Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

Host-directed therapy of tuberculosis based on interleukin-1 and type I interferon crosstalk

Subjects

Abstract

Tuberculosis remains second only to HIV/AIDS as the leading cause of mortality worldwide due to a single infectious agent1. Despite chemotherapy, the global tuberculosis epidemic has intensified because of HIV co-infection, the lack of an effective vaccine and the emergence of multi-drug-resistant bacteria2,3,4,5. Alternative host-directed strategies could be exploited to improve treatment efficacy and outcome, contain drug-resistant strains and reduce disease severity and mortality6. The innate inflammatory response elicited by Mycobacterium tuberculosis (Mtb) represents a logical host target7. Here we demonstrate that interleukin-1 (IL-1) confers host resistance through the induction of eicosanoids that limit excessive type I interferon (IFN) production 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 associated 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 manipulate the host eicosanoid network and represent feasible alternatives to conventional chemotherapy.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: IL-1 triggers PGE2 synthesis during Mtb infection.
Figure 2: IL-1 and PGE2 negatively regulate type I IFNs.
Figure 3: IL-1, interferon and eicosanoid pathways are engaged in active TB disease.
Figure 4: HDT targeting eicosanoids confers protection in highly susceptible mice associated with high type I IFN responses.

Similar content being viewed by others

References

  1. WHO. Tuberculosis Fact sheet N°104. http://www.who.int/mediacentre/factsheets/fs104/en/index.html (2012)

  2. Russell, D. G., Barry, C. E., III & Flynn, J. L. Tuberculosis: what we don’t know can, and does, hurt us. Science 328, 852–856 (2010)

    Article  ADS  CAS  Google Scholar 

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

    Article  Google Scholar 

  4. Lawn, S. D. & Zumla, A. I. Tuberculosis. Lancet 378, 57–72 (2011)

    Article  Google Scholar 

  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)

    Article  Google Scholar 

  6. Nathan, C. Fresh approaches to anti-infective therapies. Sci. Transl. Med. 4, 140sr142 (2012)

    Article  Google Scholar 

  7. Kaufmann, S. H. & Dorhoi, A. Inflammation in tuberculosis: interactions, imbalances and interventions. Curr. Opin. Immunol. 25, 441–449 (2013)

    Article  CAS  Google Scholar 

  8. Mayer-Barber, K. D. et al. Innate and adaptive interferons suppress IL-1α and IL-1β production by distinct pulmonary myeloid subsets during Mycobacterium tuberculosis infection. Immunity 35, 1023–1034 (2011)

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  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)

    Article  CAS  Google Scholar 

  11. Bafica, A. et al. Host control of Mycobacterium tuberculosis is regulated by 5-lipoxygenase-dependent lipoxin production. J. Clin. Invest. 115, 1601–1606 (2005)

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  14. Kirtikara, K. et al. Compensatory prostaglandin E2 biosynthesis in cyclooxygenase 1 or 2 null cells. J. Exp. Med. 187, 517–523 (1998)

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

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

    Article  MathSciNet  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

  32. Humason, G. L. in Animal Tissue Techniques Ch. 20, 355 ( W. H. Freeman, 1979)

    Google Scholar 

  33. Redford, P. S. et al. Influenza A virus impairs control of Mycobacterium tuberculosis co-infection through a type I interferon receptor dependent pathway. J. Infect. Dis. 209, 270–274 (2014)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the NIAID Intramural Research program and a Concept Acceleration Program-Award (K.D.M.-B., B.B.A. and A.S.) from DMID, NIAID. We are grateful to K. Elkins, S. Morris, M. Belcher as well as the NIAID ABSL3 support staff for facilitating our animal studies. We thank R. Chen, L. Goldfeder and 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 direct anti-mycobacterial activity assays and M. S. Jawahar, V. V. Banurekha and R. Sridhar for recruitment and clinical evaluation of patients in Chennai, India. We are grateful to F. 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 Tuberculosis department of Henan Chest Hospital in Zhengzhou, China and the Department of Clinical Research (NIRT) and Department of Thoracic Medicine (Government Stanley Medical Hospital) in Chennai, India for their participation in our clinical studies.

Author information

Authors and Affiliations

Authors

Contributions

K.D.M.-B. conceived the study, designed and performed experiments, 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. performed experiments; S.D.O., J.G., S.C.D., N.P.K., Y.C., L.E.V., provided technical or analytical assistance; S.B. recruited, sampled and collected data about patients and provided access 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 Natural History Study in Zhengzhou, provided access to Chinese patient samples and the preparation of the Chinese cohort description, A.S. provided conceptual advice and wrote the paper and all authors approved the final manuscript.

Corresponding author

Correspondence to Katrin D. Mayer-Barber.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Lipid mediator production and COX2 expression in Il1r1−/−animals and mixed bone-marrow chimaeric mice after Mtb infection.

a, IL12/23 p40, TNF-α or NO measured in supernatants of WT and Il1r1−/− BMDM after exposure to live Mtb (m.o.i. = 3). b, CD45.1/1 mice were lethally irradiated and reconstituted with equal ratios of WT (CD45.1/2) and Il1r1−/−(CD45.2/2) bone-marrow cells and infected with Mtb. c, Frequency of IL-12/23p40, TNF-α or iNOS expression in WT (white circles) or Il1r1−/−(KO, dark circles) total CD68pos mononuclear myeloid cells, gated as indicated. Each connecting line depicts an individual animal. Data in are representative of two independent experiments with 3–5 mice each. Paired t-tests P values are indicated (n.s., not significant). d, PGF2α, 15-Epi-LXA4, LXA4 or LTB4 concentrations in BALF 25 days after Mtb infection of WT or Il1r1−/− mice. e, Pulmonary single-cell suspensions from day 28 Mtb-infected WT mice were FACS-sorted based on indicated surface marker expression and 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 donor bone-marrow derived CD68pos myeloid cells 4 weeks p.i. in isolated lung cells marked by CD45.1 and CD45.2 expression for COX-2 protein expression (left, filled histogram: WT cells; thick line: Il1r1−/− cells; dotted line: isotype control) and frequency (right) of COX-2 expression by WT (white circles) or Il1r1−/−(dark circles) total CD68pos mononuclear myeloid cells. Each connecting line depicts an individual animal. AU, arbitrary units. Data are representative of two independent experiments with 3–5 mice each. Paired t-tests P values are indicated (n.s., not significant).

Extended Data Figure 2 Characterization of extracellular bacilli in WT or IL-1-deficient macrophage cultures.

a, PGE2 measured in supernatants of WT and Il1r1−/− or Il1α,Il1β−/− BMDM after exposure to live Mtb (m.o.i. = 3). b, Ratio of PGE2 to LXA4 in WT or Il1r1−/− BMDC 40 h after live Mtb (m.o.i. = 10) infection. WT, Il1r1−/− or Il1α,Il1β−/− BMDM were infected with H37Rv Mtb (m.o.i. = 3) in vitro. c, d, 5 days later free extracellular bacilli 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 total of 454 macrophages, per experimental group, were blindly scored. Data shown are the means ± 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 briefly with BMDM (right panel) to quantify free extracellular bacteria by flow cytometry. f, WT or Il1r1−/− BMDM were infected with H37Rv-RFP (m.o.i. = 3) in vitro. Free bacteria in cultures were analysed by FACS at indicated time points. g, WT, Il1r1−/− or Il1α,Il1β−/− BMDM were infected with H37Rv-RFP (m.o.i. = 3) in vitro and in the presence or absence of recombinant murine IL-1α, IL-1β or both. 5 days later cell-free bacteria were measured by FACS. ***P ≤ 0.0005 significant differences in indicated comparisons. Data shown are the means ± s.d. and are representative of 2 independent experiments.

Extended Data Figure 3 Role of COX-2 in IL-1-dependent PGE2 synthesis and bacterial control.

a, c.f.u. at 5 days after in vitro infection (m.o.i. = 3) of WT or Il1r1−/− BMDM in the presence or absence of PGE2. b, c.f.u. at 5 days after in vitro infection (m.o.i. = 3) of WT or Il1α,Il1β−/− BMDM in the presence 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 vitro infection (m.o.i. = 3) of WT or Ptgs2(Y385F) enzymatic activity-deficient BMDM in the presence or absence of the COX-2 inhibitor valdecoxib (left) and PGE2 concentrations after 48 h (right). d, PGE2 concentration in lungs (left) and serum (right) of 4-week Mtb-infected WT, Il1r1−/− or ptgs2(Y385F) enzymatic activity-deficient animals. e, LXA4 and 15-Epi-LXA4 concentrations or in serum of 4 weeks. Infected WT, Il1r1−/− 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 ± s.d. and are representative of 2 independent experiments. e, IL-1β concentration in supernatant of primary human monocyte derived macrophages from 35 healthy donors, 24 h after Mtb infection (m.o.i. = 5) in relation to LDH levels in same cultures.

Extended Data Figure 4 IL-1 type I IFN crosstalk.

a, IL-1β and IL1Ra concentrations measured in BALF of WT or Ifnar1−/− mice 4 weeks p.i. b, PGE2/15-Epi-LXA4 and PGE2/LXA4 ratios, LXA4, and 15-Epi-LXA4 concentrations in supernatants of primary human MDM from 22 healthy donors, 24 h after Mtb infection (m.o.i. = 5) in the presence or absence of rhIFN-β. Differences were compared as indicated by lines. c, PGE2 concentrations in culture supernatants of primary human MDM from 7 healthy donors, 24 h after Mtb infection (m.o.i. = 5) in the presence or absence of rhIFN-β, rhIL-1α and Il-1β or PGE2. Differences were compared as indicated by lines. d, WT (white) or Il1α,Il1β−/− (light grey) BMDM were infected with increasing m.o.i. of Mtb in vitro. mRNA expression of indicated genes was determined at 40 h. e, WT (white) or Il1α,Il1β−/− (light grey) BMDM were infected with m.o.i. = 3 of Mtb in vitro in the presence or absence of recombinant murine IL-1α, IL-1β or both. mRNA expression of indicated genes was determined at 40 h. Data shown are the means ± s.d. and are representative of 2 independent experiments.

Extended Data Figure 5 Cytokine expression in Mtb-infected pICLC-treated WT mice.

WT mice were treated with pICLC (+) or without (−, PBS) intranasally twice a week, starting day 1 after aerosol challenge with Mtb. ac, 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 eicosanoids were measured by ELISA in lung homogenates, BALF and serum 4 weeks p.i. Data are representative of a minimum of 3 independent experiments with a minimum of 4 mice per group.

Extended Data Figure 6 PGE2 administration and 5–LO inhibition in IL-1-deficient animals reduces necrotic pathology and increases bacterial control.

a, Experimental design for host-directed therapy with PGE2 and zileuton in Mtb-infected mice. b, Weight loss (travelling error bars indicate s.d., n = 6) during Mtb infection of WT (left) or Il1α,Il1β−/− (right) mice treated with or without i.n. PGE2 and zileuton. c, d, Bacterial loads in BALF (c) or FACS analysis (d) of dead cells 21 days after aerosol exposure to Mtb (H37Rv) in lungs of WT or Il1α,Il1β−/− treated with or without i.n. PGE2 and zileuton. e, Representative haematoxylin-and-eosin-staining of lung lobes of untreated Il1r1−/− or treated Il1r1−/− with i.n. PGE2 and zileuton in drinking water (top panels). Bottom panels show the relative number of acid fast bacilli, lung inflammation (% lung affected, mean granuloma sizes) and degree of necrosis scored blindly by a trained pathologist. Differences were compared as indicated by lines. Data shown are representative of two independent experiments with a minimum of 6 mice per group. f, Survival after aerosol exposure to Mtb (H37Rv) of Ifng−/− (top panel) or Tnfa−/− (bottom panel) untreated (PBS) or treated i.n. PGE2 (twice a week) and zileuton (in drinking water) starting at day 1 p.i. Data shown are representative of three independent experiments with a minimum of 4 mice per group. g, Survival after aerosol exposure to Mtb (H37Rv) of Il1r1−/− (top panel) treated with either PGE2 or zileuton. Data shown are representative of two independent experiments with a minimum of 4 mice per group. Bottom panel shows survival of Il1r1−/−, Alox5−/− or Il1r1,Alox5−/− mice. Data shown are representative of three independent experiments with a minimum of 4 mice per group.

Extended Data Figure 7 PGE2 and/or 5–LO inhibition in pICLC-treated WT mice reduces necrotic pathology and increases bacterial control.

a, WT mice were treated with pICLC or without (PBS) intranasal twice a week, starting day 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 weeks after Mtb infection. Right panels show the relative number of acid fast bacilli, lung inflammation (% lung affected, mean granuloma sizes) and degree of necrosis scored blindly by a trained pathologist. Differences were compared as indicated by lines. Data shown are representative of two independent experiments with a minimum of 4 mice per group. b, WT mice were treated as indicated in figure. Weight loss in experimental groups was normalized to time of moribundity of last remaining mouse in pICLC group (between day 40 and day 100, depending on experiment). For pICLC group (black), weight loss is shown for each animal at time of moribundity. Data shown are combined from three individual experiments with 3–7 mice per group. Differences were compared to pICLC (black) group. c, WT mice were untreated (PBS) treated with pICLC or pICLC, PGE2 and zileuton intranasally starting day 1 after aerosol challenge with Mtb. 3 weeks post infection IL1Ra was measured by ELISA in BALF and serum. Data shown are representative of two independent experiments with a minimum of 4 mice per group. d, Bacterial loads 28 days after 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 combination of both (red) as indicated in figure. e, PGE2 and IFN-β concentrations in BALF of indicated mice 4 weeks p.i. ***P ≤ 0.0005, significant differences compared to pICLC (black)-treated group. Data shown are representative of three independent experiments with a minimum of 4 mice per group.

Extended Data Figure 8 Host-directed therapy with PGE2 and zileuton does not interfere with antibiotic treatment.

Bacterial loads in C3HeB/FeJ mice 43 days p.i. with 100–150 c.f.u. H37Rv, treated or untreated with PGE2 intranasally and zileuton in drinking water. Starting day 24 p.i. antibiotic treatment with rifampicin, pyrazinamide and isoniazid was given 5× a week until 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 one experiment with 5–7 mice per group.

Extended Data Figure 9 IL-10, but not LTB4 or 12/15-LO, is required for type I IFN-driven disease exacerbation of pICLC-treated WT animals infected with Mtb.

a, WT or Ifnar1−/− mice were treated with pICLC or without (PBS) intranasal twice a week, starting day 1 after aerosol challenge with Mtb. Weight loss in experimental groups was normalized to time of moribundity of last remaining mouse in pICLC group (black) for each independent experiment. For pICLC group (black), weight loss is shown for each animal at time of moribundity. Data shown are combined from two individual experiments with 3–7 mice per group. Differences were compared as indicated by lines. b, WT or Alox5−/− 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 per group. Differences were compared as indicated by lines. c, WT or Il10−/− mice were treated with pICLC or without (PBS) intranasally twice a week. For pICLC group (black), weight loss is shown for each animal at time of moribundity. Data shown are representative of two individual experiments with 3–8 mice per group. Differences were compared as indicated by lines. Survival (right panel) of WT or Il10−/− mice treated with (dark grey lines) or without (light grey lines). Data shown are representative of two individual experiments with 4–6 mice per group. d, WT, 12/15-LO−/−, Ltb4r1−/− mice were treated with pICLC or without (PBS) intranasally twice a week. For pICLC groups, weight loss is shown for each animal at time of moribundity. Data shown are representative of two individual experiments with 5–7 mice per group. Differences were compared to PBS controls in each mouse strain. Survival of WT or 12/15-LO−/− mice (middle panel) treated with pICLC (dark blue lines) or without (light blue lines). Data shown are representative of two individual experiments with 4–6 mice per group. Survival of WT or Ltb4r1−/− mice (right panel) treated with pICLC (burgundy lines) or without (brown lines). Data shown are representative of two individual experiments with 4–6 mice per group.

Extended Data Figure 10 Schematic summary of IL-1–type I IFN counter-regulation during Mtb infection.

a, Virulent Mtb directly induces IL-1α and IL-1β as well as type I IFNs in myelophagocytic cells. Whereas IL-1 is required for bacterial control, type I IFNs are considered detrimental because they can exert pro-bacterial effects. During low-dose aerosol infection in WT B6 mice the balance of these two pathways establishes an inflammatory equilibrium that allows for containment of bacilli and chronic infection and thus models certain aspects associated with latency in TB patients. This is achieved by an antagonist relationship whereby type I IFNs inhibit IL-1α/β directly as well as through induction of IL-10 and IL1Ra, while in the opposing direction IL-1α/β limit type 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 bacterial replication and pathology, thus modelling the type I IFN-associated uncontrolled active disease that occurs in a subset of TB patients20,21,22. c, Likewise, experimental induction of type I IFNs by pICLC or viral co-infections33 creates a detrimental environment with uncontrolled bacterial growth and extracellular bacteria, ultimately resulting in loss of host resistance and increased mortality. d, HDT targeting PGE2, either directly or indirectly by enhancing PGE2 levels via 5-LO inhibition with zileuton, returns the system to a non-pathological steady state by limiting type I IFN-driven disease exacerbation, bacterial replication and pulmonary necrosis. Green arrows indicate inductive and red lines inhibitory pathways.

Supplementary information

Supplementary Information

This file contains Supplementary Tables 1-8 containing clinically relevant data and parameters for clinical cohorts. (PDF 322 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mayer-Barber, K., Andrade, B., Oland, S. et al. Host-directed therapy of tuberculosis based on interleukin-1 and type I interferon crosstalk. Nature 511, 99–103 (2014). https://doi.org/10.1038/nature13489

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature13489

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing