Open Access

Mechanistic exploration of hexokinase 2 and metabolism in diabetic cardiomyopathy

  • Authors:
    • Bo Li
    • Xu Zhao
    • Liming Ma
    • Xiaoying Wang
    • Yan Ding
    • Yi Zhang
  • View Affiliations

  • Published online on: May 26, 2025     https://doi.org/10.3892/mmr.2025.13576
  • Article Number: 211
  • Copyright: © Li et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The pathogenesis of diabetic cardiomyopathy (DCM) remains incompletely understood. The present study employed weighted gene co‑expression network analysis to analyze the DCM transcriptome dataset from the Gene Expression Omnibus (GEO) database to identify genes associated with this disease. Subsequently, both internal and external validation of the expression of the characterized genes was performed using additional GEO datasets. Key DCM genes were validated at both the in vitro and in vivo levels by Western blot and immunohistochemistry, IHC). Furthermore, the mechanisms of gene and metabolite co‑expression in DCM were investigated through transcriptome sequencing of cells overexpressing disease‑associated genes, combined with quantitative measurements of metabolites. Notably, hexokinase 2 (HK2) was downregulated in both DCM cell and db/db mouse models. Low expression of HK2 was implicated in the disruption of organic acids and their derivatives, as well as the receptor for advanced glycation end‑products pathway.

Introduction

Diabetes mellitus (DM) is a prevalent chronic disease worldwide, with type 2 diabetes (T2D) accounting for >90% of cases. The International Diabetes Federation estimates that by 2040, 624 million individuals worldwide will be affected by T2D (1). Diabetic cardiomyopathy (DCM), a severe cardiovascular complication of DM, accounts for 50–60% of DM-associated mortality, representing the leading cause of death in diabetic populations (2). DCM is associated with immune-inflammatory infiltration, dysregulation of glucolipid metabolism (2), mitochondrial energy metabolism disorder, oxidative stress (3), focal myocardial cell death (4) and programmed necrosis (5). Metabolic remodeling is a manifestation of DCM, characterized by the loss of cardiomyocyte capacity for carbohydrate and fatty acid metabolism, which is supplanted by mitochondrial fatty acid β-oxidation. This shift leads to cardiac steatosis, lipotoxicity and apoptosis of cardiomyocytes (6).

DCM is a pathophysiological condition associated with DM that manifests as cardiac decompensation or heart failure (3). It typically presents with thickening of the left ventricular wall, impaired left ventricular diastolic function (7) and heart failure resulting from non-coronary artery disease, hypertension and valvular heart disease. Decreased cardiomyocyte function is an important contributor to the development of heart failure in DCM (8), with early signs including abnormal diastolic relaxation (9). Chronic hyperglycemia (HG) and HG-induced oxidative stress lead to ultrastructural changes in cardiomyocytes, characterized by mitochondrial swelling, a decreased number of mitochondria and defective collagen fibers (10). These alterations may result in myocardial fibrosis, mitochondrial dysfunction, myocyte hypertrophy and myogenic fibrosis disorder, all of which disrupt the homeostasis of cardiomyocytes and may ultimately lead to heart failure (10).

Metabolic disorders induced by HG and insulin resistance are associated with the development of DCM (9). Protein glycosylation resulting from HG leads to an increase in advanced glycation end products (AGEs), which promotes collagen cross-linking, myocardial fibrosis and impaired diastolic function, triggering the onset of DCM (9). HG, insulin resistance, AGEs, collagen cross-linking, myocardial fibrosis and impaired diastolic function serve key roles in the progression of DCM (9). Emerging evidence highlights the potential of bariatric surgery, particularly Roux-en-Y gastric bypass and sleeve gastrectomy, in ameliorating DCM (11). Insulin resistance is a key pathogenic driver of DCM, leading to impaired myocardial insulin signaling, mitochondrial dysfunction and endoplasmic reticulum (ER) stress. In insulin resistance or hyperinsulinemia, the heart experiences notable metabolic dysregulation, characterized by diminished glucose uptake and use alongside heightened dependence on fatty acid oxidation. These metabolic shifts exacerbate cardiac dysfunction by fostering oxidative stress, chronic inflammation and cell injury, collectively contributing to the progression of DCM (12). The energy and lipid metabolism associated with DCM, along with its regulatory mechanisms, are complex and not fully understood. Therefore, it is important to investigate the pathogenesis of DCM through a combined analysis of genomics and metabolomics, as the underlying mechanisms of DCM at the metabolic level.

Materials and methods

Ethics approval

All experimental procedures involving db/db and db/m mice were conducted in accordance with the ARRIVE guidelines and were approved by the Institutional Animal Care and Use Committee of Hubei University of Medicine (Shiyan, China; (approval no. 2024S071). Humane endpoints were as follows: i) Body weight loss ≥20% within 48 h (relative to peak body weight); ii) severe hypoglycemia (<40 mg/dl) or hyperglycemia (>600 mg/dl) refractory to clinical stabilization (for example, dietary adjustment or insulin administration); iii) functional impairment (inability to access food/water independently or lethargy persisting >24 h) and iv) overt signs of distress (labored breathing, hunched posture lasting >12 h or ulcerative lesions affecting >5% body surface area). No animals met criteria for early euthanasia. Animals were humanely euthanized via intraperitoneal injection of sodium pentobarbital at a dose of 150 mg/kg body weight. Animal death was confirmed by the absence of heartbeat, breathing and corneal reflex. These methods align with the American Veterinary Medical Association guidelines to ensure minimal animal suffering (13).

Data source

Key genes associated with DCM were investigated using The National Center for Biotechnology Information Gene Expression Omnibus (GEO) database (ncbi.nlm.nih.gov/geo/). A total of three rat transcriptome expression profiling datasets (accession nos. GSE4745, GSE5606 and GSE6880) were retrieved from the GEO database (Table I). To eliminate batch effects and other sources of variance, surrogate variable analysis (14) and limma packages (version 3.54.2, http://bioconductor.org/packages/release/bioc/html/limma.html) were employed in R (version 4.2.3; http://cran.r-project.org/bin/windows/base/old/4.2.3/) to merge the GSE4745 and GSE6880 datasets, and the resulting merged dataset served as the internal training set. For independent validation, GSE5606 dataset as an external validation cohort. This strategic approach of combining GSE4745 and GSE6880 not only increased statistical power but also maintained platform diversity within the training set. Importantly, all three datasets investigated identical species and disease models, ensuring biological consistency while allowing rigorous assessment of model generalizability across different experimental platforms.

Table I.

Gene Expression Omnibus datasets of mRNA expression in rat heart tissue.

Table I.

Gene Expression Omnibus datasets of mRNA expression in rat heart tissue.

Accession no.PlatformControl, nDiabetic cardiomyopathy, n
GSE4745GPL851212
GSE5606GPL135567
GSE6880GPL34133
Screening for differentially expressed genes (DEGs)

GEO datasets were corrected using the limma package. The expression data files of the combined dataset were investigated for deviation analysis on the basis of |log2 (fold-change)|≥1 and adjusted P-value <0.05. DEGs were visualized using heatmaps generated by the heatmap package (version 1.0.12; cran.r-project.org/web/packages/pheatmap/index.html) and volcano plots created with the ggplot2 package (version 3.5.1; http://ggplot2.tidyverse.org/).

Functional enrichment analysis

Gene Ontology (GO) (15) and Kyoto Encyclopedia of Genes and Genomes (KEGG) (16) pathway enrichment analyses were performed using the clusterProfiler R package (version 4.7.1; bioconductor.org/packages/release/bioc/html/clusterProfiler.html). GO enrichment analysis provides a comprehensive description of the gene and protein functions associated with biological processes (BPs), molecular functions and cellular components. Adjusted P<0.05 was considered to indicate a statistically significant difference. For the Gene Set Enrichment Analysis (GSEA), the clusterProfiler package (version 4.7.1; bioconductor.org/packages/release/bioc/html/clusterProfiler.html) was utilized with adjusted P<0.05 to indicate statistically significant enrichment. The results of enrichment analyses were visualized using the ggplot2 package (version 3.5.1; ggplot2.tidyverse.org/), and chord plots were generated using the Xian Tao Academic Online Analysis platform (xiantaozi.com/products/apply/c0b6febb-52dd-4525-970a-61bbe9e263ff/analyse/f9496965-0e1a-4249-b123-e6a00c924021).

Weighted gene co-expression network analysis (WGCNA)-mediated core gene screening

WGCNA is a bioinformatics tool that calculates gene expression levels and clusters genes with similar expression patterns into modules. These functional gene clusters are used to explore interactions between genes and biological phenotypes within the modules (17). The present study employed the WGCNA package (version 1.72; cran.r-project.org/web/packages/WGCNA/index.html) to identify gene modules with similar expression patterns by constructing a gene co-expression network using the mean chained hierarchical clustering algorithm. These modules were further distinguished through hierarchical clustering based on proximity. Additionally, the association of each module was inferred by calculating the association between different modules and biological phenotype. Finally, core genes from key modules were identified. Diagnostic potential was evaluated by ROC curve analysis (AUC >0.7), with differential expression visualized through boxplots (Wilcoxon test, P<0.05).

Least absolute shrinkage and selection operator (LASSO) regression screening for key DCM genes

Expression of DEGs intersecting with modal genes significantly associated with DCM, as identified through WGCNA, were analyzed. Intersection analysis was performed using the Venn package (version 1.12; cran.r-project.org/web/packages/venn/index.html) The GLMNET package (version 4.1; cran.r-project.org/web/packages/glmnet/index.html) was employed to construct a LASSO regression model for the intersected genes and DCM feature genes were identified through cross-validation. The genes corresponding to the points with the lowest cross-validation error were designated key genes associated with DCM.

Validation of key genes

To analyze the expression of key genes in both internal and external datasets, the Limma package was used. P<0.05 was considered to indicate a statistically significant difference. Additionally, the ggpubr package (version 0.6.0; http://cran.r-project.org/web/packages/ggpubr/index.html) was employed to create a diverging box plot to visualize significant differences in key gene expression levels between the disease and control groups.

Receiver operating characteristic (ROC) curve

ROC curves were used to validate the accuracy of key genes in diagnosing DCM. R software (version 4.2.3; cran.r-project.org/bin/windows/base/old/4.2.3/) was used to analyze key gene expression and the pROC (version 1.18.5; http://xrobin.github.io/pROC/) package was used to calculate and plot the area under the curve (AUC), with a larger AUC indicating a greater diagnostic value of the key gene.

Cell culture and model

The HL-1 murine cardiomyocyte cell line (Unico) was maintained at 37°C in a humidified atmosphere of 5% CO2, 21% O2 and 74% N2. The cells were maintained in high-glucose DMEM (cat. no. SH30022.01; Cytiva) supplemented with 10% fetal bovine serum (FBS; Cat. No. SA101.02, Cellmax, Beijing, China). The medium was changed every 2 days, and cells in mid-logarithmic growth phase were collected for experiments. Cultured HL-1 cells were divided into low- (5.5 mM) and high-glucose (33.3 mM) groups, and incubated at 37°C for 48 h (18,19).

Animals

All experimental protocols were approved by the Animal Ethics Committee of Hubei University of Medicine (Shiyan, Hubei, China). A total of six male db/db diabetic mice (age, 6 weeks; body weight 27.5±1.6 g at study initiation; Certificate No. 202341294-202341295, Jiangsu Huachuang Sino Pharmaceutical Technology Co., Ltd, China) were used to establish a model of DCM, with six db/m mice serving as controls. Mice were maintained under a 12/12-h light/dark cycle at a controlled temperature of 22±1°C and 50±10% relative humidity, with ad libitum access to food and water. All animals were acclimatized for 2 weeks prior to experiments. Body weight and blood glucose levels were measured. Blood glucose levels were monitored via tail vein sampling. Briefly, the distal 1–2 mm of the tail tip was sterilized with 70% ethanol and air-dried. A sterile lancet was used to make an incision, and a single blood droplet (~1 µl) was collected. Glucose concentration was analyzed using a Sinocare Blood Glucose Monitoring System (Sinocare Inc., Changsha). Triplicate measurements were averaged per mouse to ensure reliability. All measurements were performed between 9:00 and 11:00 AM under non-fasting conditions to minimize circadian variation. In longitudinal studies, sequential sampling was restricted to the same tail region to avoid tissue damage. The animals were monitored in SPF-grade animal laboratories for 24 weeks, with echocardiograms performed every 4 weeks. Sampling was conducted at 32 weeks of age.

Lentivirus transfection and hexokinase 2 (HK2) overexpression

HL-1 cardiomyocytes were transduced with recombinant lentiviral particles carrying the human HK2 gene (Lenti-HK2; Vigene Biosciences, cat. no. FC-6073) following the manufacturer's recommended protocol. HL-1 cells were seeded in 12-well plates at 1×105 cells/well and cultured at 37°C until reaching 40–50% confluence. Cells were maintained at 37°C in medium, which was replaced with 420 µl fresh complete DMEM supplemented with 10% FBS [cat. no. SA101.02; CellTech (Beijing) Co., Ltd.], supplemented with 20 µl LV-Enhance (50X, Vigene Biosciences) and 60 µl/ml lentivirus overexpressing human HK2 (LV-hHK2-OE) (Vigene Biosciences, cat. no. FBE0001, Shandong, China; http://www.wzbio.com.cn/). LV-ZsGreen-Puro-CON (Vigene Biosciences) served as negative controls. Subsequent experiments were performed 48 h post-transfection. The transfection was conducted at 37°C in a 5% CO2 atmosphere for 18–24 h, followed by the addition of 500 µl complete DMEM (supplemented with 10% FBS [cat. no. SA101.02; CellTech (Beijing) Co., Ltd.]) for an additional 24 h under the same culture conditions. Green fluorescence was monitored for 24–48 h post-transfection to assess transfection efficiency. Once green fluorescence was detected in the cells, puromycin (3 g/ml; cat. no. HY-K1057; MedChemExpress) was added to each well, and the same concentration of puromycin was maintained for 7 days at 37°C with 5% CO. The cells were collected for further experiments and cryopreserved (−196°C) in liquid nitrogen.

Western blotting

Total protein was extracted from heart tissue and HL-1 cells using RIPA lysis buffer (Servicebio Technology Co., Wuhan, China; cat. no. G2002) containing 50 mM Tris-HCl (pH 7.4), 150 mM NaCl, 1 mM EDTA-2Na, 1% sodium deoxycholate, 0.1% SDS and 1% Triton X-100. Protein concentration was determined using the BCAk method. Equal amounts of protein (20 µg/lane) were separated by 10% SDS-PAGE and transferred to a PVDF membrane (Immobilon-PSQ Blotting Membranes; 0.2 µm; Merck KGaA). Following 1-hour blocking at room temperature with 5% skimmed milk, the membranes were incubated overnight at 4°C with the following antibodies: Anti-GAPDH mouse monoclonal (cat. no. 60004-1-Ig; 1:5,000; Proteintech Group, Inc.), anti-microsomal glutathione S-transferase 1 (MGST1) rabbit monoclonal (cat. no. A0880; 1:500; ABclonal Biotech Co., Ltd.), anti-HK2 rabbit polyclonal (1:1,000; cat. no. A0994; ABclonal Biotech Co., Ltd,) and anti-Phosphatidylinositol-4-Phosphate 3-Kinase C2 Domain-Containing Subunit Gamma (PIK3C2G) polyclonal antibody (cat. no. 25904-1-AP; 1:2,000; Proteintech Group, Inc.). Following three washes with TBST (0.05% Tween-20), the membranes were incubated with horseradish peroxidase-conjugated secondary antibody (1:1,000; Beyotime Institute of Biotechnology, cat. no. A0208) at room temperature for 1 h. The membranes were washed three additional times with TBST, and chemiluminescent signals were detected using an ultra-sensitive ECL substrate (cat. no. BL520B, Biosharp, China) and imaged with a gel imaging system (Bio-Rad ChemiDoc XRS+; Bio-Rad Laboratories, Inc.). Data were analyzed using Image Lab 6.0 software (Bio-Rad), and semi-quantification of band intensities was performed with ImageJ 1.54d (National Institutes of Health, USA).

Echocardiography

Echocardiography was performed using a Visualsonic Vevo 2100 (VisualSonics, Inc.) equipped with a 30 MHz linear array ultrasound transducer (20). Echocardiography was performed under inhalant anesthesia using isoflurane delivered via a nose cone connected to an anesthetic vaporizer. Induction was achieved with 4% isoflurane and maintenance was performed using 1.5–2.0% isoflurane. The internal diameter of the left ventricle (LV) was measured in M-mode over ≥3 cardiac cycles, and the mean value was subsequently calculated. The left ventricular end-diastolic inner diameter (LVEDd) and left ventricular end-systolic inner diameter (LVESd) were measured at the maximum and minimum LV areas, respectively. The left ventricular shortening fraction (LVFS) and left ventricular ejection fraction (LVEF) were calculated as follows: LVFS %=(LVEDd-LVESd)/LVEDd ×100%.

Immunohistochemical staining

The myocardial tissues were fixed in 4% paraformaldehyde (PFA, cat. no. BL539A, Biosharp, China) at 4°C for 24–48 h, processed through graded ethanol, cleared in xylene, and embedded in paraffin. Samples were sliced into 5-µm-thick serial sections were prepared, dewaxed in xylene at room temperature (25°C) for 30 min and hydrated with graded alcohol. Antigens were retrieved using microwave heating at 95–100°C with sodium citrate buffer (pH 6.0; cat. no. 36311-B, Yeasen, China) for 30 min. Endogenous peroxidase activity was blocked with 3% hydrogen peroxide for 10 min at room temperature. Next, 5% bovine serum albumin (cat. no. HY-D0842; MedChemExpress) was applied at 37°C for 1 h. The sections were incubated overnight at 4°C with anti-MGST1 rabbit monoclonal (1:100) and anti-HK2 rabbit polyclonal antibody (1:200), followed by incubation with liquid 3,3′-diaminobenzidine (cat. no. 36311-H and 36311-I, Yeasen) substrate for color development according to the manufacturer's instructions. Sections were stained with hematoxylin at room temperature for 3 min to visualize nuclei, differentiated using 1% hydrochloric acid alcohol, dehydrated through ascending ethanol series (70, 95, and 100% ethanol; 2 min each at room temperature) and cleared in xylene (two changes, 3 min each at room temperature) prior to mounting with neutral balsam.

Stained sections were visualized and digitized using an Aperio CS2 digital slide scanner (Leica Biosystems, Germany) under bright-field microscopy.

Hematoxylin and eosin (H&E) and Masson's trichrome staining

Cardiac tissue was fixed in 4% paraformaldehyde (PFA, cat. no. BL520B, Biosharp, China) at 4°C for 24 h. Following fixation, the samples were washed in phosphate-buffered saline (PBS) and processed through a graded ethanol series (70, 85, 95, and 100%), cleared in xylene, and embedded in paraffin using an automated tissue processor (Leica TP1020; Leica Biosystems). Serial sections (5 µm thickness) were cut using a rotary microtome (Leica RM2235; Leica Biosystems) and mounted on poly-L-lysine-coated glass slides for subsequent histological staining. The sections were stained with H&E following deparaffinization and dehydration as previously described (21). Following deparaffinization and rehydration, the sections were stained with hematoxylin solution for 5 min, rinsed in running tap water for 5 min, differentiated in 1% acid alcohol for 30 sec, and blued in 0.2% ammonia water. Subsequently, the sections were counterstained with eosin for 3 min, dehydrated through a graded ethanol series (70, 95, and 100%), cleared in xylene, and mounted with neutral balsam. Additionally, sections were stained using a modified Masson's Trichrome Stain kit (cat. no. G1340; Beijing Solarbio Science & Technology Co., Ltd.) according to the manufacturer's instructions. Sections were visualized using an Aperio CS2 digital slide scanner (Leica Biosystems, Germany) under bright-field microscopy.

Transcriptomic assay

To investigate the regulatory effect of HK2 overexpression on downstream gene expression in HL-1 cells, cultured wild-type (WT) HL-1 cells (HK2-WT) and lentivirus-transfected HK2 overexpressing HL-1 cells (HK2-OE) were washed with a sterile PBS solution, lysed by adding TRIzol (cat. no. 15596026CN; Thermo Fisher Scientific, USA) lysis solution and incubated for 5 min at room temperature. The lysed cells were mixed and transferred to 1.5 ml sterile Eppendorf tubes for storage at −80°C. The samples were preserved and transported on dry ice to Metware Biotechnology Co., Ltd. (Wuhan, China) for analysis. Total RNA was processed with the NEBNext Ultra II RNA Library Prep kit (Cat. No. E7770; New England Biolabs) for Illumina sequencing. RNA integrity was verified using the Qsep400 Bioanalyzer (Bioptic Inc.) with all samples exhibiting RNA Integrity Number >7.0. Sequencing was performed on the Illumina NovaSeq 6000 platform using the NovaSeq 6000 SP Reagent Kit (300 cycles; Cat. No. 20027464; Illumina Inc.) for paired-end 150 bp sequencing. Final libraries were quantified by qPCR using the KAPA Library Quantification Kit (Cat. No. KK4824; Roche) and loaded at 12.5 pM concentration. Unsupervised principal component analysis) was performed by statistics function prcomp within R (version 3.5.1; r-project.org). The data was unit variance scaled before unsupervised PCA.

Quantitative analysis of energy metabolism and lipid metabolism

To investigate the effects of HK2 gene expression on energy and lipid metabolism in HL-1 cells, targeted quantification of metabolites was performed using liquid chromatography-tandem mass spectrometry (LC-MS/MS) as previously described (22). All metabolites were detected through MetWare (metware.cn/) using the AB Sciex QTRAP 6500 LC-MS/MS platform. The analysis was performed in both positive and negative ionization modes, with the following parameters: Ion spray voltage set at 5,500 V (positive) and −4,500 V (negative), curtain gas at 35 psi, and source temperature maintained at 550°C. Multiple reaction monitoring (MRM) transitions were optimized for each metabolite, with specific precursor and product ion pairs (m/z) selected for quantification. The nebulizer gas flow rate was maintained at 1.5 l/min, and the collision gas pressure was set to medium. Pearson correlation analysis was performed using quantitative values of genes and metabolites from all samples. Fold-change differences in correlation coefficients with absolute values >0.8 and P<0.05 were analyzed.

Sample preparation and extraction

The cells were pelleted by centrifugation at 1,000 × g for 10 min at 4°C, and the resulting cell pellets were stored at −80°C until metabolomic analysis. The collected cell pellet samples for metabolomic analysis were thawed on ice and 100 µl ultrapure water was added to resuspend the cell pellet. A 50-µl aliquot of the cell suspension was added to 200 µl methanol (precooled to −20°C). The mixture was vortexed for 2 min at 1,000 × g at 4°C. The sample was flash-frozen in liquid nitrogen (−196°C) for 5 min, thawed on ice (0–4°C) for 5 min and vortexed at maximum speed (700 × g) at room temperature for 2 min. The freeze-thaw-vortex cycle was repeated three times. The sample was centrifuged at 12,000 × g for 10 min at 4°C and 200 µl supernatant was transferred to a new centrifuge tube and stored at −20°C for 30 min. The supernatant was centrifuged again at 12,000 × g for 10 min at 4°C. A total of 180 µl supernatant was filtered through a protein precipitation plate for LC-MS analysis, while the 50 µl cell suspension underwent three freeze-thaw cycles, followed by centrifugation at 12,000 × g for 10 min at 4°C. The protein concentration in the supernatant was determined using a BCA protein assay kit.

Quantitative PCR

Total RNA was extracted from cells using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) and reverse-transcribed into cDNA using HiScript IV RT SuperMix for qPCR (+gDNA wiper) (cat. no. R423-01; Vazyme Biotech Co., Ltd., Nanjing, China). Quantitative PCR was performed using the CFX Connect™ Real-Time PCR Detection System (Bio-Rad Laboratories, Inc., Hercules, CA, USA) with Taq Pro Universal SYBR qPCR Master Mix (cat. no. Q712; Vazyme Biotech Co., Ltd.). The thermal cycling conditions consisted of an initial denaturation at 95°C for 30 sec, followed by 40 cycles of 95°C for 10 sec and 60°C for 30 sec. Gene expression levels were normalized to GAPDH reference genes and calculated using the 2-ΔΔCq method (23). All primers were synthesized by Sangon Biotech Co., Ltd. (Shanghai, China; Table SI).

Protein-protein interaction (PPI) network analysis

PPI networks were constructed using the STRING database (version 12.0; minimum required interaction score, 0.4; http://cn.string-db.org/) with the differentially expressed genes as input. The networks were visualized and analyzed using Cytoscape software (version 3.9.0; National Institute of General Medical Sciences), with the CytoHubba plugin (version 0.1, http://apps.cytoscape.org/apps/cytohubba) used to identify hub genes based on topological parameters (degree centrality and betweenness centrality).

Statistical analysis

All data are presented as the mean ± standard deviation from three independent experimental repeats. All analyses were conducted using GraphPad Prism 9.0 software (Dotmatics). Two samples were compared using unpaired two-tailed Student's t-test. For >2 groups, one-way ANOVA followed by Tukey's post hoc test was used if the data met the assumptions of normality (assessed by the Shapiro-Wilk test) and homogeneity of variance (verified by Levene's test). If these assumptions were violated, the non-parametric Kruskal-Wallis test, followed by Dunn's post hoc test, was applied. P<0.05 was considered to indicate a statistically significant difference.

Results

Identification of DEGs

To identify DEGs, a fold-change threshold of |log2 (fold-change)|≥1 and adjusted P<0.05 were employed. In the merged dataset of GSE4745 and GSE6880, 47 DEGs were identified, comprising 25 up- and 22 downregulated genes (Fig. 1A and B). To explore the biological functions of the DEGs and the potential biological pathways associated with DCM, GO functional and KEGG pathway enrichment analysis were performed (Tables SII and SIII).

Functional characterization of DEGs

GO enrichment analysis indicated DEGs were involved in BPs, including ‘fatty acid metabolic process’, ‘pyruvate metabolic process’, ‘small molecule catabolic process’, ‘response to toxic substance’, ‘response to starvation’, ‘response to hypoxia’ and ‘response to insulin’ (Fig. 1D). Additionally, the DEGs were localized to cellular components such as ‘banded collagen fibril’, ‘fibrillar collagen trimer’, ‘organelle outer membrane’, ‘sarcolemma’, ‘organelle inner membrane’, ‘mitochondrial inner membrane’ and ‘mitochondrial matrix’. Furthermore, DEGs associated with ‘hexose transmembrane transporter activity’, ‘glucose transmembrane transporter activity’, ‘neuropeptide hormone activity’, ‘CoA hydrolase activity’, ‘acyl-CoA hydrolase activity’, ‘glutathione peroxidase activity’, ‘platelet-derived growth factor binding’, ‘palmitoyl-CoA hydrolase activity’ and ‘phospholipid binding’ were significantly enriched (Fig. 1C-E).

Pathway enrichment analysis

KEGG enrichment analyses indicated that the DEGs were significantly enriched in ‘fatty acid elongation’, ‘drug metabolism-other enzymes’, ‘butanoate metabolism’, ‘arachidonic acid metabolism’, ‘metabolism of xenobiotics by cytochrome P450’, ‘drug metabolism-cytochrome P450’, ‘adipocytokine signaling pathway’, ‘glycolysis/gluconeogenesis’, ‘diabetic cardiomyopathy’, ‘carbon metabolism’, ‘linoleic acid metabolism’, ‘biosynthesis of amino acids’ and ‘HIF-1 signaling pathway’ (Fig. 1F and G). Additionally, GSEA demonstrated enrichment of DCM-associated genes in ‘collagen biosynthesis and modifying enzymes’ and ‘metabolism of lipids’ (Fig. 1H).

Identification of the most relevant module genes to DCM through WGCNA

After conducting a WGCNA, the blue module was the most relevant to DCM (Fig. 2A-D). This module contained 19 functionally diverse hub genes that represent critical pathways in cardiac pathophysiology. The metabolic regulators included acyl-CoA thioesterases (ACOT1 and ACOT2) and acyl-CoA synthetase long-chain family member 6 (ACSL6), which coordinate lipid metabolism, along with β-hydroxybutyrate dehydrogenase (BDH1) and 3-hydroxy-3-methylglutaryl-CoA synthase 2 (HMGCS2), key enzymes in ketone body metabolism. Extracellular matrix remodeling was represented by collagen type I α2 chain (COL1A2) and collagen type III α1 chain (COL3A1), while immune modulation involved CD74 molecule (CD74) and cathepsin K. Metabolic enzymes included cytochrome P450 2E1 (CYP2E1), hexokinase 2 (HK2), and microsomal glutathione S-transferase 1 (MGST1). Additional significant genes were natriuretic peptide A, pyruvate dehydrogenase phosphatase catalytic subunit 2 (PDP2), proenkephalin (PENK), phosphatidylinositol-4-phosphate 3-kinase catalytic subunit type 2 gamma (PIK3C2G), phospholipase A2 group IIA (PLA2G2A), WAP four-disulfide core domain 1 (WFDC1), and the uncharacterized protein LOC102553868. These molecular networks provide crucial insights into the complex interplay of metabolic dysregulation, structural remodeling, and inflammatory responses in DCM development.

Identification and characterization of DCM signature genes

Using Venn analysis of DEGs and WGCNA module genes, 17 overlapping genes were identified (Fig. 3A). A LASSO regression model was constructed based on the expression matrix of these genes. Using the GLMNET package, the cross-validation curves (Fig. 3B) and LASSO coefficient path diagrams (Fig. 3C) of the model were visualized. Ultimately, four disease signature genes (HMGCS2, PIK3C2G, HK2 and MGST1) were identified. A comparative analysis of the expression of these genes was conducted in both the internal and external datasets using the limma package. Next, the ggpubr package was employed to visualize the differences in gene expression between the disease and control groups using boxplots (Fig. 3D and F). Compared with the control group, HK2 expression was downregulated in the DCM group, while the expression of HMGCS2, PIK3C2G and MGST1 was upregulated. Finally, the diagnostic value of signature genes for DCM was assessed using the pROC package. All four genes demonstrated high diagnostic value for DCM, with AUC >0.9 (Fig. 3E and G).

Signature gene expression and validation in HL-1 cardiomyocytes

The surface area of HL-1 cardiomyocytes cultured in a high-glucose environment was notably increased compared with that of cardiomyocytes cultured in a low-glucose environment (Fig. 4A). A significant increase in cell surface area was confirmed by quantitative analysis following 48 h high-glucose treatment (Fig. 4B). The number of apoptotic cells was significantly higher following high-glucose compared with low-glucose treatment (Fig. 4D and E).

Western blot analysis revealed that MGST1 protein expression increased, while HK2 protein expression decreased in HL-1 cells treated with high- compared with low-glucose (Fig. 4E and F). PIK3C2G expression did not significantly differ between low-glucose and high-glucose treatment groups (Fig. 4E and F)

ROS production was significantly elevated in HL-1 cells treated with high-glucose, whereas ROS production was significantly decreased in HL-1 cells following HK2-OE (Fig. 4G and H). The increase in HK2 protein expression after HK2-OE was validated by a lentiviral transfection assay (Fig. 4I and J).

The present study found that HK2 was abundantly expressed in the nuclei of HL-1 cells cultured under high-glucose conditions (Fig. S1), suggesting it may influence the regulation of disease-associated genes in DCM. LLPS was detected in both HK2-WT and HK2-OE HL-1 cardiomyocytes by immunofluorescence analysis (Fig. S2).

Mouse model validation and histological testing

In the present study, db/db mice were selected as a model for T2D, whereas db/m mice served as the control group. Western blot analysis of cardiac tissue revealed that HK2 was expressed at lower levels in the cardiac tissue of db/db compared with db/m mice. The difference in MGST1 and PIK3C2G cardiac tissue expression between the two groups was not significant (Fig. 4K and L). The heart-to-body weight ratio and the heart weight-to-tibia length ratio did not significantly differ between the two groups (Fig. 5A). Gross examination of the cardiac tissue indicated that the hearts of db/m mice were larger than those of db/db mice (Fig. 5B). A total of one db/db mouse died at ~32 weeks of age, and four of the surviving db/db mice exhibited significant frontal bladder overfill (4/5), suggesting the possibility of neurogenic cystitis with urinary retention (Fig. 5C).

To evaluate the effects of the intervention, blood glucose levels and body weight were measured at 8 and 32 weeks of age. At 8 weeks, the body weight of the db/db mice was significantly greater than that of the control mice, with a mean body weight ranging from 35 to 40 g. However, at 32 weeks, the db/db mice exhibited a significant decrease in body weight, and their mean weight was notably lower compared with that of the control mice (Fig. 5D). Blood glucose levels were significantly elevated in db/db mice compared with control mice at 8 and 32 weeks (Fig. 5E). Echocardiography data indicated that the LVEF and LVFS were significantly lower in db/db compared with db/m mice at 32 weeks (Fig. 5F). Additionally, the left intraventricular diameter was greater in db/db mice (Figs. 5G and S3), and diastolic function was impaired in these mice (Fig. 5H). Histological staining revealed increased cardiac fibrosis and cardiomyocyte hypertrophy in db/db mice (Fig. 5I and J).

Immunohistochemistry staining revealed no notable difference in MGST1 expression. By contrast with db/db mice, expression of HK2 was higher in the cardiac tissue of db/m mice. Similarly, the expression levels of HK2 in both db/m and db/db mice were greater than those in the PBS-treated controls (Fig. 5J).

Transcriptomic analysis and metabolic data

Transcriptomic sequencing analysis was conducted on HK2-WT and HK2-OE cells. Energy and lipid metabolisms were quantitatively analyzed in six samples from each group. Transcriptomic sequencing was performed using Illumina high-throughput sequencing platform, which included three HK2-WT and three HK2-OE samples. Similarities between the samples were assessed through principal component analysis (PCA) (Fig. 6A). Gene expression was quantified using fragments per kilobase of transcript per million fragments mapped as a measure of gene expression levels (Fig. 6B). DEGs were identified with |log2 (fold-change)|≥1 and a false discovery rate <0.05.

A total of 1,292 DEGs were identified, comprising 432 up- and 860 downregulated genes (Fig. 6C). Energy metabolomics analysis was performed using a LC-MS/MS detection platform in conjunction with the MetWare database, resulting in the detection of 57 metabolites (Fig. 6D; Table SIV). Differences in metabolites within sample groups were analyzed using PCA (24) (Fig. 6E). In total, 11 differentially expressed metabolites were identified based on fold-change ≥2 and ≤0.5 (Fig. 6F and G; Table SV). Lipid metabolomics was used to quantify 1,269 lipids, and PCA was used to elucidate the overall metabolic differences and variability between samples within groups (Fig. 6H). Lipid cluster analysis revealed significant differential expression of genes associated with lipid metabolism (Fig. 6I). After screening for variable importance in projection value >1 and P<0.05, 471 differential lipids were identified (Fig. 6J; Table SVI).

Co-analysis of transcriptomics and energy metabolism

KEGG pathway enrichment was compared to identify co-enriched KEGG pathways (Fig. 7A). DEGs and metabolites were enriched in ‘purine metabolism’, ‘nucleotide metabolism’ and ‘biosynthesis of cofactors’. KEGG enrichment analysis was conducted using the P-values from the transcriptome to identify common pathways within the top 50 most significantly enriched pathways. Transcriptomic analysis revealed significant enrichment of pathways associated with ‘glutathione metabolism’, ‘cholinergic synapse’, ‘HIF-1 signaling pathway’, ‘terpenoid backbone biosynthesis’, ‘insulin resistance’, ‘alcoholic liver disease’ and ‘purine metabolism’ (P<0.05). Additionally, metabolites involved in ‘nucleotide metabolism’ were significantly enriched (Fig. 7B). Both histologies exhibited significant enrichment in ‘purine metabolism’ (Fig. 7C).

Gene and metabolite expression patterns showed consistent differential regulation in quadrants 3 and 7, but opposing patterns in quadrants 1 and 9 (demarcated by black dashed lines), which visualizes gene-metabolite fold-change relationships 1 and 9 (Fig. 7D). DEGs and metabolites enriched in ‘purine metabolism’ were selected for correlation analysis. A total of four energy-associated metabolites were significantly associated with 14 genes involved in ‘purine metabolism’ (Fig. 7E). Finally, by constructing a correlation network of genes and metabolites, 114 genes associated with seven energy metabolites were identified (Fig. 7F).

Co-analysis of transcriptomic and lipid metabolism data

Transcriptomics and lipid metabolomics of the co-enriched KEGG pathways were compared. Metabolites associated with ‘glycerolipid metabolism’, ‘lipid and atherosclerosis’, ‘sphingolipid signaling pathway’, ‘insulin resistance’, ‘ether lipid metabolism’ and ‘glycerophospholipid metabolism’ were enriched (Fig. 8A). Transcriptomic data revealed enrichment in ‘vascular smooth muscle contraction’, ‘Rap1 signaling pathway’, ‘human cytomegalovirus infection’, ‘cAMP signaling pathway’, ‘MAPK signaling pathway’, ‘amoebiasis’ and ‘calcium signaling pathway’ (Fig. 8A). Downstream genes of HK2, which are involved in various BPs, were significantly enriched in ‘calcium signaling pathway’, ‘thyroid hormone synthesis’, ‘vascular smooth muscle contraction’, ‘insulin secretion’, ‘MAPK signaling pathway’, ‘regulation of lipolysis in adipocytes’, ‘aldosterone synthesis and secretion’, ‘Rap1 signaling pathway’, ‘cAMP signaling pathway’, ‘inflammatory mediator regulation of TRP channels’ and ‘AGE-RAGE signaling pathway in diabetic complications’. Additionally, metabolites associated with ‘AGE-RAGE signaling pathway in diabetic complications’ and ‘sphingolipid signaling pathway’ were also significantly enriched (Fig. 8B and C). Numerous metabolites correlated with specific genes (Fig. 8D). Based on this analysis, regulatory network maps for genes and metabolites associated with ‘AGE-RAGE signaling pathway in diabetic complications’ and ‘sphingolipid signaling pathway’ were constructed (Fig. 8E and F).

‘AGE-RAGE signaling pathway in diabetic complications’

A total of 14 genes [I16, I11A, thrombomodulin (THBD), angiotensinogen (AGT), COL1A1, COL3A1, endothelin-1 (EDN1), phospholipase C (Plc)b1 (PLCB1), COL4A2, CCL2, TGFB2, SERPINE1, PIK3CD and protein kinase Cβ (PRKCB)] and 32 lipid metabolites were identified by combined analysis of transcriptomics and lipid metabolomics within ‘AGE-RAGE signaling pathway in diabetic complications’ and significantly enriched (Fig. 8E). Of these, I16 and I11A were excluded due to the absence of specific primers (Table SI). Quantitative PCR analysis of the mRNAs of the remaining 12 genes in HL-1 cardiomyocytes from the WT and HK2-OE strains indicated that the difference between the subgroups was not significant for COL3A1. PRKCB exhibited high expression levels in HL-1 cardiomyocytes from the HK2-OE strain, while the other genes were expressed at low levels (Fig. 9A-C). Additionally, protein-protein interaction network maps were constructed for the remaining 12 genotypes (Fig. 9D).

Discussion

DCM is a distinct form of cardiomyopathy associated with diabetes, characterized by ventricular dysfunction in the absence of traditional cardiovascular risk factors such as coronary atherosclerosis, hypertension or valvular disease (1). DCM typically presents with early diastolic dysfunction, which can progress to late systolic dysfunction accompanied by fibrosis. This multifaceted disease involves various underlying mechanisms, including mitochondrial dysfunction, ER and oxidative stress, alterations in the extracellular matrix, disruptions in the insulin signaling pathway, inflammation, microvascular dysfunction and cardiometabolic abnormality (7). Despite extensive research (6,8), the underlying mechanisms of DCM remain poorly understood, and, to the best of our knowledge, there are currently no targeted treatments or preventive strategies available.

In the present study, bioinformatics analysis revealed that HK2, HMGSC2, MGST1 and PIK3C2G were characteristic genes associated with DCM. Both in vitro and in vivo experiments confirmed that HK2 was associated with DCM, exhibiting significantly decreased expression levels in affected tissues. Apoptosis and ROS levels were notably elevated in HL-1 cardiomyocytes treated with high-glucose. HK2-OE resulted in a significant decrease in ROS production and apoptosis in HL-1 cardiomyocytes.

Oxidative stress serves a key role in the progression of DCM. ROS interact with protein, lipids and DNA, leading to detrimental remodeling of cardiac tissue (25). Mitochondrial ROS production is a key factor in the pathogenic mechanisms triggered by elevated blood sugar levels; excessive ROS can cause DNA damage as well as oxidative modifications of proteins and lipid (26). The upregulation of heme oxygenase induced by diabetes contributes to oxidative stress in the myocardium (27). The activation of acidic sphingomyelinase (ASMase) in cardiomyocytes due to a high-fat diet may exacerbate oxidative stress and apoptosis by upregulating the expression of NADPH oxidase 4, leading to DCM (28).

The present analysis revealed that the HK2 gene regulates the expression of 12 genes in ‘sphingolipid signaling pathway’ (including sphingosine-1-phosphate receptors 1/S1PR3, kinases SPHK1/PIK3CD/PRKCB, metabolic enzymes CERS4/SGPP1, and kallikrein-kinin system components KNG1/KNG2/BDKRB2) and 49 lipid metabolites, including glycerolipids, ceramides and sphingomyelins (SMs). SM serves as a precursor to ceramide, a metabolite generated by the cleavage of SM by neutral sphingomyelinase or ASMase (29). SM is a key component of cell membranes, while ceramide functions as a second messenger of lipids, regulating phosphatase, kinases and transcription factors (30), and is associated with cell proliferation and apoptosis. Lower concentrations of SM and ceramide promote cell proliferation and function, whereas elevated levels lead to cell dysfunction and death (31). Diacylglycerol (DG) and SM are produced by SM synthase 1 (SMS1) from phosphatidylcholine and ceramide as substrates. SMS1 catalyzes the biosynthesis of DG, which modulates SMS1 activity (32). Therefore, HK2 may influence the function of cardiomyocytes in DCM through the metabolism of ceramide and SM.

In the present study, db/db mice revealed a notable accumulation of white adipose tissue on the epicardial surface. The bladder of these mice exhibited excessive urinary storage, suggesting that the 32-week-old db/db mice may have experienced severe diabetic neurogenic cystitis and abnormal lipid accumulation as comorbidities. By contrast with Li et al (33), db/db mice in the present study displayed significant emaciation after 24 weeks of continuous feeding from baseline (8 weeks). There was no notable difference in heart size or weight or cardiac decompensation. This may be associated with the severe protrusion of the enlarged bladder into the gastrointestinal tract, which affects feeding and inflammatory responses in the mice. Cardiac tissue staining indicated increased myocardial fibrosis, myocardial hypertrophy, reduced myofibrillar interstitial space and low expression of HK2, which was consistent with the present bioinformatics analysis.

Previous studies have reported decreased HK2 expression in diabetic mice (3436). HK2-OE has been demonstrated to improve cardiomyocyte contractile function and elevate ATP levels in hypoxic diabetic mice (37). During aging, methyltransferase-like 3 induces HK2 expression, leading to metabolic reprogramming that promotes liquid-liquid phase separation (LLPS) (38). LLPS is a biophysical phenomenon characterized by the condensation of biomolecules, such as protein or nucleic acids, into membraneless, liquid-like droplets within cells. This process is primarily driven by multivalent interactions and the presence of intrinsically disordered regions within proteins (39). These dynamic condensates serve a key role in cellular function by spatially organizing molecules and promoting specific biochemical reactions (40). Notably, LLPS was more prominent in HK2-OE compared with HK2-WT cells. This suggests HK2-OE may facilitate the formation of biomolecular condensates, potentially via enhanced interactions with protein or nucleic acids that drive phase separation. Previous studies have demonstrated that metabolic enzymes, including HK2, engage in LLPS, thereby influencing cellular metabolism and stress responses (41,42). The increased LLPS in HK2-OE cells may indicate a role for HK2 in the assembly of metabolic compartments or stress granules, which may be vital for maintaining cardiomyocyte function under both physiological and pathological conditions (43). The detection of LLPS in cardiomyocytes underscores its potential role in cardiac biology. For example, LLPS is associated with the regulation of sarcomere organization, calcium signaling and cellular stress responses in the heart (44). Further exploration of the molecular mechanisms governing LLPS in cardiomyocytes, particularly in the context of HK2-OE, may yield valuable insights into the pathogenesis of cardiac disorders, including hypertrophy, heart failure and ischemia-reperfusion injury. Purines serve a key role in cellular metabolism, serving as an energy source and essential component in ATP synthesis. HK2 is a key enzyme that regulates purine metabolism, and is involved in the metabolic processes of AMP, ADP, ATP, inosine monophosphate, adenosine and inosine (38). The present energy analysis revealed that HK2 modulated the purine metabolites adenine, dAMP, guanosine and IMP through 14 downstream genes. In a normal resting state, 70% of total cardiac energy production from ATP is derived from fatty acid oxidation. However, in hearts affected by DCM, glucose use is notably altered, and the energy source of the heart predominantly relies on fatty acids (45).

T2D is a chronic inflammatory disease characterized by inflammation and immune senescence (46). Metabolic stress-driven immune senescence is a key factor contributing to cardiac dysfunction in DCM (47). AGEs are formed when protein, lipid and nucleic acids undergo non-enzymatic modification through glycosylation (involving glucose, fructose and pentose) via the Maillard reaction (48). The interaction of AGEs with RAGE triggers oxidative stress, inflammatory response and increased extracellular matrix accumulation (49). Blocking RAGE prevents cardiac dysfunction in db/db mice (50). The present study identified that HK2 regulated 30 lipid submetabolites (including glycerolipids and ceramides) through 14 downstream genes (I16, I11A, THBD, AGT, COL1A1, COL3A1, EDN1, PLCB1, COL4A2, CCL2, TGFB2, SERPINE1, PIK3CD, PRKCB). The accumulation of toxic lipid metabolites is associated with mitochondrial function, bioenergetics and metabolic regulation, leading to metabolic remodeling of the DCM heart (51) and supporting alterations in the cardiomyocyte microenvironment (52).

THBD is implicated in coagulation, innate immunity, inflammation and cell proliferation, and is primarily located in the vascular endothelium (53). THBD-dependent activated protein C formation protects glomerular cells and podocytes from glucose-induced injury (54). Through its lectin-like domain, THBD mitigates ischemia-reperfusion injury in cardiomyocytes (55).

EDN1 may mediate cardiomyocyte hypertrophy in DCM via the MAPK pathway, while EDN1 antagonists prevent glucose-induced cardiomyocyte hypertrophy and AGT expression (56). AGT, the precursor of all angiotensin peptides, not only serves as a renin-angiotensin system substrate but also promotes macrophage infiltration, enhances adipocyte metabolic activity (57) and accelerates cancer progression (58). Hepatocytes are the primary source of AGT, with adipose tissue contributing ~25% of plasma AGT levels. Elevated AGT expression is associated with cardiovascular disease and obesity (59). The upregulation of COL1A1 and COL3A1 promotes myocardial fibrosis in DCM, facilitated by an increase in circular RNA homeodomain-Interacting Protein Kinase 3 (circHIPK3) in DCM mice (60). COL4A2 encodes type IV collagen fibers, which are key for the integrity and function of the basement membrane (61) and are poorly expressed in DCM (62).

The PLC enzyme hydrolyzes phosphatidylinositol diphosphate into DG and inositol triphosphate, which are key lipid mediators involved in cardiomyocyte regulatory signaling as second messengers (63). Elevated PLCB1 expression is associated with atrial dilation in both humans and mice (64). High-glucose induces an increase in CCL2 and IL-6 expression, with glycemic control decreasing IL-6 but not CCL2 levels (65). This may explain why partial glycemic control is ineffective in alleviating DCM progression. High-glucose-induced expression of TGFB and receptor activation promotes myocardial fibrosis in DCM (66).

SERPINE1 is a key gene in DCM and may be associated with impaired mitochondrial function, although the exact mechanism remains unclear (67). Hepatic PIK3CD levels are significantly decreased in mouse models of diabetes, and PIK3CD is associated with insulin resistance (68). High-glucose triggers the activation of PRKCB and increases DG levels. Overexpression of PRKCB induces cardiac hypertrophy and heart failure in mice. HG-induced oxidative stress may promote diabetic complications via the DG/PRKCB pathway (69). Consequently, downstream HK2-regulated genes involved in the AGE/RAGE pathway are associated with DCM.

Although the present study provided insight into the role of HK2 in DCM and its potential as a therapeutic target, there are limitations. First, the findings are primarily derived from preclinical models, including HL-1 cardiomyocytes and db/db mice, which may not fully replicate the complexity of the human DCM pathophysiology. Second, the present study lacked transcriptomic and metabolomic analyses of myocardial tissue, as well as investigation of the functional effects of altered metabolite levels on DCM progression. Third, the lack of validation using clinical samples limits the direct translation of the present results to human patients. Future studies should incorporate patient-derived samples or clinical data to validate the therapeutic potential of HK2 and explore the mechanisms of HK2 by manipulating key genes or metabolites within the metabolic pathway to facilitate clinical application.

In conclusion, HK2 is a key enzyme in glycolysis and associated with the pathogenesis of DCM. HK2-OE in DCM cardiomyocytes markedly protects against apoptosis and decreased ROS levels. Additionally, HK2 modulates glucose and lipid metabolism in cardiomyocytes through numerous metabolic pathways. Metabolic intermediates and end products may inversely regulate gene and protein expression. The present study suggested that the AGE/RAGE signaling pathway is a notable mechanism by which HK2 regulates lipid metabolism and is associated with diabetic complications.

Supplementary Material

Supporting Data
Supporting Data

Acknowledgements

Not applicable.

Funding

The present study was supported by National Natural Science Foundation of China (grant no. 82270530) and Research Fund of the Science and Technology Program of Fujian Province (grant no. 2023J011776).

Availability of data and materials

The data generated in the present study may be found in the National Center for Biotechnology Information BioProject and European Molecular Biology Laboratory's European Bioinformatics Institute MetaboLights under accession number PRJNA1234119 and MTBLS12315 and MTBLS12314, respectively, or at the following URL: (https://www.ncbi.nlm.nih.gov/sra/?term=PRJNA1234119; https://www.ebi.ac.uk/metabolights/MTBLS12315; http://www.ebi.ac.uk/metabolights/MTBLS12314).

Authors' contributions

YD and YZ conceived the study. BL and XZ designed experiments, analyzed data and constructed figures. BL, XZ, LM, XW, YD and YZ performed experiments. YD supervised the study. BL, XZ, LM and XW wrote the manuscript. BL, XZ, YD and YZ edited the manuscript. YD and YZ confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

The animal experiments were approved by the Center's Experimental Animal Ethics Committee of Hubei University of Medicine (approval no 2024S071; Shiyan, China) and were conducted in accordance with the Declaration of Helsinki.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ogurtsova K, da Rocha Fernandes JD, Huang Y, Linnenkamp U, Guariguata L, Cho NH, Cavan D, Shaw JE and Makaroff LE: IDF diabetes atlas: Global estimates for the prevalence of diabetes for 2015 and 2040. Diabetes Res Clin Pract. 128:40–50. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Peng C and Zhang Y, Lang X and Zhang Y: Role of mitochondrial metabolic disorder and immune infiltration in diabetic cardiomyopathy: New insights from bioinformatics analysis. J Transl Med. 21:662023. View Article : Google Scholar : PubMed/NCBI

3 

Fuentes-Antrás J, Picatoste B, Ramírez E, Egido J, Tuñón J and Lorenzo Ó: Targeting metabolic disturbance in the diabetic heart. Cardiovasc Diabetol. 14:172015. View Article : Google Scholar : PubMed/NCBI

4 

Yuan Q, Sun Y, Yang F, Yan D, Shen M, Jin Z, Zhan L, Liu G, Yang L, Zhou Q, et al: CircRNA DICAR as a novel endogenous regulator for diabetic cardiomyopathy and diabetic pyroptosis of cardiomyocytes. Signal Transduct Target Ther. 8:992023. View Article : Google Scholar : PubMed/NCBI

5 

Tan Y, Zhang Z, Zheng C, Wintergerst KA, Keller BB and Cai L: Mechanisms of diabetic cardiomyopathy and potential therapeutic strategies: Preclinical and clinical evidence. Nat Rev Cardiol. 17:585–607. 2020. View Article : Google Scholar : PubMed/NCBI

6 

Murtaza G, Virk HUH, Khalid M, Lavie CJ, Ventura H, Mukherjee D, Ramu V, Bhogal S, Kumar G, Shanmugasundaram M and Paul TK: Diabetic cardiomyopathy-A comprehensive updated review. Prog Cardiovasc Dis. 62:315–326. 2019. View Article : Google Scholar : PubMed/NCBI

7 

Jia G, Hill MA and Sowers JR: Diabetic cardiomyopathy: An update of mechanisms contributing to this clinical entity. Circ Res. 122:624–638. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Dillmann WH: Diabetic cardiomyopathy. Circ Res. 124:1160–1162. 2019. View Article : Google Scholar : PubMed/NCBI

9 

Jia G, Whaley-Connell A and Sowers JR: Diabetic cardiomyopathy: A hyperglycaemia- and insulin-resistance-induced heart disease. Diabetologia. 61:21–28. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Adeghate E and Singh J: Structural changes in the myocardium during diabetes-induced cardiomyopathy. Heart Fail Rev. 19:15–23. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Song K, Liang D, Xiao D, Kang A and Ren Y: Role of bariatric surgery in improving diabetic cardiomyopathy: Molecular mechanisms and therapeutic perspectives (Review). Mol Med Rep. 30:1992024. View Article : Google Scholar : PubMed/NCBI

12 

Jia G, DeMarco VG and Sowers JR: Insulin resistance and hyperinsulinaemia in diabetic cardiomyopathy. Nat Rev Endocrinol. 12:144–153. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Leary S, Anthony R, Gwaltney-Brant S, Cartner S, Dewell R, Webb P, Plummer P, Hoenig DE, Moyer W, Smith SA, et al: AVMA guidelines for the depopulation of animals. 2019.

14 

Leek JT, Johnson WE, Parker HS, Jaffe AE and Storey JD: The sva package for removing batch effects and other unwanted variation in high-throughput experiments. Bioinformatics. 28:882–883. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, et al: Gene ontology: Tool for the unification of biology. The gene ontology consortium. Nat Genet. 25:25–29. 2000. View Article : Google Scholar : PubMed/NCBI

16 

Kanehisa M and Goto S: KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28:27–30. 2000. View Article : Google Scholar : PubMed/NCBI

17 

Langfelder P and Horvath S: WGCNA: An R package for weighted correlation network analysis. BMC Bioinformatics. 9:5592008. View Article : Google Scholar : PubMed/NCBI

18 

Gao P, Cao M, Jiang X, Wang X, Zhang G, Tang X, Yang C, Komuro I, Ge J, Li L and Zou Y: Cannabinoid receptor 2-centric molecular feedback loop drives necroptosis in diabetic heart injuries. Circulation. 147:158–174. 2023. View Article : Google Scholar : PubMed/NCBI

19 

Sun HJ, Xiong SP, Wu ZY, Cao L, Zhu MY, Moore PK and Bian JS: Induction of caveolin-3/eNOS complex by nitroxyl (HNO) ameliorates diabetic cardiomyopathy. Redox Biol. 32:1014932020. View Article : Google Scholar : PubMed/NCBI

20 

Hu L, Wei J, Zhang Y, Wang Z, Tang J, Tang J, Gao Y, Zhang X, Li Y, Liu Y, et al: ANGPTL8 is a negative regulator in pathological cardiac hypertrophy. Cell Death Dis. 13:6212022. View Article : Google Scholar : PubMed/NCBI

21 

Wu Y, Wang J, Zhao T, Sun M, Xu M, Che S, Pan Z, Wu C and Shen L: Polystyrenenanoplastics lead to ferroptosis in the lungs. J Adv Res. 56:31–41. 2024. View Article : Google Scholar : PubMed/NCBI

22 

An Z, Hu T, Lv Y, Li P and Liu L: Targeted amino acid and related amines analysis based on iTRAQ®-LC-MS/MS for discovering potential hepatotoxicity biomarkers. J Pharm Biomed Anal. 178:1128122020. View Article : Google Scholar : PubMed/NCBI

23 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

24 

Chen W, Gong L, Guo Z, Wang W, Zhang H, Liu X, Yu S, Xiong L and Luo J: A novel integrated method for large-scale detection, identification, and quantification of widely targeted metabolites: Application in the study of rice metabolomics. Mol Plant. 6:1769–1780. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Kaludercic N and Di Lisa F: Mitochondrial ROS formation in the pathogenesis of diabetic cardiomyopathy. Front Cardiovasc Med. 7:122020. View Article : Google Scholar : PubMed/NCBI

26 

Sitte N, Huber M, Grune T, Ladhoff A, Doecke WD, Von Zglinicki T and Davies KJ: Proteasome inhibition by lipofuscin/ceroid during postmitotic aging of fibroblasts. FASEB J. 14:1490–1498. 2000. View Article : Google Scholar : PubMed/NCBI

27 

Farhangkhoee H, Khan ZA, Mukherjee S, Cukiernik M, Barbin YP, Karmazyn M and Chakrabarti S: Heme oxygenase in diabetes-induced oxidative stress in the heart. J Mol Cell Cardiol. 35:1439–1448. 2003. View Article : Google Scholar : PubMed/NCBI

28 

Liu R, Duan T, Yu L, Tang Y, Liu S, Wang C and Fang WJ: Acid sphingomyelinase promotes diabetic cardiomyopathy via NADPH oxidase 4 mediated apoptosis. Cardiovasc Diabetol. 22:252023. View Article : Google Scholar : PubMed/NCBI

29 

Xiang H, Jin S, Tan F, Xu Y, Lu Y and Wu T: Physiological functions and therapeutic applications of neutral sphingomyelinase and acid sphingomyelinase. Biomed Pharmacother. 139:1116102021. View Article : Google Scholar : PubMed/NCBI

30 

Imgrund S, Hartmann D, Farwanah H, Eckhardt M, Sandhoff R, Degen J, Gieselmann V, Sandhoff K and Willecke K: Adult ceramide synthase 2 (CERS2)-deficient mice exhibit myelin sheath defects, cerebellar degeneration, and hepatocarcinomas. J Biol Chem. 284:33549–33560. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Cutler RG and Mattson MP: Sphingomyelin and ceramide as regulators of development and lifespan. Mech Ageing Dev. 122:895–908. 2001. View Article : Google Scholar : PubMed/NCBI

32 

Suzuki R, Murakami C, Dilimulati K, Atsuta-Tsunoda K, Kawai T and Sakane F: Human sphingomyelin synthase 1 generates diacylglycerol in the presence and absence of ceramide via multiple enzymatic activities. FEBS Lett. 597:2672–2686. 2023. View Article : Google Scholar : PubMed/NCBI

33 

Li X, Wu Y, Zhao J, Wang H, Tan J, Yang M, Li Y, Deng S, Gao S, Li H, et al: Distinct cardiac energy metabolism and oxidative stress adaptations between obese and non-obese type 2 diabetes mellitus. Theranostics. 10:2675–2695. 2020. View Article : Google Scholar : PubMed/NCBI

34 

Da Silva D, Ausina P, Alencar EM, Coelho WS, Zancan P and Sola-Penna M: Metformin reverses hexokinase and phosphofructokinase downregulation and intracellular distribution in the heart of diabetic mice. IUBMB Life. 64:766–774. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Palsgaard J, Brøns C, Friedrichsen M, Dominguez H, Jensen M, Storgaard H, Spohr C, Torp-Pedersen C, Borup R, De Meyts P and Vaag A: Gene expression in skeletal muscle biopsies from people with type 2 diabetes and relatives: Differential regulation of insulin signaling pathways. PLoS One. 4:e65752009. View Article : Google Scholar : PubMed/NCBI

36 

Osawa H, Sutherland C, Robey RB, Printz RL and Granner DK: Analysis of the signaling pathway involved in the regulation of hexokinase II gene transcription by insulin. J Biol Chem. 271:16690–16694. 1996. View Article : Google Scholar : PubMed/NCBI

37 

Ye G, Donthi RV, Metreveli NS and Epstein PN: Overexpression of hexokinase protects hypoxic and diabetic cardiomyocytes by increasing ATP generation. Cardiovasc Toxicol. 5:293–300. 2005. View Article : Google Scholar : PubMed/NCBI

38 

Wang C, Tanizawa H, Hill C, Havas A, Zhang Q, Liao L, Hao X, Lei X, Wang L, Nie H, et al: METTL3-mediated chromatin contacts promote stress granule phase separation through metabolic reprogramming during senescence. Nat Commun. 15:54102024. View Article : Google Scholar : PubMed/NCBI

39 

Banani SF, Lee HO, Hyman AA and Rosen MK: Biomolecular condensates: Organizers of cellular biochemistry. Nat Rev Mol Cell Biol. 18:285–298. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Alberti S and Hyman AA: Biomolecular condensates at the nexus of cellular stress, protein aggregation disease and ageing. Nat Rev Mol Cell Biol. 22:196–213. 2021. View Article : Google Scholar : PubMed/NCBI

41 

Zhang H, Ji X, Li P, Liu C, Lou J, Wang Z, Wen W, Xiao Y, Zhang M and Zhu X: Liquid-liquid phase separation in biology: Mechanisms, physiological functions and human diseases. Sci China Life Sci. 63:953–985. 2020. View Article : Google Scholar : PubMed/NCBI

42 

Nott TJ, Craggs TD and Baldwin AJ: Membraneless organelles can melt nucleic acid duplexes and act as biomolecular filters. Nat Chem. 8:569–575. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Riback JA, Katanski CD, Kear-Scott JL, Pilipenko EV, Rojek AE, Sosnick TR and Drummond DA: Stress-Triggered phase separation is an adaptive, evolutionarily tuned response. Cell. 168:1028–1040.e19. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Mo Y, Feng Y, Huang W, Tan N, Li X, Jie M, Feng T, Jiang H and Jiang L: Liquid-liquid phase separation in cardiovascular diseases. Cells. 11:30402022. View Article : Google Scholar : PubMed/NCBI

45 

Bayeva M, Sawicki KT and Ardehali H: Taking diabetes to heart-deregulation of myocardial lipid metabolism in diabetic cardiomyopathy. J Am Heart Assoc. 2:e0004332013. View Article : Google Scholar : PubMed/NCBI

46 

Barbé-Tuana F, Funchal G, Schmitz CRR, Maurmann RM and Bauer ME: The interplay between immunosenescence and age-related diseases. Semin Immunopathol. 42:545–557. 2020. View Article : Google Scholar : PubMed/NCBI

47 

Henson SM and Aksentijevic D: Senescence and type 2 diabetic cardiomyopathy: How young can you die of old age? Front Pharmacol. 12:7165172021. View Article : Google Scholar : PubMed/NCBI

48 

Shen CY, Lu CH, Wu CH, Li KJ, Kuo YM, Hsieh SC and Yu CL: The development of maillard reaction, and advanced glycation end product (AGE)-receptor for AGE (RAGE) signaling inhibitors as novel therapeutic strategies for patients with AGE-related diseases. Molecules. 25:55912020. View Article : Google Scholar : PubMed/NCBI

49 

Bodiga VL, Eda SR and Bodiga S: Advanced glycation end products: Role in pathology of diabetic cardiomyopathy. Heart Fail Rev. 19:49–63. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Nielsen JM, Kristiansen SB, Nørregaard R, Andersen CL, Denner L, Nielsen TT, Flyvbjerg A and Bøtker HE: Blockage of receptor for advanced glycation end products prevents development of cardiac dysfunction in db/db type 2 diabetic mice. Eur J Heart Fail. 11:638–647. 2009. View Article : Google Scholar : PubMed/NCBI

51 

Chong CR, Clarke K and Levelt E: Metabolic remodeling in diabetic cardiomyopathy. Cardiovasc Res. 113:422–430. 2017. View Article : Google Scholar : PubMed/NCBI

52 

Heather LC, Gopal K, Srnic N and Ussher JR: Redefining diabetic cardiomyopathy: Perturbations in substrate metabolism at the heart of its pathology. Diabetes. 73:659–670. 2024. View Article : Google Scholar : PubMed/NCBI

53 

Loghmani H and Conway EM: Exploring traditional and nontraditional roles for thrombomodulin. Blood. 132:148–158. 2018. View Article : Google Scholar : PubMed/NCBI

54 

Isermann B, Vinnikov IA, Madhusudhan T, Herzog S, Kashif M, Blautzik J, Corat MA, Zeier M, Blessing E, Oh J, et al: Activated protein C protects against diabetic nephropathy by inhibiting endothelial and podocyte apoptosis. Nat Med. 13:1349–1358. 2007. View Article : Google Scholar : PubMed/NCBI

55 

Herzog C, Lorenz A, Gillmann HJ, Chowdhury A, Larmann J, Harendza T, Echtermeyer F, Müller M, Schmitz M, Stypmann J, et al: Thrombomodulin's lectin-like domain reduces myocardial damage by interfering with HMGB1-mediated TLR2 signalling. Cardiovasc Res. 101:400–410. 2014. View Article : Google Scholar : PubMed/NCBI

56 

Chen S, Khan ZA, Karmazyn M and Chakrabarti S: Role of endothelin-1, sodium hydrogen exchanger-1 and mitogen activated protein kinase (MAPK) activation in glucose-induced cardiomyocyte hypertrophy. Diabetes Metab Res Rev. 23:356–367. 2007. View Article : Google Scholar : PubMed/NCBI

57 

Wu M, Zhou Y, Pei D and Gao S: Unveiling the role of AGT in lipid metabolism and regulated cell death in colon cancer. Neoplasia. 54:1010092024. View Article : Google Scholar : PubMed/NCBI

58 

Wu F, Zhang L, Wang L and Zhang D: AGT may serve as a prognostic biomarker and correlated with immune infiltration in gastric cancer. Int J Gen Med. 15:1865–1878. 2022. View Article : Google Scholar : PubMed/NCBI

59 

Cassis LA, Police SB, Yiannikouris F and Thatcher SE: Local adipose tissue renin-angiotensin system. Curr Hypertens Rep. 10:93–98. 2008. View Article : Google Scholar : PubMed/NCBI

60 

Wang W, Zhang S, Xu L, Feng Y, Wu X, Zhang M, Yu Z and Zhou X: Involvement of circHIPK3 in the pathogenesis of diabetic cardiomyopathy in mice. Diabetologia. 64:681–692. 2021. View Article : Google Scholar : PubMed/NCBI

61 

Yurchenco PD: Basement membranes: Cell scaffoldings and signaling platforms. Cold Spring Harb Perspect Biol. 3:a0049112011. View Article : Google Scholar : PubMed/NCBI

62 

Lu Y, Huo H, Liang F, Xue J, Fang L, Miao Y, Shen L and He B: Role of pericytes in cardiomyopathy-associated myocardial infarction revealed by multiple single-cell sequencing analysis. Biomedicines. 11:28962023. View Article : Google Scholar : PubMed/NCBI

63 

Fazio A, Evangelisti C, Cappellini A, Mongiorgi S, Koufi FD, Neri I, Marvi MV, Russo M, Ghigo A, Manzoli L, et al: Emerging roles of phospholipase C beta isozymes as potential biomarkers in cardiac disorders. Int J Mol Sci. 24:130962023. View Article : Google Scholar : PubMed/NCBI

64 

Woodcock EA, Grubb DR, Filtz TM, Marasco S, Luo J, McLeod-Dryden TJ, Kaye DM, Sadoshima J, Du XJ, Wong C, et al: Selective activation of the ‘b’ splice variant of phospholipase Cbeta1 in chronically dilated human and mouse atria. J Mol Cell Cardiol. 47:676–683. 2009. View Article : Google Scholar : PubMed/NCBI

65 

Bernal-Lopez MR, Llorente-Cortes V, Calleja F, Lopez-Carmona D, Mayas MD, Gomez-Huelgas R, Badimon L and Tinahones FJ: Effect of different degrees of impaired glucose metabolism on the expression of inflammatory markers in monocytes of patients with atherosclerosis. Acta Diabetol. 50:553–562. 2013. View Article : Google Scholar : PubMed/NCBI

66 

Yue Y, Meng K, Pu Y and Zhang X: Transforming growth factor beta (TGF-β) mediates cardiac fibrosis and induces diabetic cardiomyopathy. Diabetes Res Clin Pract. 133:124–130. 2017. View Article : Google Scholar : PubMed/NCBI

67 

Tuersuntuoheti M, Zhou L, Li J, Yang S, Zhou S and Gong H: Investigation of crucial genes and mitochondrial function impairment in diabetic cardiomyopathy. Gene. 923:1485632024. View Article : Google Scholar : PubMed/NCBI

68 

Du X, Li X, Chen L, Zhang M, Lei L, Gao W, Shi Z, Dong Y, Wang Z, Li X and Liu G: Hepatic miR-125b inhibits insulin signaling pathway by targeting PIK3CD. J Cell Physiol. 233:6052–6066. 2018. View Article : Google Scholar : PubMed/NCBI

69 

Koya D and King GL: Protein kinase C activation and the development of diabetic complications. Diabetes. 47:859–866. 1998. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2025
Volume 32 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li B, Zhao X, Ma L, Wang X, Ding Y and Zhang Y: Mechanistic exploration of hexokinase 2 and metabolism in diabetic cardiomyopathy. Mol Med Rep 32: 211, 2025.
APA
Li, B., Zhao, X., Ma, L., Wang, X., Ding, Y., & Zhang, Y. (2025). Mechanistic exploration of hexokinase 2 and metabolism in diabetic cardiomyopathy. Molecular Medicine Reports, 32, 211. https://doi.org/10.3892/mmr.2025.13576
MLA
Li, B., Zhao, X., Ma, L., Wang, X., Ding, Y., Zhang, Y."Mechanistic exploration of hexokinase 2 and metabolism in diabetic cardiomyopathy". Molecular Medicine Reports 32.2 (2025): 211.
Chicago
Li, B., Zhao, X., Ma, L., Wang, X., Ding, Y., Zhang, Y."Mechanistic exploration of hexokinase 2 and metabolism in diabetic cardiomyopathy". Molecular Medicine Reports 32, no. 2 (2025): 211. https://doi.org/10.3892/mmr.2025.13576