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.

  • Article
  • Published:

Lactate production by Staphylococcus aureus biofilm inhibits HDAC11 to reprogramme the host immune response during persistent infection

Abstract

Staphylococcus aureus is a leading cause of biofilm-associated prosthetic joint infection (PJI), resulting in considerable disability and prolonged treatment. It is known that host leukocyte IL-10 production is required for S. aureus biofilm persistence in PJI. An S. aureus bursa aurealis Tn library consisting of 1,952 non-essential genes was screened for mutants that failed to induce IL-10 in myeloid-derived suppressor cells (MDSCs), which identified a critical role for bacterial lactic acid biosynthesis. We generated an S. aureus ddh/ldh1/ldh2 triple Tn mutant that cannot produce d- or l-lactate. Co-culture of MDSCs or macrophages with ddh/ldh1/ldh2 mutant biofilm produced substantially less IL-10 compared with wild-type S. aureus, which was also observed in a mouse model of PJI and led to reduced biofilm burden. Using MDSCs recovered from the mouse PJI model and in vitro leukocyte–biofilm co-cultures, we show that bacterial-derived lactate inhibits histone deacetylase 11, causing unchecked HDAC6 activity and increased histone 3 acetylation at the Il-10 promoter, resulting in enhanced Il-10 transcription in MDSCs and macrophages. Finally, we show that synovial fluid of patients with PJI contains elevated amounts of d-lactate and IL-10 compared with control subjects, and bacterial lactate increases IL-10 production by human monocyte-derived macrophages.

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

Fig. 1: Lactate biosynthesis pathways induce MDSC and macrophage IL-10 production.
Fig. 2: Role for Ddh, Ldh1 and Ldh2 during S. aureus orthopaedic implant infection.
Fig. 3: S. aureus lactate promotes IL-10 production and regulates leukocyte infiltrates during PJI.
Fig. 4: S. aureus-derived lactate inhibits HDAC activity to regulate histone 3 acetylation and gene expression.
Fig. 5: S. aureus-derived lactate inhibits the negative regulator HDAC11 to augment leukocyte IL-10 production in an HDAC6-dependent manner.
Fig. 6: S. aureus-derived lactate induces IL-10 production by human monocyte-derived macrophages.

Similar content being viewed by others

Data availability

The ChIP–seq and RNA-seq datasets are available in the GEO repository (accession number GSE135496). Source data are provided with this paper.

Code availability

All codes used are published programs, with citations for each provided in the references.

References

  1. Percival, S. L., Suleman, L., Vuotto, C. & Donelli, G. Healthcare-associated infections, medical devices and biofilms: risk, tolerance and control. J. Med. Microbiol. 64, 323–334 (2015).

    PubMed  Google Scholar 

  2. Pulido, L., Ghanem, E., Joshi, A., Purtill, J. J. & Parvizi, J. Periprosthetic joint infection: the incidence, timing, and predisposing factors. Clin. Orthop. Relat. Res. 466, 1710–1715 (2008).

    PubMed  PubMed Central  Google Scholar 

  3. Arciola, C. R., Campoccia, D. & Montanaro, L. Implant infections: adhesion, biofilm formation and immune evasion. Nat. Rev. Microbiol. 16, 397–409 (2018).

    CAS  PubMed  Google Scholar 

  4. Flemming, H. C. et al. Biofilms: an emergent form of bacterial life. Nat. Rev. Microbiol. 14, 563–575 (2016).

    CAS  PubMed  Google Scholar 

  5. Balaban, N. Q. et al. Definitions and guidelines for research on antibiotic persistence. Nat. Rev. Microbiol. 17, 441–448 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Cameron, D. R., Shan, Y., Zalis, E. A., Isabella, V. & Lewis, K. A genetic determinant of persister cell formation in bacterial pathogens. J. Bacteriol. 200, e00303-18 (2018).

    PubMed  PubMed Central  Google Scholar 

  7. Scherr, T. D. et al. Staphylococcus aureus biofilms induce macrophage dysfunction through leukocidin AB and alpha-toxin. mBio 6, e01021-15 (2015).

    PubMed  PubMed Central  Google Scholar 

  8. Koziel, J. et al. The Janus face of ɑ-toxin: a potent mediator of cytoprotection in staphylococci-infected macrophages. J. Innate Immun. 7, 187–198 (2015).

    CAS  PubMed  Google Scholar 

  9. Schommer, N. N. et al. Staphylococcus epidermidis uses distinct mechanisms of biofilm formation to interfere with phagocytosis and activation of mouse macrophage-like cells 774A.1. Infect. Immun. 79, 2267–2276 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Thurlow, L. R. et al. Staphylococcus aureus biofilms prevent macrophage phagocytosis and attenuate inflammation in vivo. J. Immunol. 186, 6585–6596 (2011).

    CAS  PubMed  Google Scholar 

  11. Ricciardi, B. F. et al. Staphylococcus aureus evasion of host immunity in the setting of prosthetic joint infection: biofilm and beyond. Curr. Rev. Musculoskelet. Med. 11, 389–400 (2018).

    PubMed  PubMed Central  Google Scholar 

  12. Le, K. Y., Park, M. D. & Otto, M. Immune evasion mechanisms of Staphylococcus epidermidis biofilm infection. Front. Microbiol. 9, 359 (2018).

    PubMed  PubMed Central  Google Scholar 

  13. He, L. et al. Resistance to leukocytes ties benefits of quorum sensing dysfunctionality to biofilm infection. Nat. Microbiol. 4, 1114–1119 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Heim, C. E. et al. Myeloid-derived suppressor cells contribute to Staphylococcus aureus orthopedic biofilm infection. J. Immunol. 192, 3778–3792 (2014).

    CAS  PubMed  Google Scholar 

  15. Heim, C. E. et al. IL-12 promotes myeloid-derived suppressor cell recruitment and bacterial persistence during Staphylococcus aureus orthopedic implant infection. J. Immunol. 194, 3861–3872 (2015).

    CAS  PubMed  Google Scholar 

  16. Heim, C. E., Vidlak, D. & Kielian, T. Interleukin-10 production by myeloid-derived suppressor cells contributes to bacterial persistence during Staphylococcus aureus orthopedic biofilm infection. J. Leukoc. Biol. 98, 1003–1013 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Tebartz, C. et al. A major role for myeloid-derived suppressor cells and a minor role for regulatory T cells in immunosuppression during Staphylococcus aureus infection. J. Immunol. 194, 1100–1111 (2015).

    CAS  PubMed  Google Scholar 

  18. Bernthal, N. M. et al. Protective role of IL-1β against post-arthroplasty Staphylococcus aureus infection. J. Orthop. Res. 29, 1621–1626 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Ouyang, W. & O’Garra, A. IL-10 family cytokines IL-10 and IL-22: from basic science to clinical translation. Immunity 50, 871–891 (2019).

    CAS  PubMed  Google Scholar 

  20. Kessler, B. et al. Interleukin 10 inhibits pro-inflammatory cytokine responses and killing of Burkholderia pseudomallei. Sci. Rep. 7, 42791 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Leech, J. M., Lacey, K. A., Mulcahy, M. E., Medina, E. & McLoughlin, R. M. IL-10 plays opposing roles during Staphylococcus aureus systemic and localized infections. J. Immunol. 198, 2352–2365 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Alter, G. et al. IL-10 induces aberrant deletion of dendritic cells by natural killer cells in the context of HIV infection. J. Clin. Invest. 120, 1905–1913 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Smith, L. K. et al. Interleukin-10 directly inhibits CD8+ T cell function by enhancing N-glycan branching to decrease antigen sensitivity. Immunity 48, 299–312 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Akdis, C. A., Joss, A., Akdis, M., Faith, A. & Blaser, K. A molecular basis for T cell suppression by IL-10: CD28-associated IL-10 receptor inhibits CD28 tyrosine phosphorylation and phosphatidylinositol 3-kinase binding. FASEB J. 14, 1666–1668 (2000).

    CAS  PubMed  Google Scholar 

  25. Liu, B., Tonkonogy, S. L. & Sartor, R. B. Antigen-presenting cell production of IL-10 inhibits T-helper 1 and 17 cell responses and suppresses colitis in mice. Gastroenterology 141, 653–662 (2011).

    CAS  PubMed  Google Scholar 

  26. Sinha, P., Clements, V. K., Bunt, S. K., Albelda, S. M. & Ostrand-Rosenberg, S. Cross-talk between myeloid-derived suppressor cells and macrophages subverts tumor immunity toward a type 2 response. J. Immunol. 179, 977–983 (2007).

    CAS  PubMed  Google Scholar 

  27. Beury, D. W. et al. Cross-talk among myeloid-derived suppressor cells, macrophages, and tumor cells impacts the inflammatory milieu of solid tumors. J. Leukoc. Biol. 96, 1109–1118 (2014).

    PubMed  PubMed Central  Google Scholar 

  28. Fey, P. D. et al. A genetic resource for rapid and comprehensive phenotype screening of nonessential Staphylococcus aureus genes. mBio 4, e00537-12 (2013).

    PubMed  PubMed Central  Google Scholar 

  29. Fuller, J. R. et al. Identification of a lactate-quinone oxidoreductase in Staphylococcus aureus that is essential for virulence. Front. Cell. Infect. Microbiol. 1, 19 (2011).

    PubMed  PubMed Central  Google Scholar 

  30. Stockland, A. E. & San Clemente, C. L. Multiple forms of lactate dehydrogenase in Staphylococcus aureus. J. Bacteriol. 100, 347–353 (1969).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Kondoh, Y., Kawase, M., Kawakami, Y. & Ohmori, S. Concentrations of d-lactate and its related metabolic intermediates in liver, blood, and muscle of diabetic and starved rats. Res. Exp. Med. (Berl.) 192, 407–414 (1992).

    CAS  Google Scholar 

  32. Puig-Kroger, A. et al. Peritoneal dialysis solutions inhibit the differentiation and maturation of human monocyte-derived dendritic cells: effect of lactate and glucose-degradation products. J. Leukoc. Biol. 73, 482–492 (2003).

    CAS  PubMed  Google Scholar 

  33. Husain, Z., Huang, Y., Seth, P. & Sukhatme, V. P. Tumor-derived lactate modifies antitumor immune response: effect on myeloid-derived suppressor cells and NK cells. J. Immunol. 191, 1486–1495 (2013).

    CAS  PubMed  Google Scholar 

  34. Zhang, D. et al. Metabolic regulation of gene expression by histone lactylation. Nature 574, 575–580 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Ratter, J. M. et al. In vitro and in vivo effects of lactate on metabolism and cytokine production of human primary PBMCs and monocytes. Front. Immunol. 9, 2564 (2018).

    PubMed  PubMed Central  Google Scholar 

  36. Latham, T. et al. Lactate, a product of glycolytic metabolism, inhibits histone deacetylase activity and promotes changes in gene expression. Nucleic Acids Res. 40, 4794–4803 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Wagner, W., Ciszewski, W. M. & Kania, K. D. l- and d-lactate enhance DNA repair and modulate the resistance of cervical carcinoma cells to anticancer drugs via histone deacetylase inhibition and hydroxycarboxylic acid receptor 1 activation. Cell Commun. Signal. 13, 36 (2015).

    PubMed  PubMed Central  Google Scholar 

  38. Strahl, B. D. & Allis, C. D. The language of covalent histone modifications. Nature 403, 41–45 (2000).

    CAS  PubMed  Google Scholar 

  39. Shakespear, M. R., Halili, M. A., Irvine, K. M., Fairlie, D. P. & Sweet, M. J. Histone deacetylases as regulators of inflammation and immunity. Trends Immunol. 32, 335–343 (2011).

    CAS  PubMed  Google Scholar 

  40. Cheng, F. et al. Divergent roles of histone deacetylase 6 (HDAC6) and histone deacetylase 11 (HDAC11) on the transcriptional regulation of IL10 in antigen presenting cells. Mol. Immunol. 60, 44–53 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Villagra, A. et al. The histone deacetylase HDAC11 regulates the expression of interleukin 10 and immune tolerance. Nat. Immunol. 10, 92–100 (2009).

    CAS  PubMed  Google Scholar 

  42. Garvie, E. I. Bacterial lactate dehydrogenases. Microbiol. Rev. 44, 106–139 (1980).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Gaspar, P., Al-Bayati, F. A., Andrew, P. W., Neves, A. R. & Yesilkaya, H. Lactate dehydrogenase is the key enzyme for pneumococcal pyruvate metabolism and pneumococcal survival in blood. Infect. Immun. 82, 5099–5109 (2014).

    PubMed  PubMed Central  Google Scholar 

  44. Bunch, P. K., Mat-Jan, F., Lee, N. & Clark, D. P. The ldhA gene encoding the fermentative lactate dehydrogenase of Escherichia coli. Microbiology 143, 187–195 (1997).

    CAS  PubMed  Google Scholar 

  45. Feldman-Salit, A. et al. Regulation of the activity of lactate dehydrogenases from four lactic acid bacteria. J. Biol. Chem. 288, 21295–21306 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Spahich, N. A., Vitko, N. P., Thurlow, L. R., Temple, B. & Richardson, A. R. Staphylococcus aureus lactate- and malate-quinone oxidoreductases contribute to nitric oxide resistance and virulence. Mol. Microbiol. 100, 759–773 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Bola, B. M. et al. Inhibition of monocarboxylate transporter-1 (MCT1) by AZD3965 enhances radiosensitivity by reducing lactate transport. Mol. Cancer Ther. 13, 2805–2816 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Wang, Q. et al. Characterization of monocarboxylate transport in human kidney HK-2 cells. Mol. Pharm. 3, 675–685 (2006).

    CAS  PubMed  Google Scholar 

  49. Polanski, R. et al. Activity of the monocarboxylate transporter 1 inhibitor AZD3965 in small cell lung cancer. Clin. Cancer Res. 20, 926–937 (2014).

    CAS  PubMed  Google Scholar 

  50. Corbet, C. et al. Interruption of lactate uptake by inhibiting mitochondrial pyruvate transport unravels direct antitumor and radiosensitizing effects. Nat. Commun. 9, 1208 (2018).

    PubMed  PubMed Central  Google Scholar 

  51. Garcia-Castillo, V. et al. Targeting metabolic remodeling in triple negative breast cancer in a murine model. J. Cancer 8, 178–189 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Zhao, Z., Han, F., Yang, S., Wu, J. & Zhan, W. Oxamate-mediated inhibition of lactate dehydrogenase induces protective autophagy in gastric cancer cells: involvement of the Akt-mTOR signaling pathway. Cancer Lett. 358, 17–26 (2015).

    CAS  PubMed  Google Scholar 

  53. Zhai, X., Yang, Y., Wan, J., Zhu, R. & Wu, Y. Inhibition of LDH-A by oxamate induces G2/M arrest, apoptosis and increases radiosensitivity in nasopharyngeal carcinoma cells. Oncol. Rep. 30, 2983–2991 (2013).

    CAS  PubMed  Google Scholar 

  54. Askarian, F., Wagner, T., Johannessen, M. & Nizet, V. Staphylococcus aureus modulation of innate immune responses through Toll-like (TLR), (NOD)-like (NLR) and C-type lectin (CLR) receptors. FEMS Microbiol. Rev. 42, 656–671 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Hanzelmann, D. et al. Toll-like receptor 2 activation depends on lipopeptide shedding by bacterial surfactants. Nat. Commun. 7, 12304 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Kawasaki, T. & Kawai, T. Toll-like receptor signaling pathways. Front. Immunol. 5, 461 (2014).

    PubMed  PubMed Central  Google Scholar 

  57. Larsson, L., Thorbert-Mros, S., Rymo, L. & Berglundh, T. Influence of epigenetic modifications of the interleukin-10 promoter on IL10 gene expression. Eur. J. Oral Sci. 120, 14–20 (2012).

    CAS  PubMed  Google Scholar 

  58. Yuan, Z. L., Guan, Y. J., Chatterjee, D. & Chin, Y. E. Stat3 dimerization regulated by reversible acetylation of a single lysine residue. Science 307, 269–273 (2005).

    CAS  PubMed  Google Scholar 

  59. Qin, G. et al. Metformin blocks myeloid-derived suppressor cell accumulation through AMPK-DACH1-CXCL1 axis. Oncoimmunology 7, e1442167 (2018).

    PubMed  PubMed Central  Google Scholar 

  60. Ouzounova, M. et al. Monocytic and granulocytic myeloid derived suppressor cells differentially regulate spatiotemporal tumour plasticity during metastatic cascade. Nat. Commun. 8, 14979 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Zhang, C. X. et al. STING signaling remodels the tumor microenvironment by antagonizing myeloid-derived suppressor cell expansion. Cell Death Differ. 26, 2314–2328 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Sundaram, K. et al. IκBζ regulates human monocyte pro-inflammatory responses induced by Streptococcus pneumoniae. PLoS ONE 11, e0161931 (2016).

    PubMed  PubMed Central  Google Scholar 

  63. Katoh, H. et al. CXCR2-expressing myeloid-derived suppressor cells are essential to promote colitis-associated tumorigenesis. Cancer Cell 24, 631–644 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Li, W., Mao, Z., Fan, X., Cui, L. & Wang, X. Cyclooxygenase 2 promoted the tumorigenecity of pancreatic cancer cells. Tumour Biol. 35, 2271–2278 (2014).

    CAS  PubMed  Google Scholar 

  65. Lucas, M., Zhang, X., Prasanna, V. & Mosser, D. M. ERK activation following macrophage FcγR ligation leads to chromatin modifications at the IL-10 locus. J. Immunol. 175, 469–477 (2005).

    CAS  PubMed  Google Scholar 

  66. Turner, B. M. Cellular memory and the histone code. Cell 111, 285–291 (2002).

    CAS  PubMed  Google Scholar 

  67. Youn, J. I. et al. Epigenetic silencing of retinoblastoma gene regulates pathologic differentiation of myeloid cells in cancer. Nat. Immunol. 14, 211–220 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Rosborough, B. R., Castellaneta, A., Natarajan, S., Thomson, A. W. & Turnquist, H. R. Histone deacetylase inhibition facilitates GM-CSF-mediated expansion of myeloid-derived suppressor cells in vitro and in vivo. J. Leukoc. Biol. 91, 701–709 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Sahakian, E. et al. Histone deacetylase 11: a novel epigenetic regulator of myeloid derived suppressor cell expansion and function. Mol. Immunol. 63, 579–585 (2015).

    CAS  PubMed  Google Scholar 

  70. Eskandarian, H. A. et al. A role for SIRT2-dependent histone H3K18 deacetylation in bacterial infection. Science 341, 1238858 (2013).

    PubMed  Google Scholar 

  71. Kincaid, E. Z. & Ernst, J. D. Mycobacterium tuberculosis exerts gene-selective inhibition of transcriptional responses to IFN-γ without inhibiting STAT1 function. J. Immunol. 171, 2042–2049 (2003).

    CAS  PubMed  Google Scholar 

  72. Wang, Y., Curry, H. M., Zwilling, B. S. & Lafuse, W. P. Mycobacteria inhibition of IFN-γ induced HLA-DR gene expression by up-regulating histone deacetylation at the promoter region in human THP-1 monocytic cells. J. Immunol. 174, 5687–5694 (2005).

    CAS  PubMed  Google Scholar 

  73. Pathak, S. K. et al. TLR4-dependent NF-κB activation and mitogen- and stress-activated protein kinase 1-triggered phosphorylation events are central to Helicobacter pylori peptidyl prolyl cis-, trans-isomerase (HP0175)-mediated induction of IL-6 release from macrophages. J. Immunol. 177, 7950–7958 (2006).

    CAS  PubMed  Google Scholar 

  74. Cao, J. et al. HDAC11 regulates type I interferon signaling through defatty-acylation of SHMT2. Proc. Natl Acad. Sci. USA 116, 5487–5492 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Moreno-Yruela, C., Galleano, I., Madsen, A. S. & Olsen, C. A. Histone deacetylase 11 is an ε-N-myristoyllysine hydrolase. Cell Chem. Biol. 25, 849–856 e848 (2018).

    CAS  PubMed  Google Scholar 

  76. Kutil, Z. et al. Histone deacetylase 11 is a fatty-acid deacylase. ACS Chem. Biol. 13, 685–693 (2018).

    CAS  PubMed  Google Scholar 

  77. Korkmaz, B., Horwitz, M. S., Jenne, D. E. & Gauthier, F. Neutrophil elastase, proteinase 3, and cathepsin G as therapeutic targets in human diseases. Pharm. Rev. 62, 726–759 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Lau, D. et al. Myeloperoxidase mediates neutrophil activation by association with CD11b/CD18 integrins. Proc. Natl Acad. Sci. USA 102, 431–436 (2005).

    CAS  PubMed  Google Scholar 

  79. Kumar, V., Patel, S., Tcyganov, E. & Gabrilovich, D. I. The nature of myeloid-derived suppressor cells in the tumor microenvironment. Trends Immunol. 37, 208–220 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Obermajer, N. & Kalinski, P. Key role of the positive feedback between PGE(2) and COX2 in the biology of myeloid-derived suppressor cells. Oncoimmunology 1, 762–764 (2012).

    PubMed  PubMed Central  Google Scholar 

  81. Dufait, I. et al. Perforin and granzyme B expressed by murine myeloid-derived suppressor cells: a study on their role in outgrowth of cancer cells. Cancers (Basel) 11, 808 (2019).

    CAS  Google Scholar 

  82. Sawant, A. et al. Myeloid-derived suppressor cells function as novel osteoclast progenitors enhancing bone loss in breast cancer. Cancer Res. 73, 672–682 (2013).

    CAS  PubMed  Google Scholar 

  83. Heim, C. E. et al. Human prosthetic joint infections are associated with myeloid-derived suppressor cells (MDSCs): implications for infection persistence. J. Orthop. Res. 36, 1605–1613 (2018).

    CAS  PubMed  Google Scholar 

  84. Manning Fox, J. E., Meredith, D. & Halestrap, A. P. Characterisation of human monocarboxylate transporter 4 substantiates its role in lactic acid efflux from skeletal muscle. J. Physiol. 529, 285–293 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Vitko, N. P., Grosser, M. R., Khatri, D., Lance, T. R. & Richardson, A. R. Expanded glucose import capability affords Staphylococcus aureus optimized glycolytic flux during infection. mBio 7, e00296-16 (2016).

    PubMed  PubMed Central  Google Scholar 

  86. Vitko, N. P., Spahich, N. A. & Richardson, A. R. Glycolytic dependency of high-level nitric oxide resistance and virulence in Staphylococcus aureus. mBio 6, e00045-15 (2015).

    PubMed  PubMed Central  Google Scholar 

  87. Yermak, K., Karbysheva, S., Perka, C., Trampuz, A. & Renz, N. Performance of synovial fluid d-lactate for the diagnosis of periprosthetic joint infection: a prospective observational study. J. Infect. 79, 123–129 (2019).

    PubMed  Google Scholar 

  88. Zhang, Q. & Cao, X. Epigenetic regulation of the innate immune response to infection. Nat. Rev. Immunol. 19, 417–432 (2019).

    CAS  PubMed  Google Scholar 

  89. Turner, N. A. et al. Methicillin-resistant Staphylococcus aureus: an overview of basic and clinical research. Nat. Rev. Microbiol. 17, 203–218 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Sun, L. et al. Loss of HDAC11 ameliorates clinical symptoms in a multiple sclerosis mouse model. Life Sci. Alliance 1, e201800039 (2018).

    PubMed  PubMed Central  Google Scholar 

  91. Mootz, J. M., Malone, C. L., Shaw, L. N. & Horswill, A. R. Staphopains modulate Staphylococcus aureus biofilm integrity. Infect. Immun. 81, 3227–3238 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Gries, C. M. et al. Cyclic di-AMP released from Staphylococcus aureus biofilm induces a macrophage type I interferon response. Infect. Immun. 84, 3564–3574 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Yamada, K. J. et al. Arginase-1 expression in myeloid cells regulates Staphylococcus aureus planktonic but not biofilm infection. Infect. Immun. 86, e00206-18 (2018).

    PubMed  PubMed Central  Google Scholar 

  94. Heim, C. E., West, S. C., Ali, H. & Kielian, T. Heterogeneity of Ly6G+ Ly6C+ myeloid-derived suppressor cell infiltrates during Staphylococcus aureus biofilm infection. Infect. Immun. 86, e00684-18 (2018).

    PubMed  PubMed Central  Google Scholar 

  95. Niska, J. A. et al. Vancomycin-rifampin combination therapy has enhanced efficacy against an experimental Staphylococcus aureus prosthetic joint infection. Antimicrob. Agents Chemother. 57, 5080–5086 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Pribaz, J. R. et al. Mouse model of chronic post-arthroplasty infection: noninvasive in vivo bioluminescence imaging to monitor bacterial burden for long-term study. J. Orthop. Res. 30, 335–340 (2012).

    PubMed  Google Scholar 

  97. Niska, J. A. et al. Monitoring bacterial burden, inflammation and bone damage longitudinally using optical and muCT imaging in an orthopaedic implant infection in mice. PLoS ONE 7, e47397 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Yamada, K. J. et al. Monocyte metabolic reprogramming promotes pro-inflammatory activity and Staphylococcus aureus biofilm clearance. PLoS Pathog. 16, e1008354 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Picard Toolkit (Broad Institute, GitHub repository, 2019); http://broadinstitute.github.io/picard/

  101. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    PubMed  PubMed Central  Google Scholar 

  102. Allhoff, M., Sere, K., J, F. P., Zenke, M. & I, G. C. Differential peak calling of ChIP-seq signals with replicates with THOR. Nucleic Acids Res. 44, e153 (2016).

    PubMed  PubMed Central  Google Scholar 

  103. Allhoff, M. et al. Detecting differential peaks in ChIP-seq signals with ODIN. Bioinformatics 30, 3467–3475 (2014).

    CAS  PubMed  Google Scholar 

  104. Ramírez, F. et al. deepTools: a flexible platform for exploring deep-sequencing data. Nucleic Acids Res. 42, W187–W191 (2014).

    PubMed  PubMed Central  Google Scholar 

  105. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Lin, X., Tirichine, L. & Bowler, C. Protocol: chromatin immunoprecipitation (ChIP) methodology to investigate histone modifications in two model diatom species. Plant Methods 8, 48 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Pfaffl, M. W. A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res. 29, e45 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Trim Galore (Babraham Institute, GitHub repository, 2019); https://github.com/FelixKrueger/TrimGalore

  109. Trapnell, C., Pachter, L. & Salzberg, S. L. TopHat: discovering splice junctions with RNA-seq. Bioinformatics 25, 1105–1111 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Trapnell, C. et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat. Protocols 7, 562–578 (2012).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by the National Institutes of Health/National Institute of Allergy and Infectious Diseases grant no. P01AI083211 (Project 4 to T.K.) and grant no. R01AI125588 (to V.C.T.). The authors thank R. Fallet for managing the mouse colony. The UNMC DNA Sequencing Core receives partial support from the National Institute for General Medical Science (grant nos. INBRE–P20GM103427-14 and COBRE–1P30GM110768-01). Both the UNMC DNA Sequencing and Flow Cytometry Research Cores receive support from The Fred & Pamela Buffett Cancer Center Support Grant (grant no. P30CA036727).

Author information

Authors and Affiliations

Authors

Contributions

C.E.H. and T.K. conceived the study; C.E.H., M.E.B., K.J.Y., A.L.A. and T.K. designed experiments; and A.R.K. and D.K. provided expertise in the design, execution and data analysis for the ChIP–seq, ChIP–PCR and scRNA-seq experiments. S.S.C., A.A.A., C.M.G. and V.C.T. created the S. aureus lactate mutants used in the study. C.E.H., M.E.B., K.J.Y. and A.L.A. conducted the in vivo mouse PJI experiments. C.E.H. performed the in vitro biofilm–leukocyte co-culture experiments. E.S. and Y.L. provided the HDAC11 KO mice. C.E.H., D.K. and T.K. performed data analysis. T.K. procured funding for this work. C.E.H. and T.K. wrote the manuscript. All authors edited and approved the submission of this work.

Corresponding author

Correspondence to Tammy Kielian.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 S. aureus lactate mutants do not display growth defects in liquid broth or biofilm in vitro.

a, S. aureus strains used in this study. b, The growth rate of S. aureus WT, Δddh, Δldh1/ldh2, and Δddh/ldh1/ldh2 was determined in brain-heart infusion broth over a 24 h period with constant agitation using a TECAN (7 biological replicates/strain). c, Strains were transduced with a sarA-GFP plasmid and grown for 4 days under static growth conditions in RPMI-1640 supplemented with 10% FBS, whereupon biofilm formation was visualized by confocal microscopy. Results are representative of two independent experiments, each with 4 biological replicates. Scale bars, 100 µm.

Source data

Extended Data Fig. 2 Intracellular pH of MDSCs and macrophages is not dramatically altered by S. aureus-derived lactate during biofilm co-culture.

MDSCs or macrophages were labeled with BCECF-AM (10 µM) prior to co-culture with WT (n = 4 biological replicates/group) or Δddh/ldh1/ldh2 (n = 4 and 3 biological replicates for MDSCs and macrophages, respectively) biofilm for 2 h, whereupon intracellular pH was determined by flow cytometry based on a standard curve of known pH. Results shown are from one experiment.

Source data

Extended Data Fig. 3 D- and L-lactate production during S. aureus orthopaedic infection.

(a) L- and (b) D-lactate were quantified in the implant-associated tissue of mice infected with WT, Δddh, Δldh1/ldh2, or Δddh/ldh1/ldh2 at days 3, 14, and 28 post-infection (mean ± SD; n = 5/group). The dashed line represents background in the assay as determined with tissues collected from animals receiving sterile implants at the same time points (n = 5 at days 3 and 28 and n = 4 at day 14). Results are representative of three independent experiments. *, p < 0.05; **, p < 0.01; ***, p < 0.001; One-way ANOVA.

Source data

Extended Data Fig. 4 The expression of select inflammatory mediators is independent of bacterial burden during S. aureus orthopaedic infection.

Cytokine levels were quantified in implant-associated tissue of mice infected with WT, Δddh/ldh1/ldh2, or a 1:1 ratio of WT and Δddh/ldh1/ldh2 at days 3, 14, and 28 post-infection. Results are combined from two independent experiments (mean ± SD; n = 8/group). *, p < 0.05; One-way ANOVA.

Source data

Extended Data Fig. 5 IL-10 production during S. aureus orthopaedic infection is not influenced by host lactate.

Mice received daily i.p. injections of sodium oxamate (500 mg/kg/day) dissolved in 0.5% Hydroxypropyl Methylcellulose or vehicle (0.5% Hydroxypropyl Methylcellulose) beginning one day prior to infection with S. aureus WT (n = 10/group) or Δddh/ldh1/ldh2 (n = 10 or 9 for vehicle and oxamate, respectively). Mice were killed at day 14 post-infection to quantify (a) D-lactate, (b) L-lactate, and (c) IL-10 in implant-associated tissue. Results are combined from two independent experiments (mean ± SD). D- and L-lactate measurements are also reported at day 14 for mice that received sterile implants (n = 4/group). *, p < 0.05; **, p < 0.01; ***, p < 0.001; One-way ANOVA. NS, not significant.

Source data

Extended Data Fig. 6 Sodium oxamate does not affect S. aureus growth or lactate production.

a, D- and (b) L-lactate were quantified in S. aureus WT and Δddh/ldh1/ldh2 biofilm in 96-well plates under static growth conditions in RPMI-1640 supplemented with 10% FBS over a 4 d period. The dotted lines represent daily medium changes. Results are from one experiment with 5 biological replicates. S. aureus was exposed to various concentrations of sodium oxamate during (c) growth in liquid broth (brain-heart infusion) beginning at time 0 (n = 6 biological replicates/group) or (d) throughout the 4-day biofilm maturation period (n = 5 biological replicates/group). Biofilm cultures were replenished daily with fresh medium (RPMI-1640 + 10% FBS) containing sodium oxamate. Results are presented as (c) OD600 or (d) Log10 colony forming units (CFU) per well. e, Quantification of D- and L-lactate from biofilm throughout the 4-day growth period, where the dotted lines represent daily medium changes (n = 4 biological replicates/group). All results are reported as mean ± SD.

Source data

Extended Data Fig. 7 Effects of S. aureus lactate on orthopaedic implant biofilm infection are IL-10-dependent.

WT and IL-10 KO mice were infected with WT (n = 10) or Δddh/ldh1/ldh2 (n = 9) S. aureus, whereupon bacterial burden in (a) implant-associated tissue and (b) femur as well as (c) MDSC and (d) monocyte infiltrates were assessed at day 14 post-infection. Results represent the mean ± SEM of two independent experiments. *, p < 0.05; **, p < 0.01; ***, p < 0.001; One-way ANOVA.

Source data

Extended Data Fig. 8 S. aureus-derived lactate inhibits the negative regulator HDAC11 to augment leukocyte IL-10 production in a HDAC6-dependent manner.

MDSCs and macrophages from WT or HDAC11 KO mice were co-cultured with (a) WT or (b) Δddh/ldh1/ldh2 biofilm for 2 h ± HDAC6i (36 nM). IL-10 production was measured by cytometric bead array. Results represent the mean ± SEM of two independent experiments (n = 8 and 12 biological replicates for MDSCs and macrophages, respectively). **, p < 0.01; ***, p < 0.001; ****, p < 0.0001; One-way ANOVA. Values for untreated leukocytes are the same as those presented in Fig. 5a,b because both HDAC6i and tubastatin A were tested at the same time.

Source data

Extended Data Fig. 9 S. aureus-derived lactate preferentially inhibits HDAC11.

Purified active HDAC11 or HDAC6 were exposed to conditioned medium from WT or Δddh/ldh1/ldh2 biofilm for 30 min, whereupon HDAC activity was determined using a fluorescent HDAC substrate (deAc-FdL). Results are from one experiment with 4 biological replicates and are expressed as the percent change in HDAC activity compared to purified enzyme. **, p < 0.01; ***, p < 0.001; ****, p < 0.0001; One-way ANOVA. NS, not significant.

Source data

Extended Data Fig. 10 Gating strategy to quantitate leukocyte populations in S. aureus implant-associated soft tissue.

Single cells were gated from the (a) total events using (b) FSC-A vs. FSC-H, followed by (c) exclusion of dead cells. (d) Live, CD45+ leukocytes were separated into (e) Ly6G+Ly6C+ vs. Ly6GLy6C+. (f) MDSC and neutrophil populations were identified based on CD11b expression, while (g) monocyte and macrophage populations were identified based on Ly6C and F4/80 expression, respectively.

Supplementary information

Supplementary Information

Supplementary Figs. 1–4 and Tables 1 and 2.

Reporting Summary

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Heim, C.E., Bosch, M.E., Yamada, K.J. et al. Lactate production by Staphylococcus aureus biofilm inhibits HDAC11 to reprogramme the host immune response during persistent infection. Nat Microbiol 5, 1271–1284 (2020). https://doi.org/10.1038/s41564-020-0756-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-020-0756-3

This article is cited by

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