Hypoxia and P. gingivalis synergistically induce HIF-1 and NF-κB activation in PDL cells and periodontal diseases

L Gölz, S Memmert, B Rath-Deschner, A Jäger, T Appel, G Baumgarten, W Götz, S Frede, L Gölz, S Memmert, B Rath-Deschner, A Jäger, T Appel, G Baumgarten, W Götz, S Frede

Abstract

Periodontitis is characterized by deep periodontal pockets favoring the proliferation of anaerobic bacteria like Porphyromonas gingivalis (P. gingivalis), a periodontal pathogen frequently observed in patients suffering from periodontal inflammation. Therefore, the aim of the present study was to investigate the signaling pathways activated by lipopolysaccharide (LPS) of P. gingivalis (LPS-PG) and hypoxia in periodontal ligament (PDL) cells. The relevant transcription factors nuclear factor-kappa B (NF-κB) and hypoxia inducible factor-1 (HIF-1) were determined. In addition, we analyzed the expression of interleukin- (IL-) 1β, matrix metalloproteinase-1 (MMP-1), and vascular endothelial growth factor (VEGF) in PDL cells on mRNA and protein level. This was accomplished by immunohistochemistry of healthy and inflamed periodontal tissues. We detected time-dependent additive effects of LPS-PG and hypoxia on NF-κB and HIF-1α activation in PDL cells followed by an upregulation of IL-1β, MMP-1, and VEGF expression. Immunohistochemistry performed on tissue samples of gingivitis and periodontitis displayed an increase of NF-κB, HIF-1, and VEGF immunoreactivity in accordance with disease progression validating the importance of the in vitro results. To conclude, the present study underlines the significance of NF-κB and HIF-1α and their target genes VEGF, IL-1β, and MMP-1 in P. gingivalis and hypoxia induced periodontal inflammatory processes.

Figures

Figure 1
Figure 1
Induction of IL-1β mRNA in PDL cells. PDL cells were cultured under normoxic or hypoxic condition and stimulated with LPS-PG (1 μg/mL). Cells cultured in normoxia without LPS stimulation served as control. IL-1β mRNA expression was analyzed by real-time PCR after 2, 4, 8, 24, and 48 h. Statistical differences were analyzed by one-way ANOVA and Dunnett's test as well as Tukey's multiple comparison test; *P < 0.05 indicates a significant difference between the groups (means ± SD; n = 9).
Figure 2
Figure 2
Expression of MMP-1 in PDL cells. (a) PDL cells were cultured under normoxic or hypoxic condition and stimulated with LPS-PG (1 μg/mL). Untreated cells cultured in normoxia served as control. MMP-1 mRNA expression was quantitated by real-time PCR after 2, 4, 8, 24, and 48 h. Statistical differences were analyzed by one-way ANOVA followed by different post hoc tests (Dunnett's and Tukey's multiple comparison test); a P value < 0.05 was assumed as significant. #P < 0.05 depicts a significant difference with respect to the time-matched control, *P < 0.05 indicates a significant difference between groups (means ± SD; n = 9). (b) MMP-1 protein secretion by PDL fibroblasts was determined using the Quantikine enzyme-linked immunoabsorbent assay (ELISA) system according to the manufacturer's instruction. Supernatants of PDL cells stimulated with or without LPS-PG (1 μg/mL) under normoxic or hypoxic condition were collected after varying time points (2, 4, 8, 24, and 48 h). Statistical differences were analyzed by one-way ANOVA followed by different post hoc tests (Dunnett's and Tukey's multiple comparison test); a P value < 0.05 was assumed as significant. *P < 0.05 indicates a significant difference between groups (means ± SD; n = 9).
Figure 3
Figure 3
Activation of NF-κB. (a) NF-κB was visualized by immunofluorescence staining. PDL cells were cultured under normoxic (control) or hypoxic condition (Hox) and stimulated with LPS-PG (1 μg/mL). NF-κB activation was visible as enhanced nuclear staining. Representative pictures of control cells and a 1 hour LPS-PG treatment under hypoxic conditions are shown. The scale bars indicate 100 μm. (b) Density of NF-κB nuclear staining after 1, 2, and 4 h treatment was determined in relation to total cell area using the freely available image-processing software ImageJ 1.43 (http://rsb.info.nih.gov/ij). Statistical analysis of the processed immunofluorescence data were performed by one-way ANOVA followed by Dunnett's test and Tukey's multiple comparison test; a P value < 0.05 was assumed as significant. #P < 0.05 depicts a significant difference with respect to time-matched control, *P < 0.05 indicates a significant difference between groups (means ± SD; n = 6). (c) Tissue samples from healthy gingiva (I), gingivitis (II), healthy periodontal ligament (III), and periodontitis (IV) were obtained after approval of the Ethics Committee of the University of Bonn and parental as well as patients' written consent (n = 3). Polyclonal primary antibody raised against the p65 subunit of NF-κB was used in a concentration of 1 : 100 for immunohistochemistry. We observed an increase in NF-κB immunostaining in accordance to the progression of periodontal inflammation. Arrows indicate the nuclear staining of NF-κB in endothelial cells as well as immune cells in tissue samples of periodontitis. The scale bars indicate 50, 100, or 200 μm in dependence of the magnifications.
Figure 4
Figure 4
Activation of HIF-1α. (a) PDL cells were incubated for the indicated time periods with LPS-PG under normoxic or hypoxic conditions. HIF-1α mRNA expression was quantitated by real-time PCR, and expression levels were calculated as n-fold induction of the respective time matched untreated normoxic control. Statistical differences were analyzed by one-way ANOVA followed by different post hoc tests (Dunnett's and Tukey's multiple comparison test); #P < 0.05 depicts a significant difference with respect to time-matched control, *P < 0.05 indicates a significant difference between groups (means ± SD; n = 6). (b) PDL cells were cultured under normoxic (control) or hypoxic conditions (Hox) and stimulated with LPS-PG (1 μg/mL). HIF-1α protein was visualized by immunofluorescence staining. Representative pictures of cells under normoxic control conditions and after a 2-hour LPS-PG treatment under hypoxic conditions are shown. The scale bars indicate 100 μm. (c) PDL cells were incubated for 1, 2, and 4 hours. HIF-1α protein was detected by immunofluorescence. Density of nuclear HIF-1α staining was determined as in relation to total cell area using the freely available image-processing software ImageJ 1.43 (http://rsb.info.nih.gov/ij). Statistical analysis of the processed data were performed by one-way ANOVA followed by Tukey's multiple comparison test; *P < 0.05 indicates a significant difference between groups (means ± SD; n = 6). (d) Tissue samples from healthy gingiva (I), gingivitis (II), healthy periodontal ligament (III), and periodontitis (IV) were obtained after approval of the Ethics Committee of the University of Bonn and parental as well as patients' written consent (n = 3). Polyclonal primary antibody raised against HIF-1 was used in a concentration of 1 : 100 for immunohistochemistry. We observed an increase of HIF-1α immunostaining in accordance to the progression of periodontal inflammation. The scale bars indicate 50, 100, or 200 μm in dependence of the magnifications.
Figure 5
Figure 5
VEGF protein expression. (a) VEGF protein expression in PDL cells was measured using ELISA. Supernatants of PDL cells, stimulated with or without LPS-PG (1 μg/mL) under normoxic or hypoxic condition, were collected after varying time points (2, 4, 8, 24, and 48 h). Statistical differences were analyzed by one-way ANOVA and the post hoc Dunnett's and Tukey's multiple comparison test; #P < 0.05 depicts a significant difference with respect to the time-matched control, *P < 0.05 indicates a significant difference between groups (means ± SD; n = 9). (b) Healthy gingiva (I), gingivitis (II), healthy periodontal ligament (III), and periodontitis (IV) were obtained after the approval of the Ethics Committee of the University of Bonn and parental as well as patients' written consent (n = 3). A polyclonal primary antibody against VEGF was used in a concentration of 1 : 100 for immunohistochemistry. The scale bars indicate 50, 100, or 200 μm in dependence of the magnifications.

References

    1. Holtfreter B., Kocher T., Hoffmann T., Desvarieux M., Micheelis W. Prevalence of periodontal disease and treatment demands based on a German dental survey (DMS IV) Journal of Clinical Periodontology. 2010;37(3):211–219. doi: 10.1111/j.1600-051x.2009.01517.x.
    1. Thornton-Evans G., Eke P., Wei L., et al. Centers for disease control and prevention (CDC). Periodontitis among adults aged ≥30 years—United States, 2009-2010. MMWR Surveillance Summaries. 2013;62(supplement 3):129–135.
    1. Roth G. A., Moser B., Roth-Walter F., et al. Infection with a periodontal pathogen increases mononuclear cell adhesion to human aortic endothelial cells. Atherosclerosis. 2007;190(2):271–281. doi: 10.1016/j.atherosclerosis.2006.03.018.
    1. Meyer M. S., Joshipura K., Giovannucci E., Michaud D. S. A review of the relationship between tooth loss, periodontal disease, and cancer. Cancer Causes and Control. 2008;19(9):895–907. doi: 10.1007/s10552-008-9163-4.
    1. Hosomi N., Aoki S., Matsuo K., et al. Association of serum anti-periodontal pathogen antibody with ischemic stroke. Cerebrovascular Diseases. 2012;34(5-6):385–392. doi: 10.1159/000343659.
    1. Hayashi C., Gudino C. V., Gibson F. C., III, Genco C. A. Pathogen-induced inflammation at sites distant from oral infection: bacterial persistence and induction of cell-specific innate immune inflammatory pathways. Molecular Oral Microbiology. 2010;25(5):305–316. doi: 10.1111/j.2041-1014.2010.00582.x.
    1. Tonetti M. S., van Dyke T. E. Periodontitis and atherosclerotic cardiovascular disease: consensus report of the Joint EFP/AAP Workshop on Periodontitis and Systemic Diseases. Journal of Clinical Periodontology. 2013;40(14, supplement):S24–S29. doi: 10.1111/jcpe.12089.
    1. Laine M. L., Loos B. G., Crielaard W. Gene polymorphisms in chronic periodontitis. International Journal of Dentistry. 2010;2010:22. doi: 10.1155/2010/324719.324719
    1. Socransky S. S., Haffajee A. D., Cugini M. A., Smith C., Kent R. L., Jr. Microbial complexes in subgingival plaque. Journal of Clinical Periodontology. 1998;25(2):134–144. doi: 10.1111/j.1600-051x.1998.tb02419.x.
    1. Murphy K. M., Travers P., Walport M., editors. Janeway Immunobiology. 7th. Garland Science; 2007.
    1. Kawai T., Akira S. The role of pattern-recognition receptors in innate immunity: update on toll-like receptors. Nature Immunology. 2010;11(5):373–384. doi: 10.1038/ni.1863.
    1. Medzhitov R., Preston-Hurlburt P., Janeway C. A., Jr. A human homologue of the Drosophila toll protein signals activation of adaptive immunity. Nature. 1997;388(6640):394–397. doi: 10.1038/41131.
    1. Darveau R. P., Pham T.-T. T., Lemley K., et al. Porphyromonas gingivalis lipopolysaccharide contains multiple lipid A species that functionally interact with both toll-like receptors 2 and 4. Infection and Immunity. 2004;72(9):5041–5051. doi: 10.1128/iai.72.9.5041-5051.2004.
    1. Yoshimura A., Kaneko T., Kato Y., Golenbock D. T., Hara Y. Lipopolysaccharides from periodontopathic bacteria Porphyromonas gingivalis and Capnocytophaga ochracea are antagonists for human toll-like receptor 4. Infection and Immunity. 2002;70(1):218–225. doi: 10.1128/iai.70.1.218-225.2002.
    1. Hayden M. S., Ghosh S. Shared principles in NF-kappaB signaling. Cell. 2008;132(3):344–362. doi: 10.1016/j.cell.2008.01.020.
    1. Oeckinghaus A., Hayden M. S., Ghosh S. Crosstalk in NF-κB signaling pathways. Nature Immunology. 2011;12(8):695–708. doi: 10.1038/ni.2065.
    1. Gölz L., Memmert S., Rath-Deschner B., et al. LPS from P. gingivalis and hypoxia increases oxidative stress in periodontal ligament fibroblasts and contributes to periodontitis. Mediators of Inflammation. 2014;2014:13. doi: 10.1155/2014/986264.986264
    1. Frede S., Freitag P., Otto T., Heilmaier C., Fandrey J. The proinflammatory cytokine interleukin 1β and hypoxia cooperatively induce the expression of adrenomedullin in ovarian carcinoma cells through hypoxia inducible factor 1 activation. Cancer Research. 2005;65(11):4690–4697. doi: 10.1158/0008-5472.can-04-3877.
    1. Frede S., Stockmann C., Freitag P., Fandrey J. Bacterial lipopolysaccharide induces HIF-1 activation in human monocytes via p44/42 MAPK and NF-κB. Biochemical Journal. 2006;396(3):517–527. doi: 10.1042/BJ20051839.
    1. Frede S., Stockmann C., Winning S., Freitag P., Fandrey J. Hypoxia-Inducible Factor (HIF) 1α accumulation and HIF target gene expression are impaired after induction of endotoxin tolerance. Journal of Immunology. 2009;182(10):6470–6476. doi: 10.4049/jimmunol.0802378.
    1. Höpfl G., Ogunshola O., Gassmann M. HIFs and tumors-causes and consequences. The American Journal of Physiology—Regulatory Integrative and Comparative Physiology. 2004;286(4):R608–R623. doi: 10.1152/ajpregu.00538.2003.
    1. Bonello S., Zähringer C., BelAiba R. S., et al. Reactive oxygen species activate the HIF-1α promoter via a functional NFκB site. Arteriosclerosis, Thrombosis, and Vascular Biology. 2007;27(4):755–761. doi: 10.1161/01.atv.0000258979.92828.bc.
    1. Rath-Deschner B., Deschner J., Reimann S., Jager A., Gotz W. Regulatory effects of biomechanical strain on the insulin-like growth factor system in human periodontal cells. Journal of Biomechanics. 2009;42(15):2584–2589. doi: 10.1016/j.jbiomech.2009.07.013.
    1. Chomczynski P., Sacchi N. Single-step method of RNA isolation by acid guanidinium thiocyanate-phenol-chloroform extraction. Analytical Biochemistry. 1987;162(1):156–159.
    1. Kassebaum N. J., Bernabe E., Dahiya M., Bhandari B., Murray C. J. L., Marcenes W. Global burden of severe periodontitis in 1990–2010: a systematic review and meta-regression. Journal of Dental Research. 2014;93(11):1045–1053. doi: 10.1177/0022034514552491.
    1. Bainbridge B. W., Coats S. R., Darveau R. P. Porphyromonas gingivalis lipopolysaccharide displays functionally diverse interactions with the innate host defense system. Annals of Periodontology. 2002;7(1):29–37. doi: 10.1902/annals.2002.7.1.29.
    1. Li J.-P., Li F. Y. L., Xu A., et al. Lipopolysaccharide and hypoxia-induced HIF-1 activation in human gingival fibroblasts. Journal of Periodontology. 2012;83(6):816–824. doi: 10.1902/jop.2011.110458.
    1. Ng K. T., Li J. P., Ng K. M., Tipoe G. L., Leung W. K., Fung M. L. Expression of hypoxia-inducible factor-1α in human periodontal tissue. Journal of Periodontology. 2011;82(1):136–141. doi: 10.1902/jop.2010.100100.
    1. Jian C., Li C., Ren Y., et al. Hypoxia augments lipopolysaccharide-induced cytokine expression in periodontal ligament cells. Inflammation. 2014;37(5):1413–1423. doi: 10.1007/s10753-014-9865-6.
    1. Piccioli P., Rubartelli A. The secretion of IL-1β and options for release. Seminars in Immunology. 2013;25(6):425–429. doi: 10.1016/j.smim.2013.10.007.
    1. Song G. G., Kim J.-H., Lee Y. H. Toll-like receptor (TLR) and matrix metalloproteinase (MMP) polymorphisms and periodontitis susceptibility: a meta-analysis. Molecular Biology Reports. 2013;40(8):5129–5141. doi: 10.1007/s11033-013-2616-1.
    1. Zhou J., Windsor L. J. Porphyromonas gingivalis affects host collagen degradation by affecting expression, activation, and inhibition of matrix metalloproteinases. Journal of Periodontal Research. 2006;41(1):47–54. doi: 10.1111/j.1600-0765.2005.00835.x.
    1. Sapna G., Gokul S., Bagri-Manjrekar K. Matrix metalloproteinases and periodontal diseases. Oral Diseases. 2014;20(6):538–550. doi: 10.1111/odi.12159.
    1. Li W., Xiao L., Hu J. Matrix metalloproteinase-1 promoter -1607 1G/2G polymorphism and chronic periodontitis susceptibility: a meta-analysis and systematic review. Journal of Clinical Periodontology. 2013;40(12):1095–1103. doi: 10.1111/jcpe.12166.
    1. Kraus D., Winter J., Jepsen S., Jäger A., Meyer R., Deschner J. Interactions of adiponectin and lipopolysaccharide from Porphyromonas gingivalis on human oral epithelial cells. PLoS ONE. 2012;7(2) doi: 10.1371/journal.pone.0030716.e30716
    1. Kong L., Qi X., Huang S., Chen S., Wu Y., Zhao L. Theaflavins inhibits pathogenic properties of P. gingivalis and MMPs production in P. gingivalis-stimulated human gingival fibroblasts. Archives of Oral Biology. 2015;60(1):12–22. doi: 10.1016/j.archoralbio.2014.08.019.
    1. Song A.-M., Hou C., Chen J. F., Sun J., Tian T., Li S. Effect of hypoxia on the expression of matrix metalloproteinase and tissue inhibitors of matrix metalloproteinase mRNA in human periodontal ligament fibroblasts in vitro. Zhonghua Kou Qiang Yi Xue Za Zhi. 2012;47(10):599–604.
    1. Sempere M. C., Fanjul V. R., Pérez I. S., Perona R. The role of the NFκB signalling pathway in cancer. Clinical and Translational Oncology. 2008;10(3):143–147. doi: 10.1007/s12094-008-0171-3.
    1. Lentsch A. B., Ward P. A. Activation and regulation of NFkappaB during acute inflammation. Clinical Chemistry and Laboratory Medicine. 1999;37(3):205–208.
    1. Winning S., Splettstoesser F., Fandrey J., Frede S. Acute hypoxia induces HIF-independent monocyte adhesion to endothelial cells through increased intercellular adhesion molecule-1 expression: the role of hypoxic inhibition of prolyl hydroxylase activity for the induction of NF-κB. Journal of Immunology. 2010;185(3):1786–1793. doi: 10.4049/jimmunol.0903244.
    1. Oliver K. M., Taylor C. T., Cummins E. P. Regulation of NFκB signalling during inflammation: the role of hydroxylases. Arthritis Research and Therapy. 2009;11(1, article 215) doi: 10.1186/ar2575.
    1. Rius J., Guma M., Schachtrup C., et al. NF-κB links innate immunity to the hypoxic response through transcriptional regulation of HIF-1α . Nature. 2008;453(7196):807–811. doi: 10.1038/nature06905.
    1. Luo D., Wang Z., Wu J., Jiang C., Wu J. The role of hypoxia inducible factor-1 in hepatocellular carcinoma. BioMed Research International. 2014;2014:11. doi: 10.1155/2014/409272.409272
    1. Cheng Z.-X., Wang D.-W., Liu T., et al. Effects of the HIF-1α and NF-κB loop on epithelial-mesenchymal transition and chemoresistance induced by hypoxia in pancreatic cancer cells. Oncology Reports. 2014;31(4):1891–1898. doi: 10.3892/or.2014.3022.
    1. Ramakrishnan S., Anand V., Roy S. Vascular endothelial growth factor signaling in hypoxia and inflammation. Journal of Neuroimmune Pharmacology. 2014;9(2):142–160. doi: 10.1007/s11481-014-9531-7.
    1. Goddard L. M., Luisa Iruela-Arispe M. Cellular and molecular regulation of vascular permeability. Thrombosis and Haemostasis. 2013;109(3):407–415. doi: 10.1160/TH12-09-0678.
    1. DeNiro M., Al-Mohanna F. A. Nuclear factor kappa-B signaling is integral to ocular neovascularization in ischemia-independent microenvironment. PLoS ONE. 2014;9(7) doi: 10.1371/journal.pone.0101602.e101602
    1. Ahluwalia A., Tarnawski A. S. Critical role of hypoxia sensor—HIF-1α in VEGF gene activation. Implications for angiogenesis and tissue injury healing. Current Medicinal Chemistry. 2012;19(1):90–97. doi: 10.2174/092986712803413944.
    1. Artese L., Piattelli A., Cardoso L. A. D. G., et al. Immunoexpression of angiogenesis, nitric oxide synthase, and proliferation markers in gingival samples of patients with aggressive and chronic periodontitis. Journal of Periodontology. 2010;81(5):718–726. doi: 10.1902/jop.2010.090524.
    1. Bletsa A., Virtej A., Berggreen E. Vascular endothelial growth factors and receptors are up-regulated during development of apical periodontitis. Journal of Endodontics. 2012;38(5):628–635. doi: 10.1016/j.joen.2012.01.005.
    1. Kasprzak A., Surdacka A., Tomczak M., et al. Expression of angiogenesis-stimulating factors (VEGF, CD31, CD105) and angiogenetic index in gingivae of patients with chronic periodontitis. Folia Histochemica et Cytobiologica. 2012;50(4):554–564. doi: 10.5603/FHC.2012.0078.

Source: PubMed

3
Sottoscrivi