Identification of cardiomyocyte-enriched USP20 as a downregulated factor in mouse and human cardiac hypertrophy
Recent studies suggest that deubiquitinating enzymes (DUBs) contribute to the development of pathological cardiac hypertrophy.34 We first analyzed the mRNA profiles of DUBs from seven families in the hypertrophic myocardium of mice challenged with Angiotensin (Ang II), and found significant alterations in the gene expression of Otud1, Usp28, and Usp20 (Fig. 1a). As the effects of OTUD1 and USP28 in diseased hearts have been previously documented.24,31 therefore, we sought to examine the unknown role of USP20 in cardiac pathophysiology. We confirmed Usp20 mRNA (Fig. 1b, c) and protein (Fig. 1d, e) expression levels were both attenuated in mice hearts induced by Ang II and transverse aortic constriction (TAC), compared to the control or sham group respectively. Importantly, similar results of USP20 were obtained from human hypertrophic myocardium of patients with heart failure (Fig. 1f, g).
To determine the cellular source of downregulated USP20 in the hypertrophic myocardium, we conducted single-cell RNA sequencing (scRNA-Seq) on approximately 16928 single cells from Ang II-administered mice hearts. Based on specific marker genes expression, we categorized nine cell types: cardiomyocytes (CMs), endothelial cells (ECs), fibroblasts (FBs), neutrophil (NPs), macrophages (MPs), T/NK cells, smooth muscle cells (SMCs), Lymphatic endothelial cells (Lym Ecs) and B cells. Significantly, Usp20 mRNA expression was predominantly observed in cardiomyocytes (Fig. 1h, i). Similarly, USP20 protein expression was significantly higher in cardiomyocytes compared to non-cardiomyocytes (Fig. 1j). In addition, the protein expression of USP20 in neonatal rat cardiomyocytes (NRCMs) was downregulated upon Ang II treatment in a time-dependent manner (Supplementary Fig. 1a, b). Furthermore, immunofluorescence staining of mouse heart stimulated by Ang II and TAC showed that the downregulated USP20 was primarily localized in α-actinin+ cardiomyocytes, rather than in CD68+ macrophages or vimentin+ fibroblasts (Fig. 1k, l and supplementary Fig. 2c, d). Taken together, we identified USP20 as a cardiomyocyte-enriched factor, downregulated in mouse and human hypertrophic hearts.
Cardiomyocyte-specific deficiency of USP20 exacerbates cardiac hypertrophy and dysfunction induced by Ang II
We generated cardiomyocyte-specific USP20 knockout mice (USP20 CKO) by breeding USP20fl/fl mice with Myh6-Cre mice (Supplementary Fig. 2a) and assessed the expression levels of USP20 across various organs and tissues in USP20 CKO mice (Supplementary Fig. 2b, c). USP20 CKO mice, along with littermate USP20fl/fl mice (as control counterparts), were implanted subcutaneously with osmotic mini-pumps delivering Ang II for 4 weeks (Fig. 2a). During Ang II-induced cardiac hypertrophy, serum Ang II levels did not affect the body weight (BW) of the mice (Supplementary Fig. 3a, b). Both USP20 CKO and USP20fl/fl mice exhibited a significant increase in systolic blood pressure (SBP), but no significant differences between the two groups (Supplementary Fig. 3c, d). Non-invasive echocardiography results showed that cardiomyocyte deficiency of USP20 caused more pronounced cardiac dysfunction in the mice administrated with Ang II (Fig. 2b). This was characterized by decreased ejection fraction and fractional shortening (Fig. 2c, d and Supplementary Table 3). USP20 CKO mice showed exacerbated Ang II-induced heart enlargement (Fig. 2e), and increased the ratio of heart weight (HW) to BW (Fig. 2f), as well as the ratio of HW to tibial length (TL) (Fig. 2g). Cardiomyocyte size, as shown by H&E staining (Fig. 2h) and wheat germ agglutinin (WGA) staining (Fig. 2i, j), exhibited similar patterns of hypertrophy. Furthermore, cardiac fibrosis was significantly increased in Ang II-administered USP20 CKO mice compared to USP20fl/fl mice assessed with masson's trichrome staining (Fig. 2k, l and Supplementary Fig. 3e, f). Additionally, the mRNA levels of hypertrophic markers Myh7 and Anp were significantly increased in the heart tissues of Ang II-administered USP20 CKO mice compared to USP20fl/fl mice (Fig. 2m). These findings reveal that cardiomyocyte-specific knockout of USP20 exacerbates cardiac hypertrophy and dysfunction induced by Ang II.
We then determined the effect of USP20 on cardiomyocyte hypertrophy in vitro. We showed that increasing protein level of USP20 further reduced the expression of hypertrophic markers (β-MYHC, ANP in protein; Myh7, Anp in mRNA) (Supplementary Fig. 4a, b, c). The Ang II-induced increase in cardiomyocyte surface area was also decreased (Supplementary Fig. 4d, e). In contrast, decreasing USP20 protein expression using siRNA (si-USP20) led to opposite effects, with an increase in hypertrophic marker expression and cardiomyocyte surface area (Supplementary Fig. 4f-j).
Deficiency of cardiomyocyte-specific USP20 aggravates cardiac hypertrophy and dysfunction induced by TAC
Next, we investigated whether USP20 played an important role in transverse aortic constriction (TAC)-induced cardiac hypertrophy in both USP20 CKO mice and USP20fl/fl mice (Fig. 3a). Post-procedure, we assessed the aortic blood flow velocity at the construction site using flow doppler ultrasound, observing a significant increase in blood flow velocity following TAC compared to the sham group (Supplementary Fig. 5a, b). Echocardiography showed that TAC-induced USP20 CKO mice exhibited more severe cardiac dysfunction than TAC-induced USP20fl/fl control mice (Fig. 3b, c, d; Supplementary Table 4). Moreover, USP20 CKO mice appeared exacerbated cardiac hypertrophic phenotype in response to TAC, such as gross heart size (Fig. 3e), HW/BW (Fig. 3f), and HW/TL (Fig. 3g). Consistently, pathological examination further confirmed that the deficiency of USP20 resulted in more pronounced myocardial hypertrophy and fibrosis (Fig. 3i-l; Supplementary Fig. 5c, d). Furthermore, the mRNA levels of Myh7 and Anp in USP20 CKO mice were significantly higher than those in USP20fl/fl mice subjected to TAC (Fig. 3m). These findings (Fig. 2 and Fig. 3) suggest that USP20 exerts a protective role in both TAC-induced and Ang II-induced cardiac hypertrophy in cardiomyocytes, suggesting that USP20 is a potential therapeutic target for preventing pathological cardiac hypertrophy.
Identification of STAT3 as a substrate of USP20 in cardiomyocytes
DUBs influence biological activities by modulating the degradation or function of substrates.20 To identify the candidate substrates modulated by USP20 in cardiac hypertrophy, we utilized myocardial tissues from Ang II-induced mice in vivo and mouse-derived HL-1 cells in vitro to perform co-immunoprecipitation (Co-IP) coupled with liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS) to screen potential USP20 substrates, the workflow for this interactome analysis is illustrated in Fig. 4a. Excluding peptides related to antibody light and heavy chains and selecting target proteins with a fold change greater than 2.0, we identified that 55 USP20 physically binding proteins in HL-1 cells (dataset I, Fig. 4b), and 43 ones in heart tissues (dataset II, Fig. 4c). By comparing these two interactomes, we pinpointed potential substrates of USP20: STAT3, DSP, JUP, XRRA1. Interestingly, KEGG enrichment analysis indicated that the JAK-STAT pathway is significantly involved in USP20-mediated cardiac hypertrophy (Supplementary Fig. 6a). Therefore, our results suggest that USP20 may prevent cardiac hypertrophy through STAT3 as a substrate.
We next performed validation of the mass spectrometry and found that USP20 interacts with STAT3 in cardiac tissues and neonatal rat cardiomyocytes (NRCMs) following Ang II treatment (Fig. 4d, e), suggesting that USP20 directly binds to the STAT3 protein in cardiomyocytes. Next, we verified the interaction between USP20 and STAT3 by co-transfection of Flag-USP20 and His-STAT3 plasmids into NIH3T3 cells (Fig. 4f). Additionally, immunofluorescence analysis confirmed the co-localization of both exogenous and endogenous USP20 and STAT3 in NIH3T3 cells (Fig. 4g, h) and NRCMs (Supplementary Fig. 6b, d). USP20 is composed of four distinct structural domains: the N-terminal zinc-finger ubiquitin-binding domain (ZnF-UBP), the catalytic (USP) domain, and two tandem DUSP domains (DUSP1, DUSP2).35 Given that the ZnF-UBP domain of USP20 may play a physiological role unrelated to its ubiquitin-binding capacity.35 we generated three USP20 mutants to delineate which domain interacts with STAT3 (Fig. 4i). By co-transfecting STAT3 and the mutated USP20 plasmids into NIH3T3 cells, we found that USP20 could not bind to STAT3 when amino acids 791 to 894 were deleted, whereas mutations in other domains did not affect binding (Fig. 4j). Taken together, we identified STAT3 is a substrate of USP20 in cardiomyocytes, in which USP20 directly binds to STAT3 through its DUSP2 domain.
USP20 attenuates the K63-linked deubiquitination of STAT3 at residue K177 through the active site H645
Previous studies have demonstrated that STAT3 can be modified by K63-linked ubiquitin chains, which in turn affects its phosphorylation and protein function.36 This post-translational modification plays a crucial role in regulating the activity of STAT3 in various cellular processes, including its stability, localization, and interaction with other proteins.37 Given that USP20 is a deubiquitinase that directly binds to STAT3, we determined whether USP20 could modulate the K63 ubiquitination of STAT3, thereby influencing its biological activity. Overexpression of Flag-USP20 did not increase the STAT3 protein level in NIH3T3 cells (Fig. 5a, b). The same phenomenon was observed in Ang II-induced USP20 CKO mice (Fig. 5c, d). We then generated USP20-knockout NIH3T3 cells (gUSP20) by using CRISPR/Cas9 technology. When cycloheximide (CHX) was added to USP20-knockout cells to inhibit new protein synthesis, the degradation rate of STAT3 protein remained unchanged (Fig. 5e, f). Ubiquitin has seven Lys residues (K6, K11, K27, K29, K33, K48, and K63) that can form polyubiquitin chains, with K48 and K63 linkages being the most prevalent.21 Thus, we transfected gUSP20 cells with mutated ubiquitin plasmids containing only active K48 (HA-K48) or K63 (HA-K63) sites and observed that the ubiquitin molecules on STAT3 were significantly increased in gUSP20 cells transfected with the HA-K63 plasmid compared to wild-type cells (Fig. 5g). Moreover, the K63 regulated ubiquitination level of STAT3 protein was much higher in myocardium tissues of Ang II-induced USP20 CKO mice than those of USP20fl/fl mice (Fig. 5h). Together, these results demonstrate that USP20 removes K63-linked ubiquitin molecules from STAT3.
DUBs can cleave the amide bonds between ubiquitin molecules and substrate using active sites such as cysteine and histidine.38 To explore the specific roles of these active sites in USP20, we generated mutants by substituting glycine for cysteine at position 154 and histidine at position 645 (Fig. 5i), both of which are highly conserved across species (Supplementary Fig. 7a). Then the mutants, USP20C154A and USP20H645A, were evaluated for their ability to bind and deubiquitinate STAT3. Our results showed that both mutants could still bind to STAT3 (Supplementary Fig. 7b). However, the USP20C154A mutant retained its deubiquitination activity, while the USP20H645A could no longer remove ubiquitn molecules from STAT3 (Fig. 5k). To further elucidate the role of the H645 site in cardiomyocytes, we assessed the impact of the H645 mutation on USP20's protective effects against Ang II-induced cardiomyocyte hypertrophy. The mutation of H645 abolished USP20's protective effect against Ang II-induced cardiomyocyte hypertrophy (Supplementary Fig. 7c-g). These findings reveal that USP20 promotes STAT3 deubiquitination and function through the active site H645, and this regulation is crucial for its protective role against cardiac hypertrophy.
STAT3 consists of six domains: N domain (ND), Coil-coil domain (CCD), DNA binding domain (DBD), Linker domain (LD), SH2 domain (SH2D), and TAD domain (TAD).39 We created six STAT3 truncation mutants, each lacking one of these domains (Supplementary Fig. 8a), to determine which domain is responsible for binding USP20. After co-transfection of these mutants with Flag-USP20 plasmid in HEK293T cells, it showed that the absence of amino acids 130–320, which constitute the CCD domain, disrupted the binding between STAT3 and USP20 (Supplementary Fig. 8b). These results suggest that STAT3 binds to USP20 through its CCD domain, which is known to regulate the phosphorylation and nuclear translocation of STAT3 via allosteric regulation of SH2.40 As for the ubiquitination sites on STAT3 involved in pathological cardiac hypertrophy have not been reported, we utilized affinity-based ubiquitin peptide enrichment ubiquitomics to investigate the specific ubiquitination sites on STAT3 modulated by USP20 (Supplementary Fig. 9a). Through analyzed the results, we identified 15 potential ubiquitination lysine residues K87, K97, K161, K177, K244, K294, K348, K354, K548, K551, K601, K615, K626, K631 and K707 in STAT3 (Supplementary Fig. 9b). Given that USP20 binds to the CCD domain (130-320aa) of STAT3, we focused on lysine within this region (K161, K177, K244, and K294) and constructed mutant plasmids with lysine-to-arginine substitutions to simulate the deubiquitinated state of STAT3. Interestingly, the expression of p-STAT3 in nuclear is significantly decreased when K177, but not the other lysine residues, was mutated (Supplementary Fig. 9c, d). We then selected the K177 and examined its role in the USP20-mediated STAT3 ubiquitination in Ang II-incubated cardiomyocytes. We observed that the K177R mutation did not disrupt USP20-STAT3 binding but resulted in significantly lower STAT3 ubiquitination compared to the wild-type, and this reduction was not further decreased by USP20 overexpression (Fig. 5k). Taken together, USP20 eliminates the K63-linked deubiquitination of STAT3 at residue K177 through its active site H645 (Fig. 5l).
USP20 protects heart from cardiac hypertrophy by inhibiting STAT3 nuclear transcription to promote CARM1 expression
STAT3 serves multiple biological functions, primarily acting as a transcription factor that regulates the expression of numerous genes coding for various proteins.28 Therefore, we further evaluated whether USP20 could enhance the nuclear translocation of STAT3. Previous study reported that STAT3 activation is controlled by phosphorylation at tyrosine 705 (pTyr705).41,42 Therefore, we determined the level of p-STAT3 at Try705 following USP20 overexpression in Ang II-stimulated NRCMs. Overexpression of USP20 remarkably decreased the level of p-STAT3 without affecting the total STAT3 expression (Fig. 6a, b). Notably, USP20 overexpression reduced the nuclear translocation of p-STAT3 in Ang II-incubated NRCMs, whereas silencing USP20 significantly increased p-STAT3 level in nuclear (Fig. 6c, d). Interestingly, inhibition STAT3 by STAT3 inhibitor stattic attenuates STAT3 phosphorylation and nuclear translocation as well (Supplementary Fig. 10a, b). Karyopherin alpha 3 (KPNA3) has been identified as an auxiliary protein to facilitate STAT3 nuclear translocation43, we then determined whether USP20 binding to STAT3 could interfere with the STAT3-KPNA3 association, thereby reducing nuclear translocation of STAT3. IP assay showed that Ang II stimulation enhanced the interaction between STAT3 and KPNA3, however, this interaction was diminished when USP20 was overexpressed in NRCMs (Supplementary Fig. 10c).
To identify the potential downstream targets of STAT3 following Ang II stimuli, we performed cleavage under targets and tagmentation (CUT&Tag) assay with high-throughput sequencing. The results showed that STAT3 peak-related genes significantly decreased within 1000 bp (Promotor 1k) of the gene transcriptional start site (TSS) (Fig. 6E, from 17.45–13.86%). Subsequently, we analyzed the up-regulated genes in EV + Ang II vs. USP20OE + Ang II conduction and identified significant differences in the expression of cardiac function-related genes such as Stk3, Pten, Eno1, Atf4, Alkbh5, coactivator-associated arginine methyltransferase 1 (Carm1) and Pcna (Supplementary Fig. 10d). RT-qPCR analysis showed that, apart from increased expression of Carm1 and Atf4, the expression levels of other genes did not show significant alterations in USP20OE-Ang II-incubated NRCMs (Supplementary Fig. 10e). CARM1 is a crucial factor for cardiac homeostasis and regulates multiple aspects of cardiomyocyte maturation, including cellular hypertrophic growth and myofibril expansion.44 We hypothesized that USP20-STAT3 axis prevents from cardiac hypertrophy through transcriptional regulation of Carm1. To determine the interaction motif between STAT3 and Carm1, we predicted the binding sites of STAT3 to the Carm1 promoter regions by JASPAR (Fig. 6f). We analyzed the potential binding site which clustered in the − 1659 ~ 1650 bp of the Carm1 promoter regions. Using CUT&Tag and qPCR assays, we found that STAT3 directly bound to the promoter regions of Carm1, which was significantly increased by USP20 silencing (Fig. 6g, h). On the other hand, this binding was decreased when USP20 was overexpressed (Supplementary Fig. 10f, g). We also predicted that STAT3 peak enriched to the promoter region of Carm1 by using ENCODE database (Fig. 6i). Luciferase assays were then performed by cloning the mutated − 1659 to -1650 bp fragments of the Carm1 promoter (Mut-Carm1), which lacked the putative STAT3-binding site, and using the wild-type fragment as a control (Fig. 6j). Transfection of STAT3-overexpressing in NIH3T3 cells with either wild-type or mutant Carm1 promoter plasmids showed increased luciferase expression in vectors containing WT-Carm1, but not in those with Mut-Carm1 (Fig. 6k). Taken together, these results suggest that USP20 inhibits STAT3 transcriptional activity in nuclear and promotes Carm1 expression in cardiomyocytes (Fig. 6l).
Cardiomyocyte-specific overexpression of USP20 improves cardiac hypertrophy and dysfunction induced by Ang II
We next evaluated whether USP20 overexpression has a therapeutic effect on myocardial hypertrophy. We prepared a cardiomyocyte-targeting adeno-associated virus serovar 9 (AAV9) vector and the wide type mice were subjected to AAV9 carrying USP20 (USP20OE) or empty vehicle (EV) respectively via tail vein injection. After 4 weeks, the mice were administered with Ang II to induce hypertrophy and cardiac dysfunction (Fig. 7a) and succeeded (Supplementary Fig. 11a). AAV9 administration did not alter both body weight (Supplementary Fig. 11b) and blood pressure in Ang II-challenged mice (Supplementary Fig. 11c, d). USP20 overexpression strikingly alleviated cardiac dysfunction induced by Ang II (Fig. 7b-g; Supplementary Table 5), and also resulted in reductions in cardiomyocyte size (Fig. 7h-j), fibrosis (Fig. 7k-l; Supplementary Fig. 11e-h), and hypertrophic markers (Fig. 7m), suggesting that overexpression of USP20 prevents Ang II-induced heart remodeling. More importantly, USP20 overexpression did not change the protein level of STAT3 (Supplementary Fig. 11g), and as expected, USP20 overexpression decreased the K63-ubiquitination of STAT3 in heart tissues (Supplementary Fig. 11h). Taken together, these results suggest that USP20 overexpression improves cardiac hypertrophy and dysfunction.
USP20 ameliorates cardiac hypertrophy and dysfunction by inhibiting STAT3
To assess whether USP20 can improve cardiac hypertrophy by regulating STAT3, we administered wild type and USP20 CKO mice with or without STAT3 inhibitor stattic (10mg/kg, i.g.). Stattic is a cell-permeable inhibitor that specifically targets the STAT3 protein, preventing its activation and dimerization.45 By blocking the SH2 domain, stattic prevents STAT3 from translocating to the nucleus and initiating the transcription of target genes. USP20 CKO mice was injected with the AAV9-USP20 and the empty vehicle for 4 weeks to overexpressed the level of USP20 in the hearts. Then these mice were implanted with Ang II osmotic pumps to induce cardiac hypertrophy for 4 weeks, and administered with static every day (10mg/kg, i.g., Fig. 8a). Ang II levels rose in all mice administered with Ang II (Supplementary Fig. 12a), leading to increased systolic blood pressure (Supplementary Fig. 12b, c). Stattic did not alter the body weight (Supplementary Fig. 12d). Cardiac function assessment demonstrated that STAT3 inhibitor stattic protected heart from dysfunction caused by USP20 deficiency, and more importantly, the USP20 expression in the heart failed to ameliorate Ang II-induced cardiac dysfunction when the stattic was administered (Fig. 8b, c, d; Supplementary Table 6). Similar results were observed for hypertrophic morphology (Fig. 8e-g, the cardiomyocyte size (Fig. 8h-j), the fibrosis level (Fig. 8k-l; Supplementary Fig. 12e, f), and hypertrophic markers (Fig. 8m). These results reveal that USP20 exerts a therapeutic effect on cardiac hypertrophy and dysfunction by abolishing STAT3. It was also confirmed in vitro that USP20 affects cardiomyocyte hypertrophy by regulating STAT3 (Supplementary Fig. 12g-k). Therefore, these data demonstrate that USP20 ameliorates cardiac hypertrophy by inhibiting STAT3.