Heat shock proteins (HSP) play key roles in protein folding, quality control pathways and are potent cardioprotective agents against ischemia/reperfusion injury. Yet, a fundamental but unresolved question in the field is whether selective members of the HSP family have evolved specialized roles in a tissue-specific manner (such as the heart) in mammals. We hypothesize that HSPB2, a member of small MW HSP superfamily that includes CRYAB and HSP25, exerts specialized properties for cardiac systolic performance, mitochondrial energetics and protection against apoptosis and necrosis. There are, however, several fundamental issues that need to be resolved in order to test the validity of this idea: What functions and properties of cytosolic HSPB2 expression mediate mitochondrial energetics? Would such effects on cardiac energetics and function observed in DKO/mCryAB Tg mice be recapitulated in a novel tissue-specific knockout of hspb2? What molecular mechanisms and signaling pathways of hspb2 deficiency contribute to increased resistance and paradoxical ischemic cardioprotection? Do the duration of ischemic insult and/or the ischemic milieu of DKO deficiency mimic phenotypic responses of hspb2 KO mice in the mammalian heart? Does HSPB2 expression exert direct or indirect consequences on mitochondrial (dys)function? Results of the proposed studies should help elucidate the mechanism by which the HSPB2 mediates the `classical'stress response in vivo. Using tissue-specific gain-of-function and loss-of-function models of mouse mutants, we aim to understand the mechanisms of selective HSPB2 expression in the pathogenesis of cardiomyocyte toxicity and ischemic cardioprotection.

Public Health Relevance

Cellular proteins are constantly being exposed to oxidative and metabolic stresses, which alter their basic structure and ultimately function. Heat shock protein are evolutionarily conserved protein whose main functions as molecular chaperones prevent protein misfolding and aggregation, conditions that increase considerably following an ischemic insult. This research proposal will examine the specific roles of HSPB2 for mitochondrial function and cardiac energetics using genetically engineered mouse models of human diseases.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
5R01HL074370-02
Application #
7617735
Study Section
Myocardial Ischemia and Metabolism Study Section (MIM)
Program Officer
Liang, Isabella Y
Project Start
2008-05-01
Project End
2012-04-30
Budget Start
2009-05-01
Budget End
2010-04-30
Support Year
2
Fiscal Year
2009
Total Cost
$381,011
Indirect Cost
Name
University of Utah
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
009095365
City
Salt Lake City
State
UT
Country
United States
Zip Code
84112
Grose, Julianne H; Langston, Kelsey; Wang, Xiaohui et al. (2015) Characterization of the Cardiac Overexpression of HSPB2 Reveals Mitochondrial and Myogenic Roles Supported by a Cardiac HspB2 Interactome. PLoS One 10:e0133994
Hussein, Rasha M; Benjamin, Ivor J; Kampinga, Harm H (2015) Rescue of ?B Crystallin (HSPB5) Mutants Associated Protein Aggregation by Co-Expression of HSPB5 Partners. PLoS One 10:e0126761
Lopez-Izquierdo, Angelica; Warren, Mark; Riedel, Michael et al. (2014) A near-infrared fluorescent voltage-sensitive dye allows for moderate-throughput electrophysiological analyses of human induced pluripotent stem cell-derived cardiomyocytes. Am J Physiol Heart Circ Physiol 307:H1370-7
Riedel, Michael; Jou, Chuanchau J; Lai, Shuping et al. (2014) Functional and pharmacological analysis of cardiomyocytes differentiated from human peripheral blood mononuclear-derived pluripotent stem cells. Stem Cell Reports 3:131-41
Banerjee Mustafi, Soumyajit; Grose, Julianne H; Zhang, Huali et al. (2014) Aggregate-prone R120GCRYAB triggers multifaceted modifications of the thioredoxin system. Antioxid Redox Signal 20:2891-906
Christians, Elisabeth S; Mustafi, Soumyajit B; Benjamin, Ivor J (2014) Chaperones and cardiac misfolding protein diseases. Curr Protein Pept Sci 15:189-204
Squires, Shayne; Christians, Elisabeth; Riedel, Michael et al. (2013) Effects of redox state on the efficient uptake of cell permeable Peptide in Mammalian cells. Open Biochem J 7:54-65
Xie, Heng B; Cammarato, Anthony; Rajasekaran, Namakkal S et al. (2013) The NADPH metabolic network regulates human ?B-crystallin cardiomyopathy and reductive stress in Drosophila melanogaster. PLoS Genet 9:e1003544
Brewer, Alison C; Mustafi, Soumyajit Banerjee; Murray, Thomas V A et al. (2013) Reductive stress linked to small HSPs, G6PD, and Nrf2 pathways in heart disease. Antioxid Redox Signal 18:1114-27
Limphong, Pattraranee; Zhang, Huali; Christians, Elisabeth et al. (2013) Modeling human protein aggregation cardiomyopathy using murine induced pluripotent stem cells. Stem Cells Transl Med 2:161-6

Showing the most recent 10 out of 15 publications