Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
J Periodontal Res ; 43(2): 246-54, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18221294

ABSTRACT

BACKGROUND AND OBJECTIVE: Endothelial cells have a substantial role in maintaining vascular homeostasis, and their dysregulation can contribute to the development of pathology. The plasminogen activators and their inhibitors may, arguably, be the single most important proteolytic system of the endothelium for vascular maintenance by controlling plasminogen activation and other proteolytic cascades that impact on clotting, hemodynamics, angiogenesis and the character of the vascular wall. In chronic periodontal disease, significant changes to the microvasculature occur in association with the severity of the disease. Investigation of the role played by endothelial cells in periodontal health and disease has been limited to in situ immunolocalization or to the use of endothelial cells of nongingival origin, such as human umbilical vein endothelial cells. The objective of this research was to establish a replicable protocol for isolating microvascular endothelial cells from the gingiva. MATERIAL AND METHODS: From inflamed gingiva, isolated cells were characterized by morphology, the expression of factor VIII-related antigen, the expression of UEA-1 ligand, the uptake of acetylated low-density lipoprotein, network formation on Matrigel, and by the expression levels of urokinase plasminogen activator, tissue plasminogen activator, plasminogen activator inhibitor-1 and collagen IV. RESULTS AND CONCLUSION: Gingival endothelial cells were most readily obtained from inflamed gingival tissues, and these endothelial cells, when isolated by the protocol established herein, demonstrated endothelial characteristics and constitutively secreted plasminogen activators and plasminogen activator inhibitor-1 in culture.


Subject(s)
Cell Culture Techniques , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Gingiva/blood supply , Microcirculation/metabolism , Cell Separation , Cells, Cultured , Collagen , Collagen Type IV/biosynthesis , Drug Combinations , Endothelial Cells/cytology , Endothelial Cells/metabolism , Fluorescent Antibody Technique , Gingivitis/pathology , Humans , Laminin , Lipoproteins, LDL/metabolism , Microcirculation/cytology , Plasminogen Activator Inhibitor 1/biosynthesis , Platelet Endothelial Cell Adhesion Molecule-1 , Proteoglycans , Tissue Plasminogen Activator/biosynthesis , Tissue Scaffolds , Urokinase-Type Plasminogen Activator/biosynthesis , von Willebrand Factor/isolation & purification
2.
Clin Exp Immunol ; 150(2): 217-29, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17937677

ABSTRACT

Cysteine proteinases, termed gingipains, of Porphyromonas gingivalis are able to inactivate a broad range of host proteins involved in cellular responses and have been implicated as key virulence factors in the onset and progression of adult periodontitis. In the present study, the high molecular weight Arg-gingipain, RgpA, produced a time- and concentration-dependent hydrolysis of the tumour necrosis factor (TNF)-alpha receptor family member CD27 on resting T cells. As a consequence of CD27 degradation, a reduction in CD27-ligation dependent co-stimulatory CD40L expression was observed. Concomitantly, RgpA activated the protease-activated receptors (PAR)-1, PAR-2 and PAR-4 and induced CD69 and CD25 expression on T cells, thereby demonstrating T cell activation. The Lys-gingipain Kgp demonstrated a low capacity to degrade CD27 but the ability to affect CD27 expression and biological activity was increased when T cells were pretreated with blocking peptide against PAR-2. CD70, the ligand for CD27 induced on activated B cells, was significantly reduced by RgpA treatment and weakly affected by Kgp. These findings suggest that while RgpA can activate T cells through PARs, the parallel action of direct hydrolysis of membrane CD27 as well as CD70 indicates a potential down-regulatory effect through inhibition of CD27/CD70-mediated cell activation in periodontitis.


Subject(s)
Adhesins, Bacterial/pharmacology , CD4-Positive T-Lymphocytes/immunology , Cysteine Endopeptidases/pharmacology , Porphyromonas gingivalis/immunology , Receptors, Proteinase-Activated/antagonists & inhibitors , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , CD27 Ligand/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD40 Ligand/metabolism , Cell Cycle/drug effects , Cells, Cultured , Dose-Response Relationship, Immunologic , Extracellular Fluid/immunology , Formaldehyde/pharmacology , Gingipain Cysteine Endopeptidases , Humans , Interleukin-2 Receptor alpha Subunit/metabolism , Lectins, C-Type , Lymphocyte Activation/immunology , Polymers/pharmacology , Receptors, Proteinase-Activated/metabolism , Serum , Solubility
3.
J Periodontal Res ; 42(4): 305-10, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17559626

ABSTRACT

BACKGROUND AND OBJECTIVE: The plasminogen activating system is a protease/inhibitor system central to extracellular matrix remodeling with a suggested role in periodontal disease pathology. A few studies have reported polymorphisms in the genes of plasminogen activator inhibitors to be associated with periodontal disease severity. Two gene polymorphisms - a BamHI restriction fragment length polymorphism in the urokinase plasminogen activator gene (uPA) and a HindIII restriction fragment length polymorphism in the plasminogen activator inhibitor type 1 gene (PAI-1) - have been associated with conditions having a vascular component, and our objective was to assess the association of these gene polymorphisms with alveolar bone loss in chronic periodontal disease of adults. MATERIAL AND METHODS: Genotype was determined by polymerase chain reaction amplification of whole blood, pertinent histories were obtained by interview, and alveolar bone loss was assessed from current radiographs. RESULTS: In 77 elderly patients with a normal distribution of alveolar bone loss, we demonstrated a significant association between levels of alveolar bone loss and these polymorphisms in the uPA and PAI-1 genes. Controlling for the contributions of smoking or diabetes to periodontal bone loss, estimated odds ratios for predicting lower levels of alveolar bone loss, associated with a greater degree of periodontal health, were strongest when defined by the concurrent presence of a homozygous urokinase plasminogen activator genotype and the nuclease-sensitive plasminogen activator inhibitor type 1 (HindIII) allele (odds ratio = 2.6; 95% confidence interval: 5.8-1.3). CONCLUSION: The urokinase plasminogen activator (BamHI) and plasminogen activator inhibitor type 1 (HindIII) genotypes may serve as useful markers for heritability of bone loss associated with periodontal disease.


Subject(s)
Alveolar Bone Loss/genetics , Plasminogen Activator Inhibitor 1/genetics , Polymorphism, Genetic , Urokinase-Type Plasminogen Activator/genetics , Aged , Aged, 80 and over , Alleles , Alveolar Bone Loss/diagnostic imaging , Chronic Disease , Deoxyribonuclease BamHI/analysis , Diabetes Mellitus, Type 2/complications , Epidemiologic Methods , Female , Humans , Male , Middle Aged , Plasminogen Activator Inhibitor 1/analysis , Radiography , Site-Specific DNA-Methyltransferase (Adenine-Specific)/analysis , Smoking/adverse effects , Urokinase-Type Plasminogen Activator/analysis
4.
J Dent Res ; 85(2): 122-32, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16434729

ABSTRACT

Tissue engineering, grafting procedures, regeneration, and tissue remodeling are developing therapeutic modalities with great potential medical value, but these regenerative modalities are not as effective or predictable as clinicians and patients would like. Greater understanding of growth factors, cytokines, extracellular matrix molecules, and their roles in cell-mediated healing processes have made these regenerative therapies more clinically viable and will continue advancing the fields of tissue engineering and grafting. However, millions of oral and non-oral bone-grafting procedures are performed annually, and only a small percentage yield the most desirable results. Here we review the heparan-sulfate-decorated extracellular biomolecule named perlecan and the research relating to its potential as an adjunct in bone-regenerative procedures. The review includes an overview of bone graft substitutes and biological adjuncts to bone-regenerative procedures in medicine as they apply to periodontal disease, alveolar ridge augmentation, and barrier membrane therapy. Perlecan is discussed as a potential biological adjunct in terms of growth factor sequestration and delivery, and promoting cell adhesion, proliferation, differentiation, and angiogenesis. Further, we propose delivery and application schemes for perlecan and/or its domains in bone-regenerative procedures, with particular emphasis on its heparan-sulfate-decorated domain I. The perlecan molecule, with its heparan sulfate glycosylation, may provide a multi-faceted approach for the delivery of a more comprehensive stimulus than other single potential adjuncts currently available for bone-regenerative procedures.


Subject(s)
Alveolar Bone Loss/surgery , Bone Regeneration/drug effects , Heparan Sulfate Proteoglycans/pharmacology , Animals , Bone Substitutes , Cell Adhesion/drug effects , Cell Adhesion/physiology , Growth Substances/physiology , Guided Tissue Regeneration, Periodontal , Heparan Sulfate Proteoglycans/administration & dosage , Heparan Sulfate Proteoglycans/chemistry , Heparan Sulfate Proteoglycans/physiology , Humans , Protein Structure, Tertiary
5.
Clin Exp Immunol ; 139(2): 328-37, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15654832

ABSTRACT

We previously reported evidence that patients with periodontitis have serum antibodies to oral Gram positive bacteria that are cross-reactive with epithelial antigens. In the present report cross-reactive epithelial antigens including CD24, lactate dehydrogenase A [LDM-A], antioxidant protein 2 [AOP 2] and nuclear factor of activated T cells 5 [NFAT 5], were identified by screening a cDNA expression library with pooled patient sera. Titres of antibodies to CD24 peptide correlated negatively with indices of periodontal disease severity. Strong expression of CD24 in the reactive periodontal epithelium and inflamed gingival attachment contrasted with low to undetectable expression in the external gingival epithelium. In periodontitis, a local action of these auto-reactive antibodies could modulate the regulatory potential associated with expression of CD24 in this epithelium.


Subject(s)
Antigens, Bacterial/immunology , Antigens, CD/immunology , Autoantigens/immunology , Membrane Glycoproteins/immunology , Mouth Mucosa/immunology , Periodontitis/immunology , CD24 Antigen , Cross Reactions , Humans , Immunohistochemistry/methods , Isoenzymes/immunology , L-Lactate Dehydrogenase/immunology , Lactate Dehydrogenase 5 , Periodontitis/microbiology , Peroxidases/immunology , Peroxiredoxin VI , Peroxiredoxins , Polymerase Chain Reaction , Sequence Analysis, DNA , Trans-Activators/immunology , Transcription Factors
6.
Clin Exp Immunol ; 134(2): 295-302, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14616790

ABSTRACT

Porphyromonas gingivalis cysteine proteinases (gingipains) have been associated with virulence in destructive periodontitis, a disease process that has been linked with Th2 pathways. Critical in maintaining Th2 activity is the response of B lymphocytes to environmental interleukin (IL)-4, a cytokine that also counteracts Th1-cell differentiation. Here we demonstrate that while the gingipains effectively degrade interleukin (IL)-4 under serum-free conditions, limited hydrolysis was observed in the presence of serum even after prolonged incubation. Gingipains up-regulated CD69 expression directly in purified peripheral blood B cell preparations. Further, the induction of IL-4 receptor expression on B cells by gingipains correlates with B cell activation, which is also manifested by a mitogenic response. These results suggest that the gingipains of P. gingivalis act during the early stage of B-cell growth as a competence signal, whereby sensitized B cells might become more responsive to further challenge in the disease-susceptible individual.


Subject(s)
B-Lymphocytes/immunology , Cysteine Endopeptidases/immunology , Hemagglutinins/immunology , Lymphocyte Activation/immunology , Porphyromonas gingivalis/immunology , Th2 Cells/immunology , Adhesins, Bacterial/immunology , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Cell Division/immunology , Cells, Cultured , Gingipain Cysteine Endopeptidases , Humans , Hydrolysis , Interleukin-4/metabolism , Lectins, C-Type , Receptors, Interleukin-4/metabolism , Up-Regulation/drug effects
7.
Infect Immun ; 71(1): 562-6, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12496212

ABSTRACT

In a rat periodontitis model, preinoculation with the Porphyromonas gingivalis HA2 binding domain for hemoglobin provided protection from disease. Protection was associated with induced anti-HA2 immunoglobulin G (IgG) humoral antibodies. The IgG subclass ratios suggested that relatively lower Th2/Th1-driven responses were directly associated with protection when rHA2 was administered in saline.


Subject(s)
Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/immunology , Periodontitis/prevention & control , Porphyromonas gingivalis/immunology , Animals , Antibodies, Bacterial/blood , Bacterial Proteins/administration & dosage , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/immunology , Bacteroidaceae Infections/prevention & control , Cysteine Endopeptidases/administration & dosage , Cysteine Endopeptidases/genetics , Disease Models, Animal , Hemoglobins/metabolism , Immunization , Immunoglobulin G/blood , Immunoglobulin G/classification , Rats , Rats, Inbred F344 , Recombinant Proteins/immunology
8.
Infect Immun ; 69(9): 5650-60, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11500441

ABSTRACT

Porphyromonas gingivalis cysteine proteinases (gingipains) have been associated with virulence in destructive periodontitis, a disease process variously considered to represent an unregulated stimulation of either T helper type 1 (Th1)- or Th2-type cells. Critical in maintaining Th1 activity is the response of T lymphocytes to environmental interleukin 12 (IL-12) in the form of up-regulation of gamma interferon (IFN-gamma) production. Here we demonstrate that in the presence or absence of serum, gingipains were able to hydrolyze IL-12 and reduce the IL-12-induced IFN-gamma production from CD4+ T cells. However, the induction of IL-12 receptors on T cells by gingipains did not correlate with the enhancement of IFN-gamma production. The gingipains cleaved IL-12 within the COOH-terminal region of the p40 and p35 subunit chains, which leads to IL-12 inactivity, whereas IL-2 in these assays was not affected. Inactivation of IL-12 by the gingipains could disrupt the cytokine balance or favor Th2 activities in the progression of periodontitis.


Subject(s)
Cysteine Endopeptidases/metabolism , Hemagglutinins/metabolism , Interleukin-12/metabolism , Periodontitis/immunology , Porphyromonas gingivalis/enzymology , Porphyromonas gingivalis/pathogenicity , Adhesins, Bacterial , Cells, Cultured , Gingipain Cysteine Endopeptidases , Humans , Hydrolysis , Interferon-gamma/metabolism , Lymphocyte Activation , Periodontitis/microbiology , Th1 Cells/immunology , Th2 Cells/immunology
9.
J Dent Res ; 78(7): 1354-61, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10403463

ABSTRACT

Previous studies have shown a correlation between the production of certain matrix metalloproteinases (MMPs), especially the gelatinases, by malignant tumors and the progression of these cancers as they invade and metastasize through the extracellular matrix and basement membranes. However, very few of these studies examined this relationship in human oral cancer in vivo, and none addressed the issue of how combinations of the MMPs may further enhance tumor progression. To determine which MMPs are produced in vivo by human oral cancers, we used specific anti-human-MMP antibodies and immunocytochemistry (ICC) methods to examine oral cancer tissue specimens from 20 surgery patients. The ICC data indicated that 72-kDa (72K-GL) and 92-kDa gelatinases (92K-GL) were produced in vivo by discreet clusters of tumor cells and by stromal fibroblasts, vascular endothelial cells (72K-GL), and PMNs (92K-GL). Some stromal fibroblasts near the tumors also appeared to produce fibroblast-type collagenase (FIB-CL), a finding confirmed by Western blot analysis of media conditioned by oral tumor explant cultures. ICC results indicated that 5 of the 20 tumors coincidentally produced all three MMPs. To examine how the two gelatinases and FIB-CL may interact in vitro to degrade fibrillar type I collagen, a major structural component of the extracellular matrix, we used a modified FIB-CL activity assay. Combinations of the gelatinases and FIB-CL were incubated with a 3H-collagen substrate, with the results compared with the combination of stromelysin-1 (SL-1, a superactivator of FIB-CL) and FIB-CL. 92K-GL caused a nine-fold increase in collagenase activity, equivalent to SL-1, while 72K-GL produced a four-fold increase. These results indicate that human oral cancers produce 92K-GL, 72K-GL, and FIB-CL in vivo and that the gelatinases and FIB-CL cooperate to enhance collagen degradation greatly in vitro.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Collagenases/biosynthesis , Collagenases/metabolism , Gelatinases/biosynthesis , Metalloendopeptidases/biosynthesis , Mouth Neoplasms/enzymology , Antibodies , Basement Membrane/pathology , Blotting, Western , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/secondary , Collagen/metabolism , Disease Progression , Endothelium, Vascular/enzymology , Endothelium, Vascular/pathology , Extracellular Matrix/ultrastructure , Fibroblasts/enzymology , Fibroblasts/pathology , Gelatinases/metabolism , Humans , Immunohistochemistry , Matrix Metalloproteinase 1 , Matrix Metalloproteinase 2 , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase 9 , Metalloendopeptidases/metabolism , Neoplasm Invasiveness , Neutrophils/enzymology , Neutrophils/pathology , Radiopharmaceuticals , Tritium
10.
J Bacteriol ; 181(12): 3784-91, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10368154

ABSTRACT

Heme binding and uptake are considered fundamental to the growth and virulence of the gram-negative periodontal pathogen Porphyromonas gingivalis. We therefore examined the potential role of the dominant P. gingivalis cysteine proteinases (gingipains) in the acquisition of heme from the environment. A recombinant hemoglobin-binding domain that is conserved between two predominant gingipains (domain HA2) demonstrated tight binding to hemin (Kd = 16 nM), and binding was inhibited by iron-free protoporphyrin IX (Ki = 2.5 microM). Hemoglobin binding to the gingipains and the recombinant HA2 (rHA2) domain (Kd = 2.1 nM) was also inhibited by protoporphyrin IX (Ki = 10 microM), demonstrating an essential interaction between the HA2 domain and the heme moiety in hemoglobin binding. Binding of rHA2 with either hemin, protoporphyrin IX, or hematoporphyrin was abolished by establishing covalent linkage of the protoporphyrin propionic acid side chains to fixed amines, demonstrating specific and directed binding of rHA2 to these protoporphyrins. A monoclonal antibody which recognizes a peptide epitope within the HA2 domain was employed to demonstrate that HA2-associated hemoglobin-binding activity was expressed and released by P. gingivalis cells in a batch culture, in parallel with proteinase activity. Cysteine proteinases from P. gingivalis appear to be multidomain proteins with functions for hemagglutination, erythrocyte lysis, proteolysis, and heme binding, as demonstrated here. Detailed understanding of the biochemical pathways for heme acquisition in P. gingivalis may allow precise targeting of this critical metabolic aspect for periodontal disease prevention.


Subject(s)
Cysteine Endopeptidases/metabolism , Hemagglutinins/metabolism , Hemoglobins/metabolism , Porphyrins/metabolism , Porphyromonas gingivalis/physiology , Adhesins, Bacterial , Amino Acid Sequence , Binding Sites , Cysteine Endopeptidases/chemistry , Gingipain Cysteine Endopeptidases , Hemagglutinins/chemistry , Hematoporphyrins/metabolism , Hemin/metabolism , Kinetics , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Porphyromonas gingivalis/enzymology , Protoporphyrins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
11.
Infect Immun ; 67(6): 2986-95, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10338509

ABSTRACT

Cysteine proteinases have been emphasized in the virulence of Porphyromonas gingivalis in chronic periodontitis. These hydrolases may promote the degradation of extracellular matrix proteins and disrupt components of the immune system. In this study it was shown that purified Arg-gingipain and Lys-gingipain inhibited expression of class II major histocompatibility complex (MHC) proteins in response to the stimulation of endothelial cells with human gamma interferon (IFN-gamma). Treatment with the cysteine proteinases resulted in a rapid shift in the apparent molecular size of IFN-gamma from 17 to 15 kDa, as shown by Western blot analysis, a response which also occurred in the presence of serum. Further, glycosylated natural IFN-gamma from human leukocytes and unglycosylated recombinant IFN-gamma from Escherichia coli were both digested by the cysteine proteinases. Immunoblot analysis indicated that cleavage within the carboxyl terminus of recombinant IFN-gamma correlated with the loss of induction of MHC class II expression as monitored by analytical flow cytometry. No hydrolysis of MHC class II molecules or human IFN-gamma receptor by these proteinases was detected by Western blot analysis. These findings suggest that P. gingivalis cysteine proteinases may alter the cytokine network at the point of infection through the cleavage of IFN-gamma. Degradation of IFN-gamma could have important consequences for the recruitment and activation of leukocytes and therefore may contribute significantly to the destruction of the periodontal attachment.


Subject(s)
Adhesins, Bacterial/metabolism , Cysteine Endopeptidases/metabolism , HLA-DR Antigens/biosynthesis , Hemagglutinins/metabolism , Interferon-gamma/metabolism , Porphyromonas gingivalis/immunology , Adhesins, Bacterial/pharmacology , Cells, Cultured , Cysteine Endopeptidases/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Endothelium, Vascular/cytology , Epitopes, B-Lymphocyte/immunology , Gingipain Cysteine Endopeptidases , Hemagglutinins/pharmacology , Humans , Interferon-gamma/pharmacology , Leupeptins/pharmacology , Porphyromonas gingivalis/enzymology , Proteins/metabolism , Proteins/pharmacology , Recombinant Proteins
12.
J Periodontal Res ; 33(7): 408-20, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9842506

ABSTRACT

Periodontitis is characterized by advancement of a narrow band of epithelium (1-10 cells wide) through the collagenous periodontal ligament in response to bacterial accumulation and infection. A modulating role by epithelial cells in the progression of periodontitis was hypothesized due to the close proximity of the advancing epithelium to both the etiological bacteria and to the collagen fibers of the ligament. We demonstrate that rat mucosal epithelial cells and human fibroblasts are similarly stimulated to degrade a collagen type I cellular substrate by thiol-dependent activity released by the major periodontal pathogen Porphyromonas gingivalis. A purified, extracellular bacterial thiol-proteinase from P. gingivalis ATCC 33277 stimulated mucosal epithelial cells to upregulate expression of collagenase and stromelysin, and to degrade a collagen type I fibril matrix. Stimulation of the epithelial cells with this purified proteinase was associated with morphological changes in the cells and with accumulation of secreted latent procollagenase throughout the culture medium. Release of active collagenase was minimal and collagen degradation by the epithelial cells was discreet and localized subcellularly suggesting the possibility that activation of secreted procollagenase was cell-associated. We conclude that a collagen-degrading phenotype can be stimulated in relatively quiescent mucosal epithelial cells and fibroblasts by the presence of bacterial proteinase. These experiments suggest roles for the P. gingivalis thiol-proteinase and the epithelial cell in the pathogenesis of periodontal disease and demonstrate the potential for dysregulation of extracellular matrix remodeling events during healing of other bacterially infected wounds.


Subject(s)
Cysteine Endopeptidases/metabolism , Metalloendopeptidases/biosynthesis , Periodontal Ligament/metabolism , Periodontitis/enzymology , Porphyromonas gingivalis/enzymology , Adhesins, Bacterial , Animals , Bacterial Proteins/metabolism , Base Sequence , Cells, Cultured , Collagen/metabolism , Culture Media, Conditioned , Epithelial Cells/metabolism , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Gingipain Cysteine Endopeptidases , Hemagglutinins/metabolism , Humans , Molecular Sequence Data , Mouth Mucosa/metabolism , Periodontal Ligament/cytology , Periodontitis/microbiology , Porphyromonas gingivalis/pathogenicity , RNA, Messenger/analysis , Rats , Up-Regulation
13.
J Dent Res ; 76(6): 1260-70, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9168859

ABSTRACT

A critical outcome of periodontal disease is degradation of the collagenous periodontal ligament that connects teeth to bone in the dental arch. Periodontal diseases occur in response to bacterial colonization of the teeth, but their molecular pathogenesis is still speculative. One family of enzymes, known as the matrix metalloproteinases (MMPs), has been implicated in the degradation of the periodontal ligament. MMPs, which are also suspected to play a role in many other physiologic and pathologic remodeling processes, can be secreted by epithelial cells surrounding the teeth and are found in relative abundance in tissues and fluids near periodontally diseased sites. Since most MMPs are secreted as inactive zymogens which may be activated by limited proteolysis, it has been suggested that proteinases expressed by the infecting periodontal pathogens might activate latent host MMPs to initiate or accelerate degradation of the collegenous periodontal ligament. The aim of this work was to examine interactions between purified host MMPs and bacterial proteinase. In this article, we demonstrate that a proteinase isolated from the periodontopathogen Porphyromonas gingivalis can activate MMP-1, MMP-3, and MMP-9 and can catalyze the superactivation of MMP-1 by MMP-3. Activation of these MMPs is demonstrated to result from initial hydrolysis within their propeptide. Also, for MMP-1 and MMP-9, the P. gingivalis proteinase cleaves the MMP propeptide following a lysine residue at a previously unreported site which, for both MMPs, is one residue NH2-terminal to the known autocatalytic cleavage site. These data describe a mode of virulence for the periodontopathogen Porphyromonas gingivalis that involves activation of host-degradative enzymes.


Subject(s)
Bacterial Proteins/metabolism , Cysteine Endopeptidases/metabolism , Extracellular Matrix Proteins/metabolism , Metalloendopeptidases/metabolism , Porphyromonas gingivalis/enzymology , Amino Acid Sequence , Collagenases/metabolism , Culture Media, Conditioned , Enzyme Activation , Glycoproteins/metabolism , Matrix Metalloproteinase 1 , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase 9 , Molecular Sequence Data , Porphyromonas gingivalis/pathogenicity , Protease Inhibitors/metabolism , Tissue Inhibitor of Metalloproteinases , Virulence
14.
Oral Microbiol Immunol ; 12(1): 47-56, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9151644

ABSTRACT

Thiol-dependent proteinases that are expressed and released by Porphyromonas gingivalis are considered virulence factors in periodontitis because of their potential to effect matrix degradation and inflammation. A number of P. gingivalis thiol-proteinases have been described, however, with similar biochemical characteristics. In this report we demonstrate that an isolate P. gingivalis proteinase consists of noncovalently associated peptides and that slight variations in the association pattern of these peptides could result in different proteinases with different affinities and activities. We also describe the co-purification of thiol-proteinase activity with hemagglutinin activity and demonstrate that each type of activity has similar inhibition profiles. With the use of monoclonal antibodies against the P. gingivalis proteinase we follow proteinase released into the culture medium over the course of 10 days and, by Western blot analysis, demonstrate that many of the proteinases with varying molecular weight are related. The identification of a single, immunoreactive, 140 kDa proteinase detected early in the culture and in association with the P. gingivalis cells suggests that multiple proteinase may originate from a single 140 kDa proteinase.


Subject(s)
Bacterial Proteins/chemistry , Cysteine Endopeptidases/chemistry , Hemagglutinins/chemistry , Porphyromonas gingivalis/enzymology , Amino Acid Sequence , Antibodies, Monoclonal , Cysteine Endopeptidases/metabolism , Electrophoresis, Polyacrylamide Gel , Evolution, Molecular , Hemagglutination Inhibition Tests , Hemagglutinins/metabolism , Protease Inhibitors/metabolism , Structure-Activity Relationship , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL