The overall objective of this K01 proposal is to determine the significance of visceral adipose tissue (VAT) derived microRNAs (miR) in the genesis of systemic insulin resistance (IR) associated with obesity and type 2 diabetes mellitus (DM2). In this proposal, we posit a key role for miR-223 in the pathogenesis of obesity/DM2. Our overarching hypothesis is driven by three key sets of preliminary data: a) miR-223 is the most upregulated miR in VAT of humans, correlating strongly with markers of IR and inflammation, b) miR-223 regulates multiple targets involved in insulin signaling, and c) plasma exosomes derived from obese/IR patients are enriched in miR-223 compared to controls. We propose to test the significance of visceral adipose macrophages (VAT-?) miR-223 as part of an ongoing inter-disciplinary investigation that will significantly enhance my training and propel my career towards independence.
In Aim 1, we will compare miR-223 expression in VAT-? with plasma exosome-miR-223 levels in obese patients undergoing bariatric surgery vs. non-obese controls, following post- surgery/post-weight loss exosome miR-223 levels compared to markers of BMI, IR, etc. We hypothesize that VAT-? and exosome miR-223 strongly correlate with markers of glycemic control, IR, and inflammation. Next, we will test the in vitro effects of miR-223 overexpression on target genes relevant to IR as predicted by computational bioinformatics.
In Aim 2, we hypothesize that VAT-? are an important source of plasma exosome miR-223. We will assess the temporal course of miR-223 expression in various compartments [VAT- ?, liver ? (Kupffer cells), and plasma exosomes] and further hypothesize that exosome-miR derived from VAT-? may contribute to regional (paracrine) and endocrine (liver) disruption of insulin signaling. To investigate this, we will assess in vitro transport of fluorescently labeled macrophage-derived exosomes containing miR-223 to human hepatocytes and adipocytes in co-culture assays. The effect of exosome-miR-223 on validated targets in human hepatocytes and adipocytes will be assayed. Finally in Aim 3, we hypothesize that miR-223 antagonism in VAT-? accomplished via regional delivery of targeted antagomiR-223 nanoparticles will decrease functional plasma exosome miR-223, beneficially impacting whole body insulin sensitivity. Collectively, the experimental findings from this proposal should help drive new understanding of VAT-derived miRs as pleiotropic modulators of processes in IR/DM2.

Public Health Relevance

Obesity brings with it problems in processing sugars from food. We believe that improperly functioning immune cells in the fat tissues around the body's major organs plays an important role in this obesity-related sugar processing problem. Our study proposes strategies to characterize and test this belief.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Scientist Development Award - Research & Training (K01)
Project #
1K01DK099475-01A1
Application #
8767954
Study Section
Digestive Diseases and Nutrition C Subcommittee (DDK)
Program Officer
Hyde, James F
Project Start
2014-09-01
Project End
2017-07-31
Budget Start
2014-09-01
Budget End
2015-07-31
Support Year
1
Fiscal Year
2014
Total Cost
Indirect Cost
Name
University of Maryland Baltimore
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
City
Baltimore
State
MD
Country
United States
Zip Code
21201
Gangwar, Roopesh S; Rajagopalan, Sanjay; Natarajan, Rama et al. (2018) Noncoding RNAs in Cardiovascular Disease: Pathological Relevance and Emerging Role as Biomarkers and Therapeutics. Am J Hypertens 31:150-165
Bagalkot, Vaishali; Deiuliis, Jeffrey A; Rajagopalan, Sanjay et al. (2016) ""Eat me"" imaging and therapy. Adv Drug Deliv Rev 99:2-11
Deiuliis, J A (2016) MicroRNAs as regulators of metabolic disease: pathophysiologic significance and emerging role as biomarkers and therapeutics. Int J Obes (Lond) 40:88-101
Bagalkot, Vaishali; Badgeley, Marcus A; Kampfrath, Thomas et al. (2015) Hybrid nanoparticles improve targeting to inflammatory macrophages through phagocytic signals. J Control Release 217:243-55