Efficient removal of damaged sub-cellular organelles via the autophagy-lysosome machinery is critical for maintaining cardiac myocyte viability in homeostasis and with ischemia-reperfusion (IR) injury. Emerging evidence implicates an essential role for innate immunity proteins as intracellular `damage sensors' to orchestrate their autophagic removal. One such protein, TRAF2 (Tumor necrosis factor Receptor-Associated Factor-2), activates cytoprotective signaling downstream of both TNF receptors to prevent IR-induced cardiomyocyte death. We have recently discovered a novel role for TRAF2 as an E3 ubiquitin ligase in autophagic removal of damaged mitochondria in cardiac myocytes. Mitochondrial damage triggers stabilization of PINK1, a serine-threonine kinase, on the outer mitochondrial membrane, which recruits PARKIN, an E3 ubiquitin ligase to ubiquitin- tag mitochondrial proteins for degradation. Whether PINK1 signaling also recruits TRAF2 to damaged mitochondria; and whether TRAF2, plays a non-redundant role vis--vis PARKIN in mitochondrial autophagy, in vivo, is not known. In parallel studies, we have also uncovered evidence for endoplasmic reticulum (ER) permeabilization under conditions of ER stress and in cardiac IR injury. ER damage activates IRE1?, an ER-localized serine-threonine kinase and RNAase, which interacts with TRAF2; and in-vitro studies indicate that IRE1?-TRAF2 signaling at the mitochondria associated membranes (MAM) orchestrates autophagy of damaged mitochondria and ER. In this proposal, we will test the hypothesis that TRAF2 signaling mediates selective autophagy of endoplasmic reticulum and mitochondria to regulate cardiac myocyte survival in homeostasis and under stress; under three specific aims.
In aim 1, we will determine the role of TRAF2 in cardiac myocyte survival in homeostasis and under stress.
In aim 2, we will examine the role of TRAF2 vis--vis PARKIN in mitochondrial autophagy in cardiac myocytes.
In aim 3, we will determine the role of IRE1?-TRAF2 signaling axis in ER-phagy in cardiac myocytes. These studies will elucidate the cellular basis for targeting TRAF2 as an innate immunity sensor that coordinates autophagic removal of damaged organelles at the mitochondria-ER interface (MAM) to enhance cardiomyocyte survival in myocardial infarction and prevent heart failure, a key mission of the NIH.

Public Health Relevance

Heart failure, the most common cause of death in the United States, is triggered by myocardial infarction and coronary artery disease in the majority of cases. Activation of the innate immunity system, a first line of defense against injury; and autophagy, a process for removal of damaged intracellular material, are essential for cell survival under these conditions. This proposal critically examines the role of TRAF2, an innate immunity signaling protein, in removal of damaged essential organelles in the setting of ischemic injury to ensure heart muscle cell survival and develop strategies to salvage cardiac muscle and prevent heart failure.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
2R01HL107594-06
Application #
9106064
Study Section
Myocardial Ischemia and Metabolism Study Section (MIM)
Program Officer
Schwartz, Lisa
Project Start
2011-07-01
Project End
2021-04-30
Budget Start
2016-05-01
Budget End
2017-04-30
Support Year
6
Fiscal Year
2016
Total Cost
Indirect Cost
Name
Washington University
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
068552207
City
Saint Louis
State
MO
Country
United States
Zip Code
63130
Mani, Kartik; Diwan, Abhinav (2018) Drugging the Hippo (Pathway): A Strategy to Stimulate Cardiac Regeneration? JACC Basic Transl Sci 3:654-656
Liss, Kim H H; McCommis, Kyle S; Chambers, Kari T et al. (2018) The impact of diet-induced hepatic steatosis in a murine model of hepatic ischemia/reperfusion injury. Liver Transpl 24:908-921
Liu, Haiyan; Javaheri, Ali; Godar, Rebecca J et al. (2017) Intermittent fasting preserves beta-cell mass in obesity-induced diabetes via the autophagy-lysosome pathway. Autophagy 13:1952-1968
Hartupee, Justin; Szalai, Gabor D; Wang, Wei et al. (2017) Impaired Protein Quality Control During Left Ventricular Remodeling in Mice With Cardiac Restricted Overexpression of Tumor Necrosis Factor. Circ Heart Fail 10:
Sergin, Ismail; Evans, Trent D; Zhang, Xiangyu et al. (2017) Exploiting macrophage autophagy-lysosomal biogenesis as a therapy for atherosclerosis. Nat Commun 8:15750
Klionsky, Daniel J (see original citation for additional authors) (2016) Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition). Autophagy 12:1-222
Lampropoulou, Vicky; Sergushichev, Alexey; Bambouskova, Monika et al. (2016) Itaconate Links Inhibition of Succinate Dehydrogenase with Macrophage Metabolic Remodeling and Regulation of Inflammation. Cell Metab 24:158-66
He, Li; Weber, Kassandra J; Diwan, Abhinav et al. (2016) Inhibition of mTOR reduces lipotoxic cell death in primary macrophages through an autophagy-independent mechanism. J Leukoc Biol 100:1113-1124
Yang, Kai-Chun; Ma, Xiucui; Liu, Haiyan et al. (2015) Tumor necrosis factor receptor-associated factor 2 mediates mitochondrial autophagy. Circ Heart Fail 8:175-87
Kanekura, Kohsuke; Ma, Xiucui; Murphy, John T et al. (2015) IRE1 prevents endoplasmic reticulum membrane permeabilization and cell death under pathological conditions. Sci Signal 8:ra62

Showing the most recent 10 out of 20 publications