Abstract
BACKGROUND:
Clearance of damaged mitochondria via mitophagy is crucial for cellular homeostasis. Apart from PARKIN, little is known about additional ubiquitin (Ub) ligases that mediate mitochondrial ubiquitination and turnover, particularly in highly metabolically active organs such as the heart.
METHODS:
In this study, we have combined in silico analysis and biochemical assay to identify Cullin-RING Ub ligase 5 (CRL5) as a mitochondrial Ub ligase. We generated cardiomyocytes and mice lacking RBX2 (also known as SAG), a catalytic subunit of CRL5, to understand the effects of RBX2 depletion on mitochondrial ubiquitination, mitophagy and cardiac function. We also performed proteomics analysis and RNAseq analysis to define the impact of loss of RBX2 on proteome and transcriptome.
RESULTS:
RBX2 and Cullin 5, two core components of CRL5, localize to mitochondria. Depletion of RBX2 inhibited mitochondrial ubiquitination and turnover, impaired mitochondrial membrane potential and respiration, increased cardiomyocyte cell death, and has a global impact on the mitochondrial proteome. In vivo, deletion of the Rbx2 gene in adult mouse hearts suppressed mitophagic activity, provoked accumulation of damaged mitochondria in the myocardium, and disrupted myocardial metabolism, leading to the rapid development of dilated cardiomyopathy and heart failure. Similarly, ablation of RBX2 in the developing heart resulted in dilated cardiomyopathy and heart failure. The action of RBX2 in mitochondria is not dependent on PARKIN, and PARKIN gene deletion had no impact on the onset and progression of cardiomyopathy in RBX2-deficient hearts. Furthermore, RBX2 controls the stability of PINK1 in mitochondria.
CONCLUSIONS:
These findings identify RBX2-CRL5 as a mitochondrial Ub ligase that regulates mitophagy and cardiac homeostasis in a PARKIN-independent, PINK1-dependent manner.
Keywords: mitophagy, RBX2, ubiquitination, PARKIN, heart failure, Cardiomyopathy, Remodeling
Graphical Abstract

INTRODUCTION
The removal of damaged mitochondria through mitophagy, a selective form of autophagy, is critical for maintaining mitochondrial health and thus cellular hemostasis. Labeling damaged mitochondria with a phosphorylated ubiquitin (Ub) chain facilitates the recruitment of mitophagy receptors (i.e., p62, NBR1) and LC3-incorporated autophagosomes to eliminate dysfunctional mitochondria 1–3. This process is reportedly driven by two key proteins: the E3 Ub ligase PARKIN and the kinase PINK1 1,2. Following mitochondrial depolarization, PINK1 is stabilized and activated on the mitochondrial outer membrane (MOM), where it phosphorylates Ub at serine 65 (pS65-Ub) and activates PARKIN4–6. Active PARKIN, in turn, mediates the ubiquitination of other mitochondrial proteins for subsequent PINK1 phosphorylation, thereby resulting in a feed-forward loop and signal amplification.
Mitochondria play a crucial role in providing energy required for the continuously beating heart. In adult cardiomyocytes, mitochondria are constitutively turned over, as indicated by a half-life of around 6–17 days 7,8 and a high mitophagy activity in unstressed adult mouse hearts 9,10. Known as a master regulator of mitophagy, PARKIN is expressed at very low levels in unstressed hearts 11, and germline or cardiac-specific deletion of Parkin had no impact on mitochondrial or cardiac function at baseline and during ageing12–14. These findings suggest the existence of PARKIN-independent mitophagy in the heart, which has been linked to receptor-mediated mitophagy15,16 and compensation via alternative Ub ligases such as HUWE1, ARIH1, MITOL and SIAH117–20. However, germline deletion of Bnip3 gene, or cardiac-specific ablation of Bnip3l, did not produce cardiac dysfunction 21,22, arguing against a dominant role for receptor-mediated mitophagy in mitochondrial turnover in cardiomyocytes. While deletion of Huwe1 and Mitol in the heart caused cardiomyopathy and heart failure23,24, neither has been directly implicated in mitochondrial ubiquitination and turnover in cardiomyocytes. More recently, the ubiquitin ligase TRAF2 was shown to regulate mitophagy and cardiac homeostasis likely through a PARKIN-dependent mechanism25. Therefore, the ubiquitin ligases that control mitochondrial ubiquitination and clearance in cardiomyocytes under physiological conditions remain unidentified.
Cullin-RING Ub ligase 5 (CRL5) belongs to the Cullin-RING Ub ligase family, the largest family of Ub ligases 26,27. CRL5 consists of four components: the scaffold protein Cul5, RING protein RBX2 (also known as SAG/ROC2/RNF7), adaptor proteins Elongin B/C and one of many substrate-recognizing receptors 28. Neddylation of cullins is required for the assembly and activation of functional CRLs29. As a core component of CRL5, RBX2 facilitates CUL5 neddylation and bridges Ub E2 to its substrates 28, which is essential for CRL5 activity. While RBX1, the other RING finger protein, shares 56% homology with RBX2, it primarily binds to other cullins (CULLIN 1, 2, 3, 4A and 4B) to activates CRLs 1–4. RBX2-CRL5 targets various substrates for proteolysis to regulate embryonic development, inflammation, viral infection, vasculogenesis and tumorigenesis 30–36. RBX2 is highly expressed in the heart of human and mice 37, but the role of RBX2-CRL5 in mitochondrial turnover in the heart has not been explored.
In this study, we identify an association of CRLs with mitochondria. We show that RBX2 and Cul5, two key components of CRL5, localize to the mitochondria and are required for mitophagy in cardiomyocytes. Deletion of Rbx2 in mouse hearts provokes accumulation of damaged mitochondria leading to the rapid development of heart failure and premature lethality. Mechanistically, loss of RBX2 inhibits mitochondrial ubiquitination and turnover and impairs mitochondrial respiration and function in cardiomyocytes in a PARKIN-independent, PINK1-dependent manner. Our results support a pivotal role for the RBX2-CRL5 axis in maintaining mitochondrial and cardiac integrity mostly likely through directly ubiquitinating mitochondrial proteins.
METHODS
Data Availability
All supporting data and the expanded Methods section are available within the article and its online Supplemental Material. RNA sequencing data are accessible in the Gene Expression Omnibus (GSE265777). The proteomics data are available in the Center for Computational Mass Spectrometry (MSV000094583).
Animals
Mice carrying a Rbx2Flox allele were previously reported36. αMHCCre/+ mice (# 005657), αMHC-MerCreMer mice (#011038) and Parkin knockout mice (#006582) were obtained from the Jackson Laboratory. Tamoxifen (Sigma, Cat# T5648) was administered to 12 to 16-week-old mice by intraperitoneal injection at a dosage of 50 mg/kg/day for 5 days or 20 mg/kg/day for 10 days (5 consecutive days with a 2-day interval in between). All mice were maintained on a C57BL/6J background. Both male and female mice were used in this study. All animal experiments were approved by the Augusta University Institutional Animal Care and Use Committee.
Statistics
Statistical analyses were performed with GraphPad Prism software 10.1. All values are presented as mean±SEM. The Shapiro-Wilk normality test was used to test for normal distribution prior to data analysis. Statistical significance tests used are specified in each figure legend. Values of P<0.05 were considered statistically significant.
RESULTS
Identification of CRL5 as a mitochondria-associated Ub ligase
Using peroxidase (APEX2)-mediated proximity biotinylation, a recent study defined the mitochondrial outer membrane (MOM)-interacting proteome38. Cross referencing this MOM-interactome with the E3 Ub ligase database (ESI Network 39) identified 261 Ub ligase proteins (Figure 1A). Among these, 59 (~22.6%) belong to the CRL family, including RBX1, RBX2, cullin proteins, and their substrate receptors (Figure S1A–B). Several neddylation enzymes, such as NAE1, UBA3, UBE2M, and multiple subunits of deneddylase COP9 signalosome (CSN), were also identified as MOM-interacting proteins (Figure S1B). Similarly, overlap of the MOM-interactome with Ub ligases included in UbiNet 2.0 40 confirmed the enrichment of CRLs in mitochondria (Figure S1C).
Figure 1. RBX2 and Cul5 localize to the mitochondria.

A, Schematic of APEX2-catalyzed biotinylation at the mitochondrial outer membrane (MOM) via fusion to a mitochondrial targeted peptide derived from MAVS (mitochondrial antiviral-signaling protein). Overlap of APEX2-MOM data with the ESI (E3-substrate interaction) network reveals the association of CRLs with mitochondria. B, Representative Western blot of biotinylated proteins in neonatal rat ventricular cardiomyocytes (NRVCs) with adenoviral (Ad) expression of APEX2-MOM. NRVCs were treated with CCCP (10 μM) before H2O2 activation. The resultant biotinylated proteins were enriched by streptavidin beads. C, Representative Western blot of cytosol and mitochondrial fractions from NRVCs treated with CCCP (10 μM) for the indicated times. Arrowhead, neddylated CUL5. Tubulin and VDAC serve as cytosol and mitochondrial markers, respectively. D, Immuno-gold electron microscopic images showing the localization of RBX2 on mitochondrial membranes (arrowheads) in NRVCs expressing HA-RBX2. NRVCs infected with Ad-GFP and Ad-HA-OMP25 (MOM protein) serve as negative and positive controls, respectively. Scale bars, 0.5 μm. E, Representative Western blot of mitochondria with or without proteinase K (PK) treatment for 30 min on ice after isolation from NRVCs. F, Confocal images showing the colocalization (arrowhead) of RBX2 with mitochondria in CCCP-treated cardiomyocytes. NRVCs with adenoviral expression of HA-RBX2 were treated with CCCP for 3 hours and stained or immunostained as indicated. HA, green. Mitotracker, red. TOMM20, blue. Line scan co-localization analysis was done for all channels. Scale bars, 50 μm.
To validate the association of CRLs with mitochondria, neonatal rat ventricular cardiomyocytes (NRVCs) were infected with adenovirus expressing APEX2-MOM and treated with H2O2 to trigger biotinylation of MOM-interacting proteins. We detected the presence of MOM proteins (TOMM20, CISD1, TOMM40, SAMM50 and VDAC) and discernible levels of RBX2 in enriched biotinylated proteins at baseline (Figure 1B and Figure S1D). Interestingly, treatment with the mitochondrial uncoupler carbonyl cyanide chlorophenylhydrazone (CCCP) increased the biotinylated forms of RBX2 and CUL5, but not those of RBX1, indicating enhanced association of RBX2 and CRL5 with mitochondria (Figure 1B). Western blot following subcellular fractionation detected the presence of RBX2 and CUL5, as well as neddylation enzymes, in crude mitochondrial lysates (Figure 1C). Notably, neddylated CUL5 was predominant in mitochondria versus the cytosol, implying a role for active CRL5 in mitochondria. Immuno-gold electron microscopy detected the expression of exogenous RBX2 on mitochondrial membranes, consistent with the expression pattern of MOM protein OMP25 (Figure 1D). Moreover, in isolated mitochondria, RBX2 and CUL5, like the MOM protein TOMM20, were more sensitive to proteolysis induced by proteinase K (PK), compared with mitochondrial inner membrane proteins (ATP5a and UQCRC2) and matrix protein (SOD2) (Figure 1E). Immunostaining showed that CCCP treatment induced RBX2 recruitment to mitochondria (Figure 1F). Moreover, immunostaining of myocardium section also revealed the presence of RBX2 in mitochondria (Figure S1E). Together, these findings suggest RBX2 and CUL5 proteins partially localize to the mitochondrial outer membrane, and mitochondrial stress promotes their mitochondrial translocation.
RBX2 mediates mitochondrial ubiquitination and turnover
To define the functional role of CRL5 in mitochondria, RBX2 was silenced in NRVCs with two different siRNAs, which reduced CUL5 protein levels likely due to the disassembly and degradation of the CRL5 complex (Figure 2A and Figure S2A). Silencing of RBX2 markedly attenuated CCCP-induced pS65-Ub in both total cell lysates and mitochondrial fractions, in conjunction with reduced levels of P62 (autophagy receptor) and the accumulation of mitochondrial proteins (ATP5A, SDHB, MCQRC2, NDUFB8) (Figures 2A and 2B). Inhibition of proteasome function with Bortezomib (BZM) did not restore the levels of pS65-Ub and total ubiquitinated proteins in mitochondrial fractions in CCCP-treated, RBX2-deficient cardiomyocytes (Figure S2B), indicating that RBX2 does not target these mitochondrial proteins for proteasomal degradation. Assessment of mitolysosome numbers with cell permeable Mtphagy dye 41 revealed a significant reduction of mitophagic vesicles in RBX2-deficient cardiomyocytes at baseline and following CCCP treatment (Figure 2C and 2D). Moreover, the mitophagy flux assay showed a diminished turnover of LC3-II and P62 in mitochondria in RBX2-deficient cardiomyocytes (Figure 2E and S2C). Together, these data demonstrate that RBX2 is necessary for mitochondrial ubiquitination and turnover in cardiomyocytes.
Figure 2. RBX2 mediates mitochondrial ubiquitination and mitophagy.

A, Western blot of total cell lysates. NRVCs were transfected with indicated siRNAs, followed by CCCP (10 μM) treatment for the indicated times. B, Western blot of mitochondrial (mito) and cytosol (cyto) extracts. Cells were treated as described in A. C, Representative confocal images of live NRVCs stained with Mtphagy dye (red) and LysoTracker (blue) showing mitophagic vesicles. D, Quantification of mitophagic puncta per cell. E, Western blot of mitochondrial extracts. NRVCs were transfected with indicated siRNAs and treated with lysosomal protease inhibitors, E64d (E, 10 μg/ml) and pepstatin A (P, 10 μg/ml) for 6 hours before harvest. Fold-change of LC3-II and p62 levels between cells treated with and without lysosome inhibitors, indicative of mitophagy flux, is quantified on the right. F, Oxygen consumption rate (OCR) assessed by Seahorse analysis. Basal (Bas.) and maximal (Max.) respiration (resp.) are quantified on the right. siLuci: n=6, siRBX2: n=5 biological replicates. Data are representative of 3 repeats. G, Representative confocal images of NRVC stained with TMRM (1 μM, red) and MitoTracker (1 μM, green). The mean fluorescent intensity per view from 7–8 views (over 100 cells) per group is quantified on the right. H, Representative confocal images of NRVC stained with MitoSOX (red). The mean fluorescent intensity per view from 10 views (over 100 cells) per group is quantified on the right. Two-way ANOVA with the Tukey multiple comparisons test was used in D, the Mann-Whitney test in E-F, and unpaired t test in G-H.
Mitophagy maintains mitochondrial fitness and cellular health. Seahorse analysis demonstrated that depletion of RBX2 significantly decreased basal and maximal mitochondrial respiration and ATP production (Figure 2F). Furthermore, RBX2-deficient cardiomyocytes exhibited decreased mitochondrial membrane potential and elevated mitochondrial reactive oxygen species (Figure 2G–H). Consequently, RBX2-deficient cardiomyocytes were more susceptible to CCCP-induced cellular injury and cell death (Figure S2D–S2F). Thus, these data suggest that RBX2 plays an important role in maintaining cardiomyocyte mitochondrial integrity.
RBX2 is indispensable for the function of adult mouse heart
We next generated tamoxifen (TAM)-inducible, cardiomyocyte-specific RBX2 knockout (RBX2iCKO, hereafter iCKO) mice by crossing Rbx2Flox mice with αMHCMerCreMer (MCM) mice (Figure 3A). Tamoxifen administration (50 mg/kg/d for 5 days) induced efficient cardiac RBX2 depletion and a concomitant reduction of CUL5 proteins in the iCKO hearts, with no change in CUL3 or CUL4a (Figure 3B and 3C). Nearly 40% of iCKO mice died within two weeks after tamoxifen injection (Figure 3D). Compared with MCM and F/F mice, iCKO mice promptly developed dilated cardiomyopathy and heart failure at 12 days after tamoxifen injections, as evidenced by enlarged heart size, increased ratios of heart weight to tibial length and lung weight to tibial length, left ventricular (LV) wall thinning, significant LV chamber dilatation and severely impaired cardiac contractility (Figures 3E–3H, Table S1). Consistently, deletion of RBX2 led to significantly increased cardiomyocyte cell size, marked cardiomyocyte apoptosis, upregulated expression of myocardial stress markers (Acta1, Myh7) and collagen (Col1a, Col3a), and reduced expression of Myh6 (Figure 3I–K). To control for cardiotoxicity resulting from the MCM transgene42, we employed a two-week regimen of tamoxifen injections at lower doses (20 mg/kg/d for 10 days with a 2-day interval after the first 5 injections). This regimen also resulted in cardiomyopathy in iCKO mice in comparison with MCM and F/F mice (Figure S3A–S3E, Table S2). Cardiac dysfunction was not as pronounced following the low-dose tamoxifen injection protocol, possibly due to less efficient deletion of Rbx2. Collectively, these data demonstrate that RBX2 is necessary to maintain normal function of the adult heart.
Figure 3. Deletion of Rbx2 in adult heart leads to heart failure and lethality.

A, Schematics of creation of tamoxifen-inducible, cardiac-specific RBX2 knockout (iCKO) mice. B, Western blot of indicated proteins in mouse hearts at 12 days after tamoxifen injection. C, Quantification of B. D, Survival curve. E, Gross morphology of mouse heart (top) and hematoxylin and eosin staining of myocardium section (bottom) at 12 days after tamoxifen injection. F, Heart weight to tibial length ratio and lung weight to tibial length ratio. F/F: n=13, MCM: n=5, iCKO: n=18 mice. G, Representative B-mode images. H, Quantification of echocardiographic parameters before (F/F: n=17, MCM: n=6, iCKO: n=23 mice) and after (F/F: n=12, MCM: n=6, iCKO: n=12 mice) tamoxifen treatment. I, Wheat germ agglutinin (WGA) staining (left) of myocardium sections and quantification of cardiomyocyte (CM) cross-sectional area (right). More than 100 cells/heart and 3 and 12 hearts from F/F and iCKO mice, respectively, were quantified. J, Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining of myocardium sections (left) and quantification (right). Three fields per heart, and 3 hearts per group, were quantified. K, qPCR analysis of the indicated genes. F/F: n=4, iCKO: n=4 mice. The Kruskal-Wallis test with the Dunn multiple comparisons test was used in C, F and H, Log-rank (Mantel-Cox) test in D, the Mann-Whitney U test in I-K.
Perinatal deletion of RBX2 results in cardiomyopathy during ageing
To determine the functional importance of RBX2 in the developing heart, we created αMHCCre-mediated RBX2 knockout (RBX2CKO, hereafter CKO) mice (Figure S4A). Deletion of Rbx2 in postnatal hearts reduced CUL5 expression but increased the levels of RBX1 protein (Figure S4B–S4D). While endothelial-specific deletion of RBX2 causes embryonic lethality 35, CKO mice were viable and morphologically indistinguishable from their littermate controls. Serial echocardiography showed that compared with F/F and αMHCCre mice, RBX2CKO mice exhibited discernible cardiac dysfunction (EF%: 65.9% of CKO vs 73.9% of Rbx2F/F) as early as 1 month of age and severe deterioration by 8 months of age (Figure S4E). αMHCCre mice develop cardiomyopathy during ageing43,44. At 8 months of age, CKO hearts were enlarged compared with the hearts from F/F and αMHCCre mice and exhibited more pronounced LV wall thinning and systolic dysfunction (Figure S4E–H, Table S3). Moreover, CKO hearts exhibited more myocardial interstitial fibrosis and heightened expression of cardiac stress maker genes such as Nppa, Nppb, Mhy7 and Acta1, and downregulation of Atp2a2 expression (Figure S4I–J), indicative of adverse cardiac remodeling. These data support a crucial role of RBX2 in maintenance of cardiac homeostasis.
RBX2 deficiency has a major impact on the mitochondrial proteome
To gain mechanistic insight into how RBX2 regulates cardiomyocyte function, we performed tandem mass tag (TMT)-based quantitative proteomics analyses on total cell lysates from control (CTL) and RBX2-deficient (KD) cardiomyocytes in the absence and presence of CCCP treatment (Figure 4A). Principal component analysis (PCA) of protein abundances showed distinct separation between the different groups (Figure 4B). Mass spectrometry (MS) analysis did not identify any differentially expressed proteins (DEPs) between CTL and KD groups, between CTL+CCCP and CTL groups, and between KD+CCCP and KD groups (FDR<0.1, FC>1.3 or <0.77) (Figure 4C). These data indicate a negligible impact of RBX2 deficiency and CCCP treatment (for 6 hours) on the global proteome. In contrast, following CCCP treatment, ~18.5% of the quantitively detected proteins were dysregulated (180 downregulated and 269 upregulated) in RBX2-deficient cardiomyocytes compared with CCCP-treated CTL cardiomyocytes (Figure 4C and 4D, Table S4). Gene ontology analysis identified the enrichment of upregulated proteins in the pathways relevant to oxidative phosphorylation, diabetic cardiomyopathy, cardiac muscle contraction, electron transportation chain and in mitochondrial subcompartments such as matrix and outer membrane, whereas the downregulated proteins are enriched in pathways related to actin cytoskeleton organization and the sarcomere (Figure S5A–5B). Notably, 127 (47.2%) of the 269 upregulated proteins are annotated in MitoCarta 3.0 (Figure 4E), whereas only 10 (5.6%) of the 180 downregulated proteins are in mitochondria. These dysregulated mitochondrial proteins localize in different mitochondrial subcompartments (Figure S5C and 5D). These data are consistent with a critical role for RBX2 in the regulation of mitochondrial turnover.
Figure 4. RBX2-regulated mitochondrial proteome.

A, Scheme of procedures for identification Rbx2-regulated proteome in NRVCs. NRVCs were transfected with indicated siRNAs, followed by CCCP (10 μM) treatment for 6 hours. Cell lysates were collected for trypsin digestion. The resultant peptides were labeled by TMT before mass spectrometry analysis. B, Principal component analysis (PCA) of normalized protein expression in whole cell proteome. CTL, siLuci. KD, siRBX2. C, Analysis of the percentage of differential expression of proteins (DEPs) in each group. D, Volcano plot of differentially expressed proteins (Sig., blue and red) in CCCP-treated RBX2-deficient CMs (KD) compared with CCCP-treated CTL. E, Venn diagram showing overlap of RBX2-regulated proteome and mitochondrial proteins annotated in MitoCarta 3.0. F, Venn diagram showing the identification of RBX2-regulated MOMs. G, Heatmap showing the relative expression of MOM proteins amongst the groups of cells. H, Western blot of MOM proteins in control and RBX2-deficient cardiomyocytes. N=4 biological replicates per group. Multiple unpaired t test with the Holm-Šídák correction was used. I, Western blot of WT and RBX2KO Hela cells treated with cycloheximide (CHX, 100 nM) for indicated times. RBX2 was deleted in Hela cells via CRISPR/Cas9-mediated gene editing. J, WT and RBX2KO Hela cells were transfected with plasmids expressing HA-Ub (pCDNA3-HA-Ub), treated with a proteasome inhibitor Bortezomib (BZM, 100 nM) for 6 hours, and subjected to immunoprecipitation followed by Western blot. The Mann-Whitney U test was used in I.
Crossing the mitochondrial DEPs with MOMs annotated in MitoCarta 3.0 and the APEX2-MOM study 38 identified 21 MOMs as potential RBX2 substrates (Figure 4F). All of these MOMs except ITGA3 were upregulated in the KD+CCCP group compared with the CTL+CCCP group and showed a trend towards upregulation, though not statistically significant, in the KD group compared with the CTL group (Figure 4G). Furthermore, among 112 MOM proteins annotated in MitoCarta 3.0, 37 were detected by our assay and 10 were significantly upregulated in RBX2-deficient cardiomyocytes (Figure S5E). Immunoblotting confirmed the upregulation of these MOM proteins (TOMM20, TOMM40, VDAC1, CISD1, RHOT2, SAMM50, ACSL1) and MIM proteins (HSP60, AGK, CYC1, NUDUFS5), without discernable changes in their transcript levels in RBX2-deficient cardiomyocytes (Figure 4H, Figure S5F–5G). Cycloheximide-based pulse chase assays showed reduced degradation rate of ASCL1, VDAC, SAM50 and TOMM40 in RBX2-deficient Hela cells (Figure 4I). Moreover, immunoprecipitation showed that RBX2 deficiency inhibits the ubiquitination of SAM50 and ASCL1 (Figure 4J). Together, these data suggest that RBX2 controls the ubiquitination and degradation of a subset of MOMs.
RBX2 deficiency inhibits mitochondrial ubiquitination and mitophagic activity in the heart
We next examined the impact of RBX2 deficiency on cardiac mitochondrial turnover in iCKO hearts. Detection of phosphorylated ubiquitin in tissues is challenging45. Indeed, we did detect the myocardial levels of phosphorylated ubiquitin by western blot, likely due to a relatively low percentage of mitochondria undergoing mitophagy in the normal heart at a given time point. Immunostaining of pUb on myocardium sections from MCM detected pUb-positive mitochondria, which were significantly reduced in iCKO hearts (Figure 5A and 5B). Western blot showed a trend of reduction in mitochondrial P62, an adaptor protein that delivers mitochondria to autophagosomes for degradation46, but not those in cytosol, in iCKO hearts (Figure 5C and 5D). Consistently, immunostaining also detected diminished co-localization of p62 with mitochondria in iCKO hearts (Figure S6A). We further performed autophagy flux assay in mouse hearts by treating MCM and iCKO mice with a lysosome inhibitor bafilomycin A1 (BFA). Western blot showed that loss of RBX2 suppressed LC3-II turnover in mitochondrial fraction (Figure 5E and 5F). Examination of mitophagic activity via AAV9-mediated transduction of mt-Keima reporter47 demonstrated marked suppression of mitophagy in iCKO hearts (Figure 5G and 5H). Assessment of mitophagic activity in 2-month-old CKO hearts, in which cardiac function is largely maintained, also demonstrated decreased mitophagy activity (Figure S6B–S6C). In isolated adult cardiomyocytes, RBX2 deficiency reduced the levels of phosphorylated ubiquitin (Figure 5I) and CCCP-induced mitophagic vesicles (Figure S6D–S6E). Together, these in vivo and ex vivo data suggest that RBX2 is required for the ubiquitination of mitochondria proteins and mitochondrial turnover in the heart.
Figure 5. Impaired mitochondrial ubiquitination and mitophagy in RBX2-deficient hearts.

Adult MCM and RBX2iCKO (iCKO) mice were administered with tamoxifen (50 mg/kg/d for 5 days). Tissues were collected for indicated analyses (A-H) at 12 days after tamoxifen injections. A, Representative confocal images (left) of MCM and RBX2iCKO myocardium sections immunostained with pUb (green), HSP60 (red, mitochondrial marker) and DAPI. Scale bars, 10 μm. B, Quantification of pUb+ foci normalized by mitochondria (HSP60+) area. A total of 16 views from two hearts per group were quantified. C-D, Western blot (C) and quantification (D) of mitochondrial (mito) and cytosolic (cyto) P62 in mouse hearts. E-F, Western blot (E) and quantification (F) of mitochondrial (mito) and cytosolic (cyto) LC3-II in mouse hearts. Mice at 12 days after tamoxifen administration were intraperitoneally injected with bafilomyocin A1 (BFA, 3 μmol/kg) for 3 hours before tissue harvest. 4 hearts per group were quantified. G, Representative confocal images of mt-Keima at 488 nm and 568 nm, respectively, in epicardial cardiomyocytes and the derived heatmaps. Neonatal MCM and RBX2iCKO mice were transduced with AAV9-mt-Keima (1X1011 GC/pup). At 10 weeks of age, mice were treated with tamoxifen and intact mouse hearts were excised 12 days later and scanned for mt-Keima signals in epicardial cardiomyocytes in situ with confocal microscope. Scale bars, 20 μm. H, Quantification of relative 568/488 ratio. 20–40 views per heart, 3 hearts per group were quantified. I, Western blot of indicated proteins in adult cardiomyocytes isolated from 2-month-old CTL (RBX2F/F) or RBX2CKO (CKO) mouse hearts. Results from two different batches of cells are shown. The Mann-Whitney test was used in B, D and F, and nested unpaired t test in H.
Loss of RBX2 disrupts metabolic pathways and impairs mitochondrial homeostasis
To gain additional insights into how RBX2 regulates cardiac function, we performed transcriptomic analyses of CTL (F/F) and iCKO hearts. A total of 453 downregulated and 881 upregulated genes were identified in RBX2-deficient hearts (FC > or < 1.5, Padj < 0.05. Figure S7A–S7B, Table S5). Pathway enrichment analysis revealed enrichment of downregulated genes in fatty acid degradation, PPAR signaling and cardiac muscle contraction, while upregulated genes were enriched in extracellular matrix remodeling, relaxin signaling and PI3K-Akt signaling, among others (Figure 6A, Figure S7C). Specifically, many mitochondrial genes were downregulated (Figure 6B, Figure S7D). Transmission electron microscopy demonstrated ultrastructural abnormalities in RBX2-deficient cardiomyocytes, including focal myofibrillar lysis, enlarged and swollen mitochondria with disrupted cristae associated with lysosomes, massive mitophagic vesicles, and dilated endoplasmic reticulum (ER) (Figure 6C). Immunostaining of myocardium sections revealed a substantial increase of LAMP1 (a lysosome marker)-positive mitochondria in iCKO hearts compared with MCM hearts (Figure 6D), indicative of accumulation of damaged mitochondria. Despite the diminished mitochondrial ubiquitination, the increased mitophagic vesicles in RBX2-deficient hearts suggest the possible contribution of additional Ub ligases and receptor-mediated mitophagy to mitochondrial clearance (Figure S7E). Seahorse analysis further confirmed compromised mitochondrial bioenergetics in RBX2-deficient hearts (Figure 6E). Consistent with the transcriptomics analysis, immunoblotting showed that loss of RBX2 resulted in substantial downregulation of mitochondrial proteins (ATP5A, UQCRC2, SDHB, NDUFB8) in the heart (Figure 6F). Moreover, Parkin and PINK1 were upregulated and downregulated in RBX2-deficient hearts, respectively (Figure 6F). Collectively, these in vivo data, coupled with our in vitro findings, are consistent with the critical role for RBX2 in maintaining mitochondrial homeostasis and function in the heart.
Figure 6. Alterations in metabolic pathways and mitochondrial homeostasis in RBX2-deficient hearts.

A-B, Bulk RNA sequencing of mouse hearts at 12 days after tamoxifen (50 mg/kg/day for 5 days) treatment. A, KEGG pathways enriched in downregulated (top) and upregulated (bottom) genes in iCKO hearts. B, Chord plot showing downregulated genes involved in the indicated metabolic processes. C, Transmission electron microscopic images showing myofibril lysis (*), degenerating mitochondria (yellow arrowheads) surrounding by lysosomes (arrows), and abundant mitophagic vesicles (red arrowheads) in mutant cardiomyocytes. The number of mitophagic vesicles was quantified. Over 50 cardiomyocytes per group were quantified. D, Representative confocal images of myocardium sections immunostained with LAMP1 (green), TOMM20 (red) and DAPI (blue). Quantification of LAMP1+ puncta in mitochondria is shown. 5–6 views per heart, 2 hearts per group were quantified. E, Seahorse analysis of mitochondria isolated from mice at 8 days after tamoxifen injections. n=4 technical replicates/heart, 3 hearts/group were analyzed. Basal and maximal oxygen consumption rates are shown. F, Western blot of mitochondrial proteins in mouse hearts. *, cleaved form. Mann-Whitney test was used in C and D, and nested t test in E.
PARKIN is dispensable in RBX2-mediated mitophagy
Given the central role of PARKIN in mitochondrial ubiquitination, we asked whether RBX2 acts through PARKIN to mediate mitophagy. Silencing of RBX2 did not alter Parkin transcriptional levels (Figure S8A). PARKIN protein expression is very low in neonatal cardiomyocytes. In cardiomyocytes infected with Ad-PARKIN, RBX2 depletion insignificantly reduced PARKIN protein levels (Figure 7A). Moreover, deletion of RBX2 did not alter endogenous PARKIN expression at baseline but resulted in accumulation of PARKIN protein following CCCP treatment in SH-SY5Y cells (Figure S8B). Furthermore, we detected increased PARKIN protein levels in RBX2-deficient mouse hearts (Figure 6G). Thus, loss of RBX2 does not lead to reduced PARKIN expression. In cardiomyocytes isolated from neonatal wild-type and PARKIN null mice 48, deletion of Rbx2 exhibited similar inhibitory effects on CCCP-induced pS65-Ub in wild-type and PARKIN null cardiomyocytes and cardiac fibroblasts (Figure 7B and S8C). Similarly, depletion of RBX2 exerted a greater repressive effect on pS65-Ub than depletion of PARKIN via siRNA and remained effective in PARKIN-depleted cardiomyocytes (Figure 7C and S8D). Moreover, PARKIN overexpression dose-dependently promoted pS65-Ub following CCCP treatment, which was attenuated by siRNA-mediated depletion of RBX2 (Figure 7D), suggesting that RBX2 is required for PARKIN-mediated mitochondrial protein ubiquitination. We compared the RBX2-regulated 137 mitochondrial proteins with 47 previously identified PARKIN mitochondrial substrates in cancer cell lines 49. We detected only 9 overlapping proteins (Figure S8E), suggesting that RBX2 and PARKIN have distinct mitochondrial substrates. These data suggest that RBX2 does not require PARKIN to mediate mitochondrial ubiquitination.
Figure 7. The role of Parkin in RBX2-regulated mitochondrial ubiquitination and cardiac homeostasis.

A, Western blot of Parkin in NRVCs. Cells were infected with Ad-Parkin and transfected with indicated siRNAs. B, Representative Western blots of three independent repeats. Neonatal mouse ventricular CMs (NMVCs) were isolated from WT or Parkin−/− mouse hearts, transfected with siRNA, and treated with CCCP (10 μM) for 12 hours. C, Western blots of cell lysates from NRVCs transfected with siRNAs and treated with CCCP (10 μM). D, Western blots of cell lysates from NRVCs infected with Ad-Parkin, transfected with siRNAs, and treated with CCCP (10 μM). E, Schematics of generation of RBX2 and Parkin double knockout (RBX2CKO/Parkin−/−) mice. F, Ejection fraction and fractional shortening at 5 (Parkin−/−: n=5, RBX2Het/Parkin+/−: n=8, RBX2CKO: n=12. RBX2CKO/Parkin−/−, n=8 mice) and 8 (Parkin−/−: n=7, RBX2Het/Parkin+/−: n=7, RBX2CKO: n=11. RBX2CKO/Parkin−/−, n=8 mice) months of age. G, Survival curves of indicated mice. Mann-Whitney test was used in A, one-way ANOVA followed by post hoc Tukey test in F, and Log-rank (Mantel-Cox) test in G.
To examine potential interactions between RBX2 and PARKIN in the heart, we generated PARKIN and RBX2 double knockout (DKO) mice (Figure 7E). Consistent with previous reports12–14, PARKIN deficiency did not impair cardiac contractility in 8-month-old mice. Importantly, deletion of Parkin in CKO mice did not alter the onset or progression of dilated cardiomyopathy as demonstrated by temporal echocardiography, nor did it affect the survival rate of the CKO mice (Figure 7F, 7G, and S8F, Table S6). Together, these data suggest that RBX2 regulates cardiac homeostasis in PARKIN-independent mechanism.
RBX2 stabilizes PINK1
Phosphorylation of Ub is coordinated by PINK1 and phosphatases such as PPEF2, PGAM5 and PTEN-L 9–11. PINK1 is readily degraded and was barely detected in cultured cardiomyocytes infected with adenovirus expressing PINK1 under basal condition (Figure 8A). Depletion of RBX2 significantly inhibited CCCP-induced PINK1 stabilization without affecting the expression of PPEF2, PGAM5 and PTEN (Figure 8A). Moreover, deletion of RBX2 in Hela cells via CRISPR/Cas9 also reduced CCCP-induced expression of endogenous PINK1 (Figure 8B). RBX2 deficiency did not alter the levels of PINK1 transcripts, and the reduction of PINK1 in RBX2-deficient cardiomyocytes was attenuated by the proteasome inhibitor bortezomib (Figure 8C and 8D). To determine whether RBX2 regulates PINK1 degradation, cardiomyocytes were treated with CCCP for 3 hours to promote PINK1 accumulation, and following removal of CCCP, PINK1 degradation was assessed by cycloheximide-based pulse chase assay. Under these conditions, loss of RBX2 shortened the half-life of PINK1 (Figure 8E). Moreover, RBX2 depletion increased the ubiquitinated forms of PINK1, which was blunted by proteasome inhibition and abolished CCCP-induced PARKIN phosphorylation (Figure 8F and 8G), indicating enhanced PINK1 ubiquitination and diminished PINK1 activity. Consistently, PINK1 expression was reduced in RBX2iCKO hearts (Figure 6G). Together, these data suggest that RBX2 is required for PINK1 stabilization upon mitochondrial depolarization, which in turn enhances phosphorylation of Ub on mitochondria and thus mitochondrial ubiquitination.
Figure 8. RBX2 stabilizes PINK1.

A, Western blots of indicated proteins in NRVCs. Cells were infected with Ad-PINK1, transfected with indicated siRNAs and treated with or without CCCP (10 μM) for 12 hours. B, Western blots. RBX2 was deleted in Hela cells via CRISPR/Cas9 using a single guided RNA against RBX2 (gRBX2). WT and RBX2KO cells with treated with CCCP (10 μM) for indicated times before harvest. C, Analysis of Pink1 transcript levels in NRVCs transfected with indicated siRNAs by qPCR. D, Western blots. NRVCs were infected with Ad-PINK1, transfected with siRNAs, and treated with Bortezomib (BZM, 100 nM) for 6 hours. E, Cycloheximide-based pulse chase assay. NRVCs were transfected with siRNAs, treated with CCCP (10 μM) for 3 hours, followed by removal of CCCP, and then chased for the indicated time in the presence of cycloheximide (CHX, 100 nM). F, Immunoprecipitation of PINK1, followed by Western blots. NRVCs were transfected with indicated siRNAs and treated with or without Bortezomib (BZM, 100 nM) for 6 hours. G, Western blots of cell lysates from NRVCs transfected with indicated siRNAs and treated with CCCP (10 μM) for 3 hours. H, A proposed model showing the role of RBX2-CRL5 in regulation of physiological mitophagy and cardiac homeostasis. Mann-Whitney test was used in C.
DISCUSSION
In summary, our work identifies RBX2-CRL5 ubiquitin ligase as a novel regulator of mitophagy and cardiac homeostasis. Our evidence supports a model (Figure 8H) in which RBX2 translocates to dysfunctional mitochondria of cardiomyocytes, where it mediates the ubiquitination of mitochondrial outer membrane proteins and the clearance of damaged mitochondria in a PARKIN-independent manner. Meanwhile, RBX2 also enhances the stability of PINK1 to promote phosphorylation of Ub on depolarized mitochondria, thereby amplifying mitochondrial ubiquitination and mitophagy. Consequently, loss of RBX2 in mouse hearts impairs physiological mitochondrial turnover and provokes severe mitochondrial stress, leading to rapid development of dilated cardiomyopathy and heart failure. Our findings provide a new mechanism regulating mitochondrial quality control through targeting both mitochondrial ubiquitination and phosphorylation of ubiquitin on mitochondria. These findings may also explain why PARKIN is dispensable for physiological mitochondrial turnover in the heart.
RBX2-CRL5 is a mitochondrial Ub ligase
The role of RBX2-CRL5 in mitochondria has not been explored previously. Our studies identify RBX2-CRL5 as a mitochondrial Ub ligase. This is supported by the translocation of RBX2 and CUL5 to the mitochondrial outer mitochondrial membrane following mitochondrial depolarization, the necessity of RBX2 in mitochondrial ubiquitination and turnover, the global impact of RBX2 deficiency in mitochondrial proteome, and a crucial role of RBX2 in maintenance of mitochondrial integrity and function (Figure 1–2, Figure S1–2). Retention of neddylated CUL5, an active form of CRL5 (Figure 1C) further supports its role in mitochondrial. Interestingly, CCCP treatment induced pronounced accumulation RBX2 and CUL5 in mitochondria CCCP-treated in APEX2-MOM pulldown experiments but not in crude subcellular mitochondrial fractions (Figure 1B–1C). The discrepancy may be due to the weak, transient interactions of RBX2-CRL5 with mitochondria that are more effectively captured by biotinylation. RBX2 and CUL5 do not contain a mitochondrial targeting sequence. Such non-covalent binding of CRL5 to mitochondria may offer flexibility for CRL5 to recognize and ubiquitinate different MOM proteins in damaged mitochondria by pairing with different substrate-recognizing receptors. RBX2 was identified to be a redox sensitive protein50. Whether increased mitochondrial ROS induces translocation of RBX2 to the mitochondria is unknown and worthy of future investigation. In line with the involvement of CRLs in mitophagy, two CUL3-associating adaptors, Keap1 and Klhl10, and the core component of CRL1–4, RBX1, were reported to associate with mitochondria51,52. Therefore, it is possible that other CRLs may also regulate mitophagy in cardiomyocytes.
Essential role of RBX2 in regulation of physiological mitochondrial turnover and cardiac homeostasis
Mounting evidence demonstrates an essential role of mitophagy in the heart under the conditions of stress 53,54. In contrast, much less is known about how physiological mitophagy (mitochondrial clearance under steady-state conditions) is initiated and regulated in the heart. We noted that in normal hearts, only a small fraction of mitochondria undergo mitophagy at any given time (Figure 5A, 5G, 6D, and S6), indicating a relatively low mitophagy activity in homeostatic hearts, possibly as a safeguard to maintain overall mitochondrial homeostasis. However, accumulation of damaged mitochondria over time due to defective mitophagy can have catastrophic effects on mitochondrial and cardiac function, as seen in VPS34-, ATG5-, and PINK1-deficient hearts 55–57. In agreement with its pivotal role in mitophagy, loss of RBX2 in the postnatal and adult heart causes cardiac dysfunction and heart failure (Figure 3, S3 and S4). The severe cardiac phenotype is attributable to mitochondria dysfunction arising from defective mitochondrial turnover (Figure 5–6). Notably, RBX2 deficiency results in a rapid development of cardiomyopathy with much more pronounced cardiac dysfunction in adult hearts compared with developing hearts. The difference of disease progression rates in the two models could be due to a higher demand for energy production and thus mitochondrial homeostasis in adult hearts, the plasticity in adapting to mitochondrial dysfunction in young hearts, and perhaps tamoxifen-induced cardiotoxicity. Indeed, as seen in the cases of two ubiquitin ligases involved in mitophagy, TRAF2 and MITOL, and the key regulator ATG5, loss of these mitophagy regulators in adult hearts has a more profound impact on cardiac function, and typically leads to higher mortality, in adult mice than in developing mice24,56,58 59. Notably, RBX1, the other RING-box family protein, was upregulated by 3-fold in CKO hearts but not in iCKO hearts (Figure 3B and S4B), suggesting the possibility of compensation by RBX1 in the CKO heart. Collectively, our data identify RBX2-CRL5 as an important regulator of physiological mitophagy and cardiac integrity, although we cannot rule out the involvement of other RBX2-regulated substrates and pathways 28.
Distinct actions of RBX2 and PARKIN in mitochondrial quality control
Despite its well-established role in mitophagy, PARKIN appears non-essential for mitochondrial turnover in normal hearts11–14. Conceivably, multiple ubiquitin ligases may cooperatively survey the fitness of highly abundant mitochondrial in the heart. Among the reported Ub ligases involved in mitophagy, TRAF2 and MITOL have been shown to facilitate PARKIN recruitment to mitochondria and regulate mitophagy in the heart25,60. We presented evidence demonstrating that RBX2 does not require PARKIN to mediate mitochondrial ubiquitination and possibly even acts downstream of PARKIN (Figure 7 and S8). Moreover, loss of PARKIN does not impact cardiomyopathy induced by RBX2 deficiency, supporting a key role of RBX2 in physiological mitophagy in parallel to PARKIN. Several reports have also implicated PARKIN-independent roles for other CRLs in mitophagy. ARIH1 was shown to mediate mitophagy in PARKIN-deficient cancer cells 19. CUL9 ubiquitin ligase regulates mitochondrial quality control61, and loss of CUL9 and PARKIN together does not exaggerate mitochondria damage induced by CUL9 deficiency in neuronal cells 62. Given the well-documented role of PARKIN under stress conditions, it is possible that RBX2 and PARKIN may recognize different substrate pools (Figure S8E) to regulate mitophagy in cardiomyocytes under physiological and pathological conditions, respectively.
RBX2 is a new regulator of PINK1 expression
An interesting finding of this study is that RBX2 is required for PINK1 stabilization following mitochondrial depolarization (Figure 8). PINK1 is imported into mitochondria and cleaved by the mitochondrial protease PARL63. Cleaved PINK1 is then degraded by the proteasome through the ubiquitin ligases UBR1/UBR2/UBR464. It will be interesting in the future to explore whether RBX2 acts as a ubiquitin ligase to control the degradation of PARL or UBR1/2/4, thereby increasing PINK1 stability in damaged mitochondria. Loss of PINK1 promotes mitochondrial dysfunction in the heart eventually leading to cardiac dysfunction and heart failure57. The dual role of RBX2 in regulation of mitochondrial ubiquitination and PINK1 expression explains the severe mitochondrial and cardiac phenotypes observed in adult RBX2-deficient hearts.
Supplementary Material
NOVELTY AND SIGNIFICANCE:
What Is Known?
Defects in mitochondrial quality control are associated with heart failure and cardiac disease.
PARKIN mediates mitochondrial ubiquitination but is not essential for mitochondrial turnover in homeostatic hearts.
RBX2 is the catalytic subunit of Cullin-RING ubiquitin ligase 5 known to regulate tumorigenesis and viral infection.
What New Information Does This Article Contribute?
RBX2 localizes to the mitochondria.
RBX2 regulates the ubiquitination of mitochondrial outer membrane proteins and mitophagy.
RBX2 is indispensable for the maintenance of mitochondrial and cardiac function in adult hearts.
RBX2 requires PINK1, but not PARKIN, to mediate mitophagy.
Which ubiquitin ligases mediate mitochondrial turnover in the heart remains obscure. The current study identified RBX2 as a mitochondrial ubiquitin ligase that has a dual role in regulation of mitochondrial ubiquitination and PINK1 expression. Using mice and cardiomyocytes deficient of RBX2, we demonstrated that RBX2 is crucial for mitochondrial quality control and the normal function of adult heart, potentially providing a new target to prevent and treat cardiac diseases associated with mitochondrial dysfunction.
SOURCES OF FUNDING
This study was in part supported by the US National Institutes of Health grants (R01HL124248 and R01HL165205 to H.S. and R01HL146807A1 to J.L.) and the American Heart Association grant (959479 to H.S.).
Non-standard abbreviations and acronyms:
- RBX2
RING-Box Protein 2
- SAG
Sensitive to Apoptosis Gene
- Ub
Ubiquitin
- pS65-Ub
phosphorylated Ub at serine 65
- MAVS
mitochondrial antiviral-signaling protein
- AAV
adeno-associated virus
- AV
adenovirus
- siRNA
Small interfering RNA
- GFP
green fluorescent protein
- CUL
cullin
- RING
Really Interesting New Gene
- CRLs
cullin-RING ligases
- CSN
COP9 signalosome
- APEX2
ascorbate peroxidase 2
- mito
mitochondrial; cyto, cytosolic
- MOM
mitochondrial outer membrane
- CCCP
Carbonyl Cyanide Chlorophenylhydrazone
- OMP25
Outer membrane protein 25
- PK
proteinase K
- HA
hemagglutinin
- TMRM
Tetramethylrhodamine methyl ester perchlorate
- αMHC
α-myosin heavy chain
- CKO
cardiomyocyte-specific knockout
- TAM
tamoxifen
- TMT
tandem mass tag
- KD
knockdown
- CTL
control
- MCM
MerCreMer
- iCKO
inducible cardiomyocyte-specific knockout
- BFA
bafilomycin A1
- PCA
principle component analysis
- MS
Mass spectrometry
- DEPs
differentially expressed proteins
- FC
fold change
- FDR
False Discovery Rate
- KEGG
Kyoto encyclopedia of genes and genomes
- ER
endoplasmic reticulum
- DKO
double knockout
- CM
cardiomyocyte
- cTnT
cardiac troponin T
- NRVCs
neonatal rat ventricular cardiomyocytes
- NRVMs
neonatal mouse ventricular cardiomyocytes
- NMVFs
neonatal mouse ventricular fibroblasts
- HF
heart failure; KO, knockout
- MF
Molecular Functions
- CC
Cellular Components
- BP
Biological Process
- TUNEL
Terminal deoxynucleotidyl transferase dUTP nick end labeling
- SCF
Skp1-Cullin 1-F-box
Footnotes
Disclosure
None.
REFERENCES
- 1.McWilliams TG, Muqit MM. PINK1 and Parkin: emerging themes in mitochondrial homeostasis. Curr Opin Cell Biol. 2017;45:83–91. [DOI] [PubMed] [Google Scholar]
- 2.Dorn GW, 2nd. Parkin-dependent mitophagy in the heart. J Mol Cell Cardiol. 2016;95:42–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Harper JW, Ordureau A, Heo JM. Building and decoding ubiquitin chains for mitophagy. Nat Rev Mol Cell Biol. 2018;19:93–108. [DOI] [PubMed] [Google Scholar]
- 4.Narendra D, Tanaka A, Suen DF, Youle RJ. Parkin is recruited selectively to impaired mitochondria and promotes their autophagy. J Cell Biol. 2008;183:795–803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Narendra DP, Jin SM, Tanaka A, Suen DF, Gautier CA, . . . Youle RJ. PINK1 is selectively stabilized on impaired mitochondria to activate Parkin. PLoS Biol. 2010;8:e1000298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Narendra D, Kane LA, Hauser DN, Fearnley IM, Youle RJ. p62/SQSTM1 is required for Parkin-induced mitochondrial clustering but not mitophagy; VDAC1 is dispensable for both. Autophagy. 2010;6:1090–1106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Lau E, Cao Q, Ng DC, Bleakley BJ, Dincer TU, . . . Ping P A large dataset of protein dynamics in the mammalian heart proteome. Sci Data. 2016;3:160015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Menzies RA, Gold PH. The turnover of mitochondria in a variety of tissues of young adult and aged rats. J Biol Chem. 1971;246:2425–2429. [PubMed] [Google Scholar]
- 9.Stotland A, Gottlieb RA. alpha-MHC MitoTimer mouse: In vivo mitochondrial turnover model reveals remarkable mitochondrial heterogeneity in the heart. J Mol Cell Cardiol. 2016;90:53–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Sun N, Yun J, Liu J, Malide D, Liu C, . . . Finkel T Measuring In Vivo Mitophagy. Mol Cell. 2015;60:685–696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Song M, Gong G, Burelle Y, Gustafsson AB, Kitsis RN, . . . Dorn GW, 2nd. Interdependence of Parkin-Mediated Mitophagy and Mitochondrial Fission in Adult Mouse Hearts. Circ Res. 2015;117:346–351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Kubli DA, Zhang X, Lee Y, Hanna RA, Quinsay MN, . . . Gustafsson AB. Parkin protein deficiency exacerbates cardiac injury and reduces survival following myocardial infarction. J Biol Chem. 2013;288:915–926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Piquereau J, Godin R, Deschenes S, Bessi VL, Mofarrahi M, . . . Burelle Y. Protective role of PARK2/Parkin in sepsis-induced cardiac contractile and mitochondrial dysfunction. Autophagy. 2013;9:1837–1851. [DOI] [PubMed] [Google Scholar]
- 14.Woodall BP, Orogo AM, Najor RH, Cortez MQ, Moreno ER, . . . Gustafsson AB. Parkin does not prevent accelerated cardiac aging in mitochondrial DNA mutator mice. JCI Insight. 2019;5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Zhu Y, Massen S, Terenzio M, Lang V, Chen-Lindner S, . . . Brady NR. Modulation of serines 17 and 24 in the LC3-interacting region of Bnip3 determines pro-survival mitophagy versus apoptosis. J Biol Chem. 2013;288:1099–1113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Novak I, Kirkin V, McEwan DG, Zhang J, Wild P, . . . Dikic I. Nix is a selective autophagy receptor for mitochondrial clearance. EMBO Rep. 2010;11:45–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Di Rita A, Peschiaroli A, P DA, Strobbe D, Hu Z, . . . Cecconi F. HUWE1 E3 ligase promotes PINK1/PARKIN-independent mitophagy by regulating AMBRA1 activation via IKKalpha. Nat Commun. 2018;9:3755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Yun J, Puri R, Yang H, Lizzio MA, Wu C, . . . Guo M. MUL1 acts in parallel to the PINK1/parkin pathway in regulating mitofusin and compensates for loss of PINK1/parkin. Elife. 2014;3:e01958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Villa E, Proics E, Rubio-Patino C, Obba S, Zunino B, . . . Ricci JE. Parkin-Independent Mitophagy Controls Chemotherapeutic Response in Cancer Cells. Cell Rep. 2017;20:2846–2859. [DOI] [PubMed] [Google Scholar]
- 20.Szargel R, Shani V, Abd Elghani F, Mekies LN, Liani E, . . . Engelender S. The PINK1, synphilin-1 and SIAH-1 complex constitutes a novel mitophagy pathway. Hum Mol Genet. 2016;25:3476–3490. [DOI] [PubMed] [Google Scholar]
- 21.Diwan A, Krenz M, Syed FM, Wansapura J, Ren X, . . . Dorn GW. Inhibition of ischemic cardiomyocyte apoptosis through targeted ablation of Bnip3 restrains postinfarction remodeling in mice. J Clin Invest. 2007;117:2825–2833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Diwan A, Wansapura J, Syed FM, Matkovich SJ, Lorenz JN, . . . Dorn GW, 2nd. Nix-mediated apoptosis links myocardial fibrosis, cardiac remodeling, and hypertrophy decompensation. Circulation. 2008;117:396–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Dadson K, Hauck L, Hao Z, Grothe D, Rao V, . . . 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Tokuyama T, Uosaki H, Sugiura A, Nishitai G, Takeda K, . . . Yanagi S. Protective roles of MITOL against myocardial senescence and ischemic injury partly via Drp1 regulation. iScience. 2022;25:104582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Yang KC, Ma X, Liu H, Murphy J, Barger PM, . . . Diwan A. Tumor necrosis factor receptor-associated factor 2 mediates mitochondrial autophagy. Circ Heart Fail. 2015;8:175–187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kandala S, Kim IM, Su H. Neddylation and deneddylation in cardiac biology. Am J Cardiovasc Dis. 2014;4:140–158. [PMC free article] [PubMed] [Google Scholar]
- 27.Petroski MD, Deshaies RJ. Function and regulation of cullin-RING ubiquitin ligases. Nat Rev Mol Cell Biol. 2005;6:9–20. [DOI] [PubMed] [Google Scholar]
- 28.Zhang S, Sun Y. Cullin RING Ligase 5 (CRL-5): Neddylation Activation and Biological Functions. Adv Exp Med Biol. 2020;1217:261–283. [DOI] [PubMed] [Google Scholar]
- 29.Li J, Zou J, Littlejohn R, Liu J, Su H. Neddylation, an Emerging Mechanism Regulating Cardiac Development and Function. Front Physiol. 2020;11:612927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Zhu Z, Wang L, Hao R, Zhao B, Sun L, . . . Ye RD. Cutting Edge: A Cullin-5-TRAF6 Interaction Promotes TRAF6 Polyubiquitination and Lipopolysaccharide Signaling. J Immunol. 2016;197:21–26. [DOI] [PubMed] [Google Scholar]
- 31.Teckchandani A, Laszlo GS, Simo S, Shah K, Pilling C, . . . Cooper JA. Cullin 5 destabilizes Cas to inhibit Src-dependent cell transformation. J Cell Sci. 2014;127:509–520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Okumura F, Joo-Okumura A, Nakatsukasa K, Kamura T. The role of cullin 5-containing ubiquitin ligases. Cell Div. 2016;11:1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Feng L, Allen NS, Simo S, Cooper JA. Cullin 5 regulates Dab1 protein levels and neuron positioning during cortical development. Genes Dev. 2007;21:2717–2730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Fairchild CL, Hino K, Han JS, Miltner AM, Peinado Allina G, . . . Simo S. RBX2 maintains final retinal cell position in a DAB1-dependent and -independent fashion. Development. 2018;145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Tan M, Zhao Y, Kim SJ, Liu M, Jia L, . . . Sun Y. SAG/RBX2/ROC2 E3 ubiquitin ligase is essential for vascular and neural development by targeting NF1 for degradation. Dev Cell. 2011;21:1062–1076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Tan M, Li H, Sun Y. Endothelial deletion of Sag/Rbx2/Roc2 E3 ubiquitin ligase causes embryonic lethality and blocks tumor angiogenesis. Oncogene. 2014;33:5211–5220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Sun Y, Tan M, Duan H, Swaroop M. SAG/ROC/Rbx/Hrt, a zinc RING finger gene family: molecular cloning, biochemical properties, and biological functions. Antioxid Redox Signal. 2001;3:635–650. [DOI] [PubMed] [Google Scholar]
- 38.Hung V, Lam SS, Udeshi ND, Svinkina T, Guzman G, . . . Ting AY. Proteomic mapping of cytosol-facing outer mitochondrial and ER membranes in living human cells by proximity biotinylation. Elife. 2017;6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Chen D, Liu X, Xia T, Tekcham DS, Wang W, . . . Piao HL. A Multidimensional Characterization of E3 Ubiquitin Ligase and Substrate Interaction Network. iScience. 2019;16:177–191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Li Z, Chen S, Jhong JH, Pang Y, Huang KY, . . . Lee TY. UbiNet 2.0: a verified, classified, annotated and updated database of E3 ubiquitin ligase-substrate interactions. Database (Oxford). 2021;2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Iwashita H, Torii S, Nagahora N, Ishiyama M, Shioji K, . . . Okuma K. Live Cell Imaging of Mitochondrial Autophagy with a Novel Fluorescent Small Molecule. ACS Chem Biol. 2017;12:2546–2551. [DOI] [PubMed] [Google Scholar]
- 42.Koitabashi N, Bedja D, Zaiman AL, Pinto YM, Zhang M, . . . Kass DA. Avoidance of transient cardiomyopathy in cardiomyocyte-targeted tamoxifen-induced MerCreMer gene deletion models. Circ Res. 2009;105:12–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Pugach EK, Richmond PA, Azofeifa JG, Dowell RD, Leinwand LA. Prolonged Cre expression driven by the alpha-myosin heavy chain promoter can be cardiotoxic. J Mol Cell Cardiol. 2015;86:54–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Rehmani T, Salih M, Tuana BS. Cardiac-Specific Cre Induces Age-Dependent Dilated Cardiomyopathy (DCM) in Mice. Molecules. 2019;24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Watzlawik JO, Hou X, Fricova D, Ramnarine C, Barodia SK, . . . Springer W. Sensitive ELISA-based detection method for the mitophagy marker p-S65-Ub in human cells, autopsy brain, and blood samples. Autophagy. 2021;17:2613–2628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Geisler S, Holmstrom KM, Skujat D, Fiesel FC, Rothfuss OC, . . . Springer W. PINK1/Parkin-mediated mitophagy is dependent on VDAC1 and p62/SQSTM1. Nat Cell Biol. 2010;12:119–131. [DOI] [PubMed] [Google Scholar]
- 47.Sun N, Malide D, Liu J, Rovira II, Combs CA, . . . Finkel T. A fluorescence-based imaging method to measure in vitro and in vivo mitophagy using mt-Keima. Nat Protoc. 2017;12:1576–1587. [DOI] [PubMed] [Google Scholar]
- 48.Goldberg MS, Fleming SM, Palacino JJ, Cepeda C, Lam HA, . . . Shen J. Parkin-deficient mice exhibit nigrostriatal deficits but not loss of dopaminergic neurons. J Biol Chem. 2003;278:43628–43635. [DOI] [PubMed] [Google Scholar]
- 49.Sarraf SA, Raman M, Guarani-Pereira V, Sowa ME, Huttlin EL, . . . Harper JW. Landscape of the PARKIN-dependent ubiquitylome in response to mitochondrial depolarization. Nature. 2013;496:372–376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Duan H, Wang Y, Aviram M, Swaroop M, Loo JA, . . . Sun Y. SAG, a novel zinc RING finger protein that protects cells from apoptosis induced by redox agents. Mol Cell Biol. 1999;19:3145–3155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Yamada T, Murata D, Adachi Y, Itoh K, Kameoka S, . . . Sesaki H. Mitochondrial Stasis Reveals p62-Mediated Ubiquitination in Parkin-Independent Mitophagy and Mitigates Nonalcoholic Fatty Liver Disease. Cell Metab. 2018;28:588–604 e585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Aram L, Braun T, Braverman C, Kaplan Y, Ravid L, . . . Arama E. A Krebs Cycle Component Limits Caspase Activation Rate through Mitochondrial Surface Restriction of CRL Activation. Dev Cell. 2016;37:15–33. [DOI] [PubMed] [Google Scholar]
- 53.Saito T, Hamano K, Sadoshima J. Molecular mechanisms and clinical implications of multiple forms of mitophagy in the heart. Cardiovasc Res. 2021;117:2730–2741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Diao RY, Gustafsson AB. Mitochondrial quality surveillance: mitophagy in cardiovascular health and disease. Am J Physiol Cell Physiol. 2022;322:C218–C230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Jaber N, Dou Z, Chen JS, Catanzaro J, Jiang YP, . . . Zong WX. Class III PI3K Vps34 plays an essential role in autophagy and in heart and liver function. Proc Natl Acad Sci U S A. 2012;109:2003–2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Nakai A, Yamaguchi O, Takeda T, Higuchi Y, Hikoso S, . . . Otsu K. The role of autophagy in cardiomyocytes in the basal state and in response to hemodynamic stress. Nat Med. 2007;13:619–624. [DOI] [PubMed] [Google Scholar]
- 57.Billia F, Hauck L, Konecny F, Rao V, Shen J, . . . Mak TW. PTEN-inducible kinase 1 (PINK1)/Park6 is indispensable for normal heart function. Proc Natl Acad Sci U S A. 2011;108:9572–9577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Guo X, Yin H, Li L, Chen Y, Li J, . . . Liu Q. Cardioprotective Role of Tumor Necrosis Factor Receptor-Associated Factor 2 by Suppressing Apoptosis and Necroptosis. Circulation. 2017;136:729–742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Ma X, Rawnsley DR, Kovacs A, Islam M, Murphy JT, . . . Diwan A. TRAF2, an Innate Immune Sensor, Reciprocally Regulates Mitophagy and Inflammation to Maintain Cardiac Myocyte Homeostasis. JACC Basic Transl Sci. 2022;7:223–243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Koyano F, Yamano K, Kosako H, Tanaka K, Matsuda N. Parkin recruitment to impaired mitochondria for nonselective ubiquitylation is facilitated by MITOL. J Biol Chem. 2019;294:10300–10314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Gama V, Swahari V, Schafer J, Kole AJ, Evans A, . . . Deshmukh M. The E3 ligase PARC mediates the degradation of cytosolic cytochrome c to promote survival in neurons and cancer cells. Sci Signal. 2014;7:ra67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Hollville E, Joers V, Nakamura A, Swahari V, Tansey MG, . . . Deshmukh M. Characterization of a Cul9-Parkin double knockout mouse model for Parkinson’s disease. Sci Rep. 2020;10:16886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Spinazzi M, De Strooper B. PARL: The mitochondrial rhomboid protease. Semin Cell Dev Biol. 2016;60:19–28. [DOI] [PubMed] [Google Scholar]
- 64.Yamano K, Youle RJ. PINK1 is degraded through the N-end rule pathway. Autophagy. 2013;9:1758–1769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Udeshi ND, Mani DC, Satpathy S, Fereshetian S, Gasser JA, . . . Carr SA. Rapid and deep-scale ubiquitylation profiling for biology and translational research. Nat Commun. 2020;11:359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi AH, . . . Chanda SK. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat Commun. 2019;10:1523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Zou J, Wang W, Lu Y, Ayala J, Dong K, . . . Su H. Neddylation is required for perinatal cardiac development through stimulation of metabolic maturation. Cell Rep. 2023;42:112018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Simmerman E, Qin X, Marshall B, Perry L, Cai L, . . . Baban B. Innate lymphoid cells: a paradigm for low SSI in cleft lip repair. J Surg Res. 2016;205:312–317. [DOI] [PubMed] [Google Scholar]
- 69.Ackers-Johnson M, Li PY, Holmes AP, O’Brien SM, Pavlovic D, . . . Foo RS. A Simplified, Langendorff-Free Method for Concomitant Isolation of Viable Cardiac Myocytes and Nonmyocytes From the Adult Mouse Heart. Circ Res. 2016;119:909–920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Kabaeva Z, Zhao M, Michele DE. Blebbistatin extends culture life of adult mouse cardiac myocytes and allows efficient and stable transgene expression. Am J Physiol Heart Circ Physiol. 2008;294:H1667–1674. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
All supporting data and the expanded Methods section are available within the article and its online Supplemental Material. RNA sequencing data are accessible in the Gene Expression Omnibus (GSE265777). The proteomics data are available in the Center for Computational Mass Spectrometry (MSV000094583).
