement-mediated lysis: expression on the ­Piffanelli, A., 2002. Cytokine imbalance in pregnancies with fetal
cells of three complement inhibitory membrane proteins. Immunology chromosomal abnormalities. Hum. Reprod. 17 (3), 803–808.
71, 308–311. Vosburgh, G.J., Flexner, L.B., Cowie, D.B., Hellman, L.M., Proctor, N.K.,
Samet, A., Sledzinska, A., Krawzcyk, B., Hellmann, A., Nowicki, S., Kur, Wilde, W.S., 1948. The rate of renewal in women of the water and
J., Nowicki, J., 2013. Leukemia and risk of recurrent E. coli bactere- sodium of the amniotic fluid as determined by tracer techniques. Am.
mia: genotyping implicates E. coli translocation from the colon to the J. Obstet. Gynecol. 56, 1156–1159.
bloodstream. Eur. J. Clin. Microbiol. Infect. Dis. 32 (11), 1393–1400. Wang, S., Kallichanda, N., Song, W., Ramirez, B.A., Ross, M.G., 2001.
Samet, A., Krawzcyk, B., Nowicki, B., Sledzinska, A., Nowicki, S., Kur, J., Expression of aquaporin-8 in human placenta and chorioamniotic
2013. A Subset of Two Adherence Systems, Acute Pro-Inflammatory membranes: evidence of molecular mechanism for intramembra-
pap Genes and Invasion Coding dra, fim or sfa, Increase the Risk of nous amniotic fluid resorption. Am. J. Obstet. Gynecol. 185 (5),­
Escherichia coli Translocation to the Bloodstream. Eur. J. Clin. Micro- 1226–1231.
biol. Infect. Dis. (32), 1579–1582. Weimann, E., Reisbach, G., Reinsberg, J., Lentze, M.J., 1995. IL-6 and
Santhanam, U., Avila, C., Romero, R., Viguet, H., Ida, N., Sakurai, S., G-CSF levels in amniotic fluid during the second trimester in normal
Sehgal, P.B., 1991. Cytokines in normal and abnormal parturition: and abnormal pregnancies. Arch. Gynecol. Obstet. 256, 125–130.
Elevated amniotic fluid interleukin-6 levels in women with premature Wladimiroff, J.W., Campbell, S., 1974. Fetal urine-production rates in nor-
rupture of membranes associated with intrauterine infection. Cytokine mal and complicated pregnancy. Lancet 1, 151–154.
3, 155–163. Wlodek, M.E., Koutsis, K., Westcott, K.T., Ho, P.W., Di Nicolantonio, R.,
Schlievert, P., Johnson, W., Galask, R.P., 1976. Isolation of a low-­ Moseley, J.M., 2001. The spontaneously hypertensive rat fetus, not the
molecular-weight antibacterial system from human amniotic fluid. mother, is responsible for the reduced amniotic fluid PTHrP concen-
Infect. Immun. 14, 1156–1166. trations and growth restriction. Placenta 22 (7), 646–651.
Schmidt, A.P., Tort, A.B., Amaral, O.B., Schmidt, A.P., Waltz, R., Wroblewska-Seniuk, K., Selvarangan, R., Hart, A., Pladzyk, R., Goluszko,
­Vettorazzi-Stuckzynski, J., Martins-Costa, S.H., Ramos, J.G., Souza, P., Jafari, A., du Merle, L., Nowicki, S., Yallampalli, C., Le Bou-
D.O., 2004. Serum S100B in pregnancy-related hypertensive disor- guenec, C., Nowicki, B., 2005. Dra/AfaE Adhesin of Uropathogenic
ders: a case-control study. Clin. Chem. 50 (2), 435–438. Dr/Afa+ Escherichia coli Mediates Mortality in Pregnant Rats. Infect.
Sedin, G., Hammarlund, K., Nilsson, G.E., Oberg, P.A., Stromberg, B., Immun. (73), 7597–7601.
1981. Water transport through the skin of newborn infants. Ups. J. Yoon, B.H., Oh, S.Y., Romero, R., Shim, S.S., Han, S.Y., Park, J.S., Jun,
Med. Sci. 86, 27–31. J.K., 2001. An elevated amniotic fluid matrix metalloproteinase-8
Silver, H.M., Sperling, R.S., St Clair, P.J., Gibbs, R.S., 1989. Evidence level at the time of mid-trimester genetic amniocentesis is a risk factor
relating bacterial vaginosis to intraamniotic infection. Am. J. Obstet. for spontaneous preterm delivery. Am. J. Obstet. Gynecol. 185 (5),
Gynecol. 161 (3), 808–812. 1162–1167.
Sledzinska, A., Mielech, A., Krawczyk, B., Samet, A., Nowicki, B.J., Zhang, Q., Shimoya, K., Moriyama, A., Yamanaka, K., Nakajima, A.,
Nowicki, S., Jankowski, Z., Kur, J., 2010. Fatal sepsis in a 23-year-old Nobunaga, T., Koyama, M., Azuma, C., Murata, Y., 2001. Production
pregnant woman with pyelonephritis caused by Escherichia coli bear- of secretory leukocyte protease inhibitor by human amniotic mem-
ing Dr and P adhesins: diagnostic value of postmortem genotyping of branes and regulation of its concentration in amniotic fluid. Mol. Hum.
isolated strains. British J. of Obs. Gyn. (118), 266–269. Reprod. 7 (6), 573–579.

You might also like