Network


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

Hotspot


Dive into the research topics where Keith T. Wilson is active.

Publication


Featured researches published by Keith T. Wilson.


Clinical Microbiology Reviews | 2010

Helicobacter pylori and Gastric Cancer: Factors That Modulate Disease Risk

Lydia E. Wroblewski; Richard M. Peek; Keith T. Wilson

SUMMARY Helicobacter pylori is a gastric pathogen that colonizes approximately 50% of the worlds population. Infection with H. pylori causes chronic inflammation and significantly increases the risk of developing duodenal and gastric ulcer disease and gastric cancer. Infection with H. pylori is the strongest known risk factor for gastric cancer, which is the second leading cause of cancer-related deaths worldwide. Once H. pylori colonizes the gastric environment, it persists for the lifetime of the host, suggesting that the host immune response is ineffective in clearing this bacterium. In this review, we discuss the host immune response and examine other host factors that increase the pathogenic potential of this bacterium, including host polymorphisms, alterations to the apical-junctional complex, and the effects of environmental factors. In addition to host effects and responses, H. pylori strains are genetically diverse. We discuss the main virulence determinants in H. pylori strains and the correlation between these and the diverse clinical outcomes following H. pylori infection. Since H. pylori inhibits the gastric epithelium of half of the world, it is crucial that we continue to gain understanding of host and microbial factors that increase the risk of developing more severe clinical outcomes.


Gastroenterology | 1999

Increased expression and cellular localization of inducible nitric oxide synthase and cyclooxygenase 2 in Helicobacter pylori gastritis

Sidong Fu; Kalathur S. Ramanujam; Annie Wong; George T. Fantry; Cinthia B. Drachenberg; Stephen P. James; Stephen J. Meltzer; Keith T. Wilson

BACKGROUND & AIMS Inducible nitric oxide synthase (iNOS) and cyclooxygenase (COX)-2 are important regulators of mucosal inflammation and epithelial cell growth. To determine the role of iNOS and COX-2 in Helicobacter pylori-induced tissue injury, we compared their gene expression in H. pylori-induced gastritis with that in normal gastric mucosa and in non-H. pylori gastritis. METHODS In 43 patients, we assessed H. pylori infection status, histopathology, messenger RNA (mRNA) and protein expression, and cellular localization of iNOS and COX-2. RESULTS By reverse-transcription polymerase chain reaction (RT-PCR), antral iNOS and COX-2 mRNA expression was absent to low in normal mucosa (n = 10), significantly increased in H. pylori-negative gastritis (n = 13), and even more markedly increased in H. pylori-positive gastritis (n = 20). Increased iNOS and COX-2 levels were confirmed by Northern and Western blot analysis and were both greater in the gastric antrum than in the gastric body of infected patients. Immunohistochemistry also showed increased expression of both genes in H. pylori gastritis: iNOS protein was detected in epithelium, endothelium, and lamina propria inflammatory cells, and COX-2 protein localized to mononuclear and fibroblast cells in the lamina propria. CONCLUSIONS iNOS and COX-2 are induced in H. pylori-positive gastritis and thus may modulate the inflammation and alterations in epithelial cell growth that occur in this disease. Higher levels of iNOS and COX-2 in H. pylori-positive vs. -negative gastritis and in gastric antrum, where bacterial density is greatest, suggest that expression of these genes is a direct response to H. pylori infection.


Proceedings of the National Academy of Sciences of the United States of America | 2001

Helicobacter pylori arginase inhibits nitric oxide production by eukaryotic cells: A strategy for bacterial survival

Alain P. Gobert; David J. McGee; Mahmood Akhtar; George L. Mendz; Jamie C. Newton; Yulan Cheng; Harry L. T. Mobley; Keith T. Wilson

The antimicrobial effect of nitric oxide (NO) is an essential part of innate immunity. The vigorous host response to the human gastric pathogen Helicobacter pylori fails to eradicate the organism, despite up-regulation of inducible NO synthase (iNOS) in the gastric mucosa. Here we report that wild-type strains of H. pylori inhibit NO production by activated macrophages at physiologic concentrations of l-arginine, the common substrate for iNOS and arginase. Inactivation of the gene rocF, encoding constitutively expressed arginase in H. pylori, restored high-output NO production by macrophages. By using HPLC analysis, we show that l-arginine is effectively consumed in the culture medium by wild-type but not arginase-deficient H. pylori. The substantially higher levels of NO generated by macrophages cocultured with rocF-deficient H. pylori resulted in efficient killing of the bacteria, whereas wild-type H. pylori exhibited no loss of survival under these conditions. Killing of the arginase-deficient H. pylori was NO-dependent, because peritoneal macrophages from iNOS−/− mice failed to affect the survival of the rocF mutant. Thus, bacterial arginase allows H. pylori to evade the immune response by down-regulating eukaryotic NO production.


Journal of Clinical Investigation | 2011

Colon-specific delivery of a probiotic-derived soluble protein ameliorates intestinal inflammation in mice through an EGFR-dependent mechanism

Fang Yan; Hanwei Cao; Timothy L. Cover; M. Kay Washington; Yan Shi; LinShu Liu; Rupesh Chaturvedi; Richard M. Peek; Keith T. Wilson; D. Brent Polk

Probiotic bacteria can potentially have beneficial effects on the clinical course of several intestinal disorders, but our understanding of probiotic action is limited. We have identified a probiotic bacteria-derived soluble protein, p40, from Lactobacillus rhamnosus GG (LGG), which prevents cytokine-induced apoptosis in intestinal epithelial cells. In the current study, we analyzed the mechanisms by which p40 regulates cellular responses in intestinal epithelial cells and p40s effects on experimental colitis using mouse models. We show that the recombinant p40 protein activated EGFR, leading to Akt activation. Activation of EGFR by p40 was required for inhibition of cytokine-induced apoptosis in intestinal epithelial cells in vitro and ex vivo. Furthermore, we developed a pectin/zein hydrogel bead system to specifically deliver p40 to the mouse colon, which activated EGFR in colon epithelial cells. Administration of p40-containing beads reduced intestinal epithelial apoptosis and disruption of barrier function in the colon epithelium in an EGFR-dependent manner, thereby preventing and treating DSS-induced intestinal injury and acute colitis. Furthermore, p40 activation of EGFR was required for ameliorating colon epithelial cell apoptosis and chronic inflammation in oxazolone-induced colitis. These data define what we believe to be a previously unrecognized mechanism of probiotic-derived soluble proteins in protecting the intestine from injury and inflammation.


The American Journal of Gastroenterology | 2010

Pathology of Gastric Intestinal Metaplasia: Clinical Implications

Pelayo Correa; M. Blanca Piazuelo; Keith T. Wilson

Intestinal metaplasia (IM) of the gastric mucosa is a relatively frequent precancerous lesion (1). The inclusion of IM in a gastric biopsy pathology report often creates uncertainty for the gastroenterologist about the appropriate management. Although the risk of gastric cancer is increased in the presence of IM, the overall risk of gastric cancer in a patient with IM is extremely low compared with the risk of adenocarcinoma in a patient with Barrett’s esophagus (BE)(2). Although the incidence of gastric cancer is high in certain regions, such as Asia and Latin America, it is much lower in the United States and other Western countries, making it difficult to justify broad surveillance programs. The aims of this article are (i) to assist the clinician in identifying subgroups of patients with IM at increased risk for gastric cancer and (ii) to propose an algorithm for gastric IM management, considering the lack of universally accepted guidelines that can be applied to any population.


Gastroenterology | 1996

Helicobacter pylori stimulates inducible nitric oxide synthase expression and activity in a murine macrophage cell line

Keith T. Wilson; Kalathur S. Ramanujam; Harry L. T. Mobley; Robert Musselman; Stephen P. James; Stephen J. Meltzer

BACKGROUND & AIMS Helicobacter pylori uniquely colonizes the human stomach and produces gastric mucosal inflammation. High-output nitric oxide production by inducible nitric oxide synthase (iNOS) is associated with immune activation and tissue injury. Because mononuclear cells comprise a major part of the cellular inflammatory response to H. pylori infection, the ability of H. pylori to induce iNOS in macrophages was assessed. METHODS H. pylori preparations were added to RAW 264.7 murine macrophages, and iNOS expression was assessed by Northern blot analysis, enzyme activity assay, and NO2- release. RESULTS Both whole H. pylori and French press lysates induced concentration-dependent NO2- production, with peak levels 20-fold above control. These findings were paralleled by marked increases in iNOS messenger RNA and enzyme activity levels. iNOS expression was synergistically increased with interferon gamma, indicating that the H. pylori effect can be amplified by other macrophage-activating factors. Studies of lipopolysaccharide (LPS) content and polymyxin B inhibition of LPS suggested that the H. pylori effect was attributable to both LPS-dependent and -independent mechanisms. CONCLUSIONS iNOS expression in macrophages is activated by highly stable H. pylori products and may play an important role in the pathogenesis of H. pylori-associated gastric mucosal disease.


Oncogene | 2000

Distinct methylation patterns of two APC gene promoters in normal and cancerous gastric epithelia

Takashi Tsuchiya; Gen Tamura; Kiyoshi Sato; Yasushi Endoh; Ken Sakata; Zhe Jin; Teiichi Motoyama; Osamu Usuba; Wataru Kimura; Satoshi Nishizuka; Keith T. Wilson; Stephen P. James; Jing Yin; A. Steven Fleisher; Tongtong Zou; Steven G. Silverberg; Dehe Kong; Stephen J. Meltzer

The adenomatous polyposis coli (APC) tumor suppressor gene is mutationally inactivated in both familial and sporadic forms of colorectal cancers. In addition, hypermethylation of CpG islands in the upstream portion of APC, a potential alternative mechanism of tumor suppressor gene inactivation, has been described in colorectal cancer. Because a subset of both gastric and colorectal cancers display the CpG island methylator phenotype, we hypothesized that epigenetic inactivation of APC was likely to occur in at least some gastric cancers. APC exhibits two forms of transcripts from exons 1A and 1B in the stomach. Therefore, we investigated CpG island methylation in the sequences upstream of exons 1A and 1B, i.e., promoters 1A and 1B, respectively. We evaluated DNAs from 10 gastric cancer cell lines, 40 primary gastric cancers, and 40 matching non-cancerous gastric mucosae. Methylated alleles of promoter 1A were present in 10 (100%) of 10 gastric cancer cell lines, 33 (82.5%) of 40 primary gastric cancers, and 39 (97.5%) of 40 non-cancerous gastric mucosae. In contrast, promoter 1B was unmethylated in all of these same samples. APC transcripts from exon 1A were not expressed in nine of the 10 methylated gastric cancer cell lines, whereas APC transcripts were expressed from exon 1B. Thus, expression from a given promoter correlated well with its methylation status. We conclude that in contrast to the colon, methylation of promoter 1A is a normal event in the stomach; moreover, promoter 1B is protected from methylation in the stomach and thus probably does not participate in this form of epigenetic APC inactivation.


Journal of Immunology | 2002

Helicobacter pylori Induces Macrophage Apoptosis by Activation of Arginase II

Alain P. Gobert; Yulan Cheng; Jian Ying Wang; Jean Luc Boucher; Ramaswamy K. Iyer; Stephen D. Cederbaum; Robert A. Casero; Jamie C. Newton; Keith T. Wilson

Helicobacter pylori infection induces innate immune responses in macrophages, contributing to mucosal inflammation and damage. Macrophage apoptosis is important in the pathogenesis of mucosal infections but has not been studied with H. pylori. NO derived from inducible NO synthase (iNOS) can activate macrophage apoptosis. Arginase competes with iNOS by converting l-arginine to l-ornithine. Since we reported that H. pylori induces iNOS in macrophages, we now determined whether this bacterium induces arginase and the effect of this activation on apoptosis. NF-κB-dependent induction of arginase II, but not arginase I, was observed in RAW 264.7 macrophages cocultured with H. pylori. The time course of apoptosis matched those of both arginase and iNOS activities. Surprisingly, apoptosis was blocked by the arginase inhibitors Nω-hydroxy-l-arginine or Nω-hydroxy-nor-l-arginine, but not by the iNOS inhibitor N-iminoethyl-l-lysine. These findings were confirmed in peritoneal macrophages from iNOS-deficient mice and were not dependent on bacterial-macrophage contact. Ornithine decarboxylase (ODC), which metabolizes l-ornithine to polyamines, was also induced in H. pylori-stimulated macrophages. Apoptosis was abolished by inhibition of ODC and was restored by the polyamines spermidine and spermine. We also demonstrate that arginase II expression is up-regulated in both murine and human H. pylori gastritis tissues, indicating the likely in vivo relevance of our findings. Therefore, we describe arginase- and ODC-dependent macrophage apoptosis, which implicates polyamines in the pathophysiology of H. pylori infection.


Physiological Reviews | 2010

Role of Innate Immunity in Helicobacter pylori-Induced Gastric Malignancy

Richard M. Peek; Chris Fiske; Keith T. Wilson

Helicobacter pylori colonizes the majority of persons worldwide, and the ensuing gastric inflammatory response is the strongest singular risk factor for peptic ulceration and gastric cancer. However, only a fraction of colonized individuals ever develop clinically significant outcomes. Disease risk is combinatorial and can be modified by bacterial factors, host responses, and/or specific interactions between host and microbe. Several H. pylori constituents that are required for colonization or virulence have been identified, and their ability to manipulate the host innate immune response will be the focus of this review. Identification of bacterial and host mediators that augment disease risk has profound ramifications for both biomedical researchers and clinicians as such findings will not only provide mechanistic insights into inflammatory carcinogenesis but may also serve to identify high-risk populations of H. pylori-infected individuals who can then be targeted for therapeutic intervention.


Cancer Research | 2004

Spermine Oxidation Induced by Helicobacter pylori Results in Apoptosis and DNA Damage Implications for Gastric Carcinogenesis

Hangxiu Xu; Rupesh Chaturvedi; Yulan Cheng; Françoise I. Bussière; Mohammad Asim; Micheal D. Yao; Darryn Potosky; Stephen J. Meltzer; Juong G. Rhee; Sung S. Kim; Steven F. Moss; Amy Hacker; Yanlin Wang; Robert A. Casero; Keith T. Wilson

Oxidative stress is linked to carcinogenesis due to its ability to damage DNA. The human gastric pathogen Helicobacter pylori exerts much of its pathogenicity by inducing apoptosis and DNA damage in host gastric epithelial cells. Polyamines are abundant in epithelial cells, and when oxidized by the inducible spermine oxidase SMO(PAOh1) H2O2 is generated. Here, we report that H. pylori up-regulates mRNA expression, promoter activity, and enzyme activity of SMO(PAOh1) in human gastric epithelial cells, resulting in DNA damage and apoptosis. H. pylori-induced H2O2 generation and apoptosis in these cells was equally attenuated by an inhibitor of SMO(PAOh1), by catalase, and by transient transfection with small interfering RNA targeting SMO(PAOh1). Conversely, SMO(PAOh1) overexpression induced apoptosis to the same levels as caused by H. pylori. Importantly, in H. pylori-infected tissues, there was increased expression of SMO(PAOh1) in both human and mouse gastritis. Laser capture microdissection of human gastric epithelial cells demonstrated expression of SMO(PAOh1) that was significantly attenuated by H. pylori eradication. These results identify a pathway for oxidative stress-induced epithelial cell apoptosis and DNA damage due to SMO(PAOh1) activation by H. pylori that may contribute to the pathogenesis of the infection and development of gastric cancer.

Collaboration


Dive into the Keith T. Wilson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

M. Blanca Piazuelo

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Mohammad Asim

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Alain P. Gobert

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Richard M. Peek

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Daniel P. Barry

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Kshipra Singh

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Lori A. Coburn

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Pelayo Correa

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge