1
|
M D, P A, Smiline Girija A, A P, Priyadharsini J V. Differential gene expression profile in Porphyromonas gingivalis treated human gingival keratinocytes and their role in the development of HNSCC. J Oral Biol Craniofac Res 2025; 15:48-56. [PMID: 39717877 PMCID: PMC11664403 DOI: 10.1016/j.jobcr.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 08/28/2024] [Accepted: 11/25/2024] [Indexed: 12/25/2024] Open
Abstract
Background Periodontitis is considered to be one of the major risk factors associated with cancers of the oral cavity. Periodontogenic pathogens such as Porphyromonas gingivalis, Treponema denticola, Tannerella forsythia, Fusobacterium nucleatum and Aggregatibacter actinomycetemcomitans are the important pathogens associated with periodontitis. Chronic exposure to bacterial components induces changes in the nearby cells. Hence, the present study has been designed to identify the molecular mechanisms that could be associated with the two disease conditions viz., periodontitis and head and neck cancer. Objective The present study investigated the differential gene expression profile in human gingival keratinocytes treated with P. gingivalis (Pg), a bacterium associated with periodontal disease, and its possible association with the development of Head and Neck Squamous Cell Carcinoma (HNSCC). Methods The study followed a computational design. Multiple tools and databases, such as GEOmnibus, STRING, Metascape, PANTHER, and UALCAN, cBioportal, were used to derive an association between gene expression during infection with P. gingivalis, and the resulting gene expression profiles were analyzed in the HNSCC dataset. Results The study revealed 29 genes from a pool of transcripts acquired after comparing the Pg-HIGK and Sham-HIGK. Among them, 3 genes i.e., FST, VRK3, and SGK1, were found to be overexpressed and significantly influenced patient survival. The upregulation of FST was found to correlate with poor prognosis in HNSCC patients. Conclusion The study provided insights into the possible association of FST, VRK3 and SGK1 in the development of HNSCC. Further investigations are warranted to confirm the functional role of these genes in establishing the cancer phenotype in patients with chronic infection with Pg.
Collapse
Affiliation(s)
- Dakshinya M
- Clinical Genetics Lab, Centre for Cellular and Molecular Research, Saveetha Dental College & Hospital, Saveetha Institute of Medical and Technical Sciences [SIMATS], Saveetha University, Chennai, India
- Department of Microbiology, Centre for Infectious Diseases, Saveetha Dental College & Hospital, Saveetha Institute of Medical and Technical Sciences [SIMATS], Saveetha University, Chennai, India
| | - Anitha P
- Clinical Genetics Lab, Centre for Cellular and Molecular Research, Saveetha Dental College & Hospital, Saveetha Institute of Medical and Technical Sciences [SIMATS], Saveetha University, Chennai, India
| | - A.S. Smiline Girija
- Department of Microbiology, Centre for Infectious Diseases, Saveetha Dental College & Hospital, Saveetha Institute of Medical and Technical Sciences [SIMATS], Saveetha University, Chennai, India
| | - Paramasivam A
- Clinical Genetics Lab, Centre for Cellular and Molecular Research, Saveetha Dental College & Hospital, Saveetha Institute of Medical and Technical Sciences [SIMATS], Saveetha University, Chennai, India
| | - Vijayashree Priyadharsini J
- Clinical Genetics Lab, Centre for Cellular and Molecular Research, Saveetha Dental College & Hospital, Saveetha Institute of Medical and Technical Sciences [SIMATS], Saveetha University, Chennai, India
| |
Collapse
|
2
|
Li Y, Ma Y, Yu J, Li C, Yu D, Dai R, Li Q, Cao CY. A dual functional polypeptide with antibacterial and anti-inflammatory properties for the treatment of periodontitis. Int J Biol Macromol 2023; 242:124920. [PMID: 37196724 DOI: 10.1016/j.ijbiomac.2023.124920] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/29/2023] [Accepted: 05/14/2023] [Indexed: 05/19/2023]
Abstract
Periodontitis has been reported as the sixth most prevalent disease in human beings. This destructive disease is closely related to systemic diseases. Existing local drug delivery systems for periodontitis suffer from poor antibacterial effect and drug resistance. Inspired by the pathogenesis of periodontitis, we implemented a strategy to construct a dual functional polypeptide LL37-C15, which exhibited remarkable antibacterial effect against P. gingivalis and A. actinomycetemcomitans. In addition, LL37-C15 inhibits the release of pro-inflammatory cytokines by controlling the inflammatory pathway and reversing macrophage M1. Furthermore, the anti-inflammatory effect of LL37-C15 was also verified in vivo in a periodontitis rat model through the morphometry and histological observations of alveolar bone, hematoxylin-eosin, and Trap staining in gingival tissue. The results of molecular dynamics simulations showed that LL37-C15 could selectively destroy the bacterial cell membrane and protect the animal cell membrane in a self-destructive manner. The results showed that the polypeptide LL37-C15, as a novel promising therapeutic agent, exhibited a great potential for the periodontitis management. What's more, this dual functional polypeptide provides a promising strategy for building a multifunctional therapeutic platform against the inflammation and other diseases.
Collapse
Affiliation(s)
- Yuexiang Li
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China
| | - Yunfeng Ma
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China
| | - Jianan Yu
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China
| | - Cancan Li
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China
| | - Da Yu
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China
| | - Ruoxi Dai
- Tufts University School of Dental Medicine, Department of Comprehensive Care, Boston, MA 02111, USA
| | - Quanli Li
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China
| | - Chris Ying Cao
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China.
| |
Collapse
|
3
|
Lee EJ, Kim Y, Salipante P, Kotula AP, Lipshutz S, Graves DT, Alimperti S. Mechanical Regulation of Oral Epithelial Barrier Function. Bioengineering (Basel) 2023; 10:bioengineering10050517. [PMID: 37237587 DOI: 10.3390/bioengineering10050517] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/18/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
Epithelial cell function is modulated by mechanical forces imparted by the extracellular environment. The transmission of forces onto the cytoskeleton by modalities such as mechanical stress and matrix stiffness is necessary to address by the development of new experimental models that permit finely tuned cell mechanical challenges. Herein, we developed an epithelial tissue culture model, named the 3D Oral Epi-mucosa platform, to investigate the role mechanical cues in the epithelial barrier. In this platform, low-level mechanical stress (0.1 kPa) is applied to oral keratinocytes, which lie on 3D fibrous collagen (Col) gels whose stiffness is modulated by different concentrations or the addition of other factors such as fibronectin (FN). Our results show that cells lying on intermediate Col (3 mg/mL; stiffness = 30 Pa) demonstrated lower epithelial leakiness compared with soft Col (1.5 mg/mL; stiffness = 10 Pa) and stiff Col (6 mg/mL; stiffness = 120 Pa) gels, indicating that stiffness modulates barrier function. In addition, the presence of FN reversed the barrier integrity by inhibiting the interepithelial interaction via E-cadherin and Zonula occludens-1. Overall, the 3D Oral Epi-mucosa platform, as a new in vitro system, will be utilized to identify new mechanisms and develop future targets involved in mucosal diseases.
Collapse
Affiliation(s)
- Eun-Jin Lee
- Department of Biochemistry and Molecular & Cellular Biology, School of Medicine, Georgetown University, Washington, DC 20057, USA
- Microsystems and Nanotechnology Division, Physical Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
- Department of Chemistry and Biochemistry, College of Computer, Mathematical and Natural Sciences, University of Maryland, College Park, MD 20742, USA
| | - Yoontae Kim
- Department of Biochemistry and Molecular & Cellular Biology, School of Medicine, Georgetown University, Washington, DC 20057, USA
| | - Paul Salipante
- Materials Science and Engineering Division, Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
| | - Anthony P Kotula
- Materials Science and Engineering Division, Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
| | - Sophie Lipshutz
- Department of Biochemistry and Molecular & Cellular Biology, School of Medicine, Georgetown University, Washington, DC 20057, USA
| | - Dana T Graves
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stella Alimperti
- Department of Biochemistry and Molecular & Cellular Biology, School of Medicine, Georgetown University, Washington, DC 20057, USA
| |
Collapse
|
4
|
Predicting Key Genes and Therapeutic Molecular Modelling to Explain the Association between Porphyromonas gingivalis (P. gingivalis) and Alzheimer’s Disease (AD). Int J Mol Sci 2023; 24:ijms24065432. [PMID: 36982508 PMCID: PMC10049565 DOI: 10.3390/ijms24065432] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/01/2023] [Accepted: 03/03/2023] [Indexed: 03/14/2023] Open
Abstract
The association between Porphyromonas gingivalis (P. gingivalis) and Alzheimer’s disease (AD) remains unclear. The major aim of this study was to elucidate the role of genes and molecular targets in P. gingivalis-associated AD. Two Gene Expression Omnibus (GEO) datasets, GSE5281 for AD (n = 84 Alzheimer’s, n = 74 control) and GSE9723 (n = 4 P. gingivalis, n = 4 control), were downloaded from the GEO database. Differentially expressed genes (DEGs) were obtained, and genes common to both diseases were drawn. Additionally, Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analysis was performed from the top 100 genes (50 upregulated and 50 downregulated genes). We then proceeded with CMap analysis to screen for possible small drug molecules targeting these genes. Subsequently, we performed molecular dynamics simulations. A total of 10 common genes (CALD1, HES1, ID3, PLK2, PPP2R2D, RASGRF1, SUN1, VPS33B, WTH3DI/RAB6A, and ZFP36L1) were identified with a p-value < 0.05. The PPI network of the top 100 genes showed UCHL1, SST, CHGB, CALY, and INA to be common in the MCC, DMNC, and MNC domains. Out of the 10 common genes identified, only 1 was mapped in CMap. We found three candidate small drug molecules to be a fit for PLK2, namely PubChem ID: 24971422, 11364421, and 49792852. We then performed molecular docking of PLK2 with PubChem ID: 24971422, 11364421, and 49792852. The best target, 11364421, was used to conduct the molecular dynamics simulations. The results of this study unravel novel genes to P. gingivalis-associated AD that warrant further validation.
Collapse
|
5
|
Lamont RJ, Fitzsimonds ZR, Wang H, Gao S. Role of Porphyromonas gingivalis in oral and orodigestive squamous cell carcinoma. Periodontol 2000 2022; 89:154-165. [PMID: 35244980 DOI: 10.1111/prd.12425] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Oral and esophageal squamous cell carcinomas harbor a diverse microbiome that differs compositionally from precancerous and healthy tissues. Though causality is yet to be definitively established, emerging trends implicate periodontal pathogens such as Porphyromonas gingivalis as associated with the cancerous state. Moreover, infection with P. gingivalis correlates with a poor prognosis, and P. gingivalis is oncopathogenic in animal models. Mechanistically, properties of P. gingivalis that have been established in vitro and could promote tumor development include induction of a dysbiotic inflammatory microenvironment, inhibition of apoptosis, increased cell proliferation, enhanced angiogenesis, activation of epithelial-to-mesenchymal transition, and production of carcinogenic metabolites. The microbial community context is also relevant to oncopathogenicity, and consortia of P. gingivalis and Fusobacterium nucleatum are synergistically pathogenic in oral cancer models in vivo. In contrast, oral streptococci, such as Streptococcus gordonii, can antagonize protumorigenic epithelial cell phenotypes induced by P. gingivalis, indicating functionally specialized roles for bacteria in oncogenic communities. Consistent with the notion of the bacterial community constituting the etiologic unit, metatranscriptomic data indicate that functional, rather than compositional, properties of the tumor-associated communities have more relevance to cancer development. A consistent association of P. gingivalis with oral and orodigestive carcinoma could have diagnostic potential for early detection of these conditions that have a high incidence and low survival rates.
Collapse
Affiliation(s)
- Richard J Lamont
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky, USA
| | - Zackary R Fitzsimonds
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky, USA
| | - Huizhi Wang
- Department of Oral and Craniofacial Molecular Biology, VCU School of Dentistry, Richmond, Virginia, USA
| | - Shegan Gao
- Henan Key Laboratory of Cancer Epigenetics, Cancer Institute, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
6
|
Molecular Mechanisms Leading from Periodontal Disease to Cancer. Int J Mol Sci 2022; 23:ijms23020970. [PMID: 35055157 PMCID: PMC8778447 DOI: 10.3390/ijms23020970] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/07/2022] [Accepted: 01/14/2022] [Indexed: 12/12/2022] Open
Abstract
Periodontitis is prevalent in half of the adult population and raises critical health concerns as it has been recently associated with an increased risk of cancer. While information about the topic remains somewhat scarce, a deeper understanding of the underlying mechanistic pathways promoting neoplasia in periodontitis patients is of fundamental importance. This manuscript presents the literature as well as a panel of tables and figures on the molecular mechanisms of Porphyromonas gingivalis and Fusobacterium nucleatum, two main oral pathogens in periodontitis pathology, involved in instigating tumorigenesis. We also present evidence for potential links between the RANKL–RANK signaling axis as well as circulating cytokines/leukocytes and carcinogenesis. Due to the nonconclusive data associating periodontitis and cancer reported in the case and cohort studies, we examine clinical trials relevant to the topic and summarize their outcome.
Collapse
|
7
|
Comprehensive analysis of transcriptional profiles in oral epithelial-like cells stimulated with oral probiotic Lactobacillus spp. Arch Oral Biol 2020; 118:104832. [PMID: 32739629 DOI: 10.1016/j.archoralbio.2020.104832] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/05/2020] [Accepted: 07/07/2020] [Indexed: 12/29/2022]
Abstract
OBJECTIVE The mechanisms of action of probiotics can vary among species and among strains of a single species; thus, they can affect host cells in a complex manner. In the present study, Lactobacillus spp. were evaluated for their ability to adhere to gingival epithelial-like cells. Comprehensive analyses of transcriptional profiles of mouse gingival epithelial GE1 cells treated with L. rhamnosus L8020 were performed to assess the putative in vivo probiotic potential of this strain. METHODS Five Lactobacillus spp., isolated from the oral cavity, traditional Bulgarian yoghurt, and the feces of a healthy human, were each co-cultured with GE1 cells. Adhesion assays with serial dilution plating and DNA microarray analysis were performed to identify differentially expressed genes (DEGs) in GE1 cells grown in co-culture with L. rhamnosus L8020. RESULTS The oral isolates L. rhamnosus L8020, L. casei YU3, and L. paracasei YU4 demonstrated significantly greater adhesion compared with the non-oral isolates. In total, 536 genes in GE1 cells exhibited more than twofold upregulation or downregulation, compared with the 0 h timepoint, during co-culture with L. rhamnosus L8020. Gene ontology enrichment analysis revealed that DEGs were differentially enriched in a time-dependent manner. Early responses involved widespread changes in gene expression. CONCLUSIONS This study reveals changes in expression of genes involved in the epithelial physical barrier and immune response in gingival epithelial-like cells co-cultured with L. rhamnosus L8020. Further investigations regarding the molecular mechanisms by which L. rhamnosus L8020 serves as a probiotic may provide evidence to support clinical use.
Collapse
|
8
|
In Vitro Effects of Streptococcus oralis Biofilm on Peri-Implant Soft Tissue Cells. Cells 2020; 9:cells9051226. [PMID: 32429151 PMCID: PMC7290395 DOI: 10.3390/cells9051226] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 05/12/2020] [Indexed: 12/13/2022] Open
Abstract
Human gingival epithelial cells (HGEps) and fibroblasts (HGFs) are the main cell types in peri-implant soft tissue. HGEps are constantly exposed to bacteria, but HGFs are protected by connective tissue as long as the mucosa-implant seal is intact. Streptococcus oralis is one of the commensal bacteria, is highly abundant at healthy implant sites, and might modulate soft tissue cells-as has been described for other streptococci. We have therefore investigated the effects of the S. oralis biofilm on HGEps and HGFs. HGEps or HGFs were grown separately on titanium disks and responded to challenge with S. oralis biofilm. HGFs were severely damaged after 4 h, exhibiting transcriptional inflammatory and stress responses. In contrast, challenge with S. oralis only induced a mild transcriptional inflammatory response in HGEps, without cellular damage. HGFs were more susceptible to the S. oralis biofilm than HGEps. The pro-inflammatory interleukin 6 (IL-6) was attenuated in HGFs, as was interleukin 8 (CXCL8) in HGEps. This indicates that S. oralis can actively protect tissue. In conclusion, commensal biofilms can promote homeostatic tissue protection, but only if the implant-mucosa interface is intact and HGFs are not directly exposed.
Collapse
|
9
|
Hanel AN, Herzog HM, James MG, Cuadra GA. Effects of Oral Commensal Streptococci on Porphyromonas gingivalis Invasion into Oral Epithelial Cells. Dent J (Basel) 2020; 8:E39. [PMID: 32370286 PMCID: PMC7345648 DOI: 10.3390/dj8020039] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 04/25/2020] [Accepted: 04/30/2020] [Indexed: 02/07/2023] Open
Abstract
The objective of this study was to determine if the interaction between common oral commensal bacteria and oral epithelial cells would provide protective effects against the invasion of periodontopathogen Porphyromonas gingivalis. Oral epithelial OKF6/Tert cells were used in co-cultures with Streptococcus gordonii, Streptococcus oralis, Streptococcus mitis, and Streptococcus intermedius. The viability of OKF6/Tert cells following a bacterial challenge was evaluated by trypan blue exclusion. The adherence of commensal species was determined by CFU counts. P. gingivalis invasion in OKF6/Tert cells was assessed before and after exposure to commensal species according to CFU counts. Viability assays show that only S. gordonii and S. intermedius display low toxicity toward OKF6/Tert cells. Both commensals adhere to OKF6/Tert cells at an average ratio of 1 CFU to 10 cells. P. gingivalis invasion into host cells is significantly reduced by 25% or 60% after exposure to S. gordonii or S. intermedius, respectively. The results suggest that these commensal species bind to host cells and diminish P. gingivalis invasion. This is important in the context of periodontal disease since P. gingivalis primarily acts on the host by invading it. Therefore, efforts to decrease invasion will eventually lead to future therapies harnessing the mechanisms employed by oral commensal bacteria.
Collapse
Affiliation(s)
- Alyssa N. Hanel
- Department of Biology, Muhlenberg College, 2400 W. Chew Street, Allentown, PA 18104, USA; (A.N.H.); (H.M.H.); (M.G.J.)
- College of Dental Medicine, Columbia University, 622 W 168th St, New York, NY 10032, USA
| | - Hannah M. Herzog
- Department of Biology, Muhlenberg College, 2400 W. Chew Street, Allentown, PA 18104, USA; (A.N.H.); (H.M.H.); (M.G.J.)
| | - Michelle G. James
- Department of Biology, Muhlenberg College, 2400 W. Chew Street, Allentown, PA 18104, USA; (A.N.H.); (H.M.H.); (M.G.J.)
| | - Giancarlo A. Cuadra
- Department of Biology, Muhlenberg College, 2400 W. Chew Street, Allentown, PA 18104, USA; (A.N.H.); (H.M.H.); (M.G.J.)
| |
Collapse
|
10
|
Distinct Signaling Pathways Between Human Macrophages and Primary Gingival Epithelial Cells by Aggregatibacter actinomycetemcomitans. Pathogens 2020; 9:pathogens9040248. [PMID: 32230992 PMCID: PMC7238148 DOI: 10.3390/pathogens9040248] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 03/23/2020] [Accepted: 03/23/2020] [Indexed: 12/12/2022] Open
Abstract
In aggressive periodontitis, the dysbiotic microbial community in the subgingival crevice, which is abundant in Aggregatibacter actinomycetemcomitans, interacts with extra- and intracellular receptors of host cells, leading to exacerbated inflammation and subsequent tissue destruction. Our goal was to understand the innate immune interactions of A. actinomycetemcomitans with macrophages and human gingival epithelial cells (HGECs) on the signaling cascade involved in inflammasome and inflammatory responses. U937 macrophages and HGECs were co-cultured with A. actinomycetemcomitans strain Y4 and key signaling pathways were analyzed using real-time PCR, Western blotting and cytokine production by ELISA. A. actinomycetemcomitans infection upregulated the transcription of TLR2, TLR4, NOD2 and NLRP3 in U937 macrophages, but not in HGECs. Transcription of IL-1β and IL-18 was upregulated in macrophages and HGECs after 1 h interaction with A. actinomycetemcomitans, but positive regulation persisted only in macrophages, resulting in the presence of IL-1β in macrophage supernatant. Immunoblot data revealed that A. actinomycetemcomitans induced the phosphorylation of AKT and ERK1/2, possibly leading to activation of the NF-κB pathway in macrophages. On the other hand, HGEC signaling induced by A. actinomycetemcomitans was distinct, since AKT and 4EBP1 were phosphorylated after stimulation with A. actinomycetemcomitans, whereas ERK1/2 was not. Furthermore, A. actinomycetemcomitans was able to induce the cleavage of caspase-1 in U937 macrophages in an NRLP3-dependent pathway. Differences in host cell responses, such as those seen between HGECs and macrophages, suggested that survival of A. actinomycetemcomitans in periodontal tissues may be favored by its ability to differentially activate host cells.
Collapse
|
11
|
Fitzsimonds ZR, Rodriguez-Hernandez CJ, Bagaitkar J, Lamont RJ. From Beyond the Pale to the Pale Riders: The Emerging Association of Bacteria with Oral Cancer. J Dent Res 2020; 99:604-612. [PMID: 32091956 DOI: 10.1177/0022034520907341] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Oral cancer, predominantly oral squamous cell carcinoma (OSCC), is the eighth-most common cancer worldwide, with a 5-y survival rate <50%. There are numerous risk factors for oral cancer, among which periodontal disease is gaining increasing recognition. The creation of a sustained dysbiotic proinflammatory environment by periodontal bacteria may serve to functionally link periodontal disease and oral cancer. Moreover, traditional periodontal pathogens, such as Porphyromonas gingivalis, Fusobacterium nucleatum, and Treponema denticola, are among the species most frequently identified as being enriched in OSCC, and they possess a number of oncogenic properties. These organisms share the ability to attach and invade oral epithelial cells, and from there each undergoes its own unique molecular dialogue with the host epithelium, which ultimately converges on acquired phenotypes associated with cancer, including inhibition of apoptosis, increased proliferation, and activation of epithelial-to-mesenchymal transition leading to increased migration of epithelial cells. Additionally, emerging properties of structured bacterial communities may increase oncogenic potential, and consortia of P. gingivalis and F. nucleatum are synergistically pathogenic within in vivo oral cancer models. Interestingly, however, some species of oral streptococci can antagonize the phenotypes induced by P. gingivalis, indicating functionally specialized roles for bacteria in oncogenic communities. Transcriptomic data support the concept that functional, rather than compositional, properties of oral bacterial communities have more relevance to cancer development. Collectively, the evidence is consistent with a modified polymicrobial synergy and dysbiosis model for bacterial involvement in OSCC, with driver mutations generating a conducive microenvironment on the epithelial boundary, which becomes further dysbiotic by the synergistic action of bacterial communities.
Collapse
Affiliation(s)
- Z R Fitzsimonds
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, KY, USA
| | - C J Rodriguez-Hernandez
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, KY, USA
| | - J Bagaitkar
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, KY, USA
| | - R J Lamont
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, KY, USA
| |
Collapse
|
12
|
The Msp Protein of Treponema denticola Interrupts Activity of Phosphoinositide Processing in Neutrophils. Infect Immun 2019; 87:IAI.00553-19. [PMID: 31481407 DOI: 10.1128/iai.00553-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 08/27/2019] [Indexed: 12/15/2022] Open
Abstract
Periodontal disease is a significant health burden, causing tooth loss and poor oral and overall systemic health. Dysbiosis of the oral biofilm and a dysfunctional immune response drive chronic inflammation, causing destruction of soft tissue and alveolar bone supporting the teeth. Treponema denticola, a spirochete abundant in the plaque biofilm of patients with severe periodontal disease, perturbs neutrophil function by modulating appropriate phosphoinositide (PIP) signaling. Through a series of immunoblotting and quantitative PCR (qPCR) experiments, we show that Msp does not alter the gene transcription or protein content of key enzymes responsible for PIP3 signaling: 3' phosphatase and tensin homolog (PTEN), phosphatidylinositol 3-kinase (PI3K), or 5' Src homology 2 domain-containing inositol phosphatase 1 (SHIP1). Instead, using immunoblotting and enzyme-linked immunosorbent assays (ELISAs), we found that Msp activates PTEN through dephosphorylation specifically at the S380 site. Msp in intact organisms or outer membrane vesicles also restricts PIP signaling. SHIP1 phosphatase release was assessed using chemical inhibition and immunoprecipitation to show that Msp moderately decreases SHIP1 activity. Msp also prevents secondary activation of the PTEN/PI3K response. We speculate that this result is due to the redirection of the PIP3 substrate away from SHIP1 to PTEN. Immunofluorescence microscopy revealed a redistribution of PTEN from the cytoplasm to the plasma membrane following exposure to Msp, which may contribute to PTEN activation. Mechanisms of how T. denticola modulates and evades the host immune response are still poorly described, and here we provide further mechanistic evidence of how spirochetes modify PIP signaling to dampen neutrophil function. Understanding how oral bacteria evade the immune response to perpetuate the cycle of inflammation and infection is critical for combating periodontal disease to improve overall health outcomes.
Collapse
|
13
|
Ingendoh‐Tsakmakidis A, Mikolai C, Winkel A, Szafrański SP, Falk CS, Rossi A, Walles H, Stiesch M. Commensal and pathogenic biofilms differently modulate peri-implant oral mucosa in an organotypic model. Cell Microbiol 2019; 21:e13078. [PMID: 31270923 PMCID: PMC6771885 DOI: 10.1111/cmi.13078] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 06/14/2019] [Accepted: 06/28/2019] [Indexed: 02/06/2023]
Abstract
The impact of oral commensal and pathogenic bacteria on peri-implant mucosa is not well understood, despite the high prevalence of peri-implant infections. Hence, we investigated responses of the peri-implant mucosa to Streptococcus oralis or Aggregatibacter actinomycetemcomitans biofilms using a novel in vitro peri-implant mucosa-biofilm model. Our 3D model combined three components, organotypic oral mucosa, implant material, and oral biofilm, with structural assembly close to native situation. S. oralis induced a protective stress response in the peri-implant mucosa through upregulation of heat shock protein (HSP70) genes. Attenuated inflammatory response was indicated by reduced cytokine levels of interleukin-6 (IL-6), interleukin-8 (CXCL8), and monocyte chemoattractant protein-1 (CCL2). The inflammatory balance was preserved through increased levels of tumor necrosis factor-alpha (TNF-α). A. actinomycetemcomitans induced downregulation of genes important for cell survival and host inflammatory response. The reduced cytokine levels of chemokine ligand 1 (CXCL1), CXCL8, and CCL2 also indicated a diminished inflammatory response. The induced immune balance by S. oralis may support oral health, whereas the reduced inflammatory response to A. actinomycetemcomitans may provide colonisation advantage and facilitate later tissue invasion. The comprehensive characterisation of peri-implant mucosa-biofilm interactions using our 3D model can provide new knowledge to improve strategies for prevention and therapy of peri-implant disease.
Collapse
Affiliation(s)
| | - Carina Mikolai
- Department of Prosthetic Dentistry and Biomedical Materials ScienceHannover Medical SchoolHannoverGermany
| | - Andreas Winkel
- Department of Prosthetic Dentistry and Biomedical Materials ScienceHannover Medical SchoolHannoverGermany
| | - Szymon P. Szafrański
- Department of Prosthetic Dentistry and Biomedical Materials ScienceHannover Medical SchoolHannoverGermany
| | - Christine S. Falk
- Institute of Transplant ImmunologyHannover Medical SchoolHannoverGermany
| | - Angela Rossi
- Translational Center for Regenerative TherapiesFraunhofer Institute of Silicate Research ISCWürzburgGermany
| | - Heike Walles
- Translational Center for Regenerative TherapiesFraunhofer Institute of Silicate Research ISCWürzburgGermany
- Chair of Tissue Engineering and Regenerative MedicineUniversity Hospital of WürzburgWürzburgGermany
| | - Meike Stiesch
- Department of Prosthetic Dentistry and Biomedical Materials ScienceHannover Medical SchoolHannoverGermany
| |
Collapse
|
14
|
Zhang D, Hou J, Wu Y, Liu Y, Li R, Xu T, Liu J, Pan Y. Distinct gene expression characteristics in epithelial cell- Porphyromonas gingivalis interactions by integrating transcriptome analyses. Int J Med Sci 2019; 16:1320-1327. [PMID: 31692996 PMCID: PMC6818190 DOI: 10.7150/ijms.33728] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 08/24/2019] [Indexed: 12/28/2022] Open
Abstract
Porphyromonas gingivalis is a pivotal periodontal pathogen, and the epithelial cells serve as the first physical barrier to defend the host from bacterial attack. Within this host-bacteria interaction, P. gingivalis can modify the host immune reaction and adjust the gene expression, which is associated with periodontitis pathogenesis and developing strategies. Herein, a meta-analysis was made to get the differential gene expression profiles in epithelial cells with or without P. gingivalis infection. The network-based meta-analysis program for gene expression profiling was used. Both the gene ontology analysis and the pathway enrichment analysis of the differentially expressed genes were conducted. Our results determined that 290 genes were consistently up-regulated in P. gingivalis infected epithelial cells. 229 gene ontology biological process terms of up-regulated genes were discovered, including "negative regulation of apoptotic process" and "positive regulation of cell proliferation/migration/angiogenesis". In addition to the well-known inflammatory signaling pathways, the pathway associated with a transcriptional misregulation in cancer has also been increased. Our findings indicated that P. gingivalis benefited from the survival of epithelial cells, and got its success as a colonizer in oral epithelium. The results also suggested that infection of P. gingivalis might contribute to oral cancer through chronic inflammation. Negative regulation of the apoptotic process and transcriptional misregulation in cancer pathway are important contributors to the cellular physiology changes during infection development, which have particular relevance to the pathogenesis and progressions of periodontitis, even to the occurrence of oral cancer.
Collapse
Affiliation(s)
- Dongmei Zhang
- Department of Periodontics and Oral Biology, School of Stomatology, China Medical University, Shenyang 110002, China
| | - Jingya Hou
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang 110002, China
| | - Yun Wu
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang 110002, China
| | - Yanqing Liu
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang 110002, China
| | - Rong Li
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang 110002, China
| | - Tong Xu
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang 110002, China
| | - Junchao Liu
- Department of Periodontics and Oral Biology, School of Stomatology, China Medical University, Shenyang 110002, China
| | - Yaping Pan
- Department of Periodontics and Oral Biology, School of Stomatology, China Medical University, Shenyang 110002, China
| |
Collapse
|
15
|
Zhu H, Lu S, Wei M, Cai X, Wang G. Identification of novel genes involved in gingival epithelial cells responding to Aggregatibacter actinomycetemcomitans and Porphyromonas gingivalis infections. Arch Oral Biol 2018; 96:113-121. [PMID: 30223242 DOI: 10.1016/j.archoralbio.2018.08.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 08/28/2018] [Accepted: 08/29/2018] [Indexed: 12/22/2022]
Abstract
OBJECTIVE This study aimed to identify the differentially expressed genes (DEGs) in gingiva epithelial cells responding to Aggregatibacter actinomycetemcomitans and Porphyromonas gingivalis infections using bioinformatics method. STUDY DESIGN GSE9723 dataset was downloaded from Gene Expression Omnibus, and DEGs between the infected cells and controls were identified using unpaired t-test. Overlapping DEGs in responding to Aggregatibacter actinomycetemcomitans and Porphyromonas gingivalis infections were extracted. Protein-protein interaction networks were constructed and functional modules were isolated using Molecular Complex Detection algorithm. Key genes in protein-protein interaction network and Molecular Complex Detection modules were subjected to functional enrichment analyses. In addition, the transcriptional factors were predicted. RESULTS A total of 533 co-up-regulated and 202 co-down-regulated genes were identified. The up-regulated genes, including IL6, CCL19, EDN1, ADCY9, and BCL2 and the down-regulated genes, including CCNB1, PLK1, and CCNA2 were the key genes in the protein-protein interaction network and modules. They were intensively enriched in chemokine signaling pathway, calcium signaling pathway and cell cycle. Finally, two transcriptional factors, E12 and NRSF, targeting to the up-regulated genes and one transcriptional factor, NRP1, targeting the down-regulated genes, were predicted. CONCLUSIONS CCNB1, PLK1, and CCNA2 might play important roles in the response of host epithelial cells to Aggregatibacter actinomycetemcomitans and Porphyromonas gingivalis.
Collapse
Affiliation(s)
- Hongguang Zhu
- School of Stomatology of Shandong University, Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan, 250012, China; Department of Dental Medicine, Weifang People's Hospital, Weifang 261041, China
| | - Shouyi Lu
- Department of Dentistry, Weifang People's Hospital, Weifang Medical University, Weifang, 261041, China
| | - Meirong Wei
- Department of Dentistry, Weifang People's Hospital, Weifang Medical University, Weifang, 261041, China
| | - Xiaoshan Cai
- Department of Pathology, Second People's Hospital of Weifang, Weifang, Shandong 261041, China
| | - Guoyou Wang
- Department of Dentistry, Weifang People's Hospital, Weifang Medical University, Weifang, 261041, China.
| |
Collapse
|
16
|
Bloch S, Zwicker S, Bostanci N, Sjöling Å, Boström EA, Belibasakis GN, Schäffer C. Immune response profiling of primary monocytes and oral keratinocytes to different Tannerella forsythia strains and their cell surface mutants. Mol Oral Microbiol 2018; 33:155-167. [PMID: 29235255 DOI: 10.1111/omi.12208] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2017] [Indexed: 12/18/2022]
Abstract
The oral pathogen Tannerella forsythia possesses a unique surface (S-) layer with a complex O-glycan containing a bacterial sialic acid mimic in the form of either pseudaminic acid or legionaminic acid at its terminal position. We hypothesize that different T. forsythia strains employ these stereoisomeric sugar acids for interacting with the immune system and resident host tissues in the periodontium. Here, we show how T. forsythia strains ATCC 43037 and UB4 displaying pseudaminic acid and legionaminic acid, respectively, and selected cell surface mutants of these strains modulate the immune response in monocytes and human oral keratinocytes (HOK) using a multiplex immunoassay. When challenged with T. forsythia, monocytes secrete proinflammatory cytokines, chemokines and vascular endothelial growth factor (VEGF) with the release of interleukin-1β (IL-1β) and IL-7 being differentially regulated by the two T. forsythia wild-type strains. Truncation of the bacteria's O-glycan leads to significant reduction of IL-1β and regulates macrophage inflammatory protein-1. HOK infected with T. forsythia produce IL-1Ra, chemokines and VEGF. Although the two wild-type strains elicit preferential immune responses for IL-8, both truncation of the O-glycan and deletion of the S-layer result in significantly increased release of IL-8, granulocyte-macrophage colony-stimulating factor and monocyte chemoattractant protein-1. Through immunofluorescence and confocal laser scanning microscopy of infected HOK we additionally show that T. forsythia is highly invasive and tends to localize to the perinuclear region. This indicates, that the T. forsythia S-layer and attached sugars, particularly pseudaminic acid in ATCC 43037, contribute to dampening the response of epithelial tissues to initial infection and hence play a pivotal role in orchestrating the bacterium's virulence.
Collapse
Affiliation(s)
- S Bloch
- Department of NanoBiotechnology, NanoGlycobiology unit, Universität für Bodenkultur Wien, Vienna, Austria
| | - S Zwicker
- Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - N Bostanci
- Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Å Sjöling
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - E A Boström
- Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - G N Belibasakis
- Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - C Schäffer
- Department of NanoBiotechnology, NanoGlycobiology unit, Universität für Bodenkultur Wien, Vienna, Austria
| |
Collapse
|
17
|
Kelsen JR, Albenberg L. Does Poor Oral Health Protect Against Inflammatory Bowel Disease? Clin Gastroenterol Hepatol 2017; 15:532-534. [PMID: 28043930 DOI: 10.1016/j.cgh.2016.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 12/21/2016] [Accepted: 12/21/2016] [Indexed: 02/07/2023]
Affiliation(s)
- Judith R Kelsen
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Lindsey Albenberg
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
18
|
Groeger S, Jarzina F, Domann E, Meyle J. Porphyromonas gingivalis activates NFκB and MAPK pathways in human oral epithelial cells. BMC Immunol 2017; 18:1. [PMID: 28056810 PMCID: PMC5217430 DOI: 10.1186/s12865-016-0185-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 12/16/2016] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The bacterial biofilm at the gingival margin induces a host immune reaction. In this local inflammation epithelial cells defend the host against bacterial challenge. Porphyromonas gingivalis (P. gingivalis), a keystone pathogen, infects epithelial cells. The aim of this study was to investigate the activation of signaling cascades in primary epithelial cells and oral cancer cell lines by a profiler PCR array. RESULTS After infection with P. gingivalis membranes the RNA of 16 to 33 of 84 key genes involved in the antibacterial immune response was up-regulated, amongst them were IKBKB (NF-κB signaling pathway), IRF5 (TLR signaling) and JUN, MAP2K4, MAPK14 and MAPK8 (MAPK pathway) in SCC-25 cells and IKBKB, IRF5, JUN, MAP2K4, MAPK14 and MAPK8 in PHGK. Statistically significant up-regulation of IKBKB (4.7 ×), MAP2K4 (4.6 ×), MAPK14 (4.2 ×) and IRF5 (9.8 ×) (p < 0.01) was demonstrated in SCC-25 cells and IKBKB (3.1 ×), MAP2K4 (4.0 ×) MAPK 14 (3.0 ×) (p < 0.05), IRF5 (3.0 ×) and JUN (7.7 ×) (p < 0.01) were up-regulated in PHGK. CONCLUSIONS P. gingivalis membrane up-regulates the expression of genes involved in downstream TLR, NFκB and MAPK signaling pathways involved in the pro-inflammatory immune response in primary and malignant oral epithelial cells.
Collapse
Affiliation(s)
- Sabine Groeger
- Department of Periodontology, Justus-Liebig-University of Giessen, Giessen, Germany.
| | - Fabian Jarzina
- Department of Periodontology, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Eugen Domann
- Institute for Medical Microbiology - German Center for Infection Research, DZIF Partner Site Giessen-Marburg-Langen - Justus-Liebig-University of Giessen, Giessen, Germany
| | - Joerg Meyle
- Department of Periodontology, Justus-Liebig-University of Giessen, Giessen, Germany
| |
Collapse
|
19
|
Zhao H, Rieger S, Abe K, Hewison M, Lisse TS. DNA Damage-Inducible Transcript 4 Is an Innate Surveillant of Hair Follicular Stress in Vitamin D Receptor Knockout Mice and a Regulator of Wound Re-Epithelialization. Int J Mol Sci 2016; 17:ijms17121984. [PMID: 27898044 PMCID: PMC5187784 DOI: 10.3390/ijms17121984] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 11/07/2016] [Accepted: 11/22/2016] [Indexed: 01/06/2023] Open
Abstract
Mice and human patients with impaired vitamin D receptor (VDR) signaling have normal developmental hair growth but display aberrant post-morphogenic hair cycle progression associated with alopecia. In addition, VDR–/– mice exhibit impaired cutaneous wound healing. We undertook experiments to determine whether the stress-inducible regulator of energy homeostasis, DNA damage-inducible transcript 4 (Ddit4), is involved in these processes. By analyzing hair cycle activation in vivo, we show that VDR−/− mice at day 14 exhibit increased Ddit4 expression within follicular stress compartments. At day 29, degenerating VDR−/− follicular keratinocytes, but not bulge stem cells, continue to exhibit an increase in Ddit4 expression. At day 47, when normal follicles and epidermis are quiescent and enriched for Ddit4, VDR−/− skin lacks Ddit4 expression. In a skin wound healing assay, the re-epithelialized epidermis in wildtype (WT) but not VDR−/− animals harbor a population of Ddit4- and Krt10-positive cells. Our study suggests that VDR regulates Ddit4 expression during epidermal homeostasis and the wound healing process, while elevated Ddit4 represents an early growth-arresting stress response within VDR−/− follicles.
Collapse
Affiliation(s)
- Hengguang Zhao
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Sandra Rieger
- Kathryn W. Davis Center for Regenerative Biology and Medicine, Mount Desert Island Biological Laboratory, 159 Old Bar Harbor Road, Salisbury Cove, ME 04672, USA.
| | - Koichiro Abe
- Department of Molecular Life Science, Tokai University School of Medicine, Isehara, Kanagawa 259-1193, Japan.
| | - Martin Hewison
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, The University of Birmingham, Birmingham B15 2TH, UK.
| | - Thomas S Lisse
- Kathryn W. Davis Center for Regenerative Biology and Medicine, Mount Desert Island Biological Laboratory, 159 Old Bar Harbor Road, Salisbury Cove, ME 04672, USA.
- The Jackson Laboratory, Bar Harbor, ME 04609, USA.
| |
Collapse
|
20
|
Abstract
As our knowledge of host-microbial interactions within the oral cavity increases, future treatments are likely to be more targeted. For example, efforts to target a single species or key virulence factors that they produce, while maintaining the natural balance of the resident oral microbiota that acts to modulate the host immune response would be an advantage. Targeted approaches may be directed at the black-pigmented anaerobes, Porphyromonas gingivalis and Prevotella intermedia, associated with periodontitis. Such pigments provide an opportunity for targeted phototherapy with high-intensity monochromatic light. Functional inhibition approaches, including the use of enzyme inhibitors, are also being explored to control periodontitis. More general disruption of dental plaque through the use of enzymes and detergents, alone and in combination, shows much promise. The use of probiotics and prebiotics to improve gastrointestinal health has now led to an interest in using these approaches to control oral disease. More recently the potential of antimicrobial peptides and nanotechnology, through the application of nanoparticles with biocidal, anti-adhesive and delivery capabilities, has been explored. The aim of this review is to consider the current status as regards non-conventional treatment approaches for oral infections with particular emphasis on the plaque-related diseases.
Collapse
Affiliation(s)
- Robert P Allaker
- a Oral Microbiology; Barts and The London School of Medicine & Dentistry; Queen Mary University of London ; London, UK
| | | |
Collapse
|
21
|
Sztukowska MN, Ojo A, Ahmed S, Carenbauer AL, Wang Q, Shumway B, Jenkinson HF, Wang H, Darling DS, Lamont RJ. Porphyromonas gingivalis initiates a mesenchymal-like transition through ZEB1 in gingival epithelial cells. Cell Microbiol 2016; 18:844-58. [PMID: 26639759 DOI: 10.1111/cmi.12554] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 11/18/2015] [Accepted: 12/01/2015] [Indexed: 12/28/2022]
Abstract
The oral anaerobe Porphyromonas gingivalis is associated with the development of cancers including oral squamous cell carcinoma (OSCC). Here, we show that infection of gingival epithelial cells with P. gingivalis induces expression and nuclear localization of the ZEB1 transcription factor, which controls epithelial-mesenchymal transition. P. gingivalis also caused an increase in ZEB1 expression as a dual species community with Fusobacterium nucleatum or Streptococcus gordonii. Increased ZEB1 expression was associated with elevated ZEB1 promoter activity and did not require suppression of the miR-200 family of microRNAs. P. gingivalis strains lacking the FimA fimbrial protein were attenuated in their ability to induce ZEB1 expression. ZEB1 levels correlated with an increase in expression of mesenchymal markers, including vimentin and MMP-9, and with enhanced migration of epithelial cells into matrigel. Knockdown of ZEB1 with siRNA prevented the P. gingivalis-induced increase in mesenchymal markers and epithelial cell migration. Oral infection of mice by P. gingivalis increased ZEB1 levels in gingival tissues, and intracellular P. gingivalis were detected by antibody staining in biopsy samples from OSCC. These findings indicate that FimA-driven ZEB1 expression could provide a mechanistic basis for a P. gingivalis contribution to OSCC.
Collapse
Affiliation(s)
- Maryta N Sztukowska
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky, USA
| | - Akintunde Ojo
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky, USA
| | - Saira Ahmed
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky, USA
| | - Anne L Carenbauer
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky, USA
| | - Qian Wang
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky, USA
| | - Brain Shumway
- Department of Surgical and Hospital Dentistry, University of Louisville School of Dentistry, Louisville, Kentucky, USA
| | | | - Huizhi Wang
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky, USA
| | - Douglas S Darling
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky, USA
| | - Richard J Lamont
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky, USA
| |
Collapse
|
22
|
Coelho ED, Arrais JP, Oliveira JL. Uncovering microbial duality within human microbiomes: A novel algorithm for the analysis of host-pathogen interactions. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2016; 2015:3254-7. [PMID: 26736986 DOI: 10.1109/embc.2015.7319086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Microbial species thrive within human hosts by establishing complex associations between themselves and the host. Even though species diversity can be measured (alpha- and beta-diversity), a methodology to estimate the impact of microorganisms in human pathways is still lacking. In this work we propose a computational approach to estimate which human pathways are targeted the most by microorganisms, while also identifying which microorganisms are prominent in this targeting. Our results were consistent with literature evidence, and thus we propose this methodology as a new prospective approach to be used for screening potentially impacted pathways.
Collapse
|
23
|
Kelsen J, Bittinger K, Pauly-Hubbard H, Posivak L, Grunberg S, Baldassano R, Lewis JD, Wu GD, Bushman FD. Alterations of the Subgingival Microbiota in Pediatric Crohn's Disease Studied Longitudinally in Discovery and Validation Cohorts. Inflamm Bowel Dis 2015; 21:2797-805. [PMID: 26288001 PMCID: PMC4950860 DOI: 10.1097/mib.0000000000000557] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Oral manifestations are common in Crohn's disease (CD). Here we characterized the subgingival microbiota in pediatric patients with CD initiating therapy and after 8 weeks to identify microbial community features associated with CD and therapy. METHODS Pediatric patients with CD were recruited from The Children's Hospital of Pennsylvania. Healthy control subjects were recruited from primary care or orthopedics clinic. Subgingival plaque samples were collected at initiation of therapy and after 8 weeks. Treatment exposures included 5-ASAs, immunomodulators, steroids, and infliximab. The microbiota was characterized by 16S rRNA gene sequencing. The study was repeated in separate discovery (35 CD, 43 healthy) and validation cohorts (43 CD, 31 healthy). RESULTS Most subjects in both cohorts demonstrated clinical response after 8 weeks of therapy (discovery cohort 88%, validation cohort 79%). At week 0, both antibiotic exposure and disease state were associated with differences in bacterial community composition. Seventeen genera were identified in the discovery cohort as candidate biomarkers, of which 11 were confirmed in the validation cohort. Capnocytophaga, Rothia, and TM7 were more abundant in CD relative to healthy controls. Other bacteria were reduced in abundance with antibiotic exposure among CD subjects. CD-associated genera were not enriched compared with healthy controls after 8 weeks of therapy. CONCLUSIONS Subgingival microbial community structure differed with CD and antibiotic use. Results in the discovery cohort were replicated in a separate validation cohort. Several potentially pathogenic bacterial lineages were associated with CD but were not diminished in abundance by antibiotic treatment, suggesting targets for additional surveillance.
Collapse
Affiliation(s)
- Judith Kelsen
- *Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; †Department of Microbiology; and ‡Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Gölz L, Buerfent BC, Hofmann A, Hübner MP, Rühl H, Fricker N, Schmidt D, Johannes O, Jepsen S, Deschner J, Hoerauf A, Nöthen MM, Schumacher J, Jäger A. Genome-wide transcriptome induced by Porphyromonas gingivalis LPS supports the notion of host-derived periodontal destruction and its association with systemic diseases. Innate Immun 2015; 22:72-84. [PMID: 26608307 DOI: 10.1177/1753425915616685] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 10/13/2015] [Indexed: 11/17/2022] Open
Abstract
Chronic periodontitis (CP) is a prevalent pathogen-associated inflammatory disorder characterized by the destruction of tooth-supporting tissues, and linked to several systemic diseases. Both the periodontopathogen Porphyromonas gingivalis (Pg), and the genetically determined host immune response, are hypothesized to play a crucial role in this association. To identify new target genes for CP and its associated systemic diseases, we investigated the transcriptome induced by Pg in human monocytes using a genome-wide approach. Monocytes were isolated from healthy male volunteers of European origin and challenged with the Pg virulence factor LPS. Array-based gene expression analysis comprising >47,000 transcripts was performed followed by pathway analyses. Transcriptional data were validated by protein and cell surface markers. LPS Pg challenge led to the significant induction of 902 transcripts. Besides known periodontitis-associated targets, several new candidates were identified (CCL23↑, INDO↑, GBP 1/4↑, CFB↑, ISG20↑, MIR155HG↑, DHRS9↓). Moreover, various transcripts correspond to the host immune response, and have been linked to cancer, atherosclerosis and arthritis, thus highlighting the systemic impact of CP. Protein data of immunological markers validated our results. The present findings expand understanding of Pg elicited immune responses, and indicate new target genes and pathways of relevance to diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Lina Gölz
- Center of Dento-Maxillo-Facial Medicine, University Hospital of Bonn, Bonn, Germany
| | - Benedikt C Buerfent
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital of Bonn, Bonn, Germany
| | - Andrea Hofmann
- Institute of Human Genetics, University Hospital of Bonn, Bonn, Germany
| | - Marc P Hübner
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital of Bonn, Bonn, Germany
| | - Heiko Rühl
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital of Bonn, Bonn, Germany
| | - Nadine Fricker
- Institute of Human Genetics, University Hospital of Bonn, Bonn, Germany
| | - David Schmidt
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital of Bonn, Bonn, Germany
| | - Oldenburg Johannes
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital of Bonn, Bonn, Germany
| | - Søren Jepsen
- Center of Dento-Maxillo-Facial Medicine, University Hospital of Bonn, Bonn, Germany
| | - James Deschner
- Center of Dento-Maxillo-Facial Medicine, University Hospital of Bonn, Bonn, Germany
| | - Achim Hoerauf
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital of Bonn, Bonn, Germany German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, University Hospital of Bonn, Bonn, Germany
| | | | - Andreas Jäger
- Center of Dento-Maxillo-Facial Medicine, University Hospital of Bonn, Bonn, Germany
| |
Collapse
|
25
|
Coelho ED, Santiago AM, Arrais JP, Oliveira JL. Computational methodology for predicting the landscape of the human–microbial interactome region level influence. J Bioinform Comput Biol 2015; 13:1550023. [DOI: 10.1142/s0219720015500237] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Microbial communities thrive in close association among themselves and with the host, establishing protein–protein interactions (PPIs) with the latter, and thus being able to benefit (positively impact) or disturb (negatively impact) biological events in the host. Despite major collaborative efforts to sequence the Human microbiome, there is still a great lack of understanding their impact. We propose a computational methodology to predict the impact of microbial proteins in human biological events, taking into account the abundance of each microbial protein and its relation to all other microbial and human proteins. This alternative methodology is centered on an improved impact estimation algorithm that integrates PPIs between human and microbial proteins with Reactome pathway data. This methodology was applied to study the impact of 24 microbial phyla over different cellular events, within 10 different human microbiomes. The results obtained confirm findings already described in the literature and explore new ones. We believe the Human microbiome can no longer be ignored as not only is there enough evidence correlating microbiome alterations and disease states, but also the return to healthy states once these alterations are reversed.
Collapse
Affiliation(s)
- Edgar D. Coelho
- Department of Electronics, Telecommunications and Informatics (DETI), Institute of Electronics and Informatics Engineering of Aveiro (IEETA), University of Aveiro, Campus Universitário de Santiago, Aveiro 3810-193, Portugal
| | - André M. Santiago
- Department of Informatics Engineering (DEI), Centre for Informatics and Systems of the University of Coimbra (CISUC), University of Coimbra, Polo2, Pinhal de Marrocos, 3030-290 Coimbra, Portugal
| | - Joel P. Arrais
- Department of Informatics Engineering (DEI), Centre for Informatics and Systems of the University of Coimbra (CISUC), University of Coimbra, Polo2, Pinhal de Marrocos, 3030-290 Coimbra, Portugal
| | - José Luís Oliveira
- Department of Electronics, Telecommunications and Informatics (DETI), Institute of Electronics and Informatics Engineering of Aveiro (IEETA), University of Aveiro, Campus Universitário de Santiago, Aveiro 3810-193, Portugal
| |
Collapse
|
26
|
Soto C, Bugueño I, Hoare A, Gonzalez S, Venegas D, Salinas D, Melgar-Rodríguez S, Vernal R, Gamonal J, Quest AFG, Pérez-Donoso JM, Bravo D. The Porphyromonas gingivalis O antigen is required for inhibition of apoptosis in gingival epithelial cells following bacterial infection. J Periodontal Res 2015; 51:518-28. [PMID: 26530544 DOI: 10.1111/jre.12331] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2015] [Indexed: 01/10/2023]
Abstract
BACKGROUND AND OBJECTIVE Porphyromonas gingivalis infection induces apoptosis inhibition in gingival epithelial cells; however, it is not fully understood which bacterial effectors are involved in this process. The aim of this study is to evaluate whether the P. gingivalis lipopolysaccharide (LPS), specifically the O-antigen region, affects adherence, invasion, viability and apoptosis of gingival epithelial cells. MATERIAL AND METHODS Gingival epithelial cells (OKF6/TERT2 line) were infected by different freshly prepared P. gingivalis clinical isolates, obtained from subjects with chronic periodontitis (CP3 and CP4) and healthy individuals (H1 and H3). Periodontitis and healthy isolates show differences in O-antigen production, as healthy isolates lack the O-antigen region. In addition, cells were infected by a site-specific mutant lacking the O-antigen portion. After 24 h postinfection, cell proliferation, viability and apoptosis were evaluated by Trypan blue, MTS and annexin V assays, respectively. Bacterial invasion, adhesion and proliferation were measured by gentamicin/metronidazole protection assays. Finally, toll-like receptor (TLR)2 and TLR4 mRNA expression was evaluated by quantitative reverse transcription-polymerase chain reaction. Statistical analysis was performed using ANOVA, Tukey's or Dunnett's tests (p < 0.05). RESULTS At 24 h postinfection, strains lacking the O-antigen region (healthy isolates and O-antigen ligase-deficient strain) were unable to increase proliferation and viability, or decrease apoptosis as compared with strains producing intact LPS (periodontitis isolates and reference strain). However, the presence of the O-antigen neither contributed to changes in the ability of the bacteria to adhere to or invade cells, nor to intracellular survival. The presence of O-antigen also increased the expression of TLR4 (nearly sixfold), which correlated with inhibition of apoptosis. CONCLUSION The O-antigen region of P. gingivalis LPS is required to increase gingival epithelial cell viability upon infection by bacteria and this increase is attributable to a reduction in apoptosis. Moreover, although bacterial internalization is required, the effects observed are not due to alterations in P. gingivalis adherence, invasion or intracellular survival. Interestingly, inhibition of apoptosis correlates with increased TLR4 expression, suggesting a role for TLR4 in this process.
Collapse
Affiliation(s)
- C Soto
- Oral Microbiology Laboratory, Department of Pathology and Oral Medicine, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - I Bugueño
- Oral Microbiology Laboratory, Department of Pathology and Oral Medicine, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - A Hoare
- Oral Microbiology Laboratory, Department of Pathology and Oral Medicine, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - S Gonzalez
- Oral Microbiology Laboratory, Department of Pathology and Oral Medicine, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - D Venegas
- Oral Microbiology Laboratory, Department of Pathology and Oral Medicine, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - D Salinas
- Oral Microbiology Laboratory, Department of Pathology and Oral Medicine, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - S Melgar-Rodríguez
- Laboratory of Periodontal Biology, Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - R Vernal
- Laboratory of Periodontal Biology, Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - J Gamonal
- Laboratory of Periodontal Biology, Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - A F G Quest
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago, Chile.,Laboratory of Cell Communication, Center for Molecular Studies of the Cell, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - J M Pérez-Donoso
- BioNanotechnology and Microbiology Laboratory, Center for Bioinformatics and Integrative Biology (CBIB), Faculty of Biological Sciences, Universidad Andres Bello, Santiago, Chile
| | - D Bravo
- Oral Microbiology Laboratory, Department of Pathology and Oral Medicine, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| |
Collapse
|
27
|
Olsen I, Progulske-Fox A. Invasion of Porphyromonas gingivalis strains into vascular cells and tissue. J Oral Microbiol 2015; 7:28788. [PMID: 26329158 PMCID: PMC4557090 DOI: 10.3402/jom.v7.28788] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 08/06/2015] [Accepted: 08/07/2015] [Indexed: 01/06/2023] Open
Abstract
Porphyromonas gingivalis is considered a major pathogen in adult periodontitis and is also associated with multiple systemic diseases, for example, cardiovascular diseases. One of its most important virulence factors is invasion of host cells. The invasion process includes attachment, entry/internalization, trafficking, persistence, and exit. The present review discusses these processes related to P. gingivalis in cardiovascular cells and tissue. Although most P. gingivalis strains invade, the invasion capacity of strains and the mechanisms of invasion including intracellular trafficking among them differ. This is consistent with the fact that there are significant differences in the pathogenicity of P. gingivalis strains. P. gingivalis invasion mechanisms are also dependent on types of host cells. Although much is known about the invasion process of P. gingivalis, we still have little knowledge of its exit mechanisms. Nevertheless, it is intriguing that P. gingivalis can remain viable in human cardiovascular cells and atherosclerotic plaque and later exit and re-enter previously uninfected host cells.
Collapse
Affiliation(s)
- Ingar Olsen
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway;
| | - Ann Progulske-Fox
- Department of Oral Biology and Center for Molecular Microbiology, University of Florida College of Dentistry, Gainesville, FL, USA
| |
Collapse
|
28
|
Noncanonical activation of β-catenin by Porphyromonas gingivalis. Infect Immun 2015; 83:3195-203. [PMID: 26034209 DOI: 10.1128/iai.00302-15] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 05/20/2015] [Indexed: 01/01/2023] Open
Abstract
Porphyromonas gingivalis is an established pathogen in periodontal disease and an emerging pathogen in serious systemic conditions, including some forms of cancer. We investigated the effect of P. gingivalis on β-catenin signaling, a major pathway in the control of cell proliferation and tumorigenesis. Infection of gingival epithelial cells with P. gingivalis did not influence the phosphorylation status of β-catenin but resulted in proteolytic processing. The use of mutants deficient in gingipain production, along with gingipain-specific inhibitors, revealed that gingipain proteolytic activity was required for β-catenin processing. The β-catenin destruction complex components Axin1, adenomatous polyposis coli (APC), and GSK3β were also proteolytically processed by P. gingivalis gingipains. Cell fractionation and Western blotting demonstrated that β-catenin fragments were translocated to the nucleus. The accumulation of β-catenin in the nucleus following P. gingivalis infection was confirmed by immunofluorescence microscopy. A luciferase reporter assay showed that P. gingivalis increased the activity of the β-catenin-dependent TCF/LEF promoter. P. gingivalis did not increase Wnt3a mRNA levels, a finding consistent with P. gingivalis-induced proteolytic processing causing the increase in TCF/LEF promoter activity. Thus, our data indicate that P. gingivalis can induce the noncanonical activation of β-catenin and disassociation of the β-catenin destruction complex by gingipain-dependent proteolytic processing. β-Catenin activation in epithelial cells by P. gingivalis may contribute to a proliferative phenotype.
Collapse
|
29
|
Wang Q, Sztukowska M, Ojo A, Scott DA, Wang H, Lamont RJ. FOXO responses to Porphyromonas gingivalis in epithelial cells. Cell Microbiol 2015; 17:1605-17. [PMID: 25958948 DOI: 10.1111/cmi.12459] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 05/06/2015] [Accepted: 05/07/2015] [Indexed: 12/25/2022]
Abstract
Porphyromonas gingivalis is a prominent periodontal, and emerging systemic, pathogen that redirects host cell signalling pathways and modulates innate immune responses. In this study, we show that P. gingivalis infection induces the dephosphorylation and activation of forkhead box-O (FOXO)1, 3 and 4 in gingival epithelial cells. In addition, immunofluorescence showed that FOXO1 accumulated in the nucleus of P. gingivalis-infected cells. Quantitative reverse transcription PCR demonstrated that transcription of genes involved in protection against oxidative stress (Cat, Sod2, Prdx3), inflammatory responses (IL1β) and anti-apoptosis (Bcl-6) was induced by P. gingivalis, while small-interfering RNA (siRNA)-mediated knockdown of FOXO1 suppressed the transcriptional activation of these genes. P. gingivalis-induced secretion of interleukin (IL)-1β and inhibition of apoptosis were also impeded by FOXO1 knockdown. Neutralization of reactive oxygen species (ROS) by N-acetyl-l-cysteine blocked the activation of FOXO1 by P. gingivalis and concomitantly suppressed the activation of oxidative stress responses, anti-apoptosis programmes and IL-β production. Inhibition of c-Jun-N-terminal kinase (JNK) either pharmacologically or by siRNA, reduced FOXO1 activation and downstream FOXO1-dependent gene regulation in response to P. gingivalis. The results indicate that P. gingivalis-induced ROS activate FOXO transcription factors through JNK signalling, and that FOXO1 controls oxidative stress responses, inflammatory cytokine production and cell survival. These data position FOXO as an important signalling node in the epithelial cell-P. gingivalis interaction, with particular relevance to cell fate and dysbiotic host responses.
Collapse
Affiliation(s)
- Qian Wang
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| | - Maryta Sztukowska
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| | - Akintunde Ojo
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| | - David A Scott
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| | - Huizhi Wang
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| | - Richard J Lamont
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| |
Collapse
|
30
|
Sol A, Wang G, Blotnick E, Golla R, Bachrach G, Muhlrad A. Interaction of the core fragments of the LL-37 host defense peptide with actin. RSC Adv 2014; 5:9361-9367. [PMID: 26726303 DOI: 10.1039/c4ra13007c] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Host defense peptides are effector molecules of the innate immunity that possess antimicrobial and health-promoting properties. Due to their potential therapeutic activities, host defense peptides are being explored as alternatives for antibiotics. The human LL-37 and its shorter, cost-effective, bactericidal core peptide derivates have been suggested for their therapeutic potential. Bacteria evade host defense peptides by proteolytic inactivation. Actin released from necrotized cells and abundant in infected sites was shown to bind and protect LL-37 from microbial proteolytic degradation, and to enable the peptide's antimicrobial action despite the presence of the proteases. Here, we characterized the interactions of the 10-13 residues long LL-37 core peptides with actin. We show that the LL-37 core peptides associate with actin with a lower affinity than that of LL-37. Their association with actin, which is very ionic strength sensitive, is mainly based on electrostatic interactions. Likewise, the antimicrobial activity against Escherichia coli of the minimal antimicrobial peptide KR-12 but not FK-13 nor LL-37 is also very sensitive to salts. In addition, the antimicrobial activity of the FK-13 core peptide is protected by actin against the tested bacterial proteases in a similar manner to that of LL-37, supporting its potential for therapeutic use.
Collapse
Affiliation(s)
- Asaf Sol
- Institute of Dental Sciences, The Hebrew University-Hadassah School of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel. ; Tel: +972 2675 7117
| | - Guangshun Wang
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, USA
| | - Edna Blotnick
- Department of Medical Neurobiology, The Institute for Medical Research-Israel-Canada, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Radha Golla
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, USA
| | - Gilad Bachrach
- Institute of Dental Sciences, The Hebrew University-Hadassah School of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel. ; Tel: +972 2675 7117
| | - Andras Muhlrad
- Institute of Dental Sciences, The Hebrew University-Hadassah School of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel. ; Tel: +972 2675 7117
| |
Collapse
|
31
|
Lamont RJ, Hajishengallis G. Polymicrobial synergy and dysbiosis in inflammatory disease. Trends Mol Med 2014; 21:172-83. [PMID: 25498392 DOI: 10.1016/j.molmed.2014.11.004] [Citation(s) in RCA: 372] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 11/14/2014] [Accepted: 11/17/2014] [Indexed: 12/17/2022]
Abstract
Uncontrolled inflammation of the periodontal area may arise when complex microbial communities transition from a commensal to a pathogenic entity. Communication among constituent species leads to polymicrobial synergy between metabolically compatible organisms that acquire functional specialization within the developing community. Keystone pathogens, even at low abundance, elevate community virulence, and the resulting dysbiotic community targets specific aspects of host immunity to further disable immune surveillance while promoting an overall inflammatory response. Inflammophilic organisms benefit from proteinaceous substrates derived from inflammatory tissue breakdown. Inflammation and dysbiosis reinforce each other, and the escalating environmental changes further select for a pathobiotic community. We have synthesized the polymicrobial synergy and dysbiotic components of the process into a new model for inflammatory diseases.
Collapse
Affiliation(s)
- Richard J Lamont
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, KY, USA.
| | - George Hajishengallis
- Department of Microbiology, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
32
|
RETRACTED ARTICLE: Distinctive pathways characterize A. actinomycetemcomitans and P. gingivalis. Mol Biol Rep 2014; 42:441-9. [DOI: 10.1007/s11033-014-3785-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 09/30/2014] [Indexed: 10/24/2022]
|
33
|
Belibasakis G, Bao K, Bostanci N. Transcriptional profiling of human gingival fibroblasts in response to multi-speciesin vitrosubgingival biofilms. Mol Oral Microbiol 2014; 29:174-83. [DOI: 10.1111/omi.12053] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2014] [Indexed: 12/31/2022]
Affiliation(s)
- G.N. Belibasakis
- Oral Microbiology and Immunology; Institute of Oral Biology; Centre of Dental Medicine; University of Zürich; Zürich Switzerland
| | - K. Bao
- Oral Translational Research; Institute of Oral Biology; Centre of Dental Medicine; University of Zürich; Zürich Switzerland
| | - N. Bostanci
- Oral Translational Research; Institute of Oral Biology; Centre of Dental Medicine; University of Zürich; Zürich Switzerland
| |
Collapse
|
34
|
Hajishengallis G, Lamont RJ. Breaking bad: manipulation of the host response by Porphyromonas gingivalis. Eur J Immunol 2014; 44:328-38. [PMID: 24338806 DOI: 10.1002/eji.201344202] [Citation(s) in RCA: 249] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 12/02/2013] [Accepted: 12/09/2013] [Indexed: 02/06/2023]
Abstract
Recent metagenomic and mechanistic studies are consistent with a new model of periodontal pathogenesis. This model proposes that periodontal disease is initiated by a synergistic and dysbiotic microbial community rather than by a select few bacteria traditionally known as "periopathogens." Low-abundance bacteria with community-wide effects that are critical for the development of dysbiosis are now known as keystone pathogens, the best-documented example of which is Porphyromonas gingivalis. Here, we review established mechanisms by which P. gingivalis interferes with host immunity and enables the emergence of dysbiotic communities. We integrate the role of P. gingivalis with that of other bacteria acting upstream and downstream in pathogenesis. Accessory pathogens act upstream to facilitate P. gingivalis colonization and co-ordinate metabolic activities, whereas commensals-turned pathobionts act downstream and contribute to destructive inflammation. The recent concepts of keystone pathogens, along with polymicrobial synergy and dysbiosis, have profound implications for the development of therapeutic options for periodontal disease.
Collapse
Affiliation(s)
- George Hajishengallis
- Department of Microbiology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
35
|
Porphyromonas gingivalis modulates Pseudomonas aeruginosa-induced apoptosis of respiratory epithelial cells through the STAT3 signaling pathway. Microbes Infect 2013; 16:17-27. [PMID: 24140557 DOI: 10.1016/j.micinf.2013.10.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 09/22/2013] [Accepted: 10/04/2013] [Indexed: 12/31/2022]
Abstract
Pseudomonas aeruginosa is an important opportunistic bacterial pathogen, causing infections of respiratory and other organ systems in immunocompromised hosts that may invade and proliferate in mucosal epithelial cells to induce apoptosis. Previous studies suggest that oral bacteria, especially gram-negative periodontal pathogens, may enhance P. aeruginosa invasion into respiratory epithelial cells to augment tissue destruction. In this study, we investigated the effect of the periodontopathogen Porphyromonas gingivalis on P. aeruginosa-induced epithelial cell apoptosis. P. gingivalis invasion transiently inhibited P. aeruginosa-induced apoptosis in respiratory epithelial cells via the signal transducer and activator of transcription 3 (STAT3) signaling pathway. The activated STAT3 up-regulated the downstream anti-apoptotic moleculars survivin and B-cell leukemia-2 (bcl-2). This process was accompanied by down-regulation of pro-apoptosis molecular Bcl-2-associated death promoter (bad) and caspase-3 activity inhibition. In addition, the activation of the STAT3 pathway was affected by P. gingivalis in a dose-dependent manner. Finally, co-invasion of P. aeruginosa and P. gingivalis led to greater cell death compared with P. aeruginosa challenge alone. These results suggest that regulation of P. aeruginosa-induced apoptosis by P. gingivalis contributes to the pathogenesis of respiratory disease. Interference with this process may provide a potential therapeutic strategy for the treatment and prevention of respiratory disease.
Collapse
|
36
|
Nissen L, Sgorbati B, Biavati B, Belibasakis GN. Lactobacillus salivarius and L. gasseri down-regulate Aggregatibacter actinomycetemcomitans exotoxins expression. ANN MICROBIOL 2013; 64:611-617. [PMID: 24860281 PMCID: PMC4028514 DOI: 10.1007/s13213-013-0694-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 07/11/2013] [Indexed: 12/17/2022] Open
Abstract
Beneficial microbes, such as lactobacilli establish a symbiosis with the host and confer health-associated effects, by limiting the growth of indigenous pathogens and challenging microbes introduced by altered foods. Nevertheless, there is scarce information on the effects of beneficial microbes on the virulence properties of bacterial species associated with oral diseases, such as periodontitis. Aggregatibacter actinomycetemcomitans is a Gram-negative species highly implicated in the etiology of localized aggressive periodontitis. The objective of this study was to investigate the effect of lactobacilli on the expression of the two major virulence factors of A. actinomycetemcomitans. Lactobacillus salivarius and L. gasseri were selected as beneficial species. The gene expressions of leukotoxin (LtxA) and cytolethal distending toxin (CdtB) by A. actinomycetemcomitans were analyzed in response to challenge by lactobacilli cell-free supernatants. Neither lactobacilli affected the growth, but strongly attenuated the expressions of both CdtB and LtxA in the two A. actinomycetemcomitans strains tested. This reduction of the expression of these two exotoxins was time-dependent. These fundamental findings may indicate that lactobacilli can reduce the virulence of putative opportunistic oral pathogens, and may provide insights to future therapeutic approaches for the respective diseases.
Collapse
Affiliation(s)
- Lorenzo Nissen
- />Microbiology Area, Department. Agricultural Sciences, Alma Mater Studiorum, Università di Bologna, V.le Fanin 44, 40127 Bologna, Italy
| | - Barbara Sgorbati
- />Microbiology Area, Department. Agricultural Sciences, Alma Mater Studiorum, Università di Bologna, V.le Fanin 44, 40127 Bologna, Italy
| | - Bruno Biavati
- />Microbiology Area, Department. Agricultural Sciences, Alma Mater Studiorum, Università di Bologna, V.le Fanin 44, 40127 Bologna, Italy
| | - Georgios N. Belibasakis
- />Oral Microbiology and Immunology, Institute of Oral Biology, Center of Dental Medicine, University of Zürich, Plattenstrasse 11, 8032 Zürich, Switzerland
| |
Collapse
|
37
|
Abstract
Porphyromonas gingivalis is a major pathogen in periodontal disease and is associated with immune dysbiosis. In this study, we found that P. gingivalis did not induce the expression of the T-cell chemokine IP-10 (CXCL10) from neutrophils, peripheral blood mononuclear cells (PBMCs), or gingival epithelial cells. Furthermore, P. gingivalis suppressed gamma interferon (IFN-γ)-stimulated release of IP-10, ITAC (CXCL11), and Mig (CXCL9) from epithelial cells and inhibited IP-10 secretion in a mixed infection with the otherwise stimulatory Fusobacterium nucleatum. Inhibition of chemokine expression occurred at the level of gene transcription and was associated with downregulation of interferon regulatory factor 1 (IRF-1) and decreased levels of Stat1. Ectopic expression of IRF-1 in epithelial cells relieved P. gingivalis-induced inhibition of IP-10 release. Direct contact between P. gingivalis and epithelial cells was not required for IP-10 inhibition. These results highlight the immune-disruptive potential of P. gingivalis. Suppression of IP-10 and other Th1-biasing chemokines by P. gingivalis may perturb the balance of protective and destructive immunity in the periodontal tissues and facilitate the pathogenicity of oral microbial communities.
Collapse
|
38
|
Binding of Streptococcus gordonii to oral epithelial monolayers increases paracellular barrier function. Microb Pathog 2013; 56:53-9. [DOI: 10.1016/j.micpath.2012.11.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Revised: 10/25/2012] [Accepted: 11/08/2012] [Indexed: 11/22/2022]
|
39
|
Pan C, Xu X, Tan L, Lin L, Pan Y. The effects of Porphyromonas gingivalis on the cell cycle progression of human gingival epithelial cells. Oral Dis 2013; 20:100-8. [PMID: 23444918 DOI: 10.1111/odi.12081] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 01/01/2013] [Accepted: 01/23/2013] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Porphyromonas gingivalis is a major pathogen in the development and progression of periodontal disease. The interactions or cross-talk between bacteria and gingival epithelial cells drive bacteria to manipulate the cell cycle to favor bacterial survival and virulence expression within the host. This study aims to dissect the effects of P. gingivalis on the cell cycle in human gingival epithelial cells. MATERIALS AND METHODS We established a model of P. gingivalis invading IHGE cells. The cell cycle distribution of human gingival epithelial cells was analyzed by flow cytometry. Cyclin D and cyclin E mRNA and protein were detected by real-time PCR and Western blot, respectively. RESULTS Porphyromonas gingivalis-induced facilitation of cell growth was correlated with the acceleration of G1 phase of cell cycle. Cyclin D1 mRNA levels were significantly upregulated from 6 to 12 h after infection. Cyclin E protein and mRNA levels were elevated at 10 and 12 h after invasion. CONCLUSIONS We confirmed that P. gingivalis significantly enhances IHGE cell proliferation by promoting the G1/S transition, involving the up-regulation of cyclin D and cyclin E.
Collapse
Affiliation(s)
- C Pan
- Periodontics, School of Stomatology, China Medical University, Shenyang, China
| | | | | | | | | |
Collapse
|
40
|
Moffatt-Jauregui CE, Robinson B, de Moya AV, Brockman RD, Roman AV, Cash MN, Culp DJ, Lamont RJ. Establishment and characterization of a telomerase immortalized human gingival epithelial cell line. J Periodontal Res 2013; 48:713-21. [PMID: 23441958 DOI: 10.1111/jre.12059] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2013] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND OBJECTIVE Gingival keratinocytes are used in model systems to investigate the interaction between periodontal bacteria and the epithelium in the initial stages of the periodontal disease process. Primary gingival epithelial cells (GECs) have a finite lifespan in culture before they enter senescence and cease to replicate, while epithelial cells immortalized with viral proteins can exhibit chromosomal rearrangements. The aim of this study was to generate a telomerase immortalized human gingival epithelial cell line and compare its in vitro behaviour to that of human GECs. MATERIAL AND METHODS Human primary GECs were immortalized with a bmi1/hTERT combination to prevent cell cycle triggers of senescence and telomere shortening. The resultant cell-line, telomerase immortalized gingival keratinocytes (TIGKs), were compared to GECs for cell morphology, karyotype, growth and cytokeratin expression, and further characterized for replicative lifespan, expression of toll-like receptors and invasion by P. gingivalis. RESULTS TIGKs showed morphologies, karyotype, proliferation rates and expression of characteristic cytokeratin proteins comparable to GECs. TIGKs underwent 36 passages without signs of senescence and expressed transcripts for toll-like receptors 1-6, 8 and 9. A subpopulation of cells underwent stratification after extended time in culture. The cytokeratin profiles of TIGK monolayers were consistent with basal cells. When allowed to stratify, cytokeratin profiles of TIGKs were consistent with suprabasal cells of the junctional epithelium. Further, TIGKs were comparable to GECs in previously reported levels and kinetics of invasion by wild-type P. gingivalis and an invasion defective ΔserB mutant. CONCLUSION Results confirm bmi1/hTERT immortalization of primary GECs generated a robust cell line with similar characteristics to the parental cell type. TIGKs represent a valuable model system for the study of oral bacteria interactions with host gingival cells.
Collapse
Affiliation(s)
- C E Moffatt-Jauregui
- Center for Oral Health and Systemic Disease, School of Dentistry, University of Louisville, Louisville, KY, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Lang ML, Zhu L, Kreth J. Keeping the bad bacteria in check: interactions of the host immune system with oral cavity biofilms. ACTA ACUST UNITED AC 2012. [DOI: 10.1111/j.1601-1546.2012.00278.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
42
|
Irshad M, van der Reijden WA, Crielaard W, Laine ML. In vitro invasion and survival of Porphyromonas gingivalis in gingival fibroblasts; role of the capsule. Arch Immunol Ther Exp (Warsz) 2012; 60:469-76. [PMID: 22949096 DOI: 10.1007/s00005-012-0196-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 07/23/2012] [Indexed: 11/28/2022]
Abstract
Porphyromonas gingivalis is a Gram-negative, anaerobic bacterium involved in periodontitis and peri-implantitis that can invade and survive inside host cells in vitro. P. gingivalis can invade human gingival fibroblasts (GF), but no data are available about the role of P. gingivalis' capsule in GF invasion. In the current study, we aimed to determine the ability of three strains of P. gingivalis (encapsulated wild type W83, non-encapsulated HG91 and the non-encapsulated insertional isogenic knockout mutant of W83, ΔEpsC) to invade GF and the ability of internalized P. gingivalis to survive in vitro antibiotic treatment. The ability of P. gingivalis strains to invade GF was tested using an antibiotic protection assay at multiplicity of infection (MOI) 100 and 1000. The survival of internalized P. gingivalis cells was further analyzed by subsequent in vitro treatment with either metronidazole or amoxicillin alone or a combination of metronidazole and amoxicillin and anaerobic culture viability counts. All strains of P. gingivalis used in this study were able to invade GFs. The non-encapsulated mutant of W83 (ΔEpsC mutant) was significantly more invasive than the wild type W83 at MOI 100 (p value 0.025) and MOI 1000 (p value 0.038). Furthermore, internalized P. gingivalis was able to resist in vitro antibiotic treatment. As demonstrated by the differences in invasion efficiencies of P. gingivalis strain W83 and its isogenic mutant ΔEpsC, the capsule of P. gingivalis makes it less efficient in invading gingival fibroblasts. Moreover, internalized P. gingivalis can survive antibiotic treatment in vitro.
Collapse
Affiliation(s)
- Muhammad Irshad
- Section of Preventive Dentistry, Department of Conservative and Preventive Dentistry, Academic Centre for Dentistry Amsterdam, ACTA, University of Amsterdam and Vrije University Amsterdam, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
43
|
Pflughoeft KJ, Versalovic J. Human microbiome in health and disease. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2011; 7:99-122. [PMID: 21910623 DOI: 10.1146/annurev-pathol-011811-132421] [Citation(s) in RCA: 325] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mammals are complex assemblages of mammalian and bacterial cells organized into functional organs, tissues, and cellular communities. Human biology can no longer concern itself only with human cells: Microbiomes at different body sites and functional metagenomics must be considered part of systems biology. The emergence of metagenomics has resulted in the generation of vast data sets of microbial genes and pathways present in different body habitats. The profound differences between microbiomes in various body sites reveal how metagenomes contribute to tissue and organ function. As next-generation DNA-sequencing methods provide whole-metagenome data in addition to gene-expression profiling, metaproteomics, and metabonomics, differences in microbial composition and function are being linked to health and disease states in different organs and tissues. Global parameters of microbial communities may provide valuable information regarding human health status and disease predisposition. More detailed knowledge of the human microbiome will yield next-generation diagnostics and therapeutics for various acute, chronic, localized, and systemic human diseases.
Collapse
Affiliation(s)
- Kathryn J Pflughoeft
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas 77030, USA.
| | | |
Collapse
|
44
|
Abstract
AIMS The goal of this review is to identify the antimicrobial proteins in the oral fluids, saliva and gingival crevicular fluid and identify functional families and candidates for antibacterial treatment. RESULTS Periodontal biofilms initiate a cascade of inflammatory and immune processes that lead to the destruction of gingival tissues and ultimately alveolar bone loss and tooth loss. Treatment of periodontal disease with conventional antibiotics does not appear to be effective in the absence of mechanical debridement. An alternative treatment may be found in antimicrobial peptides and proteins, which can be bactericidal and anti-inflammatory and block the inflammatory effects of bacterial toxins. The peptides have co-evolved with oral bacteria, which have not developed significant peptide resistance. Over 45 antibacterial proteins are found in human saliva and gingival crevicular fluid. The proteins and peptides belong to several different functional families and offer broad protection from invading microbes. Several antimicrobial peptides and proteins (AMPs) serve as templates for the development of therapeutic peptides and peptide mimetics, although to date none have demonstrated efficacy in human trials. CONCLUSIONS Existing and newly identified AMPs may be developed for therapeutic use in periodontal disease or can serve as templates for peptide and peptide mimetics with improved therapeutic indices.
Collapse
Affiliation(s)
- Sven-Ulrik Gorr
- Department of Diagnostic and Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, MN 55455, USA.
| | | |
Collapse
|
45
|
Abe D, Kubota T, Morozumi T, Shimizu T, Nakasone N, Itagaki M, Yoshie H. Altered gene expression in leukocyte transendothelial migration and cell communication pathways in periodontitis-affected gingival tissues. J Periodontal Res 2011; 46:345-53. [PMID: 21382035 DOI: 10.1111/j.1600-0765.2011.01349.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND AND OBJECTIVE Gene expression is related to the pathogenesis of periodontitis and plays a crucial role in local tissue destruction and disease susceptibility. The aims of the present study were to identify the expression of specific genes and biological pathways in periodontitis-affected gingival tissue using microarray and quantitative real-time RT-PCR analyses. MATERIAL AND METHODS Healthy and periodontitis-affected gingival tissues were taken from three patients with severe chronic periodontitis. Total RNAs from six gingival tissue samples were used for microarray analyses. Data-mining analyses, such as comparisons, gene ontology and pathway analyses, were performed and biological pathways with a significant role in periodontitis were identified. In addition, quantitative real-time RT-PCR analysis was performed on samples obtained from 14 patients with chronic periodontitis and from 14 healthy individuals in order to confirm the results of the pathway analysis. RESULTS Comparison analyses found 15 up-regulated and 13 down-regulated genes (all of which showed a change of more than twofold in expression levels) in periodontitis-affected gingival tissues. Pathway analysis identified 15 up-regulated biological pathways, including leukocyte transendothelial migration, and five down-regulated pathways, including cell communication. Quantitative real-time RT-PCR verified that five genes in the leukocyte transendothelial migration pathway were significantly up-regulated, and four genes in the cell communication pathway were significantly down-regulated, which was consistent with pathway analysis. CONCLUSION We identified up-regulated genes (ITGB-2, MMP-2, CXCL-12, CXCR-4 and Rac-2) and down-regulated genes (connexin, DSG-1, DSC-1 and nestin) in periodontitis-affected gingival tissues; these genes may be related to the stimulation of leukocyte transendothelial migration and to the the impairment of cell-to-cell communication in periodontitis.
Collapse
Affiliation(s)
- D Abe
- Division of Periodontology, Department of Oral Biological Science, Niigata University Graduate School of Medical and Dental Sciences, Japan
| | | | | | | | | | | | | |
Collapse
|
46
|
Porphyromonas gingivalis induction of microRNA-203 expression controls suppressor of cytokine signaling 3 in gingival epithelial cells. Infect Immun 2011; 79:2632-7. [PMID: 21536793 DOI: 10.1128/iai.00082-11] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Porphyromonas gingivalis is a pathogen in severe periodontal disease. Able to exploit an intracellular lifestyle within primary gingival epithelial cells (GECs), a reservoir of P. gingivalis can persist within the gingival epithelia. This process is facilitated by manipulation of the host cell signal transduction cascades which can impact cell cycle, cell death, and cytokine responses. Using microarrays, we investigated the ability of P. gingivalis 33277 to regulate microRNA (miRNA) expression in GECs. One of several miRNAs differentially regulated by GECs in the presence of P. gingivalis was miRNA-203 (miR-203), which was upregulated 4-fold compared to uninfected controls. Differential regulation of miR-203 was confirmed by quantitative reverse transcription-PCR (qRT-PCR). Putative targets of miR-203, suppressor of cytokine signaling 3 (SOCS3) and SOCS6, were evaluated by qRT-PCR. SOCS3 and SOCS6 mRNA levels were reduced >5-fold and >2-fold, respectively, in P. gingivalis-infected GECs compared to controls. Silencing of miR-203 using a small interfering RNA construct reversed the inhibition of SOCS3 expression. A dual luciferase assay confirmed binding of miR-203 to the putative target binding site of the SOCS3 3' untranslated region. Western blot analysis demonstrated that activation of signal transducer and activator of transcription 3 (Stat3), a downstream target of SOCS, was diminished following miR-203 silencing. This study shows that induction of miRNAs by P. gingivalis can modulate important host signaling responses.
Collapse
|
47
|
Damek-Poprawa M, Haris M, Volgina A, Korostoff J, DiRienzo JM. Cytolethal distending toxin damages the oral epithelium of gingival explants. J Dent Res 2011; 90:874-9. [PMID: 21471326 DOI: 10.1177/0022034511403743] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
UNLABELLED The cytolethal distending toxin (Cdt), expressed by the periodontal pathogen Aggregatibacter actinomycetemcomitans, inhibits the proliferation of cultured epithelial cells by arresting the cell cycle. The gingival epithelium is an early line of defense against microbial assault. When damaged, bacteria collectively gain entry into underlying connective tissue where microbial products can affect infiltrating inflammatory cells, leading to the destruction of the attachment apparatus. Histological evaluation of rat and healthy human gingival tissue exposed ex vivo to the Cdt for 36 and 18 hours, respectively, revealed extensive detachment of the keratinized outer layer and distention of spinous and basal cells in the oral epithelium. Treated human tissue also exhibited disruption of rete pegs and dissolution of cell junctions. Cells in the connective tissue appeared unaffected. Primary gingival epithelial cells, but not gingival fibroblasts, isolated from the same healthy human tissue were cell-cycle-arrested when treated with the toxin. These findings provide new evidence that the Cdt severely damages the oral epithelium, ex vivo, by specifically targeting epithelial cells, in situ. The Cdt shows preferential targeting of the epithelium as opposed to connective tissue in animal and human gingival explant models. ABBREVIATIONS cytolethal distending toxin (Cdt), connective tissue (CT), 4',6-diamidino-2-phenylindole (DAPI), human gingival epithelial cells (HGEC), human gingival explants (HGX), human gingival fibroblasts (HGF), junctional epithelium (JE), oral epithelium (OE), rete pegs (RP), sulcular epithelium (SE).
Collapse
Affiliation(s)
- M Damek-Poprawa
- Department of Microbiology, School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
48
|
Dickinson BC, Moffatt CE, Hagerty D, Whitmore SE, Brown TA, Graves DT, Lamont RJ. Interaction of oral bacteria with gingival epithelial cell multilayers. Mol Oral Microbiol 2011; 26:210-20. [PMID: 21545698 DOI: 10.1111/j.2041-1014.2011.00609.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Primary gingival epithelial cells were cultured in multilayers as a model for the study of interactions with oral bacteria associated with health and periodontal disease. Multilayers maintained at an air-liquid interface in low-calcium medium displayed differentiation and cytokeratin properties characteristic of junctional epithelium. Multilayers were infected with fluorescently labeled Porphyromonas gingivalis, Aggregatibacter actinomycetemcomitans, Fusobacterium nucleatum or Streptococcus gordonii, and bacterial association was determined by confocal microscopy and quantitative image analysis. Porphyromonas gingivalis invaded intracellularly and spread from cell to cell; A. actinomycetemcomitans and F. nucleatum remained extracellular and showed intercellular movement through the multilayer; whereas S. gordonii remained extracellular and predominantly associated with the superficial cell layer. None of the bacterial species disrupted barrier function as measured by transepithelial electrical resistance. P. gingivalis did not elicit secretion of proinflammatory cytokines. However, A. actinomycetemcomitans and S. gordonii induced interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), IL-6 and IL-8 secretion; and F. nucleatum stimulated production of IL-1β and TNF-α. Aggregatibacter actinomycetemcomitans, F. nucleatum and S. gordonii, but not P. gingivalis, increased levels of apoptosis after 24 h infection. The results indicate that the organisms with pathogenic potential were able to traverse the epithelium, whereas the commensal bacteria did not. In addition, distinct host responses characterized the interaction between the junctional epithelium and oral bacteria.
Collapse
Affiliation(s)
- B C Dickinson
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Growth of oral bacteria in situ requires adhesion to a surface because the constant flow of host secretions thwarts the ability of planktonic cells to grow before they are swallowed. Therefore, oral bacteria evolved to form biofilms on hard tooth surfaces and on soft epithelial tissues, which often contain multiple bacterial species. Because these biofilms are easy to study, they have become the paradigm of multispecies biofilms. In this Review we describe the factors involved in the formation of these biofilms, including the initial adherence to the oral tissues and teeth, cooperation between bacterial species in the biofilm, signalling between the bacteria and its role in pathogenesis, and the transfer of DNA between bacteria. In all these aspects distance between cells of different species is integral for oral biofilm growth.
Collapse
|
50
|
Kebschull M, Papapanou PN. Periodontal microbial complexes associated with specific cell and tissue responses. J Clin Periodontol 2011; 38 Suppl 11:17-27. [DOI: 10.1111/j.1600-051x.2010.01668.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|