Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Seth R. Ogden is active.

Publication


Featured researches published by Seth R. Ogden.


Gastroenterology | 2009

Helicobacter pylori Dysregulation of Gastric Epithelial Tight Junctions by Urease-Mediated Myosin II Activation

Lydia E. Wroblewski; Le Shen; Seth R. Ogden; Judith Romero–Gallo; Lynne A. Lapierre; Dawn A. Israel; Jerrold R. Turner; Richard M. Peek

BACKGROUND & AIMS Helicobacter pylori-induced gastritis predisposes to the development of gastric cancer. Increased epithelial tight junction permeability and alterations in apical-junctional complexes are also associated with an increased risk of carcinogenesis. Phosphorylation of myosin regulatory light chain (MLC) by MLC kinase (MLCK) regulates tight junction function. We determined whether MLCK was activated by H pylori and defined the mechanisms through which such activation dysregulates gastric epithelial barrier function. METHODS MKN28 gastric epithelial cells were cocultured with the H pylori cag(+) strain 60190 or cagA(-), cagE(-), ureB(-), or vacA(-) mutants. MLC phosphorylation and barrier integrity were determined by immunoblot analysis and transepithelial electrical resistance measurements, respectively. Localization of the tight junction protein occludin was determined by immunocytochemistry in MKN28 cells and INS-GAS mice. RESULTS H pylori induced a progressive loss of barrier function that was attenuated by inactivation of ureB, but not cagA, cagE, or vacA. Reductions in transepithelial electrical resistance were also dependent on functional urease activity. H pylori increased MLC phosphorylation in epithelial monolayers; this was significantly decreased by inhibition of MLCK or Rho kinase or by loss of UreB. H pylori infection of either cultured monolayers or hypergastrinemic INS-GAS mice induced occludin endocytosis, reflecting cytoskeletally mediated disruption of tight junctions. CONCLUSIONS H pylori increases MLC phosphorylation, occludin internalization and barrier dysfunction in gastric epithelial cells. This process requires functional urease activity and is independent of the cag pathogenicity island or VacA. These data provide new insights into the mechanisms by which H pylori disrupts gastric barrier function.


Gastroenterology | 2010

Regulation of p53 Tumor Suppressor by Helicobacter pylori in Gastric Epithelial Cells

Jinxiong Wei; Toni Nagy; Anna Vilgelm; Elena Zaika; Seth R. Ogden; Judith Romero–Gallo; Maria B. Piazuelo; Pelayo Correa; Mary Kay Washington; Wael El–Rifai; Richard M. Peek; Alexander Zaika

BACKGROUND & AIMS Infection with the gastric mucosal pathogen Helicobacter pylori is the strongest identified risk factor for distal gastric cancer. These bacteria colonize a significant part of the worlds population. We investigated the molecular mechanisms of p53 regulation in H pylori-infected cells. METHODS Mongolian gerbils were challenged with H pylori and their gastric tissues were analyzed by immunohistochemistry and immunoblotting with p53 antibodies. Gastric epithelial cells were co-cultured with H pylori and the regulation of p53 was assessed by real-time polymerase chain reaction, immunoblotting, immunofluorescence, and cell survival assays. Short hairpin RNA and dominant-negative mutants were used to inhibit activities of Human Double Minute 2 (HDM2) and AKT1 proteins. RESULTS We found that in addition to previously reported up-regulation of p53, H pylori can also negatively regulate p53 by increasing ubiquitination and proteasomal degradation via activation of the serine/threonine kinase AKT1, which phosphorylates and activates the ubiquitin ligase HDM2. These effects were mediated by the bacterial virulence factor CagA; ectopic expression of CagA in gastric epithelial cells increased phosphorylation of HDM2 along with the ubiquitination and proteasomal degradation of p53. The decrease in p53 levels increased survival of gastric epithelial cells that had sustained DNA damage. CONCLUSIONS H pylori is able to inhibit the tumor suppressor p53. H pylori activates AKT1, resulting in phosphorylation and activation of HDM2 and subsequent degradation of p53 in gastric epithelial cells. H pylori-induced dysregulation of p53 is a potential mechanism by which the microorganism increases the risk of gastric cancer in infected individuals.


Molecular Biology of the Cell | 2008

p120 and Kaiso Regulate Helicobacter pylori-induced Expression of Matrix Metalloproteinase-7

Seth R. Ogden; Lydia E. Wroblewski; Christiane Weydig; Judith Romero-Gallo; Daniel P. O'Brien; Dawn A. Israel; Uma Krishna; Barbara Fingleton; Albert B. Reynolds; Silja Wessler; Richard M. Peek

Helicobacter pylori is the strongest known risk factor for gastric adenocarcinoma, yet only a fraction of infected persons develop cancer. One H. pylori constituent that augments disease risk is the cytotoxin-associated gene (cag) pathogenicity island, which encodes a secretion system that translocates bacterial effector molecules into host cells. Matrix metalloproteinase (MMP)-7, a member of a family of enzymes with tumor-initiating properties, is overexpressed in premalignant and malignant gastric lesions, and H. pylori cag(+) strains selectively increase MMP-7 protein levels in gastric epithelial cells in vitro and in vivo. We now report that H. pylori-mediated mmp-7 induction is transcriptionally regulated via aberrant activation of p120-catenin (p120), a component of adherens junctions. H. pylori increases mmp-7 mRNA levels in a cag- and p120-dependent manner and induces translocation of p120 to the nucleus in vitro and in a novel ex vivo gastric gland culture system. Nuclear translocation of p120 in response to H. pylori relieves Kaiso-mediated transcriptional repression of mmp-7, which is implicated in tumorigenesis. These results indicate that selective and coordinated induction of mmp-7 expression by H. pylori cag(+) isolates may explain in part the augmentation in gastric cancer risk associated with these strains.


Gastroenterology | 2011

β-Catenin and p120 Mediate PPARδ-Dependent Proliferation Induced by Helicobacter pylori in Human and Rodent Epithelia

Toni Nagy; Lydia E. Wroblewski; Dingzhi Wang; M. Blanca Piazuelo; Alberto G. Delgado; Judith Romero–Gallo; Jennifer M. Noto; Dawn A. Israel; Seth R. Ogden; Pelayo Correa; Timothy L. Cover; Richard M. Peek

BACKGROUND & AIMS Colonization of gastric mucosa by Helicobacter pylori leads to epithelial hyperproliferation, which increases the risk for gastric adenocarcinoma. One H pylori virulence locus associated with cancer risk, cag, encodes a secretion system that transports effectors into host cells and leads to aberrant activation of β-catenin and p120-catenin (p120). Peroxisome proliferator-activated receptor (PPAR)δ is a ligand-activated transcription factor that affects oncogenesis in conjunction with β-catenin. We used a carcinogenic H pylori strain to define the role of microbial virulence constituents and PPARδ in regulating epithelial responses that mediate development of adenocarcinoma. METHODS Gastric epithelial cells or colonies were co-cultured with the H pylori cag(+) strain 7.13 or cagE(-), cagA(-), soluble lytic transglycosylase(-), or cagA(-)/soluble lytic transglycosylase(-) mutants. Levels of PPARδ and cyclin E1 were determined by real-time, reverse-transcription polymerase chain reaction, immunoblot analysis, or immunofluorescence microscopy; proliferation was measured in 3-dimensional culture. PPARδ and Ki67 expression were determined by immunohistochemical analysis of human biopsies and rodent gastric mucosa. RESULTS H pylori induced β-catenin- and p120-dependent expression and activation of PPARδ in gastric epithelial cells, which were mediated by the cag secretion system substrates CagA and peptidoglycan. H pylori stimulated proliferation in vitro, which required PPARδ-mediated activation of cyclin E1; H pylori did not induce expression of cyclin E1 in a genetic model of PPARδ deficiency. PPARδ expression and proliferation in rodent and human gastric tissue was selectively induced by cag(+) strains and PPARδ levels normalized after eradication of H pylori. CONCLUSIONS The H pylori cag secretion system activates β-catenin, p120, and PPARδ, which promote gastric epithelial cell proliferation via activation of cyclin E1. PPARδ might contribute to gastric adenocarcinoma development in humans.


Journal of Biological Chemistry | 2006

The role of decay-accelerating factor as a receptor for Helicobacter pylori and a mediator of gastric inflammation.

Daniel P. O'Brien; Dawn A. Israel; Uma Krishna; Judith Romero-Gallo; John G. Nedrud; M. Edward Medof; Feng Lin; Raymond W. Redline; Douglas M. Lublin; Bogdan Nowicki; Aime T. Franco; Seth R. Ogden; Amanda D. Williams; D. Brent Polk; Richard M. Peek

Persistent gastritis induced by Helicobacter pylori is the strongest known risk factor for peptic ulcer disease and distal gastric adenocarcinoma, a process for which adherence of H. pylori to gastric epithelial cells is critical. Decay-accelerating factor (DAF), a protein that protects epithelial cells from complement-mediated lysis, also functions as a receptor for several microbial pathogens. In this study, we investigated whether H. pylori utilizes DAF as a receptor and the role of DAF within H. pylori-infected gastric mucosa. In vitro studies showed that H. pylori adhered avidly to Chinese hamster ovary cells expressing human DAF but not to vector controls. In H. pylori, disruption of the virulence factors vacA, cagA, and cagE did not alter adherence, but deletion of DAF complement control protein (CCP) domains 1-4 or the heavily O-glycosylated serine-threonine-rich COOH-terminal domain reduced binding. In cultured gastric epithelial cells, H. pylori induced transcriptional up-regulation of DAF, and genetic deficiency of DAF attenuated the development of inflammation among H. pylori-infected mice. These results indicate that DAF may regulate H. pylori-epithelial cell interactions relevant to pathogenesis.


Journal of Biological Chemistry | 2008

Regulation of the Helicobacter pylori Cellular Receptor Decay-accelerating Factor

Daniel P. O'Brien; Judith Romero-Gallo; Barbara G. Schneider; Rupesh Chaturvedi; Alberto G. Delgado; Elizabeth J. Harris; Uma Krishna; Seth R. Ogden; Dawn A. Israel; Keith T. Wilson; Richard M. Peek

Chronic gastritis induced by Helicobacter pylori is the strongest known risk factor for peptic ulceration and distal gastric cancer, and adherence of H. pylori to gastric epithelial cells is critical for induction of inflammation. One H. pylori constituent that increases disease risk is the cag pathogenicity island, which encodes a secretion system that translocates bacterial effector molecules into host cells. Decay-accelerating factor (DAF) is a cellular receptor for H. pylori and a mediator of the inflammatory response to this pathogen. H. pylori induces DAF expression in human gastric epithelial cells; therefore, we sought to define the mechanism by which H. pylori up-regulates DAF and to extend these findings into a murine model of H. pylori-induced injury. Co-culture of MKN28 gastric epithelial cells with the wild-type H. pylori cag+ strain J166 induced transcriptional expression of DAF, which was attenuated by disruption of a structural component of the cag secretion system (cagE). H. pylori-induced expression of DAF was dependent upon activation of the p38 mitogen-activated protein kinase pathway but not NF-κB. Hypergastrinemic INS-GAS mice infected with wild-type H. pylori demonstrated significantly increased DAF expression in gastric epithelium versus uninfected controls or mice infected with an H. pylori cagE- isogenic mutant strain. These results indicate that H. pylori cag+ strains induce up-regulation of a cognate cellular receptor in vitro and in vivo in a cag-dependent manner, representing the first evidence of regulation of an H. pylori host receptor by the cag pathogenicity island.


Cancer Research | 2010

Matrix Metalloproteinase-7 and Premalignant Host Responses in Helicobacter pylori–Infected Mice

Seth R. Ogden; Jennifer M. Noto; Shannon Sedberry Allen; Dilan A. Patel; Judith Romero-Gallo; M. Kay Washington; Barbara Fingleton; Dawn A. Israel; Nuruddeen D. Lewis; Keith T. Wilson; Rupesh Chaturvedi; Zhiguo Zhao; Yu Shyr; Richard M. Peek

Helicobacter pylori-induced gastritis is the strongest singular risk factor for gastric adenocarcinoma. Matrix metalloproteinase-7 (MMP-7) is a proteolytic enzyme that can modify the intestinal microbial replicative niche as well as affect tumorigenesis, and H. pylori stimulates expression of MMP-7 in gastric epithelial cells in vitro. Utilizing a transgenic murine model of H. pylori-mediated injury, our experiments now show that gastric inflammation is increased within the context of MMP-7 deficiency, which involves both Th1- and Th17-mediated pathways. Enhanced gastritis in H. pylori-infected mmp-7-/- mice is strongly linked to accelerated epithelial cellular turnover. However, more severe inflammation and heightened proliferation and apoptosis are not dependent on MMP-7-mediated bacterial eradication. Collectively, these studies indicate that H. pylori-mediated induction of MMP-7 may serve to protect the gastric mucosa from pathophysiologic processes that promote carcinogenesis.


Gastroenterology | 2009

1042 Helicobacter pylori Transactivation of EGFR Induces COX-2 Expression in Gastric Epithelial Cells

Stuart S. Hobbs; Seth R. Ogden; Richard M. Peek; Robert H. Whitehead; Peter J. Dempsey; D. Brent Polk

ectodomain shedding of receptor tyrosine kinase ligands such as heparin-binding (HB)-EGF. We tested the hypothesis that H. pylori CagL displaces ADAM17 from α5β1 integrin and that increased ligand shedding by activated ADAM17 induces receptor tyrosine kinase signaling to repress HKα promoter activity. Gastric epithelial cells (AGS) were inoculated with wild-type (wt) H. pylori or a cagL-deficient isogenic mutant (P12ΔcagL). H. pylori wt induced a 3-fold increase in ADAM17 activity which declined to 1.2-fold in AGS cells subjected to siRNA-mediated ADAM17 knock-down. In contrast, H. pylori P12ΔcagL stimulated ADAM17 activity by only 1.2-fold. Immunoprecipitation of wt H. pylori-infected AGS cell lysates with ADAM17 antibody followed by β1 integrin immunoblotting showed 50% attenuation of ADAM17/β1 complex compared to control. ELISA measurements of AGS cellconditioned media showed that wt H. pylori infection stimulated HB-EGF shedding 7.3-fold compared to uninfected controls, while P12ΔcagL infection caused only 2.7-fold stimulation. AGS cells transiently-transfected with a 2179 bp HKα promoter-reporter construct showed dose-dependent (up to 50%) inhibition of HKα promoter activity when incubated with recombinant ADAM17. AGS cell HKα promoter activity was also repressed 62% by infection with wt H. pylori, and was unaffected by P12ΔcagL infection. siRNA-mediated knock-down of ADAM17 in transiently-transfected AGS cells abrogated wt H. pylori-mediated repression of HKα promoter. Finally, both HKα mRNA (RT-PCR assay) and HKα protein content (Immunoblotting assay) of human gastric biopsies were down-regulated 13-23 fold and 70% respectively by wt H. pylori inoculation; both HKα mRNA and protein were unaffected by inoculation with P12ΔcagL. The data are consistent with H. pylori up-regulating gastric epithelial cell ADAM17 activity by CagL-mediated dissociation of ADAM17 from α5β1 integrin. Consequent release of HB-EGF activates a receptor tyrosine kinase signaling cascade culminating in NF-κB p50 homodimer-mediated down-regulation of HKα gene expression. (Supported by NIH DK070054 and ACS-IRG to M.G.)


Gastroenterology | 2010

882 Selective Activation of PPARδ by Helicobacter pylori cag Island Substrates Regulates Epithelial Cell Hyperproliferation

Toni Nagy; Dingzhi Wang; Lydia E. Wroblewski; Seth R. Ogden; Shannon Sedberry Allen; Dawn A. Israel; Richard M. Peek


Gastroenterology | 2009

892 Helicobacter pylori Reciprocally Alter Expression of JAM-A and Claudin-7 in Gastric Epithelial Cells

Lydia E. Wroblewski; Seth R. Ogden; Toni Nagy; Shannon Sedberry Allen; Judith Romero-Gallo; Dawn A. Israel; Richard M. Peek

Collaboration


Dive into the Seth R. Ogden's collaboration.

Top Co-Authors

Avatar

Richard M. Peek

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Judith Romero-Gallo

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Toni Nagy

Vanderbilt University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge