Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Indian Soc Pedod Prev Dent ; 19(4): 143-4, 2001 Dec.
Article in English | MEDLINE | ID: mdl-12396090

ABSTRACT

Occurrence of a supernumerary tooth in the mandibular incisor region is uncommon. This report describes a rare case of mandibular midline supernumerary tooth causing crowding in the anterior region. The supernumerary tooth was extracted.


Subject(s)
Tooth, Supernumerary , Child , Humans , Incisor/abnormalities , Male , Mandible
2.
J Immunol ; 160(5): 2401-7, 1998 Mar 01.
Article in English | MEDLINE | ID: mdl-9498783

ABSTRACT

In vivo, gastric infection with Helicobacter pylori leads to substantial production of the inflammatory cytokines IL-1, IL-6, TNF-alpha, and IL-8. H. pylori strains that contain the cag pathogenicity island (cag+) and are associated with ulceration and gastric carcinoma induce greater cytokine production than cag- strains. Expression of these cytokines is often regulated by the transcription factor complex, nuclear factor-kappa B (NF-kappa B) through kappa B-binding elements in the enhancer/promoter regions of their genes. We report that more virulent cag+ H. pylori strains induce increased NF-kappa B-DNA binding activity, which elevates IL-8 expression in AGS gastric epithelial cells. The cag+ H. pylori strains induce significant stimulation of IL-8 promoter-driven reporter activity, while cag- strains do not. Furthermore, mutation of specific genes within the cag island (picA1 and picB) ablates enhanced NF-kappa B activation and IL-8 transcription. Increased IL-8 expression is inhibited by mutation in either the NF-kappa B or NF-IL-6 binding element. The cag+ strains, compared with the cag- strains, induce enhanced nuclear localization of a RelA-containing NF-kappa B binding complex, but no increase in NF-IL-6 binding activity. These studies demonstrate that the ability of different types of H. pylori strains to activate NF-kappa B correlates with their ability to induce IL-8 transcription and indicate a mechanism for the heightened inflammatory response seen in subjects infected with cag+ H. pylori strains.


Subject(s)
Epithelial Cells/immunology , Gastric Mucosa/immunology , Gene Expression Regulation/immunology , Helicobacter pylori/immunology , Interleukin-8/genetics , NF-kappa B/physiology , Bacterial Proteins/genetics , Bacterial Proteins/physiology , CCAAT-Enhancer-Binding Proteins , DNA/metabolism , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/physiology , Epithelial Cells/metabolism , Gastric Mucosa/cytology , Gastric Mucosa/metabolism , Gene Expression Regulation/drug effects , Helicobacter pylori/pathogenicity , Humans , Interleukin-6/biosynthesis , Interleukin-6/physiology , Interleukin-8/biosynthesis , NF-kappa B/metabolism , Nuclear Proteins/biosynthesis , Nuclear Proteins/physiology , Phenotype , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Protein Binding/immunology , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Stomach Neoplasms/microbiology , Transcription, Genetic/immunology , Tumor Cells, Cultured
3.
Clin Diagn Lab Immunol ; 4(4): 440-6, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9220161

ABSTRACT

For Helicobacter pylori, the hsp60 heat shock protein encoded by hspB is being considered as a potential candidate for subunit vaccines. We investigated the humoral and cellular responses to H. pylori hsp60 and its cross-reactivity with the homologous Mycobacterium bovis p65 protein and autologous human hsp60 protein. H. pylori-infected persons had significantly higher levels than uninfected persons of serum immunoglobulin G antibodies recognizing H. pylori hsp60, but not M. bovis p65 or human hsp60, as determined by enzyme-linked immunosorbent assay. In contrast, immunoblotting demonstrated cross-reactivity of H. pylori hsp60 with human hsp60. T-cell recognition of H. pylori hsp60 was found in both infected and uninfected subjects, and there was no recognition of human hsp60. T cells from infected and uninfected subjects that had been activated in response to H. pylori hsp60 or M. bovis p65 were phenotypically similar but appeared to secrete different levels of gamma interferon and interleukin-10. These results demonstrate an apparent difference in the epitopes recognized by the T and B cells responding to H. pylori hsp60 in H. pylori-infected persons. In contrast to the T-cell responses, which were highly variable in all subjects and showed no recognition of autologous proteins, a specific B-cell response that may have cross-reactivity to human hsp60 is evident in some infected subjects.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Chaperonin 60/immunology , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Adult , Antibodies, Bacterial/blood , Biomarkers , CD4-Positive T-Lymphocytes/chemistry , CD4-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/chemistry , CD8-Positive T-Lymphocytes/microbiology , Cross Reactions , Enzyme-Linked Immunosorbent Assay , HLA-DR Antigens/immunology , Humans , Immunization , Immunoblotting , Immunophenotyping , Interferon-gamma/immunology , Interleukin-10/immunology , Receptors, Interleukin-2/immunology
4.
Mol Microbiol ; 18(5): 867-76, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8825091

ABSTRACT

Approximately 60% of Helicobacter pylori strains are cagA+ and this genotype is more frequently associated with duodenal ulcer disease. Although most wild-type cagA+ strains are both cytotoxigenic and induce enhanced Interleukin-8 (IL-8) secretion in gastric epithelial cells, isogenic cagA- mutants retain full activity in these assays; thus, cagA appears to be a marker of enhanced virulence. Delineation of the nucleotide sequence of a 4 kb region upstream of cagA allowed the identification of 966 bp (picA) and 2655 bp (picB) open reading frames encoding 36 kDa and 101 kDa polypeptides, respectively. picA and picB constitute an operon in opposite orientation to cagA. The deduced picB product showed significant homology (26% identity and 50% similarity) with the Bordetella pertussis toxin secretion protein (PtlC). Of 55 H. pylori clinical isolates, the picA and picB segment was conserved exclusively in cagA+ strains and present in all isolates from patients with duodenal ulceration, versus 59% of isolates from patients with gastritis alone (P = 0.01). Using gene-replacement techniques, we constructed picA and picB mutant H. pylori strains and demonstrated that the picB gene product is involved in the induction of IL-8 expression in gastric epithelial cells. Further, Northern blot hybridization and RT-PCR data showed that picA and picB are co-transcribed and an insertional mutation in picA ablates picB expression. These studies indicate a role of picA and picB in the induction of an inflammatory response in gastric epithelial cells either directly or by enabling secretion of an unidentified product, and suggest a mechanism for the overrepresentation of strains possessing these genes in patients with peptic ulceration.


Subject(s)
Bacterial Proteins/genetics , Gastric Mucosa/metabolism , Helicobacter pylori/genetics , Interleukin-8/genetics , Pertussis Toxin , Virulence Factors, Bordetella/genetics , Amino Acid Sequence , Bacterial Proteins/metabolism , Base Sequence , Blotting, Northern , Conserved Sequence , DNA Primers , Interleukin-8/biosynthesis , Molecular Sequence Data , Mutation , Operon , Polymerase Chain Reaction , Sequence Homology, Amino Acid
5.
Infect Immun ; 63(5): 1681-87, 1995 May.
Article in English | MEDLINE | ID: mdl-7729872

ABSTRACT

Gastric infection with Helicobacter pylori activates a mucosal inflammatory response by mononuclear cells and neutrophils that includes expression of cytokines interleukin-1 beta (IL-1 beta), IL-6, tumor necrosis factor alpha, and IL-8. In this study, we analyzed the IL-8 response of human gastric cancer cell lines (Kato III, AGS, and MKN28) to H. pylori infection in vitro. IL-8 mRNA expression was detected by reverse transcription-PCR amplification of RNA extracted from epithelial cells after incubation with different H. pylori wild-type and mutant strains, and IL-8 secretion was measured by an enzyme-linked immunosorbent assay. Exposure to viable H. pylori induced IL-8 mRNA and protein synthesis in all three gastric cell lines but not in nongastric epithelial cell lines. Heat-killed H. pylori and a crude cytotoxin preparation did not induce significant IL-8 secretion. IL-8 mRNA peaked between 2 and 4 h postinfection, and IL-8 protein production was maximal 24 h postinfection. Exposure of gastric carcinoma cells to other gastrointestinal bacteria, such as Pseudomonas aeruginosa, Campylobacter jejuni, and Escherichia coli, but not Campylobacter fetus, induced IL-8 synthesis. Wild-type strains that expressed the vacuolating cytotoxin (Tox+) and a cytotoxin-associated gene (cagA) product (CagA+) induced significantly more IL-8 than did CagA- Tox- strains. However, there was no decrease in IL-8 induction by isogenic mutants of CagA-, Tox-, or Cag- Tox- strains or by a mutant lacking the urease subunits. These results indicate that exposure to H. pylori and other gram-negative organisms that do not colonize the gastric mucosa induces IL-8 production by gastric carcinoma cells in vitro. Although the CagA+ Tox+ phenotype of H. pylori is associated with enhanced IL-8 production by gastric cell lines, other bacterial constituents are clearly essential.


Subject(s)
Antigens, Bacterial , Gastric Mucosa/immunology , Helicobacter pylori/immunology , Interleukin-8/biosynthesis , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Base Sequence , Cytotoxins/genetics , Cytotoxins/metabolism , Epithelium/metabolism , Gene Expression , Humans , Interleukin-8/genetics , Molecular Sequence Data , Mutation , RNA, Messenger/analysis , Species Specificity , Stomach Neoplasms , Tissue Distribution , Tumor Cells, Cultured
6.
Clin Exp Immunol ; 97(1): 126-32, 1994 Jul.
Article in English | MEDLINE | ID: mdl-8033409

ABSTRACT

Helicobacter pylori is a major cause of chronic antral gastritis and peptic ulcer disease. Further definition is needed of the factors that determine whether infected individuals remain asymptomatic, or ultimately develop ulceration of the mucosa or transformation to malignancy. To explore the possibility that host response to H. pylori may play a role in the outcome of this infection, we have examined humoral and cellular recognition of several H. pylori proteins by seropositive and seronegative persons. A complex mixture of water-extractable cell proteins, which did not include lipopolysaccharide (LPS), was recognized by serum antibodies only in seropositive or infected individuals. IgG from seropositive subjects also bound to urease and to a heat shock protein (hsp)60 that is homologous to the 65-kD mycobacterial heat shock protein, while sera from uninfected individuals were negative. Although antibody responses to these antigens were restricted to seropositive subjects, T cell recognition of the same proteins was found in both seropositive and seronegative subjects. The water extract of H. pylori stimulated peripheral blood mononuclear cells (PBMC) from all subjects, while purified proteins activated lymphocytes of only some seropositive and seronegative subjects. PBMC that were activated by the H. pylori hsp60 did not respond to the autologous human p60 heat shock protein. These results demonstrate that, in contrast to antibody responses, T cell recognition of H. pylori proteins may occur in non-infected persons. In addition, the data suggest that in these subjects, peripheral lymphocytes that are activated by bacterial heat shock proteins do not mediate tissue damage by recognition of human heat shock homologues.


Subject(s)
Antibodies, Bacterial/biosynthesis , Bacterial Proteins/immunology , Helicobacter pylori/immunology , Immunity, Cellular , Adult , Aged , Antigens, Bacterial/isolation & purification , Autoantigens , B-Lymphocytes/immunology , Female , Heat-Shock Proteins/immunology , Humans , In Vitro Techniques , Lymphocyte Activation , Male , Middle Aged , T-Lymphocytes/immunology
7.
J Leukoc Biol ; 51(6): 579-85, 1992 Jun.
Article in English | MEDLINE | ID: mdl-1613394

ABSTRACT

Alveolar macrophages (AMs) are important in the host response to aerogenous pulmonary bacterial infections, such as Pasteurella haemolytica-induced pneumonia in cattle. Previous work has shown that AMs enhance P. haemolytica-mediated pulmonary endothelial cell (EC) damage in vitro. The purpose of this study was to determine the mechanism of AM-enhanced EC damage using an in vitro AM-EC coculture system consisting of AMs cultured on culture plate insert membranes and ECs in the underlying chamber. The addition of lipopolysaccharide (LPS) to the culture plate insert chamber resulted in EC damage indicated by 51Cr release, which was enhanced in the presence of AMs. To determine the role of AM-secreted cytokines, recombinant human interleukin 1 alpha (IL-1) or tumor necrosis factor alpha (TNF) was added to ECs simultaneously with varying concentrations of LPS. Although TNF and IL-1 alone had only marginal toxic effects on ECs, the simultaneous treatment of TNF or IL-1 with LPS greatly increased the LPS cytotoxic effect on ECs. In addition, IL-1 receptor antagonist eliminated the IL-1 enhancement of LPS-mediated EC toxicity. These results suggest that macrophage-secreted cytokines synergistically enhance LPS-mediated pulmonary EC damage.


Subject(s)
Endothelium, Vascular/drug effects , Interleukin-1/pharmacology , Lipopolysaccharides/toxicity , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cattle , Cells, Cultured , Drug Synergism , Endothelium, Vascular/pathology , Macrophages, Alveolar/physiology
8.
Infect Immun ; 60(5): 1946-51, 1992 May.
Article in English | MEDLINE | ID: mdl-1563786

ABSTRACT

Helicobacter pylori is associated with gastritis and peptic ulcer disease in humans. We have identified a homolog of the chaperonin cpn60 family of heat shock proteins in H. pylori, referred to as Hp54K. Hp54K, purified from water-extractable H. pylori proteins, migrated as a single band at 54 kDa by sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Its native molecular mass was 740 kDa; thus, Hp54K apparently comprises a 14-mer. The N-terminal 33 residues of Hp54K exhibited 60.6, 57.6, 54.5, 54.5, 51.5, and 51.5% identity with corresponding sequences in the following cpn60 homologs: HtpB (Legionella pneumophila), P1 (human mitochondria), GroEL (Escherichia coli), BA60K (Brucella abortus), HypB (Chlamydia trachomatis), and the 65-kDa immunodominant protein of Mycobacterium bovis BCG, respectively. Hp54K was the only protein recognized in whole-cell preparations of H. pylori by immunoblotting using monospecific antisera against cpn60 homologs from L. pneumophila, E. coli, C. trachomatis, and M. bovis BCG. Antiserum against Hp54K recognized proteins with molecular masses of 50 to 60 kDa in a large number of gram-negative bacteria, consistent with the known highly conserved nature of cpn60 proteins. Hp54K is a major protein and is immunogenic in humans infected with H. pylori. Thus, Hp54K shares many similarities with known cpn60 homologs. On the basis of the proposed role of other cpn60 proteins in induction of chronic inflammation, immune cross-reactivity between Hp54K and gastric tissue may provide an important link between H. pylori infection and gastritis.


Subject(s)
Heat-Shock Proteins/isolation & purification , Helicobacter pylori/chemistry , Amino Acid Sequence , Cross Reactions , Heat-Shock Proteins/immunology , Molecular Sequence Data
9.
J Infect Dis ; 165(4): 651-7, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1552194

ABSTRACT

Pasteurella haemolytica, the cause of fibrinous pleuropneumonia in cattle, produces extensive microvascular endothelial cell damage. This study investigated endothelial cell-leukocyte interactions by using a Millicell coculture assay system that modeled the bovine pulmonary alveolar defense system and showed that P. haemolytica-mediated endothelial cell damage was enhanced by the presence of alveolar macrophages, presumably due to soluble alveolar macrophage products. The alveolar macrophage-enhanced endothelial cell damage occurred regardless of the presence of anti-P. haemolytica immune serum; however, neutrophils and immune serum effectively prevented endothelial cell damage. These results suggest that alveolar macrophages are ineffective in controlling P. haemolytica growth and actually promote endothelial cell damage.


Subject(s)
Endothelium, Vascular/microbiology , Macrophages, Alveolar/immunology , Mannheimia haemolytica/physiology , Neutrophils/immunology , Animals , Cattle , Cells, Cultured , Endothelium, Vascular/pathology , Immune Sera/immunology , Mannheimia haemolytica/immunology , Pulmonary Artery
SELECTION OF CITATIONS
SEARCH DETAIL
...