Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

STATE-OF-THE-ART REVIEW

NOD-like receptors interfacing the immune and


reproductive systems
Hanne Van Gorp1,2*, Anna Kuchmiy1,2*, Filip Van Hauwermeiren1,2 and Mohamed Lamkanfi1,2
1 Department of Medical Protein Research, VIB, Ghent, Belgium
2 Department of Biochemistry, Ghent University, Ghent, Belgium

Keywords Nucleotide-binding oligomerization domain receptors (NOD-like receptors,


disease, embryogenesis, imprinting, NLRs) are intracellular proteins that are chiefly known for their critical
inflammasomes, maternal effect gene,
functions in inflammatory responses and host defense against microbial
mole, NOD-like receptors, oocyte,
pathogens. Several NLRs have been demonstrated to assemble inflamma-
reproduction, subcortical maternal complex
somes or to engage transcriptional signaling cascades that result in the pro-
duction of pro-inflammatory cytokines and bactericidal factors. In recent
Correspondence years, NLRs have also emerged as key regulators of early mammalian
M. Lamkanfi, Department of Medical embryogenesis and reproduction. A subset of phylogenetically related
Protein Research, VIB, Albert Baertsoenkaai NLRs represents a new class of maternal effect genes that are highly
3, B-9000 Ghent, Belgium
expressed in maturing oocytes and pre-implantation embryos. Mutations in
Fax: +32 9 264 9490
several of these NLRs have been linked to hereditary reproductive defects
Tel: +32 9 264 9341
E-mail: mohamed.lamkanfi@vib-ugent.be and imprinting diseases. In this review, we discuss the expression profiles,
the emerging functions and molecular mode of action of these NLRs with
*These authors contributed equally newly recognized roles at the interfaces of the immune and reproductive
systems. In addition, we provide an overview of coding mutations in NLRs
(Received 30 June 2014, revised 13 August that have been associated with human reproductive diseases, and outline
2014, accepted 19 August 2014)
crucial outstanding questions in this emerging research field.
doi:10.1111/febs.13014

Introduction
Nucleotide-binding oligomerization domain receptors and often guided by innate immune cells [1,2].
(NOD-like receptors, NLRs) are intracellular proteins Macrophages, dendritic cells, epithelial cells and innate
that have mainly been characterized in the context of lymphoid cells that contribute to innate immunity are
mammalian immunity. The vertebrate immune system equipped with a defined set of germline-encoded pat-
relies on the combined action of germline-encoded tern recognition receptors (PRRs). PRRs together with
(innate) and gene-rearrangement-based (adaptive) anti- the complement system enable innate effector cells to
gen receptors to recognize microbial pathogens and mount fast and broadly effective protection against
environmental challenges that threaten homeostasis. external and internal threats. This is accomplished
Antigen receptor gene recombination in single T and B through engagement of signaling cascades that result
cell clones empowers them to selectively recognize and in the production of inflammatory cytokines, microbi-
eliminate the threat. But clonal expansion of individual cidal agents, the activation and recruitment of addi-
antigen-specific T and B cells is temporally delayed tional immune cells to the affected tissue, and by

Abbreviations
BWS, BeckwithWiedemann syndrome; CARD, caspase-recruitment domain; IL, interleukin; LPS, lipopolysaccharide; NF-jB, nuclear factor
jB; NLR, NOD-like receptor; NOD-like receptor, nucleotide-binding oligomerization domain receptor; PRR, pattern recognition receptor; PYD,
pyrin domain; SCMC, subcortical maternal complex; siRNA, small interfering RNA; SRS, SilverRussell syndrome.

4568 FEBS Journal 281 (2014) 45684582 2014 FEBS


H. Van Gorp et al. NLRs in immunity and reproduction

tagging the foreign agents for degradation by profes-


sional phagocytes [3].
Surface-bound PRRs of the Toll-like receptor and
C-type lectin receptor family continuously monitor the
extracellular environment, while different classes of
more recently discovered intracellular PRRs safeguard
the intracellular space. The latter group comprises
members of the RIG-I-like receptor, AIM2-like recep-
tor, NLR protein families, along with a growing
collection of cytosolic nucleic acid sensors [3,4]. In the
last decade, NLRs have emerged as key components
of the innate immune system where they steer host
defense by recognizing pathogen-associated molecular
patterns (PAMPs) and danger-associated molecular
patterns (DAMPs) [5]. PAMPs constitute essential
microbial components, while DAMPs are generally
regarded as host-derived agents that accumulate under Fig. 1. Domain structure of human NLR subfamilies. NOD-like
conditions of (sterile) stress and tissue damage [4]. receptors are composed of several protein domains, each fulfilling
NLRs are subdivided according to their amino-ter- a specific function. The different NLR subfamilies share a central
nucleotide-binding and oligomerization (NACHT) domain, and
minal effector domain, which generally comprises a
carboxy-terminal leucine-rich repeats (LRRs). The NACHT domain is
caspase-recruitment (CARD), a pyrin (PYD) or a an oligomerization domain that possesses NTPase activity, while
baculovirus inhibitor repeat motif (Fig. 1). As we learn the LRRs are commonly involved in proteinprotein interactions.
more about their biological roles, NLRs can also be Different subfamilies can be defined based on the presence of a
grouped according to their known physiological func- particular amino-terminal proteinprotein interaction domain. The
tions in the immune system. NLRP1b, NLRC4 and NLRA subfamily is equipped with a CARD supplemented with an
NLRP3 are NLRs with well-established roles in the acid transactivation domain (AD), while the NLRB subfamily can be
identified based on the presence of a baculovirus inhibitory repeat
assembly of inflammasomes [6]. These cytosolic multi-
domain (BIR). The NLRC/X subfamily groups the proteins
protein complexes recruit and activate the pro-inflam- containing either a CARD domain or a still unidentified domain, and
matory cysteine proteases caspase-1 and -11, which in finally the NLRP subfamily is supplied with a PYD.
their turn promote secretion of the inflammatory cyto-
kines interleukin (IL) 1b and IL-18, and induce a pro- organ systems and signaling pathways. A significant
grammed cell death mode (pyroptosis) that restricts body of recent work has provided compelling evidence
pathogen replication [6,7]. On the other hand, the that links a subset of NLRs to key functions in repro-
NLRs NOD1 and NOD2 are chiefly known for their duction and during early stages of embryonic develop-
roles in promoting activation of nuclear factor jB ment. In this paper, we review recently gained insight,
(NF-jB), MAPK and TRAF3 signaling cascades spe- based on in vitro as well as in vivo models, into the
cifically upon cytosolic detection of bacterial peptido- roles and mechanisms by which these NLRs regulate
glycan fragments. Notably, mutations in the latter early embryonic development. We argue for the exis-
NLRs are closely linked to inflammatory syndromes of tence of a clearly defined subset of NLRs that operates
the intestinal tract [8]. Finally, CIITA is a transcrip- at the interfaces of the immune and reproductive sys-
tional co-activator that is critical for major histocom- tems and that can be segregated from other NLRs
patibility complex II expression in antigen-presenting based on their distinctive phylogeny, expression pro-
cells, and mutations in CIITA cause bare lymphocyte files in gametes and the clustered genomic localization
syndrome in humans [9]. Unlike the former NLRs, the of their genes. Finally, we also reflect on the functional
immune roles of other family members are less clear. evidence linking mutations in these NLRs to reproduc-
NLRP6, NLRP7 and NLRP12 have been implicated tion-related syndromes in patients.
in inflammasome signaling and are proposed to dam-
pen NF-jB-mediated cytokine production [1012], but
The ABC of reproduction: on maternal
further analysis is required to clarify their molecular
effect genes and imprinting disorders
modes of action.
While new and diverse roles exerted by NLRs in Humans like nearly all multi-cellular organisms in the
immunity are rapidly emerging, many questions animal and plant kingdoms generate offspring
remain regarding their potential involvement in other through sexual reproduction. At the first stage, this

FEBS Journal 281 (2014) 45684582 2014 FEBS 4569


NLRs in immunity and reproduction H. Van Gorp et al.

process requires meiosis to produce gametes, reproduc- single allele. Which of the two alleles is expressed in
tive cells that bear half the number of chromosomes of the developing embryo is determined by the parent-of-
somatic cells. The 23 chromosomes of each haploid origin of the considered allele [20]. For maternally
human oocyte and sperm cell thus carry a unique com- imprinted genes, the maternal allele is repressed to
bination of genes obtained after homologous recombi- ensure that transcription occurs uniquely from the
nation of parental DNA strands. During fertilization, paternally inherited allele. In contrast, transcription
the two gametes fuse to generate a unique diploid cell from the paternal allele is silenced in the case of pater-
(zygote) that develops into a new organism. Apart from nally imprinted genes. The methylation status of
their own unique genetic material, the sperm cell and imprinted regions is conserved in all somatic cells of
oocyte also supply cytoplasmic factors such as mRNA the adult organism, but primordial germ cells in the
or proteins that are required for initial, vital stages of gonads of the developing embryo remove the parental
embryogenesis. In particular, the products of maternal imprinting patterns in order to allow oocytes and
effect genes are critical to guide the complex processes sperm cells to subsequently re-establish proper gender-
that are needed for the fertilized oocyte to correctly specific imprinting patterns [21].
transition into a developing embryo by full transcrip- Other maternal effect proteins may ensure normal
tional activation of the zygotic genome [1315]. early embryogenesis as components of intracellular
In vertebrates, maternal effect genes may be protein complexes in maturing oocytes. In this regard,
involved in properly combining the maternal and the subcortical maternal complex (SCMC) is a large
paternal haploid genomes in zygotes, in facilitating multiprotein complex that was recently discovered in
embryonic genome activation shortly thereafter, and in the subcortex of oocytes and pre-implantation
ensuring correct pre-implantation development [15]. embryos. Zygotes originating from mutant oocytes
Given their roles in these key pre- and post-fertiliza- that are defective in SCMC assembly failed to progress
tion processes, it is not surprising that mutations in beyond the first embryonic cell divisions, suggesting an
maternal effect genes almost invariably lead to early essential role for the SCMC in early embryonic devel-
embryonic arrest [15,16]. Maternal effect gene products opment [14]. However, these mutant oocytes also
are produced by the oocyte prior to fertilization, and lacked oocyte cytoplasmic lattices, oocyte-specific cyto-
embryonic genome activation coincides with a steep plasmic cytoskeletal elements that were suggested to
decline in the amount of maternal effect gene tran- support maternal ribosome transport and organelle
scripts. The kinetics of this distinctive expression pat- (re)distribution during oocyte maturation [22]. Clarifi-
tern is species-specific, since the precise moment in the cation of the relative importance of the SCMC and
developmental plan at which embryonic genome acti- cytoplasmic lattice protein complexes in oocyte matu-
vation occurs varies greatly among animal species. ration and early embryonic development requires addi-
Murine embryos undergo genome activation at the tional analysis.
two-cell stage, cattle at the 816-cell stage and humans Although the molecular modes of action of mater-
when the embryo comprises foureight cells [13,17]. nal effect genes are still vague, it has become clear in
Several maternal effect proteins are of critical recent years that mutations in some of these genes
importance for fetal growth because they orchestrate and the resulting imprinting defects are associated
epigenetic reprogramming in primordial germ cells and with multiple fetal growth disorders and reproductive
zygotes. After gametogenesis, terminally differentiated wastage in humans [21,23]. The BeckwithWiedemann
male and female haploid genomes acquire a con- syndrome (BWS) is an imprinting disease that is
densed, transcriptionally quiescent structure. This is associated with congenital overgrowth, undescended
achieved by targeted DNA methylation and histone testes in male progeny, abdominal wall defects, sei-
modifications, which are reversible epigenetic mecha- zures and predisposition to embryonic tumorigenesis
nisms that play key roles in switching transcriptional [24]. SilverRussell syndrome (SRS) on the other
activity on and off [18]. At fertilization, the embryonic hand is characterized by growth retardation prior to
genome is globally demethylated until the developing and after birth, and patients display colored spots on
embryo reaches the morula stage. However, a few the skin, excessive sweating and inward curving of
genomic loci, called imprinted regions, escape this gen- the fifth fingers. Hydatidiform moles are another
ome-wide epigenetic reprogramming and maintain the example of a human disorder caused by hypermethy-
methylation patterns that were set in the oocyte and lation or hypomethylation of imprinted control
sperm cell [19]. As a consequence, transcription of regions. Moles constitute abnormal or molar preg-
imprinted genes a subset of an estimated few hun- nancies in which a non-viable fertilized egg implants
dred autosomal genes in mammals occurs from a in the uterus, consequently leading to absent or

4570 FEBS Journal 281 (2014) 45684582 2014 FEBS


H. Van Gorp et al. NLRs in immunity and reproduction

abnormal embryonic development with abnormal A phylogenetic NLR cluster implicated


trophoblastic growth [25]. Abnormal molar tissues in reproductive functions
have different genotypes (Fig. 2AG) [26,27]. Unlike
most types of moles, familial biparental hydatidiform The NLR gene family is evolutionarily ancient and
moles have a diploid genotype originating from both predates the emergence of mammals. Large sets of
parents (Fig. 2E). Notwithstanding the presence of a NLR genes are found in several invertebrates with
paternal as well as a maternal genome, the embryo sequenced genomes, including Hydra (simple freshwa-
does not develop properly because of multiple ter animals with radial symmetry), Strongylocentrotus
imprinting defects. Familial cases of biparental hyda- (sea urchins) and Amphimedon (sponges) [3538]. The
tidiform moles are typically caused by autosomal apparent absence of NLR genes in the genomes of
recessive mutations in maternal effect genes that Drosophila melanogaster (fruitfly) and Apis mellifera
affect oocyte sufficiency [28,29], while the paternal (honey bee) form interesting exceptions that suggest
genotype does not contribute to the pathogenesis that a common insect ancestor has probably lost the
[30,31]. Molar pregnancies may also develop into an NLR repertoire during speciation. Also Caenorhabd-
invasive form, with persistent trophoblastic disease itis elegans (roundworm) is devoid of NLR genes,
(PTD) being strongly associated with familial bipa- which is consistent with the particularly high levels
rental hydatidiform moles and developing in an esti- of gene loss observed in the model organisms D. mel-
mated 1015% of all hydatidiform mole cases anogaster and C. elegans [36,39]. The NLR gene sets
[32,33]. PTD can further lead to the development of in mammals are highly plastic, with the human gen-
choriocarcinomas that necessitate chemotherapeutic ome encoding 22 NLR genes and the mouse genome
treatment for their eradication [34]. 34 NLR genes [40]. The evolutionary bifurcation of
Each of the above developmental disorders has been rodents and primates about 80100 million years ago
associated with mutations in a subset of NLR genes. is thought to have coincided with gene duplication
In the following, we highlight recent evidence suggest- events that gave rise to novel and altered protein
ing that this NLR subset may represent a novel group functions [41,42]. In agreement, mice express three
of maternal effect genes, and review our current under- orthologs of human NLRP1, four orthologs of
standing of the molecular mechanisms by which they human NAIP, seven orthologs of human NLRP4
exert their functions at the interfaces of the immune and three orthologs of human NLRP9. Gene duplica-
and reproductive systems. tion events are also suggested to account for the

Fig. 2. Genetics of hydatidiform moles


(HM). Genetically, sporadic complete HM C
are androgenic diploid conceptions (AnHM)
hypothetically originating (A) from a
duplicated paternal haploid genome (46,
XX) or (B) after fertilization of an empty D
egg with two haploid sperm cells (46, XX
and 46, XY). (C) Sporadic partial HM
(PHM) are dispermic triploid conceptions E
and usually originate from dispermic
fertilization of an oocyte (69, XXX; 69,
XXY; 69, XYY). (D) Digynic triploid
conceptions originate from duplication of a F
maternal haploid set of chromosomes. (E)
Biparental HM are diploid and contain a
maternal and paternal set of genes. (F, G) G
Hypothetical schemes of conception
mosaicism.

FEBS Journal 281 (2014) 45684582 2014 FEBS 4571


NLRs in immunity and reproduction H. Van Gorp et al.

ys
da
A C

D E

existence of primate-specific NLRP7. Apart from Notably, phylogenetic analysis of the protein
being duplicated, genes might also get lost from the sequences of the complete human NLR set along with
genome during speciation. Indeed, rodents lack or- their murine orthologs reveals that NLRP2, 4, 5, 7, 8,
thologs of human NLRP8, 11 and 13, most probably 9, 11, 13 and 14 form a phylogenetic cluster that sepa-
reflecting loss of those genes during rodent evolution rates from other NLR clusters with clearly established
[40,43,44]. immune roles (Fig. 3A) [43,44]. Interestingly, within

4572 FEBS Journal 281 (2014) 45684582 2014 FEBS


H. Van Gorp et al. NLRs in immunity and reproduction

the PYD-containing NLR subfamily, the immune- Nlrp4a, 4b, 4c, 4e, 4f, 4 g, 5, 9ac, and 14 levels dur-
related NLRs NLRP1, 3, 6, 10 and 12 form a separate ing early-stage embryogenesis (Fig. 3C). Immune-
cluster. In addition, the CARD-containing NLRC4 related NLRs do not show similar kinetics of mRNA
clusters together with the NAIPs with which it forms decay during early developmental stages (Fig. 3C).
an inflammasome complex in response to bacteria [45]. Another marked observation is that, apart from
This suggests that NLRs may be divided in several NLRP14, all reproduction-related NLRP genes in the
phylogenetic subgroups that might have distinct func- human genome are located on chromosome 19, while
tions [43,44,46]. In this regard, the cluster containing the murine syntenic region on chromosome 7 contains
NLRP2, 4, 5, 7, 8, 9, 11, 13 and 14 coincides remark- all murine reproduction-related NLR genes with the
ably with the emerging reproduction-associated NLR notable exceptions of Nlrp4f, Nlrp4g and Nlrp14
subset. Meta-analysis of publicly available expression (Fig. 3D,E). Moreover, the genes coding for these
data demonstrates that members of this reproduction- reproduction-associated NLRs are physically clustered
associated human NLR gene cluster are expressed at together in chromosomal loci that contain other genes
high levels in oocytes and ovaries (Fig. 3B) [47]. At that are important for reproductive traits [55]. A good
the protein level, human NLRP7, NLRP5 and NLRP9 example is the genomic localization of Nlrp4 and
are expressed at all stages of developing follicles Nlrp9 paralogs on mouse chromosome 7 in the vicinity
[31,48]. The murine orthologs belonging to this cluster, of the V1r genes which belong to a chemosensory
namely Nlrp2, 4a-g, 5, 9a-c and 14, are similarly receptor family and are involved in pheromone detec-
expressed at high levels in oocytes and ovaries, while tion (Fig. 3D) [44]. Also, NLRP9 in cattle and NLRP5
immune-related NLRs are not (Fig. 3B) [49]. The fact in pigs were mapped to a quantitative trait locus
that expressed sequence tags corresponding to mouse region for reproductive traits [56,57]. In agreement,
Nlrp4a, 4c, 4e, 4f, 9b and 9c were found in cDNA mutations in some of these NLRs have been associated
libraries of oocytes, zygotes and two-cell stage with mice sterility [51] and human imprinting diseases
embryos supports this notion [50,51]. Several published (see above) [58]. However, it should be mentioned
reports validated the high expression levels of mouse that possibly not all NLRs in these syntenic regions
Nlrp2, Nlrp4a-f [52], Nlrp5, Nlrp9a-c [53] and Nlrp14 are important for reproduction. The NLRP12 gene is
in female and male gonads and maturing gametes located in this region but is most probably not
[50,54]. They also showed that mRNA expression lev- involved in reproduction since Nlrp12-deficient mice
els of these NLRs drop steeply shortly after fertiliza- develop normally without apparent reproductive
tion towards the blastocyst stage, a characterizing defects [59,60]. In addition, the protein clusters phylo-
feature of maternal effect genes [15,16]. Our meta- genetically with the immune-associated NLRs
analysis along 12 developmental stages in mouse (Fig. 3A) [44] and is known to play a role in immu-
indeed nicely re-confirms the gradual decrease of nity based on hyper-responsive reactions to Toll-like

Fig. 3. Reproduction-related NLR cluster in humans and mice: phylogeny, chromosomal clustering and expression. (A) Full-length NLR
protein sequences from both human and mouse were retrieved from the Uniprot database and analyzed with the MEGA6 package hereby
using default settings [110]. Alignment was performed with MUSCLE, while MAXIMUM LIKELIHOOD was used for tree building. The resulting
phylogenetic tree file was uploaded to EVOLVIEW for visualization [111]. A reproduction-related NLR cluster (light blue) separates from the
immune-related NLRs. The whole NLRP subfamily is indicated in blue, with immune- and reproduction-related NLRPs in dark and light blue,
respectively. The NAIP subfamily forms a separate cluster together with NLRC4 (red), while most of the NLRC/X subfamily clusters
together with the NLRA subfamily (grey). (B) GENEVESTIGATOR [47] was used to explore NLR expression in human (left panel) and mouse (right
panel) revealing strong expression of reproduction-related NLRs in oocytes. In contrast, low expression levels were detected in
reproduction-related tissues for established immune-related NLRs. The darkest red color represents the maximum level of expression for a
given gene across all measurements available in the database for this gene. Adult but not embryonic or early postnatal tissue samples were
selected for analysis. The values for mouse Nod2, Naip4, Naip7, Nlrc4, Nlrp1a, Nlrp1b, Nlrp1c, Nlrp2, Nlrp4d are absent in the database. (C)
GENEVESTIGATOR [47] was used to explore expression of immune- and reproduction-related NLRs along different stages of mouse

embryogenesis. Reproduction-related NLRs are highly expressed during prenatal period day 01 (corresponding to two-cell stage embryos)
and their levels strongly decrease during prenatal period days 24 (corresponding to four-cell embryos and blastocysts). This particular
profile matches the expression pattern of maternal effect genes. (D) Reproduction-related NLRPs are clustered on chromosome 19 in
humans and on chromosome 7 in mice. (E) Based on Uniprot sequences, the domain structure of different reproduction-related NLRPs is
depicted: blue box, PYD; green box, nucleotide-binding and oligomerization (NACHT) domain; yellow circles, leucine-rich repeats. In humans,
nine different reproduction-related NLRPs could be identified, four of which are absent in mouse. In mouse, however, duplication events
occurred leading to an NLRP4 and NLRP9 cluster.

FEBS Journal 281 (2014) 45684582 2014 FEBS 4573


NLRs in immunity and reproduction H. Van Gorp et al.

Fig. 4. Mutations in human NLRs associated with reproductive diseases. (A) Schematic view of the NLRP2 protein with indication of
harmful mutations (Uniprot Q9NX02-1; Unigene NP_001167552.1). (B) Schematic view of the NLRP7 protein with indication of harmful
mutations (Uniprot Q8WX94-3; Unigene NP_001120727.1). (C) Schematic view of the NLRP14 protein with indication of harmful mutations
(Uniprot Q86W24-1; Unigene NP_789792.1). NLR domains are represented as follows: blue box, PYD; green box, nucleotide-binding and
oligomerization (NACHT) domain; yellow circles, leucine-rich repeats. Mutations are grouped to a certain level corresponding to particular
diseases. Different colors indicate zygosity of the mutation, and cases with live birth are underlined. Mutations are presented based on the
nomenclature recommendations of the Human Genome Variation Society.

receptor stimulation in Nlrp12-deficient immune cells functional inflammasome upon its ectopic expression
[59,61]. with ASC and caspase-1 in 293T cells [70]. In addi-
In the following sections, we review the evidence tion, endogenous NLRP2 was found to co-immuno-
linking NLRP2, NLRP5 and NLRP7 to reproductive precipitate with the inflammasome adaptor ASC in
functions and immunity. For most other reproduction- lipopolysaccharide (LPS) stimulated THP-1 cells, and
related NLRs, however, much less is known and the siRNA-mediated knockdown of NLRP2 reduced the
molecular mechanisms employed still need to be levels of secreted IL-1b from these cells [70,71]. Apart
unraveled. Small interfering RNA (siRNA) mediated from its suggested role in inflammasome signaling,
knockdown of Nlrp4e and Nlrp14 in fertilized eggs led ectopically expressed NLRP2 was demonstrated to
to early embryonic arrest indicating that they may be inhibit activation of the pro-inflammatory transcrip-
maternal effect genes [50,62,63]. In addition, NLRP14 tion factor NF-jB in 293T cells [7173]. Human
has been implicated in spermatogenesis (Fig. 4C) NLRP2 transcripts were readily detectable under
[64,65], while human NLRP4 was shown to play a role steady-state conditions in thymus and spleen. In addi-
in immunity [6669]. Further research is clearly needed tion, NLRP2 mRNA levels in THP-1 cells were upreg-
to fully appreciate the role of all under-investigated ulated by diverse pro-inflammatory stimuli, including
NLRs in reproduction and immunity. LPS and IL-1b, supporting a potential role for
NLRP2 in immune signaling [71,73]. Moreover,
NLRP2 mRNA levels were upregulated in CD34+
NLRP2: at the interface of immunity
progenitor cells during differentiation in macrophages
and reproduction
and neutrophils, and in mesenchymal stem cells under-
Together with NLRP3 and NLRP1, NLRP2 was one going adipogenesis [72]. Interestingly, common genetic
of the first NLR family members shown to assemble a variants in NLRP2 were suggested to correlate with

4574 FEBS Journal 281 (2014) 45684582 2014 FEBS


H. Van Gorp et al. NLRs in immunity and reproduction

the outcome of HLA-identical allogeneic stem cell syndrome/neonatal onset multisystem inflammatory
transplantation. Donor cells with homozygous NLRP2 disease [78,79]) and related inflammasome components
variants rs1043673, corresponding to missense muta- such as pyrin (familial Mediterranean fever) [80].
tion A1052Q, and rs1043684, corresponding to a Undoubtedly, clarification of the molecular functions
mutation in the non-coding 30 -UTR, were associated and signaling partners of NLRP2 in maturing oocytes
with non-relapse mortality and overall survival [74]. and developing zygotes will shed light on this intrigu-
Further work is needed to determine how these ing possibility.
NLRP2-associated single nucleotide polymorphisms
may alter susceptibility to severe acute graft-versus-
NLRP5: a pioneering maternal effect
host disease and infections [74].
gene
While NLRP2 is expressed in immune cells and
other somatic cells in humans, in mice the highest NLRP5 was one of the first maternal effect genes
expression levels were noted in developing oocytes and reported in mammals [51]. It was originally cloned as
associated granulosa cells [49]. siRNA-mediated down- an oocyte-specific factor targeted by autoantibodies in
regulation of Nlrp2 expression in oocytes did not inhi- a mouse model of autoimmune oophoritis, a syndrome
bit oocyte fertilization [63], but these cells failed to characterized by premature ovarian failure that results
progress normally after fertilization and most zygotes in menopausal symptoms and infertility in young
arrested at the first and second cleavage events [49]. women [81,82]. Autoantibodies against NLRP5 were
A similar early blockade of embryonic development also described as a marker for hypoparathyroidism
has been shown to occur upon disruption of Nlrp5 that is detected in about half the patients suffering
(see later) [51] and other maternal effect genes such as from autoimmune polyendocrine syndrome type 1, a
Floped [14], Filia [75] and Stella [76] among others. rare autosomal recessive disorder that develops in
The functional relevance of NLRP2 in early human early childhood and affects a number of endocrine
embryonic development was inferred from a limited organs such as the adrenal cortex, ovaries and para-
set of disease-association studies (Fig. 4A). One of thyroid glands [83]. The development of mice with a
these mutations R493SerfsX32 was discovered in a targeted hypomorph allele of Nlrp5 that reduced Nlrp5
familial case of BWS and causes a frameshift in expression by 9095% (Nlrp5tm/tm mice) formally
NLRP2 mRNA that is predicted to result in a trun- established the role of Nlrp5 in early embryogenesis
cated protein [24]. However, analysis of another 11 [51,84]. Both male and female Nlrp5tm/tm mice devel-
cases of non-familial BWS failed to reveal mutations oped normally, and fertility of male Nlrp5tm/tm mice
in NLRP2, suggesting that defective NLRP2 expres- was unaffected. In contrast, female Nlrp5tm/tm mice
sion is not the principal cause of sporadic BWS [24]. A produced oocytes that could be fertilized but the
homozygous missense (Q884R) mutation in NLRP2 resulting zygotes arrested at the two-cell stage [51].
was also described in a female patient with biparental Consequently, female Nlrp5tm/tm mice were incapable
hydatidiform mole [77]. In addition to the homozygous of generating offspring and presented with a sterile
mutations described above, a heterozygous (I352S) phenotype.
missense mutation in NLRP2 was found in an SRS While the precise molecular function of NLRP5 in
patient presenting with defective methylation at six maturing oocytes and zygotes remains enigmatic, it
maternally imprinted regions [25]. Screening larger was shown to localize primarily to the subcortex in
cohorts of BWS, biparental hydatidiform mole and human oocytes [48]. In mice, Nlrp5 was shown to
SRS patients is recommended to confirm the associa- assemble the SCMC, a large multiprotein complex that
tion of NLRP2 mutations with these reproductive dis- lines the subcortex of mature oocytes and pre-implan-
orders, and to estimate the fraction of these patients tation embryos (Fig. 5) [14]. In the latter, the subcellu-
presenting with homozygous NLRP2 mutations. It is lar localization of the SCMC appears to result from its
tempting to speculate that some etiological forms of active exclusion from regions of cellcell contact, thus
reproductive syndromes may have a common underly- confining its localization to the cytocortex of the outer
ing mechanism that involves defective activation of morula cells and to the trophectoderm of blastocysts,
NLRP2 and/or its signaling pathways. This might while the inner cell mass of the pre-implantation
resemble the causal association of several hereditary embryos are devoid of the SCMC [14,84]. The molecu-
autoinflammatory syndromes with mutations in the lar mechanism guiding this peculiar intracellular distri-
inflammasome adaptor NLRP3 (familial cold auto- bution at the subcortex remains to be addressed.
inflammatory syndrome, MuckleWells syndrome and However, it is clear that Nlrp5 is crucial for assembly
chronic infantile neurological cutaneous and articular of the SCMC because Nlrp5tm/tm oocytes are devoid of

FEBS Journal 281 (2014) 45684582 2014 FEBS 4575


NLRs in immunity and reproduction H. Van Gorp et al.

due to protein degradation because mRNA expression


of SCMC components was not affected in Floped/
and Nlrp5tm/tm oocytes. In contrast, Nlrp5tm/tm oocytes
expressed normal levels of Padi6 [85], suggesting that
Padi6 stability is not critically dependent on the
SCMC complex or that the protein has a significantly
longer half-life. Whether Padi6 deletion disrupts
assembly of the SCMC remains to be shown. How-
ever, Filia deficiency did not reduce expression of
other SCMC proteins in mature oocytes, nor was
assembly of the SCMC affected [75]. These observa-
tions suggest that Filia is not essential for SCMC
assembly. In agreement, Filia/ female mice were not
sterile, although they produced significantly fewer off-
spring than Filia-sufficient controls [75].
Fig. 5. Subcellular localization of Nlrp5 in germinal vesicle oocytes. The role of NLRP5 in early embryogenesis is
Nlrp5 is part of the SCMC, which is a high molecular weight likely to be conserved in primates because RNAi-
multiprotein complex localized at the subcortex of oocytes and pre-
based downregulation of NLRP5 expression led to
implantation embryos. The complex assembles during oocyte
growth and is essential for zygotes to progress beyond the first
embryonic arrest of in vitro fertilized rhesus macaque
embryonic cell divisions. Apart from Nlrp5, four additional monkey oocytes between the eight- and 16-cell stages
maternally encoded proteins have been identified to be part of the [90]. It would thus be of major interest to establish
SCMC: Floped, Tle6, Filia and Padi6. Nlrp5, Tle6 and Floped the functional roles of the SCMC complex, and to
interact with each other, while Filia binds to Nlrp5 independently. determine whether mutations in the human homologs
The precise interaction partners of Padi6 in the SCMC remain to of Nlrp5, Floped, Filia and other genes encoding
be identified. In addition, Nlrp5, Padi6 and Floped co-localize at the
other SCMC components are clinically associated
oocyte cytoplasmic lattices, a unique oocyte cytoskeletal element
involved in maternal ribosomal storage and transport, as well as in
with fetal wastage in humans. It is noteworthy in
organelle positioning and (re)distribution. this regard that mutations in the human ortholog of
mouse Figla (factor in germline alpha) have been
identified in two patients with premature ovarian
these ooplasm structures [14]. In addition to Nlrp5, failure [91]. Figla is a basic helixloophelix tran-
the maternal effect gene products Floped, Tle6 and scription factor that drives expression of Floped,
Filia also localized to the SCMC (Fig. 5) [14]. Floped Nlrp5, Tle6, Padi6 and possibly other SCMC compo-
and Nlrp5 also co-localized with the oocyte-specific nents in mice [14,92], suggesting that defective
peptidylarginine deiminase Padi6 in oocyte cytoplasmic SCMC function may be clinically associated with
lattices, suggesting that these structures visible in elec- female sterility in at least a subset of patients pre-
tron micrographs may correspond to the SCMC senting with infertility or recurrent spontaneous
[22,8588]. Indeed, electron micrographs of Padi6/, abortion.
Nlrp5tm/tm and Floped/ oocytes contained markedly
reduced cytoplasmic lattice structures [85,87,88]. Of
NLRP7: a mystery of molar
note, Floped was also reported to be diffusely
pregnancies
expressed in the cytosol of agarose-embedded mature
oocytes and pre-implantation embryos, suggesting that Unlike primates, rodents lack NLRP7. The human
staining procedures may differentially affect the locali- gene is thought to have emerged from the duplication
zation of SCMC components [88,89]. of an ancestral NLRP2/7 gene after the split of the
Importantly, both Floped/ and Padi6/ mice phe- rodent and primate lineages [40,43,44]. In agreement,
nocopied Nlrp5tm/tm mice because oocytes lacking our phylogenetic analysis clustered mouse Nlrp2
either Floped or Padi6 also failed to progress beyond together with human NLRP2 and NLRP7, with the
the initial cleavage events after fertilization [14,16,87]. human paralogs being more closely related to each
Defective expression of Floped or Nlrp5 disrupted other than to mouse Nlrp2 (Fig. 3A). While many
assembly of the SCMC complex and led to virtual questions remain regarding the functions of this pri-
depletion of SCMC components such as Tle6, Filia mate-specific NLR, recent work has illuminated some
and each others cellular pools in Floped/ and aspects of the mechanisms by which it regulates
Nlrp5tm/tm ovaries, respectively [14]. This was probably immune and reproductive functions.

4576 FEBS Journal 281 (2014) 45684582 2014 FEBS


H. Van Gorp et al. NLRs in immunity and reproduction

Human NLRP7 mRNA is detected in the spleen, biparental hydatidiform moles appear to be causally
thymus and in peripheral blood leukocytes (PBMCs), linked to homozygous and compound heterozygous
suggesting that NLRP7 contributes to immune signal- NLRP7 mutations, it is not clear whether single het-
ing [73,93]. In agreement, NLRP7 mRNA levels were erozygous variants of NLRP7 lead to familial biparen-
further upregulated in LPS- and IL-1b-stimulated tal hydatidiform moles [27,30,93,103,104]. These
PBMCs and macrophages [73]. Initial studies with homozygous and compound heterozygous NLRP7
ectopically expressed NLRP7 suggested it negatively mutations are often are located in coding exons and
regulates extracellular IL-1b levels by inhibiting splice junctions, and are predicted to affect the protein
inflammasome-mediated maturation of IL-1b or by by introducing deleterious amino acid changes, inser-
reducing cellular proIL-1b levels or by affecting its tions, intragenic duplications and deletions that lead to
secretion [73,94]. However, another report failed to a mutated or truncated protein product (Fig. 4B)
observe defective inflammasome-driven IL-1b secretion [105]. The identification of homozygous or compound
upon transient expression of NLRP7 [69]. More heterozygous NLRP7 missense and non-sense muta-
recently, siRNA-mediated knockdown of endogenous tions in men with apparently normal reproductive
NLRP7 in human primary macrophages and THP-1 functions suggests that NLRP7 may specifically regu-
cells suggested that NLRP7 assembles a canonical late female reproduction [30,31].
inflammasome that in contrast drives IL-1b secretion So far, the molecular mechanisms of NLRP7 in
upon cytosolic detection of bacterial lipopeptides [12]. imprinting defects underlying familial biparental
NLRP7 downregulation failed to affect secretion of molar pregnancies are not clear [106108], and this
the inflammasome-independent cytokines IL-6 and is complicated by the wide spectrum of disease phe-
tumor necrosis factor, suggesting that NLRP7 specifi- notypes observed. NLRP7 does not contain any
cally regulated inflammasome-driven IL-1b secretion DNA-binding domains and is localized in the cyto-
[12]. Notably, lipopeptide-induced NLRP7 activation plasm. Seemingly, the immunological function of
did not trigger pyroptosis, a lytic cell death mode that NLRP7 to control IL-1b secretion is not involved
usually accompanies IL-1b secretion by other canoni- since IL-1b-deficient mice are fertile. Besides, mono-
cal inflammasomes [7]. Determining how NLRP7 spe- cytes from some patients with familial biparental
cifically engages inflammasome-dependent cytokine moles with NLRP7 mutations show reduced IL-1b
secretion may reveal novel mechanisms that suppress secretion [94], whereas others show increased IL-1b
pyroptosis induction. secretion [104]. It should be noted, however, that
Apart from its expression in hematopoietic cells, even the general population shows varying IL-1b
NLRP7 transcripts were found to be abundantly secretion levels [109]. Additional work is clearly
expressed in testes and ovaries, suggesting that it exerts needed to fully understand the pathophysiological
reproductive functions (Fig. 3B) [73,95]. Notably, role of NLRP7 in familial biparental molar pregnan-
NLRP7 transcript levels were shown to decrease pro- cies, in order to comprehend its contribution to nor-
gressively during oocyte maturation, and to rise signifi- mal embryonic development.
cantly 35 days after fertilization, when the embryo
has reached the blastocyst stage [96]. In contrast, tran-
Conclusion and perspectives
script levels of the reproduction-associated NLRs dis-
cussed above decline irreversibly starting from the In addition to their well-established roles in the
zygote stage [50]. In addition, NLRP7 expression was immune system, the existence of an NLR subset that
detected in endometrial cancer tissues and testicular operates at the interface of the immune and reproduc-
seminomas, showing the trend to be associated with tive systems has emerged during the past 15 years. The
cancer pathogenesis [95,97]. formal demonstration that NLRP5 acts as a maternal
NLRP7 was predicted to function as a maternal effect gene product in mammals [51] has paved the way
effect gene based on the causal association of NLRP7 for recognizing the critical importance of several addi-
mutations with familial biparental hydatidiform moles tional NLR family members in the reproductive sys-
(Figs 2E,D, and 4B) [29,31]. NLRP7 mutations are tem. Members of the reproduction-associated NLR
found in as many as 60% of clinical cases of familial subset have in common that their sequences cluster in a
biparental molar pregnancies [93]. It is currently separate phylogenetic branch, that they are highly
debated whether mutations in NLRP7 also contribute expressed in mature oocytes and pre-implantation
to the pathogenesis of other forms of molar pregnan- embryos, and that deletions and mutations in their
cies (Fig. 2AD,F,G) and reproductive wastage genes are associated with human wastage syndromes.
syndromes [77,93,98102]. While familial diploid However, many important questions remain to be

FEBS Journal 281 (2014) 45684582 2014 FEBS 4577


NLRs in immunity and reproduction H. Van Gorp et al.

addressed in the coming years, including how pre- 6 Lamkanfi M & Dixit VM (2012) Inflammasomes and
cisely NLRP2, NLRP5 and NLRP7 control early their roles in health and disease. Annu Rev Cell Dev
embryonic development, what roles NLRP4, NLRP9 Biol 28, 137161.
and other less-characterized NLRs play in reproduc- 7 Lamkanfi M & Dixit VM (2014) Mechanisms and
tion, and how mutations in these genes affect human functions of inflammasomes. Cell 157, 10131022.
fertility. Another question is whether there is func- 8 Philpott DJ, Sorbara MT, Robertson SJ, Croitoru K
tional redundancy between members of the reproduc- & Girardin SE (2014) NOD proteins: regulators of
tion-related NLR cluster. Additional insight in their inflammation in health and disease. Nat Rev Immunol
14, 923.
biological roles will undoubtedly also come from
9 Steimle V, Otten LA, Zufferey M & Mach B (1993)
addressing whether these NLRs too contribute to
Complementation cloning of an MHC class II
immune and host defense responses against microbial
transactivator mutated in hereditary MHC class II
pathogens that infect reproductive organs. Answering
deficiency (or bare lymphocyte syndrome). Cell 75,
these and other questions will markedly increase our
135146.
understanding of the importance of NLRs in human 10 Elinav E, Strowig T, Kau AL, Henao-Mejia J, Thaiss
reproduction, and may lead to the development of CA, Booth CJ, Peaper DR, Bertin J, Eisenbarth SC,
improved diagnostics advancing patient stratification Gordon JI et al. (2011) NLRP6 inflammasome
as well as to better therapeutic strategies for repro- regulates colonic microbial ecology and risk for colitis.
ductive ailments and assisted reproductive technolo- Cell 145, 745757.
gies. 11 Vladimer GI, Weng D, Paquette SW, Vanaja SK,
Rathinam VA, Aune MH, Conlon JE, Burbage JJ,
Proulx MK, Liu Q et al. (2012) The NLRP12
Acknowledgements
inflammasome recognizes Yersinia pestis. Immunity 37,
The authors apologize to those whose citations were 96107.
omitted owing to space limitations. H.V.G. and 12 Khare S, Dorfleutner A, Bryan NB, Yun C, Radian
F.V.H. are postdoctoral fellows with the Research AD, de Almeida L, Rojanasakul Y & Stehlik C (2012)
Foundation Flanders (FWO grant 1.2.449.13N and An NLRP7-containing inflammasome mediates
1.2.E99.14N). M.L. is supported in part by the Ghent recognition of microbial lipopeptides in human
University Concerted Research Actions (grant BOF14/ macrophages. Immunity 36, 464476.
GOA/013) and by grants from the European Research 13 Seydoux G (1996) Mechanisms of translational control
Council (Grant 281600) and the Research Foundation in early development. Curr Opin Genet Dev 6, 555561.
Flanders (FWO grant G030212N). 14 Li L, Baibakov B & Dean J (2008) A subcortical
maternal complex essential for preimplantation mouse
embryogenesis. Dev Cell 15, 416425.
Author contributions 15 Li L, Lu X & Dean J (2013) The maternal to zygotic
transition in mammals. Mol Aspects Med 34, 919938.
HVG, AK and ML wrote the paper. AK performed
16 Li L, Zheng P & Dean J (2010) Maternal control of
meta-analysis of expression data, while FVH per-
early mouse development. Development 137, 859870.
formed the phylogenetic analysis. 17 Vallee M, Aiba K, Piao Y, Palin MF, Ko MSH &
Sirard MA (2008) Comparative analysis of oocyte
transcript profiles reveals a high degree of conservation
References
among species. Reproduction 135, 439448.
1 Palm NW & Medzhitov R (2009) Pattern recognition 18 Lee HJ, Hore TA & Reik W (2014) Reprogramming
receptors and control of adaptive immunity. Immunol the methylome: erasing memory and creating diversity.
Rev 227, 221233. Cell Stem Cell 14, 710719.
2 Cyster JG (2010) B cell follicles and antigen encounters 19 McGrath J & Solter D (1984) Completion of mouse
of the third kind. Nat Immunol 11, 989996. embryogenesis requires both the maternal and paternal
3 Takeuchi O & Akira S (2010) Pattern recognition genomes. Cell 37, 179183.
receptors and inflammation. Cell 140, 805820. 20 Lee JT & Bartolomei MS (2013) X-inactivation,
4 Broz P & Monack DM (2013) Newly described pattern imprinting, and long noncoding RNAs in health and
recognition receptors team up against intracellular disease. Cell 152, 13081323.
pathogens. Nat Rev Immunol 13, 551565. 21 Swales AK & Spears N (2005) Genomic imprinting
5 Geddes K, Magalhaes JG & Girardin SE (2009) and reproduction. Reproduction 130, 389399.
Unleashing the therapeutic potential of NOD-like 22 Yurttas P, Vitale AM, Fitzhenry RJ, Cohen-Gould L,
receptors. Nat Rev Drug Discov 8, 465479. Wu W, Gossen JA & Coonrod SA (2008) Role for

4578 FEBS Journal 281 (2014) 45684582 2014 FEBS


H. Van Gorp et al. NLRs in immunity and reproduction

PADI6 and the cytoplasmic lattices in ribosomal genetics of post-molar choriocarcinomas in Senegal.
storage in oocytes and translational control in the Mol Hum Reprod 18, 5256.
early mouse embryo. Development 135, 26272636. 35 Zhang Q, Zmasek CM & Godzik A (2010) Domain
23 Surani MA (2001) Reprogramming of genome architecture evolution of pattern-recognition receptors.
function through epigenetic inheritance. Nature 414, Immunogenetics 62, 263272.
122128. 36 Lange C, Hemmrich G, Klostermeier UC, Lopez-
24 Meyer E, Lim D, Pasha S, Tee LJ, Rahman F, Yates Quintero JA, Miller DJ, Rahn T, Weiss Y, Bosch TC
JR, Woods CG, Reik W & Maher ER (2009) & Rosenstiel P (2011) Defining the origins of the
Germline mutation in NLRP2 (NALP2) in a familial NOD-like receptor system at the base of animal
imprinting disorder (BeckwithWiedemann syndrome). evolution. Mol Biol Evol 28, 16871702.
PLoS Genet 5, e1000423. 37 Hentschel U, Piel J, Degnan SM & Taylor MW (2012)
25 Court F, Martin-Trujillo A, Romanelli V, Garin I, Genomic insights into the marine sponge microbiome.
Iglesias-Platas I, Salafsky I, Guitart M, Perez de Nat Rev Microbiol 10, 641654.
Nanclares G, Lapunzina P & Monk D (2013) 38 Yuen B, Bayes JM & Degnan SM (2014) The
Genome-wide allelic methylation analysis reveals characterization of sponge NLRs provides insight into
disease-specific susceptibility to multiple methylation the origin and evolution of this innate immune gene
defects in imprinting syndromes. Hum Mutat 34, 595 family in animals. Mol Biol Evol 31, 106120.
602. 39 Miller DJ, Ball EE & Technau U (2005) Cnidarians
26 Sunde L, Niemann I, Hansen ES, Hindkjaer J, Degn and ancestral genetic complexity in the animal
B, Jensen UB & Bolund L (2011) Mosaics and moles. kingdom. Trends Genet 21, 536539.
Eur J Hum Genet 19, 10261031. 40 Ariffin JK & Sweet MJ (2013) Differences in the
27 Williams D, Hodgetts V & Gupta J (2010) Recurrent repertoire, regulation and function of Toll-like
hydatidiform moles. Eur J Obstet Gynecol Reprod Biol receptors and inflammasome-forming Nod-like
150, 37. receptors between human and mouse. Curr Opin
28 Reddy R, Akoury E, Phuong Nguyen NM, Abdul- Microbiol 16, 303310.
Rahman OA, Dery C, Gupta N, Daley WP, Ao A, 41 She XW, Cheng Z, Zollner S, Church DM & Eichler
Landolsi H, Ann Fisher R et al. (2013) Report of four EE (2008) Mouse segmental duplication and copy
new patients with protein-truncating mutations in number variation. Nat Genet 40, 909914.
C6orf221/KHDC3L and colocalization with NLRP7. 42 Huminiecki L & Wolfe KH (2004) Divergence of
Eur J Hum Genet 21, 957964. spatial gene expression profiles following species-
29 Murdoch S, Djuric U, Mazhar B, Seoud M, Khan R, specific gene duplications in human and mouse.
Kuick R, Bagga R, Kircheisen R, Ao A, Ratti B et al. Genome Res 14, 18701879.
(2006) Mutations in NALP7 cause recurrent 43 Duenez-Guzman EA & Haig D (2014) The evolution
hydatidiform moles and reproductive wastage in of reproduction-related NLRP genes. J Mol Evol 78,
humans. Nat Genet 38, 300302. 194201.
30 Qian JH, Deveault C, Bagga R, Xie X & Slim R 44 Tian X, Pascal G & Monget P (2009) Evolution and
(2007) Women heterozygous for NALP7/NLRP7 functional divergence of NLRP genes in mammalian
mutations are at risk for reproductive wastage: report reproductive systems. BMC Evol Biol 9, 202.
of two novel mutations. Hum Mutat 28, 741. 45 Kofoed EM & Vance RE (2011) Innate immune
31 Wang CM, Dixon PH, Decordova S, Hodges MD, recognition of bacterial ligands by NAIPs determines
Sebire NJ, Ozalp S, Fallahian M, Sensi A, Ashrafi F, inflammasome specificity. Nature 477, 592595.
Repiska V et al. (2009) Identification of 13 novel 46 Schroder K & Tschopp J (2010) The inflammasomes.
NLRP7 mutations in 20 families with recurrent Cell 140, 821832.
hydatidiform mole; missense mutations cluster in the 47 Hruz T, Laule O, Szabo G, Wessendorp F, Bleuler S,
leucine-rich region. J Med Genet 46, 569575. Oertle L, Widmayer P, Gruissem W & Zimmermann P
32 Altieri A, Franceschi S, Ferlay J, Smith J & La (2008) Genevestigator v3: a reference expression
Vecchia C (2003) Epidemiology and aetiology of database for the meta-analysis of transcriptomes. Adv
gestational trophoblastic diseases. Lancet Oncol 4, Bioinformatics 2008, 420747.
670678. 48 Sena P, Riccio M, Marzona L, Nicoli A, Marsella T,
33 Sebire NJ, Savage PM, Seckl MJ & Fisher RA (2013) Marmiroli S, Bertacchini J, Fano RA, La Sala GB &
Histopathological features of biparental complete De Pol A (2009) Human Mater localization in specific
hydatidiform moles in women with NLRP7 mutations. cell domains of oocytes and follicular cells. Reprod
Placenta 34, 5056. Biomed Online 18, 226234.
34 Slim R, Coullin P, Diatta AL, Chebaro W, Courtin D, 49 Peng H, Chang BH, Lu CL, Su JM, Wu YY, Lv P,
Abdelhak S & Garcia A (2012) NLRP7 and the Wang YS, Liu J, Zhang BW, Quan FS et al. (2012)

FEBS Journal 281 (2014) 45684582 2014 FEBS 4579


NLRs in immunity and reproduction H. Van Gorp et al.

Nlrp2, a maternal effect gene required for early dependent innate immune signaling to suppress host
embryonic development in the mouse. PLoS One 7, defenses during infection. Proc Natl Acad Sci USA
e30344. 111, 385390.
50 Hamatani T, Falco G, Carter MG, Akutsu H, Stagg 62 Chang BH, Liu X, Liu J, Quan FS, Guo ZK & Zhang
CA, Sharov AA, Dudekula DB, VanBuren V & Ko Y (2013) Developmental expression and possible
MS (2004) Age-associated alteration of gene expression functional roles of mouse Nlrp4e in preimplantation
patterns in mouse oocytes. Hum Mol Genet 13, embryos. In Vitro Cell Dev Biol Anim 49, 548553.
22632278. 63 Peng H, Zhang W, Xiao T & Zhang Y (2014) Nlrp4g
51 Tong ZB, Gold L, Pfeifer KE, Dorward H, Lee E, is an oocyte-specific gene but is not required for
Bondy CA, Dean J & Nelson LM (2000) Mater, a oocyte maturation in the mouse. Reprod Fertil Dev 26,
maternal effect gene required for early embryonic 758768.
development in mice. Nat Genet 26, 267268. 64 Westerveld GH, Korver CM, van Pelt AM, Leschot
52 Peng H, Zhuang Y, Wu X, Li H, Hong Z, Zhang X, NJ, van der Veen F, Repping S & Lombardi MP
Lin X & Zhang W (2013) Expression analysis of (2006) Mutations in the testis-specific NALP14 gene in
Nlrp4a-Nlrp4f during mouse development. J Anim Vet men suffering from spermatogenic failure. Hum Reprod
Adv 12, 754759. 21, 31783184.
53 Peng H, Zhang W, Xiao T & Zhang Y (2014) 65 Horikawa M, Kirkman NJ, Mayo KE, Mulders SM,
Expression patterns of Nlrp9a, Nlrp9b and Nlrp9c Zhou J, Bondy CA, Hsu SY, King GJ & Adashi EY
during mouse development. Biologia 69, 107112. (2005) The mouse germ-cell-specific leucine-rich repeat
54 Dade S, Callebaut I, Paillisson A, Bontoux M, protein NALP14: a member of the NACHT nucleoside
Dalbies-Tran R & Monget P (2004) In silico triphosphatase family. Biol Reprod 72, 879889.
identification and structural features of six new genes 66 Fiorentino L, Stehlik C, Oliveira V, Ariza ME, Godzik
similar to Mater specifically expressed in the oocyte. A & Reed JC (2002) A novel PAAD-containing
Biochem Biophys Res Commun 324, 547553. protein that modulates NF-kappa B induction by
55 Stolk L, Perry JR, Chasman DI, He C, Mangino M, cytokines tumor necrosis factor-alpha and interleukin-
Sulem P, Barbalic M, Broer L, Byrne EM, Ernst F 1beta. J Biol Chem 277, 3533335340.
et al. (2012) Meta-analyses identify 13 loci associated 67 Cui J, Li Y, Zhu L, Liu D, Songyang Z, Wang HY &
with age at menopause and highlight DNA repair and Wang RF (2012) NLRP4 negatively regulates type I
immune pathways. Nat Genet 44, 260268. interferon signaling by targeting the kinase TBK1 for
56 Ponsuksili S, Brunner RM, Goldammer T, Kuhn C, degradation via the ubiquitin ligase DTX4. Nat
Walz C, Chomdej S, Tesfaye D, Schellander K, Immunol 13, 387395.
Wimmers K & Schwerin M (2006) Bovine NALP5, 68 Jounai N, Kobiyama K, Shiina M, Ogata K, Ishii KJ
NALP8, and NALP9 genes: assignment to a QTL & Takeshita F (2011) NLRP4 negatively regulates
region and the expression in adult tissues, oocytes, and autophagic processes through an association with
preimplantation embryos. Biol Reprod 74, 577584. beclin1. J Immunol 186, 16461655.
57 Ma J, Milan D & Rocha D (2009) Chromosomal 69 Grenier JM, Wang L, Manji GA, Huang WJ,
assignment of the porcine NALP5 gene, a candidate Al-Garawi A, Kelly R, Carlson A, Merriam S, Lora
gene for female reproductive traits. Anim Reprod Sci JM, Briskin M et al. (2002) Functional screening of
112, 397401. five PYPAF family members identifies PYPAF5 as a
58 Van den Veyver IB & Al-Hussaini TK (2006) novel regulator of NF-kappaB and caspase-1. FEBS
Biparental hydatidiform moles: a maternal effect Lett 530, 7378.
mutation affecting imprinting in the offspring. Hum 70 Agostini L, Martinon F, Burns K, McDermott MF,
Reprod Update 12, 233242. Hawkins PN & Tschopp J (2004) NALP3 forms an
59 Zaki MH, Vogel P, Malireddi RK, Body-Malapel M, IL-1beta-processing inflammasome with increased
Anand PK, Bertin J, Green DR, Lamkanfi M & activity in MuckleWells autoinflammatory disorder.
Kanneganti TD (2011) The NOD-like receptor Immunity 20, 319325.
NLRP12 attenuates colon inflammation and 71 Bruey JM, Bruey-Sedano N, Newman R, Chandler S,
tumorigenesis. Cancer Cell 20, 649660. Stehlik C & Reed JC (2004) PAN1/NALP2/PYPAF2,
60 Arthur JC, Lich JD, Ye Z, Allen IC, Gris D, Wilson an inducible inflammatory mediator that regulates NF-
JE, Schneider M, Roney KE, OConnor BP, Moore jB and caspase-1 activation in macrophages. J Biol
CB et al. (2010) Cutting edge: NLRP12 controls Chem 279, 5189751907.
dendritic and myeloid cell migration to affect contact 72 Fontalba A, Gutierrez O & Fernandez-Luna JL (2007)
hypersensitivity. J Immunol 185, 45154519. NLRP2, an inhibitor of the NF-jB pathway, is
61 Zaki MH, Man SM, Vogel P, Lamkanfi M & transcriptionally activated by NF-jB and exhibits a
Kanneganti TD (2014) Salmonella exploits NLRP12- nonfunctional allelic variant. J Immunol 179, 85198524.

4580 FEBS Journal 281 (2014) 45684582 2014 FEBS


H. Van Gorp et al. NLRs in immunity and reproduction

73 Kinoshita T, Wang Y, Hasegawa M, Imamura R & 84 Ohsugi M, Zheng P, Baibakov B, Li L & Dean J
Suda T (2005) PYPAF3, a PYRIN-containing APAF- (2008) Maternally derived FILIAMATER complex
1-like protein, is a feedback regulator of caspase-1- localizes asymmetrically in cleavage-stage mouse
dependent interleukin-1b secretion. J Biol Chem 280, embryos. Development 135, 259269.
2172021725. 85 Kim B, Kan R, Anguish L, Nelson LM & Coonrod
74 Granell M, Urbano-Ispizua A, Pons A, Arostegui JI, SA (2010) Potential role for Mater in cytoplasmic
Gel B, Navarro A, Jansa S, Artells R, Gaya A, Talarn lattice formation in murine oocytes. PLoS One 5,
C et al. (2008) Common variants in NLRP2 and e12587.
NLRP3 genes are strong prognostic factors for the 86 Yurttas P, Morency E & Coonrod SA (2010) Use of
outcome of HLA-identical sibling allogeneic stem cell proteomics to identify highly abundant maternal
transplantation. Blood 112, 43374342. factors that drive the egg-to-embryo transition.
75 Zheng P & Dean J (2009) Role of Filia, a maternal Reproduction 139, 809823.
effect gene, in maintaining euploidy during cleavage- 87 Esposito G, Vitale AM, Leijten FP, Strik AM,
stage mouse embryogenesis. Proc Natl Acad Sci USA Koonen-Reemst AM, Yurttas P, Robben TJ, Coonrod
106, 74737478. S & Gossen JA (2007) Peptidylarginine deiminase
76 Payer B, Saitou M, Barton SC, Thresher R, Dixon JP, (PAD) 6 is essential for oocyte cytoskeletal sheet
Zahn D, Colledge WH, Carlton MB, Nakano T & formation and female fertility. Mol Cell Endocrinol
Surani MA (2003) Stella is a maternal effect gene 273, 2531.
required for normal early development in mice. Curr 88 Tashiro F, Kanai-Azuma M, Miyazaki S, Kato M,
Biol 13, 21102117. Tanaka T, Toyoda S, Yamato E, Kawakami H,
77 Andreasen L, Bolund L, Niemann I, Hansen ES & Miyazaki T & Miyazaki J (2010) Maternal-effect gene
Sunde L (2012) Mosaic moles and non-familial Ces5/Ooep/Moep19/Floped is essential for oocyte
biparental moles are not caused by mutations in NLRP7, cytoplasmic lattice formation and embryonic
NLRP2 or C6orf221. Mol Hum Reprod 18, 593598. development at the maternal-zygotic stage transition.
78 Hoffman HM, Mueller JL, Broide DH, Wanderer AA Genes Cells 15, 813828.
& Kolodner RD (2001) Mutation of a new gene 89 Morency E, Anguish L & Coonrod S (2011)
encoding a putative pyrin-like protein causes familial Subcellular localization of cytoplasmic lattice-
cold autoinflammatory syndrome and MuckleWells associated proteins is dependent upon fixation and
syndrome. Nat Genet 29, 301305. processing procedures. PLoS One 6, e17226.
79 Feldmann J, Prieur AM, Quartier P, Berquin P, 90 Wu X (2009) Maternal depletion of NLRP5
Certain S, Cortis E, Teillac-Hamel D, Fischer A & de blocks early embryogenesis in rhesus macaque
Saint Basile G (2002) Chronic infantile neurological monkeys (Macaca mulatta). Hum Reprod 24,
cutaneous and articular syndrome is caused by 415424.
mutations in CIAS1, a gene highly expressed in 91 Zhao H, Chen ZJ, Qin Y, Shi Y, Wang S, Choi Y,
polymorphonuclear cells and chondrocytes. Am J Hum Simpson JL & Rajkovic A (2008) Transcription factor
Genet 71, 198203. FIGLA is mutated in patients with premature ovarian
80 The International FMF Consortium (1997) Ancient failure. Am J Hum Genet 82, 13421348.
missense mutations in a new member of the RoRet 92 Joshi S, Davies H, Sims LP, Levy SE & Dean J (2007)
gene family are likely to cause familial Mediterranean Ovarian gene expression in the absence of FIGLA, an
fever. Cell 90, 797807. oocyte-specific transcription factor. BMC Dev Biol 7,
81 Tong ZB & Nelson LM (1999) A mouse gene 67.
encoding an oocyte antigen associated with 93 Slim R & Wallace EP (2013) NLRP7 and the genetics
autoimmune premature ovarian failure. Endocrinology of hydatidiform moles: recent advances and new
140, 37203726. challenges. Front Immunol 4, 242.
82 Ruan QG, Tung K, Eisenman D, Setiady Y, 94 Messaed C, Akoury E, Djuric U, Zeng J, Saleh M,
Eckenrode S, Yi B, Purohit S, Zheng WP, Zhang Y, Gilbert L, Seoud M, Qureshi S & Slim R (2011)
Peltonen L et al. (2007) The autoimmune regulator NLRP7, a nucleotide oligomerization domain-like
directly controls the expression of genes critical for receptor protein, is required for normal cytokine
thymic epithelial function. J Immunol 178, 71737180. secretion and co-localizes with Golgi and the
83 Alimohammadi M, Bjorklund P, Hallgren A, microtubule-organizing center. J Biol Chem 286,
Pontynen N, Szinnai G, Shikama N, Keller MP, 4331343323.
Ekwall O, Kinkel SA & Husebye ES et al. (2008) 95 Okada K, Hirota E, Mizutani Y, Fujioka T, Shuin T,
Autoimmune polyendocrine syndrome type 1 and Miki T, Nakamura Y & Katagiri T (2004) Oncogenic
NALP5, a parathyroid autoantigen. N Engl J Med role of NALP7 in testicular seminomas. Cancer Sci 95,
358, 10181028. 949954.

FEBS Journal 281 (2014) 45684582 2014 FEBS 4581


NLRs in immunity and reproduction H. Van Gorp et al.

96 Zhang P, Dixon M, Zucchelli M, Hambiliki F, Levkov non-synonymous variants confer genetic susceptibility
L, Hovatta O & Kere J (2008) Expression analysis of to recurrent reproductive wastage. J Med Genet 48,
the NLRP gene family suggests a role in human 540548.
preimplantation development. PLoS One 3, e2755. 105 Milhavet F, Cuisset L, Hoffman HM, Slim R, El-
97 Ohno S, Kinoshita T, Ohno Y, Minamoto T, Suzuki Shanti H, Aksentijevich I, Lesage S, Waterham H,
N, Inoue M & Suda T (2008) Expression of NLRP7 Wise C, Sarrauste de Menthiere C et al. (2008) The
(PYPAF3, NALP7) protein in endometrial cancer infevers autoinflammatory mutation online registry:
tissues. Anticancer Res 28, 24932497. update with new genes and functions. Hum Mutat 29,
98 Deveault C, Qian JH, Chebaro W, Ao A, Gilbert L, 803808.
Mehio A, Khan R, Tan SL, Wischmeijer A, Coullin P 106 Hayward BE, De Vos M, Talati N, Abdollahi MR,
et al. (2009) NLRP7 mutations in women with diploid Taylor GR, Meyer E, Williams D, Maher ER, Setna
androgenetic and triploid moles: a proposed F, Nazir K et al. (2009) Genetic and epigenetic
mechanism for mole formation. Hum Mol Genet 18, analysis of recurrent hydatidiform mole. Hum Mutat
888897. 30, E629E639.
99 Slim R, Ao A, Surti U, Zhang L, Hoffner L, Arseneau 107 Kou YC, Shao L, Peng HH, Rosetta R, del Gaudio
J, Cheung A, Chebaro W & Wischmeijer A (2011) D, Wagner AF, Al-Hussaini TK & Van den Veyver IB
Recurrent triploid and dispermic conceptions in (2008) A recurrent intragenic genomic duplication,
patients with NLRP7 mutations. Placenta 32, 409412. other novel mutations in NLRP7 and imprinting
100 Dixon PH, Trongwongsa P, Abu-Hayyah S, Ng SH, defects in recurrent biparental hydatidiform moles.
Akbar SA, Khawaja NP, Seckl MJ, Savage PM & Mol Hum Reprod 14, 3340.
Fisher RA (2012) Mutations in NLRP7 are associated 108 El-Maarri O, Seoud M, Coullin P, Herbiniaux U,
with diploid biparental hydatidiform moles, but not Oldenburg J, Rouleau G & Slim R (2003) Maternal
androgenetic complete moles. J Med Genet 49, 206211. alleles acquiring paternal methylation patterns in
101 Manokhina I, Hanna CW, Stephenson MD, biparental complete hydatidiform moles. Hum Mol
McFadden DE & Robinson WP (2013) Maternal Genet 12, 14051413.
NLRP7 and C6orf221 variants are not a common risk 109 Gattorno M, Tassi S, Carta S, Delfino L, Ferlito F,
factor for androgenetic moles, triploidy and recurrent Pelagatti MA, DOsualdo A, Buoncompagni A,
miscarriage. Mol Hum Reprod 19, 539544. Alpigiani MG, Alessio M et al. (2007) Pattern of
102 Fallahian M, Sebire NJ, Savage PM, Seckl MJ & interleukin-1b secretion in response to
Fisher RA (2013) Mutations in NLRP7 and KHDC3L lipopolysaccharide and ATP before and after
confer a complete hydatidiform mole phenotype on interleukin-1 blockade in patients with CIAS1
digynic triploid conceptions. Hum Mutat 34, 301308. mutations. Arthritis Rheum 56, 31383148.
103 Nguyen NM & Slim R (2014) Genetics and 110 Tamura K, Stecher G, Peterson D, Filipski A &
epigenetics of recurrent hydatidiform moles: basic Kumar S (2013) MEGA6: Molecular Evolutionary
science and genetic counselling. Curr Obstet Gynecol Genetics Analysis version 6.0. Mol Biol Evol 30,
Rep 3, 5564. 27252729.
104 Messaed C, Chebaro W, Di Roberto RB, Rittore C, 111 Zhang H, Gao S, Lercher MJ, Hu S & Chen WH
Cheung A, Arseneau J, Schneider A, Chen MF, (2012) EvolView, an online tool for visualizing,
Bernishke K, Surti U et al. (2011) NLRP7 in the annotating and managing phylogenetic trees. Nucleic
spectrum of reproductive wastage: rare Acids Res 40, W569W572.

4582 FEBS Journal 281 (2014) 45684582 2014 FEBS

You might also like