1
|
Dou Y, Mishra A, Fletcher HM. Involvement of PG1037 in the repair of 8-oxo-7,8-dihydroguanine caused by oxidative stress in Porphyromonas gingivalis. Mol Oral Microbiol 2024; 39:507-520. [PMID: 39206509 DOI: 10.1111/omi.12482] [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: 06/07/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND The PG1037 gene is part of the uvrA-PG1037-pcrA operon in Porphyromonas gingivalis. It encodes for a protein of unknown function upregulated under hydrogen peroxide (H2O2)-induced oxidative stress. Bioinformatic analysis shows that PG1037 has a zinc-finger motif, two peroxidase motifs, and one cytidylate kinase domain. The aim of this study is to characterize further the role of the PG1037 recombinant protein in the unique 8-oxoG repair system in P. gingivalis. MATERIALS AND METHODS PG1037 recombinant proteins with deletions in the zinc-finger or peroxidase motifs were created. Electrophoretic mobility shift assays were used to evaluate the ability of the recombinant proteins to bind 8-oxoG-containing oligonucleotides. Zinc binding, peroxidase, and Fenton reaction assays were used to assess the functional roles of the rPG1037 protein. A bacterial adenylate cyclase two-bride assay was used to identify the partner protein of PG1037 in the repair of 8-oxoG. RESULTS The recombinant PG1037 (rPG1037) protein carrying an N-terminal His-tag demonstrated an ability to recognize and bind 8-oxoG-containing oligonucleotide. In contrast to the wild-type rPG1037 protein, the zinc-finger motif deletion resulted in the loss of zinc and 8-oxoG binding activities. A deletion of the peroxidase motif-1 showed a decrease in peroxidase activity. Using a bacterial adenylate cyclase two-hybrid system, there was no observed protein-protein interaction of PG1037 with UvrA (PG1036), PcrA (PG1038), or mismatch repair system proteins. CONCLUSIONS Taken together, the results show that PG1037 is an important member of a novel mechanism that recognizes and repairs oxidative stress-induced DNA damage in P. gingivalis.
Collapse
Affiliation(s)
- Yuetan Dou
- Division of Microbiology and Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Arunima Mishra
- Division of Microbiology and Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Hansel M Fletcher
- Division of Microbiology and Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California, USA
| |
Collapse
|
2
|
Tang Y, Qi Y, Chen Y, Wang YQ, Zhang C, Sun Y, Huang C, Zhang XZ. Erythrocyte-Mimicking Nanovesicle Targeting Porphyromonas gingivalis for Periodontitis. ACS NANO 2024. [PMID: 39088785 DOI: 10.1021/acsnano.4c02316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/03/2024]
Abstract
Porphyromonas gingivalis has been demonstrated to have the strongest association with periodontitis. Within the host, P. gingivalis relies on acquiring iron and heme through the aggregation and lysis of erythrocytes, which are important factors in the growth and virulence of P. gingivalis. Additionally, the excess obtained heme is deposited on the surface of P. gingivalis, protecting the cells from oxidative damage. Based on these biological properties of the interaction between P. gingivalis and erythrocytes, this study developed an erythrocyte membrane nanovesicle loaded with gallium porphyrins to mimic erythrocytes. The nanovesicle can target and adhere with P. gingivalis precisely, being lysed and utilized by P. gingivalis as erythrocytes. Ingested gallium porphyrin replaces iron porphyrin in P. gingivalis, causing intracellular metabolic disruption. Deposited porphyrin generates a large amount of reactive oxygen species (ROS) under blue light, causing oxidative damage, and its lethality is enhanced by bacterial metabolic disruption, synergistically killing P. gingivalis. Our results demonstrate that this strategy can target and inhibit P. gingivalis, reduce its invasion of epithelial cells, and alleviate the progression of periodontitis.
Collapse
Affiliation(s)
- Ying Tang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yongdan Qi
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Yang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yu-Qiang Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Cheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Yunxia Sun
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Cui Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, China
| |
Collapse
|
3
|
Lamont RJ, Kuboniwa M. The polymicrobial pathogenicity of Porphyromonas gingivalis. FRONTIERS IN ORAL HEALTH 2024; 5:1404917. [PMID: 38736461 PMCID: PMC11082793 DOI: 10.3389/froh.2024.1404917] [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: 03/21/2024] [Accepted: 04/12/2024] [Indexed: 05/14/2024] Open
Abstract
Accumulating microbiome data and mechanistic studies in vitro and in vivo have refined our understanding of the oral microbiota as a functionally integrated polymicrobial community. Emergent properties of these communities are driven to a large extent by interspecies communication which can be based on physical association, secreted small molecules or nutritional exchange. Porphyromonas gingivalis is a consensus periodontal pathogen; however, virulence is only expressed in the context of a polymicrobial community. Multivalent fimbriae mediate attachment to other oral species which can initiate a distinct transcriptional program in both constituents of the binding pair. P. gingivalis also responds to small molecules and nutritional cues produced by partner organisms. Physiological interdependence forms the basis of complex networks of cooperating organisms which begin to resemble an organismal entity exhibiting a spectrum of pathogenic potential.
Collapse
Affiliation(s)
- Richard J. Lamont
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, KY, United States
| | - Masae Kuboniwa
- Department of Preventive Dentistry, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| |
Collapse
|
4
|
Irfan M, Solbiati J, Duran-Pinedo A, Rocha FG, Gibson FC, Frias-Lopez J. A Porphyromonas gingivalis hypothetical protein controlled by the type I-C CRISPR-Cas system is a novel adhesin important in virulence. mSystems 2024; 9:e0123123. [PMID: 38323815 PMCID: PMC10949514 DOI: 10.1128/msystems.01231-23] [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: 11/17/2023] [Accepted: 12/26/2023] [Indexed: 02/08/2024] Open
Abstract
The ability of many human pathogens to infect requires their ability to adhere to the host surfaces as a first step in the process. Porphyromonas gingivalis, a keystone oral pathogen, uses adhesins to adhere to the surface of the gingival epithelium and other members of the oral microbiome. In a previous study, we identified several proteins potentially linked to virulence whose mRNA levels are regulated by CRISPR-Cas type I-C. Among those, PGN_1547 was highly upregulated in the CRISPR-Cas 3 mutant. PGN_1547 is annotated as a hypothetical protein. Employing homology searching, our data support that PGN_1547 resembles an auto-transporter adhesin of P. gingivalis based on containing the DUF2807 domain. To begin to characterize the function of PGN_1547, we found that a deletion mutant displayed a significant decrease in virulence using a Galleria mellonela model. Furthermore, this mutant was significantly impaired in forming biofilms and attaching to the macrophage-like cell THP-1. Luminex revealed that the PGN_1547 mutant elicited a less robust cytokine and chemokine response from THP-1 cells, and TLR2 predominantly sensed that recombinant PGN_1547. Taken together, these findings broaden our understanding of the toolbox of virulence factors possessed by P. gingivalis. Importantly, PGN_1547, a hypothetical protein, has homologs in another member of the order Bacteroidales whose function is unknown, and our results could shed light on the role of this family of proteins as auto-transport adhesins in this phylogenetic group.IMPORTANCEPeriodontal diseases are among humans' most common infections, and besides their effect on the oral cavity, they have been associated with systemic inflammatory conditions. Among members of the oral microbiome implicated in the development of periodontitis, Porphyromonas gingivalis is considered a keystone pathogen. We have identified a new adhesin that acts as a virulence factor, PGN_1547, which contains the DUF2807 domain, which belongs to the putative auto-transporter adhesin, head GIN domain family. Deletion of this gene lowers the virulence of P. gingivalis and impacts the ability of P. gingivalis to form biofilm and attach to host cells. Furthermore, the broad distribution of these receptors in the order Bacteroidales suggests their importance in colonization by this important group of organisms.
Collapse
Affiliation(s)
- Muhammad Irfan
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, USA
| | - Jose Solbiati
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, USA
| | - Ana Duran-Pinedo
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, USA
| | - Fernanda Godoy Rocha
- Department of Periodontology, College of Dentistry, University of Florida, Gainesville, Florida, USA
| | - Frank C. Gibson
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, USA
| | - Jorge Frias-Lopez
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
5
|
Luo S, Xu T, Zheng Q, Jiang A, Zhao J, Ying Y, Liu N, Pan Y, Zhang D. Mitochondria: An Emerging Unavoidable Link in the Pathogenesis of Periodontitis Caused by Porphyromonas gingivalis. Int J Mol Sci 2024; 25:737. [PMID: 38255811 PMCID: PMC10815845 DOI: 10.3390/ijms25020737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/02/2024] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Porphyromonas gingivalis (P. gingivalis) is a key pathogen of periodontitis. Increasing evidence shows that P. gingivalis signals to mitochondria in periodontal cells, including gingival epithelial cells, gingival fibroblast cells, immune cells, etc. Mitochondrial dysfunction affects the cellular state and participates in periodontal inflammatory response through the aberrant release of mitochondrial contents. In the current review, it was summarized that P. gingivalis induced mitochondrial dysfunction by altering the mitochondrial metabolic state, unbalancing mitochondrial quality control, prompting mitochondrial reactive oxygen species (ROS) production, and regulating mitochondria-mediated apoptosis. This review outlines the impacts of P. gingivalis and its virulence factors on the mitochondrial function of periodontal cells and their role in periodontitis.
Collapse
Affiliation(s)
- Shiyin Luo
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang 110002, China; (S.L.); (T.X.); (Q.Z.); (A.J.); (J.Z.); (Y.Y.); (N.L.)
| | - Tong Xu
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang 110002, China; (S.L.); (T.X.); (Q.Z.); (A.J.); (J.Z.); (Y.Y.); (N.L.)
| | - Qifan Zheng
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang 110002, China; (S.L.); (T.X.); (Q.Z.); (A.J.); (J.Z.); (Y.Y.); (N.L.)
| | - Aijia Jiang
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang 110002, China; (S.L.); (T.X.); (Q.Z.); (A.J.); (J.Z.); (Y.Y.); (N.L.)
| | - Jiahui Zhao
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang 110002, China; (S.L.); (T.X.); (Q.Z.); (A.J.); (J.Z.); (Y.Y.); (N.L.)
| | - Yue Ying
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang 110002, China; (S.L.); (T.X.); (Q.Z.); (A.J.); (J.Z.); (Y.Y.); (N.L.)
| | - Nan Liu
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang 110002, China; (S.L.); (T.X.); (Q.Z.); (A.J.); (J.Z.); (Y.Y.); (N.L.)
| | - Yaping Pan
- Department of Periodontics and Oral Biology, School of Stomatology, China Medical University, Shenyang 110002, China;
| | - Dongmei Zhang
- Department of Periodontics and Oral Biology, School of Stomatology, China Medical University, Shenyang 110002, China;
| |
Collapse
|
6
|
Blancas-Luciano BE, Zamora-Chimal J, da Silva-de Rosenzweig PG, Ramos-Mares M, Fernández-Presas AM. Macrophages immunomodulation induced by Porphyromonas gingivalis and oral antimicrobial peptides. Odontology 2023; 111:778-792. [PMID: 36897441 PMCID: PMC10492884 DOI: 10.1007/s10266-023-00798-w] [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: 08/25/2022] [Accepted: 02/21/2023] [Indexed: 03/11/2023]
Abstract
Porphyromonas gingivalis is a keystone pathogen associated with periodontitis development, a chronic inflammatory pathology characterized by the destruction of the supporting teeth structure. Macrophages are recruited cells in the inflammatory infiltrate from patients with periodontitis. They are activated by the P. gingivalis virulence factors arsenal, promoting an inflammatory microenvironment characterized by cytokine production (TNF-α, IL-1β, IL-6), prostaglandins, and metalloproteinases (MMPs) that foster the tissular destruction characteristic of periodontitis. Furthermore, P. gingivalis suppresses the generation of nitric oxide, a potent antimicrobial molecule, through its degradation, and incorporating its byproducts as a source of energy. Oral antimicrobial peptides can contribute to controlling the disease due to their antimicrobial and immunoregulatory activity, which allows them to maintain homeostasis in the oral cavity. This study aimed to analyze the immunopathological role of macrophages activated by P. gingivalis in periodontitis and suggested using antimicrobial peptides as therapeutic agents to treat the disease.
Collapse
Affiliation(s)
- Blanca Esther Blancas-Luciano
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Col. Universidad Nacional Autónoma de México, Av. Universidad 3000, CP 04510, Mexico City, Mexico
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Ciudad Universitaria, Edificio D, 1° Piso, Mexico City, Mexico
| | - Jaime Zamora-Chimal
- Unidad de Investigación en Medicina Experimental, Universidad Nacional Autónoma de México, Hospital General de México, Dr. Balmis, 148 Col. Doctores, Del. Cuauhtémoc, C.P. 06726, Mexico City, Mexico
| | - Pablo Gomes da Silva-de Rosenzweig
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, Huixquilucan, State of Mexico, Mexico
| | - Mariana Ramos-Mares
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, Huixquilucan, State of Mexico, Mexico
| | - Ana María Fernández-Presas
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Col. Universidad Nacional Autónoma de México, Av. Universidad 3000, CP 04510, Mexico City, Mexico.
| |
Collapse
|
7
|
Böcher S, Meyer HL, Dafni E, Conrads G. Prevalence and Phylogenetic Analysis of Lipoprotein-Gene ragB-1 of Porphyromonas gingivalis-A Pilot Study. Antibiotics (Basel) 2023; 12:1458. [PMID: 37760754 PMCID: PMC10525598 DOI: 10.3390/antibiotics12091458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/15/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Porphyromonas gingivalis (P.g.) is a key pathogen involved in periodontal diseases. The aim of this study was to investigate the prevalence and phylogenetic origin of the lipoprotein-gene ragB in its most virulent variant, ragB-1 (co-transcribed with ragA-1 as locus rag-1), in different P.g. strains collected worldwide. A total of 138 P.g. strains were analyzed for the presence of ragB-1 by pooled analysis and subsequently individual PCRs. Sequencing a core fragment of ragB-1 of the individual strains made it possible to carry out a phylogenetic classification using sequence alignment. In total, 22 of the 138 P.g. strains tested positive for ragB-1, corresponding to a prevalence of 16%. The fragment investigated was highly conserved, with variations in the base sequence detected in only three strains (OMI 1072, OMI 1081, and OMI 1074). In two strains, namely OMI 1072 (original name: I-433) and OMI 1081 (original name: I-372), which originate from monkeys, two amino-acid alterations were apparent. Since ragB-1 has also been found in animal strains, it may be concluded that rag-1 was transferred from animals to humans and that this originally virulent variant was weakened by mutations over time so that new, less virulent, adapted commensal versions of rag (rag-2, -3, and -4), with P.g. as the host, evolved.
Collapse
Affiliation(s)
- Sarah Böcher
- Department of Operative Dentistry, Periodontology and Preventive Dentistry, Rheinisch-Westfälische Technische Hochschule (RWTH) University Hospital, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Hendrik L. Meyer
- Division of Oral Microbiology and Immunology, Department of Operative Dentistry, Periodontology and Preventive Dentistry, Rheinisch-Westfälische Technische Hochschule (RWTH) University Hospital, Pauwelsstrasse 30, 52074 Aachen, Germany (G.C.)
| | - Evdokia Dafni
- Division of Oral Microbiology and Immunology, Department of Operative Dentistry, Periodontology and Preventive Dentistry, Rheinisch-Westfälische Technische Hochschule (RWTH) University Hospital, Pauwelsstrasse 30, 52074 Aachen, Germany (G.C.)
| | - Georg Conrads
- Division of Oral Microbiology and Immunology, Department of Operative Dentistry, Periodontology and Preventive Dentistry, Rheinisch-Westfälische Technische Hochschule (RWTH) University Hospital, Pauwelsstrasse 30, 52074 Aachen, Germany (G.C.)
| |
Collapse
|
8
|
Boutrin MC, Mishra A, Wang C, Dou Y, Fletcher HM. The involvement of CdhR in Porphyromonas gingivalis during nitric oxide stress. Mol Oral Microbiol 2023; 38:289-308. [PMID: 37134265 PMCID: PMC11018363 DOI: 10.1111/omi.12414] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 05/05/2023]
Abstract
Porphyromonas gingivalis, the causative agent of adult periodontitis, must gain resistance to frequent oxidative and nitric oxide (NO) stress attacks from immune cells in the periodontal pocket to survive. Previously, we found that, in the wild-type and under NO stress, the expression of PG1237 (CdhR), the gene encoding for a putative LuxR transcriptional regulator previously called community development and hemin regulator (CdhR), was upregulated 7.7-fold, and its adjacent gene PG1236 11.9-fold. Isogenic mutants P. gingivalis FLL457 (ΔCdhR::ermF), FLL458 (ΔPG1236::ermF), and FLL459 (ΔPG1236-CdhR::ermF) were made by allelic exchange mutagenesis to determine the involvement of these genes in P. gingivalis W83 NO stress resistance. The mutants were black pigmented and β hemolytic and their gingipain activities varied with strains. FLL457 and FLL459 mutants were more sensitive to NO compared to the wild type, and complementation restored NO sensitivity to that of the wild type. DNA microarray analysis of FLL457 showed that approximately 2% of the genes were upregulated and over 1% of the genes downregulated under NO stress conditions compared to the wild type. Transcriptome analysis of FLL458 and FLL459 under NO stress showed differences in their modulation patterns. Some similarities were also noticed between all mutants. The PG1236-CdhR gene cluster revealed increased expression under NO stress and may be part of the same transcriptional unit. Recombinant CdhR showed binding activity to the predicted promoter regions of PG1459 and PG0495. Taken together, the data indicate that CdhR may play a role in NO stress resistance and be involved in a regulatory network in P. gingivalis.
Collapse
Affiliation(s)
- Marie-Claire Boutrin
- Department of Biological Sciences, School of Arts and Sciences, Oakwood University, Huntsville, Alabama, USA
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Arunima Mishra
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Charles Wang
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Yuetan Dou
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Hansel M. Fletcher
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California, USA
| |
Collapse
|
9
|
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
|
10
|
Poormoradi B, Dehghani S, Zare H, Alikhani MY, Haghgoo JM, Farmany A. Enhanced antibacterial activity of cadmium telluride nanocrystals in combination with 940-nm laser diode on anaerobic bacteria P. gingivalis: an in vitro study. Lasers Med Sci 2023; 38:112. [PMID: 37103664 DOI: 10.1007/s10103-023-03773-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 04/10/2023] [Indexed: 04/28/2023]
Abstract
Periodontal disease is one of the most common chronic diseases in the oral cavity that causes tooth loss. Root scaling and leveling cannot eliminate all periodontal pathogens, and the use of antibacterial agents or lasers can increase the efficiency of mechanical methods. The aim of this study was to evaluate and compare the antibacterial activity of cadmium telluride nanocrystals in combination with 940-nm laser diode. Cadmium telluride nanocrystals were prepared by a green route of synthesis in aqueous medium. The results of this study showed that cadmium telluride nanocrystals significantly inhibit the growth of P. gingivalis. The antibacterial property of this nanocrystal increases with increasing its concentration, laser diode 940-nm irradiation and with increasing the time. It was shown that the antibacterial activity of combination of 940-nm laser diode and cadmium telluride nanocrystals is greater than the effect of either alone and can have a similar effect with its long-term presence of microorganisms. This is very important because it is not possible to use these nanocrystals in the mouth and in the periodontal bag for a long time.
Collapse
Affiliation(s)
- Banafsheh Poormoradi
- Department of Periodontics, School of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Safoora Dehghani
- Department of Periodontics, School of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Mohammad Yousef Alikhani
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Janet Moradi Haghgoo
- Department of Periodontics, School of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Abbas Farmany
- Dental Research Center, School of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran.
- Dental Implant Research Center, School of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
11
|
Yuan L, Wang Y, Zong Y, Dong F, Zhang L, Wang G, Dong H, Wang Y. Response of genes related to iron and porphyrin transport in Porphyromonas gingivalis to blue light. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2023; 241:112670. [PMID: 36841175 DOI: 10.1016/j.jphotobiol.2023.112670] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/22/2023] [Accepted: 02/13/2023] [Indexed: 02/16/2023]
Abstract
BACKGROUND Antimicrobial blue light (aBL) kills a variety of bacteria, including Porphyromonas gingivalis. However, little is known about the transcriptomic response of P. gingivalis to aBL therapy. This study was designed to evaluate the selective cytotoxicity of aBL against P. gingivalis over human cells and to further investigate the genetic response of P. gingivalis to aBL at the transcriptome level. METHODS Colony forming unit (CFU) testing, confocal laser scanning microscopy (CLSM), and scanning electron microscopy (SEM) were used to investigate the antimicrobial effectiveness of blue light against P. gingivalis. The temperatures of the irradiated targets were measured to prevent overheating. Multiple fluorescent probes were used to quantify reactive oxygen species (ROS) generation after blue-light irradiation. RNA sequencing (RNA-seq) was used to investigate the changes in global gene expression. Following the screening of target genes, real-time quantitative polymerase chain reaction (RT-qPCR) was performed to confirm the regulation of gene expression. RESULTS A 405 nm aBL at 100 mW/cm2 significantly killed P. gingivalis within 5 min while sparing human gingival fibroblasts (HGFs). No obvious temperature changes were detected in the irradiated surface under our experimental conditions. RNA-seq showed that the transcription of multiple genes was regulated, and RT-qPCR revealed that the expression levels of the genes RgpA and RgpB, which may promote heme uptake, as well as the genes Ftn and FetB, which are related to iron homeostasis, were significantly upregulated. The expression levels of the FeoB-2 and HmuR genes, which are related to hydroxyl radical scavenging, were significantly downregulated. CONCLUSIONS aBL strengthens the heme uptake and iron export gene pathways while reducing the ROS scavenging pathways in P. gingivalis, thus improving the accumulation of endogenous photosensitizers and enhancing oxidative damage to P. gingivalis.
Collapse
Affiliation(s)
- Lintian Yuan
- Department of General Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, PR China; National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, PR China
| | - Yucheng Wang
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Yanni Zong
- Harvard medical school, Boston, MA02115, USA
| | - Fan Dong
- Center for Digital Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, PR China; National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, PR China
| | - Ludan Zhang
- First Clinical Division, Peking University School and Hospital of Stomatology, Beijing 100081, PR China; National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, PR China
| | - Guiyan Wang
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, PR China; National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, PR China
| | - Huihua Dong
- Center for Digital Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, PR China; National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, PR China
| | - Yuguang Wang
- Center for Digital Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, PR China; National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, PR China.
| |
Collapse
|
12
|
Knowles AA, Campbell SG, Cross NA, Stafford P. Dysregulation of Stress-Induced Translational Control by Porphyromonas gingivalis in Host Cells. Microorganisms 2023; 11:microorganisms11030606. [PMID: 36985180 PMCID: PMC10057856 DOI: 10.3390/microorganisms11030606] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/15/2023] [Accepted: 02/21/2023] [Indexed: 03/04/2023] Open
Abstract
Porphyromonas gingivalis contributes to the chronic oral disease periodontitis, triggering the activation of host inflammatory responses, inducing cellular stresses such as oxidation. During stress, host cells can activate the Integrated Stress Response (ISR), a pathway which determines cellular fate, by either downregulating protein synthesis and initiating a stress–response gene expression program, or by initiating programmed cell death. Recent studies have implicated the ISR within both host antimicrobial defenses and the pathomechanism of certain microbes. In this study, using a combination of immunofluorescence confocal microscopy and immunoblotting, the molecular mechanisms by which P. gingivalis infection alters translation attenuation during oxidative stress-induced activation of the ISR in oral epithelial cells were investigated. P. gingivalis infection alone did not result in ISR activation. In contrast, infection coupled with stress caused differential stress granule formation and composition. Infection heightened stress-induced translational repression independently of core ISR mediators. Heightened translational repression during stress was observed with both P. gingivalis–conditioned media and outer membrane vesicles, implicating a secretory factor in this exacerbated translational repression. The effects of gingipain inhibitors and gingipain-deficient P. gingivalis mutants confirmed these pathogen-specific proteases as the effector of exacerbated translational repression. Gingipains are known to degrade the mammalian target of rapamycin (mTOR) and the findings of this study implicate the gingipain-mTOR axis as the effector of host translational dysregulation during stress.
Collapse
|
13
|
Mangar M, Mishra A, Yang Z, Deivanayagam C, Fletcher HM. Characterization of FA1654: A putative DPS protein in Filifactor alocis. Mol Oral Microbiol 2023; 38:23-33. [PMID: 36412172 PMCID: PMC9905271 DOI: 10.1111/omi.12398] [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: 08/15/2022] [Revised: 10/13/2022] [Accepted: 10/26/2022] [Indexed: 11/24/2022]
Abstract
The survival/adaptation of Filifactor alocis, a fastidious Gram-positive asaccharolytic anaerobe, to the inflammatory environment of the periodontal pocket requires an ability to overcome oxidative stress. Moreover, its pathogenic characteristics are highlighted by its capacity to survive in the oxidative-stress microenvironment of the periodontal pocket and a likely ability to modulate the microbial community dynamics. There is still a significant gap in our understanding of its mechanism of oxidative stress resistance and its impact on the virulence and pathogenicity of the microbial biofilm. Coinfection of epithelial cells with F. alocis and Porphyromonas gingivalis resulted in the upregulation of several genes, including HMPREF0389_01654 (FA1654). Bioinformatics analysis indicates that FA1654 has a "di-iron binding domain" and could function as a DNA starvation and stationary phase protection (DPS) protein. We have further characterized the FA1654 protein to determine its role in oxidative stress resistance in F. alocis. In the presence of hydrogen peroxide-induced oxidative stress, there was an ∼1.3 fold upregulation of the FA1654 gene in F. alocis. Incubation of the purified FA1654 protein with DNA in the presence of hydrogen peroxide and iron resulted in the protection of the DNA from Fenton-mediated degradation. Circular dichroism and differential scanning fluorimetry studies have documented the intrinsic ability of rFA1654 protein to bind iron; however, the rFA1654 protein is missing the intrinsic ability to reduce hydrogen peroxide. Collectively, the data may suggest that FA1654 in F. alocis is involved in oxidative stress resistance via an ability to protect against Fenton-mediated oxidative stress-induced damage.
Collapse
Affiliation(s)
- Malissa Mangar
- Division of Microbiology & Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Arunima Mishra
- Division of Microbiology & Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Zhengrong Yang
- Department of Biochemistry and Molecular Genetics, University of Alabama, Birmingham, AL USA
| | - Champion Deivanayagam
- Department of Biochemistry and Molecular Genetics, University of Alabama, Birmingham, AL USA
| | - Hansel M. Fletcher
- Division of Microbiology & Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California, USA,Corresponding author: Phone: (909) 558-8497, FAX: (909) 558-4035,
| |
Collapse
|
14
|
Ximinies AD, Dou Y, Mishra A, Zhang K, Deivanayagam C, Wang C, Fletcher HM. The Oxidative Stress-Induced Hypothetical Protein PG_0686 in Porphyromonas gingivalis W83 Is a Novel Diguanylate Cyclase. Microbiol Spectr 2023; 11:e0441122. [PMID: 36719196 PMCID: PMC10101095 DOI: 10.1128/spectrum.04411-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/06/2023] [Indexed: 02/01/2023] Open
Abstract
The survival/adaptation of Porphyromonas gingivalis to the inflammatory environment of the periodontal pocket requires an ability to overcome oxidative stress. Several functional classes of genes, depending on the severity and duration of the exposure, were induced in P. gingivalis under H2O2-induced oxidative stress. The PG_0686 gene was highly upregulated under prolonged oxidative stress. PG_0686, annotated as a hypothetical protein of unknown function, is a 60 kDa protein that carries several domains including hemerythrin, PAS10, and domain of unknown function (DUF)-1858. Although PG_0686 showed some relatedness to several diguanylate cyclases (DGCs), it is missing the classical conserved, active site sequence motif (GGD[/E]EF), commonly observed in other bacteria. PG_0686-related proteins are observed in other anaerobic bacterial species. The isogenic mutant P. gingivalis FLL361 (ΔPG_0686::ermF) showed increased sensitivity to H2O2, and decreased gingipain activity compared to the parental strain. Transcriptome analysis of P. gingivalis FLL361 showed the dysregulation of several gene clusters/operons, known oxidative stress resistance genes, and transcriptional regulators, including PG_2212, CdhR and PG_1181 that were upregulated under normal anaerobic conditions. The intracellular level of c-di-GMP in P. gingivalis FLL361 was significantly decreased compared to the parental strain. The purified recombinant PG_0686 (rPG_0686) protein catalyzed the formation of c-di-GMP from GTP. Collectively, our data suggest a global regulatory property for PG_0686 that may be part of an unconventional second messenger signaling system in P. gingivalis. Moreover, it may coordinately regulate a pathway(s) vital for protection against environmental stress, and is significant in the pathogenicity of P. gingivalis and other anaerobes. IMPORTANCE Porphyromonas gingivalis is an important etiological agent in periodontitis and other systemic diseases. There is still a gap in our understanding of the mechanisms that P. gingivalis uses to survive the inflammatory microenvironment of the periodontal pocket. The hypothetical PG_0686 gene was highly upregulated under prolonged oxidative stress. Although the tertiary structure of PG_0686 showed little relatedness to previously characterized diguanylate cyclases (DGCs), and does not contain the conserved GGD(/E)EF catalytic domain motif sequence, an ability to catalyze the formation of c-di-GMP from GTP is demonstrated. The second messenger pathway for c-di-GMP was previously predicted to be absent in P. gingivalis. PG_0686 paralogs are identified in other anaerobic bacteria. Thus, PG_0686 may represent a novel class of DGCs, which is yet to be characterized. In conclusion, we have shown, for the first time, evidence for the presence of c-di-GMP signaling with environmental stress protective function in P. gingivalis.
Collapse
Affiliation(s)
- Alexia D. Ximinies
- Division of Microbiology & Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Yuetan Dou
- Division of Microbiology & Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Arunima Mishra
- Division of Microbiology & Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Kangling Zhang
- Department of Pharmacology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Champion Deivanayagam
- Department of Biochemistry and Molecular Genetics, University of Alabama, Birmingham, Alabama, USA
| | - Charles Wang
- Division of Microbiology & Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Hansel M. Fletcher
- Division of Microbiology & Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California, USA
| |
Collapse
|
15
|
Wu Y, Guo S, Chen F, Li Y, Huang Y, Liu W, Zhang G. Fn-Dps, a novel virulence factor of Fusobacterium nucleatum, disrupts erythrocytes and promotes metastasis in colorectal cancer. PLoS Pathog 2023; 19:e1011096. [PMID: 36693067 PMCID: PMC9873182 DOI: 10.1371/journal.ppat.1011096] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 01/02/2023] [Indexed: 01/25/2023] Open
Abstract
Fusobacterium nucleatum (Fn) is a critical colorectal cancer (CRC)-associated bacterium. DNA hunger/stationary phase protective proteins (Dps) are bacterial ferritins that protect DNA from oxidative stress. However, little is known about the regulatory roles of Fn-Dps towards host cellular functions. Here, we identified Fn-Dps from the culture supernatant of Fn by mass spectrometry, and prepared the recombinant of Fn-Dps protein. We show a novel virulence protein of Fn, Fn-Dps, which lyses and disrupts erythrocytes by the competition for iron acquisition. Also, Fn-Dps facilitates intracellular survival of Fn in macrophages by upregulating the expression of the chemokine CCL2/CCL7. In addition, Fn-Dps can elicit a strong humoral immune response, and mucosal immunization with Fn-Dps conferred protection against Fn in the intestinal tract. Moreover, a high level of anti-Fn-Dps antibody was prevalent in populations, and elevated anti-Fn-Dps antibody levels were observed in CRC patients. Furthermore, Fn-Dps promotes the migration of CRC cells via the CCL2/CCL7-induced epithelial-mesenchymal transition (EMT) and promotes CRC metastasis in vivo.
Collapse
Affiliation(s)
- Yixian Wu
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Songhe Guo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Fangfang Chen
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yiqiu Li
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yuying Huang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Wanli Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- * E-mail: (WL); (GZ)
| | - Ge Zhang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
- * E-mail: (WL); (GZ)
| |
Collapse
|
16
|
Blancas-Luciano BE, Becker-Fauser I, Zamora-Chimal J, Delgado-Domínguez J, Ruíz-Remigio A, Leyva-Huerta ER, Portilla-Robertson J, Fernández-Presas AM. Antimicrobial and anti-inflammatory activity of Cystatin C on human gingival fibroblast incubated with Porphyromonas gingivalis. PeerJ 2022; 10:e14232. [PMID: 36312752 PMCID: PMC9615962 DOI: 10.7717/peerj.14232] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/22/2022] [Indexed: 01/24/2023] Open
Abstract
Background Periodontal disease is considered one of the most prevalent chronic infectious diseases, often leading to the disruption of tooth-supporting tissues, including alveolar bone, causing tooth mobility and loss. Porphyromonas gingivalis is considered the major etiological agent of this disease, having a plethora of virulence factors, including, lipopolysaccharides (LPS), hemolysins, and proteinases. Antimicrobial peptides are one of the main components of the innate immune response that inhibit the growth of P. gingivalis. The aim of this study was to analyze the antimicrobial activity of cystatin C and to assess the effect on the inflammatory and anti-inflammatory cytokines, the production of reactive oxygen species, and in the release of nitric oxide by human gingival fibroblasts incubated with P. gingivalis in the presence and absence of cystatin C. Methods P. gingivalis ATCC 33277 was exposed to cystatin C for 24h and co-cultured with human gingival fibroblasts (HGFs) ATCC CRL-2014. The effect of cystatin on growth of P. gingivalis and HGFs was evaluated. Pro-inflammatory (TNFα, IL-1β) and anti-inflammatory (IL-10) cytokines were determined by ELISA in the supernatants of HGFs incubated with P. gingivalis exposed to cystatin C. Additionally, nitrites and reactive oxygen species (ROS) production were evaluated. Results Cystatin Cinhibited the growth of P. gingivalis without affecting HGFs. Incubation of HGFs with P. gingivalis led to a significant increase of TNF-α and IL-1β. In contrast, HGFs incubated with P. gingivalis exposed to cystatin C showed a decreased production of both cytokines, whereas IL-10 was enhanced. Incubation of HGFs with P. gingivalis led to an increase of nitric oxide (NO) and ROS production, which was reduced in the presence of the peptide. Conclusions Cystatin C inhibits the growth of P. gingivalis and decreases the inflammatory cytokines, ROS, and NO production during infection of HGFs with P. gingivalis. Knowledge on the antimicrobial and immunomodulatory properties of cystatin C could aid in the design of new therapeutic approaches to facilitate the elimination of this bacterium to improve the treatment of periodontal disease.
Collapse
Affiliation(s)
| | - Ingeborg Becker-Fauser
- Unidad de Investigación en Medicina Experimental, Universidad Nacional Autónoma de México, Mexico City, México
| | - Jaime Zamora-Chimal
- Unidad de Investigación en Medicina Experimental, Universidad Nacional Autónoma de México, Mexico City, México
| | - José Delgado-Domínguez
- Unidad de Investigación en Medicina Experimental, Universidad Nacional Autónoma de México, Mexico City, México
| | - Adriana Ruíz-Remigio
- Unidad de Investigación en Medicina Experimental, Universidad Nacional Autónoma de México, Mexico City, México
| | - Elba Rosa Leyva-Huerta
- Departmento de Medicina Oral y Patología, División de Posgrado, Facultad de Odontología, Universidad Nacional Autónoma de México, Mexico City, México
| | - Javier Portilla-Robertson
- Departmento de Medicina Oral y Patología, División de Posgrado, Facultad de Odontología, Universidad Nacional Autónoma de México, Mexico City, México
| | - Ana María Fernández-Presas
- Departamento de Microbiología y Parasitología, Universidad Nacional Autónoma de México, Mexico City, México,Centro de investigación en Ciencias de la Salud (CICSA), Universidad Anáhuac México Campus Norte, Mexico City, México
| |
Collapse
|
17
|
Aja E, Mishra A, Dou Y, Fletcher HM. Role of the Filifactor alocis Hypothetical Protein FA519 in Oxidative Stress Resistance. Microbiol Spectr 2021; 9:e0121221. [PMID: 34756068 PMCID: PMC8579941 DOI: 10.1128/spectrum.01212-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/14/2021] [Indexed: 11/30/2022] Open
Abstract
In the periodontal pocket, there is a direct correlation between environmental conditions, the dynamic oral microbial flora, and disease. The relative abundance of several newly recognized microbial species in the oral microenvironment has raised questions on their impact on disease development. One such organism, Filifactor alocis, is significant to the pathogenic biofilm structure. Moreover, its pathogenic characteristics are highlighted by its ability to survive in the oxidative-stress microenvironment of the periodontal pocket and alter the microbial community dynamics. There is a gap in our understanding of its mechanism(s) of oxidative stress resistance and impact on pathogenicity. Several proteins, including HMPRFF0389-00519 (FA519), were observed in high abundance in F. alocis during coinfection of epithelial cells with Porphyromonas gingivalis W83. Bioinformatics analysis shows that FA519 contains a "Cys-X-X-Cys zinc ribbon domain" which could be involved in DNA binding and oxidative stress resistance. We have characterized FA519 to elucidate its roles in the oxidative stress resistance and virulence of F. alocis. Compared to the wild-type strain, the F. alocis isogenic gene deletion mutant, FLL1013 (ΔFA519::ermF), showed significantly reduced sensitivity to hydrogen peroxide and nitric oxide-induced stress. The ability to form biofilm and adhere to and invade gingival epithelial cells was also reduced in the isogenic mutant. The recombinant FA519 protein was shown to protect DNA from Fenton-mediated damage with an intrinsic ability to reduce hydrogen peroxide and disulfide bonds. Collectively, these results suggest that FA519 is involved in oxidative stress resistance and can modulate important virulence attributes in F. alocis. IMPORTANCE Filifactor alocis is an emerging member of the periodontal community and is now proposed to be a diagnostic indicator of periodontal disease. However, due to the lack of genetic tools available to study this organism, not much is known about its virulence attributes. The mechanism(s) of oxidative stress resistance in F. alocis is unknown. Therefore, identifying the adaptive mechanisms utilized by F. alocis to survive in the oxidative stress environment of the periodontal pocket would lead to understanding its virulence regulation, which could help develop novel therapeutic treatments to combat the effects of periodontal disease. This study is focused on the characterization of FA519, a hypothetical protein in F. alocis, as a multifunctional protein that plays an important role in the reactive oxygen species-detoxification pathway. Collectively, our results suggest that FA519 is involved in oxidative stress resistance and can modulate important virulence attributes in F. alocis.
Collapse
Affiliation(s)
- Ezinne Aja
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Arunima Mishra
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Yuetan Dou
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Hansel M. Fletcher
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California, USA
| |
Collapse
|
18
|
Botelho Dinis M, Agnello M, He X, Shi W, Chaichanasakul Tran N. Pilot study on selective antimicrobial effect of a halitosis mouthrinse: monospecies and saliva-derived microbiome in an in vitro model system. J Oral Microbiol 2021; 13:1996755. [PMID: 34745444 PMCID: PMC8567964 DOI: 10.1080/20002297.2021.1996755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background Halitosis refers to malodor emanating from the oral cavity. Several mouthrinses with halitosis-reduction exist on the market, but their effect on the oral microbiome is largely unknown. In this study, we used an efficient in vitro model system to investigate a test mouthrinse's impact on the oral microbiome. Methods Single halitosis-associated species and other common oral microorganism cultures were exposed to the test mouthrinse over time, and their viability was determined by culture-based selective plating. Next, the saliva-derived microbiome from healthy and halitosis-associated individuals was cultured in the presence of the test mouthrinse over time using the previously developed in vitro model system. The microbiome composition was assessed with 16S rRNA gene sequencing and downstream bioinformatics analyses. Results The test mouthrinse displayed antimicrobial activity against known anaerobic bacterial species producing halitosis-related compounds such as Fusobacterium nucleatum, F. periodonticum, and Prevotella intermedia but not against other common oral microorganisms. In the multispecies, saliva-derived cultures, mouthrinse exposure decreased the relative abundance of the Fusobacterium and Prevotella genera while not affecting overall diversity. Conclusions The test mouthrinse had promising anti-halitosis characteristics at the microbiome level, as demonstrated by the reduction in the relative abundance of halitosis-associated taxa while maintaining microbial diversity.
Collapse
Affiliation(s)
| | | | - Xuesong He
- The Forsyth Institute, Cambridge, MA, USA
| | | | | |
Collapse
|
19
|
Masamba P, Kappo AP. Parasite Survival and Disease Persistence in Cystic Fibrosis, Schistosomiasis and Pathogenic Bacterial Diseases: A Role for Universal Stress Proteins? Int J Mol Sci 2021; 22:10878. [PMID: 34639223 PMCID: PMC8509486 DOI: 10.3390/ijms221910878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/15/2021] [Accepted: 09/28/2021] [Indexed: 12/16/2022] Open
Abstract
Universal stress proteins (USPs) were originally discovered in Escherichia coli over two decades ago and since then their presence has been detected in various organisms that include plants, archaea, metazoans, and bacteria. As their name suggests, they function in a series of various cellular responses in both abiotic and biotic stressful conditions such as oxidative stress, exposure to DNA damaging agents, nutrient starvation, high temperature and acidic stress, among others. Although a highly conserved group of proteins, the molecular and biochemical aspects of their functions are largely evasive. This is concerning, as it was observed that USPs act as essential contributors to the survival/persistence of various infectious pathogens. Their ubiquitous nature in various organisms, as well as their augmentation during conditions of stress, is a clear indication of their direct or indirect importance in providing resilience against such conditions. This paper seeks to clarify what has already been reported in the literature on the proposed mechanism of action of USPs in pathogenic organisms.
Collapse
Affiliation(s)
- Priscilla Masamba
- Molecular Biophysics and Structural Biology (MBSB) Group, Department of Biochemistry, Kingsway Campus, University of Johannesburg, Auckland Park 2006, South Africa;
| | | |
Collapse
|
20
|
Jungbauer G, Moser D, Müller S, Pfister W, Sculean A, Eick S. The Antimicrobial Effect of Cold Atmospheric Plasma against Dental Pathogens-A Systematic Review of In-Vitro Studies. Antibiotics (Basel) 2021; 10:211. [PMID: 33672690 PMCID: PMC7924351 DOI: 10.3390/antibiotics10020211] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/16/2021] [Accepted: 02/18/2021] [Indexed: 12/14/2022] Open
Abstract
Interest in the application of cold atmospheric plasma (CAP) in the medical field has been increasing. Indications in dentistry are surface modifications and antimicrobial interventions. The antimicrobial effect of CAP is mainly attributed to the generation of reactive oxygen and reactive nitrogen species. The aim of this article is to systematically review the available evidence from in-vitro studies on the antimicrobial effect of CAP on dental pathogens. A database search was performed (PubMed, Embase, Scopus). Data concerning the device parameters, experimental set-ups and microbial cultivation were extracted. The quality of the studies was evaluated using a newly designed assessment tool. 55 studies were included (quality score 31-92%). The reduction factors varied strongly among the publications although clusters could be identified between groups of set pathogen, working gases, and treatment time intervals. A time-dependent increase of the antimicrobial effect was observed throughout the studies. CAP may be a promising alternative for antimicrobial treatment in a clinically feasible application time. The introduced standardized protocol is able to compare the outcome and quality of in-vitro studies. Further studies, including multi-species biofilm models, are needed to specify the application parameters of CAP before CAP should be tested in randomized clinical trials.
Collapse
Affiliation(s)
- Gert Jungbauer
- Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland; (D.M.); (A.S.); (S.E.)
| | - Dominick Moser
- Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland; (D.M.); (A.S.); (S.E.)
| | - Steffen Müller
- Department of Cranio-Maxillofacial Surgery, Hospital of the University of Regensburg, 93053 Regensburg, Germany;
| | - Wolfgang Pfister
- Department of Hospital Hygiene, Sophien- und Hufeland-Klinikum Weimar, 99425 Weimar, Germany;
| | - Anton Sculean
- Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland; (D.M.); (A.S.); (S.E.)
| | - Sigrun Eick
- Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland; (D.M.); (A.S.); (S.E.)
| |
Collapse
|
21
|
Exacerbation of AMD Phenotype in Lasered CNV Murine Model by Dysbiotic Oral Pathogens. Antioxidants (Basel) 2021; 10:antiox10020309. [PMID: 33670526 PMCID: PMC7922506 DOI: 10.3390/antiox10020309] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 02/08/2021] [Accepted: 02/12/2021] [Indexed: 12/13/2022] Open
Abstract
Emerging evidence underscores an association between age-related macular degeneration (AMD) and periodontal disease (PD), yet the biological basis of this linkage and the specific role of oral dysbiosis caused by PD in AMD pathophysiology remains unclear. Furthermore, a simple reproducible model that emulates characteristics of both AMD and PD has been lacking. Hence, we established a novel AMD+PD murine model to decipher the potential role of oral infection (ligature-enhanced) with the keystone periodontal pathogen Porphyromonas gingivalis, in the progression of neovasculogenesis in a laser-induced choroidal-neovascularization (Li-CNV) mouse retina. By a combination of fundus photography, optical coherence tomography, and fluorescein angiography, we documented inflammatory drusen-like lesions, reduced retinal thickness, and increased vascular leakage in AMD+PD mice retinae. H&E further confirmed a significant reduction of retinal thickness and subretinal drusen-like deposits. Immunofluorescence microscopy revealed significant induction of choroidal/retinal vasculogenesis in AMD+PD mice. qPCR identified increased expression of oxidative-stress, angiogenesis, pro-inflammatory mediators, whereas antioxidants and anti-inflammatory genes in AMD+PD mice retinae were notably decreased. Through qPCR, we detected Pg and its fimbrial 16s-RrNA gene expression in the AMD+PD mice retinae. To sum-up, this is the first in vivo study signifying a role of periodontal infection in augmentation of AMD phenotype, with the aid of a pioneering AMD+PD murine model established in our laboratory.
Collapse
|
22
|
Dou Y, Rutanhira H, Schormann N, Deivanayagam C, Fletcher HM. PG1659 functions as anti-sigma factor to extracytoplasmic function sigma factor RpoE in Porphyromonas gingivalis W83. Mol Oral Microbiol 2021; 36:80-91. [PMID: 33377315 DOI: 10.1111/omi.12329] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 12/19/2022]
Abstract
Anti-sigma factors play a critical role in regulating the expression of sigma factors in response to environmental stress signals. PG1659 is cotranscribed with an upstream gene PG1660 (rpoE), which encodes for a sigma factor that plays an important role in oxidative stress resistance and the virulence regulatory network of P. gingivalis. PG1659, which is annotated as a hypothetical gene, is evaluated in this study. PG1659, composed of 130 amino acids, is predicted to be transmembrane protein with a single calcium (Ca2+ ) binding site. In P. gingivalis FLL358 (ΔPG1659::ermF), the rpoE gene was highly upregulated compared to the wild-type W83 strain. RpoE-induced genes were also upregulated in the PG1659-defective isogenic mutant. Both protein-protein pull-down and bacterial two-hybrid assays revealed that the PG1659 protein could interact with/bind RpoE. The N-terminal domain of PG1659, representing the cytoplasmic fragment of the protein, is critical for interaction with RpoE. In the presence of PG1659, the initiation of transcription by the RpoE sigma factor was inhibited. Taken together, our data suggest that PG1659 is an anti-sigma factor which plays an important regulatory role in the modulation of the sigma factor RpoE in P. gingivalis.
Collapse
Affiliation(s)
- Yuetan Dou
- Department of Microbiology and Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Hiel Rutanhira
- Department of Microbiology and Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Norbert Schormann
- Department of Biochemistry and Molecular Genetics, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Champion Deivanayagam
- Department of Biochemistry and Molecular Genetics, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hansel M Fletcher
- Department of Microbiology and Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
23
|
Zeng J, Wu L, Chen Q, Wang L, Qiu W, Zheng X, Yin X, Liu J, Ren Y, Li Y. Comprehensive profiling of protein lysine acetylation and its overlap with lysine succinylation in the Porphyromonas gingivalis fimbriated strain ATCC 33277. Mol Oral Microbiol 2020; 35:240-250. [PMID: 32939976 DOI: 10.1111/omi.12312] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 09/02/2020] [Accepted: 09/07/2020] [Indexed: 02/05/2023]
Abstract
Porphyromonas gingivalis is a pathogen closely associated with periodontal and systemic infections. Recently, lysine acetylation (Kac) and lysine succinylation (Ksuc) have been identified in bacterial proteins with diverse biological and pathological functions. The Ksuc of P. gingivalis ATCC 33277 has been characterized in our previous work, and here, we report the systematic analysis of Kac and its crosstalk with Ksuc in this bacterium. A combination of the affinity enrichment by the acetyl-lysine antibody with highly sensitive LC-MS/MS was used to identify the lysine-acetylated proteins and sites in P. gingivalis ATCC 33277. A total of 1,112 lysine-acetylated sites matching 438 proteins were identified. These proteins involved in several cellular processes, especially those proteins related to protein biosynthesis and central metabolism had a high tendency to be lysine acetylated. Moreover, lysine sites flanked by tyrosine, phenylalanine, and histidine in the +1 position, as well as residue lysine in position +4 to +5, were the targets of Kac. Additionally, proteins involved in adhesins, gingipains, black pigmentation, and oxidative stress resistance were identified as substrates of Kac. Collectively, these results suggest Kac may play a critical role in the regulation of physiology and virulence of P. gingivalis. Furthermore, we discovered that, Ksuc and Kac were extensively overlapped in P. gingivalis ATCC 33277, especially in proteins related to ribosomes and metabolism. This study provides a significant beginning for further investigating the role of Kac and Ksuc in the pathogenicity of P. gingivalis.
Collapse
Affiliation(s)
- Jumei Zeng
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Leng Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qiushi Chen
- Clinical Laboratory of BGI Health, BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Lingyun Wang
- Division of Infectious Diseases, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wei Qiu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xin Zheng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoming Yin
- Clinical Laboratory of BGI Health, BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Jie Liu
- Clinical Laboratory of BGI Health, BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Yan Ren
- Clinical Laboratory of BGI Health, BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Yuqing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
24
|
A therapeutic oxygen carrier isolated from Arenicola marina decreased P. gingivalis induced inflammation and tissue destruction. Sci Rep 2020; 10:14745. [PMID: 32901057 PMCID: PMC7479608 DOI: 10.1038/s41598-020-71593-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/17/2020] [Indexed: 12/17/2022] Open
Abstract
The control of inflammation and infection is crucial for periodontal wound healing and regeneration. M101, an oxygen carrier derived from Arenicola marina, was tested for its anti-inflammatory and anti-infectious potential based on its anti-oxidative and tissue oxygenation properties. In vitro, no cytotoxicity was observed in oral epithelial cells (EC) treated with M101. M101 (1 g/L) reduced significantly the gene expression of pro-inflammatory markers such as TNF-α, NF-κΒ and RANKL in P. gingivalis-LPS stimulated and P. gingivalis-infected EC. The proteome array revealed significant down-regulation of pro-inflammatory cytokines (IL-1β and IL-8) and chemokine ligands (RANTES and IP-10), and upregulation of pro-healing mediators (PDGF-BB, TGF-β1, IL-10, IL-2, IL-4, IL-11 and IL-15) and, extracellular and immune modulators (TIMP-2, M-CSF and ICAM-1). M101 significantly increased the gene expression of Resolvin-E1 receptor. Furthermore, M101 treatment reduced P. gingivalis biofilm growth over glass surface, observed with live/dead analysis and by decreased P. gingivalis 16 s rRNA expression (51.7%) (p < 0.05). In mice, M101 reduced the clinical abscess size (50.2%) in P. gingivalis-induced calvarial lesion concomitant with a decreased inflammatory score evaluated through histomorphometric analysis, thus, improving soft tissue and bone healing response. Therefore, M101 may be a novel therapeutic agent that could be beneficial in the management of P. gingivalis associated diseases.
Collapse
|
25
|
Kharaeva ZF, Mustafaev MS, Khazhmetov AV, Gazaev IH, Blieva LZ, Steiner L, Mayer W, De Luca C, Korkina LG. Anti-Bacterial and Anti-Inflammatory Effects of Toothpaste with Swiss Medicinal Herbs towards Patients Suffering from Gingivitis and Initial Stage of Periodontitis: from Clinical Efficacy to Mechanisms. Dent J (Basel) 2020; 8:dj8010010. [PMID: 31952199 PMCID: PMC7148460 DOI: 10.3390/dj8010010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 01/13/2020] [Indexed: 12/16/2022] Open
Abstract
Objective: To distinguish clinical effects and mechanisms of sodium monofluorophosphate plus xylitol and herbal extracts of Swiss medicinal plants (Chamomilla recutita, Arnica montana, Echinacea purpurea, and Salvia officinalis). Materials and Methods: A 2-month-long comparative clinical study of toothpaste containing 1450 ppm sodium monofluorophosphate and xylitol (control, 15 patients) and toothpaste additionally containing extracts of the medicinal herbs (experiment, 35 patients) was performed on patients with gingivitis and the initial stage of periodontitis. Clinical indices of gingivitis/periodontitis were quantified by Loe & Silness’s, CPITN, OHI-S, and PMA indexes. The pro-inflammatory and anti-inflammatory interleukins, nitrites/nitrates, total antioxidant activity, and bacterial pattern characteristic for gingivitis and periodontitis were quantified in the gingival crevicular fluid and plaque. In the in vitro tests, direct anti-bacterial effects, inhibition of catalase induction in Staphylococcus aureus, in response to oxidative burst of phagocytes, and intracellular bacterial killing were determined for the toothpastes, individual plant extracts, and their mixture. Results: Experimental toothpaste was more efficient clinically and in the diminishing of bacterial load specific for gingivitis/periodontitis. Although the control toothpaste exerted a direct moderate anti-bacterial effect, herbal extracts provided anti-inflammatory, anti-oxidant, direct, and indirect anti-bacterial actions through inhibition of bacterial defence against phagocytes. Conclusions: Chemical and plant-derived anti-bacterials to treat gingivitis and periodontitis at the initial stage should be used in combination amid their different mechanisms of action. Plant-derived actives for oral care could substitute toxic chemicals due to multiple modes of positive effects.
Collapse
Affiliation(s)
- Zaira F. Kharaeva
- Department of Microbiology, Virology and Immunology, Kabardino-Balkar Berbekov’s State University, 176 Chernishevskogo St., 360000 Nal’chik, Russia; (Z.F.K.); (L.Z.B.)
| | - Magomet Sh. Mustafaev
- Department of Dentistry & Maxillofacial Surgery, Kabardino-Balkar Berbekov’s State University, 176 Chernishevskogo St., 360000 Nal’chik, Russia; (M.S.M.); (A.V.K.)
| | - Anzor V. Khazhmetov
- Department of Dentistry & Maxillofacial Surgery, Kabardino-Balkar Berbekov’s State University, 176 Chernishevskogo St., 360000 Nal’chik, Russia; (M.S.M.); (A.V.K.)
| | - Ismail H. Gazaev
- Department of Molecular Diagnostics, Russian Federation State Reference Centre for Phyto- and Veterinary Control, 1 Ninth May St., 360000 Nal’chik, Russia;
| | - Larisa Z. Blieva
- Department of Microbiology, Virology and Immunology, Kabardino-Balkar Berbekov’s State University, 176 Chernishevskogo St., 360000 Nal’chik, Russia; (Z.F.K.); (L.Z.B.)
| | - Lukas Steiner
- Marketing Department, TRISA AG, 31 Kantonstrasse, CH-6234 Triengen, Switzerland;
| | - Wolfgang Mayer
- R&D Department, MEDENA AG, 16 Industriestrasse, 8910 Affoltern-am-Albis, Switzerland; (W.M.); (C.D.L.)
| | - Chiara De Luca
- R&D Department, MEDENA AG, 16 Industriestrasse, 8910 Affoltern-am-Albis, Switzerland; (W.M.); (C.D.L.)
| | - Liudmila G. Korkina
- Centre of Innovative Biotechnological Investigations Nanolab (CIBI-NANOLAB), 197 Vernadskiy Pr., 119571 Moscow, Russia
- Correspondence:
| |
Collapse
|
26
|
Śmiga M, Olczak T. PgRsp Is a Novel Redox-Sensing Transcription Regulator Essential for Porphyromonas gingivalis Virulence. Microorganisms 2019; 7:microorganisms7120623. [PMID: 31795139 PMCID: PMC6955866 DOI: 10.3390/microorganisms7120623] [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: 10/29/2019] [Revised: 11/20/2019] [Accepted: 11/25/2019] [Indexed: 12/14/2022] Open
Abstract
Porphyromonas gingivalis is one of the etiological agents of chronic periodontitis. Both heme and oxidative stress impact expression of genes responsible for its survival and virulence. Previously we showed that P. gingivalis ferric uptake regulator homolog affects expression of a gene encoding a putative Crp/Fnr superfamily member, termed P. gingivalis redox-sensing protein (PgRsp). Although PgRsp binds heme and shows the highest similarity to proteins assigned to the CooA family, it could be a member of a novel, separate family of proteins with unknown function. Expression of the pgrsp gene is autoregulated and iron/heme dependent. Genes encoding proteins engaged in the oxidative stress response were upregulated in the pgrsp mutant (TO11) strain compared with the wild-type strain. The TO11 strain showed higher biomass production, biofilm formation, and coaggregation ability with Tannerella forsythia and Prevotella intermedia. We suggest that PgRsp may regulate production of virulence factors, proteases, Hmu heme acquisition system, and FimA protein. Moreover, we observed growth retardation of the TO11 strain under oxidative conditions and decreased survival ability of the mutant cells inside macrophages. We conclude that PgRsp protein may play a role in the oxidative stress response using heme as a ligand for sensing changes in redox status, thus regulating the alternative pathway of the oxidative stress response alongside OxyR.
Collapse
|
27
|
Romero-Lastra P, Sánchez MC, Llama-Palacios A, Figuero E, Herrera D, Sanz M. Gene expression of Porphyromonas gingivalis ATCC 33277 when growing in an in vitro multispecies biofilm. PLoS One 2019; 14:e0221234. [PMID: 31437202 PMCID: PMC6706054 DOI: 10.1371/journal.pone.0221234] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 08/01/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Porphyromonas gingivalis, an oral microorganism residing in the subgingival biofilm, may exert diverse pathogenicity depending on the presence of specific virulence factors, but its gene expression has not been completely established. This investigation aims to compare the transcriptomic profile of this pathogen when growing within an in vitro multispecies biofilm or in a planktonic state. MATERIALS AND METHODS P. gingivalis ATCC 33277 was grown in anaerobiosis within multi-well culture plates at 37°C under two conditions: (a) planktonic samples (no hydroxyapatite discs) or (b) within a multispecies-biofilm containing Streptococcus oralis, Actinomyces naeslundii, Veillonella parvula, Fusobacterium nucleatum and Aggregatibacter actinomycetemcomitans deposited on hydroxyapatite discs. Scanning Electron Microscopy (SEM) and Confocal Laser Scanning Microscopy (CLSM) combined with Fluorescence In Situ Hybridization (FISH) were used to verify the formation of the biofilm and the presence of P. gingivalis. Total RNA was extracted from both the multispecies biofilm and planktonic samples, then purified and, with the use of a microarray, its differential gene expression was analyzed. A linear model was used for determining the differentially expressed genes using a filtering criterion of two-fold change (up or down) and a significance p-value of <0.05. Differential expression was confirmed by Reverse Transcription-quantitative Polymerase Chain Reaction (RT-qPCR). RESULTS SEM verified the development of the multispecies biofilm and FISH confirmed the incorporation of P. gingivalis. The microarray demonstrated that, when growing within the multispecies biofilm, 19.1% of P. gingivalis genes were significantly and differentially expressed (165 genes were up-regulated and 200 down-regulated), compared with planktonic growth. These genes were mainly involved in functions related to the oxidative stress, cell envelope, transposons and metabolism. The results of the microarray were confirmed by RT-qPCR. CONCLUSION Significant transcriptional changes occurred in P. gingivalis when growing in a multispecies biofilm compared to planktonic state.
Collapse
Affiliation(s)
- Patricia Romero-Lastra
- Laboratory of Dental Research, Faculty of Odontology, Complutense University of Madrid, Madrid, Spain
| | - María C. Sánchez
- Laboratory of Dental Research, Faculty of Odontology, Complutense University of Madrid, Madrid, Spain
- ETEP Research Group, Faculty of Odontology, Complutense University of Madrid, Madrid, Spain
| | - Arancha Llama-Palacios
- Laboratory of Dental Research, Faculty of Odontology, Complutense University of Madrid, Madrid, Spain
- ETEP Research Group, Faculty of Odontology, Complutense University of Madrid, Madrid, Spain
| | - Elena Figuero
- ETEP Research Group, Faculty of Odontology, Complutense University of Madrid, Madrid, Spain
- * E-mail:
| | - David Herrera
- ETEP Research Group, Faculty of Odontology, Complutense University of Madrid, Madrid, Spain
| | - Mariano Sanz
- ETEP Research Group, Faculty of Odontology, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
28
|
Are Sphingolipids and Serine Dipeptide Lipids Underestimated Virulence Factors of Porphyromonas gingivalis? Infect Immun 2018; 86:IAI.00035-18. [PMID: 29632248 DOI: 10.1128/iai.00035-18] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The keystone periodontal pathogen Porphyromonas gingivalis produces phosphorylated dihydroceramide lipids (sphingolipids) such as phosphoethanolamine dihydroceramide (PE DHC) and phosphoglycerol dihydroceramide (PG DHC) lipids. Phosphorylated DHCs (PDHCs) from P. gingivalis can affect a number of mammalian cellular functions, such as potentiation of prostaglandin secretion from gingival fibroblasts, promotion of RANKL-induced osteoclastogenesis, promotion of apoptosis, and enhancement of autoimmunity. In P. gingivalis, these lipids affect anchoring of surface polysaccharides, resistance to oxidative stress, and presentation of surface polysaccharides (anionic polysaccharides and K-antigen capsule). In addition to phosphorylated dihydroceramide lipids, serine dipeptide lipids of P. gingivalis are implicated in alveolar bone loss in chronic periodontitis through interference with osteoblast differentiation and function and promotion of osteoclast activity. As a prerequisite for designation as bacterial virulence factors, bacterial sphingolipids and serine dipeptide lipids are recovered in gingival/periodontal tissues, tooth calculus, human blood, vascular tissues, and brain. In addition to P. gingivalis, other bacteria of the genera Bacteroides, Parabacteroides, Porphyromonas, Tannerella, and Prevotella produce sphingolipids and serine dipeptide lipids. The contribution of PDHCs and serine dipeptide lipids to the pathogenesis of periodontal and extraoral diseases may be an underappreciated area in microbe-host interaction and should be more intensively investigated.
Collapse
|
29
|
Pourhajibagher M, Ghorbanzadeh R, Bahador A. Expression patterns of oxyR induced by oxidative stress from Porphyromonas gingivalis in response to photo-activated disinfection. Infect Drug Resist 2018; 11:717-725. [PMID: 29805265 PMCID: PMC5960254 DOI: 10.2147/idr.s152834] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Introduction Porphyromonas gingivalis, an important endodontic pathogen, may be exposed to sublethal doses of photo-activated disinfection (sPAD) during root canal therapy. Such an exposure can affect bacterial survival and virulence features. In this study, we evaluated the effect of sPAD-related oxidative stresses on the expression of oxidative stress response gene (oxyR) in P. gingivalis clinical isolates surviving in vitro photodynamic treatment. Materials and methods To determine the sPAD, 16 clinical P. gingivalis isolates photosensitized with toluidine blue O (TBO), methylene blue (MB), and indocyanine green (ICG) were irradiated with specific wavelength and energy density of diode laser corresponding to the photosensitizers following bacterial viability measurements. The effects of sPAD on the expression ratio of oxyR of 16 clinical P. gingivalis isolates were then assessed using quantitative real-time PCR (qRT-PCR) assay. Results Maximum values of sPAD against P. gingivalis were 6.25, 15.6, and 25 μg/mL at fluencies of 171.87, 15.6, and 93.75 J/cm2, respectively, for TBO-, ICG-, and MB-sPAD (P>0.05). ICG-, MB-, and TBO-sPAD could increase the oxyR gene expression of the clinical P. gingivalis isolates 12.3-, 5.6-, and 8.5-fold, respectively. ICG-sPAD increased the expression of oxyR gene in clinical isolates of P. gingivalis ~1.5- and 2-fold higher than TBO- and MB-sPAD, respectively. Conclusion Our results showed that upregulation of oxyR during sPAD may lead to better survival and increased pathogenicity of P. gingivalis isolates. Therefore, selection of appropriate photo-activated disinfection dosage should be considered for the successful treatment of endodontic infection.
Collapse
Affiliation(s)
- Maryam Pourhajibagher
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Dental Implant Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran.,Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Abbas Bahador
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran.,Laser Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
30
|
Meta-Analysis of the Use of 8-OHdG in Saliva as a Marker of Periodontal Disease. DISEASE MARKERS 2018; 2018:7916578. [PMID: 29854026 PMCID: PMC5954896 DOI: 10.1155/2018/7916578] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 03/29/2018] [Indexed: 01/18/2023]
Abstract
The objective was to collect the available evidence on oxidative stress marker measurements in periodontal patients, focusing specifically on 8-hydroxy-2′-deoxyguanosine (8-OHdG) as a salivary marker of periodontal disease, and to perform meta-analyses to calculate differences in concentration compared to healthy persons. A systematic search in PubMed, Cochrane Library, Embase, and Scopus identified 81 articles. Of these, 38 were duplicates. After reading the abstracts of the remaining 43, 42 were selected for full-text assessment. Finally, 17 articles were included in the qualitative synthesis. Those excluded were of low quality, did not answer the research question, or did not meet the inclusion and exclusion criteria. Of the 17 in the qualitative synthesis, 9 were included in the meta-analysis. The 9 studies in the meta-analysis were combined in a random effects model. Their heterogeneity was high (Q = 3982.02, p < 0.001, I2 = 99.8%). The difference in mean 8-OHdG concentration in saliva between periodontal and healthy subjects was estimated at 2.11 ng/ml (95% CI 1.23–2.98). The different saliva collection methods (stimulated/unstimulated) did not explain the heterogeneity. The 8-OHdG levels in saliva of periodontal patients were almost double to those of healthy patients: 8-OHdG is clearly a powerful periodontal disease marker.
Collapse
|
31
|
Dou Y, Rutanhira H, Chen X, Mishra A, Wang C, Fletcher HM. Role of extracytoplasmic function sigma factor PG1660 (RpoE) in the oxidative stress resistance regulatory network of Porphyromonas gingivalis. Mol Oral Microbiol 2017; 33:89-104. [PMID: 29059500 DOI: 10.1111/omi.12204] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2017] [Indexed: 12/27/2022]
Abstract
In Porphyromonas gingivalis, the protein PG1660, composed of 174 amino acids, is annotated as an extracytoplasmic function (ECF) sigma factor (RpoE homologue-σ24). Because PG1660 can modulate several virulence factors and responds to environmental signals in P. gingivalis, its genetic properties were evaluated. PG1660 is co-transcribed with its downstream gene PG1659, and the transcription start site was identified as adenine residue 54-nucleotides upstream of the ATG translation start codon. In addition to binding its own promoter, using the purified rPG1660 and RNAP core enzyme from Escherichia coli with the PG1660 promoter DNA as template, the function of PG1660 as a sigma factor was demonstrated in an in vitro transcription assay. Transcriptome analyses of a P. gingivalis PG1660-defective isogenic mutant revealed that under oxidative stress conditions 176 genes including genes involved in secondary metabolism were downregulated more than two-fold compared with the parental strain. The rPG1660 protein also showed the ability to bind to the promoters of the highly downregulated genes in the PG1660-deficient mutant. As the ECF sigma factor PG0162 has a 29% identity with PG1660 and can modulate its expression, the cross-talk between their regulatory networks was explored. The expression profile of the PG0162PG1660-deficient mutant (P. gingivalis FLL356) revealed that the type IX secretion system genes and several virulence genes were downregulated under hydrogen peroxide stress conditions. Taken together, we have confirmed that PG1660 can function as a sigma factor, and plays an important regulatory role in the oxidative stress and virulence regulatory network of P. gingivalis.
Collapse
Affiliation(s)
- Y Dou
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - H Rutanhira
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - X Chen
- Department of Basic Sciences, School of Medicine, Center for Genomics, Loma Linda University, Loma Linda, CA, USA
| | - A Mishra
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - C Wang
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA.,Department of Basic Sciences, School of Medicine, Center for Genomics, Loma Linda University, Loma Linda, CA, USA
| | - H M Fletcher
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA.,Institute of Oral Biology, Kyung Hee University, Seoul, Korea
| |
Collapse
|
32
|
Hu YH, Sun L. The global regulatory effect of Edwardsiella tarda Fur on iron acquisition, stress resistance, and host infection: A proteomics-based interpretation. J Proteomics 2016; 140:100-10. [PMID: 27102497 DOI: 10.1016/j.jprot.2016.04.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 03/27/2016] [Accepted: 04/07/2016] [Indexed: 02/01/2023]
Abstract
UNLABELLED Ferric uptake regulator (Fur) is an important transcriptional regulator of Gram-negative bacteria. Edwardsiella tarda is a severe fish bacterial pathogen with a broad host range that includes humans. In this study, we examined the regulatory function of Fur in E. tarda via a proteomic approach. Compared to the wild type TX01, the fur mutant TX01Δfur exhibited (i) retarded growth, (ii) enhanced siderophore production, (iii) increased acid tolerance, which is in contrast to observations in other bacterial species, (iv) decreased survival against oxidative stress and host serum, (v) impaired ability to inhibit host immune response, (vi) attenuated tissue infectivity and overall virulence. The deficiency of TX01Δfur was rescued by introduction of an exogenous fur gene. iTRAQ-based comparative proteomic analysis of TX01Δfur and TX01 identified 89 differentially expressed proteins that cover a wide range of functional categories including those affected by fur mutation. In addition, 16 proteins were identified for the first time to be regulated by Fur in Gram-negative bacteria. These results provide the first protein-based interpretation of the global impact of Fur on the physiology and infectivity of E. tarda. SIGNIFICANCE This study demonstrates that in E. tarda, Fur controls multiple aspects of bacterial life, including growth, metabolism, iron acquisition, stress response, and host infection. In line with these observations, proteomics analysis identified a large amount of proteins affected in expression by Fur, which are involved in bacterial physiology and infectivity. Hence, these results link for the first time the pleiotropic effect of Fur with global protein expression and shed new light on the function and regulatory mechanism of Fur in pathogenic bacteria.
Collapse
Affiliation(s)
- Yong-Hua Hu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Li Sun
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| |
Collapse
|
33
|
Zhang L, Butler CA, Khan HSG, Dashper SG, Seers CA, Veith PD, Zhang JG, Reynolds EC. Characterisation of the Porphyromonas gingivalis Manganese Transport Regulator Orthologue. PLoS One 2016; 11:e0151407. [PMID: 27007570 PMCID: PMC4805248 DOI: 10.1371/journal.pone.0151407] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 02/26/2016] [Indexed: 11/19/2022] Open
Abstract
PgMntR is a predicted member of the DtxR family of transcriptional repressors responsive to manganese in the anaerobic periodontal pathogen Porphyromonas gingivalis. Our bioinformatic analyses predicted that PgMntR had divalent metal binding site(s) with elements of both manganous and ferrous ion specificity and that PgMntR has unusual twin C-terminal FeoA domains. We produced recombinant PgMntR and four variants to probe the specificity of metal binding and its impact on protein structure and DNA binding. PgMntR dimerised in the absence of a divalent transition metal cation. PgMntR bound three Mn(II) per monomer with an overall dissociation constant Kd 2.0 x 10(-11) M at pH 7.5. PgMntR also bound two Fe(II) with distinct binding affinities, Kd1 2.5 x 10(-10) M and Kd2 ≤ 6.0 x 10(-8) M at pH 6.8. Two of the metal binding sites may form a binuclear centre with two bound Mn2+ being bridged by Cys108 but this centre provided only one site for Fe2+. Binding of Fe2+ or Mn2+ did not have a marked effect on the PgMntR secondary structure. Apo-PgMntR had a distinct affinity for the promoter region of the gene encoding the only known P. gingivalis manganese transporter, FB2. Mn2+ increased the DNA binding affinity of PgMntR whilst Fe2+ destabilised the protein-DNA complex in vitro. PgMntR did not bind the promoter DNA of the gene encoding the characterised iron transporter FB1. The C-terminal FeoA domain was shown to be essential for PgMntR structure/function, as its removal caused the introduction of an intramolecular disulfide bond and abolished the binding of Mn2+ and DNA. These data indicate that PgMntR is a novel member of the DtxR family that may function as a transcriptional repressor switch to specifically regulate manganese transport and homeostasis in an iron-dependent manner.
Collapse
Affiliation(s)
- Lianyi Zhang
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Catherine A. Butler
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Hasnah S. G. Khan
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Stuart G. Dashper
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Christine A. Seers
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Paul D. Veith
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Jian-Guo Zhang
- Walter and Eliza Hall Institute of Medical Research and Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Eric C. Reynolds
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Melbourne, Victoria, Australia
- * E-mail:
| |
Collapse
|
34
|
Reduced Glutathione Mediates Resistance to H2S Toxicity in Oral Streptococci. Appl Environ Microbiol 2016; 82:2078-2085. [PMID: 26801579 DOI: 10.1128/aem.03946-15] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 01/19/2016] [Indexed: 12/21/2022] Open
Abstract
Periodontal disease is associated with changes in the composition of the oral microflora, where health-associated oral streptococci decrease while Gram-negative anaerobes predominate in disease. A key feature of periodontal disease-associated anaerobes is their ability to produce hydrogen sulfide (H2S) abundantly as a by-product of anaerobic metabolism. So far, H2S has been reported to be either cytoprotective or cytotoxic by modulating bacterial antioxidant defense systems. Although oral anaerobes produce large amounts of H2S, the potential effects of H2S on oral streptococci are currently unknown. The aim of this study was to determine the effects of H2S on the survival and biofilm formation of oral streptococci. The growth and biofilm formation of Streptococcus mitis and Streptococcus oralis were inhibited by H2S. However, H2S did not significantly affect the growth of Streptococcus gordonii or Streptococcus sanguinis. The differential susceptibility of oral streptococci to H2S was attributed to differences in the intracellular concentrations of reduced glutathione (GSH). In the absence of GSH, H2S elicited its toxicity through an iron-dependent mechanism. Collectively, our results showed that H2S exerts antimicrobial effects on certain oral streptococci, potentially contributing to the decrease in health-associated plaque microflora.
Collapse
|
35
|
Holden MS, Black J, Lewis A, Boutrin MC, Walemba E, Sabir TS, Boskovic DS, Wilson A, Fletcher HM, Perry CC. Antibacterial Activity of Partially Oxidized Ag/Au Nanoparticles against the Oral Pathogen Porphyromonas gingivalis W83. JOURNAL OF NANOMATERIALS 2016; 2016:9605906. [PMID: 30245705 PMCID: PMC6146971 DOI: 10.1155/2016/9605906] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Advances in nanotechnology provide opportunities for the prevention and treatment of periodontal disease. While physicochemical properties of Ag containing nanoparticles (NPs) are known to influence the magnitude of their toxicity, it is thought that nanosilver can be made less toxic to eukaryotes by passivation of the NPs with a benign metal. Moreover, the addition of other noble metals to silver nanoparticles, in the alloy formulation, is known to alter the silver dissolution behavior. Thus, we synthesized glutathione capped Ag/Au alloy bimetallic nanoparticles (NPs) via the galvanic replacement reaction between maltose coated Ag NPs and chloroauric acid (HAuCl4) in 5% aqueous triblock F127 copolymer solution. We then compared the antibacterial activity of the Ag/Au NPs to pure Ag NPs on Porphyromonas gingivalis W83, a key pathogen in the development of periodontal disease. Only partially oxidized glutathione capped Ag and Ag/Au (Au:Ag≈0.2) NPs inhibited the planktonic growth of P. gingivalis W83. This effect was enhanced in the presence of hydrogen peroxide, which simulates the oxidative stress environment in the periodontal pocket during chronic inflammation.
Collapse
Affiliation(s)
- Megan S. Holden
- Division of Biochemistry, Loma Linda University School of
Medicine, Loma Linda, CA 92350, USA
| | - Jason Black
- Northern Caribbean University, Manchester, Jamaica
| | | | - Marie-Claire Boutrin
- Division of Microbiology and Molecular Genetics, Loma Linda
University School of Medicine, Loma Linda, CA 92350, USA
| | - Elvin Walemba
- Department of Earth and Biological Sciences, Loma Linda
University School of Medicine, Loma Linda, CA 92350, USA
| | - Theodore S. Sabir
- College of Arts and Sciences, Faulkner University,
Montgomery, AL 36109, USA
| | - Danilo S. Boskovic
- Division of Biochemistry, Loma Linda University School of
Medicine, Loma Linda, CA 92350, USA
- Department of Earth and Biological Sciences, Loma Linda
University School of Medicine, Loma Linda, CA 92350, USA
| | - Aruni Wilson
- Division of Microbiology and Molecular Genetics, Loma Linda
University School of Medicine, Loma Linda, CA 92350, USA
| | - Hansel M. Fletcher
- Division of Microbiology and Molecular Genetics, Loma Linda
University School of Medicine, Loma Linda, CA 92350, USA
| | - Christopher C. Perry
- Division of Biochemistry, Loma Linda University School of
Medicine, Loma Linda, CA 92350, USA
| |
Collapse
|
36
|
Leclerc J, Rosenfeld E, Trainini M, Martin B, Meuric V, Bonnaure-Mallet M, Baysse C. The Cytochrome bd Oxidase of Porphyromonas gingivalis Contributes to Oxidative Stress Resistance and Dioxygen Tolerance. PLoS One 2015; 10:e0143808. [PMID: 26629705 PMCID: PMC4668044 DOI: 10.1371/journal.pone.0143808] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 11/10/2015] [Indexed: 11/18/2022] Open
Abstract
Porphyromonas gingivalis is an etiologic agent of periodontal disease in humans. The disease is associated with the formation of a mixed oral biofilm which is exposed to oxygen and environmental stress, such as oxidative stress. To investigate possible roles for cytochrome bd oxidase in the growth and persistence of this anaerobic bacterium inside the oral biofilm, mutant strains deficient in cytochrome bd oxidase activity were characterized. This study demonstrated that the cytochrome bd oxidase of Porphyromonas gingivalis, encoded by cydAB, was able to catalyse O2 consumption and was involved in peroxide and superoxide resistance, and dioxygen tolerance.
Collapse
Affiliation(s)
- Julia Leclerc
- EA1254 Microbiologie—Risques Infectieux, University of Rennes1, Rennes, France
| | - Eric Rosenfeld
- UMR CNRS 7266 LIENSs, University of La Rochelle, La Rochelle, France
| | - Mathieu Trainini
- EA1254 Microbiologie—Risques Infectieux, University of Rennes1, Rennes, France
| | - Bénédicte Martin
- EA1254 Microbiologie—Risques Infectieux, University of Rennes1, Rennes, France
| | - Vincent Meuric
- EA1254 Microbiologie—Risques Infectieux, University of Rennes1, Rennes, France
- UMR CNRS 7266 LIENSs, University of La Rochelle, La Rochelle, France
- CHU Rennes, Rennes, France
| | - Martine Bonnaure-Mallet
- EA1254 Microbiologie—Risques Infectieux, University of Rennes1, Rennes, France
- UMR CNRS 7266 LIENSs, University of La Rochelle, La Rochelle, France
- CHU Rennes, Rennes, France
| | - Christine Baysse
- EA1254 Microbiologie—Risques Infectieux, University of Rennes1, Rennes, France
- * E-mail:
| |
Collapse
|
37
|
McKenzie RME, Henry LG, Boutrin MC, Ximinies A, Fletcher HM. Role of the Porphyromonas gingivalis iron-binding protein PG1777 in oxidative stress resistance. MICROBIOLOGY-SGM 2015; 162:256-267. [PMID: 26581883 DOI: 10.1099/mic.0.000213] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Whole genome sequencing of the response of Porphyromonas gingivalis W83 to hydrogen peroxide revealed an upregulation of several uncharacterized, novel genes. Under conditions of prolonged oxidative stress in P. gingivalis, increased expression of a unique transcriptional unit carrying the grpE, dnaJ and three other hypothetical genes (PG1777, PG1778 and PG1779) was observed. The transcriptional start site of this operon appears to be located 91 bp upstream of the translational start, with a potential -10 region at -3 nt and a -35 region at -39 nt. Isogenic P. gingivalis mutants FLL273 (PG1777 : : ermF-ermAM) and FLL293 (PG1779 : : ermF-ermAM) showed increased sensitivity to and decreased survival after treatment with hydrogen peroxide. P. gingivalis FLL273 showed a fivefold increase in the formation of spontaneous mutants when compared with the parent strain after exposure to hydrogen peroxide. The recombinant PG1777 protein displayed iron-binding properties when incubated with FeSO4 and Fe(NH4)2(SO4).6H2O. The rPG1777 protein protected DNA from degradation when exposed to hydrogen peroxide in the presence of iron. Taken together, the data suggest that the grpE-dnaJ-PG1777-PG1778-PG1779 transcriptional unit may play an important role in oxidative stress resistance in P. gingivalis via its ability to protect against DNA damage.
Collapse
Affiliation(s)
- Rachelle M E McKenzie
- Division of Microbiology and Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Leroy G Henry
- Division of Microbiology and Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Marie-Claire Boutrin
- Division of Microbiology and Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Alexia Ximinies
- Division of Microbiology and Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Hansel M Fletcher
- Institute of Oral Biology, Kyung Hee University, Seoul, Republic of Korea.,Division of Microbiology and Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| |
Collapse
|
38
|
Boutrin MC, Yu Y, Wang C, Aruni W, Dou Y, Shi L, Fletcher HM. A putative TetR regulator is involved in nitric oxide stress resistance in Porphyromonas gingivalis. Mol Oral Microbiol 2015; 31:340-53. [PMID: 26332057 DOI: 10.1111/omi.12128] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2015] [Indexed: 02/02/2023]
Abstract
To survive in the periodontal pocket, Porphyromonas gingivalis, the main causative agent of periodontal disease, must overcome oxidative and nitric oxide (NO) stress. Previously, we reported that, in the presence of NO comparable to stress conditions, the transcriptome of P. gingivalis was differentially expressed, and genes belonging to the PG1178-81 cluster were significantly upregulated. To further evaluate their role(s) in NO stress resistance, these genes were inactivated by allelic exchange mutagenesis. Isogenic mutants P. gingivalis FLL460 (ΔPG1181::ermF) and FLL461 (ΔPG1178-81::ermF) were black-pigmented, with gingipain and hemolytic activities comparable to that of the wild-type strain. Whereas the recovery of these isogenic mutants from NO stress was comparable to the wild-type, there was increased sensitivity to hydrogen peroxide-induced stress. RNA-Seq analysis under conditions of NO stress showed that approximately 5 and 8% of the genome was modulated in P. gingivalis FLL460 and FLL461, respectively. The PG1178-81 gene cluster was shown to be part of the same transcriptional unit and is inducible in response to NO stress. In the presence of NO, PG1181, a putative transcriptional regulator, was shown to bind to its own promoter region and that of several other NO responsive genes including PG0214 an extracytoplasmic function σ factor, PG0893 and PG1236. Taken together, the data suggest that PG1181 is a NO responsive transcriptional regulator that may play an important role in the NO stress resistance regulatory network in P. gingivalis.
Collapse
Affiliation(s)
- M-C Boutrin
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Y Yu
- School of Pharmacy, Fudan University, Shanghai, China
| | - C Wang
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - W Aruni
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Y Dou
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - L Shi
- School of Pharmacy, Fudan University, Shanghai, China
| | - H M Fletcher
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA.,Institute of Oral Biology, Kyung Hee University, Seoul, Korea
| |
Collapse
|
39
|
Dou Y, Aruni W, Muthiah A, Roy F, Wang C, Fletcher HM. Studies of the extracytoplasmic function sigma factor PG0162 in Porphyromonas gingivalis. Mol Oral Microbiol 2015. [PMID: 26216199 DOI: 10.1111/omi.12122] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PG0162, annotated as an extracytoplasmic function (ECF) sigma factor in Porphyromonas gingivalis, is composed of 193 amino acids. As previously reported, the PG0162-deficient mutant, P. gingivalis FLL350 showed significant reduction in gingipain activity compared with the parental strain. Because this ECF sigma factor could be involved in the virulence regulation in P. gingivalis, its genetic properties were further characterized. A 5'-RACE analysis showed that the start of transcription of the PG0162 gene occurred from a guanine (G) residue 69 nucleotides upstream of the ATG translation initiation codon. The function of PG0162 as a sigma factor was confirmed in a run-off in vitro transcription assay using the purified rPG0162 and RNAP core enzyme from Escherichia coli with the PG0162 promoter as template. As an appropriate PG0162 inducing environmental signal is unknown, a strain overexpressing the PG0162 gene designated P. gingivalis FLL391 was created. Compared with the wild-type strain, transcriptome analysis of P. gingivalis FLL391 showed that approximately 24% of the genome displayed altered gene expression (260 upregulated genes; 286 downregulated genes). Two other ECF sigma factors (PG0985 and PG1660) were upregulated more than two-fold. The autoregulation of PG0162 was confirmed with the binding of the rPG0162 protein to the PG0162 promoter in electrophoretic mobility shift assay. In addition, the rPG0162 protein also showed the ability to bind to the promoter region of two genes (PG0521 and PG1167) that were most upregulated in P. gingivalis FLL391. Taken together, our data suggest that PG0162 is a sigma factor that may play an important role in the virulence regulatory network in P. gingivalis.
Collapse
Affiliation(s)
- Y Dou
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - W Aruni
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - A Muthiah
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - F Roy
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - C Wang
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - H M Fletcher
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA.,Institute of Oral Biology, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
40
|
Abstract
Oral colonising bacteria are highly adapted to the various environmental niches harboured within the mouth, whether that means while contributing to one of the major oral diseases of caries, pulp infections, or gingival/periodontal disease or as part of a commensal lifestyle. Key to these infections is the ability to adhere to surfaces via a range of specialised adhesins targeted at both salivary and epithelial proteins, their glycans and to form biofilm. They must also resist the various physical stressors they are subjected to, including pH and oxidative stress. Possibly most strikingly, they have developed the ability to harvest both nutrient sources provided by the diet and those derived from the host, such as protein and surface glycans. We have attempted to review recent developments that have revealed much about the molecular mechanisms at work in shaping the physiology of oral bacteria and how we might use this information to design and implement new treatment strategies.
Collapse
|
41
|
McKenzie RME, Aruni W, Johnson NA, Robles A, Dou Y, Henry L, Boskovic DS, Fletcher HM. Metabolome variations in the Porphyromonas gingivalis vimA mutant during hydrogen peroxide-induced oxidative stress. Mol Oral Microbiol 2014; 30:111-27. [PMID: 25055986 DOI: 10.1111/omi.12075] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2014] [Indexed: 01/01/2023]
Abstract
The adaptability and survival of Porphyromonas gingivalis in the oxidative microenvironment of the periodontal pocket are indispensable for survival and virulence, and are modulated by multiple systems. Among the various genes involved in P. gingivalis oxidative stress resistance, vimA gene is a part of the 6.15-kb locus. To elucidate the role of a P. gingivalis vimA-defective mutant in oxidative stress resistance, we used a global approach to assess the transcriptional profile, to study the unique metabolome variations affecting survival and virulence in an environment typical of the periodontal pocket. A multilayered protection strategy against oxidative stress was noted in P. gingivalis FLL92 with upregulation of detoxifying genes. The duration of oxidative stress was shown to differentially modulate transcription with 94 (87%) genes upregulated twofold during 10 min and 55 (83.3%) in 15 min. Most of the upregulated genes (55%), fell in the hypothetical/unknown/unassigned functional class. Metabolome variation showed reduction in fumarate and formaldehyde, hence resorting to alternative energy generation and maintenance of a reduced metabolic state. There was upregulation of transposases, genes encoding for the metal ion binding protein transport and secretion system.
Collapse
Affiliation(s)
- R M E McKenzie
- Division of Microbiology and Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, CA, USA; Center for Dental Research, School of Dentistry, Loma Linda University, Loma Linda, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Involvement of PG2212 zinc finger protein in the regulation of oxidative stress resistance in Porphyromonas gingivalis W83. J Bacteriol 2014; 196:4057-70. [PMID: 25225267 DOI: 10.1128/jb.01907-14] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The adaptation of Porphyromonas gingivalis to H2O2-induced stress while inducible is modulated by an unknown OxyR-independent mechanism. Previously, we reported that the PG_2212 gene was highly upregulated in P. gingivalis under conditions of prolonged oxidative stress. Because this gene may have regulatory properties, its function in response to H2O2 was further characterized. PG2212, annotated as a hypothetical protein of unknown function, is a 10.3-kDa protein with a cysteine 2-histidine 2 (Cys2His2) zinc finger domain. The isogenic mutant P. gingivalis FLL366 (ΔPG_2212) showed increased sensitivity to H2O2 and decreased gingipain activity compared to the parent strain. Transcriptome analysis of P. gingivalis FLL366 revealed that approximately 11% of the genome displayed altered expression (130 downregulated genes and 120 upregulated genes) in response to prolonged H2O2-induced stress. The majority of the modulated genes were hypothetical or of unknown function, although some are known to participate in oxidative stress resistance. The promoter region of several of the most highly modulated genes contained conserved motifs. In electrophoretic mobility shift assays, the purified rPG2212 protein did not bind its own promoter region but bound a similar region in several of the genes modulated in the PG_2212-deficient mutant. A metabolome analysis revealed that PG2212 can regulate a number of genes coding for proteins involved in metabolic pathways critical for its survival under the conditions of oxidative stress. Collectively, our data suggest that PG2212 is a transcriptional regulator that plays an important role in oxidative stress resistance and virulence regulation in P. gingivalis.
Collapse
|
43
|
Huang Y, Zhu M, Li Z, Sa R, Chu Q, Zhang Q, Zhang H, Tang W, Zhang M, Yin H. Mass spectrometry-based metabolomic profiling identifies alterations in salivary redox status and fatty acid metabolism in response to inflammation and oxidative stress in periodontal disease. Free Radic Biol Med 2014; 70:223-32. [PMID: 24607715 DOI: 10.1016/j.freeradbiomed.2014.02.024] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Revised: 02/24/2014] [Accepted: 02/25/2014] [Indexed: 11/25/2022]
Abstract
Periodontal diseases represent the most common chronic inflammatory diseases in humans and a major cause of tooth loss. Combining mass spectrometry-based ionomics and targeted lipidomics on fatty acid metabolites, we identified significant alterations in redox status and fatty acid metabolism in saliva in response to chronic inflammation and oxidative stress in periodontal disease in a cohort of nonsmoker subjects with chronic periodontitis. For the first time, ionomic profiling of around 30 ions in saliva revealed significantly decreased levels of redox-active metal ions including Mn, Cu, and Zn in the periodontal group, which is consistent with decreased levels of superoxide dismutases in saliva and serum. A targeted lipidomic approach was employed to monitor the major metabolites of arachidonic acid and linoleic acid in saliva. We observed increased levels of cyclooxygenase products including PGE2, PGD2, and PGF2α and TXB2, but decreased level of PGI2 in the periodontal group. A unique pattern of the lipoxygenase products of arachidonic acid and linoleic acid was observed with increased level of 5-HETE but decreased levels of 13-HODE and 9-HODE. Levels of salivary F2-isoprostanes, free radical lipid peroxidation products, and a gold standard for oxidative stress in vivo were also significantly elevated. Taking these data together, our study using multiple powerful omics techniques demonstrates that local redox alteration contributes significantly to periodontitis through the modulation of fatty acid metabolism in response to inflammation and oxidative stress. This study highlights the importance of redox status in periodontitis and provides a rationale for preventing periodontal disease by dietary interventions aiming to restore redox balance.
Collapse
Affiliation(s)
- Yijing Huang
- Department of Clinical Nutrition, Shanghai No. 9 People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Mingjiang Zhu
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing, China
| | - Zi Li
- Mass Spectrometry Research Center, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Rina Sa
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; University of the Chinese Academy of Sciences, Shanghai, China
| | - Qianqian Chu
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing, China
| | - Qingli Zhang
- Mass Spectrometry Research Center, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Haifeng Zhang
- Department of Clinical Nutrition, Shanghai No. 9 People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wen Tang
- Department of Clinical Nutrition, Shanghai No. 9 People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Meifang Zhang
- Department of Clinical Nutrition, Shanghai No. 9 People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Huiyong Yin
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing, China; Mass Spectrometry Research Center, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
44
|
Henry LG, Boutrin MC, Aruni W, Robles A, Ximinies A, Fletcher HM. Life in a Diverse Oral Community - Strategies for Oxidative Stress Survival. J Oral Biosci 2014; 56:63-71. [PMID: 26744578 DOI: 10.1016/j.job.2014.03.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND While the oral cavity harbors more than 680 bacterial species, the interaction and association of selected bacterial species play a role in periodontal diseases. Bacterial species including Porphyromonas gingivalis, Treponema denticola and Tannerella forsythia, a consortium previously designated as the "red complex" is now being expanded to include other new emerging pathogens that are significantly associated with periodontal disease. HIGHLIGHT In addition to novel mechanisms for oxidative resistance of individual species, community dynamics may lead to an overall strategy for survival in the inflammatory environment of the periodontal pocket. Complex systems controlled by response regulators protect against oxidative and nitrosative stress. CONCLUSION The combination of these multifaceted strategies would provide a comprehensive defense and support system against the repetitive host immune response to promote microbial persistence and disease.
Collapse
Affiliation(s)
- Leroy G Henry
- Division of Microbiology and Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California 92350
| | - Marie-Claire Boutrin
- Division of Microbiology and Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California 92350
| | - Wilson Aruni
- Division of Microbiology and Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California 92350
| | - Antonette Robles
- Division of Microbiology and Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California 92350
| | - Alexia Ximinies
- Division of Microbiology and Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California 92350
| | - Hansel M Fletcher
- Division of Microbiology and Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California 92350
| |
Collapse
|
45
|
Porphyromonas gingivalis: major periodontopathic pathogen overview. J Immunol Res 2014; 2014:476068. [PMID: 24741603 PMCID: PMC3984870 DOI: 10.1155/2014/476068] [Citation(s) in RCA: 292] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 02/21/2014] [Accepted: 02/21/2014] [Indexed: 12/24/2022] Open
Abstract
Porphyromonas gingivalis is a Gram-negative oral anaerobe that is involved in the pathogenesis of periodontitis and is a member of more than 500 bacterial species that live in the oral cavity. This anaerobic bacterium is a natural member of the oral microbiome, yet it can become highly destructive (termed pathobiont) and proliferate to high cell numbers in periodontal lesions: this is attributed to its arsenal of specialized virulence factors. The purpose of this review is to provide an overview of one of the main periodontal pathogens—Porphyromonas gingivalis. This bacterium, along with Treponema denticola and Tannerella forsythia, constitute the “red complex,” a prototype polybacterial pathogenic consortium in periodontitis. This review outlines Porphyromonas gingivalis structure, its metabolism, its ability to colonize the epithelial cells, and its influence upon the host immunity.
Collapse
|
46
|
Graziano TS, Closs P, Poppi T, Franco GC, Cortelli JR, Groppo FC, Cogo K. Catecholamines promote the expression of virulence and oxidative stress genes in Porphyromonas gingivalis. J Periodontal Res 2013; 49:660-9. [DOI: 10.1111/jre.12148] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2013] [Indexed: 01/04/2023]
Affiliation(s)
- T. S. Graziano
- Department of Physiological Sciences; Area of Pharmacology, Anesthesiology and Therapeutics; Piracicaba Dental School; University of Campinas; Piracicaba SP Brazil
| | - P. Closs
- Department of Periodontology; Dentistry School University of Taubaté; Taubaté SP Brazil
| | - T. Poppi
- Department of Periodontology; Dentistry School University of Taubaté; Taubaté SP Brazil
| | - G. C. Franco
- Laboratory of Physiology and Pathophysiology; Department of General Biology; State University of Ponta Grossa; Ponta Grossa PR Brazil
| | - J. R. Cortelli
- Department of Periodontology; Dentistry School University of Taubaté; Taubaté SP Brazil
| | - F. C. Groppo
- Department of Physiological Sciences; Area of Pharmacology, Anesthesiology and Therapeutics; Piracicaba Dental School; University of Campinas; Piracicaba SP Brazil
| | - K. Cogo
- Department of Dentistry; Implantology Area; University of Santo Amaro; São Paulo SP Brazil
| |
Collapse
|
47
|
Krishnan K, Duncan MJ. Role of sodium in the RprY-dependent stress response in Porphyromonas gingivalis. PLoS One 2013; 8:e63180. [PMID: 23671672 PMCID: PMC3646045 DOI: 10.1371/journal.pone.0063180] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Accepted: 03/31/2013] [Indexed: 01/09/2023] Open
Abstract
Porphyromonas gingivalis is a Gram-negative oral anaerobe which is strongly associated with periodontal disease. Environmental changes in the gingival sulcus trigger the growth of P. gingivalis and a concurrent shift from periodontal health to disease. Bacteria adjust their physiology in response to environmental changes and gene regulation by two-component phospho-relay systems is one mechanism by which such adjustments are effected. In P. gingivalis RprY is an orphan response regulator and previously we showed that the RprY regulon included genes associated with oxidative stress and sodium metabolism. The goals of the present study were to identify environmental signals that induce rprY and clarify the role of the regulator in the stress response. In Escherichia coli an RprY-LacZ fusion protein was induced in sodium- depleted medium and a P. gingivalis rprY mutant was unable to grow in similar medium. By several approaches we established that sodium depletion induced up-regulation of genes involved in oxidative stress. In addition, we demonstrated that RprY interacted directly with the promoters of several molecular chaperones. Further, both genetic and transcription data suggest that the regulator acts as a repressor. We conclude that RprY is one of the regulators that controls stress responses in P. gingivalis, possibly by acting as a repressor since an rprY mutant showed a superstress reponse in sodium-depleted medium which we propose inhibited growth.
Collapse
Affiliation(s)
- Karthik Krishnan
- Department of Microbiology, The Forsyth Institute, Cambridge, Massachusetts, United States of America
| | | |
Collapse
|
48
|
Choi CH, Spooner R, DeGuzman J, Koutouzis T, Ojcius DM, Yilmaz Ö. Porphyromonas gingivalis-nucleoside-diphosphate-kinase inhibits ATP-induced reactive-oxygen-species via P2X7 receptor/NADPH-oxidase signalling and contributes to persistence. Cell Microbiol 2013; 15:961-76. [PMID: 23241000 DOI: 10.1111/cmi.12089] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2012] [Revised: 12/04/2012] [Accepted: 12/05/2012] [Indexed: 12/21/2022]
Abstract
Ligation of P2X7 receptors with a 'danger signal', extracellular ATP (eATP), has recently been shown to result in production of intracellular reactive-oxygen-species (ROS) in macrophages. We show that primary gingival epithelial cells (GECs) produce sustained, robust cellular ROS upon stimulation by eATP. The induction of ROS was mediated by P2X7 receptor signalling coupled with NADPH-oxidase activation, as determined by pharmacological inhibition and RNA interference. Furthermore, Porphyromonas gingivalis, an oral opportunistic pathogen, upregulated the antioxidant glutathione response, modulated eATP-induced cytosolic and mitochondrial ROS generated through P2X7 /NADPH-oxidase interactome, and subsequently blocked oxidative stress in GECs via temporal secretion of a P. gingivalis effector, nucleoside-diphosphate-kinase (Ndk). An ndk-deficient P. gingivalis mutant lacked the ability to inhibit ROS production and persist intracellularly following eATP stimulation. Treatment with recombinant Ndk significantly diminished eATP-evoked ROS production. P. gingivalis infection elicited a strong, time-dependent increase in anti-oxidativemitochondrial UCP2 levels, whereas ndk-deficient mutant did not cause any change. The results reveal a novel signalling cascade that is tightly coupled with eATP signalling and ROS regulation. Ndk by P. gingivalis counteracts these antimicrobial signalling activities by secreting Ndk, thus contributing to successful persistence of the pathogen.
Collapse
Affiliation(s)
- Chul Hee Choi
- Department of Periodontology, University of Florida, Gainesville, FL 32610, USA
| | | | | | | | | | | |
Collapse
|
49
|
Aruni AW, Robles A, Fletcher HM. VimA mediates multiple functions that control virulence in Porphyromonas gingivalis. Mol Oral Microbiol 2012; 28:167-80. [PMID: 23279905 DOI: 10.1111/omi.12017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2012] [Indexed: 12/31/2022]
Abstract
Porphyromonas gingivalis, a black-pigmented, gram-negative anaerobe, is an important etiological agent of periodontal disease. Its ability to survive in the periodontal pocket and orchestrate the microbial/host activities that can lead to disease suggest that P. gingivalis possesses a complex regulatory network involving transcriptional and post-transcriptional mechanisms. The vimA (virulence modulating) gene is part of the 6.15-kb bcp-recA-vimA-vimE-vimF-aroG locus and plays a role in oxidative stress resistance. In addition to the glycosylation and anchorage of several surface proteins including the gingipains, VimA can also modulate sialylation, acetyl coenzyme A transfer, lipid A and its associated proteins and may be involved in protein sorting and transport. In this review, we examine the multifunctional role of VimA and discuss its possible involvement in a major regulatory network important for survival and virulence regulation in P. gingivalis. It is postulated that the multifunction of VimA is modulated via a post-translational mechanism involving acetylation.
Collapse
Affiliation(s)
- A W Aruni
- Division of Microbiology and Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | | | | |
Collapse
|
50
|
McKenzie RME, Johnson NA, Aruni W, Dou Y, Masinde G, Fletcher HM. Differential response of Porphyromonas gingivalis to varying levels and duration of hydrogen peroxide-induced oxidative stress. MICROBIOLOGY-SGM 2012; 158:2465-2479. [PMID: 22745271 DOI: 10.1099/mic.0.056416-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Porphyromonas gingivalis, an anaerobic oral pathogen implicated in adult periodontitis, can exist in an environment of oxidative stress. To evaluate its adaptation to this environment, we have assessed the response of P. gingivalis W83 to varying levels and durations of hydrogen peroxide (H(2)O(2))-induced stress. When P. gingivalis was initially exposed to a subinhibitory concentration of H(2)O(2) (0.1 mM), an adaptive response to higher concentrations could be induced. Transcriptome analysis demonstrated that oxidative stress can modulate several functional classes of genes depending on the severity and duration of the exposure. A 10 min exposure to H(2)O(2) revealed increased expression of genes involved in DNA damage and repair, while after 15 min, genes involved in protein fate, protein folding and stabilization were upregulated. Approximately 9 and 2.8% of the P. gingivalis genome displayed altered expression in response to H(2)O(2) exposure at 10 and 15 min, respectively. Substantially more genes were upregulated (109 at 10 min; 47 at 15 min) than downregulated (76 at 10 min; 11 at 15 min) by twofold or higher in response to H(2)O(2) exposure. The majority of these modulated genes were hypothetical or of unknown function. One of those genes (pg1372) with DNA-binding properties that was upregulated during prolonged oxidative stress was inactivated by allelic exchange mutagenesis. The isogenic mutant P. gingivalis FLL363 (pg1372 : : ermF) showed increased sensitivity to H(2)O(2) compared with the parent strain. Collectively, our data indicate the adaptive ability of P. gingivalis to oxidative stress and further underscore the complex nature of its resistance strategy under those conditions.
Collapse
Affiliation(s)
- Rachelle M E McKenzie
- Center for Dental Research, School of Dentistry, Loma Linda University, Loma Linda, CA 92350, USA.,Division of Microbiology and Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Neal A Johnson
- Department of Oral Diagnosis, Radiology, and Pathology, School of Dentistry, Loma Linda University, Loma Linda, CA 92350, USA.,Center for Dental Research, School of Dentistry, Loma Linda University, Loma Linda, CA 92350, USA
| | - Wilson Aruni
- Division of Microbiology and Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Yuetan Dou
- Division of Microbiology and Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Godfred Masinde
- Division of Microbiology and Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Hansel M Fletcher
- Division of Microbiology and Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| |
Collapse
|