You are on page 1of 14

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/7879978

HLA-G in immune tolerance in pregnancy

Article  in  The FASEB Journal · June 2005


DOI: 10.1096/fj.04-2078rev · Source: PubMed

CITATIONS READS
363 1,816

4 authors, including:

Ramsey H McIntire
MilliporeSigma
19 PUBLICATIONS   672 CITATIONS   

SEE PROFILE

All content following this page was uploaded by Ramsey H McIntire on 28 May 2015.

The user has requested enhancement of the downloaded file.


The FASEB Journal • Review

HLA-G and immune tolerance in pregnancy


Joan S. Hunt,*,1 Margaret G. Petroff,* Ramsey H. McIntire,* and Carole Ober†
*University of Kansas Medical Center, Kansas City, Kansas, USA; and †The University
of Chicago, Chicago, Illinois, USA

ABSTRACT Multiple mechanisms underlie the sur- tation antigens are late appearing; tolerance and im-
prising willingness of mothers to tolerate genetically mune privilege at the maternal-fetal interface are
different fetal tissues during pregnancy. Chief among readily identified. Three major principles emerging
these is the choice of HLA-G, a gene with few alleles, from these studies are that 1) multiple mechanisms
rather than the highly polymorphic HLA-A and -B genes, provide protection, 2) both the fetus and the mother
for expression by the placental cells that interface contribute to development and maintenance of the
directly with maternal blood and tissues. Novel aspects pregnant uterus as an immune privileged site, and 3)
of this major histocompatibility complex class Ib gene fetal factors drive changes in maternal immune re-
include alternative splicing to permit production of sponses. In this article, we first briefly discuss a number
membrane and soluble isoforms, deletions that of conditions responsible for immune privilege and
dampen responses to interferons, and a shortened maternal tolerance for which scientific evidence is
cytoplasmic tail that affects expression at the cell strong, then focus on a central feature—a unique
surface. Placental cells migrating into the maternal capacity of placental cells to select specific genes within
uterus synthesize both membrane and soluble iso- the major histocompatibility complex (MHC) for ex-
forms, which interact with inhibitory receptors on leu- pression. We present evidence that this unique capacity
kocytes such as ILT2 and ILT4. Cytotoxic T lympho- for selection of specific MHC antigens, which in hu-
cytes either die or reduce production of one of their mans are called human leukocyte antigens (HLA), may
major coreceptor/activator cell surface molecules, be responsible in large part for the reprogramming of
CD8; natural killer cells are immobilized and mononu- local maternal immune responses that characterize
clear phagocytes are programmed into suppressive successful semiallogeneic pregnancy.
modes characterized by high production of anti-inflam-
matory cytokines. The idea that placental HLA-G pro- Strategies for protecting the semiallogeneic fetus
teins facilitate semiallogeneic pregnancy by inhibiting from maternal graft rejection responses
maternal immune responses to foreign (paternal) anti-
gens via these actions on immune cells is now well
During pregnancy, the maternal immune system is
established, and the postulate that the recombinant
clearly active, and under certain conditions may con-
counterparts of these proteins may be used as powerful
tribute to fetal damage/death. Well-defined pathologi-
tools for preventing immune rejection of transplanted
cal processes include destruction of fetal erythrocytes
organs is gaining in popularity.—Hunt, J. S., Petroff,
(Rh antigen, erythroblastosisis) and platelets (HPA-1
M. G., McIntire, R. H., Ober, C. HLA-G and immune
and -2, alloimmune thrombocytopenia) by maternal
tolerance in pregnancy. FASEB J. 19, 681– 693 (2005)
antibodies and infections of pregnancy, where activated
macrophages secreting high levels of Th1-type cyto-
Key Words: human 䡠 placenta 䡠 immune privilege kines alter the delicate cytokine balance at the mater-
nal-fetal interface (2, 3). Yet even with a demonstrably
active maternal immune system, mothers usually seem
BACKGROUND
1
The success of human pregnancy, where the fetus Correspondence: Department of Anatomy and Cell Biol-
ogy, University of Kansas Medical Center, Kansas City, Kansas,
resides comfortably within the maternal uterus for 9 66160-7400 USA. E-mail: jhunt@kumc.edu
months, defies the precepts of immunology. Medawar Supported in part by grants from the National Institutes of
was the first to attempt sorting out the strategies used in Health to J.S.H. (HD26429, HD35859, and HD39878), to
pregnancy to circumvent maternal rejection of the M.G.P. (HD045611), and to C.O. (HD21244). R.M. is sup-
embryo/fetus (1). The proposed mechanisms included ported by a fellowship from the Kansas University Medical
physical separation of maternal and fetal tissues, lack of Center Biomedical Research Training Program. The authors
fetal antigens that could stimulate graft rejection, and appreciate the assistance of S. Fernald, Kansas Reproductive
Sciences Center, in preparation of the figures. This publica-
development of tolerance. tion was made possible by NIH grant number P20 RR016475
Today certain aspects of each of these strategies have from the INBRE Program of the National Center for Re-
been identified: the blood circulations of the fetus and search Resources.
the mother are entirely separate; in the fetus, transplan- doi: 10.1096/fj.04-2078rev

0892-6638/05/0019-0681 © FASEB 681


to tolerate rather than reject their genetically disparate cytes, are identified mainly in the myometrium distal to
fetuses. Ordinarily, the mother would be expected to fetal tissues whereas members of the innate immune
generate graft-attacking antibodies and cytotoxic T system, natural killer (NK) cells and macrophages,
lymphocytes (CTL) to foreign (paternal) HLA or other predominate in the decidua (5, 6). An exception is the
antigens expressed by fetal cells. HLA antigens are Treg subset of CD4⫹/CD25⫹ cells, which produce inter-
called “transplantation” antigens because they com- leukin-10 (IL-10) and transforming growth factor-␤1
prise the most powerful stimulators of graft rejection. (TGF-␤1), and are believed to be critical to mainte-
Thus, in organ transplantation the matching of certain nance of tolerance (7, 8). These cells, whose prolifera-
donor and patient alleles is an absolute requirement tion is stimulated by estrogen (9) comprise ⬃14% of
for successful grafting. CD4⫹ cells in early decidua (10). Uterine cytokine
Even though mothers and fathers are almost invari- networks, while of great complexity and containing
ably disparate at multiple HLA loci, rejection of the many examples of Th1-type reactivities (11, 12), were
fetus as a consequence of maternal recognition of first shown in the mouse to be dominated by these and
paternal HLA as foreign is essentially undocumented. other Th2-type cytokines (13). Similar reports have
Although anti-paternal HLA antibodies are common in appeared in human pregnancy (14). Immunomodula-
pregnant women, they do no damage. Even novel HLA tory hormones such as prolactin, chorionic gonadotro-
antigens expressed in the fetal membranes are tolero- pin, and progesterone are abundant; chemokines op-
genic rather than immunogenic (4). Attack by mater- erate to control immune cell numbers and types.
nal CTL is also effectively thwarted, as described in The fetal contributions are unique. Figure 2 shows
detail below. Thus, mechanisms underlying maternal that the fetus, which is derived from the inner cell mass
tolerance are unusually effective and raise the critical of the blastocyst, is secluded within a protective shell
question of how immune privilege might be established composed of trophoblast cells that arise from the
in the natural situation of pregnancy so as to assure trophectoderm layer of the blastocyst. It is therefore the
viability of the embryo/fetus. heavy responsibility of the trophoblast cells to interact
Figure 1 illustrates the principle that maternal and appropriately with the maternal environment and pro-
fetal processes contribute to the generation of a safe tect the fetus from maternal immune attack. As illus-
environment. Within the uterus, a dramatic change in trated in Fig. 1, the trophoblast cells circumvent anti-
endometrial leukocyte subpopulations occurs as a con- body-mediated damage by exhibiting high levels of the
sequence of implantation. After a brief inflammatory complement regulatory proteins (15), and reduce cell-
reaction caused by blastocyst breaching of the uterine mediated immunity by expressing inhibitory members
epithelium, a reaction best documented in rodents, the of the B7 family (16), and apoptosis-inducing members
altered endometrium (now termed decidua) settles of the tumor necrosis factor (TNF) family of ligands
into a pattern where local protection is provided by the (17). As with cells in the decidua, fetal cells produce
innate immune system. From this point onward, the immunosuppressive cytokines, chemokines, and prosta-
major players in acquired immunity, T and B lympho- glandins that dampen T lymphocyte proliferation and
export high levels of immune suppressive hormones
such as progesterone.
Most important, trophoblast cells strictly regulate
their expression of HLA genes and the production of
their proteins. It is these proteins that, if recognized as
foreign by maternal immune cells, would stimulate
maternal anti-fetal CTL capable of destroying HLA-
expressing fetal cells (18). Instead, the antigens ex-
pressed in trophoblast cells program maternal leuko-
cytes into pathways consistent with tolerance.

HLA-G: A NOVEL GENE EXPRESSED IN


PLACENTAS

The genes encoding HLA antigens are clustered on


chromosome 6p21 at the telomeric end of the HLA
Figure 1. Multiple mechanisms underlie maternal tolerance region. Although this region contains ⬃20 to 25 HLA
of the fetus. Mothers, via changes that occur in the uterus, class I genes, relatively few are transcribed or translated;
and embryo/fetuses, via special adaptations of the placenta, most are pseudogenes or gene fragments. The ex-
contribute to the establishment of an immune privileged
environment within which the semiallogeneic fetus resides pressed class I genes are subdivided into class Ia, which
safely until termination. NK cells, natural killer cells; Treg, includes HLA-A, -B, and -C, and class Ib, which includes
CD4⫹ regulatory T cells; TNF superfamily, tumor necrosis HLA-E, -F, and -G. HLA class II (HLA-D) genes, if
factor superfamily. transcribed, are not translated in human trophoblast

682 Vol. 19 May 2005 The FASEB Journal HUNT ET AL.


ity at the HLA-C locus does not seem to be a causal
factor in infertility or termination of pregnancy. The
other HLA class I antigens expressed by trophoblast
cells are HLA-E, -F, and -G. These class Ib antigens are
distinguished by low numbers of alleles that differ at
the protein level. For example HLA-E has 2 alleles (20)
and HLA-G has five alleles (reviewed in ref 21). There
are no allelic variants of HLA-F in the published
literature, although two nonsynonymous (amino acid)
substitutions are reported in online databases (http://
genome.ucsc.edu), which are likely rare in the general
population. Moreover, most polymorphisms in the
HLA-G gene do not alter the amino acid sequence; the
few that do are not predicted to change secondary
structures of the heavy chains (4, 21).
Of the HLA class Ib molecules expressed by tropho-
blast cells, HLA-G was the first to be identified and
remains an antigen of great interest and a focus of
experimental evaluation (reviewed in refs 22–24).

Structural features of HLA-G


Figure 2. A schematic illustration of the human fetus, pla-
centa and extraplacental membranes, and modified endome-
trium known as decidua. The fetus developing within the Although the genomic structure of HLA-G is similar to
amniotic sac is surrounded and encased by trophoblast cells other class I genes, it is unique in most other respects.
in the placenta and chorion membrane (red). Upper insert: The HLA-G gene has eight exons encoding a signal
Cytotrophoblast (CTB) cells within the placental villi serve as
the progenitors for all differentiated trophoblast cell sub-
peptide (exon 1), the ␣1, ␣2, and ␣3 domains (exons 2,
populations, including the syncytiotrophoblast (sTB) layer, 3, and 4, respectively), the transmembrane domain
which is continuously exposed to maternal blood. This single (exon 5), and the intracellular domain (exons 6 and 7),
cell layer is responsible for transfer of maternal-fetal nutrients similar to other class I genes (Fig. 3). However, a
and wastes, synthesis of placental hormones, and providing a premature stop codon in exon 6 results in a truncated
physical barrier to maternal cell traffic into the fetus. CTB cytoplasmic tail that reveals a cryptic retrieval motif
cells proliferate and migrate into the decidua, attaching the (25). This results in the slower turnover and prolonged
placenta to the mother and facilitating certain crucial physi-
ological events required for successful pregnancy. Lower expression of HLA-G at the cell surface, and possibly
insert: The amnion membrane comprised of a single layer of the inefficient presentation of exogenous peptides.
epithelial cells is a strong sac holding the fetus in amnionic Park and colleagues interpret this as evidence that the
fluid. The chorion membrane CTB cells, derived from the primary function of HLA-G is not antigen presentation
migrating extravillous CTB cells, interface directly with ma- but as an inhibitory ligand for NK cells (25).
ternal decidual cells. A second unique feature of HLA-G is that it encodes
multiple isoforms as a result of alternative splicing. Five
cells even under inducing conditions (19) and will not of the seven transcripts that result from alternative
be discussed further in this review. splicing are shown in Fig. 3. The full-length isoform
One remarkable difference between the HLA class Ia HLA-G1 is structurally similar to other class I genes,
and Ib genes is that the former are highly polymorphic, except for the truncated cytoplasmic tail. The G2
with many alleles, and the latter have few variants. isoform results from the removal of exon 3 and ho-
Major differences have been observed in glycoproteins modimerizes to form an HLA class II-like structure (26,
associated with these two subsets of class I antigens. In 27). These two isoforms are expressed as soluble pro-
general, class Ia antigens are membrane bound. By teins (HLA-G5 and -G6, respectively) due to the inclu-
contrast, one member of the class Ib group, HLA-G, is sion of intron 4 sequences in the mature mRNA,
alternatively spliced. Seven alternatively spliced tran- resulting in secreted proteins with an additional 21
scripts have been identified, of which four are pre- amino acids (encoded by intron 4 sequences) following
dicted to encode membrane bound and three are the ␣3 domain (28). HLA-G3 results from the removal
predicted to encode soluble proteins. A final difference of exons 3 and 4. HLA-G4 and -G7 (not shown in Fig. 3)
is that expression of class Ia antigens is ubiquitous mRNAs are not abundant in placentas. Exon 4 (encod-
whereas expression of class Ib antigens may be tissue/ ing the ␣3 domain) is spliced out of the HLA-G4
organ-specific and/or conditional. transcript; the HLA-G7 transcript includes exon 2 and
Human trophoblast cells express one class Ia mole- part of intron 2 and is predicted to encode a small
cule (HLA-C) and all three class Ib molecules. The soluble isoform.
HLA-C gene is moderately polymorphic, and could Compared with the classical class I genes, the most
stimulate maternal anti-fetal acquired immunity if pa- polymorphic genes in the human genome, HLA-G has
ternal alleles differed from maternal. Yet allelic dispar- relatively little polymorphism in its coding region.

HLA-G AND PREGNANCY IMMUNE PRIVILEGE 683


Figure 3. Multiple HLA-G proteins re-
sult from alternative mRNA splicing.
Upper: The HLA gene is composed of 8
exons arranged in the same sequence as
other HLA class I genes. The gene is
alternatively spliced to yield 7 tran-
scripts. In two of these, a stop sequence
in intron 4 results in soluble isoforms.
Alleles encoded by the polymorphisms
and their amino acid substitutions or
deletion are shown: *0103 (Thr31Ser),
*0104 (Leu110ILe), *0105 (1597deltaC),
*0106 (Thr258Met). A 14 bp insertion/
deletion is present in exon 8 in the 3⬘
UTR. ␣1, ␣2, ␣3 extracellular domains.
Lower: Three messages encode mem-
brane isoforms (HLA-G1, -G2, -G3) and
two encode soluble isoforms (HLA-G5
and -G6, also known as sG1 and sG2,
respectively). HLA-G1 and -G5 associate
with light chain, ␤2m; the other three
do not. Isoforms HLA-G4 and -G7 re-
main poorly defined and are not illus-
trated.

Figure 3 shows the location of the 13 polymorphisms in However, this null allele has been associated with
exons 1– 4 that have been identified to date and one in increased risk for recurrent miscarriage (34, 35), sug-
the 3⬘UTR. Polymorphisms at codon 31 in the ␣1 gesting that HLA-G1 and/or -G5 proteins do indeed
domain (Thr3 Ser), at codon 110 in the ␣2 domain play an important role in the maintenance of preg-
(Leu3 Ile), and at codon 258 in the ␣3 domain nancy and that reduced levels of one or both is a risk
(Thr3 Met) result in an amino acid substitution; poly- factor for recurrent miscarriage.
morphisms at nucleotide ⫹15 and ⫹36 in exon 1, at A 14 bp insertion/deletion polymorphism in the
codons 35, 57, and 69 in exon 2, at codons 93, 100, and untranslated exon 8 was first described by Harrison and
107 in exon 3, and at codon 188 in exon 4 do not alter colleagues (36), but has recently been shown to influ-
the amino acid sequence of the protein. A single base ence mRNA transcript size and stability. The presence
pair (bp) deletion at nucleotide 1597 causes a frame- of the 14 bp insertion allele generates a 92 bp deletion
shift at amino acid 130 (29), resulting in nonfunctional in the 3⬘UTR of the G*01012 and G*01013 mRNAs,
HLA-G1 and -G5 proteins (30). The polymorphisms possibly because it acts as a cryptic splice site (37).
that alter the protein sequence define five alleles, called Transcripts with the 92 bp deletion were associated with
G*0101, G*0103, G*0104, G*0105N, and G*0106. Si- more stable mRNA in JEG-3 cells (which are homozy-
lent variation within these allelic classes defines sub- gous for G*01013) and in an M8 cell line transfected
types, referred to as G*010101, G*010102, etc. A 14 bp with G*01012, perhaps because they were less suscepti-
insertion/deletion polymorphism is present in the un- ble to degradation (38). Further, the relative abun-
translated exon 8. dance of the alternatively spliced transcripts may be
In contrast to the class Ia HLA loci, amino acid influenced by polymorphisms in HLA-G (39). For ex-
substitutions at codons 31 and 110 in the ␣1 and ␣2 ample, studying mRNA from term trophoblast cells,
domains, respectively, are conservative changes that Hviid et al. (39) showed that heterozygotes for the
occur in residues that are not predicted to interact with G*01012 allele (but not the G*01013 allele) had re-
bound peptide or T cell receptor (21). The third amino duced levels of transcripts encoding membrane-bound
acid polymorphism at codon 258 is a nonconservative isoforms (G1, G2, G3) with the 14 bp insertion, whereas
substitution in the ␣3 domain that is highly conserved heterozygotes for the G*01013 allele (but not for the
in the class Ia genes (31). It is located in the pleated G*01012 allele) had higher levels of the G2/G4 tran-
sheet structure of the ␣3 domain, where it might affect scripts than the G*01011 allele (39). Although the
recognition of CD8 in the HLA-G1 and -G5 isoforms, or significance of these findings is not clear, it is notewor-
binding to CD4 in the HLA-G2 and -G6 isoforms (4). thy that a number of studies have demonstrated re-
Last, a polymorphic 1 bp deletion of a cytosine (C) duced expression of HLA-G mRNA or protein in term
residue at codon 130 results in a null allele (called placentas of pre-eclamptic pregnancies (40 – 43). The
G*0105N), which does not encode functional HLA-G1 lack of association between the G*0105N null allele and
or -G5 protein isoforms (30). This mutation, called pre-eclampsia in one study suggested that deficiencies
1597⌬C, occurs in the homozygous form in ostensibly of the HLA-G1 and/or -G5 isoforms were not a risk
healthy individuals, indicating that HLA-G1 and -G5 factor for pre-eclampsia (44), but reduced expression
isoforms are not essential for fetal survival (30, 32). In of other isoforms, such as the short G3 transcript, may
these situations other isoforms presumably suffice (33). influence risk (45). Indeed, the relative abundance of

684 Vol. 19 May 2005 The FASEB Journal HUNT ET AL.


transcripts and noncoding polymorphisms in HLA-G extravillous cytotrophoblast cells that contact and infil-
have been associated with pre-eclampsia (45), suggest- trate the decidua (upper insert, Fig. 2). The migrating
ing that the regulation of expression of HLA-G is cells anchor the placenta to the decidua and infiltrate
influenced by genetic variation. maternal spiral arteries, thus facilitating blood flow to
This suggestion is further supported by the recent the placenta. The extravillous cytotrophoblast cells
discovery of variation in the 5⬘-upstream regulatory ultimately regress to form the chorion membrane
region of HLA-G (46, 47). In contrast to the limited (lower insert, Fig. 2).
polymorphism in the exons, the upstream region con- HLA class I antigen expression was first identified
taining all of the known promoter and regulatory (and usually is described) as being restricted to the
elements is extraordinarily polymorphic (Fig. 4). To extravillous trophoblast cell population, with proteins
date there have been 18 polymorphisms identified in particularly prominent in cells immediately adjacent to
the ⬃1300 bp upstream from exon 1. Most are very the decidua in both early and late gestation placentas
common polymorphisms with minor allele frequencies (reviewed in refs 22–24). However, the reagent used
(⬎20%), and many reside within or close to known most frequently to identify these antigens in tissue
transcription factor binding sites (Fig. 4). These poly- sections or isolated cells, the mouse monoclonal anti-
morphisms define at least eight unique haplotypes
body W6/32, requires light chain (␤2m)/heavy chain
(47). Some promoter region haplotypes are shared
association, so neither free heavy chains nor isoforms
among alleles that differ in their coding regions (for
such as HLA-G2 and -G6 that do not associate with
example, G*01012, G*0105N, and G*01061 have iden-
␤2-microglobulin (␤2m) were detected. More recent
tical promoter region sequences), while some alleles
studies using monoclonal antibodies that identify the
with identical coding regions have different promoter
region haplotypes (e.g., the common G*01011 allele is amino acid sequence derived from intron 4 nucleo-
associated with at least three different promoter region tides—16G1 generated by D. Geraghty being a good
sequences). One variant, –725G, changes the methyl- example— have supplied localization data on HLA-G5
ation status of a CpG dinucleotide in the promoter and -G6 heavy chains. New isoform-specific antibodies
region and has been associated with an increased risk show that HLA-G5 is present throughout the placenta and
for sporadic miscarriage in an unselected sample of within the chorion membrane, decidua, and maternal
healthy women, which suggest at least some of these blood (48). In villous CTB cells, HLA-G5 is likely to be
polymorphisms may influence transcription and mRNA mainly free heavy chain (J. S. Hunt and P. Morales,
abundance (47). unpublished results) as ␤2m is missing. An antibody
recognizing HLA-G2 and -G6 shows that one or another
Patterns of expression of HLA-G of these isoforms is prominent in/on extravillous cytotro-
phoblast cells distal to the placental villi, cytotrophoblast
Figure 2 illustrates that placentas contain several dis- cells infiltrating the decidua, and some chorion mem-
tinct subpopulations of trophoblast cells that arise from brane cytotrophoblast cells (48) (Fig. 5). HLA-G2/G6 is
progenitor cells within the trophectoderm layer of the located in the same cells as HLA-G1 (43), suggesting that
blastocyst. These progenitor cells may merge to form a in women carrying the null allele (G*0105N), which does
syncytialized single cell layer that interfaces directly not encode functional HLA-G1 or -G5 protein isoforms,
with maternal blood or proliferate to form columns of HLA-G2/G6 may comprise an adequate substitute (30).

Figure 4. Variation in the 5⬘-upstream


regulatory region of HLA-G. Eight
unique haplotypes are defined by the
polymorphisms. Note that polymor-
phisms are frequently associated with
transcription factor binding sites and
could affect the efficiency of transcrip-
tion of HLA-G.

HLA-G AND PREGNANCY IMMUNE PRIVILEGE 685


rosine phosphorylation, SHP-1 association, and calcium
regulation, may be translated into expression of specific
genes in decidual leukocytes as required for program-
ming the cells into pregnancy-appropriate behavior.

FUNCTIONS OF HLA-G

Although it has been proposed that HLA-G may be an


evolutionary artifact without function (53), recent stud-
ies using HLA-G proteins from transfected cells indi-
cate that these proteins may regulate immune cells and
thus be integral to immune privilege in pregnancy.
Figure 6 shows that HLA-G proteins probably target all
of the major immune cell subsets. In the following
paragraphs we describe activities related to T and B
lymphocytes, NK cells and antigen-presenting cells
Figure 5. HLA-G2/G6 isoforms are induced during cytotro-
phoblast migration and invasion. First trimester decidua is (APC).
shown in the left panel; a cytotrophoblast column is shown in
the right panel. Immunostaining with the anti-HLA-G5 mAb HLA-G interactions with T lymphocytes
1-2C3 identifies positive cells in the maternal decidua, the
proximal cytotrophoblast column and trophoblast cells in the Wegmann and co-workers were the first to report that
villus (pink label, small arrows). Double staining with the
in pregnant mice, Th2 cytokine-producing lymphocytes
anti-HLA-G2/G6 mAb 26-2H11 identifies positive cells in the
decidua and at the leading edge of the cytotrophoblast cell flourish in preference to those producing Th1 (13).
column (brown label, large arrows). Original magnifications, Subsequent research in women has shown that failure
⫻200. to achieve this preference for Th2-type, anti-inflamma-
tory cytokines may lead to unsuccessful pregnancy (54).
Receptors for HLA-G The idea that trophoblast cell signals drive T cells into
this anti-inflammatory profile has emerged as a popular
explanation for maternal tolerance to the semialloge-
The original view of how regulation of the HLA class I
neic fetus.
antigens in fetal trophoblast cells might contribute to
Convincing evidence for an ability of HLA-G to
maternal tolerance during pregnancy was restricted to
influence T cells was first presented by Sanders et al.
the negative: a failure to express these antigens meant
(55), who showed that HLA-G-expressing cells bind to
that T lymphocytes would not recognize trophoblast
CD8␣-expressing cells. This finding has recently been
cells as foreign, thus allowing them to survive (49). The
confirmed by Shiroishi et al. (56). In both studies,
finding that subpopulations of trophoblast cells invad-
HLA-G binding to the ␣␣ CD8 homodimer, the molec-
ing the decidua express HLA class I antigens (50) drove
ular form expressed by a subset of T cells in the
investigators to revise their thinking and explore new
explanations for placental immune privilege.
Binding of trophoblast cell HLA-G to receptors that
inhibit activating signals on decidual leukocytes may be
the answer (Fig. 6). HLA-G appears to be recognized
mainly by immunoglobulin-like transcript (ILT) recep-
tors, which are expressed by T and B lymphocytes, as
well as by NK cells and mononuclear phagocytes (51),
and abrogate activating signals received by these cells.
Whether these ILT receptors or others, such as TcR
and the CD94/NKG2A HLA-E receptors on uterine
CD56bright NK cells, are more important to lymphocyte
recognition remains to be established. Early studies
suggested that ILT4 may be the main receptor for
HLA-G exhibited by monocyte/macrophages, the sec-
ond most populous leukocyte population in the human
decidua (6). However, ILT2 has not been formally
excluded as a mononuclear phagocyte recognition
entity, and recent experiments in a macrophage cell
Figure 6. Potential receptors on immune cells targeted by
line suggest these two receptors may exhibit isoform- HLA-G. The six subsets of leukocytes believed to be targeted
specific binding (52). This novel finding illustrates the by HLA-G and potential receptors for HLA-G on each cell
point that little is known of how HLA-G-activated type are shown. TcR, T cell receptor; ILT, immunoglobulin-
signaling pathways, which for ILT2/ILT4 include ty- like transcript; KIR, killer inhibitory receptor.

686 Vol. 19 May 2005 The FASEB Journal HUNT ET AL.


intestine and by NK cells (57), was evaluated. Yet most sis. The specific receptor capable of binding HLA-G is
T cells express the CD8␣␤ heterodimer, which acts as a not clearly defined, but could be either the CD8
coreceptor to the T cell receptor (TcR) and an essential molecule itself, as proposed by Contini et al. (66) and
signal transduction molecule during T cell activation. Shiroishi et al. (56), or the immunoglobulin-like tran-
Interaction between the TcR and HLA-G has not been script receptor 2 (ILT2), which transduces an inhibi-
demonstrated. Many investigators question whether tory signal (61) (Fig. 6). HLA-G1 and its soluble
binding to the TcR would be a primary function of counterpart HLA-G5 protect potential target cells from
HLA-G because of its limited polymorphism (58). How- lysis by antigen-specific cytotoxic T cells (61, 66, 72).
ever, it was recently shown that cytomegalovirus-derived The same protection by the smaller isoforms of mem-
peptides can stabilize surface HLA-G expression, lead- brane HLA-G, HLA-G2, -G3, and -G4 has now been
ing to HLA-G-restricted, cytomegalovirus-specific cyto- shown (73), which is consistent with the idea that
toxic T cell response in transgenic mice (59). This smaller isoforms compensate when HLA-G1 and -G5
strongly suggests that HLA-G can indeed act as a are absent in mothers who are homozygous for the
classical presenter of foreign peptide. In solving the G*0105N allele (74).
debate of whether there is any role for cooperative Intriguingly, HLA-G down-regulates expression of
physiological binding between HLA-G and the CD8␣ mRNA and protein in IFN-␥-treated blood mono-
␣␤TcR, it will be important to analyze HLA-G-derived nuclear cells without inducing apoptosis or altering
peptides from women with known human cytomega- CD3 expression (48). Although T cells and NK cells
lovirus infection. express CD8␣/␤, signals for CD8␣ specific message and
Functional studies have yielded even stronger evi- protein in Northern blots and immunoblots were
dence. In vitro studies have clearly demonstrated an strong, suggesting it is the more numerous T cell
ability of soluble and membrane-associated HLA-G to affected. Because the effects of HLA-G are highly
modulate cytokine release from human allogeneic concentration dependent and HLA-G-producing cells
peripheral blood mononuclear cells (60) and to have are in the placental bed, these results imply that soluble
a concentration-dependent effect on generation of isoforms of placental HLA-G might reduce the ability of
an allogeneic CTL response (61). Regarding initia- T cells to function effectively in the pregnant uterus but
tion of the acquired immune response, Le Maoult et be less potent in the periphery (48, 71).
al. (62) have shown that APC transfected with When considering the impact of HLA-G on T cells, it
HLA-G1 prevent proliferation of CD4⫹ T cells and is critical to bear in mind two facts: 1) T cells are not
direct them toward an immunosuppressive pheno- numerous in the decidua (6), and 2) trophoblast cells
type. Although Le Maoult et al. did not test CD4⫹ in normal, healthy placentas are unlikely to elicit a CTL
cells to ascertain whether they might be related to cell response. The inability of trophoblast cells to
the CD4⫹CD25bright T regulatory phenotype appar- stimulate CTL is due to the fact that polymorphic HLA
ently essential for pregnancy in mice (63) and possi- class I antigens are absent on syncytiotrophoblast and
bly in women (64), it is tempting to speculate this are scarce on other subpopulations. However, an ex-
might be the case. The idea that this phenotype ception might occur in the case of virally infected
might be the consequence of exposure to a murine extravillous trophoblast cells, where trophoblast cell-
Qa-2, which bears structural similarities to HLA-G associated HLAs might present antigen to CD8⫹ cells,
(65), is intriguing. thus becoming potential targets themselves.
Membrane-associated and/or soluble HLA-G may
play a critical role in regulating CD8⫹ T cells during HLA-G interactions with B lymphocytes
pregnancy by eliminating alloreactive (antipaternal) T
cells. Two independent groups have reported that ILTs appear to be major receptors on T lymphocytes
HLA-G induces CD8⫹ T cell apoptosis (66 – 68). In both and antigen-presenting cells that interact with HLA-G,
models, exposure of CD8⫹ T cells to soluble HLA-G was as described above. They are also present on B lympho-
shown to trigger surface expression and secretion of Fas cytes, which express ILT2 inhibitory receptors (75). So
ligand, resulting in death of the activated T cells far there is no evidence for binding of HLA-G to these
through the Fas/FasL pathway. These experiments receptors on B cells or of B cells responding directly to
were based on the well-documented ability of other HLA-G; yet the possibility exists that this occurs, be-
HLA antigens to induce Fas/FasL-related cell death, a cause production of antibodies to placental HLA-G
phenomenon that has been identified in patients un- occasionally occurs in pregnant women.
dergoing transplantation and is believed to dispose of A recent study in our laboratories demonstrated that
alloreactive CD8⫹ cells (69). In pregnancy, soluble maternal tolerance to HLA-G in terms of antibody
HLA-G might induce apoptosis in CD8⫹ cells that react production is the usual condition (4). Ninety-one per-
to paternal antigens; soluble HLA-G has been reported cent of mothers, as well as all women who had never
in maternal sera by several groups (70, 71). been pregnant and all men, lack antibodies to HLA-G
A second mechanism by which HLA-G may induce in their sera. Yet tolerance is not absolute: ⬃9% of
maternal tolerance to fetal antigens is to reduce or women who had undergone at least one pregnancy
prevent cytotoxic activity of CD8⫹ T cells against target generated anti-HLA-G antibodies that were readily
cells, possibly independent of inducing T cell apopto- identified in maternal sera by ELISA and immunoblot-

HLA-G AND PREGNANCY IMMUNE PRIVILEGE 687


ting. Whether a “mimic” antigen on microbes or other evolutionary move to avoid this risk. As noted above,
environmental molecules might stimulate these anti- these genes have few alleles and their products are
bodies is not known, but since the anti-HLA-G antibod- unlikely to be recognized as foreign by the mother.
ies were found only in women who had been pregnant, NK cells of an unusual phenotype, CD16–CD56bright,
some association with the state of pregnancy would be are abundant in first and second trimester decidua but
required. Not unexpectedly, maternal anti-HLA-G an- decline in number thereafter (6). These cells are poor
tibodies have no deleterious effect; all the women who killers of the usual NK targets, suggesting that environ-
developed these antibodies had multiple successful mental conditions in the pregnant uterus affect their
pregnancies. In this respect, circulating anti-HLA-G functions. Decidual NK cell cytotoxicity could be by-
antibodies resemble other anti-HLA antibodies, which passed by ligation of HLA class I antigens to one or
are common in pregnancy and do not damage the more NK cell surface inhibitory receptor. Transfection-
developing fetus. Because several HLA-G alleles have based assays initially suggested that interaction between
been identified, we tested the hypothesis that mothers HLA-G and the CD94/NKG2A heterodimer on the
would generate antibodies to foreign (paternal) alleles, surface of NK cells prevented cytolysis induced by NK
but found no relationship between exposure to a cells (77–79). Further investigation, however, led to the
foreign (paternal) HLA-G protein on placental cells realization that class Ib gene HLA-E utilizes a leader
and maternal production of anti-HLA-G (4). peptide derived from other class I proteins, including
How tolerance is achieved remains to be elucidated. HLA-G, to stabilize its own surface expression. Thus,
Potential routes include 1) activation of B cell ILT2 initial findings led to the erroneous conclusion that
receptors by circulating HLA-G5 or -G6, an idea that HLA-G was responsible for reducing NK-mediated cy-
remains untested, and 2) activation of ILT2 receptors tolysis via CD94/NKG2A, when in fact HLA-G expres-
on Th lymphocytes, as suggested by Le Maoult and sion only permitted expression of HLA-E, the true
colleagues (62). The potential role of APC driven into ligand for this inhibitory receptor (80, 81).
a B lymphocyte-inhibiting immunosuppressive profile Nonetheless, the idea that HLA-G serves as the major
should not be overlooked, as these cells are abundant at inhibitor of NK toxicity at the maternal fetal interface
the maternal-fetal interface where HLA-G isoforms are remains popular because experiments using HLA-G-
prominent. Alternatively, HLA-G produced in the pla- specific antibodies show that trophoblast cell HLA-G
centa during fetal life might well induce lifelong toler- inhibits NK-mediated cell death in the absence of
ance in the adult. HLA-E (82, 83). These effects could be mediated
In any event, data accumulated to date, though through the killer inhibitory receptor (KIR) 2DL4,
scarce, strongly support the idea that inhibition is through ILT2, or both (75, 84, 85). ILT2 has been
specific to HLA-G and that the tolerogenic mechanisms shown to bind HLA-G and is expressed by decidual NK
underlying specific repression of production of mater- cells, albeit the quantity of ILT2 on decidual NK cells
nal antibodies are immensely effective. The deficiency may be low (86 – 89). More controversial is a role for
of anti-HLA-G antibodies is not due to maternal, preg- KIR2DL4, which in soluble form binds to HLA-G-
nancy-associated reduction in B cell function. Mothers transfected cells (84, 85). More recently, however,
produce high levels of other types of antibodies in biochemical analysis of KIR2DL4 binding to HLA-G
order to provide defense against pathogens during monomers and dimers yielded only negative results,
pregnancy and assure transfer of protective antibodies raising the question of validity of HLA-G-KIR2DL4
to the fetus. The placenta supports this hyperproduc- interaction (90). Furthermore, decidual NK cells ap-
tion. A recent study from our laboratory shows that pear to express an abundance of CD94/NKG2A, the
human placentas produce two non-apoptosis-inducing receptor for HLA-E, suggesting this interaction may
TNF superfamily ligands: BAFF (also known as BlyS, or supercede that of HLA-G and its NK-expressed recep-
B lymphocyte stimulator, as well as TALL-1, THANK, tors (91). A final confounding observation against a
and zTNF4) and APRIL (a proliferation inducing li- role for HLA-G-mediated inhibition of NK cytolytic
gand), both of which sustain B lymphocytes (76). activity is that immortalized and primary trophoblast
Placental BAFF and APRIL therefore may bear a degree cells are resistant to NK cell-mediated lysis independent
of responsibility for maternal host defense and provi- of HLA class I expression (91–94), suggesting these
sion of antibodies to the fetus required in the postpar- cells use other strategies for self protection.
tum period. New information has recently surfaced regarding a
potential role for HLA-G/NK cell interactions at the
HLA-G interactions with NK cells maternal-fetal interface. KIR2DL4 ligation and treat-
ment of NK cells with HLA-G result in production of
A complete absence of HLA class I molecules on the cytokine interferon-␥ (IFN-␥) (95–97) but, as sug-
trophoblast cells could provide protection against CTL gested above, it remains to be determined whether this
but could target the trophoblast subpopulation migrat- commonality is causally linked. Even so, stimulation of
ing into the decidua (Fig. 2, upper insert) for destruc- NK cell production of IFN-␥ at the maternal-fetal
tion by resident NK cells, a leukocyte programmed to interface by HLA-G is an intriguing possibility. IFN-␥,
recognize and destroy HLA null cells. Expression of usually considered a proinflammatory cytokine, can
HLA class Ib genes HLA-E, -F and -G may be an drive cells into immunosuppressive profiles when

688 Vol. 19 May 2005 The FASEB Journal HUNT ET AL.


linked with other modulators, and could therefore cells have not been tested, mononuclear phagocytes and
paradoxically serve as an anti-inflammatory cytokine. decidual macrophages express the two well-described true
Whether HLA-G might stimulate IFN-␥-associated path- receptors for HLA-G: ILT2 and ILT4 (75, 121, 122). The
ways of vascular and decidual remodeling (98 –100) decidual dendritic cells are likely to exhibit these recep-
remains an intriguing possibility. tors; studies in mice show that HLA-G tetramers bind only
to dendritic cells (123, 124). The ability of the mice to
Interactions with antigen-presenting cells reject grafts is strikingly diminished in the presence of
HLA-G tetramers. In humans, an immune suppressive
Two populations of APC, macrophages and dendritic effect of HLA-G on APC or other immune cell function is
cells, reside in the human decidualized endometrium implied by the finding that high levels of HLA-G proteins
throughout pregnancy. These powerful, multifunctional in the sera of heart transplant recipients is positively
leukocytes are located in close proximity to invasive cy- associated with prolonged graft survival (125).
totrophoblast cells, uterine glandular epithelium, and The development of recombinant HLA-G5 (rG5)
uterine blood vessels (6, 101–104), and are proposed to and rG6 from eukaryotic cells (48) has permitted us to
play central roles in uterine and placental homeostasis as test the hypothesis that HLA-G drives mononuclear
well as immune modulation (2, 6, 101–105). phagocytes into an immune cell inhibitory profile. In
APC in pregnant endometrium include 1) CD14⫹ contrast to results of another group who reported that
macrophages (106, 107), 2) CD83⫹ mature dendritic G5 killed T cells via a Fas/FasL pathway (68), we found
cells (103, 104, 108), and 3) CD83– immature macro- that our rG5 and rG6 did not negatively affect blood
phage/dendritic cells. The immature cell type appears mononuclear cell viability (48) or the viability of APC
to be a family of related cell types, some of which are (52). This result is in accord with a study of monocyte-
dendritic cell (DC)-SIGN⫹CD14⫹ (109) and others are derived dendritic cells indicating that HLA-G does not
DC-SIGN–CD14–DEC-205⫹ (104). Alternatively, the im- decrease the viability of immature or mature dendritic
mature phenotype cluster may be composed of similar cells, alter differentiation of dendritic cells from blood
cells in different transitional stages. It has been sug- monocytes, or influence maturation of dendritic cells
gested that DC-SIGN⫹ cells are a precursor population (126). Recombinant G5 and rG6 drive mononuclear
capable of differentiating into macrophages or den- phagocytes into suppressive pathways. In particular,
dritic cells (110), an idea supported by the finding that both strongly stimulate TGF-␤1 production by activated
macrophages and immature dendritic cells have been APC. The condition of activation is extremely impor-
shown to trans-differentiate in vitro under the influ- tant, as clearly illustrated in the study by Morales et al.
ence of various cytokines (111). (48), where rG5 and rG6 reduced CD8␣ mRNA and
Decidual macrophages, which are activated in the protein in cells treated with the monocyte-activating
pregnant uterus as evidenced by expression of HLA factor IFN-␥ to levels the same as, not below, those of
class II, CD11c, and CD86 antigens (112), appear to be CD8 in unstimulated cells.
programmed for immunosuppression. These cells pro- Do APC themselves, when activated, express HLA-G?
duce the lymphocyte inhibitory molecule, prostaglan- This possibility was first suggested in a report from our
din E2 (113, 114), elicit reduced allogeneic and autol- laboratory indicating that IFN-␥-activated, but not rest-
ogous T cell responses in comparison with monocytes ing, mononuclear phagocytes contain HLA-G mRNA
(105, 115, 116), and spontaneously secrete anti-inflam- and protein (127). The idea that inflammatory condi-
matory cytokines such as interleukin (IL)-10 and trans- tions stimulate APC production of HLA-G has been
forming growth factor (TGF)-␤1 (52, 114, 117). More supported by numerous reports of HLA-G expression
recently, decidual macrophages have been reported to in macrophages and dendritic cells during cytomegalo-
express B7-H1 (16), ILT3 (117), DC-SIGN (104, 110, virus infection, lung carcinoma, nontumoral pulmo-
118), MS-1, and factor 13 (119), all of which are nary disease, and HIV infection (128 –130). Although
markers associated with immune evasion and activation immunohistochemical studies may be compromised by
of macrophages into a suppressive profile. Mature the use of antibodies that lack specificity, soluble iso-
CD83⫹ decidual dendritic cells appear to exhibit an forms of HLA-G have been identified by Western blot in
immune cell inhibitory profile. These cells secrete less the sera of male heart transplant patients (125). There
IL-12 than monocyte-derived dendritic cells and induce are indications that APC-derived HLA-G is functional.
Th2 cells when cocultured with naı̈ve CD4⫹ T cells. The LeMaoult et al. (62) showed that HLA-G1-transfected
C-type lectin, DC-SIGN, is used for immune evasion by myelomonocytic cell lines (KG1a and U937) suppress
several viral and bacterial pathogens, with ligand bind- CD4⫹ T cell proliferation in a mixed lymphocyte
ing to DC-SIGN altering cytokine production and anti- reaction, with the resultant T cells driven into an
gen presentation to the benefit of the pathogen (120). immune-suppressive phenotype. The converse idea that
Thus, this molecule may be of importance to immuno- resting APC are not producers of HLA-G is supported
suppression by the decidual CD83–/DC-SIGN⫹ macro- by a report from Laupeze et al., who were unable to
phage/dendritic cell-like cells. detect HLA-G expression in/on resting mononuclear
The question then arises as to how decidual APC are phagocytes or dendritic cells (131). It is therefore
driven into immune inhibitory profiles. Placental HLA-G puzzling that activated APC in the decidua reportedly
is a reasonable possibility. Although decidual dendritic fail to exhibit membrane or soluble HLA-G and do not

HLA-G AND PREGNANCY IMMUNE PRIVILEGE 689


contain a specific message (132). More work is needed immune suppression so as to facilitate pregnancy or
to understand local tissue conditions that might direct graft acceptance, as has been shown in the porcine
APC into production of HLA-G, as activation is clearly model (137).
not the only important factor.

PROSPECTS AND PREDICTIONS REFERENCES


1. Medawar, P. B. (1953) Some immunological and endocrino-
Human and nonhuman primate pregnancies provide logical problems raised by the evolution of viviparity in verte-
natural models for studying mechanisms of immune brates. Symp. Soc. Exp. Biol. 44, 320 –338
tolerance and features of immune privileged sites. Studies 2. Hunt, J. S. (1989) Cytokine networks in the uteroplacental
unit: macrophages as pivotal regulatory cells. J. Reprod. Immu-
conducted on pregnancy over the past half century have nol. 16, 1–17
provided immunologists with definitive proof that in 3. Marzi, M., Vigano, A., Trabattoni, D., Villa, M. L., Salvaggio, A.,
successful transplants, which include the fetus, “foreign” Clerici, E., and Clerici, M. (1996) Characterization of type 1
and type 2 cytokine production profile in physiologic and
tissue will have devised overlapping and complementary pathologic human pregnancy. Clin. Exp. Immunol. 106, 127–133
mechanisms to avoid rejection. Of these, selection of a 4. Hunt, J. S., Pace, J. L., Morales, P. J., and Ober, C. (2003)
gene with limited polymorphism, HLA-G, for expression, Immunogenicity of the soluble isoforms of HLA-G. Mol. Hum.
and derivation of an entire family of immunomodulatory Reprod. 9, 729 –735
5. Givan, A. L., White, H. D., Stern, J. E., Colby, E., Gosselin, E. J.,
glycoproteins by alternative splicing of the gene’s single Guyre, P. M., and Wira, C. R. (1997) Flow cytometric analysis of
message are among the cleverest. leukocytes in the human female reproductive tract: compari-
Studying HLA-G structure-function relationships re- son of fallopian tube, uterus, cervix, and vagina. Am. J. Reprod.
minds us of several unifying principles of immunity, Immunol. 38, 350 –359
6. Bulmer, J. N., Pace, D., and Ritson, A. (1988) Immunoregula-
including 1) the genetic variation provided by alleles, 2) tory cells in human decidua: morphology, immunohistochem-
the potential of polymorphisms outside the coding region istry and function. Reprod. Nutr. Dev. 28, 1599 –1613
to influence rates of transcription and regulate produc- 7. Sakaguchi, S. (2000) Regulatory T cells: key controllers of
immunologic self-tolerance. Cell 101, 455– 458
tion consistent with varying conditions and specific needs, 8. Read, S., and Powrie, F. (2001) CD4(⫹) regulatory T cells.
3) the power of alternative splicing, which has been Curr. Opin. Immunol. 13, 644 – 649
revealed in other systems to be directly related to func- 9. Polanczyk, M. J., Carson, B. D., Subramanian, S., Afentoulis,
tional diversity (133), 4) the central relationship between M., Vandenbark, A. A., Ziegler, S. F., and Offner, H. (2004)
Cutting edge: estrogen drives expansion of the CD4⫹CD25⫹
the concentration of an immunomodulatory molecule regulatory T cell compartment. J. Immunol. 173, 2227–2230
and its final effects, and 5) the precise, possibly cell 10. Heikkinen, J., Mottonen, M., Alanen, A., and Lassila, O. (2004)
type-specific conditions such as activation and inflamma- Phenotypic characterization of regulatory T cells in the human
decidua. Clin. Exp. Immunol. 136, 373–378
tion that may be required for stimulating production of 11. Simon, C., Pellicer, A., and Polan, M. L. (1995) Interleukin-1
the molecule and achieving desired effects. system crosstalk between embryo and endometrium in implan-
The major HLA-G function that has emerged to date tation. Hum. Reprod. 10, Suppl. 2, 43–54
is its ability to program cells into immunosuppressive 12. Hunt, J. S., Chen, H. L., Hu, X. L., and Tabibzadeh, S. (1992)
Tumor necrosis factor-alpha messenger ribonucleic acid and
phenotypes. Since HLA-G is produced in great amounts protein in human endometrium. Biol. Reprod. 47, 141–147
at the maternal-fetal interface, the implication is that 13. Wegmann, T. G., Lin, H., Guilbert, L., and Mosmann, T. R.
the immunosuppressive function must be important. (1993) Bidirectional cytokine interactions in the maternal-fetal
Yet definitive proof that HLA-G is required remains relationship: is successful pregnancy a TH2 phenomenon?
Immunol. Today 14, 353–356
elusive, as in vivo experiments are difficult to design. 14. Lysiak, J. J., Hunt, J., Pringle, G. A., and Lala, P. K. (1995)
Mice do not demonstrate a clear correlate of HLA-G, Localization of transforming growth factor beta and its natural
although Qa-2 appears to have some of the same inhibitor decorin in the human placenta and decidua through-
out gestation. Placenta 16, 221–231
structural characteristics as HLA-G and is strongly ex- 15. Hsi, B. L., Hunt, J. S., and Atkinson, J. P. (1991) Differential
pressed in placentas (65). Nonhuman primate placen- expression of complement regulatory proteins on subpopula-
tal cells transcribe and translate proteins from HLA-G- tions of human trophoblast cells. J. Reprod. Immunol. 19,
like genes, but the products are not identical to those 209 –223
16. Petroff, M. G., Chen, L., Phillips, T. A., Azzola, D., Sedlmayr, P.,
described above for HLA-G. For example, in baboons, and Hunt, J. S. (2003) B7 family molecules are favorably
no HLA-G6-like isoform is detectable (134). Thus, positioned at the human maternal-fetal interface. Biol. Reprod.
although all indications are that HLA-G serves as a key 68, 1496 –1504
17. Chen, H. L., Yang, Y. P., Hu, X. L., Yelavarthi, K. K., Fishback,
effector of the changes that occur in maternal immune J. L., and Hunt, J. S. (1991) Tumor necrosis factor alpha
cells with the onset of pregnancy, definitive in vivo mRNA and protein are present in human placental and
studies remain to be done. uterine cells at early and late stages of gestation. Am. J. Pathol.
Assessment of levels of HLA-G isoforms may well be 139, 327–335
18. Le Bouteiller, P. (1994) HLA class I chromosomal region,
useful in predicting the ability of mothers to establish genes, and products: facts and questions. Crit. Rev. Immunol. 14,
and maintain pregnancy (71, 135, 136) as well as 89 –129
determining the likelihood that patients will accept 19. Hunt, J. S., Andrews, G. K., and Wood, G. W. (1987) Normal
allografts (125). If such assessments are ultimately trophoblasts resist induction of class I HLA. J. Immunol. 138,
2481–2487
found to be reliable, recombinant HLA-G might be a 20. Grimsley, C., and Ober, C. (1997) Population genetic studies
powerful tool for inducing a desirable state of local of HLA-E: Evidence for selection. Hum. Immunol. 52, 33– 40

690 Vol. 19 May 2005 The FASEB Journal HUNT ET AL.


21. Ober, C., and Aldrich, C. L. (1997) HLA-G polymorphisms: 41. Hara, N., Fujii, T., Yamashita, T., Kozuma, S., Okai, T., and
neutral evolution or novel function? J. Reprod. Immunol. 36, Taketani, Y. (1996) Altered expression of human leukocyte
1–21 antigen G (HLA-G) on extravillous trophoblasts in preeclamp-
22. Hunt, J. S., and Orr, H. T. (1992) HLA and maternal-fetal sia: immunohistological demonstration with anti-HLA-G spe-
recognition. FASEB J. 6, 2344 –2348 cific antibody “87G” and anti-cytokeratin antibody “CAM5.2.”
23. Le Bouteiller, P., and Mallet, V. (1997) HLA-G and pregnancy. Am. J. Reprod. Immunol. 36, 349 –358
Rev. Reprod. 2, 7–13 42. Goldman-Wohl, D. S., Ariel, I., Greenfield, C., Hochner-Celni-
24. Hunt, J. S. (2002) Major histocompatibility antigens in repro- kier, D., Cross, J., Fisher, S., and Yagel, S. (2000) Lack of
duction. In The Endometrium (Glasser, S., Alpin, J., Guidice, L., human leukocyte antigen-G expression in extravillous tropho-
and Tabibzadeh, S., eds) pp. 405– 415, Taylor & Francis, blasts is associated with pre-eclampsia. Mol. Hum. Reprod. 6,
London and New York 88 –95
25. Park, B., Lee, S., Kim, E., Chang, S., Jin, M., and Ahn, K. (2001) 43. Lim, K. H., Zhou, Y., Janatpour, M., McMaster, M., Bass, K.,
The truncated cytoplasmic tail of HLA-G serves a quality- Chun, S. H., and Fisher, S. J. (1997) Human cytotrophoblast
control function in post-ER compartments. Immunity 15, 213– differentiation/invasion is abnormal in pre-eclampsia. Am. J.
224 Pathol. 151, 1809 –1818
26. Ishitani, A., and Geraghty, D. E. (1992) Alternative splicing of 44. Aldrich, C., Verp, M. S., Walker, M. A., and Ober, C. (2000) A
HLA-G transcripts yields proteins with primary structures re- null mutation in HLA-G is not associated with preeclampsia or
sembling both class I and class II antigens. Proc. Natl. Acad. Sci. intrauterine growth retardation. J. Reprod. Immunol. 47, 41– 48
USA 89, 3947–3951 45. O'Brien, M., McCarthy, T., Jenkins, D., Paul, P., Dausset, J.,
27. Morales, P. J., Pace, J. L., Platt, J. S., Phillips, T. A., Morgan, K., Carosella, E. D., and Moreau, P. (2001) Altered HLA-G tran-
Fazleabas, A. T., and Hunt, J. S. (2003) Placental cell expres- scription in pre-eclampsia is associated with allele specific
sion of HLA-G2 isoforms is limited to the invasive trophoblast inheritance: possible role of the HLA-G gene in susceptibility
phenotype. J. Immunol. 171, 6215– 6224 to the disease. Cell. Mol. Life Sci. 58, 1943–1949
28. Fujii, T., Ishitani, A., and Geraghty, D. E. (1994) A soluble form 46. Hviid, T. V., Sorensen, S., and Morling, N. (1999) Polymor-
of the HLA-G antigen is encoded by a messenger ribonucleic phism in the regulatory region located more than 1.1 kilobases
acid containing intron 4. J. Immunol. 153, 5516 –5524 5⬘ to the start site of transcription, the promoter region, and
29. Suárez, M. B., Morales, P., Castro, M. J., Fernandez, V., Varela, exon 1 of the HLA-G gene. Hum. Immunol. 60, 1237–1244
P., Alvarez, M., Martinez-Laso, J., and Arnaiz-Villena, A. (1997) 47. Ober, C., Aldrich, C. L., Chervoneva, I., Billstrand, C., Rahi-
A new HLA-G allele (HLA-G*0105N) and its distribution in the mov, F., Gray, H. L., and Hyslop, T. (2003) Variation in the
Spanish population. Immunogenetics 45, 464 – 465 HLA-G promoter region influences miscarriage rates. Am. J.
30. Ober, C., Aldrich, C., Rosinsky, B., Robertson, A., Walker, Hum. Genet. 72, 1425–1435
M. A., Willadsen, S., Verp, M. S., Geraghty, D. E., and Hunt, 48. Morales, P. J., Pace, J. L., Platt, J. S., Phillips, T. A., Morgan, K.,
J. S. (1998) HLA-G1 protein expression is not essential for fetal Fazleabas, A. T., and Hunt, J. S. (2003) Placental cell expres-
survival. Placenta 19, 127–132 sion of HLA-G2 isoforms is limited to the invasive trophoblast
31. Hviid, T. V., Christiansen, O. B., Johansen, J. K., Hviid, U. R., phenotype. J. Immunol. 171, 6215– 6224
Lundegaard, C., Moller, C., and Morling, N. (2001) Charac- 49. Faulk, W. P., and Temple, A. (1976) Distribution of beta2
terization of a new HLA-G allele encoding a nonconservative microglobulin and HLA in chorionic villi of human placentae.
amino acid substitution in the alpha3 domain (exon 4) and its Nature (London) 262, 799 – 802
relevance to certain complications in pregnancy. Immunogenet- 50. Redman, C. W., McMichael, A. J., Stirrat, G. M., Sunderland,
ics 53, 48 –53 C. A., and Ting, A. (1984) Class 1 major histocompatibility
32. Castro, M. J., Morales, P., Rojo-Amigo, R., Martinez-Laso, J., complex antigens on human extra-villous trophoblast. Immu-
Allende, L., Varela, P., Garcia-Berciano, M., Guillen-Perales, J., nology 52, 457– 468
and Arnaiz-Villena, A. (2000) Homozygous HLA-G*0105N
51. Allan, D. S., McMichael, A. J., and Braud, V. M. (2000) The ILT
healthy individuals indicate that membrane-anchored HLA-G1
family of leukocyte receptors. Immunobiology 202, 34 – 41
molecule is not necessary for survival. Tissue Antigens 56,
52. McIntire, R. H., Morales, P. J., Petroff, M. G., Colonna, M., and
232–329
Hunt, J. S. (2004) Recombinant HLA-G5 and -G6 drive U937
33. Menier, C., Riteau, B., Dausset, J., Carosella, E. D., and
myelomonocytic cell production of TGF-{beta}1. J. Leukoc. Biol.
Rouas-Freiss, N. (2000) HLA-G truncated isoforms can substi-
76, 1220 –1228
tute for HLA-G1 in fetal survival. Hum. Immunol. 61, 1118 –1125
53. Parham, P. (1996) Immunology: keeping mother at bay. Curr.
34. Aldrich, C. L., Stephenson, M. D., Karrison, T., Odem, R. R.,
Biol. 6, 638 – 641
Branch, D. W., Scott, J. R., Schreiber, J. R., and Ober, C. (2001)
HLA-G genotypes and pregnancy outcome in couples with 54. Piccinni, M. P., Beloni, L., Livi, C., Maggi, E., Scarselli, G., and
unexplained recurrent miscarriage. Mol. Hum. Reprod. 7, 1162– Romagnani, S. (1998) Defective production of both leukemia
1172 inhibitory factor and type 2 T-helper cytokines by decidual T
35. Pfeiffer, K. A., Fimmers, R., Engels, G., van Der Ven, H., and cells in unexplained recurrent abortions. Nat. Med. 4, 1020 –
van Der Ven, K. (2001) The HLA-G genotype is potentially 1024
associated with idiopathic recurrent spontaneous abortion. 55. Sanders, S. K., Giblin, P. A., and Kavathas, P. (1991) Cell-cell
Mol. Hum. Reprod. 7, 373–378 adhesion mediated by CD8 and human histocompatibility
36. Harrison, G. A., Humphrey, K. E., Jakobsen, I. B., and Cooper, leukocyte antigen G, a nonclassical major histocompatibility
D. W. (1993) A 14 bp deletion polymorphism in the HLA-G complex class 1 molecule on cytotrophoblasts. J. Exp. Med. 174,
gene. Hum. Mol. Genet. 2, 2200 737–740
37. Hiby, S. E., King, A., Sharkey, A., and Loke, Y. W. (1999) 56. Shiroishi, M., Tsumoto, K., Amano, K., Shirakihara, Y., Col-
Molecular studies of trophoblast HLA-G: polymorphism, iso- onna, M., Braud, V. M., Allan, D. S., Makadzange, A., Rowland-
forms, imprinting and expression in preimplantation embryo. Jones, S., Willcox, B., et al. (2003) Human inhibitory receptors
Tissue Antigens 53, 1–13 Ig-like transcript 2 (ILT2) and ILT4 compete with CD8 for
38. Rousseau, P., Le Discorde, M., Mouillot, G., Marcou, C., MHC class I binding and bind preferentially to HLA-G. Proc.
Carosella, E. D., and Moreau, P. (2003) The 14 bp deletion- Natl. Acad. Sci. USA 100, 8856 – 8861
insertion polymorphism in the 3⬘ UT region of the HLA-G 57. Gangadharan, D., and Cheroutre, H. (2004) The CD8 isoform
gene influences HLA-G mRNA stability. Hum. Immunol. 64, CD8alphaalpha is not a functional homologue of the TCR
1005–1010 coreceptor CD8alphabeta. Curr. Opin. Immunol. 16, 264 –270
39. Hviid, T. V., Hylenius, S., Rorbye, C., and Nielsen, L. G. (2003) 58. Ishitani, A., Sageshima, N., Lee, N., Dorofeeva, N., Hatake, K.,
HLA-G allelic variants are associated with differences in the Marquardt, H., and Geraghty, D. E. (2003) Protein expression
HLA-G mRNA isoform profile and HLA-G mRNA levels. Immu- and peptide binding suggest unique and interacting functional
nogenetics 55, 63–79 roles for HLA-E, F, and G in maternal-placental immune
40. Colbern, G. T., Chiang, M. H., and Main, E. K. (1994) recognition. J. Immunol. 171, 1376 –1384
Expression of the nonclassic histocompatibility antigen HLA-G 59. Lenfant, F., Pizzato, N., Liang, S., Davrinche, C., Le Bouteiller,
by preeclamptic placenta. Am. J. Obstet. Gynecol. 170, 1244 –1250 P., and Horuzsko, A. (2003) Induction of HLA-G-restricted

HLA-G AND PREGNANCY IMMUNE PRIVILEGE 691


human cytomegalovirus pp65 (UL83)-specific cytotoxic T lym- involved in recognition of HLA-G by decidual and peripheral
phocytes in HLA-G transgenic mice. J. Gen. Virol. 84, 307–317 blood NK cells. J. Immunol. 159, 1072–1075
60. Kanai, T., Fujii, T., Kozuma, S., Yamashita, T., Miki, A., 79. Pende, D., Sivori, S., Accame, L., Pareti, L., Falco, M., Ger-
Kikuchi, A., and Taketani, Y. (2001) Soluble HLA-G influences aghty, D., Le Bouteiller, P., Moretta, L., and Moretta, A. (1997)
the release of cytokines from allogeneic peripheral blood HLA-G recognition by human natural killer cells. Involvement
mononuclear cells in culture. Mol. Hum. Reprod. 7, 195–200 of CD94 both as inhibitory and as activating receptor complex.
61. Kapasi, K., Albert, S. E., Yie, S., Zavazava, N., and Librach, C. L. Eur. J. Immunol. 27, 1875–1880
(2000) HLA-G has a concentration-dependent effect on the 80. Lee, N., Goodlett, D. R., Ishitani, A., Marquardt, H., and
generation of an allo-CTL response. Immunology 101, 191–200 Geraghty, D. E. (1998) HLA-E surface expression depends on
62. LeMaoult, J., Krawice-Radanne, I., Dausset, J., and Carosella, binding of TAP-dependent peptides derived from certain HLA
E. D. (2004) HLA-G1-expressing antigen-presenting cells in- class I signal sequences. J. Immunol. 160, 4951– 4960
duce immunosuppressive CD4⫹ T cells. Proc. Natl. Acad. Sci. 81. Lee, N., Llano, M., Carretero, M., Ishitani, A., Navarro, F.,
USA 101, 7064 –7069 Lopez-Botet, M., and Geraghty, D. E. (1998) HLA-E is a major
63. Aluvihare, V. R., Kallikourdis, M., and Betz, A. G. (2004) ligand for the natural killer inhibitory receptor CD94/NKG2A.
Regulatory T cells mediate maternal tolerance to the fetus. Nat. Proc. Natl. Acad. Sci. USA 95, 5199 –5204
Immunol. 5, 266 –271 82. Rouas-Freiss, N., Goncalves, R. M., Menier, C., Dausset, J., and
64. Sasaki, Y., Sakai, M., Miyazaki, S., Higuma, S., Shiozaki, A., and Carosella, E. D. (1997) Direct evidence to support the role of
Saito, S. (2004) Decidual and peripheral blood CD4⫹CD25⫹ HLA-G in protecting the fetus from maternal uterine natural
regulatory T cells in early pregnancy subjects and spontaneous killer cytolysis. Proc. Natl. Acad. Sci. USA 94, 11520 –11525
abortion cases. Mol. Hum. Reprod. 10, 347–353 83. Khalil-Daher, I., Riteau, B., Menier, C., Sedlik, C., Paul, P.,
65. Cao, W., Brenner, C. A., Alikani, M., Cohen, J., and Warner, Dausset, J., Carosella, E. D., and Rouas-Freiss, N. (1999) Role of
C. M. (1999) Search for a human homologue of the mouse Ped HLA-G versus HLA-E on NK function: HLA-G is able to inhibit
gene. Mol. Hum. Reprod. 5, 541–547 NK cytolysis by itself. J. Reprod. Immunol. 43, 175–182
66. Contini, P., Ghio, M., Poggi, A., Filaci, G., Indiveri, F., Ferrone, 84. Cantoni, C., Verdiani, S., Falco, M., Pessino, A., Cilli, M.,
S., and Puppo, F. (2003) Soluble HLA-A,-B,-C and -G molecules Conte, R., Pende, D., Ponte, M., Mikaelsson, M. S., Moretta, L.,
induce apoptosis in T and NK CD8⫹ cells and inhibit cytotoxic et al. (1998) p49, a putative HLA class I-specific inhibitory NK
T cell activity through CD8 ligation. Eur. J. Immunol. 33, receptor belonging to the immunoglobulin superfamily. Eur.
125–134 J. Immunol. 28, 1980 –1990
67. Fournel, S., Aguerre-Girr, M., Huc, X., Lenfant, F., Alam, A., 85. Rajagopalan, S., and Long, E. O. (1999) A human histocom-
Toubert, A., Bensussan, A., and Le Bouteiller, P. (2000) patibility leukocyte antigen (HLA)-G-specific receptor ex-
Cutting edge: soluble HLA-G1 triggers CD95/CD95 ligand- pressed on all natural killer cells. J. Exp. Med. 189, 1093–1100
mediated apoptosis in activated CD8⫹ cells by interacting with 86. Navarro, F., Llano, M., Bellon, T., Colonna, M., Geraghty,
CD8. J. Immunol. 164, 6100 – 6104 D. E., and Lopez-Botet, M. (1999) The ILT2(LIR1) and
68. Solier, C., Aguerre-Girr, M., Lenfant, F., Campan, A., Berrebi, CD94/NKG2A NK cell receptors respectively recognize
A., Rebmann, V., Grosse-Wilde, H., and Le Bouteiller, P. HLA-G1 and HLA-E molecules co-expressed on target cells.
(2002) Secretion of pro-apoptotic intron 4-retaining soluble Eur. J. Immunol. 29, 277–283
HLA-G1 by human villous trophoblast. Eur. J. Immunol. 32, 87. Allan, D. S., Colonna, M., Lanier, L. L., Churakova, T. D.,
3576 –3586 Abrams, J. S., Ellis, S. A., McMichael, A. J., and Braud, V. M.
(1999) Tetrameric complexes of human histocompatibility
69. Zavazava, N. (1998) Soluble HLA class I molecules: biological
leukocyte antigen (HLA)-G bind to peripheral blood my-
significance and clinical implications. Mol. Med. Today 4,
elomonocytic cells. J. Exp. Med. 189, 1149 –1156
116 –121
88. Trundley, A., and Moffett, A. (2004) Human uterine leuko-
70. Rebmann, V., Pfeiffer, K., Passler, M., Ferrone, S., Maier, S.,
cytes and pregnancy. Tissue Antigens 63, 1–12
Weiss, E., and Grosse-Wilde, H. (1999) Detection of soluble
89. Moffett-King, A. (2002) Natural killer cells and pregnancy. Nat.
HLA-G molecules in plasma and amniotic fluid. Tissue Antigens
Rev. Immunol. 2, 656 – 663
53, 14 –22
90. Boyson, J. E., Erskine, R., Whitman, M. C., Chiu, M., Lau, J. M.,
71. Hunt, J. S., Jadhav, L., Chu, W., Geraghty, D. E., and Ober, C. Koopman, L. A., Valter, M. M., Angelisova, P., Horejsi, V., and
(2000) Soluble HLA-G circulates in maternal blood during Strominger, J. L. (2002) Disulfide bond-mediated dimerization
pregnancy. Am. J. Obstet. Gynecol. 183, 682– 688 of HLA-G on the cell surface. Proc. Natl. Acad. Sci. USA 99,
72. Le Gal, F. A., Riteau, B., Sedlik, C., Khalil-Daher, I., Menier, C., 16180 –16185
Dausset, J., Guillet, J. G., Carosella, E. D., and Rouas-Freiss, N. 91. King, A., Allan, D. S., Bowen, M., Powis, S. J., Joseph, S., Verma,
(1999) HLA-G-mediated inhibition of antigen-specific cyto- S., Hiby, S. E., McMichael, A. J., Loke, Y. W., and Braud, V. M.
toxic T lymphocytes. Int. Immunol. 11, 1351–1356 (2000) HLA-E is expressed on trophoblast and interacts with
73. Riteau, B., Rouas-Freiss, N., Menier, C., Paul, P., Dausset, J., CD94/NKG2 receptors on decidual NK cells. Eur. J. Immunol.
and Carosella, E. D. (2001) HLA-G2, -G3, and -G4 isoforms 30, 1623–1631
expressed as nonmature cell surface glycoproteins inhibit NK 92. Zdravkovic, M., Aboagye-Mathiesen, G., Guimond, M. J.,
and antigen-specific CTL cytolysis. J. Immunol. 166, 5018 –5026 Hager, H., Ebbesen, P., and Lala, P. K. (1999) Susceptibility of
74. Ober, C., Aldrich, C., Rosinsky, B., Robertson, A., Walker, MHC class I expressing extravillous trophoblast cell lines to
M. A., Willadsen, S., Verp, M. S., Geraghty, D. E., and Hunt, killing by natural killer cells. Placenta 20, 431– 440
J. S. (1998) HLA-G1 protein expression is not essential for fetal 93. Avril, T., Jarousseau, A. C., Watier, H., Boucraut, J., Le Bou-
survival. Placenta 19, 127–132 teiller, P., Bardos, P., and Thibault, G. (1999) Trophoblast cell
75. Colonna, M., Navarro, F., Bellon, T., Llano, M., Garcia, P., line resistance to NK lysis mainly involves an HLA class
Samaridis, J., Angman, L., Cella, M., and Lopez-Botet, M. I-independent mechanism. J. Immunol. 162, 5902–5909
(1997) A common inhibitory receptor for major histocompat- 94. Sivori, S., Parolini, S., Marcenaro, E., Millo, R., Bottino, C., and
ibility complex class I molecules on human lymphoid and Moretta, A. (2000) Triggering receptors involved in natural
myelomonocytic cells. J. Exp. Med. 186, 1809 –1818 killer cell-mediated cytotoxicity against choriocarcinoma cell
76. Phillips, T. A., Ni, J., and Hunt, J. S. (2003) Cell-specific lines. Hum. Immunol. 61, 1055–1058
expression of B lymphocyte (APRIL, BLyS) and Th2 (CD30L/ 95. Rajagopalan, S., Fu, J., and Long, E. O. (2001) Cutting edge:
CD153) -promoting tumor necrosis factor superfamily ligands induction of IFN-gamma production but not cytotoxicity by the
in human placentas. J. Leukoc. Biol. 74, 81– 87 killer cell Ig-like receptor KIR2DL4 (CD158d) in resting NK
77. Perez-Villar, J. J., Melero, I., Navarro, F., Carretero, M., Bellon, cells. J. Immunol. 167, 1877–1881
T., Llano, M., Colonna, M., Geraghty, D. E., and Lopez-Botet, 96. Kikuchi-Maki, A., Yusa, S., Catina, T. L., and Campbell, K. S.
M. (1997) The CD94/NKG2-A inhibitory receptor complex is (2003) KIR2DL4 is an IL-2-regulated NK cell receptor that
involved in natural killer cell-mediated recognition of cells exhibits limited expression in humans but triggers strong
expressing HLA-G1. J. Immunol. 158, 5736 –5743 IFN-gamma production. J. Immunol. 171, 3415–3425
78. Soderstrom, K., Corliss, B., Lanier, L. L., and Phillips, J. H. 97. van der Meer, A., Lukassen, H. G., van Lierop, M. J., Wijnands,
(1997) CD94/NKG2 is the predominant inhibitory receptor F., Mosselman, S., Braat, D. D., and Joosten, I. (2004) Mem-

692 Vol. 19 May 2005 The FASEB Journal HUNT ET AL.


brane-bound HLA-G activates proliferation and interferon- 119. Cupurdija, K., Azzola, D., Hainz, U., Gratchev, A., Heitger, A.,
gamma production by uterine natural killer cells. Mol. Hum. Takikawa, O., Goerdt, S., Wintersteiger, R., Dohr, G., and
Reprod. 10, 189 –195 Sedlmayr, P. (2004) Macrophages of human first trimester
98. Ashkar, A. A., and Croy, B. A. (1999) Interferon-gamma decidua express markers associated to alternative activation.
contributes to the normalcy of murine pregnancy. Biol. Reprod. Am. J. Reprod. Immunol. 51, 117–122
61, 493–502 120. van Kooyk, Y., and Geijtenbeek, T. B. (2003) DC-SIGN: escape
99. Ashkar, A. A., Di Santo, J. P., and Croy, B. A. (2000) Interferon mechanism for pathogens. Nat. Rev. Immunol. 3, 697–709
gamma contributes to initiation of uterine vascular modifica- 121. Petroff, M. G., Sedlmayr, P., Azzola, D., and Hunt, J. S. (2002)
tion, decidual integrity, and uterine natural killer cell matura- Decidual macrophages are potentially susceptible to inhibition by
tion during normal murine pregnancy. J. Exp. Med. 192, class Ia and class Ib HLA molecules. J. Reprod. Immunol. 56, 3–17
259 –270 122. Samaridis, J., and Colonna, M. (1997) Cloning of novel immu-
100. Li, X. F., Charnock-Jones, D. S., Zhang, E., Hiby, S., Malik, S., noglobulin superfamily receptors expressed on human my-
Day, K., Licence, D., Bowen, J. M., Gardner, L., King, A., et al. eloid and lymphoid cells: structural evidence for new stimula-
(2001) Angiogenic growth factor messenger ribonucleic acids tory and inhibitory pathways. Eur. J. Immunol. 27, 660 – 665
in uterine natural killer cells. J. Clin. Endocrinol. Metab. 86, 123. Horuzsko, A., Lenfant, F., Munn, D. H., and Mellor, A. L.
1823–1834 (2001) Maturation of antigen-presenting cells is compromised
101. Vince, G. S., and Johnson, P. M. (1996) Immunobiology of in HLA-G transgenic mice. Int. Immunol. 13, 385–394
human uteroplacental macrophages–friend and foe? Placenta 124. Liang, S., Baibakov, B., and Horuzsko, A. (2002) HLA-G
17, 191–199 inhibits the functions of murine dendritic cells via the PIR-B
102. Nehemiah, J. L., Schnitzer, J. A., Schulman, H., and Novikoff, immune inhibitory receptor. Eur. J. Immunol. 32, 2418 –2426
A. B. (1981) Human chorionic trophoblasts, decidual cells, 125. Lila, N., Amrein, C., Guillemain, R., Chevalier, P., La-
and macrophages: a histochemical and electron microscopic tremouille, C., Fabiani, J. N., Dausset, J., Carosella, E. D., and
study. Am. J. Obstet. Gynecol. 140, 261–268 Carpentier, A. (2002) Human leukocyte antigen-G expression
103. Kammerer, U., Schoppet, M., McLellan, A. D., Kapp, M., after heart transplantation is associated with a reduced inci-
Huppertz, H. I., Kampgen, E., and Dietl, J. (2000) Human dence of rejection. Circulation 105, 1949 –1954
decidua contains potent immunostimulatory CD83(⫹) den- 126. Le Friec, G., Laupeze, B., Fardel, O., Sebti, Y., Pangault, C.,
dritic cells. Am. J. Pathol. 157, 159 –169 Guilloux, V., Beauplet, A., Fauchet, R., and Amiot, L. (2003)
104. Gardner, L., and Moffett, A. (2003) Dendritic cells in the Soluble HLA-G inhibits human dendritic cell-triggered alloge-
human decidua. Biol. Reprod. 69, 1438 –1446 neic T-cell proliferation without altering dendritic differentia-
105. Hunt, J. S. (1990) Current topic: the role of macrophages in tion and maturation processes. Hum. Immunol. 64, 752–761
the uterine response to pregnancy. Placenta 11, 467– 475 127. Yang, Y., Chu, W., Geraghty, D. E., and Hunt, J. S. (1996)
106. Starkey, P. M., Sargent, I. L., and Redman, C. W. (1988) Cell Expression of HLA-G in human mononuclear phagocytes and
populations in human early pregnancy decidua: characteriza- selective induction by IFN-gamma. J. Immunol. 156, 4224 – 4231
tion and isolation of large granular lymphocytes by flow 128. Onno, M., Pangault, C., Le Friec, G., Guilloux, V., Andre, P.,
cytometry. Immunology 65, 129 –134 and Fauchet, R. (2000) Modulation of HLA-G antigens expres-
107. Vince, G. S., Starkey, P. M., Jackson, M. C., Sargent, I. L., and sion by human cytomegalovirus: specific induction in activated
Redman, C. W. (1990) Flow cytometric characterisation of cell macrophages harboring human cytomegalovirus infection.
populations in human pregnancy decidua and isolation of J. Immunol. 164, 6426 – 6434
decidual macrophages. J. Immunol. Methods 132, 181–189 129. Onno, M., Le Friec, G., Pangault, C., Amiot, L., Guilloux, V.,
108. Miyazaki, S., Tsuda, H., Sakai, M., Hori, S., Sasaki, Y., Futatani, Drenou, B., Caulet-Maugendre, S., Andre, P., and Fauchet, R.
T., Miyawaki, T., and Saito, S. (2003) Predominance of Th2- (2000) Modulation of HLA-G antigens expression in my-
promoting dendritic cells in early human pregnancy decidua. elomonocytic cells. Hum. Immunol. 61, 1086 –1094
J. Leukoc. Biol. 74, 514 –522 130. Lozano, J. M., Gonzalez, R., Kindelan, J. M., Rouas-Freiss, N.,
109. Kammerer, U., Eggert, A. O., Kapp, M., McLellan, A. D., Caballos, R., Dausset, J., Carosella, E. D., and Pena, J. (2002)
Geijtenbeek, T. B., Dietl, J., van Kooyk, Y., and Kampgen, E. Monocytes and T lymphocytes in HIV-1-positive patients ex-
(2003) Unique appearance of proliferating antigen-presenting press HLA-G molecule. AIDS 16, 347–351
cells expressing DC-SIGN (CD209) in the decidua of early 131. Laupeze, B., Fardel, O., Onno, M., Bertho, N., Drenou, B.,
human pregnancy. Am. J. Pathol. 162, 887– 896 Fauchet, R., and Amiot, L. (1999) Differential expression of
110. Soilleux, E. J., Morris, L. S., Leslie, G., Chehimi, J., Luo, Q., major histocompatibility complex class Ia, Ib, and II molecules
Levroney, E., Trowsdale, J., Montaner, L. J., Doms, R. W., on monocytes-derived dendritic and macrophagic cells. Hum.
Weissman, D., et al. (2002) Constitutive and induced expres- Immunol. 60, 591–597
sion of DC-SIGN on dendritic cell and macrophage subpopu- 132. Sedlmayr, P., Morales, P., Trummer, S., Wascher, K., Azzola,
lations in situ and in vitro. J. Leukoc. Biol. 71, 445– 457 D., Blaschitz, A., Hammer, A., Walcher, W., Wintersteiger, R.,
111. Palucka, K. A., Taquet, N., Sanchez-Chapuis, F., and Gluck- and Dohr, G. (2002) Absence of HLA-G expression in macro-
man, J. C. (1998) Dendritic cells as the terminal stage of phages of human decidua. Am. J. Reprod. Immunol. 48, 96 –102
monocyte differentiation. J. Immunol. 160, 4587– 4595 133. Trowbridge, I. S., and Thomas, M. L. (1994) CD45: an emerg-
112. Hunt, J. S., and Robertson, S. A. (1996) Uterine macrophages ing role as a protein tyrosine phosphatase required for lympho-
and environmental programming for pregnancy success. J. cyte activation and development. Annu. Rev. Immunol. 12, 85–116
Reprod. Immunol. 32, 1–25 134. Langat, D. K., Morales, P. J., Fazleabas, A. T., Mwenda, J. M.,
113. Parhar, R. S., Kennedy, T. G., and Lala, P. K. (1988) Suppres- and Hunt, J. S. (2002) Baboon placentas express soluble and
sion of lymphocyte alloreactivity by early gestational human membrane-bound Paan-AG proteins encoded by alternatively
decidua. I. Characterization of suppressor cells and suppressor spliced transcripts of the class Ib major histocompatibility
molecules. Cell. Immunol. 116, 392– 410 complex gene, Paan-AG. Immunogenetics 54, 164 –173
114. Hunt, J. S. (1994) Immunologically relevant cells in the uterus. 135. Pfeiffer, K. A., Rebmann, V., Passler, M., van der Ven, K., van
Biol. Reprod. 50, 461– 466 der Ven, H., Krebs, D., and Grosse-Wilde, H. (2000) Soluble
115. Hunt, J. S., and Pollard, J. W. (1992) Macrophages in the HLA levels in early pregnancy after in vitro fertilization. Hum.
uterus and placenta. Curr. Top. Microbiol. Immunol. 181, 39 – 63 Immunol. 61, 559 –564
116. Mizuno, M., Aoki, K., and Kimbara, T. (1994) Functions of 136. Rebmann, V., van der Ven, K., Passler, M., Pfeiffer, K., Krebs,
macrophages in human decidual tissue in early pregnancy. D., and Grosse-Wilde, H. (2001) Association of soluble HLA-G
Am. J. Reprod. Immunol. 31, 180 –188 plasma levels with HLA-G alleles. Tissue Antigens 57, 15–21
117. Heikkinen, J., Mottonen, M., Komi, J., Alanen, A., and Lassila, 137. Sasaki, H., Xu, X. C., Smith, D., Shenoy, S., Lowell, J., Howard,
O. (2003) Phenotypic characterization of human decidual T., and Mohanahumar, T. (1999) HLA-G expression protects
macrophages. Clin. Exp. Immunol. 131, 498 –505 porcine endothelial cells from xenogeneic cytotoxicity medi-
118. Soilleux, E. J., Morris, L. S., Lee, B., Pohlmann, S., Trowsdale, ated by human natural killer cells. Transplant. Proc. 31, 953–954
J., Doms, R. W., and Coleman, N. (2001) Placental expression
of DC-SIGN may mediate intrauterine vertical transmission of Received for publication September 7, 2004.
HIV. J. Pathol. 195, 586 –592 Accepted for publication December 22, 2004.

HLA-G AND PREGNANCY IMMUNE PRIVILEGE 693

View publication stats

You might also like