17
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Persistent Exposure to Porphyromonas gingivalis Promotes Proliferative and Invasion Capabilities, and Tumorigenic Properties of Human Immortalized Oral Epithelial Cells

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Recent epidemiological studies revealed a significant association between oral squamous cell carcinoma (OSCC) and Porphyromonas gingivalis, a major pathogen of periodontal disease. As a keystone pathogen of periodontitis, P. gingivalis is known not only to damage local periodontal tissues, but also to evade the host immune system and eventually affect systemic health. However, its role in OSCC has yet to be defined. To explore the underlying effect of chronic P. gingivalis infection on OSCC and to identify relevant biomarkers as promising targets for therapy and prevention, we established a novel model by exposing human immortalized oral epithelial cells (HIOECs) to P. gingivalis at a low multiplicity of infection (MOI) for 5–23 weeks. The P. gingivalis infected HIOECs were monitored for tumor biological alteration by proliferation, wound healing, transwell invasion, and gelatin zymography assays. Microarray and proteomic analyses were performed on HIOECs infected with P. gingivalis for 15 weeks, and some selected data were validated by quantitative real-time PCR and (or) western blot on cells infected for 15 and 23 weeks. Persistent exposure to P. gingivalis caused cell morphological changes, increased proliferation ability with higher S phase fraction in the cell cycle, and promoted cell migratory and invasive properties. In combining results of bioinformatics analyses and validation assays, tumor-related genes such as NNMT, FLI1, GAS6, lncRNA CCAT1, PDCD1LG2, and CD274 may be considered as the key regulators in tumor-like transformation in response to long-time exposure of P. gingivalis. In addition, some useful clinical biomarkers and novel proteins were also presented. In conclusion, P. gingivalis could promote tumorigenic properties of HIOECs, indicating that chronic P. gingivalis infection may be considered as a potential risk factor for oral cancer. The key regulators detected from the present model might be used in monitoring the development of OSCC with chronic periodontal infection.

          Related collections

          Most cited references49

          • Record: found
          • Abstract: found
          • Article: not found

          B7-H1 expression on non-small cell lung cancer cells and its relationship with tumor-infiltrating lymphocytes and their PD-1 expression.

          B7-H1/PD-L1 (B7-H1) and B7-DC/PD-L2 (B7-DC) are ligands for the receptor PD-1, which is known to negatively regulate T-cell activation. In the present study, we investigated the expression of B7-H1 and B7-DC in tumor specimens of non-small cell lung cancer and their relationships with clinicopathological variables and postoperative survival. Furthermore, we examined the correlation between B7-H1 expression on tumor cells and the number of tumor-infiltrating lymphocytes (TILs) or PD-1 expression on TILs. The expression of B7-H1 and B7-DC in 52 surgically resected specimens of non-small cell lung cancer was evaluated immunohistochemically. Expression of B7-H1 and B7-DC was focally observed in all non-small cell lung cancer tumor specimens. No relationship was found between the expression of B7-H1 or B7-DC and clinicopathological variables or postoperative survival. However, in the same sections evaluated, significantly fewer TILs were identified in B7-H1-positive tumor regions than in B7-H1-negative tumor regions in a subset of five patients (P = 0.01). Moreover, the percentage of TILs expressing PD-1 was significantly lower in B7-H1-positive tumor regions than in B7-H1-negative tumor regions (P = 0.02). The expression of B7-H1 on tumor cells in local areas reciprocally correlated with the number of TILs, and this may contribute to negative regulation in antitumor immune responses in non-small cell lung cancer.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Periodontal pathogens Porphyromonas gingivalis and Fusobacterium nucleatum promote tumor progression in an oral-specific chemical carcinogenesis model

            Oral squamous cell carcinoma (OSCC) is a lethal disease whose incidence is increasing. Epidemiologic studies demonstrate an association between periodontitis and oral cancer, and periodontal pathogens are implicated in the pathogenesis of numerous disorders, including rheumatoid arthritis, cardiovascular diseases, diabetes and gastrointestinal malignancies. Nevertheless, a causal role for periodontal pathogens in OSCC has not been shown, partly due to the lack of an appropriate animal model. Here, utilizing a newly-established murine model of periodontitis-associated oral tumorigenesis, we report that chronic bacterial infection promotes OSCC, and that augmented signaling along the IL-6-STAT3 axis underlies this effect. Our results indicate that periodontal pathogens P. gingivalis and F. nucleatum stimulate tumorigenesis via direct interaction with oral epithelial cells through Toll-like receptors. Furthermore, oral pathogens stimulate human OSCC proliferation and induce expression of key molecules implicated in tumorigenesis. To the best of our knowledge, these findings represent the first demonstration of a mechanistic role for oral bacteria in chemically induced OSCC tumorigenesis. These results are highly relevant for the design of effective prevention and treatment strategies for OSCC.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              The microflora associated with human oral carcinomas.

              Both local and systemic infections may complicate the morbidity of patients with oral malignant neoplasms, particularly those presenting intraorally. This study investigated the microbial contents of the biofilms present on the surfaces of oral squamous cell carcinomas. Biofilm samples were obtained from the central surface of the lesions in 21 patients (20 male, 1 female) aged 52.8 (+/- 8.2) years, and from contiguous healthy mucosa, before any antibiotic therapy or any tumour treatment. All lesions were keratinising squamous cell carcinomas with surface ulceration. Samples were transported in reduced brain heart infusion (BHI) broth and cultured within 1 h of removal, using aerobic and anaerobic complete and selective media. The median number of anaerobic colony forming units (CFU/ml) at the tumour sites (1.6 x 10(8)) was significantly higher than for the healthy (control) mucosa (3.0 x 10(7); P = 0.0001, Wilcoxon); the same was true for aerobes at the tumour sites (1.51 x 10(8)) relative to the controls (2.8 x 10(7); P = 0.0008, Wilcoxon). The species isolated in increased numbers at tumour sites were Veillonella, Fusobacterium, Prevotella, Porphyromonas, Actinomyces and Clostridium (anaerobes), and Haemophilus, Enterobacteriaceae and Streptococcus spp. (aerobes). Candida albicans was found at eight of the 21 tumour sites, but never at control sites. It was concluded that human oral carcinoma surface biofilms harbour significantly increased numbers of aerobes and anaerobes as compared with the healthy mucosal surface of the same patient. Candida albicans can also be present in these biofilms. These findings must be considered in relation to the known predisposition of such patients to systemic infections, and to the unpleasant complications of oral morbidity due to infected lesions.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Cell Infect Microbiol
                Front Cell Infect Microbiol
                Front. Cell. Infect. Microbiol.
                Frontiers in Cellular and Infection Microbiology
                Frontiers Media S.A.
                2235-2988
                24 February 2017
                2017
                : 7
                : 57
                Affiliations
                [1] 1Department of Periodontics, School of Stomatology, China Medical University Shenyang, China
                [2] 2Key laboratory of Liaoning Province Oral Disease, School of Stomatology, China Medical University Shenyang, China
                [3] 3Department of Oral Biology, School of Stomatology, China Medical University Shenyang, China
                [4] 4Department of Medicine, the Center for Immunity, Inflammation & Regenerative Medicine, University of Virginia Charlottesville, VA, USA
                Author notes

                Edited by: Ingar Olsen, University of Oslo, Norway

                Reviewed by: Özlem Yilmaz, Medical University of South Carolina, USA; Alexa Laheij, Academic Center for Dentistry Amsterdam, Netherlands

                *Correspondence: Yaping Pan yppan@ 123456cmu.edu.cn
                Article
                10.3389/fcimb.2017.00057
                5323389
                28286742
                e2b9a40f-f44f-4f1c-8495-7a2c220a4b9f
                Copyright © 2017 Geng, Liu, Guo, Li, Wang, Wang, Zhao and Pan.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 20 November 2016
                : 13 February 2017
                Page count
                Figures: 8, Tables: 1, Equations: 0, References: 72, Pages: 16, Words: 10529
                Funding
                Funded by: National Natural Science Foundation of China 10.13039/501100001809
                Award ID: 81470745
                Categories
                Microbiology
                Original Research

                Infectious disease & Microbiology
                porphyromonas gingivalis,oscc,long-term infection,inflammatory microenvironment,tumorigenic properties

                Comments

                Comment on this article