Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Diabetes Ther ; 15(3): 623-637, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38240875

ABSTRACT

INTRODUCTION: Real-world data provide insight into how medications perform in clinical practice. The PIONEER REAL Switzerland study aimed to understand clinical outcomes with oral semaglutide in adults with type 2 diabetes (T2D). METHODS: PIONEER REAL Switzerland was a 34-44-week, multicentre, prospective, non-interventional, single-arm study of adults with T2D naïve to injectable glucose-lowering medication who were initiated on oral semaglutide in routine clinical practice. The primary endpoint was change in glycated haemoglobin (HbA1c) from baseline (BL) to end of study (EOS); secondary endpoints included change in body weight (BW) from BL to EOS and the proportion of participants achieving HbA1c < 7.0% and the composite endpoints HbA1c reduction ≥ 1%-points with BW reduction ≥ 3% or ≥ 5% at EOS. Safety was assessed in participants who received ≥ 1 dose of oral semaglutide. RESULTS: Of the 185 participants (female/male, n = 67/118) initiating oral semaglutide, 168 (90.8%) completed the study and 143 (77.3%) remained on treatment with oral semaglutide at EOS. At BL, participants had a mean age of 62 years, diabetes duration of 6.4 years, HbA1c of 7.7%, BW of 95.6 kg and body mass index of 33.2 kg/m2; 56.2% of participants were receiving glucose-lowering medications. Significant reductions were observed for HbA1c (estimated change - 0.91%; 95% confidence interval [CI] - 1.10, - 0.71; p < 0.0001) and BW (estimated change - 4.85%; 95% CI - 5.70, - 4.00; p < 0.0001). In total, 139 adverse events (AEs) were reported in 65 (35.1%) participants; most were mild or moderate. The most frequent AEs were gastrointestinal disorders (27.0%); 31 AEs in 20 (10.8%) participants led to discontinuation of oral semaglutide. Six serious AEs were reported; all were considered unlikely to be related to oral semaglutide. CONCLUSION: People living with T2D treated with oral semaglutide in Switzerland achieved clinically significant reductions in HbA1c and BW, with no new safety signals. CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov: NCT04537624. A graphical abstract is available for this article.

2.
Nat Commun ; 11(1): 1642, 2020 04 02.
Article in English | MEDLINE | ID: mdl-32242025

ABSTRACT

Increasing energy expenditure via induction of adipose tissue browning has become an appealing strategy to treat obesity and associated metabolic complications. Herein, we identify adipocyte-expressed apoptosis signal-regulating kinase 1 (ASK1) as regulator of adipose tissue browning. High fat diet-fed adipocyte-specific ASK1 knockout mice reveal increased UCP1 protein levels in inguinal adipose tissue concomitant with elevated energy expenditure, reduced obesity and ameliorated glucose tolerance compared to control littermates. In addition, ASK1-depletion blunts LPS-mediated downregulation of isoproterenol-induced UCP1 in subcutaneous fat both in vitro and in vivo. Conversely, adipocyte-specific ASK1 overexpression in chow-fed mice attenuates cold-induced UCP1 protein levels in inguinal fat. Mechanistically, ASK1 phosphorylates interferon regulatory factor 3 (IRF3) resulting in reduced Ucp1 expression. Taken together, our studies unravel a role of ASK1 in mediating the inhibitory effect of caloric surplus or LPS-treatment on adipose tissue browning. Adipocyte ASK1 might be a pharmacological target to combat obesity and associated morbidities.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , MAP Kinase Kinase Kinase 5/metabolism , Obesity/metabolism , Animals , Energy Metabolism , Female , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , MAP Kinase Kinase Kinase 5/genetics , Male , Mice , Mice, Knockout , Obesity/genetics , Phosphorylation , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
3.
Diabetes ; 67(1): 36-45, 2018 01.
Article in English | MEDLINE | ID: mdl-29066599

ABSTRACT

We recently showed that interleukin (IL)-6-type cytokine signaling in adipocytes induces free fatty acid release from visceral adipocytes, thereby promoting obesity-induced hepatic insulin resistance and steatosis. In addition, IL-6-type cytokines may increase the release of leptin from adipocytes and by those means induce glucagon-like peptide 1 (GLP-1) secretion. We thus hypothesized that IL-6-type cytokine signaling in adipocytes may regulate insulin secretion. To this end, mice with adipocyte-specific knockout of gp130, the signal transducer protein of IL-6, were fed a high-fat diet for 12 weeks. Compared with control littermates, knockout mice showed impaired glucose tolerance and circulating leptin, GLP-1, and insulin levels were reduced. In line, leptin release from isolated adipocytes was reduced, and intestinal proprotein convertase subtilisin/kexin type 1 (Pcsk1) expression, the gene encoding PC1/3, which controls GLP-1 production, was decreased in knockout mice. Importantly, treatment with the GLP-1 receptor antagonist exendin 9-39 abolished the observed difference in glucose tolerance between control and knockout mice. Ex vivo, supernatant collected from isolated adipocytes of gp130 knockout mice blunted Pcsk1 expression and GLP-1 release from GLUTag cells. In contrast, glucose- and GLP-1-stimulated insulin secretion was not affected in islets of knockout mice. In conclusion, adipocyte-specific IL-6 signaling induces intestinal GLP-1 release to enhance insulin secretion, thereby counteracting insulin resistance in obesity.


Subject(s)
Adipocytes/drug effects , Adipocytes/metabolism , Cytokines/pharmacology , Glucagon-Like Peptide 1/metabolism , Interleukin-6/pharmacology , Animals , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/metabolism , Eating , Glucose Tolerance Test , Leptin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Proprotein Convertase 1/genetics , Proprotein Convertase 1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects
4.
Nat Commun ; 8(1): 480, 2017 09 07.
Article in English | MEDLINE | ID: mdl-28883393

ABSTRACT

Nonalcoholic fatty liver disease is one of the most prevalent metabolic disorders and it tightly associates with obesity, type 2 diabetes, and cardiovascular disease. Reduced mitochondrial lipid oxidation contributes to hepatic fatty acid accumulation. Here, we show that the Fas cell surface death receptor (Fas/CD95/Apo-1) regulates hepatic mitochondrial metabolism. Hepatic Fas overexpression in chow-fed mice compromises fatty acid oxidation, mitochondrial respiration, and the abundance of mitochondrial respiratory complexes promoting hepatic lipid accumulation and insulin resistance. In line, hepatocyte-specific ablation of Fas improves mitochondrial function and ameliorates high-fat-diet-induced hepatic steatosis, glucose tolerance, and insulin resistance. Mechanistically, Fas impairs fatty acid oxidation via the BH3 interacting-domain death agonist (BID). Mice with genetic or pharmacological inhibition of BID are protected from Fas-mediated impairment of mitochondrial oxidation and hepatic steatosis. We suggest Fas as a potential novel therapeutic target to treat obesity-associated fatty liver and insulin resistance.Hepatic steatosis is a common disease closely associated with metabolic syndrome and insulin resistance. Here Item et al. show that Fas, a member of the TNF receptor superfamily, contributes to mitochondrial dysfunction, steatosis development, and insulin resistance under high fat diet.


Subject(s)
Lipid Metabolism/physiology , Liver/metabolism , Mitochondria, Liver/metabolism , fas Receptor/metabolism , Animals , Diet, High-Fat , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Fatty Acids/metabolism , Insulin Resistance , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Mice, Transgenic , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/genetics , Triglycerides/metabolism , fas Receptor/genetics
5.
Nat Immunol ; 18(3): 283-292, 2017 03.
Article in English | MEDLINE | ID: mdl-28092375

ABSTRACT

The deleterious effect of chronic activation of the IL-1ß system on type 2 diabetes and other metabolic diseases is well documented. However, a possible physiological role for IL-1ß in glucose metabolism has remained unexplored. Here we found that feeding induced a physiological increase in the number of peritoneal macrophages that secreted IL-1ß, in a glucose-dependent manner. Subsequently, IL-1ß contributed to the postprandial stimulation of insulin secretion. Accordingly, lack of endogenous IL-1ß signaling in mice during refeeding and obesity diminished the concentration of insulin in plasma. IL-1ß and insulin increased the uptake of glucose into macrophages, and insulin reinforced a pro-inflammatory pattern via the insulin receptor, glucose metabolism, production of reactive oxygen species, and secretion of IL-1ß mediated by the NLRP3 inflammasome. Postprandial inflammation might be limited by normalization of glycemia, since it was prevented by inhibition of the sodium-glucose cotransporter SGLT2. Our findings identify a physiological role for IL-1ß and insulin in the regulation of both metabolism and immunity.


Subject(s)
Diabetes Mellitus, Type 2/immunology , Inflammation/immunology , Insulin-Secreting Cells/physiology , Interleukin-1beta/metabolism , Macrophages/physiology , Animals , Cells, Cultured , Glucose/metabolism , Humans , Inflammasomes/metabolism , Insulin/metabolism , Interleukin-1beta/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Postprandial Period , Reactive Oxygen Species/metabolism , Signal Transduction , Sodium-Glucose Transporter 2/metabolism
6.
Diabetes ; 65(1): 140-8, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26384383

ABSTRACT

Hepatic steatosis and insulin resistance are among the most prevalent metabolic disorders and are tightly associated with obesity and type 2 diabetes. However, the underlying mechanisms linking obesity to hepatic lipid accumulation and insulin resistance are incompletely understood. Glycoprotein 130 (gp130) is the common signal transducer of all interleukin 6 (IL-6) cytokines. We provide evidence that gp130-mediated adipose tissue lipolysis promotes hepatic steatosis and insulin resistance. In obese mice, adipocyte-specific gp130 deletion reduced basal lipolysis and enhanced insulin's ability to suppress lipolysis from mesenteric but not epididymal adipocytes. Consistently, free fatty acid levels were reduced in portal but not in systemic circulation of obese knockout mice. Of note, adipocyte-specific gp130 knockout mice were protected from high-fat diet-induced hepatic steatosis as well as from insulin resistance. In humans, omental but not subcutaneous IL-6 mRNA expression correlated positively with liver lipid accumulation (r = 0.31, P < 0.05) and negatively with hyperinsulinemic-euglycemic clamp glucose infusion rate (r = -0.28, P < 0.05). The results show that IL-6 cytokine-induced lipolysis may be restricted to mesenteric white adipose tissue and that it contributes to hepatic insulin resistance and steatosis. Therefore, blocking IL-6 cytokine signaling in (mesenteric) adipocytes may be a novel approach to blunting detrimental fat-liver crosstalk in obesity.


Subject(s)
Abdominal Fat/metabolism , Cytokine Receptor gp130/genetics , Fatty Liver/genetics , Insulin Resistance , Interleukin-6/genetics , Liver/metabolism , Obesity/genetics , Subcutaneous Fat/metabolism , Adipocytes/metabolism , Adipose Tissue, White/metabolism , Animals , Blotting, Western , Cytokine Receptor gp130/metabolism , Fatty Acids, Nonesterified/metabolism , Fatty Liver/metabolism , Fatty Liver/pathology , Female , Glucose Clamp Technique , Humans , Interleukin-6/metabolism , Lipolysis , Liver/pathology , Male , Mice , Mice, Knockout , Obesity/metabolism , Omentum/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
7.
Diabetes ; 64(4): 1131-41, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25325737

ABSTRACT

Reduced kidney mass and/or function may result in multiple metabolic derangements, including insulin resistance. However, underlying mechanisms are poorly understood. Herein, we aimed to determine the impact of reduced kidney mass on glucose metabolism in lean and obese mice. To that end, 7-week-old C57BL/6J mice underwent uninephrectomy (UniNx) or sham operation. After surgery, animals were fed either a chow (standard) diet or a high-fat diet (HFD), and glucose homeostasis was assessed 20 weeks after surgery. Intraperitoneal glucose tolerance was similar in sham-operated and UniNx mice. However, insulin-stimulated glucose disposal in vivo was significantly diminished in UniNx mice, whereas insulin-stimulated glucose uptake into isolated skeletal muscle was similar in sham-operated and UniNx mice. Of note, capillary density was significantly reduced in skeletal muscle of HFD-fed UniNx mice. In contrast, hepatic insulin sensitivity was improved in UniNx mice. Furthermore, adipose tissue hypoxia-inducible factor 1α expression and inflammation were reduced in HFD-fed UniNx mice. Treatment with the angiotensin II receptor blocker telmisartan improved glucose tolerance and hepatic insulin sensitivity in HFD-fed sham-operated but not UniNx mice. In conclusion, UniNx protects from obesity-induced adipose tissue inflammation and hepatic insulin resistance, but it reduces muscle capillary density and, thus, deteriorates HFD-induced skeletal muscle glucose disposal.


Subject(s)
Adipose Tissue/metabolism , Insulin Resistance/physiology , Kidney/metabolism , Liver/metabolism , Muscle, Skeletal/metabolism , Obesity/metabolism , Animals , Diet, High-Fat , Fatty Liver/metabolism , Glucose/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/metabolism , Insulin/metabolism , Kidney/surgery , Male , Mice , Mice, Obese , Triglycerides/metabolism
8.
Adipocyte ; 3(2): 115-20, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24719784

ABSTRACT

A short bout of high fat diet (HFD) impairs glucose tolerance and hepatic insulin sensitivity. We recently identified adipose tissue inflammation and resulting dysfunctional adipose tissue-liver cross-talk as an early event in the development of HFD-induced hepatic insulin resistance. In particular, reducing white adipose tissue (WAT) inflammation by adipocyte-specific depletion of Fas/CD95 protected mice from developing hepatic insulin resistance but not hepatic steatosis. Herein, we expanded our previous work and determined the impact of four days of HFD on lipolytic activity of isolated adipocytes. Compared with chow-fed mice, the degree of basal and isoproterenol-stimulated free fatty acid (FFA) and glycerol release was similar in HFD-fed animals. Moreover, insulin's ability to suppress lipolysis remained intact, suggesting retained insulin sensitivity. Despite unaltered lipolysis, circulating FFA concentrations were greatly increased in non-fasted HFD-fed mice. In conclusion, a short-term HFD challenge does not affect lipolytic function of adipocytes. The observed increase of circulating FFA levels in randomly fed animals may rather be the result of increased dietary fat supply.

9.
Am J Physiol Regul Integr Comp Physiol ; 306(11): R861-7, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24694381

ABSTRACT

Contracting muscle releases interleukin-6 (IL-6) enabling the metabolic switch from carbohydrate to fat utilization. Similarly, metabolism is switched during transition from fed to fasting state. Herein, we examined a putative role for IL-6 in the metabolic adaptation to normal fasting. In lean C57BL/6J mice, 6 h of food withdrawal increased gene transcription levels of IL-6 in skeletal muscle but not in white adipose tissue. Concomitantly, circulating IL-6 and free fatty acid (FFA) levels were significantly increased, whereas respiratory quotient (RQ) was reduced in 6-h fasted mice. In white adipose tissue, phosphorylation of hormone-sensitive lipase (HSL) was increased on fasting, indicating increased lipolysis. Intriguingly, fasting-induced increase in circulating IL-6 levels and parallel rise in FFA concentration were absent in obese and glucose-intolerant mice. A causative role for IL-6 in the physiological adaptation to fasting was further supported by the fact that fasting-induced increase in circulating FFA levels was significantly blunted in lean IL-6 knockout (KO) and lean C57BL/6J mice treated with neutralizing IL-6 antibody. Consistently, phosphorylation of HSL was significantly reduced in adipose tissue of IL-6-depleted mice. Hence, our findings suggest a novel role for IL-6 in energy supply during early fasting.


Subject(s)
Fasting/psychology , Fatty Acids, Nonesterified/metabolism , Interleukin-6/physiology , Adaptation, Physiological/physiology , Animals , Energy Metabolism/physiology , Interleukin-6/deficiency , Interleukin-6/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal
10.
EMBO Mol Med ; 6(1): 43-56, 2014 01.
Article in English | MEDLINE | ID: mdl-24203314

ABSTRACT

Low-grade inflammation in adipose tissue and liver has been implicated in obesity-associated insulin resistance and type 2 diabetes. Yet, the contribution of inflammatory cells to the pathogenesis of skeletal muscle insulin resistance remains elusive. In a large cohort of obese human individuals, blood monocyte Fas (CD95) expression correlated with systemic and skeletal muscle insulin resistance. To test a causal role for myeloid cell Fas expression in the development of skeletal muscle insulin resistance, we generated myeloid/haematopoietic cell-specific Fas-depleted mice. Myeloid/haematopoietic Fas deficiency prevented the development of glucose intolerance in high fat-fed mice, in ob/ob mice, and in mice acutely challenged by LPS. In vivo, ex vivo and in vitro studies demonstrated preservation of muscle insulin responsiveness with no effect on adipose tissue or liver. Studies using neutralizing antibodies demonstrated a role for TNFα as mediator between myeloid Fas and skeletal muscle insulin resistance, supported by significant correlations between monocyte Fas expression and circulating TNFα in humans. In conclusion, our results demonstrate an unanticipated crosstalk between myeloid cells and skeletal muscle in the development of obesity-associated insulin resistance.


Subject(s)
Gene Expression Regulation , Insulin Resistance , Monocytes/metabolism , Muscle, Skeletal/metabolism , Obesity/metabolism , fas Receptor/metabolism , Adult , Aged , Animals , Antibodies, Neutralizing/immunology , Cohort Studies , Cross-Sectional Studies , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Middle Aged , Obesity/complications , fas Receptor/deficiency , fas Receptor/genetics
11.
Am J Physiol Endocrinol Metab ; 305(3): E388-95, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23736545

ABSTRACT

High-fat feeding for 3-4 days impairs glucose tolerance and hepatic insulin sensitivity. However, it remains unclear whether the evolving hepatic insulin resistance is due to acute lipid overload or the result of induced adipose tissue inflammation and consequent dysfunctional adipose tissue-liver cross-talk. In the present study, feeding C57Bl6/J mice a fat-enriched diet [high-fat diet (HFD)] for 4 days induced glucose intolerance, hepatic insulin resistance (as assessed by hyperinsulinemic euglycemic clamp studies), and hepatic steatosis as well as adipose tissue inflammation (i.e., TNFα expression) compared with standard chow-fed mice. Adipocyte-specific depletion of the antiapoptotic/anti-inflammatory factor Fas (CD95) attenuated adipose tissue inflammation and improved glucose tolerance as well as hepatic insulin sensitivity without altering the level of hepatic steatosis induced by HFD. In summary, our results identify adipose tissue inflammation and resulting dysfunctional adipose tissue-liver cross-talk as an early event in the development of HFD-induced hepatic insulin resistance.


Subject(s)
Adipose Tissue/physiopathology , Diet, High-Fat/adverse effects , Dietary Fats/toxicity , Inflammation/physiopathology , Insulin Resistance/physiology , Liver/drug effects , Adipocytes/physiology , Animals , Area Under Curve , Blotting, Western , Cytokines/metabolism , Exons/genetics , Fatty Acids, Nonesterified/blood , Fatty Liver/metabolism , Glucose Clamp Technique , Glucose Tolerance Test , Insulin/blood , Lipid Metabolism/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA/biosynthesis , RNA/isolation & purification , Real-Time Polymerase Chain Reaction , Triglycerides/blood , fas Receptor/metabolism
12.
Diabetes ; 62(9): 3053-63, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23670971

ABSTRACT

In established obesity, inflammation and macrophage recruitment likely contribute to the development of insulin resistance. In the current study, we set out to explore whether adipose tissue infiltration by neutrophils that occurs early (3 days) after initiating a high-fat diet (HFD) could contribute to the early occurrence of hepatic insulin resistance and to determine the role of cytosolic phospholipase A2α (cPLA2α) in this process. The 3-day HFD caused a significant upregulation of cPLA2α in periepididymal fat and in the liver. A specific antisense oligonucleotide (AS) effectively prevented cPLA2α induction, neutrophil infiltration into adipose tissue (likely involving MIP-2), and protected against 3-day HFD-induced impairment in hepatic insulin signaling and glucose over-production from pyruvate. To sort out the role of adipose neutrophil infiltration independent of cPLA2α induction in the liver, mice were injected intraperitoneally with anti-intracellular adhesion molecule-1 (ICAM-1) antibodies. This effectively prevented neutrophil infiltration without affecting cPLA2α or MIP-2, but like AS, prevented impairment in hepatic insulin signaling, the enhanced pyruvate-to-glucose flux, and the impaired insulin-mediated suppression of hepatic glucose production (assessed by clamp), which were induced by the 3-day HFD. Adipose tissue secretion of tumor necrosis factor-α (TNF-α) was increased by the 3-day HFD, but not if mice were treated with AS or ICAM-1 antibodies. Moreover, systemic TNF-α neutralization prevented 3-day HFD-induced hepatic insulin resistance, suggesting its mediatory role. We propose that an acute, cPLA2α-dependent, neutrophil-dominated inflammatory response of adipose tissue contributes to hepatic insulin resistance and glucose overproduction in the early adaptation to high-fat feeding.


Subject(s)
Adipose Tissue/metabolism , Diet, High-Fat/adverse effects , Group IV Phospholipases A2/metabolism , Insulin Resistance/physiology , Liver/metabolism , Neutrophil Infiltration/physiology , Animals , Immunoblotting , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL
13.
Eur J Appl Physiol ; 113(4): 1081-90, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23086295

ABSTRACT

We previously reported that high load resistance exercise with superimposed whole-body vibration and sustained vascular occlusion (vibroX) markedly improves cycling endurance capacity, increases capillary-to-fibre ratio and skeletal muscle oxidative enzyme activity in untrained young women. These findings are intriguing, since increases in oxidative muscle phenotype and endurance capacity are typically induced by endurance but not heavy resistance exercise. Here, we tested the hypothesis that vibroX activates genes associated with mitochondrial biogenesis and angiogenesis. Eight healthy, recreationally resistance-trained young men performed either vibroX or resistance exercise (RES) in a randomised, cross-over design. Needle biopsies (M. vastus lateralis) were obtained at rest and 3 h post-exercise. Changes in relative gene expression levels were assessed by real-time quantitative PCR. After vibroX, vascular endothelial growth factor and peroxisome proliferator-activated receptor-γ coactivator 1α mRNA abundances increased to 2- and 4.4-fold, respectively, but did not significantly change above resting values after RES. Other genes involved in mitochondrial biogenesis were not affected by either exercise modality. While vibroX increased the expression of hexokinase II, xanthine dehydrogenase, and manganese superoxide dismutase mRNA, there were no changes in these transcripts after RES. This study demonstrates that high load resistance exercise with superimposed whole-body vibration and sustained vascular occlusion activates metabolic and angiogenic gene programs, which are usually activated after endurance but not resistance exercise. Thus, targeted modification of high load resistance exercise by vibration and vascular occlusion might represent a novel strategy to induce endurance-type muscle adaptations.


Subject(s)
Heat-Shock Proteins/genetics , Ischemia/genetics , Muscle Contraction , Physical Endurance , Quadriceps Muscle/blood supply , Quadriceps Muscle/metabolism , RNA, Messenger/metabolism , Resistance Training , Transcription Factors/genetics , Vascular Endothelial Growth Factor A/genetics , Vibration , Adaptation, Physiological , Adult , Analysis of Variance , Biopsy , Cross-Over Studies , Energy Metabolism/genetics , Gene Expression Regulation, Enzymologic , Humans , Ischemia/enzymology , Ischemia/physiopathology , Male , Mitochondria, Muscle/metabolism , Mitochondrial Turnover/genetics , Neovascularization, Physiologic/genetics , Oxidative Stress/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Quadriceps Muscle/enzymology , Quadriceps Muscle/physiopathology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Switzerland , Time Factors , Up-Regulation , Young Adult
14.
Arch Physiol Biochem ; 118(3): 148-55, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22515179

ABSTRACT

The insulin receptor (IR) recruits adaptor proteins, so-called insulin receptor substrates (IRS), to connect with downstream signalling pathways. A family of IRS proteins was defined based on three major common structural elements: Amino-terminal PH and PTB domains that mediate protein-lipid or protein-protein interactions, mostly carboxy-terminal multiple tyrosine residues that serve as binding sites for proteins that contain one or more SH2 domains and serine/threonine-rich regions which may be recognized by negative regulators of insulin action. The current model for the role of IRS proteins therefore combines an adaptor function with the integration of mostly negative input from other signal transduction cascades allowing for modulation of signalling amplitude. In this review we propose an extended version of the adaptor model that can explain how signalling specificity could be implemented at the level of IRS proteins.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Insulin Receptor Substrate Proteins/metabolism , Receptor, Insulin/metabolism , Signal Transduction/genetics , Animals , Binding Sites , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Gene Expression Regulation , Humans , Insulin Receptor Substrate Proteins/genetics , Insulin Resistance/genetics , Models, Biological , Protein Binding , Protein Structure, Tertiary , Receptor, Insulin/genetics , Tyrosine/metabolism
15.
Am J Physiol Regul Integr Comp Physiol ; 301(1): R60-6, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21490367

ABSTRACT

In this study, we examined whether glycemic status influences aerobic function in women with type 1 diabetes and whether aerobic function is reduced relative to healthy women. To this end, we compared several factors determining aerobic function of 29 young sedentary asymptomatic women (CON) with 9 women of similar age and activity level with type 1 diabetes [DIA, HbA1c range = 6.9-8.2%]. Calf muscle mitochondrial capacity was estimated by (31)P-magnetic resonance spectroscopy. Capillarization and muscle fiber oxidative enzyme activity were assessed from vastus lateralis and soleus muscle biopsies. Oxygen uptake and cardiac output were evaluated by ergospirometry and N(2)O/SF(6) rebreathing. Calf muscle mitochondrial capacity was not different between CON and DIA, as indicated by the identical calculated maximal rates of oxidative ATP synthesis [0.0307 (0.0070) vs. 0.0309 (0.0058) s(-1), P = 0.930]. Notably, HbA1c was negatively correlated with mitochondrial capacity in DIA (R(2) = 0.475, P = 0.040). Although HbA1c was negatively correlated with cardiac output (R(2) = 0.742, P = 0.013) in DIA, there was no difference between CON and DIA in maximal oxygen consumption [2.17 (0.34) vs. 2.21 (0.32) l/min, P = 0.764], cardiac output [12.1 (1.9) vs. 12.3 (1.8) l/min, P = 0.783], and endurance capacity [532 (212) vs. 471 (119) s, P = 0.475]. There was also no difference between the two groups either in the oxidative enzyme activity or capillary-to-fiber ratio. We conclude that mitochondrial capacity depends on HbA1c in untrained women with type 1 diabetes but is not reduced relative to untrained healthy women.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/physiopathology , Exercise/physiology , Glycated Hemoglobin/metabolism , Mitochondria, Muscle/physiology , Adult , Biopsy , Cardiac Output/physiology , Case-Control Studies , Female , Humans , Muscle, Skeletal/pathology , Muscle, Skeletal/physiology , Oxygen/blood , Oxygen Consumption/physiology , Physical Endurance/physiology
16.
PLoS One ; 5(6): e10970, 2010 Jun 04.
Article in English | MEDLINE | ID: mdl-20532042

ABSTRACT

Adequate levels of physical activity are at the center of a healthy lifestyle. However, the molecular mechanisms that mediate the beneficial effects of exercise remain enigmatic. This gap in knowledge is caused by the lack of an amenable experimental model system. Therefore, we optimized electric pulse stimulation of muscle cells to closely recapitulate the plastic changes in gene expression observed in a trained skeletal muscle. The exact experimental conditions were established using the peroxisome proliferator-activated receptor gamma coactivator 1alpha (PGC-1alpha) as a marker for an endurance-trained muscle fiber. We subsequently compared the changes in the relative expression of metabolic and myofibrillar genes in the muscle cell system with those observed in mouse muscle in vivo following either an acute or repeated bouts of treadmill exercise. Importantly, in electrically stimulated C2C12 mouse muscle cells, the qualitative transcriptional adaptations were almost identical to those in trained muscle, but differ from the acute effects of exercise on muscle gene expression. In addition, significant alterations in the expression of myofibrillar proteins indicate that this stimulation could be used to modulate the fiber-type of muscle cells in culture. Our data thus describe an experimental cell culture model for the study of at least some of the transcriptional aspects of skeletal muscle adaptation to physical activity. This system will be useful for the study of the molecular mechanisms that regulate exercise adaptation in muscle.


Subject(s)
Electric Stimulation , Gene Expression Regulation , Muscle, Skeletal/physiology , Physical Conditioning, Animal , Animals , Cell Line , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Trans-Activators/metabolism , Transcription Factors , Transcription, Genetic
17.
Exp Cell Res ; 313(4): 805-15, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17222824

ABSTRACT

Ligand-activated insulin receptor (IR) attracts and phosphorylates various substrates such as insulin receptor substrates 1-4 (IRS) and Shc. To investigate how binding affinity for substrate affects signalling we generated chimeric receptors with the beta-chain of the insulin receptor containing NPXY motives with different affinities for receptor substrates. We found that the extent of receptor tyrosine phosphorylation positively correlates with binding affinity towards IRS1/2 but not towards Shc. Moreover, overexpression of IRS1 or IRS2 but not of Shc increased IR tyrosine phosphorylation in a dose-dependent manner, also independent of insulin. Molecular truncations of IRS1 revealed that neither the isolated PH and PTB domains nor the C-terminus with the tyrosine phosphorylation sites alone are sufficient for substrate-dependent receptor activation. Overexpression of IRS1 and IRS2 impaired insulin-induced internalization of the IR in a dose-dependent manner suggesting that IRS proteins prevent endosome-associated receptor dephosphorylation/inactivation. IRS1 and IRS2 could therefore target the activated IR to different cellular compartments. Overexpression of IRS1 and IRS2 inhibited insulin-stimulated activation of the MAP kinases Erk1/2 while it increased/induced activation of Akt/PKB. Finally, overexpression of IRS1 and IRS2 but not of Shc induced DNA synthesis in starved CHO-IR cells independent of exogenous growth factors. Our results demonstrate that variations in cellular IRS1 and IRS2 concentration affect insulin signalling both upstream and downstream and that IRS proteins could play instructive rather than just permissive roles in signal transmission.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cell Proliferation , Insulin/physiology , Intracellular Signaling Peptides and Proteins/physiology , Phosphoproteins/physiology , Receptor, Insulin/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Motifs , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Humans , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1 , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...