+ All Categories
Home > Documents > Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I...

Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I...

Date post: 23-Dec-2016
Category:
Upload: maziar
View: 213 times
Download: 1 times
Share this document with a friend
15
Immunity Article Targeted Prostaglandin E 2 Inhibition Enhances Antiviral Immunity through Induction of Type I Interferon and Apoptosis in Macrophages Franc ¸ ois Coulombe, 1 Joanna Jaworska, 1 Mark Verway, 1 Fanny Tzelepis, 1 Amir Massoud, 1 Joshua Gillard, 1 Gary Wong, 2 Gary Kobinger, 2 Zhou Xing, 3 Christian Couture, 4 Philippe Joubert, 4 Jo ¨ rg H. Fritz, 5 William S. Powell, 1 and Maziar Divangahi 1, * 1 Department of Medicine, Department of Microbiology & Immunology, Department of Pathology, McGill International TB Centre, McGill University Health Centre and Research Institute, Meakins-Christie Laboratories, 3626 St. Urbain Street, Montreal, Quebec H2X 2P2, Canada 2 Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, MB R3E 3R2, Canada 3 McMaster Immunology Research Centre and Department of Pathology and Molecular Medicine, McMaster University, 1200 Main Street West, Hamilton, Ontario L8N 3Z5, Canada 4 Department of Pathology, Centre Hospitalier Universitaire de Que ´ bec, Ho ˆ tel-Dieu de Que ´ bec, 11 co ˆ te du Palais, Quebec, Quebec G1R 2J6, Canada 5 Department of Microbiology & Immunology, McGill Life Sciences Complex, Complex Traits Group, Bellini Pavilion, 3649 Promenade Sir William Osler, Montreal, Quebec H3G 0B1, Canada *Correspondence: [email protected] http://dx.doi.org/10.1016/j.immuni.2014.02.013 SUMMARY Aspirin gained tremendous popularity during the 1918 Spanish Influenza virus pandemic, 50 years prior to the demonstration of their inhibitory action on prostaglandins. Here, we show that during influenza A virus (IAV) infection, prostaglandin E 2 (PGE 2 ) was upregulated, which led to the inhibition of type I interferon (IFN) production and apoptosis in macrophages, thereby causing an increase in virus replication. This inhibitory role of PGE 2 was not limited to innate immunity, because both antigen presentation and T cell mediated immunity were also suppressed. Targeted PGE 2 suppression via genetic ablation of microsomal prostaglandin E-synthase 1 (mPGES-1) or by the pharmacological inhibition of PGE 2 receptors EP2 and EP4 substantially improved survival against lethal IAV infection whereas PGE 2 administration reversed this phenotype. These data demonstrate that the mPGES-1-PGE 2 pathway is targeted by IAV to evade host type I IFN-dependent antiviral immunity. We propose that specific inhibi- tion of PGE 2 signaling might serve as a treatment for IAV. INTRODUCTION The lung innate immune system has a critical role in limiting respiratory viral infections (Braciale et al., 2012). Alveolar macro- phages (M4) are one of the first immune cells to encounter pul- monary viruses and are indispensable for several key antiviral mechanisms including: (1) virus recognition and initiation of cytokine and chemokine production, (2) instruction of adaptive immunity, and (3) control of lung injury and resolution of inflam- mation (Kim et al., 2008; Shirey et al., 2010). Initially, upon sensing viral pathogen-associated molecular patterns (PAMPs), macrophages induce a cascade of cytokine responses including type I interferon a (IFN-a) and IFN-b production. Type I IFNs signal through the IFN-a receptor (IFNAR) and induce the upre- gulation of numerous genes that activate both innate and adap- tive immunity for rapidly controlling viral replication (Koerner et al., 2007). In addition, infected macrophages might undergo apoptosis to restrict viral dissemination (Mok et al., 2007) and to enhance T cell-mediated immunity via antigen (Ag) cross-pre- sentation (Albert et al., 1998). Recent evidence also suggests that during infection, macrophages have the capacity to trans- port Ags from the lung to the draining lymph nodes and poten- tially contribute to the induction of adaptive immune responses (Kirby et al., 2009). Although cytokines have been extensively studied in the context of viral infection, little is known about the function of the prostanoid (which includes prostaglandins [PG]) in antiviral im- munity (McCarthy and Weinberg, 2012). Among PGs, PGE 2 is the best-characterized immune modulatory lipid, acting on four subtypes of G protein-coupled E prostanoid receptors (EP1- EP4) to exert both pro- and anti-inflammatory responses de- pending on the source of inflammation (Ricciotti and FitzGerald, 2011). Biologically active PGE 2 originates from PGH 2 , which itself is the product of cyclooxygenase-1 (COX-1) and COX-2-medi- ated enzymatic conversion of membrane phospholipid-derived arachidonic acid (AA) (Figure 1A). Whereas three enzymes have been shown to independently generate PGE 2 from PGH 2 (Smyth et al., 2009), microsomal prostaglandin E-synthase 1 (mPGES-1) is the dominant isoform for PGE 2 synthesis in different mouse models of inflammation especially involving macrophages (Hara et al., 2010). Aspirin, a COX inhibitor, gained popularity during the 1918 Spanish Influenza A virus (IAV) pandemic (Starko, 2009) and has since then been used for the manage- ment of IAV symptoms. However, the loss-of-function in COX Immunity 40, 1–15, April 17, 2014 ª2014 Elsevier Inc. 1 Please cite this article in press as: Coulombe et al., Targeted Prostaglandin E 2 Inhibition Enhances Antiviral Immunity through Induction of Type I Inter- feron and Apoptosis in Macrophages, Immunity (2014), http://dx.doi.org/10.1016/j.immuni.2014.02.013
Transcript
Page 1: Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Interferon and Apoptosis in Macrophages

Please cite this article in press as: Coulombe et al., Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Inter-feron and Apoptosis in Macrophages, Immunity (2014), http://dx.doi.org/10.1016/j.immuni.2014.02.013

Immunity

Article

Targeted Prostaglandin E2 InhibitionEnhances Antiviral Immunity through Inductionof Type I Interferon and Apoptosis in MacrophagesFrancois Coulombe,1 Joanna Jaworska,1 Mark Verway,1 Fanny Tzelepis,1 Amir Massoud,1 Joshua Gillard,1 Gary Wong,2

Gary Kobinger,2 Zhou Xing,3 Christian Couture,4 Philippe Joubert,4 Jorg H. Fritz,5 William S. Powell,1

and Maziar Divangahi1,*1Department of Medicine, Department of Microbiology & Immunology, Department of Pathology, McGill International TB Centre, McGillUniversity Health Centre and Research Institute, Meakins-Christie Laboratories, 3626 St. Urbain Street, Montreal, Quebec H2X 2P2, Canada2Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg,

MB R3E 3R2, Canada3McMaster Immunology Research Centre and Department of Pathology and Molecular Medicine, McMaster University,1200 Main Street West, Hamilton, Ontario L8N 3Z5, Canada4Department of Pathology, Centre Hospitalier Universitaire de Quebec, Hotel-Dieu de Quebec, 11 cote du Palais, Quebec, Quebec G1R 2J6,

Canada5Department of Microbiology & Immunology, McGill Life Sciences Complex, Complex Traits Group, Bellini Pavilion,3649 Promenade Sir William Osler, Montreal, Quebec H3G 0B1, Canada

*Correspondence: [email protected]

http://dx.doi.org/10.1016/j.immuni.2014.02.013

SUMMARY

Aspirin gained tremendous popularity during the1918 Spanish Influenza virus pandemic, 50 yearsprior to the demonstration of their inhibitory actionon prostaglandins. Here, we show that duringinfluenza A virus (IAV) infection, prostaglandin E2

(PGE2) was upregulated, which led to the inhibitionof type I interferon (IFN) production and apoptosisin macrophages, thereby causing an increase invirus replication. This inhibitory role of PGE2 wasnot limited to innate immunity, because both antigenpresentation and T cell mediated immunity were alsosuppressed. Targeted PGE2 suppression via geneticablation of microsomal prostaglandin E-synthase 1(mPGES-1) or by the pharmacological inhibition ofPGE2 receptors EP2 and EP4 substantially improvedsurvival against lethal IAV infection whereas PGE2

administration reversed this phenotype. These datademonstrate that the mPGES-1-PGE2 pathway istargeted by IAV to evade host type I IFN-dependentantiviral immunity. We propose that specific inhibi-tion of PGE2 signaling might serve as a treatmentfor IAV.

INTRODUCTION

The lung innate immune system has a critical role in limiting

respiratory viral infections (Braciale et al., 2012). Alveolar macro-

phages (M4) are one of the first immune cells to encounter pul-

monary viruses and are indispensable for several key antiviral

mechanisms including: (1) virus recognition and initiation of

cytokine and chemokine production, (2) instruction of adaptive

immunity, and (3) control of lung injury and resolution of inflam-

mation (Kim et al., 2008; Shirey et al., 2010). Initially, upon

sensing viral pathogen-associated molecular patterns (PAMPs),

macrophages induce a cascade of cytokine responses including

type I interferon a (IFN-a) and IFN-b production. Type I IFNs

signal through the IFN-a receptor (IFNAR) and induce the upre-

gulation of numerous genes that activate both innate and adap-

tive immunity for rapidly controlling viral replication (Koerner

et al., 2007). In addition, infected macrophages might undergo

apoptosis to restrict viral dissemination (Mok et al., 2007) and

to enhance T cell-mediated immunity via antigen (Ag) cross-pre-

sentation (Albert et al., 1998). Recent evidence also suggests

that during infection, macrophages have the capacity to trans-

port Ags from the lung to the draining lymph nodes and poten-

tially contribute to the induction of adaptive immune responses

(Kirby et al., 2009).

Although cytokines have been extensively studied in the

context of viral infection, little is known about the function of the

prostanoid (which includes prostaglandins [PG]) in antiviral im-

munity (McCarthy and Weinberg, 2012). Among PGs, PGE2 is

the best-characterized immune modulatory lipid, acting on four

subtypes of G protein-coupled E prostanoid receptors (EP1-

EP4) to exert both pro- and anti-inflammatory responses de-

pending on the source of inflammation (Ricciotti and FitzGerald,

2011). Biologically active PGE2 originates fromPGH2,which itself

is the product of cyclooxygenase-1 (COX-1) and COX-2-medi-

ated enzymatic conversion of membrane phospholipid-derived

arachidonic acid (AA) (Figure 1A). Whereas three enzymes have

been shown to independently generate PGE2 from PGH2 (Smyth

et al., 2009), microsomal prostaglandin E-synthase 1 (mPGES-1)

is the dominant isoform for PGE2 synthesis in different mouse

models of inflammation especially involving macrophages

(Hara et al., 2010). Aspirin, a COX inhibitor, gained popularity

during the 1918 Spanish Influenza A virus (IAV) pandemic

(Starko, 2009) and has since then been used for the manage-

ment of IAV symptoms. However, the loss-of-function in COX

Immunity 40, 1–15, April 17, 2014 ª2014 Elsevier Inc. 1

Page 2: Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Interferon and Apoptosis in Macrophages

Figure 1. Ptges–/– Mice Have More Monocytes and Macrophages and Are More Protected against IAV Infection

(A) Schematic representation of prostanoid synthesis pathways. Membrane phospholipid-derived AA is converted to PGH2 by COX-1 and COX-2. PGH2 is then

converted by specific synthases to five major bioactive prostanoids. Highlighted in red is the specific pathway leading to the formation of PGE2 by PGES

enzymes, of which mPGES-1 is the dominant isoform.

(B–O) WT and Ptges�/� mice were infected with a sublethal dose (50 PFU) (B–D and F–O) or a lethal dose (150 PFU) (E) of IAV. Plasma (B) and BAL (C) PGE2

concentrations and pulmonary viral loads (D and E) were measured p.i. (F and G) Micrographs of H&E-stained lung sections prepared p.i. and representative of

the inflammation score depicted in (G). (H) Total lung resistance was measured upon methacholine challenge at day 12 p.i. with flexiVent. Data are presented as

fold increases in pulmonary resistance relative to baseline resistance measured prior to methacholine challenge. (I and J) Fold increase in type I IFN (a and b) (I) or

IFN-b (J) secretion in the lungs (left panels) and BAL (right panels) at day 3 p.i. was measured by B16-Blue reporter cells or ELISA. (K) Chemokines from pooled

BAL of infected mice at day 3 p.i. evaluated with a cytokine array. (L and M) Frequency of monocytes (Mono, CD11c�F4/80�CD115+) and macrophage (lungs:

CD11c+F4/80+ and CD11c�F4/80+; BAL: CD11c+F4/80+) in the lungs and BAL. (N and O) Total numbers of Mono and macrophages, neutrophils (Neutro,

CD11c�F4/80�CD115�Gr1+), NK cells (CD19�NK1.1+), and T cells (CD19�CD3+) in the lungs and BAL at day 3 p.i. (n = 3–5; ± SEM). Results are representative of

at least three independent experiments. In (H), *p < 0.05, ***p < 0.0001 versus WT uninfected controls (two-way ANOVA) and +p < 0.05 versus infected Ptges�/�

mice. In all other panels, *p < 0.05, **p < 0.001, ***p < 0.0001 versus the other genotype. See also Figure S1.

Immunity

PGE2 Inhibition Enhances Antiviral Immunity

2 Immunity 40, 1–15, April 17, 2014 ª2014 Elsevier Inc.

Please cite this article in press as: Coulombe et al., Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Inter-feron and Apoptosis in Macrophages, Immunity (2014), http://dx.doi.org/10.1016/j.immuni.2014.02.013

Page 3: Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Interferon and Apoptosis in Macrophages

Immunity

PGE2 Inhibition Enhances Antiviral Immunity

Please cite this article in press as: Coulombe et al., Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Inter-feron and Apoptosis in Macrophages, Immunity (2014), http://dx.doi.org/10.1016/j.immuni.2014.02.013

proteins have yielded controversial findings upon IAV infection

(McCarthy and Weinberg, 2012) potentially due to inhibition of

all PGs including PGE2, PGI2, PGD2, PGF2a, and thromboxane

A2 (TXA2).

We have previously shown that Mycobacterium tuberculosis

(Mtb) inhibits PGE2 production in macrophages to suppress

both innate and adaptive immunity (Chen et al., 2008; Divan-

gahi et al., 2009; Divangahi et al., 2010) and, therefore,

mPGES-1-deficient (Ptges�/�) mice, which specifically lack

PGE2, are highly susceptible to infection. In sharp contrast to

Mtb infection, we herein report that Ptges�/� mice demon-

strated remarkably enhanced protection against IAV infection.

Mechanistically, PGE2 acted via macrophage EP2 and EP4 re-

ceptors to inhibit type I IFN production and apoptosis, thereby

directly impairing the capacity of mouse and human macro-

phages, but not epithelial cells, to restrict viral replication.

T cell recruitment and Ag-specific T cell response were also

enhanced in infected Ptges�/� mice. Finally, we show that

treatment with a specific mPGES-1 inhibitor drastically reduced

the mortality rate of mice lethally infected with IAV in a type I

IFN manner.

RESULTS

Ptges–/– Mice Are More Resistant to IAV Infection andPromote Monocyte and Macrophage Recruitment intothe LungsTo assess whether there is an alteration in PGE2 during the

course of IAV infection in vivo, we first measured PGE2 amounts

in plasma and bronchoalveolar lavage (BAL) fluid of IAV-infected

mice. IAV infection markedly increased PGE2 in WT mice,

whereas such increase was blunted in Ptges�/� mice as ex-

pected (Figures 1B and 1C). Interestingly, increased PGE2 was

associated with higher viral loads in the lungs of WT mice at

day 3 and 6 postinfection (p.i.) as compared to Ptges�/� mice

(Figures 1D and 1E). Lung histological analysis showed an in-

crease in inflammation in Ptges�/� mice during the early phase

of infection while inflammation gradually and similarly increased

from day 3 to day 9 in both WT and Ptges�/�mice with a trend to

be reduced in the lungs of Ptges�/� mice at day 12 p.i. (Figures

1F–1G; see also Figure S1A available online). We next evaluated

the consequences of inflammation on lung physiology. Although

no significant difference was observed between groups at day

6 p.i. (Figure S1B), airway hyperreactivity (AHR) was reduced

in IAV-infected Ptges�/�mice at 12 days p.i. (Figure 1H; Fig-

ure S1C), which was in line with faster resolution of inflammation

in these mice. The improvement of AHR in IAV-infected Ptges�/�

mice was associated with substantial reduction of interleukin-13

(IL-13) in the lungs, a key cytokine involved in mediating AHR

(Chang et al., 2011) (Figure S1D).

In order to investigate whether the early increase in inflam-

mation at day 3 was consistent with the protective phenotype

of IAV-infected Ptges�/� mice, we next characterized the innate

immune response in wild-type (WT) and Ptges�/� mice after IAV

infection. Measurement of pulmonary cytokine and chemokine

responses revealed increases in type I IFN-a and IFN-b as well

as IL-12p40 in the lungs of Ptges�/� mice compared to WT an-

imals at day 3 p.i. (Figures 1I and 1J; Figure S1E). In addition,

monocyte chemotactic protein-1 (MCP-1) and MCP-5, impli-

cated in monocyte recruitment during IAV infection (Dessing

et al., 2007; Sarafi et al., 1997), but not chemokine (C-X-C

motif) ligand 1 (CXCL1), which is mainly involved in neutrophil

recruitment (Wareing et al., 2007), were markedly increased in

the BAL of Ptges�/� mice (Figure 1K). Consistent with the che-

mokine profile, we observed a transient increase in the fre-

quency of monocytes and macrophages at day 3 p.i. in both

the lungs and the BAL of Ptges�/� mice compared to WT

mice (Figures 1L and 1M; Figures S1F and S1G). This increase

was specific to monocytes, macrophages, and dendritic cells

(DCs) because only the number of these cells, but not neutro-

phils, natural killer (NK) cells, and T cells, were increased in

both the lungs and the BAL of Ptges�/� mice (Figures 1N and

1O; Figures S1F–S1L). The increased pool of monocytes and

macrophages recruited to the lungs of Ptges�/� mice might

have, at least in part, originated from blood-derived monocytes

as previously described (Landsman and Jung, 2007) since the

number of monocytes, but not neutrophils or NK cells, was

transiently increased in the blood circulation at day 1 p.i. (Fig-

ures S1M–S1O). Collectively, these results suggest that PGE2

plays an opposing role in immune protection against IAV infec-

tion by suppressing monocytes and macrophage recruitment

into the lungs.

PGE2-Deficient Macrophages Have Enhanced AntiviralActivity against IAV InfectionBecause macrophages are essential for pulmonary antiviral

immunity by producing type I IFN and undergoing apoptosis

to restrict IAV dissemination (Kim et al., 2008; Mok et al.,

2007; Xing et al., 2009), we next examined the effect of PGE2

on macrophage-mediated immunity to IAV infection. Interest-

ingly, bone-marrow-derived macrophages (BMDM) obtained

from Ptges�/� mice produced more IFN-b than WT macro-

phages upon IAV infection (Figures 2A and 2B). Strikingly,

Ptges�/� macrophages infected with IAV underwent increased

apoptosis and produced considerably more apoptotic vesicles

than WT macrophages (Figures 2C and 2D), whereas exoge-

nous PGE2 suppressed apoptosis in IAV-infected THP-1 mac-

rophages (Figure S2A). Following in vivo infection, macrophage

apoptosis but not necrosis was also increased in the lungs of

Ptges�/� mice relative to WT mice (Figure S2B). To determine

the mechanism(s) by which PGE2 inhibits macrophage type I

IFN production and apoptosis following IAV infection, we

first measured the expression of mRNA encoding mPGES-1

and PGE2 receptors EP1-4 in infected WT and Ptges�/� macro-

phages by using quantitative RT-PCR. IAV infection signifi-

cantly (p < 0.05) increased the expression of mRNA encoding

mPGES-1, EP2, and EP4, but not EP1, and EP3 in WT, but

not Ptges�/� macrophages (Figures 2E and 2F; Figure S2C).

We next used antagonists for EP2 and EP4, as well as inhibitors

of their downstream signaling kinases phosphotidylinositol-3

kinase (PI3K), Akt, and protein kinase A (PKA) (Regan, 2003),

to determine the specific pathway(s) through which PGE2

inhibited type I IFN secretion and apoptosis. Reconstitution

of PGE2 in IAV-infected Ptges�/� macrophages reduced both

type I IFN secretion (Figure 2G, upper panel) and apoptosis

(Figure 2G, lower panel). Inhibition of EP2, EP4, or both EP2

and EP4 in IAV-infected Ptges�/� macrophages exposed to

PGE2 restored type I IFN induction indicating that both EP2

Immunity 40, 1–15, April 17, 2014 ª2014 Elsevier Inc. 3

Page 4: Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Interferon and Apoptosis in Macrophages

Figure 2. PGE2 Inhibits Type I IFN Production and Apoptosis by Macrophages

(A–K)WT and/or Ptges�/�BMDMwere infected or not with IAV. (A) Fold induction in IfnbmRNA and (B) IFN-b secretion weremeasuredwith qRT-PCR and ELISA.

(C) Cell death ELISA at 24 hr p.i. (D) Fold increase in apoptotic blebs formation relative to uninfected or unstimulated cells at 24 hr p.i. (E and F) Fold induction in

Ep1, Ep2, Ep3, and Ep4 mRNA in WT (E) and Ptges�/� (F) BMDM were measured by qRT-PCR. (G) Ptges�/� BMDM were pretreated or not with various

combinations of PGE2, EP2 antagonist, EP4 antagonist, PI3K-Akt inhibitor, or PKA inhibitor for 1 hr and then infected or not for 24 hr. Fold increase in type I IFN

(a and b) (top panel) and apoptosis (bottom panel) relative to uninfected and unstimulated cells were measured. (H) pIRF3 and total IRF3 proteins were measured

in whole-cell protein extracts of WT and Ptges�/� BMDM. Macrophages were pretreated or not with PGE2 for 1 hr prior to IAV infection (5 MOI) or stimulated with

1 mg/ml LPS. Actin was used as a loading control. Densitometry analysis (bottom) of pIRF3 was relative to IRF3 expression. (I and J) BMDMwere pretreated or not

with various concentrations of PGE2 for 1 hr and then infected for 24 hr. Fold increase in type I IFN secretion (I) and viral load (J) was measured. (K) Ptges�/�

BMDM were pretreated or not with PGE2, PGD2, PGF2a, PGI2, or carbocyclic TXA2 for 1 hr, infected for 24 hr, and viral load was then measured.

(legend continued on next page)

Immunity

PGE2 Inhibition Enhances Antiviral Immunity

4 Immunity 40, 1–15, April 17, 2014 ª2014 Elsevier Inc.

Please cite this article in press as: Coulombe et al., Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Inter-feron and Apoptosis in Macrophages, Immunity (2014), http://dx.doi.org/10.1016/j.immuni.2014.02.013

Page 5: Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Interferon and Apoptosis in Macrophages

Immunity

PGE2 Inhibition Enhances Antiviral Immunity

Please cite this article in press as: Coulombe et al., Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Inter-feron and Apoptosis in Macrophages, Immunity (2014), http://dx.doi.org/10.1016/j.immuni.2014.02.013

and EP4 are the major PGE2 receptors responsible for inhibition

of type I IFN (Figure 2G, upper panel). We furthermore

confirmed that downstream signaling kinases PI3K-Akt as

well as PKA are involved in mediating the inhibitory effects of

PGE2 on type I IFN production (Figure 2G, upper panel). How-

ever, during apoptosis, the inhibitory action of PGE2 was

dependent only on EP2 and PKA but not EP4 and PI3K-Akt

in IAV-infected Ptges�/� macrophages (Figure 2G, lower panel).

We next evaluated the phosphorylation of the IRF3 (pIRF3),

which is a key transcription factor for induction of type I IFN

in IAV-infected cells. Ptges�/� macrophages exhibited an early

increase in pIRF3 as compared to WT macrophages following

IAV infection (Figure 2H). Importantly, PGE2 reconstitution in

Ptges�/� macrophages drastically inhibited pIRF3 following

IAV infection. Consistent with the inhibitory effects of PGE2

on IRF3-mediated type I IFN signaling, Ptges�/� macrophages

showed increased type I IFN and decreased viral load, while

addition of exogenous PGE2 in a dose-dependent manner

reduced type I IFN and enhanced viral replication in both WT

and Ptges�/� macrophages (Figures 2I and 2J). Unlike PGE2,

addition of other prostanoids (PGD2, PGF2a, PGI2, TXA2) to

IAV-infected Ptges�/� macrophages had no effect on viral repli-

cation (Figure 2K; Figure S2D). Moreover, selective antagonists

for individual PG receptors showed no changes in either type I

IFN production or viral load in IAV-infected Ptges�/� macro-

phages (Figure S2E). These data collectively rule out the possi-

bility that, in the absence of mPGES-1, PGH2 is converted to

other PGs that then contribute to antiviral immunity. Further-

more, similar to another report (Trebino et al., 2005) demon-

strating that in the absence of mPGES-1 there is no shunting

to the leukotriene pathway, we also found no difference in

kinetics of leukotriene B4 production in either plasma or BAL

of IAV-infected WT and Ptges�/� mice (Figures S2F and

S2G). To further dissect the mechanism of how IAV inhibits

type I IFN via PGE2 in macrophages, we stimulated WT and

Ptges�/� macrophages with UV-treated replication-deficient

IAV or the RIG-I ligands 50pppRNA and PolyI:C. Under these

conditions, mPGES-1-dependent type I IFN production was

abolished indicating that active viral replication is required for

IAV to inhibit type I IFN via PGE2 (Figure 2L; Figures S2H–

S2J). Consistent with this observation, replication-competent

IAV induced more mRNA encoding mPGES-1 than UV IAV or

RIG-I ligands and only the replication competent virus induced

PGE2 secretion in WT macrophages (Figures 2M and 2N).

These data collectively indicate that IAV specifically targets

the mPGES-1 pathway and that PGE2 was the main mediator

inhibiting macrophage type I IFN-dependent antiviral immunity

to IAV infection.

Considering BMDM are not an ideal representative of pulmo-

nary macrophages, we next extended our study to investigate

the potential role of PGE2 in alveolar macrophages (AM) in-

fected with IAV. Similar to BMDM, Ptges�/� AM had increased

type I IFN and decreased viral titer as compared to WT AM

(L–N) WT and/or Ptges�/� BMDM were infected or stimulated or not with IAV (MO

increase in type I IFN (a and b) secretion wasmeasured (L). (M) Fold induction in Pt

measured at 4 hr following infection or stimulation with ELISA. Results are ± SEM a

***p < 0.0001: in (A–D and L), versus WT; in (E–G and N), versus uninfected or

unstimulated BMDM of the same genotype unless indicated otherwise (two-way

(Figures 3A and 3B). Pretreatment of AM of both genotypes

with exogenous PGE2 decreased type I IFN and increased viral

load (Figures 3A and 3B). Supporting these findings, intracel-

lular staining of AM with an antibody against IAV nucleoprotein

(NP) revealed that PGE2 deficiency decreased the frequency of

infected AM (Figure 3C), whereas exogenous PGE2 increased

the frequency of both infected WT and Ptges�/� AM. A similar

result was obtained with BMDM (data not shown). Importantly,

in human macrophages infected with a clinical strain of IAV,

gene targeting of mPGES-1 with small interfering RNA (Fig-

ure 3D) or administration of exogenous PGE2 (Figure 3E; Fig-

ure S3A) confirmed the detrimental role of the PGE2 pathway

in the control of viral replication and recapitulated our findings

in murine macrophages. To directly test whether the increased

antiviral activity of Ptges�/� macrophages translated to a better

control of IAV infection in vivo, AM derived from WT and

Ptges�/� mice were infected with IAV in vitro and adoptively

transferred intratracheally (i.t.) to Rag1�/� mice (lacking T and

B cells) (Figure 3F). Although both the frequency of infected

cells and the number of viruses in WT and Ptges�/� macro-

phages was similar prior to transfer (Figures S3B and S3C),

the mice that received infected Ptges�/� macrophages showed

increased type I IFN (Figure 3G; Figure S3C) and decreased

pulmonary viral titer (Figure 3H) as compared to the mice

receiving infected WT macrophages. To further support this

observation, we next adoptively transferred (i.t.) either naive

WT or Ptges�/� macrophages into naive WT recipient mice.

Then after adoptive transfer, mice were infected with IAV. Simi-

larly, adoptive transfer of Ptges�/� macrophages into WT recip-

ients led to increased type I IFN production, decreased viral

replication and increased survival (Figures 3I–3L; Figure S3E).

Because PGE2 inhibited the production of type I IFN in IAV-

infected macrophages, we hypothesized that the ability of a

specific murine mPGES-1 inhibitor (CAY10526) to limit viral

replication is directly linked to the type I IFN system. To test

this hypothesis, we first assessed the capacity of the

mPGES-1 inhibitor to reduce viral replication in WT macro-

phages, Ptges�/� macrophages, and in macrophages lacking

the type I IFN receptor (Ifnar�/�), which are unable to respond

to type I IFN. As expected, Ptges�/� macrophages infected

with IAV revealed a reduction in viral load, whereas Ifnar�/�

macrophages showed increased viral load in comparison to

IAV-infected WT macrophages (Figure 3M). The mPGES-1 in-

hibitor reduced viral load in IAV-infected WT macrophages

to a similar level as in IAV-infected Ptges�/� macrophages

but had no effect in Ptges�/� macrophages. Importantly, the

protective effect of the mPGES-1 inhibitor in controlling viral

replication was completely abrogated in Ifnar�/� macrophages

(Figure 3M). We next considered whether neutralizing anti-

bodies (Abs) against both IFN-a and IFN-b would recapitulate

the phenotype of Ifnar�/� macrophages infected with IAV.

Treatment of IAV-infected macrophages with neutralizing Abs

against both IFN-a and IFN-b completely abolished the

I = 1) of UV-inactivated IAV, 50pppRNA (1 mg/ml), or PolyI:C (1 mg/ml) and fold

gesmRNA inWT BMDMwasmeasured with qRT-PCR. (N) PGE2 secretion was

nd are representative of three independent experiments. *p < 0.05, **p < 0.001,

unstimulated BMDM (two-way ANOVA); in (I–K and M), versus infected but

ANOVA). See also Figure S2.

Immunity 40, 1–15, April 17, 2014 ª2014 Elsevier Inc. 5

Page 6: Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Interferon and Apoptosis in Macrophages

Figure 3. PGE2 Impairs the Control of Viral Replication by Macrophages via Type I IFN

(A–C) AM from naive WT and Ptges�/� mice were pretreated or not with PGE2 for 1 hr and infected or not with IAV. IFN-b secretion (A) and viral load (B) were

measured from the supernatant at 24 hr p.i. (C) Kinetics of the frequency of infected AM (CD11c+F4/80+NP+). Zebra plots are representative of the 24 hr time-point

and numbers adjacent to gates indicate percent of macrophages stained with intracellular anti-NP.

(D) Viral load in the supernatant of THP-1 cells transfected with a noncoding sequence transfection control (NC1) ormPGES-1-targetting DsiRNA and infected for

24 hr with IAV H3N2. Immunoblot analysis of mPGES-1 gene silencing in THP-1 macrophages is shown.

(E) Viral load in the supernatant of primary human macrophages pretreated or not with PGE2 for 1 hr and infected for 24 hr with IAV H3N2.

(F–H) AM from naive WT and Ptges�/� mice were isolated and infected with IAV in vitro and then adoptively transferred (i.t.) to Rag1�/� mice (n = 5). IFN-b

concentration (G) and viral loads (H) were measured in the lungs of Rag1�/� mice 3 days after adoptive transfer.

(I–L) BMDM fromWT and Ptges�/�mice were adoptively transferred (i.t.) toWTmice, which were then infected with IAV after 2 hr (n = 5–10). Fold increase in type I

IFN (a and b) (J), viral loads (K), and survival (L) were assessed 3 days after adoptive transfer.

(M and N)WT, Ptges�/�, and Ifnar�/�BMDMwere treated or not with a specificmurinemPGES-1 inhibitor and/or anti-IFN-a and anti-IFN-b and infected with IAV.

Viral load was measured 24 hr p.i. Results are shown as means ± SEM and are representative of at least two independent experiments. *p < 0.05, **p < 0.001,

***p < 0.0001. In (C), (two-way ANOVA). See also Figure S3.

Immunity

PGE2 Inhibition Enhances Antiviral Immunity

Please cite this article in press as: Coulombe et al., Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Inter-feron and Apoptosis in Macrophages, Immunity (2014), http://dx.doi.org/10.1016/j.immuni.2014.02.013

protective effect of both the mPGES-1 inhibitor and Ptges dele-

tion on viral replication (Figure 3N). Therefore, IAV infection

specifically activates the PGE2 pathway in macrophages to

6 Immunity 40, 1–15, April 17, 2014 ª2014 Elsevier Inc.

inhibit type I IFN and apoptosis through EP2 and EP4-depen-

dent mechanisms, thereby impairing macrophage capacity to

restrict viral replication.

Page 7: Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Interferon and Apoptosis in Macrophages

Immunity

PGE2 Inhibition Enhances Antiviral Immunity

Please cite this article in press as: Coulombe et al., Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Inter-feron and Apoptosis in Macrophages, Immunity (2014), http://dx.doi.org/10.1016/j.immuni.2014.02.013

Antiviral Immunity of Airway Epithelial Cells Is NotDirectly Affected by PGE2

Airway epithelial cells are known to constitutively secrete PGE2

and to contribute to the modulation of DCs under homeostatic

conditions (Schmidt et al., 2011). To investigate the potential

contribution of epithelial cells as a source of PGE2, we purified

CD45�CD326+ primary airway epithelial cells from WT and

Ptges�/� mice (Figure S4A). Initially, we measured the induction

of mRNA encoding mPGES-1 by IAV infection in epithelial cells

derived from WT mice and found that its induction was almost

negligible relative to macrophages (4.0-fold ± 0.6 SEM versus

42.4-fold ± 5.6 SEM, Figure 4A). To further investigate the poten-

tial contribution of PGE2-dependent macrophage immunity to

IAV infection, we next depleted alveolar phagocytes from WT

andPtges�/�mice prior to IAV infection by using clodronate lipo-

somes. Similar to other studies (Huang et al., 2011), �75% AM

were depleted prior to infection (data not shown) and these cells

were reduced by �50% at day 3 p.i. (Figure 4B). As expected,

Ptges�/� mice treated with control liposomes had lower viral

load in the lungs compared to control liposome-treated WT

mice (Figure 4C). Macrophage depletion increased viral replica-

tion in bothWT andPtges�/� lungs. Importantly, the level of repli-

cation in macrophage-depleted Ptges�/�mice was similar to the

levels of viral replication in macrophage-competent WT mice,

suggesting that the ability of Ptges�/� mice to restrict viral repli-

cation is mediated by macrophages and not other cell types,

especially pulmonary epithelial cells. In addition, there was no

difference in type I IFN expression or viral load between IAV-in-

fected WT and Ptges�/� mouse primary airway epithelial cells

(Figures 4D–4G) and between mouse (Figures 4H–4K; Figures

S4B–S4D) or human (Figures 4L–4O) airway epithelial cell lines

treated or not with exogenous PGE2 or the mPGES-1 inhibitor.

Furthermore, the loss of mPGES-1 function in primary human

bronchial epithelial cells had no effect on viral load (Figure 4P).

Thus, although the constitutive production of PGE2 by the airway

epitheliummight contribute to the inhibition of macrophage anti-

viral activity through a paracrine pathway, it does not directly

affect type I IFN or viral replication in epithelial cells.

PGE2 Inhibits T Cell Recruitment and Delays the Onsetof IAV Ag-Specific T Cell Response in the LungsMacrophage apoptosis provides an important link to adaptive

immunity as viral Ag contained in apoptotic vesicles can be taken

up by DCs and efficiently presented to naive T cells leading to

generation and maintenance of protective virus-specific T cell

responses (Albert et al., 1998). Because IAV-infected Ptges�/�

macrophages revealed increased apoptosis and apoptotic ves-

icles, we next examined the generation and kinetics of T cell

mediated immunity in WT and Ptges�/�mice following IAV infec-

tion. The total T cell numbers as well as CD4+ and CD8+ T cells

were increased in the mediastinal lymph nodes (LN) (Figures

S5A andS5C), BAL (Figure 5A; Figure S5C), and lungs (Figure 5B;

Figures S5B and S5C) of Ptges�/� mice compared to WT. This is

consistent with an increase in pulmonary CXCL9 and CXCL10,

chemokines implicated in T cell recruitment during IAV infection

(Piqueras et al., 2006) (Figure S5D). Additionally, the Ag-specific

CD8+ T cell response to the immunodominant NP Ag was higher

in the BAL (Figure 5C) and lungs (Figures 5D and 5E) of Ptges�/�

mice. Pulmonary T cells from infected Ptges�/� mice produced

more IFN-g after being stimulated with whole UV-inactivated

IAV, NP peptide (specific for CD8+ T cells) or hemagglutinin

(HA) peptide (specific for CD4+ T cells) than did T cells from in-

fected WT mice (Figures 5F and 5G). Consistent with the

increased frequency of IFN-g-secreting CD4+ T cells in Ptges�/�

mice and with the previous finding that PGE2 negatively regu-

lates MHC class II expression in macrophages (Zaslona et al.,

2012), the surface expression of MHC class II was increased in

Ptges�/� compared to WT macrophages following IAV infection

(Figure S5E).

Because the increased T cell response in Ptges�/� mice was

not due to enhanced intrinsic capacity of T cells to proliferate

as assessed by an in vitro assay of T cell proliferation (Fig-

ure S6A), we next determined whether PGE2 directly inhibits

Ag presentation during IAV infection in a type I IFN-dependent

manner. WT, Ptges�/�, and Ifnar�/� macrophages were pre-

loaded with ovalbumin (OVA) and cocultured with CFSE-labeled

OVA-specific CD8+ T cells derived from OT-I mice (TCR-trans-

genic CD8+ T cells specific for the OVA SIINFEKL epitope) in

the presence or absence of IAV infection and/or themPGES-1 in-

hibitor. In the absence of infection, WT, Ptges�/�, and Ifnar�/�

macrophages loaded with OVA induced similar levels of OT-I

CD8+ T cell proliferation whether themPGES-1 inhibitor was pre-

sent or not (Figures 6A and 6B). However, in the presence of IAV

infection, the proliferation of OT-I CD8+ T cells was reduced in

WT and Ifnar�/� macrophages compared to Ptges�/� macro-

phages. Similarly, addition of the mPGES-1 inhibitor to IAV-

infected WT macrophages restored their ability to induce OT-I

CD8+ T cell proliferation up to a similar level to Ptges�/� macro-

phages. Importantly and consistent with the notion that type I

IFN are known to enhance cross-presentation (Le Bon and

Tough, 2008), the effect of the mPGES-1 inhibitor in enhancing

OT-I CD8+ T cell proliferation was absent in Ifnar�/� macro-

phages. Therefore, these data collectively indicate that PGE2

suppresses antiviral T cell-mediated immunity by dampening

both cross- and direct priming of T cells via inhibition of the

type I IFN system during IAV infection.

To investigate whether the enhanced T cell response in

Ptges�/� mice was due to an intrinsic property of Ptges�/� mac-

rophages, AM derived fromWT and Ptges�/�mice were infected

with IAV in vitro and adoptively transferred (i.t.) into WT recipient

mice (Figure 6C). Although the number of viruses in WT and

Ptges�/�macrophages was similar prior to transfer (Figure S6B),

the WT mice that received IAV-infected Ptges�/� macrophages

showed increased frequency and absolute number of Ag-spe-

cific CD8+ T cells in the mediastinal LN (Figure 6D), as well as

decreased pulmonary viral titer at day 7 posttransfer (Figure 6E).

Strikingly, all the WT mice that received IAV-infected Ptges�/�

macrophages survived, whereas 80% of mice receiving IAV-in-

fected WT macrophages succumbed to death by day 9 post-

transfer (Figure 6F). Thus, the absence of PGE2 production in

IAV-infected Ptges�/� macrophages correlates with greater

T cell response, enhanced protection, and survival during infec-

tion in vivo.

Because PGE2 negatively regulated both the innate and adap-

tive arms of immunity to IAV infection, we next delineated the

contributions of each arm to protection against lethal IAV infec-

tion. Initially, we used monoclonal antibodies (Abs) to deplete

CD4+ and CD8+ T cells in WT and Ptges�/� mice (Figure S6C).

Immunity 40, 1–15, April 17, 2014 ª2014 Elsevier Inc. 7

Page 8: Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Interferon and Apoptosis in Macrophages

Figure 4. PGE2 Does Not Directly Impair Antiviral Immunity in Airway Epithelial Cells

(A) BMDM andmouse primary airway epithelial cells derived fromWTmice were infected or not with IAV for 24 hr to assess PtgesmRNA expression by qRT-PCR.

*p < 0.05 versus IAV-infected WT epithelial cells.

(B and C)WT and Ptges�/�micewere administered control liposomes or clodronate liposomes to deplete alveolar phagocytes prior to infection with IAV (50 PFU,

n = 5 mice per group). (B) Representative dot blots depicting the analysis of the frequency of AM in PBS-treated WT mice or liposome-treated WT and Ptges�/�

mice at day 3 p.i. Numbers adjacent to gate indicate percent of AM. (C) Viral loadwasmeasured in the lungs of control liposome- and clodronate liposome-treated

mice at day 3 pi. *p < 0.05, **p < 0.001. Data are representative of three independent experiments.

(D–G) WT and Ptges�/� murine primary airway epithelial cells were infected or not with IAV as in (A) to assess Ifnb (D) or Ifna4 (E) mRNA by qRT-PCR, IFN-b

secretion (F) by ELISA, and viral load (G).

(H–K)Murine epithelial (MLE12) cells were infected or not with IAV as in (A) in the presence or absence of exogenous PGE2 or themPGES-1 inhibitor to assess Ifnb

mRNA by qRT-PCR (H), type I IFN (a and b) secretion using B16 cells (I and J), and viral load at 24 hr p.i. (K).

(legend continued on next page)

Immunity

PGE2 Inhibition Enhances Antiviral Immunity

8 Immunity 40, 1–15, April 17, 2014 ª2014 Elsevier Inc.

Please cite this article in press as: Coulombe et al., Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Inter-feron and Apoptosis in Macrophages, Immunity (2014), http://dx.doi.org/10.1016/j.immuni.2014.02.013

Page 9: Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Interferon and Apoptosis in Macrophages

Figure 5. Ptges–/– Mice Have Enhanced T Cell Mediated Immunity to IAV Infection

(A–G)WT and Ptges�/�mice were infected with a sublethal dose of IAV. Kinetics of total T cell (CD19�CD3+) numbers in the BAL (A) and lungs (B) were evaluated.

(C–E) Kinetics of the frequency of IAV H-2Db-NP CD8+ T cells wasmeasured in the BAL (C) and lungs (D and E). Numbers adjacent to gates indicate percent CD8+

T cells stained with the NP-loaded H-2Db tetramer (NPTet). (F and G) The frequency of Ag-specific IFN-g-releasing T cells from lungs or BAL was determined

following 10 hr stimulation with UV-inactivated IAV (1MOI) or 5 mg/ml IAVNP366–374 or IAVHA110–120 peptides by intracellular cytokine staining and flow cytometry.

(F) Representative dot blots depicting the analysis of frequency of IAV NP366–374-specific IFN-g-secreting CD8+ T cells in the lungs of IAV-infected WT and

Ptges�/� mice at day 11 pi. Numbers adjacent to gates indicate percent IFN-g-secreting CD8+ T cells following ex vivo restimulation with MHC class I-restricted

IAV NP366–374 peptide. (G) Total number of CD8+ (left panels) and CD4+ (right panels) IFN-g-secreting T cells specific for either UV-inactivated IAV or MHC class

I-restricted NP366–374 and MHC class II-restricted HA110–120 peptide respectively in the lungs (top panels) and BAL (bottom panels) of IAV-infected WT and

Ptges�/� mice at day 9 and 11 p.i. (n = 3–5; ± SEM). Results are representative of three independent experiments. *p < 0.05 versus WT. See also Figure S5.

Immunity

PGE2 Inhibition Enhances Antiviral Immunity

Please cite this article in press as: Coulombe et al., Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Inter-feron and Apoptosis in Macrophages, Immunity (2014), http://dx.doi.org/10.1016/j.immuni.2014.02.013

WTmice depleted or not of T cells all died within 10 days of infec-

tion (Figure 6G; Figure S6D). Although Ptges�/�mice depleted of

T cells had increased susceptibility relative to their T cell-compe-

tent Ptges�/� counterparts, they still demonstrated improved

survival over WT mice. Moreover, pharmacological inhibition of

mPGES-1 in Rag1�/� mice again caused an increase in survival

against lethal IAV challenge (Figure 6H; Figure S6E). Because the

protective effect of the mPGES-1 inhibitor was less pronounced

in Rag1�/� mice compared to Ptges�/� mice depleted of T cells,

we next examined the potential contribution of PGE2 in B cell

(L–O) Human epithelial (A549) cells were infected or not with IAV H3N2 in the pres

expression. The frequency of infected cells was evaluated by intracellular stainin

(P) Viral load in human bronchial epithelial cells (NHBE) transfected with a nonco

infected for 24 hr with IAV H3N2. Immunoblot analysis ofmPGES-1 gene silencing

and are representative of three independent experiments. See also Figure S4.

production of IAV specific Ab. In the serum, the levels of both

IAV-specific immunoglobulin G1 (IgG1) and IgG2a were

increased in Ptges�/� mice at day 11 post-IAV infection (Figures

6I and 6J). These data indicate that although T cell mediated im-

munity is essential for protection against IAV; targeted PGE2 in-

hibition enhances innate immune protection to IAV, independent

of adaptive immune components. Overall, our data suggest that

enhanced macrophage-mediated antiviral immunity through

PGE2 inhibition positively regulates both innate and adaptive

immunity to pulmonary IAV infection.

ence or absence of exogenous PGE2 to assess Ifnb (L) and Ifna PAN (M) mRNA

g with anti-NP (N), and viral load was determined (O).

ding sequence transfection control (NC1) or mPGES-1-targetting DsiRNA and

in NHBE cells infected with IAV is shown. Results are shown as means ± SEM

Immunity 40, 1–15, April 17, 2014 ª2014 Elsevier Inc. 9

Page 10: Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Interferon and Apoptosis in Macrophages

Figure 6. PGE2 Suppresses the Ag Presentation Machinery in a Type I IFN-Dependent Manner

(A and B) CD8+ T cell proliferation was measured after CFSE-labeled OT-I CD8+ T cells were cocultured for 72 hr with WT or Ptges�/� BMDM (2 T cells: 1

macrophage) preloaded or not with OVA, infected or not with IAV, and in the presence or absence of the mPGES-1 inhibitor. Numbers adjacent to gates indicate

percent CD8+ T cell proliferation (CFSE dilution).

(C–F) AM from naiveWT andPtges�/�micewere isolated and infectedwith IAV in vitro and then adoptively transferred (i.t.) toWTmice. (D, top) Representative dot

blots depicting the frequency of IAV H-2Db-NP CD8+ T cells in the mediastinal LN of WT mice 7 days following adoptive transfer of IAV-infected WT or Ptges�/�

macrophages. (D, bottom) The frequency (left panel) and total number (right panel) of IAV H-2Db-NP CD8+ T cells in the mediastinal LN of WT mice 7 days

following adoptive transfer of IAV-infected WT or Ptges�/� macrophages. Pulmonary viral load (E) and survival (F) were assessed 7 days after transfer.

(G and H) Survival of WT, Ptges�/�, or Rag1�/� mice infected or not with a lethal dose (150 PFU) of IAV. (G) Mice received control rat IgG or CD4+ and CD8+ T cell

depletingmonoclonal antibody (mAb) regimen (DT cells). (H)Rag1�/�micewere treated intraperitoneally (i.p.) with saline or with themPGES-1 inhibitor from day 1

to day 7 pi.

(I and J) Kinetics of serum anti-IAV IgG1 (I) and IgG2a (J) inWT andPtges�/�mice infected or not with a sublethal dose of IAVwasmeasured by ELISA. (n = 5–10; ±

SEM). Results are representative of at least three independent experiments. *p < 0.05, **p < 0.001. See also Figure S6.

Immunity

PGE2 Inhibition Enhances Antiviral Immunity

Please cite this article in press as: Coulombe et al., Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Inter-feron and Apoptosis in Macrophages, Immunity (2014), http://dx.doi.org/10.1016/j.immuni.2014.02.013

Targeted PGE2 Inhibition Enhances Resistance to aLethal Dose of IAV InfectionConsidering that Ptges�/�mice weremore resistant to IAV infec-

tion, we next examined whether specific inhibition of mPGES-1

would be beneficial as a treatment strategy during IAV infection

10 Immunity 40, 1–15, April 17, 2014 ª2014 Elsevier Inc.

in mice. Upon infection of Ptges�/� mice with a lethal dose of

IAV, the pulmonary viral load was decreased, the survival rate

increased and weight loss reduced compared to WT animals

(Figure 7A-B; Figure S7A). Furthermore, systemic administration

of PGE2 in Ptges�/� mice tended to increase their susceptibility

Page 11: Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Interferon and Apoptosis in Macrophages

Figure 7. Inhibition of PGE2 Enhances Resistance to IAV Infection

(A–F) Survival of WT, Ptges�/�, or Ifnar�/� mice infected or not with a lethal dose (150 PFU) of IAV. (A and B) Mice were administered or not i.p. with exogenous

stable PGE2 analog 16,16-dimethyl PGE2 at day 0 and every 2 days for up to 12 days. One group of infected WT mice was also treated i.p. with the mPGES-1

inhibitor from day 1 to day 7 pi. (A) Pulmonary viral load was measured at day 3 p.i. *p < 0.05, **p < 0.001, ***p < 0.0001 (two-way ANOVA). In (B), *p < 0.05 versus

WTmice infected with IAV only (WT; IAV). (C) Infected mice were treated i.p. with saline or with the mPGES-1 inhibitor starting at different days and up to day 7 pi.

*p < 0.05, **p < 0.001 versus WT mice treated with saline. (D) Infected Ptges�/� mice were administered ip. with 16,16-dimethyl PGE2 as in (A). One group of

infected Ptges�/� mice was also treated i.p. with EP2 antagonist and EP4 antagonist 15 min prior to PGE2 administration. Pulmonary viral load was measured at

day 3 pi. *p < 0.05, **p < 0.001 (two-way ANOVA). (E) Infected WT mice were treated i.p. with saline or with EP2 and EP4 antagonists from day 1 to day 7 p.i.

*p < 0.05. (F) Infected Ifnar�/�micewere treated i.p. with saline or with themPGES-1 inhibitor as in (B). (n = 5–10; ± SEM). Results are representative of three to five

independent experiments. See also Figure S7.

Immunity

PGE2 Inhibition Enhances Antiviral Immunity

Please cite this article in press as: Coulombe et al., Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Inter-feron and Apoptosis in Macrophages, Immunity (2014), http://dx.doi.org/10.1016/j.immuni.2014.02.013

to infection. Strikingly, specific pharmacological inhibition of

mPGES-1 in WT mice from day 1 to day 7 p.i. increased protec-

tion (Figure 7A) and survival (Figure 7B; Figure S7A) to a level

similar to Ptges�/� mice. The beneficial effects of the mPGES-

1 inhibitor on survival were still observed when treatment was

initiated as late as 2 or 3 days p.i. (Figure 7C; Figure S7B). Impor-

tantly, treatment with both EP2 and EP4 antagonists completely

reversed the ability of exogenous PGE2 to increase viral replica-

tion in Ptges�/� mice (Figure 7D) and decreased mortality of

lethally infected WT animals (Figure 7E; Figure S7C). Finally,

the protection provided by mPGES-1 inhibition against a lethal

dose of IAV was entirely dependent on type I IFN because

Ifnar�/� mice were unresponsive to treatment and succumbed

to death (Figure 7F; Figure S7D). Thus, these results demon-

strate that targeted PGE2 inhibition provides protection against

IAV and is dependent on both EP2 and EP4 receptor signaling

and a functional type I IFN response.

DISCUSSION

During IAV infection, pulmonary macrophages convert into

highly active cells and become the major source of inflammatory

cytokines and chemokines including type I IFNs, which restrict

viral replication in airway epithelial cells and dissemination.

These signals orchestrate the recruitment of inflammatory cells

in order to mount a potent antiviral immune response (Braciale

et al., 2012). The data presented here provide evidence that

mPGES-1 and PGE2 play a central role in mediating susceptibil-

ity to IAV infection. IAV virions initially replicate in airway epithelial

cells and are then released in the airspace where they encounter

alveolar macrophages. Despite the fact that bronchial epithelial

cells are known to produce PGE2 (Schmidt et al., 2011), our

data demonstrate that neither the loss nor the gain of PGE2 func-

tion affected type I IFN production or viral replication directly in

this cell type. In sharp contrast, PGE2 had a strong inhibitory ef-

fect on the recruitment and antiviral properties of macrophages.

We identified two distinct and essential pathways through which

PGE2 paralyzed macrophage antiviral immunity via both EP2

and EP4 receptors: type I IFNs and apoptosis. This severely

impaired innate immunity because the IFN system directly coun-

teracts viral replication and apoptosis blocks the cellular

machinery crucial for viral amplification and dissemination. In

addition, we found that PGE2 inhibited T cell mediated immunity

to IAV infection. Thus, IAV specifically hijacks the mPGES-1/

PGE2 pathway to subvert macrophage function and suppress

both innate and adaptive immune responses in a type I IFN-

dependent manner.

The specific mechanism(s) used by the virus to increase PGE2

levels and evade host antiviral responses remain(s) unknown.

Interestingly, the nonstructural protein 1 (NS1) IAV effector pro-

tein activates PI3K, providing antiapoptotic signals (Ehrhardt

et al., 2007) and inhibits production of type I IFN throughmultiple

Immunity 40, 1–15, April 17, 2014 ª2014 Elsevier Inc. 11

Page 12: Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Interferon and Apoptosis in Macrophages

Immunity

PGE2 Inhibition Enhances Antiviral Immunity

Please cite this article in press as: Coulombe et al., Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Inter-feron and Apoptosis in Macrophages, Immunity (2014), http://dx.doi.org/10.1016/j.immuni.2014.02.013

strategies including direct inhibition of IRF3 (Kochs et al., 2007).

This study reports PGE2-mediated suppression of macrophage

type I IFN production and apoptosis during viral infection. In

IAV-infected macrophages, PGE2 inhibited type I IFN via both

EP2 and EP4 receptors and downstream kinases PI3K-Akt and

PKA. Akt has been shown to be activated following TLR stimula-

tion and positively regulate type I IFN (Joung et al., 2011). How-

ever, it is also known that PI3K acts as a negative regulator of

TLR signaling in the later phase of TLR activation (Fukao and

Koyasu, 2003). Thus, the duration and/or magnitude of Akt acti-

vation might be critical in determining the level of type I IFN pro-

duction, and the combined action of NS1 and PGE2 on PI3K-Akt

activation might explain the inhibitory action of Akt in this

context. Alternatively, inhibition of apoptosis by PGE2 required

EP2 and PKA, but not the EP4 and PI3K-Akt pathway. This sug-

gests that the nonredundant role of EP receptors represents a

complementary strategy used by IAV to further inhibit type I

IFN and apoptosis. More studies are required to determine the

molecular basis of PGE2 induction during IAV infection and

whether viral effector molecule(s) actively modulate the level of

PGE2 and other PGs.

Although the essential role of type I IFNs in immunity to viral

infection is well established, their protective activity against bac-

terial infection is more ambiguous. In fact, it has been suggested

that type I IFN production during IAV infection is a mechanism

that sensitizes the host to bacterial superinfection. In line with

this emerging concept, the type I IFN system increases host sus-

ceptibility to Streptococcus pneumoniae (Nakamura et al., 2011;

Shahangian et al., 2009) and Mtb (Stanley et al., 2007). We also

have recently demonstrated that PGE2 enhances protection

against Mtb infection by preventing macrophage necrosis, a

form of cell death providing an exit mechanism forMtb to initiate

a new infection cycle, and inducing macrophage apoptosis

(Chen et al., 2008; Divangahi et al., 2009; Divangahi et al.,

2010). We speculate that because type I IFNs increase the path-

ogenesis ofMtb infection (Stanley et al., 2007) and PGE2 inhibits

type I IFN induction in macrophages, another independent or in-

tegrated protective mechanism of PGE2 in tuberculosis might be

the blockade of the type I IFN system. Although this could poten-

tially explain the opposing function of PGE2 in Mtb versus IAV

infection, the biology of PGE2 in the broader context of infectious

diseases, such as chronic versus acute or bacterial versus viral

infections in addition to its pathogen-specific effects remain to

be determined.

Macrophage permissiveness to IAV replication has been the

subject of recent debate (Friesenhagen et al., 2012; Lee et al.,

2012). In this study, we carefully evaluated viral replication in

macrophages from mice (BMDM and AM) and humans (THP-1

cells and primary monocyte-derived macrophages) by using

two different virus strains (Influenza A/PR/8/34 [H1N1] and Influ-

enza A/HK/1/68 [H3N2]) and two different techniques (viral load

by plaque assay in MDCK cells and intracellular staining for influ-

enza NP protein which is a direct measurement of an active IAV

replication). We found that IAV infects and productively repli-

cates in these cells and that PGE2 was the only prostanoid family

member to impair the control of viral replication in macrophages.

Whereas the biological meaning of productive IAV replication

within macrophages undoubtedly needs more scrutiny, we

anticipate that their permissiveness to IAV replication might be

12 Immunity 40, 1–15, April 17, 2014 ª2014 Elsevier Inc.

critical for their ability of generating Ags for activation and main-

tenance of adaptive immunity (Honke et al., 2012).

The capacity of CD4+ and CD8+ T cells to produce type 1 cyto-

kines such as IFN-g and the cytolytic activity of CD8+ T cells are

crucial mechanisms to control viral replication and to promote

clearance in the infected lungs. Consistent with the previously

published work demonstrating reduction in levels of type 1 cyto-

kines (IFN-g and IL-12) by PGE2 (Betz and Fox, 1991), we found

that PGE2 deficiency led to enhanced CD4+ and CD8+ T cell re-

sponses. We anticipate that at least three mechanisms

contribute to the inhibitory effect of PGE2 on T cell-mediated im-

munity. First, PGE2 reduced the number of bloodmonocytes and

their recruitment into the lungs. A recent study reported that hu-

man blood derived monocytes were highly susceptible to infec-

tion by various IAV strains, which promoted their differentiation

into DCs with enhanced Ag presentation capacity (Hou et al.,

2012). Given the important role of DCs in priming T cells, PGE2-

mediated suppression of monocyte recruitment represents a

mechanism throughwhich IAVeffectively interrupts thispathway.

Second, PGE2 disrupted the crosstalk between macrophages

and DCs by inhibiting macrophage apoptosis. Macrophages

potentially contribute to the direct priming of CD8+ T cells (Kirby

et al., 2009); however, DCs are themajor antigen-presenting cells

(APCs) implicated in this process because they can acquire viral

Ags through the uptake of apoptotic vesicles derived from IAV-

infected macrophages and cross-present these Ags to CD8+

Tcells (Albert et al., 1998). Althoughmacrophage responsiveness

to type I IFN was required for their ability to enhance CD8+ T cell

proliferation in the absence of PGE2, the direct mechanistic link

between PGE2-dependent reduction in macrophage type I IFN

production, apoptosis, and cross-priming of CD8+ T cells re-

mains to be established. Third, PGE2 directly suppressed the

intracellular processing machinery of IAV-infected APCs, as

well as their MHC class II expression impairing their capacity to

present Ags to T cells. Collectively, these findings indicate how

PGE2 can act sequentially and at multiple levels to negatively

regulate the generation of protective type 1 T cell-mediated

immunity to IAV infection.

The observation that IAV causes hyperinduction of COX-2

in human macrophages and epithelial cells has led to several

studies investigating the effect of COX-1 and/or COX-2

deficiency or inhibition during IAV infection (Lee et al., 2008;

McCarthy and Weinberg, 2012). Interestingly, COX-1 or COX-2

deficiency had contrasting effects on host response to IAV infec-

tion (Carey et al., 2005). Ptgs2�/� mice exhibited lower mortality

despite elevated viral titers in the lungs and equivalent produc-

tion of PGE2 in the BAL relative toWTmice. In contrast, Ptgs1�/�

mice produced less PGE2 but did not show any difference in sur-

vival and pulmonary viral load as compared to WT animals. In

another study, the nonselective COX inhibitor paracetamol

(acetaminophen) and the selective COX-2 inhibitor celecoxib

were shown to reduce lung immunopathology following IAV

infection without having any effect on viral clearance or genera-

tion of adaptive immunity (Lauder et al., 2011). We envision that

the reasoning behind these controversial studies is the fact that

inhibition of COX enzymatic activity leads to the suppression of

all PGs and their respective derivatives. These PGs have distinct

properties and therefore are likely to have diverse roles during

viral infection. For instance, treatment with the PGD2 derivative

Page 13: Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Interferon and Apoptosis in Macrophages

Immunity

PGE2 Inhibition Enhances Antiviral Immunity

Please cite this article in press as: Coulombe et al., Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Inter-feron and Apoptosis in Macrophages, Immunity (2014), http://dx.doi.org/10.1016/j.immuni.2014.02.013

15-deoxy-D12,14-PGJ2 reduces lung inflammation, decreases

viral load, and protects mice against lethal IAV infection (Cloutier

et al., 2012). Our study, alternatively, demonstrated that inhibi-

tion of PGE2 per se decreased pulmonary viral load, improved

lung function, and enhanced survival following IAV infection.

This highlights the importance of considering PGs as a group

of individual lipid mediators with distinct functions in antiviral

immunity.

Despite the worldwide application of vaccination and other

antiviral interventions, IAV infection remains a persistent threat

to human health especially due to the emergence of novel

pandemic strains. Following the increased popularity of aspirin

during the 1918 pandemic, one century of NSAID usage has

taught us that inhibition of COX and subsequently all PGs might

have beneficial effects for the management of IAV symptoms. It

is also important to consider that aspirin overuse might have

increased the severity of the 1918 IAV, and the use of salicylates

in children with IAV or other viral illnesses has also been associ-

ated with mortality due to the development of Reye’s syndrome

(Starko, 2009). However, we herein report that the selective inhi-

bition of PGE2 using a nonsalicylate compound that does not

inhibit COX activity enhances immunity and protects against

IAV infection. Preclinical studies in animals have identified a

number of mPGES-1 inhibitory compounds that are orally active

at relatively high doses in animal models of inflammatory disease

and lead to the development of selective humanmPGES-1 inhib-

itors such as MF63 (Xu et al., 2008). Thus, specific suppression

of PGE2 represents a feasible and immediate therapeutic avenue

for the treatment and prevention of influenza and potentially

other viral infections.

EXPERIMENTAL PROCEDURES

See the Supplemental Information for details.

Mice

Six- to ten-week-old C57BL/6 mice, OT-I T cell antigen receptor-transgenic

mice (Taconic 4175) and Rag1�/� mice (Jax 002096) were from the Jackson

Laboratory. Ptges�/�mice (Chen et al., 2008; Divangahi et al., 2009; Divangahi

et al., 2010) (backcrossed to the C57BL/6 background, kindly provided by

B. Koller, University of North Carolina) and Ifnar�/� mice on C57BL/6 back-

ground (kindly provided by J.H. Fritz) were bred at McGill University. Experi-

ments were conducted in accordance with the guidelines of animal research

ethics board of McGill University.

Viruses and Infections

All in vivo and in vitro infections were performed with mouse-adapted IAV

H1N1 strain A/Puerto Rico/8/34 (kindly provided by J.A. McCullers) except

for in vitro infection of human cells that were performed with the clinical strain

H3N2 A/Hong-Kong/1/68 (kindly provided by S. Qureshi). UV IAV for in vitro

stimulation was generated with the H1N1 strain. In vivo infections were

performed intranasally (25 ml), and the lethal dose of IAV H1N1 used for survival

experiments was 150–200 PFU unless indicated otherwise, whereas the

sublethal dose was 50 PFU. Viruses were propagated and isolated from

Madin-Darby canine kidney (MDCK) cells and titrated with standard plaque

assay in MDCK cells (Gaush and Smith, 1968).

Flow Cytometry Analysis

Single-cell suspensions obtained from BAL and collagenase- and DNase-

treated lungs; blood or LN were stained with different combinations of Abs

(Divangahi et al., 2009; Divangahi et al., 2010). Flow cytometry was performed

with BD LSR II (BDBiosciences) and analysis was performed with FlowJo Soft-

ware (Tree Star).

Quantitative RT-PCR

Total RNA was isolated from mouse and human cells using TRIzol reagent

(Invitrogen) and processed with Moloney murine leukemia virus reverse

transcriptase (QuantiTect Reverse Transcription Kit, QIAGEN) for mRNA anal-

ysis by qRT-PCR with Bio-Rad Icycler Software (Bio-Rad Laboratories).

Expression of mRNA was shown as fold induction of infected cells relative

to uninfected or unstimulated cells of the same genotype unless stated

otherwise.

Cell Death Assays

For in vitro experiments, apoptosis was measured by the cell-death detection

ELISAPLUS (Roche Applied Science) (Divangahi et al., 2009; Divangahi et al.,

2010). The amount of apoptotic bleb formation was also measured with the

Apoptotic Blebs Assay Kit (Cayman Chemical). For in vivo infections, necrosis

and apoptosis were quantified with the Apoptosis Detection Kit I (BD Biosci-

ences) and analyzed by flow cytometry.

Adoptive Transfer Models of Infection

AM were collected from the BAL of naive WT and Ptges�/� mice and sus-

pended macrophages were infected for 2 hr in vitro with 0.5 (Figure 3F) or 5

MOI (Figure 6C) of IAV H1N1 A/PR/8/34. Free viruses were then removed by

three washes with PBS, each followed by centrifugation for 10 min at 200g

and 4�C. Cells were resuspended in PBS at a density of 0.5 3 106 cells per

40 ml and then transferred by the i.t. route into naive Rag1�/� mice or WT

mice (Divangahi et al., 2009; Divangahi et al., 2010). For BMDMadoptive trans-

fer (Figure 3I), 1 3 106 uninfected WT or Ptges�/� cells were transferred (i.t.)

into naive WT mice, which were infected i.n. with IAV H1N1 A/PR/8/34 (600

PFU) 2 hr later.

Statistics

Data was analyzed with PRISM 6.0 Software (GraphPad Software). Student’s

t test and two-way ANOVA (when indicated) were performed except for sur-

vival experiments where data was analyzed by the log rank test. Differences

were considered significant at p < 0.05.

SUPPLEMENTAL INFORMATION

Supplemental Information includes Supplemental Experimental Procedures,

seven figures, and one table and can be found with this article online at

http://dx.doi.org/10.1016/j.immuni.2014.02.013.

ACKNOWLEDGMENTS

This work was supported by the CIHR operating grant (MOP-106488) to M.D.,

and M.D. holds CIHR New Investigator Award. F.C. is supported by a F. Bant-

ing and C. Best CGS from CIHR. The authors would like to thank E. Fixman for

helpful comments and valuable discussions.

Received: October 24, 2013

Accepted: February 25, 2014

Published: April 10, 2014

REFERENCES

Albert, M.L., Sauter, B., and Bhardwaj, N. (1998). Dendritic cells acquire anti-

gen from apoptotic cells and induce class I-restricted CTLs. Nature 392,

86–89.

Betz, M., and Fox, B.S. (1991). Prostaglandin E2 inhibits production of Th1

lymphokines but not of Th2 lymphokines. J. Immunol. 146, 108–113.

Braciale, T.J., Sun, J., and Kim, T.S. (2012). Regulating the adaptive immune

response to respiratory virus infection. Nat. Rev. Immunol. 12, 295–305.

Carey, M.A., Bradbury, J.A., Seubert, J.M., Langenbach, R., Zeldin, D.C., and

Germolec, D.R. (2005). Contrasting effects of cyclooxygenase-1 (COX-1) and

COX-2 deficiency on the host response to influenza A viral infection.

J. Immunol. 175, 6878–6884.

Chang, Y.J., Kim, H.Y., Albacker, L.A., Baumgarth, N., McKenzie, A.N., Smith,

D.E., Dekruyff, R.H., and Umetsu, D.T. (2011). Innate lymphoid cells mediate

Immunity 40, 1–15, April 17, 2014 ª2014 Elsevier Inc. 13

Page 14: Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Interferon and Apoptosis in Macrophages

Immunity

PGE2 Inhibition Enhances Antiviral Immunity

Please cite this article in press as: Coulombe et al., Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Inter-feron and Apoptosis in Macrophages, Immunity (2014), http://dx.doi.org/10.1016/j.immuni.2014.02.013

influenza-induced airway hyper-reactivity independently of adaptive immunity.

Nat. Immunol. 12, 631–638.

Chen, M., Divangahi, M., Gan, H., Shin, D.S., Hong, S., Lee, D.M., Serhan,

C.N., Behar, S.M., and Remold, H.G. (2008). Lipid mediators in innate immu-

nity against tuberculosis: opposing roles of PGE2 and LXA4 in the induction

of macrophage death. J. Exp. Med. 205, 2791–2801.

Cloutier, A., Marois, I., Cloutier, D., Verreault, C., Cantin, A.M., and Richter,

M.V. (2012). The prostanoid 15-deoxy-D12,14-prostaglandin-j2 reduces lung

inflammation and protects mice against lethal influenza infection. J. Infect.

Dis. 205, 621–630.

Dessing, M.C., van der Sluijs, K.F., Florquin, S., and van der Poll, T. (2007).

Monocyte chemoattractant protein 1 contributes to an adequate immune

response in influenza pneumonia. Clin. Immunol. 125, 328–336.

Divangahi, M., Chen, M., Gan, H., Desjardins, D., Hickman, T.T., Lee, D.M.,

Fortune, S., Behar, S.M., and Remold, H.G. (2009). Mycobacterium tubercu-

losis evades macrophage defenses by inhibiting plasma membrane repair.

Nat. Immunol. 10, 899–906.

Divangahi, M., Desjardins, D., Nunes-Alves, C., Remold, H.G., and Behar, S.M.

(2010). Eicosanoid pathways regulate adaptive immunity to Mycobacterium

tuberculosis. Nat. Immunol. 11, 751–758.

Ehrhardt, C., Wolff, T., Pleschka, S., Planz, O., Beermann, W., Bode, J.G.,

Schmolke, M., and Ludwig, S. (2007). Influenza A virus NS1 protein activates

the PI3K/Akt pathway to mediate antiapoptotic signaling responses. J. Virol.

81, 3058–3067.

Friesenhagen, J., Boergeling, Y., Hrincius, E., Ludwig, S., Roth, J., and

Viemann, D. (2012). Highly pathogenic avian influenza viruses inhibit effective

immune responses of human blood-derivedmacrophages. J. Leukoc. Biol. 92,

11–20.

Fukao, T., and Koyasu, S. (2003). PI3K and negative regulation of TLR

signaling. Trends Immunol. 24, 358–363.

Gaush, C.R., and Smith, T.F. (1968). Replication and plaque assay of influ-

enza virus in an established line of canine kidney cells. Appl. Microbiol. 16,

588–594.

Hara, S., Kamei, D., Sasaki, Y., Tanemoto, A., Nakatani, Y., and Murakami, M.

(2010). Prostaglandin E synthases: Understanding their pathophysiological

roles through mouse genetic models. Biochimie 92, 651–659.

Honke, N., Shaabani, N., Cadeddu, G., Sorg, U.R., Zhang, D.E., Trilling, M.,

Klingel, K., Sauter, M., Kandolf, R., Gailus, N., et al. (2012). Enforced viral repli-

cation activates adaptive immunity and is essential for the control of a cyto-

pathic virus. Nat. Immunol. 13, 51–57.

Hou,W., Gibbs, J.S., Lu, X., Brooke, C.B., Roy, D., Modlin, R.L., Bennink, J.R.,

and Yewdell, J.W. (2012). Viral infection triggers rapid differentiation of human

blood monocytes into dendritic cells. Blood 119, 3128–3131.

Huang, F.F., Barnes, P.F., Feng, Y., Donis, R., Chroneos, Z.C., Idell, S., Allen,

T., Perez, D.R., Whitsett, J.A., Dunussi-Joannopoulos, K., and Shams, H.

(2011). GM-CSF in the lung protects against lethal influenza infection. Am. J.

Respir. Crit. Care Med. 184, 259–268.

Joung, S.M., Park, Z.Y., Rani, S., Takeuchi, O., Akira, S., and Lee, J.Y. (2011).

Akt contributes to activation of the TRIF-dependent signaling pathways of

TLRs by interacting with TANK-binding kinase 1. J. Immunol. 186, 499–507.

Kim, H.M., Lee, Y.W., Lee, K.J., Kim, H.S., Cho, S.W., van Rooijen, N., Guan,

Y., and Seo, S.H. (2008). Alveolar macrophages are indispensable for control-

ling influenza viruses in lungs of pigs. J. Virol. 82, 4265–4274.

Kirby, A.C., Coles, M.C., and Kaye, P.M. (2009). Alveolar macrophages trans-

port pathogens to lung draining lymph nodes. J. Immunol. 183, 1983–1989.

Kochs, G., Garcıa-Sastre, A., and Martınez-Sobrido, L. (2007). Multiple anti-

interferon actions of the influenza A virus NS1 protein. J. Virol. 81, 7011–7021.

Koerner, I., Kochs, G., Kalinke, U., Weiss, S., and Staeheli, P. (2007).

Protective role of beta interferon in host defense against influenza A virus.

J. Virol. 81, 2025–2030.

Landsman, L., and Jung, S. (2007). Lung macrophages serve as obligatory

intermediate between blood monocytes and alveolar macrophages.

J. Immunol. 179, 3488–3494.

14 Immunity 40, 1–15, April 17, 2014 ª2014 Elsevier Inc.

Lauder, S.N., Taylor, P.R., Clark, S.R., Evans, R.L., Hindley, J.P., Smart, K.,

Leach, H., Kidd, E.J., Broadley, K.J., Jones, S.A., et al. (2011). Paracetamol

reduces influenza-induced immunopathology in a mouse model of infection

without compromising virus clearance or the generation of protective immu-

nity. Thorax 66, 368–374.

Le Bon, A., and Tough, D.F. (2008). Type I interferon as a stimulus for cross-

priming. Cytokine Growth Factor Rev. 19, 33–40.

Lee, S.M., Cheung, C.Y., Nicholls, J.M., Hui, K.P., Leung, C.Y., Uiprasertkul,

M., Tipoe, G.L., Lau, Y.L., Poon, L.L., Ip, N.Y., et al. (2008). Hyperinduction

of cyclooxygenase-2-mediated proinflammatory cascade: a mechanism

for the pathogenesis of avian influenza H5N1 infection. J. Infect. Dis. 198,

525–535.

Lee, S.M., Dutry, I., and Peiris, J.S. (2012). Editorial: macrophage heterogene-

ity and responses to influenza virus infection. J. Leukoc. Biol. 92, 1–4.

McCarthy, M.K., and Weinberg, J.B. (2012). Eicosanoids and respiratory viral

infection: coordinators of inflammation and potential therapeutic targets.

Mediators Inflamm. 2012, 236345.

Mok, C.K., Lee, D.C., Cheung, C.Y., Peiris,M., and Lau, A.S. (2007). Differential

onset of apoptosis in influenza A virus H5N1- and H1N1-infected human blood

macrophages. J. Gen. Virol. 88, 1275–1280.

Nakamura, S., Davis, K.M., and Weiser, J.N. (2011). Synergistic stimulation of

type I interferons during influenza virus coinfection promotes Streptococcus

pneumoniae colonization in mice. J. Clin. Invest. 121, 3657–3665.

Piqueras, B., Connolly, J., Freitas, H., Palucka, A.K., and Banchereau, J.

(2006). Upon viral exposure, myeloid and plasmacytoid dendritic cells produce

3 waves of distinct chemokines to recruit immune effectors. Blood 107, 2613–

2618.

Regan, J.W. (2003). EP2 and EP4 prostanoid receptor signaling. Life Sci. 74,

143–153.

Ricciotti, E., and FitzGerald, G.A. (2011). Prostaglandins and inflammation.

Arterioscler. Thromb. Vasc. Biol. 31, 986–1000.

Sarafi, M.N., Garcia-Zepeda, E.A., MacLean, J.A., Charo, I.F., and Luster, A.D.

(1997). Murine monocyte chemoattractant protein (MCP)-5: a novel CC che-

mokine that is a structural and functional homologue of human MCP-1.

J. Exp. Med. 185, 99–109.

Schmidt, L.M., Belvisi, M.G., Bode, K.A., Bauer, J., Schmidt, C., Suchy, M.T.,

Tsikas, D., Scheuerer, J., Lasitschka, F., Grone, H.J., and Dalpke, A.H. (2011).

Bronchial epithelial cell-derived prostaglandin E2 dampens the reactivity of

dendritic cells. J. Immunol. 186, 2095–2105.

Shahangian, A., Chow, E.K., Tian, X., Kang, J.R., Ghaffari, A., Liu, S.Y.,

Belperio, J.A., Cheng, G., and Deng, J.C. (2009). Type I IFNs mediate develop-

ment of postinfluenza bacterial pneumonia in mice. J. Clin. Invest. 119, 1910–

1920.

Shirey, K.A., Pletneva, L.M., Puche, A.C., Keegan, A.D., Prince, G.A., Blanco,

J.C., and Vogel, S.N. (2010). Control of RSV-induced lung injury by alterna-

tively activated macrophages is IL-4R alpha-, TLR4-, and IFN-beta-depen-

dent. Mucosal Immunol. 3, 291–300.

Smyth, E.M., Grosser, T., Wang, M., Yu, Y., and FitzGerald, G.A. (2009).

Prostanoids in health and disease. J. Lipid Res. Suppl. 50, S423–S428.

Stanley, S.A., Johndrow, J.E., Manzanillo, P., and Cox, J.S. (2007). The Type I

IFN response to infection with Mycobacterium tuberculosis requires ESX-

1-mediated secretion and contributes to pathogenesis. J. Immunol. 178,

3143–3152.

Starko, K.M. (2009). Salicylates and pandemic influenza mortality, 1918-1919

pharmacology, pathology, and historic evidence. Clinical infectious diseases:

an official publication of the Infectious Diseases Society of America 49, 1405–

1410.

Trebino, C.E., Eskra, J.D., Wachtmann, T.S., Perez, J.R., Carty, T.J., and

Audoly, L.P. (2005). Redirection of eicosanoid metabolism in mPGES-1-defi-

cient macrophages. J. Biol. Chem. 280, 16579–16585.

Wareing, M.D., Lyon, A., Inglis, C., Giannoni, F., Charo, I., and Sarawar, S.R.

(2007). Chemokine regulation of the inflammatory response to a low-dose

influenza infection in CCR2-/- mice. J. Leukoc. Biol. 81, 793–801.

Page 15: Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Interferon and Apoptosis in Macrophages

Immunity

PGE2 Inhibition Enhances Antiviral Immunity

Please cite this article in press as: Coulombe et al., Targeted Prostaglandin E2 Inhibition Enhances Antiviral Immunity through Induction of Type I Inter-feron and Apoptosis in Macrophages, Immunity (2014), http://dx.doi.org/10.1016/j.immuni.2014.02.013

Xing, Z., Cardona, C.J., Adams, S., Yang, Z., Li, J., Perez, D., and Woolcock,

P.R. (2009). Differential regulation of antiviral and proinflammatory cytokines

and suppression of Fas-mediated apoptosis by NS1 of H9N2 avian influenza

virus in chicken macrophages. J. Gen. Virol. 90, 1109–1118.

Xu, D., Rowland, S.E., Clark, P., Giroux, A., Cote, B., Guiral, S., Salem, M.,

Ducharme, Y., Friesen, R.W., Methot, N., et al. (2008). MF63 [2-(6-chloro-

1H-phenanthro[9,10-d]imidazol-2-yl)-isophthalonitrile], a selective micro-

somal prostaglandin E synthase-1 inhibitor, relieves pyresis and pain in

preclinical models of inflammation. J. Pharmacol. Exp. Ther. 326, 754–763.

Zaslona, Z., Serezani, C.H., Okunishi, K., Aronoff, D.M., and Peters-Golden,M.

(2012). Prostaglandin E2 restrains macrophage maturation via E prostanoid

receptor 2/protein kinase A signaling. Blood 119, 2358–2367.

Immunity 40, 1–15, April 17, 2014 ª2014 Elsevier Inc. 15


Recommended