Increased secretion of amylin by pancreatic beta-cells (hyperamylinemia) is common in obese and insulin resistant patients, coincides with hyperinsulinemia, and promotes formation of toxic amylin oligomers. Oligomeric amylin induces beta-cell apoptosis contributing to the development of type-2 diabetes. Recent studies demonstrate that amylin oligomers also affect the vascular system, kidneys, and heart. Our data show large deposits of oligomerized amylin in failing hearts from obese and T2D patients, but not in hearts from controls. Oligomeric amylin was found in myocyte injury areas suggesting a role in the mechanism of injury. Indeed, our pilot study on a humanized rat model of hyperamylinemia (the HIP rat) indicates that amylin oligomers attach to cardiac myocytes and induce oxidative stress and Ca2+ dysregulation leading to diastolic dysfunction and hypertrophy. The pilot study also suggests that endogenous molecules with anti-aggregation properties, such as plasmin and epoxyeicosanoids, limit cardiac accumulation of amylin and its myopathic response. Based on these preliminary results, our research proposal will test the hypotheses that 1) cardiac accumulation of oligomerized amylin accelerates diabetic heart injury by inducing sarcolemmal damage and oxidative stress, and 2) limiting amylin deposition in the heart may reduce/ delay the onset of diabetic heart failure. These hypotheses will mechanistically be assessed by using transgenic rat models overexpressing either the amyloido- genic human amylin (HIP rats) or the non-amyloidogenic rat amylin isoform (UCD rats). Specifically, planned studies will determine how accumulation of oligomerized amylin in the HIP rat heart a) disrupts sarcolemmal processes, b) induces oxidative stress and myocyte Ca2+ dysregulation, and c) activates Ca2+-mediated CaMKII-HDAC and calcineurin-NFAT hypertrophy signaling pathways. Based on the results of our pilot study, cardiac dysfunction in HIP rats is expected to develop even in pre-diabetes, as often observed in humans. In contrast, our pilot study predicts that UCD rats matched for age and glucose, but lacking cardiac amylin deposition, may show signs of cardiac dysfunction after the onset of diabetes. Our research will also determine if disrupting deposition of oligomeric amylin in the heart and recovering sarcolemmal integrity improve cardiac function in the HIP rat model. This innovative concept will be explored in longitudinal studies using membrane sealants and scavengers of circulating amylin oligomers. Hence, our research project proposes that amylin buildup is a key contributor to the multifactorial pathogenesis of diabetic heart injury and that mitigating amylin oligomer accumulation could delay the onset of diabetic heart failure. If our hypothesis of cardiotoxic amylin oligomer is proven, then circulating amylin oligomers are a feasible therapeutic target to reduce diabetic heart injury.

Public Health Relevance

Obesity and type-2 diabetes have reached epidemic proportions in the US1 and are associated with a high risk of heart failure. The pathogenesis of heart dysfunction in obesity and T2D is multifactorial and not fully elucidated, which makes diagnosis and treatment of diabetic heart disease challenging. Our study aims at understanding how hyperamylinemia, a common condition in obese and insulin resistant patients, promotes toxic accumulation of oligomerized amylin in the heart and how the buildup of amylin affects cardiac function. Planned studies also attempt to find therapeutic tools that can mitigate cardiac accumulation of oligomerized amylin and its deleterious effects. The focus is on modulating the endogenous system of scavenging the amylin oligomers circulating in the blood as a possible mean to reduce cardiotoxicity of hyperamylinemia. If our hypothesis of cardiotoxic amylin oligomer is proven, then hyperamylinemia and the oligomerized amylin circulating in the blood will be feasible therapeutic targets to limit the risk of diabetic heart injury.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
4R01HL118474-04
Application #
9109039
Study Section
Myocardial Ischemia and Metabolism Study Section (MIM)
Program Officer
Buxton, Denis B
Project Start
2013-08-23
Project End
2017-07-31
Budget Start
2016-08-01
Budget End
2017-07-31
Support Year
4
Fiscal Year
2016
Total Cost
Indirect Cost
Name
University of Kentucky
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
939017877
City
Lexington
State
KY
Country
United States
Zip Code
40506
Stewart, Bradley D; Scott, Caitlin E; McCoy, Thomas P et al. (2018) Computational modeling of amylin-induced calcium dysregulation in rat ventricular cardiomyocytes. Cell Calcium 71:65-74
Popescu, Iuliana; Yin, Guo; Velmurugan, Sathya et al. (2018) Lower sarcoplasmic reticulum Ca2+ threshold for triggering afterdepolarizations in diabetic rat hearts. Heart Rhythm :
Liu, Miao; Hoskins, Amanda; Verma, Nirmal et al. (2018) Amylin and diabetic cardiomyopathy - amylin-induced sarcolemmal Ca2+ leak is independent of diabetic remodeling of myocardium. Biochim Biophys Acta Mol Basis Dis 1864:1923-1930
Van Steenbergen, Anne; Balteau, Magali; Ginion, Audrey et al. (2017) Sodium-myoinositol cotransporter-1, SMIT1, mediates the production of reactive oxygen species induced by hyperglycemia in the heart. Sci Rep 7:41166
Ly, Han; Verma, Nirmal; Wu, Fengen et al. (2017) Brain microvascular injury and white matter disease provoked by diabetes-associated hyperamylinemia. Ann Neurol 82:208-222
Liu, Miao; Verma, Nirmal; Peng, Xiaoli et al. (2016) Hyperamylinemia Increases IL-1? Synthesis in the Heart via Peroxidative Sarcolemmal Injury. Diabetes 65:2772-83
Ilaiwy, Amro; Liu, Miao; Parry, Traci L et al. (2016) Human amylin proteotoxicity impairs protein biosynthesis, and alters major cellular signaling pathways in the heart, brain and liver of humanized diabetic rat model in vivo. Metabolomics 12:
Verma, Nirmal; Ly, Han; Liu, Miao et al. (2016) Intraneuronal Amylin Deposition, Peroxidative Membrane Injury and Increased IL-1? Synthesis in Brains of Alzheimer's Disease Patients with Type-2 Diabetes and in Diabetic HIP Rats. J Alzheimers Dis 53:259-72
Guan, Hanjun; Chow, K Martin; Song, Eunsuk et al. (2015) The Mitochondrial Peptidase Pitrilysin Degrades Islet Amyloid Polypeptide in Beta-Cells. PLoS One 10:e0133263
Ly, Han; Despa, Florin (2015) Hyperamylinemia as a risk factor for accelerated cognitive decline in diabetes. Expert Rev Proteomics 12:575-7

Showing the most recent 10 out of 16 publications