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.

  • Letter
  • Published:

Mycobacteria manipulate macrophage recruitment through coordinated use of membrane lipids

Abstract

The evolutionary survival of Mycobacterium tuberculosis, the cause of human tuberculosis, depends on its ability to invade the host, replicate, and transmit infection. At its initial peripheral infection site in the distal lung airways, M. tuberculosis infects macrophages, which transport it to deeper tissues1. How mycobacteria survive in these broadly microbicidal cells is an important question. Here we show in mice and zebrafish that M. tuberculosis, and its close pathogenic relative Mycobacterium marinum, preferentially recruit and infect permissive macrophages while evading microbicidal ones. This immune evasion is accomplished by using cell-surface-associated phthiocerol dimycoceroserate (PDIM) lipids2 to mask underlying pathogen-associated molecular patterns (PAMPs). In the absence of PDIM, these PAMPs signal a Toll-like receptor (TLR)-dependent recruitment of macrophages that produce microbicidal reactive nitrogen species. Concordantly, the related phenolic glycolipids (PGLs)2 promote the recruitment of permissive macrophages through a host chemokine receptor 2 (CCR2)-mediated pathway. Thus, we have identified coordinated roles for PDIM, known to be essential for mycobacterial virulence3, and PGL, which (along with CCR2) is known to be associated with human tuberculosis4,5. Our findings also suggest an explanation for the longstanding observation that M. tuberculosis initiates infection in the relatively sterile environment of the lower respiratory tract, rather than in the upper respiratory tract, where resident microflora and inhaled environmental microbes may continually recruit microbicidal macrophages through TLR-dependent signalling.

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

Access options

Buy this article

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

Figure 1: PDIM-mediated evasion of MyD88-dependent macrophage recruitment.
Figure 2: Increased iNOS-dependent microbicidal activity of macrophages recruited to PDIM-deficient mycobacteria.
Figure 3: Elevated frequencies of iNOS-expressing inflammatory monocytes in mice infected with PDIM-deficient M. tuberculosis.
Figure 4: Macrophage recruitment and subsequent infectivity is mediated by mycobacterial PGL and host CCR2.
Figure 5: MyD88-dependent macrophage recruitment elicited by other bacterial pathogens and commensals attenuates pathogenic mycobacteria.

Similar content being viewed by others

References

  1. Philips, J. A. & Ernst, J. D. Tuberculosis pathogenesis and immunity. Annu. Rev. Pathol. 7, 353–384 (2012)

    Article  CAS  Google Scholar 

  2. Onwueme, K. C., Vos, C. J., Zurita, J., Ferreras, J. A. & Quadri, L. E. N. The dimycocerosate ester polyketide virulence factors of mycobacteria. Prog. Lipid Res. 44, 259–302 (2005)

    Article  CAS  Google Scholar 

  3. Murry, J. P., Pandey, A. K., Sassetti, C. M. & Rubin, E. J. Phthiocerol dimycocerosate transport is required for resisting interferon-γ-independent immunity. J. Infect. Dis. 200, 774–782 (2009)

    Article  CAS  Google Scholar 

  4. Flores-Villanueva, P. O. et al. A functional promoter polymorphism in monocyte chemoattractant protein-1 is associated with increased susceptibility to pulmonary tuberculosis. J. Exp. Med. 202, 1649–1658 (2005)

    Article  CAS  Google Scholar 

  5. Reed, M. B. et al. A glycolipid of hypervirulent tuberculosis strains that inhibits the innate immune response. Nature 431, 84–87 (2004)

    Article  ADS  CAS  Google Scholar 

  6. Medzhitov, R. Recognition of microorganisms and activation of the immune response. Nature 449, 819–826 (2007)

    Article  ADS  CAS  Google Scholar 

  7. Weiser, J. N. The pneumococcus: why a commensal misbehaves. J. Mol. Med. 88, 97–102 (2010)

    Article  ADS  Google Scholar 

  8. Yang, C.-T. et al. Neutrophils exert protection in the early tuberculous granuloma by oxidative killing of mycobacteria phagocytosed from infected macrophages. Cell Host Microbe 12, 301–312 (2012)

    Article  CAS  Google Scholar 

  9. Mayer-Barber, K. D. et al. Cutting edge: caspase-1 independent IL-1 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. Ramakrishnan, L. Revisiting the role of the granuloma in tuberculosis. Nature Rev. Immunol. 12, 352–366 (2012)

    Article  CAS  Google Scholar 

  11. Rosenthal, S. & Tager, I. B. Prevalence of Gram-negative rods in the normal pharyngeal flora. Ann. Intern. Med. 83, 355–357 (1975)

    Article  CAS  Google Scholar 

  12. Wertheim, H. F. L. et al. The role of nasal carriage in Staphylococcus aureus infections. Lancet Infect. Dis. 5, 751–762 (2005)

    Article  Google Scholar 

  13. Eddens, T. & Kolls, J. K. Host defenses against bacterial lower respiratory tract infection. Curr. Opin. Immunol. 24, 424–430 (2012)

    Article  CAS  Google Scholar 

  14. de Mendonça-Lima, L. et al. The allele encoding the mycobacterial Erp protein affects lung disease in mice. Cell. Microbiol. 5, 65–73 (2003)

    Article  Google Scholar 

  15. van der Vaart, M., van Soest, J. J., Spaink, H. P. & Meijer, A. H. Functional analysis of a zebrafish myd88 mutant identifies key transcriptional components of the innate immune system. Dis. Model. Mech. 6, 841–854 (2013)

    Article  CAS  Google Scholar 

  16. 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 

  17. Chan, J., Xing, Y., Magliozzo, R. S. & Bloom, B. R. Killing of virulent Mycobacterium tuberculosis by reactive nitrogen intermediates produced by activated murine macrophages. J. Exp. Med. 175, 1111–1122 (1992)

    Article  CAS  Google Scholar 

  18. Kröncke, K. D., Fehsel, K. & Kolb-Bachofen, V. Inducible nitric oxide synthase in human diseases. Clin. Exp. Immunol. 113, 147–156 (1998)

    Article  Google Scholar 

  19. Serbina, N. V., Jia, T., Hohl, T. M. & Pamer, E. G. Monocyte-mediated defense against microbialpathogens. Annu. Rev. Immunol. 26, 421–452 (2008)

    Article  CAS  Google Scholar 

  20. Antonelli, L. R. V. 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 

  21. Ordway, D. et al. The hypervirulent Mycobacterium tuberculosis strain HN878 induces a potent TH1 response followed by rapid down-regulation. J. Immunol. 179, 522–531 (2007)

    Article  CAS  Google Scholar 

  22. Bates, J. H., Potts, W. E. & Lewis, M. Epidemiology of primary tuberculosis in an industrial school. N. Engl. J. Med. 272, 714–717 (1965)

    Article  CAS  Google Scholar 

  23. Wells, W. F., Ratcliffe, H. L. & Grumb, C. On the mechanics of droplet nuclei infection; quantitative experimental air-borne tuberculosis in rabbits. Am. J. Hyg. 47, 11–28 (1948)

    CAS  PubMed  Google Scholar 

  24. Sinsimer, D. et al. The phenolic glycolipid of Mycobacterium tuberculosis differentially modulates the early host cytokine response but does not in itself confer hypervirulence. Infect. Immun. 76, 3027–3036 (2008)

    Article  CAS  Google Scholar 

  25. Scott, H. M. & Flynn, J. L. Mycobacterium tuberculosis in chemokine receptor 2-deficient mice: influence of dose on disease progression. Infect. Immun. 70, 5946–5954 (2002)

    Article  CAS  Google Scholar 

  26. Feng, W. X. et al. CCL2−2518 (A/G) polymorphisms and tuberculosis susceptibility: a meta-analysis. Int. J. Tuberc. Lung Dis. 16, 150–156 (2012)

    Article  Google Scholar 

  27. Gagneux, S. Variable host-pathogen compatibility in Mycobacterium tuberculosis. Proc. Natl Acad. Sci. USA 103, 2869–2873 (2006)

    Article  ADS  CAS  Google Scholar 

  28. Charlson, E. S. et al. Topographical continuity of bacterial populations in the healthy human respiratory tract. Am. J. Respir. Crit. Care Med. 184, 957–963 (2011)

    Article  Google Scholar 

  29. von Bernuth, H., Picard, C., Puel, A. & Casanova, J.-L. Experimental and natural infections in MyD88- and IRAK-4-deficient mice and humans. Eur. J. Immunol. 42, 3126–3135 (2012)

    Article  CAS  Google Scholar 

  30. Comas, I. et al. Out-of-Africa migration and Neolithic coexpansion of Mycobacterium tuberculosis with modern humans. Nature Genet. 45, 1176–1182 (2013)

    Article  CAS  Google Scholar 

  31. Cosma, C. L., Klein, K., Kim, R., Beery, D. & Ramakrishnan, L. Mycobacterium marinum Erp is a virulence determinant required for cell wall integrity and ntracellular survival. Infect. Immun. 74, 3125–3133 (2006)

    Article  CAS  Google Scholar 

  32. Takaki, K., Cosma, C. L., Troll, M. A. & Ramakrishnan, L. An in vivo platform for rapid high-throughput antitubercular drug discovery. Cell Rep. 2, 175–184 (2012)

    Article  CAS  Google Scholar 

  33. Brannon, M. K. et al. Pseudomonas aeruginosa Type III secretion system interacts with phagocytes to modulate systemic infection of zebrafish embryos. Cell. Microbiol. 11, 755–768 (2009)

    Article  CAS  Google Scholar 

  34. Yu, J. et al. Both phthiocerol dimycocerosates and phenolic glycolipids are required for virulence of Mycobacterium marinum. Infect. Immun. 80, 1381–1389 (2012)

    Article  CAS  Google Scholar 

  35. Takaki, K., Davis, J. M., Winglee, K. & Ramakrishnan, L. Evaluation of the pathogenesis and treatment of Mycobacterium marinum infection in zebrafish. Nature Protocols 8, 1114–1124 (2013)

    Article  Google Scholar 

  36. Davis, J. M. & Ramakrishnan, L. The role of the granuloma in expansion and dissemination of early tuberculous infection. Cell 136, 37–49 (2009)

    Article  CAS  Google Scholar 

  37. Clay, H. et al. Dichotomous role of the macrophage in early Mycobacterium marinum infection of the zebrafish. Cell Host Microbe 2, 29–39 (2007)

    Article  CAS  Google Scholar 

  38. Lepiller, S. et al. Imaging of nitric oxide in a living vertebrate using a diaminofluorescein probe. Free Radic. Biol. Med. 43, 619–627 (2007)

    Article  CAS  Google Scholar 

  39. Maruyama, I. N., Rakow, T. L. & Maruyama, H. I. cRACE: a simple method for identification of the 5′ end of mRNAs. Nucleic Acids Res. 23, 3796–3797 (1995)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank S. Falkow and P. Edelstein for sharing their knowledge and insights, P. Donald for discussions about human infectivity in tuberculosis, B. Cormack for manuscript review and editing, J. Bubeck-Wardenberg for the fluorescent S. aureus strain, K. Hicks for initial MyD88 experiments, T.-Y. Chen, B. Moody, P. Manzanillo and J. Cox for help with lipid analyses, and J. Cameron for fish facility management. Supported by a National Science Foundation predoctoral fellowship to C.J.C., a Senior Research Training Fellowship from the American Lung Association and the National Institutes of Health (NIH) Training Grant “Training Clinical and Basic Immunologists” to R.P.L., a NIH “Academic Pediatric Infectious Disease” Training Grant award to R.E.H., an American Cancer Society Postdoctoral Fellowship and NIH Bacterial Pathogenesis Training Grant Award to D.M.T., and NIH grants to K.B.U. and L.R. D.M.T. is a recipient of the NIH Director’s New Innovator Award and L.R. is a recipient of the NIH Director’s Pioneer Award.

Author information

Authors and Affiliations

Authors

Contributions

C.J.C., C.L.C., K.K.T., D.M.T., R.E.H. and L.R. conceived and designed M. marinum/zebrafish experiments and analysed data; C.J.C., C.L.C., K.K.T., D.M.T. and R.E.H. performed these experiments; R.P.L. and K.B.U. designed the M. tuberculosis/mouse experiments and analysed the data; R.P.L. performed the mouse experiments; C.J.C., C.L.C. and L.R. wrote the paper; C.J.C., R.P.L. and K.K.T. prepared the figures; and all authors edited the paper.

Corresponding author

Correspondence to Lalita Ramakrishnan.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Coordinate use of PDIM-mediated immune evasion and PGL-mediated recruitment by pathogenic mycobacteria.

Models for infection with wild-type (WT) and PDIM-deficient mycobacteria are shown in the context of the relatively sterile lower airway versus the upper airway, with its higher levels of resident microflora and inhaled environmental organisms.

Extended Data Figure 2 ΔmmpL7 bacteria are attenuated in zebrafish larvae.

a, Kaplan–Meier graph showing daily survival of larvae infected via caudal vein injection with medium (mock), 29 wild-type or 70 ΔmmpL7 M. marinum. N = 25 (mock), 31 (wild-type), or 29 (ΔmmpL7) larvae per group. Mean time to death (days): mock (11), wild type (7.6) and ΔmmpL7 (11.2). Survival was compared by log-rank test: wild type versus mock and wild type versus ΔmmpL7, P < 0.0001; mock versus ΔmmpL7, P = 0.5601. b, c, Larvae were infected via caudal vein injection 1 dpf with 550 wild-type, 650 ΔmmpL7, or 700Δerp, fluorescent M. marinum. b, Infection burdens were measured by Fluorescent Pixel Count (FPC; mean ± s.e.m.). c, Representative images at 7 dpi. N = 29 (wild-type and ΔmmpL7) or 30 (Δerp) larvae per group. Scale bar, 500 μm. At 3, 5 and 7 dpi, log10 FPC was compared by ANOVA, with Dunnett’s post-test. ***P < 0.001. d, e, Representative images from wild-type (d) and ΔmmpL7 (e) M. marinum HBV infections quantified in Fig. 1d. N = 18 (wild-type) or 16 (ΔmmpL7) larvae per group. HBVs are outlined with a dashed white line. Scale bar, 100 µm.

Extended Data Figure 3 Knockdown of MyD88 results in a late, dose-dependent hypersusceptibility to M. marinum systemic infection.

a, RT–PCR for actin (top) and myd88 (bottom), demonstrating that that the majority of myd88 transcripts at 7 dpf are abnormal in MyD88 morphants. Lanes marked ‘b’ and ‘c’ correspond to morphants from the same experiments depicted in panels b and c, respectively. The abnormal larger transcript (indicated by an asterisk) results from the inclusion of intron 2 in the final transcript, incorporating a premature stop codon that truncates the protein before the TIR (Toll/interleukin receptor) domain. b, c, Caudal vein infection of MyD88 morphants with 141 (b) or 325 (c) c.f.u. M. marinum/larva. Bacterial burden was assessed by FPC, values plotted represent the mean ± s.e.m. Time points were compared by one-way ANOVA and Bonferroni’s post-tests. *** P < 0.001. d, Representative images of larvae at 5 dpi from experiment in c, N = 30 control, 15 MyD88 morphant. Scale bar, 500 μm.

Extended Data Figure 4 Characteristics of macrophages recruited to wild-type and PDIM-deficient bacteria.

a, Mean Mpeg1-positive macrophages recruited at 3 hpi into the HBV of wild-type fish after infection with 80 wild-type or ΔmmpL7 M. marinum. b, Data from Fig. 2c expressed as mean numbers of total infected macrophages and iNOS-expressing infected macrophages after HBV infection with 80 wild-type, ΔmmpL7, or Δerp M. marinum. c, Bacterial burdens after L-NAME treatment. Mean bacterial burdens of 2 dpf control (CTRL)- or iNOS inhibitor (L-NAME)-treated fish after HBV infection with 80 wild-type or ΔmmpL7 M. marinum. NS, not significant.

Extended Data Figure 5 Wild-type bacterial burdens after co-infection with wild-type or ΔmmpL7 bacteria.

Representative images from the HBV co-infections quantified in Fig. 2e. a, b, Red fluorescent wild-type (WT) M. marinum co-infected with green fluorescent wild-type (a) or ΔmmpL7 (b) M. marinum. N = 18 (wild-type) and 19 (ΔmmpL7) larvae per group. Scale bar, 50 µm. c, Wild-type bacterial burdens after co-infection with wild-type or ΔmmpL7 M. marinum with and without L-NAME treatment. Significance tested by one-way ANOVA with Bonferroni’s post-test for comparisons shown.

Extended Data Figure 6 MyD88-dependent macrophage recruitment occurs in response to PDIM deficiency rather than being due to loss of another MmpL7-exported product.

a, Mean macrophage recruitment at 3 hpi into the HBV of wild-type or MyD88-morphant (MO) larvae after infection with 80 Δmas M. marinum. Student’s unpaired t-test. b, Mean surviving bacterial volume of red fluorescent wild-type M. marinum (initial infection dose of 30–40 c.f.u.) when co-infected with 30–40 green fluorescent wild-type, ΔmmpL7 or Δmas M. marinum at 3 dpi. Representative of two separate experiments. Significance tested by one-way ANOVA with Tukey’s post-test.

Extended Data Figure 7 Gating strategy and isotype controls for iNOS staining of mouse lung.

a, Representative gating strategy for isolation of inflammatory monocytes. A dump channel containing anti-CD4, CD8 and CD11c was plotted against a channel exhibiting autofluorescence and also containing anti-Ly6G. Using these markers, T cell, dendritic cell, alveolar macrophage, and neutrophil cell populations were excluded from the double-negative gate. Inflammatory monocytes were identified within the double-negative population by their co-expression of Ly6C and CD11b. These cells were then evaluated for intracellular iNOS expression. a, N = 4 per group (Fig. 3a, b) or b, with isotype control antibodies, N = 4 per group.

Extended Data Figure 8 Specificity of CCL2-mediated macrophage recruitment in wild-type and CCR2-morphant larvae

a, Mean macrophage recruitment at 3 hpi into the HBV of control (ctrl), or CCR2-morphant (CCR2) larvae after injection of vehicle control (‘mock’; 0.1% BSA in PBS), human CCL2 (hCCL2), human CCL4 (hCCL4), or human CCL5 (hCCL5). b, c, Mean macrophage (b) and neutrophil (c) recruitment at 3 hpi into the HBV of control (CTRL), CCR2-morphant (CCR2), or MyD88-morphant (MyD88) larvae after injection of vehicle control (mock), murine CCL2 (mCCL2), human CCL2 (hCCL2), human IL-8 (hIL-8), or human LTB4 (hLTB4). Representative of three separate experiments. Significance assessed by one-way ANOVA with Bonferroni’s post-test for the comparisons shown. *P < 0.05; ***P < 0.001.

Extended Data Figure 9 Identification of zebrafish CCL2 orthologue.

a, mRNA levels of potential CCL2 orthologues (mean ± s.e.m. of four biological replicates) induced at 3 h after caudal vein infection of 2 dpf larvae with 250–300 wild-type M. marinum. These assays were performed on the same cDNA pools as the data presented in Fig. 4b. b, Mean macrophage recruitment at 3 hpi into the HBV of wild-type or CCL2 morphant (MO) fish following infection with 80 M. marinum. Representative of two separate experiments.

Extended Data Figure 10 Infectivity assay.

a, b, Representative 5 hpi images from Fig. 4d following HBV infection with one (a) or three (b) M. marinum. Scale bar, 100 µm. N values for fish represented in a and b (that is, those found to be infected with 1–3 bacteria) are presented in Fig. 4d (18, 22, 28, 28, 28, 28, 22, 22 for the respective conditions as specified in the figure). c, Mean bacterial burdens 5 h after HBV infection with 1–3 wild-type (WT), ΔmmpL7 or Δpks15 M. marinum.

Supplementary information

Supplementary Tables

This file contains Supplementary Table 1. (PDF 244 kb)

Mpeg1+ cells recruited towards WT Mycobacterium marinum

Red Mpeg1+ cells in the HBV, 1-9 hours post infection with wasabi-expressing WT M. marinum, imaged every three minutes. (MOV 10564 kb)

Mpeg1+ cells recruited towards ∆mmpL7 Mycobacterium marinum

Red Mpeg1+ cells in the HBV, 1-9 hours post infection with wasabi-expressing ∆mmpL7 M. marinum, imaged every three minutes. (MOV 10536 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Cambier, C., Takaki, K., Larson, R. et al. Mycobacteria manipulate macrophage recruitment through coordinated use of membrane lipids. Nature 505, 218–222 (2014). https://doi.org/10.1038/nature12799

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature12799

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 Microbiology

Sign up for the Nature Briefing: Microbiology newsletter — what matters in microbiology research, free to your inbox weekly.

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