Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
BMC Immunol ; 12: 9, 2011 Jan 24.
Article in English | MEDLINE | ID: mdl-21261944

ABSTRACT

BACKGROUND: Immunologic responses of the tooth to caries begin with odontoblasts recognizing carious bacteria. Inflammatory propagation eventually leads to tooth pulp necrosis and danger to health. The present study aims to determine cytokine gene expression profiles generated within human teeth in response to dental caries in vivo and to build a mechanistic model of these responses and the downstream signaling network. RESULTS: We demonstrate profound differential up-regulation of inflammatory genes in the odontoblast layer (ODL) in human teeth with caries in vivo, while the pulp remains largely unchanged. Interleukins, chemokines, and all tested receptors thereof were differentially up-regulated in ODL of carious teeth, well over one hundred-fold for 35 of 84 genes. By interrogating reconstructed protein interaction networks corresponding to the differentially up-regulated genes, we develop the hypothesis that pro-inflammatory cytokines highly expressed in ODL of carious teeth, IL-1ß, IL-1α, and TNF-α, carry the converged inflammatory signal. We show that IL1ß amplifies antimicrobial peptide production in odontoblasts in vitro 100-fold more than lipopolysaccharide, in a manner matching subsequent in vivo measurements. CONCLUSIONS: Our data suggest that ODL amplifies bacterial signals dramatically by self-feedback cytokine-chemokine signal-receptor cycling, and signal convergence through IL1R1 and possibly others, to increase defensive capacity including antimicrobial peptide production to protect the tooth and contain the battle against carious bacteria within the dentin.


Subject(s)
Cytokines/genetics , Dental Caries/genetics , Dental Caries/immunology , Gene Regulatory Networks/genetics , Odontoblasts/immunology , Tooth/immunology , Tooth/pathology , Antimicrobial Cationic Peptides/biosynthesis , Cytokines/metabolism , DNA, Complementary/genetics , Dental Caries/pathology , Dental Pulp/immunology , Dental Pulp/pathology , Gene Expression Profiling , Gene Expression Regulation , Humans , Immunity, Innate/genetics , Inflammation Mediators/metabolism , Models, Immunological , Odontoblasts/pathology , Oligonucleotide Array Sequence Analysis , Protective Agents/metabolism , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , Signal Transduction/genetics , Transcription, Genetic
2.
BMC Immunol ; 11: 53, 2010 Oct 28.
Article in English | MEDLINE | ID: mdl-21029417

ABSTRACT

BACKGROUND: Protease-Activated Receptors (PARs), members of G-protein-coupled receptors, are activated by proteolytic activity of various proteases. Activation of PAR1 and PAR2 triggers innate immune responses in human oral keratinocytes (HOKs), but the signaling pathways downstream of PAR activation in HOKs have not been clearly defined. In this study, we aimed to determine if PAR1- and PAR2-mediated signaling differs in the induction of innate immune markers CXCL3, CXCL5 and CCL20 via ERK, p38 and PI3K/Akt. RESULTS: Our data show the induction of innate immunity by PAR1 requires both p38 and ERK MAP kinases, while PAR2 prominently signals via p38. However, inhibition of PI3K enhances expression of innate immune markers predominantly via suppressing p38 phosphorylation signaled by PAR activation. CONCLUSION: Our data indicate that proteases mediating PAR1 and PAR2 activation differentially signal via MAP kinase cascades. In addition, the production of chemokines induced by PAR1 and PAR2 is suppressed by PI3K/Akt, thus keeping the innate immune responses of HOK in balance. The results of our study provide a novel insight into signaling pathways involved in PAR activation.


Subject(s)
Keratinocytes/metabolism , Periodontitis/immunology , Periodontitis/metabolism , Receptor, PAR-1/metabolism , Receptor, PAR-2/metabolism , Cells, Cultured , Chemokine CCL20/biosynthesis , Chemokine CCL20/genetics , Chemokine CCL20/immunology , Chemokine CXCL5/biosynthesis , Chemokine CXCL5/genetics , Chemokine CXCL5/immunology , Chemokines, CXC/biosynthesis , Chemokines, CXC/genetics , Chemokines, CXC/immunology , Down-Regulation , Humans , Immunity, Innate , Keratinocytes/immunology , Keratinocytes/pathology , Mitogen-Activated Protein Kinase 3/metabolism , Mouth/pathology , Periodontitis/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, PAR-1/immunology , Receptor, PAR-2/immunology , Signal Transduction/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
3.
BMC Immunol ; 11: 37, 2010 Jul 09.
Article in English | MEDLINE | ID: mdl-20618959

ABSTRACT

BACKGROUND: Epithelial cells and dendritic cells (DCs) both initiate and contribute to innate immune responses to bacteria. However, much less is known about the coordinated regulation of innate immune responses between GECs and immune cells, particularly DCs in the oral cavity. The present study was conducted to investigate whether their responses are coordinated and are bacteria-specific in the oral cavity. RESULTS: The beta-defensin antimicrobial peptides hBD1, hBD2 and hBD3 were expressed by immature DCs as well as gingival epithelial cells (GECs). HBD1, hBD2 and hBD3 are upregulated in DCs while hBD2 and hBD3 are upregulated in GECs in response to bacterial stimulation. Responses of both cell types were bacteria-specific, as demonstrated by distinctive profiles of hBDs mRNA expression and secreted cytokines and chemokines in response to cell wall preparations of various bacteria of different pathogenicity: Fusobacterium nucleatum, Actinomyces naeslundii and Porphyromonas gingivalis. The regulation of expression of hBD2, IL-8, CXCL2/GRObeta and CCL-20/MIP3alpha by GECs was greatly enhanced by conditioned medium from bacterially activated DCs. This enhancement was primarily mediated via IL-1beta, since induction was largely attenuated by IL-1 receptor antagonist. In addition, the defensins influence DCs by eliciting differential cytokine and chemokine secretion. HBD2 significantly induced IL-6, while hBD3 induced MCP-1 to approximately the same extent as LPS, suggesting a unique role in immune responses. CONCLUSIONS: The results suggest that cytokines, chemokines and beta-defensins are involved in interaction of these two cell types, and the responses are bacteria-specific. Differential and coordinated regulation between GECs and DCs may be important in regulation of innate immune homeostasis and response to pathogens in the oral cavity.


Subject(s)
Bacteria/metabolism , Cytokines/metabolism , Dendritic Cells/microbiology , Gene Expression Regulation , Gingiva/cytology , Gingiva/microbiology , beta-Defensins/genetics , Bacteria/drug effects , Biomarkers/metabolism , Cells, Cultured , Chemokines/metabolism , Culture Media, Conditioned/pharmacology , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Immunity, Innate/drug effects , Immunity, Innate/genetics , Interleukin 1 Receptor Antagonist Protein/pharmacology , Lipopolysaccharides/pharmacology , Models, Immunological , beta-Defensins/metabolism
4.
J Oral Microbiol ; 22010 May 04.
Article in English | MEDLINE | ID: mdl-21523231

ABSTRACT

OBJECTIVE: Secretory leukocyte peptidase inhibitors (SLPI), elafin, squamous cell carcinoma antigen 1 and 2 (SCCA1 and SCCA2) are specific endogenous serine protease inhibitors expressed by epithelial cells that prevent tissue damage from excessive proteolytic enzyme activity due to inflammation. To determine the effects of various periopathogens on these protease inhibitors, we utilized human gingival epithelial cells (GECs) challenged with cell-free bacteria supernatants of various periopathogens Porphyromonas gingivalis, Tannerella forsythia, Aggregatibacter actinomycetemcomitans, and Fusobacterium nucleatum. DESIGN: The gene expression and secretion of SLPI, elafin, SCCA1, and SCCA2 were determined using real-time PCR and ELISA, respectively. The direct effects of periopathogens and P. gingivalis gingipain mutants on these inhibitors were examined in vitro by Western Blot. The effect on the innate immune response of GECs was measured by expression of antimicrobial peptides: human beta-defenisin-2 (hBD2) and chemokine (C-C motif) ligand 20 (CCL20). RESULTS: We found that SLPI, SCCA2, elafin, hBD2, and CCL20 gene expression levels were significantly induced (p<0.001) in response to P. gingivalis, whose virulence factors include cysteine proteases, but not in response to stimulation by other bacteria. P. gingivalis reduced the secretion of SLPI and elafin significantly in GECs, and degraded recombinant SLPI, elafin, SCCA1, and SCCA2. Differential degradation patterns of SLPI, elafin, SCCA1, and SCCA2 were observed with different bacteria as well as P. gingivalis mutants associated with the loss of specific gingipains secreted by P. gingivalis. In addition, pretreatment of GECs with SLPI, SCCA1, or SCCA2 partially blocked hBD2 and CCL20 mRNA expression in response to P. gingivalis, suggesting a protective effect. CONCLUSION: Our results suggest that different periopathogens affect the host protease inhibitors in a different manner, suggesting host susceptibility may differ in the presence of these pathogens. The balance between cellular protease inhibitors and their degradation may be an important factor in susceptibility to periodontal infection.

5.
Innate Immun ; 16(4): 226-34, 2010 Aug.
Article in English | MEDLINE | ID: mdl-19710093

ABSTRACT

Macrophage inflammatory protein-3alpha/C-C chemokine ligand 20 (MIP-3alpha/CCL20) is an antimicrobial peptide that plays an important role in innate immunity. In addition to direct microbicidal effects, MIP-3alpha/CCL20 also exhibits cytokine-like functions that are critical during dendritic cell activation. The aim of the present study was to investigate further which signaling pathways are involved in the MIP-3alpha/CCL20 mRNA expression in response to whole-cell Porphyromonas gingivalis. Primary gingival epithelial cells (GECs) and the immortalized oral keratinocyte cell-line OKF6/TERT-2 were stimulated with whole-cell P. gingivalis. Prior to stimulation, GECs and OKF6/TERT-2 cells were pretreated with specific inhibitors for nuclear-factor-kappaB (NF-kappaB), mitogen-activated protein kinase (MAPK), phospholipase C (PLC), and phosphatidylinositol-3-kinase (PI3K). In GECs and OKF6/TERT-2 cells, activation of NF-kappaB was examined after exposure to P. gingivalis. The gene expression of MIP-3alpha/CCL20 was significantly induced in response to P. gingivalis (P

Subject(s)
Chemokine CCL20/biosynthesis , NF-kappa B/physiology , Porphyromonas gingivalis/metabolism , Type C Phospholipases/physiology , p38 Mitogen-Activated Protein Kinases/physiology , Chemokine CCL20/genetics , Epithelial Cells/physiology , Gingiva/cytology , Humans , Indicators and Reagents , Interleukin-6/biosynthesis , Interleukin-6/genetics , NFATC Transcription Factors/physiology , Phosphatidylinositol 3-Kinases/physiology , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor RelA/biosynthesis , Transcription Factor RelA/genetics , Transcription Factor RelA/physiology , Transfection
6.
Innate Immun ; 16(2): 104-14, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19567485

ABSTRACT

Protease-activated receptors (PARs) are G-protein-coupled receptors with an active role in host defense. The two most highly expressed members of the PAR family in gingival epithelial cells (GECs) are PAR1 and PAR2. The major virulence factors of periodontal pathogen Porphyromonas gingivalis are its proteases which can activate PAR2. However, little is known about the function of PARs in GECs when they are activated by their endogenous agonist enzymes. The purpose of this study was to characterize how the expression of innate immune markers is modulated when PAR1 and PAR2 are activated by their agonist enzymes, thrombin and trypsin, respectively. Here, we report that activation of PAR1 and PAR2 induces cell proliferation at low concentration. Activation of PAR via proteolytic activity of thrombin and trypsin induces expression of CXCL5/ENA-78 and CCL20/MIP3alpha in a concentration-dependent manner. Induction of CXCL5 via PAR1 was inhibited in the presence of PAR1 cleavage blocking antibodies and by PAR1 siRNA. The induction of CXCL5 and CCL20 via PAR2 was inhibited by PAR2 siRNA. These findings indicate an active role in innate immune responses by PAR1 and PAR2 in GECs. Modulation of innate immunity by PARs may contribute to co-ordinated and balanced immunosurveillance in GECs.


Subject(s)
Chemokine CCL20/biosynthesis , Chemokine CXCL5/biosynthesis , Epithelial Cells/metabolism , Receptor, PAR-1/metabolism , Receptor, PAR-2/metabolism , Antibodies, Blocking/metabolism , Cell Culture Techniques , Cell Proliferation/drug effects , Cells, Cultured , Chemokine CCL20/genetics , Chemokine CXCL5/genetics , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/pathology , Gingiva/pathology , Humans , Immunity, Innate , RNA, Small Interfering/genetics , Receptor, PAR-1/genetics , Receptor, PAR-2/genetics , Thrombin/pharmacology , Trypsin/pharmacology
7.
AIDS Res Hum Retroviruses ; 25(10): 1013-21, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19842793

ABSTRACT

The oral mucosa is relatively resistant to human immunodeficiency virus type 1 (HIV-1) transmission. The mechanisms contributing to this resistance remain incompletely understood, but may include HIV-induced synthesis of innate immune factors. We used fully differentiated oral epithelium as a surrogate for the oral mucosa in vivo, exposed it to X4- and R5-tropic HIV-1 in culture, and quantified mRNA expression of six innate immune factors. Neither virus increased expression of human beta defensin 2 (hBD-2) mRNA over supernatants from uninfected lymphoblast controls. HIV-1 also failed to induce mRNA of four additional innate immunity-related genes. Similar results were obtained with oral monolayer epithelial cells. Interestingly, the X4-tropic virus inhibited mRNA expression of hBD-2, and of three of the other factors, at higher dosages in the differentiated oral epithelium but not the monolayers. The failure of HIV-1 to induce innate immune factors in the differentiated epithelium was not due to a lack of tissue penetration, as we detected fluorescence-tagged virions up to 30 mum deep from the apical surface. HIV-1 does not trigger de novo innate immune factor synthesis in oral epithelium, pointing to the role of a constitutive innate immunity for protection against HIV-1 in the oral cavity.


Subject(s)
Epithelium/immunology , HIV-1/immunology , Immunologic Factors/biosynthesis , Mouth Mucosa/immunology , Cells, Cultured , Gene Expression Profiling , Humans
8.
BMC Oral Health ; 9: 21, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19712472

ABSTRACT

BACKGROUND: Human beta-defensins (hBDs) are antimicrobial peptides with a role in innate immune defense. Our laboratory previously showed that a single nucleotide polymorphism (SNP) in the 5' untranslated region of the hBD1 gene (DEFB1), denoted -44 (rs1800972), is correlated with protection from oral Candida. Because this SNP alters the putative mRNA structure, we hypothesized that it alters hBD1 expression. METHODS: Transfection of reporter constructs and evaluation of antimicrobial activity and mRNA expression levels in keratinocytes from multiple donors were used to evaluate the effect of this SNP on constitutive and induced levels of expression. RESULTS: Transfection of CAT reporter constructs containing the 5' untranslated region showed that the -44 G allele yielded a 2-fold increase in CAT protein compared to other common haplotypes suggesting a cis effect on transcription or translation. The constitutive hBD1 mRNA level in human oral keratinocytes was significantly greater in cells from donors with the -44 GG genotype compared to those with the common CC genotype. Surprisingly, the hBD3 mRNA level as well as antimicrobial activity of keratinocyte extracts also correlated with the -44 G allele. Induced levels of hBD1, hBD2, and hBD3 mRNA were evaluated in keratinocytes challenged with Toll-like receptor 2 and 4 ligands, interleukin-1beta, TNFalpha, and interferon-gamma (IFNgamma). In contrast to constitutive expression levels, IFNgamma-induced keratinocyte hBD1 and hBD3 mRNA expression was significantly greater in cells with the common CC genotype, but there was no clear correlation of genotype with hBD2 expression. CONCLUSION: The DEFB1 -44 G allele is associated with an increase in overall constitutive antimicrobial activity and expression of hBD1 and hBD3 in a manner that is consistent with protection from candidiasis, while the more common C allele is associated with IFNgamma inducibility of these beta-defensins and is likely to be more protective in conditions that enhance IFNgamma expression such as chronic periodontitis. These results suggest a complex relationship between genetics and defensin expression that may influence periodontal health and innate immune responses.

9.
J Med Microbiol ; 56(Pt 7): 976-987, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17577065

ABSTRACT

Oral epithelia are constantly exposed to non-pathogenic (commensal) bacteria, but generally remain healthy and uninflamed. Fusobacterium nucleatum, an oral commensal bacterium, strongly induces human beta-defensin-2 (hBD2), an antimicrobial and immunomodulatory peptide, in gingival epithelial cells (GECs). hBD2 is also expressed in normal oral tissue leading to the hypothesis that oral epithelia are in an activated state with respect to innate immune responses under normal in vivo conditions. In order to test this hypothesis, global gene expression was evaluated in GECs in response to stimulation by an F. nucleatum cell wall (FnCW) preparation and to hBD2 peptide. FnCW treatment altered 829 genes, while hBD2 altered 209 genes (P<0.005, ANOVA). Many induced genes were associated with the gene ontology categories of immune responses and defence responses. Consistent with the hypothesis, similar responses were activated by commensal bacteria and hBD2. These responses included up-regulation of common antimicrobial effectors and chemokines, and down-regulation of proliferation markers. In addition, FnCW up-regulated multiple protease inhibitors, and suppressed NF-kappaB function and the ubiquitin/proteasome system. These global changes may protect the tissue from inflammatory damage. Both FnCW and hBD2 also up-regulated genes that may enhance the epithelial barrier. The findings suggest that both commensal bacteria and hBD2 activate protective responses of GECs and play an important role in immune modulation in the oral cavity.


Subject(s)
Epithelial Cells/immunology , Fusobacterium nucleatum/immunology , Gene Expression Regulation , Gingiva/microbiology , beta-Defensins/pharmacology , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Epithelial Cells/microbiology , Gene Expression Regulation/drug effects , Gingiva/cytology , Gingiva/immunology , Humans , Immunity, Innate , Inflammation , Oligonucleotide Array Sequence Analysis , Protease Inhibitors/metabolism , Proteins/genetics , Proteins/metabolism , beta-Defensins/metabolism
10.
Infect Immun ; 75(9): 4326-33, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17591792

ABSTRACT

The oral pathogen Porphyromonas gingivalis secretes proteases such as Arg-gingipain B (RgpB) that activate protease-activated receptors (PARs). Human beta-defensins (hBDs) and the macrophage inflammatory protein 3alpha/CC chemokine ligand 20 (CCL20) produced by epithelial cells are antimicrobial peptides that provide cytokine function and play an important role in innate immunity. The aim of the present study was to determine whether specific members of the PAR family mediate the expression of these innate immunity markers in gingival epithelial cells (GECs) when exposed to P. gingivalis cell-free culture supernatant or purified RgpB. hBD-2 mRNA in GECs was induced in response to supernatant and purified RgpB from P. gingivalis (P = 0.02 and P = 0.016, respectively). This effect was abrogated by the protease inhibitor tosyl-l-lysine chloromethyl ketone (TLCK) (P < 0.05). In response to P. gingivalis supernatant and to purified RgpB, the hBD-2 mRNA expression was significantly decreased in PAR-2 gene knockdown cells, whereas no change was detected in PAR-1 gene knockdown cells. CCL20 mRNA expression also increased in response to the supernatant of P. gingivalis, and this effect was blocked by the protease inhibitor, TLCK (P = 0.05 and P = 0.024, respectively), and was blocked in PAR-2 gene knockdown cells. Our data indicate that hBD-2 and CCL20 mRNA up-regulation by P. gingivalis supernatant and purified RgpB was mediated via PAR-2, but not via PAR-1, and that proteases play a role in the regulation of innate immune responses in GECs. GECs use PARs to recognize P. gingivalis and mediate cell responses involved in innate immunity.


Subject(s)
Chemokines, CC/genetics , Chemokines, CC/metabolism , Macrophage Inflammatory Proteins/genetics , Macrophage Inflammatory Proteins/metabolism , Peptide Hydrolases/metabolism , Porphyromonas gingivalis/enzymology , RNA, Messenger/biosynthesis , Receptor, PAR-2/physiology , beta-Defensins/genetics , beta-Defensins/metabolism , Cells, Cultured , Chemokine CCL20 , Chemokines, CC/biosynthesis , Epithelial Cells/enzymology , Epithelial Cells/microbiology , Gene Expression Regulation, Bacterial/physiology , Gingiva/cytology , Gingiva/enzymology , Gingiva/microbiology , Humans , Macrophage Inflammatory Proteins/biosynthesis , Peptide Hydrolases/physiology , Porphyromonas gingivalis/physiology , Up-Regulation/genetics , beta-Defensins/biosynthesis
11.
J Invest Dermatol ; 126(11): 2408-13, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16902423

ABSTRACT

Harlequin ichthyosis (HI) is the most severe form of autosomal-recessive, congenital ichthyosis. Affected infants have markedly impaired barrier function and are more susceptible to infection. Abnormalities in the localization of epidermal lipids as well as abnormal lamellar granule formation are features of HI skin. Previously, we and others have shown that mutations in the ABCA12 gene encoding an adenosine triphosphate-binding cassette (ABC) transporter underlie the skin disease HI. In this study, we have sequenced the ABCA12 gene in an additional 14 patients and show that all contain mutations, with the majority being either nonsense substitution or frameshift mutations. Eleven HI patients had bi-allelic ABCA12 mutations, whereas in the remaining three HI patients in this study, ABCA12 mutations were detected on only one allele by sequencing. In addition, the one patient from the previous study where no sequence mutations were detected was screened for heterozygous deletions. A combination of oligonucleotide arrays, multiplex PCR analysis and single-nucleotide polymorphism genotyping revealed a heterozygous intragenic deletion in exon 8. These mutation data establish ABCA12 as the major HI gene.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Ichthyosis, Lamellar/genetics , Codon, Nonsense , DNA Mutational Analysis , Exons/genetics , Female , Frameshift Mutation , Humans , Male , Mutation , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Sequence Deletion
12.
BMC Oral Health ; 6 Suppl 1: S13, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16934114

ABSTRACT

The presence of antimicrobial peptides (AMPs) in saliva may be a biological factor that contributes to susceptibility or resistance to caries. This manuscript will review AMPs in saliva, consider their antimicrobial and immunomodulatory functions, and evaluate their potential role in the oral cavity for protection of the tooth surface as well as the oral mucosa. These AMPs are made in salivary gland and duct cells and have broad antimicrobial activity. Alpha-defensins and LL37 are also released by neutrophils into the gingival crevicular fluid. Both sources may account for their presence in saliva. A recent study in middle school children aimed to determine a possible correlation between caries prevalence in children and salivary concentrations of the antimicrobial peptides human beta-defensin-3 (hBD-3), the cathelicidin, LL37, and the alpha-defensins. The levels of these AMPs were highly variable in the population. While levels of LL37 and hBD-3 did not correlate with caries experience, the mean alpha-defensin level was significantly higher in children with no caries than in children with caries (p < 0.005). We conclude that several types of AMPs that may have a role in oral health are present in unstimulated saliva. Low salivary levels of alpha-defensin may represent a biological factor that contributes to caries susceptibility. Our observation could lead to new ways to prevent caries and to a new tool for caries risk assessment.

13.
Arch Oral Biol ; 51(9): 775-83, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16815238

ABSTRACT

OBJECTIVE: Oral epithelia function as a microbial barrier and are actively involved in recognizing and responding to bacteria. Our goal was to examine a tissue engineered model of buccal epithelium for its response to oral bacteria and proinflammatory cytokines and compare the tissue responses with those of a submerged monolayer cell culture. DESIGN: The tissue model was characterized for keratin and beta-defensin expression. Altered expression of beta-defensins was evaluated by RT-PCR after exposure of the apical surface to oral bacteria and after exposure to TNF-alpha in the medium. These were compared to the response in traditional submerged oral epithelial cell culture. RESULTS: The buccal model showed expression of differentiation specific keratin 13, hBD1 and hBD3 in the upper half of the tissue; hBD2 was not detected. hBD1 mRNA was constitutively expressed, while hBD2 mRNA increased 2-fold after exposure of the apical surface to three oral bacteria tested and hBD3 mRNA increased in response to the non-pathogenic bacteria tested. In contrast, hBD2 mRNA increased 3-600-fold in response to bacteria in submerged cell culture. HBD2 mRNA increased over 100-fold in response to TNF-alpha in the tissue model and 50-fold in submerged cell culture. Thus, the tissue model is capable of upregulating hBD2, however, the minimal response to bacteria suggests that the tissue has an effective antimicrobial barrier due to its morphology, differentiation, and defensin expression. CONCLUSIONS: The oral mucosal model is differentiated, expresses hBD1 and hBD3, and has an intact surface with a functional antimicrobial barrier.


Subject(s)
Gingivitis/microbiology , Mouth Mucosa/microbiology , Cells, Cultured , Fusobacterium nucleatum , Gene Expression Regulation , Gingivitis/metabolism , Humans , Immunohistochemistry , Keratin-13/analysis , Keratin-13/metabolism , Keratin-14/analysis , Keratin-14/metabolism , Mouth Mucosa/chemistry , Porphyromonas gingivalis , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Streptococcus , Tissue Culture Techniques , Tumor Necrosis Factor-alpha/pharmacology , beta-Defensins/analysis , beta-Defensins/genetics
14.
Curr Issues Mol Biol ; 7(2): 119-33, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16053246

ABSTRACT

The oral cavity is a unique environment in which antimicrobial peptides play a key role in maintaining health and may have future therapeutic applications. Present evidence suggests that alpha-defensins, beta-defensins, LL-37, histatin, and other antimicrobial peptides and proteins have distinct but overlapping roles in maintaining oral health and preventing bacterial, fungal, and viral adherence and infection. The expression of the inducible hBD-2 in normal oral epithelium, in contrast to other epithelia, and the apparent differential signaling in response to commensal and pathogenic organisms, provides new insights into innate immunity in this body site. Commensal bacteria are excellent inducers of hBD-2 in oral epithelial cells, suggesting that the commensal bacterial community acts in a manner to benefit the overall innate immune readiness of oral epithelia. This may have major significance for understanding host defense in the complex oral environment.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Antimicrobial Cationic Peptides/metabolism , Mouth Mucosa/metabolism , Animals , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/physiology , Epithelium/immunology , Epithelium/metabolism , Epithelium/microbiology , Gingiva/immunology , Gingiva/metabolism , Immunity, Innate , Mice , Mouth Mucosa/immunology , Mouth Mucosa/microbiology
15.
Antimicrob Agents Chemother ; 49(9): 3883-8, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16127066

ABSTRACT

Dental caries is a major worldwide oral disease problem in children. Although caries are known to be influenced by dietary factors, the disease results from a bacterial infection; thus, caries susceptibility may be affected by host factors such as salivary antimicrobial peptides. This study aimed to determine a possible correlation between caries prevalence in children and salivary concentrations of the antimicrobial peptides human beta-defensin-3 (hBD-3), the cathelicidin LL37, and the alpha-defensins HNP1-3 (a mixture of HNP1, 2, 3). Oral examinations were performed on 149 middle school children, and unstimulated whole saliva was collected for immunoassays of the three peptides and for assay of caries-causing bacteria in saliva. The median salivary levels of hBD-3, LL37, and HNP1-3 were in the microgram/ml range but were highly variable in the population. While levels of LL37 and hBD-3 did not correlate with caries experience, the median HNP1-3 levels were significantly higher in children with no caries than in children with caries. Children with high caries levels did not have high levels of salivary Streptococcus mutans, and the HNP1-3 level was not correlated with salivary S. mutans. By immunohistochemistry we localized HNP1-3 in submandibular salivary duct cells. HNPs are also released by neutrophils into the gingival crevicular fluid. Both sources may account for their presence in saliva. Low salivary levels of HNP1-3 may represent a biological factor that contributes to caries susceptibility. This observation could lead to new ways to screen for caries susceptibility and to new means of assessing the risk for this common oral problem.


Subject(s)
Anti-Infective Agents/metabolism , Dental Caries/epidemiology , Dental Caries/microbiology , Saliva/metabolism , Adolescent , Antimicrobial Cationic Peptides/metabolism , Child , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , Defensins/metabolism , Dental Caries/pathology , Female , Humans , Immunohistochemistry , Male , Risk Assessment , Saliva/microbiology , Salivary Proteins and Peptides/metabolism , Streptococcus mutans/metabolism , alpha-Defensins/metabolism , Cathelicidins
16.
Am J Hum Genet ; 76(5): 794-803, 2005 May.
Article in English | MEDLINE | ID: mdl-15756637

ABSTRACT

Harlequin ichthyosis (HI) is the most severe and frequently lethal form of recessive congenital ichthyosis. Although defects in lipid transport, protein phosphatase activity, and differentiation have been described, the genetic basis underlying the clinical and cellular phenotypes of HI has yet to be determined. By use of single-nucleotide-polymorphism chip technology and homozygosity mapping, a common region of homozygosity was observed in five patients with HI in the chromosomal region 2q35. Sequencing of the ABCA12 gene, which maps within the minimal region defined by homozygosity mapping, revealed disease-associated mutations, including large intragenic deletions and frameshift deletions in 11 of the 12 screened individuals with HI. Since HI epidermis displays abnormal lamellar granule formation, ABCA12 may play a critical role in the formation of lamellar granules and the discharge of lipids into the intercellular spaces, which would explain the epidermal barrier defect seen in this disorder. This finding paves the way for early prenatal diagnosis. In addition, functional studies of ABCA12 will lead to a better understanding of epidermal differentiation and barrier formation.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Ichthyosis, Lamellar/genetics , Base Sequence , Chromosome Mapping , Chromosomes, Human, Pair 2 , Humans , Infant, Newborn , Microsatellite Repeats , Molecular Sequence Data , Mutation , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide
17.
J Immunol ; 173(8): 5165-70, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15470061

ABSTRACT

Epithelial tissues provide both a physical barrier and an antimicrobial barrier. Antimicrobial peptides of the human beta-defensin (hBD) family are part of the innate immune responses that play a role in mucosal defense. hBDs are made in epithelia including oral epithelium where the bacterial load is particularly great. hBD-2 and hBD-3 are up-regulated in response to bacterial stimuli. Previous studies show that hBD-2 expression in human gingival epithelial cells (GEC) is stimulated by both nonpathogenic and pathogenic bacteria, including Porphyromonas gingivalis, a Gram-negative pathogen associated with periodontitis. Present evidence suggests that hBD-2 expression in GEC uses several signaling pathways, including an NF-kappaB-mediated pathway but without apparent LPS-TLR4 signaling. Protease-activated receptors (PAR) are G-protein-coupled receptors that mediate cellular responses to extracellular proteinases. P. gingivalis secretes multiple proteases that contribute to its virulence mechanisms. To determine whether PAR signaling is used in hBD-2 induction, GEC were stimulated with wild-type P. gingivalis or mutants lacking one or more proteases. hBD-2 mRNA expression was reduced in GEC stimulated with single protease mutants (11-67% compared with wild type), strongly reduced in double mutants (0.1-16%), and restored to wild-type levels (93%) in mutant with restored protease activity. Stimulation by wild type was partially blocked by inhibitors of phospholipase C, a main signaling pathway for PARs. Expression of hBD-3 was unaffected. Peptide agonist of PAR-2, but not PAR-1 activator, also induced hBD-2 in GEC. Thus, P. gingivalis proteases are directly involved in regulation of hBD-2 in cultured GEC, and this induction partially uses the PAR-2 receptor and signaling pathway.


Subject(s)
Cysteine Endopeptidases/physiology , Gene Expression Regulation , Gingiva/metabolism , Hemagglutinins/physiology , Porphyromonas gingivalis/enzymology , Receptor, PAR-1/physiology , Receptor, PAR-2/physiology , Signal Transduction/physiology , beta-Defensins/genetics , Adhesins, Bacterial , Epithelial Cells/metabolism , Gingipain Cysteine Endopeptidases , Humans , Interleukin-8/genetics , Polymerase Chain Reaction
18.
Infect Immun ; 72(1): 352-8, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14688115

ABSTRACT

The innate immune response is critical for the epithelial antimicrobial barrier. The human beta-defensins are small, cationic antimicrobial peptides that are made by epithelial cells and that play a role in mucosal and skin defenses. Human beta-defensin 1 (hBD-1) is expressed constitutively in epithelial tissues, whereas hBD-2 and hBD-3 are expressed in response to bacterial stimuli or inflammation. Previous studies showed that hBD-2 was induced by Fusobacterium nucleatum cell wall extract without the involvement of the NF-kappaB transcription factors, which typically are associated with innate immunity and inflammation. The goal of this study was to characterize signaling pathways involved in hBD-2 induction in response to commensal and pathogenic bacteria. Cultured human oral and foreskin keratinocytes were treated separately with inhibitors of NF-kappaB, c-Jun N-terminal kinase (JNK), and p38 and then stimulated with oral commensal Streptococcus gordonii, oral pathogens Porphyromonas gingivalis and Actinobacillus actinomycetemcomitans, skin commensal Staphylococcus epidermidis, or skin pathogen Streptococcus pyogenes. Different bacteria induced different levels of hBD-2 and in response to the various inhibitors tested, although certain common patterns were observed for commensal- and pathogen-stimulated cells. hBD-2 induction by all bacteria tested was partially or completely blocked by inhibitors of the JNK and p38 pathways. However, in addition, hBD-2 induction by pathogenic bacteria in both oral and foreskin keratinocytes was blocked by inhibitors of NF-kappaB. The results indicate that commensal and pathogenic bacteria utilize different pathways in hBD-2 induction and suggest that epithelial cells from different body sites have common signaling mechanisms to distinguish between commensal and pathogenic bacteria.


Subject(s)
Bacteria/pathogenicity , Gingiva/immunology , JNK Mitogen-Activated Protein Kinases , Keratinocytes/immunology , Signal Transduction , Skin/immunology , beta-Defensins/metabolism , Bacteria/immunology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Gingiva/cytology , Gingiva/microbiology , Humans , Immunity, Innate , Infant, Newborn , Keratinocytes/microbiology , MAP Kinase Kinase 4 , Male , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Skin/cytology , Skin/microbiology , beta-Defensins/genetics , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...