Background/Rationale: Intestinal epithelial barrier integrity is critical for most gastrointestinal (GI) functions including digestion of food, as well as absorption and secretion of nutrients. The gastrointestinal mucosal layer is very vulnerable to injury resulting in loss of barrier integrity, and this occurs very commonly in critically ill Veteran patients in response to surgical stress. Preventing it would likely have a major impact on outcomes and costs for all critically ill Veteran populations, though the molecular controls that regulate barrier integrity are incompletely understood. Current treatment for barrier dysfunction is conservative. This application will explore a promising approach to preserve, protect, and restore epithelial barrier function by manipulating the fundamental regulation of intestinal epithelial cells; while improving our overall understanding of their function and regulation. The bioactive sphingolipid sphingosine-1-phosphate (S1P) has recently demonstrated significant effects on endothelial and epithelial cells in promoting important biological functions, such as survival of injured cells and expression of barrier proteins. We have demonstrated in previously published work and our preliminary data that S1P has significant effects on intestinal epithelial cells and the intestinal barrier; both in physiologic and pathologic conditions, though the details of this mechanistically have not been fully elucidated. Objectives: The primary objective of this study is to demonstrate that S1P protects the intestinal epithelial barrier in both physiological and pathological states with the eventual goal of having a therapeutic use for S1P in critically ill patients. Methods: This proposal includes in vitro studies using cultured intestinal epithelial cells, ex vivo studies of tissues from mice and humans, and in vivo studies in mice. Expected Results: We hypothesize that S1P will have significant effects as both an intracellular 2nd messenger and external receptor ligand to promote barrier integrity in cultured cells and in vivo, during physiological conditions and in pathological states. Status: Cells exposed to S1P show increased levels of barrier proteins and improved barrier function. Cells overexpressing Sphingosine Kinase 1, and expressing higher S1P content, show significantly increased proliferation and a decrease in microRNA (mir) 542 among other findings. Mice exposed to S1P show improved barrier function in control states and during ischemia- reperfusion, and also decreased levels of mir 542. MicroRNA 495 inhibits Sphingosine Kinase 1 and shows decreased proliferation and increased apoptosis. Impact: The proposed research addresses critical mechanisms in maintenance of an intact intestinal mucosal barrier, and in S1P, a potential therapy to preserve this function in critically ill patients. Critical illness, particularly in surgical patients, is very common and a high percentage of patients in the intensive care units incur barrier dysfunction. Despite the medical advances of the past few decades, therapies for these gastrointestinal injuries are, for the most part, conservative as there is no current pharmacologic therapy available. This proposal explores such a pharmacologic therapy that could potentially prevent injury and loss of the intestinal epithelial cells and lead to improved outcomes of critically ill Veteran patients.

Public Health Relevance

Veteran surgical patients commonly suffer critical illness, and a high percentage of these patients in the intensive care units incur stress-related injuries to their very vulnerable intestinal lining. These injuries can result in malnutrition, overwhelming bacterial infection, and death. Despite the medical advances of the past few decades, therapies for these stress injuries are, for the most part, conservative, as there is no current pharmacologic therapy available. This proposal explores the use of a new molecule that could potentially prevent injury and loss of the intestinal epithelial cells, and lead to improved outcomes of critically ill Veteran patients.

Agency
National Institute of Health (NIH)
Institute
Veterans Affairs (VA)
Type
Non-HHS Research Projects (I01)
Project #
2I01BX000113-08
Application #
9562680
Study Section
Surgery (SURG)
Project Start
2009-04-01
Project End
2022-09-30
Budget Start
2018-10-01
Budget End
2019-09-30
Support Year
8
Fiscal Year
2019
Total Cost
Indirect Cost
Name
Baltimore VA Medical Center
Department
Type
DUNS #
796532609
City
Baltimore
State
MD
Country
United States
Zip Code
21201
Hansraj, Natasha Z; Xiao, Lan; Wu, Jing et al. (2016) Posttranscriptional regulation of 14-3-3? by RNA-binding protein HuR modulating intestinal epithelial restitution after wounding. Physiol Rep 4:
Phatak, P; Byrnes, K A; Mansour, D et al. (2016) Overexpression of miR-214-3p in esophageal squamous cancer cells enhances sensitivity to cisplatin by targeting survivin directly and indirectly through CUG-BP1. Oncogene 35:2087-97
Byrnes, Kimberly A; Phatak, Pornima; Mansour, Daniel et al. (2016) Overexpression of miR-199a-5p decreases esophageal cancer cell proliferation through repression of mitogen-activated protein kinase kinase kinase-11 (MAP3K11). Oncotarget 7:8756-70
Chung, Hee Kyoung; Chen, Yu; Rao, Jaladanki N et al. (2015) Transgenic Expression of miR-222 Disrupts Intestinal Epithelial Regeneration by Targeting Multiple Genes Including Frizzled-7. Mol Med 21:676-687
Ouyang, Miao; Su, Weijie; Xiao, Lan et al. (2015) Modulation by miR-29b of intestinal epithelium homoeostasis through the repression of menin translation. Biochem J 465:315-23
Rathor, Navneeta; Chung, Hee K; Wang, Shelley R et al. (2014) Caveolin-1 enhances rapid mucosal restitution by activating TRPC1-mediated Ca2+ signaling. Physiol Rep 2:
Chung, Hee Kyoung; Rao, Jaladanki N; Zou, Tongtong et al. (2014) Jnk2 deletion disrupts intestinal mucosal homeostasis and maturation by differentially modulating RNA-binding proteins HuR and CUGBP1. Am J Physiol Cell Physiol 306:C1167-75
Cao, Shan; Xiao, Lan; Rao, Jaladanki N et al. (2014) Inhibition of Smurf2 translation by miR-322/503 modulates TGF-?/Smad2 signaling and intestinal epithelial homeostasis. Mol Biol Cell 25:1234-43
Rathor, Navneeta; Zhuang, Ran; Wang, Jian-Ying et al. (2014) Src-mediated caveolin-1 phosphorylation regulates intestinal epithelial restitution by altering Ca(2+) influx after wounding. Am J Physiol Gastrointest Liver Physiol 306:G650-8