Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Dent Res ; 99(5): 568-576, 2020 05.
Article in English | MEDLINE | ID: mdl-32105543

ABSTRACT

Porphyromonas gingivalis, like other bacteria belonging to the phylum Bacteroidetes, synthesizes sphingolipids (SLs). However, their exact roles in microbial physiology and their potential role in mediating interactions with their eukaryotic host are unclear. Our working hypothesis for this study was that synthesis of SLs (host-like lipids) affords a mechanism that allows P. gingivalis to persist in homeostasis with its host. In a previous study, we deleted a gene (PG1780 in strain W83) predicted to encode a serine palmitoyl transferase (SPT)-the enzyme that catalyzes the first conserved step in the synthesis of SLs-and we determined that the mutant was unable to synthesize SLs. Here, we characterized the SPT enzyme encoded by PG1780, analyzed the impact of SPT deletion on P. gingivalis gene expression (RNA-Seq analysis), and began to define the impact of SL synthesis on its interactions with host cells. Enzymatic analysis verified that the protein encoded by PG1780 is indeed an SPT. RNA-Seq analysis determined that a lack of SL synthesis results in differential expression of extracytoplasmic function sigma factors, components of the type IX secretion system (T9SS), and CRISPR and cas genes. Our data demonstrate that when human THP1 macrophage-like cells were challenged with the wild type (W83) and the SL-null mutant (W83 ΔSPT), the SL-null strain elicited a robust inflammatory response (elevated IL-1ß, IL-6, IL-10, IL-8, RANTES, and TNFα) while the response to the parent strain W83 was negligible. Interestingly, we also discovered that SLs produced by P. gingivalis can be delivered to host cells independent of cell-to-cell contact. Overall, our results support our working hypothesis that synthesis of SLs by P. gingivalis is central to its ability to manipulate the host inflammatory response, and they demonstrate the integral importance of SLs in the physiology of P. gingivalis.


Subject(s)
Porphyromonas gingivalis , Humans , Macrophages , Sphingolipids
2.
Mol Oral Microbiol ; 32(3): 250-261, 2017 06.
Article in English | MEDLINE | ID: mdl-27346827

ABSTRACT

Macrophages adapt both phenotypically and functionally to the cytokine balance in host tissue microenvironments. Recent studies established that macrophages contribute an important yet poorly understood role in the development of infection-elicited oral bone loss. We hypothesized that macrophage adaptation to inflammatory signals encountered before pathogen interaction would significantly influence the subsequent immune response of these cells to the keystone oral pathobiont Porphyromonas gingivalis. Employing classically activated (M1) and alternatively activated (M2) murine bone-marrow-derived macrophage (BMDMø), we observed that immunologic activation of macrophages before P. gingivalis challenge dictated phenotype-specific changes in the expression of inflammation-associated molecules important to sensing and tuning host response to bacterial infection including Toll-like receptors 2 and 4, CD14, CD18 and CD11b (together comprising CR3), major histocompatibility complex class II, CD80, and CD86. M2 cells responded to P. gingivalis with higher expression of tumor necrosis factor-α, interleukin-6, monocyte chemoattractant protein-1, macrophage inflammatory protein-1α, regulated on activation normal T cell expressed and secreted, and KC than M1 cells. M1 BMDMø expressed higher levels of interleukin-10 to P. gingivalis than M2 BMDMø. Functionally, we observed that M2 BMDMø bound P. gingivalis more robustly than M1 BMDMø. These data describe an important contribution of macrophage skewing in the subsequent development of the cellular immune response to P. gingivalis.


Subject(s)
Immunity, Cellular , Macrophage Activation , Macrophages/immunology , Porphyromonas gingivalis/immunology , Animals , B7-1 Antigen/genetics , B7-1 Antigen/immunology , B7-2 Antigen/genetics , B7-2 Antigen/immunology , Bacterial Adhesion , CD11b Antigen/genetics , CD11b Antigen/immunology , CD18 Antigens/genetics , CD18 Antigens/immunology , Chemokines/genetics , Chemokines/immunology , Cytokines/genetics , Cytokines/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Inflammation , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/immunology , Macrophages/physiology , Mice , Periodontal Diseases/immunology , Periodontal Diseases/microbiology , Porphyromonas gingivalis/pathogenicity , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
3.
Mol Oral Microbiol ; 30(6): 438-50, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25946408

ABSTRACT

Periodontal diseases are chronic oral inflammatory diseases that are polymicrobial in nature. The presence of specific bacteria in subgingival plaque such as Porphyromonas gingivalis is associated with microbial dysbiosis and the modulation of host immune response. Bacterially elicited innate immune activation and inflammation are key elements implicated in the destruction of soft and hard tissues supporting the teeth. Liver X receptors (LXRs) are nuclear hormone receptors with important function in lipid homeostasis, inflammation, and host response to infection; however, their contribution to chronic inflammatory diseases such as periodontal disease is not understood. The aim of this study was to define the contribution of LXRs in the development of immune response to P. gingivalis and to assess the roles that LXRs play in infection-elicited oral bone loss. Employing macrophages, we observed that P. gingivalis challenge led to reduced LXRα and LXRß gene expression compared with that observed with unchallenged wild-type cells. Myeloid differentiation primary response gene 88 (MyD88)-independent, Toll/interleukin-1 receptor-domain-containing adapter-inducing interferon-ß (TRIF)-dependent signaling affected P. gingivalis-mediated reduction in LXRα expression, whereas neither pathway influenced the P. gingivalis effect on LXRß expression. Employing LXR agonist and mice deficient in LXRs, we observed functional effects of LXRs in the development of a P. gingivalis-elicited cytokine response at the level of the macrophage, and participation of LXRs in P. gingivalis-elicited oral bone loss. These findings identify novel importance for LXRs in the pathogenesis of P. gingivalis infection-elicited inflammation and oral bone loss.


Subject(s)
Alveolar Bone Loss/immunology , Alveolar Bone Loss/microbiology , Bacteroidaceae Infections/immunology , Bacteroidaceae Infections/metabolism , Macrophages/immunology , Macrophages/microbiology , Orphan Nuclear Receptors/genetics , Porphyromonas gingivalis/immunology , Adaptor Proteins, Vesicular Transport/genetics , Alveolar Bone Loss/physiopathology , Animals , Bacteroidaceae Infections/microbiology , Cells, Cultured , Cytokines/genetics , Immunity, Innate , Inflammation , Interferon Regulatory Factor-3/genetics , Liver X Receptors , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Orphan Nuclear Receptors/agonists , Orphan Nuclear Receptors/metabolism , Periodontal Diseases , Signal Transduction/genetics , Toll-Like Receptor 2/genetics , Tumor Necrosis Factor-alpha/genetics
4.
J Periodontal Res ; 50(1): 89-102, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24862405

ABSTRACT

BACKGROUND AND OBJECTIVE: Periodontal disease is a highly complex chronic inflammatory disease of the oral cavity. Multiple factors influence periodontal disease, including socio-economic status, genetics and age; however, inflammation elicited by the presence of specific bacteria in the subgingival space is thought to drive the majority of soft- and hard-tissue destruction. Porphyromonas gingivalis is closely associated with periodontal disease. Toll-like receptors (TLRs) and their intracellular signaling pathways play roles in the host response to P. gingivalis. The focus of the current study was to use microarray analysis to define the contributions of the TLR adaptor molecules myeloid differentiation factor 88 (MyD88) and Toll/interleukin-1 receptor domain-containing adaptor inducing interferon-beta (TRIF), and aging, on the expression of TLR pathway-associated mRNAs in response to P. gingivalis. MATERIAL AND METHODS: Bone marrow-derived macrophages (BMØ) from wild-type (Wt), MyD88 knockout (MyD88-KO) and Trif(Lps2) [i.e. containing a point mutation in the lipopolysaccharide 2 (Lps2) gene rendering the Toll/interleukin (IL)-1 receptor domain-containing adaptor inducing interferon-beta (TRIF) protein nonfunctional] mice, at 2-and 12-mo of age, were cultured with P. gingivalis. Expression of genes in BMØ cultured with P. gingivalis was determined in comparison with expression of genes in BMØ cultured in medium only. RESULTS: Using, as criteria, a twofold increase or decrease in mRNA expression, differential expression of 32 genes was observed when Wt BMØ from 2-mo-old mice were cultured with P. gingivalis compared with the medium-only control. When compared with 2-mo-old Wt mice, 21 and 12 genes were differentially expressed (p < 0.05) as a result of the mutations in MyD88 or TRIF, respectively. The expression of five genes was significantly (p < 0.05) reduced in Wt BMØ from 12-mo-old mice compared with those from 2-mo-old mice following culture with P. gingivalis. Age also influenced the expression of genes in MyD88-KO and Trif(Lps2) mice challenged with P. gingivalis. CONCLUSIONS: Our results indicate that P. gingivalis induces differential expression of TLR pathway-associated genes, and both MyD88 and TRIF play roles in the expression of these genes. Age also played a role in the expression of TLR-associated genes following stimulation of BMØ with P. gingivalis.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Aging/genetics , Myeloid Differentiation Factor 88/genetics , Porphyromonas gingivalis/immunology , Signal Transduction/genetics , Toll-Like Receptors/genetics , Aging/immunology , Animals , Bone Marrow Cells/immunology , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation/genetics , Gene Knockout Techniques , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Lipopolysaccharides/genetics , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Point Mutation/genetics
5.
Mol Oral Microbiol ; 28(1): 28-39, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23194377

ABSTRACT

Clinical studies and experimental modeling identify a potential link between periodontal disease and periodontal pathogens such as Porphyromonas gingivalis and atherosclerosis and formation of macrophage foam cells. Toll-like receptors and molecules governing their intracellular signaling pathways such as MyD88 play roles in atherosclerosis, as well as host response to P. gingivalis. The aim of this study was to define roles of MyD88 and TRIF during macrophage foam cell formation in response to P. gingivalis. In the presence of human low-density lipoprotein (LDL) mouse bone-marrow-derived macrophages (BMφ) cultured with P. gingivalis responded with significant reduction in tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). The BMφ stained strongly with oil red O, regardless of whether bacterial challenge occurred concurrent with or before LDL treatment. Heat-killed P. gingivalis stimulated foam cell formation in a similar way to live bacteria. The BMφ from MyD88-knockout and Lps2 mice revealed a significant role for MyD88, and a minor role for TRIF in P. gingivalis-elicited foam cell formation. Porphyromonas gingivalis-elicited TNF-α and IL-6 were affected by MyD88 ablation and to a lesser extent by TRIF status. These data indicate that LDL affects the TNF-α and IL-6 response of macrophages to P. gingivalis challenge and that MyD88 and TRIF play important roles in P. gingivalis-elicited foam cell formation.


Subject(s)
Foam Cells/microbiology , Macrophages/microbiology , Myeloid Differentiation Factor 88/immunology , Porphyromonas gingivalis/immunology , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/immunology , Animals , Azo Compounds , Bacterial Proteins/genetics , Bone Marrow Cells , Cells, Cultured , Coculture Techniques , Coloring Agents , Foam Cells/immunology , Humans , Inflammation Mediators/immunology , Interleukin-6/immunology , Lipoproteins, LDL/pharmacology , Macrophages/immunology , Mannosyltransferases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Point Mutation/genetics , Tumor Necrosis Factor-alpha/immunology
6.
Mol Oral Microbiol ; 25(5): 305-16, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20883220

ABSTRACT

A hallmark of infection with the gram-negative pathogen Porphyromonas gingivalis is the induction of a chronic inflammatory response. P. gingivalis induces a local chronic inflammatory response that results in oral inflammatory bone destruction, which manifests as periodontal disease. In addition to chronic inflammation at the initial site of infection, mounting evidence has accumulated supporting a role for P. gingivalis-mediated periodontal disease as a risk factor for several systemic diseases including, diabetes, preterm birth, stroke, and atherosclerotic cardiovascular disease. A growing number of in vitro studies have demonstrated that P. gingivalis infection stimulates cell activation commensurate with expected responses paralleling inflammatory atherosclerotic-type responses. Furthermore, various mouse models have been used to examine the ability of P. gingivalis to stimulate chronic inflammatory plaque accumulation and recent studies have pointed to a pivotal role for innate immune signaling via the Toll-like receptors in the chronic inflammation associated with P. gingivalis infection. In this review we discuss the pathogen and host cell specificity of these responses and discuss possible mechanisms by which this oral pathogen can induce and maintain a chronic state of inflammation at sites distant from oral infection.


Subject(s)
Atherosclerosis/etiology , Bacteroidaceae Infections/immunology , Chronic Periodontitis/complications , Focal Infection, Dental/immunology , Focal Infection, Dental/microbiology , Porphyromonas gingivalis/immunology , Animals , Atherosclerosis/immunology , Atherosclerosis/microbiology , Chronic Periodontitis/immunology , Chronic Periodontitis/microbiology , Disease Models, Animal , Endothelium, Vascular/microbiology , Foam Cells/immunology , Humans , Immunity, Cellular , Immunity, Innate , Mice , Organ Specificity , Toll-Like Receptors/immunology
7.
Oral Microbiol Immunol ; 24(6): 456-63, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19832797

ABSTRACT

INTRODUCTION: Porphyromonas gingivalis is a periodontopathic bacterium closely associated with generalized aggressive periodontal disease. Pattern recognition receptors (PRRs) participate in host response to this organism. It is likely that PRRs not previously recognized as part of the host response to P. gingivalis also participate in host response to this organism. METHODS AND RESULTS: Employing qRT-PCR, we observed increased msr1 gene expression at 2, 6, and 24 h of culture with P. gingivalis strain 381. Flow cytometry revealed increased surface expression of SR-A protein by the 24 h time point. Macrophages cultured with an attachment impaired P. gingivalis fimA- mutant (DPG3) expressed intermediate levels of SR-A expression. Heat-killed P. gingivalis stimulated SR-A expression similar to live bacteria, and purified P. gingivalis capsular polysaccharide stimulated macrophage SR-A expression, indicating that live whole organisms are not necessary for SR-A protein expression in macrophage response. As SR-A is known to play a role in lipid uptake by macrophages, we tested the ability of low-density lipoprotein (LDL) to influence the SR-A response of macrophages to P. gingivalis, and observed no effect of LDL on P. gingivalis-elicited SR-A expression. Lastly, we observed that SR-A knockout (SR-A(-/-)) mouse macrophages produced significantly more tumor necrosis factor (TNF)-alpha than wild type mouse macrophages cultured with P. gingivalis. CONCLUSION: These data identify that SR-A is expressed by macrophages in response to P. gingivalis, and support that this molecule plays a role in TNF-alpha production by macrophages to this organism.


Subject(s)
Macrophages/metabolism , Porphyromonas gingivalis/immunology , Scavenger Receptors, Class A/biosynthesis , Scavenger Receptors, Class A/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Cells, Cultured , Fimbriae Proteins/immunology , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Scavenger Receptors, Class A/genetics , Virulence Factors/immunology
8.
J Dent Res ; 85(2): 106-21, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16434728

ABSTRACT

Periodontal diseases are a group of diseases that lead to erosion of the hard and soft tissues of the periodontium, which, in severe cases, can result in tooth loss. Anecdotal clinical observations have suggested that poor oral health may be associated with poor systemic health; however, only recently have appropriate epidemiological studies been initiated, with defined clinical endpoints of periodontal disease, to address the association of periodontal disease with increased risk for cardiovascular and cerebrovascular disease. Although conflicting reports exist, these epidemiological studies support this connection. Paralleling these epidemiological studies, emerging basic scientific studies also support that infection may represent a risk factor for atherosclerosis. With P. gingivalis as a model pathogen, in vitro studies support that this organism can activate host innate immune responses associated with atherosclerosis, and in vivo studies demonstrate that this organism can accelerate atheroma deposition in animal models. In this review, we focus primarily on the basic scientific studies performed to date which support that infection with bacteria, most notably P. gingivalis, accelerates atherosclerosis. Furthermore, we attempt to bring together these studies to provide an up-to-date framework of emerging theories into the mechanisms underlying periodontal disease and increased risk for atherosclerosis, as well as identify intervention strategies to reduce the incidence of periodontal disease in humans, in an attempt to decrease risk for systemic complications of periodontal disease such as atherosclerotic cardiovascular disease.


Subject(s)
Atherosclerosis/immunology , Atherosclerosis/microbiology , Immunity, Innate , Periodontal Diseases/immunology , Porphyromonas gingivalis/pathogenicity , Animals , Atherosclerosis/etiology , Disease Models, Animal , Endothelial Cells/immunology , Endothelial Cells/microbiology , Humans , Inflammation Mediators/physiology , Macrophages/immunology , Macrophages/microbiology , Mice , Periodontal Diseases/complications , Periodontal Diseases/microbiology , Porphyromonas gingivalis/immunology , Signal Transduction , Toll-Like Receptors/immunology
9.
Bone ; 36(3): 472-9, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15777672

ABSTRACT

The role of inducible nitric oxide synthase (iNOS) in bone development and bacterially induced periodontal bone loss was examined using mice with targeted mutation of the iNOS gene. Femurs of iNOS KO mice showed 30% and 9% higher bone mineral density compared to wild type (WT) at 4 and 9 weeks of age, respectively. Micro-computed tomography revealed that cortical thickness and cortical bone density is increased in the absence of iNOS, while trabecular bone thickness and bone density remains unchanged. Histochemical analysis using TRAP staining showed that osteoclast numbers are lower by 25% in iNOS KO femurs compared to WT femurs. When bone marrow cells were stimulated with M-CSF and RANKL in vitro, iNOS KO cultures developed 51% fewer TRAP-positive multinuclear cells compared to WT cultures. When similar cultures were grown on dentine discs, resorption pit area was decreased by 54% in iNOS KO cultures. Gene expression studies showed that iNOS expression is induced by M-CSF and RANKL in WT bone marrow cultures, while no iNOS transcript was detected in iNOS KO. No compensatory change was detected in the expression of neuronal or endothelial NOS isoforms. There was no difference in RANK and osteoprotegerin expression between iNOS KO and WT bone marrow cultures after M-CSF and RANKL-treatment, while Traf6 expression was significantly lower in the absence of iNOS. In the alveolar bone of the maxilla, the distance between the cementoenamel junction and the alveolar bone crest was larger in iNOS KO compared to WT mice from 6 to 14 weeks of age, indicating a developmental effect of iNOS in oral tissues. Oral administration of the periodontal pathogen Porphyromonas gingivalis caused alveolar bone loss in the maxilla of WT mice, but failed to do so in iNOS KO mice. Expression of the osteoclast marker cathepsin K was 25% lower in iNOS KO alveolar bone. These data indicate that iNOS promotes bone resorption during bone development as well as after bacterial infection, and that iNOS is an important signal for normal osteoclast differentiation.


Subject(s)
Alveolar Bone Loss/enzymology , Alveolar Bone Loss/microbiology , Bone Development/physiology , Nitric Oxide Synthase/physiology , Porphyromonas gingivalis/pathogenicity , Alveolar Bone Loss/genetics , Animals , Bacteroidaceae Infections/enzymology , Bacteroidaceae Infections/genetics , Bacteroidaceae Infections/microbiology , Bone Development/genetics , Cells, Cultured , Female , Gene Expression Regulation/physiology , Maxillary Diseases/enzymology , Maxillary Diseases/genetics , Maxillary Diseases/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/physiology , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase/deficiency , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II
10.
Infect Immun ; 69(12): 7959-63, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11705986

ABSTRACT

The arginine gingipains RgpA and RgpB of Porphyromonas gingivalis are well-documented virulence factors of this organism. Structurally, RgpA and RgpB have nearly identical catalytic domains, while RgpA possesses an additional hemagglutinin domain. In this study, we examined the abilities of these proteins to elicit protection against P. gingivalis-mediated oral bone loss in a murine oral challenge model. Mice immunized subcutaneously with heat-killed P. gingivalis or purified RgpA or RgpB possessed elevated levels of P. gingivalis-specific immunoglobulin G; however, only the animals immunized with P. gingivalis whole cells or RgpA were protected from maxillary bone loss. These data suggest that immunization with RgpA stimulates the production of hemagglutinin domain-specific antibodies, which contribute to the prevention of P. gingivalis-mediated periodontal disease.


Subject(s)
Alveolar Bone Loss/prevention & control , Bacterial Vaccines/immunology , Bacteroidaceae Infections/prevention & control , Cysteine Endopeptidases/immunology , Hemagglutinins/immunology , Porphyromonas gingivalis/immunology , Adhesins, Bacterial , Animals , Antibodies, Bacterial , Gingipain Cysteine Endopeptidases , Mice , Mice, Inbred BALB C , Porphyromonas gingivalis/enzymology , Vaccination
11.
Infect Immun ; 69(9): 5840-8, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11500462

ABSTRACT

In this study we utilized immortalized morphologically and functionally distinct epithelial cell lines from normal human endocervix, ectocervix, and vagina to characterize gonococcal epithelial interactions pertinent to the lower female genital tract. Piliated, but not nonpiliated, N. gonorrhoeae strain F62 variants actively invaded these epithelial cell lines, as demonstrated by an antibiotic protection assay and confocal microscopy. Invasion of these cells by green fluorescent protein-expressing gonococci was characterized by colocalization of gonococci with F actin, which were initially detected 30 min postinfection. In all three cell lines, upregulation of interleukin 8 (IL-8) and IL-6, intercellular adhesion molecule 1 (CD54), and the nonspecific cross-reacting antigen (CD66c) were detected 4 h after infection with piliated and nonpiliated gonococci. Furthermore, stimulation of all three cell lines with gonococcal whole-cell lysates resulted in a similar upregulation of IL-6 and IL-8, confirming that bacterial uptake is not essential for this response. Increased levels of IL-1 were first detected 8 h after infection with gonococci, suggesting that the earlier IL-8 and IL-6 responses were not mediated through the IL-1 signaling pathway. The IL-1 response was limited to cultures infected with piliated gonococci and was more vigorous in the endocervical epithelial cells. The ability of gonococci to stimulate distinct proinflammatory host responses in these morphologically and functionally different compartments of the lower female genital tract may contribute directly to the inflammatory signs and symptoms characteristic of disease caused by N. gonorrhoeae.


Subject(s)
Cervix Uteri/microbiology , Epithelial Cells/microbiology , Inflammation Mediators/metabolism , Inflammation/immunology , Neisseria gonorrhoeae/immunology , Vagina/microbiology , Cell Line, Transformed , Cell Transformation, Viral , Cervix Uteri/cytology , Cervix Uteri/immunology , Epithelial Cells/immunology , Female , Gonorrhea/immunology , Gonorrhea/microbiology , Humans , Intercellular Adhesion Molecule-1/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Neisseria gonorrhoeae/pathogenicity , Up-Regulation , Vagina/cytology , Vagina/immunology
12.
J Immunol ; 163(2): 893-7, 1999 Jul 15.
Article in English | MEDLINE | ID: mdl-10395684

ABSTRACT

Little is known regarding the mechanism by which T cells control intraabdominal abscess formation. Treating animals with polysaccharide A (PS A) from Bacteroides fragilis shortly before or after challenge protects against abscess formation subsequent to challenge with different abscess-inducing bacteria. Although bacterial polysaccharides are considered to be T cell-independent Ags, T cells from PS A-treated animals mediate this protective activity. In the present study, we demonstrate that CD4+ T cells transfer PS A-mediated protection against abscess formation, and that a soluble mediator produced by these cells confers this activity. Cytokine mRNA analysis showed that T cells from PS A-treated animals produced transcript for IL-2, IFN-gamma, and IL-10, but not for IL-4. The addition of IL-2-specific Ab to T cell lysates taken from PS A-treated animals abrogated the ability to transfer protection, whereas the addition of Abs specific for IFN-gamma and IL-10 did not affect protection. Finally, administration of rIL-2 to animals at the time of bacterial challenge prevented abscess formation in a dose-dependent manner. These data demonstrate that PS A-mediated protection against abscess formation is dependent upon a CD4+ T cell-dependent response, and that IL-2 is essential to this immune mechanism.


Subject(s)
Abdominal Abscess/immunology , Interleukin-2/physiology , Sepsis/immunology , Abdominal Abscess/prevention & control , Adoptive Transfer , Animals , Antibody Specificity , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/transplantation , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Cytokines/genetics , Cytokines/immunology , Immune Sera/pharmacology , Injections, Intraperitoneal , Interleukin-2/therapeutic use , Male , Polysaccharides, Bacterial/administration & dosage , Polysaccharides, Bacterial/immunology , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Recombinant Proteins/therapeutic use , Sepsis/prevention & control , Solubility
13.
J Infect Dis ; 178(1): 200-6, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9652441

ABSTRACT

Two immunomodulating polysaccharides, poly-(1-6)-beta-glucotriosyl-(1-3)-beta-glucopyranose (PGG)-glucan and Bacteroides fragilis polysaccharide A (PS A), were evaluated for the prevention of mortality and abscess formation associated with experimental intraabdominal sepsis. Prophylactic treatment with a combination of these compounds significantly reduced mortality (8% vs. 44% in the saline-treated control group) and the incidence of abscesses (30% vs. 100% in the saline-treated control group) after challenge with rat cecal contents. These compounds were also effective when administered therapeutically after bacterial contamination of the peritoneal cavity. PS A treatment conferred long-term protection against abscess formation and resulted in significantly fewer total aerobes and anaerobes in the peritoneal fluid of animals challenged with cecal contents. These data demonstrate the usefulness of two immunomodulatory polysaccharides in preventing experimental intraabdominal sepsis in the absence of antimicrobial therapy and may represent a new adjunct to antibiotic regimens currently used to prevent clinical cases of this disease.


Subject(s)
Abdominal Abscess/prevention & control , Adjuvants, Immunologic/therapeutic use , Bacteroides Infections/prevention & control , Glucans/therapeutic use , Polysaccharides, Bacterial/therapeutic use , Sepsis/prevention & control , beta-Glucans , Abdominal Abscess/immunology , Animals , Bacteroides Infections/immunology , Bacteroides fragilis/immunology , Disease Models, Animal , Glucans/immunology , Male , Peritoneal Cavity/microbiology , Polysaccharides, Bacterial/immunology , Rats , Rats, Wistar , Sepsis/immunology
14.
J Immunol ; 160(10): 5000-6, 1998 May 15.
Article in English | MEDLINE | ID: mdl-9590249

ABSTRACT

We investigated the cellular mechanism by which Bacteroides fragilis promotes the development of intraabdominal abscesses in experimental models of sepsis. B. fragilis, as well as purified capsular polysaccharide complex (CPC) from this organism, adhered to primary murine mesothelial cells (MMCs) in vitro. The binding of CPC to murine peritoneal macrophage stimulated TNF-alpha production, which when transferred to monolayers of MMCs elicited significant ICAM-1 expression by these cells. This response resulted in enhanced polymorphonuclear leukocyte attachment to MMCs that could be inhibited by Abs specific for TNF-alpha or ICAM-1. Mice treated with TNF-alpha- or ICAM-1-specific Abs failed to develop intraabdominal abscesses following challenge with purified CPC. These results illustrated the role of the CPC in promoting adhesion of B. fragilis to the peritoneal wall and coordinating the cellular events leading to the development of abscesses associated with experimental intraabdominal sepsis.


Subject(s)
Abdominal Abscess/etiology , Bacteroides Infections/etiology , Bacteroides fragilis/physiology , Animals , Bacterial Adhesion , Epithelial Cells/microbiology , Humans , Intercellular Adhesion Molecule-1/physiology , Mice , Mice, Inbred C57BL , Polysaccharides, Bacterial/physiology , Tumor Necrosis Factor-alpha/physiology
15.
Infect Immun ; 64(6): 2201-5, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8675327

ABSTRACT

Previous studies have established the efficacy of soluble polymers of poly-(1-6)-beta-glucotriosyl-(1-3)-beta-glucopyranose (PGG) glucan, a biological-response modifier, in protecting against mortality associated with experimentally induced peritonitis in a rat model. PGG glucan-treated animals showed increases in total leukocyte counts and enhanced bacterial clearance from blood. To further explore the mechanisms) by which this agent confers protection, studies were performed to examine whether protection could be transferred from PGG glucan-treated animals to naive recipients via spleen cells (SC), SC lysates, or serum. Passive-transfer experiments indicated that the responsible factor(s) was transferable by whole SC and SC lysates, as well as by peripheral leukocytes or serum from animals treated with PGG glucan. The transferable factor(s) was resistant to pronase and trypsin digestion, was heat stable at 56 or 80 degrees C, and was not removed by NH4SO4 precipitation. The protective effect of PGG glucan was abrogated by treatment with indomethacin, a potent inhibitor of prostaglandin synthesis. Administration of a purified prostaglandin extract from the sera of PGG glucan-treated animals protected against mortality in the peritonitis model. Furthermore, treatment of rats with exogenous synthetic prostaglandin E2 also conferred protection against mortality. These results suggest that the protective effect exhibited by PGG glucan in the rat peritonitis model is mediated, at least in part, by prostaglandins.


Subject(s)
Adjuvants, Immunologic/pharmacology , Glucans/pharmacology , Immunotherapy, Adoptive , Sepsis/prevention & control , beta-Glucans , Animals , Dinoprostone/pharmacology , Indomethacin/pharmacology , Male , Prostaglandins/physiology , Rats , Rats, Wistar
16.
Infect Immun ; 64(3): 1065-9, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8641762

ABSTRACT

To stimulate early-phase immunologic events following Bacteroides fragilis infection in the peritoneal cavity, we examined the cytokine response of several cell types to purified capsular polysaccharide complex (CPC) and lipopolysaccharide (LPS) of this organism. Cytokines were produced from murine resident peritoneal (MRP) cells as well as human peripheral blood leukocytes. MRP cells cocultured with either B. fragilis CPC of LPS in vitro produced tumor necrosis factor alpha and interleukin-1alpha (IL-1alpha). In addition, MRP cells challenged with CPC produced IL-10. Human peripheral blood monocytes and polymorphonuclear leukocytes secreted IL-8 when cultured in the presence of CPC.


Subject(s)
Bacteroides fragilis/immunology , Cytokines/biosynthesis , Phagocytes/metabolism , Polysaccharides, Bacterial/toxicity , Animals , Bacteroides Infections/immunology , Cells, Cultured , Humans , Interleukin-1/biosynthesis , Interleukin-10/biosynthesis , Interleukin-8/biosynthesis , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha/biosynthesis
17.
Can J Microbiol ; 40(10): 865-72, 1994 Oct.
Article in English | MEDLINE | ID: mdl-8000965

ABSTRACT

In the absence of serum, Legionella pneumophila demonstrated wash-resistant adherence to U-937 cells, primary guinea-pig alveolar macrophages, and MRC-5 cells. Neither complement nor antibody was required for binding. The dynamics of adherence following inoculation of L. pneumophila at increasing 10-fold multiplicities of infection to each of the three host cell types resulted in a first-order kinetic relationship of binding, indicative of one bacterial adhesin molecule recognized by one host cell receptor moiety. Host cell receptor saturation studies showed that depending on the cell type, 2-8% of the bacterial inoculum adhered to cells under these nonopsonic conditions. Preliminary adhesin and receptor characterization studies were performed to define the chemical composition of the binding structures on both the organism and the three different host cell surfaces. The adherence phenomenon was investigated using competitive binding assays in the presence of putative adhesin analogs as well as following treatments modifying the microbial and host cell surface membranes. Attachment was evaluated both by viable bacterial cell colony counts and by indirect immunofluorescent assay. With the exception of aldehyde treatments, the various membrane-modifying regimes and the presence of the adhesin analogs were shown to have no effect on organism or host cell viability. Data suggested that the L. pneumophila adhesin responsible for opsonin-independent binding to these host cells was a protein structure with lectin-like properties. Furthermore, this protein would appear to be intimately associated with carbohydrate or lipid structures located on the bacterial outer membrane.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Adhesins, Bacterial/metabolism , Bacterial Adhesion/physiology , Legionella pneumophila/physiology , Macrophages, Alveolar/microbiology , Receptors, Immunologic/metabolism , Animals , Antibodies, Monoclonal , Antibody Specificity , Binding, Competitive , Cell Line , Fibroblasts , Guinea Pigs , Humans , Kinetics , Legionella pneumophila/metabolism , Receptors, Complement/physiology , Tumor Cells, Cultured
18.
Can J Microbiol ; 39(7): 718-22, 1993 Jul.
Article in English | MEDLINE | ID: mdl-8364804

ABSTRACT

Legionella pneumophila adhered to and multiplied intracellularly in the human histiocytic lymphoma U-937 cell line. The infectious process was evaluated by viable bacterial cell colony counts and documented by transmission and scanning electron microscopy. In the absence of opsonins, wash-resistant bacterial adherence to host cells occurred within 1 h and attachment of 1 or 2 organisms per U-937 host cell involved close surface interactions at the prokaryotic and eukaryotic membranes. Intracellular multiplication of bacteria was maximal by 24 h after inoculation of cell monolayers. Release of L. pneumophila from these cells appeared as a lytic process that resulted in an increase in the numbers of microorganisms in the extracellular fluids and a concomitant decline in the number of intracellular bacteria. The course of cellular infection was completed by 72 h. The cellular and ultrastructural events of L. pneumophila adherence and uptake by U-937 cells in the absence of antibody or complement have been defined. In addition, this work further establishes the U-937 cell as a suitable model for investigating Legionella--host cell interactions.


Subject(s)
Bacterial Adhesion/physiology , Legionella pneumophila/growth & development , Legionella pneumophila/ultrastructure , Lymphoma, Large B-Cell, Diffuse/ultrastructure , Opsonin Proteins/physiology , Colony Count, Microbial , Humans , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...