Next Article in Journal
Loss of Individual Mitochondrial Ribonuclease P Complex Proteins Differentially Affects Mitochondrial tRNA Processing In Vivo
Next Article in Special Issue
The Role of Autophagy in Chemical Proteasome Inhibition Model of Retinal Degeneration
Previous Article in Journal
Mutations in bdcA and valS Correlate with Quinolone Resistance in Wastewater Escherichia coli
Previous Article in Special Issue
Advances in the Development Ubiquitin-Specific Peptidase (USP) Inhibitors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of HECT-Type E3 Ligase in the Development of Cardiac Disease

Department of Cardiology, Pulmonology and Nephrology, Yamagata University School of Medicine, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(11), 6065; https://doi.org/10.3390/ijms22116065
Submission received: 14 May 2021 / Revised: 26 May 2021 / Accepted: 1 June 2021 / Published: 4 June 2021
(This article belongs to the Special Issue Ubiquitination in Health and Diseases: Volume II)

Abstract

:
Despite advances in medicine, cardiac disease remains an increasing health problem associated with a high mortality rate. Maladaptive cardiac remodeling, such as cardiac hypertrophy and fibrosis, is a risk factor for heart failure; therefore, it is critical to identify new therapeutic targets. Failing heart is reported to be associated with hyper-ubiquitylation and impairment of the ubiquitin–proteasome system, indicating an importance of ubiquitylation in the development of cardiac disease. Ubiquitylation is a post-translational modification that plays a pivotal role in protein function and degradation. In 1995, homologous to E6AP C-terminus (HECT) type E3 ligases were discovered. E3 ligases are key enzymes in ubiquitylation and are classified into three families: really interesting new genes (RING), HECT, and RING-between-RINGs (RBRs). Moreover, 28 HECT-type E3 ligases have been identified in human beings. It is well conserved in evolution and is characterized by the direct attachment of ubiquitin to substrates. HECT-type E3 ligase is reported to be involved in a wide range of human diseases and health. The role of HECT-type E3 ligases in the development of cardiac diseases has been uncovered in the last decade. There are only a few review articles summarizing recent advancements regarding HECT-type E3 ligase in the field of cardiac disease. This study focused on cardiac remodeling and described the role of HECT-type E3 ligases in the development of cardiac disease. Moreover, this study revealed that the current knowledge could be exploited for the development of new clinical therapies.

1. Introduction

Despite advances in medicine, cardiovascular disease remains a significant public health problem associated with high mortality [1,2]. Heart failure (HF) is a major cause of cardiovascular deaths. Maladaptive cardiac remodeling caused by hypertension, ischemic heart disease, and other cardiac diseases is accompanied by complex mechanisms that lead to the development of HF [3,4]. Further studies are needed to prevent maladaptive cardiac remodeling and subsequent heart failure.
The modification of eukaryotic proteins with ubiquitin, named ubiquitylation, controls their lifetimes, abundance, localization, interactions, and activities, thereby regulating protein function at all levels. Thus, ubiquitylation plays a pivotal role in a wide range of cellular processes, such as signal transduction, transcriptional regulation, and maintenance of homeostasis. Failing hearts from patients with dilated cardiomyopathy and those with ischemic heart disease show hyper-ubiquitylation compared to donor hearts [5]. An increase in key components of ubiquitylation (ubiquitin, ubiquitin-activating enzyme (E1), ubiquitin-conjugating enzyme (E2), and some ubiquitin ligase (E3)) and a decrease in deubiquitinating enzymes have been observed [5,6,7]. Furthermore, several cardiac diseases, such as cardiac amyloidosis, hypertrophic cardiomyopathy, and hereditary cardiomyopathy, impair the ubiquitin–proteasome system [8,9]. The overall observed change in the ubiquitylation cascade in failing hearts is considered an adaptive response to an increased protein burden derived from increased protein synthesis that accompanies the hypertrophic response or an excess of damaged or modified proteins to be targeted for proteasomal degradation.
Ubiquitin E3 ligase is pivotal in conferring specificity to ubiquitylation and provides particularly interesting targets for therapeutic interventions. In the 2000s, many studies focused on the cardiac ubiquitin E3 ligases to clarify the role of ubiquitylation in the development of cardiac diseases, such as the carboxyl terminus of Hsp70 interacting protein (CHIP), atrogen-1, muscle ring finger (MuRF) family, mouse double mutant 2 homolog (MDM2), cellular inhibitor of apoptosis, casitas b-lineage lymphoma, and E6-associated protein (E6AP) [10,11,12,13,14,15,16,17]. Cardiac ubiquitin E3 ligase plays several roles in protein turnover, energy metabolism, receptor internalization, hypertrophic response, apoptosis, and tolerance to ischemia/reperfusion (I/R) in cardiomyocytes [18,19]. Although elevated expression levels of E6AP were observed in mice after pressure overload [16], the functional role of E6AP has never been examined. Thus, our knowledge of the molecular mechanism of HECT-type E3 ligase in the development of cardiac disease is still lacking.
HECT-type E3 ligase is reported to be involved in a wide range of human diseases and health including neurodegenerative diseases, neurological syndromes, and cancers [20,21,22]. Notably, it is considered as an intriguing target in drug discovery in the context of cancer biology [23]. HECT-type E3 ligases are highly conserved between cells and tissues; as a result, it is tempting to speculate that they also contribute to human cardiac health and disease. The role of HECT-type E3 ligases in the development of cardiac diseases has been examined and uncovered in the last decade. There are only a few review articles summarizing recent advancements regarding HECT-type E3 ligase in the field of cardiac disease [24]. This study focused on cardiac remodeling and described the role of HECT-type E3 ligases in the development of cardiac disease. Moreover, this study revealed that the current knowledge can be exploited for the development of new clinical therapies.

2. Ubiquitylation

2.1. Ubiquitylation

Ubiquitylation is a post-translational modification that covalently conjugates the ubiquitin molecule through the C-terminus to a lysine residue on a substrate protein. Ubiquitylation results in the turnover of the ubiquitylated substrate protein by either the proteasome or lysosome, a change in subcellular localization of the substrate protein, or alteration of substrate protein function [25]. Ubiquitylation is mediated by three enzymes and scaffolding proteins: E1, E2, and E3. There are only few E1s and several E2s; however, E3 ligases constitute a large class of proteins with the human genome encoding more than 600 putative E3 ligases and E3 ligase complexes [26,27,28,29]. Therefore, the specificity of ubiquitylation is determined by the numerous E3 ligases that recognize a specific substrate protein [30]. E3 ligases are also modulators of the rate-limiting step in this enzymatic cascade, participating in substrate protein recognition and catalytic transfer of ubiquitin.
As shown in Figure 1, E3 ligases are classified into three groups: really interesting new genes (RING), homologous to E6AP C-terminus (HECT), and RING-between-RINGs (RBRs). The domain architecture and mechanism of ubiquitylation depend on the class of E3 ligases [31].
Substrate proteins are modified by a single ubiquitin moiety on one or multiple sites, giving rise to mono- and multi-mono-ubiquitylated proteins, respectively. In addition, a wide variety of polyubiquitin chains can be formed on substrate proteins, in which the ubiquitin moieties can be linked through either one of the seven internal lysine residues (Lys6, Lys11, Lys27, Lys29, Lys33, Lys48, and Lys63) in ubiquitin or through its N-terminal amino group.
Polyubiquitylation through the Lsy48-linked ubiquitin chain is generally used for the ubiquitin–proteasomal degradation pathway. Substrate proteins that receive Lys48-linked polyubiquitin chains migrate to and are degraded by the 26S proteasome. The ubiquitin–proteasome system is a protein quality and quantity control system that mediates approximately 80–90% of intracellular protein degradation under optimal nutritional conditions [32,33,34,35,36]. Furthermore, mono-ubiquitylation of lysine residues or polyubiquitylation through Lys63-linked ubiquitin chains are used for nonproteolytic pathways such as DNA repair, relocalization, modifying activity (signal transcriptional activity), or endocytosis [37,38,39]. The consequences for the modified substrate are determined by the type of ubiquitin modification it receives [40].
RING E3 ligases catalyze the direct transfer of ubiquitin from the E2 conjugating enzyme to the substrate, suggesting that the linkage type of the ubiquitin chain is determined by the E2 conjugating enzyme. In contrast to RING-type E3 ligases, HECT-type E3 ligases include an active-site cysteine in the HECT domain, which forms an intermediate thioester bond with ubiquitin before it is conjugated to the substrate protein [41,42]. HECT-type E3 ligase has enzymatic activity and directly catalyzes the covalent attachment of ubiquitin to substrate proteins; therefore, it could determine the linkage type of ubiquitin chain preferred [43].

2.2. HECT-Type E3 Ligase

In 1995, HECT-type E3 ligases were found in all eukaryotic organisms and ubiquitin [41]. E6AP transcribed from the ubiquitin–protein ligase E3A gene was the first identified HECT-type E3 ligase, leading to the discovery of the HECT-type E3 ligase family [41,44,45]. There are 28 types of HECT-type E3 ligases in humans [46], which are commonly grouped into three groups based on the presence of distinct amino acid sequence motifs or domains within the N-terminal: NEDD4 subfamily, HERC subfamily, and other HECT-type E3 ligases [47] (Figure 1). The HECT domain is an approximately 40 kDa domain positioned at the C-terminus of the E3 ligases, consisting of the N-lobe and C-lobe. The N-lobe represents the E2 binding domain, whereas the C-lobe contains an active site cysteine to receive ubiquitin. In the HECT family, 16–92% amino acid identity was found for this domain [48]. The domain architecture of the HECT-type E3 ligases is shown in Figure 1.

2.2.1. NEDD4 Subfamily

The NEDD4 subfamily member includes nine types of HECT-type E3 ligases and accounts for approximately 30% of HECT-type E3 ligases [22]. The NEDD4 family is characterized by the presence of C2 and 2–4 WW domains. The N-terminal C2 domain is defined as a Ca+ phospholipid binder [49]. The WW domains are responsible for recognizing substrates and have also been found to form intramolecular interactions with the HECT domain of the E3 ligases [50,51]. Some NEDD4 subfamily members are often expressed as alternative splice isoforms [47].

2.2.2. HERC Subfamily

The HERC subfamily is characterized by a HECT domain and one or more regulators of chromosome condensation-like domains (RLDs), an effector protein domain that was first identified as a regulator of chromosome condensation 1 [52]. In humans, the HERC subfamily comprises six members, which can be further organized into two large and four small HERCs. Large HERCs (HERC1 and 2) have two or three RLDs; however, small HERCs (HERCs 3, 4, 5, and 6) have one RLD. RLD has dual functions: one side of the domain acts as a guanine nucleotide exchange factor for the small GTPase Ran, whereas the opposite side interacts with chromatin through histones H2A and H2B [53,54].

2.2.3. Other HECT E3s

Each member of another HECT E3 ligase lacks WW or RLD domains and has a distinct variety of N-terminal domains. There are several N-terminal domains of other HECT E3 ligases, such as WWE and armadillo repeats (HUWE1 and TRIP12), UBA domain (HUWE1 and UBR5), ankyrin repeats (HACE1 and HECTD1), and IQ motifs (UBE3B and UBE3C) [55]. In addition, the MIB domain in HECTD1 and the DOC domain in HECTD3 were common in the N-terminus of HERC2.

3. Importance of Ubiquitylation in Cardiac Disease

The adult heart endures a wide range of physiological and pathophysiological stresses during life. Accumulating evidence indicates that ubiquitylation is involved in developing cardiac diseases [17,56,57,58,59,60]. Cardiac proteins are in a dynamic state of continual degradation and resynthesis and are thought to replace all in 30 days under normal circumstances. Protein turnover is critical to cardiomyocytes as post-mitotic cells with minimal regenerative capacity because protein aggravation is cytotoxic [61,62]. Eukaryotic cells have developed multilayered protein quality control mechanisms primarily carried out by chaperones, lysosomal autophagy, and the ubiquitin–proteasome system [63]. More than 70% of the protein turnover is regulated by the ubiquitin–proteasome system [64,65,66]. An experimental study demonstrated that the balance of protein turnover could lead to protein accumulation and aggravation during cardiac remodeling [67]. The discovery that cardiac ubiquitin E3 ligases, such as muscle-specific ubiquitin ligase atrogin-1 and MuRF family, yields cardiac growth, and remodeling through sarcomeric protein turnover, indicated that the ubiquitylation cascade is fundamental to the maintenance of normal cardiac function through protein quality control [17,68,69].
As previously mentioned, ubiquitylation is involved in most aspects of eukaryotic cell biology, such as intracellular signaling, transcriptional control, and regulation of cell death. Research regarding the role of cardiac ubiquitin E3 ligases has developed from protein turnover to cellular processes such as signal transduction, transcriptional regulation, maintenance of homeostasis, mitochondrial dynamics, receptor turnover, and energy metabolism [17,19]. In the following paragraph, we will limit ourselves to a discussion of HECT-type E3 ligases that have been associated with cardiac diseases.

4. Cardiac Hypertrophy and HECT-Type E3 Ligase

Pathological cardiac hypertrophy caused by hypertension, aortic stenosis, and other disease-related stresses is an early milestone during the clinical course of HF [70]. A study detected an accumulation of ubiquitylated proteins and abnormal aggregation in cardiomyocytes collected from patients with decompensated cardiac hypertrophy [71]. Pressure overload-induced cardiac hypertrophy is associated with a marked increase in protein synthesis at a rate exceeding degradation with an increase in ubiquitin, E2, and several E3 ligases [16,72,73,74], indicating hyper-ubiquitylation in rat and mouse models of hypertrophy.
At the cellular level, cardiomyocyte hypertrophy is characterized by an increase in cell size, enhanced protein synthesis, multiplication of sarcomeres, a switch of proteins and enzymes to fetal isoforms, changes in intracellular Ca2+ handling, metabolic alterations, and increased rates of apoptosis [75,76]. Numerous key regulatory pathways that promote cardiac hypertrophy are either targets or components of the ubiquitylation cascade. Several signaling pathways contribute to cardiac hypertrophy, such as Wnt/β catenin, calcineurin/nuclear factor of activated T-cells (NFAT), the Janus kinase (JAK)/signal transduction and the activator of transcription (STAT) pathway [77,78,79].
This study shows the current understanding of HECT-type E3 ligases in the development of cardiac hypertrophy. An overview of HECT-type E3 ligases in cardiac hypertrophy is summarized in Table 1.

4.1. ITCH

The ubiquitin E3 ligase ITCH is a 903 amino acid residue protein that contains an N-terminal C2 domain, four WW domains, and an HECT domain [55]. ITCH was originally identified after genetic analysis of a mutant mouse with aberrant immunological phenotypes and constant skin scratching [94]. The WW domain recognizes the proline-rich PPXY consensus sequence in substrate proteins, and the HECT domain attaches ubiquitin molecules to substrates [95]. Thioredoxin-interacting protein, disheveled (Dvl), c-jun, Jun-B, Notch, Gli, p73, p63, cellular FLICE-inhibitory protein, B cell lymphoma/leukemia 10, Forkhead box O1, transforming growth factor-β-activated kinase 1, and ITCH are known substrates of ITCH for degradative ubiquitylation. Frizzled receptor 4, phospholipase C gamma 1, protein kinase C-θ, Deltex, ErbB4, CXC chemokine receptor 4, and transient receptor potential vanilloid 4 are known substrates of ITCH for degradative ubiquitylation by the non-ubiquitin–proteasome system. Furthermore, small mothers against decapentaplegic (SMAD2) are known substrates of ITCH for non-degradative ubiquitylation [96,97].
Accumulating evidence demonstrates that Wnt/β-catenin signaling is activated after transverse aortic constriction (TAC) and induces pathological cardiac hypertrophy [79,98,99,100]. Dvl is an intracellular principle component of signaling [101]. Dvl has approximately 700 amino acids, harboring conserved DIX, basic and serine threonine-rich region, PDZ, proline-rich PPXY consensus sequence region, and DEP domains. We previously reported that key molecules of the Wnt/β-catenin signaling pathway after TAC are inhibited in cardiac-specific overexpression of ITCH transgenic mice. In this study, ITCH targets Dvl proteins for ubiquitin–proteasome degradation in cardiomyocytes and attenuates cardiac hypertrophy by suppressing the Wnt/β-catenin signaling pathway (Figure 2) [80].
ITCH potentially mediates the degradation of non-Dvl substrates during cardiac hypertrophy [102,103]; therefore, further studies are needed to clarify the effect of ITCH on other substrates during cardiac hypertrophy. Overall, ITCH may be a therapeutic target for cardiac hypertrophy.

4.2. NEDD4-2

NEDD4-2, the most ancient member of the NEDD4 subfamily, was originally identified in a screening of genes downregulated during development of the central nervous system [104]. NEDD4-2 consists of a C2 domain, four WW domains, and an HECT domain. NEDD4-2 is a multifunctional protein whose mutations are associated with development disorders, hypertension, epilepsy, and end-stage renal disease [105,106]. NEDD4-2 has been suggested to function as an E3 ligase for several PY motif-containing proteins such as SMADs, Dvl2, epithelial Na+ channel (ENaC), voltage-dependent cardiac Na+ channel (Nav1.5), KCNQ1 potassium channel [107], and human ether-a-go-go-related gene [108,109].
It was reported that high salt diet-induced cardiac hypertrophy and systolic function were exacerbated in NEDD4-2 deficient mice with higher expression levels of ENaC in the kidney [82]. Furthermore, Galiana-Simal et al. demonstrated that NEDD4-2 is phosphorylated; therefore, NEDD4-2 functions to ubiquitylate ENaC in hypertrophied myocardium of aldosterone-treated rats [81]. These findings suggest an association between NEDD4-2 and cardiac hypertrophy.

4.3. WWP2

The WW domain-containing E3 ubiquitin protein ligase 2 (WWP2), also known as atrophin-1-interacting protein 2, was originally identified in screening for WW domain-containing proteins [110]. WWP2 is ubiquitously expressed in the heart, placenta, lung, liver, muscle, kidney, pancreas, and brain [111]. There are three isoforms in WWP2: a full-length WWP2, an N-terminal isoform presumably generated by failure to splice-out intron 9-10, and a C-terminal isoform possibly generated from a second promoter within introns 10-11. WWP2 interacts with multiple substrates, such as phosphatase and tensin homolog deleted from chromosome 10 (PTEN), SMADs, Oct4, and ENaC [112,113]. WWP2 is a multifunctional E3 ubiquitin ligase, which is involved in palatogenesis, craniofacial development, innate immune response, tumorigenesis, and cell death [114,115,116,117,118,119]. WWP2 was also reported to regulate PTEN/ phosphatidylinositol 3-kinase (PI3K)/Akt signaling and transforming growth factor-beta (TGF-β)/SMAD signaling [119].
Isoproterenol is a chronic infusion of β-stimulant, which induces cardiac hypertrophy and fibrosis, leading to heart failure. Poly(ADP-ribose) polymerase-1 (PARP1) is a critical injury factor in cardiac remodeling. Increases in PARP1 and poly(ADP-ribosyl)ation (PARylatin; PARP1 activity) have been observed in cardiac remodeling, leading to extreme energy consumption by myocardial cells [120,121]. PARP1 is activated to generate PARylation via recognition of damaged DNA fragments and induces cardiomyocyte damage, leading to apoptosis and necrosis [122]. Zhang N et al. reported that full-length WWP2 knockout mice exacerbated cardiac systolic function, hypertrophy, and fibrosis compared to WT mice after isoproterenol infusion. In addition, the protein expression of WWP2 decreased; however, PARP1 increased in response to isoproterenol in vitro and in vivo. WWP2 interacts with PARP1 and degrades it through the ubiquitin–proteasome system by regulating ubiquitylation of PARP1 K418 and K249 sites in the BRCT domain, leading to the inhibition of isoproterenol-induced cardiac remodeling (Figure 3) [84]. WWP2 is a protective factor against isoproterenol-induced cardiac remodeling.

4.4. HUWE1

The ubiquitin E3 ligase HECT, UBA, and WWE domain containing E3 ubiquitin protein ligase 1 (HUWE1), also termed UREB1, HECTH9, ARF-BP1, MULE, E3 Histone, and LASU1, is a 4347 amino acid residue protein that contains an N-terminal armadillo repeat domain, a UBA domain, a WWE domain, a BH3 domain, and a HECT domain [55]. c-Myc, p53, and myeloid cell leukemia-1 are known substrates of HUWE1 for degradative ubiquitylation [123]. In contrast, Dvl and herpesvirus-associated ubiquitin-specific proteases are known substrates of HUWE1 for non-degradative ubiquitylation [124,125]. HUWE1 is associated with proliferation, differentiation, apoptosis, DNA repair, and stress response [124]. HUWE1 has a general preference for attaching the K48 polyubiquitin chain to substrate proteins [126,127].
Recently, Dadson et al. reported that HUWE1 expression was reduced in the left ventricle of patients with end-stage heart failure. Furthermore, conditional cardiac-specific HUWE1 knockout mice develop spontaneous cardiac hypertrophy, left ventricular dysfunction, and premature death with an increase in c-Myc in the heart (Figure 3) [85]. In addition, conditional cardiac-specific HUWE1 knockout mice showed impaired mitochondrial energy metabolism and reactive oxygen species defense, accompanied by reduced protein expression levels of key regulators such as Pgc-1α and Pink1 [128,129,130]. Transcriptomic analysis of HUWE1 knockout mice revealed that elevated c-Myc directly inhibits the transcription of Pgc-1α and Pink1. Cardiac hypertrophy and left ventricular dysfunction were diminished in conditional cardiac-specific HUWE1 and c-Myc double knockout mice. Therefore, HUWE1 in the heart could be a potential therapeutic target for cardiac hypertrophy through its interaction with c-Myc.

4.5. HACE1

The ubiquitin E3 ligase HECT domain and ankyrin repeat containing E3 ubiquitin protein ligase 1 (HACE1) is a 909 amino acid residue protein that contains an N-terminal ankyrin repeat domain and an HECT domain [55]. HACE1 was discovered as a chromosome 6q21 tumor-suppressor gene in Wilms’ tumors [131]. Subsequently, HACE1 is noted as a tumor suppressor in multiple cancers [132]. As a mechanism of tumor suppression, it has been shown to catalyze the ubiquitylation of Ras-related C3 botulinum toxin substrate 1, a potent oncogene [133].
Zhang et al. demonstrated that HACE1 gene expression is upregulated in dilated cardiomyopathy compared with non-failing hearts in silico. They demonstrated that HACE1 deficiency in mice exacerbated heart failure and increased mortality after severe TAC. HACE1 knockout mice show abnormal cardiac hypertrophy, left ventricular dysfunction, accumulation of LC3, p62, and ubiquitylated proteins enriched for cytoskeletal species (Figure 3) [86]. This study reveals that HACE1 is required to fuse autophagosomes with lysosomes, independent of its E3 ligase activity. Hence, HACE1 may play an important role in suppressing protein accumulation and aggravation by regulating autophagy during cardiac remodeling.

4.6. HECTD3

The ubiquitin E3 ligase HECT domain E3 ubiquitin protein ligase 3 (HECTD3) is an 861 amino acid residue protein that contains a DOC domain and an HECT domain. Trio-associated repeats on actin and RAF proto-oncogene serine/threonine-protein kinase are known substrates of HECTD3 for degradative ubiquitylation by the ubiquitin–proteasome system. Furthermore, mucosa-associated lymphoid tissue proteins 1, caspase-8, caspase-9, and STAT3 are substrates of HECTD3 for non-degradative ubiquitylation [134,135].
Small ubiquitin-like modifier 2 (SUMO2) is the most efficient activator of calcineurin/NFAT signaling to induce cardiac hypertrophy [136]. Furthermore, the inflammatory response is associated with myocardial fibrosis and hypertrophy [78,137]. In cardiac and other systems, the JAK/STAT signaling pathway is associated with the inflammatory response [138]. Recently, Rangrez et al. demonstrated that HECTD3 targets SUMO2 for ubiquitin–proteasome degradation and suppresses calcineurin/NFAT signaling (Figure 3). In addition, they showed that HECTD3 reduced the activation of STAT1 by attenuating its phosphorylation through the induction of its polyubiquitylation. In this study, AAV9-mediated overexpression of HECTD3 in mice reduced cardiac SUMO2/STAT1 levels, pathological hypertrophy, largely abolished macrophage infiltration, and fibrosis induced by pressure overload [87]. Thus, HECTD3 may be a potential therapeutic target for cardiac hypertrophy.

5. Cardiac Fibrosis and HECT-Type E3 Ligase

Pathological cardiac fibrosis is a process characterized by excessive deposition of extracellular matrix (ECM), leading to the development of cardiac dysfunction, arrhythmia, and HF [139,140,141]. Several pathophysiological conditions induce cardiac fibrosis, such as pressure overload, volume overload, myocardial infarction, dilated and hypertrophied cardiomyopathy, various toxic insults, metabolic disturbances, and aging [142,143,144,145].
Cardiac fibroblasts are key effector cells in cardiac fibrosis and are responsible for ECM homeostasis in the heart [139]. After cardiac fibroblasts are activated by regulators of tissue fibrosis, such as angiotensin II, connective tissue growth factor, bone morphogenetic protein (BMP), Wnt ligands, cytokines, and TGF-β superfamily, they differentiate into myofibroblasts with an increase in ECM protein [146]. TGF-β1 contributes to cardiac fibrosis development through SMAD-dependent and SMAD-independent pathways. TGF-β1 generally exerts its biological effects by activating downstream mediators, including SMAD2 and SMAD3, while negatively regulated by SMAD7 expression [147,148,149]. SMADs have been reported to play a pivotal role in the transcription of ECM proteins [150].
Although the pathophysiological conditions leading to cardiac fibrosis are different from those of cardiac diseases, it is valuable to explore the common mechanisms involved in cardiac fibrosis. This study shows the current understanding of the role of HECT-type E3 ligases in cardiac fibrosis. An overview of HECT-type E3 ligases in cardiac fibrosis is summarized in Table 1.

5.1. WWP2

An outline of the domain architecture, function, and substrates of WWP2 is described in the previous paragraph. Chen et al. identified the WWP2 N-terminal isoform as a positive regulator of the pro-fibrotic gene network associated with cardiac fibrosis using systems genetics in human and murine dilated cardiomyopathy and repaired tetralogy of Fallot. The WWP2 N-terminal isoform consists of a C2 domain and a WW domain, indicating the absence of the HECT domain. The left ventricular single-cell RNA sequence indicated that WWP2 is mainly expressed in fibroblasts, immune cells, and endothelial cells. WWP2mut/mut mice lacking the N-terminal isoform and full-length WWP2 attenuated cardiac fibrosis and preserved cardiac function after angiotensin II infusion or myocardial infarction. The N-terminal region of WWP2 interacts with SMADs potentially through its mono-ubiquitylation and mediates the TGF-β1-induced nucleocytoplasmic shuttling and transcriptional activity of SMAD2 [88,151]. Thus, WWP2 is an important regulator of pro-fibrotic and ECM genes. These findings provide new understanding into the role of HECT-type E3 ligases independently of the HECT domain.

5.2. SMURF1

SMAD ubiquitin regulatory factor (SMURF) was initially identified as a regulator of SMAD1 stability [152]. SMURFs are composed of two members, SMURF1 and SMURF2. SMURFs have been implicated in determining the competence of cells in response to the TGF-β/BMP signaling pathway [153]. SMURF1 consists of a C2 domain, two WW domains, and an HECT domain. SMURF is a multifunctional protein that is involved in cell cycle progression, cell proliferation, differentiation, DNA damage response, and maintenance of genomic stability. SMURF1 targets Dvl2, SMADs, RhoA, and Runx2, 3 [154]. SMURF1 modulates several signal transduction pathways, such as the TGF-β/BMP signaling pathway, Wnt signaling, mitogen-activated protein kinase signaling, and RhoA/Rho-associated kinase signaling [155]. SMURF1 plays an important role in heart development, including outflow tract septation and cell-type specification, by controlling cilium-associated BMP signaling [156,157].
BMP-2, as a novel fibrosis-antagonizing cytokine, have a potential beneficial effect in attenuating pressure overload-induced cardiac fibrosis. Wang S et al. demonstrated that SMURF1 interacted with SMAD6 and that this SMURF1/SMAD6 complex was involved in BMP2 antagonization of TGF-β1 mediated protein kinase C-δ and SMAD3 signaling in cardiomyocytes [89]. This finding suggests that SMURF1 may contribute to cardiac fibrosis development.
Endothelial colony-forming cells have been reported to reduce cardiac fibrosis in myocardial infarction due to their proliferation and secretion of exosomes, which transfer microRNAs. Cardiac fibroblast activation is ameliorated by exosomes from endothelial colony-forming cells treated with normoxia compared to those treated with hypoxia. Liu et al. found that miR-10b-5p was enriched in exosomes from normoxia and targeted SMURF1 and histone deacetylase 4 using next-generation RNA sequencing. Thus, inhibition of mRNA expression of SMURF1 by miR-10b-5p was suggested to participate in the antifibrotic effects of exosomes derived from endothelial colony-forming cells treated with normoxia [90].

5.3. SMURF2

SMURF2 consists of a C2 domain, three WW domains, and an HECT domain. SMURF2 targets SMADs, heat shock proteins 27, and p53 [154]. SMURF2 was reported to be downregulated in DCM [158]. SMURF functions as a mediator of TGF-β signaling via interaction with SMAD7 containing PY motif during cardiac fibrosis [91].
Meng et al. examined the effect of a SMAD3 inhibitor on angiotensin II-induced cardiac fibrosis. The protein expression level of SMURF2 in the mouse heart increased, while that of SMAD7 decreased after angiotensin II administration. However, this effect was reversed by the SMAD3 inhibitor, suggesting that the SMAD3 inhibitor protected cardiac SMAD7 from SMURF2-mediated ubiquitin–proteasome degradation. Since SMAD7 functions as an inhibitor of both TGF-β/SMAD and NF-κB signaling, an increase in cardiac SMAD7 could be another mechanism through which SMAD3 inhibitor blocked SMAD3-mediated cardiac fibrosis and NF-κB-driven cardiac inflammation [92]. This finding suggests that SMRUF2 may contribute to the development of cardiac fibrosis.

6. HECT-Type E3 Ligase and HF with Preserved Ejection Fraction

HF is classified into three groups: HF with reduced ejection fraction, HF with mid-range ejection fraction, and HF with preserved ejection fraction (HFpEF) [159]. The prevalence rate of HFpEF has been estimated to range from one-third to one-half of all HF patients and is projected to increase [160,161]. Aging is associated with progressive fibrosis, leading to the development of HFpEF [162]. HFpEF in older persons is typified by a broad range of cardiac and non-cardiac abnormalities and reduced reserve capacity in multiple organ systems [163]. To date, there are no approved therapies available for reducing mortality or hospitalization for these patients due to the heterogeneity of HFpEF [164]. This study demonstrates the role of HECT-type E3 ligases in HFpEF development.

WWP1

The WW domain containing E3 ubiquitin protein ligase 1 (WWP1) is a multifunctional protein containing an N-terminal C2 domain, four tandem WW domains for substrate binding, and a C-terminal catalytic HECT domain for ubiquitin transfer. WWP1 was reported to be associated with cancer, aging, neurological disorders, and bone homeostasis [165,166]. WWP1 has been suggested to function as an E3 ligase for several PY motif-containing proteins such as connexin 43, large tumor suppressor 1 and 2, SMAD2, Krüppel-like transcription factor 5, p63, ErbB4/HER4, Runx2, JunB, atrophin-1, and several non-PY motif-containing proteins such as TGF-β receptor 1, SMAD4, Krüppel-like transcription factor 2, and p53 [167]. WWP1 modulates several signal transduction pathways, such as TGF-β signaling, epidermal growth factor signaling, and apoptosis signaling [166].
WWP1 is reported to be highly expressed in the heart [111,168,169] and increases with aging [166]. It was reported that the WWP1 overexpressed mice showed left ventricular hypertrophy, extracellular matrix remodeling, diastolic dysfunction, except systolic dysfunction, indicating the HFpEF phenotype [93]. The precise mechanism by which HFpEF develops has not yet been fully elucidated. Gene analysis data from RNA sequencing using right ventricular endomyocardial biopsies indicated enrichment in mitochondrial adenosine triphosphate synthesis/electron transport and a decrease in endoplasmic reticulum stress, autophagy, and angiogenesis [170]. The NEDD4 family is involved in the development of HFpEF, suggesting the importance of post-translational modification by ubiquitylation in HFpEF as well as HF with reduced ejection fraction. This knowledge provides new insights into HFpEF physiology. WWP1 may have potential therapeutic relevance in the context of HFpEF.

7. Ischemia/Reperfusion Injury and HECT-Type E3 Ligase

Early reperfusion of the ischemic myocardium plays a vital role in minimizing myocardial infarction. However, the effects of reperfusion are complex and include harmful effects, collectively referred to as reperfusion injury [171,172]. The underlying mechanisms of I/R injury are associated with reactive oxygen species generation, intracellular Ca2+ disturbance, rapid pH restoration, and inflammation [173]. Therefore, I/R injury is accompanied by detrimental manifestations, including myocardial necrosis and apoptosis [174]. Cell death contributes to an increase in infarct size, and regulation of this mechanism contributes to improved cardiac function [175]. The ubiquitin–proteasome system plays a pivotal role in I/R injury protection in the heart, organ transplantation, and cerebral ischemia [176,177,178]. CHIP has been reported to be required for cardioprotection after myocardial infarction in mice [179]. Furthermore, MDM2, which targets p53 for degradative ubiquitylation, demonstrated a protective role against hypoxia/reoxygenation-induced cell death [15]. Thus, the role of cardiac E3 ligases in the injured myocardium is important. Recently, some HECT-type E3 ligases have been reported to be involved in I/R injury. This study shows the current understanding of the role of HECT-type E3 ligases in I/R injury. An overview of HECT-type E3 ligases in I/R injury is summarized in Table 2.

7.1. NEDD4-1

Neural precursor cell expressed developmentally downregulated protein 4-1 (NEDD4-1), also termed NEDD4 and RPF1, is a 1319 amino acid residue protein that contains an N-terminal C2 domain, four WW domains, and an HECT domain [116]. In 1992, NEDD4-1 was discovered in mouse neural precursor cells, whose mRNA levels were downregulated during mouse brain development [104]. To date, the role of NEDD4-1 has been identified as an oncogene, tumor suppressor gene, autophagy regulation, and anti-Parkinson’s disease effect [188,189,190,191]. PTEN, RNA polymerase II, and N-Myc are known substrates of NEDD4-1 for degradative ubiquitylation. In contrast, Akt, MDM2, and α-Synuclein are known substrates of NEDD4-1 for non-degradative ubiquitylation [192]. NEDD4-1 is required for heart development [193]. Activation of the PI3K/Akt pathway regulating cellular processes involved in the cell cycle has been shown to protect the heart from I/R injury [194,195]. PTEN is a tumor suppressor that inhibits PI3K/Akt signaling [196]. NEDD4-1 negatively regulates PTEN stability by catalyzing PTEN polyubiquitylation [188]. Furthermore, NEDD4-1 has been shown to positively regulate the nuclear trafficking of phospho-Akt by K63-linked polyubiquitylation [197]. NEDD4-1 was reported to be downregulated in rat hearts after I/R injury [198]. Recently, it was reported that overexpression of NEDD4-1 ameliorated myocardial apoptosis after I/R injury in rats injected with NEDD4-1 lentivirus vector via activation of PI3K/Akt signaling, suggesting the cardioprotective role of NEDD4-1 [180]. However, NEDD4-1 has numerous other substrates that need to be taken into account when interpreting experimental results because it does not directly demonstrate substrates of NEDD4-1 in the heart.

7.2. SMURF2

An outline of the domain architecture, function, and substrates of SMURF2 is described in the previous paragraph. Enhancer of zeste homolog 2 (EZH2) is reported to be one of the SMURF2 substrates. EZH2 is a polycomb group protein associated with pivotal functions, including cell division, embryonic development, and cancer development [199]. EZH2 binds to PTEN, negatively regulates its expression, and upregulates PI3K/Akt signaling [200]. Recently, Dong et al. demonstrated that miR-322/503 plays a beneficial role in myocardial I/R injury. Inhibition of SMURF2 translation induces EZH2 expression and activates PI3K/Akt signaling via miR-322/503, thereby protecting cells from I/R injury [181]. Therefore, SMURF2 might be a potential target for improving the prognosis of myocardial I/R injury.

8. Other Cardiac Disease and HECT-Type E3 Ligase

An overview of HECT-type E3 ligases in other cardiac diseases such as doxorubicin cardiotoxicity and arrhythmia is briefly summarized in Table 2.

8.1. ITCH and Doxorubicin Cardiotoxicity

The outline of the domain architecture, function, and substrates of ITCH is described in the previous paragraph. Doxorubicin is one of the most widely used anticancer therapies for malignant lymphoma (CHOP therapy), breast cancer (AC therapy), and uterine cancer (AP therapy) [201,202,203]. Although doxorubicin has a significant effect on reducing mortality of these cancer patients, its cumulative and dose-dependent toxicity harms the heart [204]. The mechanisms of doxorubicin cardiotoxicity are involved in inflammation, oxidative stress, apoptosis, mitochondrial impairment, and dysregulation of autophagy [205]. We previously reported that ITCH targets the thioredoxin-interacting protein for ubiquitin–proteasome degradation in cardiomyocytes and ameliorates cardiotoxicity induced by reactive oxygen species, including doxorubicin cardiotoxicity and myocardial infarction (Figure 2). After doxorubicin injection or myocardial infarction, the survival rates were significantly higher in cardiac-specific overexpression of ITCH transgenic mice than in WT littermates [182]. In addition, Zhang et al. demonstrated that miR-34b/c targeted ITCH and suppressed its expression, promoting NFκB and subsequent inflammatory cytokine expression in doxorubicin-treated HL-1 cells [183].
Circular RNAs (circRNAs) are important modulators of cardiac development and disease. Recently, it was reported that circular RNA ITCH derived from 7–14 exons in ITCH acts as a sponge of miR-330-5p, thereby upregulating sirtuin 6, baculoviral IAP repeat containing 5, and ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporting 2 to alleviate doxorubicin cardiotoxicity [185] (Figure 2). Therefore, ITCH and circular RNA ITCH have cardioprotective roles against doxorubicin and could be novel therapeutic targets in doxorubicin cardiotoxicity.

8.2. NEDD4-2 and Arrhythmia

An outline of the domain architecture, function, and substrates of NEDD4-2 is described in the previous paragraph. The activity of NEDD4-2 is regulated by Ca2+ concentration in cells, and an increase in Ca2+ concentration disrupts the autoinhibitory conformation of NEDD4-2, leading to its activation [206].
Nav1.5, which is responsible for the action potential upstroke, is essential for maintaining an adequate conduction velocity of the electrical impulse [207]. It has been reported that protein expression of Nav1.5 is reduced in HF without any change in its mRNA level [208]. Luo et al. uncovered the mechanism of downregulating Nav1.5 in HF. The protein expression of Nav1.5 decreased, while that of NEDD4-2 increased after ionomycin-induced intracellular Ca2+ increase. This was reversed by the calcium chelator BAPTA-AM, suggesting an inverse relationship between Nav1.5 and NEDD4-2 expression in neonatal rat cardiomyocytes. Nav1.5 was ubiquitylated and downregulated in NEDD4-2 transfected neonatal rat cardiomyocytes. Reduced expression of Nav1.5 and augmented expression of NEDD4-2 were observed in a volume overload rat heart failure model and cardiomyocytes treated with isoproterenol and angiotensin II. Therefore, calcium-mediated increase in NEDD4-2 induced Nav1.5 ubiquitylation with a resultant reduction in Nav1.5 cardiomyocyte membrane density in HF [186]. Mutations in the gene encoding Nav1.5, SCN5A, have been associated with various arrhythmic disorders such as long QT syndrome, Brugada syndrome, and sick sinus syndrome, indicating the association of NEDD4-2 with cardiac arrhythmia [209,210].

8.3. WWP1 and Arrhythmia

An outline of the domain architecture, function, and substrates of WWP1 is described in the previous paragraph. Sudden cardiac death is a tragedy and the most common cause of death worldwide. The cause of sudden cardiac death in most instances is ventricular arrhythmias such as ventricular fibrillation and ventricular tachycardia [211,212]. Connexin 43 is a main component of gap junctions in the ventricular myocardium and has a PY motif in its C-terminus. Reduced expression or altered subcellular localization of connexin 43 has been reported to be associated with contractile dysfunction, aging, and arrhythmia [213,214]. Basheer et al. reported that WWP1 targets connexin 43 for ubiquitin–proteasome degradation. Both global- and cardiomyocyte-specific overexpression of WWP1 mice die around 8 weeks of age due to lethal ventricular arrhythmia accompanied by a dramatic reduction in connexin 43. Therefore, these findings suggest an association between WWP1 and ventricular arrhythmia [187].

9. Drug Discovery

The most successful clinically applied drug in this field is a proteasome inhibitor for multiple myeloma [215]. However, proteasome inhibitors attenuate cardiac remodeling after pressure overload [73,216], it showed unexpected cardiac complications, including HF [217]. Therefore, the clinical application of proteasome inhibitors for cardiac diseases appears to be limited. Since E3 ligases are often focal points of cellular regulation [218,219,220], targeting E3 ligases is thought to yield higher specificity and less toxicity than other ubiquitylation cascades [221,222]. HECT-type E3 ligases have recently been described as druggable by peptide and small molecule inhibitors, and several compounds targeting NEDD4-1, NEDD subfamily, ITCH, SMURF1, E6AP, and HUWE1 have been identified [223].
Rossi et al. identified clomipramine, a commonly used drug in treating depression, as an inhibitor of ITCH, by using high-throughput screening of ITCH auto-ubiquitylation [224]. Clomipramine binds to the SH residue irreversibly in the C-lobe of the HECT domain required for ubiquitin translocation. The therapeutic potential of clomipramine in cancer has also been examined [225].
The E3 ligase activity of ITCH is modulated by several factors such as intra- and intermolecular interactions, post-translational modification, adaptor proteins, and ITCH expression itself [80,182,226,227]. Lithium is widely used as a drug for acute mania in bipolar disorder [228]. Recently, Wang X et al. demonstrated that lithium induces upregulation of ITCH and promotes ubiquitylation and degradation of Gli1, a regulator of the Hedgehog signaling pathway, in PANC-1 cells [229]. A recent report demonstrated that low-dose lithium feeding increases sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2a (SERCA2a) to phospholamban ratio and improved SERCA function in the murine left ventricle [230], suggesting a protective role in the heart. Although the precise mechanism by which lithium upregulates ITCH is unclear, there is a possibility that ITCH target therapy is beneficial in the heart. Further studies are required to examine whether HECT-type E3 ligase target drugs can be clinically applied to treat cardiac disease.

10. Limitation

This study shows the current understanding of the role of HECT-type E3 ligases in the development of cardiac disease. However, this study had several limitations. First, only a limited number of HECT-type E3 ligases have been examined to date. There have been no reports regarding the HERC subfamily in the field of cardiac disease. Second, previous studies regarding HECT-type E3 ligases in cardiac hypertrophy were limited to left ventricular hypertrophy. Recent research highlighted the importance of right ventricular hypertrophy and suggested that the ubiquitin–proteasome system is activated in the right ventricle [231]; therefore, further studies are required to elucidate the role of HCET-type E3 ligase in cardiac hypertrophy. Third, there is still no evidence for the clinical application of drugs targeting HECT-type E3 ligases in cardiac disease. However, preclinical evidence published to date supports the idea that HECT-type E3 ligases in the myocardium may be an effective interventional strategy for cardiac disease. Finally, although our understanding of HECT-type E3 ligase in cardiac disease is developing, many more are yet to be found.

11. Conclusions

Regulation of HECT-type E3 ligases plays an important role in determining cell fate, and HECT-type E3 ligase function is implicated in cardiac disease. Recent experimental studies have emphasized the role of HECT-type E3 ligases in the development of cardiac remodeling.
Cardiac hypertrophy and fibrosis have been proposed as important therapeutic targets in patients with HF [232], the identification of druggable targets that regulate maladaptive remodeling may provide a new avenue to control the progression of HF. HECT-type E3 ligases are often the focal point of cellular regulation; this feature makes them attractive as a therapeutic target. The continuation of research to elucidate HECT-type E3 ligases is critical for increasing our knowledge and discovering new therapeutic targets for the myriad of cardiac diseases that plague humans.

Author Contributions

Conceptualization, Y.O.; writing—original draft preparation, J.G. and Y.O.; writing—review and editing, T.W. and M.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviation

BMPbone morphogenetic protein
CHIPcarboxyl terminus of Hsp70 interacting protein
Dvldishevelled
ECMextracellular matrix
E6APE6-associated protein
EZH2enhancer of zeste homolog 2
HACEHECT domain and ankyrin repeat containing E3 ubiquitin protein ligase
HECThomologous to E6AP C-terminus
HECTDHECT domain E3 ubiquitin protein ligase
HERCHECT and RLD domain containing E3 ubiquitin protein ligase
HFheart failure
HFpEFheart failure with preserved ejection fraction
HUWEHECT, UBA and WWE domain containing E3 ubiquitin protein ligase
I/Rischemia/reperfusion
JAKJanus kinase
MDM2mouse double mutant 2 homolog
MuRFmuscle ring finger
Nav1.5voltage-dependent Na+ channel
NEDD4neural precursor cell expressed developmentally downregulated protein 4
NFATnuclear factor of activated T-cells
NF-nuclear factor kappa B
PARP1poly(ADP-ribose) polymerase-1
PI3Kphosphatidylinositol 3-kinase
PTENphosphatase and tensin homolog deleted from chromosome 10
RBRsRING-between-RINGs
RINGreally interesting new genes
RLDsregulator of chromosome condensation-like domains
SERCA2sarcoplasmic/endoplasmic reticulum calcium ATPase 2
SMADsmall mother against decapentaplegic
SMURFSMAD specific E3 ubiquitin regulatory factor
STATsignal transduction and activator of transcription
SUMO2small ubiquitin-like modifier 2
TACtransverse aortic constriction
TGF-βtransforming growth factor beta
TRIP12thyroid hormone receptor interactor 12
UBAubiquitin-associated
UBEubiquitin–protein ligase E3
UBR5ubiquitin protein ligase E3 component N-recognin 5
WTwild type
WWPWW domain containing E3 ubiquitin protein ligase

References

  1. Chen, J.; Normand, S.L.; Wang, Y.; Krumholz, H.M. National and regional trends in heart failure hospitalization and mortality rates for Medicare beneficiaries, 1998–2008. JAMA 2011, 306, 1669–1678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Lloyd-Jones, D.M.; Larson, M.G.; Leip, E.P.; Beiser, A.; D’Agostino, R.B.; Kannel, W.B.; Murabito, J.M.; Vasan, R.S.; Benjamin, E.J.; Levy, D. Lifetime risk for developing congestive heart failure: The Framingham Heart Study. Circulation 2002, 106, 3068–3072. [Google Scholar] [CrossRef] [Green Version]
  3. Nakamura, M.; Sadoshima, J. Mechanisms of physiological and pathological cardiac hypertrophy. Nat. Rev. Cardiol. 2018, 15, 387–407. [Google Scholar] [CrossRef]
  4. Shimizu, I.; Minamino, T. Physiological and pathological cardiac hypertrophy. J. Mol. Cell Cardiol. 2016, 97, 245–262. [Google Scholar] [CrossRef] [PubMed]
  5. Weekes, J.; Morrison, K.; Mullen, A.; Wait, R.; Barton, P.; Dunn, M.J. Hyperubiquitination of proteins in dilated cardiomyopathy. Proteomics 2003, 3, 208–216. [Google Scholar] [CrossRef]
  6. Birks, E.J.; Latif, N.; Enesa, K.; Folkvang, T.; Luong, L.A.; Sarathchandra, P.; Khan, M.; Ovaa, H.; Terracciano, C.M.; Barton, P.J.; et al. Elevated p53 expression is associated with dysregulation of the ubiquitin-proteasome system in dilated cardiomyopathy. Cardiovasc. Res. 2008, 79, 472–480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Kostin, S.; Pool, L.; Elsasser, A.; Hein, S.; Drexler, H.C.; Arnon, E.; Hayakawa, Y.; Zimmermann, R.; Bauer, E.; Klovekorn, W.P.; et al. Myocytes die by multiple mechanisms in failing human hearts. Circ. Res. 2003, 92, 715–724. [Google Scholar] [CrossRef] [Green Version]
  8. Schlossarek, S.; Frey, N.; Carrier, L. Ubiquitin-proteasome system and hereditary cardiomyopathies. J. Mol. Cell Cardiol. 2014, 71, 25–31. [Google Scholar] [CrossRef] [Green Version]
  9. Predmore, J.M.; Wang, P.; Davis, F.; Bartolone, S.; Westfall, M.V.; Dyke, D.B.; Pagani, F.; Powell, S.R.; Day, S.M. Ubiquitin proteasome dysfunction in human hypertrophic and dilated cardiomyopathies. Circulation 2010, 121, 997–1004. [Google Scholar] [CrossRef] [Green Version]
  10. Naito, A.T.; Okada, S.; Minamino, T.; Iwanaga, K.; Liu, M.L.; Sumida, T.; Nomura, S.; Sahara, N.; Mizoroki, T.; Takashima, A.; et al. Promotion of CHIP-mediated p53 degradation protects the heart from ischemic injury. Circ. Res. 2010, 106, 1692–1702. [Google Scholar] [CrossRef] [PubMed]
  11. Schisler, J.C.; Rubel, C.E.; Zhang, C.; Lockyer, P.; Cyr, D.M.; Patterson, C. CHIP protects against cardiac pressure overload through regulation of AMPK. J. Clin. Investig. 2013, 123, 3588–3599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Le, N.T.; Takei, Y.; Shishido, T.; Woo, C.H.; Chang, E.; Heo, K.S.; Lee, H.; Lu, Y.; Morrell, C.; Oikawa, M.; et al. p90RSK targets the ERK5-CHIP ubiquitin E3 ligase activity in diabetic hearts and promotes cardiac apoptosis and dysfunction. Circ. Res. 2012, 110, 536–550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Arya, R.; Kedar, V.; Hwang, J.R.; McDonough, H.; Li, H.H.; Taylor, J.; Patterson, C. Muscle ring finger protein-1 inhibits PKC{epsilon} activation and prevents cardiomyocyte hypertrophy. J. Cell Biol. 2004, 167, 1147–1159. [Google Scholar] [CrossRef]
  14. Willis, M.S.; Schisler, J.C.; Li, L.; Rodriguez, J.E.; Hilliard, E.G.; Charles, P.C.; Patterson, C. Cardiac muscle ring finger-1 increases susceptibility to heart failure in vivo. Circ. Res. 2009, 105, 80–88. [Google Scholar] [CrossRef] [Green Version]
  15. Toth, A.; Nickson, P.; Qin, L.L.; Erhardt, P. Differential regulation of cardiomyocyte survival and hypertrophy by MDM2, an E3 ubiquitin ligase. J. Biol. Chem. 2006, 281, 3679–3689. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Balasubramanian, S.; Mani, S.; Shiraishi, H.; Johnston, R.K.; Yamane, K.; Willey, C.D.; Cooper, G., 4th; Tuxworth, W.J.; Kuppuswamy, D. Enhanced ubiquitination of cytoskeletal proteins in pressure overloaded myocardium is accompanied by changes in specific E3 ligases. J. Mol. Cell Cardiol. 2006, 41, 669–679. [Google Scholar] [CrossRef]
  17. Willis, M.S.; Bevilacqua, A.; Pulinilkunnil, T.; Kienesberger, P.; Tannu, M.; Patterson, C. The role of ubiquitin ligases in cardiac disease. J. Mol. Cell Cardiol. 2014, 71, 43–53. [Google Scholar] [CrossRef] [Green Version]
  18. Zolk, O.; Schenke, C.; Sarikas, A. The ubiquitin-proteasome system: Focus on the heart. Cardiovasc. Res. 2006, 70, 410–421. [Google Scholar] [CrossRef] [Green Version]
  19. Parry, T.L.; Willis, M.S. Cardiac ubiquitin ligases: Their role in cardiac metabolism, autophagy, cardioprotection and therapeutic potential. Biochim. Biophys. Acta 2016, 1862, 2259–2269. [Google Scholar] [CrossRef]
  20. Wang, Y.; Argiles-Castillo, D.; Kane, E.I.; Zhou, A.; Spratt, D.E. HECT E3 ubiquitin ligases-emerging insights into their biological roles and disease relevance. J. Cell Sci. 2020, 133. [Google Scholar] [CrossRef]
  21. Mao, X.; Sethi, G.; Zhang, Z.; Wang, Q. The Emerging Roles of the HERC Ubiquitin Ligases in Cancer. Curr. Pharm. Des. 2018, 24, 1676–1681. [Google Scholar] [CrossRef]
  22. Scheffner, M.; Kumar, S. Mammalian HECT ubiquitin-protein ligases: Biological and pathophysiological aspects. Biochim. Biophys. Acta 2014, 1843, 61–74. [Google Scholar] [CrossRef]
  23. Weber, J.; Polo, S.; Maspero, E. HECT E3 Ligases: A Tale With Multiple Facets. Front. Physiol. 2019, 10, 370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Zhang, Y.; Qian, H.; Wu, B.; You, S.; Wu, S.; Lu, S.; Wang, P.; Cao, L.; Zhang, N.; Sun, Y. E3 Ubiquitin ligase NEDD4 familyregulatory network in cardiovascular disease. Int. J. Biol. Sci. 2020, 16, 2727–2740. [Google Scholar] [CrossRef]
  25. Popovic, D.; Vucic, D.; Dikic, I. Ubiquitination in disease pathogenesis and treatment. Nat. Med. 2014, 20, 1242–1253. [Google Scholar] [CrossRef]
  26. Metzger, M.B.; Hristova, V.A.; Weissman, A.M. HECT and RING finger families of E3 ubiquitin ligases at a glance. J. Cell Sci. 2012, 125, 531–537. [Google Scholar] [CrossRef] [Green Version]
  27. Varshavsky, A. The ubiquitin system, an immense realm. Annu. Rev. Biochem. 2012, 81, 167–176. [Google Scholar] [CrossRef]
  28. Kerscher, O.; Felberbaum, R.; Hochstrasser, M. Modification of proteins by ubiquitin and ubiquitin-like proteins. Annu. Rev. Cell Dev. Biol. 2006, 22, 159–180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Hutchins, A.P.; Liu, S.; Diez, D.; Miranda-Saavedra, D. The repertoires of ubiquitinating and deubiquitinating enzymes in eukaryotic genomes. Mol. Biol. Evol. 2013, 30, 1172–1187. [Google Scholar] [CrossRef] [Green Version]
  30. Hicke, L.; Dunn, R. Regulation of membrane protein transport by ubiquitin and ubiquitin-binding proteins. Annu. Rev. Cell Dev. Biol. 2003, 19, 141–172. [Google Scholar] [CrossRef] [PubMed]
  31. Zheng, N.; Shabek, N. Ubiquitin Ligases: Structure, Function, and Regulation. Annu. Rev. Biochem. 2017, 86, 129–157. [Google Scholar] [CrossRef]
  32. Lee, D.H.; Goldberg, A.L. Proteasome inhibitors: Valuable new tools for cell biologists. Trends Cell Biol. 1998, 8, 397–403. [Google Scholar] [CrossRef]
  33. Lyon, R.C.; Lange, S.; Sheikh, F. Breaking down protein degradation mechanisms in cardiac muscle. Trends Mol. Med. 2013, 19, 239–249. [Google Scholar] [CrossRef] [Green Version]
  34. Portbury, A.L.; Willis, M.S.; Patterson, C. Tearin’ up my heart: Proteolysis in the cardiac sarcomere. J. Biol. Chem. 2011, 286, 9929–9934. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Goldberg, A.L. Protein degradation and protection against misfolded or damaged proteins. Nature 2003, 426, 895–899. [Google Scholar] [CrossRef]
  36. Schubert, U.; Antón, L.C.; Gibbs, J.; Norbury, C.C.; Yewdell, J.W.; Bennink, J.R. Rapid degradation of a large fraction of newly synthesized proteins by proteasomes. Nature 2000, 404, 770–774. [Google Scholar] [CrossRef] [PubMed]
  37. Peralta, D.A.; Araya, A.; Busi, M.V.; Gomez-Casati, D.F. The E3 ubiquitin-ligase SEVEN IN ABSENTIA like 7 mono-ubiquitinates glyceraldehyde-3-phosphate dehydrogenase 1 isoform in vitro and is required for its nuclear localization in Arabidopsis thaliana. Int. J. Biochem. Cell Biol. 2016, 70, 48–56. [Google Scholar] [CrossRef] [PubMed]
  38. Xie, F.; Zhang, Z.; van Dam, H.; Zhang, L.; Zhou, F. Regulation of TGF-β Superfamily Signaling by SMAD Mono-Ubiquitination. Cells 2014, 3, 981–993. [Google Scholar] [CrossRef]
  39. Welchman, R.L.; Gordon, C.; Mayer, R.J. Ubiquitin and ubiquitin-like proteins as multifunctional signals. Nat. Rev. Mol. Cell Biol. 2005, 6, 599–609. [Google Scholar] [CrossRef] [PubMed]
  40. Komander, D.; Rape, M. The ubiquitin code. Annu. Rev. Biochem. 2012, 81, 203–229. [Google Scholar] [CrossRef] [Green Version]
  41. Huibregtse, J.M.; Scheffner, M.; Beaudenon, S.; Howley, P.M. A family of proteins structurally and functionally related to the E6-AP ubiquitin-protein ligase. Proc. Natl. Acad. Sci. USA 1995, 92, 2563–2567. [Google Scholar] [CrossRef] [Green Version]
  42. Scheffner, M.; Nuber, U.; Huibregtse, J.M. Protein ubiquitination involving an E1-E2-E3 enzyme ubiquitin thioester cascade. Nature 1995, 373, 81–83. [Google Scholar] [CrossRef] [Green Version]
  43. Kim, H.C.; Huibregtse, J.M. Polyubiquitination by HECT E3s and the determinants of chain type specificity. Mol. Cell Biol. 2009, 29, 3307–3318. [Google Scholar] [CrossRef] [Green Version]
  44. Huibregtse, J.M.; Scheffner, M.; Howley, P.M. A cellular protein mediates association of p53 with the E6 oncoprotein of human papillomavirus types 16 or 18. EMBO J. 1991, 10, 4129–4135. [Google Scholar] [CrossRef]
  45. Scheffner, M.; Huibregtse, J.M.; Vierstra, R.D.; Howley, P.M. The HPV-16 E6 and E6-AP complex functions as a ubiquitin-protein ligase in the ubiquitination of p53. Cell 1993, 75, 495–505. [Google Scholar] [CrossRef]
  46. Scheffner, M.; Staub, O. HECT E3s and human disease. BMC Biochem. 2007, 8 (Suppl. 1), S6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Rotin, D.; Kumar, S. Physiological functions of the HECT family of ubiquitin ligases. Nat. Rev. Mol. Cell Biol. 2009, 10, 398–409. [Google Scholar] [CrossRef] [PubMed]
  48. Zhang, W.; Wu, K.P.; Sartori, M.A.; Kamadurai, H.B.; Ordureau, A.; Jiang, C.; Mercredi, P.Y.; Murchie, R.; Hu, J.; Persaud, A.; et al. System-Wide Modulation of HECT E3 Ligases with Selective Ubiquitin Variant Probes. Mol. Cell 2016, 62, 121–136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Rizo, J.; Sudhof, T.C. C2-domains, structure and function of a universal Ca2+-binding domain. J. Biol. Chem. 1998, 273, 15879–15882. [Google Scholar] [CrossRef] [Green Version]
  50. Ingham, R.J.; Colwill, K.; Howard, C.; Dettwiler, S.; Lim, C.S.; Yu, J.; Hersi, K.; Raaijmakers, J.; Gish, G.; Mbamalu, G.; et al. WW domains provide a platform for the assembly of multiprotein networks. Mol. Cell Biol. 2005, 25, 7092–7106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Riling, C.; Kamadurai, H.; Kumar, S.; O’Leary, C.E.; Wu, K.P.; Manion, E.E.; Ying, M.; Schulman, B.A.; Oliver, P.M. Itch WW Domains Inhibit Its E3 Ubiquitin Ligase Activity by Blocking E2-E3 Ligase Trans-thiolation. J. Biol. Chem. 2015, 290, 23875–23887. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Bischoff, F.R.; Ponstingl, H. Catalysis of guanine nucleotide exchange on Ran by the mitotic regulator RCC1. Nature 1991, 354, 80–82. [Google Scholar] [CrossRef] [PubMed]
  53. Zhang, C.; Clarke, P.R. Chromatin-independent nuclear envelope assembly induced by Ran GTPase in Xenopus egg extracts. Science 2000, 288, 1429–1432. [Google Scholar] [CrossRef] [PubMed]
  54. Nemergut, M.E.; Mizzen, C.A.; Stukenberg, T.; Allis, C.D.; Macara, I.G. Chromatin docking and exchange activity enhancement of RCC1 by histones H2A and H2B. Science 2001, 292, 1540–1543. [Google Scholar] [CrossRef] [PubMed]
  55. Sluimer, J.; Distel, B. Regulating the human HECT E3 ligases. Cell Mol. Life Sci. 2018, 75, 3121–3141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Willis, M.S.; Patterson, C. Proteotoxicity and cardiac dysfunction--Alzheimer’s disease of the heart? N. Engl. J. Med. 2013, 368, 455–464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Patterson, C.; Ike, C.; Willis, P.W., 4th; Stouffer, G.A.; Willis, M.S. The bitter end: The ubiquitin-proteasome system and cardiac dysfunction. Circulation 2007, 115, 1456–1463. [Google Scholar] [CrossRef] [Green Version]
  58. Willis, M.S.; Patterson, C. Into the heart: The emerging role of the ubiquitin-proteasome system. J. Mol. Cell Cardiol. 2006, 41, 567–579. [Google Scholar] [CrossRef]
  59. Powell, S.R. The ubiquitin-proteasome system in cardiac physiology and pathology. Am. J. Physiol. Heart Circ. Physiol. 2006, 291, H1–H19. [Google Scholar] [CrossRef] [Green Version]
  60. Brown, D.I.; Parry, T.L.; Willis, M.S. Ubiquitin Ligases and Posttranslational Regulation of Energy in the Heart: The Hand that Feeds. Compr. Physiol. 2017, 7, 841–862. [Google Scholar] [CrossRef]
  61. Kelly, S.M.; Vanslyke, J.K.; Musil, L.S. Regulation of ubiquitin-proteasome system mediated degradation by cytosolic stress. Mol. Biol. Cell 2007, 18, 4279–4291. [Google Scholar] [CrossRef] [Green Version]
  62. Bucciantini, M.; Calloni, G.; Chiti, F.; Formigli, L.; Nosi, D.; Dobson, C.M.; Stefani, M. Prefibrillar amyloid protein aggregates share common features of cytotoxicity. J. Biol. Chem. 2004, 279, 31374–31382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Grune, T.; Merker, K.; Sandig, G.; Davies, K.J. Selective degradation of oxidatively modified protein substrates by the proteasome. Biochem. Biophys. Res. Commun. 2003, 305, 709–718. [Google Scholar] [CrossRef]
  64. Young, G.W.; Wang, Y.; Ping, P. Understanding proteasome assembly and regulation: Importance to cardiovascular medicine. Trends Cardiovasc. Med. 2008, 18, 93–98. [Google Scholar] [CrossRef] [Green Version]
  65. Su, H.; Wang, X. The ubiquitin-proteasome system in cardiac proteinopathy: A quality control perspective. Cardiovasc. Res. 2010, 85, 253–262. [Google Scholar] [CrossRef] [Green Version]
  66. Dantuma, N.P.; Lindsten, K. Stressing the ubiquitin-proteasome system. Cardiovasc. Res. 2010, 85, 263–271. [Google Scholar] [CrossRef] [Green Version]
  67. Tsukamoto, O.; Minamino, T.; Kitakaze, M. Functional alterations of cardiac proteasomes under physiological and pathological conditions. Cardiovasc. Res. 2010, 85, 339–346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Bodine, S.C.; Latres, E.; Baumhueter, S.; Lai, V.K.; Nunez, L.; Clarke, B.A.; Poueymirou, W.T.; Panaro, F.J.; Na, E.; Dharmarajan, K.; et al. Identification of ubiquitin ligases required for skeletal muscle atrophy. Science 2001, 294, 1704–1708. [Google Scholar] [CrossRef]
  69. Li, H.H.; Kedar, V.; Zhang, C.; McDonough, H.; Arya, R.; Wang, D.Z.; Patterson, C. Atrogin-1/muscle atrophy F-box inhibits calcineurin-dependent cardiac hypertrophy by participating in an SCF ubiquitin ligase complex. J. Clin. Investig. 2004, 114, 1058–1071. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Aimo, A.; Gaggin, H.K.; Barison, A.; Emdin, M.; Januzzi, J.L., Jr. Imaging, Biomarker, and Clinical Predictors of Cardiac Remodeling in Heart Failure With Reduced Ejection Fraction. JACC Heart Fail. 2019, 7, 782–794. [Google Scholar] [CrossRef] [PubMed]
  71. Hein, S.; Arnon, E.; Kostin, S.; Schönburg, M.; Elsässer, A.; Polyakova, V.; Bauer, E.P.; Klövekorn, W.P.; Schaper, J. Progression from compensated hypertrophy to failure in the pressure-overloaded human heart: Structural deterioration and compensatory mechanisms. Circulation 2003, 107, 984–991. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Razeghi, P.; Baskin, K.K.; Sharma, S.; Young, M.E.; Stepkowski, S.; Essop, M.F.; Taegtmeyer, H. Atrophy, hypertrophy, and hypoxemia induce transcriptional regulators of the ubiquitin proteasome system in the rat heart. Biochem. Biophys. Res. Commun. 2006, 342, 361–364. [Google Scholar] [CrossRef]
  73. Depre, C.; Wang, Q.; Yan, L.; Hedhli, N.; Peter, P.; Chen, L.; Hong, C.; Hittinger, L.; Ghaleh, B.; Sadoshima, J.; et al. Activation of the cardiac proteasome during pressure overload promotes ventricular hypertrophy. Circulation 2006, 114, 1821–1828. [Google Scholar] [CrossRef] [Green Version]
  74. Sugden, P.H.; Clerk, A. Cellular mechanisms of cardiac hypertrophy. J. Mol. Med. 1998, 76, 725–746. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Allard, M.F.; Schonekess, B.O.; Henning, S.L.; English, D.R.; Lopaschuk, G.D. Contribution of oxidative metabolism and glycolysis to ATP production in hypertrophied hearts. Am. J. Physiol. 1994, 267, H742–H750. [Google Scholar] [CrossRef] [PubMed]
  76. Friehs, I.; Barillas, R.; Vasilyev, N.V.; Roy, N.; McGowan, F.X.; del Nido, P.J. Vascular endothelial growth factor prevents apoptosis and preserves contractile function in hypertrophied infant heart. Circulation 2006, 114, I290–I295. [Google Scholar] [CrossRef] [Green Version]
  77. Rohini, A.; Agrawal, N.; Koyani, C.N.; Singh, R. Molecular targets and regulators of cardiac hypertrophy. Pharmacol. Res. 2010, 61, 269–280. [Google Scholar] [CrossRef] [PubMed]
  78. Booz, G.W.; Day, J.N.; Baker, K.M. Interplay between the cardiac renin angiotensin system and JAK-STAT signaling: Role in cardiac hypertrophy, ischemia/reperfusion dysfunction, and heart failure. J. Mol. Cell Cardiol. 2002, 34, 1443–1453. [Google Scholar] [CrossRef]
  79. Bergmann, M.W. WNT signaling in adult cardiac hypertrophy and remodeling: Lessons learned from cardiac development. Circ. Res. 2010, 107, 1198–1208. [Google Scholar] [CrossRef] [Green Version]
  80. Goto, J.; Otaki, Y.; Watanabe, T.; Kobayashi, Y.; Aono, T.; Watanabe, K.; Wanezaki, M.; Kutsuzawa, D.; Kato, S.; Tamura, H.; et al. HECT (Homologous to the E6-AP Carboxyl Terminus)-Type Ubiquitin E3 Ligase ITCH Attenuates Cardiac Hypertrophy by Suppressing the Wnt/beta-Catenin Signaling Pathway. Hypertension 2020, 76, 1868–1878. [Google Scholar] [CrossRef]
  81. Galiana-Simal, A.; Olivares-Alvaro, E.; Klett-Mingo, M.; Ruiz-Roso, M.B.; Ballesteros, S.; de Las Heras, N.; Fuller, P.J.; Lahera, V.; Martin-Fernandez, B. Proanthocyanidins block aldosterone-dependent up-regulation of cardiac gamma ENaC and Nedd4-2 inactivation via SGK1. J. Nutr. Biochem. 2016, 37, 13–19. [Google Scholar] [CrossRef]
  82. Shi, P.P.; Cao, X.R.; Sweezer, E.M.; Kinney, T.S.; Williams, N.R.; Husted, R.F.; Nair, R.; Weiss, R.M.; Williamson, R.A.; Sigmund, C.D.; et al. Salt-sensitive hypertension and cardiac hypertrophy in mice deficient in the ubiquitin ligase Nedd4-2. Am. J. Physiol. Renal. Physiol. 2008, 295, F462–F470. [Google Scholar] [CrossRef]
  83. Yang, M.H.; Wang, H.; Han, S.N.; Jia, X.; Zhang, S.; Dai, F.F.; Zhou, M.J.; Yin, Z.; Wang, T.Q.; Zang, M.X.; et al. Circular RNA expression in isoproterenol hydrochloride-induced cardiac hypertrophy. Aging 2020, 12, 2530–2544. [Google Scholar] [CrossRef]
  84. Zhang, N.; Zhang, Y.; Qian, H.; Wu, S.; Cao, L.; Sun, Y. Selective targeting of ubiquitination and degradation of PARP1 by E3 ubiquitin ligase WWP2 regulates isoproterenol-induced cardiac remodeling. Cell Death Differ. 2020, 27, 2605–2619. [Google Scholar] [CrossRef] [PubMed]
  85. Dadson, K.; Hauck, L.; Hao, Z.; Grothe, D.; Rao, V.; Mak, T.W.; Billia, F. The E3 ligase Mule protects the heart against oxidative stress and mitochondrial dysfunction through Myc-dependent inactivation of Pgc-1alpha and Pink1. Sci. Rep. 2017, 7, 41490. [Google Scholar] [CrossRef] [Green Version]
  86. Zhang, L.; Chen, X.; Sharma, P.; Moon, M.; Sheftel, A.D.; Dawood, F.; Nghiem, M.P.; Wu, J.; Li, R.K.; Gramolini, A.O.; et al. HACE1-dependent protein degradation provides cardiac protection in response to haemodynamic stress. Nat. Commun. 2014, 5, 3430. [Google Scholar] [CrossRef]
  87. Rangrez, A.Y.; Borlepawar, A.; Schmiedel, N.; Deshpande, A.; Remes, A.; Kumari, M.; Bernt, A.; Christen, L.; Helbig, A.; Jungmann, A.; et al. The E3 ubiquitin ligase HectD3 attenuates cardiac hypertrophy and inflammation in mice. Commun. Biol. 2020, 3, 562. [Google Scholar] [CrossRef] [PubMed]
  88. Chen, H.; Moreno-Moral, A.; Pesce, F.; Devapragash, N.; Mancini, M.; Heng, E.L.; Rotival, M.; Srivastava, P.K.; Harmston, N.; Shkura, K.; et al. WWP2 regulates pathological cardiac fibrosis by modulating SMAD2 signaling. Nat. Commun. 2019, 10, 3616. [Google Scholar] [CrossRef] [Green Version]
  89. Wang, S.; Sun, A.; Li, L.; Zhao, G.; Jia, J.; Wang, K.; Ge, J.; Zou, Y. Up-regulation of BMP-2 antagonizes TGF-β1/ROCK-enhanced cardiac fibrotic signalling through activation of Smurf1/Smad6 complex. J. Cell Mol. Med. 2012, 16, 2301–2310. [Google Scholar] [CrossRef] [PubMed]
  90. Liu, W.; Zhang, H.; Mai, J.; Chen, Z.; Huang, T.; Wang, S.; Chen, Y.; Wang, J. Distinct Anti-Fibrotic Effects of Exosomes Derived from Endothelial Colony-Forming Cells Cultured Under Normoxia and Hypoxia. Med. Sci. Monit. 2018, 24, 6187–6199. [Google Scholar] [CrossRef]
  91. Cunnington, R.H.; Nazari, M.; Dixon, I.M. c-Ski, Smurf2, and Arkadia as regulators of TGF-beta signaling: New targets for managing myofibroblast function and cardiac fibrosis. Can. J. Physiol. Pharmacol. 2009, 87, 764–772. [Google Scholar] [CrossRef] [PubMed]
  92. Meng, J.; Qin, Y.; Chen, J.; Wei, L.; Huang, X.R.; Yu, X.; Lan, H.Y. Treatment of Hypertensive Heart Disease by Targeting Smad3 Signaling in Mice. Mol. Ther. Methods Clin. Dev. 2020, 18, 791–802. [Google Scholar] [CrossRef] [PubMed]
  93. Matesic, L.E.; Freeburg, L.A.; Snyder, L.B.; Duncan, L.A.; Moore, A.; Perreault, P.E.; Zellars, K.N.; Goldsmith, E.C.; Spinale, F.G. The ubiquitin ligase WWP1 contributes to shifts in matrix proteolytic profiles and a myocardial aging phenotype with diastolic heart. Am. J. Physiol. Heart Circ. Physiol. 2020, 319, H765–H774. [Google Scholar] [CrossRef]
  94. Perry, W.L.; Hustad, C.M.; Swing, D.A.; O’Sullivan, T.N.; Jenkins, N.A.; Copeland, N.G. The itchy locus encodes a novel ubiquitin protein ligase that is disrupted in a18H mice. Nat. Genet. 1998, 18, 143–146. [Google Scholar] [CrossRef]
  95. Infante, P.; Lospinoso Severini, L.; Bernardi, F.; Bufalieri, F.; Di Marcotullio, L. Targeting Hedgehog Signalling through the Ubiquitylation Process: The Multiple Roles of the HECT-E3 Ligase Itch. Cells 2019, 8, 98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Melino, G.; Gallagher, E.; Aqeilan, R.I.; Knight, R.; Peschiaroli, A.; Rossi, M.; Scialpi, F.; Malatesta, M.; Zocchi, L.; Browne, G.; et al. Itch: A HECT-type E3 ligase regulating immunity, skin and cancer. Cell Death Differ. 2008, 15, 1103–1112. [Google Scholar] [CrossRef] [Green Version]
  97. Aki, D.; Zhang, W.; Yun-Cai, L. The E3 ligase Itch in immune regulation and beyond. Immunol. Rev. 2015, 266, 6–26. [Google Scholar] [CrossRef] [PubMed]
  98. Zhang, Y.; Zhang, L.; Fan, X.; Yang, W.; Yu, B.; Kou, J.; Li, F. Captopril attenuates TAC-induced heart failure via inhibiting Wnt3a/beta-catenin and Jak2/Stat3 pathways. Biomed. Pharmacother. 2019, 113, 108780. [Google Scholar] [CrossRef] [PubMed]
  99. van de Schans, V.A.; van den Borne, S.W.; Strzelecka, A.E.; Janssen, B.J.; van der Velden, J.L.; Langen, R.C.; Wynshaw-Boris, A.; Smits, J.F.; Blankesteijn, W.M. Interruption of Wnt signaling attenuates the onset of pressure overload-induced cardiac hypertrophy. Hypertension 2007, 49, 473–480. [Google Scholar] [CrossRef] [Green Version]
  100. Zhao, Y.; Wang, C.; Hong, X.; Miao, J.; Liao, Y.; Hou, F.F.; Zhou, L.; Liu, Y. Wnt/beta-catenin signaling mediates both heart and kidney injury in type 2 cardiorenal syndrome. Kidney Int. 2019, 95, 815–829. [Google Scholar] [CrossRef]
  101. Gao, C.; Chen, Y.G. Dishevelled: The hub of Wnt signaling. Cell Signal 2010, 22, 717–727. [Google Scholar] [CrossRef]
  102. Mossinger, J.; Wieffer, M.; Krause, E.; Freund, C.; Gerth, F.; Krauss, M.; Haucke, V. Phosphatidylinositol 4-kinase IIalpha function at endosomes is regulated by the ubiquitin ligase Itch. EMBO Rep. 2012, 13, 1087–1094. [Google Scholar] [CrossRef] [Green Version]
  103. Li, L.; Chen, Y.; Doan, J.; Murray, J.; Molkentin, J.D.; Liu, Q. Transforming growth factor beta-activated kinase 1 signaling pathway critically regulates myocardial survival and remodeling. Circulation 2014, 130, 2162–2172. [Google Scholar] [CrossRef]
  104. Kumar, S.; Tomooka, Y.; Noda, M. Identification of a set of genes with developmentally down-regulated expression in the mouse brain. Biochem. Biophys. Res. Commun. 1992, 185, 1155–1161. [Google Scholar] [CrossRef]
  105. Ishigami, T.; Kino, T.; Minegishi, S.; Araki, N.; Umemura, M.; Ushio, H.; Saigoh, S.; Sugiyama, M. Regulators of Epithelial Sodium Channels in Aldosterone-Sensitive Distal Nephrons (ASDN): Critical Roles of Nedd4L/Nedd4-2 and Salt-Sensitive Hypertension. Int. J. Mol. Sci. 2020, 21, 3871. [Google Scholar] [CrossRef]
  106. Manning, J.A.; Kumar, S. Physiological Functions of Nedd4-2: Lessons from Knockout Mouse Models. Trends Biochem. Sci. 2018, 43, 635–647. [Google Scholar] [CrossRef] [PubMed]
  107. Jespersen, T.; Membrez, M.; Nicolas, C.S.; Pitard, B.; Staub, O.; Olesen, S.P.; Baro, I.; Abriel, H. The KCNQ1 potassium channel is down-regulated by ubiquitylating enzymes of the Nedd4/Nedd4-like family. Cardiovasc. Res. 2007, 74, 64–74. [Google Scholar] [CrossRef]
  108. Guo, J.; Wang, T.; Li, X.; Shallow, H.; Yang, T.; Li, W.; Xu, J.; Fridman, M.D.; Yang, X.; Zhang, S. Cell surface expression of human ether-a-go-go-related gene (hERG) channels is regulated by caveolin-3 protein via the ubiquitin ligase Nedd4-2. J. Biol. Chem. 2012, 287, 33132–33141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Goel, P.; Manning, J.A.; Kumar, S. NEDD4-2 (NEDD4L): The ubiquitin ligase for multiple membrane proteins. Gene 2015, 557, 1–10. [Google Scholar] [CrossRef]
  110. Pirozzi, G.; McConnell, S.J.; Uveges, A.J.; Carter, J.M.; Sparks, A.B.; Kay, B.K.; Fowlkes, D.M. Identification of novel human WW domain-containing proteins by cloning of ligand targets. J. Biol. Chem. 1997, 272, 14611–14616. [Google Scholar] [CrossRef] [Green Version]
  111. Wood, J.D.; Yuan, J.; Margolis, R.L.; Colomer, V.; Duan, K.; Kushi, J.; Kaminsky, Z.; Kleiderlein, J.J.; Sharp, A.H.; Ross, C.A. Atrophin-1, the DRPLA gene product, interacts with two families of WW domain-containing proteins. Mol. Cell Neurosci. 1998, 11, 149–160. [Google Scholar] [CrossRef]
  112. Chen, W.; Jiang, X.; Luo, Z. WWP2: A multifunctional ubiquitin ligase gene. Pathol. Oncol. Res. 2014, 20, 799–803. [Google Scholar] [CrossRef]
  113. Li, H.; Zhang, Z.; Wang, B.; Zhang, J.; Zhao, Y.; Jin, Y. Wwp2-mediated ubiquitination of the RNA polymerase II large subunit in mouse embryonic pluripotent stem cells. Mol. Cell Biol. 2007, 27, 5296–5305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Nakamura, Y.; Yamamoto, K.; He, X.; Otsuki, B.; Kim, Y.; Murao, H.; Soeda, T.; Tsumaki, N.; Deng, J.M.; Zhang, Z.; et al. Wwp2 is essential for palatogenesis mediated by the interaction between Sox9 and mediator subunit 25. Nat. Commun. 2011, 2, 251. [Google Scholar] [CrossRef]
  115. Yang, Y.; Liao, B.; Wang, S.; Yan, B.; Jin, Y.; Shu, H.B.; Wang, Y.Y. E3 ligase WWP2 negatively regulates TLR3-mediated innate immune response by targeting TRIF for ubiquitination and degradation. Proc. Natl. Acad. Sci. USA 2013, 110, 5115–5120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Chen, Z.; Jiang, H.; Xu, W.; Li, X.; Dempsey, D.R.; Zhang, X.; Devreotes, P.; Wolberger, C.; Amzel, L.M.; Gabelli, S.B.; et al. A Tunable Brake for HECT Ubiquitin Ligases. Mol. Cell 2017, 66, 345–357.e6. [Google Scholar] [CrossRef]
  117. Maddika, S.; Kavela, S.; Rani, N.; Palicharla, V.R.; Pokorny, J.L.; Sarkaria, J.N.; Chen, J. WWP2 is an E3 ubiquitin ligase for PTEN. Nat. Cell Biol. 2011, 13, 728–733. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Fukumoto, C.; Nakashima, D.; Kasamatsu, A.; Unozawa, M.; Shida-Sakazume, T.; Higo, M.; Ogawara, K.; Yokoe, H.; Shiiba, M.; Tanzawa, H.; et al. WWP2 is overexpressed in human oral cancer, determining tumor size and poor prognosis in patients: Downregulation of WWP2 inhibits the AKT signaling and tumor growth in mice. Oncoscience 2014, 1, 807–820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Zhang, R.; Zhang, J.; Luo, W.; Luo, Z.; Shi, S. WWP2 Is One Promising Novel Oncogene. Pathol. Oncol. Res. 2019, 25, 443–446. [Google Scholar] [CrossRef] [PubMed]
  120. Pillai, J.B.; Russell, H.M.; Raman, J.; Jeevanandam, V.; Gupta, M.P. Increased expression of poly(ADP-ribose) polymerase-1 contributes to caspase-independent myocyte cell death during heart failure. Am. J. Physiol. Heart Circ. Physiol. 2005, 288, H486–H496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Lu, J.; Zhang, R.; Hong, H.; Yang, Z.; Sun, D.; Sun, S.; Guo, X.; Ye, J.; Li, Z.; Liu, P. The poly(ADP-ribosyl)ation of FoxO3 mediated by PARP1 participates in isoproterenol-induced cardiac hypertrophy. Biochim. Biophys. Acta 2016, 1863, 3027–3039. [Google Scholar] [CrossRef] [PubMed]
  122. Virag, L.; Robaszkiewicz, A.; Rodriguez-Vargas, J.M.; Oliver, F.J. Poly(ADP-ribose) signaling in cell death. Mol. Asp. Med. 2013, 34, 1153–1167. [Google Scholar] [CrossRef] [PubMed]
  123. Zhong, Q.; Gao, W.; Du, F.; Wang, X. Mule/ARF-BP1, a BH3-only E3 ubiquitin ligase, catalyzes the polyubiquitination of Mcl-1 and regulates apoptosis. Cell 2005, 121, 1085–1095. [Google Scholar] [CrossRef] [Green Version]
  124. Kao, S.H.; Wu, H.T.; Wu, K.J. Ubiquitination by HUWE1 in tumorigenesis and beyond. J. Biomed. Sci. 2018, 25, 67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Gong, X.; Du, D.; Deng, Y.; Zhou, Y.; Sun, L.; Yuan, S. The structure and regulation of the E3 ubiquitin ligase HUWE1 and its biological functions in cancer. Investig. New Drugs 2020, 38, 515–524. [Google Scholar] [CrossRef]
  126. Chen, D.; Kon, N.; Li, M.; Zhang, W.; Qin, J.; Gu, W. ARF-BP1/Mule is a critical mediator of the ARF tumor suppressor. Cell 2005, 121, 1071–1083. [Google Scholar] [CrossRef] [Green Version]
  127. Bernassola, F.; Karin, M.; Ciechanover, A.; Melino, G. The HECT family of E3 ubiquitin ligases: Multiple players in cancer development. Cancer Cell 2008, 14, 10–21. [Google Scholar] [CrossRef]
  128. Zhang, H.; Gao, P.; Fukuda, R.; Kumar, G.; Krishnamachary, B.; Zeller, K.I.; Dang, C.V.; Semenza, G.L. HIF-1 inhibits mitochondrial biogenesis and cellular respiration in VHL-deficient renal cell carcinoma by repression of C-MYC activity. Cancer Cell 2007, 11, 407–420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Li, F.; Wang, Y.; Zeller, K.I.; Potter, J.J.; Wonsey, D.R.; O’Donnell, K.A.; Kim, J.W.; Yustein, J.T.; Lee, L.A.; Dang, C.V. Myc stimulates nuclearly encoded mitochondrial genes and mitochondrial biogenesis. Mol. Cell Biol. 2005, 25, 6225–6234. [Google Scholar] [CrossRef] [Green Version]
  130. Ahuja, P.; Zhao, P.; Angelis, E.; Ruan, H.; Korge, P.; Olson, A.; Wang, Y.; Jin, E.S.; Jeffrey, F.M.; Portman, M.; et al. Myc controls transcriptional regulation of cardiac metabolism and mitochondrial biogenesis in response to pathological stress in mice. J. Clin. Investig. 2010, 120, 1494–1505. [Google Scholar] [CrossRef] [Green Version]
  131. Anglesio, M.S.; Evdokimova, V.; Melnyk, N.; Zhang, L.; Fernandez, C.V.; Grundy, P.E.; Leach, S.; Marra, M.A.; Brooks-Wilson, A.R.; Penninger, J.; et al. Differential expression of a novel ankyrin containing E3 ubiquitin-protein ligase, Hace1, in sporadic Wilms’ tumor versus normal kidney. Hum. Mol. Genet. 2004, 13, 2061–2074. [Google Scholar] [CrossRef] [Green Version]
  132. Zhang, L.; Anglesio, M.S.; O’Sullivan, M.; Zhang, F.; Yang, G.; Sarao, R.; Mai, P.N.; Cronin, S.; Hara, H.; Melnyk, N.; et al. The E3 ligase HACE1 is a critical chromosome 6q21 tumor suppressor involved in multiple cancers. Nat. Med. 2007, 13, 1060–1069. [Google Scholar] [CrossRef] [PubMed]
  133. Torrino, S.; Visvikis, O.; Doye, A.; Boyer, L.; Stefani, C.; Munro, P.; Bertoglio, J.; Gacon, G.; Mettouchi, A.; Lemichez, E. The E3 ubiquitin-ligase HACE1 catalyzes the ubiquitylation of active Rac1. Dev. Cell 2011, 21, 959–965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Seipel, K.; O’Brien, S.P.; Iannotti, E.; Medley, Q.G.; Streuli, M. Tara, a novel F-actin binding protein, associates with the Trio guanine nucleotide exchange factor and regulates actin cytoskeletal organization. J. Cell Sci. 2001, 114, 389–399. [Google Scholar] [CrossRef] [PubMed]
  135. Jiang, Q.; Li, F.; Cheng, Z.; Kong, Y.; Chen, C. The role of E3 ubiquitin ligase HECTD3 in cancer and beyond. Cell Mol. Life Sci. 2020, 77, 1483–1495. [Google Scholar] [CrossRef] [PubMed]
  136. Bernt, A.; Rangrez, A.Y.; Eden, M.; Jungmann, A.; Katz, S.; Rohr, C.; Muller, O.J.; Katus, H.A.; Sossalla, S.T.; Williams, T.; et al. Sumoylation-independent activation of Calcineurin-NFAT-signaling via SUMO2 mediates cardiomyocyte hypertrophy. Sci. Rep. 2016, 6, 35758. [Google Scholar] [CrossRef] [Green Version]
  137. Kuusisto, J.; Karja, V.; Sipola, P.; Kholova, I.; Peuhkurinen, K.; Jaaskelainen, P.; Naukkarinen, A.; Yla-Herttuala, S.; Punnonen, K.; Laakso, M. Low-grade inflammation and the phenotypic expression of myocardial fibrosis in hypertrophic cardiomyopathy. Heart 2012, 98, 1007–1013. [Google Scholar] [CrossRef] [Green Version]
  138. Kishore, R.; Verma, S.K. Roles of STATs signaling in cardiovascular diseases. JAKSTAT 2012, 1, 118–124. [Google Scholar] [CrossRef] [Green Version]
  139. Travers, J.G.; Kamal, F.A.; Robbins, J.; Yutzey, K.E.; Blaxall, B.C. Cardiac Fibrosis: The Fibroblast Awakens. Circ. Res. 2016, 118, 1021–1040. [Google Scholar] [CrossRef] [Green Version]
  140. Berk, B.C.; Fujiwara, K.; Lehoux, S. ECM remodeling in hypertensive heart disease. J. Clin. Investig. 2007, 117, 568–575. [Google Scholar] [CrossRef]
  141. Frangogiannis, N.G. Cardiac fibrosis. Cardiovasc. Res. 2020. [Google Scholar] [CrossRef] [PubMed]
  142. Ashrafian, H.; McKenna, W.J.; Watkins, H. Disease pathways and novel therapeutic targets in hypertrophic cardiomyopathy. Circ. Res. 2011, 109, 86–96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Bernaba, B.N.; Chan, J.B.; Lai, C.K.; Fishbein, M.C. Pathology of late-onset anthracycline cardiomyopathy. Cardiovasc. Pathol. 2010, 19, 308–311. [Google Scholar] [CrossRef]
  144. Asbun, J.; Villarreal, F.J. The pathogenesis of myocardial fibrosis in the setting of diabetic cardiomyopathy. J. Am. Coll Cardiol. 2006, 47, 693–700. [Google Scholar] [CrossRef] [Green Version]
  145. Kong, P.; Christia, P.; Frangogiannis, N.G. The pathogenesis of cardiac fibrosis. Cell Mol. Life Sci. 2014, 71, 549–574. [Google Scholar] [CrossRef] [Green Version]
  146. Baum, J.; Duffy, H.S. Fibroblasts and myofibroblasts: What are we talking about? J. Cardiovasc. Pharmacol. 2011, 57, 376–379. [Google Scholar] [CrossRef]
  147. Watanabe, K.; Narumi, T.; Watanabe, T.; Otaki, Y.; Takahashi, T.; Aono, T.; Goto, J.; Toshima, T.; Sugai, T.; Wanezaki, M.; et al. The association between microRNA-21 and hypertension-induced cardiac remodeling. PLoS ONE 2020, 15, e0226053. [Google Scholar] [CrossRef]
  148. Walton, K.L.; Johnson, K.E.; Harrison, C.A. Targeting TGF-beta Mediated SMAD Signaling for the Prevention of Fibrosis. Front. Pharmacol. 2017, 8, 461. [Google Scholar] [CrossRef] [Green Version]
  149. Hu, H.H.; Chen, D.Q.; Wang, Y.N.; Feng, Y.L.; Cao, G.; Vaziri, N.D.; Zhao, Y.Y. New insights into TGF-beta/Smad signaling in tissue fibrosis. Chem. Biol. Interact. 2018, 292, 76–83. [Google Scholar] [CrossRef] [Green Version]
  150. Ross, S.; Hill, C.S. How the Smads regulate transcription. Int. J. Biochem. Cell Biol. 2008, 40, 383–408. [Google Scholar] [CrossRef]
  151. Moreno-Moral, A.; Pesce, F.; Behmoaras, J.; Petretto, E. Systems Genetics as a Tool to Identify Master Genetic Regulators in Complex Disease. Methods Mol. Biol. 2017, 1488, 337–362. [Google Scholar] [CrossRef]
  152. Zhu, H.; Kavsak, P.; Abdollah, S.; Wrana, J.L.; Thomsen, G.H. A SMAD ubiquitin ligase targets the BMP pathway and affects embryonic pattern formation. Nature 1999, 400, 687–693. [Google Scholar] [CrossRef]
  153. Lin, X.; Liang, M.; Feng, X.H. Smurf2 is a ubiquitin E3 ligase mediating proteasome-dependent degradation of Smad2 in transforming growth factor-beta signaling. J. Biol. Chem. 2000, 275, 36818–36822. [Google Scholar] [CrossRef] [Green Version]
  154. David, D.; Nair, S.A.; Pillai, M.R. Smurf E3 ubiquitin ligases at the cross roads of oncogenesis and tumor suppression. Biochim. Biophys. Acta 2013, 1835, 119–128. [Google Scholar] [CrossRef] [PubMed]
  155. Xia, Q.; Li, Y.; Han, D.; Dong, L. SMURF1, a promoter of tumor cell progression? Cancer Gene Ther. 2020. [Google Scholar] [CrossRef]
  156. Duenas, A.; Exposito, A.; Munoz, M.D.M.; de Manuel, M.J.; Camara-Morales, A.; Serrano-Osorio, F.; Garcia-Padilla, C.; Hernandez-Torres, F.; Dominguez, J.N.; Aranega, A.; et al. MiR-195 enhances cardiomyogenic differentiation of the proepicardium/septum transversum by Smurf1 and Foxp1 modulation. Sci. Rep. 2020, 10, 9334. [Google Scholar] [CrossRef] [PubMed]
  157. Koefoed, K.; Skat-Rørdam, J.; Andersen, P.; Warzecha, C.B.; Pye, M.; Andersen, T.A.; Ajbro, K.D.; Bendsen, E.; Narimatsu, M.; Vilhardt, F.; et al. The E3 ubiquitin ligase SMURF1 regulates cell-fate specification and outflow tract septation during mammalian heart development. Sci. Rep. 2018, 8, 9542. [Google Scholar] [CrossRef]
  158. Molina-Navarro, M.M.; Trivino, J.C.; Martinez-Dolz, L.; Lago, F.; Gonzalez-Juanatey, J.R.; Portoles, M.; Rivera, M. Functional networks of nucleocytoplasmic transport-related genes differentiate ischemic and dilated cardiomyopathies. A new therapeutic opportunity. PLoS ONE 2014, 9, e104709. [Google Scholar] [CrossRef]
  159. Ponikowski, P.; Voors, A.A.; Anker, S.D.; Bueno, H.; Cleland, J.G.F.; Coats, A.J.S.; Falk, V.; Gonzalez-Juanatey, J.R.; Harjola, V.P.; Jankowska, E.A.; et al. 2016 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: The Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC)Developed with the special contribution of the Heart Failure Association (HFA) of the ESC. Eur. Heart J. 2016, 37, 2129–2200. [Google Scholar] [CrossRef] [PubMed]
  160. Andersson, C.; Vasan, R.S. Epidemiology of heart failure with preserved ejection fraction. Heart Fail. Clin. 2014, 10, 377–388. [Google Scholar] [CrossRef] [Green Version]
  161. Dunlay, S.M.; Roger, V.L.; Redfield, M.M. Epidemiology of heart failure with preserved ejection fraction. Nat. Rev. Cardiol. 2017, 14, 591–602. [Google Scholar] [CrossRef]
  162. Lu, L.; Guo, J.; Hua, Y.; Huang, K.; Magaye, R.; Cornell, J.; Kelly, D.J.; Reid, C.; Liew, D.; Zhou, Y.; et al. Cardiac fibrosis in the ageing heart: Contributors and mechanisms. Clin. Exp. Pharmacol. Physiol. 2017, 44 (Suppl. 1), 55–63. [Google Scholar] [CrossRef] [Green Version]
  163. Upadhya, B.; Taffet, G.E.; Cheng, C.P.; Kitzman, D.W. Heart failure with preserved ejection fraction in the elderly: Scope of the problem. J. Mol. Cell Cardiol. 2015, 83, 73–87. [Google Scholar] [CrossRef] [Green Version]
  164. Butler, J.; Fonarow, G.C.; Zile, M.R.; Lam, C.S.; Roessig, L.; Schelbert, E.B.; Shah, S.J.; Ahmed, A.; Bonow, R.O.; Cleland, J.G.; et al. Developing therapies for heart failure with preserved ejection fraction: Current state and future directions. JACC Heart Fail. 2014, 2, 97–112. [Google Scholar] [CrossRef] [PubMed]
  165. Shu, L.; Zhang, H.; Boyce, B.F.; Xing, L. Ubiquitin E3 ligase Wwp1 negatively regulates osteoblast function by inhibiting osteoblast differentiation and migration. J. Bone Miner. Res. 2013, 28, 1925–1935. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Zhi, X.; Chen, C. WWP1: A versatile ubiquitin E3 ligase in signaling and diseases. Cell Mol. Life Sci. 2012, 69, 1425–1434. [Google Scholar] [CrossRef] [PubMed]
  167. Yeung, B.; Ho, K.C.; Yang, X. WWP1 E3 ligase targets LATS1 for ubiquitin-mediated degradation in breast cancer cells. PLoS ONE 2013, 8, e61027. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Komuro, A.; Imamura, T.; Saitoh, M.; Yoshida, Y.; Yamori, T.; Miyazono, K.; Miyazawa, K. Negative regulation of transforming growth factor-beta (TGF-beta) signaling by WW domain-containing protein 1 (WWP1). Oncogene 2004, 23, 6914–6923. [Google Scholar] [CrossRef]
  169. Mosser, E.A.; Kasanov, J.D.; Forsberg, E.C.; Kay, B.K.; Ney, P.A.; Bresnick, E.H. Physical and functional interactions between the transactivation domain of the hematopoietic transcription factor NF-E2 and WW domains. Biochemistry 1998, 37, 13686–13695. [Google Scholar] [CrossRef]
  170. Hahn, V.S.; Knutsdottir, H.; Luo, X.; Bedi, K.; Margulies, K.B.; Haldar, S.M.; Stolina, M.; Yin, J.; Khakoo, A.Y.; Vaishnav, J.; et al. Myocardial Gene Expression Signatures in Human Heart Failure With Preserved Ejection Fraction. Circulation 2021, 143, 120–134. [Google Scholar] [CrossRef]
  171. Kadowaki, S.; Shishido, T.; Sasaki, T.; Sugai, T.; Narumi, T.; Honda, Y.; Otaki, Y.; Kinoshita, D.; Takahashi, T.; Nishiyama, S.; et al. Deficiency of Senescence Marker Protein 30 Exacerbates Cardiac Injury after Ischemia/Reperfusion. Int. J. Mol. Sci. 2016, 17, 542. [Google Scholar] [CrossRef] [PubMed]
  172. Kukan, M. Emerging roles of proteasomes in ischemia-reperfusion injury of organs. J. Physiol. Pharmacol. 2004, 55, 3–15. [Google Scholar]
  173. Yellon, D.M.; Hausenloy, D.J. Myocardial reperfusion injury. N. Engl. J. Med. 2007, 357, 1121–1135. [Google Scholar] [CrossRef]
  174. Eefting, F.; Rensing, B.; Wigman, J.; Pannekoek, W.J.; Liu, W.M.; Cramer, M.J.; Lips, D.J.; Doevendans, P.A. Role of apoptosis in reperfusion injury. Cardiovasc. Res. 2004, 61, 414–426. [Google Scholar] [CrossRef]
  175. Zhao, Z. Inhibition of myocardial apoptosis reduces infarct size and improves regional contractile dysfunction during reperfusion. Cardiovasc. Res. 2003, 59, 132–142. [Google Scholar] [CrossRef] [Green Version]
  176. Shukla, S.K.; Rafiq, K. Proteasome biology and therapeutics in cardiac diseases. Transl. Res. 2019, 205, 64–76. [Google Scholar] [CrossRef]
  177. Alva, N.; Panisello-Roselló, A.; Flores, M.; Roselló-Catafau, J. Teresa Carbonell Ubiquitin-proteasome system and oxidative stress in liver transplantation. World J. Gastroenterol. 2018, 21, 3521–3530. [Google Scholar] [CrossRef] [PubMed]
  178. Hochrainer, K. Protein Modifications with Ubiquitin as Response to Cerebral Ischemia-Reperfusion Injury. Transl. Stroke Res. 2018, 9, 157–173. [Google Scholar] [CrossRef]
  179. Zhang, C.; Xu, Z.; He, X.R.; Michael, L.H.; Patterson, C. CHIP, a cochaperone/ubiquitin ligase that regulates protein quality control, is required for maximal cardioprotection after myocardial infarction in mice. Am. J. Physiol. Heart Circ. Physiol. 2005, 288, H2836–H2842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Hu, W.; Zhang, P.; Gu, J.; Yu, Q.; Zhang, D. NEDD4-1 protects against ischaemia/reperfusion-induced cardiomyocyte apoptosis via the PI3K/Akt pathway. Apoptosis 2017, 22, 437–448. [Google Scholar] [CrossRef]
  181. Dong, W.; Xie, F.; Chen, X.Y.; Huang, W.L.; Zhang, Y.Z.; Luo, W.B.; Chen, J.; Xie, M.T.; Peng, X.P. Inhibition of Smurf2 translation by miR-322/503 protects from ischemia-reperfusion injury by modulating EZH2/Akt/GSK3beta signaling. Am. J. Physiol. Cell Physiol. 2019, 317, C253–C261. [Google Scholar] [CrossRef]
  182. Otaki, Y.; Takahashi, H.; Watanabe, T.; Funayama, A.; Netsu, S.; Honda, Y.; Narumi, T.; Kadowaki, S.; Hasegawa, H.; Honda, S.; et al. HECT-Type Ubiquitin E3 Ligase ITCH Interacts With Thioredoxin-Interacting Protein and Ameliorates Reactive Oxygen Species-Induced Cardiotoxicity. J. Am. Heart Assoc. 2016, 5. [Google Scholar] [CrossRef] [Green Version]
  183. Zhang, W.C.; Yang, J.H.; Liu, G.H.; Yang, F.; Gong, J.L.; Jia, M.G.; Zhang, M.J.; Zhao, L.S. miR-34b/c regulates doxorubicin-induced myocardial cell injury through ITCH. Cell Cycle 2019, 18, 3263–3274. [Google Scholar] [CrossRef]
  184. Zhang, N.; Wang, X. Circular RNA ITCH mediates H(2)O(2)-induced myocardial cell apoptosis by targeting miR-17-5p via wnt/β-catenin signalling pathway. Int. J. Exp. Pathol. 2020, 102, 22–31. [Google Scholar] [CrossRef]
  185. Han, D.; Wang, Y.; Wang, Y.; Dai, X.; Zhou, T.; Chen, J.; Tao, B.; Zhang, J.; Cao, F. The Tumor-Suppressive Human Circular RNA CircITCH Sponges miR-330-5p to Ameliorate Doxorubicin-Induced Cardiotoxicity Through Upregulating SIRT6, Survivin, and SERCA2a. Circ. Res. 2020, 127, e108–e125. [Google Scholar] [CrossRef] [PubMed]
  186. Luo, L.; Ning, F.; Du, Y.; Song, B.; Yang, D.; Salvage, S.C.; Wang, Y.; Fraser, J.A.; Zhang, S.; Ma, A.; et al. Calcium-dependent Nedd4-2 upregulation mediates degradation of the cardiac sodium channel Nav1.5: Implications for heart failure. Acta Physiol. 2017, 221, 44–58. [Google Scholar] [CrossRef]
  187. Basheer, W.A.; Harris, B.S.; Mentrup, H.L.; Abreha, M.; Thames, E.L.; Lea, J.B.; Swing, D.A.; Copeland, N.G.; Jenkins, N.A.; Price, R.L.; et al. Cardiomyocyte-specific overexpression of the ubiquitin ligase Wwp1 contributes to reduction in Connexin 43 and arrhythmogenesis. J. Mol. Cell Cardiol. 2015, 88, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Wang, X.; Trotman, L.C.; Koppie, T.; Alimonti, A.; Chen, Z.; Gao, Z.; Wang, J.; Erdjument-Bromage, H.; Tempst, P.; Cordon-Cardo, C.; et al. NEDD4-1 is a proto-oncogenic ubiquitin ligase for PTEN. Cell 2007, 128, 129–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Liu, P.Y.; Xu, N.; Malyukova, A.; Scarlett, C.J.; Sun, Y.T.; Zhang, X.D.; Ling, D.; Su, S.P.; Nelson, C.; Chang, D.K.; et al. The histone deacetylase SIRT2 stabilizes Myc oncoproteins. Cell Death Differ. 2013, 20, 503–514. [Google Scholar] [CrossRef] [Green Version]
  190. Platta, H.W.; Abrahamsen, H.; Thoresen, S.B.; Stenmark, H. Nedd4-dependent lysine-11-linked polyubiquitination of the tumour suppressor Beclin 1. Biochem. J. 2012, 441, 399–406. [Google Scholar] [CrossRef] [Green Version]
  191. Tofaris, G.K.; Kim, H.T.; Hourez, R.; Jung, J.W.; Kim, K.P.; Goldberg, A.L. Ubiquitin ligase Nedd4 promotes alpha-synuclein degradation by the endosomal-lysosomal pathway. Proc. Natl. Acad. Sci. USA 2011, 108, 17004–17009. [Google Scholar] [CrossRef] [Green Version]
  192. Huang, X.; Chen, J.; Cao, W.; Yang, L.; Chen, Q.; He, J.; Yi, Q.; Huang, H.; Zhang, E.; Cai, Z. The many substrates and functions of NEDD4-1. Cell Death Dis. 2019, 10, 904. [Google Scholar] [CrossRef] [Green Version]
  193. Fouladkou, F.; Lu, C.; Jiang, C.; Zhou, L.; She, Y.; Walls, J.R.; Kawabe, H.; Brose, N.; Henkelman, R.M.; Huang, A.; et al. The ubiquitin ligase Nedd4-1 is required for heart development and is a suppressor of thrombospondin-1. J. Biol. Chem. 2010, 285, 6770–6780. [Google Scholar] [CrossRef] [Green Version]
  194. Duan, Q.; Madan, N.D.; Wu, J.; Kalisz, J.; Doshi, K.Y.; Haldar, S.M.; Liu, L.; Pierre, S.V. Role of phosphoinositide 3-kinase IA (PI3K-IA) activation in cardioprotection induced by ouabain preconditioning. J. Mol. Cell Cardiol. 2015, 80, 114–125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Xing, X.; Guo, S.; Zhang, G.; Liu, Y.; Bi, S.; Wang, X.; Lu, Q. miR-26a-5p protects against myocardial ischemia/reperfusion injury by regulating the PTEN/PI3K/AKT signaling pathway. Braz. J. Med. Biol. Res. 2020, 53, e9106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Abdulkareem, I.H.; Blair, M. Phosphatase and tensin homologue deleted on chromosome. Niger. Med. J. 2013, 54, 79–86. [Google Scholar] [CrossRef] [PubMed]
  197. Fan, C.D.; Lum, M.A.; Xu, C.; Black, J.D.; Wang, X. Ubiquitin-dependent regulation of phospho-AKT dynamics by the ubiquitin E3 ligase, NEDD4-1, in the insulin-like growth factor-1 response. J. Biol. Chem. 2013, 288, 1674–1684. [Google Scholar] [CrossRef] [Green Version]
  198. Kim, H.K.; Kang, S.W.; Jeong, S.H.; Kim, N.; Ko, J.H.; Bang, H.; Park, W.S.; Choi, T.H.; Ha, Y.R.; Lee, Y.S.; et al. Identification of potential target genes of cardioprotection against ischemia-reperfusion injury by express sequence tags analysis in rat hearts. J. Cardiol. 2012, 60, 98–110. [Google Scholar] [CrossRef] [Green Version]
  199. Li, C.H.; Chen, Y. Targeting EZH2 for cancer therapy: Progress and perspective. Curr. Protein Pept. Sci. 2015, 16, 559–570. [Google Scholar]
  200. Gan, L.; Xu, M.; Hua, R.; Tan, C.; Zhang, J.; Gong, Y.; Wu, Z.; Weng, W.; Sheng, W.; Guo, W. The polycomb group protein EZH2 induces epithelial-mesenchymal transition and pluripotent phenotype of gastric cancer cells by binding to PTEN promoter. J. Hematol. Oncol. 2018, 11, 9. [Google Scholar] [CrossRef]
  201. Bertrand Coiffier, E.L.; Briere, J.; Herbrecht, R.; Tilly, H.; Bouabdallah, R.; Morel, P.; van den Neste, E.; Salles, G.; Gaulard, P.; Reyes, F.; et al. CHOP chemotherapy plus rituximab compared with CHOP alone in elderly patients with diffuse large-B-cell lymphoma. N. Engl. J. Med. 2002, 346, 235–242. [Google Scholar] [CrossRef] [PubMed]
  202. Hortobagyi, G.N. Treatment of breast cancer. N. Engl. J. Med. 1998, 339, 974–984. [Google Scholar] [CrossRef] [PubMed]
  203. van Wijk, F.H.; Aapro, M.S.; Bolis, G.; Chevallier, B.; van der Burg, M.E.; Poveda, A.; de Oliveira, C.F.; Tumolo, S.; Scotto di Palumbo, V.; Piccart, M.; et al. Doxorubicin versus doxorubicin and cisplatin in endometrial carcinoma: Definitive results of a randomised study (55872) by the EORTC Gynaecological Cancer Group. Ann. Oncol. 2003, 14, 441–448. [Google Scholar] [CrossRef] [PubMed]
  204. Zamorano, J.L.; Lancellotti, P.; Rodriguez Munoz, D.; Aboyans, V.; Asteggiano, R.; Galderisi, M.; Habib, G.; Lenihan, D.J.; Lip, G.Y.H.; Lyon, A.R.; et al. 2016 ESC Position Paper on cancer treatments and cardiovascular toxicity developed under the auspices of the ESC Committee for Practice Guidelines: The Task Force for cancer treatments and cardiovascular toxicity of the European Society of Cardiology (ESC). Eur. Heart J. 2016, 37, 2768–2801. [Google Scholar] [CrossRef]
  205. Prathumsap, N.; Shinlapawittayatorn, K.; Chattipakorn, S.C.; Chattipakorn, N. Effects of doxorubicin on the heart: From molecular mechanisms to intervention strategies. Eur. J. Pharmacol. 2020, 866, 172818. [Google Scholar] [CrossRef]
  206. Escobedo, A.; Gomes, T.; Aragon, E.; Martin-Malpartida, P.; Ruiz, L.; Macias, M.J. Structural basis of the activation and degradation mechanisms of the E3 ubiquitin ligase Nedd4L. Structure 2014, 22, 1446–1457. [Google Scholar] [CrossRef] [Green Version]
  207. Rook, M.B.; Evers, M.M.; Vos, M.A.; Bierhuizen, M.F. Biology of cardiac sodium channel Nav1.5 expression. Cardiovasc. Res. 2012, 93, 12–23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Shang, L.L.; Pfahnl, A.E.; Sanyal, S.; Jiao, Z.; Allen, J.; Banach, K.; Fahrenbach, J.; Weiss, D.; Taylor, W.R.; Zafari, A.M.; et al. Human heart failure is associated with abnormal C-terminal splicing variants in the cardiac sodium channel. Circ. Res. 2007, 101, 1146–1154. [Google Scholar] [CrossRef] [Green Version]
  209. Minegishi, S.; Ishigami, T.; Kawamura, H.; Kino, T.; Chen, L.; Nakashima-Sasaki, R.; Doi, H.; Azushima, K.; Wakui, H.; Chiba, Y.; et al. An Isoform of Nedd4-2 Plays a Pivotal Role in Electrophysiological Cardiac Abnormalities. Int. J. Mol. Sci. 2017, 18, 1268. [Google Scholar] [CrossRef] [Green Version]
  210. Sanguinetti, M.C.; Tristani-Firouzi, M. hERG potassium channels and cardiac arrhythmia. Nature 2006, 440, 463–469. [Google Scholar] [CrossRef]
  211. Fishman, G.I.; Chugh, S.S.; Dimarco, J.P.; Albert, C.M.; Anderson, M.E.; Bonow, R.O.; Buxton, A.E.; Chen, P.S.; Estes, M.; Jouven, X.; et al. Sudden cardiac death prediction and prevention: Report from a National Heart, Lung, and Blood Institute and Heart Rhythm Society Workshop. Circulation 2010, 122, 2335–2348. [Google Scholar] [CrossRef] [PubMed]
  212. Kuriachan, V.P.; Sumner, G.L.; Mitchell, L.B. Sudden cardiac death. Curr. Probl. Cardiol. 2015, 40, 133–200. [Google Scholar] [CrossRef] [PubMed]
  213. Remo, B.F.; Giovannone, S.; Fishman, G.I. Connexin43 cardiac gap junction remodeling: Lessons from genetically engineered murine models. J. Membr. Biol. 2012, 245, 275–281. [Google Scholar] [CrossRef] [Green Version]
  214. Severs, N.J.; Bruce, A.F.; Dupont, E.; Rothery, S. Remodelling of gap junctions and connexin expression in diseased myocardium. Cardiovasc. Res. 2008, 80, 9–19. [Google Scholar] [CrossRef] [Green Version]
  215. Einsele, H. Bortezomib. Recent Results Cancer Res. 2014, 201, 325–345. [Google Scholar] [CrossRef]
  216. Hedhli, N.; Lizano, P.; Hong, C.; Fritzky, L.F.; Dhar, S.K.; Liu, H.; Tian, Y.; Gao, S.; Madura, K.; Vatner, S.F.; et al. Proteasome inhibition decreases cardiac remodeling after initiation of pressure overload. Am. J. Physiol. Heart Circ. Physiol. 2008, 295, H1385–H1393. [Google Scholar] [CrossRef] [Green Version]
  217. Hacihanefioglu, A.; Tarkun, P.; Gonullu, E. Acute severe cardiac failure in a myeloma patient due to proteasome inhibitor bortezomib. Int. J. Hematol. 2008, 88, 219–222. [Google Scholar] [CrossRef] [PubMed]
  218. Nalepa, G.; Rolfe, M.; Harper, J.W. Drug discovery in the ubiquitin-proteasome system. Nat. Rev. Drug Discov. 2006, 5, 596–613. [Google Scholar] [CrossRef] [PubMed]
  219. Petroski, M.D. The ubiquitin system, disease, and drug discovery. BMC Biochem. 2008, 9 (Suppl. 1), S7. [Google Scholar] [CrossRef] [Green Version]
  220. Zhang, W.; Sidhu, S.S. Development of inhibitors in the ubiquitination cascade. FEBS Lett. 2014, 588, 356–367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  221. Jia, L.; Sun, Y. SCF E3 ubiquitin ligases as anticancer targets. Curr. Cancer Drug Targets 2011, 11, 347–356. [Google Scholar] [CrossRef] [Green Version]
  222. Huang, X.; Dixit, V.M. Drugging the undruggables: Exploring the ubiquitin system for drug development. Cell Res. 2016, 26, 484–498. [Google Scholar] [CrossRef]
  223. Mund, T.; Lewis, M.J.; Maslen, S.; Pelham, H.R. Peptide and small molecule inhibitors of HECT-type ubiquitin ligases. Proc. Natl. Acad. Sci. USA 2014, 111, 16736–16741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Rossi, M.; Rotblat, B.; Ansell, K.; Amelio, I.; Caraglia, M.; Misso, G.; Bernassola, F.; Cavasotto, C.N.; Knight, R.A.; Ciechanover, A.; et al. High throughput screening for inhibitors of the HECT ubiquitin E3 ligase ITCH identifies antidepressant drugs as regulators of autophagy. Cell Death Dis. 2014, 5, e1203. [Google Scholar] [CrossRef] [Green Version]
  225. Bongiorno-Borbone, L.; Giacobbe, A.; Compagnone, M.; Eramo, A.; De Maria, R.; Peschiaroli, A.; Melino, G. Anti-tumoral effect of desmethylclomipramine in lung cancer stem cells. Oncotarget 2015, 6, 16926–16938. [Google Scholar] [CrossRef]
  226. Azakir, B.A.; Angers, A. Reciprocal regulation of the ubiquitin ligase Itch and the epidermal growth factor receptor signaling. Cell Signal 2009, 21, 1326–1336. [Google Scholar] [CrossRef] [PubMed]
  227. Santini, S.; Stagni, V.; Giambruno, R.; Fianco, G.; Di Benedetto, A.; Mottolese, M.; Pellegrini, M.; Barila, D. ATM kinase activity modulates ITCH E3-ubiquitin ligase activity. Oncogene 2014, 33, 1113–1123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  228. Malhi, G.S.; Gessler, D.; Outhred, T. The use of lithium for the treatment of bipolar disorder: Recommendations from clinical practice guidelines. J. Affect. Disord. 2017, 217, 266–280. [Google Scholar] [CrossRef]
  229. Wang, X.; Fang, Z.; Wang, A.; Luo, C.; Cheng, X.; Lu, M. Lithium Suppresses Hedgehog Signaling via Promoting ITCH E3 Ligase Activity and Gli1-SUFU Interaction in PDA Cells. Front. Pharmacol. 2017, 8, 820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  230. Hamstra, S.I.; Kurgan, N.; Baranowski, R.W.; Qiu, L.; Watson, C.J.F.; Messner, H.N.; MacPherson, R.E.K.; MacNeil, A.J.; Roy, B.D.; Fajardo, V.A. Low-dose lithium feeding increases the SERCA2a-to-phospholamban ratio, improving SERCA function in murine left ventricles. Exp. Physiol. 2020, 105, 666–675. [Google Scholar] [CrossRef]
  231. Heitmeier, T.; Sydykov, A.; Lukas, C.; Vroom, C.; Korfei, M.; Petrovic, A.; Klingel, K.; Gunther, A.; Eickelberg, O.; Weissmann, N.; et al. Altered proteasome function in right ventricular hypertrophy. Cardiovasc. Res. 2020, 116, 406–415. [Google Scholar] [CrossRef] [PubMed]
  232. Segura, A.M.; Frazier, O.H.; Buja, L.M. Fibrosis and heart failure. Heart Fail. Rev. 2014, 19, 173–185. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Classification of ubiquitin E3 ligases and the domain architecture of HECT-type E3 ligases. E3 ligases are classified into three groups: RING family, HECT E3 family, RBRs family. HECT E3 family are grouped into three subfamilies: NEDD4 subfamily, HERC subfamily, other E3 subfamily. RING, really interesting new genes; HECT, homologous to E6AP C-terminus; RBRs, RING-between-RINGs; NEDD4, neural precursor cell expressed developmentally downregulated 4; HERC, HECT and RLD domain containing E3 ubiquitin protein ligase; WWP, WW domain containing E3 ubiquitin protein ligase; SMURF, SMAD ubiquitin regulatory factor; HECW, HECT, C2, and WW domain containing E3 protein ligase; E6AP, E6-associated protein; HUWE1, HECT, UBA, and WWE domain containing E3 ubiquitin protein ligase 1; HACE1, HECT domain and ankyrin repeat containing E3 ubiquitin protein ligase 1; TRIP12, thyroid hormone receptor interactor 12; UBR5, ubiquitin protein ligase E3 component N-recognin 5; UBE3B, ubiquitin–protein ligase E3B; UBE3C, ubiquitin protein ligase E3C; HECTD, HECT domain E3 ubiquitin protein ligase; G2E3, G2/M phase-specific E3 ubiquitin protein ligase; AREL1, apoptosis-resistance E3 ubiquitin protein ligase 1; C2, C2 domain; WW, WW domain; RLD, RCC-like domain; ARM, armadillo repeat; UBA, UBA domain; WWE, WWE domain; ANK, ankyrin repeat.
Figure 1. Classification of ubiquitin E3 ligases and the domain architecture of HECT-type E3 ligases. E3 ligases are classified into three groups: RING family, HECT E3 family, RBRs family. HECT E3 family are grouped into three subfamilies: NEDD4 subfamily, HERC subfamily, other E3 subfamily. RING, really interesting new genes; HECT, homologous to E6AP C-terminus; RBRs, RING-between-RINGs; NEDD4, neural precursor cell expressed developmentally downregulated 4; HERC, HECT and RLD domain containing E3 ubiquitin protein ligase; WWP, WW domain containing E3 ubiquitin protein ligase; SMURF, SMAD ubiquitin regulatory factor; HECW, HECT, C2, and WW domain containing E3 protein ligase; E6AP, E6-associated protein; HUWE1, HECT, UBA, and WWE domain containing E3 ubiquitin protein ligase 1; HACE1, HECT domain and ankyrin repeat containing E3 ubiquitin protein ligase 1; TRIP12, thyroid hormone receptor interactor 12; UBR5, ubiquitin protein ligase E3 component N-recognin 5; UBE3B, ubiquitin–protein ligase E3B; UBE3C, ubiquitin protein ligase E3C; HECTD, HECT domain E3 ubiquitin protein ligase; G2E3, G2/M phase-specific E3 ubiquitin protein ligase; AREL1, apoptosis-resistance E3 ubiquitin protein ligase 1; C2, C2 domain; WW, WW domain; RLD, RCC-like domain; ARM, armadillo repeat; UBA, UBA domain; WWE, WWE domain; ANK, ankyrin repeat.
Ijms 22 06065 g001
Figure 2. A simplified model depicting the function of ITCH in the development of cardiac hypertrophy, myocardial infarction, and doxorubicin-induced cardiomyopathy. ITCH targets disheveled protein for ubiquitin-proteasome degradation, resulting in inhibition of the Wnt/β-catenin signaling pathway; ITCH attenuates cardiac hypertrophy induced by pressure overload. ITCH targets thioredoxin-interacting protein for ubiquitin–proteasome degradation and ameliorates reactive oxygen species-induced cardiotoxicity through the thioredoxin system in myocardial infarction and doxorubicin-induced cardiomyopathy models. Circ ITCH absorbs miR-330-5p, resulting in the activation of miR-330-5p targets (SIRT6, BIRC5, and ATP2A2), thereby ameliorating doxorubicin-induced cardiomyopathy by reducing DNA damage, cell death, and calcium handling defects. LRP5/6, low-density lipoprotein receptor-related protein 5/6; Circ ITCH, circular RNA ITCH; miR, microRNA; SIRT6, sirtuin 6; BIRC5, baculoviral IAP repeat containing 5; ATP2A2, ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporting 2; Ca2+, calcium.
Figure 2. A simplified model depicting the function of ITCH in the development of cardiac hypertrophy, myocardial infarction, and doxorubicin-induced cardiomyopathy. ITCH targets disheveled protein for ubiquitin-proteasome degradation, resulting in inhibition of the Wnt/β-catenin signaling pathway; ITCH attenuates cardiac hypertrophy induced by pressure overload. ITCH targets thioredoxin-interacting protein for ubiquitin–proteasome degradation and ameliorates reactive oxygen species-induced cardiotoxicity through the thioredoxin system in myocardial infarction and doxorubicin-induced cardiomyopathy models. Circ ITCH absorbs miR-330-5p, resulting in the activation of miR-330-5p targets (SIRT6, BIRC5, and ATP2A2), thereby ameliorating doxorubicin-induced cardiomyopathy by reducing DNA damage, cell death, and calcium handling defects. LRP5/6, low-density lipoprotein receptor-related protein 5/6; Circ ITCH, circular RNA ITCH; miR, microRNA; SIRT6, sirtuin 6; BIRC5, baculoviral IAP repeat containing 5; ATP2A2, ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporting 2; Ca2+, calcium.
Ijms 22 06065 g002
Figure 3. Schematics depicting the protective role of HECT-type E3 ligases for cardiac hypertrophy. WWP2 targets PARP1 for ubiquitin-proteasome degradation. HUWE1 targets c-Myc for ubiquitin–proteasome degradation. HECTD3 targets SUMO2 for ubiquitin–proteasome degradation. HACE1 ameliorates autophagy impairment. These HECT-type E3 ligases are a protective role for cardiac hypertrophy. WWP2, WW domain-containing E3 ubiquitin protein ligase 2; PARP1, Poly (ADP-ribose) polymerase-1; HUWE1, HECT, UBA, and WWE domain containing E3 ubiquitin protein ligase 1; HECTD3, HECT domain E3 ubiquitin protein ligase 3; SUMO2, small ubiquitin-like modifier 2; NFAT, nuclear factor of activated T-cells; HACE1, HECT domain and ankyrin repeat containing E3 ubiquitin protein ligase 1.
Figure 3. Schematics depicting the protective role of HECT-type E3 ligases for cardiac hypertrophy. WWP2 targets PARP1 for ubiquitin-proteasome degradation. HUWE1 targets c-Myc for ubiquitin–proteasome degradation. HECTD3 targets SUMO2 for ubiquitin–proteasome degradation. HACE1 ameliorates autophagy impairment. These HECT-type E3 ligases are a protective role for cardiac hypertrophy. WWP2, WW domain-containing E3 ubiquitin protein ligase 2; PARP1, Poly (ADP-ribose) polymerase-1; HUWE1, HECT, UBA, and WWE domain containing E3 ubiquitin protein ligase 1; HECTD3, HECT domain E3 ubiquitin protein ligase 3; SUMO2, small ubiquitin-like modifier 2; NFAT, nuclear factor of activated T-cells; HACE1, HECT domain and ankyrin repeat containing E3 ubiquitin protein ligase 1.
Ijms 22 06065 g003
Table 1. HECT-type E3 ligase and cardiac remodeling.
Table 1. HECT-type E3 ligase and cardiac remodeling.
HECT-Type E3 LigaseSubstrate/TargetMain FindingsReference
Cardiac hypertrophy
ITCHDishevelledCardiac-specific ITCH transgenic mice inhibited maladaptive hypertrophy via Wnt/β catenin signal inhibition.[80]
NEDD4-2ENaC in kidneyCardiac hypertrophy was observed in NEDD4-2 null mice on chronic high-salt diet.[81,82]
Circular RNA WWP1ANF and miR-23aCircular RNA WWP1 was dysregulated in the heart treated with isoproterenol. [83]
WWP2PARP1WWP2 conditional knockout mice (MycCre+;WWP2Fl/Fl) exacerbated isoproterenol-induced cardiac hypertrophy.[84]
E6AP Increased myocardium E6AP expression after pressure overload.[16]
HUWE1c-mycHUWE1 conditional knockout mice spontaneously developed cardiac hypertrophy.[85]
HACE1UnknownHACE1 conditional knockout mice spontaneously developed cardiac hypertrophy.[86]
HECTD3SUMO2/STAT1AAV9-medited overexpression of HECTD3 inhibited pathological hypertrophy in mice.[87]
Cardiac fibrosis
WWP2SMAD2WWP2mut/mut mice attenuated cardiac fibrosis after angiotensin II infusion and myocardial infarction.[88]
SMURF1 SMURF1 was involved in BMP-2 antagonization for TGF-β1 signal.
SMURF1 was a target of miR-10b-5p, which inhibits cardiac fibroblast activation.
[89,90]
SMURF2SMAD7Mediator of TGF-β signal.
SMURF2 mediated SMAD7 degradation was inhibited by SMAD3 inhibitor.
[91,92]
HFpEF
WWP1Not describedCardiac-specific overexpression of WWP1 developed cardiac hypertrophy with diastolic dysfunction.[93]
ANF, atrial natriuretic factor; BMP, bone morphogenic protein; ENaC, epithelial Na+ channel; HFpEF, heart failure with preserved ejection fraction; NEDD4-2, neural precursor cell expressed developmentally downregulated 4-2; PARP1, poly(ADP-ribose)polymerase-1; SMAD, small mother against decapentaplegic; SMURF, SMAD ubiquitin regulatory factor; STAT, signal transduction and activator of transcription; SUMO2, small ubiquitin-like modifier 2.
Table 2. HECT-type E3 ligase and ischemia reperfusion injury, doxorubicin cardiotoxicity, and arrhythmia.
Table 2. HECT-type E3 ligase and ischemia reperfusion injury, doxorubicin cardiotoxicity, and arrhythmia.
HECT-Type E3 LigaseSubstrate/TargetMain FindingsReference
I/R injury
NEDD4-1p-Akt, PTENOverexpression of NEDD4-1 ameliorated myocardial apoptosis after I/R injury in rat injected with NEDD4-1 lentivirus vector.[180]
SMURF2EZH2miR-322/503 ameliorated I/R injury via inhibition of SMURF2 translation.[181]
Doxorubicin cardiotoxicity
ITCHTXNIP
Unknown
Cardiac specific ITCH transgenic mice attenuated doxorubicin cardiotoxicity and myocardial infarction.
miR-34b/c inhibited myocardial injury through ITCH.
[182,183]
Circular RNA ITCHmiR-17-5p
miR-330-5p
Inhibition of apoptosis caused by H2O2.
Inhibition of doxorubicin cardiotoxicity.
[184]
[185]
Arrhythmia
NEDD4-2Nav1.5Contribution to Nav1.5 downregulation in HF.[186]
WWP1Connexin 43Cardiac-specific overexpression of WWP1 die due to ventricular arrhythmia.[187]
H2O2, hydrogen peroxide; I/R, ischemia reperfusion injury; Nav1.5, voltage-dependent Na+ channel; NEDD, neural precursor cell expressed developmentally downregulated; SMAD, small mother against decapentaplegic; SMURF, SMAD ubiquitin regulatory factor; TXNIP, thioredoxin-interacting protein.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Goto, J.; Otaki, Y.; Watanabe, T.; Watanabe, M. The Role of HECT-Type E3 Ligase in the Development of Cardiac Disease. Int. J. Mol. Sci. 2021, 22, 6065. https://doi.org/10.3390/ijms22116065

AMA Style

Goto J, Otaki Y, Watanabe T, Watanabe M. The Role of HECT-Type E3 Ligase in the Development of Cardiac Disease. International Journal of Molecular Sciences. 2021; 22(11):6065. https://doi.org/10.3390/ijms22116065

Chicago/Turabian Style

Goto, Jun, Yoichiro Otaki, Tetsu Watanabe, and Masafumi Watanabe. 2021. "The Role of HECT-Type E3 Ligase in the Development of Cardiac Disease" International Journal of Molecular Sciences 22, no. 11: 6065. https://doi.org/10.3390/ijms22116065

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop