1
|
Zheng H, Cai Y, Liu K, Xiang J, Han W, Wang Y, Chen R. Visualize the time dynamics and research trends of macrophage associated periodontitis research from 2004 to 2023: Bibliometrix analysis. Medicine (Baltimore) 2024; 103:e40450. [PMID: 39560581 PMCID: PMC11576026 DOI: 10.1097/md.0000000000040450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Macrophages play an important role in the symptoms and structural progression of periodontitis, and are receiving increasing attention. In recent years, research has shown significant progress in macrophage associated periodontitis. However, there is still lack of comprehensive and methodical bibliometric analysis in this domain. Therefore, this research aims to describe the state of the research and current research hotspots of macrophage associated periodontitis from the perspective of bibliometrics. METHODS This study collected and screened a total of 1424 articles on macrophage associated periodontitis retrieved between 2004 and 2023 from Web of Science Core Collection database. Use Citespace (6.1. R6), Bibliometrix-R (4.1.3), VOSviewer (1.6.19), and Graphpad Prism8 software to analyze and plot countries/regions, institutions, journals, authors, literature, and keywords to explore the research hotspots and development trends of macrophage associated periodontitis. RESULT After analysis, the amount of macrophage associated periodontitis publications has been rising consistently over time, with China having the most publications (29.32%). 3 countries accounted for 65.57% of the total publications: the United States, China, and Japan, occupying a dominant position in this research field. China publications have the fastest growth rate and played a driving role. The most productive institution is the Sichuan University in China. Journal of Periodontal Research is highly popular in the field of macrophage associated periodontitis, with the highest number of publications. Grenier, Daniel is the most prolific author. Inflammation and Bone Loss in Periodontal Disease are the most cited literature. "Biological pathogenic factors," "immune regulation," "mechanism research," "susceptibility factor research," "pathological processes and molecular correlation," "pathological characteristics," "inflammatory response" are the main keyword groups in this field. CONCLUSION This study systematically analyzes and describes the development process, direction, and hotspots of macrophage associated periodontitis using bibliometric methods, providing a reference for future researchers who continue to study macrophage associated periodontitis.
Collapse
Affiliation(s)
- Hu Zheng
- Key Laboratory of Oral Diseases Research of Anhui Province, College and Hospital of Stomatology, Anhui Medical University, Hefei, China
| | | | | | | | | | | | | |
Collapse
|
2
|
Tanum J, Kim H, Lee S, Kim A, Korostoff J, Hwang G. Photobiomodulation of Gingival Cells Challenged with Viable Oral Microbes. J Dent Res 2024; 103:745-754. [PMID: 38700089 PMCID: PMC11191660 DOI: 10.1177/00220345241246529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2024] Open
Abstract
The oral cavity, a unique ecosystem harboring diverse microorganisms, maintains health through a balanced microflora. Disruption may lead to disease, emphasizing the protective role of gingival epithelial cells (GECs) in preventing harm from pathogenic oral microbes. Shifting GECs' response from proinflammatory to antimicrobial could be a novel strategy for periodontitis. Photobiomodulation therapy (PBMT), a nonpharmacologic host modulatory approach, is considered an alternative to drugs. While the host cell response induced by a single type of pathogen-associated molecular patterns (PAMPs) was widely studied, this model does not address the cellular response to intact microbes that exhibit multiple PAMPs that might modulate the response. Inspired by this, we developed an in vitro model that simulates direct interactions between host cells and intact pathogens and evaluated the effect of PBMT on the response of human gingival keratinocytes (HGKs) to challenge viable oral microbes at both the cellular and molecular levels. Our data demonstrated that LED pretreatment on microbially challenged HGKs with specific continuous wavelengths (red: 615 nm; near-infrared: 880 nm) induced the production of various antimicrobial peptides, enhanced cell viability and proliferation, promoted reactive oxygen species scavenging, and down-modulated proinflammatory activity. The data also suggest a potential explanation regarding the superior efficacy of near-infrared light treatment compared with red light in enhancing antimicrobial activity and reducing cellular inflammation of HGKs. Taken together, the findings suggest that PBMT enhances the overall barrier function of gingival epithelium while minimizing inflammation-mediated breakdown of the underlying structures.
Collapse
Affiliation(s)
- J. Tanum
- Department of Preventive and Restorative Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - H.E. Kim
- Department of Preventive and Restorative Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - S.M. Lee
- Department of Endodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - A. Kim
- Department of Medical Engineering, College of Engineering and Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - J. Korostoff
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - G. Hwang
- Department of Preventive and Restorative Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Innovation & Precision Dentistry, School of Dental Medicine, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
- Chemical and Biomolecular Engineering College of Engineering, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
3
|
Luo S, Xu T, Zheng Q, Jiang A, Zhao J, Ying Y, Liu N, Pan Y, Zhang D. Mitochondria: An Emerging Unavoidable Link in the Pathogenesis of Periodontitis Caused by Porphyromonas gingivalis. Int J Mol Sci 2024; 25:737. [PMID: 38255811 PMCID: PMC10815845 DOI: 10.3390/ijms25020737] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/02/2024] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Porphyromonas gingivalis (P. gingivalis) is a key pathogen of periodontitis. Increasing evidence shows that P. gingivalis signals to mitochondria in periodontal cells, including gingival epithelial cells, gingival fibroblast cells, immune cells, etc. Mitochondrial dysfunction affects the cellular state and participates in periodontal inflammatory response through the aberrant release of mitochondrial contents. In the current review, it was summarized that P. gingivalis induced mitochondrial dysfunction by altering the mitochondrial metabolic state, unbalancing mitochondrial quality control, prompting mitochondrial reactive oxygen species (ROS) production, and regulating mitochondria-mediated apoptosis. This review outlines the impacts of P. gingivalis and its virulence factors on the mitochondrial function of periodontal cells and their role in periodontitis.
Collapse
Affiliation(s)
- Shiyin Luo
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang 110002, China; (S.L.); (T.X.); (Q.Z.); (A.J.); (J.Z.); (Y.Y.); (N.L.)
| | - Tong Xu
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang 110002, China; (S.L.); (T.X.); (Q.Z.); (A.J.); (J.Z.); (Y.Y.); (N.L.)
| | - Qifan Zheng
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang 110002, China; (S.L.); (T.X.); (Q.Z.); (A.J.); (J.Z.); (Y.Y.); (N.L.)
| | - Aijia Jiang
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang 110002, China; (S.L.); (T.X.); (Q.Z.); (A.J.); (J.Z.); (Y.Y.); (N.L.)
| | - Jiahui Zhao
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang 110002, China; (S.L.); (T.X.); (Q.Z.); (A.J.); (J.Z.); (Y.Y.); (N.L.)
| | - Yue Ying
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang 110002, China; (S.L.); (T.X.); (Q.Z.); (A.J.); (J.Z.); (Y.Y.); (N.L.)
| | - Nan Liu
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang 110002, China; (S.L.); (T.X.); (Q.Z.); (A.J.); (J.Z.); (Y.Y.); (N.L.)
| | - Yaping Pan
- Department of Periodontics and Oral Biology, School of Stomatology, China Medical University, Shenyang 110002, China;
| | - Dongmei Zhang
- Department of Periodontics and Oral Biology, School of Stomatology, China Medical University, Shenyang 110002, China;
| |
Collapse
|
4
|
Ming J, Zeng X, Zhou R. Portable biosensor-based oral pathogenic bacteria detection for community and family applications. Anal Bioanal Chem 2023:10.1007/s00216-023-04809-1. [PMID: 37389598 DOI: 10.1007/s00216-023-04809-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 07/01/2023]
Abstract
Detection of oral pathogens is essential in the management of oral diseases, as their occurrence and progression are closely linked to an imbalance in these microorganisms. Detection techniques such as microbial cultures, enzyme-linked immunosorbent assays and polymerase chain reactions are highly dependent on complex testing procedures and specialized laboratory equipment, making prevention and early diagnosis of oral diseases difficult. To comprehensively implement oral disease prevention and early diagnosis in social groups, there is an urgent need for portable testing methods for oral pathogenic bacteria that can be applied in community and home settings. In this review, several common portable biosensors for pathogenic bacteria are first described. Based on the goal of achieving primary prevention and diagnosis of oral diseases, we elaborate and summarize portable biosensors for common oral pathogenic bacteria in terms of how to achieve portability of the technique. This review aims to reflect the current status of portable biosensors for common oral pathogens and to lay the foundation for the further realization of portable detection of oral pathogens.
Collapse
Affiliation(s)
- Jieyu Ming
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xin Zeng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China.
| | - Ronghui Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China.
| |
Collapse
|
5
|
Liu M, Shao J, Zhao Y, Ma B, Ge S. Porphyromonas gingivalis Evades Immune Clearance by Regulating Lysosome Efflux. J Dent Res 2023; 102:555-564. [PMID: 36800907 DOI: 10.1177/00220345221146097] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2023] Open
Abstract
Porphyromonas gingivalis, a major periodontal pathogen, invades autophagosomes of cells, including gingival epithelial cells, endothelial cells, gingival fibroblasts, macrophages, and dendritic cells, to escape antimicrobial autophagy and lysosome fusion. However, it is not known how P. gingivalis resists autophagic immunity, survives within cells, and induces inflammation. Thus, we investigated whether P. gingivalis could escape antimicrobial autophagy by promoting lysosome efflux to block autophagic maturation, leading to intracellular survival, and whether the growth of P. gingivalis within cells results in cellular oxidative stress, causing mitochondrial damage and inflammatory responses. P. gingivalis invaded human immortalized oral epithelial cells in vitro and mouse oral epithelial cells of gingival tissues in vivo. The production of reactive oxygen species (ROS) increased upon bacterial invasion, as well as mitochondrial dysfunction-related parameters with downregulated mitochondrial membrane potential and intracellular adenosine triphosphate (ATP), upregulated mitochondrial membrane permeability, intracellular Ca2+ influx, mitochondrial DNA expression, and extracellular ATP. Lysosome excretion was elevated, the number of intracellular lysosomes was diminished, and lysosomal-associated membrane protein 2 was downregulated. Expression of autophagy-related proteins, microtubule-associated protein light chain 3, sequestosome-1, the NLRP3 inflammasome, and interleukin-1β increased with P. gingivalis infection. P. gingivalis may survive in vivo by promoting lysosome efflux, blocking autophagosome-lysosome fusion, and destroying autophagic flux. As a result, ROS and damaged mitochondria accumulated and activated the NLRP3 inflammasome, which recruited the adaptor protein ASC and caspase 1, leading to the production of proinflammatory factor interleukin-1β and inflammation.
Collapse
Affiliation(s)
- M Liu
- Department of Periodontology & Tissue Engineering and Regeneration, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - J Shao
- Department of Periodontology & Tissue Engineering and Regeneration, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Y Zhao
- Department of Periodontology & Tissue Engineering and Regeneration, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - B Ma
- Department of Periodontology & Tissue Engineering and Regeneration, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - S Ge
- Department of Periodontology & Tissue Engineering and Regeneration, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| |
Collapse
|
6
|
Yu YL, Ma JR, Li SN, Liao MQ, Xu S, Chen HE, Dai SH, Peng XL, Zhao D, Lou YM, Yu XX, Gao XP, Liu YH, Liu J, Ke XY, Ping Z, Wang L, Wang CY, Zeng FF. Association between Periodontitis and Aortic Calcification: A Cohort Study. Angiology 2023; 74:129-138. [PMID: 35503367 DOI: 10.1177/00033197221094713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The present study investigated the association between the presence of periodontitis and aortic calcification (AC) risk among Chinese adults. A total of 6059 individuals who underwent regular health check-ups and received a diagnosis of periodontitis between 2009 and 2016 were included. The outcome was AC, assessed by a chest low-dose spiral CT scan. Cox proportional hazards regression analysis was used to assess the association between periodontitis and AC risk after adjusting for several confounders. After a median follow-up period of 2.3 years (interquartile range: 1.03-4.97 years), 843 cases of AC were identified, with 532 (12.13%) and 311 (18.59%) patients in the non-periodontitis group and periodontitis group, respectively. Multivariate analyses demonstrated that, compared with those without periodontitis, the hazard ratio and 95% confidence interval for AC risk in participants with periodontitis was 1.18 (1.02-1.36) (P = .025) in the fully adjusted model. Stratified analyses showed that the positive relationship between periodontitis and AC was more evident in males and participants <65 years of age (pinteraction = .005 and .004, respectively). Our results show that the presence of periodontitis was positively associated with AC among Chinese adults, especially among males and younger participants.
Collapse
Affiliation(s)
- Ying-Lin Yu
- Department of Epidemiology, School of Medicine, 47885Jinan University, Guangdong, China
| | - Jun-Rong Ma
- Department of Epidemiology, School of Medicine, 47885Jinan University, Guangdong, China
| | - Shu-Na Li
- Department of Epidemiology, School of Medicine, 47885Jinan University, Guangdong, China
| | - Min-Qi Liao
- Department of Epidemiology, School of Medicine, 47885Jinan University, Guangdong, China
| | - Shan Xu
- Department of Non-communicable Disease Prevention and Control, 557960Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, China
| | - Hong-En Chen
- Department of Non-communicable Disease Prevention and Control, 557960Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, China
| | - Shu-Hong Dai
- Department of Non-communicable Disease Prevention and Control, 557960Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, China
| | - Xiao-Lin Peng
- Department of Non-communicable Disease Prevention and Control, 557960Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, China
| | - Dan Zhao
- Department of Non-communicable Disease Prevention and Control, 557960Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, China
| | - Yan-Mei Lou
- Department of Health Management, Beijing Xiao Tang Shan Hospital, Beijing, China
| | - Xiao-Xuan Yu
- Department of Epidemiology, School of Medicine, 47885Jinan University, Guangdong, China
| | - Xu-Ping Gao
- Department of Child and Adolescent Psychiatry, 74577Peking University Sixth Hospital (Institute of Mental Health), Beijing, China
| | - Yan-Hua Liu
- Department of Nutrition, 191599The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jun Liu
- Experimental Teaching Demonstration Center for Preventive Medicine of Guizhou Province, 66367Zunyi Medical University, Zunyi, China
| | - Xing-Yao Ke
- Department of Epidemiology, School of Medicine, 47885Jinan University, Guangdong, China
| | - Zhao Ping
- Department of Health Management, Beijing Xiao Tang Shan Hospital, Beijing, China
| | - Li Wang
- Department of Non-communicable Disease Prevention and Control, 557960Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, China
| | - Chang-Yi Wang
- Department of Non-communicable Disease Prevention and Control, 557960Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, China
| | - Fang-Fang Zeng
- Department of Epidemiology, School of Medicine, 47885Jinan University, Guangdong, China
| |
Collapse
|
7
|
Soto C, Rojas V, Yáñez L, Hidalgo A, Olivera M, Pacheco M, Venegas D, Salinas D, Bravo D, Quest AF. Porphyromonas gingivalis-Helicobacter pylori co-incubation enhances Porphyromonas gingivalis virulence and increases migration of infected human oral keratinocytes. J Oral Microbiol 2022; 14:2107691. [PMID: 35978839 PMCID: PMC9377229 DOI: 10.1080/20002297.2022.2107691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background Porphyromonas gingivalis is part of the subgingival biofilm and a keystone species in the development of periodontitis. Interactions between P.gingivalis and other bacteria in biofilms have been shown to affect bacterial virulence. Helicobacter pylori also inhabits the subgingival biofilm, but the consequences of interactions there with P.gingivalis remain unknown. Here, we investigated how the pre-incubation of P.gingivalis with H.pylori affects P.gingivalis virulence. Methods We assayed P.gingivalis internalization by oral keratinocytes (OKs), hemagglutination and biofilm formation to identify alterations in virulence after pre-incubation with H. pylori. Also, we evaluated viability and migration of OKs infected with P. gingivalis, as well as the role of toll-like receptor 4 (TLR4). In addition, we quantified the mRNA of genes associated with P.gingivalis virulence. Results Pre-incubation of P.gingivalis with H.pylori enhanced P.gingivalis biofilm formation, bacterial internalization into OKs and hemagglutination. Infection with pre-incubated P.gingivalis increased OK migration in a manner dependent on the O-antigen and linked to increased expression of the gingipain RgpB. Also, OK TLR4 participates in these events, because upon TLR4 knock-down, pre-incubated P.gingivalis no longer stimulated OK migration. Discussion We provide here for the first time insight to the consequences of direct interaction between P.gingivalis and H.pylori. In doing so, we shed light on the mechanism by which H. pylori presence in the oral cavity increases the severity or progression of periodontitis.
Collapse
Affiliation(s)
- Cristopher Soto
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Advanced Center for Chronic Diseases (Accdis), Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Oral Microbiology Laboratory, Department of Pathology and Oral Medicine, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Victoria Rojas
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Advanced Center for Chronic Diseases (Accdis), Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Oral Microbiology Laboratory, Department of Pathology and Oral Medicine, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Lucas Yáñez
- Oral Microbiology Laboratory, Department of Pathology and Oral Medicine, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Antonio Hidalgo
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Advanced Center for Chronic Diseases (Accdis), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Marcela Olivera
- Oral Microbiology Laboratory, Department of Pathology and Oral Medicine, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Martín Pacheco
- Oral Microbiology Laboratory, Department of Pathology and Oral Medicine, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Darna Venegas
- Oral Microbiology Laboratory, Department of Pathology and Oral Medicine, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Daniela Salinas
- Oral Microbiology Laboratory, Department of Pathology and Oral Medicine, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Denisse Bravo
- Oral Microbiology Laboratory, Department of Pathology and Oral Medicine, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Andrew F.G. Quest
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Advanced Center for Chronic Diseases (Accdis), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
8
|
Aleksijević LH, Aleksijević M, Škrlec I, Šram M, Šram M, Talapko J. Porphyromonas gingivalis Virulence Factors and Clinical Significance in Periodontal Disease and Coronary Artery Diseases. Pathogens 2022; 11:pathogens11101173. [PMID: 36297228 PMCID: PMC9609396 DOI: 10.3390/pathogens11101173] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/27/2022] [Accepted: 10/10/2022] [Indexed: 11/17/2022] Open
Abstract
Porphyromonas gingivalis is a gram-negative, anaerobic bacterium that lives in the oral cavity. It is an integral part of the oral microbiome, which includes more than 500 types of bacteria. Under certain circumstances, as a consequence of virulence factors, it can become very destructive and proliferate to many cells in periodontal lesions. It is one of the causative agents present extremely often in dental plaque and is the main etiological factor in the development of periodontal disease. During various therapeutic procedures, P. gingivalis can enter the blood and disseminate through it to distant organs. This primarily refers to the influence of periodontal agents on the development of subacute endocarditis and can facilitate the development of coronary heart disease, atherosclerosis, and ischemic infarction. The action of P. gingivalis is facilitated by numerous factors of virulence and pathogenicity such as fimbriae, hemolysin, hemagglutinin, capsules, outer membrane vesicles, lipopolysaccharides, and gingipains. A special problem is the possibility of biofilm formation. P. gingivalis in a biofilm is 500 to 1000 times less sensitive to antimicrobial drugs than planktonic cells, which represents a significant problem in the treatment of infections caused by this pathogen.
Collapse
Affiliation(s)
- Lorena Horvat Aleksijević
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Correspondence: (L.H.A.); (J.T.)
| | - Marko Aleksijević
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Ivana Škrlec
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Marko Šram
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Miroslav Šram
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Cardiology, Clinical Hospital Center Osijek, 31000 Osijek, Croatia
| | - Jasminka Talapko
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Correspondence: (L.H.A.); (J.T.)
| |
Collapse
|
9
|
Bai H, Yang J, Meng S, Liu C. Oral Microbiota-Driven Cell Migration in Carcinogenesis and Metastasis. Front Cell Infect Microbiol 2022; 12:864479. [PMID: 35573798 PMCID: PMC9103474 DOI: 10.3389/fcimb.2022.864479] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/04/2022] [Indexed: 12/20/2022] Open
Abstract
The oral cavity harbors approximately 1,000 microbial species, and both pathogenic and commensal strains are involved in the development of carcinogenesis by stimulating chronic inflammation, affecting cell proliferation, and inhibiting cell apoptosis. Moreover, some substances produced by oral bacteria can also act in a carcinogenic manner. The link between oral microbiota and chronic inflammation as well as cell proliferation has been well established. Recently, increasing evidence has indicated the association of the oral microbiota with cell migration, which is crucial in regulating devastating diseases such as cancer. For instance, increased cell migration induced the spread of highly malignant cancer cells. Due to advanced technologies, the mechanistic understanding of cell migration in carcinogenesis and cancer metastasis is undergoing rapid progress. Thus, this review addressed the complexities of cell migration in carcinogenesis and cancer metastasis. We also integrate recent findings on the molecular mechanisms by which the oral microbiota regulates cell migration, with emphasis on the effect of the oral microbiota on adhesion, polarization, and guidance. Finally, we also highlight critical techniques, such as intravital microscopy and superresolution microscopy, for studies in this field.
Collapse
Affiliation(s)
- Huimin Bai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China School & Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China School & Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shu Meng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China School & Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Shu Meng, ; Chengcheng Liu,
| | - Chengcheng Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China School & Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Shu Meng, ; Chengcheng Liu,
| |
Collapse
|
10
|
The Periodontopathic Pathogen, Porphyromonas gingivalis, Involves a Gut Inflammatory Response and Exacerbates Inflammatory Bowel Disease. Pathogens 2022; 11:pathogens11010084. [PMID: 35056032 PMCID: PMC8779656 DOI: 10.3390/pathogens11010084] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/07/2022] [Accepted: 01/08/2022] [Indexed: 12/13/2022] Open
Abstract
Periodontal disease (PD) is one of the most prevalent disorders globally and is strongly associated with many other diseases. Inflammatory bowel disease (IBD), an inflammatory condition of the colon and the small intestine, is reported to be associated with PD through undetermined mechanisms. We analyzed taxonomic assignment files from the Crohn’s Disease Viral and Microbial Metagenome Project (PRJEB3206). The abundance of Porphyromonadaceae in fecal samples was significantly different between patients with Crohn’s disease and control volunteers. Dextran sulfate sodium was used to induce colitis in mice to reveal the effect of this periodontopathic pathogen in vivo. After intrarectal implantation of Porphyromonas gingivalis (Pg)—the primary pathogen causing PD—the disease activity index score, colonic epithelial loss, and inflammatory cell infiltration were intensified. In addition, tumor necrosis factor-α and interleukin-6 showed the highest levels in Pg-infected colons. This revealed the importance of Pg in the exacerbation of IBD. Thus, simultaneous treatment of PD should be considered for people with IBD. Moreover, implantation of Pg in the rectum worsened the clinical symptoms of colitis in mice. Because Pg participates in the pathogenesis of IBD, reducing the chances of it entering the intestine might prevent the worsening of this disorder.
Collapse
|
11
|
Li M, Huang S, Zhang Y, Song Z, Fu H, Lin Z, Huang X. Regulation of the unfolded protein response transducer IRE1α by SERPINH1 aggravates periodontitis with diabetes mellitus via prolonged ER stress. Cell Signal 2022; 91:110241. [PMID: 34998932 DOI: 10.1016/j.cellsig.2022.110241] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 12/31/2021] [Accepted: 12/31/2021] [Indexed: 12/18/2022]
Abstract
The hyperglycemic microenvironment induced by diabetes mellitus aggravates the inflammatory response, in which the IRE1α signal transduction pathway of the unfolded protein response (UPR) participates. However, the mechanism by which hyperglycemia regulates the IRE1α signaling pathway and affects endoplasmic reticulum (ER) homeostasis in human gingival epithelium in periodontitis with diabetes mellitus remains unknown. Our current data provide evidence that diabetes mellitus causes a hyperinflammatory response in the gingival epithelium, which accelerates periodontal inflammation. Next, we assessed UPR-IRE1α signaling in periodontitis with diabetes mellitus by examining human clinical gingival epithelium samples from healthy subjects, subjects with periodontitis and subjects with periodontitis with diabetes mellitus and by in vitro challenge of human epithelial cells with a hyperglycemic microenvironment. The results showed that a hyperglycemic microenvironment inhibited the IRE1α/XBP1 axis, decreased the expression of a UPR target gene (GRP78), and ultimately impaired the UPR, causing ER stress to be prolonged or more severe in human gingival epithelium. Subsequently, RNA sequencing (RNA-seq) data was analyzed to investigate the expression of ER-related genes in human gingival epithelium. Experiments verified that the mechanism by which periodontitis is aggravated in individuals with diabetes mellitus may involve decreased SERPINH1 expression. Furthermore, experiments in SERPINH1-knockdown and SERPINH1-overexpression models established in vitro indicated that SERPINH1 might act as an activator of IRE1α, maintaining human gingival epithelium homeostasis and reducing proinflammatory cytokine expression by preventing prolonged ER stress induced by high-glucose conditions. In conclusion, regulation of the UPR transducer IRE1α by SERPINH1 alleviates periodontitis with diabetes mellitus by mitigating prolonged ER stress. This finding provides evidence for the further study of periodontitis with diabetes mellitus.
Collapse
Affiliation(s)
- Mengdi Li
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Shuheng Huang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Yong Zhang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Zhi Song
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Haijun Fu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Zhengmei Lin
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Xin Huang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
12
|
Singhi A, Sharma A, Nath J, Sharma S, Marri R, Ekka R. A comparative clinical study to assess the role of antibiotics in periodontal flap surgery. J Pharm Bioallied Sci 2022; 14:S841-S844. [PMID: 36110722 PMCID: PMC9469438 DOI: 10.4103/jpbs.jpbs_16_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/28/2022] [Accepted: 02/11/2022] [Indexed: 11/04/2022] Open
|
13
|
Knowles A, Campbell S, Cross N, Stafford P. Bacterial Manipulation of the Integrated Stress Response: A New Perspective on Infection. Front Microbiol 2021; 12:645161. [PMID: 33967983 PMCID: PMC8100032 DOI: 10.3389/fmicb.2021.645161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/16/2021] [Indexed: 11/13/2022] Open
Abstract
Host immune activation forms a vital line of defence against bacterial pathogenicity. However, just as hosts have evolved immune responses, bacteria have developed means to escape, hijack and subvert these responses to promote survival. In recent years, a highly conserved group of signalling cascades within the host, collectively termed the integrated stress response (ISR), have become increasingly implicated in immune activation during bacterial infection. Activation of the ISR leads to a complex web of cellular reprogramming, which ultimately results in the paradoxical outcomes of either cellular homeostasis or cell death. Therefore, any pathogen with means to manipulate this pathway could induce a range of cellular outcomes and benefit from favourable conditions for long-term survival and replication. This review aims to outline what is currently known about bacterial manipulation of the ISR and present key hypotheses highlighting areas for future research.
Collapse
Affiliation(s)
- Alex Knowles
- Biomolecular Sciences Research Centre, Department of Biosciences and Chemistry, Faculty of Health and Wellbeing, Sheffield Hallam University, Sheffield, United Kingdom
| | - Susan Campbell
- Biomolecular Sciences Research Centre, Department of Biosciences and Chemistry, Faculty of Health and Wellbeing, Sheffield Hallam University, Sheffield, United Kingdom
| | - Neil Cross
- Biomolecular Sciences Research Centre, Department of Biosciences and Chemistry, Faculty of Health and Wellbeing, Sheffield Hallam University, Sheffield, United Kingdom
| | - Prachi Stafford
- Biomolecular Sciences Research Centre, Department of Biosciences and Chemistry, Faculty of Health and Wellbeing, Sheffield Hallam University, Sheffield, United Kingdom
| |
Collapse
|
14
|
Rowińska I, Szyperska-Ślaska A, Zariczny P, Pasławski R, Kramkowski K, Kowalczyk P. The Influence of Diet on Oxidative Stress and Inflammation Induced by Bacterial Biofilms in the Human Oral Cavity. MATERIALS (BASEL, SWITZERLAND) 2021; 14:1444. [PMID: 33809616 PMCID: PMC8001659 DOI: 10.3390/ma14061444] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 12/12/2022]
Abstract
The article is a concise compendium of knowledge on the etiology of pathogenic microorganisms of all complexes causing oral diseases. The influence of particular components of the diet and the role of oxidative stress in periodontal diseases were described. The study investigated the bacteriostatic effect of the diet of adults in in vivo and in vitro tests on the formation of bacterial biofilms living in the subgingival plaque, causing diseases called periodontitis. If left untreated, periodontitis can damage the gums and alveolar bones. Anaerobic bacteria, called periopathogens or periodontopathogens, play a key role in the etiopathogenesis of periodontitis. The most important periopathogens of the oral microbiota are bacteria of all complexes, including the red complex. The obtained results suggest the possibility of using a specific diet in the prevention and treatment of periodontal diseases-already treated as a disease of civilization. The quoted article is an innovative compilation of knowledge on this subject and it can be a valuable source of knowledge for professional hygienists, dentists, peridontologists, dentistry students and anyone who cares about proper oral hygiene. The obtained results suggest the possibility of using this type of diet in the prophylaxis of the oral cavity in order to avoid periodontitis.
Collapse
Affiliation(s)
- Ilona Rowińska
- The Medical and Social Center for Vocational and Continuing Education in Toruń, St. Jana 1/3, 87-100 Toruń, Poland; (I.R.); (A.S.-Ś.)
| | - Adrianna Szyperska-Ślaska
- The Medical and Social Center for Vocational and Continuing Education in Toruń, St. Jana 1/3, 87-100 Toruń, Poland; (I.R.); (A.S.-Ś.)
| | - Piotr Zariczny
- Toruń City Hall, Business Support Center in Toruń, ul. Marii Konopnickiej 13, 87-100 Toruń, Poland;
| | - Robert Pasławski
- Veterinary Insitute, Nicolaus Copernicus University in Toruń, str. Gagarina 7, 87-100 Toruń, Poland;
| | - Karol Kramkowski
- Department of Physical Chemistry, Medical University of Bialystok, Kilińskiego 1str, 15-089 Bialystok, Poland;
| | - Paweł Kowalczyk
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110 Jabłonna, Poland
| |
Collapse
|
15
|
Miya C, Cueno ME, Suzuki R, Maruoka S, Gon Y, Kaneko T, Yonehara Y, Imai K. Porphyromonas gingivalis gingipains potentially affect MUC5AC gene expression and protein levels in respiratory epithelial cells. FEBS Open Bio 2020; 11:446-455. [PMID: 33332733 PMCID: PMC7876492 DOI: 10.1002/2211-5463.13066] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/02/2020] [Accepted: 12/15/2020] [Indexed: 01/08/2023] Open
Abstract
Porphyromonas gingivalis (Pg) is a periodontopathic pathogen that may affect MUC5AC‐related mucus hypersecretion along airway epithelial cells. Here, we attempted to establish whether Pg virulence factors (lipopolysaccharide, FimA fimbriae, gingipains) affect MUC5AC in immortalized and primary bronchial cells. We report that MUC5AC gene expression and protein levels are affected by Pg culture supernatant, but not by lipopolysaccharide or FimA fimbriae. Cells treated with either Pg single (Kgp or Rgp) or double (Kgp/Rgp) mutants had altered levels of MUC5AC gene expression and protein levels, and MUC5AC staining of double mutant‐treated mouse lung cells showed that MUC5AC protein levels were unaffected. Taken together, we propose that Pg gingipains may be the primary virulence factor that influences both MUC5AC gene expression and protein levels.
Collapse
Affiliation(s)
- Chihiro Miya
- Department of Oral and Maxillofacial Surgery II, Nihon University School of Dentistry, Tokyo, Japan.,Department of Microbiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Marni E Cueno
- Department of Microbiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Ryuta Suzuki
- Department of Oral and Maxillofacial Surgery II, Nihon University School of Dentistry, Tokyo, Japan.,Department of Microbiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Shuichiro Maruoka
- Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Yasuhiro Gon
- Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Tadayoshi Kaneko
- Department of Oral and Maxillofacial Surgery II, Nihon University School of Dentistry, Tokyo, Japan
| | - Yoshiyuki Yonehara
- Department of Oral and Maxillofacial Surgery II, Nihon University School of Dentistry, Tokyo, Japan
| | - Kenichi Imai
- Department of Microbiology, Nihon University School of Dentistry, Tokyo, Japan
| |
Collapse
|
16
|
Gürsoy UK, Fteita D, Bikker FJ, Grande MA, Nazmi K, Gürsoy M, Könönen E, Belstrøm D. Elevated Baseline Salivary Protease Activity May Predict the Steadiness of Gingival Inflammation During Periodontal Healing: A 12-Week Follow-Up Study on Adults. Pathogens 2020; 9:pathogens9090751. [PMID: 32942694 PMCID: PMC7558121 DOI: 10.3390/pathogens9090751] [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: 08/05/2020] [Revised: 09/08/2020] [Accepted: 09/11/2020] [Indexed: 12/26/2022] Open
Abstract
Aim was to profile salivary total protease, Porphyromonas gingivalis gingipain, and neutrophil elastase activities in relation to the resolution of periodontal inflammation, salivary macrophage-derived chemokine (MDC), and macrophage inflammatory protein-1α concentrations. Nonsurgical periodontal treatment was performed in 24 periodontitis patients in a prospective interventional study design. Periodontal clinical parameters were recorded, and stimulated saliva samples were collected at baseline and 2, 6, and 12 weeks after treatment. Salivary total protease and gingipain activities were determined using fluorogenic substrates, elastase activity by chromogenic substrates, and cytokine concentrations by Luminex immunoassay. For statistical analyses, generalized linear mixed models for repeated measures were used. Salivary total protease activity elevated, while gingival inflammation and plaque accumulation decreased 2 and 6 weeks after periodontal therapy. Salivary MDC concentration was elevated 12 weeks after periodontal treatment. Patients with elevated protease activities at baseline in comparison to patients with low baseline total protease activities, had higher levels of gingival inflammation before and after periodontal treatment. In conclusion, elevations in salivary total protease activity seem to be part of periodontal healing at its early phases. Higher levels of salivary total protease activities before periodontal treatment may predict the severity and steadiness of unresolved gingival inflammation.
Collapse
Affiliation(s)
- Ulvi Kahraman Gürsoy
- Department of Periodontology, Institute of Dentistry, University of Turku, 20520 Turku, Finland; (D.F.); (M.G.); (E.K.)
- Correspondence:
| | - Dareen Fteita
- Department of Periodontology, Institute of Dentistry, University of Turku, 20520 Turku, Finland; (D.F.); (M.G.); (E.K.)
| | - Floris J. Bikker
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam, Free University and University of Amsterdam, 1081LA Amsterdam, The Netherlands; (F.J.B.); (K.N.)
| | - Maria Anastasia Grande
- Section for Clinical Oral Microbiology, Periodontology, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; (M.A.G.); (D.B.)
| | - Kamran Nazmi
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam, Free University and University of Amsterdam, 1081LA Amsterdam, The Netherlands; (F.J.B.); (K.N.)
| | - Mervi Gürsoy
- Department of Periodontology, Institute of Dentistry, University of Turku, 20520 Turku, Finland; (D.F.); (M.G.); (E.K.)
| | - Eija Könönen
- Department of Periodontology, Institute of Dentistry, University of Turku, 20520 Turku, Finland; (D.F.); (M.G.); (E.K.)
| | - Daniel Belstrøm
- Section for Clinical Oral Microbiology, Periodontology, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; (M.A.G.); (D.B.)
| |
Collapse
|
17
|
Ben Lagha A, Yang Y, Trivedi HM, Masters JG, Grenier D. A Dual Zinc plus Arginine formulation attenuates the pathogenic properties of Porphyromonas gingivalis and protects gingival keratinocyte barrier function in an in vitro model. J Oral Microbiol 2020; 12:1798044. [PMID: 32944154 PMCID: PMC7482904 DOI: 10.1080/20002297.2020.1798044] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Background and objectives Porphyromonas gingivalis, a late colonizer of the periodontal biofilm, has been strongly associated with the chronic form of periodontitis. The aim of this study was to investigate the effects of a Dual Zinc plus Arginine formulation (aqueous solution and dentifrice) on the pathogenic properties of P. gingivalis and the barrier function of an in vitro gingival epithelium model. Results The Dual Zinc plus Arginine aqueous solution and dentifrice inhibited the hemolytic and proteolytic activities of P. gingivalis. The Dual Zinc plus Arginine aqueous solution and dentifrice enhanced the barrier function of an in vitro gingival epithelium model as determined by a time-dependent increase in transepithelial electrical resistance and decrease in paracellular permeability. This was associated with an increased immunolabeling of two important tight junction proteins: zonula occludens-1 and occludin. The deleterious effects of P. gingivalis on keratinocyte barrier function as well as the ability of the bacterium to translocate through a gingival epithelium model were attenuated in the presence of either Dual Zinc plus Arginine aqueous solution or dentifrice. Conclusions The Dual Zinc plus Arginine formulation may offer benefits for patients affected by periodontal disease through its ability to attenuate the pathogenic properties of P. gingivalis and promote keratinocyte barrier function.
Collapse
Affiliation(s)
- Amel Ben Lagha
- Oral Ecology Research Group, Faculty of Dentistry, Université Laval, Quebec City, QC, Canada
| | - Ying Yang
- Colgate-Palmolive Technology Center, Piscataway, NJ, USA
| | | | | | - Daniel Grenier
- Oral Ecology Research Group, Faculty of Dentistry, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
18
|
Aral K, Milward MR, Gupta D, Cooper PR. Effects of Porphyromonas gingivalis and Fusobacterium nucleatum on inflammasomes and their regulators in H400 cells. Mol Oral Microbiol 2020; 35:158-167. [PMID: 32516848 DOI: 10.1111/omi.12302] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Inflammasomes are multiprotein complexes that regulate immune processes in response to infections and tissue damage. They modulate Interleukin-1beta (IL-1β) expression, a major proinflammatory cytokine. The inflammasome/IL-1β pathway is involved in head and neck squamous cell carcinoma (HNSCC) progression and the periodontal pathogens Fusobacterium nucleatum (Fn) and Porphyromonas gingivalis (Pg) have been reported to cause chronic inflammation in HNSCC. The aim of this study was to characterise the role of these pathogens in regulating inflammasome activity and the IL-1β response in HNSCC in vitro. METHODS An HNSCC cell line (H400) was exposed to Fn and Pg individually or in combination for 24h, ± incubation for 30 min with 5 mM adenosine triphosphate (ATP). Transcript levels of inflammasomes, NLRP3 and AIM2; inflammasome-regulatory proteins, POP1, CARD16 and TRIM16; and inflammasome-component, ASC and caspase 1 and IL-1β, were assayed by RT-PCR. Expression of IL-1β was by immunocytochemistry and ELISA. RESULTS NLRP3 expression was significantly upregulated in response to Pg, Fn + Pg, Pg + ATP and Fn + Pg + ATP. AIM2 was significantly upregulated by Fn, Pg and Fn + Pg + ATP exposure. All conditions significantly upregulated IL-1β gene expression. POP1 expression was significantly downregulated by Pg or Fn exposure but not by Fn + Pg. Intracellular pro- and mature IL-1β were significantly higher following Fn and Pg + ATP exposure. CONCLUSION Pg alone increased IL-1β by upregulating AIM2, NLRP3 and downregulating POP1. Fn promoted IL-1β by increasing AIM2 and downregulating POP1. Pg + ATP with or without Fn upregulated NLRP3, IL-1β by downregulating POP1. Periodontal pathogens may contribute to HNSCC pathogenesis by increasing the IL-1β response due to inflammasome dysregulation.
Collapse
Affiliation(s)
- Kübra Aral
- Research Fellow, School of Dentistry, University of Birmingham, Birmingham, UK.,Republic of Turkey Ministry of Health, Ankara, Turkey
| | | | - Dhanak Gupta
- Research Fellow, School of Dentistry, University of Birmingham, Birmingham, UK
| | - Paul R Cooper
- School of Dentistry, University of Birmingham, Birmingham, UK.,Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| |
Collapse
|
19
|
Abdulkareem AA, Abdulbaqi HR, Milward MR. In Vitro Homeostasis of Rat Oral Epithelial Cell Cultures Following Withdrawal of Periodontal Pathogens. Braz Dent J 2020; 31:135-142. [PMID: 32556012 DOI: 10.1590/0103-6440202002561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 11/25/2019] [Indexed: 11/22/2022] Open
Abstract
Inflammation of periodontal tissues is the consequence of interaction between periodontal pathogens and immune system. This is associated with increased expression of inflammatory cytokines, which may exert destructive effect to the periodontal tissues when released over long period. The aim of this study was to chronologically track the homeostasis of oral keratinocytes following removal of periodontal pathogens. This was done by investigating expression of selected inflammatory markers and integrity of epithelial monolayers in vitro. Rat oral keratinocytes were stimulated with heat-killed Fusobacterium nucleatum and Porphyromonas gingivalis over 7-days then bacteria were washed away and epithelial cells re-cultured for 3-days. Expression of IL-1β, IL-6, and IL-8 was measured by ELISA while transcription of tissue inhibitor of metalloproteinase-1 (TIMP-1) and matrix metalloproteinase -8 (MMP-8) was measured by polymerase chain reaction before and after removal of bacteria. Integrity of epithelial sheet was investigated by using transepithelial electrical resistance. Data showed general downregulation of IL-1b, IL-6, and IL-8 associated with restoring transcription of TIMP-1 and MMP-8 to normal level following removal of bacteria from epithelial cultures. However, expression of IL-8 and MMP-8 remained significantly higher than unstimulated epithelial cells despite withdrawal of F. nucleatum and P. gingivalis respectively from oral keratinocytes cultures. In addition, integrity of epithelial barrier function remained compromised even after removal of P. gingivalis. Results suggest that even after three days following removal of periodontal pathogens, oral keratinocytes sustained persistent upregulation of certain inflammatory markers that could compromise integrity of epithelial barrier function.
Collapse
Affiliation(s)
- Ali A Abdulkareem
- Department of Periodontics, College of Dentistry, University of Baghdad, Bagdad, Iraq
| | - Hayder R Abdulbaqi
- Department of Periodontics, College of Dentistry, University of Baghdad, Bagdad, Iraq
| | - Michael R Milward
- Department of Periodontology, School of Dentistry, University of Birmingham, Birmingham, UK
| |
Collapse
|
20
|
Metronidazole-Treated Porphyromonas gingivalis Persisters Invade Human Gingival Epithelial Cells and Perturb Innate Responses. Antimicrob Agents Chemother 2020; 64:AAC.02529-19. [PMID: 32205352 DOI: 10.1128/aac.02529-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/01/2020] [Indexed: 11/20/2022] Open
Abstract
Periodontitis as a biofilm-associated inflammatory disease is highly prevalent worldwide. It severely affects oral health and yet closely links to systemic diseases like diabetes and cardiovascular disease. Porphyromonas gingivalis as a "keystone" periodontopathogen drives the shift of microbe-host symbiosis to dysbiosis and critically contributes to the pathogenesis of periodontitis. Persisters represent a tiny subset of biofilm-associated microbes highly tolerant to lethal treatment of antimicrobials, and, notably, metronidazole-tolerant P. gingivalis persisters have recently been identified by our group. This study further explored the interactive profiles of metronidazole-treated P. gingivalis persisters (M-PgPs) with human gingival epithelial cells (HGECs). P. gingivalis cells (ATCC 33277) at stationary phase were treated with a lethal dosage of metronidazole (100 μg/ml, 6 h) for generating M-PgPs. The interaction of M-PgPs with HGECs was assessed by microscopy, flow cytometry, cytokine profiling, and quantitative PCR (qPCR). We demonstrated that the overall morphology and ultracellular structure of M-PgPs remained unchanged. Importantly, M-PgPs maintained the capabilities to adhere to and invade HGECs. Moreover, M-PgPs significantly suppressed proinflammatory cytokine expression in HGECs at a level comparable to that seen with the untreated P. gingivalis cells, through the thermosensitive components. The present report reveals that P. gingivalis persisters induced by lethal treatment of antibiotics were able to maintain their capabilities to adhere to and invade human gingival epithelial cells and to perturb the innate host responses. Novel strategies and approaches need to be developed for tackling P. gingivalis and favorably modulating the dysregulated immunoinflammatory responses for oral/periodontal health and general well-being.
Collapse
|
21
|
Brown JL, Johnston W, Delaney C, Rajendran R, Butcher J, Khan S, Bradshaw D, Ramage G, Culshaw S. Biofilm-stimulated epithelium modulates the inflammatory responses in co-cultured immune cells. Sci Rep 2019; 9:15779. [PMID: 31673005 PMCID: PMC6823452 DOI: 10.1038/s41598-019-52115-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 10/14/2019] [Indexed: 12/13/2022] Open
Abstract
The gingival epithelium is a physical and immunological barrier to the microbiota of the oral cavity, which interact through soluble mediators with the immune cells that patrol the tissue at the gingival epithelium. We sought to develop a three-dimensional gingivae-biofilm interface model using a commercially available gingival epithelium to study the tissue inflammatory response to oral biofilms associated with “health”, “gingivitis” and “periodontitis”. These biofilms were developed by sequential addition of microorganisms to mimic the formation of supra- and sub-gingival plaque in vivo. Secondly, to mimic the interactions between gingival epithelium and immune cells in vivo, we integrated peripheral blood mononuclear cells and CD14+ monocytes into our three-dimensional model and were able to assess the inflammatory response in the immune cells cultured with and without gingival epithelium. We describe a differential inflammatory response in immune cells cultured with epithelial tissue, and more so following incubation with epithelium stimulated by “gingivitis-associated” biofilm. These results suggest that gingival epithelium-derived soluble mediators may control the inflammatory status of immune cells in vitro, and therefore targeting of the epithelial response may offer novel therapies. This multi-cellular interface model, both of microbial and host origin, offers a robust in vitro platform to investigate host-pathogens at the epithelial surface.
Collapse
Affiliation(s)
- Jason L Brown
- Institute of Biomedical and Environmental Health Research, School of Science and Sport, University of the West of Scotland, Paisley, PA1 2BE, UK.,Oral Sciences Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - William Johnston
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Chris Delaney
- Institute of Biomedical and Environmental Health Research, School of Science and Sport, University of the West of Scotland, Paisley, PA1 2BE, UK
| | - Ranjith Rajendran
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - John Butcher
- Institute of Biomedical and Environmental Health Research, School of Science and Sport, University of the West of Scotland, Paisley, PA1 2BE, UK.,Department of Life Sciences, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow, G4 0BA, UK
| | - Shaz Khan
- Oral Healthcare R&D, GlaxoSmithKline Consumer Healthcare, Weybridge, KT13 0DE, UK
| | - David Bradshaw
- Oral Healthcare R&D, GlaxoSmithKline Consumer Healthcare, Weybridge, KT13 0DE, UK
| | - Gordon Ramage
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK.
| | - Shauna Culshaw
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK.
| |
Collapse
|
22
|
Kaushik NK, Ghimire B, Li Y, Adhikari M, Veerana M, Kaushik N, Jha N, Adhikari B, Lee SJ, Masur K, von Woedtke T, Weltmann KD, Choi EH. Biological and medical applications of plasma-activated media, water and solutions. Biol Chem 2019; 400:39-62. [PMID: 30044757 DOI: 10.1515/hsz-2018-0226] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/11/2018] [Indexed: 01/28/2023]
Abstract
Non-thermal atmospheric pressure plasma has been proposed as a new tool for various biological and medical applications. Plasma in close proximity to cell culture media or water creates reactive oxygen and nitrogen species containing solutions known as plasma-activated media (PAM) or plasma-activated water (PAW) - the latter even displays acidification. These plasma-treated solutions remain stable for several days with respect to the storage temperature. Recently, PAM and PAW have been widely studied for many biomedical applications. Here, we reviewed promising reports demonstrating plasma-liquid interaction chemistry and the application of PAM or PAW as an anti-cancer, anti-metastatic, antimicrobial, regenerative medicine for blood coagulation and even as a dental treatment agent. We also discuss the role of PAM on cancer initiation cells (spheroids or cancer stem cells), on the epithelial mesenchymal transition (EMT), and when used for metastasis inhibition considering its anticancer effects. The roles of PAW in controlling plant disease, seed decontamination, seed germination and plant growth are also considered in this review. Finally, we emphasize the future prospects of PAM, PAW or plasma-activated solutions in biomedical applications with a discussion of the mechanisms and the stability and safety issues in relation to humans.
Collapse
Affiliation(s)
- Nagendra Kumar Kaushik
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Electrical and Biological Physics and Department of Plasma-Bio Display, Kwangwoon University, Seoul 01897, Republic of Korea
| | - Bhagirath Ghimire
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Electrical and Biological Physics and Department of Plasma-Bio Display, Kwangwoon University, Seoul 01897, Republic of Korea
| | - Ying Li
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Electrical and Biological Physics and Department of Plasma-Bio Display, Kwangwoon University, Seoul 01897, Republic of Korea
| | - Manish Adhikari
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Electrical and Biological Physics and Department of Plasma-Bio Display, Kwangwoon University, Seoul 01897, Republic of Korea
| | - Mayura Veerana
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Electrical and Biological Physics and Department of Plasma-Bio Display, Kwangwoon University, Seoul 01897, Republic of Korea
| | - Neha Kaushik
- Department of Life Science, Hanyang University, Seoul 04763, Republic of Korea
| | - Nayansi Jha
- Graduate School of Clinical Dentistry, Korea University, Seoul 02841, Republic of Korea
| | - Bhawana Adhikari
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Electrical and Biological Physics and Department of Plasma-Bio Display, Kwangwoon University, Seoul 01897, Republic of Korea
| | - Su-Jae Lee
- Department of Life Science, Hanyang University, Seoul 04763, Republic of Korea
| | - Kai Masur
- Leibniz Institute for Plasma Science and Technology, D-17489 Greifswald, Germany
| | - Thomas von Woedtke
- Leibniz Institute for Plasma Science and Technology, D-17489 Greifswald, Germany
| | | | - Eun Ha Choi
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Electrical and Biological Physics and Department of Plasma-Bio Display, Kwangwoon University, Seoul 01897, Republic of Korea
| |
Collapse
|
23
|
Palioto DB, Finoti LS, Kinane DF, Benakanakere M. Epigenetic and inflammatory events in experimental periodontitis following systemic microbial challenge. J Clin Periodontol 2019; 46:819-829. [PMID: 31131910 PMCID: PMC6641985 DOI: 10.1111/jcpe.13151] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/10/2019] [Accepted: 05/20/2019] [Indexed: 02/06/2023]
Abstract
AIM The purpose of this study was to determine inflammatory and epigenetic features following induction of oral and gut dysbiosis in experimental periodontitis in order to examine the interplay between oral and systemic infection. MATERIALS AND METHODS Periodontitis was induced in 6- to 8-week-old C57BL/6 mice by (a) Ligature placement (Lig group) (oral challenge); (b) P. gingivalis gavage (Pg group) (systemic challenge); and (c) the combination of the two models oral and systemic challenge (Pg + Lig). The duration of the experiment was 60 days, and the animals were then sacrificed for analyses. Alveolar bone loss was assessed, and a multiplex immunoassay was performed. Maxillae and gut tissues were immunostained for DNMT3b (de novo methylation marker), B and T lymphocyte attenuator (BTLA) and IL-18R1 (inflammation markers). RESULTS Pg and Pg + Lig groups exhibited higher bone loss when compared to Sham. BAFF, VEGF, RANKL, RANTES and IP-10 were significantly higher with Pg gavage. Likewise, DNMT3b was overexpressed in both gut and maxilla after the Pg administration. The same pattern was observed for BTLA and IL-18R1 in gut tissues. CONCLUSIONS The systemic microbial challenge either alone or in combination with local challenge leads to distinct patterns of inflammatory and epigenetic features when compared to simply locally induced experimental periodontitis.
Collapse
Affiliation(s)
- Daniela B. Palioto
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of OMS and Periodontology, University of São Paulo - School of Dentistry of Ribeirão Preto. Brazil
| | - Livia S. Finoti
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Denis F. Kinane
- Division of Periodontology, School of Dental Medicine, University of Geneva, Switzerland
| | - Manjunatha Benakanakere
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
24
|
Esteban‐Fernández A, Ferrer MD, Zorraquín‐Peña I, López‐López A, Moreno‐Arribas MV, Mira A. In vitro beneficial effects of
Streptococcus dentisani
as potential oral probiotic for periodontal diseases. J Periodontol 2019; 90:1346-1355. [DOI: 10.1002/jper.18-0751] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 03/20/2019] [Accepted: 04/17/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Adelaida Esteban‐Fernández
- Department of Biotechnology and MicrobiologyInstitute of Food Science Research (CIAL)CSIC‐UAM Madrid Spain
| | - Maria D. Ferrer
- Department of Health and GenomicsCenter for Advanced Research in Public HealthFISABIO Foundation Valencia Spain
| | - Irene Zorraquín‐Peña
- Department of Biotechnology and MicrobiologyInstitute of Food Science Research (CIAL)CSIC‐UAM Madrid Spain
| | - Arantxa López‐López
- Department of Health and GenomicsCenter for Advanced Research in Public HealthFISABIO Foundation Valencia Spain
| | - M. Victoria Moreno‐Arribas
- Department of Biotechnology and MicrobiologyInstitute of Food Science Research (CIAL)CSIC‐UAM Madrid Spain
| | - Alex Mira
- Department of Health and GenomicsCenter for Advanced Research in Public HealthFISABIO Foundation Valencia Spain
| |
Collapse
|
25
|
Castro AB, Herrero ER, Slomka V, Pinto N, Teughels W, Quirynen M. Antimicrobial capacity of Leucocyte-and Platelet Rich Fibrin against periodontal pathogens. Sci Rep 2019; 9:8188. [PMID: 31160643 PMCID: PMC6547693 DOI: 10.1038/s41598-019-44755-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 05/22/2019] [Indexed: 01/05/2023] Open
Abstract
Various studies have described the biological properties of the Leucocyte- and Platelet Rich Fibrin (L-PRF) such as the antimicrobial effect against wound bacteria, but less is known about the effect against periodontal pathogens. The aim of this study was to evaluate the antibacterial properties of the L-PRF membrane and L-PRF exudate against the main periopathogens cultured on agar plates and in planktonic solution. This study demonstrated the antibacterial effect of the L-PRF membrane against P. intermedia, F. nucleatum, and A. actinomycetemcomitans, but especially against P. gingivalis. The L-PRF exudate also showed a strong inhibition against P. gingivalis on agar plates. No inhibition could be observed for the other bacterial strains. Moreover, L-PRF exudate decreased the number of viable P.gingivalis in a planktonic solution in a dose-dependent way. However, A. actinomycetemcomitans showed an increased growth in planktonic solution when in contact with the L-PRF exudate.
Collapse
Affiliation(s)
- Ana B Castro
- Department of Oral Health Sciences, Periodontology, KU Leuven & Dentistry, University Hospitals Leuven, Leuven, Belgium.
| | - Esteban R Herrero
- Department of Oral Health Sciences, Periodontology, KU Leuven & Dentistry, University Hospitals Leuven, Leuven, Belgium
| | - Vera Slomka
- Department of Oral Health Sciences, Periodontology, KU Leuven & Dentistry, University Hospitals Leuven, Leuven, Belgium
| | - Nelson Pinto
- Faculty of Dentistry, Postgraduate Implant Program, University of the Andes, Santiago, Chile
| | - Wim Teughels
- Department of Oral Health Sciences, Periodontology, KU Leuven & Dentistry, University Hospitals Leuven, Leuven, Belgium
| | - Marc Quirynen
- Department of Oral Health Sciences, Periodontology, KU Leuven & Dentistry, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
26
|
Sánchez MC, Romero-Lastra P, Ribeiro-Vidal H, Llama-Palacios A, Figuero E, Herrera D, Sanz M. Comparative gene expression analysis of planktonic Porphyromonas gingivalis ATCC 33277 in the presence of a growing biofilm versus planktonic cells. BMC Microbiol 2019; 19:58. [PMID: 30866810 PMCID: PMC6417203 DOI: 10.1186/s12866-019-1423-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 02/19/2019] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Porphyromonas gingivalis, a microorganism residing in the oral cavity within complex multispecies biofilms, is one of the keystone pathogens in the onset and progression of periodontitis. In this in vitro study, using DNA microarray, we investigate the differential gene expression of Porphyromonas gingivalis ATCC 33277 when growing in the presence or in absence of its own monospecies biofilm. RESULTS Approximately 1.5% of genes (28 out of 1909 genes, at 1.5 fold change or more, p-value < 0.05) were differentially expressed by P. gingivalis cells when in the presence of a biofilm. These genes were predominantly related to the metabolism of iron, bacterial adhesion, invasion, virulence and quorum-sensing system. The results from microarrays were consistent with those obtained by RT-qPCR. CONCLUSION This study provides insight on the transcriptional changes of planktonic P. gingivalis cells when growing in the presence of a biofilm. The resulting phenotypes provide information on changes occurring in the gene expression of this pathogen.
Collapse
Affiliation(s)
- María C. Sánchez
- Laboratory of Dental Research, University Complutense, Madrid, Spain
- Etiology and Therapy of Periodontal Diseases (ETEP) Research Group, University Complutense, Madrid, Spain
| | | | - Honorato Ribeiro-Vidal
- Laboratory of Dental Research, University Complutense, Madrid, Spain
- Etiology and Therapy of Periodontal Diseases (ETEP) Research Group, University Complutense, Madrid, Spain
| | - Arancha Llama-Palacios
- Laboratory of Dental Research, University Complutense, Madrid, Spain
- Etiology and Therapy of Periodontal Diseases (ETEP) Research Group, University Complutense, Madrid, Spain
| | - Elena Figuero
- Etiology and Therapy of Periodontal Diseases (ETEP) Research Group, University Complutense, Madrid, Spain
| | - David Herrera
- Etiology and Therapy of Periodontal Diseases (ETEP) Research Group, University Complutense, Madrid, Spain
| | - Mariano Sanz
- Etiology and Therapy of Periodontal Diseases (ETEP) Research Group, University Complutense, Madrid, Spain
- Department of Dental Clinical Specialities (DDCS), Faculty of Odontology, Plaza Ramón y Cajal s/n Ciudad Universitaria, 28040 Madrid, Spain
| |
Collapse
|
27
|
Ebersole JL, Peyyala R, Gonzalez OA. Biofilm-induced profiles of immune response gene expression by oral epithelial cells. Mol Oral Microbiol 2019; 34. [PMID: 30407731 DOI: 10.1111/omi.12251] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2018] [Indexed: 12/12/2022]
Abstract
This study examined the oral epithelial immunotranscriptome response patterns modulated by oral bacterial planktonic or biofilm challenge. We assessed gene expression patterns when epithelial cells were challenged with a multispecies biofilm composed of Streptococcus gordonii, Fusobacterium nucleatum, and Porphyromonas gingivalis representing a type of periodontopathic biofilm compared to challenge with the same species of planktonic bacteria. Of the 579 human immunology genes, a substantial signal of the epithelial cells was observed to 181 genes. Biofilm challenged stimulated significant elevations compared to planktonic bacteria for IL32, IL8, CD44, B2M, TGFBI, NFKBIA, IL1B, CD59, IL1A, CCL20 representing the top 10 signals comprising 55% of the overall signal for the epithelial cell responses. Levels of PLAU, CD9, IFITM1, PLAUR, CD24, TNFSF10, and IL1RN were all elevated by each of the planktonic bacterial challenge vs the biofilm responses. While the biofilms up-regulated 123/579 genes (>2-fold), fewer genes were increased by the planktonic species (36 [S gordonii], 30 [F nucleatum], 44 [P gingivalis]). A wide array of immune genes were regulated by oral bacterial challenge of epithelial cells that would be linked to the local activity of innate and adaptive immune response components in the gingival tissues. Incorporating bacterial species into a structured biofilm dramatically altered the number and level of genes expressed. Additionally, a specific set of genes were significantly decreased with the multispecies biofilms suggesting that some epithelial cell biologic pathways are down-regulated when in contact with this type of pathogenic biofilm.
Collapse
Affiliation(s)
- Jeffrey L Ebersole
- Department of Biomedical Sciences, School of Dental Medicine, University of Nevada Las Vegas, Las Vegas, Nevada.,College of Dentistry, Center for Oral Health Research, University of Kentucky, Lexington, Kentucky
| | - Rebecca Peyyala
- College of Dentistry, Center for Oral Health Research, University of Kentucky, Lexington, Kentucky
| | - Octavio A Gonzalez
- College of Dentistry, Center for Oral Health Research, University of Kentucky, Lexington, Kentucky.,Division of Periodontology, College of Dentistry, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
28
|
Jiang M, Li Z, Zhu G. The role of autophagy in the pathogenesis of periodontal disease. Oral Dis 2019; 26:259-269. [PMID: 30674085 DOI: 10.1111/odi.13045] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 01/13/2019] [Accepted: 01/16/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Ming Jiang
- Department of Stomatology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology Wuhan China
| | - Zhuoneng Li
- Centers for Disease Control and Prevention of Wuhan Wuhan China
| | - Guangxun Zhu
- Department of Stomatology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology Wuhan China
| |
Collapse
|
29
|
Xiao S, Wang H, Liang K, Tay F, Weir MD, Melo MAS, Wang L, Wu Y, Oates TW, Ding Y, Xu HHK. Novel multifunctional nanocomposite for root caries restorations to inhibit periodontitis-related pathogens. J Dent 2018; 81:17-26. [PMID: 30552930 DOI: 10.1016/j.jdent.2018.12.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/02/2018] [Accepted: 12/08/2018] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES The objectives of this study were to: (1) develop a novel multifunctional composite with nanoparticles of silver (NAg), 2-methacryloyloxyethyl phosphorylcholine (MPC), dimethylaminohexadecyl methacrylate (DMAHDM) and nanoparticles of amorphous calcium phosphate (NACP); and (2) investigate biofilm-inhibition via the multifunctional nanocomposite against three species of periodontal pathogens for the first time. METHODS The multifunctional nanocomposite was fabricated by incorporating NAg, MPC, DMAHDM and NACP into the resin consisting of pyromellitic glycerol dimethacrylate (PMDGDM) and ethoxylated bisphenol A dimethacrylate (EBPADMA). Three species (Porphyromonas gingivalis, Aggregatibacter actinomycetemcomitans and Fusobacterium nucleatum) were tested for metabolic activity (MTT), live/dead staining, polysaccharide production and colony-forming units (CFU) of biofilms grown on resins. RESULTS Incorporation of 0.08% to 0.12% NAg, 3% MPC, 3% DMAHDM and 30% NACP did not compromise the mechanical properties of the composite (p > 0.1). The multifunctional nanocomposite reduced protein adsorption to nearly 1/10 of that of a commercial control (p < 0.05). For all three species, the biofilm CFU was reduced by about 5 and 1 orders of magnitude via the nanocomposite containing NAg + MPC + DMAHDM, compared to commercial control and the composite with MPC + DMAHDM, respectively. CONCLUSIONS The novel multifunctional nanocomposite achieved the greatest reduction in metabolic activity, polysaccharide and biofilm growth of three periodontal pathogens. CLINICAL SIGNIFICANCE The strongly-antibacterial, multifunctional composite is promising for treating root lesions, alleviating periodontitis and protecting the periodontal tissues.
Collapse
Affiliation(s)
- Shimeng Xiao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Dept. of Periodontology, West China Hospital of Stomatology, Sichuan University, China; Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, MD, 21201, USA
| | - Haohao Wang
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, MD, 21201, USA; State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Dept. of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, China
| | - Kunneng Liang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Dept. of Periodontology, West China Hospital of Stomatology, Sichuan University, China; Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, MD, 21201, USA.
| | - Franklin Tay
- Department of Endodontics, Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Michael D Weir
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, MD, 21201, USA
| | - Mary Anne S Melo
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, MD, 21201, USA
| | - Lin Wang
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, MD, 21201, USA; VIP Integrated Department, School and Hospital of Stomatology, Jilin University, Changchun, 130011, China
| | - Yafei Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Dept. of Periodontology, West China Hospital of Stomatology, Sichuan University, China
| | - Thomas W Oates
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, MD, 21201, USA
| | - Yi Ding
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Dept. of Periodontology, West China Hospital of Stomatology, Sichuan University, China.
| | - Hockin H K Xu
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, MD, 21201, USA; Center for Stem Cell Biology & Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA; Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
30
|
Lv K, Wang G, Shen C, Zhang X, Yao H. Role and mechanism of the nod-like receptor family pyrin domain-containing 3 inflammasome in oral disease. Arch Oral Biol 2018; 97:1-11. [PMID: 30315987 DOI: 10.1016/j.archoralbio.2018.10.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 10/02/2018] [Accepted: 10/03/2018] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To summarize evidence and data from experimental studies regarding the role and mechanism of the Nod-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome in the pathogenesis of several representative oral diseases. MATERIALS AND METHODS A literature search of PubMed and EBSCO was performed. The literature was searched using a combination of keywords, e.g., NLRP3 inflammasome, inflammation, microorganisms, oral inflammatory diseases, and oral immunological diseases. RESULTS The initiation and activation of the NLRP3 inflammasome are associated with the pathogenesis and progression of several representative oral diseases, including periodontitis, oral lichen planus, dental pulp disease, and oral cavity squamous cell carcinoma. CONCLUSIONS The NLRP3 inflammasome plays a crucial role in the progression of inflammatory and adaptive immune responses. The possible role of the NLRP3 inflammasome in several oral diseases, including not only periodontitis and pulpitis but also mucosal diseases and oral cavity squamous cell carcinoma, may involve the aberrant regulation of inflammatory and immune responses. Understanding the cellular and molecular biology of the NLRP3 inflammasome is necessary because the NLRP3 inflammasome may be a potential therapeutic target for the treatment and prevention of oral inflammatory and immunological diseases.
Collapse
Affiliation(s)
- Kejia Lv
- Department of Stomatology, First Affiliated Hospital, College of Medicine, Zhejiang University, China
| | - Guohua Wang
- Department of Stomatology, First Affiliated Hospital, College of Medicine, Zhejiang University, China
| | - Chenlu Shen
- Department of Stomatology, First Affiliated Hospital, College of Medicine, Zhejiang University, China
| | - Xia Zhang
- Department of Stomatology, Affiliated Yinzhou People Hospital, College of Medicine, Ningbo University, China
| | - Hua Yao
- Department of Stomatology, First Affiliated Hospital, College of Medicine, Zhejiang University, China.
| |
Collapse
|
31
|
Peyyala R, Emecen-Huja P, Ebersole JL. Environmental lead effects on gene expression in oral epithelial cells. J Periodontal Res 2018; 53:961-971. [PMID: 30152021 DOI: 10.1111/jre.12594] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 06/20/2018] [Accepted: 07/04/2018] [Indexed: 01/02/2023]
Abstract
BACKGROUND AND OBJECTIVE Host responses in periodontitis span a range of local and emigrating cell types and biomolecules. Accumulating evidence regarding the expression of this disease across the population suggests some component of genetic variation that controls onset and severity of disease, in concert with the qualitative and quantitative parameters of the oral microbiome at sites of disease. However, there remains little information regarding the capacity of accruing environmental stressors or modifiers over a lifespan at both the host genetic and microbial ecology levels to understand fully the population variation in disease. This study evaluated the impact of environmental lead exposure on the responses of oral epithelial cells to challenge with a model pathogenic oral biofilm. METHODS AND RESULTS Using NanoString technology to quantify gene expression profiles of an array of 511 host response-associated genes in the epithelial cells, we identified an interesting primary panel of basal responses of the cells with numerous genes not previously considered as major response markers for epithelial cells, eg, interleukin (IL)-32, CTNNB1, CD59, MIF, CD44 and CD99. Even high levels of environment lead had little effect on these constitutive responses. Challenge of the cells with the biofilms (Streptococcus gordonii/Fusobacterium nucleatum/Porphyromonas gingivalis) resulted in significant increases in an array of host immune-related genes (134 of 511). The greatest magnitude in differential expression was observed with many genes not previously described as major response genes in epithelial cells, including IL-32, CD44, NFKBIA, CTSC, TNFAIP3, IL-1A, IL-1B, IL-8 and CCL20. The effects of environmental lead on responses to the biofilms were mixed, although levels of IL-8, CCL20 and CD70 were significantly decreased at lead concentrations of 1 and/or 5 μmol/L. CONCLUSION The results provided new information on a portfolio of genes expressed by oral epithelial cells, targeted substantial increases in an array of immune-related genes post-biofilm challenge, and a focused impact of environmental lead on these induced responses.
Collapse
Affiliation(s)
- Rebecca Peyyala
- Center for Oral Health Research and Division of Periodontology, College of Dentistry, University of Kentucky, Lexington, Kentucky
| | - Pinar Emecen-Huja
- Center for Oral Health Research and Division of Periodontology, College of Dentistry, University of Kentucky, Lexington, Kentucky
| | - Jeffrey L Ebersole
- Center for Oral Health Research and Division of Periodontology, College of Dentistry, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
32
|
An early report: a modified porphyrin-linked metronidazole targeting intracellular Porphyromonas gingivalis in cultured oral epithelial cells. Int J Oral Sci 2018; 9:167-173. [PMID: 28960193 PMCID: PMC5709547 DOI: 10.1038/ijos.2017.31] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/03/2017] [Indexed: 11/08/2022] Open
Abstract
Porphyromonas gingivalis (P. gingivalis) has a strong association with the pathogenesis of periodontal disease. Recurrence of periodontal disease following therapy is attributed to numerous factors, and of growing interest is the potential problem of intracellular bacteria that are able to persist and multiply within the host cell, thereby facilitating relapse of infection. The effect of antibiotic therapy in controlling P. gingivalis is questionable. Accordingly, while metronidazole is very effective against anaerobic extracellular P. gingivalis by disrupting the DNA of anaerobic microbial cells, this antibiotic does not effectively penetrate into mammalian cells to inhibit intracellular bacteria. Therefore in the present study, a modified porphyrin-linked metronidazole adducts, developed in our laboratory, was used to kill intracellular P. gingivalis. A series of experiments were performed, including cytotoxicity assays and cellular uptake of adducts by flow cytometry coupled with live cell imaging analysis, P. gingivalis invasion and elimination assays, and the analysis of colocalization of P. gingivalis and porphyrin-linked metronidazole by confocal laser scanning microscopy. Findings indicated that P. gingivalis and porphyrin-linked metronidazole were colocalized in the cytoplasm, and this compound was able to kill P. gingivalis intracellular with a sufficient culture time. This is a novel antimicrobial approach in the elimination of P. gingivalis from the oral cavity.
Collapse
|
33
|
Lagha AB, Groeger S, Meyle J, Grenier D. Green tea polyphenols enhance gingival keratinocyte integrity and protect against invasion by Porphyromonas gingivalis. Pathog Dis 2018; 76:4961135. [DOI: 10.1093/femspd/fty030] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 04/03/2018] [Indexed: 12/15/2022] Open
Affiliation(s)
- Amel Ben Lagha
- Oral Ecology Research Group, Faculty of Dentistry, Université Laval, 2420 rue de la Terrasse, Quebec City, QC G1V 0A6, Canada
| | - Sabine Groeger
- Department of Periodontology, Justus-Liebig-University of Giessen, Schlangenzahl 14, Giessen 35392, Germany
| | - Joerg Meyle
- Department of Periodontology, Justus-Liebig-University of Giessen, Schlangenzahl 14, Giessen 35392, Germany
| | - Daniel Grenier
- Oral Ecology Research Group, Faculty of Dentistry, Université Laval, 2420 rue de la Terrasse, Quebec City, QC G1V 0A6, Canada
| |
Collapse
|
34
|
Li Y, Pan J, Ye G, Zhang Q, Wang J, Zhang J, Fang J. In vitro studies of the antimicrobial effect of non-thermal plasma-activated water as a novel mouthwash. Eur J Oral Sci 2017; 125:463-470. [PMID: 29024061 DOI: 10.1111/eos.12374] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The aim of this study was to evaluate the antimicrobial effects of non-thermal plasma-activated water (PAW) as a novel mouthwash in vitro. Three representative oral pathogens - Streptococcus mutans, Actinomyces viscosus and Porphyromonas gingivalis - were treated with PAW. The inactivation effect was evaluated using the colony-forming unit (CFU) method, and the morphological and structural changes of a cell were observed by scanning electron microscopy and transmission electron microscopy (TEM). The physicochemical properties of PAW were analysed, and its influence on the leakage of intracellular proteins and DNA was evaluated. The results showed significant reduction of Streptococcus mutans within 60 s, of Actinomyces viscosus within 40 s, and of Porphyromonas gingivalis in less than 40 s. Scanning electron microscopy and TEM images showed that the normal cell morphology changed by varying degrees after treatment with PAW. Intracellular proteins (280 nm) and DNA (260 nm) leaked from all three species of bacteria after treatment with PAW. Reactive oxygen species (ROS), especially atomic oxygen (O), hydroxyl radical (˙OH), and hydrogen peroxide (H2 O2 ), were generated and led to strong oxidative stress and cell damage. These results suggest that PAW has potential use as a novel antimicrobial mouthwash.
Collapse
Affiliation(s)
- Yinglong Li
- Department of General Dentistry, Peking University School and Hospital of Stomatology, Beijing, China
| | - Jie Pan
- Department of General Dentistry, Peking University School and Hospital of Stomatology, Beijing, China
| | - Guopin Ye
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.,School of Stomatology, Lan Zhou University, Gansu, China
| | - Qian Zhang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Jing Wang
- School of Stomatology, Lan Zhou University, Gansu, China
| | - Jue Zhang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.,College of Engineering, Peking University, Beijing, China
| | - Jing Fang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.,College of Engineering, Peking University, Beijing, China
| |
Collapse
|
35
|
Wang L, Li C, Weir MD, Zhang K, Zhou Y, Xu HHK, Reynolds MA. Novel multifunctional dental bonding agent for Class-V restorations to inhibit periodontal biofilms. RSC Adv 2017; 7:29004-29014. [PMID: 29910954 PMCID: PMC5998673 DOI: 10.1039/c6ra28711e] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
We recently developed a dental bonding agent to bond restorations to teeth using nanoparticles of amorphous calcium phosphate (NACP) for remineralization with rechargeable calcium and phosphate ion release. The objectives of this study were to: (1) incorporate an antibacterial monomer dimethylaminohexadecyl methacrylate (DMAHDM) and a protein-repellent agent 2-methacryloyloxyethyl phosphorylcholine (MPC); and (2) investigate protein adsorption and periodontitis-related biofilms for the first time. A primer, used to prime tooth structures for bonding, was made with pyromellitic glycerol dimethacrylate (PMGDM) and 2-hydroxyethyl methacrylate (HEMA). An adhesive was made with PMGDM, ethoxylated bisphenol A dimethacrylate and HEMA. NACP, MPC and DMAHDM were incorporated. Streptococcus gordonii, Actinomyces naeslundii, Porphyromonas gingivalis, Fusobacterium nucleatum were cultured to form single and multi-species biofilms. Colony-forming units (CFU), live/dead, metabolic activity, and polysaccharide were measured. Adding DMAHDM, MPC and NACP into the bonding agent did not compromise the dentin bond strength (p > 0.1). Bonding agents with 5% MPC reduced protein adsorption to 1/15 that of the control (p < 0.05). Bonding agents with 5% DMAHDM + 5% MPC had much greater reduction in biofilms than DMAHDM or MPC alone (p < 0.05). Biofilm CFU was reduced by 3 to 4 log via DMAHDM + MPC. Metabolic activities and polysaccharide of biofilms were also substantially reduced (p < 0.05). In conclusion, a novel bonding agent was developed for dental restorations with inhibition of biofilms, reducing CFU by 3 to 4 log. Besides remineralizartion and acid-neutralization via NACP to inhibit caries as shown previously, the multifunctional adhesive is promising for root restorations with subgingival margins to suppress periodontal pathogens and protect the periodontium.
Collapse
Affiliation(s)
- Lin Wang
- VIP Integrated Department, School and Hospital of Stomatology, Jilin University, Changchun, China
- Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Chunyan Li
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Michael D. Weir
- Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Ke Zhang
- Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Yanmin Zhou
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Hockin H. K. Xu
- Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
- Center for Stem Cell Biology & Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Mechanical Engineering, University of Maryland Baltimore County, Baltimore County, MD 21250, USA
| | - Mark A. Reynolds
- Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| |
Collapse
|
36
|
De Filippis A, Fiorentino M, Guida L, Annunziata M, Nastri L, Rizzo A. Vitamin D reduces the inflammatory response by Porphyromonas gingivalis infection by modulating human β-defensin-3 in human gingival epithelium and periodontal ligament cells. Int Immunopharmacol 2017; 47:106-117. [DOI: 10.1016/j.intimp.2017.03.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 03/07/2017] [Accepted: 03/22/2017] [Indexed: 10/19/2022]
|
37
|
Gerits E, Verstraeten N, Michiels J. New approaches to combat Porphyromonas gingivalis biofilms. J Oral Microbiol 2017; 9:1300366. [PMID: 28473880 PMCID: PMC5405727 DOI: 10.1080/20002297.2017.1300366] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 03/01/2017] [Accepted: 02/22/2017] [Indexed: 12/14/2022] Open
Abstract
In nature, bacteria predominantly reside in structured, surface-attached communities embedded in a self-produced, extracellular matrix. These so-called biofilms play an important role in the development and pathogenesis of many infections, as they are difficult to eradicate due to their resistance to antimicrobials and host defense mechanisms. This review focusses on the biofilm-forming periodontal bacterium Porphyromonas gingivalis. Current knowledge on the virulence mechanisms underlying P. gingivalis biofilm formation is presented. In addition, oral infectious diseases in which P. gingivalis plays a key role are described, and an overview of conventional and new therapies for combating P. gingivalis biofilms is given. More insight into this intriguing pathogen might direct the development of better strategies to combat oral infections.
Collapse
Affiliation(s)
- Evelien Gerits
- Department of Microbial and Molecular Systems, KU Leuven, Centre of Microbial and Plant Genetics, Leuven, Belgium
| | - Natalie Verstraeten
- Department of Microbial and Molecular Systems, KU Leuven, Centre of Microbial and Plant Genetics, Leuven, Belgium
| | - Jan Michiels
- Department of Microbial and Molecular Systems, KU Leuven, Centre of Microbial and Plant Genetics, Leuven, Belgium
| |
Collapse
|
38
|
Sobocinski V, Dridi SM, Bisson C, Jeanne S, Gaultier F, Prost-Squarcioni C, Bernard P, Pascal F, Lefevre B, Weber P, Abasq C, Agbo-Godeau S, Joly P, Ingen-Housz-Oro S, Duvert-Lehembre S. [Oral care recommendations for patients with oral autoimmune bullous diseases]. Ann Dermatol Venereol 2016; 144:182-190. [PMID: 28011091 DOI: 10.1016/j.annder.2016.09.680] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Revised: 08/14/2016] [Accepted: 09/23/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND Autoimmune bullous diseases (AIBD) may cause chronic oral lesions that progress insidiously. AIMS To provide recommendations for optimal oral-dental management of patients presenting AIBD with oral involvement. PATIENTS AND METHODS In the absence of scientific studies with high levels of proof, these recommendations have been drawn up at two meetings by a committee of experts on AIBD comprising 7 dermatologists, 1 stomatologist, 1 maxillofacial surgeon, 2 odontologists and 4 parodontologists. RESULTS The oral lesions associated with AIBD may be classified into three grades of severity: severe (generalised erosive gingivitis affecting at least 30% of dental sites), moderate (localised erosive gingivitis affecting less than 30% of dental sites) and controlled (no erosive oral lesions). Good oral-dental hygiene suited to the severity of the oral lesions, must be practised continually by these patients so as to avoid the formation of dental plaque, which aggravates symptoms. Dental and parodontal care must be considered in accordance with the severity grade of the oral lesions: in severe cases, the dental plaque must be eliminated manually with a curette, but several types of care (descaling, treatment for tooth decay, non-urgent extractions, etc.) must be suspended until the grade of severity is moderate or until the disease is stabilised.
Collapse
Affiliation(s)
- V Sobocinski
- Clinique dermatologique, hôpital Charles-Nicolle, CHU de Rouen, 1, rue de Germont, 76031 Rouen cedex, France.
| | - S-M Dridi
- Service d'odontologie, hôpital Henri-Mondor, 51, avenue du maréchal-de-Lattre-de-Tassigny, 94010 Créteil, France
| | - C Bisson
- Service d'odontologie, CHRU de Nancy, 29, avenue de Lattre-de-Tassigny, 54000 Nancy, France
| | - S Jeanne
- Service d'odontologie, CHRU de Rennes, 2, rue Henri-Le-Guilloux, 35000 Rennes, France
| | - F Gaultier
- Service d'odontologie, hôpital Henri-Mondor, 51, avenue du maréchal-de-Lattre-de-Tassigny, 94010 Créteil, France
| | - C Prost-Squarcioni
- Service de dermatologie, hôpital Avicenne, 125, rue de Stalingrad, 93000 Bobigny, France
| | - P Bernard
- Service de dermatologie, hôpital Robert-Debré, rue du général-Koening, 51100 Reims, France
| | - F Pascal
- Service de dermatologie, hôpital Avicenne, 125, rue de Stalingrad, 93000 Bobigny, France
| | - B Lefevre
- Service d'odontologie, hôpital Robert-Debré, rue du général-Koening, 51100 Reims, France
| | - P Weber
- Service de dermatologie, hôpital Avicenne, 125, rue de Stalingrad, 93000 Bobigny, France
| | - C Abasq
- Service de dermatologie, hôpital Morvan, 2, avenue maréchal-Foch, 29200 Brest, France
| | - S Agbo-Godeau
- Service de stomatologie et chirurgie maxillo-faciale, hôpital Pitié-Salpêtrière, 47-83 boulevard de l'Hôpital, 75013 Paris, France
| | - P Joly
- Clinique dermatologique, hôpital Charles-Nicolle, CHU de Rouen, 1, rue de Germont, 76031 Rouen cedex, France
| | - S Ingen-Housz-Oro
- Service de dermatologie, hôpital Henri-Mondor, 51, avenue du maréchal-de-Lattre-de-Tassigny, 94010 Créteil, France
| | - S Duvert-Lehembre
- Service de dermatologie, hôpital de Dunkerque, 130, avenue Louis-Herbeaux, 59240 Dunkerque, France
| |
Collapse
|
39
|
Bee Venom Inhibits Porphyromonas gingivalis Lipopolysaccharides-Induced Pro-Inflammatory Cytokines through Suppression of NF-κB and AP-1 Signaling Pathways. Molecules 2016; 21:molecules21111508. [PMID: 27834922 PMCID: PMC6273372 DOI: 10.3390/molecules21111508] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 10/28/2016] [Accepted: 11/04/2016] [Indexed: 01/09/2023] Open
Abstract
Periodontitis is a chronic inflammatory disease that leads to destruction of tooth supporting tissues. Porphyromonas gingivalis (P. gingivalis), especially its lipopolysaccharides (LPS), is one of major pathogens that cause periodontitis. Bee venom (BV) has been widely used as a traditional medicine for various diseases. Previous studies have demonstrated the anti-inflammatory, anti-bacterial effects of BV. However, a direct role and cellular mechanism of BV on periodontitis-like human keratinocytes have not been explored. Therefore, we investigated the anti-inflammatory mechanism of BV against P. gingivalis LPS (PgLPS)-induced HaCaT human keratinocyte cell line. The anti-inflammatory effect of BV was demonstrated by various molecular biological methods. The results showed that PgLPS increased the expression of Toll-like receptor (TLR)-4 and pro-inflammatory cytokines, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, IL-8, and interferon (IFN)-γ. In addition, PgLPS induced activation of the signaling pathways of inflammatory cytokines-related transcription factors, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and activator protein 1 (AP-1). BV effectively inhibited those pro-inflammatory cytokines through suppression of NF-κB and AP-1 signaling pathways. These results suggest that administration of BV attenuates PgLPS-induced inflammatory responses. Furthermore, BV may be a useful treatment to anti-inflammatory therapy for periodontitis.
Collapse
|
40
|
Wang L, Xie X, Weir MD, Fouad AF, Zhao L, Xu HHK. Effect of bioactive dental adhesive on periodontal and endodontic pathogens. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2016; 27:168. [PMID: 27699574 DOI: 10.1007/s10856-016-5778-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 09/09/2016] [Indexed: 06/06/2023]
Abstract
The objectives of this study were to: (1) develop a new bioactive dental bonding agent with nanoparticles of amorphous calcium phosphate and dimethylaminohexadecyl methacrylate for tooth root caries restorations and endodontic applications, and (2) investigate biofilm inhibition by the bioactive bonding agent against eight species of periodontal and endodontic pathogens for the first time. Bonding agent was formulated with 5 % of dimethylaminohexadecyl methacrylate. Nanoparticles of amorphous calcium phosphate at 30 wt% was mixed into adhesive. Eight species of biofilms were grown on resins: Porphyromonas gingivalis, Prevotella intermedia, Prevotella nigrescens, Aggregatibacter actinomycetemcomitans, Fusobacterium nucleatum, Parvimonas micra, Enterococcus faecalis, Enterococcus faecium. Colony-forming units, live/dead assay, biomass, metabolic activity and polysaccharide of biofilms were determined. The results showed that adding dimethylaminohexadecyl methacrylate and nanoparticles of amorphous calcium phosphate into bonding agent did not decrease dentin bond strength (P > 0.1). Adding dimethylaminohexadecyl methacrylate reduced the colony-forming units of all eight species of biofilms by nearly three orders of magnitude. The killing efficacy of dimethylaminohexadecyl methacrylate resin was: P. gingivalis > A. actinomycetemcomitans > P. intermedia > P. nigrescens > F. nucleatum > P. micra > E. faecalis > E. faecium. Dimethylaminohexadecyl methacrylate resin had much less biomass, metabolic activity and polysaccharide of biofilms than those without dimethylaminohexadecyl methacrylate (P < 0.05). In conclusion, a novel dental adhesive was developed for root caries and endodontic applications, showing potent inhibition of biofilms of eight species of periodontal and endodontic pathogens, and reducing colony-forming units by three orders of magnitude. The bioactive adhesive is promising for tooth root restorations to provide subgingival margins with anti-periodontal pathogen capabilities, and for endodontic sealer applications to combat endodontic biofilms.
Collapse
Affiliation(s)
- Lin Wang
- VIP Integrated Department, Stomatological Hospital of Jilin University, Changchun, China
- Department of Endodontics, Periodontics and Prosthodontics, University of Maryland Dental School, Baltimore, MD, 21201, USA
| | - Xianju Xie
- Department of Endodontics, Periodontics and Prosthodontics, University of Maryland Dental School, Baltimore, MD, 21201, USA
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Michael D Weir
- Department of Endodontics, Periodontics and Prosthodontics, University of Maryland Dental School, Baltimore, MD, 21201, USA
| | - Ashraf F Fouad
- Department of Endodontics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Liang Zhao
- Department of Endodontics, Periodontics and Prosthodontics, University of Maryland Dental School, Baltimore, MD, 21201, USA.
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Hockin H K Xu
- Department of Endodontics, Periodontics and Prosthodontics, University of Maryland Dental School, Baltimore, MD, 21201, USA.
- Center for Stem Cell Biology & Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Mechanical Engineering, University of Maryland, Baltimore County, MD, 21250, USA.
| |
Collapse
|
41
|
Kashiwagi Y, Takedachi M, Mori K, Kubota M, Yamada S, Kitamura M, Murakami S. High glucose-induced oxidative stress increases IL-8 production in human gingival epithelial cells. Oral Dis 2016; 22:578-84. [PMID: 27171647 DOI: 10.1111/odi.12502] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 04/25/2016] [Accepted: 05/08/2016] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Diabetes is often associated with increased prevalence and severity of periodontal disease. We hypothesized that gingival epithelial cells modify periodontal disease progression and predicted that hyperglycemia would activate an inflammatory response in human gingival epithelial cells (HGECs). MATERIALS AND METHODS We tested our hypothesis in immortalized HGECs (epi 4 cells) isolated from periodontal tissue and transfected with the simian virus 40 T antigen. The epi 4 cells were cultured in high (25 mM, HG) and normal (6 mM, NG) glucose conditions. RESULTS The epi 4 cells showed increased interleukin-8 (IL-8) protein secretion and mRNA expression when cultured in HG, compared with in NG. These effects were not associated with increased cell proliferation and were not observed in a hyperosmolar control group (normal glucose with 19 mM mannitol). Increased IL-8 secretion in HG was inhibited by pretreatment with an antioxidant, N-acetylcysteine, or a protein kinase C inhibitor, Ro31-8220. Hyperglycemia did not affect IL-8 secretion by gingival fibroblasts or periodontal ligament cells. In epi 4 cells, hyperglycemia also induced expression of toll-like receptor 2 (TLR2) but not TLR4. CONCLUSION These findings suggest a potential participation of epithelial cells in periodontal disease during diabetes by evoking an excessive host inflammatory response.
Collapse
Affiliation(s)
- Y Kashiwagi
- Division of Oral Biology and Disease Control, Department of Periodontology, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - M Takedachi
- Division of Oral Biology and Disease Control, Department of Periodontology, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - K Mori
- Division of Oral Biology and Disease Control, Department of Periodontology, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - M Kubota
- Division of Oral Biology and Disease Control, Department of Periodontology, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - S Yamada
- Division of Oral Biology and Disease Control, Department of Periodontology, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - M Kitamura
- Division of Oral Biology and Disease Control, Department of Periodontology, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - S Murakami
- Division of Oral Biology and Disease Control, Department of Periodontology, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| |
Collapse
|
42
|
Savage JR, Pulsipher A, Rao NV, Kennedy TP, Prestwich GD, Ryan ME, Lee WY. A Modified Glycosaminoglycan, GM-0111, Inhibits Molecular Signaling Involved in Periodontitis. PLoS One 2016; 11:e0157310. [PMID: 27308827 PMCID: PMC4911086 DOI: 10.1371/journal.pone.0157310] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 05/30/2016] [Indexed: 01/08/2023] Open
Abstract
Background Periodontitis is characterized by microbial infection, inflammation, tissue breakdown, and accelerated loss of alveolar bone matrix. Treatment targeting these multiple stages of the disease provides ways to treat or prevent periodontitis. Certain glycosaminoglycans (GAGs) block multiple inflammatory mediators as well as suppress bacterial growth, suggesting that these GAGs may be exploited as a therapeutic for periodontitis. Methods We investigated the effects of a synthetic GAG, GM-0111, on various molecular events associated with periodontitis: growth of Porphyromonas gingivalis (P. gingivalis) and Aggregatibacter actinomycetemcomitans (A. actinomycetemcomitans) pathogenic bacteria associated with periodontitis; activation of pro-inflammatory signaling through TLR2 and TLR4 in mouse macrophage RAW 264.7 cells and heterologously expressed HEK 293 cells; osteoclast formation and bone matrix resorption in cultured mouse pre-osteoclasts. Results (1) GM-0111 suppressed the growth of P. gingivalis and A. actinomycetemcomitans even at 1% (w/v) solution. The antibacterial effects of GM-0111 were stronger than hyaluronic acid (HA) or xylitol in P. gingivalis at all concentrations and comparable to xylitol in A. actinomycetemcomitans at ≥2% (w/v) solution. We also observed that GM-0111 suppressed biofilm formation of P. gingivalis and these effects were much stronger than HA. (2) GM-0111 inhibited TLR-mediated pro-inflammatory cellular signaling both in macrophage and HEK 293 cells with higher selectivity for TLR2 than TLR4 (IC50 of 1–10 ng/mL vs. > 100 μg/mL, respectively). (3) GM-0111 blocked RANKL-induced osteoclast formation (as low as 300 ng/mL) and bone matrix resorption. While GM-0111 showed high affinity binding to RANKL, it did not interfere with RANKL/RANK/NF-κB signaling, suggesting that GM-0111 inhibits osteoclast formation by a RANKL-RANK-independent mechanism. Conclusions We report that GM-0111 inhibits multiple molecular events involved in periodontitis, spanning from the early pro-inflammatory TLR signaling, to pathways activated at the later stage component of bone loss.
Collapse
Affiliation(s)
- Justin R. Savage
- GlycoMira Therapeutics, Inc. Salt Lake City, UT, 84108, United States of America
| | - Abigail Pulsipher
- GlycoMira Therapeutics, Inc. Salt Lake City, UT, 84108, United States of America
| | - Narayanam V. Rao
- GlycoMira Therapeutics, Inc. Salt Lake City, UT, 84108, United States of America
| | - Thomas P. Kennedy
- GlycoMira Therapeutics, Inc. Salt Lake City, UT, 84108, United States of America
- Pulmonary Diseases Critical Care and Environmental Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, United States of America
| | - Glenn D. Prestwich
- GlycoMira Therapeutics, Inc. Salt Lake City, UT, 84108, United States of America
- Department of Medicinal Chemistry and Center for Therapeutic Biomaterials, University of Utah, Salt Lake City, UT, 84108, United States of America
| | - Maria E. Ryan
- Department of Oral Biology and Pathology, School of Dental Medicine, Stony Brook University, Stony Brook, NY, 11794, United States of America
| | - Won Yong Lee
- GlycoMira Therapeutics, Inc. Salt Lake City, UT, 84108, United States of America
- * E-mail:
| |
Collapse
|
43
|
Park M, Jeong S, Na H, Chung J. Porphyromonas gingivalisinduces autophagy in THP-1-derived macrophages. Mol Oral Microbiol 2016; 32:48-59. [DOI: 10.1111/omi.12153] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2016] [Indexed: 12/30/2022]
Affiliation(s)
- M.H. Park
- Department of Oral Microbiology; School of Dentistry; Pusan National University; Yangsan South Korea
| | - S.Y. Jeong
- Department of Oral Microbiology; School of Dentistry; Pusan National University; Yangsan South Korea
| | - H.S. Na
- Department of Oral Microbiology; School of Dentistry; Pusan National University; Yangsan South Korea
| | - J. Chung
- Department of Oral Microbiology; School of Dentistry; Pusan National University; Yangsan South Korea
| |
Collapse
|
44
|
Fournier-Larente J, Azelmat J, Yoshioka M, Hinode D, Grenier D. The Daiokanzoto (TJ-84) Kampo Formulation Reduces Virulence Factor Gene Expression in Porphyromonas gingivalis and Possesses Anti-Inflammatory and Anti-Protease Activities. PLoS One 2016; 11:e0148860. [PMID: 26859747 PMCID: PMC4747585 DOI: 10.1371/journal.pone.0148860] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 01/25/2016] [Indexed: 01/02/2023] Open
Abstract
Kampo formulations used in Japan to treat a wide variety of diseases and to promote health are composed of mixtures of crude extracts from the roots, bark, leaves, and rhizomes of a number of herbs. The present study was aimed at identifying the beneficial biological properties of Daiokanzoto (TJ-84), a Kampo formulation composed of crude extracts of Rhubarb rhizomes and Glycyrrhiza roots, with a view to using it as a potential treatment for periodontal disease. Daiokanzoto dose-dependently inhibited the expression of major Porphyromonas gingivalis virulence factors involved in host colonization and tissue destruction. More specifically, Daiokanzoto reduced the expression of the fimA, hagA, rgpA, and rgpB genes, as determined by quantitative real-time PCR. The U937-3xκB-LUC monocyte cell line transfected with a luciferase reporter gene was used to evaluate the anti-inflammatory properties of Daiokanzoto. Daiokanzoto attenuated the P. gingivalis-mediated activation of the NF-κB signaling pathway. It also reduced the secretion of pro-inflammatory cytokines (IL-6 and CXCL8) by lipopolysaccharide-stimulated oral epithelial cells and gingival fibroblasts. Lastly, Daiokanzoto, dose-dependently inhibited the catalytic activity of matrix metalloproteinases (-1 and -9). In conclusion, the present study provided evidence that Daiokanzoto shows potential for treating and/or preventing periodontal disease. The ability of this Kampo formulation to act on both bacterial pathogens and the host inflammatory response, the two etiological components of periodontal disease, is of high therapeutic interest.
Collapse
Affiliation(s)
- Jade Fournier-Larente
- Groupe de Recherche en Écologie Buccale (GREB), Faculté de Médecine Dentaire, Université Laval, Quebec City, QC, Canada
| | - Jabrane Azelmat
- Groupe de Recherche en Écologie Buccale (GREB), Faculté de Médecine Dentaire, Université Laval, Quebec City, QC, Canada
| | - Masami Yoshioka
- Department of Oral Health Science and Social Welfare, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Daisuke Hinode
- Department of Hygiene and Oral Health Science, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Daniel Grenier
- Groupe de Recherche en Écologie Buccale (GREB), Faculté de Médecine Dentaire, Université Laval, Quebec City, QC, Canada
- * E-mail:
| |
Collapse
|
45
|
Links between atherosclerotic and periodontal disease. Exp Mol Pathol 2016; 100:220-35. [DOI: 10.1016/j.yexmp.2016.01.006] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 01/08/2016] [Indexed: 02/06/2023]
|
46
|
Fournier-Larente J, Morin MP, Grenier D. Green tea catechins potentiate the effect of antibiotics and modulate adherence and gene expression in Porphyromonas gingivalis. Arch Oral Biol 2016; 65:35-43. [PMID: 26849416 DOI: 10.1016/j.archoralbio.2016.01.014] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 01/06/2016] [Accepted: 01/24/2016] [Indexed: 12/22/2022]
Abstract
OBJECTIVES A number of studies have brought evidence that green tea catechins may contribute to periodontal health. The objective of this study was to investigate the ability of a green tea extract and its principal constituent epigallocatechin-3-gallate (EGCG) to potentiate the antibacterial effects of antibiotics (metronidazole, tetracycline) against Porphyromonas gingivalis, and to modulate the adherence to oral epithelial cells and expression of genes coding for virulence factors and the high temperature requirement A (HtrA) stress protein in P. gingivalis. METHODS A broth microdilution assay was used to determine the antibacterial activity of the green tea extract and EGCG. The synergistic effects of either compounds in association with metronidazole or tetracycline were evaluated using the checkerboard technique. A fluorescent assay was used to determine bacterial adherence to oral epithelial cells. The modulation of gene expression in P. gingivalis was evaluated by quantitative RT-PCR. The Vibrio harveyi bioassay was used for monitoring quorum sensing inhibitory activity. RESULTS The MIC values of the green tea extract on P. gingivalis ranged from 250 to 1000 μg/ml, while those of EGCG ranged from 125 to 500 μg/ml. A marked synergistic effect on P. gingivalis growth was observed for the green tea extract or EGCG in combination with metronidazole. Both the green tea extract and EGCG caused a dose-dependent inhibition of P. gingivalis adherence to oral epithelial cells. On the one hand, green tea extract and EGCG dose-dependently inhibited the expression of several P. gingivalis genes involved in host colonization (fimA, hagA, hagB), tissue destruction (rgpA, kgp), and heme acquisition (hem). On the other hand, both compounds increased the expression of the stress protein htrA gene. The ability of the green tea extract and EGCG to inhibit quorum sensing may contribute to the modulation of gene expression. CONCLUSIONS This study explored the preventive and therapeutic potential of green tea catechins against periodontal disease. In addition to inhibit growth and adherence of P. gingivalis, a green tea extract and its main constituent EGCG was found to decrease the expression of genes coding for the major virulence factors.
Collapse
Affiliation(s)
- Jade Fournier-Larente
- Oral Ecology Research Group, Faculty of Dentistry, Université Laval, Quebec City, Quebec, Canada
| | - Marie-Pierre Morin
- Oral Ecology Research Group, Faculty of Dentistry, Université Laval, Quebec City, Quebec, Canada
| | - Daniel Grenier
- Oral Ecology Research Group, Faculty of Dentistry, Université Laval, Quebec City, Quebec, Canada.
| |
Collapse
|
47
|
Lombardo Bedran TB, Morin MP, Palomari Spolidorio D, Grenier D. Black Tea Extract and Its Theaflavin Derivatives Inhibit the Growth of Periodontopathogens and Modulate Interleukin-8 and β-Defensin Secretion in Oral Epithelial Cells. PLoS One 2015; 10:e0143158. [PMID: 26581041 PMCID: PMC4651492 DOI: 10.1371/journal.pone.0143158] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 11/02/2015] [Indexed: 12/12/2022] Open
Abstract
Over the years, several studies have brought evidence suggesting that tea polyphenols, mostly from green tea, may have oral health benefits. Since few data are available concerning the beneficial properties of black tea and its theaflavin derivatives against periodontal disease, the objective of this study was to investigate their antibacterial activity as well as their ability to modulate interleukin-8 and human β-defensin (hBD) secretion in oral epithelial cells. Among the periodontopathogenic bacteria tested, Porphyromonas gingivalis was found to be highly susceptible to the black tea extract and theaflavins. Moreover, our data indicated that the black tea extract, theaflavin and theaflavin-3,3’-digallate can potentiate the antibacterial effect of metronidazole and tetracycline against P. gingivalis. Using lipopolysaccharide-stimulated oral epithelial cells, the black tea extract (100 μg/ml), as well as theaflavin and theaflavin-3,3’-digallate (50 μg/ml) reduced interleukin-8 (IL-8) secretion by 85%, 79%, and 86%, respectively, thus suggesting an anti-inflammatory property. The ability of the black tea extract and its theaflavin derivatives to induce the secretion of the antimicrobial peptides hBD-1, hBD-2 and hBD-4 by oral epithelial cells was then evaluated. Our results showed that the black tea extract as well as theaflavin-3,3’-digallate were able to increase the secretion of the three hBDs. In conclusion, the ability of a black tea extract and theaflavins to exert antibacterial activity against major periodontopathogens, to attenuate the secretion of IL-8, and to induce hBD secretion in oral epithelial cells suggest that these components may have a beneficial effect against periodontal disease.
Collapse
Affiliation(s)
| | - Marie-Pierre Morin
- Oral Ecology Research Group, Faculty of Dentistry, Laval University, Quebec City, QC, Canada
| | - Denise Palomari Spolidorio
- Department of Oral Diagnosis and Surgery, Araraquara Dental School, State University of São Paulo, São Paulo, Brazil
| | - Daniel Grenier
- Oral Ecology Research Group, Faculty of Dentistry, Laval University, Quebec City, QC, Canada
- * E-mail:
| |
Collapse
|
48
|
Guo W, Wang P, Liu Z, Yang P, Ye P. The activation of pyrin domain-containing-3 inflammasome depends on lipopolysaccharide from Porphyromonas gingivalis and extracellular adenosine triphosphate in cultured oral epithelial cells. BMC Oral Health 2015; 15:133. [PMID: 26511096 PMCID: PMC4625523 DOI: 10.1186/s12903-015-0115-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 10/10/2015] [Indexed: 01/15/2023] Open
Abstract
Background Gingival epithelial cells are the major population of the gingival tissue, acting as the front-line defense against microbial intrusion and regulating the homeostasis of the periodontal tissue in health and disease via NLR family pyrin domain-containing-3 (NLRP3) inflammasome, which recognizes pathogen- and danger-associated molecular patterns (PAMPs and DAMPs). The aim of this study was to determine whether the activation of NLRP3 inflammasome depends on infection with the periodontal pathogen Porphyromonas gingivalis (P. gingivalis), or stimulation with P. gingivalis lipopolysaccharide (LPS), and/or extracellular adenosine triphosphate (ATP). Methods An oral epithelial cell line was treated with P. gingivalis, P. gingivalis LPS and ATP. The gene and protein expression of NLRP3 inflammasome components were quantified by real time RT-PCR and immunoblots. Production of IL-1β and IL-18 was measured by ELISA. Results There was no increase in NLRP3 inflammasome gene expression after P. gingivalis infection unless pre-stimulated by ATP. Obvious increases of NLRP3 inflammasome gene expression was observed after P. gingivalis LPS stimulation, even pre-stimulated by ATP at 2 h. Conclusions The findings indicate that the activation of NLRP3 inflammasome does not rely on P. gingivalis infection, unless stimulated by P. gingivalis LPS and/or extracellular ATP, suggesting diverse signaling pathways are involved in the host immune response.
Collapse
Affiliation(s)
- Wei Guo
- Department of Periodontology, Shandong Provincial Key Laboratory of Oral Biomedicine, Shandong University, Jinan, 250012, People's Republic of China. .,Department of Endodontics, Yantai Stomatological Hospital, Yantai, Shandong Province, China.
| | - Peng Wang
- Yantai Stomatological Hospital, Yantai, Shandong Province, China.
| | - Zhonghao Liu
- Yantai Stomatological Hospital, Yantai, Shandong Province, China.
| | - Pishan Yang
- Department of Periodontology, Shandong Provincial Key Laboratory of Oral Biomedicine, Shandong University, Jinan, 250012, People's Republic of China.
| | - Ping Ye
- Institute of Dental Research, Westmead Millennium Institute and Westmead Centre for Oral Health, Westmead Hospital, Westmead, Australia.
| |
Collapse
|
49
|
Hosn KN, Jefferson MM, Leding C, Shokouh‐Amiri S, Thomas EL. Inhibitors of bacterial protease enzymes for periodontal therapy. Clin Exp Dent Res 2015; 1:18-25. [PMID: 29744136 PMCID: PMC5839262 DOI: 10.1002/cre2.4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 07/24/2015] [Accepted: 08/04/2015] [Indexed: 11/10/2022] Open
Abstract
Locally applied therapeutic agents have become established in the treatment of periodontal disease. Inhibition of human metalloproteases by metal-chelating antibiotics contributes to the utility of local therapy. Adding inhibitors of bacterial proteases might extend and improve local therapy. The periodontal pathogen Porphyromonas gingivalis (Pg) produces two extracellular cysteine proteases (gingipains Rgp and Kgp) that are virulence factors and contribute to destruction of oral tissues. Our aims were to compare efficacy of protease inhibitors against gingipains and evaluate bactericidal activity of the inhibitors. Protease activity was measured in fluorescent assays with specific Rgp and Kgp substrates. Bacterial viability was measured with BacLight™ (Invitrogen, Inc., Carlsbad, CA) reagents. Pairs of inhibitors of Rgp and Kgp, respectively, were leupeptin and cathepsin B inhibitor II, KYT-1 and KYT-36, and PPACK and Z-FK-ck. The cysteine-protease inhibitor E64 was also tested. Rgp activity was higher than Kgp activity, and activity was higher in Pg 33277 and 49417 cell suspensions than in media. Concentrations required for 50% inhibition of Rgp in cell suspensions were 2 × 10-9, 2 × 10-9, 2 × 10-8, and 5 × 10-5 M for KYT-1, PPACK, leupeptin, and E64, respectively. Concentrations required for 50% Kgp inhibition were 5 × 10-10, 1 × 10-9, and 5 × 10-8 M for Z-FK-ck, KYT-36, and cathepsin B inhibitor II. E64 did not inhibit Kgp. Inhibition of Rgp could be accounted for by competition for binding between the arginine residue of the substrate and the guanidinobutane portion of E64. PPACK was the least selective, with a 10-fold difference in concentrations that inhibited Rgp and Kgp. KYT-1 and Z-FK-ck inhibited both Rgp and Kgp, but inhibitory concentrations differed by 10,000-fold. At up to 1 × 10-4 M, only Z-FK-ck was bactericidal. KYT-1 and KYT-36 were remarkably effective even when used in cell suspensions in which bacterial proteins could bind inhibitors or compete for binding to gingipains. These inhibitors might prove useful as an addition to locally applied therapeutic agents.
Collapse
Affiliation(s)
- Kalid N. Hosn
- Periodontology DepartmentUniversity of Tennessee Health Science CenterMemphisTennesseeUSA
| | - Mary Margaret Jefferson
- Bioscience Research Department, College of DentistryUniversity of Tennessee Health Science CenterMemphisTennesseeUSA
| | - Carlton Leding
- Bioscience Research Department, College of DentistryUniversity of Tennessee Health Science CenterMemphisTennesseeUSA
| | - Solomon Shokouh‐Amiri
- Bioscience Research Department, College of DentistryUniversity of Tennessee Health Science CenterMemphisTennesseeUSA
| | - Edwin L. Thomas
- Bioscience Research Department, College of DentistryUniversity of Tennessee Health Science CenterMemphisTennesseeUSA
- Microbiology, Immunology & Biochemistry DepartmentUniversity of Tennessee Health Science CenterMemphisTennesseeUSA
| |
Collapse
|
50
|
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
|