Explanation Mono-ADP-ribosylation is a post-translational protein modification, in which ADP-ribose is transferred from NAD to an acceptor amino acid. It was first identified as a mechanism of disease pathogenesis in the bacterial diseases such as cholera, diphtheria and pertussis, where toxins ADP-ribosylate critical regulatory and biosynthetic proteins. Mammalian tissues have enzymatic activities that mimic those of the bacterial toxins. Mammalian arginine-specific mono-ADP-ribosylation however, is a reversible modification of protein. Arginine-specific mono-ADP-ribosyltrans- ferases (ARTs) (e.g., ART1, ART5), transfer ADP-ribose from NAD to arginine residues of target proteins and ADP-ribosylarginine hydrolase 1 (ARH1) reverses the reaction by cleaving the ADP-ribose-(arginine)-protein bond. Data are consistent with ART and ARH1 serving as opposing arms of an arginine ADP-ribosylation cycle. Significant advances have led to the identification of mono-ADP-ribosylated proteins. Reanalysis of phosphoproteomic data identified 79 mono-ADP-ribosylated proteins including a muscle-specific membrane-repair protein tripartite motif 72 (TRIM72), also known as Mitsugumin 53 (MG53). Another method involved enrichment of ADP-ribose-acceptor proteins using the ADP-ribose-binding macro domain protein Af1521. Similar to other post-translational modifications, e.g., phosphorylation, mono-ADP-ribosylation can alter protein function. TRIM72 is abundant in heart and skeletal muscle and in lung and kidney epithelial cells where it is involved in membrane repair. Effective membrane repair protects cardiomyocytes from ischemic damage. Oligomerization of TRIM72 was required acutely for membrane repair, bringing TRIM72 to the site of injury. Nitrosylation of TRIM72 protected it from degradation under H2O2-induced oxidative stress. TRIM72 also countered cell damage due to ischemia-reperfusion injury. TRIM72 in complex with caveolin-3 (Cav-3) activated phosphatidylinositol-3-kinases (PI3K)-dependent reperfusion injury salvage kinase (RISK), thereby enhancing cell survival. Overexpression of TRIM72 protected cells from plasma membrane damage in muscular dystrophy, as well as lung, kidney and heart injury, suggesting that TRIM72 could be a therapeutic target in diseases involving cell membrane injury. ARH1-deficient mice developed cardiomyopathy with myocardial fibrosis, decreased myocardial function under dobutamine stress, and increased susceptibility to ischemia-reperfusion injury. We hypothesized that a mono-ADP-ribosylated protein serving as an ART1 and ARH1 substrate was involved in preserving cardiac function. Indeed, membrane-repair protein, TRIM72, was identified as a substrate for ART1 and ARH. Further, we observed in ARH1-knockout mouse heart in vivo and in the Langendorff perfused-heart model ex vivo, that the amount of mono-ADP-ribosylated TRIM72 was increased in ischemia-reperfusion injury. To understand better the role of TRIM72 and ADP-ribosylation, we used C2C12 myocytes. ARH1 knockdown in C2C12 myocytes increased ADP-ribosylation of TRIM72, and delayed wound healing in a scratch assay. Indeed, interrupting the TRIM72 mono-ADP-ribosylation cycle by knockdown of ART1, ARH1, or TRIM72 delayed membrane repair and wound healing. Effects of overexpressing mutant TRIM72 that lacks two primary ADP-ribosylation sites (R207K, R260K) and is not ADP-ribosylated, had a similar effect. We used a laser-injury model to test the role of ADP-ribosylated TRIM72 on acute membrane repair. After laser injury of C2C12 myocyte membranes, live-cell imaging revealed rapid accumulation of WT TRIM72-GFP at the site, which was markedly lower in cells depleted of ARH1 or ART1. Moreover, accumulation of double mutant TRIM72 (R207K, R260K)-GFP at the injury site was significantly less than that of WT TRIM72-GFP. The regulatory enzymes ART1 and ARH1 and their substrate TRIM72 were found in complexes, which were co-immunoprecipitated from mouse heart lysates. The complexes found in association with TRIM72 appeared to be relatively heterogeneous and could be resolved by FPLC and HPLC. Some of the complexes included Cav-3. It has been reported that TRIM72 oligomerization serves as a key mechanism for regulation of acute membrane repair. Oligomerization of TRIM72, observed in WT mice, was delayed in ARH1-deficient, heart lysates, where a dimer of TRIM72 rather than a trimer was seen. In addition, at a cellular level, oligomerization of TRIM72 at the sites of injury required the presence of the mono-ADP-ribosylation cycle, that is, ART1 and ARH1. The mono-ADP-ribosylation inhibitors, vitamin K1 and novobiocin, inhibited oligomerization of TRIM72, the mechanism by which TRIM72 is recruited to the site of injury. We propose that a mono-ADP-ribosylation cycle involving recruitment of TRIM72 and other regulatory factors to sites of membrane damage is critical for membrane repair and wound healing following myocardial injury.

Project Start
Project End
Budget Start
Budget End
Support Year
26
Fiscal Year
2018
Total Cost
Indirect Cost
Name
U.S. National Heart Lung and Blood Inst
Department
Type
DUNS #
City
State
Country
Zip Code
Stevens, Linda A; Moss, Joel (2018) Mono-ADP-Ribosylation Catalyzed by Arginine-Specific ADP-Ribosyltransferases. Methods Mol Biol 1813:149-165
Pourfarjam, Yasin; Ventura, Jessica; Kurinov, Igor et al. (2018) Structure of human ADP-ribosyl-acceptor hydrolase 3 bound to ADP-ribose reveals a conformational switch that enables specific substrate recognition. J Biol Chem 293:12350-12359
Yahiro, Kinnosuke; Nagasawa, Sayaka; Ichimura, Kimitoshi et al. (2018) Mechanism of inhibition of Shiga-toxigenic Escherichia coli SubAB cytotoxicity by steroids and diacylglycerol analogues. Cell Death Discov 4:22
Mashimo, Masato; Moss, Joel (2018) ADP-Ribosyl-Acceptor Hydrolase Activities Catalyzed by the ARH Family of Proteins. Methods Mol Biol 1813:187-204
Bu, Xiangning; Kato, Jiro; Hong, Julie A et al. (2018) CD38 knockout suppresses tumorigenesis in mice and clonogenic growth of human lung cancer cells. Carcinogenesis 39:242-251
Abplanalp, Jeannette; Leutert, Mario; Frugier, Emilie et al. (2017) Proteomic analyses identify ARH3 as a serine mono-ADP-ribosylhydrolase. Nat Commun 8:2055
Sparrer, Konstantin M J; Gableske, Sebastian; Zurenski, Matthew A et al. (2017) TRIM23 mediates virus-induced autophagy via activation of TBK1. Nat Microbiol 2:1543-1557
Ida, Chieri; Yamashita, Sachiko; Tsukada, Masaki et al. (2016) An enzyme-linked immunosorbent assay-based system for determining the physiological level of poly(ADP-ribose) in cultured cells. Anal Biochem 494:76-81
Yamashita, Sachiko; Tanaka, Masakazu; Sato, Teruaki et al. (2016) Effect of mild temperature shift on poly(ADP-ribose) and ?H2AX levels in cultured cells. Biochem Biophys Res Commun 476:594-599
Chen, Pei-Wen; Jian, Xiaoying; Heissler, Sarah M et al. (2016) The Arf GTPase-activating Protein, ASAP1, Binds Nonmuscle Myosin 2A to Control Remodeling of the Actomyosin Network. J Biol Chem 291:7517-26

Showing the most recent 10 out of 41 publications