Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2135 2023-01-30 17:01:48 |
2 update references and layout -13 word(s) 2122 2023-01-31 06:35:34 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Nuñez-Belmar, J.;  Morales-Olavarria, M.;  Vicencio, E.;  Vernal, R.;  Cortez, C.;  Cárdenas, J.P. Porphyromonas gingivalis during Periodontitis Pathogenesis. Encyclopedia. Available online: https://encyclopedia.pub/entry/40604 (accessed on 04 May 2024).
Nuñez-Belmar J,  Morales-Olavarria M,  Vicencio E,  Vernal R,  Cortez C,  Cárdenas JP. Porphyromonas gingivalis during Periodontitis Pathogenesis. Encyclopedia. Available at: https://encyclopedia.pub/entry/40604. Accessed May 04, 2024.
Nuñez-Belmar, Josefa, Mauricio Morales-Olavarria, Emiliano Vicencio, Rolando Vernal, Cristian Cortez, Juan P. Cárdenas. "Porphyromonas gingivalis during Periodontitis Pathogenesis" Encyclopedia, https://encyclopedia.pub/entry/40604 (accessed May 04, 2024).
Nuñez-Belmar, J.,  Morales-Olavarria, M.,  Vicencio, E.,  Vernal, R.,  Cortez, C., & Cárdenas, J.P. (2023, January 30). Porphyromonas gingivalis during Periodontitis Pathogenesis. In Encyclopedia. https://encyclopedia.pub/entry/40604
Nuñez-Belmar, Josefa, et al. "Porphyromonas gingivalis during Periodontitis Pathogenesis." Encyclopedia. Web. 30 January, 2023.
Porphyromonas gingivalis during Periodontitis Pathogenesis
Edit

Periodontitis is a non-communicable chronic inflammatory disease characterized by the progressive and irreversible breakdown of the soft periodontal tissues and resorption of teeth-supporting alveolar bone. The etiology of periodontitis involves dysbiotic shifts in the diversity of microbial communities inhabiting the subgingival crevice, which is dominated by anaerobic Gram-negative bacteria, including Porphyromonas gingivalis. Indeed, P. gingivalis is a keystone pathogen with a repertoire of attributes that allow it to colonize periodontal tissues and influence the metabolism, growth rate, and virulence of other periodontal bacteria. The pathogenic potential of P. gingivalis has been traditionally analyzed using classical biochemical and molecular approaches.

periodontitis microbial dysbiosis Porphyromonas gingivalis keystone pathogen

1. Introduction

Periodontitis is a non-communicable chronic inflammatory disease characterized by the progressive and irreversible destruction of the periodontium [1][2]. Periodontitis is a long-term disease with a high prevalence worldwide, being the most prevalent form of osteolytic pathology in humans [3][4]. The etiology of periodontitis lies in the constant challenge of the dysbiotic biofilm attached to the tooth in the subgingival crevice, which leads to a deregulated host immune response responsible for the clinical phenotype of the disease [5].
Porphyromonas gingivalis is a well-characterized member of the dysbiotic subgingival microbiota and a key bacterium in the aetiopathogenesis of periodontitis [6][7]. It is an anaerobe, asaccharolytic, non-motile, Gram-negative bacterium associated with destructive chronic periodontitis [8]. Its association with periodontitis is based fundamentally on its virulence properties, recurrent presence in affected sites, and higher serum antibody levels in patients diagnosed with chronic periodontitis. In addition, periodontal health indicators are inversely correlated with the presence of this periodontal pathogen [8][9][10]. Moreover, periodontitis induced by P. gingivalis may influence the course and pathogenesis of other chronic inflammatory disorders [11]. P. gingivalis expresses several virulence factors that enable it to colonize, invade, and disrupt periodontal tissues [8][12]. Despite its low-relative abundance in the subgingival biofilm, P. gingivalis operates as a keystone pathogen, being critical in promoting the host inflammatory and osteolytic response involved in periodontitis onset and progression [7][13][14]. Furthermore, P. gingivalis significantly changes the bacterial taxa that comprise symbiotic microbial communities for heterotypical communities with recognized destructive inflammatory potential [6][7] (Figure 1). P. gingivalis strain virulence varies among different types, and this diversity may alter its keystone pathogen capacity [14][15][16]. Consequently, precise analysis methods are required for a better understanding of P. gingivalis in the context of its different pathogenic potential and its modulating effect on multispecies heterotopic communities in the periodontitis-related biofilm.
Figure 1. Periodontitis pathogenesis and P. gingivalis overview. (A) During the onset and progression of periodontitis, P. gingivalis resides in the subgingival biofilm adhered to the tooth surface, where it interacts metabolically with other bacteria, inducing them to express different virulence factors with pathogenic potential. In parallel, P. gingivalis acts as a keystone pathogen, altering the regulation of the immune response in the susceptible host. The metabolic synergism and immune response subversion provide the nutritional and protective conditions required by the dysbiotic subgingival community to increase their diversity and abundance, with the concomitant induction of a strong, destructive inflammatory response. Together, all these activities cause irreversible connective tissue breakdown and resorption of the tooth-supporting alveolar bone, the critical hallmark of periodontitis that causes tooth loss. (B) To invade the periodontium, P. gingivalis uses various virulence factors that allow it to colonize, replicate, and spread in different subsets of cells to increase its progeny and generate infection. (C) In addition to causing tooth loss, P. gingivalis-induced periodontitis can also affect systemic health, influencing the course of other diseases and conditions.
Omics technologies have revolutionized modern biology, enabling simultaneous analysis of multiple molecular components in response to environmental stimuli across time [17]. These technological approaches, mainly metagenomics, metatranscriptomics, proteomics, and metabolomics, have the potential to increase the efficiency of information to elucidate complex systems [18] and have been used to understand the composition and function of the oral microbiome during health and disease [19][20][21].

2. P. gingivalis Is a Keystone Pathogen of Periodontitis

As a keystone pathogen, P. gingivalis disrupts the host’s immune response even at low relative abundances [22][23]. This microbe generates protection and the optimal nutritional conditions for the growth and development of pathobiont microorganisms, responsible for maintaining the destructive chronic inflammatory environment [24]. Therefore, P. gingivalis modulates the entire bacterial community’s composition, abundance, and adaptive fitness [25][26]. This synergistic interaction was evidenced in a mechanistic study using germ-free mice. In contrast to wild-type mice, the oral microbiota lack in germ-free mice did not induce destructive inflammation after P. gingivalis inoculation into the periodontal tissues [13].
The oral microbiome comprises over 600 prevalent taxa distributed in different ecological niches. Because many of these species cannot be cultured, omics technologies are powerful tools for assessing their composition [27]. Using clinical samples, metagenomics and metatranscriptomics studies have confirmed the prominent subgingival microbial dysbiosis in individuals with periodontitis. Furthermore, they established the presence and relative abundance of P. gingivalis and the virulence factors upregulation associated with its pathogenesis [19][28][29][30].
The synergy interactions of the resident microbiota are fundamental in this new periodontitis model. In this sense, Streptococcus gordonii is an accessory pathogen that contributes to increasing P. gingivalis virulence [7]. LuxS is a soluble mediator autoinducer 2 (AI-2) molecule of P. gingivalis involved in the uptake of hemin/inorganic iron and quorum sensing interspecies with Streptococcus gordonii. Utilizing transcriptomics was demonstrated in a LuxS-deficient strain that P. gingivalis fails to form microcolonies, confirming the significance of these molecules in the signaling involved in the formation and maturation of the biofilm [31]. The role of keystone bacteria has also been evaluated in vitro. A multispecies symbiotic biofilm (oral health) was generated in the presence or absence of P. gingivalis and A. actinomycetemcomitans. The transcriptomic analyses showed significant gene expression pattern differences when comparing both conditions. Similarly, they exhibited different gene expressions when planktonic, and biofilm conditions were compared. These findings showed a two-way modulatory effect of periodontal pathogens on multispecies heterotypic communities [32]. The keystone pathogen effect has also been evaluated in in vivo models. The ligature-induced periodontitis model in mice induces microbial dysbiosis and recreates the destructive inflammatory conditions exhibited during periodontitis in humans [33]. Sterile silk ligature alone causes alveolar bone resorption. However, when it is preincubated with P. gingivalis and subsequently tied to the tooth, the concomitant bone loss is significantly exacerbated [34]. Consistent with the systemic effects of periodontitis, oral administration of P. gingivalis significantly alters the gut microbiome. The metagenomic studies showed evident differences in the composition and relative abundance of the community members compared to mice that did not receive P. gingivalis. In addition, dysbiosis generates systemic inflammatory effects, changes in the serum metabolome, and aggravates collagen-induced arthritis [35][36][37][38]. This evidence reveals the contribution of omics disciplines to determining the keystone role of P. gingivalis in perturbing bacterial communities.

3. Proteomics and Metabolomics in P. gingivalis Pathogenesis

Proteomics corresponds to the study (Table 1) of the structure, function, and protein interaction on a large scale [39]. This technique has become a powerful tool for protein identification involved in host-P. gingivalis interactions and their relationship with other oral bacteria. For example, proteomic studies using mixed oral bacteria cultures showed that nearly 40% of P. gingivalis proteins can be regulated by the presence of other members of the oral microbiota, such as Fusobacterium nucleatum, Streptococcus gordonii, or Streptococcus oralis [40][41], suggesting an effect of the ecological community on P. gingivalis protein expression.
Table 1. Summary of metabolomics and proteomics research studies.
The identification and characterization of proteins involved in bacterial colonization and invasion is another relevant scope of P. gingivalis proteomics. Notable changes have been reported in the extracellular proteomic profiles and virulence factors expressed by different strains of P. gingivalis [42]. For instance, using mass spectrometry, alterations in the P. gingivalis O-glycoproteome are evidenced, determining that gingipains lacked these post-translational modifications [43]. On the other hand, a previously known marker of rheumatoid arthritis is the loss of tolerance to the presence of proteins that have undergone citrullination. This post-translational modification affects arginine residues and produces changes in the structure and function of proteins [49]. Interestingly, P. gingivalis contains a peptidyl-arginine deiminase (PPAD), an enzyme capable of protein citrullination, and it can modify bacterial and human proteins [42]. Moreover, the comparison of secreted protein citrullination profiles among several P. gingivalis clinical isolates with reference strains showed substantial differences, primarily focused on six to 25 proteins, mostly composed of virulence factors. The role of these modifications remains to be clarified.
Proteomics can also help to understand the role of some cellular derivates, such as Outer membrane vesicles (OMVs). P. gingivalis releases several virulence factors toward the environment via OMVs [44]. Studies covering the role of OMV-associated proteome in P. gingivalis suggest important changes according to the availability of heme, displaying an upregulation of proteins involved in heme binding and transport when this nutrient is limited [45]. Collectively, proteomic studies reveal that the P. gingivalis proteome changes depending on the interaction target and the biological context, offering another opportunity to understand P. gingivalis physiology and pathogenesis.
Another useful technique to understand the role of P. gingivalis in periodontitis is metabolomics (Table 1), which is the systematic study of metabolic content produced and consumed by specific cellular processes. Thus, it presents the collection of all metabolites within a particular sample [50]. Current evidence suggests that during periodontitis, a metabolic alteration in the oral cavity is induced by the colonization of pathogenic bacteria [48]. Additionally, metabolic changes are also reported in cells of the periodontal ligament (PDLSCs) under P. gingivalis infection, showing a metabolic reprogramming that compromises processes associated with the glycolysis, tricarboxylic acid (TCA) cycle, tryptophan, and choline metabolism [47][48]. Since gene sequence and expression profiles are essential but not sufficient to understand the mechanisms underlying P. gingivalis pathogenesis, proteomic and metabolomic studies have begun to gain more insights into periodontitis-related host-pathogen interactions.

References

  1. Wu, Y.; Dong, G.; Xiao, W.; Xiao, E.; Miao, F.; Syverson, A.; Missaghian, N.; Vafa, R.; Ortega, A.A.C.; Rossa, J.C.; et al. Effect of aging on periodontal inflammation, microbial colonization, and disease susceptibility. J. Dent. Res. 2016, 95, 460–466.
  2. van Dyke, T.E.; Sima, C. Understanding resolution of inflammation in periodontal diseases: Is chronic inflammatory periodontitis a failure to resolve? Periodontology 2020, 82, 205–213.
  3. Graves, D.T.; Oates, T.; Garlet, G.P. Review of osteoimmunology and the host response in endodontic and periodontal lesions. J. Oral Microbiol. 2011, 3, 5304.
  4. Alvarez, C.; Monasterio, G.; Cavalla, F.; Córdova, L.A.; Hernández, M.; Heymann, D.; Garlet, G.P.; Sorsa, T.; Pärnänen, P.; Lee, H.-M.; et al. Osteoimmunology of oral and maxillofacial diseases: Translational applications based on biological mechanisms. Front. Immunol. 2019, 10, 1664.
  5. Hajishengallis, G.; Korostoff, J.M. Revisiting the Page & Schroeder model: The good, the bad and the unknowns in the periodontal host response 40 years later. Periodontology 2017, 75, 116–151.
  6. Curtis, M.A.; Diaz, P.I.; van Dyke, T.E. The role of the microbiota in periodontal disease. Periodontology 2020, 83, 14–25.
  7. Hajishengallis, G. Periodontitis: From microbial immune subversion to systemic inflammation. Nat. Rev. Immunol. 2015, 15, 30–44.
  8. How, K.Y.; Song, K.P.; Chan, K.G. Porphyronomas gingivalis: An overview of periodontopathic pathogen below the gum line. Front. Microbiol. 2016, 7, 53.
  9. Casarin, R.C.V.; Ribeiro, É.D.; Mariano, F.S.; Nociti, F.H.; Casati, M.Z.; Gonçalves, R.B. Levels of Aggregatibacter actinomycetemcomitans, Porphyromonas gingivalis, inflammatory cytokines and species-specific immunoglobulin G in generalized aggressive and chronic periodontitis. J. Periodontal Res. 2010, 45, 635–642.
  10. Griffen, A.L.; Becker, M.R.; Lyons, S.R.; Moeschberger, M.L.; Leys, E.J. Prevalence of Porphyromonas gingivalis and periodontal health status. J. Clin. Microbiol. 1998, 36, 3239–3242.
  11. Hajishengallis, G.; Chavakis, T. Local and systemic mechanisms linking periodontal disease and inflammatory comorbidities. Nat. Rev. Immunol. 2021, 21, 426–440.
  12. Zenobia, C.; Hajishengallis, G. Porphyronomas gingivalis virulence factors involved in subversion of leukocytes and microbial dysbiosis. Virulence 2015, 6, 236–243.
  13. Hajishengallis, G.; Liang, S.; Payne, M.A.; Hashim, A.; Jotwani, R.; Eskan, M.A.; McIntosh, M.L.; Alsam, A.; Kirkwood, K.; Lambris, J.; et al. Low-Abundance Biofilm Species Orchestrates Inflammatory Periodontal Disease through the Commensal Microbiota and the Complement. Cell Host Microbe 2011, 10, 497–506.
  14. Hajishengallis, G.; Lamont, R.J. Beyond the red complex and into more complexity: The polymicrobial synergy and dysbiosis (PSD) model of periodontal disease etiology. Mol. Oral Microbiol. 2012, 27, 409–419.
  15. Tribble, G.; Kerr, J.; Wang, B. Genetic diversity in the oral pathogen Porphyronomas gingivalis: Molecular mechanisms and biological consequences. Futur. Microbiol. 2013, 8, 607–622.
  16. Silva, I.L.; Cascales, E. Molecular Strategies Underlying Porphyromonas gingivalis Virulence. J. Mol. Biol. 2021, 433, 166836.
  17. Gutleben, J.; de Mares, M.C.; van Elsas, J.D.; Smidt, H.; Overmann, J.; Sipkema, D. The multi-omics promise in context: From sequence to microbial isolate. Crit. Rev. Microbiol. 2018, 44, 212–229.
  18. Beale, D.J.; Kouremenos, K.A.; Palombo, E.A. Microbial Metabolomics: Applications in Clinical, Environmental, and Industrial Microbiology; Springer: Cham, Switzerland, 2016; p. 321.
  19. Wang, J.; Qi, J.; Zhao, H.; He, S.; Zhang, Y.; Wei, S.; Zhao, F. Metagenomic sequencing reveals microbiota and its functional potential associated with periodontal disease. Sci. Rep. 2013, 3, 1843.
  20. Utter, D.R.; Borisy, G.G.; Eren, A.M.; Cavanaugh, C.M.; Welch, J.L.M. Metapangenomics of the oral microbiome provides insights into habitat adaptation and cultivar diversity. Genome Biol. 2020, 21, 293.
  21. Williams, D.W.; Greenwell-Wild, T.; Brenchley, L.; Dutzan, N.; Overmiller, A.; Sawaya, A.P.; Webb, S.; Martin, D.; Hajishengallis, G.; Divaris, K.; et al. Human oral mucosa cell atlas reveals a stromal-neutrophil axis regulating tissue immunity. Cell 2021, 184, 4090–4104.
  22. Wang, M.; Krauss, J.L.; Domon, H.; Hosur, K.B.; Liang, S.; Magotti, P.; Triantafilou, M.; Triantafilou, K.; Lambris, J.D.; Hajishengallis, G. Microbial Hijacking of Complement–Toll-like Receptor Crosstalk. Sci. Signal. 2010, 3, ra11.
  23. Maekawa, T.; Krauss, J.L.; Abe, T.; Jotwani, R.; Triantafilou, M.; Triantafilou, K.; Hashim, A.; Hoch, S.; Curtis, M.A.; Nussbaum, G.; et al. Porphyronomas gingivalis manipulates complement and TLR signaling to uncouple bacterial clearance from inflammation and promote dysbiosis. Cell Host Microbe 2014, 15, 768–778.
  24. Hajishengallis, G.; Darveau, R.; Curtis, M. The keystone-pathogen hypothesis. Nat. Rev. Microbiol. 2012, 10, 717–725.
  25. Hajishengallis, G.; Lamont, R.; Graves, D. The enduring importance of animal models in understanding periodontal disease. Virulence 2015, 6, 229–235.
  26. Hajishengallis, G.; Lamont, R. The polymicrobial synergy and dysbiosis model of periodontal disease pathogenesis. In The Human Microbiota and Chronic Disease: Dysbiosis as a Cause of Human Pathology; John Wiley & Sons Inc.: New York, NY, USA, 2016; pp. 227–242.
  27. Dewhirst, F.E.; Chen, T.; Izard, J.; Paster, B.J.; Tanner, A.C.R.; Yu, W.-H.; Lakshmanan, A.; Wade, W.G. The human oral microbiome. J. Bacteriol. 2010, 192, 5002–5017.
  28. Abusleme, L.; Dupuy, A.K.; Nicol, D.; Silva, N.; Burleson, J.A.; Strausbaugh, L.D. The subgingival microbiome in health and periodontitis and its relationship with community biomass and inflammation. ISME J. 2013, 7, 1016–1025.
  29. Yost, S.; Duran-Pinedo, A.E.; Teles, R.; Krishnan, K.; Frias-Lopez, J. Functional signatures of oral dysbiosis during periodontitis progression revealed by microbial metatranscriptome analysis. Genome Med. 2015, 7, 27.
  30. Wang, G.P. Defining functional signatures of dysbiosis in periodontitis progression. Genome Med. 2015, 7, 4–6.
  31. Hirano, T.; Beck, D.A.C.; Demuth, D.R.; Hackett, M.; Lamont, R.J. Deep sequencing of Porphyronomas gingivalis and comparative transcriptome analysis of a LuxS mutant. Front. Cell. Infect. Microbiol. 2012, 2, 79.
  32. Frias-Lopez, J.; Duran-Pinedo, A. Effect of periodontal pathogens on the metatranscriptome of a healthy multispecies biofilm model. J. Bacteriol. 2012, 194, 2082–2095.
  33. Abe, T.; Hajishengallis, G. Optimization of the ligature-induced periodontitis model in mice. J. Immunol. Methods 2013, 394, 49–54.
  34. Kimura, S.; Nagai, A.; Onitsuka, T.; Koga, T.; Fujiwara, T.; Hamada, S. Induction of experimental periodontitis in mice with Porphyronomas gingivalis-adhered ligatures. J. Periodontol. 2000, 71, 1167–1173.
  35. Lin, J.; Bi, L.; Yu, X.; Kawai, T.; Taubman, M.A.; Shen, B.; Han, X. Porphyronomas gingivalis exacerbates ligature-induced, RANKLdependent alveolar bone resorption via differential regulation of Toll-like receptor 2 (TLR2) and TLR4. Infect. Immun. 2014, 82, 4127–4134.
  36. Arimatsu, K.; Yamada, H.; Miyazawa, H.; Minagawa, T.; Nakajima, M.; Ryder, M.I.; Gotoh, K.; Motooka, D.; Nakamura, S.; Iida, T.; et al. Oral pathobiont induces systemic inflammation and metabolic changes associated with alteration of gut microbiota. Sci. Rep. 2014, 4, 4828.
  37. Sato, K.; Takahashi, N.; Kato, T.; Matsuda, Y.; Yokoji, M.; Yamada, M.; Nakajima, T.; Kondo, N.; Endo, N.; Yamamoto, R.; et al. Aggravation of collagen-induced arthritis by orally administered Porphyronomas gingivalis through modulation of the gut microbiota and gut immune system. Sci. Rep. 2017, 7, 6955.
  38. Kato, T.; Yamazaki, K.; Nakajima, M.; Date, Y.; Kikuchi, J.; Hase, K.; Ohno, H.; Yamazaki, K. Oral Administration of Porphyronomas gingivalis Alters the Gut Microbiome and Serum Metabolome. mSphere 2018, 3, e00460-18.
  39. Zhu, H.; Bilgin, M.; Snyder, M. Proteomics. Annu. Rev. Biochem. 2003, 72, 783–812.
  40. Kuboniwa, M.; Hendrickson, E.L.; Xia, Q.; Wang, T.; Xie, H.; Hackett, M.; Lamont, R.J. Proteomics of Porphyronomas gingivalis within a model oral microbial community. BMC Microbiol. 2009, 9, 98.
  41. Maeda, K.; Nagata, H.; Ojima, M.; Amano, A. Proteomic and transcriptional analysis of interaction between oral microbiota Porphyronomas gingivalis and Streptococcus oralis. J. Proteome Res. 2015, 14, 82–94.
  42. Stobernack, T.; Glasner, C.; Junker, S.; Gabarrini, G.; de Smit, M.; de Jong, A.; Otto, A.; Becher, D.; van Winkelhoff, A.J.; van Dijl, J.M. The extracellular Proteome and Citrullinome of the Oral Pathogen Porphyronomas gingivalis. J. Proteome Res. 2016, 15, 4532–4543.
  43. Veith, P.D.; Shoji, M.; Scott, N.E.; Reynolds, E.C. Characterization of the O-Glycoproteome of Porphyronomas gingivalis. Microbiol. Spectr. 2022, 10, e01502-21.
  44. Veith, P.D.; Chen, Y.-Y.; Gorasia, D.G.; Chen, D.; Glew, M.D.; O’Brien-Simpson, N.M.; Cecil, J.D.; Holden, J.A.; Reynolds, E.C. Porphyronomas gingivalis outer membrane vesicles exclusively contain outer membrane and periplasmic proteins and carry a cargo enriched with virulence factors. J. Proteome Res. 2014, 13, 2420–2432.
  45. Veith, P.D.; Luong, C.; Tan, K.H.; Dashper, S.G.; Reynolds, E.C. Outer membrane vesicle proteome of Porphyronomas gingivalis Is Differentially Modulated Relative to the Outer Membrane in Response to Heme Availability. J. Proteome Res. 2018, 17, 2377–2389.
  46. Gawron, K.; Wojtowicz, W.; Łazarz-Bartyzel, K.; Łamasz, A.; Qasem, B.; Mydel, P.; Chomyszyn-Gajewska, M.; Potempa, J.; Mlynarz, P. Metabolomic status of the oral cavity in chronic periodontitis. Vivo 2019, 33, 1165–1174.
  47. Su, W.; Shi, J.; Zhao, Y.; Yan, F.; Lei, L.; Li, H. Porphyronomas gingivalis triggers inflammatory responses in periodontal ligament cells by succinate-succinate dehydrogenase–HIF–1α axis. Biochem. Biophys. Res. Commun. 2020, 522, 184–190.
  48. Dong, Z.; Lv, W.; Zhang, C.; Chen, S. Correlation analysis of gut microbiota and serum metabolome with Porphyronomas gingivalis-induced metabolic disorders. Front. Cell. Infect. Microbiol. 2022, 12, 858902.
  49. Payne, J.B.; Golub, L.M.; Thiele, G.M.; Mikuls, T.R. The link between Periodontitis and Rheumatoid Arthritis: A Periodontist’s Perspective. Curr. Oral Health Rep. 2015, 2, 20–29.
  50. Johnson, C.H.; Ivanisevic, J.; Siuzdak, G. Metabolomics: Beyond biomarkers and towards mechanisms. Mol. Cell Biol. 2016, 17, 451–459.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , ,
View Times: 397
Revisions: 2 times (View History)
Update Date: 31 Jan 2023
1000/1000