ABSTRACT
Among the branched-chain amino acids, leucine and isoleucine have been well studied for their roles in improving mitochondrial function and reducing oxidative stress. However, role of valine in mitochondrial function regulation and oxidative stress management remains elusive. This study investigated valine effect on mitochondrial function and oxidative stress in vitro. Valine increased expression of genes involved in mitochondrial biogenesis and dynamics. It upregulates mitochondrial function at complexes I, II, and IV levels of electron transport chain. Flow cytometry studies revealed, valine reduced oxidative stress by significantly lowering mitochondrial reactive oxygen species and protein expression of 4-hydroxynonenal. Functional role of valine against oxidative stress was analyzed by XFe96 Analyzer. Valine sustained oxidative phosphorylation and improved ATP generation rates during oxidative stress. In conclusion, our findings shed more light on the critical function of valine in protecting mitochondrial function thereby preventing mitochondrial/cellular damage induced by oxidative stress.
Keywords: valine, mitochondrial function, oxidative stress, reactive oxygen species
Graphical Abstract
Graphical Abstract.
Valine improves mitochondrial function and protects against oxidative stress. It modulates oxidative phosphorylation, ATP production, and inhibits mitochondrial dysfunction by reducing reactive oxygen species (ROS) production.
Branched-chain amino acids (BCAAs) are among the most prevalent essential amino acids that are pivotal for the health of the human body (Nie et al.2018). Leucine, isoleucine, and valine are α-amino acids with aliphatic side chains that are important for maintaining the body's nitrogen balance (Doi et al.2003) as well as skeletal muscle cell growth and development and have been linked to multiple metabolic processes, that are important in cellular pathways, and are central in the relationship between diet, health, and aging (Wolfe 2017; Green and Lamming 2019; Vanweert et al.2022). The activation of pathways including mammalian target of rapamycin, phosphoinositide 3-kinase-protein kinase B (PI3K-AKT), and maintenance of physiological metabolic health are all influenced by these amino acids (Viana et al.2019; Mann et al.2021). Aside from helping with protein synthesis, they also aid in translation and degradation (Tom and Nair 2006). The potential of BCAAs as dietary supplements for treating various disorders has been studied in both human and mouse models and has been shown to influence metabolic activity, improve mitochondrial bioenergetics, support muscle growth, and maintain mitochondrial health (Lynch and Adams 2014; Holecek 2018; Ruocco et al. 2021). Mitochondria are crucial for producing adenosine triphoshate (ATP) and also participate in fatty acid oxidation, cell survival, and apoptosis (Hewton et al.2021; Yoneshiro et al.2021). The activation of multiple genes that regulate mitochondrial function, including peroxisome proliferator-activated receptor gamma (PPAR-γ) coactivator-1 alpha (PGC-1α), PPAR-gamma coactivator-1 beta (PGC-1β), and the mitofusins, by BCAAs, in turn stimulates mitochondrial biogenesis. According to several studies (Hinkle et al.2022), mitochondrial dysfunction is associated with oxidative stress, cellular damage and is thought to be a common underlying mechanism of metabolic diseases (Rocha et al.2013; Walters et al.2016). Extreme reactive oxygen species (ROS) production can hinder mitochondrial membrane permeability, and produce lipid peroxidation products, which leads to mitochondrial-related apoptosis (Ren et al.2017; Zhao et al.2019). In skeletal muscle, ROS generation induces a signaling cascade that can produce muscle damage (Lee et al.2017; Pan and Chen 2021). It has been demonstrated that adding BCAA supplements lengthens the average lifespan of mice by boosting the ROS defense mechanism in the skeletal and cardiac muscle tissues and improving mitochondrial biogenesis (Valerio et al.2011). BCAAs alone or together can mediate the antioxidant defense system and reduce oxidative stress in skeletal muscles in middle-aged mice (D'Antona et al.2010; Cruzat et al.2014). Valine (Val), a necessary BCAA, can also act as an energetic molecule and is important for protein synthesis and growth of skeletal muscle (Kohlmeier 2003). Further, valine also plays a favorable function in lipid metabolism (Bishop et al.2020) and have inflammatory properties (Gart et al.2022).
While studies have been conducted on BCAAs, especially leucine and isoleucine on mitochondrial function, cellular metabolism and oxidative stress (Mattick et al.2013; Tamanna and Mahmood 2014), there is a dearth of literature on the cellular and functional importance of valine. In the present study, we evaluated the effect of valine treatment on mitochondrial function and its response to oxidative stress. Our findings demonstrate that valine administration improved oxygen consumption rate (OCR) and overall ATP generation in the mitochondria. Additionally, valine treatment also enhanced the transcriptional activity of PGC-1α, PGC-1β, and other genes that regulate mitochondrial function. Valine significantly reduced ROS production, thereby maintaining the OCR during H2O2-mediated oxidative stress. The findings from this study enhance our understanding of valine as an important nutrient to improve mitochondrial function, thereby protecting cells against oxidative damage.
Materials and methods
Cell culture and cell proliferation assay
The C2C12 mouse cell line used in this study was purchased from the American Type Culture Collection (ATCC, CRL-1772). Cell culture reagents were obtained from Thermo Fischer Scientific as previously described (Rogers et al.2013). Valine was obtained from Sigma–Aldrich, St. Louis, MO, USA, (V0500), and used following the manufacturer's protocol. C2C12 cell viability after valine treatment was determined by using MTS reagent (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) (Abcam; ab197010). A 200 µL C2C12 cell suspension (5 × 103/well) was seeded in a 96-well plate. At 70%-80% confluency, cells were treated with two different concentrations (1.0 and 1.5 m m) of valine for 24 h. A volume of 20 µL/well MTS reagent was added in each well and incubated for a period of 0.5-4 h at 37°C following the manufactures’ protocol. The absorbance was recorded at 490 nm.
Measurement of ATP and NAD/NADH levels
C2C12 cells (5 × 103 cells/well) were plated in 96-well plate, treated with 1.0 m m concentration of valine for 24 h. Intracellular ATP levels were assessed using the CellTiter-Glo 2.0 assay kit (Promega, Madison, WI, USA; G9242). Further, the NAD/NADH (nicotinamide adenine dinucleotide) levels after valine treatment were determined using a NAD/NADH Glo Assay Kit (Promega, Madison, WI, USA; G9071) following the manufacturer's protocol.
RNA isolation and quantitative reverse-transcriptase PCR
Cells were pre-treated with 1.0 m m concentration of valine for 24 h. Total cellular RNA was extracted from cells utilizing the RNeasy Kit (Qiagen) in accordance with the manufacturer's protocol. Total RNA obtained was then revere transcribed to Complementary DNA (cDNA) using cDNA Reverse Transcription Kit (Applied Biosystems, Thermo-Fischer Scientific, Waltham, MA, USA; 4368814). The real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) amplification was performed using SYBR Green master mix (Applied Biosystems, Thermo Fischer Scientific, Waltham, MA, USA; 4344463) in a QuantStudioTM 3 Real-Time PCR instrument (Applied Biosystems). The relative expression was measured using StepOne software. All experiments were performed as described previously (Rogers et al.2013). Primer set used for qPCR were listed in Table S1.
Western blot analysis
C2C12 cells were cultured and treated with valine as mentioned above and protein concentration was determined by using a BCA protein assay kit (Thermo–Fisher, Waltham, MA, USA). Western blotting was conducted as previously described (Zhang et al.2012). The antibodies used in the study were PGC-1α (sc-518025) and anti-4HNE antibody (ab46545), anti-mouse HRP (Invitrogen, Carlsbad, CA, USA; 62-6520) and anti-rabbit AP (Bio-Rad, Hercules, CA, USA; 64251130). iBright™ CL1500 (Invitrogen) was used to visualized the protein signal and ImageJ software v1.53t (National Institutes of Health, Bethesda, MD, USA) was used for quantification of protein expression.
Extracellular flux analysis
C2C12 cells were seeded in XFe96 Well plates at a density of 1.2 × 104 cells/well (Seahorse Bioscience, Billerica, MA, USA). The cells were then treated with 1.0 m m concentration of valine for 24 h respectively. After valine treatment, Seahorse XFe96 Extracellular Flux Analyzer was used to measure OCR using XF Cell Mito Stress Test Kit (Agilent, Santa Clara, CA, USA; 103015-100) and extracellular acidification rate (ECAR) using XF Glycolytic Rate Assay Kit (Agilent, Santa Clara, CA, USA; 103344-100). For determination of ATP production rate from mitochondria and glycolysis, XF-Real-time ATP Assay Kit (Agilent, Santa Clara, CA, USA; 103592-100). All metabolic assays were performed following the procedure from the manufacturer.
High-resolution respirometry
The mitochondrial respiration in the intact C2C12 cells (1 × 106/sample) after valine treatment was determined using Oxygraph-O2K high-resolution respirometer (Oroboros Instruments GmbH, Innsbruck, Austria) (Raiteri et al.2021). The mitochondrial respiratory activity at different complexes was analyzed using substrate-uncoupler-inhibitor-titration protocol as described in (Patyal et al.2022). The OCR from all mitochondrial complex was expressed as oxygen flux (pmol/s*Million Cells). DatLab 6.2 software (Innsbruck, Austria) was used to perform the data analysis.
Oxidative stress analysis
C2C12 cells were seeded at a density of 5 × 103 cells/mL (200 µL) in 96-well plate. H2O2 was obtained from Sigma–Aldrich, St. Louis, MO, USA (88 597). A volume of 200-1000 µmol/L treatment of H2O2 generates oxidative stress in C2C12 cells as previously reported (Li et al.2020). Cells were grown overnight and next day exposed to different concentrations of H2O2 (200, 400, 600, and 800 µmol/L) to stimulate oxidative stress for 6 h (Wu et al.2022). A total of 400 µmol/L concentration of H2O2 was selected as the final concentration for treatment. After H2O2 treatment, cells were treated with 1.0 m m concentration of valine for 24 h to assess cell proliferation viability as described above. Four groups used in this and subsequent experiments were control group (without any treatment), valine treated group, H2O2 treated group, H2O2 + valine treatment group.
Flow cytometry analysis
MitoSOXTM Red (Thermo Fisher, Eugene, OR, USA; M3600) was used to detect the mitochondrial ROS production. The C2C12 cells (1 × 106 cells/well), were plated in 6-well culture plate and treated with 400 µmol/L H2O2 for 6 h. Following H2O2 treatment, cells were treated with a final concentration of 1.0 m m of valine for 24 h. After 24 h, cells were incubated with 5 µm of MitoSox Red for 10 min at 37 °C as previously described (Verma et al.2023). Flow cytometer (BD LSRFortessaTM Cell Analyzer, Franklin Lakes, NJ, USA) was used to quantify the fluorescence intensity and data were processed by FlowJo_v10.8.1 software.
Statistical analysis
Results obtained from this study were described as mean ± standard deviation. The 2-tailed Student's t-test was used to determine the statistical significance between two groups. Multiple groups were ascertained by using one-way analysis of variance (ANOVA) followed Tukey's multiple comparison test. All the P values < 0.05 were considered to indicate as statistically significant. GraphPad Prism 9.11 Software Inc. (Dotmatics, Boston, MA, USA) was utilized to process the statistical analysis.
Results
Valine increases ATP and NAD/NADH production
Valine treatment at two different concentrations (1.0 and 1.5 m m) for 24 h, showed no significant effect on C2C12 cells viability as compared to control as shown in Figure S1. We next investigated the effect of valine treatment on ATP and NAD/NADH production rate. The treatment of valine at 1.0 m m concentration for 24 h, resulted in significantly higher ATP and NAD/NADH production rates (Figure S2a and b). Therefore, we selected this concentration for further experiments.
Valine impacts genes that regulate mitochondrial biogenesis and function
The mRNA expression levels of PGC-1α and PGC-1β (Figure 1a) were upregulated following valine treatment. The protein expression of PGC-1α was also found to be significantly higher in the valine-treated cells (Figure 1b). Further, valine treatment also upregulated the expression levels of mitofusin-1 (MFN1), mitofusin-2 (MFN2), and mitochondrial fission 1 (Fis1), whereas no difference was observed on optic atrophy-1 (Opa 1) gene expression (Figure 1c).
Figure 1.
Valine treatment upregulated expression of important mitochondrial genes in C2C12 cells (a) The mRNA expression levels of PGC1-α and PGC1-β were determined using Real-time RT-qPCR (b) The protein levels of PGC-1α was determined using western blotting analysis with gleyecelarldehyde 3-phosphate dehydrogenase (GAPDH) being used as loading control (c) Gene expression levels of MFN1, MFN2, and Fis1 were increased with no change in Opa1 levels (n = 3). *P < 0.05, **P < 0.01, ns: P > 0.05.
Valine distinctively regulates mitochondrial bioenergetics
Valine treatment resulted in increased OCR production and, therefore improved the mitochondrial function in C2C12 cells. Valine treatment elevated all of the different domains of OCR such as basal respiration, maximal respiratory capacity, and spare respiratory capacity (Figure 2a). ECAR analysis resulted in reduced basal glycolysis and compensatory glycolysis following valine treatment (Figure 2b). However, valine treatment significantly increased the mitochondrial ATP production rate, due to an increase in the oxidative phosphorylation (OXPHOS) activity (Figure 2c).
Figure 2.
Valine treatment increased OCR and total ATP and reduced glycolytic rate (ECAR) in C2C12 cells. (a) Valine upregulated the basal respiration, maximum respiration, and spare respiratory capacity in C2C12 cells. (b) In ECAR, both basal and compensatory glycolysis were significantly reduced following treatment. (c) Total ATP production was significantly upregulated in oxidative phosphorylation (mitochondrial-ATP) but showed no change in glycolysis (glycolysis-ATP) (n = 4). *P < 0.05, **P < 0.01, ***P < 0.001, ns: P > 0.05.
Valine specifically regulates the mitochondrial respiratory chain complex levels
Oxygraph-2 K (O2K) was used to measure the mitochondrial function by analyzing the high-resolution respiratory capacity in C2C12 cells. Valine treatment significantly increased the basal respiration and the maximal capacity of mitochondrial electron transport chain (ETC) (Figure 3a) in C2C12 cells. We then further assessed the effect of valine treatment on different complexes of the ETC. The respiration rates of complexes I (NADH/ubiquinone oxidoreductase), II (succinate dehydrogenase), and IV (cytochrome c oxidase) increased after treatment. No change was observed in complex III activity (cytochrome c reductase) (Figure 3b).
Figure 3.
High-resolution respiratory analysis showed an increased mitochondrial respiration following valine treatment. (a) C2C12 cells were treated with 0.1 m m concentration of valine for 24 h. Basal respiration and maximum ET capacity were increased following the treatment (b) Measurement of OCR at different complexes of ETC showed increase in complexes I, II, and IV activity after treatment (n = 3). * P < 0.05, **P < 0.01, ***P < 0.001, ns: P > 0.05.
Valine reduced mitochondrial ROS production induced by H2O2
We first examined the effect of H2O2 treatment on the cellular viability of C2C12 cells. The results showed that treatment of H2O2 at different concentrations for 6 h affected cell viability in dose-dependent manner (Figure S3). We selected 400 µmol/L concentration of H2O2 as our final treatment, as the recovery rate at this dose was found to be higher after valine treatment as compared to the other higher concentration used (Figure S4). The results showed that valine treatment significantly recovered the cell viability in C2C12 cells affected by H2O2 treatment (Figure S5). Further, we observed that H2O2 treatment significantly upregulate the ROS production. Our results showed that treatment of valine, along with H2O2, prevented ROS production (Figure 4a). In addition, H2O2 treatment also increased the protein expression level of 4-hydroxynonenal (4-HNE), which was prevented by valine treatment.
Figure 4.
Valine reduced mitochondrial ROS production in C2C12 cells. (a) ROS generation was evaluated using a flow cytometer. H2O2 treatment caused ROS production in 55.6% of cells, whereas, when H2O2 was used along with valine, recovery from oxidative stress was improved and only 28.8% of cells were ROS positive. (b) The protein expression of 4-HNE antibody was assessed by western blotting technique with β-actin as a loading control (n = 3). ****P < 0.0001.
Valine protects against oxidative stress and metabolic alterations
To confirm the protective role of valine against oxidative stress, we next assessed its impact on oxidative phosphorylation. Data were normalized to remove variation due to cell density before the experiment. H2O2 treatment initially repressed basal respiration, maximal respiratory capacity and spare respiratory capacity (Figure 5a). Valine treatment alone and in the presence of H2O2 significantly improved the maximal respiratory capacity and spare respiratory capacity. Additionally, we also determined the effect of H2O2 on total ATP production rate. H2O2 treatment significantly reduced the mitochondrial ATP rate, which was restored and improved by valine treatment (Figure 5b).
Figure 5.
Protective role of valine against oxidative stress. (a) OCR, Maximum respiration and spare respiratory capacity was repressed due to oxidative stress after H2O2 treatment, which was restored after valine treatment. (b) Total ATP production was decreased in both oxidative phosphorylation and glycolysis, which was recovered after valine treatment (n = 4). *P < 0.05, **P < 0.01, ***P < 0.001, **** P < 0.0001, ns: P > 0.05.
Discussion
This study has three major findings: first, valine plays a crucial role in improvement of cellular mitochondrial function; second, valine has a protective effect against oxidative stress by minimizing the production of mitochondrial ROS; and third, valine maintains oxidative phosphorylation and ATP rate during oxidative stress. Clinically, BCAAs intake have been suggested to reduce sarcopenia in older adults and possibly improve glucose metabolism, potentially by increasing mitochondrial biogenesis and muscle cellular function (Yoon 2016; Kitajima et al.2018). Despite many studies demonstrating the correlation between BCAAs and insulin resistance (Schnuck et al.2016), valine was reported not to cause insulin sensitivity or worsen insulin resistance (Rivera et al.2020; Mao et al.2022). We are among the first to report on the key role of the BCAA, valine, in enhancing mitochondrial function and reducing oxidative stress in a skeletal muscle cell line. Our study focused on exploring whether valine could be utilized to improve mitochondrial function. We analyzed the effect of valine treatment on cell viability of C2C12 cells. Valine treatment showed no change in cell viability. Our observation is consistent with no effect of varying concentrations (0.5 m m–2 m m) of valine for up to 48 h of treatment in C2C12 cells (Rivera et al.2020).
Our study suggests that mitochondrial biogenesis is improved with valine treatment. PGC-1α and PGC-1β are the main transcriptional coactivators of oxidative phosphorylation in mitochondria and also regulate mitochondrial biogenesis and function (Kang and Li 2012). Other studies have shown that leucine stimulates expression of PGC-1α and PGC-1β to promote mitochondrial biogenesis (Liang et al.2014; Johnson et al.2018; Rivera et al.2020; Ye et al.2020). Similarly, we found that valine regulates mitochondrial biogenesis and dynamics by increasing the expression levels of PGC-1α, PGC-1β, MFN1, MFN2, and Fis1. Mitochondria play a predominant role in regulating energy metabolism, ATP production and oxygen consumption and for the maintenance of cellular functions (Chu et al.2021). The increased OCR is an important indicator, which reflects the mitochondrial respiratory chain complex activity (du Plessis et al.2015). Our data showed increased OCR and reduced ECAR in C2C12 cells following valine treatment. This reduction in ECAR could be due to extreme oxidation of fatty acids, which inactivate pyruvate dehydrogenase and obstruct the normal process of glycolysis (Savage et al.2007). Others have reported that valine did not alter mitochondrial biogenesis or glycolysis (Rivera et al.2020). We also observed an increase in total ATP production in C2C12 cells following valine treatment, which emphasized the crucial role mitochondrial oxidative phosphorylation plays in sustaining energy status (Zheng 2012). Interestingly, our findings highlighted the important part that valine plays in improving mitochondrial activity at electron transport complex levels, including complex I, II, and IV. Recent studies have focused on inhibiting Complex I to target the Warburg effect and metabolic plasticity of cancer cells (Chaube et al.2015). The role of valine in regulating complex I is well demonstrated in T-cell acute lymphoblastic leukemia (Thandapani et al.2022). It is possible that valine could act as an anaplerotic metabolite and drive the tricarboxylic acid cycle to sustain mitochondrial ATP production.
Muscle tissues are most vulnerable to a high risk of oxidative stress (Li et al.2020). Oxidative stress can impair mitochondrial oxidation and worsen metabolic disorders (Lian et al.2022). Extreme levels of ROS production can impair skeletal muscle function by harming the cells through oxidation (Zhang et al.2001; Zhao et al.2021). Our results indicate that valine significantly reduced mitochondrial ROS production and protein expression of 4-HNE in C2C12 cells, and thus can help in promoting the growth of muscle and the healing of damaged tissue. Interestingly, valine also protects the cell by sustaining oxidative phosphorylation and generating more ATP for energy supplementation to counter the H2O2-induced oxidative stress (Jeong et al.2022). In accordance with our findings, cysteine was reported to reduce the H2O2-induced mitochondrial stress and restore mitochondrial function in C2C12 myoblasts (Mizugaki et al.2022). Valine could potentially be used therapeutically to support good mitochondrial activity and assist in reducing cellular and mitochondrial damage induced by ROS-driven oxidative stress.
Limitations
This study primarily focused on determining the effect of valine treatment on one cell line. Further research is required to completely delineate the mechanism (s) by which valine might help in cell survival during stress in different cell lines and in vivo.
Conclusion
In conclusion, we found that valine treatment does improve mitochondrial function, by enhancing mitochondrial oxidative phosphorylation and total ATP production. This could be due to its role in the improvement of the gene expression of PGC-1α, PGC-1β, and those involved in biogenesis and function. By lowering the H2O2-driven oxidative stress, valine apparently preserves cell viability, maximizes respiratory capacity, and lowers the formation of mitochondrial ROS. The findings of this study provide insight into valine's role in providing mitochondrial protection against oxidative stress, and suggest that it may be a future target for maintaining cellular function under stress.
Supplementary Material
Acknowledgments
We thank Amanda Pangle and Yingni Chi for their assistance. We are also thankful to the UAMS digital flow cytometry core facility for the expertise and equipment access.
Contributor Information
Shakshi Sharma, Donald W. Reynolds Department of Geriatrics, Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
Xiaomin Zhang, Donald W. Reynolds Department of Geriatrics, Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
Gohar Azhar, Donald W. Reynolds Department of Geriatrics, Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
Pankaj Patyal, Donald W. Reynolds Department of Geriatrics, Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
Ambika Verma, Donald W. Reynolds Department of Geriatrics, Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
Grishma KC, Donald W. Reynolds Department of Geriatrics, Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
Jeanne Y Wei, Donald W. Reynolds Department of Geriatrics, Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
Data Availability
The dataset generated during and/ or analyzed during the current study are available from the corresponding author on reasonable request.
Author contribution
Conceived and designed the experiments: S.S., G.A., X.Z., and J.Y.W. Performed the experiments: S.S., P.P., A.V. and G.K. Analyzed the data: S.S., X.Z. and G.A. Wrote the paper: S.S., X.Z., G.A. and J.Y.W. Statistical analysis: S.S., X.Z. and G.A. All authors have read and agreed to the published version of the manuscript.
Funding
This research was supported in part by the Claude D. Pepper Older American Independence Center Grant (P30AG28718) from National Institute on Aging (NIA).
Disclosure statement
The authors declare no conflict of interest.
References
- Bishop CA, Schulze MB, Klaus Set al. The branched-chain amino acids valine and leucine have differential effects on hepatic lipid metabolism. FASEB J 2020;34:9727-39. [DOI] [PubMed] [Google Scholar]
- Chaube B, Malvi P, Singh SVet al. Targeting metabolic flexibility by simultaneously inhibiting respiratory complex I and lactate generation retards melanoma progression. Oncotarget 2015;6:37281-99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chu YD, Lim SN, Yeh CTet al. COX5B-mediated bioenergetic alterations modulate cell growth and anti-cancer drug susceptibility by orchestrating claudin-2 expression in colorectal cancers. Biomedicines 2021;10:60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cruzat VF, Krause M, Newsholme P.. Amino acid supplementation and impact on immune function in the context of exercise. J Int Soc Sports Nutr 2014;11:61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- D'Antona G, Ragni M, Cardile Aet al. Branched-chain amino acid supplementation promotes survival and supports cardiac and skeletal muscle mitochondrial biogenesis in middle-aged mice. Cell Metab 2010;12:362-72. [DOI] [PubMed] [Google Scholar]
- Doi M, Yamaoka I, Fukunaga Tet al. Isoleucine, a potent plasma glucose-lowering amino acid, stimulates glucose uptake in C2C12 myotubes. Biochem Biophys Res Commun 2003;312:1111-7. [DOI] [PubMed] [Google Scholar]
- du Plessis S, Agarwal A, Mohanty Get al. Oxidative phosphorylation versus glycolysis: what fuel do spermatozoa use? Asian J Androl 2015;17:230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gart E, van Duyvenvoorde W, Caspers MPMet al. Intervention with isoleucine or valine corrects hyperinsulinemia and reduces intrahepatic diacylglycerols, liver steatosis, and inflammation in ldlr-/-.Leiden mice with manifest obesity-associated NASH. FASEB J 2022;36:e22435. [DOI] [PubMed] [Google Scholar]
- Green CL, Lamming DW.. Regulation of metabolic health by essential dietary amino acids. Mech Ageing Dev 2019;177:186-200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hewton KG, Johal AS, Parker SJ.. Transporters at the interface between cytosolic and mitochondrial amino acid metabolism. Metabolites 2021;11:112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hinkle JS, Rivera CN, Vaughan RA.. Branched-chain amino acids and mitochondrial biogenesis: an overview and mechanistic summary. Mol Nut Food Res 2022;66:2200109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holeček M. Branched-chain amino acids in health and disease: metabolism, alterations in blood plasma, and as supplements. Nutr Metab (Lond) 2018;15:33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jeong MJ, Lim DS, Kim SOet al. Protection of oxidative stress-induced DNA damage and apoptosis by rosmarinic acid in murine myoblast C2C12 cells. Biotechnol Bioproc E 2022;27:171-82. [Google Scholar]
- Johnson MA, Gannon NP, Schnuck JKet al. Leucine, palmitate, or leucine/palmitate cotreatment enhances myotube lipid content and oxidative preference. Lipids 2018;53:1043-57. [DOI] [PubMed] [Google Scholar]
- Kang C, Li Ji L.. Role of PGC-1α signaling in skeletal muscle health and disease. Ann NY Acad Sci 2012;1271:110-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kitajima Y, Takahashi H, Akiyama Tet al. Supplementation with branched-chain amino acids ameliorates hypoalbuminemia, prevents sarcopenia, and reduces fat accumulation in the skeletal muscles of patients with liver cirrhosis. J Gastroenterol 2018;53:427-37. [DOI] [PubMed] [Google Scholar]
- Kohlmeier M. Valine. In: Nutrient Metabolism. Cambridge, Massachusetts, USA: Elsevier, 2003: 370-7. [Google Scholar]
- Lee MH, Han MH, Lee DSet al. Morin exerts cytoprotective effects against oxidative stress in C2C12 myoblasts via the upregulation of Nrf2-dependent HO-1 expression and the activation of the ERK pathway. Int J Mol Med 2017;39:399-406. [DOI] [PubMed] [Google Scholar]
- Li J, Yang Q, Han Let al. C2C12 mouse myoblasts damage induced by oxidative stress is alleviated by the antioxidant capacity of the active substance phloretin. Front Cell Dev Biol 2020;8:541260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lian D, Chen MM, Wu Het al. The role of oxidative stress in skeletal muscle myogenesis and muscle dis-ease. Antioxidants 2022;11:755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liang C, Curry BJ, Brown PLet al. Leucine modulates mitochondrial biogenesis and SIRT1-AMPK signaling in C2C12 myotubes. J Nutr Metab2014;2014:1-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lynch CJ, Adams SH.. Branched-chain amino acids in metabolic signaling and insulin resistance. Nat Rev Endocrinol 2014;10:723-36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mann G, Mora S, Madu Get al. Branched-chain amino acids: catabolism in skeletal muscle and implications for muscle and whole-body metabolism. Front Physiol 2021;12:702826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mao H, Zhang Y, Ji Wet al. Leucine protects bovine intestinal epithelial cells from hydrogen peroxide-induced apoptosis by alleviating oxidative damage. J Sci Food Agric 2022;102:5903-12. [DOI] [PubMed] [Google Scholar]
- Mattick JSA, Kamisoglu K, Ierapetritou MGet al. Branched-chain amino acid supplementation: impact on signaling and relevance to critical illness. WIREs Mech Dis 2013;5:449-60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mizugaki A, Kato H, Takeda Tet al. Cystine reduces mitochondrial dysfunction in C2C12 myotubes under moderate oxidative stress induced by H2O2. Amino Acids 2022;54:1203-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nie C, He T, Zhang Wet al. Branched chain amino acids: beyond nutrition metabolism. Int J Mol Sci 2018;19:954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pan R, Chen Y.. Management of oxidative stress: crosstalk between brown/beige adipose tissues and skeletal muscles. Front Physiol 2021;12:712372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Patyal P, Nguyen B, Zhang Xet al. Rho/SRF inhibitor modulates mitochondrial functions. Int J Mol Sci 2022;23:11536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raiteri T, Zaggia I, Reano Set al. The atrophic effect of 1,25(OH)2 Vitamin D3 (Calcitriol) on C2C12 myotubes depends on oxidative stress. Antioxidants 2021;10:1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ren Y, Li Y, Yan Jet al. Adiponectin modulates oxidative stress-induced mitophagy and protects C2C12 myoblasts against apoptosis. Sci Rep 2017;7:3209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rivera ME, Lyon ES, Johnson MAet al. Leucine increases mitochondrial metabolism and lipid content without altering insulin signaling in myotubes. Biochimie 2020;168:124-33. [DOI] [PubMed] [Google Scholar]
- Rivera ME, Lyon ES, Johnson MAet al. Effect of valine on myotube insulin sensitivity and metabolism with and without insulin resistance. Mol Cell Biochem 2020;468:169-83. [DOI] [PubMed] [Google Scholar]
- Rocha M, Rovira-Llopis S, Banuls Cet al. Mitochondrial dysfunction and oxidative stress in insulin resistance. CPD 2013;19:5730-41. [DOI] [PubMed] [Google Scholar]
- Rogers SC, Zhang X, Azhar Get al. Exposure to high or low glucose levels accelerates the appearance of markers of endothelial cell senescence and induces dysregulation of nitric oxide synthase. J Gerontol Ser A 2013;68:1469-81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ruocco C, Segala A, Valerio Aet al. Essential amino acid formulations to prevent mitochondrial dysfunction and oxidative stress. Curr Opin Clin Nutr Metab Care 2021;24:88-95. [DOI] [PubMed] [Google Scholar]
- Savage DB, Petersen KF, Shulman GI.. Disordered lipid metabolism and the pathogenesis of insulin resistance. Physiol Rev 2007;87:507-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schnuck JK, Sunderland KL, Gannon NPet al. Leucine stimulates PPARβ/δ-dependent Mi-tochondrial biogenesis and oxidative metabolism with enhanced GLUT4 content and glucose uptake in myotubes. Biochimie 2016;128-129: 1–7. [DOI] [PubMed] [Google Scholar]
- Tamanna N, Mahmood N.. Emerging roles of branched-chain amino acid supplementation in human diseases. Int Sch Res Notices 2014;2014:1-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thandapani P, Kloetgen A, Witkowski MTet al. Valine TRNA levels and availability regulate complex I assembly in leukaemia. Nature 2022;601:428-33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tom A, Nair KS.. Assessment of branched-chain amino acid status and potential for biomarkers. J Nutr 2006;136:324S-30S. [DOI] [PubMed] [Google Scholar]
- Valerio A, D'Antona G, Nisoli E.. Branched-chain amino acids, mitochondrial biogenesis, and healthspan: an evolutionary perspective. Aging 2011;3:464-78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vanweert F, Schrauwen P, Phielix E.. Role of branched-chain amino acid metabolism in the pathogenesis of obesity and Type 2 diabetes-related metabolic disturbances BCAA metabolism in Type 2 diabetes. Nutr Diabetes 2022;12:35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Verma A, Azhar G, Zhang Xet al. P. gingivalis-LPS induces mitochondrial dysfunction mediated by neuroinflammation through oxidative stress. Int J Mol Sci 2023;24:950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Viana LR, Tobar N, Busanello ENBet al. Leucine-rich diet induces a shift in tumour metabolism from glycolytic towards oxidative phosphorylation, reducing glucose consumption and metastasis in Walker-256 tumour-bearing rats. Sci Rep 2019;9:15529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Walters JW, Amos D, Ray Ket al. Mitochondrial redox status as a target for cardiovascular disease. Curr Opin Pharmacol 2016;27:50-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wolfe RR. Branched-chain amino acids and muscle protein synthesis in humans: myth or reality? J Int Soc Sports Nutr 2017;14:30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu S, Liu X, Cheng Let al. Protective mechanism of leucine and isoleucine against H2O2-induced oxidative damage in bovine mammary epithelial cells. Oxid Med Cell Long 2022;2022:1-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ye Z, Wang S, Zhang Cet al. Coordinated modulation of energy metabolism and inflammation by branched-chain amino acids and fatty acids. Front Endocrinol 2020;11:617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yoneshiro T, Kataoka N, Walejko JMet al. Metabolic flexibility via mitochondrial BCAA carrier SLC25A44 is required for optimal fever. Cell Bio 2021;10:e66865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yoon MS. The emerging role of branched-chain amino acids in insulin resistance and metabolism. Nutrients 2016;8:405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang X, Azhar G, Nagano Ket al. Differential vulnerability to oxidative stress in rat cardiac myocytes versus fibroblasts. J Am Coll Cardiol 2001;38:2055-62. [DOI] [PubMed] [Google Scholar]
- Zhang X, Azhar G, Wei JY.. The expression of MicroRNA and MicroRNA clusters in the aging heart. PLoS One 2012;7:e34688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao MJ, Yuan S, Zi Het al. Oxidative stress links aging-associated cardiovascular diseases and prostatic diseases. Oxid Med Cell Long 2021;2021:1-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao R, Jiang S, Zhang Let al. Mitochondrial electron transport chain, ROS generation and uncoupling (review). Int J Mol Med 2019;44:3-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zheng J. Energy metabolism of cancer: glycolysis versus oxidative phosphorylation (review). Onco Letters 2012;4:1151-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The dataset generated during and/ or analyzed during the current study are available from the corresponding author on reasonable request.