You are on page 1of 11

Central Role for MyD88 in the Responses of

Microglia to Pathogen-Associated Molecular


Patterns
This information is current as Nilufer Esen and Tammy Kielian
of April 14, 2022. J Immunol 2006; 176:6802-6811; ;
doi: 10.4049/jimmunol.176.11.6802
http://www.jimmunol.org/content/176/11/6802

Downloaded from http://www.jimmunol.org/ by guest on April 14, 2022


References This article cites 76 articles, 21 of which you can access for free at:
http://www.jimmunol.org/content/176/11/6802.full#ref-list-1

Why The JI? Submit online.


• Rapid Reviews! 30 days* from submission to initial decision
• No Triage! Every submission reviewed by practicing scientists
• Fast Publication! 4 weeks from acceptance to publication
*average

Subscription Information about subscribing to The Journal of Immunology is online at:


http://jimmunol.org/subscription
Permissions Submit copyright permission requests at:
http://www.aai.org/About/Publications/JI/copyright.html
Email Alerts Receive free email-alerts when new articles cite this article. Sign up at:
http://jimmunol.org/alerts

The Journal of Immunology is published twice each month by


The American Association of Immunologists, Inc.,
1451 Rockville Pike, Suite 650, Rockville, MD 20852
Copyright © 2006 by The American Association of
Immunologists All rights reserved.
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
The Journal of Immunology

Central Role for MyD88 in the Responses of Microglia to


Pathogen-Associated Molecular Patterns1

Nilufer Esen and Tammy Kielian2


Microglia, the innate immune effector cells of the CNS parenchyma, express TLR that recognize conserved motifs of microor-
ganisms referred to as pathogen-associated molecular patterns (PAMP). All TLRs identified to date, with the exception of TLR3,
use a common adaptor protein, MyD88, to transduce activation signals. Recently, we reported that microglial activation in
response to the Gram-positive bacterium Staphylococcus aureus was not completely attenuated following TLR2 ablation, sug-
gesting the involvement of additional receptors. To assess the functional role of alternative TLRs in microglial responses to S.
aureus and its cell wall product peptidoglycan as well as the Gram-negative PAMP LPS, we evaluated primary microglia from
MyD88 knockout (KO) and wild-type mice. The induction of TNF-␣, IL-12 p40, and MIP-2 (CXCL2) expression by S. aureus- and
peptidoglycan-stimulated microglia was MyD88 dependent, as revealed by the complete inhibition of cytokine production in
MyD88 KO cells. In addition, the expression of additional pattern recognition receptors, including TLR9, pentraxin-3, and

Downloaded from http://www.jimmunol.org/ by guest on April 14, 2022


lectin-like oxidized LDL receptor-1, was regulated, in part, via a MyD88-dependent manner as demonstrated by the attenuated
expression of these receptors in MyD88 KO microglia. Microglial activation was only partially inhibited in LPS-stimulated MyD88
KO cells, suggesting the involvement of MyD88-independent pathways. Collectively, these findings reveal the complex mechanisms
for microglia to respond to diverse bacterial pathogens, which occur via both MyD88-dependent and -independent pathways. The
Journal of Immunology, 2006, 176: 6802– 6811.

M icroglia, the resident innate immune effector cells of from the cell wall of Gram-positive bacteria (4, 5). Our laboratory
the CNS parenchyma, represent a first line of defense is interested in defining the signals responsible for microglial ac-
against invading CNS pathogens (1). As such, micro- tivation and subsequent induction of proinflammatory mediator ex-
glia are armed with an extensive repertoire of pattern recognition pression in response to the Gram-positive bacterium Staphylococ-
receptors (PRRs)3 including TLRs that are likely pivotal for max- cus aureus, a prevalent CNS pathogen and frequent etiologic agent
imizing pathogen detection (2). To date, the TLR family includes of brain abscess in humans (6, 7). Understanding the mechanisms
a total of 11 receptors that are responsible for the recognition of by which Gram-positive bacteria engage CNS glia may lead to the
highly conserved structural motifs that are essential for pathogen development of novel therapeutic strategies aimed at attenuating
survival and are conserved across broad subclasses of microorgan- inappropriate innate immune activation that has been associated
isms (3, 4). These conserved microbial structural motifs are re- with several CNS infectious diseases including bacterial meningi-
ferred to as pathogen-associated molecular patterns (PAMPs), and tis and brain abscess (8 –11).
at least one agonist has been identified for each TLR with the Recent studies have established that microglia constitutively ex-
exception of TLR10. TLR2 recognizes the widest array of PAMPs press TLR2 (12–18) (reviewed in Ref. 2). With regard to TLR2
in heterodimeric complexes with either TLR1 or TLR6, including
Ags, as defined by studies investigating the functional importance
lipoproteins and peptidoglycan (PGN) that are components of all
of TLR2 in macrophage activation, microglia are capable of rec-
bacteria possessing cell walls as well as lipoteichoic acid (LTA)
ognizing numerous TLR2 ligands including PGN, LTA, and tri-
palmitoyl-S-glycerylcysteine (Pam3Cys) (13, 16, 18 –21). Con-
cerning the functional importance of TLR2 in microglial
Department of Neurobiology and Developmental Sciences, University of Arkansas
for Medical Sciences, Little Rock, AR 72205 activation, our group was the first to report that this PRR plays an
Received for publication December 14, 2005. Accepted for publication March essential role in enabling PGN recognition using primary micro-
22, 2006. glia isolated from TLR2 knockout (KO) and wild-type (WT) mice
The costs of publication of this article were defrayed in part by the payment of page (18). Interestingly, microglial responses to the Gram-positive
charges. This article must therefore be hereby marked advertisement in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
pathogen S. aureus were found to be largely TLR2 independent,
1 with the majority of proinflammatory mediators examined in re-
This work was supported by the National Institutes of Health National Institute of
Mental Health (RO1 MH65297) to T.K. and the National Institute of Neurological sponse to intact bacteria equivalently expressed in both TLR2 KO
Disorders and Stroke-supported core facility at University of Arkansas for Medical and WT cells, suggesting that microglia use alternative PRRs for
Sciences (P30 NS047546).
2
S. aureus recognition (18). One possible candidate is TLR9 which
Address correspondence and reprint requests to Dr. Tammy Kielian, Department of
Neurobiology and Developmental Sciences, University of Arkansas for Medical Sci- recognizes nonmethylated CpG motifs that are prevalent in bacte-
ences, 4301 West Markham Street, Slot 846, Little Rock, AR 72205. E-mail address: rial DNA (5). In addition, it is plausible that TLR1 and TLR6, both
KielianTammyL@uams.edu of which are expressed by microglia (13, 16), are sufficient to
3
Abbreviations used in this paper: PRR, pattern recognition receptor; PAMP, patho- account for the residual proinflammatory mediator production ob-
gen-associated molecular pattern; PGN, peptidoglycan; LTA, lipoteichoic acid; KO,
knockout; WT, wild type; TIR, TLR/IL-1R; IRAK, IL-1R-associated kinase; qRT- served in TLR2 KO microglia following bacterial stimulation (18).
PCR, quantitative real-time RT-PCR; PTX3, pentraxin-3; LOX-1, lectin-like oxidized However, although several studies have defined the array of TLRs
low density lipoprotein receptor-1; NOD2, nucleotide-binding oligomerization do-
main 2; NOS, NO synthase; iNOS, inducible NOS; MDP, muramyl dipeptide; TRIF, expressed on microglia (12, 16, 22, 23), a comprehensive assess-
TIR domain-containing adaptor inducing IFN-␤; IRF-3, IFN regulatory factor-3. ment of the functional relevance of TLRs in enabling microglia to

Copyright © 2006 by The American Association of Immunologists, Inc. 0022-1767/06/$02.00


The Journal of Immunology 6803

recognize PAMPs has not yet been performed. Therefore, the cur- Unless otherwise indicated, heat-inactivated S. aureus (107 CFU/ml), PGN
rent study was designed to directly investigate the functional im- (10 ␮g/ml), and LPS (100 ng/ml) were added to microglial cultures at the
portance of multiple TLRs in mediating microglial activation final concentrations indicated, representing doses that were previously de-
termined to be optimal for inducing maximal proinflammatory cytokine
through the use of primary microglia isolated from MyD88 KO expression in microglia without any evidence of cytotoxicity (18, 35). In
and WT mice, where MyD88 represents a common adaptor protein these studies, we used a multiplicity of infection of S. aureus to microglia
required for signaling of all TLRs identified to date, with the ex- of 50:1. In our in vivo model of brain abscess, bacterial titers reach 106- 107
ception of TLR3. CFU within the first 24 h following infection and continue to peak until day
5 (10, 37). When one considers the focal nature of brain abscess, and hence
Upon ligand binding, the intracellular portion of TLRs, referred S. aureus localization in the brain parenchyma, and the fact that bacteria
to as the Toll/IL-1R (TIR) domain due to its similarity with the double in number every 5–10 min, the ratios of S. aureus:microglia used in
cytoplasmic domain of the IL-1R, initiates the TLR signaling cas- our study are likely an underrepresentation of the actual pathogen burden
cade (24, 25). The TIR domain is highly conserved among all that microglia would encounter in vivo.
TLRs (with the exception of TLR3) and mediates its downstream ELISA
signaling effects by interacting with the TIR-containing common
adaptor protein MyD88. Interestingly, in addition to TLRs, both Comparisons in cytokine and chemokine expression between MyD88 WT
and KO microglia were performed using standard sandwich ELISA kits
the IL-1 and IL-18 receptors also use MyD88 to transduce activa- according to the manufacturer’s instructions (OptEIA mouse TNF-␣, IL-12
tion signals emanating from receptor ligation by their respective p40, IL-1␤, and MCP-1/CCL2; BD Pharmingen; DuoSet mouse MIP-2/
cytokines (26). Association of the TIR domain of MyD88 with that CXCL2; R&D Systems). The level of sensitivity for all ELISAs was 15.6
of TLRs recruits the serine/threonine kinase IL-1R-associated ki- pg/ml.
nase (IRAK). Subsequently, IRAK is activated by autophosphor- Cell viability assays
ylation and dissociates from the receptor complex to interact with

Downloaded from http://www.jimmunol.org/ by guest on April 14, 2022


To confirm that the observed changes in inflammatory mediator expression
TNFR-associated factor-6. Consequently, the association of between MyD88 WT and KO microglia were not due to differences in cell
TNFR-associated factor-6 with IRAK leads to the activation of seeding densities or potential confounding proliferative effects, a standard
two distinct signaling pathways, namely the NF-␬B and MAPK MTT assay based on the mitochondrial conversion of MTT into formazan
pathways. Both the NF-␬B and MAPK pathways are capable of crystals was performed as previously described (35).
regulating the expression of numerous immune response genes in- Quantitative real-time RT-PCR (qRT-PCR)
cluding proinflammatory cytokines and chemokines (27). Several
groups have demonstrated the importance of MyD88 in signaling Total RNA from MyD88 WT and KO microglia was isolated using the
TRIzol reagent and treated with DNase1 (both from Invitrogen Life Tech-
through numerous TLRs either in MyD88 knockout mice (26, 28 – nologies) before use in qRT-PCR studies. The experimental procedure was
31) or through the expression of a mutant MyD88 protein that no performed as previously described (18). Briefly, TLR2 and GAPDH prim-
longer is capable of transducing an activation signal (32, 33). ers and TAMRA TaqMan probes were designed as previously described
However, the role of MyD88, and as an extension all TLRs, in (18, 38) and synthesized by Applied Biosystems. ABI Assays-on-Demand
TaqMan kits were used to examine microglial IL-23 p19, TLR9, pen-
enabling microglia to recognize and respond to PAMPs of clinical
traxin-3 (PTX3), lectin-like oxidized low density lipoprotein receptor-1
importance to CNS infectious diseases has not yet been examined. (LOX-1), and nucleotide-binding oligomerization domain 2 (NOD2) ex-
To determine the functional importance of multiple TLRs in S. pression. Comparisons in gene expression between MyD88 WT and KO
aureus-dependent microglial activation, we evaluated the role of primary microglia were calculated after normalizing cycle thresholds
MyD88-dependent pathways using primary microglia isolated against the “housekeeping” gene GAPDH and are presented as the fold-
induction (2⫺⌬⌬Ct, where Ct is the cycle threshold) value relative to un-
from MyD88 KO and WT mice. stimulated microglia.

Protein extraction and Western blotting


Materials and Methods
Protein extracts were prepared and quantitated from both MyD88 WT and
Primary microglia cell culture and reagents
KO microglia as previously described (35). The effects of heat-inactivated
MyD88 gene KO mice (originally from Dr. S. Akira, Osaka University, S. aureus, PGN, and LPS on microglial TLR2, MyD88, and inducible NO
Suita, Osaka, Japan) (26) were purchased from the Centre de La Recherche synthase (iNOS) protein expression were evaluated by Western blot anal-
Scientifique and have been previously backcrossed with C57BL/6 mice for ysis. Blots were probed using goat anti-mouse TLR2 (R&D Systems), rab-
over 10 generations (29, 34). Primary microglia were isolated from MyD88 bit anti-mouse MyD88 (eBioscience), or rabbit anti-mouse NOS2 (iNOS;
KO or WT C57BL/6 pups (Harlan Laboratories; 1– 8 days of age) as pre- Santa Cruz Biotechnology) Abs followed by the appropriate IgG-HRP con-
viously described (35). The purity of microglial cultures was evaluated by jugated secondary Abs (Jackson ImmunoResearch Laboratories). Blots
immunohistochemical staining using Abs against CD11b (BD Pharmingen) were stripped and reprobed with a rabbit anti-actin polyclonal Ab (Sigma-
and glial fibrillary acidic protein (DakoCytomation). The purity of primary Aldrich) for normalization. Blots were developed using the Immobilon
microglial cultures was routinely ⬎95%. Western substrate (Millipore) and visualized by exposure to x-ray film.
Heat-inactivated S. aureus (strain RN6390, provided by Dr. A. Cheung,
Dartmouth Medical School, Hanover, NH) was prepared as previously de- Statistics
scribed (13). PGN derived from S. aureus was obtained from Fluka and Significant differences between experimental groups were determined by
Invivogen and Escherichia coli O11:B1 LPS was from List Biological the Student t test at the 95% confidence interval using Sigma Stat (SPSS
Laboratories. Heat-inactivated S. aureus was used in these studies rather Science).
than live organisms to control the standard dosing of bacteria within and
between individual experiments. Live organisms exhibit an extremely rapid
doubling time (i.e., 5–10 min) that can quickly lead to culture overgrowth,
Results
medium exhaustion, and subsequent microglial death. Therefore, we Both Gram-positive and -negative bacterial stimuli enhance
elected to use heat-inactivated bacteria throughout the course of our studies MyD88 expression in primary microglia
to avoid these potentially confounding effects. All non-LPS reagents and
culture medium were verified to have endotoxin levels ⬍0.03 endotoxin The signal transduction pathways emanating from all of the TLRs
units/ml as determined by Limulus amebocyte lysate assay (Associates of identified to date, with the exception of TLR3, use the common
Cape Cod). In addition, a phosphate assay (Pierce) was performed on PGN adaptor protein MyD88 (28, 39, 40). Recently, we have shown that
stocks to ensure reagent purity since commercial PGN preparations have both S. aureus and its cell wall component PGN significantly in-
been reported to be contaminated with LTA and bacterial DNA (36),
whereas PGN does not contain phosphate residues. The phosphate content
creased TLR2 expression at both the mRNA and protein levels
of the PGN preparation used in this study was below the limit of detection, (18). In the present study, the effects of S. aureus and PGN as well
indicating that it was not contaminated with alternative microbial products. as the Gram-negative bacterial cell wall product LPS, on MyD88
6804 MyD88 IN MICROGLIA

protein expression in primary microglia was evaluated by Western


blotting. As shown in Fig. 1, all three bacterial stimuli were found
to enhance microglial MyD88 protein levels. As we had recently
reported, TLR2 protein expression was elevated in response to S.
aureus, PGN, and LPS in WT microglia (18, 38) (Fig. 2). Inter-
estingly, compared with WT microglia, constitutive TLR2 expres-
sion was extremely low and not induced in response to either S.
aureus or PGN treatment in MyD88 KO cells (Fig. 2). In contrast,
LPS stimulation was capable of inducing TLR2 expression in
MyD88 KO microglia (Fig. 2). Collectively, these results suggest
that MyD88 is pivotal for the induction of TLR2 protein expres-
sion in response to both S. aureus and PGN, which may serve to
potentate microglial responses to Gram-positive pathogens
through receptor up-regulation. In contrast, the induction of TLR2 FIGURE 2. LPS-induced TLR2 expression is regulated by a MyD88-
independent pathway. MyD88 KO and WT primary microglia were stim-
expression in response to LPS occurs via a MyD88-independent
ulated with either 107 heat-inactivated S. aureus, 10 ␮g/ml PGN, or 100
pathway possibly via alternative adaptor proteins such as TIR do- ng/ml LPS for 24 h, whereupon protein extracts from whole cell lysates (40
main-containing adaptor protein and TIR domain-containing adap- ␮g/sample) were evaluated for TLR2 expression by Western blotting as
tor inducing IFN-␤ (TRIF). described in Materials and Methods. Following analysis, blots were
stripped and reprobed with an Ab specific for ␤-actin to verify uniformity
MyD88 regulates LPS-induced iNOS expression in gel loading. Results are representative of three independent experiments.

Downloaded from http://www.jimmunol.org/ by guest on April 14, 2022


NO is a highly reactive free radical that performs a wide variety of
cellular functions in addition to serving as an effective cytotoxic
agent used by immune cells to inactivate pathogens (41). It is
produced by any one of three enzyme isoforms termed NO syn- aureus- or PGN-stimulated microglia suggesting that these bacte-
thases (NOS). Endothelial NOS and neuronal NOS occur consti- rial stimuli are poor inducers of iNOS in primary microglia (Fig.
tutively and mediate vasodilation and neurosignaling, respectively. 3B). The inability to detect iNOS protein expression in response to
iNOS was first demonstrated in monocytes but is now known to be S. aureus and PGN by Western blots may have resulted from lim-
expressed in a variety of cell types, including microglia (42). iNOS itations in the sensitivity of this technique. Therefore, we per-
expression is stimulated in glia by a variety of inflammatory cy- formed highly sensitive qRT-PCR analysis and were able to dem-
tokines and bacterial products including LPS. Although recent onstrate that both S. aureus and PGN were capable of inducing
studies have implicated TLR4 in mediating the LPS-dependent iNOS mRNA expression in WT microglia. However, compared
production of reactive oxygen species in microglia (43), the ques- with LPS, the induction levels were very low, ⬃20- to 30-fold less
tion of whether MyD88-independent pathways are also involved in S. aureus or PGN-stimulated cells compared with the former
and the role of MyD88 in mediating NO expression in response to (Fig. 3A). In addition, iNOS mRNA levels in response to both S.
Gram-positive bacterial stimuli has not yet been investigated. aureus and PGN were significantly attenuated in MyD88 KO mi-
MyD88-dependent signals, presumably delivered via TLR4 en- croglia compared with WT cells (Fig. 3A). Therefore, the reason(s)
gagement, were found to be pivotal for iNOS induction in LPS- that iNOS protein could not be detected in primary microglia in
stimulated microglia as reflected by a significant attenuation in response to S. aureus or PGN could be due to either sensitivity
iNOS levels in MyD88 KO microglia (Fig. 3B). However, the issues with Western blots and/or the fact that alterations in mRNA
residual iNOS expression detected in MyD88 KO cells indicated levels do not always necessarily extend to differences in protein
that MyD88-independent pathways are also involved. Interest- expression.
ingly, we could not detect iNOS protein expression in either S.
MyD88-dependent signals are pivotal for the induction of
proinflammatory mediator release from S. aureus- and PGN-
stimulated microglia
Recently, we reported that TLR2 plays an important role in mi-
croglial recognition of PGN, whereas responses to intact S. aureus
were primarily TLR2 independent because TLR2 ablation did not
completely inhibit proinflammatory mediator production from S.
aureus-stimulated microglia (18). Similar findings have also been
reported in macrophages; however, macrophage activation was
completely attenuated in response to S. aureus treatment in
MyD88 KO cells, suggesting an important role for alternative
TLRs (30, 31, 44). In the present study, an essential role for TLRs
in S. aureus-dependent microglial activation was demonstrated.
Specifically, TNF-␣ production was completely abolished in
FIGURE 1. MyD88 is induced in primary microglia in response to both MyD88 KO microglia treated with PGN and minimal cytokine
Gram-positive and -negative bacterial stimuli. MyD88 KO and WT pri- expression was detected in response to intact S. aureus only at the
mary microglia were stimulated with either 107 heat-inactivated S. aureus,
highest dose examined (Fig. 4, A and B). In contrast, microglial
10 ␮g/ml PGN, or 100 ng/ml LPS for 24 h, whereupon protein extracts
from whole cell lysates (40 ␮g/sample) were evaluated for MyD88 expres-
TNF-␣ production in response to LPS was found to be mediated by
sion by Western blotting as described in Materials and Methods. Following both MyD88-dependent and -independent pathways (Fig. 4C).
analysis, blots were stripped and reprobed with an Ab specific for ␤-actin Similar results were obtained for IL-1␤ (data not shown). Exam-
to verify uniformity in gel loading. Results are representative of three in- ination of chemokine expression in MyD88 KO and WT microglia
dependent experiments. revealed a similar requirement for MyD88 in response to S. aureus
The Journal of Immunology 6805

Downloaded from http://www.jimmunol.org/ by guest on April 14, 2022


FIGURE 3. Role of MyD88-dependent signals in regulating iNOS ex-
pression. A, MyD88 KO and WT microglia were seeded at 2 ⫻ 106 cells/
well in 6-well plates and incubated overnight. The following day, cells
FIGURE 4. MyD88 is essential in mediating microglial activation in
were stimulated with 107 heat-inactivated S. aureus, 10 ␮g/ml PGN, or 100
response to S. aureus and PGN. MyD88 KO and WT microglia were
ng/ml LPS for 6 or 24 h, whereupon total RNA was isolated and examined
seeded at 2 ⫻ 105 cells/well in 96-well plates and incubated overnight. The
for iNOS expression by qRT-PCR as described in Materials and Methods. following day, cells were exposed to various concentrations of heat-inac-
Gene expression levels were calculated after normalizing iNOS signals tivated S. aureus (A), PGN (B), or LPS (C) for 24 h, whereupon condi-
against GAPDH and are presented in relative mRNA expression units tioned supernatants were collected and analyzed for TNF-␣ protein expres-
(mean ⫹ SEM of three independent experiments). Significant differences
sion by ELISA. Results are presented as the amount of TNF-␣ (nanogram)
in iNOS mRNA expression between unstimulated (data not shown) vs
per milliliter of culture supernatant (mean ⫾ SD). Microglial cell viability
PAMP-treated microglia are denoted with asterisks (ⴱ, p ⬍ 0.05), whereas
was assessed using a standard MTT assay and the raw OD570 absorbance
significant differences between MyD88 KO vs WT microglia are denoted values are reported (D; mean ⫾ SD). Significant differences in TNF-␣
with a hatched sign (#, p ⬍ 0.05). B, MyD88 KO and WT primary mi-
expression between unstimulated vs PAMP-treated microglia are denoted
croglia were stimulated with either 107 heat-inactivated S. aureus, 10
with asterisks (ⴱ, p ⬍ 0.05; ⴱⴱ, p ⬍ 0.001), whereas significant differences
␮g/ml PGN, or 100 ng/ml LPS for 24 h, whereupon protein extracts from between MyD88 KO vs WT microglia are denoted with a hatched sign (##,
whole cell lysates (40 ␮g/sample) were evaluated for iNOS expression by p ⬍ 0.001). Results are representative of three independent experiments.
Western blotting as described in Materials and Methods. Following anal-
ysis, blots were stripped and reprobed with an Ab specific for ␤-actin to
verify uniformity in gel loading. Results are representative of three inde-
pendent experiments. way. Recently, several reports have demonstrated the presence of
numerous negative regulators of TLR signaling, most of which
target the MyD88-dependent signaling cascade (45, 46). It is en-
or PGN. Specifically, production of the potent neutrophil chemoat- visioned that the multimolecular complex formed by MyD88 fol-
tractant MIP-2/CXCL2 was completely attenuated in response to lowing TLR signaling is an important target for some negative
either S. aureus or PGN in MyD88 KO microglia compared with regulators and consequently affects the fine tuning of TLR re-
WT cells (Fig. 5A). In addition, MyD88 KO microglia produced sponses (45). Indeed, in the present study we found that S. aureus
significantly lower levels of MIP-2/CXCL2 in response to LPS stimulation was not capable of inducing IL-12 p40 production in
compared with WT cells (Fig. 5A). Similar findings were observed MyD88KO microglia, similar to what was observed with PGN
for the monocyte/lymphocyte chemoattractant MCP-1/CCL2 (data (Fig. 5B). IL-12 p40 production was also attenuated in MyD88 KO
not shown). Importantly, the concentrations of all bacterial stimuli microglia in response to LPS (Fig. 5B). Therefore, it appears as if
used in this study did not adversely affect microglial cell viability, S. aureus activation of microglia induces a MyD88-dependent neg-
indicating that the attenuated responses of MyD88 KO microglia ative regulatory signal(s) that acts to attenuate the expression of
were not due to overt cell death or random differences in cell- select proinflammatory mediators such as IL-12 p40.
seeding density (Fig. 4D). Biologically active IL-12 consists of two subunits, p35 and p40,
Recent studies from our laboratory have demonstrated that, un- the latter of which is promiscuous in its ability to interact with
like other proinflammatory cytokines examined, IL-12 p40 release other members of the IL-12 cytokine family (47). One such ex-
was significantly increased in both S. aureus-stimulated TLR2 and ample is IL-23, which is composed of a unique p19 subunit and the
CD14 KO microglia compared with WT cells (18, 38). Based on common p40 subunit shared with IL-12 (48, 49). Although IL-12
these results we had suggested that TLR2 signaling, potentially in and IL-23 share some overlapping activities in terms of their abil-
cooperation with CD14, regulates IL-12 p40 release by inducing ity to impact T cell activation, IL-23 is unique in its ability to
suppressor cytokines and/or activating a direct suppressor path- induce memory T cell proliferation (49, 50). Microglia have been
6806 MyD88 IN MICROGLIA

FIGURE 5. The loss of MyD88 results in diminished MIP-2 and IL-12


p40 expression. Primary microglia from MyD88 KO and WT mice were
seeded at 2 ⫻ 105 cells/well in 96-well plates and incubated overnight. The
following day, cells were exposed to either 107 heat-inactivated S. aureus,

Downloaded from http://www.jimmunol.org/ by guest on April 14, 2022


10 ␮g/ml PGN, or 100 ng/ml LPS for 24 h, whereupon conditioned su-
pernatants were collected and analyzed for MIP-2 (A) and IL-12 p40 (B)
protein expression by ELISA. Results are presented as the amount of cy-
tokine (nanograms) per milliliter of culture supernatant (mean ⫾ SD). Sig-
nificant differences in cytokine expression between unstimulated vs
PAMP-treated microglia are denoted with asterisks (ⴱ, p ⬍ 0.05; ⴱⴱ, p ⬍
0.001), whereas significant differences between MyD88 KO vs WT micro-
glia are denoted with a hatched sign (##, p ⬍ 0.001). Results are repre-
sentative of three independent experiments.

reported to express IL-23 in response to IFN-␥ and LPS stimula-


tion (51, 52); however, the role of MyD88-dependent signals in
regulating IL-23 levels has not yet been examined. To extend our
analysis of cytokines associated with adaptive immunity, we eval-
uated the expression of IL-23 p19 mRNA in response to intact S.
aureus, PGN, and LPS in MyD88 KO and WT microglia using
qRT-PCR. All three bacterial stimuli were capable of eliciting
IL-23 p19 expression in WT microglia; however, the induction of
IL-23 p19 in response to S. aureus did not reach statistical signif-
icance (Fig. 6). Although there was no induction of IL-23 p19
mRNA expression in MyD88 KO microglia in response to S. au-
reus, the differences between S. aureus-treated WT and KO cells
was statistically insignificant (Fig. 6A). Conversely, MyD88-de-
FIGURE 6. MyD88 is involved in IL-23 p19 mRNA expression.
pendent signals were found to be important for IL-23 p19 induc- MyD88 KO and WT microglia were seeded at 2 ⫻ 106 cells/well in 6-well
tion in response to both PGN and LPS (Fig. 6, B and C). However, plates and incubated overnight. The following day, cells were stimulated
LPS was still capable of significantly augmenting IL-23 p19 with either 107 heat-inactivated S. aureus (A), 10 ␮g/ml PGN (B), or 100
mRNA expression in MyD88 KO microglia, suggesting that this ng/ml LPS (C) for 6, 12, or 24 h, whereupon total RNA was isolated and
cytokine is also influenced by MyD88-independent pathway(s). examined for IL-23 p19 expression by qRT-PCR as described in Materials
Collectively, these data demonstrate that MyD88 is essential for and Methods. Gene expression levels were calculated after normalizing
signaling the production of proinflammatory mediators in response IL-23 p19 signals against GAPDH and are presented in relative mRNA
to both S. aureus and PGN and as such, implicates the involvement expression units (mean ⫾ SEM of three independent experiments). Sig-
of multiple TLRs in mediating maximal microglial activation in nificant differences in IL-23 p19 mRNA expression between unstimulated
vs PAMP-treated microglia are denoted with asterisks (ⴱ, p ⬍ 0.05; ⴱⴱ, p ⬍
response to these Gram-positive stimuli. In contrast, LPS-induced
0.001), whereas significant differences between MyD88 KO vs WT micro-
microglial activation occurs via both MyD88-dependent and -in- glia are denoted with a hatched sign (##, p ⬍ 0.001).
dependent pathways.

Role of MyD88 on the expression of alternative PRRs in general, engagement of phagocytic receptors does not directly lead
microglia to cytokine induction, these receptors are capable of influencing
Previous studies from our laboratory have revealed that both S. the activity of cytokine signaling receptors through receptor cross-
aureus and PGN modulate the expression of alternative PRRs in talk. Therefore, we investigated the potential involvement of
microglia, including the phagocytic receptors LOX-1 and PTX3 as MyD88-dependent pathways on the modulation of phagocytic
well as TLR9 (18, 38). Because LOX-1 and PTX3 are considered PRRs. The induction of microglial LOX-1 expression in response
phagocytic receptors and can directly bind pathogens (53–55), they to all bacterial stimuli was time dependent, with maximal effects
may be responsible for S. aureus uptake by microglia. Although in observed at 6-h poststimulation (Fig. 7). MyD88 was found to play
The Journal of Immunology 6807

Downloaded from http://www.jimmunol.org/ by guest on April 14, 2022


FIGURE 7. Microglial LOX-1 expression is partially regulated by FIGURE 8. The expression of PTX3 is regulated by MyD88 in primary
MyD88. MyD88 KO and WT microglia were seeded at 2 ⫻ 106 cells/well microglia. MyD88 KO and WT microglia were seeded at 2 ⫻ 106 cells/
in 6-well plates and incubated overnight. The following day, cells were well in 6-well plates and incubated overnight. The following day, cells
stimulated with either 107 heat-inactivated S. aureus (A), 10 ␮g/ml PGN were stimulated with either 107 heat-inactivated S. aureus (A), 10 ␮g/ml
(B), or 100 ng/ml LPS (C) for 6, 12, or 24 h, whereupon total RNA was PGN (B), or 100 ng/ml LPS (C) for 6, 12, or 24 h, whereupon total RNA
isolated and examined for LOX-1 expression by qRT-PCR as described in was isolated and examined for PTX3 expression by qRT-PCR as described
Materials and Methods. Gene expression levels were calculated after nor- in Materials and Methods. Gene expression levels were calculated after
malizing LOX-1 signals against GAPDH and are presented in relative normalizing PTX3 signals against GAPDH and are presented in relative
mRNA expression units (mean ⫾ SEM of three independent experiments). mRNA expression units (mean ⫾ SEM of three independent experiments).
Significant differences in LOX-1 mRNA expression between unstimulated Significant differences in PTX3 mRNA expression between unstimulated
vs PAMP-treated microglia are denoted with asterisks (ⴱ, p ⬍ 0.05; ⴱⴱ, p ⬍ vs PAMP-treated microglia are denoted with asterisks (ⴱ, p ⬍ 0.05; ⴱⴱ, p ⬍
0.001), whereas significant differences between MyD88 KO vs WT micro- 0.001), whereas significant differences between MyD88 KO vs WT micro-
glia are denoted with a hatched sign (#, p ⬍ 0.05; ##, p ⬍ 0.001). glia are denoted with a hatched sign (#, p ⬍ 0.05; ##, p ⬍ 0.001).

an important role in regulating microglial LOX-1 expression in


response to S. aureus and PGN as evident by a reduction in LOX-1 recognizes bacterial DNA which contains high levels of unmeth-
levels in MyD88 KO cells; however, MyD88 did not appear to ylated CpG motifs and this receptor uses a MyD88-dependent sig-
modulate microglial LOX-1 induction in response to LPS (Fig. 7). naling pathway. Similar to what we have recently reported in pri-
A similar requirement for MyD88-dependent signaling on the in- mary microglia (18), TLR9 expression was attenuated in response
duction of PTX3 expression was demonstrated by the failure of to all three microbial stimuli examined; however, the inhibition re-
MyD88 KO microglia to up-regulate PTX3 levels (Fig. 8). TLR9 sulting from S. aureus treatment did not reach statistical significance
6808 MyD88 IN MICROGLIA

(Fig. 9A). Interestingly, the level of PGN- and LPS-induced suppres- ber of the nucleotide-binding oligomerization domain family of
sion of TLR9 expression was significantly less in MyD88 KO micro- proteins and considered a PRR because it recognizes the bacterial
glia compared with WT cells (Fig. 9, B and C). product muramyl dipeptide (MDP), a degradation product of bac-
To expand our analysis of alternative PRRs in microglia, we terial PGN (56, 57). A recent study has proposed that NOD2 could
evaluated changes in the expression of NOD2 in response to bac- represent a negative regulator of TLR2-dependent signaling (58).
terial stimuli in MyD88 KO and WT microglia. NOD2 is a mem- However, to our knowledge the expression of NOD2 in microglia
has not yet been reported. All three microbial stimuli tested were
effective at augmenting microglial NOD2 expression (Fig. 10). In
addition, MyD88-dependent signal(s) were found to be important,
in part, for regulating NOD2 levels as demonstrated by the atten-
uation of NOD2 expression in MyD88 KO microglia compared
with WT cells (Fig. 10).
Collectively, these findings revealed that MyD88-dependent
signaling pathways regulate, to varying extents, the expression of
distinct PRRs including LOX-1, TLR9, PTX3, and NOD2. These
findings reveal the complexity by which PRR expression is regu-
lated in microglia. The establishment of an intricate set of signal-
ing paradigms to augment PRR expression would ensure that the
loss of a particular signaling pathway would not compromise mi-
croglial recognition of pathogens with potentially devastating ef-

Downloaded from http://www.jimmunol.org/ by guest on April 14, 2022


fects on CNS integrity.

Discussion
Bacterial infections of the CNS continue to occur, despite ad-
vances made in detection and therapy. In addition, the emergence
of multidrug-resistant strains of CNS pathogens, such as S. aureus,
has become a confounding factor that is magnified by the inability
of many antibiotics to reach high therapeutic levels in brain tissue.
Therefore, understanding the mechanisms by which CNS bacterial
pathogens are recognized by resident glia and infiltrating immune
cells are critical toward the development of new therapeutic mo-
dalities for infection. The role of MyD88, the central adaptor mol-
ecule essential for signaling of the TIR family of receptors, has
been examined in various systemic and CNS infections caused by
a variety Gram-positive organisms including Streptococcus pneu-
moniae and S. aureus (31, 59 – 61). However, the functional im-
portance of MyD88 in enabling the recognition of diverse bacterial
motifs by microglia has not yet been evaluated. Therefore, in the
present study, we examined the effects of S. aureus, a common
etiological agent of brain abscesses, and the bacterial cell wall
components PGN and LPS on the activation of MyD88 KO and
WT microglia. Our results revealed that MyD88 is pivotal for mi-
croglial activation and subsequent proinflammatory mediator re-
lease in response to all three bacterial stimuli (S. aureus, PGN,
and LPS).
A recent publication has raised a debate as to whether PGN
represents a true TLR2 ligand, a tenant that is supported by nearly
200 publications from various research groups. Specifically,
Travassos et al. (36) reported that PGN is not recognized by TLR2
but instead by the intracellular PRR NOD2 because purified PGN
was capable of activating NOD2-transfected, but not TLR2-trans-
FIGURE 9. The loss of MyD88 results in differential expression of fected, HEK293 cells. However, in a recent study, Dziarski et al.
TLR9 mRNA expression in response to S. aureus, PGN, or LPS. MyD88 (62) re-evaluated the effects of highly purified PGN on TLR2-
KO and WT microglia were seeded at 2 ⫻ 106 cells/well in 6-well plates transfected HEK293 cells and found that polymeric PGN is a po-
and incubated overnight. The following day, cells were stimulated with tent TLR2 ligand, whereas muramidase treatment dramatically re-
either 107 heat-inactivated S. aureus (A), 10 ␮g/ml PGN (B), or 100 ng/ml duced the responsiveness of cells to PGN. Therefore, the authors
LPS (C) for 6, 12, or 24 h, whereupon total RNA was isolated and exam- suggested that various purification procedures could damage the
ined for TLR9 expression by qRT-PCR as described in Materials and cross-linked TLR2-activating structure of PGN which is required
Methods. Gene expression levels were calculated after normalizing TLR9
for TLR2, but not NOD activation. Our previous and current find-
signals against GAPDH and are presented in relative mRNA expression
units (mean ⫾ SEM of three independent experiments). Significant differ-
ings support the premise that PGN represents a TLR2 agonist in
ences in TLR9 mRNA expression between unstimulated vs PAMP-treated microglia, because we demonstrated that neither TLR2 nor MyD88
microglia are denoted with asterisks (ⴱ, p ⬍ 0.05; ⴱⴱ, p ⬍ 0.001), whereas KO microglia were responsive to PGN (18). Importantly, the com-
significant differences between MyD88 KO vs WT microglia are denoted mercial PGN preparations used in this study were not found to
with a hatched sign (#, p ⬍ 0.05; ##, p ⬍ 0.001). contain detectable phosphate levels, indicating that any differences
The Journal of Immunology 6809

the activation of alternative receptors (18, 38). However, in the


present work we have reported that cytokine/chemokine produc-
tion in MyD88 KO microglia was nearly abolished in response to
intact S. aureus. This finding directly implicates the functional
involvement of additional TLRs due to the absence of this key
adaptor molecule. Intact S. aureus presents microglia with a com-
plex milieu of Ags including lipoproteins, PGN, and DNA-con-
taining nonmethylated CpG motifs that serve as agonists for TLR1
and TLR6 (lipoproteins and PGN) and TLR9 (CpG DNA) (2).
Both TLR1 and TLR6 form functional heterodimers with TLR2,
which dictate ligand specificity to various bacterial lipoproteins
(63– 65). In addition, upon phagocytosis, endosomal degradation
of pathogens leads to the exposure of bacterial DNA that can di-
rectly trigger activation signals via TLR9, which is an intracellular
PRR (3, 66 – 68). We found that the ability of MyD88 KO and WT
microglia to phagocytize S. aureus was equivalent (N. Esen and T.
Kielian, unpublished observations), indicating that the failure of
MyD88-deficient cells to produce proinflammatory mediators was
not due to impaired access to bacterial Ags. Evaluation of TLR9

Downloaded from http://www.jimmunol.org/ by guest on April 14, 2022


expression revealed that this TLR was regulated by both MyD88-
dependent and -independent pathways, which was dependent on
the type of stimulus used. Specifically, TLR9 levels were signifi-
cantly different in PGN- and LPS-stimulated MyD88 KO micro-
glia compared with WT cells, whereas TLR9 levels were equiva-
lent in S. aureus-activated MyD88 KO and WT microglia.
Recently, it has been reported that various forms of CpG DNA can
activate dendritic cells differentially via TLR9-dependent and -in-
dependent signals (68). Specifically, conventional CpG DNA ac-
tivated dendritic cells via a MyD88-dependent manner whereas a
distinct CpG DNA, where polyG stretches are located at the 5⬘ and
3⬘ ends of a central CpG motif, induced IFN-␣ through IFN reg-
ulatory factor-3 (IRF-3) which did not require MyD88. Therefore,
heterogeneity in the chemical composition of bacterial DNA may
lead to activation of microglia via MyD88-dependent and -inde-
pendent mechanisms; however, this possibility remains specula-
tive. Finally, although every effort was made to ensure that endo-
toxin contamination of non-LPS reagents was not a confounding
factor in our study, we cannot completely rule out the presence of
minor LPS contaminants that were below the detection limit of our
Limulus amebocyte lysate assay. Minor LPS contaminants may
explain the low levels of residual cytokine expression observed in
MyD88 KO microglia in response to S. aureus. Another possibility
is that intact S. aureus may stimulate additional, as of yet uniden-
FIGURE 10. NOD2 mRNA is expressed in primary microglia. MyD88
KO and WT microglia were seeded at 2 ⫻ 106 cells/well in 6-well plates tified, cytokine signaling PRRs that function independently of
and incubated overnight. The following day, cells were stimulated with MyD88. It should be noted that there was no residual cytokine
either 107 heat-inactivated S. aureus (A), 10 ␮g/ml PGN (B), or 100 ng/ml expression in PGN-treated MyD88 KO microglia, suggesting that
LPS (C) for 6, 12, or 24 h, whereupon total RNA was isolated and exam- this reagent did not contain trace amounts of LPS that could func-
ined for NOD2 expression by qRT-PCR as described in Materials and tion via a MyD88-independent manner.
Methods. Gene expression levels were calculated after normalizing NOD2 Unlike proinflammatory cytokine production, which was nearly
signals against GADPH and are presented in relative mRNA expression completely attenuated in MyD88 KO microglia in response to both
units (mean ⫾ SEM of three independent experiments). Significant differ-
S. aureus and PGN, the expression of alternative PRRs was not as
ences in NOD2 mRNA expression between unstimulated vs PAMP-treated
microglia are denoted with asterisks (ⴱ, p ⬍ 0.05; ⴱⴱ, p ⬍ 0.001), whereas
affected by the loss of this TLR adaptor protein. For example,
significant differences between MyD88 KO vs WT microglia are denoted LOX-1 which has been reported to serve as a phagocytic receptor
with a hatched sign (#, p ⬍ 0.05). for S. aureus (55), was significantly increased in both MyD88 KO
and WT microglia. In addition, the repeating carbohydrate moi-
eties of PGN also make it a potential target for lectin-like mole-
observed between MyD88 KO and WT microglia throughout this cules such as LOX-1 (69). Although it was evident, to a limited
study cannot be explained by effects of contaminating LTA or extent, that MyD88-dependent pathways were involved in regu-
DNA, which may be present in various commercial PGN prepa- lating LOX-1 induction following PGN stimulation, in the major-
rations (36). ity of treatment conditions, microglial LOX-1 expression was
In our previous studies, we demonstrated that microglial proin- equivalent in MyD88 KO and WT cells. These findings are similar
flammatory mediator production was not attenuated in either TLR2 to what we have previously reported in TLR2 KO microglia (18).
or CD14 KO microglia following S. aureus stimulation, suggesting The role of LOX-1 in microglial phagocytosis has not yet been
6810 MyD88 IN MICROGLIA

examined; however, it is possible that LOX-1 is involved in bac- to impair NF-␬B activation, IRF-3 has been implicated as a me-
terial uptake and subsequent processing, providing ligands to en- diator of TNF-␣ activation via a MyD88-independent pathway.
gage TLR9 to augment antibacterial immunity. Additional studies In conclusion, our results indicate that microglial proinflamma-
examining the functional significance of LOX-1 in microglial re- tory mediator release induced by the Gram-positive stimuli S. au-
sponses to bacteria are needed to assess its role in phagocytosis. reus and PGN occurs via a MyD88-dependent pathway and in-
We also examined the role of MyD88 in regulating the expression volves the activity of multiple TLRs. In addition, the expression of
of the soluble PRR PTX3 in microglia. PTX3 expression was sig- additional phagocytic PRRs, including LOX-1 and PTX3, is reg-
nificantly enhanced following stimulation with either, S. aureus, ulated, in part, by MyD88-dependent signals. In contrast, LPS-
PGN, or LPS in WT cells; however, PTX3 levels in MyD88 KO dependent microglial activation is regulated by both MyD88-de-
microglia were similar to baseline, indicating that MyD88-depen- pendent and -independent mechanisms. Moreover, we report the
dent signal(s) are critical for inducing the expression of this sol- novel expression of the intracellular PRR, NOD2, in microglia and
uble PRR. This finding is in agreement with our previous report find that this receptor is augmented in response to Gram-positive
demonstrating that CD14-dependent signals, likely in cooperation bacteria and its cell wall product PGN. We are currently extending
with TLR2, are essential for maximal PTX3 expression in PGN- these studies to examine the role of MyD88 in S. aureus-dependent
activated microglia (18, 38). microglial activation in vivo using the experimental brain abscess
NOD2 is an intracellular PRR that recognizes MDP moieties model developed in our laboratory, where microglial responses
that are released as a result of hydrolytic or lysosomal digestion of will be quantified in FACS-purified populations from MyD88 WT
PGN (56, 57). The interaction of NOD2 with MDP leads to NF-␬B and KO mice. Collectively, these results reveal the importance of
activation and subsequent proinflammatory gene expression (70). MyD88, and by extension, multiple TLRs, in mediating microglial
One interesting finding that surfaced during the course of these activation in response to a wide array of PAMPs. However, it is

Downloaded from http://www.jimmunol.org/ by guest on April 14, 2022


studies was the ability of all three bacterial stimuli examined to apparent that alternative PRRs also play a role in enabling maxi-
induce NOD2 expression in MyD88 KO and WT microglia. To our mal microglial responses to pathogens, which is not unexpected
knowledge, this is the first report demonstrating the expression of because bacteria such as S. aureus have the potential to elicit dev-
NOD2 in primary microglia. We also observed that NOD2 levels astating consequences in a tissue that has limited regenerative ca-
were attenuated in response to both S. aureus and PGN, but not pacity such as the CNS.
LPS, in MyD88 KO microglia, suggesting a role for MyD88-de-
pendent signals in regulating the expression of this intracellular Acknowledgments
PRR. It is intriguing that NOD2 expression was found to be in- We thank Patrick Mayes for excellent technical assistance.
fluenced, in part, by MyD88 in response to stimuli that harbor
NOD2 ligands upon intercellular processing (i.e., PGN from the Disclosures
cell wall of intact S. aureus and PGN itself). The functional im- The authors have no financial conflict of interest.
portance of NOD2 in mediating microglial activation and its role
in the context of the myriad of PRRs expressed by microglia re-
References
1. Aloisi, F. 2001. Immune function of microglia. Glia 36: 165–179.
main to be determined. 2. Kielian, T. 2006. Toll-like receptors (TLR) in central nervous system glial in-
LPS is a well-established TLR4 ligand and activates macro- flammation and homeostasis. J. Neurosci. Res. 83: 711–730.
3. Kaisho, T., and S. Akira. 2004. Pleiotropic function of Toll-like receptors. Mi-
phages and dendritic cells via both MyD88-dependent and -inde- crobes Infect. 6: 1388 –1394.
pendent pathways (3, 71, 72). Specifically, the induction of clas- 4. Kopp, E., and R. Medzhitov. 2003. Recognition of microbial infection by Toll-
sical proinflammatory cytokines (i.e., TNF-␣, IL-1␤) by LPS is like receptors. Curr. Opin. Immunol. 15: 396 – 401.
5. Takeda, K., and S. Akira. 2004. TLR signaling pathways. Semin. Immunol.
mediated via MyD88-dependent signals, whereas IFN-inducible 16: 3–9.
genes such as iNOS, IFN-inducible protein 10, and IFN␣␤ are 6. Mathisen, G. E., and J. P. Johnson. 1997. Brain abscess. Clin. Infect. Dis. 25:
regulated via MyD88-independent pathways (29, 73). These IFN- 763–779; 780 –761.
7. Townsend, G. C., and W. M. Scheld. 1998. Infections of the central nervous
inducible genes are controlled by the activation of two adaptor system. Adv. Intern. Med. 43: 403– 447.
proteins, TIR domain-containing adaptor protein and TRIF, that 8. Koedel, U., W. M. Scheld, and H. W. Pfister. 2002. Pathogenesis and pathophys-
iology of pneumococcal meningitis. Lancet Infect. Dis. 2: 721–736.
act downstream to activate IRF-3 through Tank-binding kinase 1 9. Scheld, W. M., U. Koedel, B. Nathan, and H. W. Pfister. 2002. Pathophysiology
and I␬B kinase (74, 75). One interesting observation in the current of bacterial meningitis: mechanism(s) of neuronal injury. J. Infect. Dis.
study was the finding that LPS-induced proinflammatory cytokine 186(Suppl. 2): S225–S233.
10. Baldwin, A. C., and T. Kielian. 2004. Persistent immune activation associated
expression was still evident in MyD88 KO microglia and although with a mouse model of Staphylococcus aureus-induced experimental brain ab-
levels were significantly less compared with WT cells residual scess. J. Neuroimmunol. 151: 24 –32.
cytokine expression was still significantly elevated compared with 11. Kielian, T. 2004. Immunopathogenesis of brain abscess. J. Neuroinflammation
1: 16.
unstimulated microglia. This finding differs from a study by Kawai 12. Bsibsi, M., R. Ravid, D. Gveric, and J. M. van Noort. 2002. Broad expression of
et al. (29) where LPS-induced proinflammatory cytokine expres- Toll-like receptors in the human central nervous system. J. Neuropathol. Exp.
Neurol. 61: 1013–1021.
sion was abolished in MyD88 KO macrophages. The reason(s) 13. Kielian, T., P. Mayes, and M. Kielian. 2002. Characterization of microglial re-
responsible for these discrepancies are not known; however, our sponses to Staphylococcus aureus: effects on cytokine, costimulatory molecule,
results suggest that proinflammatory cytokine production in mi- and Toll-like receptor expression. J. Neuroimmunol. 130: 86 –99.
14. Laflamme, N., G. Soucy, and S. Rivest. 2001. Circulating cell wall components
croglia may be influenced by MyD88-independent signals, a find- derived from Gram-negative, not Gram-positive, bacteria cause a profound in-
ing which may be cell type specific. A recent study provides ad- duction of the gene-encoding Toll-like receptor 2 in the CNS. J. Neurochem. 79:
ditional evidence to support our findings where LPS-stimulation 648 – 657.
15. Laflamme, N., H. Echchannaoui, R. Landmann, and S. Rivest. 2003. Cooperation
was found to induce several chemokine and cytokine genes such as between Toll-like receptor 2 and 4 in the brain of mice challenged with cell wall
MIP-1␣, MIP-1␤, MIP-2, and TNF-␣ in MyD88-deficient cells as components derived from Gram-negative and Gram-positive bacteria. Eur. J. Im-
munol. 33: 1127–1138.
evaluated by microarray analysis (76). In addition, this study dem- 16. Olson, J. K., and S. D. Miller. 2004. Microglia initiate central nervous system
onstrated that the TRIF-dependent pathway activated TNF-␣ pro- innate and adaptive immune responses through multiple TLRs. J. Immunol. 173:
duction and secretion via a NF-␬B-independent manner and sug- 3916 –3924.
17. Rasley, A., J. Anguita, and I. Marriott. 2002. Borrelia burgdorferi induces inflam-
gested that secreted TNF-␣ acts in an autocrine manner to induce matory mediator production by murine microglia. J. Neuroimmunol. 130: 22–31.
delayed NF-␬B activation. Because depletion of IRF-3 was found 18. Kielian, T., N. Esen, and E. D. Bearden. 2005. Toll-like receptor 2 (TLR2) is
The Journal of Immunology 6811

pivotal for recognition of S. aureus peptidoglycan but not intact bacteria by 49. Oppmann, B., R. Lesley, B. Blom, J. C. Timans, Y. Xu, B. Hunte, F. Vega,
microglia. Glia 49: 567–576. N. Yu, J. Wang, K. Singh, et al. 2000. Novel p19 protein engages IL-12p40 to
19. Ebert, S., J. Gerber, S. Bader, F. Muhlhauser, K. Brechtel, T. J. Mitchell, and form a cytokine, IL-23, with biological activities similar as well as distinct from
R. Nau. 2005. Dose-dependent activation of microglial cells by Toll-like receptor IL-12. Immunity 13: 715–725.
agonists alone and in combination. J. Neuroimmunol. 159: 87–96. 50. Lankford, C. S., and D. M. Frucht. 2003. A unique role for IL-23 in promoting
20. Chien, H. F., K. Y. Yeh, Y. F. Jiang-Shieh, I. H. Wei, C. Y. Chang, M. L. Chang, cellular immunity. J. Leukocyte Biol. 73: 49 –56.
and C. H. Wu. 2005. Signal transduction pathways of nitric oxide release in 51. Li, J., B. Gran, G. X. Zhang, E. S. Ventura, I. Siglienti, A. Rostami, and
primary microglial culture challenged with Gram-positive bacterial constituent, M. Kamoun. 2003. Differential expression and regulation of IL-23 and IL-12
lipoteichoic acid. Neuroscience 133: 423– 436. subunits and receptors in adult mouse microglia. J. Neurol. Sci. 215: 95–103.
21. Jung, D. Y., H. Lee, B. Y. Jung, J. Ock, M. S. Lee, W. H. Lee, and K. Suk. 2005. 52. Sonobe, Y., I. Yawata, J. Kawanokuchi, H. Takeuchi, T. Mizuno, and
TLR4, but not TLR2, signals autoregulatory apoptosis of cultured microglia: a A. Suzumura. 2005. Production of IL-27 and other IL-12 family cytokines by
critical role of IFN-␤ as a decision maker. J. Immunol. 174: 6467– 6476. microglia and their subpopulations. Brain Res. 1040: 202–207.
22. Lehnardt, S., L. Massillon, P. Follett, F. E. Jensen, R. Ratan, P. A. Rosenberg, 53. Garlanda, C., E. Hirsch, S. Bozza, A. Salustri, M. De Acetis, R. Nota, A. Maccagno,
J. J. Volpe, and T. Vartanian. 2003. Activation of innate immunity in the CNS F. Riva, B. Bottazzi, G. Peri, et al. 2002. Non-redundant role of the long pentraxin
triggers neurodegeneration through a Toll-like receptor 4-dependent pathway. PTX3 in anti-fungal innate immune response. Nature 420: 182–186.
Proc. Natl. Acad. Sci. USA 100: 8514 – 8519. 54. Mantovani, A., C. Garlanda, and B. Bottazzi. 2003. Pentraxin 3, a non-redundant
23. Lehnardt, S., C. Lachance, S. Patrizi, S. Lefebvre, P. L. Follett, F. E. Jensen, soluble pattern recognition receptor involved in innate immunity. Vaccine
P. A. Rosenberg, J. J. Volpe, and T. Vartanian. 2002. The Toll-like receptor 21(Suppl. 2): S43–S47.
TLR4 is necessary for lipopolysaccharide-induced oligodendrocyte injury in the
55. Shimaoka, T., N. Kume, M. Minami, K. Hayashida, T. Sawamura, T. Kita, and
CNS. J. Neurosci. 22: 2478 –2486.
S. Yonehara. 2001. LOX-1 supports adhesion of Gram-positive and Gram-neg-
24. Akira, S. 2003. Toll-like receptor signaling. J. Biol. Chem. 278: 38105–38108.
ative bacteria. J. Immunol. 166: 5108 –5114.
25. Akira, S., and S. Sato. 2003. Toll-like receptors and their signaling mechanisms.
56. Netea, M. G., G. Ferwerda, D. J. de Jong, T. Jansen, L. Jacobs, M. Kramer,
Scand. J. Infect. Dis. 35: 555–562.
T. H. Naber, J. P. Drenth, S. E. Girardin, B. J. Kullberg, et al. 2005. Nucleotide-
26. Adachi, O., T. Kawai, K. Takeda, M. Matsumoto, H. Tsutsui, M. Sakagami,
binding oligomerization domain-2 modulates specific TLR pathways for the in-
K. Nakanishi, and S. Akira. 1998. Targeted disruption of the MyD88 gene results
duction of cytokine release. J. Immunol. 174: 6518 – 6523.
in loss of IL-1- and IL-18-mediated function. Immunity 9: 143–150.
27. Hanada, T., and A. Yoshimura. 2002. Regulation of cytokine signaling and in- 57. Girardin, S. E., I. G. Boneca, J. Viala, M. Chamaillard, A. Labigne, G. Thomas,

Downloaded from http://www.jimmunol.org/ by guest on April 14, 2022


flammation. Cytokine Growth Factor Rev. 13: 413– 421. D. J. Philpott, and P. J. Sansonetti. 2003. Nod2 is a general sensor of peptidoglycan
28. Medzhitov, R., P. Preston-Hurlburt, E. Kopp, A. Stadlen, C. Chen, S. Ghosh, and through muramyl dipeptide (MDP) detection. J. Biol. Chem. 278: 8869 – 8872.
C. A. Janeway, Jr. 1998. MyD88 is an adaptor protein in the hToll/IL-1 receptor 58. Watanabe, T., A. Kitani, P. J. Murray, and W. Strober. 2004. NOD2 is a negative
family signaling pathways. Mol. Cell 2: 253–258. regulator of Toll-like receptor 2-mediated T helper type 1 responses. Nat. Im-
29. Kawai, T., O. Adachi, T. Ogawa, K. Takeda, and S. Akira. 1999. Unresponsive- munol. 5: 800 – 808.
ness of MyD88-deficient mice to endotoxin. Immunity 11: 115–122. 59. Sugawara, I., H. Yamada, S. Mizuno, K. Takeda, and S. Akira. 2003. Mycobac-
30. Takeuchi, O., K. Takeda, K. Hoshino, O. Adachi, T. Ogawa, and S. Akira. 2000. terial infection in MyD88-deficient mice. Microbiol. Immunol. 47: 841– 847.
Cellular responses to bacterial cell wall components are mediated through 60. Koedel, U., T. Rupprecht, B. Angele, J. Heesemann, H. Wagner, H. W. Pfister,
MyD88-dependent signaling cascades. Int. Immunol. 12: 113–117. and C. J. Kirschning. 2004. MyD88 is required for mounting a robust host im-
31. Takeuchi, O., K. Hoshino, and S. Akira. 2000. Cutting edge: TLR2-deficient and mune response to Streptococcus pneumoniae in the CNS. Brain 127: 1437–1445.
MyD88-deficient mice are highly susceptible to Staphylococcus aureus infection. 61. Henneke, P., O. Takeuchi, R. Malley, E. Lien, R. R. Ingalls, M. W. Freeman,
J. Immunol. 165: 5392–5396. T. Mayadas, V. Nizet, S. Akira, D. L. Kasper, and D. T. Golenbock. 2002.
32. Underhill, D. M., A. Ozinsky, K. D. Smith, and A. Aderem. 1999. Toll-like Cellular activation, phagocytosis, and bactericidal activity against group B Strep-
receptor-2 mediates mycobacteria-induced proinflammatory signaling in macro- tococcus involve parallel myeloid differentiation factor 88-dependent and inde-
phages. Proc. Natl. Acad. Sci. USA 96: 14459 –14463. pendent signaling pathways. J. Immunol. 169: 3970 –3977.
33. Underhill, D., A. Ozinsky, A. M. Hajjar, A. Stevens, C. B. Wilson, M. Bassetti, 62. Dziarski, R., and D. Gupta. 2005. Staphylococcus aureus peptidoglycan is a Toll-
and A. Aderem. 1999. The Toll-like receptor 2 is recruited to macrophage phago- like receptor 2 activator: a reevaluation. Infect. Immun. 73: 5212–5216.
somes and discriminates between pathogens. Nature 401: 811– 815. 63. Takeuchi, O., S. Sato, T. Horiuchi, K. Hoshino, K. Takeda, Z. Dong,
34. Fremond, C. M., V. Yeremeev, D. M. Nicolle, M. Jacobs, V. F. Quesniaux, and R. L. Modlin, and S. Akira. 2002. Cutting edge: role of Toll-like receptor 1 in
B. Ryffel. 2004. Fatal Mycobacterium tuberculosis infection despite adaptive mediating immune response to microbial lipoproteins. J. Immunol. 169: 10 –14.
immune response in the absence of MyD88. J. Clin. Invest. 114: 1790 –1799. 64. Takeuchi, O., T. Kawai, P. F. Muhlradt, M. Morr, J. D. Radolf, A. Zychlinsky,
35. Kielian, T., M. McMahon, E. D. Bearden, A. C. Baldwin, P. D. Drew, and K. Takeda, and S. Akira. 2001. Discrimination of bacterial lipoproteins by Toll-
N. Esen. 2004. S. aureus-dependent microglial activation is selectively attenuated like receptor 6. Int. Immunol. 13: 933–940.
by the cyclopentenone prostaglandin 15-deoxy-␦12,14-prostaglandin J2 (15d- 65. Omueti, K. O., J. M. Beyer, C. M. Johnson, E. A. Lyle, and R. I. Tapping. 2005.
PGJ2). J. Neurochem. 90: 1163–1172. Domain exchange between human Toll-like receptors 1 and 6 reveals a region
36. Travassos, L. H., S. E. Girardin, D. J. Philpott, D. Blanot, M. A. Nahori, C. Werts, required for lipopeptide discrimination. J. Biol. Chem. 280: 36616 –36625.
and I. G. Boneca. 2004. Toll-like receptor 2-dependent bacterial sensing does not 66. Akira, S., and H. Hemmi. 2003. Recognition of pathogen-associated molecular
occur via peptidoglycan recognition. EMBO Rep. 5: 1000 –1006. patterns by TLR family. Immunol. Lett. 85: 85–95.
37. Kielian, T., B. Barry, and W. F. Hickey. 2001. CXC chemokine receptor-2 li- 67. Hemmi, H., O. Takeuchi, T. Kawai, T. Kaisho, S. Sato, H. Sanjo, M. Matsumoto,
gands are required for neutrophil-mediated host defense in experimental brain K. Hoshino, H. Wagner, K. Takeda, and S. Akira. 2000. A Toll-like receptor
abscesses. J. Immunol. 166: 4634 – 4643. recognizes bacterial DNA. Nature 408: 740 –745.
38. Esen, N., and T. Kielian. 2005. Recognition of Staphylococcus aureus-derived 68. Hemmi, H., T. Kaisho, K. Takeda, and S. Akira. 2003. The roles of Toll-like
peptidoglycan (PGN) but not intact bacteria is mediated by CD14 in microglia. receptor 9, MyD88, and DNA-dependent protein kinase catalytic subunit in the
J. Neuroimmunol. 170: 93–104. effects of two distinct CpG DNAs on dendritic cell subsets. J. Immunol. 170:
39. Wesche, H., W. J. Henzel, W. Shillinglaw, S. Li, and Z. Cao. 1997. MyD88: an 3059 –3064.
adapter that recruits IRAK to the IL-1 receptor complex. Immunity 7: 837– 847.
69. Gordon, S. 2002. Pattern recognition receptors: doubling up for the innate im-
40. Burns, K., F. Martinon, C. Esslinger, H. Pahl, P. Schneider, J. L. Bodmer,
mune response. Cell 111: 927–930.
F. Di Marco, L. French, and J. Tschopp. 1998. MyD88, an adapter protein in-
70. Inohara, N., M. Chamaillard, C. McDonald, and G. Nunez. 2004. NOD-LRR
volved in interleukin-1 signaling. J. Biol. Chem. 273: 12203–12209.
proteins: role in host-microbial interactions and inflammatory disease. Annu. Rev.
41. MacMicking, J., Q. W. Xie, and C. Nathan. 1997. Nitric oxide and macrophage
Biochem. 74: 355– 83
function. Annu. Rev. Immunol. 15: 323–350.
42. Lee, S. C., and C. F. Brosnan. 1996. Cytokine regulation of iNOS expression in 71. Akira, S., M. Yamamoto, and K. Takeda. 2003. Role of adapters in Toll-like
human glial cells. Methods 10: 31–37. receptor signalling. Biochem. Soc. Trans. 31: 637– 642.
43. Qin, L., G. Li, X. Qian, Y. Liu, X. Wu, B. Liu, J. S. Hong, and M. L. Block. 2005. 72. Takeda, K., T. Kaisho, and S. Akira. 2003. Toll-like receptors. Annu. Rev. Im-
Interactive role of the Toll-like receptor 4 and reactive oxygen species in LPS- munol. 21: 335–376.
induced microglia activation. Glia 52: 78 – 84. 73. Lee, J. Y., C. A. Lowell, D. G. Lemay, H. S. Youn, S. H. Rhee, K. H. Sohn,
44. Takeuchi, O., K. Hoshino, T. Kawai, H. Sanjo, H. Takada, T. Ogawa, K. Takeda, and B. Jang, J. Ye, J. H. Chung, and D. H. Hwang. 2005. The regulation of the
S. Akira. 1999. Differential roles of TLR2 and TLR4 in recognition of Gram-negative expression of inducible nitric oxide synthase by Src-family tyrosine kinases me-
and Gram-positive bacterial cell wall components. Immunity 11: 443– 451. diated through MyD88-independent signaling pathways of Toll-like receptor 4.
45. Liew, F. Y., D. Xu, E. K. Brint, and L. A. O’Neill. 2005. Negative regulation of Biochem. Pharmacol. 70: 1231–1240.
Toll-like receptor-mediated immune responses. Nat. Rev. Immunol. 5: 446 – 458. 74. Akira, S., and K. Hoshino. 2003. Myeloid differentiation factor 88-dependent and
46. Negishi, H., Y. Ohba, H. Yanai, A. Takaoka, K. Honma, K. Yui, T. Matsuyama, -independent pathways in toll-like receptor signaling. J. Infect. Dis. 187(Suppl.
T. Taniguchi, and K. Honda. 2005. Negative regulation of Toll-like receptor 2): S356 –S363.
signaling by IRF-4. Proc. Natl. Acad. Sci. USA 102: 15989 –15994. 75. Kawai, T., O. Takeuchi, T. Fujita, J. Inoue, P. F. Muhlradt, S. Sato, K. Hoshino,
47. Langrish, C. L., B. S. McKenzie, N. J. Wilson, R. de Waal Malefyt, and S. Akira. 2001. Lipopolysaccharide stimulates the MyD88-independent path-
R. A. Kastelein, and D. J. Cua. 2004. IL-12 and IL-23: master regulators of innate way and results in activation of IFN-regulatory factor 3 and the expression of a
and adaptive immunity. Immunol. Rev. 202: 96 –105. subset of lipopolysaccharide-inducible genes. J. Immunol. 167: 5887–5894.
48. Watford, W. T., M. Moriguchi, A. Morinobu, and J. J. O’Shea. 2003. The biology 76. Covert, M. W., T. H. Leung, J. E. Gaston, and D. Baltimore. 2005. Achieving
of IL-12: coordinating innate and adaptive immune responses. Cytokine Growth stability of lipopolysaccharide-induced NF-␬B activation. Science 309:
Factor Rev. 14: 361–368. 1854 –1857.

You might also like