Open Access

Lithium downregulates phosphorylated acetyl‑CoA carboxylase 2 and attenuates mitochondrial fatty acid utilization and oxidative stress in cardiomyocytes

  • Authors:
    • Pao-Huan Chen
    • Ting-Wei Lee
    • Shuen-Hsin Liu
    • Tin Van Huynh
    • Cheng-Chih Chung
    • Yung-Hsin Yeh
    • Yu-Hsun Kao
    • Yi-Jen Chen
  • View Affiliations

  • Published online on: February 5, 2024     https://doi.org/10.3892/etm.2024.12413
  • Article Number: 126
  • Copyright: © Chen 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

Acetyl‑CoA carboxylase 2 plays a crucial role in regulating mitochondrial fatty acid oxidation in cardiomyocytes. Lithium, a monovalent cation known for its cardioprotective potential, has been investigated for its influence on mitochondrial bioenergetics. The present study explored whether lithium modulated acetyl‑CoA carboxylase 2 and mitochondrial fatty acid metabolism in cardiomyocytes and the potential therapeutic applications of lithium in alleviating metabolic stress. Mitochondrial bioenergetic function, fatty acid oxidation, reactive oxygen species production, membrane potential and the expression of proteins involved in fatty acid metabolism in H9c2 cardiomyocytes treated with LiCl for 48 h was measured by using a Seahorse extracellular flux analyzer, fluorescence microscopy and western blotting. Small interfering RNA against glucose transporter type 4 was transfected into H9c2 cardiomyocytes for 48 h to induce metabolic stress mimicking insulin resistance. The results revealed that LiCl at a concentration of 0.3 mM (but not at a concentration of 0.1 or 1.0 mM) upregulated the expression of phosphorylated (p‑)glycogen synthase kinase‑3 beta and downregulated the expression of p‑acetyl‑CoA carboxylase 2 but did not affect the expression of adenosine monophosphate‑activated protein kinase or calcineurin. Cotreatment with TWS119 (8 µM) and LiCl (0.3 mM) downregulated p‑acetyl‑CoA carboxylase 2 expression to a similar extent as did treatment with TWS119 (8 µM) alone. Moreover, LiCl (0.3 mM) inhibited mitochondrial fatty acid oxidation, improved coupling efficiency and the cellular respiratory control ratio, hindered reactive oxygen species production and proton leakage and restored mitochondrial membrane potential in glucose transporter type 4 knockdown‑H9c2 cardiomyocytes. These findings suggested that low therapeutic levels of lithium can downregulate p‑acetyl‑CoA carboxylase 2, thus reducing mitochondrial fatty acid oxidation and oxidative stress in cardiomyocytes.

Introduction

Growing evidence suggests that disruptions in mitochondrial energy metabolism can contribute to the development of various cardiac diseases (1-5). Among the fuel sources utilized by the myocardium, increased fatty acid oxidation plays a pivotal role in driving the pathogenesis of cardiomyopathy (1-7). The inhibition of fatty acid oxidation has been demonstrated to alleviate cardiac dysfunction (2,4,6,8). Acetyl-CoA carboxylases (ACCs) are key enzymes responsible for catalyzing the carboxylation of acetyl-CoA into malonyl-CoA (9). Mammalian cells contain two isoforms of ACC, namely ACC1 and ACC2. ACC1 serves as the rate-limiting enzyme responsible for fatty acid biosynthesis in the cytoplasm of adipocytes and hepatocytes. ACC2 is located in the outer mitochondrial membrane of cardiomyocytes and governs mitochondrial fatty acid oxidation (9,10). Therapeutics targeting ACC2 have the potential to regulate mitochondrial fatty acid utilization, reduce oxidative stress and confer cardioprotective benefits.

For the preceding five decades, lithium has served as the first-line medication for treating bipolar disorder (11). Human and preclinical studies have both demonstrated that in addition to its mood-stabilizing effects, lithium can enhance the activity of mitochondrial complex I while hindering the formation of free radicals and reducing lipid peroxidation and DNA damage (12-14). Furthermore, low-dose lithium has been shown to promote longevity (15,16) and reduce the risk of heart failure (17,18). These findings suggest that low-dose lithium may possess cytoprotective properties and thus may play an essential role in regulating metabolic stress. However, whether lithium exerts cardioprotective effects through the modulation of cardiac metabolism remains unclear. Laboratory evidence has indicated that lithium directly targets glycogen synthase kinase-3 beta (GSK-3β) (19), a crucial protein kinase that inhibits the activity of ACC (20,21). The present study investigated whether lithium can regulate the activity of ACC2 and modulate mitochondrial fatty acid oxidation in cardiomyocytes, with a focus on the potential of lithium for mitigating metabolic stress. The results revealed that at a low physiological concentration (0.3 mM), LiCl upregulated the expression of phosphorylated (p-)GSK-3β and downregulated the level of p-ACC2 in H9c2 cardiomyocytes. Additionally, when used in combination with the GSK-3β inhibitor TWS119, LiCl (0.3 mM) downregulated the expression of p-ACC2, an effect comparable to treatment with TWS119 alone. Furthermore, LiCl (0.3 mM) inhibited mitochondrial fatty acid oxidation, enhanced coupling efficiency and the cellular respiratory control ratio, suppressed reactive oxygen species (ROS) production and proton leakage and restored mitochondrial membrane potential in glucose transporter type 4 (GLUT4)-knockdown H9c2 cardiomyocytes. Taken together, these findings suggest that at a low physiological concentration, lithium can inhibit mitochondrial fatty acid utilization and mitigate oxidative stress in cardiomyocytes, potentially through its upregulation of p-GSK-3β and downregulation of p-ACC2.

Materials and methods

Cell culture and treatment

The H9c2 cell line (cat. no. 60096) was purchased from the Bioresource Collection and Research Center and cultured in Dulbecco's modified Eagle's medium (DMEM; Sigma-Aldrich; Merck KGaA) supplemented with 10% fetal bovine serum (FBS; Sigma-Aldrich; Merck KGaA) at 37˚C in a humidified atmosphere with 5% CO2. To retain the differentiation capacity and mitochondrial respiratory activity, a subculture was performed when the cells reached 80% confluence. The culture medium was changed every 2-3 days. H9c2 cells were treated with LiCl (Sigma-Aldrich; Merck KGaA) at concentrations of 0.1 mM (i.e., subtherapeutic), 0.3 mM (i.e., low therapeutic), or 1.0 mM (i.e., high therapeutic) for 48 h. Additionally, these cells were cotreated with LiCl and a GSK-3β inhibitor, namely TWS119 (Sigma-Aldrich; Merck KGaA), at a concentration of 8 µM for 48 h to evaluate whether lithium downregulated p-ACC2 by modulating GSK-3β activity. Each experiment was performed at least three times.

GLUT4-knockdown cellular model

To simulate insulin resistance and induce metabolic stress in H9c2 cardiomyocytes, a GLUT4-knockdown cellular model was established. H9c2 cardiomyocytes were seeded at a density of 2x105 cells per well on a 6-well plate and transfected when reaching ~80% confluence. Transfection was performed with either 50 nM GLUT4 small interfering (si)RNA (sense 5'-GCUGUUUUCUACUAUUCAAtt-3', antisense 5'-UUGAAUAGUAGAAAACAGCat-3'; cat. no. s73928; Thermo Fisher Scientific, Inc.) or 50 nM negative control siRNA (sense 5'-UAACGACGCGACGACGUAAtt-3', antisense 5'-UUACGUCGUCGCGUCGUUAtt-3'; cat. no. 4390843; Thermo Fisher Scientific, Inc.) using Lipofectamine® RNAiMax Transfection Reagent (Thermo Fisher Scientific, Inc.) for 48 h at 37˚C. The protein knockdown efficiency of GLUT4 and protein expression levels of carnitine palmitoyl transferase 1, the mitochondrial enzyme responsible for the translocation of fatty acids from the cytosol to the mitochondrial matrix, were assessed 48 h after siRNA transfection (Fig. S1). LiCl was administered 24 h post-initiation of transfection. For experiments in which GLUT4-knockdown was combined with LiCl administration, cells were incubated for an additional 48 h. In groups without LiCl treatment, cells were incubated for an equivalent 48 h period, matching the incubation time of the LiCl-treated groups.

Measurement of mitochondrial bioenergetic function

Mitochondrial bioenergetic function was assessed using the XFe24 Extracellular Flux Analyzer (Seahorse Bioscience) and Seahorse XF Cell Mito Stress Test kit (Seahorse Bioscience). H9c2 cells were initially seeded at a density of 7,000 cells per well on a Seahorse XFe24 culture plate and cultured with DMEM supplemented with 10% FBS for 48 h. On the day of the assay, the culture medium was substituted with Seahorse assay medium supplemented with 25 mM glucose, 1 mM pyruvate and 2 mM glutamine. Subsequently, a series of injections was administered, including 1.5 µM oligomycin, 3 µM carbonyl cyanide p-trifluoromethoxy phenylhydrazone (FCCP) and 0.5 µM rotenone/antimycin A. Basal respiration (last rate measurement before oligomycin injection-minimum rate measurement after rotenone/antimycin A injection), adenosine triphosphate (ATP) production (last rate measurement before oligomycin injection-minimum rate measurement after oligomycin injection), proton leakage (minimum rate measurement after oligomycin injection-minimum rate measurement after rotenone/antimycin A injection) and maximal respiration (maximum rate measurement after FCCP injection-minimum rate measurement after rotenone/antimycin A injection) were determined using our previously described methods (22). In addition, coupling efficiency and the cell respiratory control ratio were analyzed as these measures are internally normalized bioenergetic parameters used to assess the proportion of mitochondrial respiratory activity contributing to ATP generation (i.e., coupling efficiency) and the degree of change in mitochondrial respiratory activity attributable to proton leakage (i.e., the cell respiratory control ratio) (23,24). Coupling efficiency was calculated by dividing ATP production by basal respiration. The cell respiratory control ratio was calculated by dividing maximal respiration by proton leakage.

Measurement of mitochondrial fatty acid oxidation

Mitochondrial fatty acid oxidation was assessed using the XFe24 Extracellular Flux Analyzer (Seahorse Bioscience) and Seahorse XF substrate oxidation stress test kit (Seahorse Bioscience). H9c2 cells were initially seeded at a density of 7,000 cells per well on a Seahorse XFe24 culture plate and cultured with DMEM supplemented with 10% FBS for 48 h. On the day of the assay, the culture medium was substituted with Seahorse assay medium containing 25 mM glucose, 1 mM pyruvate and 2 mM glutamine. During the substrate oxidation stress test, etomoxir was injected at a concentration of 40 µM. Mitochondrial fatty acid oxidation was estimated by monitoring the change in the oxygen consumption rate following etomoxir injection.

Measurement of mitochondrial ROS

Mitochondrial ROS levels were assessed using MitoSox Red dye (Invitrogen; Thermo Fisher Scientific, Inc.) and a fluorescence microscopy system (EVOS M5000 Imaging System; Thermo Fisher Scientific, Inc.) in accordance with our previously described methods (22). In brief, H9c2 cells were seeded at a density of 5,000 cells per well on a 96-well plate and cultured with DMEM supplemented with 10% FBS for 48 h. Prior to fluorescence microscopy, the H9c2 cells were loaded with MitoSox Red at a concentration of 5 µM and Hoechst 33423 (Sigma-Aldrich; Merck KGaA) at a concentration of 1 µg/ml for a 30-min incubation period at 37˚C. Fluorescence intensity was then measured in four randomly selected fields in each well and quantified using ImageJ 1.52a software (National Institutes of Health).

Measurement of mitochondrial membrane potential

Mitochondrial membrane potential was assessed using the TMRE Mitochondrial Membrane Potential Assay Kit (Cayman Chemical Company) and fluorescence microscopy (EVOS M5000 Imaging System; Thermo Fisher Scientific, Inc.). In brief, H9c2 cells were seeded at a density of 5,000 cells per well on a 96-well plate and cultured with DMEM supplemented with 10% FBS for 48 h. Prior to fluorescence microscopy, the H9c2 cells were loaded with tetramethylrhodamine ethyl ester at a concentration of 125 nM and HOECHST 33423 at a concentration of 1 µg/ml for a 30-min incubation period at 37˚C in accordance with the manufacturer's instructions. Fluorescence intensity was measured in four randomly selected fields within each well and quantified using ImageJ 1.52a software (National Institutes of Health).

Western blot analysis

Western blotting was performed as previously described (25). Briefly, H9c2 cells were lysed using protein extraction reagent (cat. no. 78501; Thermo Fisher Scientific, Inc.). Protein concentrations were determined using Qubit™ Protein Assay Kits (Thermo Fisher Scientific, Inc.). Subsequently, 30 µg protein/lane was separated using 8% sodium dodecyl sulfate-polyacrylamide gel electrophoresis. This process was followed by the electrophoretic transfer of the separated proteins onto equilibrated polyvinylidene difluoride membranes. These membranes were then blocked with 5% skimmed milk for 1 h at room temperature. Following this blocking procedure, the membranes were incubated overnight at 4˚C with specific antibodies against total ACC2 (1:2,000; monoclonal; cat. no. ab45174; Abcam), p-(p-)ACC2 (1:500; polyclonal; cat. no. 07303; Millipore), total AMP-activated protein kinase (AMPK; 1:500; monoclonal; cat. no. 5831; Cell Signaling), p-AMPK (1:1,000; polyclonal; cat. no. 07681; Millipore), calcineurin (1:10,000; monoclonal; cat. no. ab109412; Abcam), total GSK-3β (1:1,000; monoclonal; cat. no. 9315; Cell Signaling), p-GSK-3β (1:1,000; polyclonal; cat. no. 9336; Cell Signaling) and GLUT4 (1:500; monoclonal; cat. no. sc-53566; Santa Cruz). After washing with PBS containing Tween 20 (0.1%) for 15 min at room temperature, a peroxidase-conjugated secondary antibody (anti-rabbit IgG; 1:1,000; cat. no. HAF008; or anti-mouse IgG; 1:500; cat. no. HAF007; R&D Systems, Inc.) was added for incubation for 1 h at room temperature. Bound antibodies were detected using an enhanced chemiluminescence detection system (MilliporeSigma) and the results were analyzed using AlphaEaseFC 4.0.0.34 software (ProteinSimple). Targeted bands were normalized to glyceraldehyde 3-phosphate dehydrogenase (1:50,000; monoclonal; cat. no. M171-1; MBL) or β-actin (1:10,000; polyclonal; cat. no. ab6274; Abcam) to confirm equal protein loading.

Statistical analysis

Quantitative data are presented as mean ± standard error of the mean. Statistical significance in H9c2 cells exposed to various conditions was determined using a one-way analysis of variance followed by Tukey's post hoc test. Statistical analysis was performed using SigmaPlot 12.3 software (Systat Software, Inc.). P<0.05 was considered to indicate a statistically significant difference.

Results

Effects of lithium on ACC2 in H9c2 cardiomyocytes

In H9c2 cardiomyocytes treated with 0.3 mM LiCl for 48 h, the expression of p-ACC2 was significantly downregulated compared with the control (Fig. 1A). However, LiCl at concentrations of 0.1 and 1.0 mM had only a minimal effect on the expression of p-ACC2. Notably, no significant differences were observed in the expression of total ACC2 between the control group and H9c2 cardiomyocytes treated with LiCl at concentrations of 0.1, 0.3 and 1 mM.

Effects of lithium on GSK-3β in H9c2 cardiomyocytes

As GSK-3β is a potential regulator of ACC2, it was investigated whether lithium regulated GSK-3β activity in H9c2 cardiomyocytes. Compared with the control, LiCl at a concentration of 0.3 mM upregulated the expression of p-GSK-3β by 55.4% (Fig. 1B). By contrast, LiCl at concentrations of 0.1 and 1.0 mM did not significantly affect the expression of p-GSK-3β in H9c2 cardiomyocytes. No significant differences in the expression of total GSK-3β were observed between the control group and H9c2 cardiomyocytes treated with LiCl at concentrations of 0.1, 0.3 and 1 mM. To confirm whether lithium downregulated the expression of p-ACC2 by modulating GSK-3β activity, H9c2 cardiomyocytes were treated with TWS119, a GSK-3β inhibitor. The expression of p-ACC2 in H9c2 cardiomyocytes was downregulated to a similar extent in cells subjected to combined treatment with TWS119 (8 µM) and LiCl (0.3 mM) and in those treated with TWS119 (8 µM) alone (Fig. 2). These findings suggest that lithium downregulated the expression of p-ACC2 through the modulation of GSK-3β activity. As the activities of ACC1 and ACC2 are regulated by numerous protein kinases and phosphatases, the present study also examined the effects of lithium on AMPK and calcineurin, the two principal protein kinases involved in the regulation of ACC2 activity in H9c2 cardiomyocytes. It was observed that LiCl did not significantly affect the expression level of total or p-AMPK or calcineurin in H9c2 cardiomyocytes (Fig. S2).

Effects of lithium on mitochondrial bioenergetic function in GLUT4-knockdown H9c2 cardiomyocytes

To explore the therapeutic potential of lithium for mitigating metabolic stress, mitochondrial bioenergetic function was evaluated in GLUT4-knockdown H9c2 cardiomyocytes treated with lithium. Compared with the control cells, the GLUT4-knockdown H9c2 cardiomyocytes exhibited greater proton leakage (Fig. 3). Additionally, the GLUT4-knockdown H9c2 cardiomyocytes exhibited greater reductions in coupling efficiency and the cell respiratory control ratio compared with the control cells. Furthermore, GLUT4-knockdown H9c2 cardiomyocytes treated with LiCl (0.3 mM) exhibited a reduction in proton leakage and improvement in coupling efficiency and cell respiratory control ratio compared with cells not treated with LiCl. These findings indicated that LiCl at a concentration of 0.3 mM enhanced mitochondrial bioenergetic function in GLUT4-knockdown H9c2 cardiomyocytes.

Effects of lithium on mitochondrial ROS and membrane potential in GLUT4-knockdown H9c2 cardiomyocytes

The effects of lithium on mitochondrial ROS and membrane potential in GLUT4-knockdown H9c2 cardiomyocytes are illustrated in Fig. 4. Compared with the control cells, the GLUT4-knockdown H9c2 cardiomyocytes exhibited higher levels of mitochondrial ROS. By contrast, the GLUT4-knockdown cells treated with LiCl (0.3 mM) had mitochondrial ROS levels similar to those in the control cells. Furthermore, the GLUT4-knockdown H9c2 cardiomyocytes exhibited reduced mitochondrial membrane potential in contrast to both the control cells and the LiCl (0.3 mM)-treated GLUT4-knockdown cells. These findings suggested that LiCl at a concentration of 0.3 mM attenuated mitochondrial ROS and restored the mitochondrial membrane potential in GLUT4-knockdown H9c2 cardiomyocytes.

Effects of lithium on mitochondrial fatty acid oxidation in GLUT4-knockdown H9c2 cardiomyocytes

The effects of lithium on mitochondrial fatty acid oxidation in GLUT4-knockdown H9c2 cardiomyocytes are depicted in Fig. 5. Compared with the control cells, the GLUT4-knockdown H9c2 cardiomyocytes exhibited a greater elevation in mitochondrial fatty acid oxidation. Additionally, when GLUT4-knockdown H9c2 cardiomyocytes were treated with LiCl (0.3 mM), they exhibited a greater reduction in mitochondrial fatty acid oxidation compared with GLUT4-knockdown H9c2 cardiomyocytes not treated with LiCl. These findings suggested that LiCl at a concentration of 0.3 mM attenuated the increase in mitochondrial fatty acid oxidation in GLUT4-knockdown H9c2 cardiomyocytes.

Discussion

Studies have suggested that ACC2 can be inactivated through phosphorylation (9,10). In the present study, for the first time to the best of the authors' knowledge, LiCl at a concentration of 0.3 mM, representing a low therapeutic level but not a supraphysiological level, was observed to downregulate the expression of p-ACC2 in H9c2 cardiomyocytes but not to affect the expression of total ACC2. The downregulation of p-ACC2 induced by lithium may activate ACC2 and subsequently elevate the level of malonyl-CoA (9). Such an increase in malonyl-CoA levels can inhibit carnitine palmitoyltransferase 1, which is crucial for the transport of long-chain fatty acyl-CoAs into the mitochondria for β-oxidation (26,27). Additionally, the present study also revealed that LiCl at a low therapeutic level attenuated mitochondrial fatty acid oxidation in H9c2 cardiomyocytes.

The inhibition of fatty acid oxidation has been demonstrated to have beneficial effects related to heart failure associated with diabetes mellitus (2,4,8,28,29). In patients with diabetes, heart failure often presents a shift in cardiac fuel substrate utilization toward an increased reliance on more mitochondrial fatty acid oxidation, a change driven by insulin resistance (1,6). Such an increase in fatty acid oxidation can overwhelm mitochondria, resulting in oxidative stress (6). The subsequent generation of ROS from lipid-overburdened mitochondria may worsen insulin resistance and accelerate the progression of heart failure (6). Thus, inhibiting fatty acid oxidation in patients with diabetic cardiomyopathy may offer cardioprotection by reducing oxidative stress, promoting glucose utilization and enhancing cardiac efficiency (6,30-32). Supporting this hypothesis, a previous study found that a low dose of lithium (0.36±0.03 mM) mitigated cardiac dysfunction in an experimental animal model to investigate sleep deprivation (33). The current study corroborated the cardioprotective potential of lithium in a GLUT4-knockdown cellular model designed to simulate insulin resistance (34-36). This finding agrees with previous findings on the benefits of lithium on cardiac metabolism (12-14,37). Nonetheless, failing hearts show varying mitochondrial fatty acid oxidation patterns, which could increase or decrease depending on the heart failure type (4). For example, in heart failure associated with conditions such as hypertension and ischemia, myocardial fatty acid oxidation tends to decline. Hence, the implications of the present findings might not extend to all types of heart failure given the variability in underlying disease processes.

ROS-induced proton leakage is mediated by adenine nucleotide translocase (38,39), a protein that is a central component of the mitochondrial permeability transition pore (40). The opening of the mitochondrial permeability transition pore, particularly under pathological conditions, leads to the dissipation of mitochondrial membrane potential, which in turn triggers the mitochondrial pathway of apoptosis (41). Studies have demonstrated that lithium exerts a cardioprotective effect by enhancing the threshold at which the mitochondrial permeability transition pore is activated by ROS (42,43). The present study demonstrated that low-dose lithium hinders ROS generation and proton leakage while restoring mitochondrial membrane potential in GLUT4-knockdown H9c2 cardiomyocytes.

GSK-3β signaling plays a pivotal role in the regulation of multiple mitochondrial functions, including energy bioenergetics, biogenesis and apoptosis (44). The inhibition of GSK-3β has been shown to reduce the apoptosis of cardiomyocytes and alleviate cardiac dysfunction (45-48). In the present study, LiCl at a concentration of 0.3 mM inhibited GSK-3β and activated ACC2 in H9c2 cardiomyocytes. Although studies have reported that lithium may regulate the activity of AMPK and calcineurin (49-52), the present study revealed that lithium at a concentration of 0.3 mM had no significant effect on the expression of p-AMPK or that of calcineurin in H9c2 cardiomyocytes. These findings suggested that the biological effects of lithium may be concentration dependent. As summarized in Fig. 6, the results of the present study suggested that at a low therapeutic concentration, lithium inhibits mitochondrial fatty acid utilization and mitigates oxidative stress in cardiomyocytes. These effects are probably achieved through the inhibition of GSK-3β and the activation of ACC2.

In the present study, lithium had a biphasic dose-response effect on ACC2 and GSK-3β activities. This phenomenon may stem from interactions among multiple signaling pathways targeted by lithium (53), resulting in dose-response curves that significantly differ from the monotonic curves typically seen in pharmaceuticals acting on specific receptors (54). Numerous studies have identified the biphasic dose-response effects of lithium on a wide range of signaling pathways in multiple cell types (55-58). To gain a comprehensive understanding of the mechanisms underlying the effects of lithium on mitochondrial energy metabolism in cardiomyocytes at the system level, future research involving large-scale multiomics data is warranted.

In conclusion, the findings of the present study indicate that lithium directly regulated mitochondrial fatty acid utilization and mitigated oxidative stress in cardiomyocytes at low therapeutic concentrations. These findings suggest that lithium possessed cardioprotective properties and may attenuate metabolic stress in the myocardium. Considering the inherent limitations of in vitro research, future in vivo studies are necessary to confirm the cardioprotective effects of lithium in metabolic stress.

Supplementary Material

Expression of GLUT4 and CPT1 in H9c2 cardiomyocytes after transfection with either negative control or GLUT4 siRNA. Relative to transfection with negative control, GLUT4 expression was downregulated by 43.1% after transfection with GLUT4 siRNA at 50 nM for 48 h. Additionally, the expression of CPT1, the mitochondrial enzyme responsible for the translocation of fatty acids from the cytosol to the mitochondrial matrix, where fatty acid oxidation occurs, was upregulated by 11.6% after transfection with GLUT4 siRNA at 50 nM. A paired t-test was conducted to compare the protein expression in H9c2 cardiomyocytes after transfection with negative control with that after transfection with GLUT4 siRNA. GLUT4, glucose transporter type 4; CPT1, carnitine palmitoyltransferase 1; siRNA, small interfering RNA; CON, control; p-, phosphorylated.
Expression of AMPK and calcineurin in H9c2 cardiomyocytes treated with LiCl. The expression of calcineurin and the total and p-forms of AMPK did not significantly differ between control cells (n=5) and H9c2 cardiomyocytes treated with 0.1 mM (n=5), 0.3 mM (n=5) and 1.0 mM (n=5) of LiCl. A one-way repeated measures analysis of variance was performed to compare protein expression under multiple treatment conditions. AMPK. adenosine monophosphate-activated protein kinase; CON, control; p-, phosphorylated.

Acknowledgements

Not applicable.

Funding

Funding: The present study was supported by research grants from the National Science and Technology Council of Taiwan (grant nos. NSTC 111-2314-B-038-061 and NSTC 112-2314-B-038-050), Taipei Medical University (grant no. TMU110-AE1-B19) and Wan Fang Hospital (grant no. 108-wf-swf-06). The National Science and Technology Council of Taiwan, Taipei Medical University and Wan Fang Hospital had no involvement in the study design; collection, analysis and interpretation of data; composition of the report; or decisions regarding article publication.

Availability of data and materials

The data generated in the present study may be requested from the corresponding author.

Authors' contributions

PHC, YHY, YHK and YJC conceived and designed the study. PHC, YHK and YJC conducted the experiments and analyzed the data. PHC, YHK and YJC confirm the authenticity of all the raw data. PHC and CCC acquired funding. PHC, TWL, SHL, TVH and CCC contributed to the data interpretation and prepared the manuscript. TVH assisted with data visualization and created graphs. PHC composed the first draft of the manuscript. YHK, YHY and YJC reviewed and edited the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Bertero E and Maack C: Metabolic remodelling in heart failure. Nat Rev Cardiol. 15:457–470. 2018.PubMed/NCBI View Article : Google Scholar

2 

Fillmore N, Mori J and Lopaschuk GD: Mitochondrial fatty acid oxidation alterations in heart failure, ischaemic heart disease and diabetic cardiomyopathy. Br J Pharmacol. 171:2080–2090. 2014.PubMed/NCBI View Article : Google Scholar

3 

Lesnefsky EJ, Chen Q and Hoppel CL: Mitochondrial metabolism in aging heart. Circ Res. 118:1593–611. 2016.PubMed/NCBI View Article : Google Scholar

4 

Lopaschuk GD, Karwi QG, Tian R, Wende AR and Abel ED: Cardiac energy metabolism in heart failure. Circ Res. 128:1487–1513. 2021.PubMed/NCBI View Article : Google Scholar

5 

Marin W, Marin D, Ao X and Liu Y: Mitochondria as a therapeutic target for cardiac ischemia-reperfusion injury (Review). Int J Mol Med. 47:485–499. 2021.PubMed/NCBI View Article : Google Scholar

6 

Fukushima A and Lopaschuk GD: Cardiac fatty acid oxidation in heart failure associated with obesity and diabetes. Biochim Biophys Acta. 1861:1525–1534. 2016.PubMed/NCBI View Article : Google Scholar

7 

Lkhagva B, Lee TW, Lin YK, Chen YC, Chung CC, Higa S and Chen YJ: Disturbed cardiac metabolism triggers atrial arrhythmogenesis in diabetes mellitus: Energy substrate alternate as a potential therapeutic intervention. Cells. 11(2915)2022.PubMed/NCBI View Article : Google Scholar

8 

Zuurbier CJ, Bertrand L, Beauloye CR, Andreadou I, Ruiz-Meana M, Jespersen NR, Kula-Alwar D, Prag HA, Eric Botker H, Dambrova M, et al: Cardiac metabolism as a driver and therapeutic target of myocardial infarction. J Cell Mol Med. 24:5937–5954. 2020.PubMed/NCBI View Article : Google Scholar

9 

Tong L: Structure and function of biotin-dependent carboxylases. Cell Mol Life Sci. 70:863–891. 2013.PubMed/NCBI View Article : Google Scholar

10 

Brownsey RW, Boone AN, Elliott JE, Kulpa JE and Lee WM: Regulation of acetyl-CoA carboxylase. Biochem Soc Trans. 34(Pt 2):223–227. 2006.PubMed/NCBI View Article : Google Scholar

11 

Malhi GS, Gessler D and Outhred T: The use of lithium for the treatment of bipolar disorder: Recommendations from clinical practice guidelines. J Affect Disord. 217:266–280. 2017.PubMed/NCBI View Article : Google Scholar

12 

Machado-Vieira R: Lithium, stress and resilience in bipolar disorder: Deciphering this key homeostatic synaptic plasticity regulator. J Affect Disord. 233:92–99. 2018.PubMed/NCBI View Article : Google Scholar

13 

Ochoa ELM: Lithium as a neuroprotective agent for bipolar disorder: An overview. Cell Mol Neurobiol. 42:85–97. 2022.PubMed/NCBI View Article : Google Scholar

14 

Singulani MP, De Paula VJR and Forlenza OV: Mitochondrial dysfunction in Alzheimer's disease: Therapeutic implications of lithium. Neurosci Lett. 760(136078)2021.PubMed/NCBI View Article : Google Scholar

15 

Castillo-Quan JI, Li L, Kinghorn KJ, Ivanov DK, Tain LS, Slack C, Kerr F, Nespital T, Thornton J, Hardy J, et al: Lithium promotes longevity through GSK3/NRF2-dependent hormesis. Cell Rep. 15:638–650. 2016.PubMed/NCBI View Article : Google Scholar

16 

Zarse K, Terao T, Tian J, Iwata N, Ishii N and Ristow M: Low-dose lithium uptake promotes longevity in humans and metazoans. Eur J Nutr. 50:387–389. 2011.PubMed/NCBI View Article : Google Scholar

17 

Chen PH, Hsiao CY, Chiang SJ, Shen RS, Lin YK, Chung KH and Tsai SY: Cardioprotective potential of lithium and role of fractalkine in euthymic patients with bipolar disorder. Aust N Z J Psychiatry. 57:104–114. 2023.PubMed/NCBI View Article : Google Scholar

18 

Chen PH, Hsiao CY, Chiang SJ and Tsai SY: Association of lithium treatment with reduced left ventricular concentricity in patients with bipolar disorder. Psychiatry Clin Neurosci. 77:59–61. 2023.PubMed/NCBI View Article : Google Scholar

19 

Snitow ME, Bhansali RS and Klein PS: Lithium and therapeutic targeting of GSK-3. Cells. 10(255)2011.PubMed/NCBI View Article : Google Scholar

20 

Domoto T, Pyko IV, Furuta T, Miyashita K, Uehara M, Shimasaki T, Nakada M and Minamoto T: Glycogen synthase kinase-3β is a pivotal mediator of cancer invasion and resistance to therapy. Cancer Sci. 170:1363–1372. 2016.PubMed/NCBI View Article : Google Scholar

21 

Terrand J, Bruban V, Zhou L, Gong W, El Asmar Z, May P, Zurhove K, Haffner P, Philippe C, Woldt E, et al: LRP1 controls intracellular cholesterol storage and fatty acid synthesis through modulation of Wnt signaling. J Biol Chem. 284:381–388. 2009.PubMed/NCBI View Article : Google Scholar

22 

Lkhagva B, Kao YH, Lee TI, Lee TW, Cheng WL and Chen YJ: Activation of Class I histone deacetylases contributes to mitochondrial dysfunction in cardiomyocytes with altered complex activities. Epigenetics. 13:376–385. 2018.PubMed/NCBI View Article : Google Scholar

23 

Brand MD and Nicholls DG: Assessing mitochondrial dysfunction in cells. Biochem J. 435:297–312. 2011.PubMed/NCBI View Article : Google Scholar

24 

Nisr RB and Affourtit C: Insulin acutely improves mitochondrial function of rat and human skeletal muscle by increasing coupling efficiency of oxidative phosphorylation. Biochim Biophys Acta. 1837:270–276. 2014.PubMed/NCBI View Article : Google Scholar

25 

Chen PH, Chung CC, Lin YF, Kao YH and Chen YJ: Lithium reduces migration and collagen synthesis activity in human cardiac fibroblasts by inhibiting store-operated Ca2+ entry. Int J Mol Sci. 22(842)2021.PubMed/NCBI View Article : Google Scholar

26 

Eaton S: Control of mitochondrial beta-oxidation flux. Prog Lipid Res. 41:197–239. 2022.PubMed/NCBI View Article : Google Scholar

27 

Schlaepfer IR and Joshi M: CPT1A-mediated fat oxidation, mechanisms and therapeutic potential. Endocrinology. 161(bqz046)2020.PubMed/NCBI View Article : Google Scholar

28 

McCarthy CP, Mullins KV and Kerins DM: The role of trimetazidine in cardiovascular disease: Beyond an anti-anginal agent. Eur Heart J Cardiovasc Pharmacother. 2:266–272. 2016.PubMed/NCBI View Article : Google Scholar

29 

Zhao C, Jin C, He X and Xiang M: The efficacy of trimetazidine in non-ischemic heart failure patients: A meta-analysis of randomized controlled trials. Rev Cardiovasc Med. 22:1451–1459. 2021.PubMed/NCBI View Article : Google Scholar

30 

Karwi QG, Sun Q and Lopaschuk GD: The contribution of cardiac fatty acid oxidation to diabetic cardiomyopathy severity. Cells. 10(3259)2021.PubMed/NCBI View Article : Google Scholar

31 

Shu H, Peng Y, Hang W, Zhou N and Wang DW: Trimetazidine in heart failure. Front Pharmacol. 11(569132)2021.PubMed/NCBI View Article : Google Scholar

32 

Sung MM, Hamza SM and Dyck JR: Myocardial metabolism in diabetic cardiomyopathy: Potential therapeutic targets. Antioxid Redox Signal. 22:1606–1630. 2015.PubMed/NCBI View Article : Google Scholar

33 

Chen PH, Chung CC, Liu SH, Kao YH and Chen YJ: Lithium treatment improves cardiac dysfunction in rats deprived of rapid eye movement sleep. Int J Mol Sci. 23(11226)2022.PubMed/NCBI View Article : Google Scholar

34 

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

35 

Riehle C and Abel ED: Insulin signaling and heart failure. Circ Res. 118:1151–1169. 2016.PubMed/NCBI View Article : Google Scholar

36 

Saotome M, Ikoma T, Hasan P and Maekawa Y: Cardiac insulin resistance in heart failure: The role of mitochondrial dynamics. Int J Mol Sci. 20(3552)2019.PubMed/NCBI View Article : Google Scholar

37 

Chen PH, Chao TF, Kao YH and Chen YJ: Lithium interacts with cardiac remodeling: The fundamental value in the pharmacotherapy of bipolar disorder. Prog Neuropsychopharmacol Biol Psychiatry. 88:208–214. 2019.PubMed/NCBI View Article : Google Scholar

38 

Cheng J, Nanayakkara G, Shao Y, Cueto R, Wang L, Yang WY, Tian Y, Wang H and Yang X: Mitochondrial proton leak plays a critical role in pathogenesis of cardiovascular diseases. Adv Exp Med Biol. 982:359–370. 2017.PubMed/NCBI View Article : Google Scholar

39 

Nadtochiy SM, Tompkins AJ and Brookes PS: Different mechanisms of mitochondrial proton leak in ischaemia/reperfusion injury and preconditioning: Implications for pathology and cardioprotection. Biochem J. 395:611–618. 2006.PubMed/NCBI View Article : Google Scholar

40 

Carrer A, Laquatra C, Tommasin L and Carraro M: Modulation and pharmacology of the mitochondrial permeability transition: A journey from F-ATP synthase to ANT. Molecules. 26(6463)2021.PubMed/NCBI View Article : Google Scholar

41 

Bonora M, Giorgi C and Pinton P: Molecular mechanisms and consequences of mitochondrial permeability transition. Nat Rev Mol Cell Biol. 23:266–285. 2022.PubMed/NCBI View Article : Google Scholar

42 

Juhaszova M, Zorov DB, Kim SH, Pepe S, Fu Q, Fishbein KW, Ziman BD, Wang S, Ytrehus K, Antos CL, et al: Glycogen synthase kinase-3beta mediates convergence of protection signaling to inhibit the mitochondrial permeability transition pore. J Clin Invest. 113:1535–1549. 2004.PubMed/NCBI View Article : Google Scholar

43 

Juhaszova M, Zorov DB, Yaniv Y, Nuss HB, Wang S and Sollott SJ: Role of glycogen synthase kinase-3beta in cardioprotection. Circ Res. 104:1240–1252. 2009.PubMed/NCBI View Article : Google Scholar

44 

Yang K, Chen Z, Gao J, Shi W, Li L, Jiang S, Hu H, Liu Z, Xu D and Wu L: The key roles of GSK-3β in regulating mitochondrial activity. Cell Physiol Biochem. 44:1445–1459. 2017.PubMed/NCBI View Article : Google Scholar

45 

Hirotani S, Zhai P, Tomita H, Galeotti J, Marquez JP, Gao S, Hong C, Yatani A, Avila J and Sadoshima J: Inhibition of glycogen synthase kinase 3beta during heart failure is protective. Circ Res. 101:1164–1174. 2007.PubMed/NCBI View Article : Google Scholar

46 

Murphy E and Steenbergen C: Inhibition of GSK-3beta as a target for cardioprotection: The importance of timing, location, duration and degree of inhibition. Expert Opin Ther Targets. 9:447–456. 2005.PubMed/NCBI View Article : Google Scholar

47 

Sharma AK, Bhatia S, Al-Harrasi A, Nandave M and Hagar H: Crosstalk between GSK-3β-actuated molecular cascades and myocardial physiology. Heart Fail Rev. 26:1495–1504. 2021.PubMed/NCBI View Article : Google Scholar

48 

Sharma AK, Thanikachalam PV and Bhatia S: The signaling interplay of GSK-3β in myocardial disorders. Drug Discov Today. 25:633–641. 2020.PubMed/NCBI View Article : Google Scholar

49 

Bao H, Zhang Q, Liu X, Song Y, Li X, Wang Z, Li C, Peng A and Gong R: Lithium targeting of AMPK protects against cisplatin-induced acute kidney injury by enhancing autophagy in renal proximal tubular epithelial cells. FASEB J. 33:14370–14381. 2019.PubMed/NCBI View Article : Google Scholar

50 

Lee Y, Kim SM, Jung EH, Park J, Lee JW and Han IO: Lithium chloride promotes lipid accumulation through increased reactive oxygen species generation. Biochim Biophys Acta Mol Cell Biol Lipids. 1865(158552)2020.PubMed/NCBI View Article : Google Scholar

51 

Liu P, Zhang Z, Wang Q, Guo R and Mei W: Lithium chloride facilitates autophagy following spinal cord injury via ERK-dependent pathway. Neurotox Res. 32:535–543. 2017.PubMed/NCBI View Article : Google Scholar

52 

Qu Z, Sun D and Young W: Lithium promotes neural precursor cell proliferation: Evidence for the involvement of the non-canonical GSK-3β-NF-AT signaling. Cell Biosci. 1(18)2011.PubMed/NCBI View Article : Google Scholar

53 

Jakobsson E, Argüello-Miranda O, Chiu SW, Fazal Z, Kruczek J, Nunez-Corrales S, Pandit S and Pritchet L: Towards a unified understanding of lithium action in basic biology and its significance for applied biology. J Membr Biol. 250:587–604. 2017.PubMed/NCBI View Article : Google Scholar

54 

Parris GE: A hypothesis concerning the biphasic dose-response of tumors to angiostatin and endostatin. Dose Response 13: dose-response. 14-020. Parris, 2015.

55 

Ge W and Jakobsson E: Systems biology understanding of the effects of lithium on cancer. Front Oncol. 9(296)2019.PubMed/NCBI View Article : Google Scholar

56 

Erguven M, Oktem G, Kara AN and Bilir A: Lithium chloride has a biphasic effect on prostate cancer stem cells and a proportional effect on midkine levels. Oncol Lett. 12:2948–2955. 2016.PubMed/NCBI View Article : Google Scholar

57 

Suganthi M, Sangeetha G, Gayathri G and Ravi Sankar B: Biphasic dose-dependent effect of lithium chloride on survival of human hormone-dependent breast cancer cells (MCF-7). Biol Trace Elem Res. 150:477–486. 2012.PubMed/NCBI View Article : Google Scholar

58 

Gao XM, Fukamauchi F and Chuang DM: Long-term biphasic effects of lithium treatment on phospholipase C-coupled M3-muscarinic acetylcholine receptors in cultured cerebellar granule cells. Neurochem Int. 22:395–403. 1993.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

April-2024
Volume 27 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen P, Lee T, Liu S, Huynh T, Chung C, Yeh Y, Kao Y and Chen Y: Lithium downregulates phosphorylated acetyl‑CoA carboxylase 2 and attenuates mitochondrial fatty acid utilization and oxidative stress in cardiomyocytes. Exp Ther Med 27: 126, 2024
APA
Chen, P., Lee, T., Liu, S., Huynh, T., Chung, C., Yeh, Y. ... Chen, Y. (2024). Lithium downregulates phosphorylated acetyl‑CoA carboxylase 2 and attenuates mitochondrial fatty acid utilization and oxidative stress in cardiomyocytes. Experimental and Therapeutic Medicine, 27, 126. https://doi.org/10.3892/etm.2024.12413
MLA
Chen, P., Lee, T., Liu, S., Huynh, T., Chung, C., Yeh, Y., Kao, Y., Chen, Y."Lithium downregulates phosphorylated acetyl‑CoA carboxylase 2 and attenuates mitochondrial fatty acid utilization and oxidative stress in cardiomyocytes". Experimental and Therapeutic Medicine 27.4 (2024): 126.
Chicago
Chen, P., Lee, T., Liu, S., Huynh, T., Chung, C., Yeh, Y., Kao, Y., Chen, Y."Lithium downregulates phosphorylated acetyl‑CoA carboxylase 2 and attenuates mitochondrial fatty acid utilization and oxidative stress in cardiomyocytes". Experimental and Therapeutic Medicine 27, no. 4 (2024): 126. https://doi.org/10.3892/etm.2024.12413