Skip to main content

Abstract

BACKGROUND:

Exercise-induced physiological cardiac growth regulators may protect the heart from ischemia/reperfusion (I/R) injury. Homeobox-containing 1 (Hmbox1), a homeobox family member, has been identified as a putative transcriptional repressor and is downregulated in the exercised heart. However, its roles in exercise-induced physiological cardiac growth and its potential protective effects against cardiac I/R injury remain largely unexplored.

METHODS:

We studied the function of Hmbox1 in exercise-induced physiological cardiac growth in mice after 4 weeks of swimming exercise. Hmbox1 expression was then evaluated in human heart samples from deceased patients with myocardial infarction and in the animal cardiac I/R injury model. Its role in cardiac I/R injury was examined in mice with adeno-associated virus 9 (AAV9) vector-mediated Hmbox1 knockdown and in those with cardiac myocyte–specific Hmbox1 ablation. We performed RNA sequencing, promoter prediction, and binding assays and identified glucokinase (Gck) as a downstream effector of Hmbox1. The effects of Hmbox1 together with Gck were examined in cardiomyocytes to evaluate their cell size, proliferation, apoptosis, mitochondrial respiration, and glycolysis. The function of upstream regulator of Hmbox1, ETS1, was investigated through ETS1 overexpression in cardiac I/R mice in vivo.

RESULTS:

We demonstrated that Hmbox1 downregulation was required for exercise-induced physiological cardiac growth. Inhibition of Hmbox1 increased cardiomyocyte size in isolated neonatal rat cardiomyocytes and human embryonic stem cell–derived cardiomyocytes but did not affect cardiomyocyte proliferation. Under pathological conditions, Hmbox1 was upregulated in both human and animal postinfarct cardiac tissues. Furthermore, both cardiac myocyte–specific Hmbox1 knockout and AAV9-mediated Hmbox1 knockdown protected against cardiac I/R injury and heart failure. Therapeutic effects were observed when sh-Hmbox1 AAV9 was administered after I/R injury. Inhibition of Hmbox1 activated the Akt/mTOR/P70S6K pathway and transcriptionally upregulated Gck, leading to reduced apoptosis and improved mitochondrial respiration and glycolysis in cardiomyocytes. ETS1 functioned as an upstream negative regulator of Hmbox1 transcription, and its overexpression was protective against cardiac I/R injury.

CONCLUSIONS:

Our studies unravel a new role for the transcriptional repressor Hmbox1 in exercise-induced physiological cardiac growth. They also highlight the therapeutic potential of targeting Hmbox1 to improve myocardial survival and glucose metabolism after I/R injury.

Clinical Perspective

What Is New?

Homeobox-containing 1 (Hmbox1) is responsive to exercise training, whose downregulation mediates exercise-induced physiological cardiac hypertrophy and protects against cardiac ischemia/reperfusion injury.
Inhibition of Hmbox1 can promote cardiomyocyte mitochondrial respiration and glycolysis in ischemia/reperfusion injury through the transcriptional activation of glucokinase (Gck).
Our study adds novel insights to the exercise-responsive molecules in regulating myocardial glycolysis metabolism.

What Are the Clinical Implications?

Reducing Hmbox1 promotes survival of human embryonic stem cell–derived cardiomyocytes, and therapeutic Hmbox1 knockdown reduces cardiac ischemia/reperfusion injury, indicating Hmbox1 as a therapeutic target for myocardial protection.
Inhibition of Hmbox1 and activation of Gck are prosperous to promote glycolysis and mitochondrial respiration of cardiomyocytes in treating cardiac ischemia/reperfusion injury and heart failure.
Coronary heart disease is among the leading causes of cardiovascular disease–related death worldwide.1 Despite advancements in the interventional approach for treating acute myocardial infarction (MI), effective drugs for preventing cardiac ischemia/reperfusion (I/R) injury in clinical practice are still absent.2 Cardiac I/R injury is related to complex pathophysiological processes that cause cardiomyocyte death, cardiac remodeling, and heart failure.3–5 It has been well-recognized that exercise is beneficial for cardiovascular health.6,7 Accumulating evidence has indicated that molecules regulated by exercise in the heart may protect the heart from cardiac I/R injury,8–11 thus prospectively leading to the identification of potential targets for myocardial protection.
Homeobox-containing 1 (Hmbox1) belongs to the homeobox family and has been identified as a putative transcriptional repressor.12,13 Hmbox1 is also a telomere-associated protein that can bind to telomere repeats and participate in telomerase recruitment and telomere maintenance.14–17 Increasing evidence has demonstrated that Hmbox1 regulates different cell processes, such as cell differentiation, proliferation, apoptosis, and autophagy.18–22 Dysregulated expression patterns of Hmbox1 also contribute to the development of cancers.23–26 Moreover, Hmbox1 negatively regulates natural killer cell functions by inhibiting the production of cytolytic molecules.27,28 We previously identified Hmbox1 as a downstream target gene of microRNA-222 (miR-222). The latter plays a key role in mediating exercise-induced physiological cardiac growth and protects against cardiac I/R injury.29 However, the functional role of Hmbox1 in the heart is far from elucidated. Whether and how Hmbox1 is involved in exercise-induced cardiac growth and its putative function in cardiac I/R injury remain largely unknown.
In the present study, we first demonstrated the contribution of reducing Hmbox1 to exercise-induced physiological cardiac hypertrophy. Conversely, Hmbox1 was upregulated in human and murine postinfarct cardiac tissues. We further revealed that inhibition of Hmbox1 in vivo through adeno-associated virus 9 (AAV9) injections or cardiac myocyte–specific Hmbox1 knockout had preventive and therapeutic effects against cardiac I/R injury and heart failure. Inhibition of Hmbox1 promoted physiological hypertrophy and reduced apoptosis of cardiomyocytes through activation of Akt (protein kinase B)/mTOR (mammalian target of rapamycin)/P70S6K (70 kDa ribosomal protein S6 kinase) signaling. Furthermore, we identified glucokinase (Gck) as a transcriptional direct downstream target of Hmbox1 that mediated the protective effect of reducing Hmbox1 against myocardial I/R injury through improving cardiomyocyte glycolysis. Last, we demonstrated that increasing ETS1 could downregulate Hmbox1, thereby protecting the heart. Therefore, these functional and mechanistic studies reveal a novel exercise-responsive, downregulated transcriptional repressor, Hmbox1, and underscore the therapeutic effect of Hmbox1 inhibition in reducing cardiac I/R injury.

METHODS

All data that support the findings of this work, as well as detailed methods and materials, are available in the Supplemental Material. Additional technical information is available from the corresponding authors upon reasonable request.
Human cardiac tissue sections, obtained from the left ventricular wall of deceased patients with MI, were used for immunohistochemistry staining of C4d and Hmbox1. The usage of human cardiac tissue was approved by the medical ethics committee of the University Medical Center Utrecht, The Netherlands. The study was conducted in accordance with the Declaration of Helsinki, and all participants provided written informed consent.
A 4-week swimming exercise program induced the physiological cardiac growth in adult male C57BL/6J mice with and without AAV9-mediated Hmbox1 overexpression. The function of Hmbox1 in cardiac I/R injury was evaluated in mice with AAV9-mediated Hmbox1 knockdown and in cardiac myocyte–specific Hmbox1 conditional knockout (cKO) mice. The Hmbox1 cKO mice were generated by crossing floxed Hmbox1 (Hmbox1fl/fl) mice with transgenic mice expressing Cre recombinase under the α-MHC (α-myosin heavy chain) promoter. All animal experiments complied with the Guidelines on the Care and Use of Laboratory Animals for biomedical research as published by the National Institutes of Health (No. 85-23, revised 1996) and were approved by the committee for the ethics of animal experiments of Shanghai University.
The regulatory effects of Hmbox1 on cardiomyocyte size, proliferation, and apoptosis underwent stress through oxygen glucose deprivation and reperfusion (OGDR) were investigated in isolated neonatal rat cardiomyocytes (NRCMs) and human embryonic stem cell–derived cardiomyocytes. RNA sequencing (RNA-seq) and promoter prediction using Cistrome Data Browser were performed to identify the downstream targets of Hmbox1. The RNA-seq profiling data are available in the Gene Expression Omnibus (GEO: GSE241079). Cardiomyocyte mitochondrial respiration and glycolysis, in relation to Hmbox1 and Gck, were investigated in NRCMs using the Seahorse XF flux analyzer 96.

Statistical Analysis

All data were analyzed using SPSS20.0 or GraphPad Prism 8 and presented as mean±SD using GraphPad Prism 8. Normality distribution test was first performed for all data. For data with normality distribution, unpaired Student t test, 1-way ANOVA test, or 2-way ANOVA test followed by Tukey post hoc test was used for comparisons of difference between 2 groups or multigroups. For data that did not pass the normality distribution test, nonparametric unpaired Mann-Whitney U test, Kruskal-Wallis test with the original false discovery rate method of Benjamini and Hochberg, or robust 2-way ANOVA test followed by post hoc pairwise Median Test in the rcompanion package was applied for comparisons of difference between 2 groups or multigroups. A P value <0.05 was considered statistically significant.

RESULTS

Downregulation of Hmbox1 Contributes to Exercise-Induced Physiological Cardiac Hypertrophy

We previously reported Hmbox1 as a target gene of miR-222, a vital microRNA contributing to exercise-induced physiological cardiac growth.29 Nevertheless, the role of Hmbox1 in physiological cardiac growth remained elusive. In the present study, we identified a significant downregulation of Hmbox1 in heart tissues from a murine swimming exercise model that induces physiological cardiac growth (Figure 1A and 1B). In contrast, Hmbox1 was upregulated in transaortic constriction–induced pathological cardiac hypertrophy (Figure S1A), highlighting differential regulation of Hmbox1 in physiological versus pathological hypertrophy. Next, we isolated NRCMs and neonatal rat cardiac fibroblasts and observed that Hmbox1 was preferentially expressed in cardiomyocytes compared with cardiac fibroblasts at both mRNA and protein levels (Figure S1B and S1C). Although Hmbox1 was downregulated in both cardiomyocytes and cardiac fibroblasts isolated from heart tissue of adult mice subjected to swimming (Figure S1D), functional experiments through upregulating or knocking down Hmbox1 in neonatal rat cardiac fibroblasts showed that Hmbox1 did not influence neonatal rat cardiac fibroblast proliferation or differentiation to myofibroblasts in vitro as determined by immunofluorescent staining for EdU (5-ethynyl-2’-deoxyuridine) and α-SMA (α-smooth muscle actin; Figure S2). We subsequently delved deeper into the function of Hmbox1 in cardiomyocytes.
Figure 1. Homeobox-containing 1 (Hmbox1) downregulation is required for exercise-induced physiological cardiac hypertrophy. A, Western blot analysis of Hmbox1 in the hearts of sedentary control and swim-trained mice (n=6). B, Quantitative reverse-transcription polymerase chain reaction (qRT-PCR) assessing Hmbox1 levels in sedentary control and swim mice hearts (n=6). C, Measurements of α-actinin labeled cardiomyocyte (CM) size and the percentage of EdU (5-ethynyl-2’-deoxyuridine) and α-actinin double-positive CMs in stained neonatal rat cardiomyocytes (NRCMs) infected with either Hmbox1 knockdown (sh-Hmbox1) or overexpression (OE-Hmbox1) lentivirus and their respective control vectors (n=6). Scale bar=100 μm. D, qRT-PCR of Hmbox1 and pathological hypertrophy markers in NRCMs infected with sh-Hmbox1 or OE-Hmbox1 lentivirus and their respective control vectors (n=5 or 6). E, Schematic diagram illustrating the procedure wherein mice were injected with Hmbox1 overexpression adeno-associated virus 9 (OE-Hmbox1 AAV9) or control AAV9 (CTL AAV9), and 1 week later began a swimming regimen for 4 weeks. F, qRT-PCR of Hmbox1 in the hearts of control and swim-trained mice, both with or without Hmbox1 overexpression (n=6 for control groups; n=9 for swim groups). G, Measurements of heart weight (HW) and heart weight/tibia length (HW/TL) ratio for control and swim-trained mice with or without Hmbox1 overexpression (n=6 for control groups, n=9 for swim groups). H, Wheat germ agglutinin (WGA) staining of cardiac tissues to measure cardiomyocyte cross-sectional area (n=6). Scale bar=100 μm. I, Quantification of Ki67 and α-actinin double-positive CMs (%) in cardiac tissues (n=6). Scale bar=100 μm. J, qRT-PCR of Anp (atrial natriuretic peptide), Bnp (brain natriuretic peptide), and β-MHC (β-myosin heavy chain) in the hearts of control and swim-trained mice with or without Hmbox1 overexpression (n=6 for control groups; n=9 for swim groups). Statistical analysis was performed by unpaired Student t test for A through D, by 2-way ANOVA test followed by Tukey post hoc test for F, H, and J, and by robust 2-way ANOVA test followed by post hoc pairwise Median Test for G and I. *P<0.05; **P<0.01; ***P<0.001.
Exercise-induced physiological cardiac growth is characterized by physiological hypertrophy and increased proliferation markers in cardiomyocytes.9,29 Our experiments in NRCMs revealed that Hmbox1 knockdown increased cardiomyocyte size, whereas its overexpression decreased it; however, Hmbox1 did not affect cardiomyocyte proliferation (Figure 1C). Moreover, pathological hypertrophy markers atrial natriuretic peptide (Anp) and brain natriuretic peptide (Bnp) remained unaffected by Hmbox1 in cardiomyocytes (Figure 1D), indicating the physiological function of Hmbox1 in regulating cardiomyocyte size.
Meanwhile, we determined Hmbox1 expression in human embryonic stem cell–derived cardiomyocytes treated either by a physiological mechanical stretch using the Flexcell system or a pharmacological stimulus with IGF-1 (insulin-like growth factor 1), which simulated the effect of exercise in vitro.30,31 Our results showed that both the mechanical stretch to a physiological extent and IGF-1, a canonical mediator for exercise-induced physiological cardiac hypertrophy,32 could downregulate Hmbox1 expression at both the mRNA and protein levels in human embryonic stem cell–derived cardiomyocytes, accompanied by an increased level of Akt phosphorylations, whereas Anp and Bnp expressions were not changed, thus excluding pathological hypertrophy in these conditions (Figure S3). Thus, Hmbox1 was also downregulated in exercise-simulated treatment of human cardiomyocytes. We further determined the function of Hmbox1 in human embryonic stem cell–derived cardiomyocytes and found that reducing Hmbox1 also enlarged human cardiomyocyte size while overexpressing Hmbox1 decreased cardiomyocyte size, without influencing the Anp and Bnp expressions (Figure S4), indicating that Hmbox1 was also able to regulate human cardiomyocyte size physiologically.
In vivo examination of the role of Hmbox1 in exercise-induced physiological cardiac growth involved injecting mice with AAV9 expressing Hmbox1 (overexpressing-Hmbox1-AAV9) and then subjecting them to swimming exercise (Figure 1E). We observed that neither cardiac systolic function nor diastolic function was markedly influenced in the swimming-exercised mice with Hmbox1 overexpression or not (Tables S1 and S2). After evaluating cardiac function, mice heart tissues were harvested, and we verified the efficiency of OE-Hmbox1-AAV9 in enhancing Hmbox1 expression in mice hearts (Figure 1F). It is interesting that the swim-induced increase in heart weight and the heart weight/tibia length ratio were reduced in mice overexpressing Hmbox1 (Figure 1G). Wheat germ agglutinin staining further indicated that upregulating Hmbox1 abolished the exercise-induced enlargement of the cross-sectional area of the myocardium (Figure 1H). However, the exercise-induced increase in Ki67-labeled cardiomyocytes was not influenced by Hmbox1 overexpression (Figure 1I), which was corroborating our in vitro findings of the noninvolvement of Hmbox1 in cardiomyocyte proliferation. Similarly, pathological hypertrophy markers Anp, Bnp, and β-Mhc (β-myosin heavy chain) were unaffected by Hmbox1 overexpression in mice hearts (Figure 1J). These data indicate that although Hmbox1 regulates physiological hypertrophy, it does not affect cardiomyocyte proliferation. This supports the idea that downregulating Hmbox1 is pivotal for exercise-induced physiological cardiac hypertrophy.

Hmbox1 Is Upregulated in Postinfarct Myocardium, and Its Downregulation Reduces Cardiomyocyte Apoptosis

Considering exercise-regulated molecules might confer myocardial protection in pathological conditions, we assessed Hmbox1 expression in clinical and experimental postinfarct cardiac tissues, and then examined its function in cardiomyocytes upon pathological stress. Heart samples from 2 deceased patients with MI who had received vascular revascularization treatment were collected from the infarct area (Table S3). The myocardial slices were stained with double immunohistochemistry for C4d/Hmbox1, displaying a markedly high expression of Hmbox1 in MI zones adjacent to the necrotic cardiomyocytes (Figure 2A). These observations suggest that increased Hmbox1 may be involved in human MI disease progression.
Figure 2. Homeobox-containing 1 (Hmbox1) is upregulated in postinfarct myocardium, and downregulation of Hmbox1 inhibits cardiomyocyte apoptosis. A, Heart samples of the infarct area from 2 patients (patient Nos. 1 and 2) with myocardial infarction (MI) were collected. Myocardial slices underwent immunohistochemistry staining for C4d/Hmbox1, highlighting Hmbox1 expression (red) around the necrotic myocardium (blue). Scale bar=5 mm. B and C, Western blot analysis showing Hmbox1 levels in heart tissues from sham and cardiac ischemia/reperfusion (I/R) mice at 24 hours (B, n=3) and 3 weeks (C, n=6) after injury. D, Western blot analysis of Hmbox1 in neonatal rat cardiomyocytes (NRCMs) subjected to oxygen glucose deprivation and reperfusion (OGDR) stress (n=6). E, Quantification of terminal deoxynucleotidyl transferase mediated dUTP nick-end labeling (Tunel) and α-actinin double positive cardiomyocytes (CMs, %) in stained NRCMs infected with Hmbox1 knockdown (sh-Hmbox1) lentivirus and control vectors, with or without OGDR stress (n=6). Scale bar=100 μm. F, Western blot of Hmbox1 in human embryonic stem cell-derived cardiomyocytes (hESC-CM) either subjected to OGDR stress or not (n=3). G, Quantification of Tunel and α-actinin double-positive cardiomyocytes (CMs, %) in stained hESC-CM infected with sh-Hmbox1 lentivirus and control vectors, with or without OGDR stress (n=6). Scale bar=100 μm. Statistical analysis was performed by unpaired Student t test for C, D, and F, by 1-way ANOVA test followed by Bonferroni test for B, and by 2-way ANOVA test followed by Tukey post hoc test for E and G. **P<0.01; ***P<0.001.
In parallel, we assessed Hmbox1 expression in experimental mouse models of cardiac I/R injury. Hmbox1 was significantly upregulated in the border zone of I/R heart in the acute (24 hours) phase (Figure 2B), and in the remodeling I/R heart in the chronic (3 weeks) phase as well (Figure 2C). Likewise, Hmbox1 was upregulated in isolated NRCMs upon OGDR stress, which mimics cardiac I/R injury in vitro (Figure 2D). OGDR-induced cardiomyocyte apoptosis was then determined in Hmbox1 knocked down or overexpressed NRCMs. We first confirmed that Hmbox1 was efficiently knocked down by sh-Hmbox1 lentivirus (Figure S5A), and overexpressed by OE-Hmbox1 lentivirus in NRCMs (Figure S5C), either with OGDR stress or not. Hmbox1 knockdown inhibited OGDR-induced cardiomyocyte apoptosis, as evidenced by reduced terminal deoxynucleotidyl transferase mediated dUTP nick-end labeling (Tunel)–positive cardiomyocytes, diminished Bax/Bcl2 ratio, and decreased cleaved caspase3 expression (Figure 2E; Figure S5A). On the contrary, Hmbox1 overexpression aggravated cardiomyocyte apoptosis (Figure S5B and S5C). Meanwhile, we cultured human embryonic stem cell–derived cardiomyocytes and found an increase of Hmbox1 expression in OGDR-stressed human cardiomyocytes (Figure 2F), whereas reducing Hmbox1 prevented cardiomyocyte apoptosis (Figure 2G; Figure S6). Our data from both human and murine samples indicate that Hmbox1 is upregulated in postinfarct myocardium, whereas downregulation of Hmbox1 reduces cardiomyocyte apoptosis in both human and murine cardiomyocytes.

AAV9-Mediated Hmbox1 Knockdown Protects Against Cardiac I/R Injury

To assess the in vivo effects of targeting Hmbox1, we used a murine model of cardiac I/R injury. Mice were injected with AAV9, which mediated Hmbox1 knockdown through shRNA (sh-Hmbox1-AAV9), and then subjected to I/R injury (Figure S7A). Mouse hearts were harvested 24 hours after I/R, and the border zone of I/R hearts was used to determine the expression of Hmbox1 and to examine the apoptosis-related molecular changes. Our results showed that sh-Hmbox1-AAV9 significantly downregulated Hmbox1 in the heart tissues (Figure S7B and S7C). Functionally, Hmbox1 knockdown reduced the infarct size (Figure 3A) and inhibited myocardial apoptosis (Figure 3B; Figure S7C). At 3 weeks after I/R (Figure S8A through S8C), Hmbox1 knockdown also preserved cardiac function and inhibited cardiac apoptosis and remodeling (Figure 3C through 3F; Table S4; Figure S8C). Moreover, downregulation of Hmbox1 was associated with increased Akt phosphorylation, a key signaling pathway for cell survival, in I/R heart tissues (Figures S7D and S8D). Hmbox1 also influenced the Akt/mTOR/P70S6K pathway in cardiomyocytes in vitro (Figure S9). Our data further showed that Akt activation was necessary to mediate the effect of reduced Hmbox1 in promoting cardiomyocyte size and inhibiting cardiomyocyte apoptosis (Figure S10). This suggests that reducing Hmbox1 can activate the Akt/mTOR/P70S6K signaling, which might contribute to its protective effect against cardiac I/R injury and myocardial apoptosis.
Figure 3. Adeno-associated virus 9 (AAV9)–mediated Homeobox-containing 1 (Hmbox1) downregulation protects against cardiac ischemia/reperfusion injury. A, 2,3,5-Triphenyltetrazolium chloride (TTC) staining was used to evaluate the area at risk/left ventricle weight (AAR/LV) ratio and the infarct size/area at risk (INF/AAR) ratio in I/R mice 24 hours after injury (n=6 vs 8). B, Quantification of terminal deoxynucleotidyl transferase mediated dUTP nick-end labeling (Tunel) and α-actinin double positive cardiomyocytes (CMs, %) in stained cardiac tissues from either sham or I/R mice, 24 hours after injury (n=6, 7, 8, 10). Scale bar=100 μm. C, Echocardiography was used to measure the left ventricle ejection fraction (EF) and fractional shortening (FS) in mice 3 weeks after I/R injury (n=8, 8, 9, 9). D, Masson’s trichrome staining for assessment of cardiac fibrosis, 3 weeks after I/R injury (n=8, 7, 7, 9). Scale bar=100 μm. E and F, Quantitative reverse-transcription polymerase chain reaction was used to measure expression levels of cardiac remodeling marker genes (E, including Anp [atrial natriuretic peptide], Bnp [brain natriuretic peptide], and β-MHC [β-myosin heavy chain]) and fibrosis marker genes (F, including Col1a1, Col3a1, and Ctgf) in mouse heart tissues 3 weeks after I/R injury (n=8). Statistical analysis was performed by unpaired Student t test for A, by 2-way ANOVA test followed by Tukey post hoc test for B through D, E (Bnp and β-MHC), and F, and by robust 2-way ANOVA test followed by post hoc pairwise Median Test for E (Anp). *P<0.05; **P<0.01; ***P<0.001.

Hmbox1 Is a Transcriptional Repressor Targeting Gck to Control Cardiomyocyte Size and Apoptosis

To investigate the mechanisms underlying the protective effects of reducing Hmbox1 in myocardial tissue, we performed RNA-seq of heart tissues from both sham and 3-week-I/R mice injected with either sh-Hmbox1-AAV9 or CTL-AAV9. The RNA-seq profiling data has been uploaded to the Gene Expression Omnibus (GEO: GSE241079). We identified 119 upregulated and 38 downregulated genes in the I/R group compared with the sham group (fold change >2.0; P<0.05; Figure 4A). Concurrently, 83 genes were upregulated and 124 genes were downregulated in the I/R mice treated with sh-Hmbox1-AAV9 relative to those treated with CTL-AAV9 (fold change >2.0; P<0.05) (Figure 4B). Hmbox1 is recognized as a novel transcriptional repressor that negatively regulates target gene expression.12 From the RNA-seq data, 12 genes were found to be downregulated in I/R hearts but upregulated after sh-Hmbox1-AAV9 intervention, as shown in the Venn diagram. We further used the Cistrome Data Browser and applied the Tools “Check a putative target” to predict the potential targets that Hmbox1 might regulate at the promoter regions and found that Myl4, Pllp, and Gck had the top 3 scores among the 12 genes (Figure 4C). We continued to assess the expression of these genes in the animal I/R model and the OGDR-stressed NRCMs by quantitative reverse-transcription polymerase chain reaction. Only Gck was consistently downregulated in the myocardium after both I/R injury and OGDR stress, but was upregulated after Hmbox1 inhibition at the mRNA level (Figure 4D; Figure S11A) and the protein level as well (Figure S11B and S11C), which was consistent with the RNA-seq data. According to the quantitative reverse-transcription polymerase chain reaction validation experiments, although Myl4 was upregulated in the I/R heart tissues compared with the sham group, and was downregulated in the I/R mice injected with sh-Hmbox1 AAV9 compared with those injected with CTL-AAV9 (Figure 4D), the results from quantitative reverse-transcription polymerase chain reaction for Myl4 were not consistent with the RNA-seq analysis, which was at least in part attributed to the within-group variations of the RNA-seq data for Myl4. In addition, Hmbox1, known as a transcriptional repressor, was not able to negatively regulate Myl4 expression in NRCMs (Figure 4D). Considering the above reasons, Gck was selected to be further studied. Kyoto Encyclopedia of Genes and Genomes enrichment analysis of metabolism pathways further showed that Gck could be involved in glycolysis (Figure 4E), highlighting the possible regulatory role of Hmbox1 in myocardial metabolism upon I/R injury. Collectively, we identified Gck as the most likely downstream target of Hmbox1 during cardiac I/R injury.
Figure 4. Homeobox-containing 1 (Hmbox1) functions through glucokinase (Gck). A, Volcano plots of differentially expressed genes in heart tissues from the cardiac ischemia/reperfusion (I/R, 3 weeks after I/R) group compared with the sham group (P value<0.05 and |log2 fold change|>1). Both groups were injected with control adeno-associated virus 9 (AAV9). The top 10 genes according to the fold change and Gck were marked in the volcano plots. B, Volcano plots of differentially expressed genes in heart tissues from the cardiac I/R (3 weeks after I/R) mice injected with sh-Hmbox1 AAV9 compared with the I/R mice injected with control AAV9 (P value<0.05 and |log2 fold change|>1). The top 10 ranked genes according to the fold change and Gck were marked in the volcano plots. C, Bioinformatic analyses of RNA-sequencing data, accompanied by promoter prediction using Cistrome Data Browser (Cistrome DB), to identify potential downstream targets of Hmbox1. D, Quantitative reverse-transcription polymerase chain reaction (qRT-PCR) was used to measure Pllp, Myl4, and Gck gene expression levels in mouse heart tissues at 3 weeks after I/R injury (n=6) and in neonatal rat cardiomyocytes (NRCMs) infected with sh-Hmbox1 lentivirus with or without oxygen glucose deprivation and reperfusion (OGDR) stress (n=6). E, KEGG (Kyoto Encyclopedia of Genes and Genomes) enrichment analysis of metabolism pathways for genes that showed downregulation in I/R hearts compared with sham, whereas they were upregulated in I/R hearts after sh-Hmbox1-AAV9 intervention. F and G, qRT-PCR of Gck in NRCMs treated either with Hmbox1 overexpression (F, n=6) or its inhibition (G, n=6). H, Luciferase activity measurement in HEK293 cells transfected with a Gck promoter reporter with or without Hmbox1 overexpression (OE-Hmbox1) lentivirus treatment (n=6). I, Chromatin immunoprecipitation polymerase chain reaction was used to determine the interaction between Hmbox1 protein and Gck gene promoter in H9C2 cells (n=3). J, α-actinin–labeled cardiomyocyte (CM) size was measured in stained NRCMs treated with sh-Hmbox1 lentivirus and Gck siRNA (si-Gck) (n=6). Scale bar=100 μm. K, Quantification of terminal deoxynucleotidyl transferase mediated dUTP nick-end labeling (Tunel) and α-actinin double positive CMs (%) in stained NRCMs treated with sh-Hmbox1 lentivirus and si-GCK under OGDR condition (n=5). Scale bar=100 μm. Statistical analysis was performed by 2-way ANOVA test followed by Tukey post hoc test for D (IRI 3-week experiment and Gck in OGDR experiment), J and K, by robust 2-way ANOVA test followed by post hoc pairwise Median Test for D (Pllp and Myl4 in OGDR experiment), and by unpaired Student t test for F through I. *P<0.05; **P<0.01; ***P<0.001.
To further elucidate the regulation of Gck by Hmbox1 in cardiomyocytes, we infected primary NRCMs with either OE-Hmbox1 or sh-Hmbox1 lentivirus. We observed that Hmbox1 negatively regulated Gck at both mRNA and protein levels (Figure 4F and 4G; Figure S11D and S11E). Both the luciferase reporter experiment and the chromatin immunoprecipitation polymerase chain reaction assay confirmed the ability of Hmbox1 to directly bind to and enrich in the promoter region of Gck (Figure 4H and 4I). These findings suggest that Hmbox1 operates as a transcriptional repressor, negatively regulating Gck in cardiomyocytes.
Next, we performed function-rescue experiments to evaluate the involvement of Gck in the function of Hmbox1. In NRCMs cotreated with sh-Hmbox1 and Gck siRNA (si-Gck), we observed that downregulation of Gck significantly reversed the increased cell size and antiapoptotic effect observed with Hmbox1 knockdown (Figure S12A and S12B; Figure 4J and 4K). Using a glucokinase activator, GKA50, we observed that GKA50 could inhibit cardiomyocyte apoptosis under OGDR stress (Figure S12C) and efficiently increased Gck activity in cardiomyocytes (Figure S12D). Meanwhile, we demonstrated that Gck activation in NRCMs counteracted the negative regulation of cell size and the proapoptotic effect seen with Hmbox1 overexpression (Figure S12E and S12F). Taken together, these findings underscore the role of Hmbox1 as a transcriptional repressor targeting Gck. By reducing Hmbox1, physiological hypertrophy is promoted, and cardiomyocyte apoptosis is inhibited through Gck activation.

Reducing Hmbox1 Improves Mitochondrial Respiration and Glycolysis in OGDR-Stressed Cardiomyocytes Through Gck Activation

Gck, also known as glucokinase, encodes hexokinase-IV, a member of the hexokinase protein family. This enzyme catalyzes the initial step of glycolysis: the phosphorylation of glucose to form glucose-6-phosphate.33 Impaired glycolysis and mitochondrial respiration are critical contributors to cardiomyocyte death during cardiac hypoxic and ischemic stress.34,35 However, the role of Hmbox1 inhibition in regulating myocardium metabolism remains largely unknown. Using the XFe96 extracellular flux analyzer, we observed that OGDR stress significantly reduced mitochondrial respiration in isolated cardiomyocytes, as indicated by reduced levels of oxygen consumption rate, basal respiration, maximal respiration, and spare respiratory capacity (Figure 5A). Concurrently, ATP production also diminished in cardiomyocytes subjected to OGDR stress (Figure 5A). However, the Hmbox1 inhibition mitigated the OGDR-induced reduction of mitochondrial respiration (Figure 5A). Conversely, OGDR stress increased the glycolysis proton efflux rate and enhanced the basal and compensatory glycolysis levels in isolated cardiomyocytes (Figure 5B). We hypothesize that this metabolic shift is a self-responsive adaptation of cardiomyocytes to OGDR stress. It is interesting that Hmbox1 inhibition further amplified glycolysis in OGDR-stressed cardiomyocytes (Figure 5B).
Figure 5. Inhibition of Homeobox-containing 1 (Hmbox1) improves mitochondrial respiration and glycolysis in cardiomyocytes through activating glucokinase (Gck). A and B, Mitochondrial stress test (A, n=42–45) and glycolysis rate assay (B, n=39–46) were conducted on neonatal rat cardiomyocytes (NRCMs) that were infected with sh-Hmbox1 lentivirus or their respective control vectors and were subjected to oxygen glucose deprivation and reperfusion (OGDR) stress or not. C and D, Mitochondrial stress test (C, n=19–21) and glycolysis rate assay (D, n=12–17) were conducted in NRCMs cotreated with sh-Hmbox1 lentivirus and Gck siRNA (si-Gck) and exposed to OGDR stress. Statistical analysis was performed by 2-way ANOVA test followed by Tukey post hoc test for A (maximal respiration and spare respiratory capacity), B (basal glycolysis), and C and D (compensatory glycolysis), and by robust 2-way ANOVA test followed by post hoc pairwise Median Test for A (basal respiration and ATP production), B (compensatory glycolysis), and D (basal glycolysis). *P<0.05; **P<0.01; ***P<0.001. glycoPER indicates glycolytic proton efflux rate; OCR, oxygen consumption rate; and si-NC, siRNA negative control.
To further investigate the role of Gck activation in Hmbox1-regulated metabolism, cardiomyocytes were cotreated with si-Gck and sh-Hmbox1 lentivirus. Results demonstrated that Gck knockdown significantly diminished the beneficial effects of Hmbox1 inhibition on mitochondrial respiration and glycolysis (Figure 5C and 5D). Collectively, this evidence directly suggests that reducing Hmbox1 improves mitochondrial respiration and glycolysis in cardiomyocytes under OGDR stress by activating Gck.

Cardiac Myocyte–Specific Hmbox1 Suppression Protects Against Cardiac I/R Injury

To specifically assess the functional role of Hmbox1 in cardiomyocytes in vivo, we used cardiac myocyte–specific Hmbox1 cKO (Hmbox1fl/fl Cre+) mice and control (Hmbox1fl/fl Cre-) mice and established a cardiac I/R injury model (Figure 6A and 6B). We first confirmed efficient Hmbox1 suppression in mouse heart tissues (Figure 6C and 6I). Three weeks after I/R injury, Hmbox1 cKO mice demonstrated preserved cardiac function and diminished cardiac remodeling (Figure 6D through 6G; Table S5). Cardiac-specific Hmbox1 cKO mice exhibited a reduced Bax/Bcl2 ratio and caspase3 cleavage in heart tissues, alongside increased Akt phosphorylation (Figure 6H; Figure S13), suggesting attenuated myocardial apoptosis. It is important to note that these mice also displayed elevated Gck expression in heart tissues (Figure 6I and 6J), indicating the efficacy of cardiac-specific Hmbox1 ablation in activating Gck in vivo.
Figure 6. Cardiac myocyte–specific Homeobox-containing 1 (Hmbox1) ablation protects the heart from ischemia/reperfusion (I/R) injury. A, Floxed Hmbox1 (Hmbox1fl/fl) mice and transgenic mice expressing Cre recombinase under α-MHC (α-myosin heavy chain) promoter were used to generate cardiac myocyte–specific Hmbox1 conditional knockout (cKO, Hmbox1fl/fl Cre+) mice. These were then compared with control (CON, Hmbox1fl/fl Cre-) mice. B, A schematic representation showing Hmbox1 cKO and CON mice subjected to 3 weeks of cardiac I/R injury. C, Quantitative reverse-transcription polymerase chain reaction (qRT-PCR) determining Hmbox1 levels in mice heart tissues 3 weeks after I/R injury (n=3 for sham group; n=5 for I/R group). D, Echocardiography assessing left ventricle ejection fraction (EF) and fractional shortening (FS) in mice 3 weeks after I/R injury (n=3 for sham group, n=5 for I/R group). E, Masson’s trichrome staining was used to evaluate cardiac fibrosis 3 weeks after I/R injury (n=3–5). Scale bar=100 μm. F and G, qRT-PCR analysis of fibrosis marker genes (F) and cardiac remodeling marker genes (G) in mice heart tissues 3 weeks after I/R injury (n=3–5). H and I, Western blot showing caspase3 cleavage and Bax/Bcl2 ratio (H), along with Hmbox1 and glucokinase (Gck; I) in mice heart tissues 3 weeks after I/R injury (n=3). J, qRT-PCR analysis of Gck in mice heart tissues 3 weeks after I/R injury (n=3–5). Statistical analysis was performed by 2-way ANOVA test followed by Tukey post hoc test for C through E, F (Col1a1 and Col3a1), G (Anp [atrial natriuretic peptide] and β-MHC [β-myosin heavy chain]), and H through J, and by robust 2-way ANOVA test followed by post hoc pairwise Median Test for F (Ctgf) and G (Bnp [brain natriuretic peptide]). *P<0.05; **P<0.01; ***P<0.001.
In a separate animal experiment, we injected Hmbox1fl/fl mice with AAV9 vectors expressing cardiac-specific cTnT (cardiac troponin T) promoter–driven Cre (induced Cre-AAV9, iCre-AAV9), followed by cardiac I/R injury or sham operation a week later (Figure S14A). Three weeks after I/R injury, Hmbox1 was significantly reduced in mice heart tissues (Figure S14B and S14H). Interestingly, cardiac-specific Hmbox1 knockdown markedly improved cardiac function, alleviated cardiac apoptosis and remodeling (Figure S14C through S14G; Table S6; Figure S15), and was accompanied by an increase in cardiac Gck expression (Figure S14H and S14I). Collectively, this evidence strongly supports the protective role of cardiac myocyte–specific Hmbox1 suppression against cardiac I/R injury and its activation of Gck in vivo.

ETS1 Negatively Regulates Hmbox1 and Prevents Cardiac I/R Injury

To identify potential upstream regulators of Hmbox1, we consulted the PROMO (prediction of transcription factor binding sites) database, which predicted GR, STAT4, IK-1, and ETS1 as candidates. Given the downregulation of Hmbox1 in exercised hearts and its upregulation during cardiac I/R injury, we first examined the regulation of these genes in these conditions. Validation experiments revealed that only ETS1 exhibited downregulation in OGDR-stressed NRCMs and heart tissues after I/R injury and upregulation in the hearts of swimming-exercised mice (Figures S16, S17A, and S17B; Figure 7A and 7B). This suggests an inverse relationship between ETS1 and Hmbox1 regulation in these scenarios. Subsequent studies demonstrated that ETS1 repressed Hmbox1 expression in primary NRCMs (Figure 7C; Figure S17C and S17D). Chromatin immunoprecipitation polymerase chain reaction assay analysis revealed an enrichment of ETS1 in the promoter region of Hmbox1 (Figure 7D). Gain- and loss-of-function experiments in NRCMs showed that ETS1 overexpression increased cell size and inhibited cardiomyocyte apoptosis, whereas ETS1 knockdown had the opposite effects (Figure S18). Rescue experiments further revealed that Hmbox1 knockdown abolished the effects of reduced ETS1 on cardiomyocyte size and apoptosis (Figure S19). These findings indicate that ETS1 acts as a predominant upstream inhibitor of Hmbox1 in the myocardium.
Figure 7. ETS1 downregulates Homeobox-containing 1 (Hmbox1) and is protective during cardiac ischemia/reperfusion (I/R) injury. A and B, Western blot of ETS1 in heart tissues from sham and cardiac I/R mice at 24 hours (A, n=3) and 3 weeks (B, n=6) after injury. C, Quantitative reverse-transcription polymerase chain reaction (qRT-PCR) analysis of Hmbox1 in neonatal rat cardiomyocytes (NRCMs) infected with either ETS1 knockdown (sh-ETS1) or overexpression (OE-ETS1) lentivirus vs their respective control vectors (n=6). D, Chromatin immunoprecipitation-polymerase chain reaction assessing the interaction between ETS1 protein and Hmbox1 gene promoter in H9C2 cells (n=5). E, 2,3,5-Triphenyltetrazolium chloride (TTC) staining was used to assess the area at risk/left ventricle weight (AAR/LV) ratio and the infarct size/area at risk (INF/AAR) ratio in mice that underwent intramyocardial injection with ETS1 overexpressing (OE-ETS1) lentivirus followed by 24 hours of cardiac I/R injury (n=6). F, Echocardiographic analysis of left ventricle ejection fraction (EF) and fractional shortening (FS) in mice 3 weeks after I/R injury (n=7, 6, 10, 8). G, qRT-PCR to determine expression levels of ETS1 in mice heart tissues 3 weeks after I/R injury (n=7, 6, 10, 8). H, Masson’s trichrome staining for the assessment of cardiac fibrosis 3 weeks after I/R injury (n=6). Scale bar=100 μm. I, Western blot showing levels of Hmbox1 and glucokinase (Gck) in mice heart tissues 3 weeks after I/R injury (n=6). Statistical analysis was performed by nonparametric Kruskal-Wallis test with the original false discovery rate method of Benjamini and Hochberg for A, by unpaired Student t test for B through E, and by 2-way ANOVA test followed by Tukey post hoc test for F through I. *P<0.05; **P<0.01; ***P<0.001.
ETS1 is known to play pivotal roles in cardiac development and myocardial survival.36–38 However, its function in cardiac I/R injury in vivo remained unclear. Here, we introduced the myocardial injection of ETS1-expressing (OE-ETS1) lentivirus and subsequently established a cardiac I/R injury model in mice. We observed that ETS1 overexpression reduced infarct size and preserved cardiac function after I/R injury (Figure 7E and 7F; Table S7). Efficacy validation revealed that the OE-ETS1 lentivirus increased cardiac ETS1 expression while suppressing Hmbox1 mRNA expression in heart tissues with I/R injury (Figure 7G; Figure S20A). Mice overexpressing ETS1 exhibited diminished cardiac remodeling and reduced fibrosis- and pathological hypertrophy–associated gene expressions (Figure 7H; Figure S20B). Moreover, ETS1 overexpression resulted in reduced Hmbox1 and elevated Gck protein levels, and enhanced Akt phosphorylation levels in I/R-injured mouse hearts (Figure 7I; Figure S20C). Collectively, these findings reveal that ETS1 negatively regulates Hmbox1 in the heart. Overexpression of ETS1 effectively downregulates Hmbox1 and activates Gck, providing protection against cardiac I/R injury.

AAV9-Mediated Hmbox1 Knockdown Has Therapeutic Effects Against Cardiac I/R Injury

Considering that therapeutic approaches are mostly applied after the onset of cardiac I/R injury in clinical practice, we further investigated the therapeutic efficacy of AAV9-mediated Hmbox1 knockdown. Mice were injected with either sh-Hmbox1-AAV9 or CTL-AAV9 within 30 minutes after myocardial reperfusion (Figure 8A). After validating Hmbox1 knockdown in heart tissues (Figure 8B and 8G), we demonstrated that the sh-Hmbox1-AAV9 treatment effectively preserved cardiac function and mitigated cardiac remodeling 3 weeks after I/R injury (Figure 8C through 8F; Table S8), whereas cardiac diastolic function was unaltered in the model (Table S9). Mice treated with sh-Hmbox1-AAV9 exhibited decreased expression of apoptosis-related proteins (Figure S21A) and increased Gck expression and Akt phosphorylation in heart tissues (Figure 8G; Figure S21B and S21C). Collectively, these data suggest that AAV9-mediated Hmbox1 reduction remains a viable therapeutic strategy for protection against cardiac I/R injury.
Figure 8. Adeno-associated virus (AAV9) therapy–mediated Homeobox-containing 1 (Hmbox1) knockdown mitigates cardiac ischemia/reperfusion (I/R) injury. A, A schematic representation demonstrating that mice received either sh-Hmbox1AAV9 or control AAV9 injections within 30 minutes after myocardial reperfusion. Heart tissues were collected 3 weeks after cardiac I/R injury. B, Quantitative reverse-transcription polymerase chain reaction (qRT-PCR) measuring Hmbox1 expression levels in mice heart tissues 3 weeks after I/R injury (n=8). C, Echocardiography assessing left ventricle ejection fraction (EF) and fractional shortening (FS) in mice 3 weeks after I/R injury (n=8, 10, 10, 10). D, Masson’s trichrome staining was used to evaluate cardiac fibrosis 3 weeks after I/R injury (n=7, 10, 9, 8). Scale bar=100 μm. E and F, qRT-PCR analysis of fibrosis marker genes (E) and cardiac remodeling marker genes (F) in mice heart tissues 3 weeks after I/R injury (n=8). G, Western blots showcasing Hmbox1 and glucokinase (Gck) in mice heart tissues 3 weeks after I/R injury (n=6). H, Working model illustrating the role of Hmbox1 in exercise-induced physiological cardiac hypertrophy and the protective effect of Hmbox1 inhibition against myocardial injury. Statistical analysis was performed by 2-way ANOVA test followed by Tukey post hoc test for B through E, F (Anp [atrial natriuretic peptide] and Bnp [brain natriuretic peptide]), and G, and by robust 2-way ANOVA test followed by post hoc pairwise Median Test for F (β-MHC [β-myosin heavy chain]). *P<0.05; **P<0.01; ***P<0.001.

DISCUSSION

Previous studies have indicated that Hmbox1 is downregulated upon exercise training and is targeted by miR-222, an exercise-responsive miRNA that exerts protective effects against ischemic myocardial injury.29,39,40 However, the role of Hmbox1, a known transcriptional repressor, in exercise-induced physiological cardiac growth, and its potential regulation of myocardial injury remained unknown. In this study, we have for the first time revealed that the exercise-induced downregulation of Hmbox1 is a crucial step for the induction of exercise-triggered physiological cardiac hypertrophy. Conversely, Hmbox1 levels increase during cardiac I/R injury. Inhibiting Hmbox1 activates Gck, promoting cardiomyocyte survival and enhancing glucose metabolism. These mechanisms offer protection against cardiac I/R injury. ETS1 is an upstream negative regulator of Hmbox1 transcription, and its overexpression is protective against cardiac I/R injury (Figure 8H).
Exercise-induced physiological cardiac growth is attributed to both physiological hypertrophy of the myocardium and the increased proliferative activity of cardiomyocytes.41 Here our results showed that Hmbox1 downregulation was necessary for exercise-induced cardiomyocyte hypertrophy but did not affect cardiomyocyte proliferation. Mechanistic analysis further revealed that Hmbox1 inhibition promoted cardiomyocyte hypertrophy by activating Akt and transcriptionally upregulating Gck. Hmbox1 has previously been linked to cell differentiation, immunomodulation, and cancer development.42 To the best of our knowledge, our study is the first to elucidate the functional role of Hmbox1 in the exercised heart, revealing that its downregulation is essential for exercise-induced physiological cardiac hypertrophy.
Regulators of exercise-induced physiological cardiac growth have potential therapeutic implications for myocardial protection.6,43 We were also keen to understand whether targeting Hmbox1 could shield against myocardial injury. Hmbox1 expression was evident in the postinfarct myocardium of patients with MI, as well as in OGDR-stressed cardiomyocytes and mouse I/R heart tissues, which contrasts with its downregulation during exercise. Moreover, knockdown of Hmbox1 significantly inhibited cardiomyocyte apoptosis under OGDR stress, which was attributed to the activation of the Akt/mTOR/P70S6K signaling pathway. In experimental models of cardiac I/R injury in mice, we showed that in vivo inhibition of Hmbox1 through AAV9 injections conferred protection against myocardial apoptosis and cardiac I/R injury during both acute and chronic phases. Moreover, myocardium-specific ablation of Hmbox1 was effective to protect the heart from I/R injury. These results provide compelling evidence that targeting myocardium Hmbox1 is exceptionally effective in preventing cardiac I/R injury and heart failure.
Given the promising functional effects of reducing Hmbox1 for myocardium protection, we further explored its translational potential for future clinical applications. Consistent with our observations of increased Hmbox1 expression in human postinfarct cardiac tissues, Hmbox1 was similarly upregulated in human ESC-derived cardiomyocytes upon OGDR stress, whereas knockdown of Hmbox1 could also shield the human myocardium from apoptosis. We further showed that therapeutic knockdown of Hmbox1 after the onset of myocardial reperfusion could also protect the mouse heart from I/R injury. However, the role of Hmbox1 in larger animal models of myocardial injury warrants further research.
Although Hmbox1 is recognized as a transcriptional repressor, the molecular mechanisms underlying its role in myocardial protection were unclear. Here we identified Gck as a potential downstream target that could be transcriptionally regulated by Hmbox1. Inhibition of Hmbox1 reduced cardiomyocyte apoptosis through activating Gck. The Gck gene encodes glucokinase, also known as hexokinase-IV, an enzyme that catalyzes glucose phosphorylation to form glucose-6-phosphate. Fatty acid and glucose are the primary metabolic substrates used for cardiac ATP production. Although fatty acid oxidative phosphorylation contributes to 40% to 70% of cardiac ATP production, glucose oxidative phosphorylation adds another 20% to 30%.44 However, ischemic or overload heart stress typically instigates metabolic remodeling, shifting the energy substrate preference from fatty acid oxidation to glycolysis, the initial step toward glucose oxidation and ATP production.45 As a result, although cardiac ATP production is significantly reduced under pathological stress, the switch to glucose oxidation can be initially beneficial for cardiac energy use given the scarce oxygen availability.46 Based on the observation that Hmbox1 inhibition could upregulate Gck in cardiomyocytes, we further probed the combined regulatory effects of Hmbox1 and Gck on mitochondrial respiration and glycolysis. We found that OGDR stress significantly compromised mitochondrial respiration and ATP production in isolated cardiomyocytes. It also enhanced glycolysis, which is consistent with previous studies reporting increased glycolysis of cardiomyocytes upon ischemic or overload stress.47,48 Promoting glycolysis and mitochondrial respiration have been reported to be beneficial for myocardial protection.47,49 Interestingly, reducing Hmbox1 improved mitochondrial respiration and further promoted glycolysis in cardiomyocytes. However, these metabolic improvements were abolished by Gck knockdown. Moreover, Hmbox1 reduction effectively upregulated Gck in both AAV9-mediated Hmbox1 knockdown and cardiac myocyte–specific Hmbox1 ablation mice in vivo. Collectively, these data emphasize that Hmbox1 inhibition enhances mitochondrial respiration and glycolysis in cardiac I/R injury by activating Gck. To the best of our knowledge, this is the first study to highlight the metabolic regulatory effect of Hmbox1 reduction in myocardial protection.
To elucidate how Hmbox1 might be regulated in the heart, we used bioinformatic analysis and quantitative polymerase chain reaction to identify potential upstream transcriptional regulators of Hmbox1 across various experimental models. We demonstrated that ETS1 was a transcriptional negative regulator of Hmbox1, influencing cardiomyocyte size and apoptosis. Compared with miR-222, which has previously been reported to target Hmbox1 by binding to the 3’ untranslated region of Hmbox1, in the present study we identify ETS1 acting as a transcription factor that negatively regulates Hmbox1 expression. MiR-222 has been known as an exercise-responsive miRNA, which contributes to exercise-induced physiological cardiac adaptive growth through promoting both physiological hypertrophy and proliferation of cardiomyocytes. In addition, increasing miR-222 is protective to mitigate myocardial apoptosis and cardiac I/R injury.29 Hmbox1 is a target gene of miR-222, but it does not regulate cardiomyocyte proliferation, which is different from miR-222. Meanwhile, inhibiting Hmbox1 promotes cardiomyocyte survival and glucose metabolism through Gck activation in I/R injury, which adds novel insights to the exercise-responsive molecules in regulating myocardial glycolysis metabolism. Thus, although both miR-222 and ETS1 can be induced upon exercise and inhibit Hmbox1 expression in the heart, their regulatory mode on Hmbox1 expression and their functions in cardiomyocytes could be different. Moreover, functions of molecules that are regulated by exercise can be different in cardiomyocytes, which deserve to be analyzed differently.
ETS1 is known to directly regulate cardiac genes, promoting early cardiomyocyte development and regulating vascular remodeling.36,50,51 Emerging evidence indicates that increased ETS1 expression can promote cardiomyocyte survival and reduce apoptosis.37,38 However, the potential protective role of ETS1 overexpression in cardiac I/R injury and its influence on Hmbox1 regulation was previously uncertain. Here we further revealed that ETS1 overexpression was protective against cardiac I/R injury. Moreover, ETS1 overexpression sufficiently downregulated Hmbox1, accompanied by Gck upregulation and Akt phosphorylation. These results unravel ETS1 as a paramount negative regulator of Hmbox1, signifying a potential target for myocardial protection against cardiac I/R injury. It is plausible that additional upstream regulators of Hmbox1 will emerge in future investigations.
In conclusion, we have identified Hmbox1 as a novel transcriptional repressor engaged in both physiological and pathological cardiac alterations. Reducing Hmbox1 is pivotal for exercise-induced physiological cardiac hypertrophy and offers protection against cardiac I/R injury and heart failure. Our research also unveils a novel role of Hmbox1 reduction in modulating cardiomyocyte mitochondrial respiration and glycolysis through Gck activation. Targeting Hmbox1 and its associated pathway holds promise for enhancing myocardial survival and glucose metabolism in cardiac I/R injury. The therapeutic potential of this strategy warrants in-depth exploration in larger animal models and across other cardiovascular diseases.

ARTICLE INFORMATION

Supplemental Material

Supplemental Methods
Figures S1–S21
Tables S1–S10
References 52–54

Footnote

Nonstandard Abbreviations and Acronyms

α-MHC
α-myosin heavy chain
α-SMA
α-smooth muscle actin
β-MHC
β-myosin heavy chain
AAV9
adeno-associated virus 9
Akt
protein kinase B
Anp
atrial natriuretic peptide
Bnp
brain natriuretic peptide
cKO
conditional knockout
cTnT
cardiac troponin T
EdU
5-ethynyl-2’-deoxyuridine
Gck
glucokinase
Hmbox1
homeobox-containing 1
IGF-1
insulin-like growth factor 1
I/R
ischemia/reperfusion
MI
myocardial infarction
miR-222
microRNA-222
mTOR
mammalian target of rapamycin
NRCM
neonatal rat cardiomyocyte
OGDR
oxygen glucose deprivation and reperfusion
P70S6K
70 kDa ribosomal protein S6 kinase
RNA-seq
RNA sequencing
Tunel
terminal deoxynucleotidyl transferase mediated dUTP nick-end labeling

Supplemental Material

File (circ_circulationaha-2023-067592_supp1.pdf)
File (circ_circulationaha-2023-067592_supp2.pdf)
File (supplemental materials 2024.5.5.pdf)

REFERENCES

1.
Tsao CW, Aday AW, Almarzooq ZI, Alonso A, Beaton AZ, Bittencourt MS, Boehme AK, Buxton AE, Carson AP, Commodore-Mensah Y, et al. Heart disease and stroke statistics-2022 update: a report from the American Heart Association. Circulation. 2022;145:e153–e639. doi: 10.1161/CIR.0000000000001052
2.
Ibanez B, Heusch G, Ovize M, Van de Werf F. Evolving therapies for myocardial ischemia/reperfusion injury. J Am Coll Cardiol. 2015;65:1454–1471. doi: 10.1016/j.jacc.2015.02.032
3.
Heusch G, Gersh BJ. The pathophysiology of acute myocardial infarction and strategies of protection beyond reperfusion: a continual challenge. Eur Heart J. 2017;38:774–784. doi: 10.1093/eurheartj/ehw224
4.
Oerlemans MI, Koudstaal S, Chamuleau SA, de Kleijn DP, Doevendans PA, Sluijter JP. Targeting cell death in the reperfused heart: pharmacological approaches for cardioprotection. Int J Cardiol. 2013;165:410–422. doi: 10.1016/j.ijcard.2012.03.055
5.
Wang L, Yu P, Wang J, Xu G, Wang T, Feng J, Bei Y, Xu J, Wang H, Das S, et al. Downregulation of circ-ZNF609 promotes heart repair by modulating RNA N(6)-methyladenosine-modified Yap expression. Research (Wash D C). 2022;2022:9825916. doi: 10.34133/2022/9825916
6.
Chen H, Chen C, Spanos M, Li G, Lu R, Bei Y, Xiao J. Exercise training maintains cardiovascular health: signaling pathways involved and potential therapeutics. Signal Transduct Target Ther. 2022;7:306. doi: 10.1038/s41392-022-01153-1
7.
Wang H, Xie Y, Guan L, Elkin K, Xiao J. Targets identified from exercised heart: killing multiple birds with one stone. NPJ Regen Med. 2021;6:23. doi: 10.1038/s41536-021-00128-0
8.
Bei Y, Lu D, Bar C, Chatterjee S, Costa A, Riedel I, Mooren FC, Zhu Y, Huang Z, Wei M, et al. miR-486 attenuates cardiac ischemia/reperfusion injury and mediates the beneficial effect of exercise for myocardial protection. Mol Ther. 2022;30:1675–1691. doi: 10.1016/j.ymthe.2022.01.031
9.
Gao R, Wang L, Bei Y, Wu X, Wang J, Zhou Q, Tao L, Das S, Li X, Xiao J. Long noncoding RNA cardiac physiological hypertrophy-associated regulator induces cardiac physiological hypertrophy and promotes functional recovery after myocardial ischemia-reperfusion injury. Circulation. 2021;144:303–317. doi: 10.1161/CIRCULATIONAHA.120.050446
10.
Feng N, Yu H, Wang Y, Zhang Y, Xiao H, Gao W. Exercise training attenuates angiotensin II-induced cardiac fibrosis by reducing POU2F1 expression. J Sport Health Sci. 2023;12:464–476. doi: 10.1016/j.jshs.2022.10.004
11.
Luo Z, Zhang T, Chen S. Exercise prescription: pioneering the “third pole” for clinical health management. Research (Wash D C). 2023;6:0284. doi: 10.34133/research.0284
12.
Chen S, Saiyin H, Zeng X, Xi J, Liu X, Li X, Yu L. Isolation and functional analysis of human HMBOX1, a homeobox containing protein with transcriptional repressor activity. Cytogenet Genome Res. 2006;114:131–136. doi: 10.1159/000093328
13.
Zhang M, Chen S, Li Q, Ling Y, Zhang J, Yu L. Characterization of a novel human HMBOX1 splicing variant lacking the homeodomain and with attenuated transcription repressor activity. Mol Biol Rep. 2010;37:2767–2772. doi: 10.1007/s11033-009-9815-9
14.
Dejardin J, Kingston RE. Purification of proteins associated with specific genomic loci. Cell. 2009;136:175–186. doi: 10.1016/j.cell.2008.11.045
15.
Kappei D, Butter F, Benda C, Scheibe M, Draskovic I, Stevense M, Novo CL, Basquin C, Araki M, Araki K, et al. HOT1 is a mammalian direct telomere repeat-binding protein contributing to telomerase recruitment. EMBO J. 2013;32:1681–1701. doi: 10.1038/emboj.2013.105
16.
Feng X, Luo Z, Jiang S, Li F, Han X, Hu Y, Wang D, Zhao Y, Ma W, Liu D, et al. The telomere-associated homeobox-containing protein TAH1/HMBOX1 participates in telomere maintenance in ALT cells. J Cell Sci. 2013;126:3982–3989. doi: 10.1242/jcs.128512
17.
Lee JH, Hong J, Zhang Z, de la Pena Avalos B, Proietti CJ, Deamicis AR, Guzman GP, Lam HM, Garcia J, Roudier MP, et al. Regulation of telomere homeostasis and genomic stability in cancer by N (6)-adenosine methylation (m(6)A). Sci Adv. 2021;7:eabg7073. doi: 10.1126/sciadv.abg7073
18.
Su L, Zhao H, Sun C, Zhao B, Zhao J, Zhang S, Su H, Miao J. Role of Hmbox1 in endothelial differentiation of bone-marrow stromal cells by a small molecule. ACS Chem Biol. 2010;5:1035–1043. doi: 10.1021/cb100153r
19.
Han L, Shao J, Su L, Gao J, Wang S, Zhang Y, Zhang S, Zhao B, Miao J. A chemical small molecule induces mouse embryonic stem cell differentiation into functional vascular endothelial cells via Hmbox1. Stem Cells Dev. 2012;21:2762–2769. doi: 10.1089/scd.2012.0055
20.
Ma H, Su L, Yue H, Yin X, Zhao J, Zhang S, Kung H, Xu Z, Miao J. HMBOX1 interacts with MT2A to regulate autophagy and apoptosis in vascular endothelial cells. Sci Rep. 2015;5:15121. doi: 10.1038/srep15121
21.
Yu YL, Diao NN, Li YZ, Meng XH, Jiao WL, Feng JB, Liu ZP, Lu N. Low expression level of HMBOX1 in high-grade serous ovarian cancer accelerates cell proliferation by inhibiting cell apoptosis. Biochem Biophys Res Commun. 2018;501:380–386. doi: 10.1016/j.bbrc.2018.04.203
22.
Ma H, Su L, He X, Miao J. Loss of HMBOX1 promotes LPS-induced apoptosis and inhibits LPS-induced autophagy of vascular endothelial cells in mouse. Apoptosis. 2019;24:946–957. doi: 10.1007/s10495-019-01572-6
23.
Dai J, Zhang C, Tian Z, Zhang J. Expression profile of HMBOX1, a novel transcription factor, in human cancers using highly specific monoclonal antibodies. Exp Ther Med. 2011;2:487–490. doi: 10.3892/etm.2011.240
24.
Zhang P, Liu Q, Yan S, Yuan G, Shen J, Li G. Homeoboxcontaining protein 1 loss is associated with clinicopathological performance in glioma. Mol Med Rep. 2017;16:4101–4106. doi: 10.3892/mmr.2017.7050
25.
Chen S, Li Y, Zhi S, Ding Z, Wang W, Peng Y, Huang Y, Zheng R, Yu H, Wang J, et al. WTAP promotes osteosarcoma tumorigenesis by repressing HMBOX1 expression in an m(6)A-dependent manner. Cell Death Dis. 2020;11:659. doi: 10.1038/s41419-020-02847-6
26.
Zhou C, Wei W, Ma J, Yang Y, Liang L, Zhang Y, Wang Z, Chen X, Huang L, Wang W, et al. Cancer-secreted exosomal miR-1468-5p promotes tumor immune escape via the immunosuppressive reprogramming of lymphatic vessels. Mol Ther. 2021;29:1512–1528. doi: 10.1016/j.ymthe.2020.12.034
27.
Wu L, Zhang C, Zhang J. HMBOX1 negatively regulates NK cell functions by suppressing the NKG2D/DAP10 signaling pathway. Cell Mol Immunol. 2011;8:433–440. doi: 10.1038/cmi.2011.20
28.
Wu L, Zhang C, Zheng X, Tian Z, Zhang J. HMBOX1, homeobox transcription factor, negatively regulates interferon-gamma production in natural killer cells. Int Immunopharmacol. 2011;11:1895–1900. doi: 10.1016/j.intimp.2011.07.021
29.
Liu X, Xiao J, Zhu H, Wei X, Platt C, Damilano F, Xiao C, Bezzerides V, Bostrom P, Che L, et al. miR-222 is necessary for exercise-induced cardiac growth and protects against pathological cardiac remodeling. Cell Metab. 2015;21:584–595. doi: 10.1016/j.cmet.2015.02.014
30.
Weeks KL, Bernardo BC, Ooi JYY, Patterson NL, McMullen JR. The IGF1-PI3K-Akt signaling pathway in mediating exercise-induced cardiac hypertrophy and protection. Adv Exp Med Biol. 2017;1000:187–210. doi: 10.1007/978-981-10-4304-8_12
31.
Sanchez-Aguilera P, Lopez-Crisosto C, Norambuena-Soto I, Penannen C, Zhu J, Bomer N, Hoes MF, Van Der Meer P, Chiong M, Westenbrink BD, et al. IGF-1 boosts mitochondrial function by a Ca(2+) uptake-dependent mechanism in cultured human and rat cardiomyocytes. Front Physiol. 2023;14:1106662. doi: 10.3389/fphys.2023.1106662
32.
Bass-Stringer S, Tai CMK, McMullen JR. IGF1-PI3K-induced physiological cardiac hypertrophy: implications for new heart failure therapies, biomarkers, and predicting cardiotoxicity. J Sport Health Sci. 2021;10:637–647. doi: 10.1016/j.jshs.2020.11.009
33.
Gersing S, Cagiada M, Gebbia M, Gjesing AP, Cote AG, Seesankar G, Li R, Tabet D, Weile J, Stein A, et al. A comprehensive map of human glucokinase variant activity. Genome Biol. 2023;24:97. doi: 10.1186/s13059-023-02935-8
34.
Tachibana S, Yu NK, Li R, Fernandez-Costa C, Liang A, Choi J, Jung D, Xiao C, Kralli A, Yates JR, et al. Perm1 protects the heart from pressure overload-induced dysfunction by promoting oxidative metabolism. Circulation. 2023;147:916–919. doi: 10.1161/CIRCULATIONAHA.122.060173
35.
Song R, Dasgupta C, Mulder C, Zhang L. MicroRNA-210 controls mitochondrial metabolism and protects heart function in myocardial infarction. Circulation. 2022;145:1140–1153. doi: 10.1161/CIRCULATIONAHA.121.056929
36.
Ruan H, Liao Y, Ren Z, Mao L, Yao F, Yu P, Ye Y, Zhang Z, Li S, Xu H, et al. Single-cell reconstruction of differentiation trajectory reveals a critical role of ETS1 in human cardiac lineage commitment. BMC Biol. 2019;17:89. doi: 10.1186/s12915-019-0709-6
37.
Guo X, Zhang BF, Zhang J, Liu G, Hu Q, Chen J. The histone demthylase KDM3A protects the myocardium from ischemia/reperfusion injury via promotion of ETS1 expression. Commun Biol. 2022;5:270. doi: 10.1038/s42003-022-03225-y
38.
Wang WK, Lu QH, Zhang JN, Wang B, Liu XJ, An FS, Qin WD, Chen XY, Dong WQ, Zhang C, et al. HMGB1 mediates hyperglycaemia-induced cardiomyocyte apoptosis via ERK/Ets-1 signalling pathway. J Cell Mol Med. 2014;18:2311–2320. doi: 10.1111/jcmm.12399
39.
Zhou Q, Deng J, Yao J, Song J, Meng D, Zhu Y, Xu M, Liang Y, Xu J, Sluijter JP, et al. Exercise downregulates HIPK2 and HIPK2 inhibition protects against myocardial infarction. EBioMedicine. 2021;74:103713. doi: 10.1016/j.ebiom.2021.103713
40.
Vujic A, Lerchenmuller C, Wu TD, Guillermier C, Rabolli CP, Gonzalez E, Senyo SE, Liu X, Guerquin-Kern JL, Steinhauser ML, et al. Exercise induces new cardiomyocyte generation in the adult mammalian heart. Nat Commun. 2018;9:1659. doi: 10.1038/s41467-018-04083-1
41.
Bernardo BC, Ooi JYY, Weeks KL, Patterson NL, McMullen JR. Understanding key mechanisms of exercise-induced cardiac protection to mitigate disease: current knowledge and emerging concepts. Physiol Rev. 2018;98:419–475. doi: 10.1152/physrev.00043.2016
42.
Jiang Y, Mu H, Zhao H. HMBOX1, a member of the homeobox family: current research progress. Cent Eur J Immunol. 2023;48:63–69. doi: 10.5114/ceji.2023.126615
43.
Xiao J, Rosenzweig A. Exercise and cardiovascular protection: update and future. J Sport Health Sci. 2021;10:607–608. doi: 10.1016/j.jshs.2021.11.001
44.
Lopaschuk GD, Jaswal JS. Energy metabolic phenotype of the cardiomyocyte during development, differentiation, and postnatal maturation. J Cardiovasc Pharmacol. 2010;56:130–140. doi: 10.1097/FJC.0b013e3181e74a14
45.
Neubauer S. The failing heart--an engine out of fuel. N Engl J Med. 2007;356:1140–1151. doi: 10.1056/NEJMra063052
46.
Ingwall JS. Energy metabolism in heart failure and remodelling. Cardiovasc Res. 2009;81:412–419. doi: 10.1093/cvr/cvn301
47.
Zhang B, Jiang H, Wu J, Cai Y, Dong Z, Zhao Y, Hu Q, Hu K, Sun A, Ge J. m6A demethylase FTO attenuates cardiac dysfunction by regulating glucose uptake and glycolysis in mice with pressure overload-induced heart failure. Signal Transduct Target Ther. 2021;6:377. doi: 10.1038/s41392-021-00699-w
48.
Tian H, Zhao X, Zhang Y, Xia Z. Abnormalities of glucose and lipid metabolism in myocardial ischemia-reperfusion injury. Biomed Pharmacother. 2023;163:114827. doi: 10.1016/j.biopha.2023.114827
49.
Jiang H, Jia D, Zhang B, Yang W, Dong Z, Sun X, Cui X, Ma L, Wu J, Hu K, et al. Exercise improves cardiac function and glucose metabolism in mice with experimental myocardial infarction through inhibiting HDAC4 and upregulating GLUT1 expression. Basic Res Cardiol. 2020;115:28. doi: 10.1007/s00395-020-0787-1
50.
Zhou P, Zhang Y, Sethi I, Ye L, Trembley MA, Cao Y, Akerberg BN, Xiao F, Zhang X, Li K, et al. GATA4 regulates developing endocardium through interaction with ETS1. Circ Res. 2022;131:e152–e168. doi: 10.1161/CIRCRESAHA.120.318102
51.
Wang L, Lin L, Qi H, Chen J, Grossfeld P. Endothelial loss of ETS1 impairs coronary vascular development and leads to ventricular non-compaction. Circ Res. 2022;131:371–387. doi: 10.1161/CIRCRESAHA.121.319955
52.
Jenkins CP, Cardona DM, Bowers JN, Oliai BR, Allan RW, Normann SJ. The utility of C4d, C9, and troponin T immunohistochemistry in acute myocardial infarction. Arch Pathol Lab Med. 2010;134:256–263. doi: 10.5858/134.2.256
53.
Bei Y, Zhu Y, Wei M, Yin M, Li L, Chen C, Huang Z, Liang X, Gao J, Yao J, et al. HIPK1 inhibition protects against pathological cardiac hypertrophy by inhibiting the CREB-C/EBPbeta axis. Adv Sci (Weinh). 2023;10:e2300585. doi: 10.1002/advs.202300585
54.
Viereck J, Kumarswamy R, Foinquinos A, Xiao K, Avramopoulos P, Kunz M, Dittrich M, Maetzig T, Zimmer K, Remke J, et al. Long noncoding RNA Chast promotes cardiac remodeling. Sci Transl Med. 2016;8:326ra–32322. doi: 10.1126/scitranslmed.aaf1475

eLetters(0)

eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.

Information & Authors

Information

Published In

Go to Circulation
Go to Circulation

Versions

You are viewing the most recent version of this article.

History

Received: 14 October 2023
Accepted: 11 April 2024
Published online: 6 May 2024
Published in print: 10 September 2024

Permissions

Request permissions for this article.

Keywords

  1. apoptosis
  2. cardiac ischemia/reperfusion injury
  3. exercise
  4. Gck
  5. glycolysis
  6. Hmbox1

Subjects

Authors

Affiliations

Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People’s Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.).
Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China.
Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China.
Yujiao Zhu, PhD*
Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People’s Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.).
Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China.
Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China.
Jingwen Zhou, BS
Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People’s Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.).
Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China.
Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China.
Songwei Ai, BS
Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People’s Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.).
Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China.
Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China.
Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, China (J.Y.).
Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People’s Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.).
Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China.
Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China.
Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People’s Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.).
Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China.
Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China.
Weitong Qi, MS
Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People’s Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.).
Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China.
Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China.
Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston (M.S., G.L.).
Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People’s Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.).
Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China.
Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China.
Meng Wei, MS
Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People’s Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.).
Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China.
Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China.
Zhenzhen Huang, MS
Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People’s Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.).
Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China.
Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China.
Juan Gao, PhD
Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People’s Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.).
Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China.
Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China.
Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People’s Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.).
Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China.
Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China.
Petra H. van der Kraak, BS
Department of Pathology (P.H.v.d.K.), University Medical Center Utrecht, University Utrecht, The Netherlands.
Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston (M.S., G.L.).
Zhiyong Lei, PhD
Department of Cardiology, Laboratory of Experimental Cardiology (Z.L., J.P.G.S.), University Medical Center Utrecht, University Utrecht, The Netherlands.
Division Laboratory, Central Diagnosis Laboratory Research (Z.L.), University Medical Center Utrecht, University Utrecht, The Netherlands.
Joost P.G. Sluijter, PhD https://orcid.org/0000-0003-2088-9102
Department of Cardiology, Laboratory of Experimental Cardiology (Z.L., J.P.G.S.), University Medical Center Utrecht, University Utrecht, The Netherlands.
Utrecht Regenerative Medicine Center (J.P.G.S.), University Medical Center Utrecht, University Utrecht, The Netherlands.
Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People’s Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.).
Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China.
Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China.

Notes

*
Y. Bei and Y. Zhu contributed equally.
Supplemental Material is available at Supplemental Material.
For Sources of Funding and Disclosures, see page 865.
Circulation is available at www.ahajournals.org/journal/circ .
Correspondence to: Yihua Bei, PhD, Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Shanghai University, 333 Nan Chen Rd, Shanghai 200444, China, Email [email protected]
Junjie Xiao, MD, PhD, Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Shanghai University, 333 Nan Chen Rd, Shanghai 200444, China, Email [email protected]

Disclosures

None.

Sources of Funding

This work was supported by grants from the National Natural Science Foundation of China (82225005 and 82020108002 to J.X., 82170285 and 81970335 to Y.B., and 82170390 to J.G.), grants from the Science and Technology Commission of Shanghai Municipality (23410750100, 20DZ2255400, and 21XD1421300 to J.X., and 23010500300 to Y.B.), the “Dawn” Program of Shanghai Education Commission (19SG34 to J.X.), the Oriental Scholars of Shanghai Universities (TP2022057 to Y.B.), the Shanghai Rising-Star Program (19QA1403900 to Y.B.), the Natural Science Foundation of Shanghai (21ZR1422700 to J.G. and 20ZR1443300 to J.Y.), and the Natural Science Foundation of Tibet Autonomous Region (XZ2020ZR-ZY35[Z] and XZ202101ZR0003G to J.Y.).

Metrics & Citations

Metrics

Citations

Download Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Select your manager software from the list below and click Download.

  1. Associations between triglyceride-glucose index combined with waist circumference and heart failure in individuals with different body mass indices: a cross-sectional study using NHANES 2011–2020 data, Lipids in Health and Disease, 24, 1, (2025).https://doi.org/10.1186/s12944-025-02476-y
    Crossref
  2. A study on the molecular mechanism of cardiac protection of procarboxylpeptidase in MIRI rats based on the NLRP3 signaling pathway, Science Progress, 108, 1, (2025).https://doi.org/10.1177/00368504251322085
    Crossref
  3. Initial cardioplegic flush with crystalloid cardioplegia improves donor heart preservation and function via NR4A3 upregulation and metabolic reprogramming, Science Bulletin, (2025).https://doi.org/10.1016/j.scib.2025.03.019
    Crossref
  4. Swimming exercise induces redox-lipid crosstalk to ameliorate osteoarthritis progression, Redox Biology, 81, (103535), (2025).https://doi.org/10.1016/j.redox.2025.103535
    Crossref
  5. Salvianolic acid B drives gluconeogenesis and peroxisomal redox remodeling in cardiac ischemia/reperfusion injury: A metabolism regulation by metabolite signal crosstalk, Free Radical Biology and Medicine, 229, (399-414), (2025).https://doi.org/10.1016/j.freeradbiomed.2025.01.037
    Crossref
  6. Electrical stimulation promotes peripheral nerve regeneration by upregulating glycolysis and oxidative phosphorylation, Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease, 1871, 5, (167804), (2025).https://doi.org/10.1016/j.bbadis.2025.167804
    Crossref
  7. Regulated cell death in acute myocardial infarction: Molecular mechanisms and therapeutic implications, Ageing Research Reviews, 104, (102629), (2025).https://doi.org/10.1016/j.arr.2024.102629
    Crossref
  8. Cardiac Regeneration in Adult Zebrafish: A Review of Signaling and Metabolic Coordination, Current Cardiology Reports, 27, 1, (2025).https://doi.org/10.1007/s11886-024-02162-y
    Crossref
  9. Dexmedetomidine Ameliorates Myocardial Ischemia‐Reperfusion Injury by Inhibiting MDH2 Lactylation via Regulating Metabolic Reprogramming, Advanced Science, 11, 48, (2024).https://doi.org/10.1002/advs.202409499
    Crossref
  10. miR‐30d Attenuates Pulmonary Arterial Hypertension via Targeting MTDH and PDE5A and Modulates the Beneficial Effect of Sildenafil, Advanced Science, 11, 40, (2024).https://doi.org/10.1002/advs.202407712
    Crossref
Loading...

View Options

View options

PDF and All Supplements

Download PDF and All Supplements

PDF/EPUB

View PDF/EPUB
Login options

Check if you have access through your login credentials or your institution to get full access on this article.

Personal login Institutional Login
Purchase Options

Purchase this article to access the full text.

Purchase access to this article for 24 hours

Inhibition of Hmbox1 Promotes Cardiomyocyte Survival and Glucose Metabolism Through Gck Activation in Ischemia/Reperfusion Injury
Circulation
  • Vol. 150
  • No. 11

Purchase access to this journal for 24 hours

Circulation
  • Vol. 150
  • No. 11
Restore your content access

Enter your email address to restore your content access:

Note: This functionality works only for purchases done as a guest. If you already have an account, log in to access the content to which you are entitled.

Figures

Tables

Media

Share

Share

Share article link

Share

Comment Response