Skip to main content
Top
Published in: BMC Musculoskeletal Disorders 1/2016

Open Access 01-12-2016 | Research article

The osteoblast as an inflammatory cell: production of cytokines in response to bacteria and components of bacterial biofilms

Authors: Ulrike Dapunt, Thomas Giese, Sabine Stegmaier, Arash Moghaddam, Gertrud Maria Hänsch

Published in: BMC Musculoskeletal Disorders | Issue 1/2016

Login to get access

Abstract

Background

Implant infections are a major complication in the field of orthopaedics. Bacteria attach to the implant-surface and form biofilm-colonies which makes them difficult to treat. Not only immune cells exclusively respond to bacterial challenges, but also local tissue cells are capable of participating in defense mechanisms. The aim of this study was to evaluate the role of osteoblasts in the context of implant infections.

Methods

Primary osteoblasts were cultivated and stimulated with free-swimming bacteria at 4 °C and 37 °C. Supernatants were harvested for ELISA and expression of pro-inflammatory cytokines evaluated by RT-PCR. Bacterial binding to osteoblasts was evaluated using cytofluorometry and uptake was investigated by 3H thymidine-labelling of bacteria. Osteoblasts were additionally stimulated with the extracellular polymeric substance (EPS) of Staphylococcus epidermidis biofilms, as well as components of the EPS; the bacterial heat shock protein GroEL in particular.

Results

We demonstrated that binding of bacteria to the osteoblast cell surface leads to an increased production of pro-inflammatory cytokines. Bacteria are capable of surviving intracellular. Furthermore, osteoblasts do not only respond to free-swimming, planktonic bacteria, but also to components of the EPS, including lipoteichoic acid and the heat shock protein GroEL.

Conclusion

In conclusion, local tissue cells, specifically osteoblasts, might contribute to the persistence of the inflammatory response associated with implant-infections.
Literature
1.
go back to reference Littlewood AJ, Russell J, Harvey GR, Hughes DE, Russell RG, Gowen M. The modulation of the expression of IL-6 and its receptor in human osteoblasts in vitro. Endocrinology. 1991;129(3):1513–20.CrossRefPubMed Littlewood AJ, Russell J, Harvey GR, Hughes DE, Russell RG, Gowen M. The modulation of the expression of IL-6 and its receptor in human osteoblasts in vitro. Endocrinology. 1991;129(3):1513–20.CrossRefPubMed
2.
go back to reference Littlewood AJ, Aarden LA, Evans DB, Russell RG, Gowen M. Human osteoblastlike cells do not respond to interleukin-6. J Bone Miner Res. 1991;6(2):141–8.CrossRefPubMed Littlewood AJ, Aarden LA, Evans DB, Russell RG, Gowen M. Human osteoblastlike cells do not respond to interleukin-6. J Bone Miner Res. 1991;6(2):141–8.CrossRefPubMed
3.
go back to reference Bost KL, Ramp WK, Nicholson NC, Bento JL, Marriott I, Hudson MC. Staphylococcus aureus infection of mouse or human osteoblasts induces high levels of interleukin-6 and interleukin-12 production. J Infect Dis. 1999;180(6):1912–20.CrossRefPubMed Bost KL, Ramp WK, Nicholson NC, Bento JL, Marriott I, Hudson MC. Staphylococcus aureus infection of mouse or human osteoblasts induces high levels of interleukin-6 and interleukin-12 production. J Infect Dis. 1999;180(6):1912–20.CrossRefPubMed
4.
go back to reference Marriott I, Gray DL, Tranguch SL, Fowler Jr VG, Stryjewski M, Scott Levin L, Hudson MC, Bost KL. Osteoblasts express the inflammatory cytokine interleukin-6 in a murine model of Staphylococcus aureus osteomyelitis and infected human bone tissue. Am J Pathol. 2004;164(4):1399–406.CrossRefPubMedPubMedCentral Marriott I, Gray DL, Tranguch SL, Fowler Jr VG, Stryjewski M, Scott Levin L, Hudson MC, Bost KL. Osteoblasts express the inflammatory cytokine interleukin-6 in a murine model of Staphylococcus aureus osteomyelitis and infected human bone tissue. Am J Pathol. 2004;164(4):1399–406.CrossRefPubMedPubMedCentral
5.
go back to reference Marriott I. Osteoblast responses to bacterial pathogens: a previously unappreciated role for bone-forming cells in host defense and disease progression. Immunol Res. 2004;30(3):291–308.CrossRefPubMed Marriott I. Osteoblast responses to bacterial pathogens: a previously unappreciated role for bone-forming cells in host defense and disease progression. Immunol Res. 2004;30(3):291–308.CrossRefPubMed
6.
7.
go back to reference Schrum LW, Bost KL, Hudson MC, Marriott I. Bacterial infection induces expression of functional MHC class II molecules in murine and human osteoblasts. Bone. 2003;33(5):812–21.CrossRefPubMed Schrum LW, Bost KL, Hudson MC, Marriott I. Bacterial infection induces expression of functional MHC class II molecules in murine and human osteoblasts. Bone. 2003;33(5):812–21.CrossRefPubMed
8.
go back to reference Schrum LW, Marriott I, Butler BR, Thomas EK, Hudson MC, Bost KL. Functional CD40 expression induced following bacterial infection of mouse and human osteoblasts. Infect Immun. 2003;71(3):1209–16.CrossRefPubMedPubMedCentral Schrum LW, Marriott I, Butler BR, Thomas EK, Hudson MC, Bost KL. Functional CD40 expression induced following bacterial infection of mouse and human osteoblasts. Infect Immun. 2003;71(3):1209–16.CrossRefPubMedPubMedCentral
9.
go back to reference Warnke PH, Springer IN, Russo PA, Wiltfang J, Essig H, Kosmahl M, Sherry E, Acil Y. Innate immunity in human bone. Bone. 2006;38(3):400–8.CrossRefPubMed Warnke PH, Springer IN, Russo PA, Wiltfang J, Essig H, Kosmahl M, Sherry E, Acil Y. Innate immunity in human bone. Bone. 2006;38(3):400–8.CrossRefPubMed
10.
go back to reference Hudson MC, Ramp WK, Nicholson NC, Williams AS, Nousiainen MT. Internalization of Staphylococcus aureus by cultured osteoblasts. Microb Pathog. 1995;19(6):409–19.CrossRefPubMed Hudson MC, Ramp WK, Nicholson NC, Williams AS, Nousiainen MT. Internalization of Staphylococcus aureus by cultured osteoblasts. Microb Pathog. 1995;19(6):409–19.CrossRefPubMed
11.
go back to reference Ellington JK, Reilly SS, Ramp WK, Smeltzer MS, Kellam JF, Hudson MC. Mechanisms of Staphylococcus aureus invasion of cultured osteoblasts. Microb Pathog. 1999;26(6):317–23.CrossRefPubMed Ellington JK, Reilly SS, Ramp WK, Smeltzer MS, Kellam JF, Hudson MC. Mechanisms of Staphylococcus aureus invasion of cultured osteoblasts. Microb Pathog. 1999;26(6):317–23.CrossRefPubMed
12.
go back to reference Bayles KW, Wesson CA, Liou LE, Fox LK, Bohach GA, Trumble WR. Intracellular Staphylococcus aureus escapes the endosome and induces apoptosis in epithelial cells. Infect Immun. 1998;66(1):336–42.PubMedPubMedCentral Bayles KW, Wesson CA, Liou LE, Fox LK, Bohach GA, Trumble WR. Intracellular Staphylococcus aureus escapes the endosome and induces apoptosis in epithelial cells. Infect Immun. 1998;66(1):336–42.PubMedPubMedCentral
13.
go back to reference Mohamed W, Sommer U, Sethi S, Domann E, Thormann U, Schutz I, Lips KS, Chakraborty T, Schnettler R, Alt V. Intracellular proliferation of S. aureus in osteoblasts and effects of rifampicin and gentamicin on S. aureus intracellular proliferation and survival. Eur Cell Mater. 2014;28:258–68.PubMed Mohamed W, Sommer U, Sethi S, Domann E, Thormann U, Schutz I, Lips KS, Chakraborty T, Schnettler R, Alt V. Intracellular proliferation of S. aureus in osteoblasts and effects of rifampicin and gentamicin on S. aureus intracellular proliferation and survival. Eur Cell Mater. 2014;28:258–68.PubMed
14.
go back to reference Loffler B, Tuchscherr L, Niemann S, Peters G. Staphylococcus aureus persistence in non-professional phagocytes. Int J Med Microbiol. 2014;304(2):170–6.CrossRefPubMed Loffler B, Tuchscherr L, Niemann S, Peters G. Staphylococcus aureus persistence in non-professional phagocytes. Int J Med Microbiol. 2014;304(2):170–6.CrossRefPubMed
15.
go back to reference Tuchscherr L, Medina E, Hussain M, Volker W, Heitmann V, Niemann S, Holzinger D, Roth J, Proctor RA, Becker K et al. Staphylococcus aureus phenotype switching: an effective bacterial strategy to escape host immune response and establish a chronic infection. EMBO Mol Med. 2011;3(3):129–41.CrossRefPubMedPubMedCentral Tuchscherr L, Medina E, Hussain M, Volker W, Heitmann V, Niemann S, Holzinger D, Roth J, Proctor RA, Becker K et al. Staphylococcus aureus phenotype switching: an effective bacterial strategy to escape host immune response and establish a chronic infection. EMBO Mol Med. 2011;3(3):129–41.CrossRefPubMedPubMedCentral
16.
go back to reference Valour F, Rasigade JP, Trouillet-Assant S, Gagnaire J, Bouaziz A, Karsenty J, Lacour C, Bes M, Lustig S, Benet T et al. Delta-toxin production deficiency in Staphylococcus aureus: a diagnostic marker of bone and joint infection chronicity linked with osteoblast invasion and biofilm formation. Clin Microbiol Infect. 2015;21(6):568 e561-511. Valour F, Rasigade JP, Trouillet-Assant S, Gagnaire J, Bouaziz A, Karsenty J, Lacour C, Bes M, Lustig S, Benet T et al. Delta-toxin production deficiency in Staphylococcus aureus: a diagnostic marker of bone and joint infection chronicity linked with osteoblast invasion and biofilm formation. Clin Microbiol Infect. 2015;21(6):568 e561-511.
17.
go back to reference Costerton JW. Introduction to biofilm. Int J Antimicrob Agents. 1999;11(3–4):217–21. discussion 237–219.CrossRefPubMed Costerton JW. Introduction to biofilm. Int J Antimicrob Agents. 1999;11(3–4):217–21. discussion 237–219.CrossRefPubMed
18.
go back to reference Costerton JW, Stewart PS, Greenberg EP. Bacterial biofilms: a common cause of persistent infections. Science. 1999;284(5418):1318–22.CrossRefPubMed Costerton JW, Stewart PS, Greenberg EP. Bacterial biofilms: a common cause of persistent infections. Science. 1999;284(5418):1318–22.CrossRefPubMed
19.
go back to reference Costerton W, Veeh R, Shirtliff M, Pasmore M, Post C, Ehrlich G. The application of biofilm science to the study and control of chronic bacterial infections. J Clin Invest. 2003;112(10):1466–77.CrossRefPubMedPubMedCentral Costerton W, Veeh R, Shirtliff M, Pasmore M, Post C, Ehrlich G. The application of biofilm science to the study and control of chronic bacterial infections. J Clin Invest. 2003;112(10):1466–77.CrossRefPubMedPubMedCentral
20.
go back to reference Parsek MR, Singh PK. Bacterial biofilms: an emerging link to disease pathogenesis. Annu Rev Microbiol. 2003;57:677–701.CrossRefPubMed Parsek MR, Singh PK. Bacterial biofilms: an emerging link to disease pathogenesis. Annu Rev Microbiol. 2003;57:677–701.CrossRefPubMed
21.
go back to reference Arciola CR. Opportunistic bacteria in implant infections. Knowing them to plan their control. Int J Artif Organs. 2006;29(4):341–2.PubMed Arciola CR. Opportunistic bacteria in implant infections. Knowing them to plan their control. Int J Artif Organs. 2006;29(4):341–2.PubMed
22.
go back to reference Wagner C, Kondella K, Bernschneider T, Heppert V, Wentzensen A, Hansch GM. Post-traumatic osteomyelitis: analysis of inflammatory cells recruited into the site of infection. Shock. 2003;20(6):503–10.CrossRefPubMed Wagner C, Kondella K, Bernschneider T, Heppert V, Wentzensen A, Hansch GM. Post-traumatic osteomyelitis: analysis of inflammatory cells recruited into the site of infection. Shock. 2003;20(6):503–10.CrossRefPubMed
23.
go back to reference Gaida MM, Stegmaier S, Schirmacher P, Wagner C, Hänsch GM. Polymorphonuclear neutrophils in osteomyelitis: link to osteoclast generation and bone resorption. Eur J Inflamm. 2012;10(3):413–26. Gaida MM, Stegmaier S, Schirmacher P, Wagner C, Hänsch GM. Polymorphonuclear neutrophils in osteomyelitis: link to osteoclast generation and bone resorption. Eur J Inflamm. 2012;10(3):413–26.
24.
go back to reference Dapunt U, Giese T, Lasitschka F, Lehner B, Ewerbeck V, Hansch GM. Osteoclast generation and cytokine profile at prosthetic interfaces: a study on tissue of patients with aseptic loosening or implant-associated infections. Eur J Inflamm. 2014;12(1):147–59. Dapunt U, Giese T, Lasitschka F, Lehner B, Ewerbeck V, Hansch GM. Osteoclast generation and cytokine profile at prosthetic interfaces: a study on tissue of patients with aseptic loosening or implant-associated infections. Eur J Inflamm. 2014;12(1):147–59.
25.
go back to reference Meyle E, Brenner-Weiss G, Obst U, Prior B, Hansch GM. Immune defense against S. epidermidis biofilms: components of the extracellular polymeric substance activate distinct bactericidal mechanisms of phagocytic cells. Int J Artif Organs. 2012;35(10):700–12.CrossRefPubMed Meyle E, Brenner-Weiss G, Obst U, Prior B, Hansch GM. Immune defense against S. epidermidis biofilms: components of the extracellular polymeric substance activate distinct bactericidal mechanisms of phagocytic cells. Int J Artif Organs. 2012;35(10):700–12.CrossRefPubMed
26.
go back to reference Maurer S, Meyle E, Prior B, Hänsch GM, Dapunt U. Activation of Neutrophils by the extracellular polymeric substance of Sepidermidis biofilms is mediated by the bacterial heat shock protein Groel. J Biotechnol Biomater. 2015;5(1):176–83. Maurer S, Meyle E, Prior B, Hänsch GM, Dapunt U. Activation of Neutrophils by the extracellular polymeric substance of Sepidermidis biofilms is mediated by the bacterial heat shock protein Groel. J Biotechnol Biomater. 2015;5(1):176–83.
27.
go back to reference Liu H, Fang HH. Extraction of extracellular polymeric substances (EPS) of sludges. J Biotechnol. 2002;95(3):249–56.CrossRefPubMed Liu H, Fang HH. Extraction of extracellular polymeric substances (EPS) of sludges. J Biotechnol. 2002;95(3):249–56.CrossRefPubMed
28.
go back to reference Arciola CR, Bustanji Y, Conti M, Campoccia D, Baldassarri L, Samori B, Montanaro L. Staphylococcus epidermidis-fibronectin binding and its inhibition by heparin. Biomaterials. 2003;24(18):3013–9.CrossRefPubMed Arciola CR, Bustanji Y, Conti M, Campoccia D, Baldassarri L, Samori B, Montanaro L. Staphylococcus epidermidis-fibronectin binding and its inhibition by heparin. Biomaterials. 2003;24(18):3013–9.CrossRefPubMed
29.
30.
go back to reference Poultsides LA, Liaropoulos LL, Malizos KN. The socioeconomic impact of musculoskeletal infections. J Bone Joint Surg Am. 2010;92(11):e13.CrossRefPubMed Poultsides LA, Liaropoulos LL, Malizos KN. The socioeconomic impact of musculoskeletal infections. J Bone Joint Surg Am. 2010;92(11):e13.CrossRefPubMed
31.
go back to reference Tsukayama DT. Pathophysiology of posttraumatic osteomyelitis. Clin Orthop Relat Res. 1999;360:22–9.CrossRefPubMed Tsukayama DT. Pathophysiology of posttraumatic osteomyelitis. Clin Orthop Relat Res. 1999;360:22–9.CrossRefPubMed
32.
go back to reference Hall-Stoodley L, Stoodley P. Evolving concepts in biofilm infections. Cell Microbiol. 2009;11(7):1034–43.CrossRefPubMed Hall-Stoodley L, Stoodley P. Evolving concepts in biofilm infections. Cell Microbiol. 2009;11(7):1034–43.CrossRefPubMed
33.
go back to reference Arciola CR, Alvi FI, An YH, Campoccia D, Montanaro L. Implant infection and infection resistant materials: a mini review. Int J Artif Organs. 2005;28(11):1119–25.PubMed Arciola CR, Alvi FI, An YH, Campoccia D, Montanaro L. Implant infection and infection resistant materials: a mini review. Int J Artif Organs. 2005;28(11):1119–25.PubMed
34.
go back to reference Schommer NN, Christner M, Hentschke M, Ruckdeschel K, Aepfelbacher M, Rohde H. Staphylococcus epidermidis uses distinct mechanisms of biofilm formation to interfere with phagocytosis and activation of mouse macrophage-like cells 774A.1. Infect Immun. 2011;79(6):2267–76.CrossRefPubMedPubMedCentral Schommer NN, Christner M, Hentschke M, Ruckdeschel K, Aepfelbacher M, Rohde H. Staphylococcus epidermidis uses distinct mechanisms of biofilm formation to interfere with phagocytosis and activation of mouse macrophage-like cells 774A.1. Infect Immun. 2011;79(6):2267–76.CrossRefPubMedPubMedCentral
35.
go back to reference Vuong C, Voyich JM, Fischer ER, Braughton KR, Whitney AR, DeLeo FR, Otto M. Polysaccharide intercellular adhesin (PIA) protects Staphylococcus epidermidis against major components of the human innate immune system. Cell Microbiol. 2004;6(3):269–75.CrossRefPubMed Vuong C, Voyich JM, Fischer ER, Braughton KR, Whitney AR, DeLeo FR, Otto M. Polysaccharide intercellular adhesin (PIA) protects Staphylococcus epidermidis against major components of the human innate immune system. Cell Microbiol. 2004;6(3):269–75.CrossRefPubMed
36.
go back to reference Spiliopoulou AI, Krevvata MI, Kolonitsiou F, Harris LG, Wilkinson TS, Davies AP, Dimitracopoulos GO, Karamanos NK, Mack D, Anastassiou ED. An extracellular Staphylococcus epidermidis polysaccharide: relation to Polysaccharide Intercellular Adhesin and its implication in phagocytosis. BMC Microbiol. 2012;12:76.CrossRefPubMedPubMedCentral Spiliopoulou AI, Krevvata MI, Kolonitsiou F, Harris LG, Wilkinson TS, Davies AP, Dimitracopoulos GO, Karamanos NK, Mack D, Anastassiou ED. An extracellular Staphylococcus epidermidis polysaccharide: relation to Polysaccharide Intercellular Adhesin and its implication in phagocytosis. BMC Microbiol. 2012;12:76.CrossRefPubMedPubMedCentral
37.
go back to reference Gunther F, Wabnitz GH, Stroh P, Prior B, Obst U, Samstag Y, Wagner C, Hansch GM. Host defence against Staphylococcus aureus biofilms infection: phagocytosis of biofilms by polymorphonuclear neutrophils (PMN). Mol Immunol. 2009;46(8–9):1805–13.CrossRefPubMed Gunther F, Wabnitz GH, Stroh P, Prior B, Obst U, Samstag Y, Wagner C, Hansch GM. Host defence against Staphylococcus aureus biofilms infection: phagocytosis of biofilms by polymorphonuclear neutrophils (PMN). Mol Immunol. 2009;46(8–9):1805–13.CrossRefPubMed
38.
go back to reference Meyle E, Stroh P, Gunther F, Hoppy-Tichy T, Wagner C, Hansch GM. Destruction of bacterial biofilms by polymorphonuclear neutrophils: relative contribution of phagocytosis, DNA release, and degranulation. Int J Artif Organs. 2010;33(9):608–20.PubMed Meyle E, Stroh P, Gunther F, Hoppy-Tichy T, Wagner C, Hansch GM. Destruction of bacterial biofilms by polymorphonuclear neutrophils: relative contribution of phagocytosis, DNA release, and degranulation. Int J Artif Organs. 2010;33(9):608–20.PubMed
39.
go back to reference Stroh P, Gunther F, Meyle E, Prior B, Wagner C, Hansch GM. Host defence against Staphylococcus aureus biofilms by polymorphonuclear neutrophils: oxygen radical production but not phagocytosis depends on opsonisation with immunoglobulin G. Immunobiology. 2011;216(3):351–7.CrossRefPubMed Stroh P, Gunther F, Meyle E, Prior B, Wagner C, Hansch GM. Host defence against Staphylococcus aureus biofilms by polymorphonuclear neutrophils: oxygen radical production but not phagocytosis depends on opsonisation with immunoglobulin G. Immunobiology. 2011;216(3):351–7.CrossRefPubMed
40.
go back to reference Valour F, Trouillet-Assant S, Rasigade JP, Lustig S, Chanard E, Meugnier H, Tigaud S, Vandenesch F, Etienne J, Ferry T et al. Staphylococcus epidermidis in orthopedic device infections: the role of bacterial internalization in human osteoblasts and biofilm formation. PLoS One. 2013;8(6):e67240.CrossRefPubMedPubMedCentral Valour F, Trouillet-Assant S, Rasigade JP, Lustig S, Chanard E, Meugnier H, Tigaud S, Vandenesch F, Etienne J, Ferry T et al. Staphylococcus epidermidis in orthopedic device infections: the role of bacterial internalization in human osteoblasts and biofilm formation. PLoS One. 2013;8(6):e67240.CrossRefPubMedPubMedCentral
41.
go back to reference van der Flier M, Chhun N, Wizemann TM, Min J, McCarthy JB, Tuomanen EI. Adherence of Streptococcus pneumoniae to immobilized fibronectin. Infect Immun. 1995;63(11):4317–22.PubMedPubMedCentral van der Flier M, Chhun N, Wizemann TM, Min J, McCarthy JB, Tuomanen EI. Adherence of Streptococcus pneumoniae to immobilized fibronectin. Infect Immun. 1995;63(11):4317–22.PubMedPubMedCentral
42.
go back to reference Dapunt U, Giese T, Maurer S, Stegmaier S, Prior B, Hansch GM, Gaida MM: Neutrophil-derived MRP-14 is up-regulated in infectious osteomyelitis and stimulates osteoclast generation. J Leukoc Biol. 2015;98(4):575-82. Dapunt U, Giese T, Maurer S, Stegmaier S, Prior B, Hansch GM, Gaida MM: Neutrophil-derived MRP-14 is up-regulated in infectious osteomyelitis and stimulates osteoclast generation. J Leukoc Biol. 2015;98(4):575-82.
43.
go back to reference Dapunt U, Maurer S, Giese T, Gaida MM, Hansch GM. The macrophage inflammatory proteins MIP1alpha (CCL3) and MIP2alpha (CXCL2) in implant-associated osteomyelitis: linking inflammation to bone degradation. Mediators Inflamm. 2014;2014:728619.CrossRefPubMedPubMedCentral Dapunt U, Maurer S, Giese T, Gaida MM, Hansch GM. The macrophage inflammatory proteins MIP1alpha (CCL3) and MIP2alpha (CXCL2) in implant-associated osteomyelitis: linking inflammation to bone degradation. Mediators Inflamm. 2014;2014:728619.CrossRefPubMedPubMedCentral
Metadata
Title
The osteoblast as an inflammatory cell: production of cytokines in response to bacteria and components of bacterial biofilms
Authors
Ulrike Dapunt
Thomas Giese
Sabine Stegmaier
Arash Moghaddam
Gertrud Maria Hänsch
Publication date
01-12-2016
Publisher
BioMed Central
Published in
BMC Musculoskeletal Disorders / Issue 1/2016
Electronic ISSN: 1471-2474
DOI
https://doi.org/10.1186/s12891-016-1091-y

Other articles of this Issue 1/2016

BMC Musculoskeletal Disorders 1/2016 Go to the issue