1
|
Wu C, Fujiki J, Mathieu J, Schwarz C, Cornell C, Alvarez PJJ. Phage-based biocontrol of Porphyromonas gingivalis through indirect targeting. Appl Environ Microbiol 2024; 90:e0095124. [PMID: 39248462 PMCID: PMC11497834 DOI: 10.1128/aem.00951-24] [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] [Received: 05/17/2024] [Accepted: 08/14/2024] [Indexed: 09/10/2024] Open
Abstract
Bacteriophages offer an opportunity for chemical-free, precise control of problematic bacteria, but this approach can be limited when lytic phages are difficult to obtain for the target host. In such cases, phage-based targeting of cooperating or cross-feeding bacteria (e.g., Streptococcus gordonii) can be an effective approach to control the problematic bacteria (e.g., Porphyromonas gingivalis). Using a dual-species biofilm system, phage predation of S. gordonii (108 PFU·mL-1) decreased the abundance of pathogenic P. gingivalis by >99% compared with no-treatment controls, while also inhibiting the production of cytotoxic metabolic end products (butyric and propionic acids). Phage treatment upregulated genes associated with interspecies co-adhesion (5- to 8-fold) and quorum sensing (10-fold) in residual P. gingivalis, which is conducive to increased potential to bind to S. gordonii. Counterintuitively, lower-titer phage applications (104 PFU·mL-1) increased the production of extracellular polymeric substance (EPS) by 22% and biofilm biomass by 50%. This overproduction of EPS may contribute to the phenomenon where the biofilm separated into two distinct species layers, as observed by confocal laser scanning microscopy. Although more complex mixed-culture systems should be considered to delineate the merits and limitations of this novel biocontrol approach (which would likely require the use of phage cocktails), our results offer proof of concept that indirect phage-based targeting can expand the applicability of phage-based control of pathogenic bacteria for public health protection. IMPORTANCE Lytic phages are valuable agents for targeted elimination of bacteria in diverse applications. Nevertheless, lytic phages are difficult to isolate for some target pathogens. We offer proof of concept that this limitation may be overcome via indirect phage targeting, which involves knocking out species that interact closely with and benefit the primary problematic target bacteria. Our target (P. gingivalis) only forms a periodontal pathogenic biofilm if the pioneer colonizer (S. gordonii) offers its surface for P. gingivalis to attach. Phage predation of the co-adhesive S. gordonii significantly reduced abundance of the target pathogen by >99%, decreased the total biofilm biomass by >44%, and suppressed its production of cytotoxic metabolic byproducts. Thus, this research extends the scope of phage-based biocontrol for public health protection.
Collapse
Affiliation(s)
- Chuncheng Wu
- Department of Civil and Environmental Engineering, Rice University, Houston, Texas, USA
| | - Jumpei Fujiki
- Department of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido, Japan
| | - Jacques Mathieu
- Department of Civil and Environmental Engineering, Rice University, Houston, Texas, USA
| | - Cory Schwarz
- Department of Civil and Environmental Engineering, Rice University, Houston, Texas, USA
| | - Carolyn Cornell
- Department of Civil and Environmental Engineering, Rice University, Houston, Texas, USA
| | - Pedro J. J. Alvarez
- Department of Civil and Environmental Engineering, Rice University, Houston, Texas, USA
| |
Collapse
|
2
|
Kaintura A, Ramar K. Comparative Evaluation of Biofilm Formation on Temporization Crown Materials Used in the Rehabilitation of Primary Dentition With Different Polishing Materials: An In Vitro Study. Cureus 2024; 16:e68944. [PMID: 39381466 PMCID: PMC11460649 DOI: 10.7759/cureus.68944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 09/08/2024] [Indexed: 10/10/2024] Open
Abstract
Introduction Advancements in dental materials have enhanced aesthetic treatments for managing dental caries and injuries in primary dentition. Bis-acryl composite-based temporization materials are now preferred for restoring primary crowns due to their superior properties. However, prolonged exposure to dietary and hygienic factors can lead to discoloration and roughness, making efficient polishing essential to prevent plaque buildup. Objective This study aims to evaluate Streptococcus mutans biofilm formation on temporization material polished with different polishing systems. Methods This study tested bis-acryl methacrylate temporization material. Thirty disk-shaped specimens were prepared and divided into three groups according to the polishing system used (n = 10 per group): Shofu Super Snap mini kit (Shofu, San Marcos, CA), aluminum oxide polishing paste, and propol polishing paste. Each group's specimens were polished according to the manufacturer's instructions. Surface roughness (SR), scanning electron microscopy (SEM) morphological analysis, and Streptococcus mutans biofilm formation were assessed for each group. Results The results showed significant differences in roughness average (Ra) values among the polishing materials, with the Shofu Super Snap mini kit having the highest roughness (Ra = 2.04), followed by propol polishing paste (Ra = 1.30) and aluminum oxide paste (Ra = 0.75). Additionally, polishing methods significantly affected mean colony-forming unit (CFU) levels, with the first group having the highest mean CFU value (0.24), with SEM images showing substantial biofilm formation by Streptococcus mutans. Conclusion Bacterial biofilm formation on the aluminum oxide paste group's surface differed from that on the propol polishing paste and aluminum oxide disc groups. The polishing techniques that we tested significantly influenced surface properties and biofilm formation. These findings suggest that selecting an appropriate polishing system can reduce the risk of gingival inflammation associated with temporization materials.
Collapse
Affiliation(s)
- Akansha Kaintura
- Pediatric and Preventive Dentistry, SRM (Sri Ramaswamy Memorial) Kattankulathur Dental College, Chennai, IND
| | - Kavitha Ramar
- Pedodontics and Preventive Dentistry, SRM (Sri Ramaswamy Memorial) Kattankulathur Dental College, Chennai, IND
| |
Collapse
|
3
|
Goenka S. Exploring the effect of butyric acid, a metabolite from periodontopathic bacteria, on primary human melanocytes: An in vitro study. J Oral Biosci 2024; 66:253-259. [PMID: 38215819 DOI: 10.1016/j.job.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 01/09/2024] [Accepted: 01/09/2024] [Indexed: 01/14/2024]
Abstract
Effects of butyric acid, a bacterial metabolite implicated in periodontitis progression, have never been examined on oral melanocytes. Herein, primary human epidermal melanocytes were used as a model for oral melanocytes. Results show the adverse effects of butyric acid (sodium butyrate; NaB) on them, which comprise marked cytotoxicity at higher concentrations (>1 mM) and robust differentiation at lower nontoxic concentrations. NaB did not alter MITF protein levels; however, it stimulated tyrosinase protein synthesis and inhibited tyrosinase activity, with no changes in cellular melanin. NaB did not affect oxidative stress, although it induced significant levels of the pro-inflammatory cytokine IL-6.
Collapse
Affiliation(s)
- Shilpi Goenka
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA; Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
4
|
Suhasini PC, Shetty SS, Shetty VV, Bhat V, Roopashree PG, Kumari NS. A Comparative Study of Serum Butyric Acid in Subjects with Tongue Cancer. JOURNAL OF DATTA MEGHE INSTITUTE OF MEDICAL SCIENCES UNIVERSITY 2024; 19:104-108. [DOI: 10.4103/jdmimsu.jdmimsu_822_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 01/15/2024] [Indexed: 01/03/2025]
Abstract
Abstract
Aim:
The aim of the study was to study the level of butyric acid in tongue cancer subjects.
Materials and Methods:
Thirty controls and 30 tongue cancer subjects were recruited for the study. Serum butyric acid levels were estimated using ELISA kits. Statistical analyses were done using SPSS vs 22. The Mann–Whitney U and Kruskal–Wallis tests were used. P ≤ 0.05 was considered statistically significant.
Results:
Butyric acid levels were significantly higher in control subjects when compared to case subjects.
Conclusion:
Butyric acid could be used as a potential anticancer agent in tongue cancer treatment.
Collapse
Affiliation(s)
- P. C. Suhasini
- Department of Biochemistry, KS Hegde Medical Academy, NITTE (Deemed to be University), Mangalore, India
| | - Shilpa S. Shetty
- Department of Cellomics, Lipidomics and Molecular Genetics Division, Central Research Laboratory, KS Hegde Medical Academy, NITTE (Deemed to be University), Mangalore, India
| | - Vijith Vittal Shetty
- Department of Oncology, KS Hegde Medical Academy, NITTE (Deemed to be University), Mangalore, India
| | - Vadisha Bhat
- Department of ENT, KS Hegde Medical Academy, NITTE (Deemed to be University), Mangalore, India
| | - P. G. Roopashree
- Department of Biochemistry, KS Hegde Medical Academy, NITTE (Deemed to be University), Mangalore, India
| | - N. Suchetha Kumari
- Department of Biochemistry, KS Hegde Medical Academy, NITTE (Deemed to be University), Mangalore, India
| |
Collapse
|
5
|
Xiao X, Xiao X, Liu Y, Sun H, Liu X, Guo Z, Li Q, Sun W. Metaproteomics Characterizes the Human Gingival Crevicular Fluid Microbiome Function in Periodontitis. J Proteome Res 2023. [PMID: 37327455 DOI: 10.1021/acs.jproteome.3c00143] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Periodontitis is the leading cause of tooth loss in adults worldwide. The human proteome and metaproteome characterization of periodontitis is not clearly understood. Gingival crevicular fluid samples were collected from eight periodontitis and eight healthy subjects. Both the human and microbial proteins were characterized by liquid chromatography coupled with high-resolution mass spectrometry. A total of 570 human proteins were found differentially expressed, which were primarily associated with inflammatory response, cell death, cellular junction, and fatty acid metabolism. For the metaproteome, 51 genera were identified, and 10 genera were found highly expressed in periodontitis, while 11 genera were downregulated. The analysis showed that microbial proteins related to butyrate metabolism were upregulated in periodontitis cases. In particular, correlation analysis showed that the expression of host proteins related to inflammatory response, cell death, cellular junction, and lipid metabolism correlates with the alteration of metaproteins, which reflect the changes of molecular function during the occurrence of periodontitis. This study showed that the gingival crevicular fluid human proteome and metaproteome could reflect the characteristics of periodontitis. This might benefit the understanding of the periodontitis mechanism.
Collapse
Affiliation(s)
- Xiaolian Xiao
- Core Facility of Instrument, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, China
| | - Xiaoping Xiao
- Core Facility of Instrument, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Yaoran Liu
- Department of Stomatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Haidan Sun
- Core Facility of Instrument, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China
| | - Xiaoyan Liu
- Core Facility of Instrument, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China
| | - Zhengguang Guo
- Core Facility of Instrument, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China
| | - Qian Li
- Department of Stomatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Wei Sun
- Core Facility of Instrument, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China
| |
Collapse
|
6
|
Basic A, Dahlén G. Microbial metabolites in the pathogenesis of periodontal diseases: a narrative review. FRONTIERS IN ORAL HEALTH 2023; 4:1210200. [PMID: 37388417 PMCID: PMC10300593 DOI: 10.3389/froh.2023.1210200] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 05/31/2023] [Indexed: 07/01/2023] Open
Abstract
The purpose of this narrative review is to highlight the importance of microbial metabolites in the pathogenesis of periodontal diseases. These diseases, involving gingivitis and periodontitis are inflammatory conditions initiated and maintained by the polymicrobial dental plaque/biofilm. Gingivitis is a reversible inflammatory condition while periodontitis involves also irreversible destruction of the periodontal tissues including the alveolar bone. The inflammatory response of the host is a natural reaction to the formation of plaque and the continuous release of metabolic waste products. The microorganisms grow in a nutritious and shielded niche in the periodontal pocket, protected from natural cleaning forces such as saliva. It is a paradox that the consequences of the enhanced inflammatory reaction also enable more slow-growing, fastidious, anaerobic bacteria, with often complex metabolic pathways, to colonize and thrive. Based on complex food chains, nutrient networks and bacterial interactions, a diverse microbial community is formed and established in the gingival pocket. This microbiota is dominated by anaerobic, often motile, Gram-negatives with proteolytic metabolism. Although this alternation in bacterial composition often is considered pathologic, it is a natural development that is promoted by ecological factors and not necessarily a true "dysbiosis". Normal commensals are adapting to the gingival crevice when tooth cleaning procedures are absent. The proteolytic metabolism is highly complex and involves a number of metabolic pathways with production of a cascade of metabolites in an unspecific manner. The metabolites involve short chain fatty acids (SCFAs; formic, acetic, propionic, butyric, and valeric acid), amines (indole, scatole, cadaverine, putrescine, spermine, spermidine) and gases (NH3, CO, NO, H2S, H2). A homeostatic condition is often present between the colonizers and the host response, where continuous metabolic fluctuations are balanced by the inflammatory response. While it is well established that the effect of the dental biofilm on the host response and tissue repair is mediated by microbial metabolites, the mechanisms behind the tissue destruction (loss of clinical attachment and bone) are still poorly understood. Studies addressing the functions of the microbiota, the metabolites, and how they interplay with host tissues and cells, are therefore warranted.
Collapse
|
7
|
Ohkusa T, Nishikawa Y, Sato N. Gastrointestinal disorders and intestinal bacteria: Advances in research and applications in therapy. Front Med (Lausanne) 2023; 9:935676. [PMID: 36825261 PMCID: PMC9941163 DOI: 10.3389/fmed.2022.935676] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 12/27/2022] [Indexed: 02/09/2023] Open
Abstract
Intestinal bacteria coexist with humans and play a role in suppressing the invasion of pathogens, producing short-chain fatty acids, producing vitamins, and controlling the immune system. Studies have been carried out on culturable bacterial species using bacterial culture methods for many years. However, as metagenomic analysis of bacterial genes has been developed since the 1990s, it has recently revealed that many bacteria in the intestine cannot be cultured and that approximately 1,000 species and 40 trillion bacteria are present in the gut microbiota. Furthermore, the composition of the microbiota is different in each disease state compared with the healthy state, and dysbiosis has received much attention as a cause of various diseases. Regarding gastrointestinal diseases, dysbiosis has been reported to be involved in inflammatory bowel disease, irritable bowel syndrome, and non-alcoholic steatohepatitis. Recent findings have also suggested that dysbiosis is involved in colon cancer, liver cancer, pancreatic cancer, esophageal cancer, and so on. This review focuses on the relationship between the gut microbiota and gastrointestinal/hepatobiliary diseases and also discusses new therapies targeting the gut microbiota.
Collapse
Affiliation(s)
| | - Yuriko Nishikawa
- Department of Microbiota Research, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Nobuhiro Sato
- Department of Microbiota Research, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
8
|
Proof-of-Principle Study Suggesting Potential Anti-Inflammatory Activity of Butyrate and Propionate in Periodontal Cells. Int J Mol Sci 2022; 23:ijms231911006. [PMID: 36232340 PMCID: PMC9570314 DOI: 10.3390/ijms231911006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/14/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Short-chain fatty acids (SCFAs) are potent immune modulators present in the gingival crevicular fluid. It is therefore likely that SCFAs exert a role in periodontal health and disease. To better understand how SCFAs can module inflammation, we screened acetic acid, propionic acid, and butyric acid for their potential ability to lower the inflammatory response of macrophages, gingival fibroblasts, and oral epithelial cells in vitro. To this end, RAW 264.7 and primary macrophages were exposed to LPSs from Porphyromonas gingivalis (P. gingivalis) with and without the SCFAs. Moreover, gingival fibroblasts and HSC2 oral epithelial cells were exposed to IL1β and TNFα with and without the SCFAs. We report here that butyrate was effective in reducing the lipopolysaccharide (LPS)-induced expression of IL6 and chemokine (C-X-C motif) ligand 2 (CXCL2) in the RAW 264.7 and primary macrophages. Butyrate also reduced the IL1β and TNFα-induced expression of IL8, chemokine (C-X-C motif) ligand 1 (CXCL1), and CXCL2 in gingival fibroblasts. Likewise, butyrate lowered the induced expression of CXCL1 and CXCL2, but not IL8, in HSC2 cells. Butyrate further caused a reduction of p65 nuclear translocation in RAW 264.7 macrophages, gingival fibroblasts, and HSC2 cells. Propionate and acetate partially lowered the inflammatory response in vitro but did not reach the level of significance. These findings suggest that not only macrophages, but also gingival fibroblasts and oral epithelial cells are susceptive to the anti-inflammatory activity of butyrate.
Collapse
|
9
|
Levine M, Lohinai ZM. Resolving the Contradictory Functions of Lysine Decarboxylase and Butyrate in Periodontal and Intestinal Diseases. J Clin Med 2021; 10:jcm10112360. [PMID: 34072136 PMCID: PMC8198195 DOI: 10.3390/jcm10112360] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 05/14/2021] [Indexed: 11/16/2022] Open
Abstract
Periodontal disease is a common, bacterially mediated health problem worldwide. Mastication (chewing) repeatedly traumatizes the gingiva and periodontium, causing traces of inflammatory exudate, gingival crevicular fluid (GCF), to appear in crevices between the teeth and gingiva. Inadequate tooth cleaning causes a dentally adherent microbial biofilm composed of commensal salivary bacteria to appear around these crevices where many bacteria grow better on GCF than in saliva. We reported that lysine decarboxylase (Ldc) from Eikenella corrodens depletes the GCF of lysine by converting it to cadaverine and carbon dioxide. Lysine is an amino acid essential for the integrity and continuous renewal of dentally attached epithelium acting as a barrier to microbial products. Unless removed regularly by oral hygiene, bacterial products invade the lysine-deprived dental attachment where they stimulate inflammation that enhances GCF exudation. Cadaverine increases and supports the development of a butyrate-producing microbiome that utilizes the increased GCF substrates to slowly destroy the periodontium (dysbiosis). A long-standing paradox is that acid-induced Ldc and butyrate production support a commensal (probiotic) microbiome in the intestine. Here, we describe how the different physiologies of the respective tissues explain how the different Ldc and butyrate functions impact the progression and control of these two chronic diseases.
Collapse
Affiliation(s)
- Martin Levine
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Correspondence:
| | - Zsolt M. Lohinai
- Department of Conservative Dentistry, Semmelweis University, H-1088 Budapest, Hungary;
| |
Collapse
|
10
|
Na HS, Kim S, Kim S, Yu Y, Kim SY, Kim HJ, Lee JY, Lee JH, Chung J. Molecular subgroup of periodontitis revealed by integrated analysis of the microbiome and metabolome in a cross-sectional observational study. J Oral Microbiol 2021; 13:1902707. [PMID: 33828820 PMCID: PMC8008934 DOI: 10.1080/20002297.2021.1902707] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 01/27/2021] [Accepted: 03/10/2021] [Indexed: 11/29/2022] Open
Abstract
Background: Periodontitis (PT) is a multifactorial, chronic inflammatory disease that can have heterogeneous clinical presentations. The oral microbiome and its metabolites have been implicated as the causes and regulators of PT pathogenesis. In this study, we assessed the oral microbiome and its metabolome in PT patients to clarify the interactions between the microbiome and its metabolites.Methods: A total of 112 subjects were recruited. Buccal and supragingival samples were collected for microbiome analysis. Saliva samples were collected for metabolomic analyses. Microbiome and metabolome data were analyzed and further integrated for combined analysis using various bioinformatics approaches.Results: Oral metabolomic analysis identified 28 metabolites distinguishing the healthy (H) and PT groups. PT group were further clustered into two subgroups (PT_G1 and PT_G2) depending on metabolite profiles. Oral microbiome analysis revealed discriminatory bacterial species in the H, PT_G1, and PT_G2 microbiota. Interestingly, PT_G2 had significantly higher concentration of short chain fatty acids and higher abundance of pathogenic bacteria. Integrated analysis of the microbiome and metabolome showed close association.Conclusion: Our results provide evidence of a close interplay between the oral microbiome and metabolome. Multi-omics approach including microbiome and microbe-associated metabolites may serve as diagnostic biomarkers and enhance treatment prediction in periodontal disease.
Collapse
Affiliation(s)
- Hee Sam Na
- Department of Oral Microbiology, School of Dentistry, Pusan National University, Busan, South Korea
- Oral Genomics Research Center, Pusan National University, Busan, South Korea
| | - Suhkmann Kim
- Department of Chemistry, Center for Proteome Biophysics, and Chemistry Institute for Functional Materials, Pusan National University, Busan, South Korea
| | - Seonghye Kim
- Department of Chemistry, Center for Proteome Biophysics, and Chemistry Institute for Functional Materials, Pusan National University, Busan, South Korea
| | - Yeuni Yu
- Interdisplinary Program of Genomic Science, Pusan National University, Busan, South Korea
| | - Si Yeong Kim
- Department of Oral Microbiology, School of Dentistry, Pusan National University, Busan, South Korea
- Oral Genomics Research Center, Pusan National University, Busan, South Korea
| | - Hyun-Joo Kim
- Department of Periodontology, School of Dentistry, Pusan National University, Busan, South Korea
- Dental Research Institute, School of Dentistry, Pusan National University, Busan, South Korea
| | - Ju Youn Lee
- Department of Periodontology, School of Dentistry, Pusan National University, Busan, South Korea
- Dental Research Institute, School of Dentistry, Pusan National University, Busan, South Korea
| | - Jae-Hyung Lee
- Department of Oral Microbiology, School of Dentistry, Kyung Hee University, Seoul, South Korea
- Department of Life and Nanopharmaceutical Sciences, Kyung Hee University, Seoul, South Korea
- Kyung Hee Medical Science Research Institute, Kyung Hee University, Seoul, South Korea
| | - Jin Chung
- Department of Oral Microbiology, School of Dentistry, Pusan National University, Busan, South Korea
- Oral Genomics Research Center, Pusan National University, Busan, South Korea
| |
Collapse
|
11
|
Abstract
Although the composition of the oral human microbiome is now well studied, regulation of genes within oral microbial communities remains mostly uncharacterized. Current concepts of periodontal disease and caries highlight the importance of oral biofilms and their role as etiological agents of those diseases. Currently, there is increased interest in exploring and characterizing changes in the composition and gene-expression profiles of oral microbial communities. These efforts aim to identify changes in functional activities that could explain the transition from health to disease and the reason for the chronicity of those infections. It is now clear that the functions of distinct species within the subgingival microbiota are intimately intertwined with the rest of the microbial community. This point highlights the relevance of examining the expression profile of specific species within the subgingival microbiota in the case of periodontal disease or caries lesions, in the context of the other members of the biofilm in vivo. Metatranscriptomic analysis of the oral community is the starting point for identifying environmental signals that modulate the shift in metabolism of the community from commensal to dysbiotic. These studies give a snapshot of the expression patterns of microbial communities and also allow us to determine triggers to diseases. For example, in the case of caries, studies have unveiled a potential new pathway of sugar metabolism, namely the use of sorbitol as an additional source of carbon by Streptococcus mutans; and in the case of periodontal disease, high levels of extracellular potassium could be a signal of disease. Longitudinal studies are needed to identify the real markers of the initial stages of caries and periodontal disease. More information on the gene-expression profiles of the host, along with the patterns from the microbiome, will lead to a clearer understanding of the modulation of health and disease. This review presents a summary of these initial studies, which have opened the door to a new understanding of the dynamics of the oral community during the dysbiotic process in the oral cavity.
Collapse
Affiliation(s)
- Ana E Duran-Pinedo
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL, USA
| |
Collapse
|
12
|
Guan X, Li W, Meng H. A double-edged sword: Role of butyrate in the oral cavity and the gut. Mol Oral Microbiol 2020; 36:121-131. [PMID: 33155411 DOI: 10.1111/omi.12322] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 10/22/2020] [Accepted: 11/02/2020] [Indexed: 12/19/2022]
Abstract
Butyrate, a four-carbon short-chain fatty acid (SCFA), is a metabolite of anaerobic bacteria. Butyrate has primarily been described as an energy substance in the studies on the digestive tract. The multiple mechanisms of its protective function in the gut and on underlying diseases (including metabolic diseases, diseases of the nervous system, and osteoporosis) via interaction with intestinal epithelial cells and immune cells have been well documented. There are many butyrogenic bacteria in the oral cavity as well. As essential components of the oral microbiome, periodontal pathogens are also able to generate butyrate when undergoing metabolism. Considerable evidence has indicated that butyrate plays an essential role in the initiation and perpetuation of periodontitis. However, butyrate is considered to participate in the pro-inflammatory activities in periodontal tissue and the reactivation of latent viruses. In this review, we focused on the production and biological impact of butyrate in both intestine and oral cavity and explained the possible pathway of various diseases that were engaged by butyrate. Finally, we suggested two hypotheses, which may give a better understanding of the significantly different functions of butyrate in different organs (i.e., the expanded butyrate paradox).
Collapse
Affiliation(s)
- Xiaoyuan Guan
- Department of Periodontology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Wenjing Li
- Department of Periodontology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Huanxin Meng
- Department of Periodontology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
13
|
Kin LX, Butler CA, Slakeski N, Hoffmann B, Dashper SG, Reynolds EC. Metabolic cooperativity between Porphyromonas gingivalis and Treponema denticola. J Oral Microbiol 2020; 12:1808750. [PMID: 32944158 PMCID: PMC7482830 DOI: 10.1080/20002297.2020.1808750] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background Porphyromonas gingivalis and Treponema denticola are proteolytic periodontopathogens that co-localize in polymicrobial subgingival plaque biofilms, display in vitro growth symbiosis and synergistic virulence in animal models of disease. These symbioses are underpinned by a range of metabolic interactions including cooperative hydrolysis of glycine-containing peptides to produce free glycine, which T. denticola uses as a major energy and carbon source. Objective To characterize the P. gingivalis gene products essential for these interactions. Methods: The P. gingivalis transcriptome exposed to cell-free T. denticola conditioned medium was determined using RNA-seq. P. gingivalis proteases potentially involved in hydrolysis of glycine-containing peptides were identified using a bioinformatics approach. Results One hundred and thirty-twogenes displayed differential expression, with the pattern of gene expression consistent with succinate cross-feeding from T. denticola to P. gingivalis and metabolic shifts in the P. gingivalis folate-mediated one carbon superpathway. Interestingly, no P. gingivalis proteases were significantly up-regulated. Three P. gingivalis proteases were identified as candidates and inactivated to determine their role in the release of free glycine. P. gingivalis PG0753 and PG1788 but not PG1605 are involved in the hydrolysis of glycine-containing peptides, making free glycine available for T. denticola utilization. Conclusion Collectively these metabolic interactions help to partition resources and engage synergistic interactions between these two species.
Collapse
Affiliation(s)
- Lin Xin Kin
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Melbourne, Australia
| | - Catherine A Butler
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Melbourne, Australia
| | - Nada Slakeski
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Melbourne, Australia
| | - Brigitte Hoffmann
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Melbourne, Australia
| | - Stuart G Dashper
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Melbourne, Australia
| | - Eric C Reynolds
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
14
|
Magrin GL, Strauss FJ, Benfatti CAM, Maia LC, Gruber R. Effects of Short-Chain Fatty Acids on Human Oral Epithelial Cells and the Potential Impact on Periodontal Disease: A Systematic Review of In Vitro Studies. Int J Mol Sci 2020; 21:ijms21144895. [PMID: 32664466 PMCID: PMC7402343 DOI: 10.3390/ijms21144895] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 12/22/2022] Open
Abstract
Short-chain fatty acids (SCFA), bacterial metabolites released from dental biofilm, are supposed to target the oral epithelium. There is, however, no consensus on how SCFA affect the oral epithelial cells. The objective of the present study was to systematically review the available in vitro evidence of the impact of SCFA on human oral epithelial cells in the context of periodontal disease. A comprehensive electronic search using five databases along with a grey literature search was performed. In vitro studies that evaluated the effects of SCFA on human oral epithelial cells were eligible for inclusion. Risk of bias was assessed by the University of Bristol's tool for assessing risk of bias in cell culture studies. Certainty in cumulative evidence was evaluated using GRADE criteria (grading of recommendations assessment, development, and evaluation). Of 3591 records identified, 10 were eligible for inclusion. A meta-analysis was not possible due to the heterogeneity between the studies. The risk of bias across the studies was considered "serious" due to the presence of methodological biases. Despite these limitations, this review showed that SCFA negatively affect the viability of oral epithelial cells by activating a series of cellular events that includes apoptosis, autophagy, and pyroptosis. SCFA impair the integrity and presumably the transmigration of leucocytes through the epithelial layer by changing junctional and adhesion protein expression, respectively. SCFA also affect the expression of chemokines and cytokines in oral epithelial cells. Future research needs to identify the underlying signaling cascades and to translate the in vitro findings into preclinical models.
Collapse
Affiliation(s)
- Gabriel Leonardo Magrin
- Department of Oral Biology, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (G.L.M.); (F.J.S.)
- Department of Dentistry, Center for Education and Research on Dental Implants, Federal University of Santa Catarina, Campus Reitor João David Ferreira Lima s/n, Florianopolis 88040-900, Brazil;
| | - Franz Josef Strauss
- Department of Oral Biology, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (G.L.M.); (F.J.S.)
- Department of Conservative Dentistry, Faculty of Dentistry, University of Chile, Av. Sergio Livingstone 943, Santiago 7500566, Chile
| | - Cesar Augusto Magalhães Benfatti
- Department of Dentistry, Center for Education and Research on Dental Implants, Federal University of Santa Catarina, Campus Reitor João David Ferreira Lima s/n, Florianopolis 88040-900, Brazil;
| | - Lucianne Cople Maia
- Department of Pediatric Dentistry and Orthodontics, Federal University of Rio de Janeiro, Rua Prof. Rodolpho Paulo Rocco 325, Rio de Janeiro 21941-617, Brazil;
| | - Reinhard Gruber
- Department of Oral Biology, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (G.L.M.); (F.J.S.)
- Correspondence:
| |
Collapse
|
15
|
Llama‐Palacios A, Potupa O, Sánchez MC, Figuero E, Herrera D, Sanz M. Proteomic analysis ofFusobacterium nucleatumgrowth in biofilm versus planktonic state. Mol Oral Microbiol 2020; 35:168-180. [DOI: 10.1111/omi.12303] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 06/01/2020] [Accepted: 06/11/2020] [Indexed: 01/04/2023]
Affiliation(s)
- Arancha Llama‐Palacios
- Oral Microbiology Laboratory at the Faculty of Odontology University Complutense Madrid Spain
- ETEP (Etiology and Therapy of Periodontal Diseases) Research Group University Complutense Madrid Spain
| | - Oksana Potupa
- Oral Microbiology Laboratory at the Faculty of Odontology University Complutense Madrid Spain
| | - María C. Sánchez
- Oral Microbiology Laboratory at the Faculty of Odontology University Complutense Madrid Spain
- ETEP (Etiology and Therapy of Periodontal Diseases) Research Group University Complutense Madrid Spain
| | - Elena Figuero
- ETEP (Etiology and Therapy of Periodontal Diseases) Research Group University Complutense Madrid Spain
| | - David Herrera
- ETEP (Etiology and Therapy of Periodontal Diseases) Research Group University Complutense Madrid Spain
| | - Mariano Sanz
- ETEP (Etiology and Therapy of Periodontal Diseases) Research Group University Complutense Madrid Spain
| |
Collapse
|
16
|
Magrin GL, Di Summa F, Strauss FJ, Panahipour L, Mildner M, Magalhães Benfatti CA, Gruber R. Butyrate Decreases ICAM-1 Expression in Human Oral Squamous Cell Carcinoma Cells. Int J Mol Sci 2020; 21:ijms21051679. [PMID: 32121422 PMCID: PMC7084181 DOI: 10.3390/ijms21051679] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/20/2020] [Accepted: 02/27/2020] [Indexed: 12/15/2022] Open
Abstract
Short-chain fatty acids (SCFA) are bacterial metabolites that can be found in periodontal pockets. The expression of adhesion molecules such as intercellular adhesion molecule-1 (ICAM-1) within the epithelium pocket is considered to be a key event for the selective transmigration of leucocytes towards the gingival sulcus. However, the impact of SCFA on ICAM-1 expression by oral epithelial cells remains unclear. We therefore exposed the oral squamous carcinoma cell line HSC-2, primary oral epithelial cells and human gingival fibroblasts to SCFA, namely acetate, propionate and butyrate, and stimulated with known inducers of ICAM-1 such as interleukin-1-beta (IL1β) and tumor necrosis factor-alfa (TNFα). We report here that butyrate but not acetate or propionate significantly suppressed the cytokine-induced ICAM-1 expression in HSC-2 epithelial cells and primary epithelial cells. The G-protein coupled receptor-43 (GPR43/ FFAR2) agonist but not the histone deacetylase inhibitor, trichostatin A, mimicked the butyrate effects. Butyrate also attenuated the nuclear translocation of p65 into the nucleus on HSC-2 cells. The decrease of ICAM-1 was independent of Nrf2/HO-1 signaling and phosphorylation of JNK and p38. Nevertheless, butyrate could not reverse an ongoing cytokine-induced ICAM-1 expression in HSC-2 cells. Overall, these observations suggest that butyrate can attenuate cytokine-induced ICAM-1 expression in cells with epithelial origin.
Collapse
Affiliation(s)
- Gabriel Leonardo Magrin
- Department of Oral Biology, School of Dentistry, Medical University of Vienna, Sensengasse 2a, Vienna 1090, Austria; (G.L.M.); (F.D.S.); (F.-J.S.); (L.P.)
- Center for Education and Research on Dental Implants (CEPID), Department of Dentistry, School of Dentistry, Federal University of Santa Catarina, Campus Reitor João David Ferreira Lima s/n, Florianopolis – SC 88040-900, Brazil;
| | - Francesca Di Summa
- Department of Oral Biology, School of Dentistry, Medical University of Vienna, Sensengasse 2a, Vienna 1090, Austria; (G.L.M.); (F.D.S.); (F.-J.S.); (L.P.)
| | - Franz-Josef Strauss
- Department of Oral Biology, School of Dentistry, Medical University of Vienna, Sensengasse 2a, Vienna 1090, Austria; (G.L.M.); (F.D.S.); (F.-J.S.); (L.P.)
- Department of Conservative Dentistry, School of Dentistry, University of Chile, Av. Sergio Livingstone 943, Santiago 7500566, Chile
- Clinic of Reconstructive Dentistry, University of Zurich, 8032 Zurich, Switzerland
| | - Layla Panahipour
- Department of Oral Biology, School of Dentistry, Medical University of Vienna, Sensengasse 2a, Vienna 1090, Austria; (G.L.M.); (F.D.S.); (F.-J.S.); (L.P.)
| | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, Spitalgasse 23, Vienna 1090, Austria;
| | - Cesar Augusto Magalhães Benfatti
- Center for Education and Research on Dental Implants (CEPID), Department of Dentistry, School of Dentistry, Federal University of Santa Catarina, Campus Reitor João David Ferreira Lima s/n, Florianopolis – SC 88040-900, Brazil;
| | - Reinhard Gruber
- Department of Oral Biology, School of Dentistry, Medical University of Vienna, Sensengasse 2a, Vienna 1090, Austria; (G.L.M.); (F.D.S.); (F.-J.S.); (L.P.)
- Department of Periodontology, University Bern, Hochschulstrasse 4, 3012 Bern, Switzerland
- Correspondence:
| |
Collapse
|
17
|
Liu J, Wang Y, Meng H, Yu J, Lu H, Li W, Lu R, Zhao Y, Li Q, Su L. Butyrate rather than LPS subverts gingival epithelial homeostasis by downregulation of intercellular junctions and triggering pyroptosis. J Clin Periodontol 2019; 46:894-907. [PMID: 31241781 DOI: 10.1111/jcpe.13162] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 06/16/2019] [Accepted: 06/23/2019] [Indexed: 12/20/2022]
Abstract
AIM To investigate the effects of sodium butyrate (NaB) and lipopolysaccharide (LPS) on gingival epithelial barrier. MATERIAL AND METHODS We cultured human primary gingival epithelial cells and investigated the effects of NaB and LPS on gingival epithelial barrier and involved mechanisms at in vitro and in vivo levels by immunostaining, confocal microscopy, field emission scanning electron microscopy (FE-SEM), transmission electronic microscopy (TEM), transepithelial electrical resistance (TEER), FTIC-dextran flux, flow cytometry, real-time PCR and Western blot assays. RESULTS Our results showed that NaB, rather than LPS, destroyed the epithelial barrier by breaking down cell-cell junctions and triggering gingival epithelial cell pyroptosis with characteristic morphological changes, including swollen cells, large bubbles, pore formation in the plasma membrane and subcellular organelles changes. The upregulated expression of pyroptosis-related markers, caspase-3 and gasdermin-E (GSDME) contributed to this effect. Pyroptosis aroused by NaB is a pro-inflammatory cell death. Pyroptotic cell death provoked inflammatory responses by upregulation of IL-8 and MCP-1, and releasing intracellular contents into the extracellular microenvironment after pyroptotic rupture of the plasma membrane. CONCLUSIONS Our new findings indicate that butyrate is a potent destructive factor of gingival epithelial barrier and pro-inflammatory mediator, which shed a new light on our understanding of periodontitis initiation.
Collapse
Affiliation(s)
- Juan Liu
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yixiang Wang
- Central Laboratory, Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| | - Huanxin Meng
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Jingting Yu
- Department of General Dentistry II, Peking University School and Hospital of Stomatology, Beijing, China
| | - Hongye Lu
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Wenjing Li
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Ruifang Lu
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yibing Zhao
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Qiqiang Li
- Department of Periodontology, Capital Medical University School of Stomatology, Beijing, China
| | - Li Su
- Center of Medical and Health Analysis, Peking University, Beijing, China
| |
Collapse
|
18
|
Miller DP, Fitzsimonds ZR, Lamont RJ. Metabolic Signaling and Spatial Interactions in the Oral Polymicrobial Community. J Dent Res 2019; 98:1308-1314. [PMID: 31356756 DOI: 10.1177/0022034519866440] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Oral supra- and subgingival biofilms are complex communities in which hundreds of bacteria, viruses, and fungi reside and interact. In these social environments, microbes compete and cooperate for resources, such as living space and nutrients. The metabolic activities of bacteria can transform their microenvironment and dynamically influence the fitness and growth of cohabitating organisms. Biofilm communities are temporally and spatially organized largely due to cell-to-cell communication, which promotes synergistic interactions. Metabolic interactions maintain biofilm homeostasis through mutualistic cross-feeding, metabolic syntrophy, and cross-respiration. These interactions include reciprocal metabolite exchanges that promote the growth of physiologically compatible bacteria, processive catabolism of complex substrates, and unidirectional interactions that are globally important for the polymicrobial community. Additionally, oral bacterial interactions can lead to detoxification of oxidative compounds, which will provide protection to the community at large. It has also been established that specific organisms provide terminal electron acceptors to partner species that result in a shift from fermentation to respiration, thus increasing ATP yields and improving fitness. Indeed, many interspecies relationships are multidimensional, and the net outcome can be spatially and temporally dependent. Cross-kingdom interactions also occur as oral yeast are antagonistic to some oral bacteria, while numerous mutualistic interactions contribute to yeast-bacterial colonization, fitness in the oral community, and the pathogenesis of caries. Consideration of this social environment reveals behaviors and phenotypes that are not apparent through the study of microbes in isolation. Here, we provide a comprehensive overview of the metabolic interactions that shape the oral microbial community.
Collapse
Affiliation(s)
- D P Miller
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, KY, USA
| | - Z R Fitzsimonds
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, KY, USA
| | - R J Lamont
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, KY, USA
| |
Collapse
|
19
|
Zeller I, Malovichko MV, Hurst HE, Renaud DE, Scott DA. Cigarette smoke reduces short chain fatty acid production by a Porphyromonas gingivalis clinical isolate. J Periodontal Res 2019; 54:566-571. [PMID: 30982987 PMCID: PMC6776670 DOI: 10.1111/jre.12660] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 02/13/2019] [Accepted: 03/18/2019] [Indexed: 01/13/2023]
Abstract
Objectives We hypothesized that short chain fatty acid (SCFA) production by oral pathogens is suppressed by exposure to cigarette smoke extract (CSE). Background Tobacco smoking is a major risk factor for plaque‐induced periodontal diseases. Despite increased disease susceptibility, overt oral inflammation is suppressed in smokers, presenting a diagnostic conundrum. Bacterial‐derived SCFAs can penetrate into oral tissues where they influence multiple components of immune and healing responses. Indeed, the SCFA burden has been correlated with the inflammatory condition of the gingiva. However, the influence of cigarette consumption on SCFA production is unknown. Methods GC/MS was employed to monitor the production of several SCFAs (propionic acid, isobutyric acid, butyric acid, and isovaleric acid) by representative anaerobic oral pathogens (Filifactor alocis 35896, Fusobacterium nucleatum 25586, Porphyromonas gingivalis 33277) that were exposed, or not, to a physiologically relevant dose of CSE (2000 ng/ml nicotine equivalents) generated from 3R4F reference cigarettes. Results The growth of all three bacterial species was unaffected by CSE. The capacity to produce SCFAs by these bacteria was highly varied. F alocis produced the highest concentration of a specific SCFA (butyrate); P gingivalis provided the most robust overall SCFA signal, while F alocis and F nucleatum did not release detectable levels of isobutyrate or isovalerate. As P gingivalis 33277 was the broadest SCFA producer, three low‐passage clinical isolates (10208C, 5607, and 10512) were also examined. Compared to unconditioned microbes, reduced SCFA release was apparent in CSE‐exposed low‐passage clinical isolates of P gingivalis which reached significance for one of the three isolates (propionic, isobutyric, butyric, and isovaleric acids, all P < 0.05). Conclusions There is high disparity in the SCFA profiles of variant chronic periodontitis‐associated bacteria, while CSE exposure reduces SCFA production by a specific clinical strain of P gingivalis. If the latter phenomenon occurs in vivo, a reduced SCFA burden may help explain the reduced vascular response to dental plaque in tobacco smokers.
Collapse
Affiliation(s)
- Iris Zeller
- Departments of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, Kentucky
| | - Marina V Malovichko
- University of Louisville Superfund Research Center and Envirome Institute, University of Louisville, Louisville, Kentucky.,American Heart Association Tobacco Regulatory Science and Addiction Center, University of Louisville, Louisville, Kentucky
| | - Harrell E Hurst
- Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
| | - Diane E Renaud
- Departments of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, Kentucky
| | - David A Scott
- Departments of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, Kentucky
| |
Collapse
|
20
|
Abstract
Over half of adults experience gingivitis, a mild yet treatable form of periodontal disease caused by the overgrowth of oral microbes. Left untreated, gingivitis can progress to a more severe and irreversible disease, most commonly chronic periodontitis. While periodontal diseases are associated with a shift in the oral microbiota composition, it remains unclear how this shift impacts microbiota function early in disease progression. Here, we analyzed the transition from health to gingivitis through both 16S v4-v5 rRNA amplicon and metatranscriptome sequencing of subgingival plaque samples from individuals undergoing an experimental gingivitis treatment. Beta-diversity analysis of 16S rRNA reveals that samples cluster based on disease severity and patient but not by oral hygiene status. Significant shifts in the abundance of several genera occurred during disease transition, suggesting a dysbiosis due to development of gingivitis. Comparing taxonomic abundance with transcriptomic activity revealed concordance of bacterial diversity composition between the two quantification assays in samples originating from both healthy and diseased teeth. Metatranscriptome sequencing analysis indicates that during the early stages of transition to gingivitis, a number of virulence-related transcripts were significantly differentially expressed in individual and across pooled patient samples. Upregulated genes include those involved in proteolytic and nucleolytic processes, while expression levels of those involved in surface structure assembly and other general virulence functions leading to colonization or adaptation within the host are more dynamic. These findings help characterize the transition from health to periodontal disease and identify genes associated with early disease. Although more than 50% of adults have some form of periodontal disease, there remains a significant gap in our understanding of its underlying cause. We initiated this study in order to better characterize the progression from oral health to disease. We first analyzed changes in the abundances of specific microorganisms in dental plaque collected from teeth during health and gingivitis, the mildest form of periodontal disease. We found that the clinical score of disease and patient from whom the sample originated but not tooth brushing are significantly correlated with microbial community composition. While a number of virulence-related gene transcripts are differentially expressed in gingivitis samples relative to health, not all are increased, suggesting that the overall activity of the microbiota is dynamic during disease transition. Better understanding of which microbes are present and their function during early periodontal disease can potentially lead to more targeted prophylactic approaches to prevent disease progression.
Collapse
|
21
|
Sato M, Yoshida Y, Nagano K, Hasegawa Y, Takebe J, Yoshimura F. Three CoA Transferases Involved in the Production of Short Chain Fatty Acids in Porphyromonas gingivalis. Front Microbiol 2016; 7:1146. [PMID: 27486457 PMCID: PMC4949257 DOI: 10.3389/fmicb.2016.01146] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 07/08/2016] [Indexed: 12/11/2022] Open
Abstract
Butyryl-CoA:acetate CoA transferase, which produces butyrate and acetyl-CoA from butyryl-CoA and acetate, is responsible for the final step of butyrate production in bacteria. This study demonstrates that in the periodontopathogenic bacterium Porphyromonas gingivalis this reaction is not catalyzed by PGN_1171, previously annotated as butyryl-CoA:acetate CoA transferase, but by three distinct CoA transferases, PGN_0725, PGN_1341, and PGN_1888. Gas chromatography/mass spectrometry (GC-MS) and spectrophotometric analyses were performed using crude enzyme extracts from deletion mutant strains and purified recombinant proteins. The experiments revealed that, in the presence of acetate, PGN_0725 preferentially utilized butyryl-CoA rather than propionyl-CoA. By contrast, this preference was reversed in PGN_1888. The only butyryl-CoA:acetate CoA transferase activity was observed in PGN_1341. Double reciprocal plots revealed that all the reactions catalyzed by these enzymes follow a ternary-complex mechanism, in contrast to previously characterized CoA transferases. GC-MS analysis to determine the concentrations of short chain fatty acids (SCFAs) in culture supernatants of P. gingivalis wild type and mutant strains revealed that PGN_0725 and PGN_1888 play a major role in the production of butyrate and propionate, respectively. Interestingly, a triple deletion mutant lacking PGN_0725, PGN_1341, and PGN_1888 produced low levels of SCFAs, suggesting that the microorganism contains CoA transferase(s) in addition to these three enzymes. Growth rates of the mutant strains were mostly slower than that of the wild type, indicating that many carbon compounds produced in the SCFA synthesis appear to be important for the biological activity of this microorganism.
Collapse
Affiliation(s)
- Mitsunari Sato
- Department of Microbiology, School of Dentistry, Aichi Gakuin UniversityNagoya, Japan; Department of Removable Prosthodontics, School of Dentistry, Aichi Gakuin UniversityNagoya, Japan
| | - Yasuo Yoshida
- Department of Microbiology, School of Dentistry, Aichi Gakuin University Nagoya, Japan
| | - Keiji Nagano
- Department of Microbiology, School of Dentistry, Aichi Gakuin University Nagoya, Japan
| | - Yoshiaki Hasegawa
- Department of Microbiology, School of Dentistry, Aichi Gakuin University Nagoya, Japan
| | - Jun Takebe
- Department of Removable Prosthodontics, School of Dentistry, Aichi Gakuin University Nagoya, Japan
| | - Fuminobu Yoshimura
- Department of Microbiology, School of Dentistry, Aichi Gakuin University Nagoya, Japan
| |
Collapse
|
22
|
Nemoto TK, Ohara-Nemoto Y, Bezerra GA, Shimoyama Y, Kimura S. A Porphyromonas gingivalis Periplasmic Novel Exopeptidase, Acylpeptidyl Oligopeptidase, Releases N-Acylated Di- and Tripeptides from Oligopeptides. J Biol Chem 2016; 291:5913-5925. [PMID: 26733202 PMCID: PMC4786725 DOI: 10.1074/jbc.m115.687566] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 12/06/2015] [Indexed: 01/01/2023] Open
Abstract
Exopeptidases, including dipeptidyl- and tripeptidylpeptidase, are crucial for the growth of Porphyromonas gingivalis, a periodontopathic asaccharolytic bacterium that incorporates amino acids mainly as di- and tripeptides. In this study, we identified a novel exopeptidase, designated acylpeptidyl oligopeptidase (AOP), composed of 759 amino acid residues with active Ser(615) and encoded by PGN_1349 in P. gingivalis ATCC 33277. AOP is currently listed as an unassigned S9 family peptidase or prolyl oligopeptidase. Recombinant AOP did not hydrolyze a Pro-Xaa bond. In addition, although sequence similarities to human and archaea-type acylaminoacyl peptidase sequences were observed, its enzymatic properties were apparently distinct from those, because AOP scarcely released an N-acyl-amino acid as compared with di- and tripeptides, especially with N-terminal modification. The kcat/Km value against benzyloxycarbonyl-Val-Lys-Met-4-methycoumaryl-7-amide, the most potent substrate, was 123.3 ± 17.3 μm(-1) s(-1), optimal pH was 7-8.5, and the activity was decreased with increased NaCl concentrations. AOP existed predominantly in the periplasmic fraction as a monomer, whereas equilibrium between monomers and oligomers was observed with a recombinant molecule, suggesting a tendency of oligomerization mediated by the N-terminal region (Met(16)-Glu(101)). Three-dimensional modeling revealed the three domain structures (residues Met(16)-Ala(126), which has no similar homologue with known structure; residues Leu(127)-Met(495) (β-propeller domain); and residues Ala(496)-Phe(736) (α/β-hydrolase domain)) and further indicated the hydrophobic S1 site of AOP in accord with its hydrophobic P1 preference. AOP orthologues are widely distributed in bacteria, archaea, and eukaryotes, suggesting its importance for processing of nutritional and/or bioactive oligopeptides.
Collapse
Affiliation(s)
- Takayuki K Nemoto
- From the Department of Oral Molecular Biology, Course of Medical and Dental Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Yuko Ohara-Nemoto
- From the Department of Oral Molecular Biology, Course of Medical and Dental Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Gustavo Arruda Bezerra
- the Department of Structural and Computational Biology, Max F. Perutz Laboratories, University of Vienna, Campus Vienna Biocenter 5, 1030 Vienna, Austria, and.
| | - Yu Shimoyama
- the Division of Molecular Microbiology, Iwate Medical University, Iwate 028-3694, Japan
| | - Shigenobu Kimura
- the Division of Molecular Microbiology, Iwate Medical University, Iwate 028-3694, Japan
| |
Collapse
|
23
|
Nemoto TK, Ohara-Nemoto Y. Exopeptidases and gingipains in Porphyromonas gingivalis as prerequisites for its amino acid metabolism. JAPANESE DENTAL SCIENCE REVIEW 2016; 52:22-29. [PMID: 28408952 PMCID: PMC5382784 DOI: 10.1016/j.jdsr.2015.08.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 08/20/2015] [Accepted: 08/20/2015] [Indexed: 01/22/2023] Open
Abstract
Porphyromonas gingivalis, an asaccharolytic bacterium, utilizes amino acids as energy and carbon sources. Since amino acids are incorporated into the bacterial cells mainly as di- and tri-peptides, exopeptidases including dipeptidyl-peptidase (DPP) and tripeptidyl-peptidase are considered to be prerequisite components for their metabolism. We recently discovered DPP11, DPP5, and acylpeptidyl oligopeptidase in addition to previously reported DPP4, DPP7, and prolyl tripeptidyl peptidase A. DPP11 is a novel enzyme specific for acidic P1 residues (Asp and Glu) and distributed ubiquitously in eubacteria, while DPP5 is preferential for the hydrophobic P1 residue and the first entity identified in prokaryotes. Recently, acylpeptidyl oligopeptidase with a preference for hydrophobic P1 residues was found to release N-terminally blocked di- and tri-peptides. Furthermore, we also demonstrated that gingipains R and K contribute to P1-basic dipeptide production. These observations implicate that most, if not all, combinations of di- and tri-peptides are produced from extracellular oligopeptides even with an N-terminal modification. Here, we review P. gingivalis exopeptidases mainly in regard to their enzymatic characteristics. These exopeptidases with various substrate specificities benefit P. gingivalis for obtaining energy and carbon sources from the nutritionally limited subgingival environment.
Collapse
Affiliation(s)
- Takayuki K. Nemoto
- Department of Oral Molecular Biology, Course of Medical and Dental Sciences, Nagasaki University Graduate School of Biomedical Sciences, Japan
| | | |
Collapse
|
24
|
Intestinal microbiota and ulcerative colitis. J Infect Chemother 2015; 21:761-8. [DOI: 10.1016/j.jiac.2015.07.010] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 06/30/2015] [Accepted: 07/29/2015] [Indexed: 02/06/2023]
|
25
|
May A, Brandt BW, El-Kebir M, Klau GW, Zaura E, Crielaard W, Heringa J, Abeln S. metaModules identifies key functional subnetworks in microbiome-related disease. Bioinformatics 2015; 32:1678-85. [PMID: 26342232 DOI: 10.1093/bioinformatics/btv526] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 09/02/2015] [Indexed: 11/13/2022] Open
Abstract
MOTIVATION The human microbiome plays a key role in health and disease. Thanks to comparative metatranscriptomics, the cellular functions that are deregulated by the microbiome in disease can now be computationally explored. Unlike gene-centric approaches, pathway-based methods provide a systemic view of such functions; however, they typically consider each pathway in isolation and in its entirety. They can therefore overlook the key differences that (i) span multiple pathways, (ii) contain bidirectionally deregulated components, (iii) are confined to a pathway region. To capture these properties, computational methods that reach beyond the scope of predefined pathways are needed. RESULTS By integrating an existing module discovery algorithm into comparative metatranscriptomic analysis, we developed metaModules, a novel computational framework for automated identification of the key functional differences between health- and disease-associated communities. Using this framework, we recovered significantly deregulated subnetworks that were indeed recognized to be involved in two well-studied, microbiome-mediated oral diseases, such as butanoate production in periodontal disease and metabolism of sugar alcohols in dental caries. More importantly, our results indicate that our method can be used for hypothesis generation based on automated discovery of novel, disease-related functional subnetworks, which would otherwise require extensive and laborious manual assessment. AVAILABILITY AND IMPLEMENTATION metaModules is available at https://bitbucket.org/alimay/metamodules/ CONTACT a.may@vu.nl or s.abeln@vu.nl SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Ali May
- Centre for Integrative Bioinformatics VU (IBIVU), VU University Amsterdam, Amsterdam, The Netherlands, Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands, Amsterdam Institute for Molecules Medicines and Systems (AIMMS), VU University Amsterdam, Amsterdam, The Netherlands
| | - Bernd W Brandt
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
| | - Mohammed El-Kebir
- Centre for Integrative Bioinformatics VU (IBIVU), VU University Amsterdam, Amsterdam, The Netherlands, Department of Computer Science and Center for Computational Molecular Biology, Brown University, Providence, USA and Life Sciences, Centre for Mathematics and Computer Science (CWI), Amsterdam, The Netherlands
| | - Gunnar W Klau
- Centre for Integrative Bioinformatics VU (IBIVU), VU University Amsterdam, Amsterdam, The Netherlands, Amsterdam Institute for Molecules Medicines and Systems (AIMMS), VU University Amsterdam, Amsterdam, The Netherlands, Life Sciences, Centre for Mathematics and Computer Science (CWI), Amsterdam, The Netherlands
| | - Egija Zaura
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
| | - Wim Crielaard
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
| | - Jaap Heringa
- Centre for Integrative Bioinformatics VU (IBIVU), VU University Amsterdam, Amsterdam, The Netherlands, Amsterdam Institute for Molecules Medicines and Systems (AIMMS), VU University Amsterdam, Amsterdam, The Netherlands
| | - Sanne Abeln
- Centre for Integrative Bioinformatics VU (IBIVU), VU University Amsterdam, Amsterdam, The Netherlands, Amsterdam Institute for Molecules Medicines and Systems (AIMMS), VU University Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
26
|
Nishimata H, Ohara-Nemoto Y, Baba TT, Hoshino T, Fujiwara T, Shimoyama Y, Kimura S, Nemoto TK. Identification of Dipeptidyl-Peptidase (DPP)5 and DPP7 in Porphyromonas endodontalis, Distinct from Those in Porphyromonas gingivalis. PLoS One 2014; 9:e114221. [PMID: 25494328 PMCID: PMC4262410 DOI: 10.1371/journal.pone.0114221] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 11/04/2014] [Indexed: 11/21/2022] Open
Abstract
Dipeptidyl peptidases (DPPs) that liberate dipeptides from the N-terminal end of oligopeptides are crucial for the growth of Porphyromonas species, anaerobic asaccharolytic gram negative rods that utilize amino acids as energy sources. Porphyromonas endodontalis is a causative agent of periapical lesions with acute symptoms and Asp/Glu-specific DPP11 has been solely characterized in this organism. In this study, we identified and characterized two P. endodontalis DPPs, DPP5 and DPP7. Cell-associated DPP activity toward Lys-Ala-4-methylcoumaryl-7-amide (MCA) was prominent in P. endodontalis ATCC 35406 as compared with the Porphyromonas gingivalis strains ATCC 33277, 16-1, HW24D1, ATCC 49417, W83, W50, and HNA99. The level of hydrolysis of Leu-Asp-MCA by DPP11, Gly-Pro-MCA by DPP4, and Met-Leu-MCA was also higher than in the P. gingivalis strains. MER236725 and MER278904 are P. endodontalis proteins belong to the S9- and S46-family peptidases, respectively. Recombinant MER236725 exhibited enzymatic properties including substrate specificity, and salt- and pH-dependence similar to P. gingivalis DPP5 belonging to the S9 family. However, the kcat/Km figure (194 µM-1·sec-1) for the most potent substrate (Lys-Ala-MCA) was 18.4-fold higher as compared to the P. gingivalis entity (10.5 µM-1·sec-1). In addition, P. endodontalis DPP5 mRNA and protein contents were increased several fold as compared with those in P. gingivalis. Recombinant MER278904 preferentially hydrolyzed Met-Leu-MCA and exhibited a substrate specificity similar to P. gingivalis DPP7 belonging to the S46 family. In accord with the deduced molecular mass of 818 amino acids, a 105-kDa band was immunologically detected, indicating that P. endodontalis DPP7 is an exceptionally large molecule in the DPP7/DPP11/S46 peptidase family. The enhancement of four DPP activities was conclusively demonstrated in P. endodontalis, and remarkable Lys-Ala-MCA-hydrolysis was achieved by qualitative and quantitative potentiation of the DPP5 molecule.
Collapse
Affiliation(s)
- Haruka Nishimata
- Department of Oral Molecular Biology, Course of Medical and Dental Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Pediatric Dentistry, Course of Medical and Dental Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yuko Ohara-Nemoto
- Department of Oral Molecular Biology, Course of Medical and Dental Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Tomomi T. Baba
- Department of Oral Molecular Biology, Course of Medical and Dental Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Tomonori Hoshino
- Department of Pediatric Dentistry, Course of Medical and Dental Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Taku Fujiwara
- Department of Pediatric Dentistry, Course of Medical and Dental Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yu Shimoyama
- Division of Molecular Microbiology, Iwate Medical University, Yahaba-cho, Iwate, Japan
| | - Shigenobu Kimura
- Division of Molecular Microbiology, Iwate Medical University, Yahaba-cho, Iwate, Japan
| | - Takayuki K. Nemoto
- Department of Oral Molecular Biology, Course of Medical and Dental Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
27
|
Hendrickson EL, Wang T, Beck DAC, Dickinson BC, Wright CJ, J Lamont R, Hackett M. Proteomics of Fusobacterium nucleatum within a model developing oral microbial community. Microbiologyopen 2014; 3:729-51. [PMID: 25155235 PMCID: PMC4234264 DOI: 10.1002/mbo3.204] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 07/01/2014] [Accepted: 07/08/2014] [Indexed: 11/29/2022] Open
Abstract
Fusobacterium nucleatum is a common oral organism that can provide adhesive and metabolic support to developing periodontal bacterial communities. It is within the context of these communities that disease occurs. We have previously reported whole cell proteomics analyses of Porphyromonas gingivalis and Streptococcus gordonii in early-stage communities with each other and with F. nucleatum, modeled using 18 h pellets. Here, we report the adaptation of F. nucleatum to the same experimental conditions as measured by differential protein expression. About 1210 F. nucleatum proteins were detected in single species F. nucleatum control samples, 1192 in communities with P. gingivalis, 1224 with S. gordonii, and 1135 with all three species. Quantitative comparisons among the proteomes revealed important changes in all mixed samples with distinct responses to P. gingivalis or S. gordonii alone and in combination. The results were inspected manually and an ontology analysis conducted using DAVID (Database for annotation, visualization, and integrated discovery). Extensive changes were detected in energy metabolism. All multispecies comparisons showed reductions in amino acid fermentation and a shift toward butanoate as a metabolic byproduct, although the two organism model community with S. gordonii showed increases in alanine, threonine, methionine, and cysteine pathways, and in the three species samples there were increases in lysine and methionine. The communities with P. gingivalis or all three organisms showed reduced glycolysis proteins, but F. nucleatum paired with S. gordonii displayed increased glycolysis/gluconeogenesis proteins. The S. gordonii containing two organism model also showed increases in the ethanolamine pathway while the three species sample showed decreases relative to the F. nucleatum single organism control. All of the nascent model communities displayed reduced translation, lipopolysaccharide, and cell wall biosynthesis, DNA replication and DNA repair.
Collapse
Affiliation(s)
- Erik L Hendrickson
- Department of Chemical Engineering and Center for Microbial Proteomics, University of Washington, Box 355014, Seattle, Washington, 98195
| | | | | | | | | | | | | |
Collapse
|
28
|
Belibasakis G, Thurnheer T, Bostanci N. Porphyromonas gingivalis: a heartful oral pathogen? Virulence 2014; 5:463-4. [PMID: 24759693 PMCID: PMC4063808 DOI: 10.4161/viru.28930] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 04/16/2014] [Indexed: 12/17/2022] Open
Affiliation(s)
- Georgios Belibasakis
- Oral Microbiology and Immunology; Institute of Oral Biology; Center of Dental Medicine; University of Zürich; Zürich, Switzerland
| | - Thomas Thurnheer
- Oral Microbiology and Immunology; Institute of Oral Biology; Center of Dental Medicine; University of Zürich; Zürich, Switzerland
| | - Nagihan Bostanci
- Oral Translational Research; Institute of Oral Biology; Center of Dental Medicine; University of Zürich; Zürich, Switzerland
| |
Collapse
|
29
|
Activation and repression of Epstein-Barr Virus and Kaposi's sarcoma-associated herpesvirus lytic cycles by short- and medium-chain fatty acids. J Virol 2014; 88:8028-44. [PMID: 24807711 DOI: 10.1128/jvi.00722-14] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The lytic cycles of Epstein-Barr virus (EBV) and Kaposi's sarcoma-associated herpesvirus (KSHV) are induced in cell culture by sodium butyrate (NaB), a short-chain fatty acid (SCFA) histone deacetylase (HDAC) inhibitor. Valproic acid (VPA), another SCFA and an HDAC inhibitor, induces the lytic cycle of KSHV but blocks EBV lytic reactivation. To explore the hypothesis that structural differences between NaB and VPA account for their functional effects on the two related viruses, we investigated the capacity of 16 structurally related short- and medium-chain fatty acids to promote or prevent lytic cycle reactivation. SCFAs differentially affected EBV and KSHV reactivation. KSHV was reactivated by all SCFAs that are HDAC inhibitors, including phenylbutyrate. However, several fatty acid HDAC inhibitors, such as isobutyrate and phenylbutyrate, did not reactivate EBV. Reactivation of KSHV lytic transcripts could not be blocked completely by any fatty acid tested. In contrast, several medium-chain fatty acids inhibited lytic activation of EBV. Fatty acids that blocked EBV reactivation were more lipophilic than those that activated EBV. VPA blocked activation of the BZLF1 promoter by NaB but did not block the transcriptional function of ZEBRA. VPA also blocked activation of the DNA damage response that accompanies EBV lytic cycle activation. Properties of SCFAs in addition to their effects on chromatin are likely to explain activation or repression of EBV. We concluded that fatty acids stimulate the two related human gammaherpesviruses to enter the lytic cycle through different pathways. Importance: Lytic reactivation of EBV and KSHV is needed for persistence of these viruses and plays a role in carcinogenesis. Our direct comparison highlights the mechanistic differences in lytic reactivation between related human oncogenic gammaherpesviruses. Our findings have therapeutic implications, as fatty acids are found in the diet and produced by the human microbiota. Small-molecule inducers of the lytic cycle are desired for oncolytic therapy. Inhibition of viral reactivation, alternatively, may prove useful in cancer treatment. Overall, our findings contribute to the understanding of pathways that control the latent-to-lytic switch and identify naturally occurring molecules that may regulate this process.
Collapse
|
30
|
Mazumdar V, Amar S, Segrè D. Metabolic proximity in the order of colonization of a microbial community. PLoS One 2013; 8:e77617. [PMID: 24204896 PMCID: PMC3813667 DOI: 10.1371/journal.pone.0077617] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Accepted: 09/03/2013] [Indexed: 01/25/2023] Open
Abstract
Microbial biofilms are often composed of multiple bacterial species that accumulate by adhering to a surface and to each other. Biofilms can be resistant to antibiotics and physical stresses, posing unresolved challenges in the fight against infectious diseases. It has been suggested that early colonizers of certain biofilms could cause local environmental changes, favoring the aggregation of subsequent organisms. Here we ask whether the enzyme content of different microbes in a well-characterized dental biofilm can be used to predict their order of colonization. We define a metabolic distance between different species, based on the overlap in their enzyme content. We next use this metric to quantify the average metabolic distance between neighboring organisms in the biofilm. We find that this distance is significantly smaller than the one observed for a random choice of prokaryotes, probably reflecting the environmental constraints on metabolic function of the community. More surprisingly, this metabolic metric is able to discriminate between observed and randomized orders of colonization of the biofilm, with the observed orders displaying smaller metabolic distance than randomized ones. By complementing these results with the analysis of individual vs. joint metabolic networks, we find that the tendency towards minimal metabolic distance may be counter-balanced by a propensity to pair organisms with maximal joint potential for synergistic interactions. The trade-off between these two tendencies may create a "sweet spot" of optimal inter-organism distance, with possible broad implications for our understanding of microbial community organization.
Collapse
Affiliation(s)
- Varun Mazumdar
- Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
| | - Salomon Amar
- Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
- Center for Anti-Inflammatory Therapeutics; Boston University Goldman School of Dental Medicine, Boston, Massachusetts, United States of America
| | - Daniel Segrè
- Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
- Department of Biology and Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
| |
Collapse
|
31
|
Butyrate stimulates the early process of the osteogenic differentiation but inhibits the biomineralization in dental follicle cells (DFCs). Odontology 2013; 102:154-9. [PMID: 23836050 DOI: 10.1007/s10266-013-0117-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 05/01/2013] [Indexed: 10/26/2022]
Abstract
Dental stem cells, especially dental follicle cells (DFCs) as precursor cells for the periodontium have interesting prospects for regenerative dentistry. During periodontitis, butyrate as a bacterial metabolite and inflammatory agent is often found in millimolar concentrations in periodontal pockets. This study evaluates the effects of butyrate on the proliferation and osteogenic differentiation of DFCs. We assessed cell viability/proliferation (BCA assay) and osteogenic differentiation (ALP activity, alizarin staining and RT PCR) of DFCs in vitro after butyrate supplementation. Butyrate concentrations of 20 mM or higher are toxic for DFCs. At a non-toxic concentration, butyrate promotes the expression of alkaline phosphatase and collagen type-1 but inhibits the formation of calcified nodules and the induction of RUNX2 and osteocalcin under osteogenic differentiation conditions. In conclusion, DFCs are resistant to physiological high concentrations of butyrate. Butyrate facilitates the osteogenic differentiation of DFCs in early stages but inhibits calcification at later stages of the differentiation process.
Collapse
|
32
|
Host-bacteria crosstalk at the dentogingival junction. Int J Dent 2012; 2012:821383. [PMID: 22899931 PMCID: PMC3412119 DOI: 10.1155/2012/821383] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 05/22/2012] [Accepted: 05/22/2012] [Indexed: 12/21/2022] Open
Abstract
The dentogingival junction is of crucial importance in periodontal host defense both structurally and functionally. Oral bacteria exert a constant challenge to the host cells and tissues at the dentogingival junction. The host response is set up to eliminate the pathogens by the innate and adaptive defense mechanisms. In health, the commensal bacteria and the host defense mechanisms are in a dynamic steady state. During periodontal disease progression, the dental bacterial plaque, junctional epithelium (JE), inflammatory cells, connective tissue, and bone all go through a series of changes. The tissue homeostasis is turned into tissue destruction and progression of periodontitis. The classical study of Slots showed that in the bacterial plaque, the most remarkable change is the shift from gram-positive aerobic and facultatively anaerobic flora to a predominantly gram-negative and anaerobic flora. This has been later confirmed by several other studies. Furthermore, not only the shift of the bacterial flora to a more pathogenic one, but also bacterial growth as a biofilm on the tooth surface, allows the bacteria to communicate with each other and exert their virulence aimed at favoring their growth. This paper focuses on host-bacteria crosstalk at the dentogingival junction and the models studying it in vitro.
Collapse
|
33
|
Thorn RMS, Greenman J. Microbial volatile compounds in health and disease conditions. J Breath Res 2012; 6:024001. [PMID: 22556190 PMCID: PMC7106765 DOI: 10.1088/1752-7155/6/2/024001] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Accepted: 04/12/2012] [Indexed: 12/24/2022]
Abstract
Microbial cultures and/or microbial associated diseases often have a characteristic smell. Volatile organic compounds (VOCs) are produced by all microorganisms as part of their normal metabolism. The types and classes of VOC produced is wide, including fatty acids and their derivatives (e.g. hydrocarbons, aliphatic alcohols and ketones), aromatic compounds, nitrogen containing compounds, and volatile sulfur compounds. A diversity of ecological niches exist in the human body which can support a polymicrobial community, with the exact VOC profile of a given anatomical site being dependent on that produced by both the host component and the microbial species present. The detection of VOCs is of interest to various disciplines, hence numerous analytical approaches have been developed to accurately characterize and measure VOCs in the laboratory, often from patient derived samples. Using these technological advancements it is evident that VOCs are indicative of both health and disease states. Many of these techniques are still largely confined to the research laboratory, but it is envisaged that in future bedside 'VOC profiling' will enable rapid characterization of microbial associated disease, providing vital information to healthcare practitioners.
Collapse
Affiliation(s)
- Robin Michael Statham Thorn
- Centre for Research in Biomedicine, Department of Applied Sciences, University of the West of England, Bristol, Frenchay Campus, Coldharbour Lane, BS16 1QY, UK
| | - John Greenman
- Centre for Research in Biomedicine, Department of Applied Sciences, University of the West of England, Bristol, Frenchay Campus, Coldharbour Lane, BS16 1QY, UK
| |
Collapse
|
34
|
Qiqiang L, Huanxin M, Xuejun G. Longitudinal study of volatile fatty acids in the gingival crevicular fluid of patients with periodontitis before and after nonsurgical therapy. J Periodontal Res 2012; 47:740-9. [DOI: 10.1111/j.1600-0765.2012.01489.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
35
|
Morozumi A. High concentration of sodium butyrate suppresses osteoblastic differentiation and mineralized nodule formation in ROS17/2.8 cells. J Oral Sci 2012; 53:509-16. [PMID: 22167038 DOI: 10.2334/josnusd.53.509] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Periodontitis is a destructive disease that is likely the result of the activities of different microbial complexes, including anaerobic Gram-negative periodontopathic bacteria. Butyric acid (sodium butyrate; BA) is a major metabolic by-product of anaerobic Gram-negative periodontopathic bacteria present in subgingival plaque. This study was undertaken to examine the effect of BA on the expression of osteogenesis-related transcription factors and mineralized nodule formation in osteoblastic ROS17/2.8 cells. The cells were cultured with 0 (control), 10(-5), 10(-4), or 10(-3) M BA for up to 7 days. The gene and protein expression levels of transcription factors such as Runx2, Osterix, Dlx5, Msx2, and AJ18, as well as extracellular matrix proteins such as bone sialoprotein (BSP) and osteocalcin, were examined using real-time PCR and Western blotting, respectively. Mineralized nodule formation was detected by alizarin red staining. The expression of Runx2, Osterix, Dlx5, and Msx2 decreased significantly in the presence of 10(-3 )M BA compared to the control, whereas AJ18 expression increased significantly. Mineralized nodule formation decreased markedly in the presence of 10(-3) M BA. Alkaline phosphatase activity and the expression of bone sialoprotein and osteocalcin decreased significantly in the presence of 10(-3) M BA compared to the control. These results suggest that 10(-3) M BA suppresses osteoblastic differentiation and mineralized nodule formation in ROS17/2.8 cells.
Collapse
Affiliation(s)
- Akira Morozumi
- Department of Oral Health Sciences, Nihon University School of Dentistry, Tokyo, Japan.
| |
Collapse
|
36
|
Lohinai Z, Keremi B, Szoko E, Tabi T, Szabo C, Tulassay Z, Levine M. Bacterial lysine decarboxylase influences human dental biofilm lysine content, biofilm accumulation, and subclinical gingival inflammation. J Periodontol 2011; 83:1048-56. [PMID: 22141361 DOI: 10.1902/jop.2011.110474] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Dental biofilms contain a protein that inhibits mammalian cell growth, possibly lysine decarboxylase from Eikenella corrodens. This enzyme decarboxylates lysine, an essential amino acid for dentally attached cell turnover in gingival sulci. Lysine depletion may stop this turnover, impairing the barrier to bacterial compounds. The aims of this study are to determine biofilm lysine and cadaverine contents before oral hygiene restriction (OHR) and their association with plaque index (PI) and gingival crevicular fluid (GCF) after OHR for 1 week. METHODS Laser-induced fluorescence after capillary electrophoresis was used to determine lysine and cadaverine contents in dental biofilm, tongue biofilm, and saliva before OHR and in dental biofilm after OHR. RESULTS Before OHR, lysine and cadaverine contents of dental biofilm were similar and 10-fold greater than in saliva or tongue biofilm. After 1 week of OHR, the biofilm content of cadaverine increased and that of lysine decreased, consistent with greater biofilm lysine decarboxylase activity. Regression indicated that PI and GCF exudation were positively related to biofilm lysine after OHR, unless biofilm lysine exceeded the minimal blood plasma content, in which case PI was further increased but GCF exudation was reduced. CONCLUSIONS After OHR, lysine decarboxylase activity seems to determine biofilm lysine content and biofilm accumulation. When biofilm lysine exceeds minimal blood plasma content after OHR, less GCF appeared despite more biofilm. Lysine appears important for biofilm accumulation and the epithelial barrier to bacterial proinflammatory agents. Inhibiting lysine decarboxylase may retard the increased GCF exudation required for microbial development and gingivitis.
Collapse
Affiliation(s)
- Zsolt Lohinai
- Department of Conservative Dentistry, Semmelweis University, Budapest, Hungary
| | | | | | | | | | | | | |
Collapse
|
37
|
Iida T, Kawato T, Tanaka H, Tanabe N, Nakai K, Zhao N, Suzuki N, Ochiai K, Maeno M. Sodium butyrate induces the production of cyclooxygenases and prostaglandin E₂ in ROS 17/2.8 osteoblastic cells. Arch Oral Biol 2011; 56:678-86. [PMID: 21281931 DOI: 10.1016/j.archoralbio.2010.12.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2010] [Revised: 11/22/2010] [Accepted: 12/30/2010] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Sodium butyrate (butyric acid; BA) is a major metabolic by-product of the anaerobic periodontopathic bacteria present in subgingival plaque. We examined the effects of BA and/or indomethacin on cell proliferation, the expression of cyclooxygenases (COXs), prostaglandin (PG) receptors (EP1-4), extracellular matrix proteins, such as type I collagen and osteopontin, and PGE(2) production, using ROS17/2.8 cells as osteoblasts. METHODS The rat clonal cell line ROS 17/2.8 was cultured with 0, 10(-5), 10(-4), and 10(-3)M BA in the presence or absence of 0.5 μM indomethacin, for up to 7 days. The expression of COX-1, COX-2, EP1, EP2, EP3, EP4, type I collagen, and osteopontin was examined at the mRNA and protein levels using real-time PCR and Western blotting, respectively. The amount of PGE(2) in the culture medium was measured by ELISA. RESULTS Proliferation of ROS 17/2.8 cells was not affected by the addition of BA. However, PGE(2) production and the expression of COX-1 and COX-2 increased with the addition of BA. In contrast, indomethacin, an inhibitor of COX, blocked the stimulatory effect of BA. Furthermore, EP2 expression increased with BA treatment, whereas EP1 expression was not affected and the expression of EP3 and EP4 was not detected. The addition of BA also increased the expression of type I collagen and osteopontin. Indomethacin blocked about 50% of the stimulatory effect of BA on type I collagen, whereas it did not block the effect on osteopontin. CONCLUSIONS These results suggest that BA induces PGE(2) production by increasing the expression of COX-1 and COX-2 in osteoblasts, and that an autocrine action of the produced PGE(2), via EP1 or BA-induced EP2, is related to an increase in type I collagen expression by BA.
Collapse
Affiliation(s)
- Takafumi Iida
- Division of Oral Health Sciences, Nihon University Graduate School of Dentistry, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Ochiai K, Imai K, Tamura M, Kurita-Ochiai T. Butyric Acid Effects in the Development of Periodontitis and Systemic Diseases. J Oral Biosci 2011. [DOI: 10.1016/s1349-0079(11)80004-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
39
|
|
40
|
Tsuda H, Ochiai K, Suzuki N, Otsuka K. Butyrate, a bacterial metabolite, induces apoptosis and autophagic cell death in gingival epithelial cells. J Periodontal Res 2010; 45:626-34. [PMID: 20546110 DOI: 10.1111/j.1600-0765.2010.01277.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND OBJECTIVE Butyrate is produced by some types of anaerobic periodontal bacteria. Millimolar concentrations of butyrate are found in mature dental plaque from periodontitis patients. Although butyrate reportedly has a variety of effects in many mammalian cells, its effect on gingival epithelial cells is not well known. In this study, we investigated the effect of butyrate on gingival epithelial Ca9-22 cell death. MATERIAL AND METHODS Death of Ca9-22 cells was assessed after treating the cells with or without butyrate. A SYTOX Green dye, which exhibits strong green fluorescence once it enters dead cells through ruptured cell membranes, was used for cell death detection. Phosphatidylserine redistribution was measured using fluorescein isothiocyanate-labeled annexin V. The activity of caspase-3 was measured as the amount of cleaved substrate peptide. Anti-apoptotic bcl-2 mRNA expression was measured using real-time RT-PCR. Western blotting and fluoromicroscopic analysis with anti-microtubule-associated protein 1 light chain 3 (LC3) antibodies were performed for detection of autophagy. RESULTS Stimulation with millimolar concentrations of butyrate for 48 h induced Ca9-22 cell death. The stimulation also caused increased caspase-3 activity, phosphatidylserine redistribution and bcl-2 down-regulation, suggesting butyrate-induced apoptosis. However, the pan-caspase inhibitor, Z-VAD-FMK, did not inhibit cell death completely. This implies the existence of other types of cell death. In addition, markers of autophagy, namely, the conversion of LC3-I to LC3-II and increased LC3 accumulation, were observed. Moreover, inhibition of autophagy by 3-methyladenine suppressed the butyrate-induced cell death, suggesting that butyrate could induce cell death through autophagy. CONCLUSION These data suggest that butyrate induces apoptosis and autophagic cell death.
Collapse
Affiliation(s)
- H Tsuda
- Department of Biochemistry, Nihon University School of Dentistry, Tokyo, Japan.
| | | | | | | |
Collapse
|
41
|
Application of rpoB and zinc protease gene for use in molecular discrimination of Fusobacterium nucleatum subspecies. J Clin Microbiol 2009; 48:545-53. [PMID: 19955278 DOI: 10.1128/jcm.01631-09] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Fusobacterium nucleatum is classified into five subspecies that inhabit the human oral cavity (F. nucleatum subsp. nucleatum, F. nucleatum subsp. polymorphum, F. nucleatum subsp. fusiforme, F. nucleatum subsp. vincentii, and F. nucleatum subsp. animalis) based on several phenotypic characteristics and DNA-DNA hybridization patterns. However, the methods for detecting or discriminating the clinical isolates of F. nucleatum at the subspecies levels are laborious, expensive, and time-consuming. Therefore, in this study, the nucleotide sequences of the RNA polymerase beta-subunit gene (rpoB) and zinc protease gene were analyzed to discriminate the subspecies of F. nucleatum. The partial sequences of rpoB (approximately 2,419 bp), the zinc protease gene (878 bp), and 16S rRNA genes (approximately 1,500 bp) of the type strains of five subspecies, 28 clinical isolates of F. nucleatum, and 10 strains of F. periodonticum (as a control group) were determined and analyzed. The phylogenetic data showed that the rpoB and zinc protease gene sequences clearly delineated the subspecies of F. nucleatum and provided higher resolution than the 16S rRNA gene sequences in this respect. According to the phylogenetic analysis of rpoB and the zinc protease gene, F. nucleatum subsp. vincentii and F. nucleatum subsp. fusiforme might be classified into a single subspecies. Five clinical isolates could be delineated as a new subspecies of F. nucleatum. The results suggest that rpoB and the zinc protease gene are efficient targets for the discrimination and taxonomic analysis of the subspecies of F. nucleatum.
Collapse
|
42
|
Kinetic characterization and quaternary structure of glutamate racemase from the periodontal anaerobe Fusobacterium nucleatum. Arch Biochem Biophys 2009; 491:16-24. [DOI: 10.1016/j.abb.2009.09.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2009] [Revised: 09/11/2009] [Accepted: 09/15/2009] [Indexed: 11/17/2022]
|
43
|
|
44
|
Pruzzo, Roberta Capretti, Paola Mas C. Short Chain Fatty Acids, Menaquinones and Ubiquinones and Their Effects on the Host. MICROBIAL ECOLOGY IN HEALTH AND DISEASE 2009. [DOI: 10.1080/089106000750060468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
45
|
Scragg MA, Cannon SJ, Williams DM. Comparative Cytotoxic Effects of Short-chain Fatty Acids Produced by Periodontal Pathogens on Two Cultured Fibroblast Lines. MICROBIAL ECOLOGY IN HEALTH AND DISEASE 2009. [DOI: 10.3109/08910609409141576] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- M. A. Scragg
- Department of Oral Pathology, The London Hospital Medical College, Turner Street, London, EI 2AD, UK
| | - S. J. Cannon
- Department of Oral Pathology, The London Hospital Medical College, Turner Street, London, EI 2AD, UK
| | - D. M. Williams
- Department of Oral Pathology, The London Hospital Medical College, Turner Street, London, EI 2AD, UK
| |
Collapse
|
46
|
Shin HS, Kim MJ, Kim HS, Park SN, Kim DK, Baek DH, Kim C, Kook JK. Development of strain-specific PCR primers for the identification of Fusobacterium nucleatum subsp. fusiforme ATCC 51190(T) and subsp. vincentii ATCC 49256(T). Anaerobe 2009; 16:43-6. [PMID: 19398030 DOI: 10.1016/j.anaerobe.2009.04.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2008] [Accepted: 04/09/2009] [Indexed: 11/16/2022]
Abstract
The objective of this study was to develop the strain-specific PCR primers for Fusobacterium nucleatum subsp. fusiforme ATCC 51190(T) and F. nucleatum subsp. vincentii ATCC 49256(T) based on the nucleotide sequence of the Fs17 and Fv35 DNA probes, respectively. The strain specificity was tested against 10 type strains of Fusobacterium spp. or subsp., 21 clinical isolates of F. nucleatum from Koreans, and five type strains of distinct Fusobacterium species. Primer sensitivity was determined by testing serial dilutions (4 ng-4 fg) of the purified genomic DNA from each of the type strains. PCR showed that two pairs of PCR primers, Fs17-F14/Fs17-R14 and Fv35-F1/Fv35-R1 primers, could produce strain-specific amplicons from F. nucleatum subsp. fusiforme ATCC 51190(T) and F. nucleatum subsp. vincentii ATCC 49256(T), respectively. The two PCR primer sets could detect as little as 0.4 pg or 4 pg of the genomic DNA of each target strain. These results suggest that the two sets of PCR primers could be used to identify F. nucleatum subsp. fusiforme ATCC 51190(T) and F. nucleatum subsp. vincentii ATCC 49256(T), particularly for ascertaining the authenticity of the strain.
Collapse
Affiliation(s)
- Hwan Seon Shin
- Department of Oral Biochemistry, College of Dentistry, Chosun University, 375 Seo-Suk Dong, Dong-Ku, Gwangju 501-759, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Katono T, Kawato T, Tanabe N, Suzuki N, Iida T, Morozumi A, Ochiai K, Maeno M. Sodium butyrate stimulates mineralized nodule formation and osteoprotegerin expression by human osteoblasts. Arch Oral Biol 2008; 53:903-9. [PMID: 18406397 DOI: 10.1016/j.archoralbio.2008.02.016] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2007] [Revised: 01/29/2008] [Accepted: 02/29/2008] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Butyric acid (sodium butyrate; BA) is a major metabolic by-product of main periodontopathic bacteria present in subgingival plaque. In the present study, we examined the effects of BA on cell proliferation, alkaline phosphatase (ALPase) activity, mineralized nodule formation, extracellular matrix protein expression, macrophage colony-stimulating factor (M-CSF), and osteoprotegerin (OPG) in normal human osteoblasts. METHODS The cells were cultured with 0, 10(-8), 10(-6) or 10(-4)M BA for up to 12 days. Mineralized nodule formation was detected by alizarin red staining, and the calcium content in mineralized nodules was determined using a calcium assay kit. The gene and protein expression levels for type I collagen, bone sialoprotein (BSP), osteopontin (OPN), M-CSF, and OPG were examined using real-time PCR and ELISA, respectively. RESULTS Mineralized nodule formation and the calcium content of mineralized nodules were increased by BA in a dose-dependent manner. Cell proliferation and ALPase activity were not affected by the addition of BA. Following the addition of 10(-4)M BA, the expression levels of BSP, OPN, and OPG increased, whereas the expression levels of type I collagen and M-CSF were not markedly affected. CONCLUSION These results suggest that BA stimulates bone formation by increasing the production of BSP and OPN, whereas it suppresses osteoclast differentiation by increasing the production of OPG by human osteoblasts.
Collapse
Affiliation(s)
- Tomoko Katono
- Nihon University Graduate School of Dentistry, 1-8-13 Kanda Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Ho YC, Chang YC. Effects of a bacterial lipid byproduct on human pulp fibroblasts in vitro. J Endod 2007; 33:437-41. [PMID: 17368334 DOI: 10.1016/j.joen.2006.12.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2006] [Revised: 12/15/2006] [Accepted: 12/24/2006] [Indexed: 10/23/2022]
Abstract
Butyrate, a short chain fatty acid, is a metabolic lipid byproduct of various root canal pathogens, such as Porphyromonas endodontalis. However, little is known about the effects of butyrate on cultured human pulp fibroblasts. H33258 fluorescence, flow cytometry, and protein synthesis assays were used to investigate the pathobiologic effects of butyrate on cultured human pulp fibroblasts. Butyrate exhibited cytotoxic effects on human pulp fibroblasts in a concentration-dependent manner (p < 0.05). The addition of butyrate resulted in G2/M phase arrest (p < 0.05). Butyrate also inhibited protein synthesis in a dose-dependent manner (p < 0.05). To determine whether glutathione (GSH) levels were important in the cytotoxicity of butyrate, we pretreated cells with the GSH precursor, 2-oxothiazolidine-4-carboxylic acid (OTZ), to boost thiol levels, or buthionine sulfoximine (BSO) to deplete GSH. The addition of OTZ acted as a protective effect on the butyrate-induced cytotoxicity (p < 0.05). In contrast, the addition of BSO enhanced the butyrate-induced cytotoxicity (p < 0.05). These results indicate that butyrate is cytotoxic to human pulp fibroblasts by inhibiting cell growth, cell-cycle kinetics, and protein synthesis. These inhibitory effects were associated with intracellular GSH levels.
Collapse
Affiliation(s)
- Yung-Chuan Ho
- School of Applied Chemistry, Chung Shan Medical University, Taichung, Taiwan
| | | |
Collapse
|
49
|
Jeng JH, Kuo MYP, Lee PH, Wang YJ, Lee MY, Lee JJ, Lin BR, Tai TF, Chang MC. Toxic and metabolic effect of sodium butyrate on SAS tongue cancer cells: Role of cell cycle deregulation and redox changes. Toxicology 2006; 223:235-47. [PMID: 16737765 DOI: 10.1016/j.tox.2006.04.033] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2006] [Revised: 04/15/2006] [Accepted: 04/19/2006] [Indexed: 12/12/2022]
Abstract
Butyrate is a metabolite produced by oral and colonic microorganism. Butyrate has been shown to reduce colon cancer, whereas its role in oral carcinogenesis is not clear. Butyrate concentration in dental plaque and saliva ranged from 0.2 to 16 mM. In this study, we found that sodium butyrate inhibited the growth of SAS tongue cancer cells by 32% and 53% at concentrations of 1 and 2mM, respectively. Low concentrations of sodium butyrate (1-8mM) induced G0/G1 cell cycle arrest of SAS cells, whereas concentrations of 4-16 mM elicited G2/M arrest and a slight increase in apoptotic cell populations. These events were concomitant with induction of intracellular reactive oxygen species (ROS) production. An elevation in p21 mRNA and protein level was noted in SAS cells by sodium butyrate. On the contrary, a decline of cyclin Bl, cdc2 and cdc25C mRNA and protein expression in SAS cells was found after exposure to sodium butyrate. In addition, no evident increase in cdc2 inhibitory phosphorylation was found in sodium butyrate-treated SAS cancer cells. Inclusion of N-acetyl-l-cysteine (NAC) (3mM), catalase (1000 U/ml) and dimethylthiourea (DMT, 5mM), and also SOD (500 U/ml) attenuated the sodium butyrate-induced ROS production in SAS cells. However, they were not able to prevent the cell cycle arrest, apoptosis and growth inhibition in SAS cells induced by 1, 2 and 16 mM of sodium butyrate. These results indicate that sodium butyrate is toxic and inhibits the tongue cancer cell growth via induction of cell cycle arrest and apoptosis. Sodium butyrate mediates these events by mechanisms additional to ROS production.
Collapse
Affiliation(s)
- Jiiang-Huei Jeng
- Laboratory of Dental Pharmacology and Toxicology, Department of Dentistry, National Taiwan University Hospital and Medical College, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
In the last decade, the dogma that no bacteria could grow in the acid milieu of the stomach has been destroyed by evidence that the infective agent, H. pylori, is responsible for gastric and duodenal disease. Studies on H. pylori infection suggest that some strains of intestinal bacteria may be responsible for intestinal ulceration and inflammation concomitant with inflammatory bowel diseases (IBD), i.e., ulcerative colitis and Crohn's disease. Evidence for pathophysiological roles for certain strains of luminal bacteria result from a number of IBD animal models. Recent studies on innate immunity, including toll-like receptors and NOD isoforms, suggest that bacterial infections may contribute to intestinal inflammation in genetically susceptible hosts. This brief review focuses on the bacterial pathogenesis and the role of innate immunity in the etiology of IBD's.
Collapse
Affiliation(s)
- Toshifumi Ohkusa
- Department of Gastroenterology, Juntendo University School of Medicine, Tokyo
| | | | | |
Collapse
|