You are on page 1of 15

REVIEWS

Peptidoglycan recognition proteins:


modulators of the microbiome and
inflammation
Julien Royet*, Dipika Gupta‡ and Roman Dziarski‡
Abstract | All animals, including humans, live in symbiotic association with microorganisms.
The immune system accommodates host colonization by the microbiota, maintains
microbiota–host homeostasis and defends against pathogens. This Review analyses
how one family of antibacterial pattern recognition molecules — the peptidoglycan
recognition proteins — has evolved a fascinating variety of mechanisms to control host
interactions with mutualistic, commensal and parasitic microorganisms to benefit
both invertebrate and vertebrate hosts.

Eukaryotes
All multicellular eukaryotes live in symbiotic (mutualistic or systemic diseases such as arthritis, asthma and diabetes8,9;
Organisms (such as animals, commensal) association with microorganisms, which often these effects are mediated by microbial products such as
plants and fungi) whose cells form complex communities on and/or in the body of the short-chain fatty acids9 or ATP10. Thus, any component
contain genetic material host. These microorganisms make up the host microbiome. of the host that influences the composition of the micro-
contained within a
All eukaryotes evolved in the presence of prokaryotes, and biota can have a profound effect on the development and
membrane-enclosed nucleus.
thus eukaryotes and prokaryotes have often co-evolved well-being of the host.
Symbiotic to use each other to their advantage. Eukaryotes acquired It has recently been recognized that the immune
Symbiosis is a physical prokaryotes to form two of their organelles: mitochondria system developed not only to defend the host against
association between two and chloroplasts. Multicellular eukaryotes have developed pathogens and to react to tissue damage, but also to
organisms and includes
mutualism, commensalism
long-lasting and usually mutually beneficial relationships accommodate host colonization by symbiotic micro­
and parasitism. In common with specific microorganisms, which carry out essen- organisms and to maintain microbiota–host homeo-
(non-scientific) usage it is often tial functions needed by the multicellular eukaryotes in stasis11. As the health of the host relies on the proper
assumed to be beneficial to exchange for a stable environment with a constant supply formation and maintenance of the microbiota, there
both organisms.
of nutrients1–3. The most complex microbial communi- are multiple mechanisms that control the complex
ties have developed in the guts of mammals, in particular interactions between the microbiota and the host.
omnivores and herbivores1. For example, the human gut In this Review, we focus on one family of pattern
microbiome has ten times more microorganisms than the recognition molecules of the innate immune system,
number of cells in the human body, 100 times more genes peptido­glycan recognition proteins (PGRPs), which are
*Institut de Biologie du than the human genome, and a metabolic capacity equiva- conserved from insects to mammals and have diverse
Développement de Marseille- lent to the human liver 4. The gut microbiota provides the functions in antimicrobial defence12. We discuss how both
Luminy, UMR6216 CNRS et
Aix-Marseille Universites,
host with essential nutrients (such as vitamins), breaks invertebrate and vertebrate members of the PGRP fam-
Parc Scientifique de Luminy, down indigestible compounds (such as complex carbo- ily have developed an amazing variety of mechanisms to
Marseille, France. hydrates), defends against colonization by pathogens, and coordinate the host response to mutualistic, commensal

Indiana University School of is required for the proper development of not only the and parasitic microorganisms.
Medicine – Northwest,
immune system1,2,4, but also the brain5 and gut4.
Gary, Indiana 46408, USA.
Correspondence to J.R.  Genetic and environmental factors can result in Peptidoglycan recognition proteins
and R.D. changes in mammalian microbiomes that might predis- Genes and proteins. PGRPs were first discovered in the
e-mails: pose the host to various diseases. For example, obesity, haemolymph of a silkworm (Bombyx mori) as proteins
julien.royet@univmed.fr; inflammatory bowel disease and colon cancer are asso- that bind to bacterial peptidoglycan and activate the
rdziar@iun.edu
doi:10.1038/nri3089
ciated with (and probably, in part, caused by) changes prophenoloxidase cascade, an antimicrobial host defence
Published online in the gut microbiota4,6,7. Changes in the gut microbiota mechanism in insects13. Cloning of PGRP genes from the
11 November 2011 affect sensitivity not only to intestinal diseases, but also to fruitfly (Drosophila melanogaster)14 led to the discovery

NATURE REVIEWS | IMMUNOLOGY VOLUME 11 | DECEMBER 2011 | 837

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

of families of PGRP genes in insects and other inverte- Although most PGRPs bind to peptidoglycan12,15,20,
brates, such as molluscs and echinoderms, as well as some PGRPs also bind with high affinity to other
in vertebrates, including fish, amphibians, birds and microbial molecules, such as lipopolysaccharide (LPS)
Mutualistic mammals12,15. However, PGRPs have not been found so and lipoteichoic acid; these molecules usually bind
A form of symbiosis that is
beneficial to both organisms
far in plants or in lower metazoa, such as nematodes (for outside the peptidoglycan-binding groove23–25. In addi-
involved. example, Caenorhabditis elegans). tion, certain fungus-derived molecules can be bound by
Insects usually have many PGRP genes. For example, several PGRPs (including bovine PGLYRP1 and human
Commensal D. melanogaster has 13 PGRP genes that are transcribed PGLYRP3 and PGLYRP4)24,26.
A form of symbiosis in which
into 19 proteins14,16, and a mosquito (Anopheles gambiae)
one organism benefits and the
other derives neither benefit has seven PGRP genes that are transcribed into nine pro- Control of the microbiota by insect PGRPs
nor harm. teins17. Based on the size of the mRNA transcript, these To detect and control microorganisms, insects use a
genes are usually classified into short and long forms, relatively small number of pattern recognition mol-
Microbiome known as S and L, respectively (FIG. 1). Mammals have ecules. Work carried out mainly using D. melanogaster
The microbiome is the entire
community of microorganisms
four PGRP genes, PGLYRP1, PGLYRP2, PGLYRP3 and has highlighted the central role of the members of the
that live in or on the body of PGLYRP4, which were initially named PGRP-S, PGRP-L, PGRP family in bacterial recognition and control in
a multicellular eukaryotic PGRP-Iα and PGRP-Iβ (on the basis of their short (S), insects; indeed, PGRPs are the largest and most versatile
organism. Because in many long (L) and intermediate (I) transcript lengths) by family of pattern recognition molecules for microbial
cases these microorganisms
analogy to insect PGRPs18. products in these organisms. By contrast, vertebrates
can only be characterized by
genetic methods, the term have fewer PGRPs with a much narrower range of func-
microbiome is often used to Structure and specificity. All PGRPs have at least one tions because, in addition to expressing PGRPs, they have
refer to the collective genomes PGRP domain, which is ~165 amino acids long and evolved a vast array of other pattern recognition receptors
present in all microorganisms is structurally similar to bacteriophage and bacterial (PRRs). These include Toll-like receptors (TLRs), NOD-
in a given microbial community.
type 2 amidases (which catalyse the hydrolysis of amide like receptors (NLRs), C‑type lectin receptors (CLRs),
Prokaryotes bonds)12,14–18; the PGRP domain constitutes most of RIG-I-like receptors (RLRs), scavenger receptors and the
Simple organisms (such as the sequence of short PGRPs (FIG. 1). Some PGRPs (for mannose receptor. Moreover, they possess many secreted
bacteria) whose cells lack a example, D. melanogaster PGRP-LF and mammalian recognition molecules, such as complement, collectins,
membrane-enclosed nucleus
PGLYRP3 and PGLYRP4) have two PGRP domains, pentraxins and C‑reactive protein.
and membrane-enclosed
organelles. which are homologous but not identical. Long PGRPs
usually have one PGRP domain and an additional PGRPs are upstream PRRs of D. melanogaster immune
Pattern recognition unique amino-terminal sequence (FIG. 1). Many PGRPs cascades. The functions of D. melanogaster PGRPs as
molecules are secreted, and some (such as mammalian PGLYRP1, PRRs were initially discovered by investigating the
Proteins that recognize
molecular patterns that are
PGLYRP3 and PGLYRP4) form disulphide-linked role of these proteins as sentinel receptors upstream
characteristic of microbial homodimers or heterodimers (for example, PGLYRP3– of the pathways that regulate the systemic immune
molecules but not present PGLYRP4). Some PGRPs (such as D. melanogaster response27–30. In flies, the Toll pathway is mainly acti-
in the host, such as PGRP-LC and PGRP-LF) are transmembrane recep- vated by Gram-positive bacteria and induces the pro-
peptidoglycan (which is found
tors, and some others are intracellular proteins (such as duction of antimicrobial peptides (AMPs) by cells of the
in the cell walls of all bacteria)
or lipopolysaccharide (which D. melanogaster full-length PGRP-LE). fat body; AMPs are then released into the circulating
is found in the cell walls of The general structural design of the PGRP domain haemolymph, where they kill infectious bacteria2 (FIG. 2).
Gram-negative bacteria). is similar to that of bacteriophage type 2 amidases in Two PGRP family members (PGRP-SA and PGRP-SD)
Some pattern recognition that it has three peripheral α‑helices and several central and the misnamed Gram-negative binding protein 1
molecules are found on the
surface of host cells and
β‑strands12,15,19,20. The front face of the molecule has a cleft (GNBP1) form a circulating PRR complex upstream of
function as pattern recognition that forms a peptidoglycan-binding groove, and the back the Toll pathway 27,31–33. By binding to peptidoglycan from
receptors to activate host cells, of the molecule has a PGRP-specific segment that is not the cell walls of Gram-positive bacteria, PGRP-SA trig-
whereas others are secreted present in bacteriophage amidases (FIG. 1a); this region is gers a protease cascade that ultimately transforms the
and function to trigger
often hydrophobic and is diverse among various PGRPs, inactive form of Spätzle into an active ligand, which then
pro-inflammatory cascades
or help to remove and it might bind additional ligands or other molecules19. induces Toll dimerization and downstream signalling to
microorganisms. In some PGRPs (such as D. melanogaster PGRP-LCa nuclear factor-κB (NF-κB). D. melanogaster mutants that
and PGRP-LF), the peptidoglycan-binding groove is dis- have genetic defects in Toll pathway components or in
Parasitic rupted, which prevents direct recognition of peptidoglycan the upstream PRRs are, as a result, highly susceptible to
A form of symbiosis that is
beneficial to one organism
fragments by these PGRP family members21,22. infection with Gram-positive bacteria27,34.
and detrimental to the other. The peptidoglycan-binding groove of PGRPs typi- The D. melanogaster immune system can discrimi-
cally binds the muramyl pentapeptide or tetrapeptide nate between various types of bacteria based on their
Prophenoloxidase cascade fragment of peptidoglycan with the highest affinity peptidoglycan composition12,35,36. Whereas bacteria that
A cascade of enzymatic
(FIG. 1b), and this binding usually induces a structural have a lysine residue in the third position of the peptido­
reactions that generates
quinones, which are toxic to change in the PGRP domain or an interaction with glycan stem peptide (Lys-type peptidoglycan, which is
microorganisms, and melanin another PGRP molecule. Both of these changes lock present in most Gram-positive bacteria) elicit the Toll
pigments, which restrict the the muramyl peptide in the binding site. Many PGRPs pathway, bacteria with meso-diaminopimelic acid in this
spreading of microorganisms can also discriminate between different amino acids in position (DAP-type peptidoglycan, which is present in
within the host. It is an
important component of the
the peptide or between N‑acetylmuramic acid and its most Gram-negative bacteria and in bacilli) activate
innate immune response in anhydro form within peptidoglycan 12,20, and this is another signalling cascade upstream of NF‑κB known
invertebrates. discussed below in more detail. as the IMD pathway.

838 | DECEMBER 2011 | VOLUME 11 www.nature.com/reviews/immunol

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

C D /WT0#E
CPJ[FTQ

)NE0#E
.#NC

&)NW
OGUQ&#2

&TQUQRJKNCOGNCPQICUVGT &#NC
&TQUQRJKNCOGNCPQICUVGT $CEVGTKQRJCIG6N[UQ\[OG 2)42.%Zs6%6EQORNGZ
2)42.$
V[RGCOKFCUG
2)42FQOCKPQPN[ 6%6
2)0CPJ[FTQOQPQOGT
E
&TQUQRJKNCOGNCPQICUVGT2)42U
2)425$2)425$
0 % 5GETGVGFCOKFCUGU
2)425%2)425%
0 % 2)425# 5GETGVGF244HQT2)0CEVKXCVGU6QNN
0 % 2)425& 5GETGVGF244HQT2)0CEVKXCVGU6QNN
0 % 2)42.# &KȭGTGPVKUQHQTOUGZKUVWPMPQYPHWPEVKQP
0 % 2)42.$ 5GETGVGFCOKFCUGFKȭGTGPVKUQHQTOUGZKUV
0 % 2)42.%Z 6TCPUOGODTCPG244HQT2)0CEVKXCVGU+/&
0 % 2)42.%C 6TCPUOGODTCPGRTQVGKP2)42.%ZEQTGEGRVQT
0 % 2)42.%[ 6TCPUOGODTCPGRTQVGKPWPMPQYPHWPEVKQP
0 % 2)42.& 2WVCVKXGOKVQEJQPFTKCNRTQVGKPWPMPQYPHWPEVKQP
0 % 2)42.'ȯ +PVTCEGNNWNCT244HQT2)0
0 % 2)42.'RI 'ZVTCEGNNWNCT2)0EQTGEGRVQT
0 % 2)42.( 0QPUKIPCNNKPIVTCPUOGODTCPGTGEGRVQT
'WRT[OPCUEQNQRGU
*CYCKKCPDQDVCKNUSWKF 2)42U
0 % 2)42 +PVTCEGNNWNCTRTQVGKPRTGFKEVGFCOKFCUG
0 % 2)42 5GETGVGFCOKFCUG
2TGFKEVGF)2+NKPMGFOGODTCPGTGEGRVQT
0 % 2)42
CPFCOKFCUG
0 % 2)42 2TGFKEVGFVTCPUOGODTCPGTGEGRVQT
&CPKQTGTKQ
\GDTCȮUJ 2)42U
0 % 2IN[TR $CEVGTKEKFCNCOKFCUG
0 % 2IN[TR $CEVGTKEKFCNCOKFCUG
0 % 2IN[TR $CEVGTKEKFCNCOKFCUG
/COOCNKCP2)42U
0 % 2).;42 5GETGVGFDCEVGTKEKFCNRTQVGKP
0 % 2).;42 5GETGVGFDCEVGTKEKFCNCOKFCUG
0 % 2).;42 5GETGVGFDCEVGTKEKFCNRTQVGKP
0 % 2).;42 5GETGVGFDCEVGTKEKFCNRTQVGKP

2)42FQOCKP 2)42FQOCKPYJQUG 5RGEKȮEHQT&#2V[RG2)0


*[FTQRJQDKEFQOCKP
YKVJCOKFCUGCEVKXKV[ 2)0DKPFKPIKUWPMPQYP
2)42FQOCKP
4*+/FQOCKP +/&KPVGTCEVKPIFQOCKP 5RGEKȮEHQT.[UV[RG2)0
YKVJQWVCOKFCUGCEVKXKV[
2)42FQOCKPYKVJQWV /KVQEJQPFTKCN
2)0DKPFKPICEVKXKV[ VCTIGVKPIFQOCKP

Figure 1 | The structure of PGRPs. a | The ribbon diagrams show the domain of D. melanogaster PGRP-LCx is shown in complex with a PGN
conservation of structure between a Drosophila melanogaster fragment, tracheal cytotoxin (TCT), the structure of which is shown
0CVWTG4GXKGYU^+OOWPQNQI[
amidase (peptidoglycan recognition protein-LB (PGRP-LB)) and an on the right. Images are reproduced, with permission, from REF. 40
ancestral type 2 amidase (bacteriophage T7 lysozyme). A Zn2+ ion © (2006) The American Association for the Advancement of Science.
(green sphere), which is required for amidase activity, is shown in the c | The figure shows diagrams of the domain structure and the main
peptidoglycan (PGN)-binding groove. The PGRP-unique segment is functions of D. melanogaster, Euprymna scolopes, Danio rerio and
shown in yellow. Images are reproduced, with permission, from REF. 19 mammalian PGRPs. GPI, glycosylphosphatidylinositol; PRR, pattern
© (2003) Macmillan Publishers Ltd. All rights reserved. b | The PGRP recognition receptor; RHIM, RIP homotypic interaction motif.

NATURE REVIEWS | IMMUNOLOGY VOLUME 11 | DECEMBER 2011 | 839

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

Toll pathway The receptor proteins of the IMD pathway are various specificities. Whereas PGRP-LCa and PGRP-
A signalling cascade in insects un­related to Toll and all belong to the PGRP family. The LCx pair to form a receptor for monomeric peptido­
that regulates the expression main transmembrane receptor is PGRP-LC, which func- glycan fragments, such as tracheal cytotoxin (TCT; also
of antimicrobial peptides tions both as a microbial binding protein (through its known as anhydromonomer)21,22,38–40 (FIG. 1b), PGRP-LCx
after infection by fungi or
Gram-positive bacteria.
extra­cellular PGRP domain) and as a signalling receptor homo­dimerizes to recognize polymeric peptidoglycan. By
Detection of fungi by GNBP3 (through its intracellular tail that interacts directly with forming inactive heterodimers with PGRP-LCx, another
and of bacteria by circulating IMD, an orthologue of mammalian RIP1)28–30,37 (FIGS 2,3). member of the family — PGRP-LF (a transmembrane
peptidoglycan recognition In this way, PGRP-LC is similar to mammalian TLRs, but receptor that lacks the intracellular signalling domain)
proteins (PGRPs) triggers the
unlike the D. melanogaster Toll protein. By alternative — prevents the formation of PGRP-LCa–PGRP-LCx
cleavage of the cytokine
Spätzle, which then activates
splicing, the gene encoding PGRP-LC generates three pro- heterodimers and, hence, functions as a negative regu-
the Toll receptor, and this tein isoforms (x, y and a) that have identical intra­cellular lator of the IMD pathway by decreasing downstream
initiates the activation of the domains but different extracellular PGRP domains with signalling 41,42 (FIG. 2). DAP-type peptidoglycan is also
NF‑κB-like transcription factors
Dif and Dorsal and their
translocation into the nucleus.
This pathway also controls
+/&RCVJYC[ 6QNNRCVJYC[
)TCOPGICVKXGDCEVGTKWO )TCORQUKVKXGDCEVGTKWO
dorsoventral patterning during
QTDCEKNNWU
&#2V[RG2)0
.[UV[RG2)0
embryogenesis.

Antimicrobial peptides 2)425%


(AMPs). Polypeptides that are QT2)42.$ /QPQOGTKE 2QN[OGTKE
typically less than 100 amino
2)0
6%6 2)0 2)0
acids and that have an )0$2
important role in innate #OKFCUG 2)425#
immune responses by CEVKXKV[ QT2)425&
providing broad-spectrum 2TQVGCUGU
antimicrobial activity against
2)42.%C 2)42.( 2)42.%Z
pathogenic microorganisms. 2TQ5RÀV\NG
There are different types of
2)425$ 2)42.%Z 2)42.%Z 2)42.'RI
AMP that promote microbial
killing by various mechanisms, 5RÀV\NG
but many function by 'P\[OCVKE
disrupting the membranes FGITCFCVKQP 6QNN
of microorganisms.
!
IMD pathway
A signalling cascade in 2)425%
insects (named after 2)42.'ȯ
‘immunodeficiency’ mutant
flies) that corresponds closely +/&
to the mammalian tumour .KUVGTKC
necrosis factor receptor OQPQE[VQIGPGU 2JCIQE[VKE
signalling pathway. This TGEGRVQT
cascade is mainly activated
by Gram-negative bacteria, #WVQRJCIKE 2JCIQUQOG
which are recognized by the XGUKENG 0(κ$
upstream receptors PGRP-LC
and PGRP-LE. Intracellular #/2
signalling involves the death
domain-containing protein
IMD, IκB kinase, FADD, Dredd, 0WENGWU
TAK1 and the NF‑κB-like 2JCIQN[UQUQOG
protein Relish. In contrast to
the Toll pathway, the IMD
pathway has no known role +OOWPQEQORGVGPV&TQUQRJKNCOGNCPQICUVGTEGNN
in development.
Figure 2 | Insect PGRPs recognize peptidoglycan and regulate multiple defence pathways. Depending on their
peptidoglycan (PGN) composition, bacteria can activate either the Toll pathway or the IMD pathway in Drosophila
melanogaster cells. Extracellular bacteria that contain Lys-type PGN are recognized by peptidoglycan recognition
protein-SA (PGRP-SA), PGRP-SD and Gram-negative binding protein 1 (GNBP1). This induces a protease cascade that
ultimately cleaves pro-Spätzle into an active ligand for Toll, leading to nuclear factor-κB (NF‑κB)-dependent signalling and
antimicrobial peptide (AMP) production. Bacteria that contain DAP-type PGN elicit the IMD 0CVWTG4GXKGYU^+OOWPQNQI[
pathway in flies. Polymeric
PGN is recognized by both PGRP-LCx and an extracellular version of PGRP-LE that comprises only the PGRP domain
(PGRP-LEpg). Monomeric PGN (such as tracheal cytotoxin (TCT)) is recognized extracellularly by a PGRP-LCa–PGRP-LCx
heterodimer and intracellularly by a full-length cytoplasmic form of PGRP-LE (PGRP-LEfl). Both complexes activate AMP
production through NF‑κB signalling. It is unclear how TCT enters the cells. Extracellular IMD pathway activation is
inhibited by PGRPs with amidase activity (namely, PGRP-LB and PGRP-SC), which cleave PGN into inactive amino sugars
and peptides, and by PGRP-LF, which competes with PGRP-LCa for binding to PGRP-LCx and forms non-signalling
PGRP-LF–PGRP-LCx heterodimers. Some extracellular PGRPs also act as scavengers through enzymatic degradation of
PGN (in the case of PGRP-SB) or as opsonins for phagocytosis (in the case of PGRP-SC). Finally, PGRP-LEfl induces the
elimination of intracellular bacteria by promoting autophagy, a process that is independent of IMD.

840 | DECEMBER 2011 | VOLUME 11 www.nature.com/reviews/immunol

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

Autophagy recognized in flies by PGRP-LE, which exists in two Insect PGRPs also participate in other antibacterial
Autophagocytosis that involves forms. A full-length isoform (PGRP-LEfl) detects intra- mechanisms. In D. melanogaster, full-length PGRP-LE
the degradation of the cell’s cellular peptidoglycan, and a cleaved form (PGRP-LEpg) is required to eliminate intracellular bacteria (such as
own components through the enhances PGRP-LCx-mediated cell-surface recognition Listeria monocytogenes) by promoting autophagy43 (FIG. 2),
lysosomal machinery. This
process has a role in normal
of polymeric peptidoglycan43,44 (FIG. 2). which has a protective role against intracellular patho-
cell growth and development, In contrast to systemic immune responses, which are gens (in addition to its catabolic role in the degradation
and also has a protective role controlled by both the Toll and IMD pathways, AMP pro- of the cell’s own components). Some extracellular PGRPs
against infection with duction locally in the respiratory and intestinal epithelia also function as bactericidal proteins (for example,
intracellular pathogens.
depends on the IMD pathway only. This indicates that PGRP-SB) or as opsonins (for example, PGRP-SC)12,15,45.
Gram-negative bacteria are probably the main inducers In various insects, several extracellular PGRPs trigger the
of these epithelial immune responses in D. melanogaster. prophenoloxidase cascade, which results in the formation
of melanin and of other antimicrobial compounds that
have been less well defined12,13,15,46.
&TQUQRJKNC
OGNCPQICUVGT
PGRPs regulate the gut microbiota in D. melanogaster.
)WV AMPs are constantly produced at basal levels in the
uninfected gut of D. melanogaster, but their transcrip-
tion is highly upregulated following feeding on food
)TCOPGICVKXGDCEVGTKWO
QTDCEKNNWU
&#2V[RG2)0 that is contaminated with microorganisms (such as
#/2U rotten fruits). Consistently, various laboratories have
415
shown genetically that IMD-dependent AMP produc-
tion in the gut is essential for flies to resist infection by
numerous ingested pathogens, such as Serratia marc-
2)425%QT2)42.$ escens or Pseudomonas entomophila 47,48. Although it
2)42.%
COKFCUGCEVKXKV[ has not yet been unambiguously demonstrated, it is
2)0
assumed that PGRP-LC mediates this IMD-dependent
&71:
gut response (FIG. 3).
In addition to regulating the induction of AMP pro-
+/& duction following infection, the IMD pathway has an
important role in shaping the gut response to the endog-
enous microbiota. The commensal bacterial community
R +-- in the intestinal tract of D. melanogaster is much simpler
than in humans and consists of ~3 × 105 microbial cells of
10 to 20 different species (mainly of the Acetobacter and
4GNKUJ Lactobacillus genera)49–52. Although the D. melanogaster
microbiota is a new area of research, it is already clear
4GNKUJ 2KTM
that, as shown for vertebrates, these microorganisms
#6(
influence the physiology of the host fly. The gut micro-
&71:
4GNKUJ 2)42U biota promotes larval growth by modulating hormonal
signals53 and even triggers sexual selection preference in
D. melanogaster populations by altering the production
0WENGWU %CWFCN 4GNKUJ #/2U of sex pheromones54. This implies that a tight qualita-
tive and quantitative control of the gut microbiota is
required for the optimal adaptation of D. melanogaster
to its environment.
/KFIWVGRKVJGNKCNEGNN The constant contact between bacteria and the gut
Figure 3 | Drosophila melanogaster PGRPs protect the gut from infections and mucosal epithelium raises the question of how the epi-
maintain gut microbiome homeostasis. DAP-type peptidoglycan (PGN) from thelium manages this continuous input of immune-
0CVWTG4GXKGYU^+OOWPQNQI[
intestinal bacteria is sensed by peptidoglycan recognition protein-LC (PGRP-LC), which activating signals derived from commensal micro­
triggers the IMD and p38 mitogen-activated protein kinase pathways. The activation organisms. Recent evidence points to the important
of p38 enhances the transactivating function of ATF2, which in turn activates the role of PGRP family members in the tolerance of gut
transcription of dual oxidase (Duox). This leads to the production of reactive oxygen epithelia towards the endogenous microbiota. Of the
species (ROS) in the intestinal lumen, where they control endogenous and infectious 19 D. melanogaster PGRP proteins, seven have amidase
bacteria. PGN recognition by PGRP-LC also triggers (through the IMD pathway) the activity, which allows them to cleave pro-inflammatory
cleavage and nuclear translocation of the nuclear factor‑κB family member Relish, which peptidoglycan into non-stimulatory muropeptides55,56.
then induces increased transcription of antimicrobial peptide (AMP) genes. However, in
These seven proteins are PGRP-LB‑A, PGRP-LB‑B,
the gut epithelium, the IMD pathway is under tight negative control. Two of the genes
that are most responsive to IMD activation encode PGRP-LB, which cleaves PGN and PGRP-LB‑C, PGRP‑SB1, PGRP‑SB2, PGRP‑SC1 and
therefore blocks activation of the IMD pathway, and Pirk, which interferes with the plasma PGRP‑SC2. Through their amidase activity, PGRP-SC
membrane localization of PGRP-LC. Finally, in some gut cells, the endogenous microbiota and PGRP-LB synergize to sustain a low basal level of
causes nuclear translocation of an inhibitor (Caudal), which prevents constant AMP peptidoglycan in the gut, a mechanism that prevents
production and the destruction of the normal microbiota. IKK, IκB kinase. spontaneous overactivation of the IMD pathway by the

NATURE REVIEWS | IMMUNOLOGY VOLUME 11 | DECEMBER 2011 | 841

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

gut microbiota (B. Lemaitre, personal communication) ROS are produced at basal levels by dual oxidase
(FIG. 3). These enzymes also prevent excessive transfer (DUOX), which is associated with the gut membrane.
across the epithelial barrier (and into the haemolymph) Following infection, increases in both Duox gene activa-
of immunostimulatory peptidoglycan fragments that tion and DUOX protein expression are required to coun-
are released from gut bacteria. This function is revealed ter the increased bacterial burden in the gut 63. DUOX
by the overactivation of the systemic inflammatory enzymatic activation requires an increase in intra­cellular
response in the fat body cells of flies that have inactivated calcium levels and probably depends on signalling
catalytic PGRPs in the gut 57. through an as yet unidentified upstream G protein-
Another mechanism that flies use to render their coupled receptor (and is therefore independent of
endogenous microbiota invisible to their immune sys- PGRPs). By contrast, Duox transcriptional upregula-
tem is the modulation of PGRP-LC localization in the tion is triggered by both peptidoglycan-dependent and
epithelial cell membrane. This function is mediated -independent pathways. In the peptidoglycan-dependent
by the intracellular protein Pirk (poor IMD response pathway, peptidoglycan recognition by PGRP-LC acti-
upon knock-in; also known as Rudra or Pims), which, vates the transcription of Duox through IMD, MEKK1,
by sequestering PGRP-LC in the cytoplasm, prevents p38 and ATF2. This results in increased levels of DUOX
it from accessing extracellular peptidoglycan and thus in the epithelial cell membrane and thus enhances the
decreases IMD signalling 58–60 (FIG. 3). Accordingly, loss- release of ROS into the gut lumen63 (FIG. 3).
of-function mutations in the genes encoding PGRP-SC, The above findings show that D. melanogaster gut
PGRP-LB and Pirk are associated with an increased innate immune responses, which are Toll independent,
constitutive production of AMPs in the gut, and this rely on a finely tuned balance between the IMD and the
disappears when flies are grown in axenic conditions DUOX pathways, which are both activated by upstream
(free of all microorganisms). Because these genes are PGRP molecules. Activation or repression of these path-
also transcriptional targets of the IMD pathway, their ways allows the host to adapt its intestinal antimicrobial
protein products are constitutively produced at basal response to the gut microbial load. Phenotypic analy-
levels in the gut in response to the microbiota. This ses of flies with mutations in genes that are involved in
establishes a negative feedback loop that prevents hyper- these pathways demonstrate that controlling the quantity
activation of the IMD target genes by peptidoglycan and the quality of the intestinal microbiota is crucial for
derived from commensal bacteria (FIG. 3). It is expected, the flies, and that PGRP family members are important
although not yet demonstrated, that the dysregulated players in these defence and homeostatic mechanisms.
immune activation observed in flies with mutant forms
of PGRP-SC, PGRP-LB and Pirk will induce a shift in Control of symbiotic bacteria and parasites in other
the composition of the gut microbiota. Similar results insects. Haematophagous insects frequently encoun-
have been obtained by analysing the effects of another ter blood-borne infectious agents, including para-
IMD pathway regulator, Caudal, on the composition of sites and viruses. For example, female Anopheles spp.
the microbiota. This transcription factor directly sup- mosquitoes become infected with malaria parasites
presses IMD-dependent NF‑κB-mediated expression (Plasmodium spp.) during feeding on infected humans
of AMPs in the gut by blocking AMP gene promoters52. and can then transmit the parasites to new hosts dur-
The absence of such a control, as observed by inhibition ing subsequent bites. Blood-meal ingestion is associated
of Caudal expression, results in changes in the com- with an increase in the size of the microbial commu-
mensal microbiota52. The baseline repression of the nity in the gut of the female mosquito, and this triggers
IMD pathway mediated by these various proteins is also PGRP-LC-mediated NF‑κB signalling, which induces
essential for maintaining the fitness of the flies. The AMP production to control the microbial burden64
absence of PGRP-LB, PGRP-SC, Pirk or Caudal dis- (FIG. 4a). In mosquitoes in which PGRP-LC levels have
rupts gut homeostasis, provokes epithelial cell renewal been downregulated by RNA interference, gut bacterial
and decreases the lifespan of D.  melanogaster 52,61 loads are strongly increased compared with those in con-
Reactive oxygen species (B. Lemaitre, personal communication). trol mosquitoes. Interestingly, this PGRP-LC-mediated
(ROS). Aerobic organisms response not only affects gut bacteria but also seems
derive their energy from the Control of the microbiota through regulation of ROS to decrease the infection efficiency of protozoal para-
reduction of oxygen. The production in D. melanogaster. AMP production by the sites. Silencing of the gene encoding PGRP-LC results
metabolism of oxygen, and
in particular its reduction
gut epithelium downstream of the IMD pathway is not in an increased number of Plasmodium spp. oocysts in
through the mitochondrial the only effector function of the intestinal antimicrobial infected mosquitoes (FIG. 4a). Because this effect is no
electron-transfer chain, immune response. Consistently, flies lacking gut AMP longer observed in mosquitoes in which the gut micro-
generates by-products such expression are resistant to infection by many bacterial biota has been eliminated by antibiotic treatment, these
as superoxide (O2–), hydrogen
species and, as discussed earlier, IMD pathway activa- results indicate that PGRP-LC-mediated antibacte-
peroxide (H2O2) and hydroxyl
radicals (•OH). These three tion is under strong negative regulation in the gut to rial defence also has an indirect antiparasitic effect in
species and the unstable preserve the commensal microbiota and host health. Anopheles gambiae65. It is probable that, in addition to
intermediates that are formed Recent studies have shown that reactive oxygen species controlling the gut microbiota, some of the bacterium-
by lipid peroxidation are (ROS) are important microbicidal effectors in the fly induced defence molecules (such as AMPs and ROS)
known as ROS. ROS can
damage important intracellular
gut 62. Although ROS and AMPs are directed towards might interfere with the normal development of malaria
targets such as DNA, the same microbial targets, their production in the parasites during the midgut stages of their life cycle,
carbohydrates and proteins. D. melanogaster gut is regulated by different mechanisms. although this has not yet been clearly shown65.

842 | DECEMBER 2011 | VOLUME 11 www.nature.com/reviews/immunol

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

C #PQRJGNGUICODKCG

8GTVGDTCVGJQUV $NQQFHGGFKPI
1QE[UV 5CNKXCT[INCPFU
/KFIWVGRKVJGNKWO

5RQTQ\QKVG
1QMKPGVG !

+PETGCUGF
! DCEVGTKCN
DWTFGP
2)42.% #/2
RTQFWEVKQP
<[IQVG 2)0 $CEVGTKWO

2NCUOQFKWOURR
ICOGVQE[VG

)COGVQE[VG $NQQF 2NCUOQFKWOURR


OGCN VTCPUOKUUKQP
D /KFIWV
$NQQFOGCN

2)42.$
COKFCUG
6UGVUGȯ[ CEVKXKV[HQT2)0
2)0
&GETGCUGUFCOCIGVQ
VJGJQUVKPFWEGFD[ #PVKRTQVQ\QCN
VJGKOOWPGTGURQPUG CEVKXKV[

$CEVGTKQE[VG
6T[RCPQUQOG
9KIINGUYQTVJKC
INQUUKPKFKC

2)42.$

0WENGWU

Figure 4 | In blood-sucking insects, PGRPs maintain the balance between symbiotic bacteria and protozoal
parasites. a | Peptidoglycan recognition protein-LC (PGRP-LC) controls intestinal symbiotic and infectious bacteria in
Anopheles gambiae by inducing the production of antimicrobial peptides (AMPs). This inhibits 0CVWTG4GXKGYU^+OOWPQNQI[
the proliferation of gut
bacteria after a mosquito blood meal. PGRP-LC activation also strongly decreases the number of Plasmodium spp.
oocysts in the A. gambiae gut after a blood meal. The tripartite interaction between symbiotic bacteria, invading
parasites and the mosquito immune system is important for the balance of mosquito susceptibility and resistance to
malaria and perpetuates disease transmission. b | The tsetse fly (Glossina spp.) is the sole African vector for trypanosomes
and has co-evolved with the mutualistic endosymbiont Wigglesworthia glossinidia. W. glossinidia induces the expression
of PGRP-LB by infected midgut cells (bacteriocytes), and PGRP-LB cleaves peptidoglycan (PGN) and decreases the
damage to the host associated with the permanent presence of the endosymbiont. PGRP-LB also maintains low levels of
trypanosomes by an unknown mechanism. Elimination of W. glossinidia renders tsetse flies sterile and increases their
susceptibility to trypanosome infection.

PGRP family members also regulate interactions for this nutritionally restricted diet, tsetse flies carry
between some insects and their microbial endosymbi- the obligate endosymbiont Wigglesworthia glossinidia,
onts. Adult tsetse flies (Glossina spp.) feed exclusively on the presence of which is required for adequate diges-
vertebrate blood, a very unbalanced meal lacking vital tion and nutrition in the flies and for their successful
components, such as many vitamins. To compensate reproduction. The Gram-negative W. glossinidia bacteria

NATURE REVIEWS | IMMUNOLOGY VOLUME 11 | DECEMBER 2011 | 843

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

reside in differentiated midgut cells, where they form an These squid are nocturnal animals, and light produced
organ known as a ‘bacteriome’. W. glossinidia elimina- by the symbiotic bacteria provides counter-illumination
tion results in sterility of tsetse flies and increases their to moonlight and makes the squid less visible to
susceptibility to trypanosome infection66, demonstrating predators.
the essential role of this endosymbiont in the life cycle of E. scolopes are not associated with any bacteria
tsetse flies (FIG. 4b). at birth; however, within a few hours after hatching,
The symbiosis between the tsetse fly and W. glossi- the light organ becomes populated with V. fischeri in
nidia raises two questions: how does the tsetse fly an association that is highly specific for this bacterial
immune system tolerate the constant presence of its species75,76. The nascent light organ has a pair of pro-
gut symbiont, and what is the mechanism by which truding ciliated appendages on the opposing sides of
W. glossinidia mediates its antitrypanosome effect? three pores (FIG. 5). In response to bacteria in the sur-
Part of the answer was obtained by studying the func- rounding water, epithelial cells of the appendages
tion of the DAP-type peptidoglycan-cleaving enzyme secrete mucus that covers the pores and appendages74–76.
PGRP-LB in tsetse flies. It has been shown that W. glossi- The beating of the cilia concentrates bacteria from the
nidia triggers the expression of tsetse fly PGRP-LB in surrounding water into the mucus, which starts the
its bacteriome67 (FIG. 4b). It is thought that by scavenging ‘winnowing’ of V. fischeri from hundreds of other bac-
W. glossinidia peptidoglycan, PGRP-LB prevents consti- terial species present in the water, so that only V. fis-
tutive IMD pathway activation and, hence, establishes cheri enters through the pores into the ducts of the light
tolerance towards this symbiont. Consistently, decreas- organ. These bacteria then swim into the deep crypts,
ing the in vivo PGRP-LB level induces an increase in the where they lose their flagella and permanently colonize
level of AMPs and decreases the number of W. glossi- the host 74–76. Entry of V. fischeri into the ducts induces
nidia in the tsetse fly gut. Unexpectedly, depletion of apoptosis and morphogenesis in the light organ; this
PGRP-LB also increases the susceptibility of tsetse flies involves the loss of ciliated epithelium and the shorten-
to trypanosome parasites (FIG. 4b), and this indicates that ing and eventual loss of the appendages, which prevents
PGRP-LB might have an antiprotozoal activity that con- the entry of other bacteria into the light organ73–76.
fers a resistance to these parasites67. Bacteriomes contain- PGRPs have been proposed to participate in the
ing symbiotic bacteria are common in other insects, and winnowing and maintenance of V. fischeri in the light
the mutualistic interactions between the insects and their organ72–76, but the basis for the selectivity of this win-
endosymbionts are probably also controlled by PGRPs; nowing is still poorly understood. E. scolopes has four
for example, this has been suggested by the high level of PGRPs, and they are all expressed in the light organ72.
expression of PGRP-LB in the bacteriome of the weevil Two PGRPs have been proposed to function as recep-
Sitophilus zeamais 68. tors on the surface of light organ cells; these are PGRP3,
Thus, in insects that do not belong to the Drosophilidae which has a predicted glycosylphosphatidylinositol
family, PGRPs and the host innate immune response (GPI) membrane anchor and amidase activity, and
interact to fine-tune the homeostatic balance between PGRP4, which is predicted to have two transmembrane
tolerance to indispensable mutualistic microorganisms domains, an extracellular PGRP domain and no amidase
and resistance to pathogenic microorganisms. It is possi- activity 72 (FIG. 5D). Light organ cells also express a TLR
ble that this tripartite interaction between the gut micro- and an LPS-binding protein (LBP)72. It is proposed that
biome of insects, PGRP-mediated antibacterial defences bacterial products, such as peptidoglycan and LPS, acti-
and parasites such as Plasmodium spp. or trypanosomes vate the transcription factors NF‑κB, ETS and Krüppel
could be exploited in future interventions aiming to through PGRP3, PGRP4 and TLR. These transcrip-
control disease transmission. tion factors then induce the synthesis of antibacterial
factors — such as mucin, nitric oxide synthase (NOS)
Control of luminescent bacteria by squid PGRPs and LBP — and also of PGRP1 and PGRP2 by the
PGRPs are also present in other invertebrates. In ciliated epithelial cells72 (FIG. 5D).
molluscs — such as oysters69, scallops70,71, squid72–76 PGRP2 is secreted with the mucus and is an ami-
and snails77 — PGRPs are expressed in the mantle, dase that hydrolyses pro-inflammatory peptido­
gills, haemocytes, digestive diverticula, kidneys and glycan fragments74. PGRP2 is also secreted into the
gonads69–72,77. Some of the mollusc PGRPs are predicted lumen of the crypts74, where it helps to maintain the
or proven to be peptidoglycan hydrolases, whereas oth- homeostatic balance between E. scolopes and the per-
ers have defensin-like domains, indicating that they manently present V. fischeri. PGRP2 accomplishes
might have a direct bactericidal function69–72,77. These this by hydrolysing peptidoglycan fragments that are
mollusc PGRPs probably have a role in defence against constantly released from V. fischeri and thus prevent-
infections, because their expression is increased in ing the inflammatory effects of these peptidoglycan
response to bacterial pathogens, such as Echinostoma fragments, which allows beneficial coexistence of the
paraensei 70. symbiont with the host 74. PGRP2 might also have a low
In the Hawaiian bobtail squid, Euprymna scol- level of antibacterial activity that helps in the winnow-
opes, PGRPs are involved in highly specialized host– ing function of the appendage-derived mucus and in
microorganism interactions that result in the establish- maintaining the proper numbers of V. fischeri in the
ment and maintenance of the symbiotic luminescent crypts, although this antibacterial activity has not yet
bacterium Vibrio fischeri within the squid light organ72. been experimentally proven.

844 | DECEMBER 2011 | VOLUME 11 www.nature.com/reviews/immunol

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

# $ %
1VJGT
8ƒUEJGTK DCEVGTKC
2)42 2)42
C #RQRVQUKU
↓2)42 QHEKNKCVGF /QTRJQIGPGUKU
GRKVJGNKCNEGNNU
.QUUQHEKNKCVGF
CRRGPFCIGU
D

2QTG

E
&WEV G

%T[RV
%KNKCVGFCRRGPFCIG

&

$CEVGTKCNRTQFWEVU

2)42 2)42
2)42 #OKFCUG 2GRVKFQIN[ECP
6.4 CEVKXKV[
#OKFCUG
.$2 CEVKXKV[

! ! ! 2)42
.KIJVQTICP !
EGNN 0(κ$
'65
-TØRRGN #RQRVQUKU

0WENGWU #PVKDCEVGTKCN
HCEVQTU
0(κ$ 2)42
2)42
/WEKP
015
.$2

Figure 5 | Euprymna scolopes PGRPs control colonization and maintenance of the luminescent bacterium Vibrio
0CVWTG4GXKGYU^+OOWPQNQI[
fischeri within the light organ of the squid. A | The location of the light organ in the body cavity of E. scolopes is shown.
B | The figure shows the colonization of the crypts in the light organ by V. fischeri. Seawater contains many different
bacteria, which stimulate epithelial cells of the light organ to secrete mucus containing peptidoglycan recognition
protein 2 (PGRP2) and probably other antibacterial factors that select V. fischeri (a). As a result, only Gram-negative
bacteria aggregate between the appendages of the light organ (b), and V. fischeri becomes dominant (c). V. fischeri then
enter the ducts (d) and permanently colonize the crypts (e). C | Successful establishment of V. fischeri in the crypts results
in the loss of ciliated appendages. D | Light organ cells express Toll-like receptor (TLR) and four PGRPs with different
cellular locations. PGRP1 is intracellular, PGRP2 is secreted, PGRP3 has a predicted glycosylphosphatidylinositol
membrane anchor and PGRP4 has two predicted transmembrane domains and an extracellular PGRP domain. It is
proposed that PGRP3 and PGRP4 function as cell surface receptors for bacterial products and, together with TLR, activate
the transcription factors nuclear factor‑κB (NF‑κB), ETS and Krüppel, which induce the synthesis of mucin, nitric oxide
synthase (NOS), lipopolysaccharide binding protein (LBP), PGRP1 and PGRP2 in ciliated epithelial cells, and PGRP2 in
crypt epithelial cells. Loss of PGRP1 expression in ciliated epithelial cells after colonization of the light organ by V. fischeri
leads to apoptosis and the loss of appendages. PGRP2 is secreted by epithelial cells, first in the appendages and then in
the crypts, where it degrades peptidoglycan fragments that are released by V. fischeri. This maintains homeostasis
between the host and the symbiont. Figure parts A–C are modified, with permission, from REF. 76 © (2004) Macmillan
Publishers Ltd. All rights reserved.

NATURE REVIEWS | IMMUNOLOGY VOLUME 11 | DECEMBER 2011 | 845

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

PGRP1 is a predicted amidase, but it does not have several pathways, including immune and inflammatory
a signal peptide and is located in the nuclei and the responses, signalling pathways, transcription and metab-
cytoplasm of ciliated epithelial cells in the appendages. olism82,83. Inhibition of Pglyrp5 increases the expression
The loss of PGRP1 expression by these cells (presum- of TLR2, TLR3, MAPK-interacting serine/threonine
ably when the activating signals derived from bacteria kinase 2, the interleukin‑17 receptor and NF‑κB83 (FIG. 6a).
decrease) correlates with the initiation of their apopto- Therefore, PGRPs can directly or indirectly down­regulate
sis73, but the exact mechanism of this process is unknown. the immune response to bacteria in the surrounding
Apoptosis of epithelial cells is followed by the loss of cilia water, thus preventing a constant state of inflammation.
and eventually appendages73, and this marks the com- However, additional studies confirming these roles of
pleted colonization of the light organ with V. fischeri PGRPs are needed.
(FIG. 5). PGRP1 is also expressed in the crypts, but its role
there is unknown. Protection against colitis by mammalian PGRPs
PGRPs are also expressed by another group of Mammals have four PGRPs, three of which (PGLYRP1,
marine invertebrates, the echinoderms. In the European PGLYRP3 and PGLYRP4) are bactericidal24,26,84,85 (BOX 1)
starfish, Asterias rubens, PGRP‑S1a is expressed by the but have no amidase activity 26,85,86, which distinguishes
epithelial cells lining the coelom, and PGRP‑S2a is them from fish PGRPs, which have both activities79.
expressed by phagocytes. PGRP‑S2a hydrolyses pepti- Mammalian PGLYRP2, like fish PGRPs, has both ami-
doglycan and increases the phagocytosis of Micrococcus dase86,87 and antibacterial activities (R.D., S.‑Y. Wang and
luteus, indicating that it has a role in the defence D.G., unpublished observations). PGLYRP1 is mainly
against bacteria78. expressed in the granules of neutrophils and eosino-
phils and contributes to the antibacterial activity of these
Defence against infections by fish PGRPs cells88–90. PGLYRP1 is also expressed to a lesser extent in
PGRPs have been identified in several species of fish, the salivary glands, tongue, throat, oesophagus, jejunum,
including zebrafish 79, rockfish 80 and large yellow ileum and caecum. PGLYRP2 has the highest constitutive
croaker 81. Zebrafish have four PGRP genes, three of level of expression in the liver, from where it is secreted
which have been cloned and characterized (namely, into the blood91,92, and PGLYRP2 expression is induced
pglyrp2, pglyrp5 and pglyrp6) 79. These PGRPs are by bacteria and cytokines (such as interleukin‑1β and
expressed widely in different tissues of adult fish, includ- tumour necrosis factor) in the skin and in other epithe-
ing in the skin, gills, liver, intestine and pancreas (FIG. 6a), lial cells, including those in all segments of the gastro­
indicating that they function both in the defence against intestinal tract 26,93–95. PGLYRP3 and PGLYRP4 have
bacteria that are present in the environment and during the highest level of expression in the skin and on the
systemic infections. The high-level expression of PGRPs mucous membranes, in particular in the mouth, throat,
in the intestine also indicates a probable role of these oesophagus and salivary glands and, to a lesser extent,
proteins in the modulation of the fish gut microbiome. in the stomach and the small and large intestines26,96,97.
Zebrafish Pglyrp2, Pglyrp5 and Pglyrp6 are amidases Thus, mammalian PGRPs are strategically positioned
that cleave the bond between N‑acetylmuramic acid and in all areas of the body that come into contact with
alanine in peptidoglycan79. They are also bactericidal environmental microorganisms and that are inhabited
against both Gram-positive and Gram-negative bacte- by commensal microorganisms, and it is thought that
ria79 (FIG. 6a). Rockfish PGRPs also have both amidase through their bactericidal effects mammalian PGRPs
and bactericidal activities80. can influence the microbiota at these sites. PGRPs (in
Zebrafish Pglyrp2 is highly expressed in eggs and in particular, PGLYRP1 and PGLYRP4) are also expressed
developing oocytes, and Pglyrp5 is induced during early in the mammary glands and are present in milk97,98, and
embryogenesis and is strongly expressed at 72 hours thus these PGRPs could influence the mouth and gut
post fertilization79. These proteins protect the develop- microbiota as soon as newborns begin to drink milk.
ing embryo from infection by bacteria that are present in All four mammalian PGRPs control the acquisition
the surrounding water (FIG. 6a). Knockdown of pglyrp5 and maintenance of the normal gut microbiota, which
expression during early embryogenesis (using a specific protects the host from enhanced inflammation, tis-
morpholino) results in increased sensitivity to infections sue damage and colitis97 (FIG. 6b). Pglyrp1–/–, Pglyrp2–/–,
with Salmonella enterica and Bacillus subtilis and a high Pglyrp3–/– and Pglyrp4–/– mice are all more sensitive
level of embryonic mortality 79. Similarly, inhibition of than wild-type mice to experimental ulcerative colitis
Pglyrp6 expression increases the sensitivity and mortal- induced by dextran sulphate sodium (DSS), a com-
ity of zebrafish embryos to Flavobacterium columnare pound that damages the intestinal epithelium. The
infection82. Therefore, PGRPs have an essential role in probable sequence of events that makes PGRP-deficient
the defence against bacteria during the early stages of fish mice more sensitive to colitis is as follows (FIG. 6b). First,
development, when adaptive immunity has not yet devel- deficiencies in individual PGRPs induce dysbiosis in
oped and thus, like in invertebrates, innate immunity is the colonic microbiota; this involves shifts in the levels
the only defence mechanism. of various groups of bacteria in the colon, and these
Zebrafish Pglyrp5 and Pglyrp6 might also affect alterations are unique for mice deficient in different
multiple intracellular pathways. Inhibition of Pglyrp5 PGRPs. For example, there is a decrease in the numbers
expression in the developing embryo using small inter- of lactobacilli (which often have beneficial probiotic
fering RNA modified the expression of genes involved in effects) in Pglyrp2–/–, Pglyrp3–/– and Pglyrp4–/– mice.

846 | DECEMBER 2011 | VOLUME 11 www.nature.com/reviews/immunol

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

C&CPKQTGTKQ
5MKP +PVGUVKPG )KNNU

2IN[TR
2IN[TR
2IN[TR
CPF2IN[TR

)QPCFU .KXGT

2IN[TR
2IN[TR
$CEVGTKEKFCN
CPF2IN[TR
2IN[TR 2IN[TR
$CEVGTKWO 2IN[TR #OKFCUG
2IN[TR
KPYCVGT 2IN[TR CEVKXKV[ 2GRVKFQIN[ECP

2IN[TR
&QYPTGIWNCVG 6.4
GZRTGUUKQP 6.4
+.4
'II /-0-
2IN[TR 0(κ$
'ODT[Q

D/COOCNU
2)42FGȮEKGPV 2)42UWȰEKGPV
*CTOHWN $GPGȮEKCN
OKETQQTICPKUOU &55 OKETQQTICPKUOU &55
%QNQPKE
GRKVJGNKWO

+PLWT[ +PLWT[

.COKPC +(0γ +(0γ


RTQRTKC
0-EGNN
%:%. %:%.
0-EGNN %:%. %:%.
%:%. %:%.

%QNKVKU 2TQVGEVKQPHTQOEQNKVKU

0QDNGGFKPI
$NGGFKPI

.QUUQHEQNQPKE *GCNKPIQHEQNQPKE
GRKVJGNKWO 7NEGT GRKVJGNKWO

*[RGTRNCUKCQH 0QJ[RGTRNCUKC
NCOKPCRTQRTKC QHNCOKPCRTQRTKC

Figure 6 | Vertebrate PGRPs are bactericidal, defend against and, following intestinal injury with dextran sulphate sodium (DSS), this
infections, regulate intestinal microorganisms and protect results in increased production of natural killer (NK) cell-attracting and
0CVWTG4GXKGYU^+OOWPQNQI[
mammals against colitis. a | Danio rerio (zebrafish) peptidoglycan -activating chemokines (namely, CXCL9, CXCL10 and CXCL11). This
recognition proteins (PGRPs) are expressed in the adults, eggs and leads to the recruitment of NK cells to the colonic epithelium, the
embryos and protect against bacterial infections during early production of interferon-γ (IFNγ) by NK cells, the activation of
development. Zebrafish PGRPs have amidase, bactericidal and IFN-inducible genes (which include NK cell-attracting chemokine
immunomodulatory activities and are differentially expressed in various genes) and eventually colitis, which is manifested by hyperplasia of the
organs, including the skin, gills, intestine, liver and gonads in the adult lamina propria, loss of colonic epithelium, ulceration, bleeding and
fish. Zebrafish embryos develop in bacteria-contaminated water and death. PGRPs promote beneficial gut microorganisms that do not
rely on innate immunity for host defence. Zebrafish eggs express induce these chemokines and do not cause NK cell recruitment and
Pglyrp2 and the embryos express Pglyrp2, Pglyrp5 and Pglyrp6, and this IFNγ production, and thus they protect against colitis. This paradigm has
protects the eggs and embryos from bacterial infections. b | Mammalian been experimentally verified in a mouse model of colitis, but it probably
PGRPs promote beneficial intestinal microorganisms, which facilitate applies to all mammals, including humans. IL‑17R, interleukin‑17
healing following intestinal injury and thus protect against colitis. A lack receptor; MKNK2, MAPK-interacting serine/threonine kinase 2;
of PGRPs results in the overgrowth of harmful intestinal microorganisms NF‑κB, nuclear factor‑κB; TLR, Toll-like receptor.

NATURE REVIEWS | IMMUNOLOGY VOLUME 11 | DECEMBER 2011 | 847

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 1 | PGRPs kill bacteria by activating two-component systems


Peptidoglycan recognition proteins (PGRPs) kill bacteria by exploiting bacterial two-component stress response systems,
such as CssR–CssS in Bacillus subtilis and CpxA–CpxR in Escherichia coli85 (see the figure). These two-component systems
consist of a transmembrane sensor and a cytoplasmic regulator; they detect extracytoplasmic misfolded and aggregated
bacterial proteins, which are generated under stress, exported from the cell and degraded by proteases104,105.
In the case of Gram-positive bacteria, PGRPs bind to peptidoglycan in bacterial cell walls at the sites of hydrolysis
by LytE and LytF enzymes, which in B. subtilis separate daughter cells after cell division. In the case of Gram-negative
bacteria, PGRPs bind uniformly to the entire outer membrane85, which is composed of lipopolysaccharide (LPS) and
covers a thin peptidoglycan layer. In addition to binding peptidoglycan12,15,20, PGRPs bind to LPS and other molecules23–25
using binding sites outside the peptidoglycan-binding groove. The binding of PGRPs to peptidoglycan or LPS probably
induces PGRP oligomerization into ribbon-like structures106, which are then detected by the B. subtilis CssR–CssS or
E. coli CpxA–CpxR two-component systems85.
Activation of the protein-sensing two-component systems not only triggers the removal of the misfolded proteins,
but also signals into the cell to activate a repair and defence response, and induces membrane depolarization and the
production of toxic hydroxyl radicals (•OH). This is accompanied by the cessation of all major intracellular biosynthetic
reactions, probably because of the lack of energy generation through mechanisms that are dependent on the membrane
potential85,105. If stress is sustained or its level is too high, the bacteria die. This suicide mechanism might have evolved as
being advantageous to the entire bacterial population, because bacteria that are damaged beyond their ability to repair
themselves would die quickly and not use up scarce resources. The mammalian PGRPs (and probably other PGRPs)
trigger this bacterial defence/suicide mechanism to kill bacteria85. PGRPs use this mechanism both to defend against
pathogens79 and to control the levels of symbiotic microorganisms97; it is possible that symbiotic microorganisms
have a higher tolerance to PGRPs than other bacteria26.

$CEKNNWUUWDVKNKU 'UEJGTKEJKCEQNK
2)42
1WVGT
2)42 OGODTCPG
2GRVKFQIN[ECP
.25

%GNN %GNN
.[V' OGODTCPG OGODTCPG
.[V( /GODTCPG %RZ#
%UU5 FGRQNCTK\CVKQP
%UU4 %RZ4

r1* +PJKDKVKQPQH r1*


DKQU[PVJGUKUQH
OCETQOQNGEWNGU

$CEVGTKCNFGCVJ

0CVWTG4GXKGYU^+OOWPQNQI[
The stool microbiota from PGRP-deficient mice is ability of stools from PGRP-deficient mice, but not stools
more pro-inflammatory and more damaging to the from wild-type mice, to transfer this increased sensitivity
colon than the stool microbiota from wild-type mice. to colitis to wild-type germ-free mice97. This scenario
So, in vivo, after intestinal injury (for example, by DSS), is an experimental example of a genetic deficiency that
the more pro-inflammatory and damaging microbiota results in dysbiosis, with the clinical consequence of
in the colon of PGRP-deficient mice induces the pro- increased sensitivity to colitis following damage to the
duction of higher levels of chemokines that attract colonic epithelium. Nucleotide-binding oligomerization
innate immune cells, in particular natural killer (NK) domain 2 (Nod2) is another gene that, when mutated,
cells, to the site of injury. NK cells produce excessive induces intestinal dysbiosis and increased sensitivity to
amounts of interferon‑γ, which results in the activa- inflammatory bowel disease (IBD)99.
tion of interferon-inducible genes (including more NK Thus, in wild-type animals, PGRPs promote the
cell-attracting chemokine genes) that promote intesti- acquisition and maintenance of beneficial gut micro-
nal damage and prevent healing. This insult leads to organisms that, following intestinal injury, promote
hyperplasia of the colonic lamina propria (the layer of intestinal healing and protect the animal from devel-
cells underlying the colonic epithelium), loss of colonic oping severe and usually lethal colitis (FIG. 6b). Although
epithelium, ulceration, bleeding and eventually death97. not yet proven, PGRPs probably have a similar protec-
The role of dysregulation of the colonic microbiota tive role in all mammals, including humans, through
as the primary factor responsible for this sequence of the maintenance of beneficial microbial communities
events in PGRP-deficient mice has been shown by the within the gut.

848 | DECEMBER 2011 | VOLUME 11 www.nature.com/reviews/immunol

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

Conclusions and future directions with IBD101. The human PGLYRP2 gene is located at
PGRPs control the levels of symbiotic microorganisms position 19p13.12, within the IBD susceptibility locus
and defend against other bacteria in all animals that have 19p13, for which the susceptibility gene has not yet been
so far been studied. In some animals, they have very spe- identified101–103. Therefore, PGLYRP2 could be one of
cialized functions to control highly specialized symbi- the human IBD susceptibility genes. Mutations in other
onts, whereas in other animals they control and defend PGRP genes could also predispose patients to IBD, as
against a wide range of microorganisms, and even control many previously unknown IBD susceptibility genes
parasites. PGRPs accomplish these tasks both through are continuing to be discovered. These possibilities are
direct antibacterial activity and through indirect effects, currently being tested by genotyping patients with IBD.
including the induction of antimicrobial peptides and the If human PGRPs are associated with IBD, it should be
modulation of inflammation and immune responses. determined whether this effect is mediated through
There are many questions regarding how PGRPs changes in the intestinal microbiota and whether this
accomplish these functions. PGRPs kill bacteria by acti- predisposition could be treated by oral administration
vating their two-component systems (BOX 1). Future work of PGRPs.
should define whether this activation occurs directly or Many other applications based on the interactions
indirectly and how this activation induces bactericidal of PGRPs with the microbiota can be envisioned. New
mechanisms, such as membrane depolarization and the antimicrobial therapies could exploit the activation
inhibition of biosynthetic reactions. of bacterial and fungal two-component systems by
Although we know that PGRPs induce changes in PGRPs, PGRP fragments or PGRP mimetics. The role
the microbiota, future work should define these changes of PGRPs in maintaining beneficial microorganisms in
more precisely and should determine how alterations in the mouth and gut could be applied to develop toothpastes
the microbiota induce diseases or immunological imbal- and mouthwashes that promote this healthy microbiota.
ance. It should also be determined whether the effects of Transgenic cows that express human PGRPs in milk could
PGRPs on the gut microbial communities have systemic be produced, and this milk could be used to reduce the
consequences in other inflammatory diseases and, if so, incidence of food-borne infections and to promote
which microbial products mediate these effects. Although the maintenance of beneficial intestinal micro­organisms.
it is possible that all the in vivo effects of PGRPs are based Furthermore, PGRPs or PGRP mimetics could be used
on changes in the microbiota, it is likely that some effects to control insect-transmitted diseases, such as malaria
of PGRPs (for example, PGLYRP2‑dependent sensitivity and trypanosomiasis. Parallel dissection of PGRP
to experimental arthritis100) are based on other intrinsic functions in various invertebrates and vertebrates, and
characteristics of PGRPs, and the mechanisms responsible a more intense focus on the functions of PGRPs in
for these outcomes need to be defined. mammals will shed more light on PGRP evolution and
Increased sensitivity to colitis in PGRP-deficient mice lead to new approaches to control the microbiome and
raises the possibility that genetic defects in PGRPs might to prevent and treat infections and other diseases that are
predispose humans to IBD. Many genes are associated affected by the microbiome.

1. Ley, R. E. et al. Evolution of mammals and their gut 13. Yoshida, H., Kinoshita, K. & Ashida, M. Purification of in vitro. Proc. Natl Acad. Sci. USA 102, 6455–6460
microbes. Science 320, 1647–1651 (2008). a peptidoglycan recognition protein from hemolymph (2005).
2. Turnbaugh, P. J. et al. The human microbiome project. of the silkworm, Bombyx mori. J. Biol. Chem. 271, 22. Chang, C. I. et al. Structure of the ectodomain of
Nature 449, 804–810 (2007). 13854–13860 (1996). Drosophila peptidoglycan-recognition protein LCa
3. Lee, Y. K. & Mazmanian, S. K. Has the microbiota 14. Kang, D., Liu, G., Lundstrom, A., Gelius, E. & Steiner, H. suggests a molecular mechanism for pattern
played a critical role in the evolution of the adaptive A peptidoglycan recognition protein in innate recognition. Proc. Natl Acad. Sci. USA 102,
immune system? Science 330, 1768–1773 (2010). immunity conserved from insects to humans. 10279–10284 (2005).
4. Round, J. L. & Mazmanian, S. K. The gut microbiota Proc. Natl Acad. Sci. USA 95, 10078–10082 (1998). 23. Liu, C., Gelius, E., Liu, G., Steiner, H. & Dziarski, R.
shapes intestinal immune responses during health This was the first description of cloning of insect Mammalian peptidoglycan recognition protein binds
and disease. Nature Rev. Immunol. 9, 313–323 and mammalian PGRP genes, showing that PGRPs peptidoglycan with high affinity, is expressed in
(2009). are conserved in evolution. neutrophils and inhibits bacterial growth. J. Biol.
5. Heijtz, R. D. et al. Normal gut microbiota modulates 15. Dziarski, R. & Gupta, D. The peptidoglycan recognition Chem. 275, 24490–24499 (2000).
brain development and behavior. Proc. Natl Acad. Sci. proteins (PGRPs). Genome Biol. 7, 232 (2006). 24. Tydell, C. C., Yuan, J., Tran, P. & Selsted, M. E. Bovine
USA 108, 3047–3052 (2011). 16. Werner, T. et al. A family of peptidoglycan recognition peptidoglycan recognition protein‑S: antimicrobial
6. Ley, R. E. Obesity and the human microbiome. proteins in the fruit fly Drosophila melanogaster. activity, localization, secretion and binding properties.
Curr. Opin. Gastroenterol. 26, 5–11 (2010). Proc. Natl Acad. Sci. USA 97, 13772–13777 (2000). J. Immunol. 176, 1154–1162 (2006).
7. Chung, H. & Kasper, D. L. Microbiota-stimulated 17. Christophides, G. K. et al. Immunity-related genes and 25. Sharma, P. et al. Structural basis of recognition of
immune mechanisms to maintain gut homeostasis. gene families in Anopheles gambiae. Science 298, pathogen-associated molecular patterns and
Curr. Opin. Immunol. 22, 455–460 (2010). 159–165 (2002). inhibition of proinflammatory cytokines by camel
8. Wen, L. et al. Innate immunity and intestinal 18. Liu, C., Xu, Z., Gupta, D. & Dziarski, R. Peptidoglycan peptidoglycan recognition protein. J. Biol. Chem.
microbiota in the development of type 1 diabetes. recognition proteins: a novel family of four human 286, 16208–16217 (2011).
Nature 455, 1109–1113 (2008). innate immunity pattern recognition molecules. 26. Lu, X. et al. Peptidoglycan recognition proteins are
9. Maslowski, K. M. et al. Regulation of inflammatory J. Biol. Chem. 276, 34686–34694 (2001). a new class of human bactericidal proteins.
responses by gut microbiota and chemoattractant This study identified a family of PGRPs in mammals. J. Biol. Chem. 281, 5895–5907 (2006).
receptor GPR43. Nature 461, 1282–1286 (2009). 19. Kim, M.‑S., Byun, M. & Oh, B.‑H. Crystal structure 27. Michel, T., Reichhart, J. M., Hoffmann, J. A. &
10. Atarashi, K. et al. ATP drives lamina propria of peptidoglycan recognition protein LB from Royet, J. Drosophila Toll is activated by Gram-
TH17 cell differentiation. Nature 455, 808–812 Drosophila melanogaster. Nature Immunol. positive bacteria through a circulating peptidoglycan
(2008). 4, 787–793 (2003). recognition protein. Nature 414, 756–759 (2001).
11. Cerf-Bensussan, N. & Gaboriau-Routhiau, V. The This was the first reported crystal structure of a This study presented the first in vivo evidence that
immune system and the gut microbiota: friends or PGRP protein. PGRP molecules can function as PRRs in insects.
foes? Nature Rev. Immunol. 10, 735–744 (2010). 20. Guan, R. et al. Structural basis for peptidoglycan 28. Choe, K. M., Werner, T., Stoven, S., Hultmark, D. &
12. Royet, J. & Dziarski, R. Peptidoglycan recognition binding by peptidoglycan recognition proteins. Anderson, K. V. Requirement for a peptidoglycan
proteins: pleiotropic sensors and effectors of Proc. Natl Acad. Sci. USA 101, 17168–17173 (2004). recognition protein (PGRP) in Relish activation and
antimicrobial defences. Nature Rev. Microbiol. 21. Mellroth, P. et al. Ligand-induced dimerization of antibacterial immune responses in Drosophila.
5, 264–277 (2007). Drosophila peptidoglycan recognition proteins Science 296, 359–362 (2002).

NATURE REVIEWS | IMMUNOLOGY VOLUME 11 | DECEMBER 2011 | 849

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

29. Gottar, M. et al. The Drosophila immune response 49. Corby-Harris, V. et al. Geographical distribution and 69. Itoh, N. & Takahashi, K. G. Distribution of multiple
against Gram-negative bacteria is mediated by a diversity of bacteria associated with natural peptidoglycan recognition proteins in the tissues of
peptidoglycan recognition protein. Nature 416, populations of Drosophila melanogaster. Appl. Pacific oyster, Crassostrea gigas. Comp. Biochem.
640–644 (2002). Environ. Microbiol. 73, 3470–3479 (2007). Physiol. B, Biochem. Mol. Biol. 150, 409–417
30. Ramet, M., Manfruelli, P., Pearson, A., 50. Cox, C. R. & Gilmore, M. S. Native microbial (2008).
Mathey-Prevot, B. & Ezekowitz, R. A. Functional colonization of Drosophila melanogaster and its use 70. Su, J., Ni, D., Song, L., Zhao, J. & Qiu, L.
genomic analysis of phagocytosis and identification as a model of Enterococcus faecalis pathogenesis. Molecular cloning and characterization of a short type
of a Drosophila receptor for E. coli. Nature 416, Infect. Immun. 75, 1565–1576 (2007). peptidoglycan recognition protein (CfPGRP‑S1) cDNA
644–648 (2002). 51. Ren, C., Webster, P., Finkel, S. E. & Tower, J. from Zhikong scallop Chlamys farreri. Fish Shellfish
References 28–30 demonstrated that a Increased internal and external bacterial load Immunol. 23, 646–656 (2007).
D. melanogaster PGRP functions both as a PRR during Drosophila aging without life-span trade-off. 71. Yang, J. et al. Peptidoglycan recognition protein of
and as a signalling transmembrane receptor Cell Metab. 6, 144–152 (2007). Chlamys farreri (CfPGRP‑S1) mediates immune
upstream of the IMD pathway. 52. Ryu, J. H. et al. Innate immune homeostasis by the defenses against bacterial infection. Dev. Comp.
31. Gobert, V. et al. Dual activation of the Drosophila homeobox gene Caudal and commensal–gut mutualism Immunol. 34, 1300–1307 (2010).
Toll pathway by two pattern recognition receptors. in Drosophila. Science 319, 777–782 (2008). 72. Goodson, M. S. et al. Identifying components of the
Science 302, 2126–2130 (2003). A demonstration of immune tolerance in the NF‑κB pathway in the beneficial Euprymna scolopes–
32. Bischoff, V. et al. Function of the Drosophila pattern- D. melanogaster gut and the participation of Vibrio fischeri light organ symbiosis. Appl. Environ.
recognition receptor PGRP-SD in the detection of Caudal in this process. Microbiol. 71, 6934–6946 (2005).
Gram-positive bacteria. Nature Immunol. 53. Storelli, G. et al. Lactobacillus plantarum promotes 73. Troll, J. V. et al. Peptidoglycan induces loss of a
5, 1175–1180 (2004). Drosophila systemic growth by modulating hormonal nuclear peptidoglycan recognition protein during host
33. Wang, L. et al. Peptidoglycan recognition protein-SD signals through TOR dependent nutrient sensing. tissue development in a beneficial animal–bacterial
provides versatility of receptor formation in Cell Metab. 14, 403–414 (2011). symbiosis. Cell. Microbiol. 11, 1114–1127 (2009).
Drosophila immunity. Proc. Natl Acad. Sci. USA 105, 54. Sharon, G. et al. Commensal bacteria play a role in 74. Troll, J. V. et al. Taming the symbiont for coexistence:
11881–11886 (2008). mating preference of Drosophila melanogaster. a host PGRP neutralizes a bacterial symbiont toxin.
34. Rutschmann, S. et al. The Rel protein DIF mediates Proc. Natl Acad. Sci. USA 107, 20051–20056 (2010). Environ. Microbiol. 12, 2190–2203 (2010).
the antifungal but not the antibacterial host defense 55. Bischoff, V. et al. Downregulation of the Drosophila This study identified the role of PGRP2 in
in Drosophila. Immunity 12, 569–580 (2000). immune response by peptidoglycan-recognition Euprymna scolopes–Vibrio fischeri symbiosis.
35. Takehana, A. et al. Overexpression of a pattern- proteins SC1 and SC2. PLoS Pathog. 2, e14 (2006). 75. Koropatnick, T. A. et al. Microbial factor-mediated
recognition receptor, peptidoglycan-recognition 56. Zaidman-Remy, A. et al. The Drosophila amidase development in a host–bacterial mutualism. Science
protein-LE, activates Imd/Relish-mediated PGRP-LB modulates the immune response to bacterial 306, 1186–1188 (2004).
antibacterial defense and the prophenoloxidase infection. Immunity 24, 463–473 (2006). 76. Nyholm, S. V. & McFall-Ngai, M. J. The winnowing:
cascade in Drosophila larvae. Proc. Natl Acad. Sci. References 55 and 56 showed that PGRPs with establishing the squid–vibrio symbiosis. Nature Rev.
USA 99, 13705–13710 (2002). amidase activity dampen the D. melanogaster Microbiol. 2, 632–642 (2004).
36. Leulier, F. et al. The Drosophila immune system immune response. 77. Zhang, S. M., Zeng, Y. & Loker, E. S. Characterization
detects bacteria through specific peptidoglycan 57. Gendrin, M., Welchman, D. P., Poidevin, M., Hervé, M. of immune genes from the schistosome host snail
recognition. Nature Immunol. 4, 478–484 (2003). & Lemaitre, B. Long-range activation of systemic Biomphalaria glabrata that encode peptidoglycan
A demonstration that D. melanogaster can immunity through peptidoglycan diffusion in recognition proteins and Gram-negative bacteria
distinguish between different bacterial Drosophila. PLoS Pathog. 5, e1000694 (2009). binding protein. Immunogenetics 59, 883–898
peptidoglycans. Lys-type peptidoglycans activate 58. Kleino, A. et al. Pirk is a negative regulator of the (2007).
the Toll pathway, whereas DAP-type peptidoglycans Drosophila Imd pathway. J. Immunol. 180, 78. Coteur, G. et al. Peptidoglycan recognition proteins
activate the IMD pathway. 5413–5422 (2008). with amidase activity in early deuterostomes
37. Choe, K. M., Lee, H. & Anderson, K. V. Drosophila 59. Lhocine, N. et al. PIMS modulates immune tolerance (Echinodermata). Dev. Comp. Immunol. 31, 790–804
peptidoglycan recognition protein LC (PGRP-LC) acts by negatively regulating Drosophila innate immune (2007).
as a signal-transducing innate immune receptor. signaling. Cell Host Microbe 4, 147–158 (2008). 79. Li, X. et al. Zebrafish peptidoglycan recognition
Proc. Natl Acad. Sci. USA 102, 1122–1126 (2005). 60. Aggarwal, K. et al. Rudra interrupts receptor signaling proteins are bactericidal amidases essential for
38. Werner, T., Borge-Renberg, K., Mellroth, P., complexes to negatively regulate the IMD pathway. defense against bacterial infections. Immunity 27,
Steiner, H. & Hultmark, D. Functional diversity of the PLoS Pathog. 4, e1000120 (2008). 518–529 (2007).
Drosophila PGRP-LC gene cluster in the response to 61. Buchon, N., Broderick, N. A., Chakrabarti, S. & In this study, zebrafish PGRP genes were cloned
lipopolysaccharide and peptidoglycan. J. Biol. Chem. Lemaitre, B. Invasive and indigenous microbiota and the amidase and bactericidal activities of these
278, 26319–26322 (2003). impact intestinal stem cell activity through multiple PGRPs and their role in the protection of embryos
39. Kaneko, T. et al. Monomeric and polymeric Gram- pathways in Drosophila. Genes Dev. 23, 2333–2344 from bacterial infections were identified.
negative peptidoglycan but not purified LPS stimulate (2009). 80. Kim, M. Y., Jang, J. H., Lee, J. W. & Cho, J. H.
the Drosophila IMD pathway. Immunity 20, 637–649 62. Ha, E. M., Oh, C. T., Bae, Y. S. & Lee, W. J. A direct Molecular cloning and characterization of
(2004). role for dual oxidase in Drosophila gut immunity. peptidoglycan recognition proteins from the rockfish,
40. Chang, C. I., Chelliah, Y., Borek, D., Mengin-Lecreulx, D. Science 310, 847–850 (2005). Sebastes schlegeli. Fish Shellfish Immunol. 28,
& Deisenhofer, J. Structure of tracheal cytotoxin in A demonstration that ROS production has an 632–639 (2010).
complex with a heterodimeric pattern-recognition essential role in the gut immune response in 81. Mao, Y., Wang, J., Zhang, Z., Ding, S. & Su, Y.
receptor. Science 311, 1761–1764 (2006). D. melanogaster. Cloning, mRNA expression, and recombinant
41. Maillet, F., Bischoff, V., Vignal, C., Hoffmann, J. & 63. Ha, E. M. et al. Coordination of multiple dual oxidase- expression of peptidoglycan recognition protein II
Royet, J. The Drosophila peptidoglycan recognition regulatory pathways in responses to commensal and gene from large yellow croaker (Pseudosciaena
protein PGRP-LF blocks PGRP-LC and IMD/JNK infectious microbes in Drosophila gut. Nature crocea). Mol. Biol. Rep. 37, 3897–3908 (2010).
pathway activation. Cell Host Microbe 3, 293–303 Immunol. 10, 949–957 (2009). 82. Chang, M. X. & Nie, P. RNAi suppression of zebrafish
(2008). 64. Meister, S. et al. Anopheles gambiae PGRPLC- peptidoglycan recognition protein 6 (zfPGRP6)
42. Basbous, N. et al. The Drosophila peptidoglycan- mediated defense against bacteria modulates mediated differentially expressed genes involved in
recognition protein LF interacts with peptidoglycan- infections with malaria parasites. PLoS Pathog. Toll-like receptor signaling pathway and caused
recognition protein LC to downregulate the Imd 5, e1000542 (2009). increased susceptibility to Flavobacterium columnare.
pathway. EMBO Rep. 12, 327–333 (2011). 65. Dong, Y., Manfredini, F. & Dimopoulos, G. Implication Vet. Immunol. Immunopathol. 124, 295–301 (2008).
43. Yano, T. et al. Autophagic control of Listeria through of the mosquito midgut microbiota in the defense 83. Chang, M. X., Wang, Y. P. & Nie, P. Zebrafish
intracellular innate immune mechanism. Nature against malaria parasites. PLoS Pathog. 5, e1000423 peptidoglycan recognition protein SC (zfPGRP-SC)
Immunol. 9, 908–916 (2008). (2009). mediates multiple intracellular signaling pathways.
A demonstration that PGRP-LE functions as a PRR A demonstration that the PGRP‑LC‑mediated Fish Shellfish Immunol. 26, 264–274 (2009).
for bacteria in the cytoplasm. response to the gut microbiota in mosquitoes has 84. Wang, M. et al. Human peptidoglycan recognition
44. Kaneko, T. et al. PGRP-LC and PGRP-LE have essential an indirect effect on parasite transmission. proteins require zinc to kill both Gram-positive and
yet distinct functions in the Drosophila immune 66. Pais, R., Lohs, C., Wu, Y., Wang, J. & Aksoy, S. The Gram-negative bacteria and are synergistic with
response to monomeric DAP-type peptidoglycan. obligate mutualist Wigglesworthia glossinidia antibacterial peptides. J. Immunol. 178, 3116–3125
Nature Immunol. 7, 715–723 (2006). influences reproduction, digestion, and immunity (2007).
45. Mellroth, P. & Steiner, H. PGRP‑SB1: an processes of its host, the tsetse fly. Appl. Environ. 85. Kashyap, D. R. et al. Peptidoglycan recognition
N‑acetylmuramoyl l‑alanine amidase with antibacterial Microbiol. 74, 5965–5974 (2008). proteins kill bacteria by activating protein-sensing
activity. Biochem. Biophys. Res. Commun. 350, 67. Wang, J., Wu, Y., Yang, G. & Aksoy, S. Interactions two-component systems. Nature Med. 17, 676–683
994–999 (2006). between mutualist Wigglesworthia and tsetse (2011).
46. Park, J. W. et al. Clustering of peptidoglycan peptidoglycan recognition protein (PGRP-LB) influence A demonstration that PGRPs kill bacteria through
recognition protein-SA is required for sensing lysine- trypanosome transmission. Proc. Natl Acad. Sci. USA a novel mechanism based on the overactivation of
type peptidoglycan in insects. Proc. Natl Acad. Sci. 106, 12133–12138 (2009). bacterial two-component systems.
USA 104, 6602–6607 (2007). A demonstration of the role of PGRP-LB in 86. Wang, Z.‑M. et al. Human peptidoglycan recognition
47. Nehme, N. T. et al. A model of bacterial intestinal tolerance towards symbiotic bacteria and in protein‑L is an N‑acetylmuramoyl‑l-alanine amidase.
infections in Drosophila melanogaster. PLoS Pathog. trypanosome transmission in tsetse flies. J. Biol. Chem. 278, 49044–49052 (2003).
3, e173 (2007). 68. Anselme, C., Vallier, A., Balmand, S., Fauvarque, M. O. 87. Gelius, E., Persson, C., Karlsson, J. & Steiner, H.
48. Liehl, P., Blight, M., Vodovar, N., Boccard, F. & & Heddi, A. Host PGRP gene expression and bacterial A mammalian peptidoglycan recognition protein
Lemaitre, B. Prevalence of local immune response release in endosymbiosis of the weevil Sitophilus with N‑acetylmuramoyl‑l-alanine amidase activity.
against oral infection in a Drosophila/Pseudomonas zeamais. Appl. Environ. Microbiol. 72, 6766–6772 Biochem. Biophys. Res. Commun. 306, 988–994
infection model. PLoS Pathog. 2, e56 (2006). (2006). (2003).

850 | DECEMBER 2011 | VOLUME 11 www.nature.com/reviews/immunol

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

88. Tydell, C. C., Yount, N., Tran, D., Yuan, J. & Selsted, M. 96. Mathur, P. et al. Murine peptidoglycan recognition 104. Hyyryläinen, H. L. et al. A novel two-component
Isolation, characterization, and antimicrobial proteins PglyrpIα and PglyrpIβ are encoded in the regulatory system in Bacillus subtilis for the survival
properties of bovine oligosaccharide-binding protein. epidermal differentiation complex and are expressed of severe secretion stress. Mol. Microbiol. 41,
J. Biol. Chem. 277, 19658–19664 (2002). in epidermal and hematopoietic tissues. Genomics 83, 1159–1172 (2001).
89. Dziarski, R., Platt, K. A., Gelius, E., Steiner, H. & 1151–1163 (2004). 105. Kohanski, M. A., Dwyer, D. J., Wierzbowski, J.,
Gupta, D. Defect in neutrophil killing and increased 97. Saha, S. et al. Peptidoglycan recognition proteins Cottarel, G. & Collins, J. J. Mistranslation of
susceptibility to infection with non-pathogenic protect mice from experimental colitis by promoting membrane proteins and two-component system
Gram-positive bacteria in peptidoglycan recognition normal gut flora and preventing induction of activation trigger antibiotic-mediated cell death.
protein‑S (PGRP‑S)-deficient mice. Blood 102, interferon‑γ. Cell Host Microbe 8, 147–162 (2010). Cell 135, 679–690 (2008).
689–697 (2003). A demonstration that mammalian PGRPs protect 106. Lim, J. H. et al. Structural basis for preferential
90. Cho, J. H. et al. Human peptidoglycan recognition the host from colitis by maintaining the beneficial, recognition of diaminopimelic acid-type
protein S is an effector of neutrophil-mediated innate normal gut microbiota. peptidoglycan by a subset of peptidoglycan
immunity. Blood 106, 2552–2558 (2005). 98. Kappeler, S. R., Heuberger, C., Farah, Z. & Puhan, Z. recognition proteins. J. Biol. Chem. 281,
91. Xu, M., Wang, Z. & Locksley, R. M. Innate Expression of the peptidoglycan recognition protein, 8286–8295 (2006).
immune responses in peptidoglycan recognition PGRP, in the lactating mammary gland. J. Dairy Sci.
protein L‑deficient mice. Mol. Cell. Biol. 24, 87, 2660–2668 (2004). Acknowledgements
7949–7957 (2004). 99. Rehman, A. et al. Nod2 is essential for temporal Research in the authors’ laboratories is supported by the
92. Zhang, Y. et al. Identification of serum development of intestinal microbial communities. National Institutes of Health (D.G. and R.D.) and the Action
N‑acetylmuramoyl‑l-alanine amidase as liver Gut 60, 1354–1362 (2011). Concertée Insitative, Fondation pour la Recherche Médicale,
peptidoglycan recognition protein 2. Biochim. 100. Saha, S. et al. PGLYRP‑2 and Nod2 are both Institut Universitaire de France, and Agence Nationale de la
Biophys. Acta 1752, 34–46 (2005). required for peptidoglycan-induced arthritis and local Recherche (J.R.).
93. Wang, H., Gupta, D., Li, X. & Dziarski, R. inflammation. Cell Host Microbe 5, 137–150 (2009).
Peptidoglycan recognition protein 2 101. Budarf, M. L., Labbé, C., David, G. & Rioux, J. D. Competing interests statement
(N‑acetylmuramoyl‑l-Ala amidase) is induced in GWA studies: rewriting the story of IBD. Trends Genet. The authors declare no competing financial interests.
keratinocytes by bacteria through the p38 kinase 25, 137–146 (2009).
pathway. Infect. Immun. 73, 7216–7225 (2005). 102. Rioux, J. D. et al. Genomewide search in Canadian
94. Uehara, A. et al. Chemically synthesized pathogen- families with inflammatory bowel disease reveals two FURTHER INFORMATION
associated molecular patterns increase the expression novel susceptibility loci. Am. J. Hum. Genet. 66, Julien Royet’s homepage: http://www.ibdml.univ-mrs.fr/
of peptidoglycan recognition proteins via Toll-like 1863–1870 (2000). equipes/equipe_gb.php?id=63
receptors, NOD1 and NOD2 in human oral epithelial 103. Lakatos, P. L., Fischer, S., Lakatos, L., Gal, I. & Papp, J. Dipika Gupta’s homepage: http://iusm-nw.medicine.iu.edu/
cells. Cell. Microbiol. 7, 675–686 (2005). Current concept on the pathogenesis of inflammatory faculty/dipika-gupta-ph-d/
95. Li, X., Wang, S., Wang, H. & Gupta, D. Differential bowel disease-crosstalk between genetic and microbial Roman Dziarski’s homepage: http://iusm-nw.medicine.iu.
expression of peptidoglycan recognition protein 2 in factors: pathogenic bacteria and altered bacterial edu/faculty/roman-dziarski-ph-d/
the skin and liver requires different transcription sensing or changes in mucosal integrity take “toll”? ALL LINKS ARE ACTIVE IN THE ONLINE PDF
factors. J. Biol. Chem. 281, 20738–20748 (2006). World J. Gastroenterol. 12, 1829–1841 (2006).

NATURE REVIEWS | IMMUNOLOGY VOLUME 11 | DECEMBER 2011 | 851

© 2011 Macmillan Publishers Limited. All rights reserved

You might also like