Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Legal Med ; 122(6): 489-92, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18688634

ABSTRACT

The evaluation of four pairs of tightly linked chromosome X (ChrX) short tandem repeat (STR)s at Xp22, Xq12, Xq26 and Xq28 led to the creation of the Argus X 8 multiplex amplification kit. These eight STRs are distributed as four closely linked pairs over the entire X-chromosome, and for practical reasons, they are assigned to four linkage groups 1-4. To achieve a further considerable enhancement in discrimination power, we suggest to include additional markers. A recent paper referred to the earlier evaluation of STR clusters at Xq12, Xq26 and Xq28, and here we present the pending data of linkage group 1 at Xp22. The newly established STR updates the Xp22 STR cluster which now presents three polymorphic markers: DXS10148 (PIC = 0.8556), DXS10135 (PIC = 0.9093) and DXS 8378 (PIC = 0.6454). Typing of 398 X-chromosomes provided 278 different and 200 unique haplotypes. All the other haplotypes observed appeared with frequencies in the range between 0.005 and 0.015. Considering this STR triple in the context with the three further triple clusters Xq12, Xq26 and Xq28 published earlier, we announced the development of a next generation of a ChrX STR cluster typing kit.


Subject(s)
Chromosomes, Human, X , DNA Fingerprinting , Haplotypes , Tandem Repeat Sequences , Female , Gene Frequency , Genetic Linkage , Genetic Markers , Humans , Male , Mutation , Polymerase Chain Reaction
2.
Pathol Res Pract ; 203(6): 433-44, 2007.
Article in English | MEDLINE | ID: mdl-17509776

ABSTRACT

Helicobacter pylori infection is related to the development of diverse gastric pathologies, possibly by affecting epithelial junctional complexes that define cell polarity and play an essential role in transepithelial transport and cell-cell adhesion. Using primary gastric epithelial cell cultures, effects of H. pylori on the expression and localization of tight/adherence junction proteins and the resulting morphological changes and migratory capabilities were studied under in vivo-like conditions. Gastric epithelial cells were isolated from biopsies or gastrectomies and maintained in Quantum286 on collagen I-coated culture dishes or cover-slips. Cell cultures were characterized and further analyzed by western blot and immunofluorescent staining for ZO-1, p120ctn, and H. pylori CagA. Morphological changes and migratory response were monitored by time-lapse digital image microscopy. ZO-1 and p120ctn protein expression levels remain unaffected by H. pylori infection. Immunocytochemistry on H. pylori-infected primary cell monolayers focally showed disruption of intercellular ZO-1 staining and accumulation of ZO-1 in small vesicles. H. pylori infection recruited non-phosphorylated p120ctn to perinuclear vesicles. The fraction of phosphorylated p120ctn increased and could be detected in the nucleus, at the cell membrane, and at the leading edge of migrating cells. These alterations, triggered by H. pylori infection, are associated with an elongation phenotype and increased migration.


Subject(s)
Cell Adhesion Molecules/metabolism , Epithelial Cells/metabolism , Gastric Mucosa/metabolism , Helicobacter Infections/metabolism , Helicobacter pylori/isolation & purification , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Pyloric Antrum/metabolism , Tight Junctions/metabolism , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Catenins , Cell Movement , Cell Shape , Cells, Cultured , Epithelial Cells/microbiology , Epithelial Cells/pathology , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Gastric Mucosa/physiopathology , Gene Expression , Helicobacter Infections/genetics , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter Infections/physiopathology , Helicobacter pylori/metabolism , Humans , Immunohistochemistry , Microscopy, Video , Permeability , Phosphorylation , Pyloric Antrum/microbiology , Pyloric Antrum/pathology , Pyloric Antrum/physiopathology , Tight Junctions/pathology , Time Factors , Wound Healing , Zonula Occludens-1 Protein , Delta Catenin
3.
J Biol Chem ; 281(5): 2868-75, 2006 Feb 03.
Article in English | MEDLINE | ID: mdl-16321971

ABSTRACT

Helicobacter pylori colonizes the human gastric epithelium and induces an inflammatory response that is a trigger for gastric carcinogenesis. Matrix metalloproteinases (MMPs) have recently been shown to be up-regulated in gastric epithelial cells infected with H. pylori and might contribute to the pathogenesis of peptic ulcer. The aim of this study was to extend the knowledge about the effect of H. pylori infection on MMP-1 expression by gastric epithelial cells, the kinetics of induction, the pathogenetic properties of the bacterium, and the intracellular signaling pathways required for MMP-1 up-regulation. Expression of MMP-1 was induced more than 10-fold by co-culture of AGS+cells with H. pylori strains carrying the pathogenicity island (PAI). H. pylori strains with mutations in the PAI and a defective type IV secretion system had no effect on MMP-1. Double immunofluorescence revealed strong MMP-1 staining in epithelial cells of gastric biopsies at sites of bacterial attachment. In vitro, MMP-1 is up-regulated by interleukin-1beta and tumor necrosis factor-alpha, but these regulatory mechanisms are not operating in H. pylori infection as shown by inhibitory antibodies. Specific inhibitors of JNK kinase and ERK1/2 kinase were found to suppress the H. pylori-induced MMP-1 expression and activity. AGS cells treated with antisense MMP-1 showed a significantly reduced potential to degrade reconstituted basement membrane. Our results suggest that in gastric epithelial cells, H. pylori up-regulates MMP-1 in a type IV secretion system-dependent manner via JNK and ERK1/2. Induction of MMP-1 is further implicated in complex processes induced by H. pylori, resulting in tissue degradation and remodeling of the gastric mucosa.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Genomic Islands/genetics , Helicobacter pylori/physiology , JNK Mitogen-Activated Protein Kinases/metabolism , Matrix Metalloproteinase 1/metabolism , Stomach/pathology , Epithelium/enzymology , Epithelium/microbiology , Helicobacter pylori/pathogenicity , Humans , Kinetics , Matrix Metalloproteinase 1/genetics , Signal Transduction , Stomach/microbiology , Up-Regulation
4.
J Pathol ; 207(1): 32-42, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16025436

ABSTRACT

Recently, we identified increased cathepsin X expression in H. pylori-infected gastric mucosa. Here, we describe further up-regulation in gastric cancer and report on the role of inflammatory cytokines required for cathepsin X up-regulation in H. pylori-infected gastric mucosa, as well as on consequences for cellular invasion. Biopsy specimens were taken from the antrum, corpus and cardia of H. pylori-infected and non-infected patients. Gastric cancer samples were obtained from patients undergoing gastric surgery. Cathepsin X was detected in gastric mucosa by quantitative real-time RT-PCR, western blotting and immunohistochemistry. Induction of cathepsin X expression in epithelial and inflammatory cells caused by H. pylori infection was tested in in vitro contact and non-contact co-cultures of AGS cells and monocytic cells. Patients with H. pylori gastritis showed significantly higher cathepsin X mRNA (2.5-fold) and protein (1.6-fold) expression than H. pylori-negative patients. Cathepsin X was also up-regulated in gastric cancer (3-12-fold) compared to non-neoplastic mucosa. Cathepsin X was predominantly expressed by macrophages in the mucosal stroma and in glands of the antral mucosa. In addition, tumour cells stained for cathepsin X in 26 (68%) patients with gastric carcinoma. In general, staining was significantly more common (20 vs. 6 patients) and more intense (3.55 vs. 0.83) in intestinal type gastric cancer than in the diffuse type. In vitro cell culture experiments revealed that intercellular signalling between pathogenicity island (PAI)-positive H. pylori-infected epithelial cells and macrophages via soluble factors in the culture medium seems to be responsible for increased expression of cathepsin X in monocytes. Using antisense oligonucleotides, cathepsin X up-regulation was directly associated with higher invasiveness in vitro. Although no correlation of cathepsin X expression and TNM stage was found, our study demonstrates that cathepsin X plays a role not only in the chronic inflammation of gastric mucosa but also in the tumourigenesis of gastric cancer.


Subject(s)
Cathepsins/biosynthesis , Gastritis/enzymology , Helicobacter Infections/enzymology , Helicobacter pylori , Stomach Neoplasms/enzymology , Adult , Aged , Cardia/enzymology , Cathepsin K , Cathepsins/genetics , Cytokines/pharmacology , Female , Gastric Mucosa/enzymology , Gastritis/microbiology , Gene Expression Regulation, Enzymologic/drug effects , Humans , Immunoenzyme Techniques , Male , Middle Aged , Neoplasm Invasiveness , Protein Array Analysis , Pyloric Antrum/enzymology , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Stomach Neoplasms/pathology , Stromal Cells/enzymology , Up-Regulation/drug effects
5.
Biochem Biophys Res Commun ; 322(3): 860-6, 2004 Sep 24.
Article in English | MEDLINE | ID: mdl-15336542

ABSTRACT

The pathogenesis of Helicobacter pylori-associated disorders is strongly dependent on a specialized type IV secretion system (T4SS) encoded by the cag pathogenicity island (PAI). Cytotoxin-associated gene A (CagA) is the only known H. pylori protein translocated into the host cell followed by tyrosine phosphorylation through host protein kinases. H. pylori induces cellular processes which are either PAI- or CagA-dependent (e.g., cell motility), PAI-dependent, but CagA-independent (e.g., interleukin-8 release), or PAI- and CagA-independent (e.g., cyclooxygenase-2 release). Here, we investigated H. pylori strains mutated in single PAI genes of the wild type strain Hp26695 and their effects on cell motility. We found 17 gene products out of 27 PAI genes playing a superordinated role and five PAI-encoded proteins exhibiting a clearly critical role in motogenic host cell responses, whereas the remaining five PAI gene products had no significant influence on the motogenic response in reaction to H. pylori infection. This study clearly demonstrated that H. pylori-induced cell motility and invasive growth involve type IV secretion system-dependent signalling as well as translocated and phosphorylated CagA. These findings reveal a deeper insight in to the meaning of the T4SS of H. pylori for host cell motility.


Subject(s)
Gastric Mucosa/microbiology , Gastric Mucosa/physiology , Helicobacter pylori/pathogenicity , JNK Mitogen-Activated Protein Kinases , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Cell Movement , Cells, Cultured , Enzyme Activation , Gastric Mucosa/metabolism , Helicobacter pylori/genetics , Helicobacter pylori/physiology , Humans , MAP Kinase Kinase 1 , MAP Kinase Kinase 2 , MAP Kinase Kinase 4 , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Protein-Tyrosine Kinases/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...