host-directed therapy of tuberculosis based on interleukin-1 ......antagonize the il-1 pathway...

17
LETTER doi:10.1038/nature13489 Host-directed therapy of tuberculosis based on interleukin-1 and type I interferon crosstalk Katrin D. Mayer-Barber 1 , Bruno B. Andrade 1 , Sandra D. Oland 1 , Eduardo P. Amaral 1,2 , Daniel L. Barber 3 , Jacqueline Gonzales 4 , Steven C. Derrick 5 , Ruiru Shi 6 , Nathella Pavan Kumar 7,8 , Wang Wei 6 , Xing Yuan 6 , Guolong Zhang 9 , Ying Cai 4 , Subash Babu 7,10 , Marta Catalfamo 11 , Andres M. Salazar 12 , Laura E. Via 4 , Clifton E. Barry III 4 & Alan Sher 1 Tuberculosis remains second only to HIV/AIDS as the leading cause of mortality worldwide due to a single infectious agent 1 . Despite chemo- therapy, the global tuberculosis epidemic has intensified because of HIV co-infection, the lack of an effective vaccine and the emergence of multi-drug-resistant bacteria 2–5 . Alternative host-directed strategies could be exploited to improve treatment efficacy and outcome, con- tain drug-resistant strains and reduce disease severity and mortality 6 . The innate inflammatory response elicited by Mycobacterium tuber- culosis (Mtb) represents a logical host target 7 . Here we demonstrate that interleukin-1 (IL-1) confers host resistance through the induc- tion of eicosanoids that limit excessive type I interferon (IFN) pro- duction and foster bacterial containment. We further show that, in infected mice and patients, reduced IL-1 responses and/or excessive type I IFN induction are linked to an eicosanoid imbalance assoc- iated with disease exacerbation. Host-directed immunotherapy with clinically approved drugs that augment prostaglandin E2 levels in these settings prevented acute mortality of Mtb-infected mice. Thus, IL-1 and type I IFNs represent two major counter-regulatory classes of inflammatory cytokines that control the outcome of Mtb infection and are functionally linked via eicosanoids. Our findings establish proof of concept for host-directed treatment strategies that manip- ulate the host eicosanoid network and represent feasible alternatives to conventional chemotherapy. The mechanismsby which IL-1-driven inflammation promotes host resistance against Mtb in vivo have not been elucidated and offer novel targets for immunological intervention in tuberculosis (TB). Despite the high susceptibility of IL-1-deficient mice to Mtb infection, estab- lished mediators of host resistance, such as interferon (IFN)-c, tumour necrosis factor (TNF)-a, inducible nitric oxide synthase (iNOS) or IL- 12p40 were not diminished in the absence of IL1R1 when assessed in vitro, in vivo at the single-cell level or in bronchoalveolar lavage fluid (BALF) (Extended Data Fig. 1a–c) 8,9 . Instead, we observed that increased bacterial loads in BALF were accompanied by an eicosanoid imbalance (Fig. 1a, b, Extended Data Fig. 1d). Eicosanoids are lipid mediators derived from arachidonic acid that include prostaglandins, resolvins, lipoxins and leukotrienes 10 . Cyclooxygenase-2 (COX-2, gene also known as Ptgs2) and 5-lipoxygenase (5-LO, gene also known as Alox5) or 12/ 15-lipoxygenase (12/15-LO) compete for arachidonic acid to generate COX-dependent prostaglandins or LO-dependent lipoxins and leuko- trienes. Alox5 2/2 mice have been reported to be more resistant and prostaglandin E synthase (Ptges)-deficient mice more susceptible to Mtb infection 11–13 . In the absence of IL-1 signalling, we found that pros- taglandin E2 (PGE2) and prostaglandin F2a (PGF2a) were signifi- cantly reduced in BALF of Mtb-infected Il1r1 2/2 mice, whereas the 5-LO products LXA4 and LTB4 were increased compared to wild-type animals (Fig. 1b, Extended Data Fig. 1d). We then purified myeloid cells from infected wild-type lungs by fluorescence-activated cell sorting (FACS), and found that they pro- duced large amounts of PGE2 ex vivo (Extended Data Fig. 1e). We next used mixed bone marrow chimaeras to analyse the requirement for IL1R1 on pulmonary CD68 pos myeloid cells for COX-2 production in vivo. Il1r1 2/2 cells expressed significantly less COX-2 protein, indi- cating that IL-1 acts directly on myeloid cells for optimal COX-2 induc- tion (Extended Data Fig. 1b, f) independently of pulmonary bacterial loads. In vitro, Mtb-infected Il1r1 2/2 bone-marrow-derived macro- phages (BMDM) failed to control bacterial growth compared to wild-type BMDM (Fig. 1c) and the increased colony forming units in Il1r1 2/2 BMDM or bone-marrow dendritic cell cultures were associated with decreased PGE2 (Extended Data Fig. 2a, b). In addition, we observed a significant increase in both extracellular bacterial as well as infected macrophage numbers in Il1a,Il1b 2/2 cultures (Extended Data Fig. 2c, d). Add-back of exogenous IL-1a, IL-1b or PGE2 reduced extracellular bacteria to wild-type levels (Fig. 1d, Extended Data Fig. 2e–g) and re- stored control of bacterial growth in IL-1-deficient BMDM (Fig. 1e and Extended Data Fig. 3a). IL-1-dependent bacterial control was lost when COX-2 was inhib- ited with valdecoxib and restored upon PGE2 addition (Fig. 1e and Extended Data Fig. 3b). Of note, valdecoxib increased bacterial loads and inhibited PGE2 synthesis in infected wild-type BMDM as was ob- served in BMDM derived from B6 mice deficient in the enzymatic activity of COX-2 (Ptgs2(Y385F) mice) (Extended Data Fig. 3c). In vivo, absence of COX-2 enzymatic activity did not affect pulmonary and serum levels of PGE2 or lung bacterial loads in Ptgs2(Y385F) mice at 4 weeks after Mtb infection, consistent with a compensatory role for COX-1, which can generate PGE2 when COX-2 is absent 14 (Fig. 1f, Extended Data Fig. 3d, e). Nonetheless, at later stages of Mtb infection, Ptgs2(Y385F) mice exhibited increased susceptibility compared to wild-type animals (Fig. 1g). We next examined whether IL-1 also drives PGE2 synthesis in infected primary human monocyte-derived macrophages (MDM). Blockade of IL1R1 signalling resulted in reduced PGE2 levels, whereas addition of exogenous IL-1a or IL-1b further increased PGE2 synthesis during Mtb infection (Fig. 1h). Although additional PGE2-independ- ent mechanisms of IL-1-mediated protection are likely to exist, our data demonstrate in vitro and in vivo that IL-1a and IL-1b, acting partially through COX-2, trigger PGE2 synthesis, which in turn reg- ulates the containment and replication of bacilli during Mtb infection. In many settings type I IFNs promote bacterial virulence and disease exacerbation 15,16 . Hypervirulence of certain Mtb strains correlates with 1 Immunobiology Section, Laboratory of Parasitic Diseases (LPD), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA. 2 Department of Immunology, Biomedical Sciences Institutes, University of Sao Paulo, 05508-900 Sao Paulo, Brazil. 3 T Lymphocyte Biology Unit, LPD, NIAID, NIH, Bethesda, Maryland 20892, USA. 4 Tuberculosis Research Section, Laboratory of Clinical Infectious Disease, NIAID, NIH, Bethesda, Maryland 20892, USA. 5 Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland 20892, USA. 6 Henan Chest Hospital, 450003 Zhengzhou, China. 7 NIH, International Center for Excellence in Research, 600 031 Chennai, India. 8 National Institute for Research in Tuberculosis (NIRT), 600 031 Chennai, India. 9 Sino-US International Research Center for Tuberculosis, and Henan Public Health Center, 450003 Zhengzhou, China. 10 Helminth Immunology Section, LPD, NIAID, NIH, Bethesda, Maryland 20892, USA. 11 Clinical and Molecular Retrovirology Section, Laboratory of Immunoregulation, NIAID, NIH, Bethesda, Maryland 20892, USA. 12 Oncovir Inc., Washington, Washington DC 20008, USA. 00 MONTH 2014 | VOL 000 | NATURE | 1 Macmillan Publishers Limited. All rights reserved ©2014

Upload: others

Post on 07-Nov-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Host-directed therapy of tuberculosis based on interleukin-1 ......antagonize the IL-1 pathway during Mtb infection8, absence of type I IFN signalling resulted in increased PGE2 and

LETTERdoi:10.1038/nature13489

Host-directed therapy of tuberculosis based oninterleukin-1 and type I interferon crosstalkKatrin D. Mayer-Barber1, Bruno B. Andrade1, Sandra D. Oland1, Eduardo P. Amaral1,2, Daniel L. Barber3, Jacqueline Gonzales4,Steven C. Derrick5, Ruiru Shi6, Nathella Pavan Kumar7,8, Wang Wei6, Xing Yuan6, Guolong Zhang9, Ying Cai4, Subash Babu7,10,Marta Catalfamo11, Andres M. Salazar12, Laura E. Via4, Clifton E. Barry III4 & Alan Sher1

Tuberculosis remains second only to HIV/AIDS as the leading causeof mortality worldwide due to a single infectious agent1. Despite chemo-therapy, the global tuberculosis epidemic has intensified because ofHIV co-infection, the lack of an effective vaccine and the emergenceof multi-drug-resistant bacteria2–5. Alternative host-directed strategiescould be exploited to improve treatment efficacy and outcome, con-tain drug-resistant strains and reduce disease severity and mortality6.The innate inflammatory response elicited by Mycobacterium tuber-culosis (Mtb) represents a logical host target7. Here we demonstratethat interleukin-1 (IL-1) confers host resistance through the induc-tion of eicosanoids that limit excessive type I interferon (IFN) pro-duction and foster bacterial containment. We further show that, ininfected mice and patients, reduced IL-1 responses and/or excessivetype I IFN induction are linked to an eicosanoid imbalance assoc-iated with disease exacerbation. Host-directed immunotherapy withclinically approved drugs that augment prostaglandin E2 levels inthese settings prevented acute mortality of Mtb-infected mice. Thus,IL-1 and type I IFNs represent two major counter-regulatory classes ofinflammatory cytokines that control the outcome of Mtb infectionand are functionally linked via eicosanoids. Our findings establishproof of concept for host-directed treatment strategies that manip-ulate the host eicosanoid network and represent feasible alternativesto conventional chemotherapy.

The mechanisms by which IL-1-driven inflammation promotes hostresistance against Mtb in vivo have not been elucidated and offer noveltargets for immunological intervention in tuberculosis (TB). Despitethe high susceptibility of IL-1-deficient mice to Mtb infection, estab-lished mediators of host resistance, such as interferon (IFN)-c, tumournecrosis factor (TNF)-a, inducible nitric oxide synthase (iNOS) or IL-12p40 were not diminished in the absence of IL1R1 when assessedin vitro, in vivo at the single-cell level or in bronchoalveolar lavage fluid(BALF) (Extended Data Fig. 1a–c)8,9. Instead, we observed that increasedbacterial loads in BALF were accompanied by an eicosanoid imbalance(Fig. 1a, b, Extended Data Fig. 1d). Eicosanoids are lipid mediatorsderived from arachidonic acid that include prostaglandins, resolvins,lipoxins and leukotrienes10. Cyclooxygenase-2 (COX-2, gene also knownas Ptgs2) and 5-lipoxygenase (5-LO, gene also known as Alox5) or 12/15-lipoxygenase (12/15-LO) compete for arachidonic acid to generateCOX-dependent prostaglandins or LO-dependent lipoxins and leuko-trienes. Alox52/2 mice have been reported to be more resistant andprostaglandin E synthase (Ptges)-deficient mice more susceptible to Mtbinfection11–13. In the absence of IL-1 signalling, we found that pros-taglandin E2 (PGE2) and prostaglandin F2a (PGF2a) were signifi-cantly reduced in BALF of Mtb-infected Il1r12/2 mice, whereas the

5-LO products LXA4 and LTB4 were increased compared to wild-typeanimals (Fig. 1b, Extended Data Fig. 1d).

We then purified myeloid cells from infected wild-type lungs byfluorescence-activated cell sorting (FACS), and found that they pro-duced large amounts of PGE2 ex vivo (Extended Data Fig. 1e). We nextused mixed bone marrow chimaeras to analyse the requirement forIL1R1 on pulmonary CD68pos myeloid cells for COX-2 productionin vivo. Il1r12/2 cells expressed significantly less COX-2 protein, indi-cating that IL-1 acts directly on myeloid cells for optimal COX-2 induc-tion (Extended Data Fig. 1b, f) independently of pulmonary bacterialloads. In vitro, Mtb-infected Il1r12/2 bone-marrow-derived macro-phages (BMDM) failed to control bacterial growth compared to wild-typeBMDM (Fig. 1c) and the increased colony forming units in Il1r12/2

BMDM or bone-marrow dendritic cell cultures were associated withdecreased PGE2 (Extended Data Fig. 2a, b). In addition, we observed asignificant increase in both extracellular bacterial as well as infectedmacrophage numbers in Il1a,Il1b2/2 cultures (Extended Data Fig. 2c, d).Add-back of exogenous IL-1a, IL-1b or PGE2 reduced extracellularbacteria to wild-type levels (Fig. 1d, Extended Data Fig. 2e–g) and re-stored control of bacterial growth in IL-1-deficient BMDM (Fig. 1e andExtended Data Fig. 3a).

IL-1-dependent bacterial control was lost when COX-2 was inhib-ited with valdecoxib and restored upon PGE2 addition (Fig. 1e andExtended Data Fig. 3b). Of note, valdecoxib increased bacterial loadsand inhibited PGE2 synthesis in infected wild-type BMDM as was ob-served in BMDM derived from B6 mice deficient in the enzymatic activityof COX-2 (Ptgs2(Y385F) mice) (Extended Data Fig. 3c). In vivo, absenceof COX-2 enzymatic activity did not affect pulmonary and serum levelsof PGE2 or lung bacterial loads in Ptgs2(Y385F) mice at 4 weeks afterMtb infection, consistent with a compensatory role for COX-1, whichcan generate PGE2 when COX-2 is absent14 (Fig. 1f, Extended DataFig. 3d, e). Nonetheless, at later stages of Mtb infection, Ptgs2(Y385F)mice exhibited increased susceptibility compared to wild-type animals(Fig. 1g). We next examined whether IL-1 also drives PGE2 synthesisin infected primary human monocyte-derived macrophages (MDM).Blockade of IL1R1 signalling resulted in reduced PGE2 levels, whereasaddition of exogenous IL-1a or IL-1b further increased PGE2 synthesisduring Mtb infection (Fig. 1h). Although additional PGE2-independ-ent mechanisms of IL-1-mediated protection are likely to exist, ourdata demonstrate in vitro and in vivo that IL-1a and IL-1b, actingpartially through COX-2, trigger PGE2 synthesis, which in turn reg-ulates the containment and replication of bacilli during Mtb infection.

In many settings type I IFNs promote bacterial virulence and diseaseexacerbation15,16. Hypervirulence of certain Mtb strains correlates with

1Immunobiology Section, Laboratory of Parasitic Diseases (LPD), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA.2Department of Immunology, Biomedical Sciences Institutes, University of Sao Paulo, 05508-900 Sao Paulo, Brazil. 3T Lymphocyte Biology Unit, LPD, NIAID, NIH, Bethesda, Maryland 20892, USA.4Tuberculosis Research Section, Laboratory of Clinical Infectious Disease, NIAID, NIH, Bethesda, Maryland 20892, USA. 5Center for Biologics Evaluation and Research, Food and Drug Administration,Bethesda, Maryland 20892, USA. 6Henan Chest Hospital, 450003 Zhengzhou, China. 7NIH, International Center for Excellence in Research, 600 031 Chennai, India. 8National Institute for Research inTuberculosis (NIRT), 600 031 Chennai, India. 9Sino-US International Research Center for Tuberculosis, and Henan Public Health Center, 450003 Zhengzhou, China. 10Helminth Immunology Section, LPD,NIAID, NIH, Bethesda, Maryland 20892, USA. 11Clinical and Molecular Retrovirology Section, Laboratory of Immunoregulation, NIAID, NIH, Bethesda, Maryland 20892, USA. 12Oncovir Inc., Washington,Washington DC 20008, USA.

0 0 M O N T H 2 0 1 4 | V O L 0 0 0 | N A T U R E | 1

Macmillan Publishers Limited. All rights reserved©2014

Page 2: Host-directed therapy of tuberculosis based on interleukin-1 ......antagonize the IL-1 pathway during Mtb infection8, absence of type I IFN signalling resulted in increased PGE2 and

enhanced necrosis, type I IFN production and Mtb-infected Ifnar12/2

mice harbour fewer bacteria compared to wild-type animals8,17–19. TypeI IFNs subvert anti-tuberculous host defences by inhibiting iNOS,

IL-12p40, IL-1a and IL-1b production, while inducing the immuno-suppressive mediators IL-10 and IL1R antagonist (IL1Ra, gene also knownas Il1rn)8,19. Consistent with our previous findings that type I IFNs

***

***

PG

E2 (ng

ml–

1)

2

3

4

5

6

BA

LF

, c.f

.u.

(lo

g10)

WT

Il1r1–/

Il1a–/

Il1b–/

*** ***

***

***

a

******

******

b

rhIL-1αrhIL-1β

Mtb

Anti-hIL1R1

PG

E2 (p

g m

l–1)

101

102

103

n.s.

******

n = 23

0

1.5

3.0

4.5

rmIL-1 α+β –––

–––

+––

–+–

+–+

– – + ++ +– – – +– –– – – –– +– + – –+ –

PGE2Valdecoxib

n.s.6.0

***

*** ***

***

e

0.8

1.0

1.2

1.4

0.6

0.4

0.2

0

h

c.f

.u.

(10

6)

Il1a,I

l1b–/

WT

Il1r1–/

Il1a–/

Il1b–/

Il1a,I

l1b–/

– 5 d

n.s.

**c

0

15

3

6

9

12Il1r1–/–WT

6

7

8

9

10

n.s.

***

g***

Day 2

8,

lung

,

c.f

.u.

(lo

g10)

WT

Il1r1–/

Ptgs2

(Y38

5F)

Il1r1–/–

Ptgs2 (Y385F)

WT

Surv

ival (%

)

Time post infection (days)

250

200

150

100500

300

0

20

40

60

80

100

f

PGE2 –+– +

Extr

acellu

lar

Mtb

(fo

ld incre

ase o

ver

4 h

) **

0

1

2

3

4

5

6

*

***d

PG

E2/L

XA

4

1

10

0.1

*

Il1r1–/

WT

P = 0.0328*

Day 25 BALF day 25

c.f

.u.

(10

6)

4 h

Figure 1 | IL-1 triggers PGE2 synthesis during Mtb infection. a, Bacterialloads (colony forming units, c.f.u.); b, PGE2 concentrations and PGE2/LXAratio 25 days after aerosol exposure to Mtb in BALF of C57BL6 wild type (WT),Il1r12/2, Il1a,Il1b2/2, Il1b2/2 or Il1a2/2 mice (n 5 4–8). c, c.f.u. in Mtb-infected (m.o.i. 5 3) WT or Il1r12/2 BMDM cultures. d, Mtb-RFP-infected(m.o.i. 5 3) WT and Il1a,Il1b2/2 BMDMs cultured with or without PGE2.Extracellular bacteria were analysed by FACS (day 5). e, c.f.u. after in vitroinfection (m.o.i. 5 3) of WT or Il1a,Il1b2/2 BMDM in the presence or absenceof IL1, PGE2 or the COX-2 inhibitor valdecoxib. rmIL-1a, recombinant murineIL-1a. f, Lung c.f.u. of 4-week Mtb-infected WT, Il1r12/2 or Ptgs2(Y385F)

mice (n 5 5). a–f, Data are representative of two independent experiments.*P # 0.05; **P , 0.005; ***P , 0.0005; compared as indicatedin figure by lines or to WT control groups. Error bars denote s.d. (Mann–Whitney test). g, Survival of WT, Il1r12/2 or Ptgs2(Y385F) animals afteraerosol challenge with 25–40 c.f.u. Data shown are from one experiment (n 5 4,Mantel–Cox test). h, PGE2 concentration in supernatants of MDM from 23healthy donors, 24 h after Mtb infection in the presence or absence of rhIL-1a,rhIL-1b or anti-hIL1R1 neutralizing antibody. rhIL-1a, recombinanthuman IL-1a; anti-hIL1R1, anti-human IL-1R1. Differences were comparedas indicated by lines (Wilcoxon matched pairs test).

***

10

20

30

40

50

10

20

30

40

50

50

WT

Ifnb

mR

NA

(AU

, re

lative t

o H

PR

T)

Lung day 29

100

20

40

60

80

0

*

0

***

ec

WT

2.5

2.0

1.5

1.0

0.5

3.0

0

***

**

PGE2

(ng ml–1)

1

Mtb (m.o.i. = 5)

0

5

10

15

20

25

30

10 100

500

1,00

0

2,00

0

IFN

-β (p

g m

l–1)

0.00

0.05

0.10

0.15

0.20

0.25

***

f

g

PG

E2 (ng

ml–

1 B

ALF

)

WT

Ifnar

1–/

Il1r1–/

a

0rmIL-1αrmIL-1β

0 10 20 30 40 50 60 70 800

20

40

60

80

100

P = 0.0462*

Ifnar1–/–

WT

Il1r1, Ifnar1–/–

Il1r1–/–

3

6

9

12

0

PGE2Anti-Ifnar1

*** ******j k

Time post infection (days)

n = 7

0

2

4

6

8

IFN

-β (

IU m

l–1)

PGE2

WT Il1a, Il1b–/–

2.0

1.5

1.0

0.5

0

PG

E2 (ng

ml–

1)

rmIFN-β + –+– +

+–– +

+–– –––– +––– –––– –

–++–

–+–+

––

––

+–

–+

+–

++–

+––

– –

b

******

***

Mtb (m.o.i. = 5)

Mtb (m.o.i. = 5)

h

0

200

400

600

800

Mtb

rhIFN-β

PG

E2 (ng

ml–

1)

n = 22

P = 0.0002

P = 0.0005

***

P = 0.04 *

0

Il1r1–/

– WT

Il1r1–/

Il1a,I

l1b–/

***

d

rmIL-1α, IL-1βrmIFN-β

PGE2

Anti-Ifnar1

0

2

4

6

8

10

18

d0 d5

n.s.

i* *

*

*** ***IFN

-β (p

g m

l–1)

IFN

-β (p

g m

l–1)

IFN

-β (p

g m

l–1)

Mtb (m.o.i. = 5)

c.f

.u. (1

06)

c.f

.u. (1

06)

Surv

ival (%

)

Figure 2 | IL-1 and PGE2 negatively regulate type I IFNs. a, b, PGE2measured in BALF of indicated mouse strains 4 weeks post infection (p.i.)(n 5 5) (a) and WT or Il1a,Il1b2/2 BMDM infected for 40 h (m.o.i. 5 5) or inthe presence or absence of recombinant murine IFN-b (rmIFNb; b). c, PGE2 insupernatants of MDM from 22 healthy donors, 24 h after Mtb infection(m.o.i. 5 5) in the presence or absence of rhIFN-b (Wilcoxon matched pairstest). d, Ifnb mRNA expression and IFN-b protein in lungs of Mtb-infected WTor Il1r12/2 mice at indicated time points. AU, arbitrary units (n 5 9). e, f, IFN-b protein in supernatants of WT or Il1a,Il1b2/2 BMDM infected for 40 h (e) orWT BMDM in the presence or absence of rmIL-1a or IL-1b (f). g, IFN-bprotein concentration in supernatants WT BMDM (40 h p.i. Mtb) treated withincreasing concentrations of PGE2. h, IFN-b protein in supernatants of

MDM from 7 healthy donors, 24 h after Mtb infection with or without PGE2(Wilcoxon matched pairs test). i, c.f.u. at indicated time points of Mtb-infected(m.o.i. 5 5) WT BMDM cultures treated with anti-IFNAR1 monoclonalantibody, rmIL-1a or Il-1b, rmIFN-b or PGE2. j, c.f.u. after 5 days ofMtb infection (m.o.i. 5 5) of Il1r12/2 BMDM treated with anti-mIFNAR1mAb or PGE2. k, Survival of WT, Il1r12/2, Ifnar12/2 or Il1r1,Ifnar12/2

double-deficient animals after Mtb infection (n 5 5, Mantel–Cox test). Datashown are representative of two (a, b, d–g, i, k) or three (j) independentexperiments. *P # 0.05; **P , 0.005; ***P , 0.0005; compared as indicated infigure by lines or to WT control groups. Error bars denote s.d. (Mann–Whitneytest).

RESEARCH LETTER

2 | N A T U R E | V O L 0 0 0 | 0 0 M O N T H 2 0 1 4

Macmillan Publishers Limited. All rights reserved©2014

Page 3: Host-directed therapy of tuberculosis based on interleukin-1 ......antagonize the IL-1 pathway during Mtb infection8, absence of type I IFN signalling resulted in increased PGE2 and

antagonize the IL-1 pathway during Mtb infection8, absence of type IIFN signalling resulted in increased PGE2 and IL-1b and decreasedIL1Ra in BALF (Fig. 2a and Extended Data Fig. 4a). Mtb-infected wild-type BMDM produced significantly less PGE2 when exogenous IFN-bwas present (Fig. 2b). Addition of IFN-b to infected human MDM alsosignificantly reduced PGE2 (Fig. 2c, Extended Data Fig. 4b), yet supple-mentation of IFN-b-treated cultures with IL-1 failed to restore PGE2levels (Extended Data Fig. 4c). Together these observations suggested that,in addition to IL-1 itself8 and even in the presence of exogenous IL-1,type I IFNs potently counter-regulate the prostaglandin axis in Mtb-infected human and murine cells.

We next asked whether in the opposing direction IL-1 and PGE2inhibit the type I IFN pathway, which could represent an importantmechanism of IL-1/PGE2-dependent bacterial control. Indeed, in Mtb-infected Il1r12/2 mice pulmonary messenger RNA and protein levelsof IFN-bwere significantly elevated compared to wild-type mice (Fig. 2d).Mtb-infected Il1a,Il1b2/2 BMDM also expressed significantly elevatedIfna, Ifnb, IFN-inducible Il1rn (IL1Ra) and Il1r2 decoy receptor mRNAas well as IFN-bprotein (Extended Data Fig. 4d and Fig. 2e). ExogenousIL-1a and IL-1b inhibited IFN-b protein in wild-type macrophagesand reduced mRNA expression in Il1a,Il1b2/2 BMDM (Fig. 2f, ExtendedData Fig. 4e). Importantly, exogenous PGE2 also suppressed type I IFNproduction in Mtb-infected murine and human macrophages (Fig. 2g,h). Addition of either IL-1 or PGE2 to IFN-b-treated BMDM culturesreversed IFN-b-dependent loss of bacterial control (Fig. 2i). IL-1 failedto reverse PGE2 inhibition by type I IFNs, yet fully restored bacterial con-trol, suggesting the existence of other IL-1-driven anti-mycobacterialmechanisms in addition to PGE2 regulation.

To investigate if the IL-1-type I IFN counter-regulatory pathway hasan important function in IL-1-dependent bacterial control, we neutral-ized type I IFN in infected Il1r12/2 BMDM. Blocking type I IFN sig-nalling significantly reduced bacterial growth in Il1r12/2 macrophages(Fig. 2j). Importantly, mice doubly deficient in both IL1R1 and IFNAR1

(Il1r1, Ifnar12/2) were significantly less susceptible to Mtb infectionthan Il1r12/2 single-deficient animals (Fig. 2k). These findings revealsuppression of type I IFNs and their pro-bacterial activity as a majormechanism of IL-1- and PGE2-mediated host resistance against Mtb.

In previous studies both the development of active TB in latently in-fected individuals and the extent of radiographic disease among activeTB patients were found to be closely associated with a type I IFN genesignature20–22. Therefore we speculated that the IL-1–type I IFN balancemight be particularly relevant in active TB patients with more severedisease presentations, than in latent or asymptomatic cases23. We retro-spectively measured components of the IL-1, eicosanoid and IFN path-ways in plasma of patients from Chennai, India, stratified into healthycontrols (HC), latently infected individuals (LTBI) and pulmonary TB(PTB) cases based on their acid fast (AFB) sputum classification (Sup-plementary Tables 1, 2, 6). With increasing disease status, IL-1a wasreduced whereas PGE2, LXA4 and 15-Epi-LXA4 showed a significanttrend to increase (Supplementary Table 6). We next measured the sameparameters in a cohort from Zhengzhou, China, and found similar trendsfor IL-1a, PGE2 and 15-Epi-LXA4 (Supplementary Tables 3, 5, 7). Thedecreases in plasma IL-1a observed here diverge from previous find-ings in BALF or alveolar macrophages of active TB patients, where IL-1bwas shown to be elevated24–27. This may reflect differences in systemicversus pulmonary cytokine levels or between the IL-1 species becausewe observed a similar dichotomy in mice infected with Mtb (ExtendedData Fig. 5). The finding of increased PGE2 concentrations in PTBcompared to HC and/or LTBI patients was unanticipated based on ourmurine data. Nevertheless, when we investigated the eicosanoid bal-ance within the PTB group, as proposed above, we found a significantlinear trend for PGE2/LXA4 or PGE2/15-Epi-LXA4 ratios to be de-creased with higher sputum grades (Fig. 3a). We then further stratifiedthe PTB group into patients with mild or severe disease (Fig. 3b, Sup-plementary Table 5). Importantly, in both study sites the IL-1, eicos-anoid and IFN pathways faithfully distinguished PTB patients basedon disease severity after canonical correlation analysis (CCA)28 (Fig. 3b,Supplementary Table 8). Thus, clinical data from two independent studycohorts support the hypothesis that TB severity is associated with changesin IL-1, type I IFNs and eicosanoids thus supporting a link between thesepathways in active tuberculosis.

On the basis of these observations we proposed that a host-directedtherapy (HDT) targeting eicosanoids would be most effective in activedisease associated with necrotic lung pathology where high type I IFNconditions dominate. Indeed, when Il1a,Il1b2/2 animals were admin-istered PGE2 together with zileuton, a 5-LO inhibitor (Extended DataFig. 6a), pulmonary bacterial loads and pathology were reduced andsurvival enhanced (Fig. 4a, Extended Data Fig. 6b–e). Of note, when thesame treatment was given to wild-type animals for up to 6 months bac-terial control was not enhanced nor were negative side effects observed(Fig. 4a, Extended Data Fig. 6b, e and data not shown). Importantly,PGE2 and zileuton treatment had no effect on survival of susceptibleIfng2/2 or Tnfa2/2 animals (Extended Data Fig. 6f). Thus, the observedtreatment is specific for IL-1-mediated host resistance, rather than ow-ing to indirect effects on pathways generally shared by susceptible mice.We next administered either PGE2 or zileuton alone and found thatadd-back of PGE2 extended survival of Il1r12/2 animals, whereas ad-ministration of zileuton failed to do so (Extended Data Fig. 6g). Con-sistent with the latter finding, infected Il1r1,Alox52/2 mice died withsimilar kinetics as Il1r12/2 animals (Extended Data Fig. 6g). These dataindicate that the susceptibility of IL-1-deficient mice is linked to de-creased PGE2, rather than to an increase in 5-LO products, and thatyet unidentified functions of IL-1 contribute to host resistance againstMtb, because rescue of IL-1-deficient animals, either by PGE2 add-back or type I IFN neutralization, was incomplete.

We then tested the eicosanoid-based HDT in a model using IL-1-replete animals where induction of high type I IFN levels by intranasaladministration of polyinosinic-polycytidylic acid stabilized with poly-L-lysine (pICLC) causes uncontrolled disease leading to mortality and

n = 1

1

n = 5

8

n = 1

4

n = 1

5

n = 9

n = 4

HC LTBI 0

0

1+ 2+ 3+ 4+0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

0.8

**P = 0.0007***

PTB: AFB grade

PTB: AFB grade

0.0

0.1

0.2

0.3

0.4

0.5P = 0.0002

***P = 0.1198n.s.

b

Parameters in

canonical

correlation

analysis (CCA)

CC1:

IL-1αIL-1β

IL-1Ra

sIL-1R1

sIL-1R2

IFN−γIFN−αIFN−βIL-10

CC2:

PGE2

PGF2αLXA4

15-Epi-LXA4

Severe

AFB–; unilateral

AFB +; bilateral

HC LTBI 1+ 2+ 3+ 4+

a

Can

on

ical co

eff

cie

nt

2 (C

C2

)

Canonical coefficient 1 (CC1)

–1

0

1

2

3

4

5

6

7

0 2 4 6 8

–3

–2

–1

0

1

2

3

–1 2 3 4 5 6 7 80

–4

–51

Can

on

ical co

eff

cie

nt

2 (C

C2

)

Canonical coefficient 1 (CC1)

Zhengzhou, China

Chennai, India

HC

LTBI

Mild

Mild

Severe

HC

LTBI

Mild

Severe

PG

E2

/LX

A4

P = 0.0015

PG

E2

/15-E

pi-

LX

A4

n = 8

Figure 3 | IL-1, interferon and eicosanoid pathways are engaged in activeTB disease. a, Mann–Kendall linear trend post hoc comparison of ratios ofPGE2/LXA4 and PGE2/15-Epi-LXA4 according to AFB grade in the Chinesecohort. Each dot represents one individual with triangles depictingdirectionality of trend. b, PTB patients were stratified into mild (AFB2,unilateral lung lesions) and severe disease (AFB $ 11, and bilateral lunglesions) categories and canonical correlation analysis (CCA) was performedindependently for Indian and Chinese cohorts. Indicated parameters were usedto differentiate PTB from HC and LTBI groups as well as clinical severity withinPTB group based on combined canonical coefficients. sIL-1R, soluble IL-1R.

LETTER RESEARCH

0 0 M O N T H 2 0 1 4 | V O L 0 0 0 | N A T U R E | 3

Macmillan Publishers Limited. All rights reserved©2014

Page 4: Host-directed therapy of tuberculosis based on interleukin-1 ......antagonize the IL-1 pathway during Mtb infection8, absence of type I IFN signalling resulted in increased PGE2 and

necrotic lung pathology15. While triggering loss of bacterial control,increased cell death, IFN-a, IFN-b, IL-10 and IL1Ra expression inmouse lungs, pICLC treatment failed to overtly suppress IL-1 or PGE2(Extended Data Fig. 5), perhaps reflecting an indirect effect of the in-creased bacterial burden and/or type I IFN induction of IL1Ra. WhenPGE2 and zileuton were administered together, pICLC-treated micewere fully protected against weight loss and mortality, displaying lowercolony forming units as well as decreased necrotic lung pathology(Fig. 4b, Extended Data Fig. 7a). PGE2 or zileuton alone reduced pul-monary colony forming units, which resulted in complete protectionagainst both weight loss and mortality (Fig. 4c, Extended Data Fig. 7b).Importantly, protection directly correlated with increased PGE2 anddecreased IFN-b and IL1Ra production, even when animals were given5-LO blockade alone (Extended Data Fig. 7c, e).

We confirmed that zileuton and PGE2 did not directly inhibit growthof Mtb in vitro at concentrations as high as 75mg ml21 and 50mg ml21,respectively (data not shown), and co-administration did not interferewith standard antibiotic therapy (Extended Data Fig. 8). Mtb-infectedIfnar12/2 and Alox52/2 animals were resistant to pICLC-driven dis-ease, confirming that pICLC-driven susceptibility is type I IFN-mediatedand that zileuton protects by targeting 5-LO rather than through off-target effects (Fig. 4d, Extended Data Fig. 9a, b). IL-10 is induced bytype I IFNs and contributes to its antagonism of IL-1 during Mtb8,19,and accordingly Il10 2/2 animals survived pICLC administration(Extended Data Fig. 9c). LTB4, although 5-LO-dependent and prev-iously implicated in pathology of zebrafish infected with Mycobacteriummarinum29,30, did not mediate pICLC-driven disease, because Ltb4r12/2

mice succumbed with similar kinetics as wild-type controls (ExtendedData Fig. 9d). Likewise, 12/15-LO2/2 animals displayed the same mor-tality as wild-type animals, arguing against a role of 12/15-LO (ExtendedData Fig. 9d). Because PGE2 supplementation, either by direct additionor 5-LO inhibition with zileuton, completely prevented type I IFN-driven

mortality, it is possible that the HDT protects via PGE2-mediated bac-terial control and/or inhibition of type I IFNs rather than suppressionof 5-LO products.

Finally, we tested the therapeutic efficacy of 5-LO blockade by initi-ating Zileuton treatment at 30 days post-infection, when pICLC-treatedanimals already display 10–20% weight loss (Fig. 4e and data not shown).This resulted in enhanced survival, with 40% of the animals remainingalive at the end of the study. Thus, manipulation of the eicosanoid bal-ance towards PGE2 can both prevent and therapeutically amelioratedisease exacerbation associated with type I IFN expression (ExtendedData Fig. 10). Although eicosanoids have been suggested previously astargets for therapeutic intervention in tuberculosis, the data presentedhere provide in vivo proof of concept for this approach in a mammalianmodel of pulmonary Mtb infection.

METHODS SUMMARYEight-to-twelve-week-old C57BL/6 or gene-deficient animals were aerosol infectedwith Mtb strain H37Rv (100–150 colony forming units per mouse unless statedotherwise). In vitro infections of MDM or BMDM were performed at indicated mul-tiplicity of infection (m.o.i.). Eicosanoids and cytokines were measured between 24and 48 h after in vitro, in vivo at indicated time points or in plasma of TB patientsusing commercial enzyme immunoassay (EIA) or enzyme-linked immunosorbentassay (ELISA) kits. For in vitro colony forming units determination, supernatantswere collected day 5 post infection and cells lysed in distilled H2O for 5–10 min. Super-natants and cell lysate were then combined to evaluate total mycobacterial growth.Extracellular colony forming units were determined by microscopy or flow cytome-try as described in Methods. Antibodies, PCR primers and other pharmacologicalreagents as well as staining, RT–PCR and cell differentiation protocols are listed inMethods. C57BL/6 mice were administered pICLC intranasally twice a week (6 mgin 30ml). For HDT mice were given PGE2 intranasally twice a week (2mg in 30 ml)or zileuton in drinking water ad libitum (6 mg ml21) or both starting 1 day afterinfection, unless stated otherwise. Patient cohort descriptions and immune medi-ator measurements are provided in Supplementary Information and Methods.

0

20

40

60

80

100

0 20 40 60 80

P = 0.006**Lung

, c.f

.u. (lo

g10)

60

70

80

90

100

110

0 20 403010

Bo

dy w

eig

ht

(%)

WT + pICLC i.n.

(PGE2 + zileuton)a b

WT (PBS)

WT (PGE2 + zileuton)

WT + pICLC i.n.

pICLC

PBS

+++– +

–+–– +

–––+ –

++–– –PGE2

Zileuton

d

WT+ – + –

– + – +

Ifnar1–/– (PBS)

0 20 40 60 80

100

80

60

40

20

0

Ifnar1–/– + pICLC

Alox5–/– (PBS)

Alox5–/– + pICLC

PGE2 +

zileuton

PBS

WT (PBS)

WT(PGE2 + zileuton)

(PBS)

Il1a, Il1b–/–

(PGE2 + zileuton)

0 20 40 60 80 1000

20

40

60

80

100WT + pICLC (zileuton)

(zileuton at day 30)

P = 0.0445*

e

0 20 40 60 800

20

40

60

80

100

0

20

40

60

80

100

0 20 40 60503010

P = 0.0001***

WT + pICLC (PGE2 + zileuton)

Il1a,Il1b–/–

Surv

ival (%

)

Surv

ival (%

)

Surv

ival (%

)

WT + pICLC

WT + pICLC

WT + pICLC

P = 0.0001***

P = 0.0001***

Surv

ival (%

)

Surv

ival (%

)

Time post infection (days) Time post infection (days) Time post infection (days)

Time post infection (days) Time post infection (days) Time post infection (days)

6.5

7.5

8.5

8.0

7.0

9.0 ***

n.s.

Il1a,Il1b–/–

WT + pICLC WT + pICLC 3

4

5

6

7

8

*****

***

***

Lung

, c.f

.u. (lo

g10)

c

Figure 4 | HDT targeting eicosanoids confers protection in highlysusceptible mice associated with high type I IFN responses. a, Lung c.f.u.(day 21 p.i.) (left, n 5 10, error bars denote s.d., Mann–Whitney test) andsurvival (right, n 5 13, Mantel–Cox test between treated and untreatedIl1a,Il1b2/2 mice) after aerosol exposure to Mtb of WT or Il1a,Il1b2/2 treatedwith or without intranasal (i.n.) PGE2 (twice a week) and zileuton (in drinkingwater) starting day 1 p.i. Data shown are combined from two independentexperiments. b–e, Mice were treated i.n. with pICLC or without (PBS) twice aweek, starting day 1 after aerosol challenge with Mtb. b, Weight loss (left,travelling error bars indicate s.d., n 5 7) and survival (right) during Mtbinfection of WT (pICLC or PBS i.n.) mice treated with or without i.n. PGE2and zileuton. c, Lung c.f.u. 4 weeks p.i. of WT mice treated as indicated (left,n 5 5, error bars denote s.d. (Mann–Whitney test). Survival (right) of

pICLC-treated WT mice (black) compared to those who also received PGE2(yellow), zileuton (orange) or both (red). Differences were compared topICLC group (Mantel–Cox test). Data are representative of three independentexperiments (n 5 5). d, Survival of pICLC-treated WT mice (black), Ifnar12/2

animals (purple, without pICLC in pink) or Alox52/2 mice (dark green,without pICLC in light green). Statistical significance was compared to pICLCgroup. Data are representative of two independent experiments (n 5 4–6).e, Survival of pICLC-administered WT mice treated with zileuton in drinkingwater starting day 1 (solid orange line) compared to d30 (dotted orange line).Data shown are combined from two independent experiments (n 5 10).Statistical significance was compared to pICLC (black) group withMantel–Cox test. *P # 0.05; **P , 0.005; ***P , 0.0005.

RESEARCH LETTER

4 | N A T U R E | V O L 0 0 0 | 0 0 M O N T H 2 0 1 4

Macmillan Publishers Limited. All rights reserved©2014

Page 5: Host-directed therapy of tuberculosis based on interleukin-1 ......antagonize the IL-1 pathway during Mtb infection8, absence of type I IFN signalling resulted in increased PGE2 and

Online Content Methods, along with any additional Extended Data display itemsandSourceData, are available in the online version of the paper; references uniqueto these sections appear only in the online paper.

Received 28 September 2013; accepted 16 May 2014.

Published online 25 June 2014.

1. WHO. Tuberculosis Fact sheet Nu104. http://www.who.int/mediacentre/factsheets/fs104/en/index.html (2012).

2. Russell, D. G., Barry, C. E. III & Flynn, J. L. Tuberculosis: whatwe don’t knowcan, anddoes, hurt us. Science 328, 852–856 (2010).

3. Bishai, W., Sullivan, Z., Bloom,B.R. & Andersen, P.Bettering BCG: a tough task for aTB vaccine? Nature Med. 19, 410–411 (2013).

4. Lawn, S. D. & Zumla, A. I. Tuberculosis. Lancet 378, 57–72 (2011).5. Gandhi, N.R.et al. Multidrug-resistant and extensively drug-resistant tuberculosis:

a threat to global control of tuberculosis. Lancet 375, 1830–1843 (2010).6. Nathan, C. Fresh approaches to anti-infective therapies. Sci. Transl. Med. 4,

140sr142 (2012).7. Kaufmann, S. H. & Dorhoi, A. Inflammation in tuberculosis: interactions,

imbalances and interventions. Curr. Opin. Immunol. 25, 441–449 (2013).8. Mayer-Barber, K. D. et al. Innate and adaptive interferons suppress IL-1a and IL-1b

production by distinct pulmonary myeloid subsets during Mycobacteriumtuberculosis infection. Immunity 35, 1023–1034 (2011).

9. Mayer-Barber, K. D. et al. Caspase-1 independent IL-1b production is critical forhost resistance to Mycobacterium tuberculosis and does not require TLR signalingin vivo. J. Immunol. 184, 3326–3330 (2010).

10. Serhan, C. N., Chiang, N. & Van Dyke, T. E. Resolving inflammation: dual anti-inflammatory and pro-resolution lipid mediators. Nature Rev. Immunol. 8,349–361 (2008).

11. Bafica, A. et al. Host control of Mycobacterium tuberculosis is regulated by5-lipoxygenase-dependent lipoxin production. J. Clin. Invest. 115, 1601–1606(2005).

12. Chen, M. et al. Lipid mediators in innate immunity against tuberculosis: opposingroles of PGE2 and LXA4 in the induction of macrophage death. J. Exp. Med. 205,2791–2801 (2008).

13. Divangahi, M. et al. Mycobacterium tuberculosis evades macrophage defenses byinhibiting plasma membrane repair. Nature Immunol. 10, 899–906 (2009).

14. Kirtikara, K. et al. Compensatory prostaglandin E2 biosynthesis in cyclooxygenase1 or 2 null cells. J. Exp. Med. 187, 517–523 (1998).

15. Antonelli, L. R. et al. Intranasal Poly-IC treatment exacerbates tuberculosis in micethrough the pulmonary recruitment of a pathogen-permissive monocyte/macrophage population. J. Clin. Invest. 120, 1674–1682 (2010).

16. Rayamajhi,M.,Humann, J., Kearney, S., Hill, K. K.& Lenz, L. L. Antagonistic crosstalkbetween type I and II interferons and increased host susceptibility to bacterialinfections. Virulence 1, 418–422 (2010).

17. Manca, C. et al. Virulence of a Mycobacterium tuberculosis clinical isolate in mice isdetermined by failure to induce Th1 type immunity and is associated withinduction of IFN-a/b. Proc. Natl Acad. Sci. USA 98, 5752–5757 (2001).

18. Stanley, S. A., Johndrow, J. E., Manzanillo, P. & Cox, J. S. The type I IFN response toinfection with Mycobacterium tuberculosis requires ESX-1-mediated secretion andcontributes to pathogenesis. J. Immunol. 178, 3143–3152 (2007).

19. O’Garra, A. et al. The immune response in tuberculosis. Annu. Rev. Immunol. 31,475–527 (2013).

20. Berry, M. P. et al. An interferon-inducible neutrophil-driven blood transcriptionalsignature in human tuberculosis. Nature 466, 973–977 (2010).

21. Maertzdorf, J. et al. Human gene expression profiles of susceptibility andresistance in tuberculosis. Genes Immun. 12, 15–22 (2011).

22. Ottenhoff, T. H. et al. Genome-wide expression profiling identifies type 1 interferonresponse pathways in active tuberculosis. PLoS ONE 7, e45839 (2012).

23. Barry, C. E. III et al. The spectrum of latent tuberculosis: rethinking the biology andintervention strategies. Nature Rev. Microbiol. 7, 845–855 (2009).

24. Law, K. et al. Increased release of interleukin-1 beta, interleukin-6, and tumornecrosis factor-alpha by bronchoalveolar cells lavaged from involved sites inpulmonary tuberculosis. Am. J. Respir. Crit. Care Med. 153, 799–804 (1996).

25. Tsao, T. C. et al. Increased TNF-alpha, IL-1 beta and IL-6 levels in thebronchoalveolar lavage fluid with the upregulation of their mRNA in macrophageslavaged from patients with active pulmonary tuberculosis. Tuber. Lung. Dis. 79,279–285 (1999).

26. Kuo,H.P.et al.Nitric oxidemodulates interleukin-1betaandtumornecrosis factor-alpha synthesis by alveolar macrophages in pulmonary tuberculosis. Am. J. Respir.Crit. Care Med. 161, 192–199 (2000).

27. Wang, C. H. & Kuo, H. P. Nitric oxide modulates interleukin-1beta and tumournecrosis factor-alpha synthesis, and disease regression by alveolar macrophagesin pulmonary tuberculosis. Respirology 6, 79–84 (2001).

28. Rousu, J., Agranoff, D. D., Sodeinde, O., Shawe-Taylor, J. & Fernandez-Reyes, D.Biomarker discovery by sparse canonical correlation analysis of complex clinicalphenotypes of tuberculosis and malaria. PLOS Comput. Biol. 9, e1003018 (2013).

29. Tobin, D. M. et al. The lta4h locus modulates susceptibility to mycobacterialinfection in zebrafish and humans. Cell 140, 717–730 (2010).

30. Tobin, D. M. et al. Host genotype-specific therapies can optimize the inflammatoryresponse to mycobacterial infections. Cell 148, 434–446 (2012).

Supplementary Information is available in the online version of the paper.

Acknowledgements This work was supported by the NIAID Intramural Researchprogram and a Concept Acceleration Program-Award (K.D.M.-B., B.B.A. and A.S.) fromDMID, NIAID. We are grateful to K. Elkins, S. Morris, M. Belcher as well as the NIAIDABSL3 support staff for facilitating our animal studies. We thank R. Chen, L. Goldfederand Q. Gao for sharing their clinical trial expertise and research facilities, respectively.We also thank K. Kauffman, R. Thompson, S. Hieny, P. Dayal, D. Surman, L. Meng, Z. Li,L. Lifa, Q. Shen and Z. Huang for technical assistance, H. Boshoff for help with directanti-mycobacterial activity assays and M.S. Jawahar, V. V.Banurekha andR. Sridhar forrecruitment and clinical evaluation of patients in Chennai, India. We are grateful toF. Andrade Neto, H. Remold, K. Arora, J. Aliberti, M. Moayeri, P. Murphy, A. O’Garra,R. Germain and C. Serhan for discussion or critical reading of the manuscript. Finally,we thank the patients, volunteer participants, and clinical staff of the Tuberculosisdepartment of Henan Chest Hospital in Zhengzhou, China and the Department ofClinical Research (NIRT) and Department of Thoracic Medicine (Government StanleyMedical Hospital) in Chennai, India for their participation in our clinical studies.

Author Contributions K.D.M.-B. conceived the study, designed and performedexperiments, analysed data and wrote the paper; B.B.A. performed experiments,analysed data and prepared the Indian cohort description; E.P.A. and D.L.B. performedexperiments; S.D.O., J.G., S.C.D., N.P.K., Y.C., L.E.V., provided technical or analyticalassistance; S.B. recruited, sampled and collected data about patients and providedaccess to samples from Indian cohort, M.C. provided healthy donor material, A.M.S.provided Hiltonol (pICLC); R.S., W.W., X.Y., G.Z., L.E.V. and C.E.B. conducted the NaturalHistory Study in Zhengzhou, provided access to Chinese patient samples and thepreparation of the Chinese cohort description, A.S. provided conceptual advice andwrote the paper and all authors approved the final manuscript.

Author Information Reprints and permissions information is available atwww.nature.com/reprints. The authors declare no competing financial interests.Readers are welcome to comment on the online version of the paper. Correspondenceand requests for materials should be addressed to K.D.M.-B. ([email protected]).

LETTER RESEARCH

0 0 M O N T H 2 0 1 4 | V O L 0 0 0 | N A T U R E | 5

Macmillan Publishers Limited. All rights reserved©2014

Page 6: Host-directed therapy of tuberculosis based on interleukin-1 ......antagonize the IL-1 pathway during Mtb infection8, absence of type I IFN signalling resulted in increased PGE2 and

METHODSMice and Mtb infections. C57BL/6 mice were purchased from Taconic Farms(Hudson, NY). Il1a2/2, Il1b2/2, Il1a, Il1b2/2 mice were originally derived by Y.Iwakura (Tokyo University). Il1r12/2, Ptgs2(Y385F), Tnfa2/2, Alox52/2, Alox12/152/2, Ltb4r12/2 and C3HeB/FeJ mice were purchased from Jackson Laboratories(Bar Harbour, ME) and Il1r12/2 mice were backcrossed to B6 for at least 10 gen-erations. B6.SJL (CD45.1/1), B6.SJL (CD45.1/2), Il102/2, Ifng2/2, Ifnar12/2 micewere obtained through a supply contract between the National Institute of Allergyand Infectious Diseases (NIAID) and Taconic Farms. Il1r1,Ifnar12/2 and Il1r1,Alox52/2 mice were generated by inter-breeding of Il1r12/2 with Ifnar12/2 orAlox52/2 mice. All animals were maintained in an AALAC-accredited BSL2 orBSL3 facilities at the NIH or FDA and experiments performed in compliance withan animal study proposal approved by the NIAID or FDA Animal Care and UseCommittee. 8–12 weeks old male and female mice were used. Aerosol infectionwith the H37Rv strain of Mtb (100–150 c.f.u. per mouse unless stated otherwise) anddetermination of lung bacterial growth were performed as previously described9.Macrophage/dendritic cell differentiation and in vitro cultures. Murine bone-marrow cells were cultured for 7 days in either 15% GM-CSF media to generateBMDC or 30% L929 supernatant media to differentiate BMDM and then exposedto H37Rv at an indicated multiplicity of infection (m.o.i.) for 24–48 h and super-natants harvested. In some experiments PGE2 (2mg ml21, Sigma), zileuton (20mM,Tocris), valdecoxib (0.5mM, Tocris) or recombinant murine IFNb (20 ng ml21),IL-1aor IL-1b (10 ng ml21) or both together (5 ng ml21 1 5 ng ml21, R&D Systems,Minneapolis, MN) were added to the cultures.

Human elutriated monocytes were obtained from peripheral blood of healthycytomegalovirus-negative donors. Monocyte derived macrophages (MDM) weregenerated by culturing monocytes with media containing M-CSF (60 ng ml21) for7 days. Fresh media with indicated growth factors was added every 48 h. Cells wereexposed to H37Rv (m.o.i. 5 5) in the presence or absence of PGE2 (2mg ml21, Sigma),recombinant human IFN-b (10 ng ml21), IL-1a (10 ng ml21), IL-1b (10 ng ml21) orneutralizing anti-hIL1-R1 antibody (20mg ml21, R&D Systems, Minneapolis, MN)for 24 h. All recombinant human cytokines were from Peprotech (Rocky Hill, NJ).Cytokine and eicosanoid measurements in culture supernatants and biologicalfluids. Murine and human cytokine and lipid mediator concentrations in culturesupernatants were quantified using commercial ELISA kits (R&D Systems) andenzyme immunoassay (EIA) kits (Cayman Chemical, Ann Harbour, MI and OxfordBiomedical Research, Oxford, MI). Murine bronchoalveolar lavage (BAL) fluid andcell free lung homogenates were obtained and cytokines and nitric oxide measuredas described previously9. Eicosanoids in murine serum, BAL and lung were mea-sured with EIA kits (Cayman Chemical, Ann Harbour, MI and Oxford BiomedicalResearch, Oxford, MI).

For clinical studies in India, plasma levels of human IL-1a, IL-1b, IL-10, IL1Ra,IL1RI, IL1RII, IFN-c, IFN-a, IFN-b and TNF-a were measured using commercialELISA kits (R&D Systems, Minneapolis, MN). Levels of PGE2, PGF2a (CaymanChemical, Ann Harbour, MI), LXA4 and 15-EPI-LXA4 (Oxford Biomedical Re-search, Oxford, MI), were assessed using EIA kits following the manufacturers’instructions. For clinical studies in China, plasma levels of IL-1a, IL-1b, IL-10,IL1Ra, IL1RI, IL1RII, IFN-c, IFN-a, IFN-b and TNF-awere measured using com-mercial ELISA kits (R&D Systems, Minneapolis, MN) and Flowcytomix MultiplexArrays (eBioscience, San Diego, CA) according to the manufacturer’s instructions.Levels of PGE2, PGF2a, LXA4 and 15-EPI-LXA4 were assessed using EIA kits accord-ing to the manufacturer’s directions (Oxford Biomedical Research, Oxford, MI).Flow cytometry. Abs against mouse surface antigens and cytokines were purchasedfrom Ebioscience, Biolegend (San Diego, CA), AbD Serotec (Raleigh, NC) and BDPharmingen (San Diego, CA) and used in 12-colour flow cytometry either bioti-nylated or directly conjugated. The Abs used were directed against Ly6C (clone AL-21), Ly6G (1A8), CD11c (HL3 and N418), CD68 (FA-11), CD45.1 (A20), CD45.2(104), TCRb (H57-597), NK1.1 (PK136), IL-1aPE (ALF-161), IL-1bAPC (pro-IL-1b monoclonal NJTEN3), iNOS (polyclonal rabbit unconjugated), IL-12/23 p40(C15.6), TNF-a (MP6-XT22), CD11b (M1/70), activated caspase-3 (BD Pharmingen),COX-2 (polyclonal rabbit unconjugated, Cell Signaling). Biotinylated and uncon-jugated polyclonal rabbit antibodies were detected with streptavidin-conjugatedQdot 605, or goat anti-rabbit secondary-AF647, respectively from Molecular Probes-Invitrogen (Carlsbad, Ca). Ultraviolet fixable live/dead cell stain was purchased fromMolecular Probes-Invitrogen and used according to the manufacturer’s protocol.All samples were acquired on a LSRII flow cytometer (BD Biosciences, San Jose,CA) and analysed using FlowJo software (Three Star, Ashland, OR).Isolation and re-stimulation of myeloid cells from lung tissue. Perfused lungsfrom infected mice were cut into 1–2 mm pieces and subsequently digested withliberase Cl (0.4 mg ml21 in PBS; Roche-Diagnostics, Indianapolis, IN). The reactionwas stopped after 30–45 min. with an equal volume of fetal calf serum. Digestedlung was fully dispersed by passage through a 100-mm pore-size cell strainer and analiquot removed for bacterial load measurements. Isolated cells were then washed,

counted and subsequently surface stained for FACS sorting in a BSL3 containmentfacility under sterile conditions. In some experiments FACS-sorted cells or whole-lung single-cell suspensions from infected mice were stimulated overnight in thepresence or absence of live Mtb H37Rv (m.o.i. 5 1) at 106 cells per well in 96-wellplates. Supernatants were then examined for lipid mediators or cytokines. In otherexperiments, infected lung cells were re-suspended in media containing monensin(0.1%, Ebioscience) in the presence or absence of 100mg ml21 irradiated Mtb H37Rvat 106 cells per well in 96-well plates and incubated at 37 uC, 5% CO2. 5 h later cellswere surface-stained, fixed/permeabilized and intracellular staining (ICS) performed.Preparation of mixed bone-marrow chimaeric mice. B6.SJL (CD45.1/1) micewere lethally irradiated (950 rad) and reconstituted with a total of 107 donor bone-marrow cells from C57BL/6 CD45.1/2 wild-type (WT) mice mixed at equal partswith bone-marrow cells from mice deficient (KO) in CD45.2/2 Il1r12/2. Mice wereallowed to reconstitute for 8–10 weeks before aerosol infection with H37Rv.In vitro c.f.u. assay. BMDM were infected with virulent H37Rv at indicated m.o.i.At indicated time points, supernatants were collected and cells lysed in distilledH20 for 5–10 min. Supernatants and cell lysate were combined to evaluate total my-cobacterial growth by plating on Middlebrook 7H10 agar plates and incubating at37 uC for 21 days, after which c.f.u. were enumerated.In vitro extracellular c.f.u. assay. BMDM were infected with virulent H37Rv atm.o.i. 5 3. At indicated time points, supernatants were collected and cells trypsinizedfor 5–10 min. Supernatants and cell lysates were combined and acid-fast-stainedcytospin preparations were used to visualize and count intracellular AFB by lightmicroscopy. A minimum of 400–500 macrophages, number of bacteria per mac-rophage and percentage of infected macrophages as well as cell-free bacteria, werecounted by microscopy. Alternatively, BMDM were infected with virulent MtbH37Rv expressing the red fluorescent protein (H37Rv-RFP) (gift from J. Ernst) atm.o.i. 5 3. At indicated time points, supernatants were collected and cells trypsi-nized for 5–10 min. Supernatants and cell lysates were combined to evaluated ex-tracellular and intracellular mycobacterial growth by acquiring the samples on anLSRII inside a BSL3 containment laboratory.RT–PCR. Total RNA was isolated from cell suspension using the RNeasy mini kit(Qiagen, Valencia, CA). Briefly, cells were lysed in 400ml of RLT buffer containing2-mercaptoethanol. Lysates were then passed through QIA Shredder columns andRNA isolated on Qiagen mini columns (Qiagen), according to the manufacturer’sprotocol. Total RNA from lung tissue was isolated by homogenizing the lung tissuein TRIZOL (Life Technologies) and then using the Qiagen RNAeasy mini kit. In-dividual RNA samples (100 ng–1mg each) were reverse-transcribed using Super-script II (Invitrogen Life Technologies) and a mixture of oligo(dT) and randomprimers. Real-time PCR was performed on an ABI Prism 7900 sequence detectionsystem (Applied Biosystems). Relative quantities of mRNA for several genes weredetermined using SYBR Green PCR Master Mix (Applied Biosystems) and by thecomparative threshold cycle method, as described by Applied Biosystems for theABI Prism 7700/7900 sequence detection systems. In this method, mRNA levelsfor each sample were normalized to hypoxanthine-guanine phosphoribosyl trans-ferase (HPRT) mRNA levels and then expressed as a relative increase or decreasein arbitrary units (AU). Primers were designed using Roche Universal Probe Library(Roche). The sequences of the specific primers are as follows: Hprt: forward, cctcctcagaccgcttttt; reverse, aacctggttcatcatcgctaa; Il1r2: forward, tctggtacctacatttgcacattc;reverse, aaagaccttgagttccacagaca; Il1rn: forward, tgtgccaagtctggagatga; reverse, ttctttgttcttgctcagatcagt; Ifna: forward, aagcctgtgtgatgcagga; reverse, caggggctgtgtttcttctc; Ifnb: forward, ggaaagattgacgtgggaga; reverse, cctttgcaccctccagtaat.Histopathology. Lungs were fixed by inflating the tissues with 4% formaldehyde,sectioned and stained with haematoxylin and eosin or by the Ziehl–Neelsen methodto visualize acid-fast mycobacteria. The extent of the pulmonary inflammatoryresponse and was determined under light microscopy at magnifications of 320and 3100, respectively. The extent of necrosis in lungs was assessed microscop-ically using a scale of 1–5, with 5 representing the most severe necrotic response.All histological analyses were performed on blinded specimens by a trained pathol-ogist who scored all lesions on one lung section for each animal in the experiment.Treatment of animals. To create a high type I IFN environment and to exacerbatedisease in WT B6 mice, mice were administered pICLC intranasally (i.n.) twice aweek (6mg in 30ml), as previously described15. For host-directed therapy mice weregiven PGE2 i.n. twice a week (2mg in 30ml) or zileuton (Zyflo CR, CornerstoneTherapeutics, Cary, NC) in drinking water ad libitum (6mg ml21) or both startingone day after infection, unless stated otherwise. For antibiotic treatment mice werefed by gavage daily with isoniazid (625mg), rifampicin (250mg) and pyrazinamide(3.75 mg) during the indicated time period.Clinical cohort description for Chennai, India. Baseline plasma samples wereoriginated from clinical protocols approved by the Institutional Review Board (IRB)of the National Institute for Research in Tuberculosis (NIRT), NCT01154959 andNCT00342017. Written informed consent was obtained from all participants ortheir legally responsible guardians and all clinical investigations were conducted

RESEARCH LETTER

Macmillan Publishers Limited. All rights reserved©2014

Page 7: Host-directed therapy of tuberculosis based on interleukin-1 ......antagonize the IL-1 pathway during Mtb infection8, absence of type I IFN signalling resulted in increased PGE2 and

according to the principles expressed in the Declaration of Helsinki. Samples werecollected from a cohort of 97 subjects with active pulmonary TB (PTB), 14 indi-viduals with latent TB infection (LTBI) and 20 healthy controls (HC) recruited aspart of a TB cohort study at the Government Stanley Medical Hospital and at TBclinics supported by the National Institute for Research in Tuberculosis (NIRT),Chennai, India and described previously31.

TB diagnosis was based on culture positivity of sputum smears. Three sputumper subject were examined by fluorescence microscopy, processed by the modifiedPetroff’s method and cultured on Lowenstein–Jensen medium. Positive cultures ofM. tuberculosis were graded as 11 (20–100 colonies), 21 (more than 100 discretecolonies) and 31 (more than 100 colonies forming a confluent mass). A poster-oanterior chest X-ray was performed determine the extent of lung disease (unilat-eral vs. bilateral lesions), which was scored by three independent physicians fromNIRT. LTBI diagnosis was based on tuberculin skin test (TST) and QuantiferonTB-gold ELISA positivity, absence of chest radiography abnormalities or pulmo-nary symptoms and negative sputum smears and cultures. HC individuals wereasymptomatic with normal chest radiographies and negative sputum smears, cul-tures, Quantiferon results (,0.35) and TST induration (,5 mm in diameter). Attime of enrolment, all individuals were HIV-negative, ATT-naive, BCG-vaccinatedand had no record of prior TB disease. All individuals were negative for type-2 dia-betes on the basis of glycosylated haemoglobin (HbA1c) levels and random bloodglucose, according to the American Diabetes Association criteria (HbA1c levels.6.5% and random plasma glucose .200 mg dl21). Haematology was performedon all individuals using the Act-5 Diff-hematology analyser (Beckman Coulter).Plasma levels of C-reactive protein (CRP) were assessed using a Luminex-basedELISA system (Bio-Rad, Hercules, CA). Study groups were similar with regard toage and gender and a summary of the characteristics is shown in SupplementaryTables 1 and 2 where the patients with active TB were compared according to thesputum smear positivity. Individuals with positive sputum smears had higher CRPvalues in plasma and presented more frequently with bilateral lung disease thanthose with negative smears.Clinical cohort description for Zhengzhou, China. Persons with signs and symp-toms indicative of active TB and who were HIV2 and administered less than twoweeks of anti-tubercular chemotherapy or community controls were enrolled intoa prospective clinical protocol to assess response to chemotherapy (NCT01071603)conducted at the Henan Chest Hospital (HCH) from 2010 to 2012. IRBs from HCHand US National Institute for Allergy and Infectious Disease (Bethesda, Maryland)reviewed and approved the protocol and written informed consent was obtainedfrom all participants. The protocol was conducted according to the ethical prin-ciples embodied in the Declaration of Helsinki and local and national Chineseregulations.

Participants described here were enrolled into one of the two cohorts for thosewith pulmonary tuberculosis (sputum AFB smear1 cohort or the sputum AFBsmear2 cohort) based on the hospital’s initial results. The enrolled subjects allunderwent a chest CT, provided three sputum samples for AFB smear and cultureby both the BACTEC MGIT 960 system (Becton, Dickenson and Company) andLowenstein–Jensen medium (Chuang Xin Company; Hangzhou, China), and hadblood drawn for routine blood chemistry and cytology as well as several experimentalassays at enrolment. The HCH radiology department provided a scored assessmentof the chest CTs that included locations of disease by lobe, type of abnormalitiesand number of cavities for each CT. The sputum smears were prepared by the Zeil–Neilsen method using 1% carbolfuchsine32 and scored using the International UnionAgainst Tuberculosis and Lung Disease (IUATLD) scale. The 94 subjects included inthis analysis of the two PTB cohorts were confirmed to match their cohort definition,and to have abnormal chest CT scans consistent with active PTB. Fifteen percentof the subjects had a history of treatment for TB and less than 5% had a history ofdiabetes. Healthy community controls were recruited from Zhengzhou, China andincluded hospital personnel but excluded persons known to have a history of tu-berculosis or HIV infection. Each enrolee was given a physical examination, a chestCT, and the same microbiologic, biochemical and cytology tests administered tothe patient cohorts. Those HC and LTBI participants presented in this analysislacked radiologic and microbiological signs of active M. tuberculosis infection andhad no pulmonary symptoms of tuberculosis, 14 individuals were found to havelatent TB infection as indicated by a positive Quantiferon Gold in-the-tube testresult and the remaining 11 had a negative result. BCG vaccination is adminis-tered in China and the majority of trial enrolees had a scar from vaccination so theTST was not administered. Haematology was performed on all individuals usingthe Sysmex Xt-1800i (Sysmex co., LTD, Japan). Plasma levels of C-reactive protein(CRP) were assessed using an Olympus Chemistry Analyzer (Beckman Coulter)

and the Nanopia CRP kit (Sekisui Medical Co. LTD, Japan). A summary of thepatient characteristics of all study groups is shown in Supplementary Table 3. Sup-plementary Table 4 compares characteristics of subjects with active TB based ontheir sputum smear positivity. Supplementary Table 5 compares characteristics ofpatients within the PTB group that were further stratified into mild (AFB2, uni-lateral lung lesions) and severe disease (AFB $ 11, and bilateral lung lesions).Statistical analysis of clinical data sets. Median values with interquartile rangeswere used as measures of central tendency. First, anthropometric and haematolo-gical characteristics were compared between the groups of patients with differentclinical categorizations (HC, LTBI and PTB) using the non-parametric Kruskal–Wallis test (for continuous variables) or the chi-square test (for categorical variables)(Supplementary Table 1 for India, Supplementary Table 3 for China). Further ana-lyses compared the distribution of the same anthropometric and haematologicalparameters among PTB patients with positive identification of acid fast bacilli (AFB)in sputum smears and those with negative smears using the Mann–Whitney test (forcontinuous variables) of the Fisher’s exact test (for categorical variables) (Supplemen-tary Table 2 for India, Supplementary Table 4 for China). Mann–Whitney or Fisher’sexact tests, which compared distributions of parameters were used to infer clinicalseverity between PTB patients with unilateral lung lesions and negative sputumsmears (mild TB) and those with bilateral lesions and positive smears (severe TB).This analysis was limited to the Chinese data set, since the Indian data set did notinclude all the clinical parameters required for this specific test (SupplementaryTable 5).

Plasma concentrations of cytokines and lipid mediators were compared betweenclinical groups (HC, LTBI, PTB AFB2 and PTB AFB1) using the Kruskal–Wallistest with the non-parametric Mann–Kendall linear trend post hoc comparison. Thistype of post hoc test was specifically used to assess the general trend of variation ofthe plasma values of each plasma factor according to the degree of infectious burden(Supplementary Table 6 and 7). A similar approach was used considering the exactAFB sputum grade, where upward and downward trends were observed dependingon the threshold of AFB grade considered (Fig. 3b).

The distribution values for each plasma factor used are shown in SupplementaryTable 6 for India and Supplementary Table 7 for China.

Canonical correlation analysis (CCA) modelling was employed to assess whethercombinations of circulating factors of the IL-1, IFN and eicosanoids pathways candiscriminate between patients that present different degrees of TB severity (Fig. 3a).The CCA model was chosen as the number of variables studied was relatively high(13 variables were investigated in 131 individuals from India and 119 from China).This model is able to perform dimensionality reduction for two co-dependent datasets (CC1 and CC2) simultaneously so that the discrimination of the clinical end-points represents a combination of variables that are maximally correlated. Thus,trends of correlations between parameters in different clinical groups rather thantheir respective distribution within each group are the key components driving thediscrimination outcome. In our CCA model, simplified and adapted from prev-iously reported investigations of biomarkers for TB diagnosis28, linear regressiongraphs represent coefficients from different combinations of plasma factors. Thecombinations of parameters used were the ones resulting in maximum values forcorrelation analysis first in India, and then applied without modifications in theChinese data set. In the x-axis, the canonical coefficient (CC1) values were com-posed with data from the following variables: IL-1a, IL-1b, IFN-c, IFN-b, IFN-a,IL-10, sIL1R1, sIL1R2 and IL1Ra. In the y-axis, the variables considered for thecanonical coefficient (CC2) were: PGF2a, PGE2, LXA4 and 15-LXA4 (Fig. 3a).The diagnostic class prediction values obtained for each study site are shown inSupplementary Table 8 and were calculated using receiver operator characteristicscurve analysis. The statistical analyses were performed using JMP 10 (SAS, Cary,NC), STATA 10 (StataCrop LP, College Station, TX) and GraphPad Prism 6.0(GraphPad Software, San Diego, CA).Statistical analyses. The statistical significance of differences between data groupswas determined using the Mann–Whitney test, Wilcoxon matched pairs test or theMantel–Cox test using GraphPad Prism 6. Averages with s.d. are shown. *P # 0.05;**P , 0.005; ***P , 0.0005; significant differences compared as indicated in fig-ure by lines or compared to WT control groups.

31. Andrade, B. B. et al. Plasma heme oxygenase-1 levels distinguish latent orsuccessfully treated human tuberculosis from active disease. PLoS ONE 8,e62618 (2013).

32. Humason, G. L. in Animal Tissue Techniques Ch. 20, 355 (W. H. Freeman, 1979).33. Redford, P. S. et al. Influenza A virus impairs control of Mycobacterium

tuberculosis co-infection through a type I interferon receptor dependentpathway. J. Infect. Dis. 209, 270–274 (2014).

LETTER RESEARCH

Macmillan Publishers Limited. All rights reserved©2014

Page 8: Host-directed therapy of tuberculosis based on interleukin-1 ......antagonize the IL-1 pathway during Mtb infection8, absence of type I IFN signalling resulted in increased PGE2 and

LXA

4 (n

g/m

l)

Il1r1-/-

WT

0

0.5

1.0

1.5

2.0

**

COX-2

% o

f Max

CD45.1/2WT B6 BM

CD45.2/2Il1r1-/- BM

CD45.1/1WT B6 recipient

1:1

0

2

4

6

8

nsns

IL-1

2/23

p40

posi

tive

(%)

Mtb

KOWT KOWT

unst.

20

30

40

50

60

TN

posi

tive

(%)

Mtb

KOWT KOWT

unst.

ns

ns

0

5

10

15

20

iNO

S p

ositi

ve (

%)

Mtb

KOWT KOWT

unst.

nsns

CD45pos, autofl.neg , CD11bpos,Ly6Gneg,

CD68pos, Live

0

0.2

0.4

0.6

0.8

1.0

1.2

**

PG

F2α

(ng

/ml)

Il1r1-/-

WT

02.55.07.5

10.012.515.017.520.0

15-E

pi-L

XA

4 (n

g/m

l)

ns

Il1r1-/-W

T

Il1r1-/-

WT

LTB

4 (p

g/m

l)

0

20

40

60

80

100***

b c

e f

p=0.0018100

90

80

70

60% C

OX

-2 p

ositi

ve

**

WTCD45.1/2

Il1r1-/-

CD45.2/2

d

FA

CS

pur

ified

lu

ng c

ells

d28

p.i.

NK1.1neg,TCRbneg, Live

PG

E2

(ng/

ml)

4

3

2

1

0Mtbunst.

0

5

10

15

20

25

30

35

IL12

/23

p40

(ng/

ml)

WT Il1r1-/-

ans

0102030405060708090

NO

(uM

)

0

0.2

0.4

0.6

0.8

1.0

1.2

1.4

TN

(ng/

ml)

WT Il1r1-/- WT Il1r1-/-

ns ns

***

CD45pos,autofl.neg,CD11bpos ,Ly6G neg,CD68pos,Live

CD45pos, autofl.neg , CD11bpos,Ly6Gneg,

Extended Data Figure 1 | Lipid mediator production and COX2 expressionin Il1r12/2animals and mixed bone-marrow chimaeric mice after Mtbinfection. a, IL12/23 p40, TNF-a or NO measured in supernatants of WTand Il1r12/2 BMDM after exposure to live Mtb (m.o.i. 5 3). b, CD45.1/1 micewere lethally irradiated and reconstituted with equal ratios of WT (CD45.1/2)and Il1r12/2(CD45.2/2) bone-marrow cells and infected with Mtb.c, Frequency of IL-12/23p40, TNF-a or iNOS expression in WT (white circles)or Il1r12/2(KO, dark circles) total CD68pos mononuclear myeloid cells, gatedas indicated. Each connecting line depicts an individual animal. Data in arerepresentative of two independent experiments with 3–5 mice each. Pairedt-tests P values are indicated (n.s., not significant). d, PGF2a, 15-Epi-LXA4,LXA4 or LTB4 concentrations in BALF 25 days after Mtb infection of WT or

Il1r12/2 mice. e, Pulmonary single-cell suspensions from day 28 Mtb-infectedWT mice were FACS-sorted based on indicated surface marker expressionand cultured for 16 h, with (white bars) or without addition of Mtb (grey bars),after which PGE2 was measured in culture supernatants. f, Analysis of donorbone-marrow derived CD68pos myeloid cells 4 weeks p.i. in isolated lungcells marked by CD45.1 and CD45.2 expression for COX-2 protein expression(left, filled histogram: WT cells; thick line: Il1r12/2 cells; dotted line: isotypecontrol) and frequency (right) of COX-2 expression by WT (white circles) orIl1r12/2(dark circles) total CD68pos mononuclear myeloid cells. Eachconnecting line depicts an individual animal. AU, arbitrary units. Data arerepresentative of two independent experiments with 3–5 mice each. Pairedt-tests P values are indicated (n.s., not significant).

RESEARCH LETTER

Macmillan Publishers Limited. All rights reserved©2014

Page 9: Host-directed therapy of tuberculosis based on interleukin-1 ......antagonize the IL-1 pathway during Mtb infection8, absence of type I IFN signalling resulted in increased PGE2 and

0

100

200

300

400

PG

E2

(pg/

ml)

*** ***

WT

Il1r1-/-

Il1a,

Il1b-/-

a

0

1

2

3

4

WT

Il1r1-/-

***

PG

E2/

LXA

4

WT

Il1r1-/-

Il1a,

Il1b-/-

***

60

65

70

75

80

85

90

***

Infe

cted

mac

roph

ages

(%

)

WT

Il1r1-/-

Il1a,

Il1b-/-

******

Ext

race

llula

r M

tb (

%)

0

5

10

15

20

25

c db

Ext

race

llula

r M

tb-R

FP(fo

ld in

crea

se in

# o

ver

bas

elin

e)

WT

Il1r1-/-

Il1a,Il1b-/-

0

1

2

3

4

5

6

7

rm IL-1αrm IL-1β

SSC-H

H37Rv-RFP 4°C BMDM 4°CBMDM + H37Rv-RFP

mixed 1:1 for 2 minutes at 4°C

H37Rv-RFP

SS

C

4.96 7.23

H37Rv-RFP

SS

C

WT Il1r1-/-

4hrs37°C

5 days37°C

******

******

***

e

f g

Extracellular Bacteria

Extended Data Figure 2 | Characterization of extracellular bacilli in WT orIL-1-deficient macrophage cultures. a, PGE2 measured in supernatants ofWT and Il1r12/2 or Il1a,Il1b2/2 BMDM after exposure to live Mtb(m.o.i. 5 3). b, Ratio of PGE2 to LXA4 in WT or Il1r12/2 BMDC 40 h after liveMtb (m.o.i. 5 10) infection. WT, Il1r12/2 or Il1a,Il1b2/2 BMDM wereinfected with H37Rv Mtb (m.o.i. 5 3) in vitro. c, d, 5 days later free extracellularbacilli in each culture was assessed through microscopy on cytospin slides (c)as well as the frequency of infected macrophages (d). 4–5 view fields, a totalof 454 macrophages, per experimental group, were blindly scored. Data shownare the means 6 s.d. and are representative of 2 independent experiments.***P # 0.0005 significant differences compared to WT cells. e, H37Rv-RFP

(left panel) alone, BMDM alone (middle panel) or H37Rv-RFP mixed brieflywith BMDM (right panel) to quantify free extracellular bacteria by flowcytometry. f, WT or Il1r12/2 BMDM were infected with H37Rv-RFP(m.o.i. 5 3) in vitro. Free bacteria in cultures were analysed by FACS atindicated time points. g, WT, Il1r12/2 or Il1a,Il1b2/2 BMDM were infectedwith H37Rv-RFP (m.o.i. 5 3) in vitro and in the presence or absence ofrecombinant murine IL-1a, IL-1b or both. 5 days later cell-free bacteria weremeasured by FACS. ***P # 0.0005 significant differences in indicatedcomparisons. Data shown are the means 6 s.d. and are representative of 2independent experiments.

LETTER RESEARCH

Macmillan Publishers Limited. All rights reserved©2014

Page 10: Host-directed therapy of tuberculosis based on interleukin-1 ......antagonize the IL-1 pathway during Mtb infection8, absence of type I IFN signalling resulted in increased PGE2 and

3.0

3.5

4.0

4.5

5.0

5.5

6.0

0

1

2

3

4

05

1015202530354045

15-E

pi-L

XA

4 (n

g/m

l) **ns

ns*

LXA

4 (n

g/m

l)

WT

Il1r1-/-

nsns

a

e

CF

U (

106 )

rm IL-1α+β

PGE2Valdecoxib

0

1

2

3

4

5

6

Il1a,Il1b -/-WT

* ***ns

**

0

1

2

3

4 **** ns

ValdecoxibPtgs2-Y385FWT

b***

***

CF

U (

106 )

PG

E2

(ng/

ml

0

0.2

0.4

0.6

0.8

1.0

ns

d

PG

E2

(ng/

ml) *

ns

0

50

100

150

***

WT

Il1r1-/-

Ptgs2

-Y38

5F

Lung Serum

PG

E2

(ng/

ml

0

0.1

0.2

0.3

0.4

0.5

0.6 *** *****

Ptgs2

-Y38

5Fd28 W

T

Il1r1-/-

Ptgs2

-Y38

5F WT

Il1r1-/-

Ptgs2

-Y38

5F

0

10

2

4

6

8

Il1r1-/-WT

ns

***

CF

U (

106 )

PGE2

c

Extended Data Figure 3 | Role of COX-2 in IL-1-dependent PGE2 synthesisand bacterial control. a, c.f.u. at 5 days after in vitro infection (m.o.i. 5 3) ofWT or Il1r12/2 BMDM in the presence or absence of PGE2. b, c.f.u. at 5 daysafter in vitro infection (m.o.i. 5 3) of WT or Il1a,Il1b2/2 BMDM in thepresence or absence of IL1, PGE2 or the COX-2 inhibitor valdecoxib (left)and PGE2 concentrations after 48 h (right). c, c.f.u. at 5 days after in vitroinfection (m.o.i. 5 3) of WT or Ptgs2(Y385F) enzymatic activity-deficientBMDM in the presence or absence of the COX-2 inhibitor valdecoxib (left) andPGE2 concentrations after 48 h (right). d, PGE2 concentration in lungs (left)

and serum (right) of 4-week Mtb-infected WT, Il1r12/2 or ptgs2(Y385F)enzymatic activity-deficient animals. e, LXA4 and 15-Epi-LXA4concentrations or in serum of 4 weeks. Infected WT, Il1r12/2 or ptgs2(Y385F)enzymatic activity-deficient animals after aerosol challenge with 100–150 c.f.u.H37Rv. *P # 0.05 and **P # 0.005, significant differences compared to WT.Data shown are the means 6 s.d. and are representative of 2 independentexperiments. e, IL-1b concentration in supernatant of primary humanmonocyte derived macrophages from 35 healthy donors, 24 h after Mtbinfection (m.o.i. 5 5) in relation to LDH levels in same cultures.

RESEARCH LETTER

Macmillan Publishers Limited. All rights reserved©2014

Page 11: Host-directed therapy of tuberculosis based on interleukin-1 ......antagonize the IL-1 pathway during Mtb infection8, absence of type I IFN signalling resulted in increased PGE2 and

0

1

2

3

4

0

200

400

600

800 p=0.4914

p=0.0704

p=0.2897

p=0.4472

LXA

4 (p

g/m

l)

15-E

pi-L

XA

4 (n

g/m

l)

PG

E2/

15-E

pi-L

XA

4

0.01

0.10

1

10

p=0.1926

p=0.0075**ba

p=0.0145

PG

E2

(ng/

ml)

rh IFNβ

Mtbrh IFN-β

Mtb

4.0

3.5

3.0

2.5

2.0

1.0

0.5

0.0

1.5

***

ns

p=0.0081

*p=0.0395

p=0.004**

0

PG

E2/

LXA

4

.1

1

10 p=0.0644

p=0.0624

rh IL-1α+IL-1β

0

1000

2000

3000

4000

5000

6000

WT Ifnar1-/-0

5

10

15

20

WT Ifnar1-/-

BA

LF (p

g/m

l)IL-1RaIL-1β

** *

n=22

0

1

2

3

4

5

6

0

2

4

6

8

10

Ifna

Ifnb

***

***

0

1

2

3

4 Il1r2

*** *** ***

Ifnb

0

5

10

15

Rel

ativ

e m

RN

A (f

old

incr

ease

ove

r un

infe

cted

, rel

ativ

e to

HP

RT

)Ifna

0

1

2

3

4

5 Il1rn

0

10

20

30

WTIl1a,Il1b-/-

Il1r2

0

1

2

3

4

MOI: 0.1 1 3 10 0.1 1 3 10

*

**

**

**

0.1 1 3 10 0.1 1 3 10

Rel

ativ

e m

RN

A (f

old

incr

ease

ove

r un

infe

cted

, rel

ativ

e to

HP

RT

)

rm IL-1αrm IL-1β

rm IL-1αrm IL-1β

c d e

Extended Data Figure 4 | IL-1 type I IFN crosstalk. a, IL-1b and IL1Raconcentrations measured in BALF of WT or Ifnar12/2 mice 4 weeks p.i.b, PGE2/15-Epi-LXA4 and PGE2/LXA4 ratios, LXA4, and 15-Epi-LXA4concentrations in supernatants of primary human MDM from 22 healthydonors, 24 h after Mtb infection (m.o.i. 5 5) in the presence or absence ofrhIFN-b. Differences were compared as indicated by lines. c, PGE2concentrations in culture supernatants of primary human MDM from 7healthy donors, 24 h after Mtb infection (m.o.i. 5 5) in the presence or absence

of rhIFN-b, rhIL-1a and Il-1b or PGE2. Differences were compared asindicated by lines. d, WT (white) or Il1a,Il1b2/2 (light grey) BMDM wereinfected with increasing m.o.i. of Mtb in vitro. mRNA expression of indicatedgenes was determined at 40 h. e, WT (white) or Il1a,Il1b2/2 (light grey) BMDMwere infected with m.o.i. 5 3 of Mtb in vitro in the presence or absence ofrecombinant murine IL-1a, IL-1b or both. mRNA expression of indicatedgenes was determined at 40 h. Data shown are the means 6 s.d. and arerepresentative of 2 independent experiments.

LETTER RESEARCH

Macmillan Publishers Limited. All rights reserved©2014

Page 12: Host-directed therapy of tuberculosis based on interleukin-1 ......antagonize the IL-1 pathway during Mtb infection8, absence of type I IFN signalling resulted in increased PGE2 and

c

0

50000

100000

150000

200000

IL1Ra

0

5 00

1000

1500

2000

*

0

200

400

600

800

1000nsns

IFNβ

IL1Ra

nd0

50

100

150

200

0

2

4

6

8

0

3000

6000

9000

12000

*

0

2

4

6

0

3

6

9

12

1

10

100

1000

0

5

10

15

20

*IL-10

*

**

ns

*

IL-1β IL-1α

IL-1β IL-1α

IFNα

IL-1β

% D

ead

cells

(d31

p.i.

BA

LF, u

ngat

ed)

0

20

40

60

80***

100a

IL-1α/

IL1R

a

0.

0.

0.

0.

00

01

02

03

*

BA

LF (p

g/m

l)

3

4

5

6

7

BA

LF, C

FU

(Lo

g 10)

***

pICLC

b

d

BA

LF (p

g/m

l)

Lung

(pg/

ml)

Ser

um (p

g/m

l)

0

200

400

600

800

0

1000

2000

3000

PGE2

ns

PGE2ns

pICLC pICLC

pICLC

pICLC

Extended Data Figure 5 | Cytokine expression in Mtb-infected pICLC-treated WT mice. WT mice were treated with pICLC (1) or without (2, PBS)intranasally twice a week, starting day 1 after aerosol challenge with Mtb.a–c, c.f.u. (a), FACS analysis of dead cells (b) and indicated cytokines measured

in BALF at 4 weeks post infection (c). d, Indicated cytokines and eicosanoidswere measured by ELISA in lung homogenates, BALF and serum 4 weeks p.i.Data are representative of a minimum of 3 independent experiments with aminimum of 4 mice per group.

RESEARCH LETTER

Macmillan Publishers Limited. All rights reserved©2014

Page 13: Host-directed therapy of tuberculosis based on interleukin-1 ......antagonize the IL-1 pathway during Mtb infection8, absence of type I IFN signalling resulted in increased PGE2 and

ns0

20

40

60

80

100

Time post infection (days)

0 20 40 60

d0

100-150 CFUH37Rv by aerosol

Zileuton in drinking water

d3 d7 d10 d14 d17 d21 d24...

2x weekly PGE2 i.n.

WT (PBS)

WT (PGE2 + Zileuton)

Il1a, Il1b-/- (PGE2 + Zileuton)

Il1a, Il1b-/- (PBS)

f Ifng-/- (PBS)

Ifng-/-(PGE2 + Zileuton)

Tnfa-/- (PBS)

Tnfa-/- (PGE2 + Zileuton)

0

20

40

60

80

100

ba

e Il1r1-/- (PBS) Il1r1-/- (PGE2+ Zileuton)d22

0123456789

WT Il1r1 -/-

PGE2 + Zileuton

PBS

Lung

infla

mm

atio

n (

% a

rea

affe

cted

, AU

)

*

0

1

2

3

4

5

6

7

8

9

WT Il1r1 -/-

PGE2 + Zileuton

PBS

Aci

d F

ast B

acill

i (

Arb

itrat

y U

nits

, AU

) ***

0

1

2

3

4

5

WT Il1r1 -/-

PGE2 + Zileuton

PBS

*ns

nsns

Nec

rosi

s (

Arb

itrat

y U

nits

, AU

)

d

0

10

20

30

40

50

60

Live

/Dea

d po

sitiv

e (%

) (L

ung,

ung

ated

)

PGE2 + Zileuton

PBS

*

***

Sur

viva

l (%

)S

urvi

val (

%)

0 20 301060

70

80

90

100

110

Bod

y w

eigh

t (%

)0 20 3010

Il1a, Il1b-/-

(PGE2 + Zileuton)

Il1a, Il1b -/- (PBS)

WT (PBS)

WT (PGE2 + Zileuton)

Time post infection (days)

3.5

4.5

5.5

5.0

4.0

6.5

BA

LF, C

FU

(Lo

g 10) ***

ns

WT Il1a,Il1b-/-

PGE2 + Zileuton

PBS

c

503010

0 20 40 60503010

ns

g

Time post infection (days)

0

20

40

60

80

100

Sur

viva

l (%

)

0 20 40 60503010

Il1r1-/-

Il1r1-/-(Zileuton)

Il1r1-/-(PGE2)

(PBS)

Il1r1-/-

Il1r1,Alox5 -/-

Alox5-/-

0

20

40

60

80

100

Sur

viva

l (%

)

0 20 40 8060

p=0.0414

*

WT Il1a,Il1b-/-

Extended Data Figure 6 | PGE2 administration and 5–LO inhibition in IL-1-deficient animals reduces necrotic pathology and increases bacterialcontrol. a, Experimental design for host-directed therapy with PGE2 andzileuton in Mtb-infected mice. b, Weight loss (travelling error bars indicate s.d.,n 5 6) during Mtb infection of WT (left) or Il1a,Il1b2/2 (right) mice treatedwith or without i.n. PGE2 and zileuton. c, d, Bacterial loads in BALF (c) orFACS analysis (d) of dead cells 21 days after aerosol exposure to Mtb (H37Rv)in lungs of WT or Il1a,Il1b2/2 treated with or without i.n. PGE2 and zileuton.e, Representative haematoxylin-and-eosin-staining of lung lobes of untreatedIl1r12/2 or treated Il1r12/2 with i.n. PGE2 and zileuton in drinking water(top panels). Bottom panels show the relative number of acid fast bacilli, lunginflammation (% lung affected, mean granuloma sizes) and degree of necrosis

scored blindly by a trained pathologist. Differences were compared as indicatedby lines. Data shown are representative of two independent experiments with aminimum of 6 mice per group. f, Survival after aerosol exposure to Mtb(H37Rv) of Ifng2/2 (top panel) or Tnfa2/2 (bottom panel) untreated (PBS)or treated i.n. PGE2 (twice a week) and zileuton (in drinking water) starting atday 1 p.i. Data shown are representative of three independent experiments witha minimum of 4 mice per group. g, Survival after aerosol exposure to Mtb(H37Rv) of Il1r12/2 (top panel) treated with either PGE2 or zileuton. Datashown are representative of two independent experiments with a minimumof 4 mice per group. Bottom panel shows survival of Il1r12/2, Alox52/2 orIl1r1,Alox52/2 mice. Data shown are representative of three independentexperiments with a minimum of 4 mice per group.

LETTER RESEARCH

Macmillan Publishers Limited. All rights reserved©2014

Page 14: Host-directed therapy of tuberculosis based on interleukin-1 ......antagonize the IL-1 pathway during Mtb infection8, absence of type I IFN signalling resulted in increased PGE2 and

PBS

PGE2 Zileuton

a d21 WT (PBS)

WT + pICLC + PGE2+ Zileuton

WT + pICLC

pICLCPBS

PGE2 Zileuton

0

2

4

6

8

10

0

1

2

3

4

5

0

2

4

6

8

10

Nec

rosi

s (

Arb

itrat

y U

nits

, AU

)Lu

ng in

flam

mat

ion

(%

are

a af

fect

ed, A

U)

Aci

d F

ast B

acill

i (

Arb

itrat

y U

nits

, AU

)

0

1

2

3

4

5

6

7e

PBS

PGE2 Zileuton

*********

* *

* *

* *

0

5

10

15

IL-1

Ra

(ng/

ml)

PBS

PGE2 Zileuton

* *c Serum

*BALF

ns

d28 d28

d21

ns

PBS

PGE2 Zileuton

IFN

-β (

pg/m

l)

PG

E2

(ng/

ml)

** *

0

5

10

15

20 ** *

Bod

y w

eigh

t (%

) 130

120

110

100

90

80

70

60PBS

PGE2 Zileuton

n=19

n=21

n=10 n=17 n=15 n=15

***b ******

***

***

ns

pICLC

pICLC

pICLC pICLC pICLC

*********

0

1

2

3

4

5

6 ***

BA

LF, C

FU

(Lo

g 10)

d

Extended Data Figure 7 | PGE2 and/or 5–LO inhibition in pICLC-treatedWT mice reduces necrotic pathology and increases bacterial control. a, WTmice were treated with pICLC or without (PBS) intranasal twice a week, startingday 1 after aerosol challenge with Mtb. Representative haematoxylin-and-eosin-staining of lung lobes of untreated WT (PBS), pICLC-treated or pICLC-treated with i.n. PGE2 and zileuton in drinking water (left panels) three weeksafter Mtb infection. Right panels show the relative number of acid fast bacilli,lung inflammation (% lung affected, mean granuloma sizes) and degree ofnecrosis scored blindly by a trained pathologist. Differences were compared asindicated by lines. Data shown are representative of two independentexperiments with a minimum of 4 mice per group. b, WT mice were treated asindicated in figure. Weight loss in experimental groups was normalized totime of moribundity of last remaining mouse in pICLC group (between day 40and day 100, depending on experiment). For pICLC group (black), weight loss

is shown for each animal at time of moribundity. Data shown are combinedfrom three individual experiments with 3–7 mice per group. Differences werecompared to pICLC (black) group. c, WT mice were untreated (PBS) treatedwith pICLC or pICLC, PGE2 and zileuton intranasally starting day 1 afteraerosol challenge with Mtb. 3 weeks post infection IL1Ra was measured byELISA in BALF and serum. Data shown are representative of two independentexperiments with a minimum of 4 mice per group. d, Bacterial loads 28 daysafter aerosol exposure to Mtb (H37Rv) in BALF of pICLC-treated WT (black)mice with or without i.n. PGE2 (yellow) or zileuton (orange) or a combinationof both (red) as indicated in figure. e, PGE2 and IFN-b concentrations inBALF of indicated mice 4 weeks p.i. ***P # 0.0005, significant differencescompared to pICLC (black)-treated group. Data shown are representative ofthree independent experiments with a minimum of 4 mice per group.

RESEARCH LETTER

Macmillan Publishers Limited. All rights reserved©2014

Page 15: Host-directed therapy of tuberculosis based on interleukin-1 ......antagonize the IL-1 pathway during Mtb infection8, absence of type I IFN signalling resulted in increased PGE2 and

0

1

2

3

4

5

6

BA

LF, C

FU

(Lo

g 10),

d43

RHZ (d24-d42)PGE2 + Zileuton

ns

ns

2

3

4

5

6

7

8

Lung

, CF

U (

Log 10

), d

43

ns

nsa b

Extended Data Figure 8 | Host-directed therapy with PGE2 and zileutondoes not interfere with antibiotic treatment. Bacterial loads in C3HeB/FeJmice 43 days p.i. with 100–150 c.f.u. H37Rv, treated or untreated with PGE2intranasally and zileuton in drinking water. Starting day 24 p.i. antibiotic

treatment with rifampicin, pyrazinamide and isoniazid was given 53 a weekuntil one day before collection. a, b, Bacterial loads in BALF (a) and lungs (b).Differences were compared as indicated by lines. Data shown are from oneexperiment with 5–7 mice per group.

LETTER RESEARCH

Macmillan Publishers Limited. All rights reserved©2014

Page 16: Host-directed therapy of tuberculosis based on interleukin-1 ......antagonize the IL-1 pathway during Mtb infection8, absence of type I IFN signalling resulted in increased PGE2 and

PBS

Zileuton

a

60

80

100

120

140

WT Alox5-/-

nsns

ns***

60

70

80

90

100

110

120

***

ns

WT Ifnar1-/-

pICLCPBS

Bod

y w

eigh

t (%

)

0 20 40 60 800

20

40

60

80

100

WT + pICLC

WT (PBS)

Ltb4r1 -/-(PBS)

Ltb4r1-/- + pICLC

d

0

20

40

60

80

100

0 20 40 60 80

12/15-LO-/- + pICLC

12/15-LO -/-(PBS)

60

80

100

120

140

WT Ltb4r1-/-12/15-LO-/-

PBS

b cns

50

60

70

80

90

100

110

120ns

WT Il10-/-PBS

***

0 10 20 30 40 50 600

20

40

60

80

100Il10-/-(PBS)

Il10-/- + pICLCns

*** ***

***

Bod

y w

eigh

t (%

)

Bod

y w

eigh

t (%

)

Sur

viva

l (%

)

Sur

viva

l (%

)

Bod

y w

eigh

t (%

)

pICLC pICLC

pICLC

WT + pICLC

Time post infection (days)

Time post infection (days)

Extended Data Figure 9 | IL-10, but not LTB4 or 12/15-LO, is required fortype I IFN-driven disease exacerbation of pICLC-treated WT animalsinfected with Mtb. a, WT or Ifnar12/2 mice were treated with pICLC orwithout (PBS) intranasal twice a week, starting day 1 after aerosol challengewith Mtb. Weight loss in experimental groups was normalized to time ofmoribundity of last remaining mouse in pICLC group (black) for eachindependent experiment. For pICLC group (black), weight loss is shown foreach animal at time of moribundity. Data shown are combined from twoindividual experiments with 3–7 mice per group. Differences were compared asindicated by lines. b, WT or Alox52/2 mice were treated with pICLC or without(PBS) intranasally twice a week or administered zileuton in the drinking water.Data shown are representative of two individual experiments with 4–8 mice pergroup. Differences were compared as indicated by lines. c, WT or Il102/2 micewere treated with pICLC or without (PBS) intranasally twice a week. For pICLCgroup (black), weight loss is shown for each animal at time of moribundity.

Data shown are representative of two individual experiments with 3–8 mice pergroup. Differences were compared as indicated by lines. Survival (right panel)of WT or Il102/2 mice treated with (dark grey lines) or without (light greylines). Data shown are representative of two individual experiments with 4–6mice per group. d, WT, 12/15-LO2/2, Ltb4r12/2 mice were treated with pICLCor without (PBS) intranasally twice a week. For pICLC groups, weight loss isshown for each animal at time of moribundity. Data shown are representativeof two individual experiments with 5–7 mice per group. Differences werecompared to PBS controls in each mouse strain. Survival of WT or 12/15-LO2/2 mice (middle panel) treated with pICLC (dark blue lines) or without(light blue lines). Data shown are representative of two individual experimentswith 4–6 mice per group. Survival of WT or Ltb4r12/2 mice (right panel)treated with pICLC (burgundy lines) or without (brown lines). Data shown arerepresentative of two individual experiments with 4–6 mice per group.

RESEARCH LETTER

Macmillan Publishers Limited. All rights reserved©2014

Page 17: Host-directed therapy of tuberculosis based on interleukin-1 ......antagonize the IL-1 pathway during Mtb infection8, absence of type I IFN signalling resulted in increased PGE2 and

IL-1α/β

IFNα/β

M. tuberculosis

IL-1Ra

IL-10PGE2

IL-1α/β

PGE2M. tuberculosis

IL-1Ra

IL-10

IFNα/β

IL-1α/β

PGE2M. tuberculosis

IL-1Ra

IL-10

IL-1α/β

IFNα/β

M. tuberculosis

PGE2

Zileuton

HDTIL-1Ra

IL-10

HDT targeting PGE2:Return to non-pathological equilibrium

COX-2

COX-2 5-LO

IFNα/β

pICLC

PGE2

High Type I IFN environment-Scenario A: IL-1 deficiency

High type I IFN environment-Scenario B: pICLC driven induction

Inflammatory Equilibriuma b

c d

WT B6 mice:(models latent state in TB patients)

IL-1 deficient mice:(models uncontrolled active

disease in subset of TB patients)

WT mice - pICLC administration:(models uncontrolled active

disease in subset of TB patients and/or viral co-infections)

Extended Data Figure 10 | Schematic summary of IL-1–type I IFN counter-regulation during Mtb infection. a, Virulent Mtb directly induces IL-1a andIL-1b as well as type I IFNs in myelophagocytic cells. Whereas IL-1 is requiredfor bacterial control, type I IFNs are considered detrimental because they canexert pro-bacterial effects. During low-dose aerosol infection in WT B6 micethe balance of these two pathways establishes an inflammatory equilibrium thatallows for containment of bacilli and chronic infection and thus models certainaspects associated with latency in TB patients. This is achieved by an antagonistrelationship whereby type I IFNs inhibit IL-1a/b directly as well as throughinduction of IL-10 and IL1Ra, while in the opposing direction IL-1a/b limittype I IFNs through COX-2-mediated PGE2 induction. b, In the absence of IL-1, PGE2 fails to inhibit type I IFNs and the equilibrium is disturbed. This leads

to excessive type I IFN expression that in turn causes increased bacterialreplication and pathology, thus modelling the type I IFN-associateduncontrolled active disease that occurs in a subset of TB patients20–22.c, Likewise, experimental induction of type I IFNs by pICLC or viral co-infections33 creates a detrimental environment with uncontrolled bacterialgrowth and extracellular bacteria, ultimately resulting in loss of host resistanceand increased mortality. d, HDT targeting PGE2, either directly or indirectlyby enhancing PGE2 levels via 5-LO inhibition with zileuton, returns the systemto a non-pathological steady state by limiting type I IFN-driven diseaseexacerbation, bacterial replication and pulmonary necrosis. Green arrowsindicate inductive and red lines inhibitory pathways.

LETTER RESEARCH

Macmillan Publishers Limited. All rights reserved©2014