1
|
Chopra A, Bhuvanagiri G, Natu K, Chopra A. Role of CRISPR-Cas systems in periodontal disease pathogenesis and potential for periodontal therapy: A review. Mol Oral Microbiol 2025; 40:1-16. [PMID: 39224035 DOI: 10.1111/omi.12483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/05/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPRs) are DNA sequences capable of editing a host genome sequence. CRISPR and its specific CRISPR-associated (Cas) protein complexes have been adapted for various applications. These include activating or inhibiting specific genetic sequences or acting as molecular scissors to cut and modify the host DNA precisely. CRISPR-Cas systems are also naturally present in many oral bacteria, where they aid in nutrition, biofilm formation, inter- and intraspecies communication (quorum sensing), horizontal gene transfer, virulence, inflammation modulation, coinfection, and immune response evasion. It even functions as an adaptive immune system, defending microbes against invading viruses and foreign genetic elements from other bacteria by targeting and degrading their DNA. Recently, CRISPR-Cas systems have been tested as molecular editing tools to manipulate specific genes linked with periodontal disease (such as periodontitis) and as novel methods of delivering antimicrobial agents to overcome antimicrobial resistance. With the rapidly increasing role of CRISPR in treating inflammatory diseases, its application in periodontal disease is also becoming popular. Therefore, this review aims to discuss the different types of CRISPR-Cas in oral microbes and their role in periodontal disease pathogenesis and precision periodontal therapy.
Collapse
Affiliation(s)
- Aditi Chopra
- Department of Periodontology, Manipal College of Dental Sciences, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Geeta Bhuvanagiri
- Department of Periodontology, Manipal College of Dental Sciences, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Kshitija Natu
- School of Dentistry, University of California, Los Angeles, California, USA
| | - Avneesh Chopra
- Department of Periodontology, Oral Medicine and Oral Surgery, Institute for Dental and Craniofacial Sciences, Charité-University Medicine Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Manipal College of Dental Sciences, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
2
|
Shawkatova I, Durmanova V, Javor J. Alzheimer's Disease and Porphyromonas gingivalis: Exploring the Links. Life (Basel) 2025; 15:96. [PMID: 39860036 PMCID: PMC11766648 DOI: 10.3390/life15010096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/10/2025] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
Recent research highlights compelling links between oral health, particularly periodontitis, and systemic diseases, including Alzheimer's disease (AD). Although the biological mechanisms underlying these associations remain unclear, the role of periodontal pathogens, particularly Porphyromonas gingivalis, has garnered significant attention. P. gingivalis, a major driver of periodontitis, is recognized for its potential systemic effects and its putative role in AD pathogenesis. This review examines evidence connecting P. gingivalis to hallmark AD features, such as amyloid β accumulation, tau hyperphosphorylation, neuroinflammation, and other neuropathological features consistent with AD. Virulence factors, such as gingipains and lipopolysaccharides, were shown to be implicated in blood-brain barrier disruption, neuroinflammation, and neuronal damage. P. gingivalis-derived outer membrane vesicles may serve to disseminate virulence factors to brain tissues. Indirect mechanisms, including systemic inflammation triggered by chronic periodontal infections, are also supposed to exacerbate neurodegenerative processes. While the exact pathways remain uncertain, studies detecting P. gingivalis virulence factors and its other components in AD-affected brains support their possible role in disease pathogenesis. This review underscores the need for further investigation into P. gingivalis-mediated mechanisms and their interplay with host responses. Understanding these interactions could provide critical insights into novel strategies for reducing AD risk through periodontal disease management.
Collapse
Affiliation(s)
- Ivana Shawkatova
- Institute of Immunology, Faculty of Medicine, Comenius University in Bratislava, Odborarske nam. 14, 811 08 Bratislava, Slovakia; (V.D.); (J.J.)
| | | | | |
Collapse
|
3
|
Shoji M, Reynolds EC, Veith PD. Analyses of biosynthesis mutants reveal that the fifth and sixth sugars of the Porphyromonas gingivalis O-glycan are L-fucose and N-acetylgalactosamine respectively. Gene 2024; 939:149182. [PMID: 39701196 DOI: 10.1016/j.gene.2024.149182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/05/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024]
Abstract
The oral pathogen, Porphyromonas gingivalis has a general O-glycosylation system which it utilises to modify hundreds of proteins localised outside of the cytoplasm. The O-glycan is a heptasaccharide that includes a putative L-fucose and N-acetylgalactosamine (GalNAc) as the 5th and 6th sugar residues respectively. The putative L-fucose is expected to be synthesized as GDP-L-fucose involving the enzymes Gmd (PGN_1078) and Fcl (PGN_1079), while GalNAc is putatively epimerised from GlcNAc by GalE (PGN_1614). In this study we created mutants lacking each of these three enzymes and analysed the resultant glycosylation defects. Immunoblot analysis using antibodies against the model glycoproteins Mfa2, PGN_0742 and PGN_1037 detected bands of reduced size, consistent with glycan truncation. Mass spectrometry analysis of tryptic digests of whole cell lysate proteins revealed that O-glycans in the galE mutant were predominantly pentasaccharides consistent with the 6th sugar being GalNAc. For the gmd and fcl mutants, tetrasaccharides were observed confirming the 5th sugar as L-fucose. The confirmation of these sugars also confirmed PGN_1135 as a GalNAc transferase as the previously analysed PGN_1135 mutant produced the same O-glycan truncation as the galE mutant.
Collapse
Affiliation(s)
- Mikio Shoji
- Department of Microbiology and Oral Infection, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Eric C Reynolds
- Melbourne Dental School, Bio21 Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Paul D Veith
- Melbourne Dental School, Bio21 Institute, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
4
|
Abe K, Yahara H, Nakao R, Yamaguchi T, Akeda Y. A simple and cost-effective transformation system for Porphyromonas gingivalis via natural competence. Front Microbiol 2024; 15:1476171. [PMID: 39498132 PMCID: PMC11532111 DOI: 10.3389/fmicb.2024.1476171] [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: 08/05/2024] [Accepted: 10/03/2024] [Indexed: 11/07/2024] Open
Abstract
Porphyromonas gingivalis is a major oral bacterial pathogen responsible for severe periodontal diseases. Numerous studies have used genetic approaches to elucidate the molecular mechanisms underlying its pathogenicity. Typically, electroporation and conjugation are utilized for mutagenesis of P. gingivalis; however, these techniques require specialized equipment such as high-voltage electroporators, conjugative plasmids and donor strains. In this study, we present a simple, cost-effective transformation method for P. gingivalis without any special equipment by exploiting its natural DNA competence. P. gingivalis ATCC 33277 was grown to the early-exponential phase and mixed with a donor DNA cassette. This mixture was then spotted onto a BHI-HM blood-agar plate and incubated for one day to promote colony biofilm formation. The resulting colony biofilm was suspended in a liquid medium and spread onto antibiotic-containing agar plates. Transformants appeared within 4 to 5 days, achieving a maximum efficiency of 7.7 × 106 CFU/μg. Although we optimized the transformation conditions using a representative strain ATCC 33277, but the method was also effective for other P. gingivalis strains, W83 and TDC60. Additionally, we discovered that deletion of PGN_0421 or PGN_0519, encoding putative ComEA and ComEC, abolished competency, indicating that these gene products are essential for the natural competence.
Collapse
Affiliation(s)
- Kimihiro Abe
- Department of Bacteriology I, National Institute of Infectious Diseases, Tokyo, Japan
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Hiroko Yahara
- Genome Medical Science Project, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Ryoma Nakao
- Department of Bacteriology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Takehiro Yamaguchi
- Department of Bacteriology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yukihiro Akeda
- Department of Bacteriology I, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
5
|
Feng X, Peng D, Qiu Y, Guo Q, Zhang X, Li Z, Pan C. Identification and Validation of Aging- and Endoplasmic Reticulum Stress-Related Genes in Periodontitis Using a Competing Endogenous RNA Network. Inflammation 2024:10.1007/s10753-024-02124-0. [PMID: 39136902 DOI: 10.1007/s10753-024-02124-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/26/2024] [Accepted: 08/06/2024] [Indexed: 01/03/2025]
Abstract
Periodontitis is a multifactorial chronic inflammatory disease that destroy periodontium. Apart from microbial infection and host immune responses, emerging evidence shows aging and endoplasmic reticulum stress (ER stress) play a key role in periodontitis pathogenesis. The aim of this study is to identify aging-related genes (ARGs) and endoplasmic reticulum stress-related genes (ERGs) in periodontitis. Data were obtained from the Gene Expression Omnibus (GEO), Human Ageing Genomic Resources (HAGR) and GeneCards databases to identify differentially expressed mRNAs/miRNAs/lncRNAs (DEmRNAs/DEmiRNAs/DElncRNAs), ARGs and ERGs, respectively. We used the MultiMiR database for the reverse prediction of miRNAs and predicted miRNA-lncRNA interactions using the STARBase database. Afterwards, we constructed a mRNA-miRNA-lncRNA ceRNA network. A total of 10 hub genes, namely LCK, LYN, CXCL8, IL6, HCK, IL1B, BTK, CXCL12, GNAI1 and FCER1G, and 5 DEmRNAs-ARGs-ERGs were then discovered. Further, weighted gene co-expression network analysis (WGCNA) and single sample gene set enrichment analysis (ssGSEA) were performed to explore co-expression modules and immune infiltration respectively. Finally, we used transmission electron microscope (TEM), inverted fluorescence microscopy, quantitative real-time polymerase chain reaction (qRT-PCR) and Western Blot to verify the bioinformatic results in periodontal ligament stem cells (PDLSCs) infected with Porphyromonas gingivalis (P. gingivalis). The experimental results broadly confirmed the accuracy of bioinformatic analysis. The present study established an aging- and ER stress-related ceRNA network in periodontitis, contributing to a deeper understanding of the pathogenesis of periodontitis.
Collapse
Affiliation(s)
- Xinran Feng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Da Peng
- School and Hospital of Stomatology, China Medical University, Shenyang, 110002, China
| | - Yunjing Qiu
- School of Nursing & Midwifery, Faculty of Health, University of Technology Sydney, Sydney, 2007, Australia
| | - Qian Guo
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiaoyu Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Zhixuan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Chunling Pan
- School and Hospital of Stomatology, China Medical University, Shenyang, 110002, China.
| |
Collapse
|
6
|
Nakao R, Takatsuka A, Mandokoro K, Narisawa N, Ikeda T, Takai H, Ogata Y. Multimodal inhibitory effect of matcha on Porphyromonas gingivalis. Microbiol Spectr 2024; 12:e0342623. [PMID: 38771061 PMCID: PMC11218439 DOI: 10.1128/spectrum.03426-23] [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: 09/19/2023] [Accepted: 03/29/2024] [Indexed: 05/22/2024] Open
Abstract
Porphyromonas gingivalis has been associated with progression of periodontitis, characterized by inflammation and destruction of periodontal tissues. Here, we report that matcha, a product of Camellia sinensis, hampers the adherence and survival of P. gingivalis through multiple tactics. Matcha extract (ME) inhibited the growth not only of P. gingivalis but also of Prevotella nigrescens and Fusobacterium nucleatum, while it did not inhibit growth of nine species of oral streptococci and Aggregatibacter actinomycetemcomitans. ME-mediated P. gingivalis growth inhibition was characterized by both morphological and physiological changes at the bacterial envelope, which were accompanied by nano-particle formation and decreased membrane fluidity/permeability without loss of membrane integrity. ME also triggered autoaggregation of P. gingivalis in a major fimbriae (FimA)-dependent manner. In addition, adherence of P. gingivalis was dramatically inhibited by ME, irrespective of fimbriae. Furthermore, a structure-activity relationship study tested a series of catechins isolated from ME and identified the pyrogallol-type B-ring of catechins as essential for P. gingivalis growth inhibition. In a clinical study to assess the microbiological and therapeutic effects of matcha mouthwash in patients with periodontitis, the P. gingivalis number in saliva was significantly reduced by matcha mouthwash compared to the pre-intervention level. A tendency toward improvement in probing pocket depth was observed in the matcha group, although the difference was not statistically significant. Taken together, we present a proof of concept, based on the multimodal inhibitory effect of matcha against P. gingivalis, and that matcha may have clinical applicability for prevention and treatment of periodontitis. IMPORTANCE Periodontitis, a multifactorial inflammatory disease of the oral cavity, results in alveolar bone destruction, and is a major cause of tooth loss of humans. In addition, emerging evidence has demonstrated associations between periodontitis and a wide range of other chronic inflammation-driven disorders, including diabetes mellitus, preterm birth, cardiovascular disease, aspiration pneumonia, rheumatoid arthritis, cognitive disorder, and cancer. In the present study, we report that matcha, a product of Camellia sinensis, hampers Porphyromonas gingivalis, a major periodontal pathobiont, in not only a series of in vitro experiments but also a pilot intervention clinical trial of patients with periodontitis, in which matcha mouthwash statistically significantly reduced the P. gingivalis number in saliva, as compared to the pre-intervention level. Taken together, we suggest that matcha may have clinical applicability for prevention and treatment of periodontitis.
Collapse
Affiliation(s)
- Ryoma Nakao
- Department of Bacteriology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Ayami Takatsuka
- Department of Bacteriology I, National Institute of Infectious Diseases, Tokyo, Japan
- Department of Food Bioscience and Biotechnology, College of Bioresource Science, Nihon University, Kanagawa, Japan
| | - Kengo Mandokoro
- Department of Food Bioscience and Biotechnology, College of Bioresource Science, Nihon University, Kanagawa, Japan
| | - Naoki Narisawa
- Department of Food Bioscience and Biotechnology, College of Bioresource Science, Nihon University, Kanagawa, Japan
| | - Tsuyoshi Ikeda
- Department of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan
| | - Hideki Takai
- Department of Periodontology, Nihon University School of Dentistry at Matsudo, Chiba, Japan
| | - Yorimasa Ogata
- Department of Periodontology, Nihon University School of Dentistry at Matsudo, Chiba, Japan
| |
Collapse
|
7
|
Wint WY, Miyanohara M, Yamada H, Nakatsuka T, Okamoto M, Ryo K, Tanaka T, Hanada N, Murata T. Rapid multiplex real-time PCR assay using a portable device for the detection of oral pathogens. Diagn Microbiol Infect Dis 2024; 109:116214. [PMID: 38402755 DOI: 10.1016/j.diagmicrobio.2024.116214] [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: 12/11/2023] [Revised: 02/07/2024] [Accepted: 02/12/2024] [Indexed: 02/27/2024]
Abstract
Colonization by several oral pathogens and the onset of oral diseases, such as dental caries and periodontal diseases, are closely related. Therefore, the analysis of pathogens in oral specimens would be helpful for the risk assessment of oral diseases. We developed a rapid multiplex real-time polymerase chain reaction (PCR) method using a portable device and newly designed probe/primer sets to detect the oral pathogens Streptococcus mutans, Porphyromonas gingivalis, Treponema denticola, and Tannerella forsythia. The theoretical minimum detectable cell numbers of S. mutans, P. gingivalis, T. denticola, and T. forsythia were 1, 1, 4, and 3, respectively. The multiplex real-time PCR system simultaneously detected the colonization of S. mutans and P. gingivalis in human saliva. These results suggest that the multiplex real-time PCR system may be useful for the risk assessment of oral diseases.
Collapse
Affiliation(s)
- Wit Yee Wint
- Department of Oral Health Science, Tsurumi University School of Dental Medicine, Tsurumi, 230-8501 Yokohama, Japan
| | - Mayu Miyanohara
- Department of Oral Health Science, Tsurumi University School of Dental Medicine, Tsurumi, 230-8501 Yokohama, Japan
| | - Hidenori Yamada
- Department of Oral Health Science, Tsurumi University School of Dental Medicine, Tsurumi, 230-8501 Yokohama, Japan
| | - Takako Nakatsuka
- Department of Oral Health Science, Tsurumi University School of Dental Medicine, Tsurumi, 230-8501 Yokohama, Japan
| | - Masaaki Okamoto
- Department of Oral Health Science, Tsurumi University School of Dental Medicine, Tsurumi, 230-8501 Yokohama, Japan
| | - Koufuchi Ryo
- Department of Oral Health Science, Tsurumi University School of Dental Medicine, Tsurumi, 230-8501 Yokohama, Japan
| | - Tomoko Tanaka
- Department of Oral Health, School of Life Dentistry at Tokyo, Nippon Dental University, Fujimi, Chiyoda-ku, Tokyo, 102-8159 Japan
| | - Nobuhiro Hanada
- Department of Oral Health Science, Tsurumi University School of Dental Medicine, Tsurumi, 230-8501 Yokohama, Japan
| | - Takatoshi Murata
- Department of Oral Health Science, Tsurumi University School of Dental Medicine, Tsurumi, 230-8501 Yokohama, Japan.
| |
Collapse
|
8
|
Melgar-Rodríguez S, Polanco A, Ríos-Muñoz J, García M, Sierra-Cristancho A, González-Osuna L, Díaz-Zúñiga J, Carvajal P, Vernal R, Bravo D. Differential Response of Human Dendritic Cells upon Stimulation with Encapsulated or Non-Encapsulated Isogenic Strains of Porphyromonas gingivalis. Int J Mol Sci 2024; 25:4510. [PMID: 38674095 PMCID: PMC11049913 DOI: 10.3390/ijms25084510] [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: 01/31/2024] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
During periodontitis, the extracellular capsule of Porphyromonas gingivalis favors alveolar bone loss by inducing Th1 and Th17 patterns of lymphocyte response in the infected periodontium. Dendritic cells recognize bacterial antigens and present them to T lymphocytes, defining their activation and polarization. Thus, dendritic cells could be involved in the Th1 and Th17 response induced against the P. gingivalis capsule. Herein, monocyte-derived dendritic cells were obtained from healthy individuals and then stimulated with different encapsulated strains of P. gingivalis or two non-encapsulated isogenic mutants. Dendritic cell differentiation and maturation were analyzed by flow cytometry. The mRNA expression levels for distinct Th1-, Th17-, or T-regulatory-related cytokines and transcription factors, as well as TLR2 and TLR4, were assessed by qPCR. In addition, the production of IL-1β, IL-6, IL-23, and TNF-α was analyzed by ELISA. The encapsulated strains and non-encapsulated mutants of P. gingivalis induced dendritic cell maturation to a similar extent; however, the pattern of dendritic cell response was different. In particular, the encapsulated strains of P. gingivalis induced higher expression of IRF4 and NOTCH2 and production of IL-1β, IL-6, IL-23, and TNF-α compared with the non-encapsulated mutants, and thus, they showed an increased capacity to trigger Th1 and Th17-type responses in human dendritic cells.
Collapse
Affiliation(s)
- Samanta Melgar-Rodríguez
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago 8380492, Chile; (S.M.-R.); (A.P.); (J.R.-M.); (M.G.); (A.S.-C.); (L.G.-O.); (J.D.-Z.); (P.C.)
- Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago 8380492, Chile
| | - Alan Polanco
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago 8380492, Chile; (S.M.-R.); (A.P.); (J.R.-M.); (M.G.); (A.S.-C.); (L.G.-O.); (J.D.-Z.); (P.C.)
| | - Jearitza Ríos-Muñoz
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago 8380492, Chile; (S.M.-R.); (A.P.); (J.R.-M.); (M.G.); (A.S.-C.); (L.G.-O.); (J.D.-Z.); (P.C.)
- Biomedical Research Center, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Michelle García
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago 8380492, Chile; (S.M.-R.); (A.P.); (J.R.-M.); (M.G.); (A.S.-C.); (L.G.-O.); (J.D.-Z.); (P.C.)
| | - Alfredo Sierra-Cristancho
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago 8380492, Chile; (S.M.-R.); (A.P.); (J.R.-M.); (M.G.); (A.S.-C.); (L.G.-O.); (J.D.-Z.); (P.C.)
- Faculty of Dentistry, Universidad Andrés Bello, Santiago 8370035, Chile
| | - Luis González-Osuna
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago 8380492, Chile; (S.M.-R.); (A.P.); (J.R.-M.); (M.G.); (A.S.-C.); (L.G.-O.); (J.D.-Z.); (P.C.)
| | - Jaime Díaz-Zúñiga
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago 8380492, Chile; (S.M.-R.); (A.P.); (J.R.-M.); (M.G.); (A.S.-C.); (L.G.-O.); (J.D.-Z.); (P.C.)
- Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago 8380492, Chile
| | - Paola Carvajal
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago 8380492, Chile; (S.M.-R.); (A.P.); (J.R.-M.); (M.G.); (A.S.-C.); (L.G.-O.); (J.D.-Z.); (P.C.)
- Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago 8380492, Chile
| | - Rolando Vernal
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago 8380492, Chile; (S.M.-R.); (A.P.); (J.R.-M.); (M.G.); (A.S.-C.); (L.G.-O.); (J.D.-Z.); (P.C.)
- Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago 8380492, Chile
| | - Denisse Bravo
- Laboratorio de Interacciones Microbianas, Faculty of Dentistry, Universidad Andrés Bello, Santiago 8370035, Chile
| |
Collapse
|
9
|
Zhou Y, McClain C, Feng W. Porphyromonas gingivalis Strain W83 Infection Induces Liver Injury in Experimental Alcohol-Associated Liver Disease (ALD) in Mice. Appl Microbiol 2024; 4:620-634. [DOI: 10.3390/applmicrobiol4020043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
The liver plays a vital role in the defense against infections. Porphyromonas gingivalis (P. gingivalis), a dominant etiologic oral bacterium implicated in periodontal disease (PD), has been associated with various systemic diseases. This study aimed to investigate the influence of P. gingivalis on alcohol-associated liver diseases (ALD). Mice were fed a Lieber–DeCarli liquid diet containing 5% ethanol for 10 days after an initial adaptation period on a diet with lower ethanol content for 7 days. Two days before tissue sample collection, the mice were administered P. gingivalis strain W83 (Pg) through intraperitoneal injection (IP). Pair-fed mice with Pg infection (PF+Pg) exhibited an activated immune response to combat infections. However, alcohol-fed mice with Pg infection (AF+Pg) showed liver injury with noticeable abscess lesions and elevated serum alanine aminotransferase (ALT) levels. Additionally, these mice displayed liver infiltration of inflammatory monocytes and significant downregulation of proinflammatory cytokine gene expression levels; and AF+Pg mice also demonstrated increased intrahepatic neutrophil infiltration, as confirmed by chloroacetate esterase (CAE) staining, along with elevated gene expression levels of neutrophil cytosol factor 1 (Ncf1), neutrophilic inflammation driver lipocalin 2 (Lcn2), and complement component C5a receptor 1 (C5ar1), which are associated with neutrophilic inflammation. Interestingly, compared to PF+Pg mice, the livers of AF+Pg mice exhibited downregulation of gene expression levels of NADPH oxidase 2 (Cybb), the leukocyte adhesion molecule Cd18, and the Toll-like receptor adaptor Myd88. Consequently, impaired clearance of P. gingivalis and other bacteria in the liver, increased susceptibility to infections, and inflammation-associated hepatic necrotic cell death were observed in AF+Pg mice, which is likely to have facilitated immune cell infiltration and contributed to liver injury. Furthermore, in addition to the Srebf1/Fasn pathway induced by alcohol feeding, Pg infection also activated carbohydrate response element-binding protein (ChREBP) in AF+Pg mice. In summary, this study demonstrates that P. gingivalis infection, acting as a “second hit”, induces dysfunction of immune response and impairs the clearance of bacteria and infections in alcohol-sensitized livers. This process drives the development of liver injury.
Collapse
Affiliation(s)
- Yun Zhou
- Department of Medicine, University of Louisville, Louisville, KY 40202, USA
- Alcohol Research Center, University of Louisville, Louisville, KY 40202, USA
| | - Craig McClain
- Department of Medicine, University of Louisville, Louisville, KY 40202, USA
- Alcohol Research Center, University of Louisville, Louisville, KY 40202, USA
- Hepatobiology and Toxicology Center, University of Louisville, Louisville, KY 40202, USA
- Robley Rex VA Medical Center, Louisville, KY 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
| | - Wenke Feng
- Department of Medicine, University of Louisville, Louisville, KY 40202, USA
- Alcohol Research Center, University of Louisville, Louisville, KY 40202, USA
- Hepatobiology and Toxicology Center, University of Louisville, Louisville, KY 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
- Department of Structural and Cellular Biology, Tulane University, New Orleans, LA 70112, USA
| |
Collapse
|
10
|
Murugaiyan V, Utreja S, Hovey KM, Sun Y, LaMonte MJ, Wactawski-Wende J, Diaz PI, Buck MJ. Defining Porphyromonas gingivalis strains associated with periodontal disease. Sci Rep 2024; 14:6222. [PMID: 38485747 PMCID: PMC10940620 DOI: 10.1038/s41598-024-56849-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 03/12/2024] [Indexed: 03/18/2024] Open
Abstract
Porphyromonas gingivalis, a Gram-negative anaerobic bacterium commonly found in human subgingival plaque, is a major etiologic agent for periodontitis and has been associated with multiple systemic pathologies. Many P. gingivalis strains have been identified and different strains possess different virulence factors. Current oral microbiome approaches (16S or shotgun) have been unable to differentiate P. gingivalis strains. This study presents a new approach that aims to improve the accuracy of strain identification, using a detection method based on sequencing of the intergenic spacer region (ISR) which is variable between P. gingivalis strains. Our approach uses two-step PCR to amplify only the P. gingivalis ISR region. Samples are then sequenced with an Illumina sequencer and mapped to specific strains. Our approach was validated by examining subgingival plaque from 153 participants with and without periodontal disease. We identified the avirulent strain ATCC33277/381 as the most abundant strain across all sample types. The W83/W50 strain was significantly enriched in periodontitis, with 13% of participants harboring that strain. Overall, this approach can have significant implications not only for the diagnosis and treatment of periodontal disease but also for other diseases where P. gingivalis or its toxins have been implicated, such as Alzheimer's disease.
Collapse
Affiliation(s)
- Vijaya Murugaiyan
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Simran Utreja
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Kathleen M Hovey
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, USA
| | - Yijun Sun
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Michael J LaMonte
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, USA
| | - Jean Wactawski-Wende
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Patricia I Diaz
- UB Microbiome Center, University at Buffalo, Buffalo, NY, USA
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, NY, USA
| | - Michael J Buck
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.
- Department of Biomedical Informatics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
11
|
Pham C, Guo S, Han X, Coleman L, Sze CW, Wang H, Liu J, Li C. A pleiotropic role of sialidase in the pathogenicity of Porphyromonas gingivalis. Infect Immun 2024; 92:e0034423. [PMID: 38376159 PMCID: PMC10929438 DOI: 10.1128/iai.00344-23] [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: 01/02/2024] [Accepted: 02/01/2024] [Indexed: 02/21/2024] Open
Abstract
As one of the keystone pathogens of periodontitis, the oral bacterium Porphyromonas gingivalis produces an array of virulence factors, including a recently identified sialidase (PG0352). Our previous report involving loss-of-function studies indicated that PG0352 plays an important role in the pathophysiology of P. gingivalis. However, this report had not been corroborated by gain-of-function studies or substantiated in different P. gingivalis strains. To fill these gaps, herein we first confirm the role of PG0352 in cell surface structures (e.g., capsule) and serum resistance using P. gingivalis W83 strain through genetic complementation and then recapitulate these studies using P. gingivalis ATCC33277 strain. We further investigate the role of PG0352 and its counterpart (PGN1608) in ATCC33277 in cell growth, biofilm formation, neutrophil killing, cell invasion, and P. gingivalis-induced inflammation. Our results indicate that PG0352 and PGN1608 are implicated in P. gingivalis cell surface structures, hydrophobicity, biofilm formation, resistance to complement and neutrophil killing, and host immune responses. Possible molecular mechanisms involved are also discussed. In summary, this report underscores the importance of sialidases in the pathophysiology of P. gingivalis and opens an avenue to elucidate their underlying molecular mechanisms.
Collapse
Affiliation(s)
- Christopher Pham
- Department of Oral Craniofacial Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Shuaiqi Guo
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, Connecticut, USA
- Microbial Sciences Institute, Yale University, West Haven, Connecticut, USA
| | - Xiao Han
- Department of Oral Craniofacial Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Laurynn Coleman
- Department of Oral Craniofacial Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Ching Wooen Sze
- Department of Oral Craniofacial Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Huizhi Wang
- Department of Oral Craniofacial Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Jun Liu
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, Connecticut, USA
- Microbial Sciences Institute, Yale University, West Haven, Connecticut, USA
| | - Chunhao Li
- Department of Oral Craniofacial Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, USA
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
12
|
Śmiga M, Ślęzak P, Olczak T. Comparative analysis of Porphyromonas gingivalis A7436 and ATCC 33277 strains reveals differences in the expression of heme acquisition systems. Microbiol Spectr 2024; 12:e0286523. [PMID: 38289063 PMCID: PMC10913741 DOI: 10.1128/spectrum.02865-23] [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: 07/19/2023] [Accepted: 12/15/2023] [Indexed: 03/06/2024] Open
Abstract
Porphyromonas gingivalis strains exhibit different phenotypes in vitro, different virulence potential in animal models, and different associations with human diseases, with strains classified as virulent/more virulent (e.g., A7436 and W83) or as less virulent/avirulent (e.g., ATCC 33277). In this study, we comparatively analyzed the A7436 and ATCC 33277 strains to better understand their variability. Global gene expression analysis in response to heme and iron limitation revealed more pronounced differences in the A7436 than in the ATCC 33277 strain; however, in both strains, the largest changes were observed in genes encoding hypothetical proteins, genes whose products participate in energy metabolism, and in genes encoding proteins engaged in transport and binding proteins. Our results confirmed that variability between P. gingivalis strains is due to differences in the arrangement of their genomes. Analysis of gene expression of heme acquisition systems demonstrated that not only the availability of iron and heme in the external environment but also the ability to store iron intracellularly can influence the P. gingivalis phenotype. Therefore, we assume that differences in virulence potential may also be due to differences in the production of systems involved in iron and heme acquisition, mainly the Hmu system. In addition, our study showed that hemoglobin, in a concentration-dependent manner, differentially influences the virulence potential of P. gingivalis strains. We conclude that iron and heme homeostasis may add to the variability observed between P. gingivalis strains. IMPORTANCE Periodontitis belongs to a group of multifactorial diseases, characterized by inflammation and destruction of tooth-supporting tissues. P. gingivalis is one of the most important microbial factors involved in the initiation and progression of periodontitis. To survive in the host, the bacterium must acquire heme as a source of iron and protoporphyrin IX. P. gingivalis strains respond differently to changing iron and heme concentrations, which may be due to differences in the expression of systems involved in iron and heme acquisition. The ability to accumulate iron intracellularly, being different in more and less virulent P. gingivalis strains, may influence their phenotypes, production of virulence factors (including proteins engaged in heme acquisition), and virulence potential of this bacterium.
Collapse
Affiliation(s)
- Michał Śmiga
- Laboratory of Medical Biology, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Paulina Ślęzak
- Laboratory of Medical Biology, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Teresa Olczak
- Laboratory of Medical Biology, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| |
Collapse
|
13
|
Minnick MF. Functional Roles and Genomic Impact of Miniature Inverted-Repeat Transposable Elements (MITEs) in Prokaryotes. Genes (Basel) 2024; 15:328. [PMID: 38540387 PMCID: PMC10969869 DOI: 10.3390/genes15030328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 02/27/2024] [Accepted: 03/01/2024] [Indexed: 06/14/2024] Open
Abstract
Prokaryotic genomes are dynamic tapestries that are strongly influenced by mobile genetic elements (MGEs), including transposons (Tn's), plasmids, and bacteriophages. Of these, miniature inverted-repeat transposable elements (MITEs) are undoubtedly the least studied MGEs in bacteria and archaea. This review explores the diversity and distribution of MITEs in prokaryotes and describes what is known about their functional roles in the host and involvement in genomic plasticity and evolution.
Collapse
Affiliation(s)
- Michael F Minnick
- Program in Cellular, Molecular and Microbial Biology, Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| |
Collapse
|
14
|
Sukmana BI, Saleh RO, Najim MA, AL-Ghamdi HS, Achmad H, Al-Hamdani MM, Taher AAY, Alsalamy A, Khaledi M, Javadi K. Oral microbiota and oral squamous cell carcinoma: a review of their relation and carcinogenic mechanisms. Front Oncol 2024; 14:1319777. [PMID: 38375155 PMCID: PMC10876296 DOI: 10.3389/fonc.2024.1319777] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/15/2024] [Indexed: 02/21/2024] Open
Abstract
Oral Squamous Cell Carcinoma (OSCC) is the most common type of head and neck cancer worldwide. Emerging research suggests a strong association between OSCC and the oral microbiota, a diverse community of bacteria, fungi, viruses, and archaea. Pathogenic bacteria, in particular Porphyromonas gingivalis and Fusobacterium nucleatum, have been closely linked to OSCC. Moreover, certain oral fungi, such as Candida albicans, and viruses, like the human papillomavirus, have also been implicated in OSCC. Despite these findings, the precise mechanisms through which the oral microbiota influences OSCC development remain unclear and necessitate further research. This paper provides a comprehensive overview of the oral microbiota and its relationship with OSCC and discusses potential carcinogenic pathways that the oral microbiota may activate or modulate are also discussed.
Collapse
Affiliation(s)
| | - Raed Obaid Saleh
- Department of Medical Laboratory Techniques, Al-Maarif University College, Al-Anbar, Iraq
| | | | - Hasan S. AL-Ghamdi
- Internal Medicine Department, Division of Dermatology, Faculty of Medicine, Albaha University, Albaha, Saudi Arabia
| | - Harun Achmad
- Department of Pediatric Dentistry, Faculty of Dentistry, Hasanuddin University, Indonesia (Lecture of Pediatric Dentistry), Makassar, Indonesia
| | | | | | - Ali Alsalamy
- College of Technical Engineering, Imam Ja’afar Al‐Sadiq University, Al‐Muthanna, Iraq
| | - Mansoor Khaledi
- Department of Microbiology and Immunology, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Kasra Javadi
- Department of Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| |
Collapse
|
15
|
Cornejo Ulloa P, van der Veen MH, Brandt BW, Buijs MJ, Krom BP. The effect of sex steroid hormones on the ecology of in vitro oral biofilms. Biofilm 2023; 6:100139. [PMID: 37621393 PMCID: PMC10447177 DOI: 10.1016/j.bioflm.2023.100139] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 06/27/2023] [Accepted: 06/27/2023] [Indexed: 08/26/2023] Open
Abstract
Sex steroid hormones (SSH) such as oestrogen, progesterone and testosterone are cholesterol derived molecules that regulate various physiological processes. They are present in both blood and saliva, where they come in contact with oral tissues and oral microorganisms. Several studies have confirmed the effect of these hormones on different periodontal-disease-associated bacteria, using single-species models. Bacteria can metabolize SSH, use them as alternative for vitamin K and also use them to induce the expression of virulence factors. However, it is still unclear what the effects of SSH are on the oral microbiome. In this study, we investigated the effects of four SSH on commensal in vitro oral biofilms. Saliva-derived oral biofilms were grown in Mc Bain medium without serum or menadione using the Amsterdam Active-Attachment model. After initial attachment in absence of SSH, the biofilms were grown in medium containing either oestradiol, oestriol, progesterone or testosterone at a 100-fold physiological concentration. Menadione or ethanol were included as positive control and negative control, respectively. After 12 days with daily medium refreshments, biofilm formation, biofilm red fluorescence and microbial composition were determined. The supernatants were tested for proteolytic activity using the Fluorescence Resonance Energy Transfer Analysis (FRET). No significant differences were found in biofilm formation, red fluorescence or microbial composition in any of the tested groups. Samples grown in presence of progesterone and oestradiol showed proteolytic activity comparable to biofilms supplemented with menadione. In contrast, testosterone and oestriol showed a decreased proteolytic activity compared to biofilms grown in presence of menadione. None of the tested SSH had large effects on the ecology of in vitro oral biofilms, therefore a direct translation of our results into in vivo effects is not possible. Future experiments should include other host factors such as oral tissues, immune cells and combinations of SSH as present in saliva, in order to have a more accurate picture of the phenomena taking place in both males and females.
Collapse
Affiliation(s)
- Pilar Cornejo Ulloa
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, the Netherlands
| | - Monique H. van der Veen
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and 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
| | - Mark J. Buijs
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, the Netherlands
| | - Bastiaan P. Krom
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
16
|
Kleiboeker BA, Frankfater C, Davey ME, Hsu FF. Lipidomic analysis of Porphyromonas gingivalis reveals novel glycerol bisphosphoceramide, phosphatidyl-, and phosphoglycerol dipeptide lipid families. J Lipid Res 2023; 64:100470. [PMID: 37924978 PMCID: PMC10757044 DOI: 10.1016/j.jlr.2023.100470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/28/2023] [Accepted: 10/30/2023] [Indexed: 11/06/2023] Open
Abstract
Porphyromonas gingivalis, like other members of the phylum Bacteroidetes (synonym Bacteroidota), synthesizes several classes of dihydroceramides and peptidolipids. Using a similar strategy as that recently used to delimit the lipidome of its close relative Bacteroides fragilis, we applied linear ion trap multiple-stage mass spectrometry (linear ion trap MSn) with high-resolution mass spectrometry, to structurally characterize the complete lipidome of P. gingivalis and compare it to B. fragilis. This analysis discovered that the P. gingivalis lipidome consists of several previously unidentified lipid families, including dihydroceramide-1-phosphophate, acylated dihydroceramide-1-phosphophate, phosphoglycerol glycylserine lipid, and bis(phosphodihydroceramide) glycerol. Interestingly, we also found a novel sphingolipid family containing a polyunsaturated long-chain base, and a new lipoglycylserine phosphatic acid containing unsaturated acyl chains not reported for the lipid family. The comprehensive coverage of the lipidome of P. gingivalis conducted in this study has revealed more than 140 lipid species including several novel lipids in over 20 lipid families/subfamilies.
Collapse
Affiliation(s)
- Brian A Kleiboeker
- Mass Spectrometry Resource, Division of Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Cheryl Frankfater
- Mass Spectrometry Resource, Division of Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Mary E Davey
- Department of Microbiology, The Forsyth Institute, Cambridge, MA, USA
| | - Fong-Fu Hsu
- Mass Spectrometry Resource, Division of Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
17
|
Hayashi M, Ikenaga N, Nakata K, Luo H, Zhong P, Date S, Oyama K, Higashijima N, Kubo A, Iwamoto C, Torata N, Abe T, Yamada Y, Ohuchida K, Oda Y, Nakamura M. Intratumor Fusobacterium nucleatum promotes the progression of pancreatic cancer via the CXCL1-CXCR2 axis. Cancer Sci 2023; 114:3666-3678. [PMID: 37438965 PMCID: PMC10475786 DOI: 10.1111/cas.15901] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/13/2023] [Accepted: 06/19/2023] [Indexed: 07/14/2023] Open
Abstract
Intratumor bacteria modify the tumor immune microenvironment and influence outcomes of various tumors. Periodontal pathogen Fusobacterium nucleatum has been detected in pancreatic cancer tissues and is associated with poor prognosis. However, it remains unclear how F. nucleatum affects pancreatic cancer. Here, we compared clinical features with F. nucleatum colonization in pancreatic cancer tissues. F. nucleatum was detected in 15.5% (13/84) of pancreatic cancer patients. The tumor size was significantly larger in the F. nucleatum-positive group than in the negative group. To clarify the biological effect of intratumor F. nucleatum on pancreatic cancer progression, we performed migration/invasion assays and cytokine array analysis of cancer cells cocultured with F. nucleatum. F. nucleatum promoted CXCL1 secretion from pancreatic cancer cells, leading to cancer progression through autocrine signaling. Intratumor F. nucleatum suppressed tumor-infiltrating CD8+ T cells by recruiting myeloid-derived suppressor cells (MDSCs) to the tumor in an F. nucleatum-injected subcutaneous pancreatic cancer mouse model, resulting in tumor progression. Furthermore, tumor growth accelerated by F. nucleatum was suppressed by MDSC depletion or cytokine inhibitors. Intratumor F. nucleatum promoted pancreatic cancer progression through autocrine and paracrine mechanisms of the CXCL1-CXCR2 axis. Blockade of the CXCL1-CXCR2 axis may be a novel therapeutic approach for patients with intratumor F. nucleatum-positive pancreatic cancer.
Collapse
Affiliation(s)
- Masataka Hayashi
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Naoki Ikenaga
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Kohei Nakata
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Haizhen Luo
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - PingShan Zhong
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Satomi Date
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Koki Oyama
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Nobuhiro Higashijima
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Akihiro Kubo
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Chika Iwamoto
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Nobuhiro Torata
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Bachelor of Health ScienceKyushu UniversityFukuokaJapan
| | - Toshiya Abe
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Yutaka Yamada
- Department of Anatomical Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Kenoki Ohuchida
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Yoshinao Oda
- Department of Anatomical Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| |
Collapse
|
18
|
Morales-Olavarría M, Nuñez-Belmar J, González D, Vicencio E, Rivas-Pardo JA, Cortez C, Cárdenas JP. Phylogenomic analysis of the Porphyromonas gingivalis - Porphyromonas gulae duo: approaches to the origin of periodontitis. Front Microbiol 2023; 14:1226166. [PMID: 37538845 PMCID: PMC10394638 DOI: 10.3389/fmicb.2023.1226166] [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: 05/20/2023] [Accepted: 07/04/2023] [Indexed: 08/05/2023] Open
Abstract
Porphyromonas gingivalis is an oral human pathogen associated with the onset and progression of periodontitis, a chronic immune-inflammatory disease characterized by the destruction of the teeth-supporting tissue. P. gingivalis belongs to the genus Porphyromonas, which is characterized by being composed of Gram-negative, asaccharolytic, non-spore-forming, non-motile, obligatory anaerobic species, inhabiting niches such as the oral cavity, urogenital tract, gastrointestinal tract and infected wound from different mammals including humans. Among the Porphyromonas genus, P. gingivalis stands out for its specificity in colonizing the human oral cavity and its keystone pathogen role in periodontitis pathogenesis. To understand the evolutionary process behind P. gingivalis in the context of the Pophyoromonas genus, in this study, we performed a comparative genomics study with publicly available Porphyromonas genomes, focused on four main objectives: (A) to confirm the phylogenetic position of P. gingivalis in the Porphyromonas genus by phylogenomic analysis; (B) the definition and comparison of the pangenomes of P. gingivalis and its relative P. gulae; and (C) the evaluation of the gene family gain/loss events during the divergence of P. gingivalis and P. gulae; (D) the evaluation of the evolutionary pressure (represented by the calculation of Tajima-D values and dN/dS ratios) comparing gene families of P. gingivalis and P. gulae. Our analysis found 84 high-quality assemblies representing P. gingivalis and 14 P. gulae strains (from a total of 233 Porphyromonas genomes). Phylogenomic analysis confirmed that P. gingivalis and P. gulae are highly related lineages, close to P. loveana. Both organisms harbored open pangenomes, with a strong core-to-accessory ratio for housekeeping genes and a negative ratio for unknown function genes. Our analyses also characterized the gene set differentiating P. gulae from P. gingivalis, mainly associated with unknown functions. Relevant virulence factors, such as the FimA, Mfa1, and the hemagglutinins, are conserved in P. gulae, P. gingivalis, and P. loveana, suggesting that the origin of those factors occurred previous to the P. gulae - P. gingivalis divergence. These results suggest an unexpected evolutionary relationship between the P. gulae - P. gingivalis duo and P. loveana, showing more clues about the origin of the role of those organisms in periodontitis.
Collapse
Affiliation(s)
- Mauricio Morales-Olavarría
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
| | - Josefa Nuñez-Belmar
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
| | - Dámariz González
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
| | - Emiliano Vicencio
- Escuela de Tecnología Médica, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Jaime Andres Rivas-Pardo
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
- Escuela de Biotecnología, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
| | - Cristian Cortez
- Escuela de Tecnología Médica, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Juan P. Cárdenas
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
- Escuela de Biotecnología, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
| |
Collapse
|
19
|
Visentin D, Gobin I, Maglica Ž. Periodontal Pathogens and Their Links to Neuroinflammation and Neurodegeneration. Microorganisms 2023; 11:1832. [PMID: 37513004 PMCID: PMC10385044 DOI: 10.3390/microorganisms11071832] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/05/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Pathogens that play a role in the development and progression of periodontitis have gained significant attention due to their implications in the onset of various systemic diseases. Periodontitis is characterized as an inflammatory disease of the gingival tissue that is mainly caused by bacterial pathogens. Among them, Porphyromonas gingivalis, Treponema denticola, Fusobacterium nucleatum, Aggregatibacter actinomycetemcomitans, and Tannerella forsythia are regarded as the main periodontal pathogens. These pathogens elicit the release of cytokines, which in combination with their virulence factors induce chronic systemic inflammation and subsequently impact neural function while also altering the permeability of the blood-brain barrier. The primary objective of this review is to summarize the existing information regarding periodontal pathogens, their virulence factors, and their potential association with neuroinflammation and neurodegenerative diseases. We systematically reviewed longitudinal studies that investigated the association between periodontal disease and the onset of neurodegenerative disorders. Out of the 24 studies examined, 20 showed some degree of positive correlation between periodontal disease and neurodegenerative disorders, with studies focusing on cognitive function demonstrating the most robust effects. Therefore, periodontal pathogens might represent an exciting new approach to develop novel preventive treatments for neurodegenerative diseases.
Collapse
Affiliation(s)
- David Visentin
- Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
| | - Ivana Gobin
- Department of Microbiology and Parasitology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Željka Maglica
- Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
20
|
Nakao R, Hirayama S, Yamaguchi T, Senpuku H, Hasegawa H, Suzuki T, Akeda Y, Ohnishi M. A bivalent outer membrane vesicle-based intranasal vaccine to prevent infection of periodontopathic bacteria. Vaccine 2023; 41:4369-4383. [PMID: 37302966 DOI: 10.1016/j.vaccine.2023.05.058] [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: 12/09/2022] [Revised: 04/30/2023] [Accepted: 05/25/2023] [Indexed: 06/13/2023]
Abstract
Periodontal disease has become a serious public health problem, not only causing tooth loss, but also inducing chronic disorders of extra-oral organs. The present study assessed an intranasal vaccine strategy to prevent periodontal disease using outer membrane vesicles (OMVs) of two major periodontopathic bacteria, Porphyromonas gingivalis (Pg) and Aggregatibacter actinomycetemcomitans (Aa). We compared the morphology, composition, and immune activity between OMVs of Pg strain ATCC 33277 and Aa strain Y4. Aa OMVs had a smoother surface and stronger lipid A activity compared to Pg OMVs. The in vitro immune activity elicited by Aa OMVs in macrophage-like cells was remarkably stronger than that of Pg OMVs. Intranasal immunization of mice with Aa OMVs alone resulted in robust, humoral immune responses in blood and saliva. Despites the intrinsically low mucosal immunogenicity of Pg OMVs alone, using Aa OMVs as a mucosal adjuvant strongly enhanced Pg-specific immune responses, resulting in both serum IgG and salivary IgA, both of which aggregated Pg and Aa cells. Furthermore, Aa OMVs were found to be a more potent mucosal adjuvant than Poly(I:C) in the context of enhancing the production of Pg-specific IgG (especially IgG2a) and IgA. In addition, in a randomized, blinded study, mice oral challenged with Pg and Aa after intranasal immunization with Pg OMVs and Aa OMVs had significantly decreased numbers of both microorganisms compared to mock-immunized mice. Furthermore, in an intracerebral injection mouse model, there were no serious adverse effects on the brain even after administrating a dose of OMVs as same as that used for intranasal administration. Taken together, the bivalent OMV intranasal vaccine may be effective in preventing colonization of periodontopathic bacteria in the oral cavity and related systemic disorders associated with periodontal diseases.
Collapse
Affiliation(s)
- Ryoma Nakao
- Department of Bacteriology I, National Institute of Infectious Diseases, Tokyo 162-8640, Japan.
| | - Satoru Hirayama
- Department of Bacteriology I, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8514, Japan
| | - Takehiro Yamaguchi
- Department of Bacteriology I, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Hidenobu Senpuku
- Department of Bacteriology I, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; Department of Microbiology and Immunology, School of Dentistry at Matsudo, Nihon University, Chiba 271-8587, Japan
| | - Hideki Hasegawa
- Department of Influenza Virus Research Center, National Institute of Infectious Diseases, Tokyo 208-0011, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Yukihiro Akeda
- Department of Bacteriology I, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Makoto Ohnishi
- Department of Bacteriology I, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| |
Collapse
|
21
|
Fujimoto M, Naiki Y, Sakae K, Iwase T, Miwa N, Nagano K, Nawa H, Hasegawa Y. Structural and antigenic characterization of a novel genotype of Mfa1 fimbriae in Porphyromonas gingivalis. J Oral Microbiol 2023; 15:2215551. [PMID: 37223052 PMCID: PMC10201998 DOI: 10.1080/20002297.2023.2215551] [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: 07/26/2022] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 05/25/2023] Open
Abstract
Background Mfa1 fimbriae of the periodontal pathogen Porphyromonas gingivalis are responsible for biofilm formation and comprise five proteins: Mfa1-5. Two major genotypes, mfa170 and mfa153, encode major fimbrillin. The mfa170 genotype is further divided into the mfa170A and mfa170B subtypes. The properties of the novel mfa170B remain unclear. Methods Fimbriae were purified from P. gingivalis strains JI-1 (mfa170A), 1439 (mfa170B), and Ando (mfa153), and their components and their structures were analyzed. Protein expression and variability in the antigenic specificity of fimbrillins were compared using Coomassie staining and western blotting using polyclonal antibodies against Mfa170A, Mfa170B, and Mfa153 proteins. Cell surface expression levels of fimbriae were analyzed by filtration enzyme-linked immunosorbent assays. Results The composition and structures of the purified Mfa1 fimbriae of 1439 was similar to that of JI-1. However, each Mfa1 protein of differential subtype/genotype was specifically detected by western blotting. Mfa170B fimbriae were expressed in several strains such as 1439, JKG9, B42, 1436, and Kyudai-3. Differential protein expression and antigenic heterogeneities were detected in Mfa2-5 between strains. Conclusion Mfa1 fimbriae from the mfa170A and mfa170B genotypes indicated an antigenic difference suggesting the mfa170B, is to be utilized for the novel classification of P. gingivalis.
Collapse
Affiliation(s)
- Miyuna Fujimoto
- Department of Microbiology, School of Dentistry, Aichi Gakuin University, Nagoya, Japan
- Department of Pediatric Dentistry, School of Dentistry, Aichi Gakuin University, Nagoya, Japan
| | - Yoshikazu Naiki
- Department of Microbiology, School of Dentistry, Aichi Gakuin University, Nagoya, Japan
| | - Kotaro Sakae
- Department of Microbiology, School of Dentistry, Aichi Gakuin University, Nagoya, Japan
| | - Tomohiko Iwase
- Department of Microbiology, School of Dentistry, Aichi Gakuin University, Nagoya, Japan
| | - Naoyoshi Miwa
- Department of Pediatric Dentistry, School of Dentistry, Aichi Gakuin University, Nagoya, Japan
| | - Keiji Nagano
- Division of Microbiology, Department of Oral Biology, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, Japan
| | - Hiroyuki Nawa
- Department of Pediatric Dentistry, School of Dentistry, Aichi Gakuin University, Nagoya, Japan
| | - Yoshiaki Hasegawa
- Department of Microbiology, School of Dentistry, Aichi Gakuin University, Nagoya, Japan
| |
Collapse
|
22
|
Fujita M, Chiu CH, Nagano K. Transcriptional analysis of the mfa-cluster genes in Porphyromonas gingivalis strains with one and two mfa5 genes. Mol Oral Microbiol 2023; 38:41-47. [PMID: 36333926 DOI: 10.1111/omi.12399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 10/28/2022] [Accepted: 10/31/2022] [Indexed: 11/08/2022]
Abstract
The Porphyromonas gingivalis Mfa1 fimbria is composed of the Mfa1 to Mfa5 proteins, encoded by the mfa1 to mfa5 genes, respectively, which are tandemly arranged on chromosomes. A recent study discovered that many P. gingivalis strains possess two mfa5 genes (called herein mfa5-1 and mfa5-2), which are also in tandem. This study examined the transcriptional unit and activity of mfa-cluster genes in strains with one (the ATCC 33277 and TDC60 strains) and two (the HG66 and A7436 strains) mfa5 genes. Complementary DNA was prepared from the total RNA extracted from the bacterial cells in the logarithmic growth phase using a random primer. PCR analysis for the intergenic regions from mfa1 to mfa5 or mfa5-2 showed that mfa1 to mfa5 or mfa5-2 formed a polycistronic gene cluster. Quantitative real-time PCR showed that the mfa1 transcription was 5-10 times higher than that of mfa2 in all the strains. However, mfa2 to mfa5 mostly showed a comparable expression. Both mfa5 genes were comparably transcribed in HG66 and A7436 strains. The transcriptional levels were almost consistent with the respective protein expression levels. In silico analysis identified a transcriptional terminator structure in the intergenic region between mfa1 and mfa2 that was probably responsible for the decreased transcription rate of mfa2 and the downstream genes.
Collapse
Affiliation(s)
- Mari Fujita
- Division of Microbiology, Department of Oral Biology, School of Dentistry, Health Sciences University of Hokkaido, Tobetsu, Japan
| | - Chen-Hsuan Chiu
- Division of Microbiology, Department of Oral Biology, School of Dentistry, Health Sciences University of Hokkaido, Tobetsu, Japan
| | - Keiji Nagano
- Division of Microbiology, Department of Oral Biology, School of Dentistry, Health Sciences University of Hokkaido, Tobetsu, Japan
| |
Collapse
|
23
|
Contribution of -Omics Technologies in the Study of Porphyromonas gingivalis during Periodontitis Pathogenesis: A Minireview. Int J Mol Sci 2022; 24:ijms24010620. [PMID: 36614064 PMCID: PMC9820714 DOI: 10.3390/ijms24010620] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/10/2022] [Accepted: 11/12/2022] [Indexed: 12/31/2022] Open
Abstract
Periodontitis is a non-communicable chronic inflammatory disease characterized by the progressive and irreversible breakdown of the soft periodontal tissues and resorption of teeth-supporting alveolar bone. The etiology of periodontitis involves dysbiotic shifts in the diversity of microbial communities inhabiting the subgingival crevice, which is dominated by anaerobic Gram-negative bacteria, including Porphyromonas gingivalis. Indeed, P. gingivalis is a keystone pathogen with a repertoire of attributes that allow it to colonize periodontal tissues and influence the metabolism, growth rate, and virulence of other periodontal bacteria. The pathogenic potential of P. gingivalis has been traditionally analyzed using classical biochemical and molecular approaches. However, the arrival of new techniques, such as whole-genome sequencing, metagenomics, metatranscriptomics, proteomics, and metabolomics, allowed the generation of high-throughput data, offering a suitable option for bacterial analysis, allowing a deeper understanding of the pathogenic properties of P. gingivalis and its interaction with the host. In the present review, we revise the use of the different -omics technologies and techniques used to analyze bacteria and discuss their potential in studying the pathogenic potential of P. gingivalis.
Collapse
|
24
|
Modulatory Mechanisms of Pathogenicity in Porphyromonas gingivalis and Other Periodontal Pathobionts. Microorganisms 2022; 11:microorganisms11010015. [PMID: 36677306 PMCID: PMC9862357 DOI: 10.3390/microorganisms11010015] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/25/2022] [Accepted: 11/25/2022] [Indexed: 12/24/2022] Open
Abstract
The pathogenesis of periodontitis depends on a sustained feedback loop where bacterial virulence factors and immune responses both contribute to inflammation and tissue degradation. Periodontitis is a multifactorial disease that is associated with a pathogenic shift in the oral microbiome. Within this shift, low-abundance Gram-negative anaerobic pathobionts transition from harmless colonisers of the subgingival environment to a virulent state that drives evasion and subversion of innate and adaptive immune responses. This, in turn, drives the progression of inflammatory disease and the destruction of tooth-supporting structures. From an evolutionary perspective, bacteria have developed this phenotypic plasticity in order to respond and adapt to environmental stimuli or external stressors. This review summarises the available knowledge of genetic, transcriptional, and post-translational mechanisms which mediate the commensal-pathogen transition of periodontal bacteria. The review will focus primarily on Porphyromonas gingivalis.
Collapse
|
25
|
High Molecular Weight Hyaluronic Acid Reduces the Expression of Virulence Genes fimA, mfa1, hagA, rgpA, and kgp in the Oral Pathogen Porphyromonas gingivalis. Pharmaceutics 2022; 14:pharmaceutics14081628. [PMID: 36015254 PMCID: PMC9415305 DOI: 10.3390/pharmaceutics14081628] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/13/2022] [Accepted: 08/02/2022] [Indexed: 02/04/2023] Open
Abstract
Porphyromonas gingivalis (P. gingivalis) is a cornerstone pathogen in the development and progression of periodontal and peri-implant tissue destruction. It is capable of causing dysbiosis of the microbial biofilm and modulation of the host immune system. Hyaluronic acid (HA) is a naturally occurring glycosaminoglycan found in all living organisms. It is well known and has been used for improving tissue healing. In addition, some studies have suggested that there may be an antimicrobial potential to HA. The aim of this study was to evaluate the effect of hyaluronic acid, azithromycin (AZM), and chlorhexidine (CHX) on the expression of genes (i.e., fimA, mfa1, hagA, rgpA, rgpB, and kgp) related to the virulence and adhesion of P. gingivalis. The study groups were divided into four: (1) HA treated group; (2) AZM treated group; (3) CHX treated group; and (4) untreated group to serve as a negative control. P. gingivalis ATCC 33277 was cultured and then exposed to four different concentrations (100% MIC, 50% MIC, 25% MIC, and 12.5% MIC) of HA, AZM, and CHX for 24 h. The expression levels of the aforementioned genes were measured using quantitative reverse transcription polymerase chain reaction (RT-qPCR). Relative fold-change values were calculated and compared between groups. The fold-change values of all genes combined were 0.46 ± 0.33, 0.31 ± 0.24, and 0.84 ± 0.77 for HA, AZM, and CHX, respectively. HA has downregulated all the genes by mostly a half-fold: 0.35 ± 0.20, 0.47 ± 0.35, 0.44 ± 0.25, 0.67 ± 0.46, 0.48 ± 0.33 and 0.35 ± 0.22 with fimA, mfa1, hagA, rgpA, rgpB and kgp, respectively. The effect of HA was significant on all genes except rgpB compared to the untreated control. Lower concentrations of HA tended to exhibit greater downregulation with 1 mg/mL being the most effective. High molecular weight (1.5 MDa) hyaluronic acid has a potent effect on P. gingivalis by downregulating fimA, mfa1, hagA, rgpA, and kgp. The effect of HA was generally less than that of AZM but greater than that of CHX.
Collapse
|
26
|
Porphyromonas gingivalis outer membrane vesicles modulate cytokine and chemokine production by gingipain-dependent mechanisms in human macrophages. Arch Oral Biol 2022; 140:105453. [DOI: 10.1016/j.archoralbio.2022.105453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 04/22/2022] [Accepted: 05/07/2022] [Indexed: 11/19/2022]
|
27
|
Aduse-Opoku J, Joseph S, Devine DA, Marsh PD, Curtis MA. Molecular basis for avirulence of spontaneous variants of Porphyromonas gingivalis: genomic analysis of strains W50, BE1 and BR1. Mol Oral Microbiol 2022; 37:122-132. [PMID: 35622827 PMCID: PMC9328147 DOI: 10.1111/omi.12373] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/12/2022] [Accepted: 05/19/2022] [Indexed: 11/29/2022]
Abstract
The periodontal pathogen Porphyromonas gingivalis is genetically heterogeneous. However, the spontaneous generation of phenotypically different sub‐strains has also been reported. McKee et al. (1988) cultured P. gingivalis W50 in a chemostat during investigations into the growth and properties of this bacterium. Cell viability on blood agar plates revealed two types of non‐pigmenting variants, W50 beige (BE1), and W50 brown (BR1), in samples grown in a high‐hemin medium after day 7, and the population of these variants increased to approximately 25% of the total counts by day 21. W50, BE1 and BR1 had phenotypic alterations in pigmentation, reduced protease activity and haemagglutination and susceptibility to complement killing. Furthermore, the variants exhibited significant attenuation in a mouse model of virulence. Other investigators showed that in BE1, the predominant extracellular Arg‐gingipain was RgpB, and no reaction with an A‐lipopolysaccharide‐specific MAb 1B5 (Collinson et al., 1998; Slaney et al., 2006). In order to determine the genetic basis for these phenotypic properties, we performed hybrid DNA sequence long reads using Oxford Nanopore and the short paired‐end DNA sequence reads of Illumina HiSeq platforms to generate closed circular genomes of the parent and variants. Comparative analysis indicated loss of intact kgp in the 20 kb region of the hagA‐kgp locus in the two variants BE1 and BR1. Deletions in hagA led to smaller open reading frames in the variants, and BR1 had incurred a major chromosomal DNA inversion. Additional minor changes to the genomes of both variants were also observed. Given the importance of Kgp and HagA to protease activity and haemagglutination, respectively, in this bacterium, genomic changes at this locus may account for most of the phenotypic alterations of the variants. The homologous and repetitive nature of hagA and kgp and the features at the inverted junctions are indicative of specific and stable homologous recombination events, which may underlie the genetic heterogeneity of this species.
Collapse
Affiliation(s)
- Joseph Aduse-Opoku
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London
| | - Susan Joseph
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London
| | - Deirdre A Devine
- Division of Oral Biology, School of Dentistry, University of Leeds
| | - Philip D Marsh
- Division of Oral Biology, School of Dentistry, University of Leeds
| | - Michael A Curtis
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London
| |
Collapse
|
28
|
Han Y, Jia Z, Shi J, Wang W, He K. The active lung microbiota landscape of COVID-19 patients through the metatranscriptome data analysis. BIOIMPACTS : BI 2022; 12:139-146. [PMID: 35411293 PMCID: PMC8905590 DOI: 10.34172/bi.2021.23378] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 10/30/2020] [Accepted: 11/14/2020] [Indexed: 12/12/2022]
Abstract
![]()
Introduction: With the outbreak of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the interaction between the host and SARS-CoV-2 was widely studied. However, it is unclear whether and how SARS-CoV-2 infection affects lung microflora, which contribute to COVID-19 complications.
Methods: Here, we analyzed the metatranscriptomic data of bronchoalveolar lavage fluid (BALF) of 19 COVID-19 patients and 23 healthy controls from 6 independent projects and detailed the active microbiota landscape in both healthy individuals and COVID-19 patients.
Results: The infection of SARS-CoV-2 could deeply change the lung microbiota, evidenced by the α-diversity, β-diversity, and species composition analysis based on bacterial microbiota and virome. Pathogens (e.g., Klebsiella oxytoca causing pneumonia as well), immunomodulatory probiotics (e.g., lactic acid bacteria and Faecalibacterium prausnitzii, a butyrate producer), and Tobacco mosaic virus (TMV) were enriched in the COVID-19 group, suggesting a severe microbiota dysbiosis. The significant correlation between Rothia mucilaginosa, TMV, and SARS-CoV-2 revealed drastic inflammatory battles between the host, SARS-CoV-2, and other microbes in the lungs. Notably, TMV only existed in the COVID-19 group, while human respirovirus 3 (HRV 3) only existed in the healthy group. Our study provides insights into the active microbiota in the lungs of COVID-19 patients and would contribute to the understanding of the infection mechanism of SARS-CoV-2 and the treatment of the disease and complications.
Conclusion: SARS-COV-2 infection deeply altered the lung microbiota of COVID-19 patients. The enrichment of several other pathogens, immunomodulatory probiotics (lactic acid or butyrate producers), and TMV in the COVID-19 group suggests a complex and active lung microbiota disorder.
Collapse
Affiliation(s)
- Yang Han
- Key Laboratory of Biomedical Engineering and Translational Medicine, Ministry of Industry and Information Technology, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China.,Beijing Key Laboratory for Precision Medicine of Chronic Heart Failure, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Zhilong Jia
- Key Laboratory of Biomedical Engineering and Translational Medicine, Ministry of Industry and Information Technology, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China.,Beijing Key Laboratory for Precision Medicine of Chronic Heart Failure, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Jinlong Shi
- Key Laboratory of Biomedical Engineering and Translational Medicine, Ministry of Industry and Information Technology, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China.,Beijing Key Laboratory for Precision Medicine of Chronic Heart Failure, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Weidong Wang
- Key Laboratory of Biomedical Engineering and Translational Medicine, Ministry of Industry and Information Technology, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China.,Beijing Key Laboratory for Precision Medicine of Chronic Heart Failure, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Kunlun He
- Key Laboratory of Biomedical Engineering and Translational Medicine, Ministry of Industry and Information Technology, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China.,Beijing Key Laboratory for Precision Medicine of Chronic Heart Failure, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
29
|
Zou P, Cao P, Liu J, Li P, Luan Q. Comparisons of the killing effect of direct current partially mediated by reactive oxygen species on Porphyromonas gingivalis and Prevotella intermedia in planktonic state and biofilm state - an in vitro study. J Dent Sci 2022; 17:459-467. [PMID: 35028071 PMCID: PMC8739843 DOI: 10.1016/j.jds.2021.07.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 07/30/2021] [Indexed: 12/11/2022] Open
Abstract
Background/purpose Bacterial biofilms formed on the surface of tissues and biomaterials are major causes of chronic infections in humans. Among them, Porphyromonas gingivalis (P. gingivalis) and Prevotella intermedia (P. intermedia) are anaerobic pathogens causing dental infections associated with periodontitis. In this study, we evaluated the killing effect and underlying mechanisms of direct current (DC) as an antimicrobial method in vitro. Materials and methods We chose P. gingivalis and P. intermedia in different states to make comparisons of the killing effect of DC. By viable bacteria counting, fluorescent live/dead staining, reactive oxygen species (ROS) assay, addition of ROS scavenger DMTU and mRNA expression assay of ROS scavenging genes, the role of ROS in the killing effect was explored. Results The planktonic and biofilm states of two bacteria could be effectively killed by low-intensity DC. For the killing effect of 1000 μA DC, there were significant differences whether on planktonic P. gingivalis and P. intermedia (mean killing values: 2.40 vs 2.62 log10 CFU/mL) or on biofilm state of those (mean killing values: 0.63 vs 0.98 log10 CFU/mL). 1000 μA DC greatly induced ROS production and the mRNA expression of ROS scavenging genes. DMTU could partially decrease the killing values of DC and downregulate corresponding gene’s expression. Conclusion 1000 μA DC can kill P. gingivalis and P. intermedia in two states by promoting overproduction of ROS, and P. intermedia is more sensitive to DC than P. gingivalis. These findings indicate low-intensity DC may be a promising approach in treating periodontal infections.
Collapse
Affiliation(s)
- Peihui Zou
- Department of Periodontology, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, China
- Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, China
| | - Pei Cao
- Department of Periodontology, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, China
- Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, China
| | - Jia Liu
- Department of Periodontology, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, China
- Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, China
| | - Peng Li
- Department of Periodontology, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, China
- Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, China
| | - Qingxian Luan
- Department of Periodontology, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, China
- Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, China
- Corresponding author. Department of Periodontology, School and Hospital of Stomatology, Peking University, NO.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR, China.
| |
Collapse
|
30
|
Abstract
Porphyromonas gingivalis is an important human pathogen and also a model organism for the Bacteroidetes phylum. O-glycosylation has been reported in this phylum with findings that include the O-glycosylation motif, the structure of the O-glycans in a few species, and an extensive O-glycoproteome analysis in Tannerella forsythia. However, O-glycosylation has not yet been confirmed in P. gingivalis. We therefore used glycoproteomics approaches including partial deglycosylation with trifluoromethanesulfonic acid as well as both HILIC and FAIMS based glycopeptide enrichment strategies leading to the identification of 257 putative glycosylation sites in 145 glycoproteins. The sequence of the major O-glycan was elucidated to be HexNAc-HexNAc(P-Gro-[Ac]0-2)-dHex-Hex-HexA-Hex(dHex). Western blot analyses of mutants lacking the glycosyltransferases PGN_1134 and PGN_1135 demonstrated their involvement in the biosynthesis of the glycan while mass spectrometry analysis of the truncated O-glycans suggested that PGN_1134 and PGN_1135 transfer the two HexNAc sugars. Interestingly, a strong bias against the O-glycosylation of abundant proteins exposed to the cell surface such as abundant T9SS cargo proteins, surface lipoproteins, and outer membrane β-barrel proteins was observed. In contrast, the great majority of proteins associated with the inner membrane or periplasm were glycosylated irrespective of their abundance. The P. gingivalis O-glycosylation system may therefore function to establish the desired physicochemical properties of the periplasm. IMPORTANCEPorphyromonas gingivalis is an oral pathogen primarily associated with severe periodontal disease and further associated with rheumatoid arthritis, dementia, cardiovascular disease, and certain cancers. Protein glycosylation can be important for a variety of reasons including protein function, solubility, protease resistance, and thermodynamic stability. This study has for the first time demonstrated the presence of O-linked glycosylation in this organism by determining the basic structure of the O-glycans and identifying 257 glycosylation sites in 145 proteins. It was found that most proteins exposed to the periplasm were O-glycosylated; however, the abundant surface exposed proteins were not. The O-glycans consisted of seven monosaccharides and a glycerol phosphate with 0–2 acetyl groups. These glycans are likely to have a stabilizing role to the proteins that bear them and must be taken into account when the proteins are produced in heterologous organisms.
Collapse
|
31
|
Zou P, Li P, Liu J, Cao P, Luan Q. Direct current exerts electricidal and bioelectric effects on Porphyromonas gingivalis biofilms partially via promoting oxidative stress and antibiotic transport. J Microbiol 2021; 60:70-78. [PMID: 34826101 DOI: 10.1007/s12275-022-1238-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 10/05/2021] [Accepted: 10/12/2021] [Indexed: 01/18/2023]
Abstract
Low electric current can inhibit certain microbial biofilms and enhance the efficacy of antimicrobials against them. This study investigated the electricidal and bioelectric effects of direct current (DC) against Porphyromonas gingivalis biofilms as well as the underlying mechanisms. Here, we firstly showed that DC significantly suppressed biofilm formation of P. gingivalis in time- and intensity-dependent manners, and markedly inhibited preformed P. gingivalis biofilms. Moreover, DC enhanced the killing efficacy of metronidazole (MTZ) and amoxicillin with clavulanate potassium (AMC) against the biofilms. Notably, DC-treated biofilms displayed upregulated intracellular ROS and expression of ROS related genes (sod, feoB, and oxyR) as well as porin gene. Interestingly, DC-induced killing of biofilms was partially reversed by ROS scavenger N-dimethylthiourea (DMTU), and the synergistic effect of DC with MTZ/AMC was weakened by small interfering RNA of porin gene (si-Porin). In conclusion, DC can exert electricidal and bioelectric effects against P. gingivalis biofilms partially via promotion of oxidative stress and antibiotic transport, which offers a promising approach for effective management of periodontitis.
Collapse
Affiliation(s)
- Peihui Zou
- Department of Periodontology, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, 100081, P. R. China.,Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, P. R. China
| | - Peng Li
- Department of Periodontology, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, 100081, P. R. China.,Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, P. R. China
| | - Jia Liu
- Department of Periodontology, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, 100081, P. R. China.,Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, P. R. China
| | - Pei Cao
- Department of Periodontology, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, 100081, P. R. China.,Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, P. R. China
| | - Qingxian Luan
- Department of Periodontology, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, 100081, P. R. China. .,Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, P. R. China.
| |
Collapse
|
32
|
Bachtiar EW, Septiwidyati TR. Possible Role of Porphyromonas gingivalis in the Regulation of E2F1, CDK11, and iNOS Gene Expression in Neuronal Cell Cycle: A Preliminary Study. J Int Soc Prev Community Dent 2021; 11:582-587. [PMID: 34760804 PMCID: PMC8533040 DOI: 10.4103/jispcd.jispcd_108_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/30/2021] [Accepted: 07/26/2021] [Indexed: 12/04/2022] Open
Abstract
Objective: This study aimed at evaluating the in vitro effect of Porphyromonas gingivalis exposure in gene expression of E2F1 (family of transcription factors), cyclin-dependent kinase-1 (CDK11), and inducible nitric oxide synthase (iNOS) of the neuronal cell cycle. Materials and Methods: The culture of neuronal cell line SH-SY5Y was exposed to P. gingivalis ATCC 33277, and the gene expression of E2F1, CDK11, and iNOS was analyzed by using a real-time polymerase chain reaction. Results: It was shown that E2F1, a G1 phase biomarker and transcription factor, was upregulated in neuronal cells exposed to P. gingivalis compared with that in control cells. However, CDK11, a biomarker of G2/M checkpoint and iNOS, was downregulated in neuronal cells exposed to P. gingivalis compared with that in control cells. Conclusions: P. gingivalis can regulate the neuronal cell cycle, as indicated in the E2F1, CDK11, and iNOS gene expression.
Collapse
Affiliation(s)
- Endang W Bachtiar
- Department of Oral Biology and Oral Science Research Center, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - Tienneke R Septiwidyati
- Department of Oral Biology and Oral Science Research Center, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| |
Collapse
|
33
|
Hasegawa Y, Nagano K. Porphyromonas gingivalis FimA and Mfa1 fimbriae: Current insights on localization, function, biogenesis, and genotype. JAPANESE DENTAL SCIENCE REVIEW 2021; 57:190-200. [PMID: 34691295 PMCID: PMC8512630 DOI: 10.1016/j.jdsr.2021.09.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/16/2021] [Accepted: 09/21/2021] [Indexed: 11/16/2022] Open
Abstract
In general, the periodontal pathogen Porphyromonas gingivalis expresses distinct FimA and Mfa1 fimbriae. Each of these consists of five FimA–E and five Mfa1–5 proteins encoded by the fim and mfa gene clusters, respectively. The main shaft portion comprises FimA and Mfa1, whereas FimB and Mfa2 are localized on the basal portion and function as anchors and elongation terminators. FimC–E and Mfa3–5 participate in the assembly of an accessory protein complex on the tips of each fimbria. Hence, they serve as ligands for the receptors on host cells and other oral bacterial species. The crystal structures of FimA and Mfa1 fimbrial proteins were recently elucidated and new insights into the localization, function, and biogenesis of these proteins have been reported. Several studies indicated a correlation between P. gingivalis pathogenicity and the fimA genotype but not the mfa1 genotype. We recently revealed polymorphisms of all genes in the fim and mfa gene clusters. Intriguingly, mfa5 occurred in numerous different forms and underwent duplication. Detailed structural and functional knowledge of the fimbrial proteins in the context of the entire filament could facilitate the development of innovative therapeutic strategies for structure-based drug design.
Collapse
Affiliation(s)
- Yoshiaki Hasegawa
- Department of Microbiology, School of Dentistry, Aichi Gakuin University, Nagoya, Japan
| | - Keiji Nagano
- Division of Microbiology, Department of Oral Biology, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, Japan
| |
Collapse
|
34
|
Ali Mohammed MM, Pettersen VK, Nerland AH, Wiker HG, Bakken V. Label-free quantitative proteomic analysis of the oral bacteria Fusobacterium nucleatum and Porphyromonas gingivalis to identify protein features relevant in biofilm formation. Anaerobe 2021; 72:102449. [PMID: 34543761 DOI: 10.1016/j.anaerobe.2021.102449] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 07/24/2021] [Accepted: 09/14/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND The opportunistic pathogens Fusobacterium nucleatum and Porphyromonas gingivalis are Gram-negative bacteria associated with oral biofilm and periodontal disease. This study investigated interactions between F. nucleatum and P. gingivalis proteomes with the objective to identify proteins relevant in biofilm formation. METHODS We applied liquid chromatography-tandem mass spectrometry to determine the expressed proteome of F. nucleatum and P. gingivalis cells grown in biofilm or planktonic culture, and as mono- and dual-species models. The detected proteins were classified into functional categories and their label-free quantitative (LFQ) intensities statistically compared. RESULTS The proteomic analyses detected 1,322 F. nucleatum and 966 P. gingivalis proteins, including abundant virulence factors. Using univariate statistics, we identified significant changes between biofilm and planktonic culture (p-value ≤0.05) in 0,4% F. nucleatum, 7% P. gingivalis, and 14% of all proteins in the dual-species model. For both species, proteins involved in vitamin B2 (riboflavin) metabolism had significantly increased levels in biofilm. In both mono- and dual-species biofilms, P. gingivalis increased the production of proteins for translation, oxidation-reduction, and amino acid metabolism compared to planktonic cultures. However, when we compared LFQ intensities between mono- and dual-species, over 90% of the significantly changed P. gingivalis proteins had their levels reduced in biofilm and planktonic settings of the dual-species model. CONCLUSIONS The findings suggest that P. gingivalis reduces the production of multiple proteins because of the F. nucleatum presence. The results highlight the complex interactions of bacteria contributing to oral biofilms, which need to be considered in the design of prevention strategies.
Collapse
Affiliation(s)
| | | | - Audun H Nerland
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway.
| | - Harald G Wiker
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway.
| | - Vidar Bakken
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway.
| |
Collapse
|
35
|
Sakae K, Nagano K, Furuhashi M, Hasegawa Y. Diversity analysis of genes encoding Mfa1 fimbrial components in Porphyromonas gingivalis strains. PLoS One 2021; 16:e0255111. [PMID: 34310632 PMCID: PMC8313007 DOI: 10.1371/journal.pone.0255111] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 07/08/2021] [Indexed: 12/18/2022] Open
Abstract
Porphyromonas gingivalis, a gram-negative anaerobic bacterium, is associated with the development of periodontal disease. The genetic diversity in virulence factors, such as adhesive fimbriae, among its strains affects the bacterial pathogenicity. P. gingivalis generally expresses two distinct types of fimbriae, FimA and Mfa1. Although the genetic diversity of fimA, encoding the major FimA fimbrilin protein, has been characterized, the genes encoding the Mfa1 fimbrial components, including the Mfa1 to Mfa5 proteins, have not been fully studied. We, therefore, analyzed their genotypes in 12 uncharacterized and 62 known strains of P. gingivalis (74 strains in total). The mfa1 genotype was primarily classified into two genotypes, 53 and 70. Additionally, we found that genotype 70 could be further divided into two subtypes (70A and 70B). The diversity of mfa2 to mfa4 was consistent with the mfa1 genotype, although no subtype in genotype 70 was observed. Protein structure modeling showed high homology between the genotypes in Mfa1 to Mfa4. The mfa5 gene was classified into five genotypes (A to E) independent of other genotypes. Moreover, genotype A was further divided into two subtypes (A1 and A2). Surprisingly, some strains had two mfa5 genes, and the 2ndmfa5 exclusively occurred in genotype E. The Mfa5 protein in all genotypes showed a homologous C-terminal half, including the conserved C-terminal domain recognized by the type IX secretion system. Furthermore, the von Willebrand factor domain at the N-terminal was detected only in genotypes A to C. The mfa1 genotypes partially correlated with the ragA and ragB genotypes (located immediately downstream of the mfa gene cluster) but not with the fimA genotypes.
Collapse
Affiliation(s)
- Kotaro Sakae
- Department of Microbiology, School of Dentistry, Aichi Gakuin University, Nagoya, Japan
- Department of Endodontics, School of Dentistry, Aichi Gakuin University, Nagoya, Japan
| | - Keiji Nagano
- Division of Microbiology, Department of Oral Biology, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, Japan
- * E-mail:
| | - Miyuna Furuhashi
- Department of Pediatric Dentistry, School of Dentistry, Aichi-Gakuin University, Nagoya, Japan
| | - Yoshiaki Hasegawa
- Department of Microbiology, School of Dentistry, Aichi Gakuin University, Nagoya, Japan
| |
Collapse
|
36
|
Kajitani R, Yoshimura D, Ogura Y, Gotoh Y, Hayashi T, Itoh T. Platanus_B: an accurate de novo assembler for bacterial genomes using an iterative error-removal process. DNA Res 2021; 27:5870828. [PMID: 32658266 PMCID: PMC7433917 DOI: 10.1093/dnares/dsaa014] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 07/07/2020] [Indexed: 11/14/2022] Open
Abstract
De novo assembly of short DNA reads remains an essential technology, especially for large-scale projects and high-resolution variant analyses in epidemiology. However, the existing tools often lack sufficient accuracy required to compare closely related strains. To facilitate such studies on bacterial genomes, we developed Platanus_B, a de novo assembler that employs iterations of multiple error-removal algorithms. The benchmarks demonstrated the superior accuracy and high contiguity of Platanus_B, in addition to its ability to enhance the hybrid assembly of both short and nanopore long reads. Although the hybrid strategies for short and long reads were effective in achieving near full-length genomes, we found that short-read-only assemblies generated with Platanus_B were sufficient to obtain ≥90% of exact coding sequences in most cases. In addition, while nanopore long-read-only assemblies lacked fine-scale accuracies, inclusion of short reads was effective in improving the accuracies. Platanus_B can, therefore, be used for comprehensive genomic surveillances of bacterial pathogens and high-resolution phylogenomic analyses of a wide range of bacteria.
Collapse
Affiliation(s)
- Rei Kajitani
- School of Life Science and Technology, Tokyo Institute of Technology, Tokyo 152-8550, Japan
| | - Dai Yoshimura
- School of Life Science and Technology, Tokyo Institute of Technology, Tokyo 152-8550, Japan
| | - Yoshitoshi Ogura
- Division of Microbiology, Department of Infectious Medicine, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan.,Department of Bacteriology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Yasuhiro Gotoh
- Department of Bacteriology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Tetsuya Hayashi
- Department of Bacteriology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Takehiko Itoh
- School of Life Science and Technology, Tokyo Institute of Technology, Tokyo 152-8550, Japan
| |
Collapse
|
37
|
Witkowska E, Łasica AM, Niciński K, Potempa J, Kamińska A. In Search of Spectroscopic Signatures of Periodontitis: A SERS-Based Magnetomicrofluidic Sensor for Detection of Porphyromonas gingivalis and Aggregatibacter actinomycetemcomitans. ACS Sens 2021; 6:1621-1635. [PMID: 33792284 PMCID: PMC8155661 DOI: 10.1021/acssensors.1c00166] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
![]()
Recently, Porphyromonas gingivalis, the keystone pathogen implicated
in the development of gum disease
(periodontitis), was detected in the brains of Alzheimer’s
disease patients, opening up a fascinating possibility that it is
also involved in the pathobiology of this neurodegenerative illness.
To verify this hypothesis, an unbiased, specific, and sensitive method
to detect this pathogen in biological specimens is needed. To this end, our interdisciplinary
studies demonstrate that P. gingivalis can be easily identified by surface-enhanced Raman scattering (SERS).
Moreover, based on SERS measurements, P. gingivalis can be distinguished from another common periodontal pathogen, Aggregatibacter actinomycetemcomitans, and also from
ubiquitous oral Streptococcus spp.
The results were confirmed by principal component analysis (PCA).
Furthermore, we have shown that different P. gingivalis and A. actinomycetemcomitans strains
can easily adsorb to silver-coated magnetic nanoparticles (Fe2O3@AgNPs). Thus, it is possible to magnetically
separate investigated bacteria from other components of a specimen
using the microfluidic chip. To obtain additional enhancement of the
Raman signal, the NPs adsorbed to bacterial cells were magnetically
attracted to the Si/Ag SERS platform. Afterward, the SERS spectra
could be recorded. Such a time-saving procedure can be very helpful
in rapid medical diagnostics and thus in starting the appropriate
pharmacological therapy to prevent the development of periodontitis
and associated comorbidities, e.g., Alzheimerʼs disease.
Collapse
Affiliation(s)
- Evelin Witkowska
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| | - Anna M. Łasica
- Department of Bacterial Genetics, Institute of Microbiology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland
| | - Krzysztof Niciński
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| | - Jan Potempa
- Department of Microbiology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
- Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, 501 S. Preston Street, Louisville, Kentucky 40202, United States
| | - Agnieszka Kamińska
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| |
Collapse
|
38
|
Nara PL, Sindelar D, Penn MS, Potempa J, Griffin WST. Porphyromonas gingivalis Outer Membrane Vesicles as the Major Driver of and Explanation for Neuropathogenesis, the Cholinergic Hypothesis, Iron Dyshomeostasis, and Salivary Lactoferrin in Alzheimer's Disease. J Alzheimers Dis 2021; 82:1417-1450. [PMID: 34275903 PMCID: PMC8461682 DOI: 10.3233/jad-210448] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2021] [Indexed: 12/22/2022]
Abstract
Porphyromonas gingivalis (Pg) is a primary oral pathogen in the widespread biofilm-induced "chronic" multi-systems inflammatory disease(s) including Alzheimer's disease (AD). It is possibly the only second identified unique example of a biological extremophile in the human body. Having a better understanding of the key microbiological and genetic mechanisms of its pathogenesis and disease induction are central to its future diagnosis, treatment, and possible prevention. The published literature around the role of Pg in AD highlights the bacteria's direct role within the brain to cause disease. The available evidence, although somewhat adopted, does not fully support this as the major process. There are alternative pathogenic/virulence features associated with Pg that have been overlooked and may better explain the pathogenic processes found in the "infection hypothesis" of AD. A better explanation is offered here for the discrepancy in the relatively low amounts of "Pg bacteria" residing in the brain compared to the rather florid amounts and broad distribution of one or more of its major bacterial protein toxins. Related to this, the "Gingipains Hypothesis", AD-related iron dyshomeostasis, and the early reduced salivary lactoferrin, along with the resurrection of the Cholinergic Hypothesis may now be integrated into one working model. The current paper suggests the highly evolved and developed Type IX secretory cargo system of Pg producing outer membrane vesicles may better explain the observed diseases. Thus it is hoped this paper can provide a unifying model for the sporadic form of AD and guide the direction of research, treatment, and possible prevention.
Collapse
Affiliation(s)
| | | | - Marc S. Penn
- Summa Heart Health and Vascular Institute, Akron, OH, USA
| | - Jan Potempa
- Department of Oral Immunology and Infectious Diseases in the School of Dentistry, University of Louisville, Louisville, KY, USA
| | - W. Sue T. Griffin
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
39
|
Takizawa S, Asano R, Fukuda Y, Feng M, Baba Y, Abe K, Tada C, Nakai Y. Change of Endoglucanase Activity and Rumen Microbial Community During Biodegradation of Cellulose Using Rumen Microbiota. Front Microbiol 2020; 11:603818. [PMID: 33391225 PMCID: PMC7775302 DOI: 10.3389/fmicb.2020.603818] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/26/2020] [Indexed: 12/20/2022] Open
Abstract
Treatment with rumen microorganisms improves the methane fermentation of undegradable lignocellulosic biomass; however, the role of endoglucanase in lignocellulose digestion remains unclear. This study was conducted to investigate endoglucanases contributing to cellulose degradation during treatment with rumen microorganisms, using carboxymethyl cellulose (CMC) as a substrate. The rate of CMC degradation increased for the first 24 h of treatment. Zymogram analysis revealed that endoglucanases of 52 and 53 kDa exhibited high enzyme activity for the first 12 h, whereas endoglucanases of 42, 50, and 101 kDa exhibited high enzyme activities from 12 to 24 h. This indicates that the activities of these five endoglucanases shifted and contributed to efficient CMC degradation. Metagenomic analysis revealed that the relative abundances of Selenomonas, Eudiplodinium, and Metadinium decreased after 12 h, which was positively correlated with the 52- and 53-kDa endoglucanases. Additionally, the relative abundances of Porphyromonas, Didinium, unclassified Bacteroidetes, Clostridiales family XI, Lachnospiraceae and Sphingobacteriaceae increased for the first 24 h, which was positively correlated with endoglucanases of 42, 50, and 101 kDa. This study suggests that uncharacterized and non-dominant microorganisms produce and/or contribute to activity of 40, 50, 52, 53, and 101 kDa endoglucanases, enhancing CMC degradation during treatment with rumen microorganisms.
Collapse
Affiliation(s)
- Shuhei Takizawa
- Laboratory of Sustainable Animal Environment, Graduate School of Agricultural Science, Tohoku University, Osaki, Japan.,Japan Society for the Promotion of Science, Chiyoda-ku, Japan
| | - Ryoki Asano
- Department of Agro-Food Science, Faculty of Agro-Food Science, Niigata Agro-Food University, Tainai, Japan
| | - Yasuhiro Fukuda
- Laboratory of Sustainable Animal Environment, Graduate School of Agricultural Science, Tohoku University, Osaki, Japan
| | - Mengjia Feng
- Laboratory of Sustainable Animal Environment, Graduate School of Agricultural Science, Tohoku University, Osaki, Japan
| | - Yasunori Baba
- Research Institute for Bioresources and Biotechnology, Ishikawa Prefectural University, Nonoichi, Japan
| | - Kenichi Abe
- Department of Agro-Food Science, Faculty of Agro-Food Science, Niigata Agro-Food University, Tainai, Japan
| | - Chika Tada
- Laboratory of Sustainable Animal Environment, Graduate School of Agricultural Science, Tohoku University, Osaki, Japan
| | - Yutaka Nakai
- Department of Agro-Food Science, Faculty of Agro-Food Science, Niigata Agro-Food University, Tainai, Japan
| |
Collapse
|
40
|
Seers CA, Mahmud ASM, Huq NL, Cross KJ, Reynolds EC. Porphyromonas gingivalis laboratory strains and clinical isolates exhibit different distribution of cell surface and secreted gingipains. J Oral Microbiol 2020; 13:1858001. [PMID: 33391630 PMCID: PMC7733959 DOI: 10.1080/20002297.2020.1858001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: The cell-surface cysteine proteinases RgpA, RgpB (Arg-gingipain), and Kgp (Lys-gingipain) are major virulence factors of P. gingivalis, a keystone pathogen in the development of destructive periodontal disease. The gingipains function as proteinases and transpeptidases utilising small peptides such as glycylglycine as acceptor molecules. However, the characteristics of the gingipains from most P. gingivalis strains have not been determined. Methods: We determined the phenotypes of a panel of P. gingivalis laboratory strains and global clinical isolates with respect to growth on blood agar plus whole-cell and vesicle-free culture supernatant (VFSN) Arg- and Lys-specific proteinase activities. Results: The P. gingivalis isolates exhibited different growth characteristics and hydrolysis of haemoglobin in solid media. Whole-cell Arg-gingipain Vmax varied 5.8-fold and the whole cell Lys-gingipain Vmax varied 2.1-fold across the strains. Furthermore, the P. gingivalis strains showed more than 107-fold variance in soluble Arg-gingipain activity in VFSN and more than 371-fold variance in soluble Lys-gingipain activity in VFSN. Glycylglycine and cysteine stimulated Arg- and Lys-specific cleavage activities of all strains. The stimulation by cysteine was in addition to its redox effect consistent with both glycylglycine and cysteine promoting transpeptidation.
Conclusion: The global P. gingivalis clinical isolates exhibit different Arg- and Lys‑gingipain activities with substantial variability in the level of soluble proteinases released into the environment.
Collapse
Affiliation(s)
- Christine A Seers
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Melbourne, Australia
| | - A Sayeed M Mahmud
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Melbourne, Australia
| | - N Laila Huq
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Melbourne, Australia
| | - Keith J Cross
- 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
|
41
|
PorA, a conserved C-terminal domain-containing protein, impacts the PorXY-SigP signaling of the type IX secretion system. Sci Rep 2020; 10:21109. [PMID: 33273542 PMCID: PMC7712824 DOI: 10.1038/s41598-020-77987-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 11/19/2020] [Indexed: 01/07/2023] Open
Abstract
Porphyromonas gingivalis, a periodontal pathogen, translocates many virulence factors including the cysteine proteases referred to as gingipains to the cell surface via the type IX secretion system (T9SS). Expression of the T9SS component proteins is regulated by the tandem signaling of the PorXY two-component system and the ECF sigma factor SigP. However, the details of this regulatory pathway are still unknown. We found that one of the T9SS conserved C-terminal domain-containing proteins, PGN_0123, which we have designated PorA, is involved in regulating expression of genes encoding T9SS structural proteins and that PorA can be translocated onto the cell surface without the T9SS translocation machinery. X-ray crystallography revealed that PorA has a domain similar to the mannose-binding domain of Escherichia coli FimH, the tip protein of Type 1 pilus. Mutations in the cytoplasmic domain of the sensor kinase PorY conferred phenotypic recovery on the ΔporA mutant. The SigP sigma factor, which is activated by the PorXY two-component system, markedly decreased in the ΔporA mutant. These results strongly support a potential role for PorA in relaying a signal from the cell surface to the PorXY-SigP signaling pathway.
Collapse
|
42
|
Nemoto TK, Ohara Nemoto Y. Dipeptidyl-peptidases: Key enzymes producing entry forms of extracellular proteins in asaccharolytic periodontopathic bacterium Porphyromonas gingivalis. Mol Oral Microbiol 2020; 36:145-156. [PMID: 33006264 PMCID: PMC8048996 DOI: 10.1111/omi.12317] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/17/2020] [Accepted: 09/24/2020] [Indexed: 02/06/2023]
Abstract
Porphyromonas gingivalis, a pathogen of chronic periodontitis, is an asaccharolytic microorganism that solely utilizes nutritional amino acids as its energy source and cellular constituents. The bacterium is considered to incorporate proteinaceous nutrients mainly as dipeptides, thus exopeptidases that produce dipeptides from polypeptides are critical for survival and proliferation. We present here an overview of dipeptide production by P. gingivalis mediated by dipeptidyl-peptidases (DPPs), e.g., DPP4, DPP5, DPP7, and DPP11, serine exopeptidases localized in periplasm, which release dipeptides from the N-terminus of polypeptides. Additionally, two other exopeptidases, acylpeptidyl-oligopeptidase (AOP) and prolyl tripeptidyl-peptidase A (PTP-A), which liberate N-terminal acylated di-/tri-peptides and tripeptides with Pro at the third position, respectively, provide polypeptides in an acceptable form for DPPs. Hence, a large fraction of dipeptides is produced from nutritional polypeptides by DPPs with differential specificities in combination with AOP and PTP-A. The resultant dipeptides are then incorporated across the inner membrane mainly via a proton-dependent oligopeptide transporter (POT), a member of the major facilitator superfamily. Recent studies also indicate that DPP4 and DPP7 directly link between periodontal and systemic diseases, such as type 2 diabetes mellitus and coagulation abnormality, respectively. Therefore, these dipeptide-producing and incorporation molecules are considered to be potent targets for prevention and treatment of periodontal and related systemic diseases.
Collapse
Affiliation(s)
- Takayuki K Nemoto
- Department of Oral Molecular Biology, 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
| |
Collapse
|
43
|
Shen D, Perpich JD, Stocke KS, Yakoumatos L, Fitzsimonds ZR, Liu C, Miller DP, Lamont RJ. Role of the RprY response regulator in P. gingivalis community development and virulence. Mol Oral Microbiol 2020; 35:231-239. [PMID: 32940001 DOI: 10.1111/omi.12311] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/07/2020] [Accepted: 09/01/2020] [Indexed: 12/27/2022]
Abstract
Porphyromonas gingivalis expresses a limited number of two-component systems, including RprY, an orphan response regulator which lacks a cognate sensor kinase. In this study, we examined cross-phosphorylation of RprY on tyrosine residues and its importance for RprY function. We show that RprY reacts with phosphotyrosine antibodies, and found that the tyrosine (Y) residue at position 41 is predicted to be solvent accessible. Loss of RprY increased the level of heterotypic community development with Streptococcus gordonii, and the community-suppressive function of RprY required Y41. Expression of the Mfa1 fimbrial adhesin was increased in the rprY mutant and in the mutant complemented with rprY containing a Y41F mutation. In a microscale thermophoresis assay, recombinant RprY protein bound to the promoter region of mfa1, and binding was diminished with RprY containing the Y41F substitution. RprY was required for virulence of P. gingivalis in a murine model of alveolar bone loss. Transcriptional profiling indicated that RprY can control the expression of genes encoding the type IX secretion system (T9SS) machinery and virulence factors secreted through the T9SS, including the gingipain proteases and peptidylarginine deiminase (PPAD). Collectively, these results establish the RprY response regulator as a component of the tyrosine phosphorylation regulon in P. gingivalis, which can independently control heterotypic community development through the Mfa1 fimbriae and virulence through the T9SS.
Collapse
Affiliation(s)
- Daonan Shen
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| | - John D Perpich
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| | - Kendall S Stocke
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| | - Lan Yakoumatos
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| | - Zackary R Fitzsimonds
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| | - Chengcheng Liu
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| | - Daniel P Miller
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| | - Richard J Lamont
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| |
Collapse
|
44
|
Saiki K, Urano-Tashiro Y, Takahashi Y. Reassessment of minimal media reveals differences in growth among Porphyromonas gingivalis standard strains. J Oral Biosci 2020; 62:315-321. [PMID: 32937181 DOI: 10.1016/j.job.2020.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/04/2020] [Accepted: 09/04/2020] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Porphyromonas gingivalis is one of the etiologic agents of chronic periodontitis. Our previous study showed that the use of minimal media for P. gingivalis allowed to isolate novel inhibitors of P. gingivalis growth. However, growth of P. gingivalis in minimal media was not always reproducible. METHODS To explain this phenomenon, we analyzed the growth of seven wild-type ATCC 33277 strains and two wild-type W83 strains in 10 minimal media and three complex media. RESULTS All nine strains grew in LF (Lactalbumin-Ferric chloride), GC (bovine γ-immunoglobulin G-Calcium chloride), and newly developed mC (milk-Casein) minimal media. Therefore, LF, GC, and mC could be used as minimal media for P. gingivalis. In contrast, other six minimal media containing bovine serum albumin (BSA) supported the growth of several less strains; among these, two media also showed lack of reproducibility in growth among ATCC 33277 strains. On the other hand, four ATCC 33277 strains grew similarly in all 13 media, but two W83 and other three ATCC 33277 strains grew differently in at least one medium. CONCLUSIONS These results suggest that the lack of reproducibility of P. gingivalis growth on minimal media is caused by the presence of BSA, and by differences among the standard strains of P. gingivalis.
Collapse
Affiliation(s)
- Keitarou Saiki
- Department of Microbiology, Nippon Dental University School of Life Dentistry at Tokyo, Tokyo, Japan.
| | - Yumiko Urano-Tashiro
- Department of Microbiology, Nippon Dental University School of Life Dentistry at Tokyo, Tokyo, Japan.
| | - Yukihiro Takahashi
- Department of Microbiology, Nippon Dental University School of Life Dentistry at Tokyo, Tokyo, Japan.
| |
Collapse
|
45
|
Shoji M, Shibata S, Sueyoshi T, Naito M, Nakayama K. Biogenesis of Type V pili. Microbiol Immunol 2020; 64:643-656. [DOI: 10.1111/1348-0421.12838] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/11/2020] [Accepted: 08/13/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Mikio Shoji
- Department of Microbiology and Oral Infection, Graduate School of Biomedical Sciences Nagasaki University Nagasaki Nagasaki Japan
| | - Satoshi Shibata
- Molecular Cryo‐Electron Microscopy Unit Okinawa Institute of Science and Technology Graduate University Onna Okinawa Japan
| | - Takayuki Sueyoshi
- Department of Microbiology and Oral Infection, Graduate School of Biomedical Sciences Nagasaki University Nagasaki Nagasaki Japan
| | - Mariko Naito
- Department of Microbiology and Oral Infection, Graduate School of Biomedical Sciences Nagasaki University Nagasaki Nagasaki Japan
| | - Koji Nakayama
- Department of Microbiology and Oral Infection, Graduate School of Biomedical Sciences Nagasaki University Nagasaki Nagasaki Japan
| |
Collapse
|
46
|
Eltigani SA, Eltayeb MM, Bito T, Ichiyanagi T, Ishihara A, Arima J. Argeloside I inhibits the pathogenicity of Porphyromonas gingivalis TDC60. J Biosci Bioeng 2020; 130:644-649. [PMID: 32847740 DOI: 10.1016/j.jbiosc.2020.07.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 07/15/2020] [Accepted: 07/21/2020] [Indexed: 01/05/2023]
Abstract
Porphyromonas gingivalis, a major pathogen associated with chronic periodontitis, produces several virulence agents in the outer cell membrane, including gingipains and hemagglutinins. These virulence factors enable the bacteria to adhere to periodontal tissue and degrade host proteins to obtain the nutrients needed for dental plaque formation. P. gingivalis TDC60 was recently identified as the most aggressive P. gingivalis strain to dates. In this study, we isolated a known pregnane glycoside, argeloside I, from the aqueous extract of Solenostemma argel leaves. Argeloside I completely hindered the growth of P. gingivalis TDC60 and inhibited the production of hemagglutinins as well as Arg- and Lys-specific gingipains. Our results demonstrate a new function of pregnane glycosides. Argeloside I may be a candidate for reducing the risk associated with P. gingivalis TDC60 and its adhesion factors.
Collapse
Affiliation(s)
- Sara A Eltigani
- The United Graduate School of Agricultural Sciences, Tottori University, 4-101 Koyama-Minami, Tottori 680-8553, Japan
| | - Mohamed M Eltayeb
- Arid Land Research Center, Tottori University, 1390 Hamasaka, Tottori 680-0001, Japan; Department of Food Science and Technology, Faculty of Agriculture, University of Khartoum, Shambat, Khartoum North 14413, Sudan
| | - Tomohiro Bito
- Department of Agricultural, Life and Environmental Sciences, Faculty of Agriculture, Tottori University, 4-101 Koyama-Minami, Tottori 680-8553, Japan
| | - Tsuyoshi Ichiyanagi
- Department of Agricultural, Life and Environmental Sciences, Faculty of Agriculture, Tottori University, 4-101 Koyama-Minami, Tottori 680-8553, Japan
| | - Atsushi Ishihara
- Department of Agricultural, Life and Environmental Sciences, Faculty of Agriculture, Tottori University, 4-101 Koyama-Minami, Tottori 680-8553, Japan
| | - Jiro Arima
- Department of Agricultural, Life and Environmental Sciences, Faculty of Agriculture, Tottori University, 4-101 Koyama-Minami, Tottori 680-8553, Japan.
| |
Collapse
|
47
|
Emrizal R, Nor Muhammad NA. Phylogenetic comparison between Type IX Secretion System (T9SS) protein components suggests evidence of horizontal gene transfer. PeerJ 2020; 8:e9019. [PMID: 32617187 PMCID: PMC7323717 DOI: 10.7717/peerj.9019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 03/28/2020] [Indexed: 12/20/2022] Open
Abstract
Porphyromonas gingivalis is one of the major bacteria that causes periodontitis. Chronic periodontitis is a severe form of periodontal disease that ultimately leads to tooth loss. Virulence factors that contribute to periodontitis are secreted by Type IX Secretion System (T9SS). There are aspects of T9SS protein components that have yet to be characterised. Thus, the aim of this study is to investigate the phylogenetic relationship between members of 20 T9SS component protein families. The Bayesian Inference (BI) trees for 19 T9SS protein components exhibit monophyletic clades for all major classes under Bacteroidetes with strong support for the monophyletic clades or its subclades that is consistent with phylogeny exhibited by the constructed BI tree of 16S rRNA. The BI tree of PorR is different from the 19 BI trees of T9SS protein components as it does not exhibit monophyletic clades for all major classes under Bacteroidetes. There is strong support for the phylogeny exhibited by the BI tree of PorR which deviates from the phylogeny based on 16S rRNA. Hence, it is possible that the porR gene is subjected to horizontal transfer as it is known that virulence factor genes could be horizontally transferred. Seven genes (porR included) that are involved in the biosynthesis of A-LPS are found to be flanked by insertion sequences (IS5 family transposons). Therefore, the intervening DNA segment that contains the porR gene might be transposed and subjected to conjugative transfer. Thus, the seven genes can be co-transferred via horizontal gene transfer. The BI tree of UgdA does not exhibit monophyletic clades for all major classes under Bacteroidetes which is similar to the BI tree of PorR (both are a part of the seven genes). Both BI trees also exhibit similar topology as the four identified clusters with strong support and have similar relative positions to each other in both BI trees. This reinforces the possibility that porR and the other six genes might be horizontally transferred. Other than the BI tree of PorR, the 19 other BI trees of T9SS protein components also exhibit evidence of horizontal gene transfer. However, their genes might undergo horizontal gene transfer less frequently compared to porR because the intervening DNA segment that contains porR is easily exchanged between bacteria under Bacteroidetes due to the presence of insertion sequences (IS5 family transposons) that flank it. In conclusion, this study can provide a better understanding about the phylogeny of T9SS protein components.
Collapse
Affiliation(s)
- Reeki Emrizal
- Institute of Systems Biology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | | |
Collapse
|
48
|
Cheng T, Lai YT, Wang C, Wang Y, Jiang N, Li H, Sun H, Jin L. Bismuth drugs tackle Porphyromonas gingivalis and attune cytokine response in human cells. Metallomics 2020; 11:1207-1218. [PMID: 31179464 DOI: 10.1039/c9mt00085b] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Periodontitis is the leading cause of severe tooth loss and edentulism in adults worldwide and is closely linked to systemic conditions such as diabetes and cardiovascular disease. Porphyromonas gingivalis is the key pathogen in periodontitis. Herein, we provided the first evidence that bismuth drugs suppress P. gingivalis in its planktonic, biofilm, and intracellular states. In total, 42 bismuth-associated proteins were identified including its major virulent factors (e.g., gingipains, hemagglutinin HagA, and fimbriae). Bismuth perturbed its iron acquisition, disturbed the energy metabolism and virulence, and deactivated multiple key enzymes (e.g., superoxide dismutase and thioredoxins). Moreover, bismuth inhibited its biofilm formation and disrupted the 3-day matured biofilms. Notably, the internalized P. gingivalis in various human cells (e.g., human gingival epithelium progenitors, HGEPs) was oppressed by bismuth but not the commonly used antibiotic metronidazole. Importantly, bismuth drugs enabled the counteraction of immuno-inflammatory responses in different host cells perturbed by P. gingivalis. The production of IL-6 and IL-8 attenuated by P. gingivalis in both of native and IL-1β-stimulated HGEPs was restored, while the bacterium-enhanced expression of IL-6, IL-1β, and TNFα in THP-1 macrophages was alleviated. This proof-of-concept study brings prospects for the potential reposition of the routinely used anti-Helicobacter pylori bismuth drugs to better manage inflammatory diseases such as periodontitis and P. gingivalis-related complex systemic disorders.
Collapse
Affiliation(s)
- Tianfan Cheng
- Discipline of Periodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China.
| | - Yau-Tsz Lai
- Department of Chemistry, The University of Hong Kong, Hong Kong SAR, China.
| | - Chuan Wang
- Discipline of Periodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China.
| | - Yi Wang
- Discipline of Periodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China.
| | - Nan Jiang
- Department of Chemistry, The University of Hong Kong, Hong Kong SAR, China.
| | - Hongyan Li
- Department of Chemistry, The University of Hong Kong, Hong Kong SAR, China.
| | - Hongzhe Sun
- Department of Chemistry, The University of Hong Kong, Hong Kong SAR, China.
| | - Lijian Jin
- Discipline of Periodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
49
|
Ohara-Nemoto Y, Sarwar MT, Shimoyama Y, Kobayakawa T, Nemoto TK. Preferential dipeptide incorporation of Porphyromonas gingivalis mediated by proton-dependent oligopeptide transporter (Pot). FEMS Microbiol Lett 2020; 367:6041718. [PMID: 33338236 DOI: 10.1093/femsle/fnaa204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 11/24/2020] [Indexed: 11/13/2022] Open
Abstract
Multiple dipeptidyl-peptidases (DPPs) are present in the periplasmic space of Porphyromonas gingivalis, an asaccharolytic periodontopathic bacterium. Dipeptides produced by DPPs are presumed to be transported into the bacterial cells and metabolized to generate energy and cellular components. The present study aimed to identify a transporter responsible for dipeptide uptake in the bacterium. A real-time metabolic analysis demonstrated that P. gingivalis preferentially incorporated Gly-Xaa dipeptides, and then, single amino acids, tripeptides and longer oligopeptides to lesser extents. Heterologous expression of the P. gingivalis serine/threonine transporter (SstT; PGN_1460), oligopeptide transporter (Opt; PGN_1518) and proton-dependent oligopeptide transporter (Pot; PGN_0135) genes demonstrated that Escherichia coli expressing Pot exclusively incorporated Gly-Gly, while SstT managed Ser uptake and Opt was responsible for Gly-Gly-Gly uptake. Dipeptide uptake was significantly decreased in a P. gingivalis Δpot strain and further suppressed in a Δpot-Δopt double-deficient strain. In addition, the growth of the Δpot strain was markedly attenuated and the Δpot-Δopt strain scarcely grew, whereas the ΔsstT strain grew well almost like wild type. Consequently, these results demonstrate that predominant uptake of dipeptide in P. gingivalis is mostly managed by Pot. We thus propose that Pot is a potential therapeutic target of periodontal disease and P. gingivalis-related systemic diseases.
Collapse
Affiliation(s)
- Yuko Ohara-Nemoto
- Department of Oral Molecular Biology, Course of Medical and Dental Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| | - Mohammad Tanvir Sarwar
- Department of Oral Molecular Biology, Course of Medical and Dental Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| | - Yu Shimoyama
- Division of Molecular Microbiology, Department of Microbiology, Iwate Medical University, 1-1-1 Idai-dori, Yahaba-cho, Iwate 028-3694, Japan
| | - Takeshi Kobayakawa
- Department of Oral Molecular Biology, Course of Medical and Dental Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| | - Takayuki K Nemoto
- Department of Oral Molecular Biology, Course of Medical and Dental Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| |
Collapse
|
50
|
Xu W, Zhou W, Wang H, Liang S. Roles of Porphyromonas gingivalis and its virulence factors in periodontitis. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2020; 120:45-84. [PMID: 32085888 DOI: 10.1016/bs.apcsb.2019.12.001] [Citation(s) in RCA: 198] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Periodontitis is an infection-driven inflammatory disease, which is characterized by gingival inflammation and bone loss. Periodontitis is associated with various systemic diseases, including cardiovascular, respiratory, musculoskeletal, and reproductive system related abnormalities. Recent theory attributes the pathogenesis of periodontitis to oral microbial dysbiosis, in which Porphyromonas gingivalis acts as a critical agent by disrupting host immune homeostasis. Lipopolysaccharide, proteases, fimbriae, and some other virulence factors are among the strategies exploited by P. gingivalis to promote the bacterial colonization and facilitate the outgrowth of the surrounding microbial community. Virulence factors promote the coaggregation of P. gingivalis with other bacteria and the formation of dental biofilm. These virulence factors also modulate a variety of host immune components and subvert the immune response to evade bacterial clearance or induce an inflammatory environment. In this chapter, our focus is to discuss the virulence factors of periodontal pathogens, especially P. gingivalis, and their roles in regulating immune responses during periodontitis progression.
Collapse
Affiliation(s)
- Weizhe Xu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China; Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, United States
| | - Wei Zhou
- Department of Endodontics, Ninth People's Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, Pudong, China
| | - Huizhi Wang
- VCU Philips Institute for Oral Health Research, Department of Oral and Craniofacial Molecular Biology, Virginia Commonwealth University School of Dentistry, Richmond, VA, United States
| | - Shuang Liang
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, United States
| |
Collapse
|