Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters

Publication year range
1.
Am J Physiol Endocrinol Metab ; 327(2): E183-E193, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38895980

ABSTRACT

Elevated skeletal muscle diacylglycerols (DAGs) and ceramides can impair insulin signaling, and acylcarnitines (acylCNs) reflect impaired mitochondrial fatty acid oxidation, thus, the intramuscular lipid profile is indicative of insulin resistance. Acute (i.e., postprandial) hyperinsulinemia has been shown to elevate lipid concentrations in healthy muscle and is an independent risk factor for type 2 diabetes (T2D). However, it is unclear how the relationship between acute hyperinsulinemia and the muscle lipidome interacts across metabolic phenotypes, thus contributing to or exacerbating insulin resistance. We therefore investigated the impact of acute hyperinsulinemia on the skeletal muscle lipid profile to help characterize the physiological basis in which hyperinsulinemia elevates T2D risk. In a cross-sectional comparison, endurance athletes (n = 12), sedentary lean adults (n = 12), and individuals with obesity (n = 13) and T2D (n = 7) underwent a hyperinsulinemic-euglycemic clamp with muscle biopsies. Although there were no significant differences in total 1,2-DAG fluctuations, there was a 2% decrease in athletes versus a 53% increase in T2D during acute hyperinsulinemia (P = 0.087). Moreover, C18 1,2-DAG species increased during the clamp with T2D only, which negatively correlated with insulin sensitivity (P < 0.050). Basal muscle C18:0 total ceramides were elevated with T2D (P = 0.029), but not altered by clamp. Acylcarnitines were universally lowered during hyperinsulinemia, with more robust reductions of 80% in athletes compared with only 46% with T2D (albeit not statistically significant, main effect of group, P = 0.624). Similar fluctuations with acute hyperinsulinemia increasing 1,2 DAGs in insulin-resistant phenotypes and universally lowering acylcarnitines were observed in male mice. In conclusion, acute hyperinsulinemia elevates muscle 1,2-DAG levels with insulin-resistant phenotypes. This suggests a possible dysregulation of intramuscular lipid metabolism in the fed state in individuals with low insulin sensitivity, which may exacerbate insulin resistance.NEW & NOTEWORTHY Postprandial hyperinsulinemia is a risk factor for type 2 diabetes and may increase muscle lipids. However, it is unclear how the relationship between acute hyperinsulinemia and the muscle lipidome interacts across metabolic phenotypes, thus contributing to insulin resistance. We observed that acute hyperinsulinemia elevates muscle 1,2-DAGs in insulin-resistant phenotypes, whereas ceramides were unaltered. Insulin-mediated acylcarnitine reductions are also hindered with high-fat feeding. The postprandial period may exacerbate insulin resistance in metabolically unhealthy phenotypes.


Subject(s)
Diabetes Mellitus, Type 2 , Diglycerides , Hyperinsulinism , Insulin Resistance , Muscle, Skeletal , Phenotype , Hyperinsulinism/metabolism , Humans , Diglycerides/metabolism , Male , Muscle, Skeletal/metabolism , Adult , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/complications , Female , Cross-Sectional Studies , Middle Aged , Glucose Clamp Technique , Obesity/metabolism , Obesity/complications , Athletes , Young Adult , Acute Disease , Animals , Ceramides/metabolism , Mice , Carnitine/analogs & derivatives
2.
Diabetologia ; 64(1): 168-180, 2021 01.
Article in English | MEDLINE | ID: mdl-33128577

ABSTRACT

AIMS/HYPOTHESIS: Subcellular localisation is an important factor in the known impact of bioactive lipids, such as diacylglycerol and sphingolipids, on insulin sensitivity in skeletal muscle; yet, the role of localised intramuscular triacylglycerol (IMTG) is yet to be described. Excess accumulation of IMTG in skeletal muscle is associated with insulin resistance, and we hypothesised that differences in subcellular localisation and composition of IMTG would relate to metabolic health status in humans. METHODS: We evaluated subcellular localisation of IMTG in lean participants, endurance-trained athletes, individuals with obesity and individuals with type 2 diabetes using LC-MS/MS of fractionated muscle biopsies and insulin clamps. RESULTS: Insulin sensitivity was significantly different between each group (athletes>lean>obese>type 2 diabetes; p < 0.001). Sarcolemmal IMTG was significantly greater in individuals with obesity and type 2 diabetes compared with lean control participants and athletes, but individuals with type 2 diabetes were the only group with significantly increased saturated IMTG. Sarcolemmal IMTG was inversely related to insulin sensitivity. Nuclear IMTG was significantly greater in individuals with type 2 diabetes compared with lean control participants and athletes, and total and saturated IMTG localised in the nucleus had a significant inverse relationship with insulin sensitivity. Total cytosolic IMTG was not different between groups, but saturated cytosolic IMTG species were significantly increased in individuals with type 2 diabetes compared with all other groups. There were no significant differences between groups for IMTG concentration in the mitochondria/endoplasmic reticulum. CONCLUSIONS/INTERPRETATION: These data reveal previously unknown differences in subcellular IMTG localisation based on metabolic health status and indicate the influence of sarcolemmal and nuclear IMTG on insulin sensitivity. Additionally, these studies suggest saturated IMTG may be uniquely deleterious for muscle insulin sensitivity. Graphical abstract.


Subject(s)
Insulin Resistance/physiology , Muscle, Skeletal/chemistry , Muscle, Skeletal/ultrastructure , Triglycerides/analysis , Triglycerides/chemistry , Adult , Athletes , Cell Nucleus/chemistry , Cytosol/chemistry , Diabetes Mellitus, Type 2/metabolism , Dietary Fats/administration & dosage , Diglycerides/analysis , Endoplasmic Reticulum/chemistry , Female , Humans , Male , Middle Aged , Mitochondria, Muscle/chemistry , Obesity/metabolism , Physical Endurance , Sarcolemma/chemistry
3.
FASEB J ; 34(3): 4602-4618, 2020 03.
Article in English | MEDLINE | ID: mdl-32030805

ABSTRACT

Understanding the mechanisms regulating mitochondrial respiratory function and adaptations to metabolic challenges, such as exercise and high dietary fat, is necessary to promote skeletal muscle health and attenuate metabolic disease. Autophagy is a constitutively active degradation pathway that promotes mitochondrial turnover and transiently increases postexercise. Recent evidence indicates Bcl2 mediates exercise-induced autophagy and skeletal muscle adaptions to training during high-fat diet. We determined if improvements in mitochondrial respiration due to exercise training required Bcl2-mediated autophagy using a transgenic mouse model of impaired inducible autophagy (Bcl2AAA ). Mitochondrial adaptations to a treadmill exercise training protocol, in either low-fat or high-fat diet fed mice, did not require Bcl2-mediated autophagy activation. Instead, training increased protein synthesis rates and basal autophagy in the Bcl2AAA mice, while acute exercise activated BNIP3 and Parkin autophagy. High-fat diet stimulated lipid-specific mitochondrial adaptations. These data demonstrate increases in basal mitochondrial turnover, not transient activation with exercise, mediate adaptations to exercise and high-fat diet.


Subject(s)
Autophagy/physiology , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Physical Conditioning, Animal/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Immunoblotting , Lipid Metabolism/physiology , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/genetics , Ubiquitin-Protein Ligases/genetics
4.
Am J Physiol Cell Physiol ; 317(2): C339-C347, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31091142

ABSTRACT

Rat L6 and mouse C2C12 cell lines are commonly used to investigate myocellular metabolism. Mitochondrial characteristics of these cell lines remain poorly understood despite mitochondria being implicated in the development of various metabolic diseases. To address this need, we performed high-resolution respirometry to determine rates of oxygen consumption and H2O2 emission in suspended myoblasts during multiple substrate-uncoupler-inhibitor titration protocols. The capacity for oxidative phosphorylation supported by glutamate and malate, with and without succinate, or supported by palmitoyl-l-carnitine was lower in L6 compared with C2C12 myoblasts (all P < 0.01 for L6 vs. C2C12). Conversely, H2O2 emission during oxidative phosphorylation was greater in L6 than C2C12 myoblasts (P < 0.01 for L6 vs. C2C12). Induction of noncoupled respiration revealed a significantly greater electron transfer capacity in C2C12 compared with L6 myoblasts, regardless of the substrate(s) provided. Mitochondrial metabolism was also investigated in differentiated L6 and C2C12 myotubes. Basal rates of oxygen consumption were not different between intact, adherent L6, and C2C12 myotubes; however, noncoupled respiration was significantly lower in L6 compared with C2C12 myotubes (P = 0.01). In summary, L6 myoblasts had lower respiration rates than C2C12 myoblasts, including lesser capacity for fatty acid oxidation and greater electron leak toward H2O2. L6 cells also retain a lower capacity for electron transfer compared with C2C12 following differentiation to form fused myotubes. Intrinsic differences in mitochondrial metabolism between these cell lines should be considered when modeling and investigating myocellular metabolism.


Subject(s)
Hydrogen Peroxide/metabolism , Mitochondria, Muscle/metabolism , Myoblasts, Skeletal/metabolism , Oxidative Phosphorylation , Animals , Cell Line , Cell Respiration , Electron Transport Chain Complex Proteins/metabolism , Fatty Acids/metabolism , Mice , Oxidation-Reduction , Oxygen Consumption , Rats
5.
Am J Physiol Endocrinol Metab ; 316(5): E866-E879, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30620635

ABSTRACT

Intermuscular adipose tissue (IMAT) is negatively related to insulin sensitivity, but a causal role of IMAT in the development of insulin resistance is unknown. IMAT was sampled in humans to test for the ability to induce insulin resistance in vitro and characterize gene expression to uncover how IMAT may promote skeletal muscle insulin resistance. Human primary muscle cells were incubated with conditioned media from IMAT, visceral (VAT), or subcutaneous adipose tissue (SAT) to evaluate changes in insulin sensitivity. RNAseq analysis was performed on IMAT with gene expression compared with skeletal muscle and SAT, and relationships to insulin sensitivity were determined in men and women spanning a wide range of insulin sensitivity measured by hyperinsulinemic-euglycemic clamp. Conditioned media from IMAT and VAT decreased insulin sensitivity similarly compared with SAT. Multidimensional scaling analysis revealed distinct gene expression patterns in IMAT compared with SAT and muscle. Pathway analysis revealed that IMAT expression of genes in insulin signaling, oxidative phosphorylation, and peroxisomal metabolism related positively to donor insulin sensitivity, whereas expression of macrophage markers, inflammatory cytokines, and secreted extracellular matrix proteins were negatively related to insulin sensitivity. Perilipin 5 gene expression suggested greater IMAT lipolysis in insulin-resistant individuals. Combined, these data show that factors secreted from IMAT modulate muscle insulin sensitivity, possibly via secretion of inflammatory cytokines and extracellular matrix proteins, and by increasing local FFA concentration in humans. These data suggest IMAT may be an important regulator of skeletal muscle insulin sensitivity and could be a novel therapeutic target for skeletal muscle insulin resistance.


Subject(s)
Adipose Tissue/metabolism , Insulin Resistance/genetics , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Adult , Athletes , Diabetes Mellitus, Type 2/metabolism , Fatty Acids, Nonesterified/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation , Glucose Clamp Technique , Humans , Intra-Abdominal Fat/metabolism , Male , Middle Aged , Obesity/metabolism , Primary Cell Culture , Sedentary Behavior , Sequence Analysis, RNA , Subcutaneous Fat/metabolism
6.
J Nutr ; 149(12): 2120-2132, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31495890

ABSTRACT

BACKGROUND: Dietary nitrate improves exercise performance by reducing the oxygen cost of exercise, although the mechanisms responsible are not fully understood. OBJECTIVES: We tested the hypothesis that nitrate and nitrite treatment would lower the oxygen cost of exercise by improving mitochondrial function and stimulating changes in the availability of metabolic fuels for energy production. METHODS: We treated 9-mo-old zebrafish with nitrate (sodium nitrate, 606.9 mg/L), nitrite (sodium nitrite, 19.5 mg/L), or control (no treatment) water for 21 d. We measured oxygen consumption during a 2-h, strenuous exercise test; assessed the respiration of skeletal muscle mitochondria; and performed untargeted metabolomics on treated fish, with and without exercise. RESULTS: Nitrate and nitrite treatment increased blood nitrate and nitrite levels. Nitrate treatment significantly lowered the oxygen cost of exercise, as compared with pretreatment values. In contrast, nitrite treatment significantly increased oxygen consumption with exercise. Nitrate and nitrite treatments did not change mitochondrial function measured ex vivo, but significantly increased the abundances of ATP, ADP, lactate, glycolytic intermediates (e.g., fructose 1,6-bisphosphate), tricarboxylic acid (TCA) cycle intermediates (e.g., succinate), and ketone bodies (e.g., ß-hydroxybutyrate) by 1.8- to 3.8-fold, relative to controls. Exercise significantly depleted glycolytic and TCA intermediates in nitrate- and nitrite-treated fish, as compared with their rested counterparts, while exercise did not change, or increased, these metabolites in control fish. There was a significant net depletion of fatty acids, acyl carnitines, and ketone bodies in exercised, nitrite-treated fish (2- to 4-fold), while exercise increased net fatty acids and acyl carnitines in nitrate-treated fish (1.5- to 12-fold), relative to their treated and rested counterparts. CONCLUSIONS: Nitrate and nitrite treatment increased the availability of metabolic fuels (ATP, glycolytic and TCA intermediates, lactate, and ketone bodies) in rested zebrafish. Nitrate treatment may improve exercise performance, in part, by stimulating the preferential use of fuels that require less oxygen for energy production.


Subject(s)
Fatty Acids/metabolism , Glycolysis , Nitrates/therapeutic use , Nitrites/therapeutic use , Oxygen/metabolism , Physical Conditioning, Animal , Zebrafish/metabolism , Animals , Mitochondria/metabolism , Zebrafish/physiology
7.
Am J Physiol Endocrinol Metab ; 315(4): E425-E434, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29812987

ABSTRACT

Excess fat intake can increase lipid oxidation and expression of mitochondrial proteins, indicating remodeling of the mitochondrial proteome. Yet intermediates of lipid oxidation also accumulate, indicating a relative insufficiency to completely oxidize lipids. We investigated remodeling of the mitochondrial proteome to determine mechanisms of changes in lipid oxidation following high-fat feeding. C57BL/6J mice consumed a high-fat diet (HFD, 60% fat from lard) or a low-fat diet (LFD, 10% fat) for 12 wk. Mice were fasted for 4 h and then anesthetized by pentobarbital sodium overdose for tissue collection. A mitochondrial-enriched fraction was prepared from gastrocnemius muscles and underwent proteomic analysis by high-resolution mass spectrometry. Mitochondrial respiratory efficiency was measured as the ratio of ATP production to O2 consumption. Intramuscular acylcarnitines were measured by liquid chromatography-mass spectrometry. A total of 658 mitochondrial proteins were identified: 40 had higher abundance and 14 had lower abundance in mice consuming the HFD than in mice consuming the LFD. Individual proteins that changed with the HFD were primarily related to ß-oxidation; there were fewer changes to the electron transfer system. Gene set enrichment analysis indicated that the HFD increased pathways of lipid metabolism and ß-oxidation. Intramuscular concentrations of select acylcarnitines (C18:0) were greater in the HFD mice and reflected dietary lipid composition. Mitochondrial respiratory ATP production-to-O2 consumption ratio for lipids was not different between LFD and HFD mice. After the 60% fat diet, remodeling of the mitochondrial proteome revealed upregulation of proteins regulating lipid oxidation that was not evident for all mitochondrial pathways. The accumulation of lipid metabolites with obesity may occur without intrinsic dysfunction to mitochondrial lipid oxidation.


Subject(s)
Diet, High-Fat , Electron-Transferring Flavoproteins/metabolism , Lipid Metabolism , Mitochondria, Muscle/metabolism , Mitochondrial Proteins/metabolism , Muscle, Skeletal/metabolism , Proteome/metabolism , Adenosine Triphosphate/metabolism , Animals , Carnitine/analogs & derivatives , Carnitine/metabolism , Cell Respiration , Chromatography, Liquid , Diet, Fat-Restricted , Electron Transport , Insulin Resistance , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Oxidation-Reduction , Oxygen Consumption , Protein Processing, Post-Translational , Proteomics , Tandem Mass Spectrometry
8.
Am J Physiol Endocrinol Metab ; 313(5): E552-E562, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28698283

ABSTRACT

Skeletal muscle mitochondrial protein synthesis is regulated in part by insulin. The development of insulin resistance with diet-induced obesity may therefore contribute to impairments to protein synthesis and decreased mitochondrial respiration. Yet the impact of diet-induced obesity and insulin resistance on mitochondrial energetics is controversial, with reports varying from decreases to increases in mitochondrial respiration. We investigated the impact of changes in insulin sensitivity on long-term rates of mitochondrial protein synthesis as a mechanism for changes to mitochondrial respiration in skeletal muscle. Insulin resistance was induced in C57BL/6J mice using 4 wk of a high-fat compared with a low-fat diet. For 8 additional weeks, diets were enriched with pioglitazone to restore insulin sensitivity compared with nonenriched control low-fat or high-fat diets. Skeletal muscle mitochondrial protein synthesis was measured using deuterium oxide labeling during weeks 10-12 High-resolution respirometry was performed using palmitoyl-l-carnitine, glutamate+malate, and glutamate+malate+succinate as substrates for mitochondria isolated from quadriceps. Mitochondrial protein synthesis and palmitoyl- l-carnitine oxidation were increased in mice consuming a high-fat diet, regardless of differences in insulin sensitivity with pioglitazone treatment. There was no effect of diet or pioglitazone treatment on ADP-stimulated respiration or H2O2 emission using glutamate+malate or glutamate+malate+succinate. The results demonstrate no impairments to mitochondrial protein synthesis or respiration following induction of insulin resistance. Instead, mitochondrial protein synthesis was increased with a high-fat diet and may contribute to remodeling of the mitochondria to increase lipid oxidation capacity. Mitochondrial adaptations with a high-fat diet appear driven by nutrient availability, not intrinsic defects that contribute to insulin resistance.


Subject(s)
Diet, High-Fat , Dietary Fats/pharmacology , Hypoglycemic Agents/pharmacology , Mitochondrial Proteins/biosynthesis , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Protein Biosynthesis/drug effects , Animals , Insulin/metabolism , Insulin Resistance , Lipid Metabolism/drug effects , Male , Mice , Mice, Inbred C57BL , Mitochondria, Muscle/drug effects , Mitochondria, Muscle/metabolism , Oxidation-Reduction/drug effects , Pioglitazone , Protein Biosynthesis/physiology , Thiazolidinediones/pharmacology , Up-Regulation/drug effects
9.
Physiol Rep ; 12(11): e16093, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38845596

ABSTRACT

Regular exercise and antihyperglycemic drugs are front-line treatments for type-2 diabetes and related metabolic disorders. Leading drugs are metformin, sodium-glucose cotransporter-2 inhibitors, and glucagon-like peptide 1 receptor agonists. Each class has strong individual efficacy to treat hyperglycemia, yet the combination with exercise can yield varied results, some of which include blunting of expected metabolic benefits. Skeletal muscle insulin resistance contributes to the development of type-2 diabetes while improvements in skeletal muscle insulin signaling are among key adaptations to exercise training. The current review identifies recent advances into the mechanisms, with an emphasis on skeletal muscle, of the interaction between exercise and these common antihyperglycemic drugs. The review is written toward researchers and thus highlights specific gaps in knowledge and considerations for future study directions.


Subject(s)
Exercise , Hypoglycemic Agents , Muscle, Skeletal , Humans , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Exercise/physiology , Animals , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Insulin Resistance/physiology , Metformin/pharmacology , Metformin/therapeutic use , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use
10.
Drug Discov Today Dis Mech ; 10(1-2): e35-e40, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23997790

ABSTRACT

Obesity and obesity-related complications are epidemic issues currently plaguing much of the developed world with increasing associated morbidity, mortality, and economic burden. In this brief review, we discuss emerging evidence and remaining questions regarding the possible role for mitochondrial sirtuin 3 as a therapeutic target for the treatment of obesity-related metabolic diseases.

11.
Physiol Rep ; 11(20): e15840, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37857571

ABSTRACT

High-fat diet (HFD) and exercise remodel skeletal muscle mitochondria. The electron transfer flavoproteins (ETF) transfer reducing equivalents from ß-oxidation into the electron transfer system. Exercise may stimulate the synthesis of ETF proteins to increase lipid respiration. We determined mitochondrial remodeling for lipid respiration through ETF in the context of higher mitochondrial abundance/capacity seen in female mice. We hypothesized HFD would be a greater stimulus than exercise to remodel ETF and lipid pathways through increased protein synthesis alongside increased lipid respiration. Female C57BL/6J mice (n = 15 per group) consumed HFD or low-fat diet (LFD) for 4 weeks then remained sedentary (SED) or completed 8 weeks of treadmill training (EX). We determined mitochondrial lipid respiration, RNA abundance, individual protein synthesis, and abundance for ETFα, ETFß, and ETF dehydrogenase (ETFDH). HFD increased absolute and relative lipid respiration (p = 0.018 and p = 0.034) and RNA abundance for ETFα (p = 0.026), ETFß (p = 0.003), and ETFDH (p = 0.0003). HFD increased synthesis for ETFα and ETFDH (p = 0.0007 and p = 0.002). EX increased synthesis of ETFß and ETFDH (p = 0.008 and p = 0.006). Higher synthesis rates of ETF were not always reflected in greater protein abundance. Greater synthesis of ETF during HFD indicates mitochondrial remodeling which may contribute higher mitochondrial lipid respiration through enhanced ETF function.


Subject(s)
Diet, High-Fat , Electron-Transferring Flavoproteins , Female , Animals , Mice , Electron-Transferring Flavoproteins/genetics , Electron-Transferring Flavoproteins/metabolism , Diet, High-Fat/adverse effects , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Lipids , Respiration , RNA/metabolism
12.
J Appl Physiol (1985) ; 134(2): 339-355, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36603044

ABSTRACT

Aerobic training remodels the quantity and quality (function per unit) of skeletal muscle mitochondria to promote substrate oxidation, however, there remain key gaps in understanding the underlying mechanisms during initial training adaptations. We used short-term high-intensity interval training (HIIT) to determine changes to mitochondrial respiration and regulatory pathways that occur early in remodeling. Fifteen normal-weight sedentary adults started seven sessions of HIIT over 14 days and 14 participants completed the intervention. We collected vastus lateralis biopsies before and 48 h after HIIT to determine mitochondrial respiration, RNA sequencing, and Western blotting for proteins of mitochondrial respiration and degradation via autophagy. HIIT increased respiration per mitochondrial protein for lipid (+23% P = 0.020), complex I (+18%, P = 0.0015), complex I + II (+14%, P < 0.0001), and complex II (+24% P < 0.0001). Transcripts that increased with HIIT identified several gene sets of mitochondrial respiration, particularly for complex I, whereas transcripts that decreased identified pathways of DNA and chromatin remodeling. HIIT lowered protein abundance of autophagy markers for p62 (-19%, P = 0.012) and LC3 II/I (-20%, P = 0.004) in whole tissue lysates but not isolated mitochondria. Meal tolerance testing revealed HIIT increased the change in whole body respiratory exchange ratio and lowered cumulative plasma insulin concentrations. Gene transcripts and respiratory function indicate remodeling of mitochondria within 2 wk of HIIT. Overall changes are consistent with increased protein quality driving rapid improvements in substrate oxidation.NEW & NOTEWORTHY Aerobic training stimulates mitochondrial metabolism in skeletal muscle that is linked to improvements to whole body fuel metabolism. The mechanisms driving changes to the quantity and quality (function per unit) of mitochondria are less known. We used seven sessions of high-intensity interval training (HIIT) to determine functional changes and mechanisms of mitochondrial remodeling in skeletal muscle. HIIT increased mitochondrial respiration per mass for fatty acids, complex I, and complex II substrates. HIIT-induced remodeling pathways including gene transcripts for mitochondrial respiration (via RNA sequencing of muscle tissue) and proteins related to complex I respiration. We conclude that an early feature of aerobic training is increased mitochondrial protein quality via improved respiration and induction of mitochondrial transcriptional patterns.


Subject(s)
High-Intensity Interval Training , Adult , Humans , Muscle, Skeletal/physiology , Oxidation-Reduction , Mitochondria, Muscle/metabolism , Respiration
13.
Physiol Behav ; 255: 113940, 2022 10 15.
Article in English | MEDLINE | ID: mdl-35961609

ABSTRACT

Cephalic phase insulin release (CPIR) is a rapid pulse of insulin secreted within minutes of food-related sensory stimulation. Understanding the mechanisms underlying CPIR in humans has been hindered by its small observed effect size and high variability within and between studies. One contributing factor to these limitations may be the use of peripherally measured insulin as an indicator of secreted insulin, since a substantial portion of insulin is metabolized by the liver before delivery to peripheral circulation. Here, we investigated the use of c-peptide, which is co-secreted in equimolar amounts to insulin from pancreatic beta cells, as a proxy for insulin secretion during the cephalic phase period. Changes in insulin and c-peptide were monitored in 18 adults over two repeated sessions following oral stimulation with a sucrose-containing gelatin stimulus. We found that, on average, insulin and c-peptide release followed a similar time course over the cephalic phase period, but that c-peptide showed a greater effect size. Importantly, when insulin and c-peptide concentrations were compared across sessions, we found that changes in c-peptide were significantly correlated at the 2 min (r = 0.50, p = 0.03) and 4 min (r = 0.65, p = 0.003) time points, as well as when participants' highest c-peptide concentrations were considered (r = 0.64, p = 0.004). In contrast, no significant correlations were observed for changes in insulin measured from the sessions (r = -0.06-0.35, p > 0.05). Herein, we detail the individual variability of insulin and c-peptide concentrations measured during the cephalic phase period, and identify c-peptide as a valuable metric for insulin secretion alongside insulin concentrations when investigating CPIR.


Subject(s)
Blood Glucose , Insulin , Adult , Blood Glucose/metabolism , C-Peptide/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Sucrose
14.
Physiol Rep ; 10(24): e15543, 2022 12.
Article in English | MEDLINE | ID: mdl-36541261

ABSTRACT

High dietary fat intake induces significant whole-body and skeletal muscle adaptations in mice, including increased capacity for fat oxidation and mitochondrial biogenesis. The impact of a diet that is high in fat and simple sugars (i.e., western diet [WD]), particularly on regulation of skeletal muscle mitochondrial function, is less understood. The purpose of the current study was to determine physiologic adaptations in mitochondrial respiratory capacity in skeletal muscle during short-term consumption of WD, including if adaptive responses to WD-feeding are modified by concurrent exercise training or may be sex-specific. Male and female C57BL/6J mice were randomized to consume low-fat diet (LFD) or WD for 4 weeks, with some WD-fed mice also performing concurrent treadmill training (WD + Ex). Group sizes were n = 4-7. Whole-body metabolism was measured using in-cage assessment of food intake and energy expenditure, DXA body composition analysis and insulin tolerance testing. High-resolution respirometry of mitochondria isolated from quadriceps muscle was used to determine skeletal muscle mitochondrial respiratory function. Male mice fed WD gained mass (p < 0.001), due to increased fat mass (p < 0.001), and displayed greater respiratory capacity for both lipid and non-lipid substrates compared with LFD mice (p < 0.05). There was no effect of concurrent treadmill training on maximal respiration (WD + Ex vs. WD). Female mice had non-significant changes in body mass and composition as a function of the interventions, and no differences in skeletal muscle mitochondrial oxidative capacity. These findings indicate 4 weeks of WD feeding can increase skeletal muscle mitochondrial oxidative capacity among male mice; whereas WD, with or without exercise, had minimal impact on mass gain and skeletal muscle respiratory capacity among female mice. The translational relevance is that mitochondrial adaptation to increases in dietary fat intake that model WD may be related to differences in weight gain among male and female mice.


Subject(s)
Diet, Western , Mitochondria, Muscle , Physical Conditioning, Animal , Animals , Female , Male , Mice , Diet, High-Fat/adverse effects , Diet, Western/adverse effects , Dietary Fats/metabolism , Mice, Inbred C57BL , Mitochondria , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Respiration
15.
Clin Auton Res ; 21(2): 81-7, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21113641

ABSTRACT

PURPOSE: Electromagnetic fields have been administered, with mixed success, in order to treat a variety of ailments. Transcranial magnetic stimulation (TMS) elicits brief changes in peripheral sympathetic nervous system (SNS) activity. The purpose of this study was to explore the utility of repetitive trans-spinal magnetic stimulation (rTSMS) for acute and prolonged modulation of SNS in adult humans. METHODS: 23 healthy men and women were randomly assigned to receive either rTSMS (figure-eight coil aligned with the sixth and seventh cervical vertebrae; 10 Hz; n = 14, at 100% intensity of stimulator output) or sham stimulation (n = 13). RESULTS: Compared with sham, rTSMS did not affect skeletal muscle SNS activity (via microneurography) during the 60-s or 10-min period following stimulation. rTSMS also had no effect on R-to-R interval (RR(int)) and standard deviation of RR(int) (a marker of heart rate variability), blood pressure or plasma concentrations of norepinephrine, epinephrine, insulin and glucose (condition/time interaction, all P > 0.10). CONCLUSION: These data suggest that rTSMS does not influence SNS in adults. While rTSMS represents a novel application of TMS technology, further study and perhaps modification of the technique is required before use in clinical studies of peripheral SNS function.


Subject(s)
Magnetic Field Therapy/methods , Spinal Cord/physiology , Sympathetic Nervous System/physiology , Adult , Female , Heart Rate/physiology , Humans , Male , Muscle, Skeletal/physiology
16.
Med Sci Sports Exerc ; 53(3): 624-632, 2021 03 01.
Article in English | MEDLINE | ID: mdl-32796254

ABSTRACT

INTRODUCTION: Evidence from model systems implicates long-chain acyl-coenzyme A synthetase (ACSL) as key regulators of skeletal muscle fat oxidation and fat storage; however, such roles remain underexplored in humans. PURPOSE: We sought to determine the protein expression of ACSL isoforms in skeletal muscle at rest and in response to acute exercise and identify relationships between skeletal muscle ACSL and measures of fat metabolism in humans. METHODS: Sedentary adults (n = 14 [4 males and 10 females], body mass index = 22.2 ± 2.1 kg·m-2, V˙O2max = 32.2 ± 4.5 mL·kg-1⋅min-1) completed two study visits. Trials were identical other than completing 1 h of cycling exercise (65% V˙O2max) or remaining sedentary. Vastus lateralis biopsies were obtained 15-min postexercise (or rest) and 2-h postexercise to determine ACSL protein abundance. Whole-body fat oxidation was assessed at rest and during exercise using indirect calorimetry. Skeletal muscle triacylglycerol (TAG) was measured via lipidomic analysis. RESULTS: We detected protein expression for four of the five known ACSL isoforms in human skeletal muscle. ACSL protein abundances were largely unaltered in the hours after exercise aside from a transient increase in ACSL5 15-min postexercise (P = 0.01 vs rest). Skeletal muscle ACSL1 protein abundance tended to be positively related with whole-body fat oxidation during exercise (P = 0.07, r = 0.53), when skeletal muscle accounts for the majority of energy expenditure. No such relationship between ACSL1 and fat oxidation was observed at rest. Skeletal muscle ACSL6 protein abundance was positively associated with muscle TAG content at rest (P = 0.05, r = 0.57). CONCLUSION: Most ACSL protein isoforms can be detected in human skeletal muscle, with minimal changes in abundance after acute exercise. Our findings agree with those from model systems implicating ACSL1 and ACSL6 as possible determinants of fat oxidation and fat storage within skeletal muscle.


Subject(s)
Coenzyme A Ligases/metabolism , Lipid Metabolism , Muscle, Skeletal/enzymology , Adult , Bicycling/physiology , Female , Humans , Isoenzymes/metabolism , Lipid Peroxidation , Male , Oxygen Consumption/physiology , Sedentary Behavior , Triglycerides/analysis , Young Adult
17.
Med Sci Sports Exerc ; 53(7): 1375-1384, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34127633

ABSTRACT

INTRODUCTION: Skeletal muscle mitochondria have dynamic shifts in oxidative metabolism to meet energy demands of aerobic exercise. Specific complexes oxidize lipid and nonlipid substrates. It is unclear if aerobic exercise stimulates intrinsic oxidative metabolism of mitochondria or varies between substrates. METHODS: We studied mitochondrial metabolism in sedentary male and female adults (n = 11F/4M) who were free of major medical conditions with mean ± SD age of 28 ± 7 yr, peak aerobic capacity of 2.0 ± 0.4 L·min-1, and body mass index of 22.2 ± 2 kg·m-2. Biopsies were collected from the vastus lateralis muscle on separate study days at rest or 15 min after exercise (1 h cycling at 65% peak aerobic capacity). Isolated mitochondria were analyzed using high-resolution respirometry of separate titration protocols for lipid (palmitoylcarnitine, F-linked) and nonlipid substrates (glutamate-malate, N-linked; succinate S-linked). Titration protocols distinguished between oxidative phosphorylation and leak respiration and included the measurement of reactive oxygen species emission (H2O2). Western blotting determined the protein abundance of electron transfer flavoprotein (ETF) subunits, including inhibitory methylation site on ETF-ß. RESULTS: Aerobic exercise induced modest increases in mitochondrial respiration because of increased coupled respiration across F-linked (+13%, P = 0.08), N(S)-linked (+14%, P = 0.09), and N-linked substrates (+17%, P = 0.08). Prior exercise did not change P:O ratio. Electron leak to H2O2 increased 6% increased after exercise (P = 0.06) for lipid substrates but not for nonlipid. The protein abundance of ETF-α or ETF-ß subunit or inhibitory methylation on ETF-ß was not different between rest and after exercise. CONCLUSION: In sedentary adults, the single bout of moderate-intensity cycling induced modest increases for intrinsic mitochondrial oxidative phosphorylation that was consistent across multiple substrates.


Subject(s)
Cell Respiration/physiology , Exercise/physiology , Mitochondria, Muscle/metabolism , Oxidative Phosphorylation , Quadriceps Muscle/metabolism , Adult , Female , Humans , Hydrogen Peroxide/metabolism , Male , Sedentary Behavior , Young Adult
18.
Front Endocrinol (Lausanne) ; 12: 651211, 2021.
Article in English | MEDLINE | ID: mdl-33868178

ABSTRACT

Lipid overload of the mitochondria is linked to the development of insulin resistance in skeletal muscle which may be a contributing factor to the progression of type 2 diabetes during obesity. The targeted degradation of mitochondria through autophagy, termed mitophagy, contributes to the mitochondrial adaptive response to changes in dietary fat. Our previous work demonstrates long-term (2-4 months) consumption of a high-fat diet increases mitochondrial lipid oxidation capacity but does not alter markers of mitophagy in mice. The purpose of this study was to investigate initial stages of mitochondrial respiratory adaptations to high-fat diet and the activation of mitophagy. C57BL/6J mice consumed either a low-fat diet (LFD, 10% fat) or high-fat diet (HFD, 60% fat) for 3 or 7 days. We measured skeletal muscle mitochondrial respiration and protein markers of mitophagy in a mitochondrial-enriched fraction of skeletal muscle. After 3 days of HFD, mice had lower lipid-supported oxidative phosphorylation alongside greater electron leak compared with the LFD group. After 7 days, there were no differences in mitochondrial respiration between diet groups. HFD mice had greater autophagosome formation potential (Beclin-1) and greater activation of mitochondrial autophagy receptors (Bnip3, p62) in isolated mitochondria, but no difference in downstream autophagosome (LC3II) or lysosome (Lamp1) abundance after both 3 and 7 days compared with the LFD groups. In cultured myotubes, palmitate treatment decreased mitochondrial membrane potential and hydrogen peroxide treatment increased accumulation of upstream mitophagy markers. We conclude that several days of high-fat feeding stimulated upstream activation of skeletal muscle mitophagy, potentially through lipid-induced oxidative stress, without downstream changes in respiration.


Subject(s)
Lipids/chemistry , Mitochondria/pathology , Mitophagy/physiology , Muscle, Skeletal/physiology , Animals , Autophagy , Beclin-1/biosynthesis , Diabetes Mellitus, Type 2/genetics , Diet, Fat-Restricted , Diet, High-Fat , Fatty Acids/chemistry , Fatty Acids/metabolism , Hydrogen Peroxide/chemistry , Lipid Peroxidation , Lysosomes/metabolism , Male , Membrane Potential, Mitochondrial , Membrane Proteins/biosynthesis , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/biosynthesis , Mitochondria/metabolism , Mitochondrial Proteins/biosynthesis , Obesity/genetics , Oxidative Stress , Oxygen/chemistry , Phenotype , Reactive Oxygen Species , Time Factors
19.
JCI Insight ; 6(24)2021 12 22.
Article in English | MEDLINE | ID: mdl-34935645

ABSTRACT

Maternal obesity affects nearly one-third of pregnancies and is a major risk factor for nonalcoholic fatty liver disease (NAFLD) in adolescent offspring, yet the mechanisms behind NAFLD remain poorly understood. Here, we demonstrate that nonhuman primate fetuses exposed to maternal Western-style diet (WSD) displayed increased fibrillar collagen deposition in the liver periportal region, with increased ACTA2 and TIMP1 staining, indicating localized hepatic stellate cell (HSC) and myofibroblast activation. This collagen deposition pattern persisted in 1-year-old offspring, despite weaning to a control diet (CD). Maternal WSD exposure increased the frequency of DCs and reduced memory CD4+ T cells in fetal liver without affecting systemic or hepatic inflammatory cytokines. Switching obese dams from WSD to CD before conception or supplementation of the WSD with resveratrol decreased fetal hepatic collagen deposition and reduced markers of portal triad fibrosis, oxidative stress, and fetal hypoxemia. These results demonstrate that HSCs and myofibroblasts are sensitive to maternal WSD-associated oxidative stress in the fetal liver, which is accompanied by increased periportal collagen deposition, indicative of early fibrogenesis beginning in utero. Alleviating maternal WSD-driven oxidative stress in the fetal liver holds promise for halting steatosis and fibrosis and preventing developmental programming of NAFLD.


Subject(s)
Diet, Western/adverse effects , Liver Cirrhosis/physiopathology , Maternal Nutritional Physiological Phenomena/physiology , Animals , Female , Maternal Exposure , Pregnancy , Primates , Uterus
20.
Obesity (Silver Spring) ; 29(3): 550-561, 2021 03.
Article in English | MEDLINE | ID: mdl-33624435

ABSTRACT

OBJECTIVE: Sex differences in insulin sensitivity are present throughout the life-span, with men having a higher prevalence of insulin resistance and diabetes compared with women. Differences in lean mass, fat mass, and fat distribution-particularly ectopic fat-have all been postulated to contribute to the sexual dimorphism in diabetes risk. Emerging data suggest ectopic lipid composition and subcellular localization are most relevant; however, it is not known whether they explain sex differences in obesity-induced insulin resistance. METHODS: To address this gap, this study evaluated insulin sensitivity and subcellular localization of intramuscular triacylglycerol, diacylglycerol, and sphingolipids as well as muscle acylcarnitines and serum lipidomics in people with obesity. RESULTS: Insulin sensitivity was significantly lower in men (P < 0.05); however, no sex differences were found in localization of intramuscular triacylglycerol, diacylglycerol, or sphingolipids in skeletal muscle. In contrast, men had higher total muscle acylcarnitine (P < 0.05) and long-chain muscle acylcarnitine (P < 0.05), which were related to lower insulin sensitivity (r = -0.42, P < 0.05). Men also displayed higher serum ceramide (P = 0.05) and lysophosphatidylcholine (P < 0.01). CONCLUSIONS: These data reveal novel sex-specific associations between lipid species involved in the coupling of mitochondrial fatty acid transport, ß-oxidation, and tricarboxylic acid cycle flux that may provide therapeutic targets to improve insulin sensitivity.


Subject(s)
Carnitine/analogs & derivatives , Insulin Resistance/physiology , Muscle, Skeletal/metabolism , Adult , Carnitine/analysis , Carnitine/metabolism , Citric Acid Cycle/physiology , Cohort Studies , Female , Glucose Clamp Technique , Glucose Tolerance Test , Humans , Insulin/blood , Lipid Metabolism/physiology , Male , Mitochondria, Muscle/metabolism , Muscle, Skeletal/chemistry , Muscle, Skeletal/ultrastructure , Obesity/etiology , Obesity/metabolism , Oxidation-Reduction , Sex Characteristics , Sphingolipids/metabolism , Subcellular Fractions/chemistry , Subcellular Fractions/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL