Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
Add more filters

Publication year range
1.
Cell ; 134(1): 37-47, 2008 Jul 11.
Article in English | MEDLINE | ID: mdl-18614009

ABSTRACT

Satellite cells reside beneath the basal lamina of skeletal muscle fibers and include cells that act as precursors for muscle growth and repair. Although they share a common anatomical localization and typically are considered a homogeneous population, satellite cells actually exhibit substantial heterogeneity. We used cell-surface marker expression to purify from the satellite cell pool a distinct population of skeletal muscle precursors (SMPs) that function as muscle stem cells. When engrafted into muscle of dystrophin-deficient mdx mice, purified SMPs contributed to up to 94% of myofibers, restoring dystrophin expression and significantly improving muscle histology and contractile function. Transplanted SMPs also entered the satellite cell compartment, renewing the endogenous stem cell pool and participating in subsequent rounds of injury repair. Together, these studies indicate the presence in adult skeletal muscle of prospectively isolatable muscle-forming stem cells and directly demonstrate the efficacy of myogenic stem cell transplant for treating muscle degenerative disease.


Subject(s)
Adult Stem Cells/cytology , Muscle, Skeletal/cytology , Satellite Cells, Skeletal Muscle/cytology , Adult Stem Cells/chemistry , Animals , Cell Separation , Dystrophin/genetics , Dystrophin/metabolism , Humans , Membrane Proteins/analysis , Mice , Mice, Inbred C57BL , Muscle Contraction , Muscle, Skeletal/physiology , Muscular Dystrophy, Animal/therapy , Satellite Cells, Skeletal Muscle/chemistry , Stem Cell Transplantation
2.
J Cell Biochem ; 120(1): 685-696, 2019 01.
Article in English | MEDLINE | ID: mdl-30256437

ABSTRACT

The signaling mechanisms mediating myocardial glucose transport are not fully understood. Sucrose nonfermenting AMP-activated protein kinase (AMPK)-related kinase (SNARK) is an AMPK-related protein kinase that is expressed in the heart and has been implicated in contraction-stimulated glucose transport in mouse skeletal muscle. We first determined if SNARK is phosphorylated on Thr208 , a site critical for SNARK activity. Mice were treated with exercise, ischemia, submaximal insulin, or maximal insulin. Treadmill exercise slightly, but significantly increased SNARK Thr208 phosphorylation. Ischemia also increased SNARK Thr208 phosphorylation, but there was no effect of submaximal or maximal insulin. HL1 cardiomyocytes were used to overexpress wild-type (WT) SNARK and to knockdown endogenous SNARK. Overexpression of WT SNARK had no effect on ischemia-stimulated glucose transport; however, SNARK knockdown significantly decreased ischemia-stimulated glucose transport. SNARK overexpression or knockdown did not alter insulin-stimulated glucose transport or glycogen concentrations. To study SNARK function in vivo, SNARK heterozygous knockout mice (SNARK+/- ) and WT littermates performed treadmill exercise. Exercise-stimulated glucose transport was decreased by ~50% in hearts from SNARK+/- mice. In summary, exercise and ischemia increase SNARK Thr208 phosphorylation in the heart and SNARK regulates exercise-stimulated and ischemia-stimulated glucose transport. SNARK is a novel mediator of insulin-independent glucose transport in the heart.


Subject(s)
Coronary Vessels/metabolism , Glucose/metabolism , Ischemia/metabolism , Myocardium/metabolism , Physical Conditioning, Animal , Protein Serine-Threonine Kinases/genetics , Animals , Biological Transport , Cell Line, Tumor , Gene Knockdown Techniques , Insulin/pharmacology , Male , Mice , Mice, Inbred ICR , Mice, Knockout , Myocytes, Cardiac/metabolism , Phosphorylation , Signal Transduction/drug effects
3.
Biochem Biophys Res Commun ; 493(3): 1236-1242, 2017 11 25.
Article in English | MEDLINE | ID: mdl-28962861

ABSTRACT

Skeletal muscle atrophy is associated with a disruption in protein turnover involving increased protein degradation and suppressed protein synthesis. Although it has been well studied that the IGF-1/PI3K/Akt pathway plays an essential role in the regulation of the protein turnover, molecule(s) that triggers the change in protein turnover still remains to be elucidated. TRB3 has been shown to inhibit Akt through direct binding. In this study, we hypothesized that TRB3 in mouse skeletal muscle negatively regulates protein turnover via the disruption of Akt and its downstream molecules. Muscle-specific TRB3 transgenic (TRB3TG) mice had decreased muscle mass and fiber size, resulting in impaired muscle function. We also found that protein synthesis rate and signaling molecules, mTOR and S6K1, were significantly reduced in TRB3TG mice, whereas the protein breakdown pathway was significantly activated. In contrast, TRB3 knockout mice showed increased muscle mass and had an increase in protein synthesis rate, but decreases in FoxOs, atrogin-1, and MuRF-1. These findings indicate that TRB3 regulates protein synthesis and breakdown via the Akt/mTOR/FoxO pathways.


Subject(s)
Cell Cycle Proteins/metabolism , Muscle, Skeletal/metabolism , Animals , Cell Cycle Proteins/genetics , Female , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O3/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Muscle Proteins/genetics , Muscle, Skeletal/physiopathology , Protein Biosynthesis/genetics , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , SKP Cullin F-Box Protein Ligases/genetics , TOR Serine-Threonine Kinases/metabolism
4.
EMBO Rep ; 16(10): 1378-93, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26303948

ABSTRACT

Brown adipose tissue (BAT) dissipates chemical energy as heat and can counteract obesity. MicroRNAs are emerging as key regulators in development and disease. Combining microRNA and mRNA microarray profiling followed by bioinformatic analyses, we identified miR-455 as a new regulator of brown adipogenesis. miR-455 exhibits a BAT-specific expression pattern and is induced by cold and the browning inducer BMP7. In vitro gain- and loss-of-function studies show that miR-455 regulates brown adipocyte differentiation and thermogenesis. Adipose-specific miR-455 transgenic mice display marked browning of subcutaneous white fat upon cold exposure. miR-455 activates AMPKα1 by targeting HIF1an, and AMPK promotes the brown adipogenic program and mitochondrial biogenesis. Concomitantly, miR-455 also targets the adipogenic suppressors Runx1t1 and Necdin, initiating adipogenic differentiation. Taken together, the data reveal a novel microRNA-regulated signaling network that controls brown adipogenesis and may be a potential therapeutic target for human metabolic disorders.


Subject(s)
Adipocytes, Brown/metabolism , Adipogenesis/genetics , MicroRNAs/genetics , Signal Transduction , AMP-Activated Protein Kinases/metabolism , Adipose Tissue, White , Animals , Cell Differentiation/genetics , Cells, Cultured , Cold Temperature , Humans , Mice , Mice, Transgenic , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Organelle Biogenesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Repressor Proteins/metabolism , Thermogenesis/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Diabetologia ; 59(8): 1769-77, 2016 08.
Article in English | MEDLINE | ID: mdl-27209464

ABSTRACT

AIMS/HYPOTHESIS: Adipose tissue dysfunction is a prime risk factor for the development of metabolic disease. Bone morphogenetic proteins (BMPs) have previously been implicated in adipocyte formation. Here, we investigate the role of BMP signalling in adipose tissue health and systemic glucose homeostasis. METHODS: We employed the Cre/loxP system to generate mouse models with conditional ablation of BMP receptor 1A in differentiating and mature adipocytes, as well as tissue-resident myeloid cells. Metabolic variables were assessed by glucose and insulin tolerance testing, insulin-stimulated glucose uptake and gene expression analysis. RESULTS: Conditional deletion of Bmpr1a using the aP2 (also known as Fabp4)-Cre strain resulted in a complex phenotype. Knockout mice were clearly resistant to age-related impairment of insulin sensitivity during normal and high-fat-diet feeding and showed significantly improved insulin-stimulated glucose uptake in brown adipose tissue and skeletal muscle. Moreover, knockouts displayed significant reduction of variables of adipose tissue inflammation. Deletion of Bmpr1a in myeloid cells had no impact on insulin sensitivity, while ablation of Bmpr1a in mature adipocytes partially recapitulated the initial phenotype from aP2-Cre driven deletion. Co-cultivation of macrophages with pre-adipocytes lacking Bmpr1a markedly reduced expression of proinflammatory genes. CONCLUSIONS/INTERPRETATION: Our findings show that altered BMP signalling in adipose tissue affects the tissue's metabolic properties and systemic insulin resistance by altering the pattern of immune cell infiltration. The phenotype is due to ablation of Bmpr1a specifically in pre-adipocytes and maturing adipocytes rather than an immune cell-autonomous effect. Mechanistically, we provide evidence for a BMP-mediated direct crosstalk between pre-adipocytes and macrophages.


Subject(s)
Adipose Tissue/metabolism , Bone Morphogenetic Protein Receptors, Type I/metabolism , Insulin Resistance/physiology , Adipocytes/metabolism , Animals , Bone Morphogenetic Protein Receptors, Type I/genetics , Diet, High-Fat/adverse effects , Fatty Acids, Nonesterified/blood , Glucose/metabolism , Insulin/blood , Insulin Resistance/genetics , Interleukin-6/blood , Mice , Mice, Knockout , Tumor Necrosis Factor-alpha/blood
6.
Am J Physiol Regul Integr Comp Physiol ; 306(12): R925-33, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24740654

ABSTRACT

Increasing evidence suggests that TRB3, a mammalian homolog of Drosophila tribbles, plays an important role in cell growth, differentiation, and metabolism. In the liver, TRB3 binds and inhibits Akt activity, whereas in adipocytes, TRB3 upregulates fatty acid oxidation. In cultured muscle cells, TRB3 has been identified as a potential regulator of insulin signaling. However, little is known about the function and regulation of TRB3 in skeletal muscle in vivo. In the current study, we found that 4 wk of voluntary wheel running (6.6 ± 0.4 km/day) increased TRB3 mRNA by 1.6-fold and protein by 2.5-fold in the triceps muscle. Consistent with this finding, muscle-specific transgenic mice that overexpress TRB3 (TG) had a pronounced increase in exercise capacity compared with wild-type (WT) littermates (TG: 1,535 ± 283; WT: 644 ± 67 joules). The increase in exercise capacity in TRB3 TG mice was not associated with changes in glucose uptake or glycogen levels; however, these mice displayed a dramatic shift toward a more oxidative/fatigue-resistant (type I/IIA) muscle fiber type, including threefold more type I fibers in soleus muscles. Skeletal muscle from TRB3 TG mice had significantly decreased PPARα expression, twofold higher levels of miR208b and miR499, and corresponding increases in the myosin heavy chain isoforms Myh7 and Myb7b, which encode these microRNAs. These findings suggest that TRB3 regulates muscle fiber type via a peroxisome proliferator-activated receptor-α (PPAR-α)-regulated miR499/miR208b pathway, revealing a novel function for TRB3 in the regulation of skeletal muscle fiber type and exercise capacity.


Subject(s)
Cell Cycle Proteins/metabolism , Exercise Tolerance/physiology , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Physical Conditioning, Animal/physiology , Up-Regulation/physiology , Animals , Cell Cycle Proteins/genetics , Glucose/metabolism , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/metabolism , Models, Animal , Myosin Heavy Chains/metabolism , PPAR alpha/metabolism
7.
Nat Metab ; 6(5): 963-979, 2024 May.
Article in English | MEDLINE | ID: mdl-38693320

ABSTRACT

Subcutaneous white adipose tissue (scWAT) is a dynamic storage and secretory organ that regulates systemic homeostasis, yet the impact of endurance exercise training (ExT) and sex on its molecular landscape is not fully established. Utilizing an integrative multi-omics approach, and leveraging data generated by the Molecular Transducers of Physical Activity Consortium (MoTrPAC), we show profound sexual dimorphism in the scWAT of sedentary rats and in the dynamic response of this tissue to ExT. Specifically, the scWAT of sedentary females displays -omic signatures related to insulin signaling and adipogenesis, whereas the scWAT of sedentary males is enriched in terms related to aerobic metabolism. These sex-specific -omic signatures are preserved or amplified with ExT. Integration of multi-omic analyses with phenotypic measures identifies molecular hubs predicted to drive sexually distinct responses to training. Overall, this study underscores the powerful impact of sex on adipose tissue biology and provides a rich resource to investigate the scWAT response to ExT.


Subject(s)
Adipose Tissue, White , Physical Conditioning, Animal , Sex Characteristics , Subcutaneous Fat , Animals , Male , Female , Rats , Adipose Tissue, White/metabolism , Subcutaneous Fat/metabolism , Adipogenesis , Rats, Sprague-Dawley , Multiomics
8.
Am J Physiol Endocrinol Metab ; 304(6): E631-9, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23321478

ABSTRACT

Type 2 diabetes is characterized by reduced muscle glycogen synthesis. The key enzyme in this process, glycogen synthase (GS), is activated via proximal insulin signaling, but the exact molecular events remain unknown. Previously, we demonstrated that phosphorylation of Thr³°8 on Akt (p-Akt-Thr³°8), Akt2 activity, and GS activity in muscle were positively associated with insulin sensitivity. Here, in the same study population, we determined the influence of several upstream elements in the canonical PI3K signaling on muscle GS activation. One-hundred eighty-one nondiabetic twins were examined with the euglycemic hyperinsulinemic clamp combined with excision of muscle biopsies. Insulin signaling was evaluated at the levels of the insulin receptor, IRS-1-associated PI3K (IRS-1-PI3K), Akt, and GS employing activity assays and phosphospecific Western blotting. The insulin-stimulated GS activity was positively associated with p-Akt-Thr³°8 (P = 0.01) and Akt2 activity (P = 0.04) but not p-Akt-Ser47³ or IRS-1-PI3K activity. Furthermore, p-Akt-Thr³°8 and Akt2 activity were negatively associated with NH2-terminal GS phosphorylation (P = 0.001 for both), which in turn was negatively associated with insulin-stimulated GS activity (P < 0.001). We found no association between COOH-terminal GS phosphorylation and Akt or GS activity. Employing whole body Akt2-knockout mice, we validated the necessity for Akt2 in insulin-mediated GS activation. However, since insulin did not affect NH2-terminal phosphorylation in mice, we could not use this model to validate the observed association between GS NH2-terminal phosphorylation and Akt activity in humans. In conclusion, our study suggests that although COOH-terminal dephosphorylation is likely necessary for GS activation, Akt2-dependent NH2-terminal dephosphorylation may be the site for "fine-tuning" insulin-mediated GS activation in humans.


Subject(s)
Glycogen Synthase/metabolism , Insulin/metabolism , Muscle, Skeletal/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Adult , Aged , Animals , Cohort Studies , Cross-Sectional Studies , Enzyme Activation , Female , Humans , Insulin/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Muscle, Skeletal/enzymology , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/genetics , Threonine/metabolism , Young Adult
9.
Proc Natl Acad Sci U S A ; 107(35): 15541-6, 2010 Aug 31.
Article in English | MEDLINE | ID: mdl-20713714

ABSTRACT

The signaling mechanisms that mediate the important effects of contraction to increase glucose transport in skeletal muscle are not well understood, but are known to occur through an insulin-independent mechanism. Muscle-specific knockout of LKB1, an upstream kinase for AMPK and AMPK-related protein kinases, significantly inhibited contraction-stimulated glucose transport. This finding, in conjunction with previous studies of ablated AMPKalpha2 activity showing no effect on contraction-stimulated glucose transport, suggests that one or more AMPK-related protein kinases are important for this process. Muscle contraction increased sucrose nonfermenting AMPK-related kinase (SNARK) activity, an effect blunted in the muscle-specific LKB1 knockout mice. Expression of a mutant SNARK in mouse tibialis anterior muscle impaired contraction-stimulated, but not insulin-stimulated, glucose transport. Whole-body SNARK heterozygotic knockout mice also had impaired contraction-stimulated glucose transport in skeletal muscle, and knockdown of SNARK in C2C12 muscle cells impaired sorbitol-stimulated glucose transport. SNARK is activated by muscle contraction and is a unique mediator of contraction-stimulated glucose transport in skeletal muscle.


Subject(s)
Glucose/metabolism , Muscle Contraction/physiology , Muscle, Skeletal/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Adult , Animals , Biological Transport/drug effects , Blotting, Western , Cell Line , Enzyme Activation , Exercise/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , In Vitro Techniques , Insulin/pharmacology , Male , Mice , Mice, Knockout , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/metabolism , Phosphorylation , Physical Conditioning, Animal/physiology , Protein Serine-Threonine Kinases/genetics , RNA Interference , Sorbitol/pharmacology
10.
iScience ; 26(7): 107226, 2023 Jul 21.
Article in English | MEDLINE | ID: mdl-37485354

ABSTRACT

Physical activity is important for type 2 diabetes treatment, yet the underlying mechanisms for these beneficial effects of exercise are not fully understood. Here, we investigated the effects of exercise training on biphasic ß-cell insulin secretory function, a key factor regulating blood glucose. Adults with type 2 diabetes (7F/3M, age 49 ± 5 years, BMI 30 ± 3 kg/m2) completed a 10-week moderate-intensity exercise program and multiple components of glucose homeostasis were measured. Training improved glycemic control, insulin sensitivity, and processing of proinsulin-to-insulin. Training increased late phase ß-cell function by 38% (p = 0.01), which was correlated with changes in VO2peak suggesting training response-dependent effects. Ras-Responsive Element Binding Protein 1 (RREB1) concentrations, a protein postulated to increase type 2 diabetes risk, were inversely correlated with increases in training-induced late-phase disposition index, consistent with an inhibitory role of RREB1 on insulin secretion. Moderate-intensity exercise training improves late-phase ß-cell function and glycemic control in adults with type 2 diabetes.

11.
bioRxiv ; 2023 Oct 18.
Article in English | MEDLINE | ID: mdl-37905018

ABSTRACT

Exercise training and cold exposure both improve systemic metabolism, but the mechanisms are not well-established. We tested the hypothesis that adaptations to inguinal white adipose tissue (iWAT) are critical for these beneficial effects by determining the impact of exercise-trained and cold-exposed iWAT on systemic glucose metabolism and the iWAT proteome and secretome. Transplanting trained iWAT into sedentary mice improved glucose tolerance, while cold-exposed iWAT transplantation showed no such benefit. Compared to training, cold led to more pronounced alterations in the iWAT proteome and secretome, downregulating >2,000 proteins but also boosting iWAT's thermogenic capacity. In contrast, only training increased extracellular space and vesicle transport proteins, and only training upregulated proteins that correlate with favorable fasting glucose, suggesting fundamental changes in trained iWAT that mediate tissue-to-tissue communication. This study defines the unique exercise training- and cold exposure-induced iWAT proteomes, revealing distinct mechanisms for the beneficial effects of these interventions on metabolic health.

12.
Cell Rep ; 42(4): 112392, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37058410

ABSTRACT

Inguinal white adipose tissue (iWAT) is essential for the beneficial effects of exercise training on metabolic health. The underlying mechanisms for these effects are not fully understood, and here, we test the hypothesis that exercise training results in a more favorable iWAT structural phenotype. Using biochemical, imaging, and multi-omics analyses, we find that 11 days of wheel running in male mice causes profound iWAT remodeling including decreased extracellular matrix (ECM) deposition and increased vascularization and innervation. We identify adipose stem cells as one of the main contributors to training-induced ECM remodeling, show that the PRDM16 transcriptional complex is necessary for iWAT remodeling and beiging, and discover neuronal growth regulator 1 (NEGR1) as a link between PRDM16 and neuritogenesis. Moreover, we find that training causes a shift from hypertrophic to insulin-sensitive adipocyte subpopulations. Exercise training leads to remarkable adaptations to iWAT structure and cell-type composition that can confer beneficial changes in tissue metabolism.


Subject(s)
Adipose Tissue, White , Motor Activity , Male , Mice , Animals , Adipose Tissue, White/metabolism , Adaptation, Physiological/physiology , Acclimatization/physiology , Transcription Factors/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue/metabolism , Mice, Inbred C57BL , Cell Adhesion Molecules, Neuronal/metabolism
13.
J Biol Chem ; 286(6): 4133-40, 2011 Feb 11.
Article in English | MEDLINE | ID: mdl-21127070

ABSTRACT

Contraction and insulin promote glucose uptake in skeletal muscle through GLUT4 translocation to cell surface membranes. Although the signaling mechanisms leading to GLUT4 translocation have been extensively studied in muscle, the cellular transport machinery is poorly understood. Myo1c is an actin-based motor protein implicated in GLUT4 translocation in adipocytes; however, the expression profile and role of Myo1c in skeletal muscle have not been investigated. Myo1c protein abundance was higher in more oxidative skeletal muscles and heart. Voluntary wheel exercise (4 weeks, 8.2 ± 0.8 km/day), which increased the oxidative profile of the triceps muscle, significantly increased Myo1c protein levels by ∼2-fold versus sedentary controls. In contrast, high fat feeding (9 weeks, 60% fat) significantly reduced Myo1c by 17% in tibialis anterior muscle. To study Myo1c regulation of glucose uptake, we expressed wild-type Myo1c or Myo1c mutated at the ATPase catalytic site (K111A-Myo1c) in mouse tibialis anterior muscles in vivo and assessed glucose uptake in vivo in the basal state, in response to 15 min of in situ contraction, and 15 min following maximal insulin injection (16.6 units/kg of body weight). Expression of wild-type Myo1c or K111A-Myo1c had no effect on basal glucose uptake. However, expression of wild-type Myo1c significantly increased contraction- and insulin-stimulated glucose uptake, whereas expression of K111A-Myo1c decreased both contraction-stimulated and insulin-stimulated glucose uptake. Neither wild-type nor K111A-Myo1c expression altered GLUT4 expression, and neither affected contraction- or insulin-stimulated signaling proteins. Myo1c is a novel mediator of both insulin-stimulated and contraction-stimulated glucose uptake in skeletal muscle.


Subject(s)
Glucose Transporter Type 4/biosynthesis , Glucose/metabolism , Insulin/metabolism , Muscle Contraction/physiology , Muscle, Skeletal/metabolism , Myosins/metabolism , Animals , Catalytic Domain , Female , Gene Expression Regulation/physiology , Glucose/genetics , Glucose Transporter Type 4/genetics , Insulin/genetics , Mice , Mice, Inbred ICR , Mutation , Myosin Type I , Myosins/genetics , Time Factors
14.
Mol Metab ; 60: 101490, 2022 06.
Article in English | MEDLINE | ID: mdl-35398278

ABSTRACT

OBJECTIVE: A major factor in the growing world-wide epidemic of obesity and type 2 diabetes is the increased risk of transmission of metabolic disease from obese mothers to both first (F1) and second (F2) generation offspring. Fortunately, recent pre-clinical studies demonstrate that exercise before and during pregnancy improves F1 metabolic health, providing a potential means to disrupt this cycle of disease. Whether the beneficial effects of maternal exercise can also be transmitted to the F2 generation has not been investigated. METHODS: C57BL/6 female mice were fed a chow or high-fat diet (HFD) and housed in individual cages with or without running wheels for 2 wks before breeding and during gestation. Male F1 offspring were sedentary and chow-fed, and at 8-weeks of age were bred with age-matched females from untreated parents. This resulted in 4 F2 groups based on grandmaternal treatment: chow sedentary; chow trained; HFD sedentary; HFD trained. F2 were sedentary and chow-fed and studied up to 52-weeks of age. RESULTS: We find that grandmaternal exercise improves glucose tolerance and decreases fat mass in adult F2 males and females, in the absence of any treatment intervention of the F1 after birth. Grandmaternal exercise also improves F2 liver metabolic function, including favorable effects on gene and miRNA expression, triglyceride concentrations and hepatocyte glucose production. CONCLUSION: Grandmaternal exercise has beneficial effects on the metabolic health of grandoffspring, demonstrating an important means by which exercise during pregnancy could help reduce the worldwide incidence of obesity and type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2 , Prenatal Exposure Delayed Effects , Animals , Diabetes Mellitus, Type 2/complications , Female , Glucose/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/metabolism
15.
Diabetes ; 71(10): 2094-2105, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35838316

ABSTRACT

That maternal and paternal exercise improve the metabolic health of adult offspring is well established. Tissue and serum metabolites play a fundamental role in the health of an organism, but how parental exercise affects offspring tissue and serum metabolites has not yet been investigated. Here, male and female breeders were fed a high-fat diet and housed with or without running wheels before breeding (males) and before and during gestation (females). Offspring were sedentary and chow fed, with parents as follows: sedentary (Sed), maternal exercise (MatEx), paternal exercise (PatEx), or maternal+paternal exercise (Mat+PatEx). Adult offspring from all parental exercise groups had similar improvement in glucose tolerance and hepatic glucose production. Targeted metabolomics was performed in offspring serum, liver, and triceps muscle. Offspring from MatEx, PatEx, and Mat+PatEx each had a unique tissue metabolite signature, but Mat+PatEx offspring had an additive phenotype relative to MatEx or PatEx alone in a subset of liver and muscle metabolites. Tissue metabolites consistently indicated that the metabolites altered with parental exercise contribute to enhanced fatty acid oxidation. These data identify distinct tissue-specific adaptations and mechanisms for parental exercise-induced improvement in offspring metabolic health. Further mining of this data set could aid the development of novel therapeutic targets to combat metabolic diseases.


Subject(s)
Diet, High-Fat , Physical Conditioning, Animal , Animals , Diet, High-Fat/adverse effects , Fatty Acids , Female , Glucose/metabolism , Liver/metabolism , Male , Physical Conditioning, Animal/physiology
16.
Diabetes ; 71(6): 1170-1181, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35290440

ABSTRACT

Preclinical studies reveal maternal exercise as a promising intervention to reduce the transmission of multigenerational metabolic dysfunction caused by maternal obesity. The benefits of maternal exercise on offspring health may arise from multiple factors and have recently been shown to involve DNA demethylation of critical hepatic genes leading to enhanced glucose metabolism in offspring. Histone modification is another epigenetic regulator, yet the effects of maternal obesity and exercise on histone methylation in offspring are not known. Here, we find that maternal high-fat diet (HFD; 60% kcal from fat) induced dysregulation of offspring liver glucose metabolism in C57BL/6 mice through a mechanism involving increased reactive oxygen species, WD repeat-containing 82 (WDR82) carbonylation, and inactivation of histone H3 lysine 4 (H3K4) methyltransferase leading to decreased H3K4me3 at the promoters of glucose metabolic genes. Remarkably, the entire signal was restored if the HFD-fed dams had exercised during pregnancy. WDR82 overexpression in hepatoblasts mimicked the effects of maternal exercise on H3K4me3 levels. Placental superoxide dismutase 3 (SOD3), but not antioxidant treatment with N-acetylcysteine was necessary for the regulation of H3K4me3, gene expression, and glucose metabolism. Maternal exercise regulates a multicomponent epigenetic system in the fetal liver resulting in the transmission of the benefits of exercise to offspring.


Subject(s)
Obesity, Maternal , Prenatal Exposure Delayed Effects , Animals , Chromosomal Proteins, Non-Histone/metabolism , Diet, High-Fat , Female , Glucose/metabolism , Histones/metabolism , Humans , Mice , Mice, Inbred C57BL , Placenta/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
17.
Cell Metab ; 34(10): 1578-1593.e6, 2022 10 04.
Article in English | MEDLINE | ID: mdl-36198295

ABSTRACT

Exercise training is critical for the prevention and treatment of obesity, but its underlying mechanisms remain incompletely understood given the challenge of profiling heterogeneous effects across multiple tissues and cell types. Here, we address this challenge and opposing effects of exercise and high-fat diet (HFD)-induced obesity at single-cell resolution in subcutaneous and visceral white adipose tissue and skeletal muscle in mice with diet and exercise training interventions. We identify a prominent role of mesenchymal stem cells (MSCs) in obesity and exercise-induced tissue adaptation. Among the pathways regulated by exercise and HFD in MSCs across the three tissues, extracellular matrix remodeling and circadian rhythm are the most prominent. Inferred cell-cell interactions implicate within- and multi-tissue crosstalk centered around MSCs. Overall, our work reveals the intricacies and diversity of multi-tissue molecular responses to exercise and obesity and uncovers a previously underappreciated role of MSCs in tissue-specific and multi-tissue beneficial effects of exercise.


Subject(s)
Adipose Tissue , Mesenchymal Stem Cells , Adipose Tissue/metabolism , Animals , Diet, High-Fat , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Obesity/metabolism
18.
Cell Metab ; 3(5): 355-66, 2006 May.
Article in English | MEDLINE | ID: mdl-16679293

ABSTRACT

The evolutionarily conserved phosphoinositide 3-kinase (PI3K) signaling pathway mediates both the metabolic effects of insulin and the growth-promoting effects of insulin-like growth factor-1 (IGF-1). We have generated mice deficient in both the p85alpha/p55alpha/p50alpha and the p85beta regulatory subunits of class I(A) PI3K in skeletal muscles. PI3K signaling in the muscle of these animals is severely impaired, leading to a significant reduction in muscle weight and fiber size. These mice also exhibit muscle insulin resistance and whole-body glucose intolerance. Despite their ability to maintain normal fasting and fed blood glucose levels, these mice show increased body fat content and elevated serum free fatty acid and triglyceride levels. These results demonstrate that in vivo p85 is a critical mediator of class I(A) PI3K signaling in the regulation of muscle growth and metabolism. Our finding also indicates that compromised muscle PI3K signaling could contribute to symptoms of hyperlipidemia associated with human type 2 diabetes.


Subject(s)
Hyperlipidemias/enzymology , Muscle, Skeletal/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Animals , Blood Glucose/metabolism , Body Composition , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/pathology , Fatty Acids, Nonesterified/blood , Glucose/metabolism , Glucose Intolerance , Hyperlipidemias/blood , Hyperlipidemias/pathology , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Insulin Resistance , Insulin-Like Growth Factor I/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/drug effects , Muscle, Skeletal/growth & development , Muscle, Skeletal/pathology , Muscular Atrophy/blood , Muscular Atrophy/enzymology , Muscular Atrophy/pathology , Phosphatidylinositol 3-Kinases/deficiency , Phosphatidylinositol 3-Kinases/genetics , Triglycerides/blood
19.
Biochim Biophys Acta ; 1802(7-8): 593-600, 2010.
Article in English | MEDLINE | ID: mdl-20441792

ABSTRACT

Energy deprivation in the myocardium is associated with impaired heart function and increased morbidity. LKB1 is a kinase that is required for activation of AMP-activated protein kinase (AMPK) as well as 13 AMPK-related protein kinases. AMPK stimulates ATP production during ischemia and prevents post-ischemic dysfunction. We used the Cre-Lox system to generate mice where LKB1 was selectively knocked out in cardiomyocytes and muscle cells (LKB1-KO) to assess the role of LKB1 on cardiac function in these mice. Heart rates of LKB1-KO mice were reduced and ventricle diameter was increased. Ex vivo, cardiac function was impaired during aerobic perfusion of isolated working hearts, and recovery of function after ischemia was reduced. Although oxidative metabolism and mitochondrial function were normal, the AMP/ATP ratio was increased in LKB1-KO hearts. This was associated with a complete ablation of AMPKalpha2 activity, and a stimulation of signaling through the mammalian target of rapamycin. Our results establish a critical role for LKB1 for normal cardiac function under both aerobic conditions and during recovery after ischemia. Ablation of LKB1 leads to a decreased cardiac efficiency despite normal mitochondrial oxidative metabolism.


Subject(s)
Energy Metabolism/genetics , Heart Diseases/genetics , Heart Diseases/physiopathology , Myocardium/metabolism , Protein Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinases , Aerobiosis/physiology , Animals , Cells, Cultured , Gene Deletion , Heart/physiology , Heart Diseases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Mice , Mice, Knockout , Muscles/metabolism , Myocardial Ischemia/genetics , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Organ Culture Techniques , Organ Specificity/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Signal Transduction/genetics , Signal Transduction/physiology , TOR Serine-Threonine Kinases
20.
Am J Physiol Endocrinol Metab ; 301(1): E164-71, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21505148

ABSTRACT

Exercise and weight loss are cornerstones in the treatment and prevention of type 2 diabetes, and both interventions function to increase insulin sensitivity and glucose uptake into skeletal muscle. Studies in rodents demonstrate that the underlying mechanism for glucose uptake in muscle involves site-specific phosphorylation of the Rab-GTPase-activating proteins AS160 (TBC1D4) and TBC1D1. Multiple kinases, including Akt and AMPK, phosphorylate TBC1D1 and AS160 on distinct residues, regulating their activity and allowing for GLUT4 translocation. In contrast to extensive rodent-based studies, the regulation of AS160 and TBC1D1 in human skeletal muscle is not well understood. In this study, we determined the effects of dietary intervention and a single bout of exercise on TBC1D1 and AS160 site-specific phosphorylation in human skeletal muscle. Ten obese (BMI 33.4 ± 2.4, M-value 4.3 ± 0.5) subjects were studied at baseline and after a 2-wk dietary intervention. Muscle biopsies were obtained from the subjects in the resting (basal) state and immediately following a 30-min exercise bout (70% Vo(2 max)). Muscle lysates were analyzed for AMPK activity and Akt phosphorylation and for TBC1D1 and AS160 phosphorylation on known or putative AMPK and Akt sites as follows: AS160 Ser(711) (AMPK), TBC1D1 Ser(231) (AMPK), TBC1D1 Ser(660) (AMPK), TBC1D1 Ser(700) (AMPK), and TBC1D1 Thr(590) (Akt). The diet intervention that consisted of a major shift in the macronutrient composition resulted in a 4.2 ± 0.4 kg weight loss (P < 0.001) and a significant increase in insulin sensitivity (M value 5.6 ± 0.6), but surprisingly, there was no effect on expression or phosphorylation of any of the muscle-signaling proteins. Exercise increased muscle AMPKα2 activity but did not increase Akt phosphorylation. Exercise increased phosphorylation on AS160 Ser(711), TBC1D1 Ser(231), and TBC1D1 Ser(660) but had no effect on TBC1D1 Ser(700). Exercise did not increase TBC1D1 Thr(590) phosphorylation or TBC1D1/AS160 PAS phosphorylation, consistent with the lack of Akt activation. These data demonstrate that a single bout of exercise regulates TBC1D1 and AS160 phosphorylation on multiple sites in human skeletal muscle.


Subject(s)
Exercise/physiology , GTPase-Activating Proteins/metabolism , Muscle, Skeletal/metabolism , Protein Kinases/metabolism , AMP-Activated Protein Kinases/metabolism , Diet , Female , Glucose Transporter Type 4/analysis , Glucose Transporter Type 4/metabolism , Glycogen/analysis , Glycogen/metabolism , Humans , Isoenzymes/metabolism , Male , Phosphorylation , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-akt/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL