Diabetes is at epidemic proportions with 300 million people projected to have diabetes by 2025. Heart disease is the cause of death in 80% of diabetic patients of which the major contributing factor is coronary artery disease. Diabetics suffer from a diabetic cardiomyopathy independent of the vascular effects of hypertension or coronary artery disease. This suggests that there are contributing factors within the cardiac myocyte itself that may give rise to detrimental cardiac remodeling associated with diabetes. Diabetic cardiomyopathy is triggered by alteration in fatty acid metabolism, hyperinsulinemia, and hyperglycemia. Diabetic cardiomyopathy involves complex changes in signaling and metabolism that may be regulated at multiple levels: 1) altered cellular signaling through differential regulation of receptor/effector expression and localization;2) altered mitochondrial function and dynamics;3) altered nuclear activity leading t pathologic gene expression that may ultimately affect the latter two factors. As such, membrane/cytoplasmic signaling, mitochondria, and the nucleus could be defined as three control points that limit the ability of the heart to adapt to diabetic stress and offer novel therapeutic targets. A common molecular regulator at these three sites has not been identified. An emerging idea in signal transduction suggests signaling molecules exist as dynamic, spatially organized multi-protein complexes in lipid-rich microdomains of the plasma membrane continuously forming and dissociating under basal or stimulated conditions. Caveolae are cholesterol and sphingolipid-enriched microenvironments that form microscopically distinct flask-like invaginations of the plasma membrane. Our novel preliminary data show that heart-specific caveolin-3 overexpression (Cav-3 OE) protects hearts from diabetic cardiomyopathy in a model of Type II diabetes by modulating function of membrane/cytoplasmic signaling, mitochondria, and the nucleus. We hypothesize that the localized expression and regulatory activity of caveolin in the membrane/cytoplasm, mitochondria, and/or the nucleus are critical to protection of the heart from diabetic cardiomyopathy. Targeted expression of caveolin may provide a detailed understanding of the molecular role of caveolin in diabetes to provide more directed targeting of therapeutics. The following aims are proposed:
Aim 1 : Determine the role of caveolin in membrane/cytoplasmic signaling and the therapeutic potential of membrane-targeted caveolin expression in the progression of diabetic cardiomyopathy.
Aim 2 : Determine the role of caveolin in mitochondrial function and dynamics and the therapeutic potential of mitochondrial-targeted caveolin expression in the progression of diabetic cardiomyopathy.
Aim 3 : Determine the role of caveolin in nuclear envelope stability, modulation of gene expression, and the therapeutic potential of nuclear-targeted caveolin expression in the progression of diabetic cardiomyopathy.

Public Health Relevance

Heart disease is the cause of death in 80% of diabetic patients of which the major contributing factor is coronary artery disease. Diabetics suffer from a diabetic cardiomyopathy independent of the vascular effects of hypertension or coronary artery disease. An emerging idea in signal transduction suggests signaling molecules exist as dynamic, spatially organized multi-protein complexes in lipid-rich microdomains of the plasma membrane continuously forming and dissociating under basal or stimulated conditions. Caveolae are cholesterol and sphingolipid-enriched microenvironments that form microscopically distinct flask-like invaginations of the plasma membrane. Our novel preliminary data show that heart-specific caveolin-3 overexpression (Cav-3 OE) protects hearts from diabetic cardiomyopathy in a model of Type II diabetes by modulating function of membrane/cytoplasmic signaling, mitochondria, and the nucleus. We propose caveolin based therapeutics can limit the progression of diabetic cardiomyopathy.

Agency
National Institute of Health (NIH)
Institute
Veterans Affairs (VA)
Type
Non-HHS Research Projects (I01)
Project #
5I01BX001963-02
Application #
8669722
Study Section
Cardiovascular Studies A (CARA)
Project Start
2013-04-01
Project End
2017-03-31
Budget Start
2014-04-01
Budget End
2015-03-31
Support Year
2
Fiscal Year
2014
Total Cost
Indirect Cost
Name
VA San Diego Healthcare System
Department
Type
DUNS #
City
San Diego
State
CA
Country
United States
Zip Code
92161
Egawa, Junji; Zemljic-Harpf, Alice; Mandyam, Chitra D et al. (2018) Neuron-Targeted Caveolin-1 Promotes Ultrastructural and Functional Hippocampal Synaptic Plasticity. Cereb Cortex 28:3255-3266
Schilling, Jan M; Head, Brian P; Patel, Hemal H (2018) Caveolins as Regulators of Stress Adaptation. Mol Pharmacol 93:277-285
Kong, Cherrie H T; Bryant, Simon M; Watson, Judy J et al. (2018) The Effects of Aging on the Regulation of T-Tubular ICa by Caveolin in Mouse Ventricular Myocytes. J Gerontol A Biol Sci Med Sci 73:711-719
Haushalter, Kristofer J; Casteel, Darren E; Raffeiner, Andrea et al. (2018) Phosphorylation of protein kinase A (PKA) regulatory subunit RI? by protein kinase G (PKG) primes PKA for catalytic activity in cells. J Biol Chem 293:4411-4421
Ichikawa, Yasuhiro; Zemljic-Harpf, Alice E; Zhang, Zheng et al. (2017) Modulation of caveolins, integrins and plasma membrane repair proteins in anthracycline-induced heart failure in rabbits. PLoS One 12:e0177660
Thomas, Joanna L; Pham, Hai; Li, Ying et al. (2017) Hypoxia-inducible factor-1? activation improves renal oxygenation and mitochondrial function in early chronic kidney disease. Am J Physiol Renal Physiol 313:F282-F290
Egawa, Junji; Schilling, Jan M; Cui, Weihua et al. (2017) Neuron-specific caveolin-1 overexpression improves motor function and preserves memory in mice subjected to brain trauma. FASEB J 31:3403-3411
Busija, Anna R; Patel, Hemal H; Insel, Paul A (2017) Caveolins and cavins in the trafficking, maturation, and degradation of caveolae: implications for cell physiology. Am J Physiol Cell Physiol 312:C459-C477
Mandyam, Chitra D; Schilling, Jan M; Cui, Weihua et al. (2017) Neuron-Targeted Caveolin-1 Improves Molecular Signaling, Plasticity, and Behavior Dependent on the Hippocampus in Adult and Aged Mice. Biol Psychiatry 81:101-110
Schilling, Jan M; Patel, Hemal H (2016) Non-canonical roles for caveolin in regulation of membrane repair and mitochondria: implications for stress adaptation with age. J Physiol 594:4581-9

Showing the most recent 10 out of 36 publications