1
|
Liao L, Wang Q, Feng Y, Li G, Lai R, Jameela F, Zhan X, Liu B. Advances and challenges in the development of periodontitis vaccines: A comprehensive review. Int Immunopharmacol 2024; 140:112650. [PMID: 39079346 DOI: 10.1016/j.intimp.2024.112650] [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: 02/14/2024] [Revised: 06/28/2024] [Accepted: 07/05/2024] [Indexed: 09/01/2024]
Abstract
Periodontitis is a prevalent polymicrobial disease. It damages soft tissues and alveolar bone, and causes a significant public-health burden. Development of an advanced therapeutic approach and exploration of vaccines against periodontitis hold promise as potential treatment avenues. Clinical trials for a periodontitis vaccine are lacking. Therefore, it is crucial to address the urgent need for developing strategies to implement vaccines at the primary level of prevention in public health. A deep understanding of the principles and mechanisms of action of vaccines plays a crucial role in the successful development of vaccines and their clinical translation. This review aims to provide a comprehensive summary of potential directions for the development of highly efficacious periodontitis vaccines. In addition, we address the limitations of these endeavors and explore future possibilities for the development of an efficacious vaccine against periodontitis.
Collapse
Affiliation(s)
- Lingzi Liao
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Qi Wang
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, College of Chemistry and Chemical Engineering, Linyi University, Linyi, China
| | - Yujia Feng
- School of Stomatology, Jinan University, Guangzhou, China
| | - Guojiang Li
- School of Stomatology, Jinan University, Guangzhou, China
| | - Renfa Lai
- Hospital of Stomatology, the First Affiliated Hospital of Jinan University, Guangzhou, China; School of Stomatology, Jinan University, Guangzhou, China
| | - Fatima Jameela
- Modern American Dental Clinic, West Warren Avenue, MI, USA
| | - Xiaozhen Zhan
- Hospital of Stomatology, the First Affiliated Hospital of Jinan University, Guangzhou, China; School of Stomatology, Jinan University, Guangzhou, China.
| | - Bin Liu
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China.
| |
Collapse
|
2
|
Yu F, Xu QA, Chen W. A targeted fimA DNA vaccine prevents alveolar bone loss in mice after intra-nasal administration. J Clin Periodontol 2011; 38:334-40. [PMID: 21261672 DOI: 10.1111/j.1600-051x.2010.01700.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
AIM To construct a dendritic cell (DC)-targeted DNA vaccine against FimA of Porphyromonas gingivalis and evaluate the immunogenicity and protection in mice. MATERIALS AND METHODS A targeted DNA plasmid pCTLA4-FimA, which encodes the signal peptide and extracellular regions of mouse cytotoxic T lymphocyte-associated antigen 4 (CTLA4), the hinge and Fc regions of human Igγ1 and FimA of P. gingivalis, was constructed. Mice were immunized with pCTLA4-FimA, the non-targeted DNA plasmid pFimA, which contains only fimA gene, or pCI vector intra-nasally. Serum and saliva antibody responses were detected by enzyme-linked immunosorbent assay. The protection against P. gingivalis-induced periodontitis was evaluated by measuring alveolar bone loss in mice. RESULTS Mice immunized with pCTLA4-FimA showed elevated levels of specific serum IgG and salivary IgA antibody responses compared with mice immunized with pFimA (p<0.01). Both pFimA and pCTLA4-FimA immunized groups showed significantly lower alveolar bone loss, with the magnitude protection greater in the latter (p<0.01), compared with the pCI immunized group. CONCLUSIONS The DC-targeted DNA construct pCTLA4-FimA enhanced both systemic and mucosal immunity following intra-nasal immunization. A DNA-based immunization strategy may be an effective way to attenuate periodontitis induced by P. gingivalis.
Collapse
Affiliation(s)
- Fei Yu
- Center of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | | | | |
Collapse
|
3
|
Koh EM, Kim J, Lee JY, Kim TG. Production of Monoclonal Antibodies Specific to FimA of Porphyromonas gingivalis and Their Inhibitory Activity on Bacterial Binding. Immune Netw 2009; 9:203-7. [PMID: 20157608 PMCID: PMC2816954 DOI: 10.4110/in.2009.9.5.203] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2009] [Revised: 09/24/2009] [Accepted: 09/28/2009] [Indexed: 01/08/2023] Open
Abstract
Background The FimA of Porphyromonas gingivalis is a crucial pathogenic component of the bacteria and has been implicated as a target for vaccine development against the periodontal diseases. Methods In this study, the purified fimbriae (FimA subunit polymers) protein was used for immunization in their native form and B hybridoma clones producing antibodies specific to FimA were established. Results The monoclonal antibodies prepared from selected two clones, designated #123 (IgG2b/ kappa) and #265 (IgG1/kappa), displayed different patterns of binding activity against the cognate antigen. Both antibodies reacted with conformational epitopes expressed by partially dissociated oligomers, but not with monomer as elucidated by Western blot analysis. Ascites fluid containing the monoclonal antibodies showed the inhibitory activity against P. gingivalis to saliva-coated hydroxyapatite beads, an in vitro model for the pellicle-coated tooth surface. Conclusion These results suggest that the monoclonal antibodies could be used as vaccine material against the periodontal diseases through passive immunization.
Collapse
Affiliation(s)
- Eun-Mi Koh
- Division of Biological Sciences and The Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju 561-756, Korea
| | | | | | | |
Collapse
|
4
|
Hardham J, Sfintescu C, Evans RT. Evaluation of cross-protection by immunization with an experimental trivalent companion animal periodontitis vaccine in the mouse periodontitis model. J Vet Dent 2008; 25:23-7. [PMID: 18512622 DOI: 10.1177/089875640802500107] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Companion animal periodontal disease is one of the most prevalent diseases seen by veterinarians. The goal of this study was to evaluate the vaccine performance of a trivalent canine periodontitis vaccine in the mouse oral challenge model of periodontitis. Mice vaccinated subcutaneously with an inactivated, whole-cell vaccine preparation of Porphyromonas denticanis, Porphyromonas gulae, and Porphyromonas salivosa displayed significantly reduced alveolar bone loss in response to heterologous and cross-species challenges as compared to sham vaccinated animals. Based on the results of these studies, a periodontitis vaccine may be a useful tool in preventing the initiation and progression of periodontitis caused by the most commonly isolated pigmenting anaerobic bacteria in animals.
Collapse
Affiliation(s)
- John Hardham
- Pfizer Inc., Veterinary Medicine Research and Development, Kalamazoo, MI 49001, USA.
| | | | | |
Collapse
|
5
|
Petrunov B, Marinova S, Markova R, Nenkov P, Nikolaeva S, Nikolova M, Taskov H, Cvetanov J. Cellular and humoral systemic and mucosal immune responses stimulated in volunteers by an oral polybacterial immunomodulator "Dentavax". Int Immunopharmacol 2006; 6:1181-93. [PMID: 16714223 DOI: 10.1016/j.intimp.2006.02.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2005] [Revised: 11/21/2005] [Accepted: 02/28/2006] [Indexed: 11/17/2022]
Abstract
The oral polybacterial immunomodulator Dentavax (D), composed of killed cells from Klebsiella pneumoniae, Streptococcus pyogenes, Staphylococcus aureus, Candida albicans and Lactobacillus acidophilus and their lysates was created for immunoprophylaxis and therapy of oral mucosa and parodont inflammations. The stimulating effect of the preparation was evaluated in twelve volunteers immunized for 10 consecutive days. On days 7, 14, 21, 28 and 49 after the last immunization peripheral blood (PB) lymphocyte subsets, T lymphocyte activation and PB phagocytic activity, were studied by flow cytometry. PB lymphocyte proliferative responses to PHA, rIL-2, LPS and D were evaluated radiometrically. The production of TNF-alpha in supernatants of in vitro stimulated lymphocytes and specific IgA, IgM and IgG antibodies in serum and saliva was determined by ELISA. Ultrastructural morphologic changes in T and B lymphocyte populations were also investigated. Although no significant changes in the levels of basic lymphocyte subsets were detected, the early/late (CD57+/CD57-) CD8 T effectors ratio was increased at the end of the studied period, as were the percentage of PHA-responding (CD69+) T cells and PB phagocytizing cells. The most prominent lymphoprolipherative responses were measured upon costimulation with LPS+D and PHA+D on day 21. Electron-microscopic studies demonstrated a significant effect of D on both T and B cell activity. TNF-alpha concentration increased progressively from day 7 till the end of the investigation. Maximal concentrations were observed after stimulation with D and LPS. An increased level of specific salivary and serum antibodies against the components of D was found, with highest levels between days 7 and 21. Specific secretory IgA predominated in saliva as compared to IgM and IgG. Our results demonstrate the stimulating effect of Dentavax on PB lymphocyte functional activity and the specific humoral systemic and mucosal immunity.
Collapse
Affiliation(s)
- Bogdan Petrunov
- National Center of Infectious and Parasitic Diseases (NCIPD), Sofia, Bulgaria
| | | | | | | | | | | | | | | |
Collapse
|
6
|
Hardham J, Reed M, Wong J, King K, Laurinat B, Sfintescu C, Evans RT. Evaluation of a monovalent companion animal periodontal disease vaccine in an experimental mouse periodontitis model. Vaccine 2005; 23:3148-56. [PMID: 15837214 DOI: 10.1016/j.vaccine.2004.12.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2004] [Revised: 12/17/2004] [Accepted: 12/21/2004] [Indexed: 11/24/2022]
Abstract
Periodontal disease in companion animals is clinically similar to that of human periodontal disease. Despite the usage of veterinary procedures and antibiotic therapy, the disease still remains as one of the most highly prevalent disorders seen by veterinarians. The goal of this study was to evaluate the immunogenic properties and vaccine performance of a monovalent canine periodontal disease vaccine in the mouse oral challenge model of periodontitis. Mice vaccinated subcutaneously with inactivated, whole-cell bacterin preparations of Porphyromonas gulae displayed both high titers of anti-P. gulae specific antibodies and significantly reduced alveolar bone loss in response to homologous, heterologous, and cross-species challenge. Based on the results of these studies, a periodontal disease vaccine may be a useful tool in preventing the progression of periodontitis in animals.
Collapse
Affiliation(s)
- John Hardham
- Pfizer Inc., Veterinary Medicine Research and Development, 301 Henrietta Street, Kalamazoo, MI 49001, USA.
| | | | | | | | | | | | | |
Collapse
|
7
|
Hardham J, Dreier K, Wong J, Sfintescu C, Evans RT. Pigmented-anaerobic bacteria associated with canine periodontitis. Vet Microbiol 2005; 106:119-28. [PMID: 15737481 DOI: 10.1016/j.vetmic.2004.12.018] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2004] [Revised: 12/14/2004] [Accepted: 12/17/2004] [Indexed: 11/15/2022]
Abstract
The etiology of human periodontal disease has been the focus of considerable research, yet relatively little is known about the causative agents of companion animal periodontitis. In humans, Porphyromonas gingivalis, a black-pigmented anaerobic bacteria (BPAB), has been implicated as the primary periopathogen. It has been demonstrated that BPAB are also found in companion animal periodontal pockets. While some animal BPAB have been individually identified, a study to identify the most frequently isolated subgingival BPAB has not been completed using genetic tools. The objective of this work was to identify the types and relative frequencies of pigmented anaerobic bacteria found in the periodontal pockets of dogs. Porphyromonas salivosa, Porphyromonas denticanis (a novel species) and Porphyromonas gulae were found to be the most frequently isolated BPAB associated with canine periodontitis.
Collapse
Affiliation(s)
- John Hardham
- Pfizer Inc., Veterinary Medicine Research and Development, 301 Henrietta Street, Kalamazoo, MI 49001, USA.
| | | | | | | | | |
Collapse
|
8
|
O'Brien-Simpson NM, Veith PD, Dashper SG, Reynolds EC. Antigens of bacteria associated with periodontitis. Periodontol 2000 2004; 35:101-34. [PMID: 15107060 DOI: 10.1111/j.0906-6713.2004.003559.x] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Neil M O'Brien-Simpson
- Centre for Oral Health Science, School of Dental Science, The University of Melbourne, Victoria, Australia
| | | | | | | |
Collapse
|
9
|
Gibson FC, Gonzalez DA, Wong J, Genco CA. Porphyromonas gingivalis-specific immunoglobulin G prevents P. gingivalis-elicited oral bone loss in a murine model. Infect Immun 2004; 72:2408-11. [PMID: 15039370 PMCID: PMC375152 DOI: 10.1128/iai.72.4.2408-2411.2004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Active immunization with Porphyromonas gingivalis whole-cell preparations has been shown to prevent P. gingivalis infection and oral bone loss. Employing passive antibody transfer and opsonization, we demonstrate with this study that immunization-elicited P. gingivalis-specific immunoglobulin G facilitates clearance of P. gingivalis in a subcutaneous chamber model and prevents P. gingivalis-elicited oral bone loss.
Collapse
Affiliation(s)
- Frank C Gibson
- Department of Medicine, Section of Infectious Diseases, Boston University Medical Center, Boston, Massachusetts 02118, USA
| | | | | | | |
Collapse
|
10
|
Tanaka S, Murakami Y, Ogiwara T, Shoji M, Seto K, Nagasaki M, Fujisawa S. Frequency of reactivity for Porphyromonas gingivalis and Prevotella spp. in supra- and subgingival plaques, and periodontal clinical parameters according to subject age. J Periodontol 2002; 73:877-85. [PMID: 12211497 DOI: 10.1902/jop.2002.73.8.877] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND The present study was conducted to assess the association between selected clinical parameters and the distribution of Porphyromonas gingivalis (Pg), Prevotella intermedia (Pi), Prevotella nigrescens (Pn), and Prevotella melaninogenica (Pm) in supra- and subgingival plaque samples measured by an immunoslot blot assay (IBA) using their monoclonal antibodies. METHODS Plaque samples from 299 patients aged 6 to 69 randomly chosen from a group of dental outpatients were examined. Plaque index, gingival index, and probing depths were evaluated according to the criteria of positive (cell number > or = 10(6)) or negative (<106) reactivity to the 4 different monoclonal antibodies. RESULTS An increase in probing depth in subjects exhibiting either a positive or negative reaction for the 4 test bacteria was associated with increasing age. Comparing bacteria-positive subgingival plaque samples to their corresponding bacteria-negative counterparts, we found an increased plaque index in children positive for any of the 4 bacteria; in addition, that for Pg and Pi was increased in subjects 40 to 49 years old. The gingival index increased with increasing amount of Pi and Pn, but not with Pg and Pm in those 20 to 29 years of age. The frequency of Pg reactivity in subgingival plaque was markedly enhanced in subjects older than 30 to 39 years of age, and was significantly higher than that in supragingival plaque. The frequency of Pi and Pn reactivity was significantly increased in adults aged 20 to 29 and plateaued at older ages. The frequency of Pm reactivity was relatively low and independent of subject age. CONCLUSIONS The increase in probing depth with increasing age was not affected by the occurrence of periodontopathic bacteria. The high rate of occurrence of Pg, together with Pi and Pn, in subgingival plaque of the adult age groups was suggested to be associated with the high frequency of periodontal disease in the older age groups (above 30 to 49 years of age). The IBA appears to be useful for the efficient and rapid detection of periodontopathic bacteria.
Collapse
Affiliation(s)
- Shoji Tanaka
- Department of Oral Diagnosis, Meikai University School of Dentistry, Saitama, Japan
| | | | | | | | | | | | | |
Collapse
|
11
|
Abiko Y. Passive immunization against dental caries and periodontal disease: development of recombinant and human monoclonal antibodies. CRITICAL REVIEWS IN ORAL BIOLOGY AND MEDICINE : AN OFFICIAL PUBLICATION OF THE AMERICAN ASSOCIATION OF ORAL BIOLOGISTS 2002; 11:140-58. [PMID: 12002812 DOI: 10.1177/10454411000110020101] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Indigenous micro-organisms in the oral cavity can cause two major diseases, dental caries and periodontal diseases. There is neither agreement nor consensus as to the actual mechanisms of pathogenesis of the specific virulence factors of these micro-organisms. The complexity of the bacterial community in dental plaque has made it difficult for the single bacterial agent of dental caries to be determined. However, there is considerable evidence that Streptococcus mutans is implicated as the primary causative organism of dental caries, and the cell-surface protein antigen (SA I/II) as well as glucosyltransferases (GTFs) produced by S. mutans appear to be major colonization factors. Various forms of periodontal diseases are closely associated with specific subgingival bacteria. Porphyromonas gingivalis has been implicated as an important etiological agent of adult periodontitis. Adherence of bacteria to host tissues is a prerequisite for colonization and one of the important steps in the disease process. Bacterial coaggregation factors and hemagglutinins likely play major roles in colonization in the subgingival area. Emerging evidence suggests that inhibition of these virulence factors may protect the host against caries and periodontal disease. Active and passive immunization approaches have been developed for immunotherapy of these diseases. Recent advances in mucosal immunology and the introduction of novel strategies for inducing mucosal immune responses now raise the possibility that effective and safe vaccines can be constructed. In this regard, some successful results have been reported in animal experimental models. Nevertheless, since the public at large might be skeptical about the seriousness of oral diseases, immunotherapy must be carried out with absolute safety. For this goal to be achieved, the development of safe antibodies for passive immunization is significant and important. In this review, salient advances in passive immunization against caries and periodontal diseases are summarized, and the biotechnological approaches for developing recombinant and human-type antibodies are introduced. Furthermore, our own attempts to construct single-chain variable fragments (ScFv) and human-type antibodies capable of neutralizing virulence factors are discussed.
Collapse
Affiliation(s)
- Y Abiko
- Department of Biochemistry, Nihon University School of Dentistry at Matsudo, Chiba, Japan.
| |
Collapse
|
12
|
Abstract
This paper was prepared by the Research, Science, and Therapy Committee of the American Academy of Periodontology to provide the dental profession an overview of current and potential methods to modulate the host response in the treatment of periodontal diseases. Specifically, it discusses components of periodontal disease pathogenesis (i.e., immune and inflammatory responses, excessive production of matrix metalloproteinases and arachidonic acid metabolites, and regulation of bone metabolism) and their modulation.
Collapse
|
13
|
Sharma A, Honma K, Evans RT, Hruby DE, Genco RJ. Oral immunization with recombinant Streptococcus gordonii expressing porphyromonas gingivalis FimA domains. Infect Immun 2001; 69:2928-34. [PMID: 11292708 PMCID: PMC98244 DOI: 10.1128/iai.69.5.2928-2934.2001] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Porphyromonas gingivalis, a gram-negative anaerobe, is implicated in the etiology of adult periodontitis. P. gingivalis fimbriae are one of several critical surface virulence factors involved in both bacterial adherence and inflammation. P. gingivalis fimbrillin (FimA), the major subunit protein of fimbriae, is considered an important antigen for vaccine development against P. gingivalis-associated periodontitis. We have previously shown that biologically active domains of P. gingivalis fimbrillin can be expressed on the surface of the human commensal bacterium Streptococcus gordonii. In this study, we examined the effects of oral coimmunization of germfree rats with two S. gordonii recombinants expressing N (residues 55 to 145)- and C (residues 226 to 337)-terminal epitopes of P. gingivalis FimA to elicit FimA-specific immune responses. The effectiveness of immunization in protecting against alveolar bone loss following P. gingivalis infection was also evaluated. The results of this study show that the oral delivery of P. gingivalis FimA epitopes via S. gordonii vectors resulted in the induction of FimA-specific serum (immunoglobulin G [IgG] and IgA) and salivary (IgA) antibody responses and that the immune responses were protective against subsequent P. gingivalis-induced alveolar bone loss. These results support the potential usefulness of the S. gordonii vectors expressing P. gingivalis fimbrillin as a mucosal vaccine against adult periodontitis.
Collapse
Affiliation(s)
- A Sharma
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, New York 14214, USA.
| | | | | | | | | |
Collapse
|
14
|
Steffen MJ, Holt SC, Ebersole JL. Porphyromonas gingivalis induction of mediator and cytokine secretion by human gingival fibroblasts. ORAL MICROBIOLOGY AND IMMUNOLOGY 2000; 15:172-80. [PMID: 11154400 DOI: 10.1034/j.1399-302x.2000.150305.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We hypothesized that bacterial viability and strain characteristics of Porphyromonas gingivalis could affect the induction of pro-inflammatory mediator secretion by human gingival fibroblast cultures. Both killed and viable P. gingivalis elicited production of prostaglandin E2, interleukin-1 beta (IL-1 beta), IL-6 and IL-8, although killed P. gingivalis induced generally higher levels, particularly IL-6 and IL-8, compared with the viable bacteria. P. gingivalis strains, which exhibited wild-type levels of trypsin-like protease activity, stimulated human gingival fibroblasts to secrete increased levels of prostaglandin E2 and IL-1 beta, although minimal levels of IL-6 and IL-8 were noted in supernatants from the gingival fibroblast cells. P. gingivalis strains BEI and NG4B19, which have either decreased or undetectable levels of trypsin-like protease, respectively, induced significantly greater IL-6 and IL-8 levels in gingival fibroblast cultures compared with the other strains. The ability of antibody to P. gingivalis to alter human gingival fibroblast production of pro-inflammatory mediators was tested using nonhuman primate antisera. Both immune and nonimmune sera altered the P. gingivalis-generated pattern of mediators from the gingival fibroblasts. We conclude that: (i) viable and killed P. gingivalis were capable of inducing various pro-inflammatory cytokines from human gingival fibroblasts; (ii) strain differences in cytokine induction were noted, and the expression of a trypsin-like protease activity was related to decreased extracellular levels of IL-6 and IL-8; and (iii) the presence of serum, particularly with specific antibody to P. gingivalis, significantly altered human gingival fibroblast cytokine production compared with P. gingivalis alone.
Collapse
Affiliation(s)
- M J Steffen
- Department of Periodontics, University of Texas Health Science Center, 7703 Floyd Curl Dr., San Antonio, TX 78284, USA
| | | | | |
Collapse
|
15
|
Baker PJ, Dixon M, Evans RT, Roopenian DC. Heterogeneity of Porphyromonas gingivalis strains in the induction of alveolar bone loss in mice. ORAL MICROBIOLOGY AND IMMUNOLOGY 2000; 15:27-32. [PMID: 11155161 DOI: 10.1034/j.1399-302x.2000.150105.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
These experiments examine alveolar bone loss in a model in which specific pathogen-free mice are exposed orally with Porphyromonas gingivalis. Alveolar bone loss was induced as a result of a specific infection with P. gingivalis, rather than other environmental antigens. Infection with live P. gingivalis was required, as significant bone loss did not follow gavage with formalin-killed P. gingivalis. The virulence of different strains of P. gingivalis was compared. Two laboratory strains of the bacteria (ATCC 53977 and W50) and a mutant strain lacking the 43-kDa fimbrillin (strain DPG3) induced bone loss. P. gingivalis 381, however, did not induce bone loss. There was a strong immunoglobulin G (IgG) antibody response to infection with each strain but a significant serum IgA response only to strain 381. These studies show that in mice with a background oral microflora bone loss is induced by a specific infection with P. gingivalis and that bacterial strain variation is important in determining whether alveolar bone loss will ensue.
Collapse
Affiliation(s)
- P J Baker
- Biology Department, Bates College, Lewiston, ME 04240, USA
| | | | | | | |
Collapse
|
16
|
Naganuma K, Amano S, Takeda H, Kitano S, Hanazawa S. Role of transcriptional factor activation protein-1 in endogenous expression of the interleukin-1 beta gene involved in Porphyromonas gingivalis fimbria-stimulated bone resorption in the mouse calvarial system. ORAL MICROBIOLOGY AND IMMUNOLOGY 2000; 15:53-7. [PMID: 11155165 DOI: 10.1034/j.1399-302x.2000.150109.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We previously suggested that endogenous interleukin-1 beta (IL-1 beta) produced by Porphyromonas gingivalis fimbriae stimulation in calvarial bone cell cultures plays a role in bone resorption as a major cytokine. Therefore, in the present study, we initiated experiments to clarify the stimulatory mechanism of IL-1 beta gene expression in fimbria-stimulated bone resorption. Fimbria-stimulated bone resorption was dramatically inhibited by curcumin, a potent inhibitor of activation protein-1 (AP-1). In fact, the fimbriae induced markedly both the expression of c-fos and c-jun genes and their protein production in the calvarial cells. In addition, a mixture of antisense oligonucleotides against c-fos and c-jun significantly inhibited not only the fimbria-induced expression of the IL-1 beta gene but also the fimbria-induced bone resorption. Therefore, the present study demonstrates that transcriptional factor AP-1 plays a functional role in P. gingivalis fimbria-stimulated bone resorption via endogenous IL-1 beta in the mouse calvarial system.
Collapse
Affiliation(s)
- K Naganuma
- Department of Oral Microbiology, Meikai University School of Dentistry, Keyakidai 1-1, Sakado City, Saitama 350-0283, Japan
| | | | | | | | | |
Collapse
|
17
|
Baker PJ, Carter S, Dixon M, Evans RT, Roopenian DC. Serum antibody response to oral infection precedes but does not prevent Porphyromonas gingivalis-induced alveolar bone loss in mice. ORAL MICROBIOLOGY AND IMMUNOLOGY 1999; 14:194-6. [PMID: 10495715 DOI: 10.1034/j.1399-302x.1999.140309.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The purpose of this study was to determine whether humoral immunity prevents bacterially induced alveolar bone loss. BALB/cByJ mice were orally infected with the human periodontopathic bacterium Porphyromonas gingivalis, and were compared with sham-infected mice. Specific serum antibody titers to P. gingivalis were measured by enzyme-linked immunosorbent assay. Alveolar bone levels were measured as the distance from the cementoenamel junction to the alveolar bone crest and bone loss was defined as a change in bone levels over time or between infected and sham-infected animals. The specific humoral response was predominantly of the IgG isotype, although low levels of specific serum IgA were also present. Antibody titers in the infected animals were significantly different from those in the sham-infected animals by 18 days and remained at maximal levels at 47 days. Bone loss became significant by 26 days and continued to progress at 47 days. Thus the serum antibody response to oral infection with P. gingivalis preceded detectable bone loss and remained elevated while bone loss increased. The presence of specific antibody did not prevent the onset or progression of bone loss.
Collapse
Affiliation(s)
- P J Baker
- Biology Department, Bates College, Lewiston, Maine 04240, USA
| | | | | | | | | |
Collapse
|
18
|
Affiliation(s)
- S C Holt
- Department of Microbiology, University of Texas Health Science Center at San Antonio, Graduate School of Biomedical Sciences, USA
| | | | | | | |
Collapse
|
19
|
Sharma A, Honma K, Sojar HT, Hruby DE, Kuramitsu HK, Genco RJ. Expression of saliva-binding epitopes of the Porphyromonas gingivalis FimA protein on the surface of Streptococcus gordonii. Biochem Biophys Res Commun 1999; 258:222-6. [PMID: 10222264 DOI: 10.1006/bbrc.1999.0616] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Porphyromonas gingivalis, a gram-negative oral anaerobic bacterium, has been implicated in the onset and development of periodontitis. The P. gingivalis fimbriae which mediate bacterial adherence to host oral sites and induce host inflammatory responses have been suggested as a potential antigen candidate. for vaccine development. This study was undertaken to generate Streptococcus gordonii vectors expressing the major subunit protein (FimA) of P. gingivalis fimbriae for testing as a potential live vaccine against periodontitis. We report here the expression of the C-terminal saliva-binding epitopes of P. gingivalis FimA on the surface of S. gordonii and demonstrate that domains containing free cysteine residues are poorly expressed on the surface of S. gordonii.
Collapse
Affiliation(s)
- A Sharma
- School of Dental Medicine, State University of New York at Buffalo, Buffalo, New York, 14214, USA.
| | | | | | | | | | | |
Collapse
|
20
|
Kesavalu L, Holt SC, Ebersole JL. Virulence of a polymicrobic complex, Treponema denticola and Porphyromonas gingivalis, in a murine model. ORAL MICROBIOLOGY AND IMMUNOLOGY 1998; 13:373-7. [PMID: 9872114 DOI: 10.1111/j.1399-302x.1998.tb00694.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The effect of a polymicrobic infection employing Treponema denticola and Porphyromonas gingivalis in the murine lesion model was used to determine the synergistic virulence of these two periodontopathic bacteria. At high doses of P. gingivalis W50, addition of T. denticola in the infection mixture had no effect on the formation and size of the spreading lesion caused by this microorganism. However, at low P. gingivalis challenge doses, T. denticola significantly enhanced the virulence of P. gingivalis compared with monoinfection of this microorganism. A potential role of the trypsin-like protease enzyme activity of P. gingivalis in this synergistic virulence was tested using P. gingivalis mutants deficient (i.e., BEI) or devoid (i.e., NG4B19) of this protease activity. These findings demonstrated that T. denticola-P. gingivalis complexes exhibit enhanced virulence in this model and that even using a polymicrobic challenge infection, the trypsin-like protease activity was important to P. gingivalis virulence expression.
Collapse
Affiliation(s)
- L Kesavalu
- Department of Periodontics, University of Texas Health Science Center at San Antonio 78284, USA
| | | | | |
Collapse
|
21
|
Stashenko P, Teles R, D'Souza R. Periapical inflammatory responses and their modulation. CRITICAL REVIEWS IN ORAL BIOLOGY AND MEDICINE : AN OFFICIAL PUBLICATION OF THE AMERICAN ASSOCIATION OF ORAL BIOLOGISTS 1998; 9:498-521. [PMID: 9825224 DOI: 10.1177/10454411980090040701] [Citation(s) in RCA: 263] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Periapical inflammatory responses occur as a consequence of bacterial infection of the dental pulp, as a result of caries, trauma, or iatrogenic insult. Periapical inflammation stimulates the formation of granulomas and cysts, with the destruction of bone. These inflammatory responses are complex and consist of diverse elements. Immediate-type responses--including vasodilatation, increased vascular permeability, and leukocyte extravasation--are mediated by endogenous mediators, including prostanoids, kinins, and neuropeptides. Non-specific immune responses--including polymorphonuclear leukocyte and monocyte migration and activation, and cytokine production--are elicited in response to bacteria and their products. Interleukin-1 and prostaglandins in particular have been implicated as central mediators of periapical bone resorption. Chronic periapical inflammation further involves specific T- and B-cell-mediated anti-bacterial responses, and activates a network of regulatory cytokines which are produced by Th1- and Th2-type T-lymphocytes. Various naturally occurring and genetically engineered models of immunodeficiency are beginning to help elucidate those components of the immune system which protect the pulpal/periapical complex. Both specific and non-specific responses interface with and are regulated by the neural system. The modulation of these responses by immune response modifies, cytokine antagonists, and other novel therapeutic agents is discussed. As an experimental model, periapical inflammation has many advantages which permit it to be used in studies of microbial ecology and pathogenesis, host response, neuroimmunology, and bone resorption and regeneration.
Collapse
Affiliation(s)
- P Stashenko
- Department of Cytokine Biology, Forsyth Dental Center, Boston, Massachusetts, USA
| | | | | |
Collapse
|
22
|
Ramamurthy NS, Schroeder KL, McNamara TF, Gwinnett AJ, Evans RT, Bosko C, Golub LM. Root-surface caries in rats and humans: inhibition by a non-antimicrobial property of tetracyclines. Adv Dent Res 1998; 12:43-50. [PMID: 9972121 DOI: 10.1177/08959374980120011801] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The incidence of root caries has been found to increase as the population ages and as edentulism becomes less prevalent due to improved dental awareness and care, and as exposure of roots due to gingival recession has also increased in the elderly. The mechanism of root caries is thought to be mediated by both bacterial and mammalian proteases produced by plaque and the periodontal tissues, respectively. In the current study, a rat model of periodontal disease was used in which gnotobiotic rats were infected intra-orally with a periodontal pathogen (P. gingivalis). Infecting the rats with P. gingivalis increased the collagenase activity in the gingival tissue in association with severe alveolar bone loss. Treating P. gingivalis-infected rats with doxycycline or CMT-1 prevented the destruction of the periodontium by MMPs, thus preventing exposure of roots to subgingival bacterial plaque and host tissue collagenases and the subsequent development of root caries. In addition, a low-dose doxycycline (LDD, 20 mg bid, non-antimicrobial dose) for 3 months was used in humans predisposed to increased root caries as the result of heavy use of smokeless (chewing) tobacco, causing gingival recession, subgingival plaque accumulation with Gram-negative bacteria, increased gingival crevicular fluid flow (GCF), and elevated GCF collagenase. Daily administration of LDD in smokeless tobacco patients reduced the GCF collagenase and prevented the further development of root caries.
Collapse
Affiliation(s)
- N S Ramamurthy
- Department of Oral Biology and Pathology, School of Dental Medicine, SUNY at Stony Brook 11794-8702, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Marcotte H, Lavoie MC. Oral microbial ecology and the role of salivary immunoglobulin A. Microbiol Mol Biol Rev 1998; 62:71-109. [PMID: 9529888 PMCID: PMC98907 DOI: 10.1128/mmbr.62.1.71-109.1998] [Citation(s) in RCA: 301] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In the oral cavity, indigenous bacteria are often associated with two major oral diseases, caries and periodontal diseases. These diseases seem to appear following an imbalance in the oral resident microbiota, leading to the emergence of potentially pathogenic bacteria. To define the process involved in caries and periodontal diseases, it is necessary to understand the ecology of the oral cavity and to identify the factors responsible for the transition of the oral microbiota from a commensal to a pathogenic relationship with the host. The regulatory forces influencing the oral ecosystem can be divided into three major categories: host related, microbe related, and external factors. Among host factors, secretory immunoglobulin A (SIgA) constitutes the main specific immune defense mechanism in saliva and may play an important role in the homeostasis of the oral microbiota. Naturally occurring SIgA antibodies that are reactive against a variety of indigenous bacteria are detectable in saliva. These antibodies may control the oral microbiota by reducing the adherence of bacteria to the oral mucosa and teeth. It is thought that protection against bacterial etiologic agents of caries and periodontal diseases could be conferred by the induction of SIgA antibodies via the stimulation of the mucosal immune system. However, elucidation of the role of the SIgA immune system in controlling the oral indigenous microbiota is a prerequisite for the development of effective vaccines against these diseases. The role of SIgA antibodies in the acquisition and the regulation of the indigenous microbiota is still controversial. Our review discusses the importance of SIgA among the multiple factors that control the oral microbiota. It describes the oral ecosystems, the principal factors that may control the oral microbiota, a basic knowledge of the secretory immune system, the biological functions of SIgA, and, finally, experiments related to the role of SIgA in oral microbial ecology.
Collapse
Affiliation(s)
- H Marcotte
- Département de Microbiologie-Immunologie, Faculté de Médecine, Université Laval, Québec, Canada
| | | |
Collapse
|
24
|
Kesavalu L, Holt SC, Ebersole JL. Porphyromonas gingivalis virulence in a murine lesion model: effects of immune alterations. Microb Pathog 1997; 23:317-26. [PMID: 9441858 DOI: 10.1006/mpat.1997.0161] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
This study utilized various mouse strains with documented alterations in immune system components to assess their contribution to modify the virulence of Porphyromonas gingivalis. P. gingivalis W50 was cultivated on blood agar plates, harvested and used to challenge mice by subcutaneous injection on the dorsolateral surface of the back. Soft tissue lesion development was estimated by measuring the area of the spreading lesion formed by this microorganism over a period of 15 days. Challenge of various normal inbred and outbred mouse strains including: BALB/cN, BALB/cJ, BALB/c nu/+, ICR, B10.A(4R), B10.MBR, A/J, C57BL/6J, CBA/CaH, C.B-17/Icv Tacf DF and C3H/HeN with 2 x 10(10) bacteria showed similar lesion size among these strains (approximately 400 mm2). Genetically deficient mouse strains [C.B-17/Icr Tac (SCID); DBA/2 (C5 deficient); BALB/c nu/nu (T cell deficient); CBA/CaHN-XID/J (B cell deficient) and C3H/HeJ (LPS hyporesponsive)] demonstrated a lesion size which was similar to normal animals. C57BL/6J-BgJ (NK cell deficient) mice exhibited a significantly more severe lesion than the other strains tested. Following healing of the lesions, we initiated a secondary infection of the surviving animals to estimate the acquisition of protective immunity following recovery from the primary infection. Normal mice demonstrated a delayed onset and decrease in lesion size of 15 to 30% compared with the primary infection. In contrast, each of the immunodeficient strains appeared unable to develop immune protection to the secondary challenge. The findings suggest that protection against primary infections with P. gingivalis are mediated by innate immune mechanisms (PMN. NK cells). Additionally, it appears that T-cell-dependent humoral responses are critical to developing immunity to subsequent P. gingivalis infection.
Collapse
Affiliation(s)
- L Kesavalu
- Department of Periodontics, School of Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78284, USA
| | | | | |
Collapse
|
25
|
Sharma A, Sojar HT, Hruby DE, Kuramitsu HK, Genco RJ. Secretion of Porphyromonas gingivalis fimbrillin polypeptides by recombinant Streptococcus gordonii. Biochem Biophys Res Commun 1997; 238:313-6. [PMID: 9299501 DOI: 10.1006/bbrc.1997.7306] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The fimbriae of Porphyromonas gingivalis plays an important role in the pathogenesis of periodontal disease. A structural subunit of the P. gingivalis fimbriae, fimbrillin, has been shown to promote adherence of the bacteria to host surfaces and also induce an immune response. Biologically active domains of fimbrillin responsible for adherence or eliciting immune responses have been determined. In a previous study, we engineered the human oral commensal organism Streptococcus gordonii to express such biologically active domains on the surface of the bacteria as a vaccine delivery system. In this study we report an alternative approach of secreting fimbrillin polypeptide domains into the medium by modification of the surface-expression system described earlier. Such recombinant S. gordonii, in addition to being a source for antigen presentation to trigger a protective immune response, may have the added advantage of directly blocking the fimbriae-mediated adherence of P. gingivalis to the oral cavity following implantation. This approach can also be utilized for secreting other biologically important therapeutic molecules on mucosal surfaces for modulating local microenvironments.
Collapse
Affiliation(s)
- A Sharma
- School of Dental Medicine, State University of New York at Buffalo, Buffalo, New York 14214, USA.
| | | | | | | | | |
Collapse
|
26
|
Teles R, Wang CY, Stashenko P. Increased susceptibility of RAG-2 SCID mice to dissemination of endodontic infections. Infect Immun 1997; 65:3781-7. [PMID: 9284152 PMCID: PMC175539 DOI: 10.1128/iai.65.9.3781-3787.1997] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Specific immunity has been implicated in the pathogenesis of periapical lesions, although the extent to which these mechanisms are actually involved in either protection or destruction of the pulp-periapex complex is yet to be established. To investigate this question we compared periapical-lesion pathogenesis in RAG-2 severe combined immunodeficient (SCID) mice with immunocompetent control mice following surgical pulp exposure. In order to equalize the bacterial challenge, an infection protocol using Prevotella intermedia, Fusobacterium nucleatum, Peptostreptococcus micros, and Streptococcus intermedius was devised. The results demonstrated that after infection, the proportion of the root canal flora represented by the four pathogens was almost identical in both groups (39.9 and 42.2% for RAG-2 and immunocompetent control mice, respectively). The effects of abrogation of T- and B-cell mechanisms on periapical pathogenesis were then assessed. Approximately one-third of the RAG-2 mice developed endodontic abscesses, while no immunocompetent controls had abscesses, results which indicated regional dissemination of the infection. A similar incidence of abscesses was found in two additional experiments. Abscessed RAG-2 teeth had significantly larger periapical lesions than did nonabscessed RAG-2 teeth (P < or = 0.05) and exposed immunocompetent controls (P < or = 0.01), whereas nonabscessed RAG-2 teeth were not significantly different from those of exposed immunocompetent controls in periapical-lesion size. We conclude that B- and T-cell-mediated immunity protects the host from the dissemination of endodontic infections and that RAG-2 mice are more susceptible to infection-induced pulp-periapex destruction.
Collapse
Affiliation(s)
- R Teles
- Department of Cytokine Biology, Forsyth Dental Center, Boston, Massachusetts 02115, USA.
| | | | | |
Collapse
|
27
|
Ebersole JL, Feuille F, Kesavalu L, Holt SC. Host modulation of tissue destruction caused by periodontopathogens: effects on a mixed microbial infection composed of Porphyromonas gingivalis and Fusobacterium nucleatum. Microb Pathog 1997; 23:23-32. [PMID: 9250777 DOI: 10.1006/mpat.1996.0129] [Citation(s) in RCA: 52] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
These studies determined the ability of selected periodontopathogens to synergistically initiate soft tissue destruction in a murine abscess model. The development of immunity following recovery from infection or by active immunization was also examined. Mice were infected with P. gingivalis W50, F. nucleatum T18, or a combination of the two microorganisms. F. nucleatum caused only a localized lesion in contrast to P. gingivalis which caused a spreading suppurative inflammatory lesion of the skin and subcutaneous tissues, which, depending upon the dose, could result in death. Infection of mice with a combination of P. gingivalis and F. nucleatum elicited a significantly greater lesion size (P<0.001) and lethality compared with P. gingivalis alone. Mice infected with a subclinical dose (no visible lesion) of P. gingivalis failed to develop protective immunity to a secondary P. gingivalis challenge. Mice that had recovered from P. gingivalis lesions demonstrated partial protection against subsequent P. gingivalis challenge; however, the immunity was less protective against the mixed F. nucleatum + P. gingivalis infection. Active immunization with P. gingivalis protected against both the P. gingivalis and F. nucleatum + P. gingivalis challenges and this protection was correlated with the levels of specific serum immunoglobulin G (IgG) antibody. The results indicated that the murine model is ideally suited to examine bacterially-mediated mixed infections that result in soft tissue destruction. This destruction can be minimized, but not abrogated, with development of immunity. Challenge with sufficient numbers of the pathogens can overwhelm the acquired immunity.
Collapse
Affiliation(s)
- J L Ebersole
- Department of Periodontics, University of Texas Health Science Center at San Antonio, 78284, USA
| | | | | | | |
Collapse
|
28
|
Ishikawa I, Nakashima K, Koseki T, Nagasawa T, Watanabe H, Arakawa S, Nitta H, Nishihara T. Induction of the immune response to periodontopathic bacteria and its role in the pathogenesis of periodontitis. Periodontol 2000 1997; 14:79-111. [PMID: 9567967 DOI: 10.1111/j.1600-0757.1997.tb00193.x] [Citation(s) in RCA: 82] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- I Ishikawa
- Department of Periodontology, Faculty of Dentistry, Tokyo Medical and Dental University, Japan
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Kawata Y, Iwasaka H, Kitano S, Hanazawa S. Porphyromonas gingivalis fimbria-stimulated bone resorption is inhibited through binding of the fimbriae to fibronectin. Infect Immun 1997; 65:815-7. [PMID: 9009349 PMCID: PMC176132 DOI: 10.1128/iai.65.2.815-817.1997] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Our most recent study demonstrated that fibronectin is one of the Porphyromonas gingivalis fimbria-binding proteins. In this present study, we demonstrate with mouse embryonic calvarial cells that P. gingivalis fimbria-stimulated bone resorption is inhibited by human fibronectin. The fibronectin inhibition was dose and culture time dependent and was completely neutralized by antifibronectin antibody. The inhibitory action of fibronectin depended on fimbrial interaction with the heparin-binding and cell-attachment domains in the fibronectin structure.
Collapse
Affiliation(s)
- Y Kawata
- Department of Oral Microbiology, Meikai University School of Dentistry, Keyakidai, Sakado City, Saitama, Japan
| | | | | | | |
Collapse
|
30
|
Affiliation(s)
- J J Zambon
- Department of Periodontology, State University of New York, School of Dental Medicine, Buffalo, USA
| |
Collapse
|
31
|
Sharma A, Nagata H, Hamada N, Sojar HT, Hruby DE, Kuramitsu HK, Genco RJ. Expression of functional Porphyromonas gingivalis fimbrillin polypeptide domains on the surface of Streptococcus gordonii. Appl Environ Microbiol 1996; 62:3933-8. [PMID: 8899979 PMCID: PMC168210 DOI: 10.1128/aem.62.11.3933-3938.1996] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Genetically engineering bacteria to express surface proteins which can antagonize the colonization of other microorganisms is a promising strategy for altering bacterial environments. The fimbriae of Porphyromonas gingivalis play an important role in the pathogenesis of periodontal diseases. A structural subunit of the P. gingivalis fimbriae, fimbrillin, has been shown to be an important virulence factor, which likely promotes adherence of the bacterium to saliva-coated oral surfaces and induces host responses. Immunization of gnotobiotic rats with synthetic peptides based on the predicted amino acid sequence of fimbrillin has also been shown to elicit a specific immune response and protection against P. gingivalis-associated periodontal destruction. In this study we engineered the human oral commensal organism Streptococcus gordonii to surface express subdomains of the fimbrillin polypeptide fused to the anchor region of streptococcal M6 protein. The resulting recombinant S. gordonii strains expressing P. gingivalis fimbrillin bound saliva-coated hydroxyapatite in a concentration-dependent manner and inhibited binding of P. gingivalis to saliva-coated hydroxyapatite. Moreover, the recombinant S. gordonii strains were capable of eliciting a P. gingivalis fimbrillin-specific immune response in rabbits. These results show that functional and immunologically reactive P. gingivalis fimbrillin polypeptides can be expressed on the surface of S. gordonii. The recombinant fimbrillin-expressing S. gordonii strains may provide an effective vaccine or a vehicle for replacement therapy against P. gingivalis. These experiments demonstrated the feasibility of expressing biologically active agents (antigens or adhesin molecules) by genetically engineered streptococci. Such genetically engineered organisms can be utilized to modulate the microenvironment of the oral cavity.
Collapse
Affiliation(s)
- A Sharma
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo 14214, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Affiliation(s)
- S Offenbacher
- Dental Research Center, University of North Carolina, Chapel Hill, USA
| |
Collapse
|
33
|
Saito A, Sojar HT, Genco RJ. Porphyromonas gingivalis surface components induce interleukin-1 release and tyrosine phosphorylation in macrophages. FEMS IMMUNOLOGY AND MEDICAL MICROBIOLOGY 1996; 15:51-7. [PMID: 8871116 DOI: 10.1111/j.1574-695x.1996.tb00358.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The aim of the present study was to characterize the responses of macrophages to surface antigens of Porphyromonas gingivalis. Native fimbriae, full-length recombinant fimbrillin, and a lectin-like 12-kDa antigen all stimulated BALB/c peritoneal macrophages to secrete interleukin (IL)-1 beta. The antigens induced similar patterns of tyrosine phosphorylation; proteins in approximately 35-46 kDa range of undetermined identities were phosphorylated in the macrophages. The abilities of the surface antigens to induce IL-1 beta were markedly attenuated by tyrosine kinase inhibitors. This inhibition correlated with inhibition of the induced phosphorylation of specific macrophage proteins at tyrosine. The data suggest that tyrosine kinase(s) plays an important role in the regulatory intracellular signaling mechanisms by which P. gingivalis surface antigens can mediate certain responses in macrophages.
Collapse
Affiliation(s)
- A Saito
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo 14214-3092, USA
| | | | | |
Collapse
|
34
|
Nair SP, Meghji S, Wilson M, Reddi K, White P, Henderson B. Bacterially induced bone destruction: mechanisms and misconceptions. Infect Immun 1996; 64:2371-80. [PMID: 8698454 PMCID: PMC174085 DOI: 10.1128/iai.64.7.2371-2380.1996] [Citation(s) in RCA: 390] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Normal bone remodelling requires the coordinated regulation of the genesis and activity of osteoblast and osteoclast lineages. Any interference with these integrated cellular systems can result in dysregulation of remodelling with the consequent loss of bone matrix. Bacteria are important causes of bone pathology in common conditions such as periodontitis, dental cysts, bacterial arthritis, and osteomyelitis. It is now established that many of the bacteria implicated in bone diseases contain or produce molecules with potent effects on bone cells. Some of these molecules, such as components of the gram-positive cell walls (lipoteichoic acids), are weak stimulators of bone resorption in vitro, while others (PMT, cpn60) are as active as the most active mammalian osteolytic factors such as cytokines like IL-1 and TNF. The complexity of the integration of bone cell lineage development means that there are still question marks over the mechanism of action of many well-known bone-modulatory molecules such as parathyroid hormone. The key questions which must be asked of the now-recognized bacterial bone-modulatory molecules are as follows: (i) what cell population do they bind to, (ii) what is the nature of the receptor and postreceptor events, and (iii) is their action direct or dependent on the induction of secondary extracellular bone-modulating factors such as cytokines, eicosanoids, etc. In the case of LPS, this ubiquitous gram-negative polymer probably binds to osteoblasts or other cells in bone through the CD14 receptor and stimulates them to release cytokines and eicosanoids which then induce the recruitment and activation of osteoclasts. This explains the inhibitor effects of nonsteroidal and anticytokine agents on LPS-induced bone resorption. However, other bacterial factors such as the potent toxin PMT may act by blocking the normal maturation pathway of the osteoblast lineage, thus inducing dysregulation in the tightly regulated process of resorption and replacement of bone matrix. At the present time, it is not possible to define a general mechanism by which bacteria promote loss of bone matrix. Many bacteria are capable of stimulating bone matrix loss, and the information available would suggest that each organism possesses different factors which interact with bone in different ways. With the rapid increase in antibiotic resistance, particularly with Staphylococcus aureus and M. tuberculosis, organisms responsible for much bone pathology in developed countries only two generations ago, we would urge that much greater attention should be focused on the problem of bacterially induced bone remodelling in order to define pathogenetic mechanisms which could be therapeutic targets for the development of new treatment modalities.
Collapse
Affiliation(s)
- S P Nair
- Maxillofacial Surgery Research Unit, Eastman Dental Insitute, University College London, United Kingdom
| | | | | | | | | | | |
Collapse
|
35
|
Ebersole JL, Kesavalu L, Schneider SL, Machen RL, Holt SC. Comparative virulence of periodontopathogens in a mouse abscess model. Oral Dis 1995; 1:115-28. [PMID: 8705817 DOI: 10.1111/j.1601-0825.1995.tb00174.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
OBJECTIVE(S) This report compares the virulence of selected strains of P. gingivalis, A. actinomycetemcomitans, C. rectus, F. nucleatum and T. denticola in a murine model as a measure of pathogenic potential of these oral microorganisms. The characteristics of the tissue destruction associated with these monoinfections were then related to a potential model for bacterial synergism in progressing periodontitis. DESIGN AND METHODS All bacterial strains were grown to mid-logarithmic to early stationary growth phase, harvested and used at various doses to challenge BALB/c normal and BALB/c dexamethasone (DEX) treated mice to mimic a neutrophil dysfunction. The characteristics of tissue destruction, and overt tissue destructive capacity of these species were examined as a function of challenge dose and time. OUTCOME MEASURES The mice were examined for an interval of approximately 15 days post-challenge and the presence/absence of lesions, localized or generalized nature of the lesion (including size in mm2), and lethality of the infection were assessed. RESULTS Comparison of the virulence of the various P. gingivalis strains related to lethality and lesion size associated with destruction of the connective tissue, indicated a virulence capacity of P. gingivalis strains 53977>W50 = T22>3079>33277>381. C. rectus elicited localized necrotic lesions which were limited to the epithelial layers of the skin. The size of the lesions also indicated a graded difference in virulence, such that C. rectus strains 234>576>>33238. A. actinomycetemcomitans caused the formation of classic localized abscesses with a PMN infiltrate and inflammatory exudates. Although each of the A. actinomycetemcomitans strains exhibited a similar virulence pattern in this murine model, A. actinomycetemcomitans serotype b representative strains were potentially more pathogenic with a virulence capacity of 3113D-N = 3975A>JP2 > or = Y4>29523>33384. Both C. rectus and A. actinomycetemcomitans strains showed clear evidence that recent clinical isolates were more virulent than laboratory strains. Challenge with F. nucleatum resulted in tissue destructive responses which were different from those observed with the other strains used in this study. A rapid onset of dose-dependent lesion development, related to the formation of either closed abscesses or open lesions, was observed with F. nucleatum. Tissue involvement was also greater at lower F. nucleatum doses when compared to the other bacteria. F. nucleatum challenge of DEX-treated mice resulted in a shift to open lesions. T. denticola appeared to be more tissue invasive than the other species examined in this study. Challenge of mice with T. denticola resulted in involvement of multiple tissues, including epithelial and connective tissues, as well as appearing to invade muscle layers and deeper tissues. In addition to invading deeper tissues, the resulting lesions took considerably longer to resolve. In the DEX-treated mice (neutrophil depleted), P. gingivalis, C. rectus, and A. actinomycetemcomitans were significantly more virulent. In contrast, while DEX treatment altered the characteristics of lesions caused by F. nucleatum, the extent of lesions produced by F. nucleatum and T. denticola was not substantially enhanced. CONCLUSIONS The results obtained from this study suggest that different microorganisms have the ability to provide individual pathologies which may act in an additive/synergistic fashion contributing to the tissue destruction noted in periodontitis.
Collapse
Affiliation(s)
- J L Ebersole
- Department of Periodontics, University of Texas Health Science Center at San Antonio, USA
| | | | | | | | | |
Collapse
|
36
|
Hanazawa S, Kawata Y, Murakami Y, Naganuma K, Amano S, Miyata Y, Kitano S. Porphyromonas gingivalis fimbria-stimulated bone resorption in vitro is inhibited by a tyrosine kinase inhibitor. Infect Immun 1995; 63:2374-7. [PMID: 7768625 PMCID: PMC173315 DOI: 10.1128/iai.63.6.2374-2377.1995] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Our previous study (Y. Kawata, S. Hanazawa, S. Amano, Y. Murakami, T. Matsumoto, K. Nishida, and S. Kitano, Infect. Immun. 62:3012-3016, 1994) showed that Porphyromonas gingivalis fimbriae stimulate bone resorption in vitro. Since it has recently been demonstrated that tyrosine kinase encoded by the c-src gene plays an important role in osteoclastic bone resorption, in the present study we examined the effect of a tyrosine kinase inhibitor on the fimbria-stimulated bone resorption. Genistein, a potent inhibitor of tyrosine kinase, markedly inhibited bone resorption stimulated by the fimbriae. Genistein also inhibited induction of several tyrosine-phosphorylated proteins in the fimbria-treated calvarial bone cells from mouse embryos.
Collapse
Affiliation(s)
- S Hanazawa
- Department of Oral Microbiology, Meikai University School of Dentistry, Sakado City, Japan
| | | | | | | | | | | | | |
Collapse
|
37
|
Kawata Y, Hanazawa S, Amano S, Murakami Y, Matsumoto T, Nishida K, Kitano S. Porphyromonas gingivalis fimbriae stimulate bone resorption in vitro. Infect Immun 1994; 62:3012-6. [PMID: 7911789 PMCID: PMC302914 DOI: 10.1128/iai.62.7.3012-3016.1994] [Citation(s) in RCA: 53] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Our previous study demonstrated that Porphyromonas gingivalis fimbriae induce the expression of interleukin-1, a potent bone-resorbing cytokine, in macrophages. This demonstration suggested to use the possibility that the fimbriae may stimulate bone resorption via the generation of an inflammatory cytokine(s). The present study was performed to test this suggestion. The bone-resorbing activity was evaluated by measuring the area of resorption lacunae on bone slices incubated with calvarial bone cells taken from 14-day-old mouse embryos. Fimbriae at 0.5 micrograms of protein per ml stimulated the bone-resorbing activity significantly, and the effect was dose and treatment time dependent. Since it is well known that interleukin-1 and granulocyte macrophage colony-stimulating factor induce differentiation of osteoclast lineage cells, we examined the involvement of these cytokines in fimbria-stimulated bone resorption. Fimbria-stimulated bone resorption was abolished significantly by antisera against both cytokines. We observed by Northern (RNA) blot assay that both cytokine genes were markedly expressed in the fimbria-treated calvarial bone cells. Our present data demonstrate that P. gingivalis fimbriae stimulate bone resorption in vitro.
Collapse
Affiliation(s)
- Y Kawata
- Department of Oral Microbiology, Meikai University School of Dentistry, Saitama, Japan
| | | | | | | | | | | | | |
Collapse
|
38
|
Affiliation(s)
- A D Haffajee
- Department of Periodontology, Forsyth Dental Center, Boston, Massachusetts, USA
| | | |
Collapse
|
39
|
Affiliation(s)
- J L Ebersole
- Department of Periodontics, University of Texas Health Science Center at San Antonio, USA
| | | |
Collapse
|
40
|
Affiliation(s)
- S S Socransky
- Department of Periodontology, Forsyth Dental Center, Boston, Massachusetts, USA
| | | |
Collapse
|