The long-term goal of our work is to gain insight into mechanisms by which oxidative stress responses to H. pylori infection and other injury to the gastric epithelium lead to human disease including peptic ulcer disease and gastric carcinoma. In response to infection, phagocytes recruited to the gastric mucosa, become activated and generate reactive oxygen species (ROS) and H. pylori infection induces oxidative stress in gastric epithelial cells directly through the generation of ROS. This project has focused on ROS-induced activation of apurinic/apyrimidinic endonuclease (APE1), a multifunctional protein that is the rate-limiting enzyme in DNA base excision repair of oxidative lesions. It is also known as redox factor (Ref)-1 due to its ability to control gene expression by reductively activating transcription factors including activator protein (AP)-1, NF-?B, and p53. Our recent preliminary studies build on the understanding that ROS arises from activation of NADPH oxidase during H. pylori infection. During the course of preliminary studies, we observed that APE1 is not only activated by ROS, but its induction provides a negative feedback on the accumulation of ROS. Data suggest that this feedback is based on novel molecular interactions between APE1 and Rac1. Rac1 is an important regulator of cell function, implicated in the control of bacterial infections and the pathogenesis of chronic inflammatory diseases including IBD. One of its functions is to activate NADPH oxidase which leads to the accumulation of ROS. The hypothesis that APE1 modulates gastric epithelial cell responses by regulating the accumulation of ROS will be tested in the following Specific Aims:
Aim 1 : Define the role of APE1 in ROS accumulation in epithelial cells during H. pylori infection.
Aim 2 : Determine the mechanisms whereby APE1 regulates Rac1 activity.
Aim 3 : Determine the mechanisms whereby APE1 regulates NAPH oxidase. Despite the advances in our understanding of the pathogenesis of H. pylori, the mechanisms by which this infection leads to epithelial cell injury and disease such as malignancy remain poorly understood. The broad objective for the proposed studies is to define a novel molecular mechanism whereby APE1 regulates Rac activation and the generation of ROS through NADPH oxidase. Using H. pylori as a model system, these studies will provide new knowledge of the control of oxidative stress. By the end of 5 years, the expected outcomes and milestones include a definition of the molecular basis for the regulation of ROS accumulation by APE1 including its interaction with Rac1 and NADPH oxidase. This information will have an important positive impact by advancing our understanding of the molecular mechanisms regulating Rac activation that is relevant to diseases in the human GI tract.

Public Health Relevance

Over half the world is infected with Helicobactor pylori bacteria that cause most cases of gastric cancer, the second leading cause of cancer death worldwide. Our laboratory has shown that H. pylori infection leads to increased levels of a vital protein, a purinic/apyrimidinic endonuclease-1 (APE1) that regulates life and death modulate human responses to infection and enhance knowledge of how H. pylori-associated diseases arise with the potential to develop interventions to prevent or treat these conditions.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
2R01DK061769-11A1
Application #
8514362
Study Section
Gastrointestinal Mucosal Pathobiology Study Section (GMPB)
Program Officer
Hamilton, Frank A
Project Start
2001-09-21
Project End
2016-07-31
Budget Start
2013-08-01
Budget End
2014-07-31
Support Year
11
Fiscal Year
2013
Total Cost
$310,000
Indirect Cost
$110,000
Name
University of California San Diego
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
804355790
City
La Jolla
State
CA
Country
United States
Zip Code
92093
Butcher, Lindsay D; den Hartog, Gerco; Ernst, Peter B et al. (2017) Oxidative Stress Resulting From Helicobacter pylori Infection Contributes to Gastric Carcinogenesis. Cell Mol Gastroenterol Hepatol 3:316-322
Das, Lopamudra; Kokate, Shrikant Babanrao; Rath, Suvasmita et al. (2016) ETS2 and Twist1 promote invasiveness of Helicobacter pylori-infected gastric cancer cells by inducing Siah2. Biochem J 473:1629-40
den Hartog, Gerco; Chattopadhyay, Ranajoy; Ablack, Amber et al. (2016) Regulation of Rac1 and Reactive Oxygen Species Production in Response to Infection of Gastrointestinal Epithelia. PLoS Pathog 12:e1005382
Hunt, R H; Camilleri, M; Crowe, S E et al. (2015) The stomach in health and disease. Gut 64:1650-68
Rath, Suvasmita; Das, Lopamudra; Kokate, Shrikant Babanrao et al. (2015) Regulation of Noxa-mediated apoptosis in Helicobacter pylori-infected gastric epithelial cells. FASEB J 29:796-806
Fox, Sarah; Ryan, Kieran A; Berger, Alice H et al. (2015) The role of C1q in recognition of apoptotic epithelial cells and inflammatory cytokine production by phagocytes during Helicobacter pylori infection. J Inflamm (Lond) 12:51
Boland, Brigid S; Widjaja, Christella E; Banno, Asoka et al. (2015) Immunodeficiency and autoimmune enterocolopathy linked to NFAT5 haploinsufficiency. J Immunol 194:2551-60
Nagoya, Hiroyuki; Futagami, Seiji; Shimpuku, Mayumi et al. (2014) Apurinic/apyrimidinic endonuclease-1 is associated with angiogenesis and VEGF production via upregulation of COX-2 expression in esophageal cancer tissues. Am J Physiol Gastrointest Liver Physiol 306:G183-90
Das, Soumita; Sarkar, Arup; Ryan, Kieran A et al. (2014) Brain angiogenesis inhibitor 1 is expressed by gastric phagocytes during infection with Helicobacter pylori and mediates the recognition and engulfment of human apoptotic gastric epithelial cells. FASEB J 28:2214-24
Bhattacharyya, Asima; Chattopadhyay, Ranajoy; Mitra, Sankar et al. (2014) Oxidative stress: an essential factor in the pathogenesis of gastrointestinal mucosal diseases. Physiol Rev 94:329-54

Showing the most recent 10 out of 20 publications