Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Rev Cardiol ; 18(10): 701-711, 2021 10.
Article in English | MEDLINE | ID: mdl-33772258

ABSTRACT

Increases in calorie consumption and sedentary lifestyles are fuelling a global pandemic of cardiometabolic diseases, including coronary artery disease, diabetes mellitus, cardiomyopathy and heart failure. These lifestyle factors, when combined with genetic predispositions, increase the levels of circulating lipids, which can accumulate in non-adipose tissues, including blood vessel walls and the heart. The metabolism of these lipids produces bioactive intermediates that disrupt cellular function and survival. A compelling body of evidence suggests that sphingolipids, such as ceramides, account for much of the tissue damage in these cardiometabolic diseases. In humans, serum ceramide levels are proving to be accurate biomarkers of adverse cardiovascular disease outcomes. In mice and rats, pharmacological inhibition or depletion of enzymes driving de novo ceramide synthesis prevents the development of diabetes, atherosclerosis, hypertension and heart failure. In cultured cells and isolated tissues, ceramides perturb mitochondrial function, block fuel usage, disrupt vasodilatation and promote apoptosis. In this Review, we discuss the body of literature suggesting that ceramides are drivers - and not merely passengers - on the road to cardiovascular disease. Moreover, we explore the feasibility of therapeutic strategies to lower ceramide levels to improve cardiovascular health.


Subject(s)
Cardiovascular Diseases , Ceramides , Sphingolipids , Animals , Cardiovascular Diseases/epidemiology , Ceramides/metabolism , Mice , Rats , Sphingolipids/metabolism
2.
Biochem Biophys Res Commun ; 547: 29-35, 2021 04 02.
Article in English | MEDLINE | ID: mdl-33592376

ABSTRACT

Brown adipose tissue (BAT) and stimulating adaptive thermogenesis have been implicated as anti-obese and anti-diabetic tissues due to their ability to dissipate energy as heat by the expression of UCP1. We have recently demonstrated that TRB3 impairs differentiation of brown preadipocytes via inhibiting insulin signaling. However, the roles of the protein in BAT function and thermogenesis in vivo have not yet been established. For this study we tested the hypothesis that TRB3 mediates obesity- and diabetes-induced impairments in BAT differentiation and function, and that inhibition of TRB3 improves BAT function. TRB3 expression was increased in BAT from high-fat fed mice and ob/ob mice, which was associated with decreased UCP1 expression. Incubation of brown adipocytes with palmitate increased TRB3 expression and decreased UCP1. Knockout of TRB3 in mice displayed higher UCP1 expression in BAT and cold resistance. Incubation of brown adipocytes with ER stressors increased TRB3 but decreased UCP1 and ER stress markers were elevated in BAT from high-fat fed mice and ob/ob mice. Finally, high-fat feeding in TRB3KO mice were protected from obesity-induced glucose intolerance and displayed cold resistance and higher expression of BAT-specific markers. These data demonstrate that high-fat feeding and obesity increase TRB3 in BAT, resulting in impaired tissue function.


Subject(s)
Adipose Tissue, Brown/metabolism , Cell Cycle Proteins/metabolism , Obesity/metabolism , Uncoupling Protein 1/metabolism , Adipose Tissue, Brown/pathology , Adipose Tissue, Brown/physiology , Animals , Cells, Cultured , Diet, High-Fat , Disease Models, Animal , Endoplasmic Reticulum Stress , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Obesity/pathology , Signal Transduction , Thermogenesis
3.
Biochem Biophys Res Commun ; 519(3): 633-638, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31540695

ABSTRACT

AMP-activated protein kinase (AMPK) is a member of Ser/Thr kinases that has been shown to regulate energy balance. Although recent studies have demonstrated the function of AMPK in adipose tissue using different fat-specific AMPK knockout mouse models, the results were somewhat inconsistent. For this study, we tested the hypothesis that AMPK in adipose tissue regulates whole body glucose metabolism. To determine the role of adipose tissue AMPK in vivo, we generated fat-specific AMPKα1/α2 knockout mice (AMPKFKO) using the Cre-loxP system. Body weights of AMPKFKO mice were not different between 8 and 27 weeks of age. Furthermore, tissue weights (liver, kidney, muscle, heart and white and brown adipose tissue) were similar to wild type littermates and DEXA scan analysis revealed no differences in percentages of body fat and lean mass. Knockout of AMPKα1/α2 in adipose tissue abolished basal and AICAR-stimulated phosphorylation of AMPK and Acetyl-CoA Carboxylase, a downstream of AMPK. Despite of the ablation of AICAR-stimulated AMPK phosphorylation, the blood glucose-lowering effect of AICAR injection (i.p.) was normal in AMPKFKO mice. In addition, AMPKFKO displayed normal fasting blood glucose concentration, glucose tolerance, insulin tolerance, and insulin signaling, indicating normal whole body glucose metabolism. These data demonstrate that adipose tissue AMPK plays a minimum role in whole body glucose metabolism on a chow diet.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adipose Tissue/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Glucose/metabolism , Ribonucleotides/metabolism , AMP-Activated Protein Kinases/deficiency , Aminoimidazole Carboxamide/administration & dosage , Aminoimidazole Carboxamide/metabolism , Animals , Diet , Injections, Intraperitoneal , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ribonucleotides/administration & dosage
4.
FASEB J ; 33(4): 5654-5666, 2019 04.
Article in English | MEDLINE | ID: mdl-30681896

ABSTRACT

Tribbles 3 (TRB3) is a pseudokinase that has been found in multiple tissues in response to various stress stimuli, such as nutrient deprivation and endoplasmic reticulum (ER) stress. We recently found that TRB3 has the potential to regulate skeletal muscle mass at the basal state. However, it has not yet been explored whether TRB3 regulates skeletal muscle mass under atrophic conditions. Here, we report that food deprivation for 48 h in mice significantly reduces muscle mass by ∼15% and increases TRB3 expression, which is associated with increased ER stress. Interestingly, inhibition of ER stress in C2C12 myotubes reduces food deprivation-induced expression of TRB3 and muscle-specific E3-ubiquitin ligases. In further in vivo experiments, muscle-specific TRB3 transgenic mice increase food deprivation-induced muscle atrophy compared with wild-type (WT) littermates presumably by the increased proteolysis. On the other hand, TRB3 knockout mice ameliorate food deprivation-induced atrophy compared with WT littermates by preserving a higher protein synthesis rate. These results indicate that TRB3 plays a pivotal role in skeletal muscle mass regulation under food deprivation-induced muscle atrophy and TRB3 could be a pharmaceutical target to prevent skeletal muscle atrophy.-Choi, R. H., McConahay, A., Silvestre, J. G., Moriscot, A. S., Carson, J. A., Koh, H.-J. TRB3 regulates skeletal muscle mass in food deprivation-induced atrophy.


Subject(s)
Cell Cycle Proteins/metabolism , Food Deprivation/physiology , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Animals , Cell Line , Endoplasmic Reticulum Stress/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Muscle Fibers, Skeletal/metabolism , Signal Transduction/physiology , Ubiquitin-Protein Ligases/metabolism
5.
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
6.
Biochem Biophys Res Commun ; 470(4): 783-91, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26801556

ABSTRACT

Recent studies have demonstrated that adult humans have substantial amounts of functioning brown adipose tissue (BAT). Since BAT has been implicated as an anti-obese and anti-diabetic tissue, it is important to understand the signaling molecules that regulate BAT function. There has been a link between insulin signaling and BAT metabolism as deletion or pharmaceutical inhibition of insulin signaling impairs BAT differentiation and function. Tribbles 3 (TRB3) is a pseudo kinase that has been shown to regulate metabolism and insulin signaling in multiple tissues but the role of TRB3 in BAT has not been studied. In this study, we found that TRB3 expression was present in BAT and overexpression of TRB3 in brown preadipocytes impaired differentiation and decreased expression of BAT markers. Furthermore, TRB3 overexpression resulted in significantly lower oxygen consumption rates for basal and proton leakage, indicating decreased BAT activity. Based on previous studies showing that deletion or pharmaceutical inhibition of insulin signaling impairs BAT differentiation and function, we assessed insulin signaling in brown preadipocytes and BAT in vivo. Overexpression of TRB3 in cells impaired insulin-stimulated IRS1 and Akt phosphorylation, whereas TRB3KO mice displayed improved IRS1 and Akt phosphorylation. Finally, deletion of IRS1 abolished the function of TRB3 to regulate BAT differentiation and metabolism. These data demonstrate that TRB3 inhibits insulin signaling in BAT, resulting in impaired differentiation and function.


Subject(s)
Adipocytes, Brown/cytology , Adipocytes, Brown/metabolism , Adipogenesis/physiology , Cell Cycle Proteins/metabolism , Insulin/physiology , Animals , Cell Differentiation/physiology , Cells, Cultured , Down-Regulation , Mice , Mice, Inbred C57BL , Signal Transduction/physiology
7.
Amino Acids ; 47(7): 1389-98, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25837301

ABSTRACT

Sustamine™ (SUS) is a dipeptide composed of alanine and glutamine (AlaGln). Glutamine has been suggested to increase muscle protein accretion; however, the underlying molecular mechanisms of glutamine on muscle protein metabolism following resistance exercise have not been fully addressed. In the present study, 2-month-old rats climbed a ladder 10 times with a weight equal to 75 % of their body mass attached at the tail. Rats were then orally administered one of four solutions: placebo (PLA-glycine = 0.52 g/kg), whey protein (WP = 0.4 g/kg), low dose of SUS (LSUS = 0.1 g/kg), or high dose of SUS (HSUS = 0.5 g/kg). An additional group of sedentary (SED) rats was intubated with glycine (0.52 g/kg) at the same time as the ladder-climbing rats. Blood samples were collected immediately after exercise and at either 20 or 40 min after recovery. The flexor hallucis longus (FHL), a muscle used for climbing, was excised at 20 or 40 min post exercise and analyzed for proteins regulating protein synthesis and degradation. All supplements elevated the phosphorylation of FOXO3A above SED at 20 min post exercise, but only the SUS supplements significantly reduced the phosphorylation of AMPK and NF-kB p65. SUS supplements had no effect on mTOR signaling, but WP supplementation yielded a greater phosphorylation of mTOR, p70S6k, and rpS6 compared with PLA at 20 min post exercise. However, by 40 min post exercise, phosphorylation of mTOR and rpS6 in PLA had risen to levels not different than WP. These results suggest that SUS blocks the activation of intracellular signals for MPB, whereas WP accelerates mRNA translation.


Subject(s)
Dipeptides/pharmacology , Gene Expression Regulation/drug effects , Proteolysis , Adenylate Kinase/metabolism , Animals , Drug Evaluation, Preclinical , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Male , Muscle Proteins/genetics , NF-kappa B/metabolism , Phosphorylation , Protein Biosynthesis , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Resistance Training , Ribosomal Protein S6/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Whey Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...