For unknown reasons, the incidence of allergies and asthma continues to increase in the US. It is increasingly clear that asthma is a heterogeneous disease with differing endotypes which suggest discrete pathophysiology. In humans, asthma exists in allergic and non-allergic forms. It is an unmet need in the field to better understand ho different mechanisms contribute to asthma endotypes. Genome wide array analysis has identified many asthma relevant genes. However, due to poor correlation between steady state mRNA levels and protein, transcriptomic approaches may overlook critical genes. Posttranscriptional gene regulation by RNA binding proteins (RBPs) and microRNAs (miRNAs) are increasingly recognized as important control mechanisms for proinflammatory genes. RBPs, such as HuR (elav1) which bind to AU-rich elements (AREs) play critical roles in coordinately regulating proinflammatory genes in asthma by stabilizing target gene mRNAs and increasing translatability. Methods used by our lab and others, called RNA immunoprecipitation applied to microarrays (RIP-Chip) have identified how RBPs are coordinately regulating inflammation. Posttranscriptional gene regulation plays an important role in CD4+ T differentiation, yet these processes are poorly understood. Our long term goal is to understand posttranscriptional gene regulation in airway inflammation. The objective of this application, which is our next step in pursuit of that goal, is to understand how HuR is regulating key molecules, such as Th2/Th17 cytokines and IL-2 during allergen challenge. The central esis is that the RBP, HuR, is permissive for development of CD4+ Th2 mediated allergic airway inflammation and required for normal IL-2 homeostatic expression. The rationale for this proposal is that our work has demonstrated that HuR controls both Th2 and Th17 differentiation. HuR KO mice do not develop airway inflammation due to suppression of Th2 cytokine production and do not have the ability to turn off IL-2 expression following T cell activation. Understanding posttranscriptional mechanisms of IL-2 and Th2/Th17 cytokine gene regulation will allow the field to modulate and affect outcomes of inflammatory responses in allergen driven asthma and also perhaps aid in better defining the heterogeneity amongst asthma endotypes. We plan to test the central esis and accomplish these objectives by the following four specific aims: 1) Does HuR ablation alter CD4+ Th subset differentiation?; 2) Determine whether HuR is required for allergic airway inflammation in vivo; 3) Mechanistic determinants of IL-2 and Th2 cytokine expression; 4) Determine whether human lymphocytes have dysregulated HuR expression. We believe our study is innovative, because such approaches will provide novel mechanistic insights into T cell posttranscriptional cytokine regulation. The proposed research is significant, because it will elucidate how airway responses connect at the molecular level with both adaptive and innate immunity to control lung inflammation.

Public Health Relevance

The proposed research is relevant to public health because elucidation of posttranscriptional gene control mechanisms in lungs is ultimately expected to increase understanding of pulmonary inflammation in asthma. This is especially the case, since there appears to be disease heterogeneity in human asthma and not all patients respond equally to commonly used medications to treat pulmonary inflammation. Thus, the proposed research is relevant to the part of the NIH's mission to increase knowledge that will help to reduce morbidity and mortality from pulmonary diseases such as asthma.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Research Project (R01)
Project #
5R01AI080870-10
Application #
9697436
Study Section
Hypersensitivity, Autoimmune, and Immune-mediated Diseases Study Section (HAI)
Program Officer
Dong, Gang
Project Start
2009-08-14
Project End
2020-08-31
Budget Start
2019-03-01
Budget End
2020-08-31
Support Year
10
Fiscal Year
2019
Total Cost
Indirect Cost
Name
University of Michigan Ann Arbor
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
073133571
City
Ann Arbor
State
MI
Country
United States
Zip Code
48109
Amatya, Nilesh; Childs, Erin E; Cruz, J Agustin et al. (2018) IL-17 integrates multiple self-reinforcing, feed-forward mechanisms through the RNA binding protein Arid5a. Sci Signal 11:
Chen, Jing; Adamiak, William; Huang, Ganlei et al. (2017) Interaction of RNA-binding protein HuR and miR-466i regulates GM-CSF expression. Sci Rep 7:17233
Chen, Jing; Martindale, Jennifer L; Cramer, Carole et al. (2017) The RNA-binding protein HuR contributes to neuroinflammation by promoting C-C chemokine receptor 6 (CCR6) expression on Th17 cells. J Biol Chem 292:14532-14543
Techasintana, Patsharaporn; Ellis, Jason S; Glascock, Jacqueline et al. (2017) The RNA-Binding Protein HuR Posttranscriptionally Regulates IL-2 Homeostasis and CD4+ Th2 Differentiation. Immunohorizons 1:109-123
Techasintana, Patsharaporn; Davis, J Wade; Gubin, Matthew M et al. (2015) Transcriptomic-Wide Discovery of Direct and Indirect HuR RNA Targets in Activated CD4+ T Cells. PLoS One 10:e0129321
Miller, Daniel L; Davis, J Wade; Taylor, Kristen H et al. (2015) Identification of a human papillomavirus-associated oncogenic miRNA panel in human oropharyngeal squamous cell carcinoma validated by bioinformatics analysis of the Cancer Genome Atlas. Am J Pathol 185:679-92
Chen, Jing; Cascio, Jason; Magee, Joseph D et al. (2013) Posttranscriptional gene regulation of IL-17 by the RNA-binding protein HuR is required for initiation of experimental autoimmune encephalomyelitis. J Immunol 191:5441-50
Dahm, Garrett M; Gubin, Matthew M; Magee, Joseph D et al. (2012) Method for the isolation and identification of mRNAs, microRNAs and protein components of ribonucleoprotein complexes from cell extracts using RIP-Chip. J Vis Exp :
Stellato, Cristiana; Gubin, Matthew M; Magee, Joseph D et al. (2011) Coordinate regulation of GATA-3 and Th2 cytokine gene expression by the RNA-binding protein HuR. J Immunol 187:441-9
Gubin, Matthew M; Calaluce, Robert; Davis, J Wade et al. (2010) Overexpression of the RNA binding protein HuR impairs tumor growth in triple negative breast cancer associated with deficient angiogenesis. Cell Cycle 9:3337-46