Complement split product C3c in saliva as biomarker for periodontitis and response to periodontal treatment

Maria Anastasia Grande, Daniel Belstrøm, Christian Damgaard, Palle Holmstrup, Sai Sindhu Thangaraj, Claus Henrik Nielsen, Yaseelan Palarasah, Maria Anastasia Grande, Daniel Belstrøm, Christian Damgaard, Palle Holmstrup, Sai Sindhu Thangaraj, Claus Henrik Nielsen, Yaseelan Palarasah

Abstract

Background and objective: The complement system is engaged in inflammatory reactions both in the periodontal pockets and in the periodontium itself, where it can mediate tissue destruction. The aim of this study was, first, to compare salivary levels of the total complement system protein C3 and its split product, fluid-phase C3c in patients with periodontitis and periodontally healthy controls. Next, to determine if C3 and C3c levels had biomarker potential in diagnosing and monitoring periodontitis and its treatment. We hypothesized that salivary levels of total C3 and the split product C3c associated with the severity of periodontitis and reflected decreased inflammatory activity after periodontal treatment.

Methods: At baseline, stimulated saliva samples were collected from patients with periodontitis (n = 18) and periodontally healthy controls (n = 15). Subsequently, non-surgical periodontal treatment was performed in the patients, and saliva sampling from patients was repeated two-, six-, and twelve weeks post-treatment (NCT02913248 at clinicaltrials.gov). The patients were grouped as good and poor responders to treatment according to the achieved reduction in bleeding on probing (BOP). Salivary levels of C3 and C3c were quantified using sandwich ELISA.

Results: Patients with periodontitis had higher baseline levels of both total C3 and the split product C3c in saliva than did periodontally healthy controls (P < .0001). Receiver operating curve (ROC) analyses discriminated patients with periodontitis from controls based on both C3 (AUC (area under curve) = 0.91, P < .001) and C3c levels (AUC = 0.84, P < .001) in saliva. Periodontal treatment improved all clinical parameters (P < .01). Good responders (n = 10) had lower baseline levels of C3c than poor responders (n = 8), (P < .05), and baseline levels of C3c discriminated between good and poor responders (AUC = 0.80, P < .05).

Conclusion: In conclusion, patients with periodontitis had higher salivary levels of C3c, and the C3c levels were predictive of reductions in BOP, that is, the poor responders. This suggests that salivary C3c levels possess potential to serve as a biomarker predicting the clinical response to non-surgical periodontal treatment.

Keywords: complement component 3; intervention; periodontitis; saliva.

Conflict of interest statement

All authors declare no conflict of interest.

© 2020 The Authors. Journal of Periodontal Research published by John Wiley & Sons Ltd.

Figures

Figure 1
Figure 1
Baseline salivary levels of C3 and C3c in patients with periodontitis and periodontally healthy controls. The scatter dot‐plots illustrate salivary levels of (A) C3 (μg/mL) and (B) C3c (μg/mL) in patients with periodontitis (n = 18) and periodontally healthy controls (n = 15), (P < .0001). The horizontal lines show median levels
Figure 2
Figure 2
Area under curve (AUC) for C3 and C3c in identifying periodontitis from periodontal health at baseline. Receiver operating curves (ROC) of salivary A = C3 and B = C3c levels discriminating between patients with periodontitis and periodontally healthy controls at baseline. (A) C3, AUC = 0.91, CI = [0.81;1.00], (P < .001), (B) C3c, AUC = 0.84, CI = [0.69;1.00], (P < .001)
Figure 3
Figure 3
Pre‐ and post‐treatment levels of C3 and C3c in patients with periodontitis. The scatter dot‐plots illustrate salivary levels of (A) C3 (μg/mL) and (B) C3c (μg/mL) recorded at baseline and two‐, six‐, and twelve weeks post‐treatment in patients with periodontitis (n = 18). The horizontal lines show the median levels
Figure 4
Figure 4
Baseline salivary C3 and C3c levels in good and poor responders to non‐surgical periodontal treatment. The scatter dot‐plots illustrate baseline saliva levels of (A) C3 (P > .05) and (B) C3c (P < .05) in good responders (n = 10) compared with poor responders (n = 8). The horizontal lines show the median levels in the two groups
Figure 5
Figure 5
AUC for total C3 and C3c at baseline in identifying poor responders to non‐surgical periodontal treatment. ROC analysis of salivary baseline levels of A = C3 and B = C3c to discriminate between the good (n = 10) and the poor (n = 8) responders to periodontal treatment. (A) C3, AUC = 0.65, CI = [0.36;0.94], (P > .05) and (B) C3c, AUC = 0.80, CI = [0.59;1.00], (P < .05)

References

    1. Eke PI, Thornton‐Evans GO, Wei L, Borgnakke WS, Dye BA, Genco RJ. Periodontitis in US Adults: National Health and Nutrition Examination Survey 2009–2014. J Am Dental Assoc. 2018;149(7):576‐588. e576.
    1. Hajishengallis G, Darveau RP, Curtis MA. The keystone‐pathogen hypothesis. Nat Rev Microbiol. 2012;10(10):717‐725.
    1. Holmstrup P, Damgaard C, Olsen I, et al. Comorbidity of periodontal disease: two sides of the same coin? An introduction for the clinician. J Oral Microbiol. 2017;9(1):1332710.
    1. Chapple IL, Genco R. Diabetes and periodontal diseases: consensus report of the Joint EFP/AAP Workshop on periodontitis and systemic diseases. J Periodontol. 2013;84(4 Suppl):S106‐112.
    1. Tonetti MS, Van Dyke TE. Periodontitis and atherosclerotic cardiovascular disease: consensus report of the Joint EFP/AAPWorkshop on periodontitis and systemic diseases. J Periodontol. 2013;84(Suppl 4S):S24‐S29.
    1. Hajishengallis G, Maekawa T, Abe T, Hajishengallis E, Lambris JD. Complement involvement in periodontitis: molecular mechanisms and rational therapeutic approaches. Adv Exp Med Biol. 2015;865:57‐74.
    1. Damgaard C, Holmstrup P, Van Dyke TE, Nielsen CH. The complement system and its role in the pathogenesis of periodontitis: current concepts. J Periodontal Res. 2015;50(3):283‐293.
    1. Janssen BJC, Huizinga EG, Raaijmakers HCA, et al. Structures of complement component C3 provide insights into the function and evolution of immunity. Nature. 2005;437(7058):505‐511.
    1. Hajishengallis G, Liang S, Payne M, et al. Low‐abundance biofilm species orchestrates inflammatory periodontal disease through the commensal microbiota and complement. Cell Host Microbe. 2011;10(5):497‐506.
    1. Sahu A, Lambris JD. Structure and biology of complement protein C3, a connecting link between innate and acquired immunity. Immunol Rev. 2001;180:35‐48.
    1. Leffler J, Bengtsson AA, Blom AM. The complement system in systemic lupus erythematosus: an update. Ann Rheum Dis. 2014;73(9):1601‐1606.
    1. Rasmussen KJ, Skjoedt MO, Vitved L, Skjoedt K, Palarasah Y. A novel antihuman C3d monoclonal antibody with specificity to the C3d complement split product. J Immunol Methods. 2017;444:51‐55.
    1. Slots J. Periodontitis: facts, fallacies and the future. Periodontol 2000. 2017;75(1):7‐23.
    1. Riis JL, Bryce CI, Ha T, et al. Adiponectin: Serum‐saliva associations and relations with oral and systemic markers of inflammation. Peptides. 2017;91:58‐64.
    1. Schenkein HA, Genco RJ. Gingival fluid and serum in periodontal diseases. I. Quantitative study of immunoglobulins, complement components, and other plasma proteins. J Periodontol. 1977;48(12):772‐777.
    1. Lally ET, McArthur WP, Baehni PC. Biosynthesis of complement components in chronically inflamed gingiva. J Periodontal Res. 1982;17(3):257‐262.
    1. Attstrom R, Laurel AB, Lahsson U, Sjoholm A. Complement factors in gingival crevice material from healthy and inflamed gingiva in humans. J Periodontal Res. 1975;10(1):19‐27.
    1. Athina A, Papaconstantinou N, Johannessen AC, Kristoffersen T. Deposits of immunoglobulins, complement, and immune complexes in inflamed human gingiva. Acta Odontol Scand. 1987;45(3):187‐193.
    1. Toto PD, Lin L, Gargiulo A. Identification of C3a, IgG, IgM in inflamed human gingiva. J Dent Res. 1978;57(5–6):696.
    1. Maekawa T, Abe T, Hajishengallis E, et al. Genetic and intervention studies implicating complement C3 as a major target for the treatment of periodontitis. J Immunol. 2014;192(12):6020‐6027.
    1. Pickering MC, D'Agati VD, Nester CM, et al. C3 glomerulopathy: consensus report. Kidney Int. 2013;84(6):1079‐1089.
    1. Palarasah Y, Skjodt K, Brandt J, et al. Generation of a C3c specific monoclonal antibody and assessment of C3c as a putative inflammatory marker derived from complement factor C3. J Immunol Methods. 2010;362(1–2):142‐150.
    1. Teisner B, Brandslund I, Grunnet N, Hansen LK, Thellesen J, Svehag SE. Acute complement activation during an anaphylactoid reaction to blood transfusion and the disappearance rate of C3c and C3d from the circulation. J Clin Lab Immunol. 1983;12(2):63‐67.
    1. Belstrøm D, Grande MA, Sembler‐Møller ML, et al. Influence of periodontal treatment on subgingival and salivary microbiotas. J Periodontol. 2018;89(5):531‐539.
    1. Grande MA, Belstrøm D, Damgaard C, et al. Salivary concentrations of macrophage activation‐related chemokines are influenced by non‐surgical periodontal treatment: a 12‐week follow‐up study. J Oral Microbiol. 2020;12(1):1694383.
    1. American academy of periodontology task force report on the update to the 1999 classification of periodontal diseases and conditions. J Periodontol. 2015;86(7):835‐838.
    1. Papapanou PN, Sanz M, Buduneli N, et al. Periodontitis: consensus report of workgroup 2 of the 2017 world workshop on the classification of periodontal and peri‐implant diseases and conditions. J Periodontol. 2018;89(Suppl 1):S173‐S182.
    1. Whitcomb BW, Schisterman EF. Assays with lower detection limits: implications for epidemiological investigations. Paediatr Perinat Epidemiol. 2008;22(6):597‐602.
    1. Croghan CaP P.Methods of Dealing with Values Below the Limit of Detection using SAS. . Accessed February 21, 2019.
    1. Aurer A, Jorgic‐Srdjak K, Plancak D, Stavljenic‐Rukavina A, Aurer‐Kozelj J. Proinflammatory factors in saliva as possible markers for periodontal disease. Coll Antropol. 2005;29(2):435‐439.
    1. Boackle RJ. The interaction of salivary secretions with the human complement system–a model for the study of host defense systems on inflamed mucosal surfaces. Crit Rev Oral Biol Med. 1991;2(3):355‐367.
    1. Niekrash CE, Patters MR. Simultaneous assessment of complement components C3, C4, and B and their cleavage products in human gingival fluid. II. Longitudinal changes during periodontal therapy. J Periodontal Res. 1985;20(3):268‐275.
    1. Niekrash CE, Patters MR, Lang NP. The relationship of complement cleavage in gingival fluid to periodontal diseases. J Periodontal Res. 1984;19(6):622‐627.
    1. Beikler T, Peters U, Prior K, Eisenacher M, Flemmig TF. Gene expression in periodontal tissues following treatment. BMC Med Genomics. 2008;1:30.
    1. Bostanci N, Bao K, Li X, et al. Gingival exudatome dynamics implicate inhibition of the alternative complement pathway in the protective action of the C3 inhibitor Cp40 in nonhuman primate periodontitis. J Proteome Res. 2018;17(9):3153‐3175.
    1. Maekawa T, Briones RA, Resuello RRG, et al. Inhibition of pre‐existing natural periodontitis in non‐human primates by a locally administered peptide inhibitor of complement C3. J Clin Periodontol. 2016;43(3):238‐249.
    1. Frank J, Courts RJB, Hugh Fudenberg H, Silverman MS. Detection of functional complement components in gingival creviular fluid from humans with periodontal disease. J Dent Res. 1976;56:327‐331.
    1. Talungchit S, Buajeeb W, Lerdtripop C, et al. Putative salivary protein biomarkers for the diagnosis of oral lichen planus: a case‐control study. BMC Oral Health. 2018;18(1):42.

Source: PubMed

3
Prenumerera