Abstract
BACKGROUND:
HIF (hypoxia inducible factor) regulates many aspects of cardiac function. We and others previously showed that chronic HIF activation in the heart in mouse models phenocopies multiple features of ischemic cardiomyopathy in humans, including mitochondrial loss, lipid accumulation, and systolic cardiac dysfunction. In some settings, HIF also causes the loss of peroxisomes. How, mechanistically, HIF promotes cardiac dysfunction is an open question.
METHODS:
We used mice lacking cardiac pVHL (von Hippel-Lindau protein) to investigate how chronic HIF activation causes multiple features of ischemic cardiomyopathy, such as autophagy induction and lipid accumulation. We performed immunoblot assays, RNA sequencing, mitochondrial and peroxisomal autophagy flux measurements, and live cell imaging on isolated cardiomyocytes. We used CRISPR-Cas9 gene editing in mice to validate a novel mediator of cardiac dysfunction in the setting of chronic HIF activation.
RESULTS:
We identify a previously unknown pathway by which cardiac HIF activation promotes the loss of mitochondria and peroxisomes. We found that DEPP1 (decidual protein induced by progesterone 1) is induced under hypoxia in a HIF-dependent manner and localizes inside mitochondria. DEPP1 is both necessary and sufficient for hypoxia-induced autophagy and triglyceride accumulation in cardiomyocytes ex vivo. DEPP1 loss increases cardiomyocyte survival in the setting of chronic HIF activation ex vivo, and whole-body Depp1 loss decreases cardiac dysfunction in hearts with chronic HIF activation caused by VHL loss in vivo.
CONCLUSIONS:
Our findings identify DEPP1 as a key component in the cardiac remodeling that occurs with chronic ischemia.
Keywords: autophagy, cardiomyocyte, DEPP1, HIF, hypoxia, mitochondria, peroxisome, VHL
Clinical Perspective.
What Is New?
We investigated how chronic HIF (hypoxia inducible factor) activation leads to many features of ischemic cardiomyopathy and identified a previously unknown pathway by which cardiac HIF activation promotes the loss of mitochondria and peroxisomes through autophagy.
We discovered that DEPP1 (decidual protein induced by progesterone 1) is a key component in the cardiac remodeling that occurs with chronic ischemia and that DEPP1 loss decreases cardiac dysfunction in the setting of chronic HIF activation.
What Are the Clinical Implications?
Our findings provide mechanistic insights that underlie key molecular changes that occur in the ischemic myocardium.
Strategies that increase mitochondrial and peroxisome abundance, such as by inhibiting DEPP1, might help preserve cardiac function in the setting of chronic ischemia.
Editorial, see p 787
Chronic HIF (hypoxia inducible factor) activation in mouse models is sufficient to produce many of the hallmarks of ischemic cardiomyopathy.1–3 For example, HIF causes the loss of cardiac mitochondria, lipid accumulation, and systolic cardiac dysfunction, which are features of human ischemic cardiomyopathy.1–3 How HIF induces these changes is poorly understood.
The HIF transcription factor, which consists of a labile α subunit (HIFα) and a stable β subunit (HIFβ/ARNT [aryl hydrocarbon nuclear translocator]), accumulates during hypoxia and activates genes that orchestrate the cellular adaptation to hypoxia.4 In the presence of oxygen, HIFα becomes hydroxylated on 1 (or both) of 2 conserved proline residues by members of the PHD1-3 (prolyl hydroxylase domain), also known as the EglN (Egl9 homolog), family. PHD enzymatic activity requires oxygen, reduced iron, and 2-oxoglutarate and is sensitive to other inputs that indirectly reflect oxygen availability, such as reactive oxygen species (ROS) and Krebs cycle intermediates. Thus, the PHD proteins act as oxygen sensors upstream of the HIFα transcription factor. Once hydroxylated, HIFα is polyubiquitinylated by an E3 ubiquitin ligase complex containing pVHL (von Hippel-Lindau protein), leading to its proteasomal degradation.4
Systematic inactivation of PHD2, especially when combined with PHD3 loss, leads to dilated cardiomyopathy, suggesting that impaired PHD activity, as would occur in the setting of chronic ischemia, causes cardiac dysfunction.5,6 Although these studies were potentially confounded by the polycythemia and volume overload that occurs with systemic PHD2 loss, cardiac-specific VHL deletion in mice is also sufficient to promote cardiomyopathy, which is completely prevented by concomitant deletion of HIF1α.2 Moreover, we and others showed that cardiomyocyte-intrinsic HIF activation is sufficient to account for many of the electron microscopic, histological, and functional changes in hearts with ischemic cardiomyopathy.1–3 Collectively these findings suggest that chronic HIF activation in cardiomyocytes plays a causal role in the pathogenesis of ischemic cardiomyopathy.
How chronic HIF activation promotes heart failure, however, remains unclear. Chronic HIF activation leads to decreased mitochondrial biogenesis, increased autophagy, increased lipid accumulation, and subsequent metabolic dysfunction.1,7,8 HIF can selectively induce mitochondria and peroxisome autophagy, 2 organelles that play major roles in oxygen, redox, energy, and lipid metabolism.9 BNIP3 (Bcl-2/adenovirus E1B 19-kDa interacting protein 3), a canonical HIF target gene, plays a role in hypoxia-induced mitochondrial autophagy and is increased in mouse hearts lacking PHD or pVHL function.10 BNIP3 loss minimizes ventricular remodeling and cardiomyocyte apoptosis after acute ischemic reperfusion injury, which suggests that inhibiting autophagy would be therapeutically useful in this acute setting.11 Whether increased autophagy is an adaptive or deleterious response to chronic cardiac ischemia and whether other specific HIF-responsive proteins are necessary for HIF to promote autophagy in the heart remains to be determined. Whether autophagy inhibition would alter the natural history of heart failure in the setting of chronic HIF activation is therefore an open question.
Here, we show that hypoxia and chronic HIF activation promotes mitochondrial and peroxisome autophagy and lipid accumulation in cardiomyocytes ex vivo and in vivo. We identify DEPP1 (decidual protein induced by progesterone 1) as a direct transcriptional target of HIF that is necessary for mitochondrial and peroxisome autophagy mediated by HIF activation, but not by other autophagy inducers, such as mammalian Target of Rapamycin (mTOR) inhibition. Upon expression, DEPP1 localizes inside mitochondria and is sufficient to promote cardiomyocyte autophagy and lipid accumulation. DEPP1 loss increases cardiomyocyte survival in the setting chronic HIF activation, and global Depp1 loss decreases cardiac dysfunction in mouse hearts lacking VHL. These studies provide new insights into how HIF activation promotes autophagy and suggest that inhibiting autophagy in the setting of chronic ischemia would prevent the loss of mitochondria and peroxisomes and preserve cardiac function.
METHODS
Detailed methods are provided in the Supplemental Material. All data needed to evaluate the conclusions in the article are present in the article or the Supplemental Material. All plasmids described herein are available from the authors on request.
Experimental Animals
All care and treatment of experimental animals were carried out in strict accordance with good animal practice as defined by the US Office of Laboratory Animal Welfare and approved by the Dana-Farber Cancer Institute (protocols 04-019 and 04-020) institutional animal care and use committee.
Human Heart Biopsy Samples
Hearts were obtained under a protocol approved by the University of Utah institutional review board.
Statistical Analysis
GraphPad Prism versions 9 and 10 were used for statistical analysis included in the main and supplemental figures. Statistical significance was calculated using the unpaired, 2-tailed Student t test or 1-way ANOVA with Šidák multiple comparisons test for post hoc pairwise comparisons. When 2 independent factors were used in the experimental design, statistical significance was calculated using a 2-way ANOVA with Šidák multiple comparisons test. Data were evaluated for normality before parametric statistics were applied using the Shapiro-Wilk test. For groups of unequal variances, data were reevaluated by t test or ANOVA with Welch correction. For comparisons for 2 groups with nonnormal distribution, the Mann-Whitney test was used. For comparisons of >2 groups with nonnormal distribution, the Kruskal-Wallis test was used. Survival curve analysis was performed using the log-rank test. P values were considered statistically significant if the P value was <0.05. For all figures, an asterisk indicates *P < 0.05 unless otherwise indicated. Error bars represent SD unless otherwise indicated. No statistical methods were used to predetermine the sample size.
RESULTS
Chronic HIF Activation Caused by Cardiac VHL Loss or Hypoxia Promotes Autophagy-Dependent Mitochondria and Peroxisome Loss
To understand how chronic HIF activation promotes the development of cardiomyopathy, we inactivated the VHL gene in the mouse heart. VHLfl/fl and VHL+/+ mice were crossed with mice expressing Cre recombinase in cardiomyocytes under the control of the myosin heavy chain promoter (αMHC-Cre). As expected, cardiac VHL loss increased the accumulation of HIF and proteins encoded by well-studied HIF-responsive mRNAs, such as BNIP3, in mouse hearts, but not in skeletal muscle, as determined by immunoblot analysis (Figure 1A; Figure S1A).
We previously showed that mice lacking VHL in the heart succumb to heart failure in ≈10 weeks and display dysregulated metabolism, mitochondrial abnormalities, and increased autophagy.1 Previous studies showed that HIF promotes mitochondrial and peroxisomal autophagy, at least in some settings, presumably to maintain O2 homeostasis under hypoxia.9 Mitochondria and peroxisomes play essential roles in redox, lipid, and energy metabolism.12 Whether the loss of these 2 organelles causes, rather than merely correlates with, heart failure in the setting of chronic ischemia has not been determined. Cardiac VHL loss decreased peroxisomal and mitochondrial protein abundance, peroxisomal number and size, and mitochondrial area in whole tissue lysates and isolated mouse cardiomyocytes compared with wild-type counterparts (Figure 1A; Figure S1B through S1F). Similarly, hypoxia and VHL loss decreased mitochondrial and peroxisomal function as measured by mitochondrial oxygen consumption through Seahorse analysis and oxidation of the mitochondria-specific substrate palmitic acid and the peroxisome-specific substrate lignoceric acid, respectively (Figure 1B and 1C; Figure S1G).
We asked whether VHL loss decreased cardiac mitochondria and peroxisome abundance through autophagy-dependent degradation. We confirmed cardiomyocytes isolated from VHLfl/fl; αMHC-Cre hearts or VHL knockout cardiomyocytes generated using CRISPR-Cas9 displayed increased autophagy as measured by increased microtubule-associated proteins 1A/1B light chain 3B (LC3B), WD repeat domain phosphoinositide-interacting protein 1 (WIPI-I), and Sequestosome-1 (SQSTM1/p62) puncta compared with wild-type counterparts by confocal microscopy (Figure S2A through S2C). Similarly, hypoxia promoted mitochondria and peroxisome colocalization with LAMP1 (Lysosome-associated membrane glycoprotein 1)-positive lysosomes, the final destination of autophagy cargo, in fixed cardiomyocytes by confocal microscopy (Figure S3A through S3B). In addition, hypoxia promoted biotin labeling of LAMP1, as measured by confocal microscopy using streptavidin-568 as a probe, in cardiomyocytes expressing PEX26 (Peroxisome assembly protein 26), a resident peroxisome membrane protein, fused to a biotin ligase (APEX [ascorbate peroxidase]) (Figure S4A and S4B). These results were autophagy-dependent because they were abrogated by loss of Beclin1, a core autophagy component, through CRISPR-Cas9 (Figure S3A and S3B; Figure S4B). To test whether autophagy inhibition could rescue mitochondria or peroxisome abundance in VHLfl/fl; αMHC-Cre hearts, we treated VHLfl/fl; αMHC-Cre mice with chloroquine, a pharmacologic inhibitor of lysosomal degradation. Chloroquine treatment of VHLfl/fl; αMHC-Cre mice increased mitochondrial and peroxisomal protein abundance in heart lysates (Figure 1D).
To facilitate the study of organelle trafficking during autophagy, we used Keima fusion proteins.13 Keima provides 2 important advantages. First, Keima itself is stable to lysosomal proteases. Accordingly, when a Keima fusion enters the lysosome, it liberates “processed” Keima (Keima and any peptidic remnants left behind after proteolysis of the fusion partner), which can be readily detected by immunoblot analysis. Second, Keima is also a pH-responsive reporter that undergoes a chromophore resting charge state change upon trafficking to the lysosome (pH ≈4.5). This enables autophagy flux measurements by quantifying the ratio of 561 nm/442 nm excitation by flow cytometry or confocal microscopy. We generated 2 different peroxisome-Keima reporters: one in which Keima was fused to the N terminus of amino acids 237–305 of human PEX26, which is sufficient for localization to and insertion into the peroxisomal membrane, and the other in which Keima was fused to the N terminus of full-length human PEX11A (Peroxisomal membrane protein 11A).14,15 In tandem, we generated a mitochondrially targeted Keima by fusing Keima to the N terminus of the well-characterized, outer mitochondrial membrane localization sequence of OMP25 (Keima-OMP25).16 We validated that the PEX26 and OMP25 sequences were sufficient to localize to PEX14 (Peroxisomal membrane protein PEX14)-mCherry peroxisomes or MitoTracker DeepRed–positive mitochondria, respectively, when fused to a fluorescent protein (Figure S5A and S5B).
We stably infected human AC16 cardiomyocytes to express either of the 2 Keima-peroxisome reporters (Keima-PEX26 or Keima-PEX11A) or the Keima-OMP25 reporter. Conditions that activate the HIF pathway such as hypoxia, VHL loss, and pharmacologic EglN inhibition with FG-4592 promoted Keima-PEX processing by immunoblotting (Figure 1E; Figure S5C through S5E). This processing was inhibited by SAR405, a VPS34 inhibitor that blocks the autophagy system, CRISPR-Cas9–mediated Beclin1 loss, and chloroquine (Figure 1E; Figure S5C through S5E).17 As expected, other well-studied autophagy inducers, such as mTOR inhibition with Torin1 or amino acid starvation, also promoted Keima-PEX processing in a phosphatidylinositol 3-kinase, catalytic subunit type 3 (VPS34)- and Beclin1- dependent manner, consistent with autophagy-mediated peroxisome degradation (Figure 1E; Figure S5C and S5D). Similarly, hypoxia, FG-4592, Torin1, and mitophagy activators such as carbonyl cyanide p-trifluoro-methoxyphenyl hydrazone (FCCP) and oligomycin promoted VPS34- and Beclin1-dependent Keima-OMP25 processing by immunoblotting, consistent with autophagy-mediated mitochondrial degradation under these conditions (Figure 1F; Figure S5F).
To ask whether the HIF pathway specifically regulates the autophagy-mediated degradation of mitochondria and peroxisomes in cardiomyocytes, we inactivated ARNT using CRISPR-Cas9 in our Keima-PEX or OMP25 reporter cardiomyocytes to block HIF-mediated transcription. ARNT loss completely abrogated Keima-PEX or -OMP25 processing under hypoxia or FG-4592, as measured by immunoblotting. In contrast, ARNT loss did not abrogate Keima-PEX or -OMP25 processing induced by Torin1 (Figure 1E and 1G). Therefore, HIF is selectively required for the autophagy-dependent degradation of mitochondria and peroxisomes under hypoxia, rather than being generally required for autophagy.
DEPP1 Is a Direct Transcriptional Target of HIF
Our results with ARNT knockout cardiomyocytes suggested that 1 or more HIF-responsive gene products are necessary to promote mitochondrial and peroxisomal autophagy under conditions that activate the HIF pathway. To identify the responsible factor, we performed RNA sequencing in wild-type and ARNT knockout human induced pluripotent stem (hIPS) cell–derived cardiomyocytes (hIPS-CMs) grown under normoxic or hypoxia conditions (Figure 2A). As expected, hypoxia significantly increased the mRNA abundance of established HIF target genes, such as CA9, EGLN3, BNIP3, and NDRG1, in an ARNT-dependent manner. Hypoxia did not alter (positively or negatively) mRNA abundance of known mitochondrial or peroxisome autophagy factors, such as NBR1, PEX3, PHB2, or FUNDC1 (Figure 2A).18–21
To narrow our search for candidate factor(s), we hypothesized that the mediator(s) of HIF-regulated mitochondrial and peroxisomal autophagy would have a mitochondrial or peroxisomal targeting sequence. Similarly, because several proteins have been identified that regulate both mitochondrial and peroxisomal compartments, we reasoned HIF would regulate autophagy of both organelles through a shared mechanism. Among the hypoxia-inducible, ARNT-dependent genes was DEPP1 (Decidual protein induced by progesterone 1). DEPP1 was of interest because it contains a conserved PTS2 (peroxisomal targeting sequence type 2) nested within an N-terminal t-snare domain (Figure 2B). Moreover, DEPP1 has been linked with autophagy before through the nutrient sensitive FOXO (Forkhead box O) 3 transcription factor.22–24 We confirmed that DEPP1 mRNA and protein abundance were induced by hypoxia and FG-4592 in an ARNT-dependent manner by real-time polymerase chain reaction and immunoblot assays in hIPS-CMs (Figure 2C) and U2OS cells (Figure 2D). DEPP1 contains a conserved hypoxia response element (HRE) upstream of the DEPP1 transcriptional start site (Figure 2E).25 We confirmed that HIF1α bound the DEPP1 HRE during hypoxia in an ARNT-dependent manner using chromatin immunoprecipitation assays in human AC16 cardiomyocytes (Figure 2F). CRISPR-Cas9 DEPP1 HRE editing, like CRISPR-Cas9 targeting of the DEPP1 open reading frame, abrogated hypoxia-induced DEPP1 protein abundance in hIPS-CMs (Figure 2G). DEPP1 loss also damped the induction of HIF1α and the product of HIF-responsive mRNAs, such as NDRG1, under hypoxia, suggesting that the HIF and DEPP1 participate in a positive feedback loop.
DEPP1 Is Localized Inside Mitochondria
Previous overexpression studies showed that DEPP1 localizes to peroxisomes.24 To study this further, we made fluorescent versions of DEPP1 and a DEPP1 variant that lacks the N-terminal t-snare domain (ΔTsnare) by fusing them to either mCherry or mNeonGreen. These proteins were then introduced into DEPP1 knockout cardiomyocytes by lentiviral infection followed by fluorescence cell sorting for cells in which the levels of the exogenous proteins approximated endogenous DEPP1 levels. We found, unexpectedly, that wild-type DEPP1 localized to mitochondria rather than peroxisomes in cardiomyocytes by confocal microscopy irrespective of the fluorophore (Figure 3A through 3C). Mitochondrial localization was confirmed using TOM70 (Mitochondrial import receptor subunit TOM70)-GFP (green fluorescent protein) and MitoTracker, 2 markers of mitochondria. In contrast, the ΔTsnare variant gave rise to diffuse cytosolic staining.
DEPP1-mCherry did not overlap with peroxisomes marked by GFP fused to PTS1 (GFP-PTS1) (Figure S6A). DEPP1-mCherry–positive mitochondria did, however, associate with GFP-PTS1, consistent with direct mitochondrial-peroxisome contacts between these organelles (Figure S6A).
We next tested whether DEPP1 is present inside or outside of mitochondria. Confocal microscopy with Airyscan revealed that DEPP1 resides in a diffuse pattern that completely overlaps with MitoTracker encased within the TOM70 outer mitochondrial membrane (Figure 3B and 3C; Figure S6B). Stimulated emission depletion super-resolution microscopy imaging, which provides an optical resolution superior to diffraction-limited confocal microscopy, revealed a similar diffuse DEPP1 pattern, whereas outer mitochondrial membrane protein TOM20 (Mitochondrial import receptor subunit TOM20) was distributed in a punctated pattern throughout the outer mitochondrial membrane (Figure 3D; Figure S6C). To ask whether DEPP1 resides within the mitochondrial inner membrane space or within the mitochondrial matrix, we fused DEPP1 or DEPP1 ΔTsnare to a biotin ligase (APEX) and measured biotinylation by confocal microscopy using streptavidin-568 as a probe. A mitochondrial matrix–localized APEX-generated biotin-phenoxyl radical will generate a restricted biotin-labeling pattern because the matrix is surrounded by the impermeable inner mitochondrial membrane. Conversely, an inner membrane space–localized APEX-generated biotin-phenoxyl radical will generate a diffuse biotinylation pattern because the outer mitochondrial matrix is porous to small molecules, including biotin-phenoxyl radicals.26 We introduced DEPP1-APEX or DEPP1ΔTsnare-APEX into DEPP1 knockout U2OS cells by lentiviral infection. We performed single-cell sorting to identify clones in which the exogenous DEPP1 levels approximated endogenous levels. DEPP1-APEX, in a t-snare–dependent manner, caused a restricted biotin-labeling pattern that completely overlapped with citrate synthase, a mitochondrial matrix enzyme (Figure 3E; Figure S6D). PMP70 (peroxisomal membrane protein 70)–positive peroxisomes associated with DEPP1-mediated streptavidin-568 consistent with direct mitochondrial-peroxisome contacts (Figure S6E). Therefore, DEPP1 localizes inside mitochondria, likely within the matrix compartment.
DEPP1 Is Necessary for HIF-Mediated Autophagy and Triglyceride Accumulation
To test the role of DEPP1 in mitochondrial and peroxisomal autophagy in the setting of chronic HIF activation, we generated whole-body Depp1-/- mice using CRISPR-Cas9–based gene editing of mouse embryos and crossed them to VHL+/+ or VHLfl/fl; αMHC-Cre mice (Figure S7A). The Depp1-/- mice were viable, did not display gross phenotypes, and were normotensive, consistent with previous reports of similar mice (Figure S7B).27,28 DEPP1 loss did not affect peroxisomal protein abundance or mitochondrial membrane potential by immunoblot and microscopy analysis (Figure S7C through S7E). We stably infected cardiomyocytes derived from the hearts of these mice with EGFP (enhanced GFP)–LC3, a reporter for autophagy induction, and Mito-RFP (red fluorescent protein), to visualize mitochondria. As expected, cardiomyocyte VHL loss promoted EGFP-LC3 puncta, indicative of autophagy induction, which colocalized with Mito-RFP, consistent with mitochondrial autophagy (Figure 4A; Figure S8A). Depp1 loss in the setting of VHLfl/fl; αMHC-Cre abrogated EGFP-LC3 puncta, suggesting Depp1 is necessary for autophagy induction upon VHL loss (Figure 4A; Figure S8A). Consistent with these observations, DEPP1 loss inhibited LC3B lipidation and p62 degradation mediated by either VHL loss or hypoxia in cells and heart tissue lysates in immunoblot assays (Figure S8B through S8D). DEPP1 loss decreased mitochondrial swelling and increased mitochondrial oxygen consumption in the setting of hypoxia or VHL loss as measured by electron microscopy and Seahorse analysis (Figure S9A and S9B). Similarly, DEPP1 loss reduced accumulation of triglycerides of chain lengths C50 to C60 in the setting of hypoxia and increased peroxisomal number in cardiomyocytes lacking VHL as measured by lipidomic analysis and confocal microscopy (Figure 4B; Figure S10A and S10B). DEPP1 loss did not affect, positively or negatively, HIF stabilization in the setting of VHL loss or after FG-4592 treatment in isolated cells or in vivo (Figure 4C; Figure S8B and S8D).
To directly examine the role of DEPP1 in HIF-mediated peroxisome and mitochondrial autophagy, we stably expressed our Keima-PEX or OMP25 reporters in wild-type and Depp1-/- cardiomyocytes. As expected, HIF activation through hypoxia or FG-4592, like mTOR inhibition by Torin1 or amino acid starvation, promoted Keima-PEX and Keima-OMP25 processing in wild-type cardiomyocytes as measured by immunoblot analysis (Figure 4C and 4D; Figure S11A through S11C). Depp1 loss abrogated the Keima-PEX and Keima-OMP25 processing caused by HIF activation but did not affect the Keima processing caused by mTOR inhibition (Figure 4C and 4D; Figure S11A). Similarly, hypoxia and FG-4592 promoted Depp1-dependent mitochondrial (Mito-RFP or ATP5H) and peroxisomal (mRFP-PEX26 or PEX14) colocalization with LAMP1-positive lysosomes and LC3B-positive autophagosomes in cardiomyocytes by confocal microscopy (Figure S12A and S12B; Figure S13A and S13B; Figure S14A; Figure S15A). In addition, hypoxia promoted Depp1-dependent biotin-labeling of LAMP1 by APEX-PEX26, as measured by confocal microscopy using streptavidin-568 as a probe (Figure S15B).
We postulated the direct regulation of DEPP1 by HIF was necessary to promote autophagy under hypoxia. Indeed, DEPP1 HRE gene editing by CRISPR-Cas9 inhibited Keima-PEX processing, Keima-OMP25 processing, and p62 degradation under hypoxia (Figure 4E and 4F; Figure S16A). Similarly, DEPP1 HRE gene editing inhibited autophagosome formation and their association with mitochondria under hypoxia but not Torin1 treatment, as measured by GFP-WIPI-I colocalization with Mito-RFP by live cell imaging (Figure S16B and S16C). Therefore, DEPP1, and its regulation by the HIF transcription factor, is necessary for HIF-mediated mitochondrial and peroxisome autophagy and triglyceride accumulation.
DEPP1 Is Sufficient to Induce Autophagy, Triglyceride Accumulation, and Mitochondrial Permeability Transition Pore Activation
Hypoxia induces HIF, and HIF activation in mice is sufficient to promote many features of ischemic cardiomyopathy, including autophagy induction and lipid accumulation.1–3,29 To ask whether DEPP1 is sufficient to induce autophagy, we stably expressed DEPP1, DEPP1 ΔTsnare, or empty vector in cardiomyocytes (Figure 5A). Consistent with VHL loss, DEPP1, but not DEPP1 ΔTsnare, increased LC3B lipidation, as measured by fluorescent microscopy (Figure 5B; Figure S17A). These changes were not a result of exogenous DEPP1 indirectly inducing HIF (Figure 5A). We did reproducibly note that VHL loss caused more LC3B-positive puncta than exogenous DEPP1 in cardiomyocytes, although the endogenous DEPP1 levels achieved with VHL loss were similar to the exogenous levels. Among several possibilities, this could reflect a contribution of additional HIF target genes to autophagy after VHL loss.
To ask whether DEPP1 is sufficient to promote autophagy in vivo, we used CAG-RFP-EGFP-LC3 transgenic mice in which a CAG promotor sequence drives the expression of RFP and an EGFP fused to LC3.30 These mice have been widely used to monitor autophagy in tissues as determined by the formation of RFP- and EGFP-positive puncta.30 We generated adeno-associated viruses serotype 9 (AAV9) expressing DEPP1 or DEPP1 ΔTsnare and then injected these viruses, or the empty vector, into the tail veins of CAG-RFP-EGFP-LC3 mice and removed their hearts 7 days later. CAG-RFP-EGFP-LC3 mice treated with FG-4592 served as controls. As seen with HIF activation by FG-4592, DEPP1, but not DEPP1 ΔTsnare, increased RFP and EGFP puncta in mouse hearts (Figure 5C). Ultrastructural analysis by electron microscopy of mouse hearts derived from mice treated with FG-4592 or expressing AAV9-DEPP1, but not DEPP1 ΔTsnare, revealed increased autophagosome formation and instances of mitochondrial engulfment by autophagosomes, suggestive of mitophagy (Figure 5D).
HIF promotes glucose and fatty acid uptake, aerobic glycolysis, and glucose to lipid conversion, while repressing oxidative phosphorylation and β-oxidation. Because HIF is necessary and sufficient for the accumulation of lipids in hearts lacking pVHL and chronic hypoxia increases triglyceride abundance,8,31–33 we tested whether DEPP1 is sufficient to promote cardiomyocyte triglyceride accumulation. Indeed, DEPP1, but not DEPP1 ΔTsnare, increased triacylglyceride abundance of chain lengths C50 to C60 in cardiomyocytes by lipidomic profiling (Figure 5E).
Chronic hypoxia is sufficient to induce mitochondrial permeability transition pore (MPTP) opening leading to cell death.34–36 To ask whether DEPP1 is sufficient to induce MPTP opening, we stably expressed DEPP1, DEPP1 ΔTsnare, in cardiomyocytes and treated these cells with and without ionomycin to trigger MPTP activation (Figure 5F). Like VHL loss, DEPP1, but not DEPP1 ΔTsnare, increased MPTP opening at baseline, as measured by confocal microscopy and image analysis (Figure 5F). MPTP opening was sensitive to cyclosporin A treatment, a classic MPTP inhibitor (Figure 5F).36 MPTP opening promotes apoptosis and cell death.37 We asked whether DEPP1 is sufficient to promote apoptosis. Although DEPP1 expression did not activate apoptosis at baseline, as measured by caspase-3 and PARP (Poly [ADP-ribose] polymerase 1) cleavage in immunoblot assays, DEPP1, but not DEPP1 ΔTsnare, increased the PARP and caspase-3 cleavage in response to staurosporine (Figure S17B).
DEPP1 Loss Inhibits Hypoxia-Mediated ROS Production and HIF Stabilization
We frequently detected decreased HIF stabilization under hypoxia after DEPP1 loss or disruption of the DEPP1 HRE (Figure 2G; Figure 4C through 4F; Figure S11B; Figure S16A). DEPP1 loss inhibited HIF stabilization and LC3B lipidation in U2OS cells grown under hypoxia for 24 to 36 hours (Figure 6A). The EglN-pVHL axis appeared to be functionally intact because cells lacking DEPP1 or DEPP1 HRE stabilized HIF when treated with FG-4592 or the prolyl hydroxylase inhibitor dimethyloxalylglycine, or after VHL inactivation (Figure 6B and 6C). Inhibition of hypoxia-mediated HIF stabilization in DEPP1 knockout cells correlated with decreased abundance of the HIF-responsive gene product NDRG1 (Figure 6B and 6C). Mitochondrial ROS is essential for proper O2 sensing and subsequent HIF stabilization.38–40 We hypothesized that DEPP1 loss reduces hypoxia-induced ROS production, thereby suppressing HIF. Indeed, DEPP1 loss suppressed hypoxia-induced ROS production and oxidative stress, as measured by CellRox Green and protein carbonylation (Figure 6D and 6E). Therefore, DEPP1 is necessary for hypoxia-induced ROS and modulates HIF stabilization.
DEPP1 Loss Increases Cardiomyocyte Survival After Chronic HIFα Activation and Decreases Cardiac Dysfunction Upon VHL Loss
DEPP1 loss inhibits HIF-mediated autophagy of mitochondria and peroxisomes and triglyceride accumulation. To ask whether DEPP1 loss would be beneficial or deleterious in the setting of chronic HIF activation, we used CRISPR-Cas9 to generate hIPS cells lacking DEPP1 and then infected them or wild-type counterparts with a lentivirus encoding a doxycycline inducible non-hydroxylatable HIF2α variant (HIF2α-dPA). These cells were then induced to become cardiomyocytes (hiPS-CMs) and treated with doxycycline (Figure S18A).41 We previously showed that HIF2α-dPA is sufficient to cause cardiomyopathy in vivo.1 HIF2α-dPA increased hIPS-CM DEPP1 abundance and decreased hIPS-CM survival (Figure S18A and S18B). DEPP1 loss increased hIPS-CM survival in the setting of HIF2α-dPA (Figure S18B). Consistent with this, whole-body Depp1 loss decreased cardiac dysfunction in mice lacking pVHL in the heart as measured by myocardial thickness, fractional shortening, and cardiomegaly (Figure 7A through 7D). Depp1 loss increased peroxisomal and mitochondrial fatty acid oxidation in isolated cardiomyocytes from hearts lacking pVHL (Figure 7E through 7F). Similarly, whole-body Depp1 loss reduced cardiac fibrosis and apoptosis in hearts lacking pVHL (Figure 7G and 7H). These changes were associated with increased survival (median survival of ≈10 weeks for mice with hearts lacking pVHL and ≈22 weeks for mice with hearts lacking Depp1 and pVHL; Figure 7I).
HIF1α loss has been shown to completely rescue the cardiomyopathy and premature mortality in mice lacking cardiac pVHL.2 In contrast, Depp1 loss extended, but did not fully rescue, the survival defect in mice lacking pVHL in the heart. Among multiple possibilities, this would suggest the contribution of additional HIF target genes to cardiomyopathy in the context of chronic HIF activation. Indeed, pVHL loss has been shown to result in cardiac inflammation, apoptotic cell death, and pathological hypertrophy through NF-κB (nuclear factor-κB) activation, and NF-κB inhibition is protective in the setting of heart failure.42,43 Whole-body Depp1 loss did not reduce NF-κB activation in hearts lacking pVHL, as measured by NF-κB p62 S536 and IKKαβS176/S177 phosphorylation in immunoblot assays (Figure S19A). Similarly, pVHL-defective hearts had increased proinflammatory cytokine abundance, such as IL (interleukin)–6 and IL1β, that was unaffected by whole-body Depp1 loss (Figure S19A). These results suggest that although Depp1 loss decreases cardiac dysfunction in the setting of chronic HIF activation, pVHL loss still compromises cardiac function and longevity, at least in part, because of the Depp1-independent chronic activation of inflammation through NF-kB.
Because HIF activation is a predictable consequence of impaired oxygen delivery, we asked whether DEPP1 levels are increased in human ischemic cardiomyopathy. In the limited number of autopsy samples available to us from patients with chronic ischemic cardiomyopathy (n=3), DEPP1 and HIF abundance was increased, and this correlated with deregulated mitochondrial and peroxisomal protein abundance and increased LC3B lipidation by immunoblot analysis and decreased mitochondrial SDHA (succinate dehydrogenase) and PEX11B (peroxisomal biogenesis factor 11B) immunohistochemical reactivity (Figure S20A through S20C). Collectively, these results suggest that DEPP1 induction plays a key role in the pathogenesis of chronic ischemic cardiomyopathy and that its loss would decrease cardiac dysfunction in the setting of chronic HIF activation.
DISCUSSION
Chronic HIF activation is a predictable consequence of chronic ischemia. HIF regulates many aspects of cardiac function, and the induction of cardiomyopathy by HIF is likely multifactorial. Therefore, understanding the contribution of HIF-regulated processes to heart failure and whether manipulation of HIF, or its downstream targets, could alter the natural course of this disease remains a key question. Here, we identify a mechanism by which hypoxia activates autophagy through the HIF pathway. We show in both cardiomyocytes as well as in noncardiac-derived cells, such as U2OS cells, that DEPP1 is under direct control of the HIF transcription factor, localizes inside mitochondria, and is necessary and sufficient for HIF-mediated autophagy. DEPP1 loss is protective in the setting of chronic HIF activation, and whole-body Depp1 loss in mice decreases cardiac dysfunction and maladaptive metabolic remodeling after pVHL loss.
Patients with heart failure display systemic perturbations in energy-related metabolites, especially those reflecting mitochondrial and peroxisomal fatty acid metabolism, and derangement in cardiac substrate use is thought to contribute to contractile dysfunction and disease progression.44,45 These changes may reflect altered organelle biogenesis, increased autophagy, direct effects on fatty acid oxidation within mitochondria or peroxisomes, or a combination of all three. Mitochondrial autophagy plays a clear role in cardiac homeostasis, whereas the role of the peroxisome and its quality control remain understudied. Our studies identify DEPP1 as necessary and sufficient for HIF-mediated autophagy in cardiomyocytes. DEPP1 loss increases mitochondrial and peroxisomal fatty acid oxidation and decreases cardiac dysfunction in the setting of chronic HIF activation. Similarly, DEPP1 loss decreases triglyceride accumulation under hypoxia. Moreover, DEPP1 abundance is increased in ischemic cardiomyopathy patient biopsies, and this correlates with dysregulated mitochondrial and peroxisomal protein abundance.
Our work suggests increasing mitochondrial and peroxisomal abundance may provide a therapeutic strategy for ischemic cardiomyopathy. In this regard, activation of PPARα (peroxisome proliferator-activated receptor α), a key transcriptional activator of mitochondrial and peroxisomal fatty acid metabolism, improves cardiac function after pressure overload, ischemia, and pacing-induced heart failure in mice.46–50 Although PPARα agonists have not been directly tested in human heart failure trials, they were shown to reduce the risk of major cardiovascular events in the setting of type 2 diabetes, and this effect correlated with the degree to which they lowered serum lipids.51,52 Although cardioprotection mediated by PPARα activation presumably reflects, at least in part, reduced cardiotoxic lipid accumulation through increased extracardiac tissue lipid use, our results suggest that it could also confer cardiomyocyte-intrinsic protection in the setting of chronic ischemia by preserving mitochondrial and peroxisome abundance. Furthermore, it would be of interest to test whether PPARα agonists reduce cardiac dysfunction in the setting of chronic HIFα activation. In line with this possibility, peroxisomes are selectively essential under hypoxia, and peroxisomal fatty acid metabolism activation improves cardiac recovery after ischemia in mouse models.53–55
We find DEPP1 is necessary and sufficient for HIF-mediated autophagy, and Depp1 inhibition decreases cardiac dysfunction in the setting of chronic HIF activation. Whether increased autophagy is an adaptive or maladaptive response to chronic myocardial ischemia remains a debate.56,57 In this regard, cardiomyocyte death in hearts from patients with end-stage heart failure is mediated most prominently through autophagy.58 Consistent with this, autophagy inhibition through heterozygous disruption of Beclin 1 decreases cardiomyocyte autophagy and pathological cardiac remodeling after severe pressure overload in mice. Conversely, Beclin 1 overexpression increases autophagy and pathological cardiac remodeling.59 Similarly, activation of Tfeb (transcription factor EB), a master regulator of lysosomal biogenesis and autophagy, causes heart failure in mice in the setting of pressure overload.60 It will be important to confirm experimentally whether increasing autophagy of mitochondrial and peroxisomes is sufficient to induce cardiac dysfunction. Further studies are necessary to understand whether autophagy inhibition is beneficial in the setting of chronic ischemia and heart failure.
Our studies do not preclude a protective role of HIF activation in the setting of acute myocardial ischemia. Indeed, HIF activates many genes that would be predicted to promote survival under conditions of acute hypoxia. PHD inhibition with pharmacologic inhibitors or genetic strategies, and transgenic expression of HIF1α in the heart, have been shown to be beneficial in animal models of acute myocardial infarction.61–66 Similarly, autophagy induction in the setting of acute ischemia reperfusion injury is believed to be beneficial rather than maladaptive. Autophagosome number increases in the heart during both the ischemia and reperfusion phases.67 Increased autophagy correlates with recovery and salvage of the myocardium in animal models of ischemia reperfusion injury and cardiomyocyte-specific ATG7 (Ubiquitin-like modifier-activating enzyme ATG7) loss, which enfeebles autophagy and aggravates ischemia reperfusion injury, leading to increased cardiac hypertrophy, dysfunction, and fibrosis.68 In addition, complete abrogation of autophagy is sufficient to trigger rapid-onset heart failure in mice, supporting a key role of basal levels of cardiomyocyte autophagy in cardiac homeostasis.69
DEPP1 appears to be a critical component of the metabolic control of autophagy in multiple contexts. We found that DEPP1 is directly regulated by the HIF transcription factor, thereby connecting DEPP1 expression to cellular oxygen levels and autophagy. In addition, DEPP1 mRNA is induced after insulin or nutrient deprivation, fasting, or H2O2 through the FOXO1/3 transcription factors and is necessary for FOXO3-mediated autophagy.24,70–72 FOXO activation is necessary and sufficient to induce fasting-mediated skeletal and cardiac muscle autophagy and atrophy.73–76 Because chronic exposure to high altitude leads to hypoxia-induced muscle wasting, it will be important to learn whether DEPP1 acts as a muscle atrophy–related factor. Similarly, autophagy-mediated mitochondrial clearance controls beige adipocyte maintenance, and autophagy inhibition prevents beige adipocyte loss and protects against diet-induced insulin resistance in mice.77 Because Depp1 deficiency induces white adipocyte browning (“beiging”) and protects against diet-induced insulin resistance in mice, it would be of interest to understand DEPP1 and its role in adipocyte autophagy and adaptive thermogenesis.28
DEPP1 function remains to be determined. Mechanistically, DEPP1 is not a core component of the autophagy machinery because autophagy induction by mTOR inhibition is unaffected by DEPP1 loss (Figure 4C and 4D). Instead, DEPP1 localizes inside mitochondria (Figure 3A through 3E). A recent study showed DEPP1 inhibition suppressed MPTP opening in the setting of mitochondrial Ca2+ overload, a key step in cardiomyocyte death in the setting of ischemia, suggesting DEPP1 may play a role in mitochondrial Ca2+ homeostasis.78,79 In line with this, we find DEPP1 is sufficient to induce MPTP opening (Figure 5F). We hypothesize that DEPP1 regulates mitochondrial function to coordinate autophagy induction under conditions of low oxygen. Because DEPP1 is necessary and sufficient for hypoxia-mediated reactive oxygen species production, and mitochondrial ROS plays a clear role in hypoxia sensing and HIF pathway activation, precise understanding of how DEPP1 controls mitochondrial function and ROS production, how this is coordinated with associated peroxisomes, and how these signals regulate the autophagy machinery will need to be determined.
Study Limitations
Our work demonstrates that DEPP1 loss reduces cardiac dysfunction in the context of chronic HIF activation modeled through loss of pVHL in the heart. Although chronic HIF activation is a predictable consequence of chronic ischemia, the effects of pVHL loss and of chronic ischemia on the heart are unlikely to be entirely congruent. It will therefore be of interest to determine whether DEPP1 loss is cardioprotective in other models of acute or chronic ischemia as well in the setting of other cardiomyopathies. Our work was restricted to isolated cardiomyocytes, immortalized U2OS osteosarcoma cells, and whole-body Depp1 knockout mice. Because communication between cardiac resident cell types is known to provide cardioprotection, it will be of interest in future experiments to use mice in which Depp1 is specifically deleted in cardiomyocytes or other cardiac resident cell populations, such as endothelial cells, epicardial cells, cardiac fibroblasts, and pericytes. Our mouse studies were restricted to 1 genetic background (C57/BL6) and 1 animal model (mouse). Our studies used male mice only because of their increased sensitivity to heart failure.80 It will be important to explore other strains and to test female mice. Because the mouse is an imperfect model of cardiovascular physiology, it will also be important to explore our findings in larger animal models.
ARTICLE INFORMATION
Acknowledgments
We thank the members of the Kaelin laboratory, J. Moslehi (University of California San Francisco), and R. Malhotra (Massachusetts General Hospital) for helpful discussions and critical reading of the article. Special thanks to the Cardiovascular Physiology Core at Brigham and Women's Hospital/Harvard Medical School (HMS) for assistance with cardiac physiology experiments, S. Drakos (University of Utah) for access to human heart samples and critical reading of the article, the Transgenic Mouse Core (HMS) for assistance with generation of CRISPR editing of mouse embryos, the HMS Electron Microscopy Facility for consultation and sample preparation, and the Beth Israel Deaconess Medical Center Mass Spectrometry Core facility for assistance with lipidomic profiling and analysis. This article is subject to HHMI’s Open Access to Publications policy. HHMI laboratory heads have previously granted a nonexclusive CC BY 4.0 license to the public and a sublicensable license to HHMI in their research articles. Pursuant to those licenses, the author-accepted article of this article can be made freely available under a CC BY 4.0 license immediately upon publication. G.A.W. performed experiments and, together with W.G.K., designed experiments, analyzed data, and assembled and wrote the article. Q.J. assisted in RNA sequencing sample preparation, sequence analysis, and experimental design. M.S. performed immunohistochemistry and assisted in confocal imaging. S.Q. performed quantitative polymerase chain reaction and apoptosis analysis. C.L. assisted in experimental design. B.A.M. assisted in human biopsy collection and sample preparation.
Sources of Funding
W.G.K. is supported by the National Institutes of Health (grant No. 5R35CA210068) and is an HHMI investigator. G.A.W. is supported by the National Institutes of Health (grant No. 4R00HL163396; National Heart, Lung, and Blood Institute) and a Hassenfeld Award (MGH).
Disclosures
W.G.K. has financial interests in Lilly Pharmaceuticals, Fibrogen, Nextech Invest, Tango Therapeutics, LifeMine Therapeutics, Circle Pharma, IconOVir Bio, and Casdin Capital. The other authors report no conflicts.
Supplemental Material
Expanded Methods
Figures S1–S20
Supplementary Material
Nonstandard Abbreviations and Acronyms
- αMHC
- α myosin heavy chain
- ΔTsnare
- N-terminal t-snare domain
- AAV9
- adeno-associated viruses serotype 9
- APEX
- ascorbate peroxidase
- ARNT
- aryl hydrocarbon nuclear translocator
- BNIP3
- Bcl-2/adenovirus E1B 19-kDa interacting protein 3
- CM
- cardiomyocyte
- DEPP1
- decidual protein induced by progesterone 1
- EGFP
- enhanced green fluorescent protein
- EGLN
- Egl 9 homolog
- FOXO
- Forkhead box O
- GFP
- green fluorescent protein
- HIF
- hypoxia inducible factor
- hIPS
- human induced pluripotent stem
- HRE
- hypoxia response element
- IL
- interleukin
- LC3
- microtubule-associated protein 1 light chain 3 α
- MPTP
- mitochondrial permeability transition pore
- mTOR
- mammalian target of rapamycin
- NF-κB
- nuclear factor-κB
- PEX11B
- peroxisomal biogenesis factor 11B
- PHD
- prolyl hydroxylase domain
- PMP70
- peroxisomal membrane protein 70
- PPARα
- peroxisome proliferator-activated receptor α
- PTS
- peroxisomal targeting sequence
- pVHL
- von Hippel-Lindau protein
- RFP
- red fluorescent protein
- ROS
- reactive oxygen species
- SDHA
- succinate dehydrogenase
- Tfeb
- transcription factor EB
Supplemental Material, the podcast, and transcript are available with this article at https://www.ahajournals.org/doi/suppl/10.1161/CIRCULATIONAHA.123.066628.
For Sources of Funding and Disclosures, see page 784.
Circulation is available at www.ahajournals.org/journal/circ.
Contributor Information
Madhu Singh, Email: msingh21@mgh.harvard.edu.
Shariq Qayyum, Email: sqayyum1@mgh.harvard.edu.
REFERENCES
- 1.Moslehi J, Minamishima YA, Shi J, Neuberg D, Charytan DM, Padera RF, Signoretti S, Liao R, Kaelin WG, Jr. Loss of hypoxia-inducible factor prolyl hydroxylase activity in cardiomyocytes phenocopies ischemic cardiomyopathy. Circulation. 2010;122:1004–1016. doi: 10.1161/CIRCULATIONAHA.109.922427 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Lei L, Mason S, Liu D, Huang Y, Marks C, Hickey R, Jovin IS, Pypaert M, Johnson RS, Giordano FJ. Hypoxia-inducible factor-dependent degeneration, failure, and malignant transformation of the heart in the absence of the von Hippel-Lindau protein. Mol Cell Biol. 2008;28:3790–3803. doi: 10.1128/MCB.01580-07 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Bekeredjian R, Walton CB, MacCannell KA, Ecker J, Kruse F, Outten JT, Sutcliffe D, Gerard RD, Bruick RK, Shohet RV. Conditional HIF-1alpha expression produces a reversible cardiomyopathy. PLoS One. 2010;5:e11693. doi: 10.1371/journal.pone.0011693 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Kaelin WG, Jr, Ratcliffe PJ. Oxygen sensing by metazoans: the central role of the HIF hydroxylase pathway. Mol Cell. 2008;30:393–402. doi: 10.1016/j.molcel.2008.04.009 [DOI] [PubMed] [Google Scholar]
- 5.Minamishima YA, Moslehi J, Bardeesy N, Cullen D, Bronson RT, Kaelin WG, Jr. Somatic inactivation of the PHD2 prolyl hydroxylase causes polycythemia and congestive heart failure. Blood. 2008;111:3236–3244. doi: 10.1182/blood-2007-10-117812 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Minamishima YA, Moslehi J, Padera RF, Bronson RT, Liao R, Kaelin WG, Jr. A feedback loop involving the Phd3 prolyl hydroxylase tunes the mammalian hypoxic response in vivo. Mol Cell Biol. 2009;29:5729–5741. doi: 10.1128/MCB.00331-09 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Zhang H, Gao P, Fukuda R, Kumar G, Krishnamachary B, Zeller KI, Dang CV, Semenza GL. 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. doi: 10.1016/j.ccr.2007.04.001 [DOI] [PubMed] [Google Scholar]
- 8.Ackerman D, Tumanov S, Qiu B, Michalopoulou E, Spata M, Azzam A, Xie H, Simon MC, Kamphorst JJ. Triglycerides promote lipid homeostasis during hypoxic stress by balancing fatty acid saturation. Cell Rep. 2018;24:2596–2605.e5. doi: 10.1016/j.celrep.2018.08.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Walter KM, Schonenberger MJ, Trotzmuller M, Horn M, Elsasser HP, Moser AB, Lucas MS, Schwarz T, Gerber PA, Faust PL, et al. Hif-2alpha promotes degradation of mammalian peroxisomes by selective autophagy. Cell Metab. 2014;20:882–897. doi: 10.1016/j.cmet.2014.09.017 [DOI] [PubMed] [Google Scholar]
- 10.Bellot G, Garcia-Medina R, Gounon P, Chiche J, Roux D, Pouyssegur J, Mazure NM. Hypoxia-induced autophagy is mediated through hypoxia-inducible factor induction of BNIP3 and BNIP3L via their BH3 domains. Mol Cell Biol. 2009;29:2570–2581. doi: 10.1128/MCB.00166-09 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Diwan A, Krenz M, Syed FM, Wansapura J, Ren X, Koesters AG, Li H, Kirshenbaum LA, Hahn HS, Robbins J, et al. Inhibition of ischemic cardiomyocyte apoptosis through targeted ablation of Bnip3 restrains postinfarction remodeling in mice. J Clin Invest. 2007;117:2825–2833. doi: 10.1172/JCI32490 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Fransen M, Lismont C, Walton P. The peroxisome-mitochondria connection: how and why? Int J Mol Sci. 2017;18:1126. doi: 10.3390/ijms18061126 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Engedal N, Sonstevold T, Beese CJ, Selladurai S, Melcher T, Simensen JE, Frankel LB, Urbanucci A, Torgersen ML. Measuring autophagic cargo flux with Keima-based probes. Methods Mol Biol. 2022;2445:99–115. doi: 10.1007/978-1-0716-2071-7_7 [DOI] [PubMed] [Google Scholar]
- 14.Hosoi KI, Miyata N, Mukai S, Furuki S, Okumoto K, Cheng EH, Fujiki Y. The VDAC2-BAK axis regulates peroxisomal membrane permeability. J Cell Biol. 2017;216:709–722. doi: 10.1083/jcb.201605002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Ray GJ, Boydston EA, Shortt E, Wyant GA, Lourido S, Chen WW, Sabatini DM. A PEROXO-tag enables rapid isolation of peroxisomes from human cells. iScience. 2020;23:101109. doi: 10.1016/j.isci.2020.101109 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Nemoto Y, De Camilli P. Recruitment of an alternatively spliced form of synaptojanin 2 to mitochondria by the interaction with the PDZ domain of a mitochondrial outer membrane protein. EMBO J. 1999;18:2991–3006. doi: 10.1093/emboj/18.11.2991 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Ronan B, Flamand O, Vescovi L, Dureuil C, Durand L, Fassy F, Bachelot MF, Lamberton A, Mathieu M, Bertrand T, et al. A highly potent and selective Vps34 inhibitor alters vesicle trafficking and autophagy. Nat Chem Biol. 2014;10:1013–1019. doi: 10.1038/nchembio.1681 [DOI] [PubMed] [Google Scholar]
- 18.Deosaran E, Larsen KB, Hua R, Sargent G, Wang Y, Kim S, Lamark T, Jauregui M, Law K, Lippincott-Schwartz J, et al. NBR1 acts as an autophagy receptor for peroxisomes. J Cell Sci. 2013;126:939–952. doi: 10.1242/jcs.114819 [DOI] [PubMed] [Google Scholar]
- 19.Yamashita S, Abe K, Tatemichi Y, Fujiki Y. The membrane peroxin PEX3 induces peroxisome-ubiquitination-linked pexophagy. Autophagy. 2014;10:1549–1564. doi: 10.4161/auto.29329 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Wei Y, Chiang WC, Sumpter R, Jr, Mishra P, Levine B. Prohibitin 2 is an inner mitochondrial membrane mitophagy receptor. Cell. 2017;168:224–238.e10. doi: 10.1016/j.cell.2016.11.042 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Liu L, Feng D, Chen G, Chen M, Zheng Q, Song P, Ma Q, Zhu C, Wang R, Qi W, et al. Mitochondrial outer-membrane protein FUNDC1 mediates hypoxia-induced mitophagy in mammalian cells. Nat Cell Biol. 2012;14:177–185. doi: 10.1038/ncb2422 [DOI] [PubMed] [Google Scholar]
- 22.Salcher S, Hermann M, Kiechl-Kohlendorfer U, Ausserlechner MJ, Obexer P. C10ORF10/DEPP-mediated ROS accumulation is a critical modulator of FOXO3-induced autophagy. Mol Cancer. 2017;16:95. doi: 10.1186/s12943-017-0661-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Stepp MW, Folz RJ, Yu J, Zelko IN. The c10orf10 gene product is a new link between oxidative stress and autophagy. Biochim Biophys Acta. 2014;1843:1076–1088. doi: 10.1016/j.bbamcr.2014.02.003 [DOI] [PubMed] [Google Scholar]
- 24.Salcher S, Hagenbuchner J, Geiger K, Seiter MA, Rainer J, Kofler R, Hermann M, Kiechl-Kohlendorfer U, Ausserlechner MJ, Obexer P. C10ORF10/DEPP, a transcriptional target of FOXO3, regulates ROS-sensitivity in human neuroblastoma. Mol Cancer. 2014;13:224. doi: 10.1186/1476-4598-13-224 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Klee K, Storti F, Maggi J, Todorova V, Karademir D, Berger W, Samardzija M, Grimm C. The expression of decidual protein induced by progesterone (DEPP) is controlled by three distal consensus hypoxia responsive element (HRE) in hypoxic retinal epithelial cells. Genes (Basel). 2020;11:111. doi: 10.3390/genes11010111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Lee SY, Kang MG, Park JS, Lee G, Ting AY, Rhee HW. APEX fingerprinting reveals the subcellular localization of proteins of interest. Cell Rep. 2016;15:1837–1847. doi: 10.1016/j.celrep.2016.04.064 [DOI] [PubMed] [Google Scholar]
- 27.Kuwahara M, Akasaki Y, Kurakazu I, Sueishi T, Toya M, Uchida T, Tsutsui T, Hirose R, Tsushima H, Teramura T, et al. C10orf10/DEPP activates mitochondrial autophagy and maintains chondrocyte viability in the pathogenesis of osteoarthritis. FASEB J. 2022;36:e22145. doi: 10.1096/fj.202100896R [DOI] [PubMed] [Google Scholar]
- 28.Guo F, Zhu Y, Han Y, Feng X, Pan Z, He Y, Li Y, Jin L. DEPP deficiency contributes to browning of white adipose tissue. Int J Mol Sci. 2022;23:6563. doi: 10.3390/ijms23126563 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Menendez-Montes I, Escobar B, Palacios B, Gomez MJ, Izquierdo-Garcia JL, Flores L, Jimenez-Borreguero LJ, Aragones J, Ruiz-Cabello J, Torres M, et al. Myocardial VHL-HIF signaling controls an embryonic metabolic switch essential for cardiac maturation. Dev Cell. 2016;39:724–739. doi: 10.1016/j.devcel.2016.11.012 [DOI] [PubMed] [Google Scholar]
- 30.Li L, Wang ZV, Hill JA, Lin F. New autophagy reporter mice reveal dynamics of proximal tubular autophagy. J Am Soc Nephrol. 2014;25:305–315. doi: 10.1681/ASN.2013040374 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Kim WY, Safran M, Buckley MR, Ebert BL, Glickman J, Bosenberg M, Regan M, Kaelin WG, Jr. Failure to prolyl hydroxylate hypoxia-inducible factor alpha phenocopies VHL inactivation in vivo. EMBO J. 2006;25:4650–4662. doi: 10.1038/sj.emboj.7601300 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Rankin EB, Rha J, Selak MA, Unger TL, Keith B, Liu Q, Haase VH. Hypoxia-inducible factor 2 regulates hepatic lipid metabolism. Mol Cell Biol. 2009;29:4527–4538. doi: 10.1128/MCB.00200-09 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Krishnan J, Suter M, Windak R, Krebs T, Felley A, Montessuit C, Tokarska-Schlattner M, Aasum E, Bogdanova A, Perriard E, et al. Activation of a HIF1alpha-PPARgamma axis underlies the integration of glycolytic and lipid anabolic pathways in pathologic cardiac hypertrophy. Cell Metab. 2009;9:512–524. doi: 10.1016/j.cmet.2009.05.005 [DOI] [PubMed] [Google Scholar]
- 34.Nazareth W, Yafei N, Crompton M. Inhibition of anoxia-induced injury in heart myocytes by cyclosporin A. J Mol Cell Cardiol. 1991;23:1351–1354. doi: 10.1016/0022-2828(91)90181-k [DOI] [PubMed] [Google Scholar]
- 35.Susin SA, Zamzami N, Castedo M, Hirsch T, Marchetti P, Macho A, Daugas E, Geuskens M, Kroemer G. Bcl-2 inhibits the mitochondrial release of an apoptogenic protease. J Exp Med. 1996;184:1331–1341. doi: 10.1084/jem.184.4.1331 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Halestrap AP, Pasdois P. The role of the mitochondrial permeability transition pore in heart disease. Biochim Biophys Acta. 2009;1787:1402–1415. doi: 10.1016/j.bbabio.2008.12.017 [DOI] [PubMed] [Google Scholar]
- 37.Bauer TM, Murphy E. Role of mitochondrial calcium and the permeability transition pore in regulating cell death. Circ Res. 2020;126:280–293. doi: 10.1161/CIRCRESAHA.119.316306 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Brunelle JK, Bell EL, Quesada NM, Vercauteren K, Tiranti V, Zeviani M, Scarpulla RC, Chandel NS. Oxygen sensing requires mitochondrial ROS but not oxidative phosphorylation. Cell Metab. 2005;1:409–414. doi: 10.1016/j.cmet.2005.05.002 [DOI] [PubMed] [Google Scholar]
- 39.Guzy RD, Hoyos B, Robin E, Chen H, Liu L, Mansfield KD, Simon MC, Hammerling U, Schumacker PT. Mitochondrial complex III is required for hypoxia-induced ROS production and cellular oxygen sensing. Cell Metab. 2005;1:401–408. doi: 10.1016/j.cmet.2005.05.001 [DOI] [PubMed] [Google Scholar]
- 40.Mansfield KD, Guzy RD, Pan Y, Young RM, Cash TP, Schumacker PT, Simon MC. Mitochondrial dysfunction resulting from loss of cytochrome c impairs cellular oxygen sensing and hypoxic HIF-alpha activation. Cell Metab. 2005;1:393–399. doi: 10.1016/j.cmet.2005.05.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Sharma A, Toepfer CN, Schmid M, Garfinkel AC, Seidman CE. Differentiation and contractile analysis of GFP-sarcomere reporter hiPSC-cardiomyocytes. Curr Protoc Hum Genet. 2018;96:21.12.1–21.12.12. doi: 10.1002/cphg.53 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Hamid T, Guo SZ, Kingery JR, Xiang X, Dawn B, Prabhu SD. Cardiomyocyte NF-kappaB p65 promotes adverse remodelling, apoptosis, and endoplasmic reticulum stress in heart failure. Cardiovasc Res. 2011;89:129–138. doi: 10.1093/cvr/cvq274 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Lin Q, Huang Y, Booth CJ, Haase VH, Johnson RS, Celeste Simon M, Giordano FJ, Yun Z. Activation of hypoxia-inducible factor-2 in adipocytes results in pathological cardiac hypertrophy. J Am Heart Assoc. 2013;2:e000548. doi: 10.1161/JAHA.113.000548 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Ruiz M, Labarthe F, Fortier A, Bouchard B, Thompson Legault J, Bolduc V, Rigal O, Chen J, Ducharme A, Crawford PA, et al. Circulating acylcarnitine profile in human heart failure: a surrogate of fatty acid metabolic dysregulation in mitochondria and beyond. Am J Physiol Heart Circ Physiol. 2017;313:H768–H781. doi: 10.1152/ajpheart.00820.2016 [DOI] [PubMed] [Google Scholar]
- 45.Bertero E, Maack C. Metabolic remodelling in heart failure. Nat Rev Cardiol. 2018;15:457–470. doi: 10.1038/s41569-018-0044-6 [DOI] [PubMed] [Google Scholar]
- 46.Kaimoto S, Hoshino A, Ariyoshi M, Okawa Y, Tateishi S, Ono K, Uchihashi M, Fukai K, Iwai-Kanai E, Matoba S. Activation of PPAR-alpha in the early stage of heart failure maintained myocardial function and energetics in pressure-overload heart failure. Am J Physiol Heart Circ Physiol. 2017;312:H305–H313. doi: 10.1152/ajpheart.00553.2016 [DOI] [PubMed] [Google Scholar]
- 47.Brigadeau F, Gele P, Wibaux M, Marquie C, Martin-Nizard F, Torpier G, Fruchart JC, Staels B, Duriez P, Lacroix D. The PPARalpha activator fenofibrate slows down the progression of the left ventricular dysfunction in porcine tachycardia-induced cardiomyopathy. J Cardiovasc Pharmacol. 2007;49:408–415. doi: 10.1097/FJC.0b013e3180544540 [DOI] [PubMed] [Google Scholar]
- 48.Duhaney TA, Cui L, Rude MK, Lebrasseur NK, Ngoy S, De Silva DS, Siwik DA, Liao R, Sam F. Peroxisome proliferator-activated receptor alpha-independent actions of fenofibrate exacerbates left ventricular dilation and fibrosis in chronic pressure overload. Hypertension. 2007;49:1084–1094. doi: 10.1161/HYPERTENSIONAHA.107.086926 [DOI] [PubMed] [Google Scholar]
- 49.Yue TL, Bao W, Jucker BM, Gu JL, Romanic AM, Brown PJ, Cui J, Thudium DT, Boyce R, Burns-Kurtis CL, et al. Activation of peroxisome proliferator-activated receptor-alpha protects the heart from ischemia/reperfusion injury. Circulation. 2003;108:2393–2399. doi: 10.1161/01.CIR.0000093187.42015.6C [DOI] [PubMed] [Google Scholar]
- 50.Montaigne D, Butruille L, Staels B. PPAR control of metabolism and cardiovascular functions. Nat Rev Cardiol. 2021;18:809–823. doi: 10.1038/s41569-021-00569-6 [DOI] [PubMed] [Google Scholar]
- 51.Jun M, Foote C, Lv J, Neal B, Patel A, Nicholls SJ, Grobbee DE, Cass A, Chalmers J, Perkovic V. Effects of fibrates on cardiovascular outcomes: a systematic review and meta-analysis. Lancet. 2010;375:1875–1884. doi: 10.1016/S0140-6736(10)60656-3 [DOI] [PubMed] [Google Scholar]
- 52.Ferreira JP, Vasques-Novoa F, Ferrao D, Saraiva F, Falcao-Pires I, Neves JS, Sharma A, Rossignol P, Zannad F, Leite-Moreira A. Fenofibrate and heart failure outcomes in patients with type 2 diabetes: analysis from ACCORD. Diabetes Care. 2022;45:1584–1591. doi: 10.2337/dc21-1977 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Jain IH, Calvo SE, Markhard AL, Skinner OS, To TL, Ast T, Mootha VK. Genetic screen for cell fitness in high or low oxygen highlights mitochondrial and lipid metabolism. Cell. 2020;181:716–727.e11. doi: 10.1016/j.cell.2020.03.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Maulik N, Tosaki A, Engelman RM, Cordis GA, Das DK. Myocardial salvage by 1-O-hexadecyl-Sn-glycerol: possible role of peroxisomal dysfunction in ischemia reperfusion injury. J Cardiovasc Pharmacol. 1994;24:486–492. doi: 10.1097/00005344-199409000-00018 [DOI] [PubMed] [Google Scholar]
- 55.Liepinsh E, Skapare E, Kuka J, Makrecka M, Cirule H, Vavers E, Sevostjanovs E, Grinberga S, Pugovics O, Dambrova M. Activated peroxisomal fatty acid metabolism improves cardiac recovery in ischemia-reperfusion. Naunyn Schmiedebergs Arch Pharmacol. 2013;386:541–550. doi: 10.1007/s00210-013-0849-0 [DOI] [PubMed] [Google Scholar]
- 56.Yan L, Vatner DE, Kim SJ, Ge H, Masurekar M, Massover WH, Yang G, Matsui Y, Sadoshima J, Vatner SF. Autophagy in chronically ischemic myocardium. Proc Natl Acad Sci U S A. 2005;102:13807–13812. doi: 10.1073/pnas.0506843102 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Goswami SK, Das DK. Autophagy in the myocardium: dying for survival? Exp Clin Cardiol. 2006;11:183–188. [PMC free article] [PubMed] [Google Scholar]
- 58.Kostin S, Pool L, Elsasser A, Hein S, Drexler HC, Arnon E, Hayakawa Y, Zimmermann R, Bauer E, Klovekorn WP, et al. Myocytes die by multiple mechanisms in failing human hearts. Circ Res. 2003;92:715–724. doi: 10.1161/01.RES.0000067471.95890.5C [DOI] [PubMed] [Google Scholar]
- 59.Zhu H, Tannous P, Johnstone JL, Kong Y, Shelton JM, Richardson JA, Le V, Levine B, Rothermel BA, Hill JA. Cardiac autophagy is a maladaptive response to hemodynamic stress. J Clin Invest. 2007;117:1782–1793. doi: 10.1172/jci27523 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Wundersitz S, Pablo Tortola C, Schmidt S, Oliveira Vidal R, Kny M, Hahn A, Zanders L, Katus HA, Sauer S, Butter C, et al. The transcription factor EB (TFEB) sensitizes the heart to chronic pressure overload. Int J Mol Sci . 2022;23:5943. doi: 10.3390/ijms23115943 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Eckle T, Kohler D, Lehmann R, El Kasmi K, Eltzschig HK. Hypoxia-inducible factor-1 is central to cardioprotection: a new paradigm for ischemic preconditioning. Circulation. 2008;118:166–175. doi: 10.1161/CIRCULATIONAHA.107.758516 [DOI] [PubMed] [Google Scholar]
- 62.Huang M, Chan DA, Jia F, Xie X, Li Z, Hoyt G, Robbins RC, Chen X, Giaccia AJ, Wu JC. Short hairpin RNA interference therapy for ischemic heart disease. Circulation. 2008;118:S226–S233. doi: 10.1161/CIRCULATIONAHA.107.760785 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Philipp S, Jurgensen JS, Fielitz J, Bernhardt WM, Weidemann A, Schiche A, Pilz B, Dietz R, Regitz-Zagrosek V, Eckardt KU, et al. Stabilization of hypoxia inducible factor rather than modulation of collagen metabolism improves cardiac function after acute myocardial infarction in rats. Eur J Heart Fail. 2006;8:347–354. doi: 10.1016/j.ejheart.2005.10.009 [DOI] [PubMed] [Google Scholar]
- 64.Nwogu JI, Geenen D, Bean M, Brenner MC, Huang X, Buttrick PM. Inhibition of collagen synthesis with prolyl 4-hydroxylase inhibitor improves left ventricular function and alters the pattern of left ventricular dilatation after myocardial infarction. Circulation. 2001;104:2216–2221. doi: 10.1161/hc4301.097193 [DOI] [PubMed] [Google Scholar]
- 65.Olenchock BA, Moslehi J, Baik AH, Davidson SM, Williams J, Gibson WJ, Chakraborty AA, Pierce KA, Miller CM, Hanse EA, et al. EGLN1 inhibition and rerouting of alpha-ketoglutarate suffice for remote ischemic protection. Cell. 2016;164:884–895. doi: 10.1016/j.cell.2016.02.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Wyant GA, Yu W, Doulamis IP, Nomoto RS, Saeed MY, Duignan T, McCully JD, Kaelin WG, Jr. Mitochondrial remodeling and ischemic protection by G protein-coupled receptor 35 agonists. Science. 2022;377:621–629. doi: 10.1126/science.abm1638 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Matsui Y, Takagi H, Qu X, Abdellatif M, Sakoda H, Asano T, Levine B, Sadoshima J. Distinct roles of autophagy in the heart during ischemia and reperfusion: roles of AMP-activated protein kinase and Beclin 1 in mediating autophagy. Circ Res. 2007;100:914–922. doi: 10.1161/01.RES.0000261924.76669.36 [DOI] [PubMed] [Google Scholar]
- 68.Xie M, Cho GW, Kong Y, Li DL, Altamirano F, Luo X, Morales CR, Jiang N, Schiattarella GG, May HI, et al. Activation of autophagic flux blunts cardiac ischemia/reperfusion injury. Circ Res. 2021;129:435–450. doi: 10.1161/CIRCRESAHA.120.318601 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Nakai A, Yamaguchi O, Takeda T, Higuchi Y, Hikoso S, Taniike M, Omiya S, Mizote I, Matsumura Y, Asahi M, et al. The role of autophagy in cardiomyocytes in the basal state and in response to hemodynamic stress. Nat Med. 2007;13:619–624. doi: 10.1038/nm1574 [DOI] [PubMed] [Google Scholar]
- 70.Oyabu M, Takigawa K, Mizutani S, Hatazawa Y, Fujita M, Ohira Y, Sugimoto T, Suzuki O, Tsuchiya K, Suganami T, et al. FOXO1 cooperates with C/EBPdelta and ATF4 to regulate skeletal muscle atrophy transcriptional program during fasting. FASEB J. 2022;36:e22152. doi: 10.1096/fj.202101385RR [DOI] [PubMed] [Google Scholar]
- 71.Chen S, Gai J, Wang Y, Li H. FoxO regulates expression of decidual protein induced by progesterone (DEPP) in human endothelial cells. FEBS Lett. 2011;585:1796–1800. doi: 10.1016/j.febslet.2011.04.024 [DOI] [PubMed] [Google Scholar]
- 72.Kuroda Y, Kuriyama H, Kihara S, Kishida K, Maeda N, Hibuse T, Nishizawa H, Matsuda M, Funahashi T, Shimomura I. Insulin-mediated regulation of decidual protein induced by progesterone (DEPP) in adipose tissue and liver. Horm Metab Res. 2010;42:173–177. doi: 10.1055/s-0029-1241841 [DOI] [PubMed] [Google Scholar]
- 73.Milan G, Romanello V, Pescatore F, Armani A, Paik JH, Frasson L, Seydel A, Zhao J, Abraham R, Goldberg AL, et al. Regulation of autophagy and the ubiquitin-proteasome system by the FoxO transcriptional network during muscle atrophy. Nat Commun. 2015;6:6670. doi: 10.1038/ncomms7670 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Zhao J, Brault JJ, Schild A, Cao P, Sandri M, Schiaffino S, Lecker SH, Goldberg AL. FoxO3 coordinately activates protein degradation by the autophagic/lysosomal and proteasomal pathways in atrophying muscle cells. Cell Metab. 2007;6:472–483. doi: 10.1016/j.cmet.2007.11.004 [DOI] [PubMed] [Google Scholar]
- 75.Mammucari C, Milan G, Romanello V, Masiero E, Rudolf R, Del Piccolo P, Burden SJ, Di Lisi R, Sandri C, Zhao J, et al. FoxO3 controls autophagy in skeletal muscle in vivo. Cell Metab. 2007;6:458–471. doi: 10.1016/j.cmet.2007.11.001 [DOI] [PubMed] [Google Scholar]
- 76.Schips TG, Wietelmann A, Hohn K, Schimanski S, Walther P, Braun T, Wirth T, Maier HJ. FoxO3 induces reversible cardiac atrophy and autophagy in a transgenic mouse model. Cardiovasc Res. 2011;91:587–597. doi: 10.1093/cvr/cvr144 [DOI] [PubMed] [Google Scholar]
- 77.Altshuler-Keylin S, Shinoda K, Hasegawa Y, Ikeda K, Hong H, Kang Q, Yang Y, Perera RM, Debnath J, Kajimura S. Beige adipocyte maintenance is regulated by autophagy-induced mitochondrial clearance. Cell Metab. 2016;24:402–419. doi: 10.1016/j.cmet.2016.08.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Bonora M, Giorgi C, Pinton P. Molecular mechanisms and consequences of mitochondrial permeability transition. Nat Rev Mol Cell Biol. 2022;23:266–285. doi: 10.1038/s41580-021-00433-y [DOI] [PubMed] [Google Scholar]
- 79.Shanmughapriya S, Rajan S, Hoffman NE, Higgins AM, Tomar D, Nemani N, Hines KJ, Smith DJ, Eguchi A, Vallem S, et al. SPG7 is an essential and conserved component of the mitochondrial permeability transition pore. Mol Cell. 2015;60:47–62. doi: 10.1016/j.molcel.2015.08.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Tamura T, Said S, Gerdes AM. Gender-related differences in myocyte remodeling in progression to heart failure. Hypertension. 1999;33:676–680. doi: 10.1161/01.hyp.33.2.676 [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.