Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Mol Cell Endocrinol ; 559: 111800, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36270542

ABSTRACT

PURPOSE: Type 2 diabetes is characterized by reduced insulin sensitivity which correlates with increased circulating BCAA. These experiments investigated the effects of insulin resistance with and without excess BCAA on myotube insulin sensitivity and L-type amino acid transporter-1 (LAT1). METHODS: C2C12 myotubes were treated with or without excess BCAA for 1 or 6 days, both with and without insulin resistance. Western blot was used to assess insulin sensitivity and LAT1 content. Liquid chromatography-mass spectrometry was used to evaluate BCAA media content. RESULTS: Insulin resistance was associated with significantly increased extracellular BCAA accumulation independent of LAT1 content. Conversely, prior BCAA treatment was not associated with extracellular BCAA accumulation regardless of level of insulin sensitivity. CONCLUSION: These data suggest insulin resistance, but not BCAA treatment, promotes extracellular BCAA accumulation independent of changes in LAT1 content, implicating insulin resistance as a causal agent of extracellular BCAA accumulation.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Humans , Insulin Resistance/physiology , Amino Acids, Branched-Chain/metabolism , Diabetes Mellitus, Type 2/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism
2.
Amino Acids ; 54(3): 455-468, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35112170

ABSTRACT

Glutamine is an amino acid previously linked with improved skeletal muscle metabolism and insulin signaling, however, past observations often use cell culture models with only supraphysiological concentrations. Additionally, past reports have yet to simultaneously investigate both metabolic outcomes and insulin signaling. The present report utilized cell culture experiments and measured the effects of both physiological and supraphysiological levels of glutamine on myotube metabolism and insulin signaling/resistance. It was hypothesized the addition of glutamine at any level would increase cell metabolism and related gene expression, as well as improve insulin signaling versus respective control cells. C2C12 myotubes were treated with glutamine ranging from 0.25 mM-4 mM (or media control) for 24 h to capture a range of physiological and supraphysiological concentrations. qRT-PCR was used to measure metabolic gene expression. Mitochondrial and glycolytic metabolism were measured via oxygen consumption and extracellular acidification rate, respectively. Insulin sensitivity (indicated by pAkt:Akt) and metabolism following glucose/insulin infusion were also assessed. Glutamine treatment consistently increased mitochondrial and glycolytic metabolism versus true controls (cells treated with media void of glutamine), however, supraphysiological glutamine did not enhance metabolism beyond that of cells with physiological levels of glutamine. Neither physiological nor supraphysiological levels of glutamine altered insulin signaling regardless of insulin stimulation or insulin resistance when compared with respective controls. These data demonstrate excess glutamine does not appear to alter myotube metabolism or glucose disposal when base levels of glutamine are present. Moreover, glutamine does not appear to alter insulin sensitivity regardless of level of insulin resistance or presence of insulin stimulation.


Subject(s)
Insulin Resistance , Glutamine/metabolism , Humans , Insulin/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism
3.
Diabetes Metab Res Rev ; 38(2): e3490, 2022 02.
Article in English | MEDLINE | ID: mdl-34397159

ABSTRACT

AIMS: Branched-chain amino acids (BCAA) are often emphasized in the diets of avid exercisers, yet population data demonstrates a correlation between circulating BCAA and insulin resistance. However, it is unclear if BCAA independently promote insulin resistance in otherwise healthy cells. The purpose of this study is to examine the effect of a BCAA mixture on muscle insulin signaling in vitro in both insulin resistant and sensitive cells. MATERIALS AND METHODS: C2C12 myotubes were treated with a BCAA mixture containing leucine:isoleucine:valine at a ratio of 2:1:1 at 0.2, 2, or 20 mM (based on leucine content) for either 30 min, 1 day, or 6 days. Western blot was used to assess insulin sensitivity of cells treated with BCAA both with and without concurrent insulin resistance, and, with and without insulin stimulation. RESULTS: BCAA treatment for 1 day significantly reduced basal, but not insulin-stimulated pAkt expression. BCAA treatment for 6 days resulted in significantly reduced basal insulin signaling in healthy cells and insulin-stimulated insulin signaling in insulin resistant (but not insulin sensitive) cells. CONCLUSION: Similar to previous observations demonstrating BCAA may correlate with insulin resistance during metabolically stressed conditions, we demonstrate excessively high BCAA exposure can negatively influence basal insulin signaling, as well as insulin sensitivity in insulin resistant myotubes. However, given the intentionally high concentrations of BCAA used in this study, the extent to which these observations translate to in vivo models is unclear and warrants further investigation.


Subject(s)
Insulin Resistance , Amino Acids, Branched-Chain/pharmacology , Humans , Insulin/metabolism , Insulin Resistance/physiology , Muscle Fibers, Skeletal/metabolism , Signal Transduction
4.
Endocrine ; 76(1): 18-28, 2022 04.
Article in English | MEDLINE | ID: mdl-34811646

ABSTRACT

PURPOSE: Branched-chain amino acids (BCAA) have been shown to enhance several cellular signaling pathways including protein synthesis and mitochondrial biogenesis, yet population data demonstrate a correlation between circulating BCAA and severity of insulin resistance which has been hypothesized to be, in part, a byproduct of BCAA inhibition of mitochondrial function. The purpose of this study is to examine the effect of a BCAA mixture on muscle metabolism and related gene expression in vitro. METHODS: C2C12 myotubes were treated with a BCAA mixture containing leucine:isoleucine:valine at a ratio of 2:1:1 at 0.2, 2, or 20 mM (based on leucine content) for 6 days. qRT-PCR was used to measure metabolic gene expression. Oxygen consumption and extracellular acidification were used to assess mitochondrial and glycolytic metabolism, respectively. Mitochondrial content was determined via mitochondrial-specific staining. RESULTS: Despite significantly elevated mitochondrial staining, 6-day BCAA treatment reduced basal mitochondrial metabolism at a supraphysiological concentration (20 mM) in both insulin sensitive and resistant cells. Peak mitochondrial capacity was also reduced in insulin-resistant (but not insulin sensitive) cells. Conversely, basal glycolytic metabolism was elevated following 20 mM BCAA treatment, regardless of insulin resistance. In addition, insulin-resistant cells treated with 20 mM BCAA exhibited reduced gene expression of Ppargc1a, Cytc, Atp5b, Glut4, and several glycolytic enzymes versus insulin sensitive cells treated with 20 mM BCAA. CONCLUSIONS: Collectively, these findings suggest BCAA at supraphysiologically high levels may negatively alter mitochondrial metabolism, and concurrent insulin resistance may also diminish peak mitochondrial capacity, as well as impede molecular adaptations that support a transition to a glycolytic preference/compensation.


Subject(s)
Insulin Resistance , Amino Acids, Branched-Chain , Humans , Insulin/metabolism , Insulin Resistance/physiology , Leucine/metabolism , Leucine/pharmacology , Muscle Fibers, Skeletal
5.
Lipids ; 56(6): 563-578, 2021 11.
Article in English | MEDLINE | ID: mdl-34382222

ABSTRACT

Previous studies have shown various metabolic stressors such as saturated fatty acids (SFA) and excess insulin promote insulin resistance in metabolically meaningful cell types (such as skeletal muscle). Additionally, these stressors have been linked with suppressed mitochondrial metabolism, which is also a common characteristic of skeletal muscle of diabetics. This study characterized the individual and combined effects of excess lipid and excess insulin on myotube metabolism and related metabolic gene and protein expression. C2C12 myotubes were treated with either 500 µM palmitate (PAM), 100 nM insulin (IR), or both (PAM-IR). qRT-PCR and western blot were used to measure metabolic gene and protein expression, respectively. Oxygen consumption was used to measure mitochondrial metabolism. Glycolytic metabolism and insulin-mediated glucose uptake were measured via extracellular acidification rate. Cellular lipid and mitochondrial content were measured using Nile Red and NAO staining, respectively. IR and PAM-IR treatments led to reductions in p-Akt expression. IR treatment reduced insulin mediated glucose metabolism while PAM and PAM-IR treatment showed increases with concurrent reductions in mitochondrial metabolism. All three treatments showed suppression in mitochondrial metabolism. PAM and PAM-IR also showed increases in glycolytic metabolism. While PAM and PAM-IR significantly increased lipid content, expression of inflammatory and lipogenic proteins were unaltered. Lastly, PAM-IR reduced BCAT2 protein expression, a regulator of BCAA metabolism. Both stressors independently reduced insulin signaling, mitochondrial function, and cell metabolism, however, only PAM-IR co-treatment significantly reduced the expression of regulators of metabolism not seen with individual stressors, suggesting an additive effect of stressors on metabolic programming.


Subject(s)
Insulin Resistance , Insulin , Humans , Muscle Fibers, Skeletal , Muscle, Skeletal , Palmitates
6.
Mol Cell Biochem ; 468(1-2): 169-183, 2020 May.
Article in English | MEDLINE | ID: mdl-32222880

ABSTRACT

Population data have consistently demonstrated a correlation between circulating branched-chain amino acids (BCAA) and insulin resistance. Most recently valine catabolite, 3-hydroxyisobutyrate, has emerged as a potential cause of BCAA-mediated insulin resistance; however, it is unclear if valine independently promotes insulin resistance. It is also unclear if excess valine influences the ability of cells to degrade BCAA. Therefore, this study investigated the effect of valine on muscle insulin signaling and related metabolism in vitro. C2C12 myotubes were treated with varying concentrations (0.5 mM-2 mM) of valine for up to 48 h. qRT-PCR and western blot were used to measure metabolic gene and protein expression, respectively. Insulin sensitivity (indicated by pAkt:Akt), metabolic gene and protein expression, and cell metabolism were also measured following valine treatment both with and without varying levels of insulin resistance. Mitochondrial and glycolytic metabolism were measured via oxygen consumption and extracellular acidification rate, respectively. Valine did not alter regulators of mitochondrial biogenesis or glycolysis; however, valine reduced branched-chain alpha-keto acid dehydrogenase a (Bckdha) mRNA (but not protein) expression which was exacerbated by insulin resistance. Valine treatment had no effect on pAkt:Akt following either acute or 48-h treatment, regardless of insulin stimulation or varying levels of insulin resistance. In conclusion, despite consistent population data demonstrating a relationship between circulating BCAA (and related metabolites) and insulin resistance, valine does not appear to independently alter insulin sensitivity or worsen insulin resistance in the myotube model of skeletal muscle.


Subject(s)
Amino Acids, Branched-Chain/drug effects , Insulin Resistance , Insulin/metabolism , Muscle Fibers, Skeletal/drug effects , Muscle, Skeletal/drug effects , Valine/pharmacology , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/genetics , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/metabolism , Amino Acids, Branched-Chain/metabolism , Animals , Cell Line , Cell Survival/drug effects , Glycolysis/drug effects , Insulin/pharmacology , Mice , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
7.
J Physiol Biochem ; 76(1): 169-178, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32056134

ABSTRACT

Uncarboxylated osteocalcin (uOC) is a circulating bone matrix protein, which has previously been shown to regulate glucose uptake and systemic metabolism. However, the cellular mechanism by which uOC acts has yet to be elucidated. C2C12 mouse myotubes were treated for 72 h with uOC (1-100 ng/mL). Cellular metabolism was analyzed using oxygen consumption and extracellular acidification rate. Metabolic gene and protein expression were measured via quantitative real-time polymerase chain reaction and Western blot, respectively. Additionally, C2C12 myotubes were treated with 10 ng/mL uOC to examine glucose uptake and activation of insulin signaling with or without insulin resistance. Finally, cellular lipid content was measured via Oil Red O and Nile Red staining. uOC treatment resulted in dose-dependent alterations of oxygen consumption with little effect on regulators of mitochondrial metabolism. Basal expression of regulators of glucose uptake were unaffected by uOC treatment. However, insulin-stimulated glucose uptake was blunted by uOC treatment with no concurrent alterations in insulin signaling. While chronic insulin treatment resulted in suppressed activation of Akt, concurrent uOC treatment was unable to prevent these detrimental effects on insulin signaling. uOC treatment had no effect on markers of lipogenesis and cellular lipid content. These findings suggest that 72-h uOC treatment may alter oxygen consumption without effect on regulators of mitochondrial biogenesis. Additionally, uOC treatment suppressed insulin-stimulated glucose uptake in cultured myotubes but had little effect on insulin signaling or regulators of cellular metabolism and was unable to mitigate insulin resistance.


Subject(s)
Glucose/metabolism , Insulin/metabolism , Mitochondria , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Osteocalcin/pharmacology , Animals , Cell Line , Insulin/pharmacology , Insulin Resistance , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Muscle Fibers, Skeletal/cytology , Organelle Biogenesis , Oxygen/metabolism , Oxygen Consumption
8.
J Cell Biochem ; 121(1): 816-827, 2020 01.
Article in English | MEDLINE | ID: mdl-31385363

ABSTRACT

Metformin has antihyperglycemic properties and is a commonly prescribed drug for type II diabetes mellitus. Metformin functions in part by activating 5'-AMP-activated protein kinase, reducing hepatic gluconeogenesis and blood glucose. Metformin also upregulates peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α). Several population studies have shown levels of circulating branched-chain amino acids (BCAA) positively correlate with insulin resistance. Because BCAA catabolic enzyme content is regulated by PGC-1α, we hypothesized metformin may alter BCAA catabolism. Therefore, the purpose of this work was to investigate the effect of metformin at varying concentrations on myotube metabolism and related gene and protein expression. C2C12 myotubes were treated with metformin at 30 uM (physiological) or 2 mM (supraphysiological) for up to 24 hours. Metabolic gene expression was measured via quantitative real time polymerase chain reaction, protein expression was measured using Western blot, and mitochondrial and glycolytic metabolism were measured via oxygen consumption and extracellular acidification rate, respectively. Supraphysiological metformin upregulated PGC-1α mRNA expression along with related downstream targets, yet the reduced expression of electron transport chain components as well as basal and peak cell metabolism. Supraphysiological metformin also suppressed branched-chain aminotransferase 2 (BCAT2) and branched-chain-alpha-keto acid dehydrogenase E1a (BCKDHa) mRNA expression as well as BCAT2 protein expression and BCKDHa activity, which was accompanied by decreased Kruppel-like factor 15 protein expression. Physiological levels of metformin suppressed BCKDHa and cytochrome c oxidase mRNA expression at early time points (4-12 hours) but had no effect on any other outcomes. Together these data suggest metformin may suppress BCAA catabolic enzyme expression or activity, possibly reducing levels of circulating gluconeogenic substrates.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Amino Acids, Branched-Chain/metabolism , Fibroblasts/metabolism , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Muscle Fibers, Skeletal/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Animals , Fibroblasts/drug effects , Glycolysis , Mice , Muscle Fibers, Skeletal/drug effects , Organelle Biogenesis
9.
Biochimie ; 168: 124-133, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31682874

ABSTRACT

Elevated circulating branched-chain amino acids (BCAA) such as leucine have been consistently correlated with increasing severity of insulin resistance across numerous populations. BCAA may promote insulin resistance through either mTOR-mediated suppression of insulin receptor substrate-1 or through the accumulation of toxic BCAA catabolites. Although the link between circulating BCAA and insulin resistance has been consistent, it has yet to be concluded if BCAA causally contribute to the development or worsening of insulin resistance. This work investigated the effect of leucine both with and without varying levels of insulin resistance on metabolism, metabolic gene expression, and insulin signaling. C2C12 myotubes were treated with and without varied concentrations of leucine up to 2 mM for 24 h both with and without varied levels of insulin resistance. Gene and protein expression were measured via qRT-PCR and Western blot, respectively. Mitochondrial metabolism was measured via O2 consumption. Leucine at 2 mM increased oxidative metabolism as well as gene expression of mitochondrial biogenesis, which was associated with increased cellular lipid content. Despite increased lipid content of leucine-treated cells, neither acute nor chronic leucine treatment at 2 mM affected insulin signaling in insulin sensitive, mildly insulin resistant, or severely insulin resistant cells. Similarly, leucine at lower concentrations (0.25 mM, 0.5 mM, and 1 mM) did not alter insulin signaling either, regardless of insulin resistance. Leucine appears to improve myotube oxidative metabolism and related metabolic gene expression. And despite increased lipid content of leucine-treated cells, leucine does not appear to alter insulin sensitivity either acutely or chronically, regardless of level of insulin resistance.


Subject(s)
Insulin/metabolism , Leucine/pharmacology , Mitochondria/metabolism , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Animals , Cell Line , Insulin Resistance , Lipid Metabolism/drug effects , Muscle Fibers, Skeletal/cytology , Organelle Biogenesis
10.
Nutr Res ; 66: 22-31, 2019 06.
Article in English | MEDLINE | ID: mdl-31051319

ABSTRACT

Branched-chain amino acids (BCAAs) are essential in the diet and may provide benefit for those who partake in regular physical activity and resistance training, yet circulating BCAAs have been repeatedly shown to correlate with severity of insulin resistance in obese/diseased populations. Recently, the valine catabolite 3-hydroxyisobuterate (3HIB) was shown to promote insulin resistance in skeletal muscle by increasing lipid content in vivo. The purpose of this study was to investigate the mechanistic effects of 3HIB on skeletal muscle insulin signaling, metabolism, and related gene expression in vitro. Given these previous observations, we hypothesized that 3HIB would depress skeletal muscle metabolism and insulin sensitivity. C2C12 myotubes were treated with 3HIB for up to 48 hours using both physiological (25-100 µmol/L) and supraphysiological (5 mmol/L) concentrations. Metabolic gene expression was measured via quantitative real-time polymerase chain reaction, mitochondrial metabolism was measured via O2 consumption, and glycolytic metabolism was quantified using extracellular acidification rate. Western blot was used to assess insulin sensitivity following insulin stimulation (indicated by phospho-AKT expression). 3HIB did not alter expressional indicators of mitochondrial biogenesis, glycolysis, BCAA catabolism, or lipogenesis. Chronic physiological 3HIB treatment significantly increased peak oxygen consumption, whereas supraphysiological 3HIB treatment suppressed basal and peak mitochondrial and glycolytic metabolism. Both physiological and supraphysiological 3HIB reduced pAkt expression during insulin stimulation. These findings suggest that 3HIB may reduce muscle insulin sensitivity in cultured myotubes, supporting a potentially causal role of 3HIB in the development of insulin resistance in highly metabolic cell types.


Subject(s)
Hydroxybutyrates/administration & dosage , Insulin/metabolism , Mitochondria, Muscle/drug effects , Mitochondria, Muscle/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Amino Acids, Branched-Chain/metabolism , Animals , Cell Line , Dose-Response Relationship, Drug , Gene Expression/drug effects , Glycolysis/drug effects , Insulin Resistance , Lipid Metabolism/drug effects , Mice , Muscle Fibers, Skeletal/ultrastructure , Muscle, Skeletal/drug effects , Muscle, Skeletal/ultrastructure , Myoblasts , Oxygen Consumption/drug effects , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...