Obesity, type 2 diabetes (T2DM), and insulin resistance are independent risk factors for heart failure. The long- term goal of this proposal is to understand the relationship between hyperinsulinemia and cardiac dysfunction in these populations. Hyperinsulinemia may also accelerate adverse LV remodeling in pressure overload hypertrophy and genetic reduction of insulin signaling in cardiomyocytes limits hypertrophic remodeling and reduces apoptosis in pressure overload, thereby preserving LV function. Our recent studies reveal that hyperinsulinemia desensitizes -AR-mediated stimulation of cardiac contractility by promoting 2ARGi-biased signaling and by inducing the phosphodiesterase (PDE4D), which represents a novel mechanism linking insulin resistance, hyperinsulinemia and LV dysfunction. This proposal will test the hypothesis that hyperinsulinemia attenuates LV contractility by directly impairing ?AR signaling via two distinct mechanisms: (1) Increased ?2AR/Gi coupling that inhibits adenylyl cyclase (AC) and cAMP production, and (2) Increased expression of PDE4D that increases cAMP degradation. This multi PI proposal reflects an active collaboration by the laboratories of Evan Dale Abel (University of Iowa) and Yang Kevin Xiang (University of California -Davis). Our combined expertise in myocardial insulin signaling and myocardial adrenergic signaling, using novel molecular biosensors to define subcellular adrenergic signaling domains in cardiomyocytes and a comprehensive array of mutant mouse models with perturbed IR or ?AR signaling, will address this hypothesis in the following three specific aims.
Aim 1 (Xiang): Will define the molecular mechanisms for and consequences of PDE4 induction in response to chronic hyperinsulinemia. Hypothesis: Insulin signaling reduces cAMP levels by increasing cardiac PDE4 levels for cAMP hydrolysis via ?2AR-ERK dependent modulation of PDE4 transcription and protein turnover.
Aim 2 (Abel): Will determine the mechanisms by which modulation of IR-??2AR signaling may attenuate obesity associated LV dysfunction. Hypothesis: Acute or chronic hyperinsulinemia will impair myocardial ?AR signaling and reduce contractility or inotropic reserve by promoting cAMP degradation and genetic or pharmacological inhibition of this crosstalk will preserve LV function in hyperinsulinemic states.
Aim 3 (Abel). Will determine if IR-??2AR crosstalk contributes to LV dysfunction in heart failue or in subjects with insulin resistance. Hypothesis: LV or atrial tissue from subjects with heart failure will reveal molecular signatures consistent with increased IR-??2AR ERK activation and PDE4 induction, and insulin resistant subjects will exhibit impaired heart rate responses to sub-maximal exercise. These studies will comprehensively dissect the mechanism for IR-??AR crosstalk that limits myocardial contractility in insulin resistant states, and may lead to novel approaches for treating or preventing heart failure in the high risk population with insulin resistance and the metabolic syndrome.

Public Health Relevance

Heart failure is a leading cause of mortality in the United States. Obesity and insulin resistance are independent risk factors for heart failure. The present study will address the critical question of the role of altered beta-adrenergic signaling in the pathophysiology of cardiac dysfunction in obesity, type 2 diabetes, and other insulin-resistant states. We will determine if augmented myocardial insulin signaling, as a consequence of the hyperinsulinemia that develops in obesity and type 2 diabetes, directly impairs the adrenergic signaling in the heart leading to reduced cardiac contractility. These studies will provide novel information regarding modulation of beta-adrenergic signaling pathways in the prevention or treatment of cardiac dysfunction in obesity and type 2 diabetes.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
1R01HL127764-01
Application #
8899989
Study Section
Cardiac Contractility, Hypertrophy, and Failure Study Section (CCHF)
Program Officer
Buxton, Denis B
Project Start
2015-04-01
Project End
2019-01-31
Budget Start
2015-04-01
Budget End
2016-01-31
Support Year
1
Fiscal Year
2015
Total Cost
Indirect Cost
Name
University of Iowa
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
062761671
City
Iowa City
State
IA
Country
United States
Zip Code
52246
Patriarchi, Tommaso; Shen, Ao; He, Wei et al. (2018) Nanodelivery of a functional membrane receptor to manipulate cellular phenotype. Sci Rep 8:3556
Zhang, Jianyi; Abel, E Dale (2018) Effective Metabolic Approaches for the Energy Starved Failing Heart: Bioenergetic Resiliency via Redundancy or Something Else? Circ Res 123:329-331
Reddy, Gopireddy Raghavender; West, Toni M; Jian, Zhong et al. (2018) Illuminating cell signaling with genetically encoded FRET biosensors in adult mouse cardiomyocytes. J Gen Physiol 150:1567-1582
Steinhorn, Benjamin; Sartoretto, Juliano L; Sorrentino, Andrea et al. (2017) Insulin-dependent metabolic and inotropic responses in the heart are modulated by hydrogen peroxide from NADPH-oxidase isoforms NOX2 and NOX4. Free Radic Biol Med 113:16-25
Fu, Qin; Wang, Qingtong; Xiang, Yang K (2017) Insulin and ? Adrenergic Receptor Signaling: Crosstalk in Heart. Trends Endocrinol Metab 28:416-427
Fu, Qin; Hu, Yuting; Wang, Qingtong et al. (2017) High-fat diet induces protein kinase A and G-protein receptor kinase phosphorylation of ?2 -adrenergic receptor and impairs cardiac adrenergic reserve in animal hearts. J Physiol 595:1973-1986
Park, Misun; Reddy, Gopireddy R; Wallukat, Gerd et al. (2017) ?1-adrenergic receptor O-glycosylation regulates N-terminal cleavage and signaling responses in cardiomyocytes. Sci Rep 7:7890
Pires, Karla Maria; Buffolo, Marcio; Schaaf, Christin et al. (2017) Activation of IGF-1 receptors and Akt signaling by systemic hyperinsulinemia contributes to cardiac hypertrophy but does not regulate cardiac autophagy in obese diabetic mice. J Mol Cell Cardiol 113:39-50
Wang, Qingtong; Liu, Yongming; Fu, Qin et al. (2017) Inhibiting Insulin-Mediated ?2-Adrenergic Receptor Activation Prevents Diabetes-Associated Cardiac Dysfunction. Circulation 135:73-88
Shi, Qian; Li, Minghui; Mika, Delphine et al. (2017) Heterologous desensitization of cardiac ?-adrenergic signal via hormone-induced ?AR/arrestin/PDE4 complexes. Cardiovasc Res 113:656-670

Showing the most recent 10 out of 18 publications