Skip to main content
Erschienen in: Diabetologia 6/2010

Open Access 01.06.2010 | Article

Nitric oxide increases cyclic GMP levels, AMP-activated protein kinase (AMPK)α1-specific activity and glucose transport in human skeletal muscle

verfasst von: A. S. Deshmukh, Y. C. Long, T. de Castro Barbosa, H. K. R. Karlsson, S. Glund, W. J. Zavadoski, E. M. Gibbs, H. A. Koistinen, H. Wallberg-Henriksson, J. R. Zierath

Erschienen in: Diabetologia | Ausgabe 6/2010

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Aims/hypothesis

We investigated the direct effect of a nitric oxide donor (spermine NONOate) on glucose transport in isolated human skeletal muscle and L6 skeletal muscle cells. We hypothesised that pharmacological treatment of human skeletal muscle with N-(2-aminoethyl)-N-(2-hydroxy-2-nitrosohydrazino)-1,2-ethylenediamine (spermine NONOate) would increase intracellular cyclic GMP (cGMP) levels and promote glucose transport.

Methods

Skeletal muscle strips were prepared from vastus lateralis muscle biopsies obtained from seven healthy men. Muscle strips were incubated in the absence or presence of 5 mmol/l spermine NONOate or 120 nmol/l insulin. The L6 muscle cells were treated with spermine NONOate (20 µmol/l) and incubated in the absence or presence of insulin (120 nmol/l). The direct effect of spermine NONOate and insulin on glucose transport, cGMP levels and signal transduction was determined.

Results

In human skeletal muscle, spermine NONOate increased glucose transport 2.4-fold (p < 0.05), concomitant with increased cGMP levels (80-fold, p < 0.001). Phosphorylation of components of the canonical insulin signalling cascade was unaltered by spermine NONOate exposure, implicating an insulin-independent signalling mechanism. Consistent with this, spermine NONOate increased AMP-activated protein kinase (AMPK)-α1-associated activity (1.7-fold, p < 0.05). In L6 muscle cells, spermine NONOate increased glucose uptake (p < 0.01) and glycogen synthesis (p < 0.001), an effect that was in addition to that of insulin. Spermine NONOate also elicited a concomitant increase in AMPK and acetyl-CoA carboxylase phosphorylation. In the presence of the guanylate cyclase inhibitor LY-83583 (10 µmol/l), spermine NONOate had no effect on glycogen synthesis and AMPK-α1 phosphorylation.

Conclusions/interpretation

Pharmacological treatment of skeletal muscle with spermine NONOate increases glucose transport via insulin-independent signalling pathways involving increased intracellular cGMP levels and AMPK-α1-associated activity.
Abkürzungen
ACC
Acetyl-CoA carboxylase
AICAR
5′-Aminoimidazole-4-carboxamide ribonucleoside
AMPK
AMP-activated protein kinase
CaMK
Ca2+-calmodulin-dependent protein kinase
cGMP
Cyclic GMP
GSK
Glycogen synthase kinase
KHB
Krebs–Henseleit buffer
NOS
Nitric oxide synthase
PAS
Phospho-Akt substrate
Rab
Rev/Rex activation domain-binding protein
Spermine NONOate
N-(2-Aminoethyl)-N-(2-hydroxy-2-nitrosohydrazino)-1,2-ethylenediamine
TBC1
Tre-2/BUB2/CDC16 domain-containing protein family, member 1

Introduction

Nitric oxide is implicated in a wide array of signalling networks [1]. In rodents and humans, exercise increases skeletal muscle nitric oxide production, concomitant with increased glucose uptake [24]. Nitric oxide biogenesis is catalysed by different isoforms of nitric oxide synthase (NOS) [5], of which neuronal-type NOS and endothelial-type NOS isoforms are produced in skeletal muscle [1]. Neuronal-type NOS is produced at higher levels in human skeletal muscle than brain [6] and therefore is likely to be the major isoform involved in the effect of nitric oxide on glucose metabolism [1]. Acute administration of the NOS inhibitor NG-monomethyl-l-arginine during exercise in humans reduces leg muscle glucose uptake [4], implicating nitric oxide signalling in the mechanism by which exercise controls glucose homeostasis.
The intracellular mechanism by which nitric oxide increases skeletal muscle glucose uptake in humans is incompletely resolved. Intra-femoral artery infusion of a nitric oxide donor increased glucose uptake in healthy participants [7], as well as in type 2 diabetic patients [8]. Furthermore, exposure of isolated rat skeletal muscle to the nitric oxide donor sodium nitroprusside increased glucose transport in a dose-dependent manner [9, 10]. Stimulation of glucose transport by nitric oxide involves the activation of a calcium/contraction- and phosphatidylinositol-3-kinase-independent pathway that acutely increases GLUT4 content at the cell surface [11]. Nitric oxide seems to exert its action on muscle glucose transport partly via activation of guanylate cyclase, leading to elevation of cyclic GMP (cGMP) levels [10, 12]. Indeed, the cGMP analogue 8-bromo-cGMP also increases glucose uptake in isolated rat skeletal muscle [10]. Thus, nitric oxide/cGMP signalling may be part of a novel pathway that regulates skeletal muscle glucose uptake.
The effect of exercise on the acute regulation of skeletal muscle glucose transport has been attributed to several signalling nodes including Ca2+-calmodulin-dependent protein kinase (CaMK)II, AMP-activated protein kinase (AMPK) and nitric oxide [13]. In this regard, the interaction between AMPK and nitric oxide signalling pathways is especially intriguing. AMPK is a serine/threonine protein kinase, which acts as a sensor of cellular energy status and regulates a wide variety of gene regulatory and metabolic pathways, including glucose uptake and fatty acid oxidation in skeletal muscle [14]. AMPK subunits (α1, α2, β1, β2, γ1, γ2, γ3) form a heterotrimeric enzyme consisting of α (catalytic), and β and γ (regulatory) isoforms. In rodent skeletal muscle, sodium nitroprusside increases glucose transport, concomitant with α1-associated AMPK activation [15]. Moreover, chronic exposure of L6 muscle cells to sodium nitroprusside increases Glut4 (also known as SLC2A4) mRNA expression by an AMPK-dependent mechanism [16], positioning AMPK downstream of nitric oxide signalling. AMPK is also considered to be an upstream kinase for NOS, since it phosphorylates and activates endothelial and neuronal NOS [1719]. Thus, a positive feedback interaction between AMPK and NOS in the control of skeletal muscle metabolism is implicated [16].
In the present study, we determined the effect of the nitric oxide donor spermine N-(2-aminoethyl)-N-(2-hydroxy-2-nitrosohydrazino)-1,2-ethylenediamine NONOate on glucose transport and intracellular signalling in isolated human skeletal muscle. Using L6 skeletal muscle cells, we also determined whether spermine NONOate and insulin have additive effects on glucose uptake and intracellular signalling. We hypothesised that pharmacological treatment of human skeletal muscle with a compound that increases cGMP levels may promote glucose uptake.

Methods

Study participants
The study protocol was approved by the regional Ethics Committee at Karolinska Institutet. Informed consent was received from all participants (n = 7) before participation. The clinical characteristics of the healthy male volunteers are presented in Table 1. Glucose, insulin and HbA1c values were within the normal range, with no elevation of serum aminotransferase levels. None of the participants used tobacco products or reported taking any medication. The participants were asked to refrain from strenuous exercise for 48 h before the study and to report to the laboratory after an overnight fast.
Table 1
Clinical characteristics of the study participants
Characteristics
Mean ± SE
Age (year)
50.1 ± 3.8
Weight (kg)
78.6 ± 2.5
Height (cm)
178.6 ± 2.4
BMI (kg/m2)
24.8 ± 0.88
Insulin (pmol/l)
30 ± 7.3
Plasma glucose (mmol/l)
5.2 ± 0.2
HbA1c (%)
4.4 ± 0.1
Total cholesterol (mmol/l)
4.9 ± 0.24
HDL (mmol/l)
1.5 ± 0.1
LDL (mmol/l)
3.4 ± 0.2
Triacylglycerol (mmol/l)
1.0 ± 0.3
ASAT (μkat/l)
0.40 ± 0.04
ALAT (μkat/l)
0.41 ± 0.05
γ-GT (μkat/l)
0.48 ± 0.19
Results are presented as mean ± SEM for n = 7 participants
ALAT, alanine aminotransferase; ASAT, aspartate aminotransferase; γ-GT, gamma-glutamyltransferase
Muscle biopsy procedure
Skeletal muscle (∼1 g) was obtained by means of an open biopsy. Biopsies were taken under local anaesthesia (mepivakain chloride 5 mg/ml) from the vastus lateralis portion of the quadriceps femoris muscle [20]. An incision (∼4 cm) was made 15 cm above the proximal border of patella and the muscle fascia was exposed. Thereafter, four to five muscle fibre bundles were excised and placed in oxygenated Krebs–Henseleit buffer (KHB), which contained 5 mmol/l glucose, 15 mmol/l mannitol and 0.1% (wt/vol.) BSA (RIA Grade; Sigma, St Louis, MO, USA).
Muscle incubation procedure
Smaller skeletal muscle strips were dissected from the biopsy specimen, mounted on Plexiglass clamps (9 mm in width) and incubated in vitro in pregassed (95% O2 and 5% CO2) KHB in a shaking water bath at 35°C for 30 min. The gas phase in the vials was maintained during the incubation procedure. Thereafter, skeletal muscle strips were incubated for 30 min at 35°C in KHB in the absence (basal) or presence of either 5 mmol/l spermine NONOate (Calbiochem, San Diego, CA, USA) or 120 nmol/l insulin (Insulin Actrapid; Novo Nordisk, Bagsværd, Denmark). The concentrations of spermine NONOate and insulin were maintained throughout all remaining incubation procedures.
Glucose transport
Skeletal muscle strips were transferred to fresh KHB containing 20 mmol/l mannitol and incubated at 35°C for 10 min. Thereafter, muscles were incubated for 20 min in KHB containing 5 mmol/l 3-O-methyl [3H]glucose (29.6 MBq/mmol) and 15 mmol/l [14C]mannitol (2.0 MBq/mmol). Thus, muscle strips were exposed to either spermine NONOate or insulin for a total of 60 min. At the end of the incubation protocol, skeletal muscle specimens were blotted of excess fluid, snap-frozen in liquid nitrogen and stored at −80°C until further analysis. Glucose transport was determined by the accumulation of intracellular 3-O-methyl-[3H]glucose as previously described [21].
Cyclic guanosine monophosphate measurement
Skeletal muscle strips were incubated in the absence or presence of spermine NONOate as described above for 60 min and then rapidly clamp frozen in liquid N2 cooled tongs. Frozen muscles were homogenised on ice in 1 ml 10% (vol./vol.) trichloroacetic acid using a polytron. Homogenates were subjected to centrifugation for 10 min at 14,000 g. Thereafter, the trichloroacetic acid was extracted from the supernatant fraction with 2H2O-saturated diethyl ether. Samples were then immediately frozen in liquid N2 and concentrated in a speed vacuum. The concentrated samples were resuspended in assay buffer and acetylated, after which cGMP levels were measured by immunoassay according to manufacturer’s instructions (cGMP BioTRAK, RPN 226; Amersham, Pittsburgh, PA, USA). cGMP is expressed as pmol/g wet weight.
AMP kinase activity
Muscles were homogenised in ice-cold lysis buffer containing 50 mmol/l Tris–HCl (pH 7.5), 1 mmol/l EDTA, 1 mmol/l dithiothreitol, 10% (vol./vol.) glycerol, 50 mmol/l NaF, 5 mmol/l Na pyrophosphate, 1 mmol/l benzamidine, 0.1 mmol/l phenylmethyl sulfonyl fluoride and 1% (vol./vol.) Triton X-100. Muscle homogenates were subjected to centrifugation for 10 min at 14,000 g and 4°C. Supernatant fractions were removed and used to determine protein content using a commercially available kit based on the Bradford method (Bio-Rad, Hercules, CA, USA). Aliquots (200 μg protein) were incubated overnight at 4°C with antibodies against α1- and α2-AMPK subunits (provided by D. G. Hardie, Division of Molecular Physiology, College of Life Sciences, University of Dundee, Dundee, UK). Human muscle lysates from rested and exercised conditions were also incubated overnight at 4°C with antibodies against the AMPK-α1- and α2-subunits, and were used as positive control for the assay. The immunoprecipitates were washed once in lysis buffer, once in 480 mmol/l HEPES (pH 7.0) and 240 mmol/l NaCl, and twice in 240 mmol/l HEPES (pH 7.0) and 120 mmol/l NaCl, leaving 10 µl of buffer with the Sepharose after the last wash. To determine isoform-specific AMPK activity, the immunoprecipitates were incubated for 30 min at 30°C in a total volume of 30 µl containing 833 µmol/l diothiothreitol, 200 µmol/l AMP, 100 µmol/l AMARA-peptide (Upstate Millipore, Billerica, MA, USA), 5 mmol/l MgCl2, 200 µmol/l ATP and 74 kBq of [γ-32P]ATP. The reaction was stopped by spotting 25 µl on to a piece of P81 filter paper, which was then washed four times at 15 min per time in 1% (vol./vol.) phosphoric acid. The dried filter paper was analysed for activity using liquid scintillation counting.
Cell culture
L6 muscle cells (provided by A. Klip, The Hospital for Sick Children, Toronto, ON, Canada) were grown in αMEM supplemented with 10% (vol./vol.) FBS, 1% (vol./vol.) penicillin and streptomycin (100 U/ml penicillin, 100 µg/ml streptomycin), and 1% (vol./vol.) fungizone in 5% CO2 95% O2 humidified air at 37°C. The L6 cells were differentiated into myotubes for 6 days in αMEM supplemented with 2% (vol./vol.) FBS. Myotubes were then serum-starved for 3 h before the start of the experiment. Myotubes were incubated under basal conditions or in the presence or absence of spermine NONOate (20 µmol/l) or a guanylate cyclase inhibitor (10 µmol/l LY-83583) and glucose incorporation into glycogen (glycogen synthesis), glucose uptake and protein signalling were determined. To establish insulin-stimulated conditions, insulin (120 nmol/l) was added during the last 20 min for protein phosphorylation and glucose uptake analysis or during the last 120 min for glycogen synthesis analysis.
Glucose incorporation to glycogen (glycogen synthesis)
Glycogen synthesis was assessed in L6 cells by measuring the incorporation of d-[U-14C]glucose into glycogen as described [22]. Overnight serum-starved myotubes were incubated in 6 well plates and treated for 1 h with spermine NONOate (20 µmol/l) and LY-83583 (10 µmol/l), in the absence or presence of insulin (120 nmol/l). During the last 90 min, the cells were incubated with medium supplemented with glucose (5 mmol/l) and d-[U-14C]glucose (final specific activity, 37 kBq/ml). Each experiment was performed on duplicate wells. Glucose uptake and incorporation to glycogen was determined as mmol glucose per mg protein per h.
Glucose uptake
Myotubes were serum-starved for 3 to 5 h and incubated with or without spermine NONOate (20 µmol/l) in the absence or presence of insulin (120 nmol/l). 2-Deoxyglucose uptake was determined as described [23]. Each experiment was performed on duplicate wells.
Western blot analysis
A portion of the muscle specimen incubated for the glucose transport assay was homogenised in 0.3 ml ice-cold lysis buffer using a glass-on-glass system with a motor pestle. The buffer contained 20 mmol/l Tris (pH 8.0), 137 mmol/l NaCl, 2.7 mmol/l KCl, 10 mmol/l NaF, 1 mmol/l MgCl2, 1 mmol/l Na3VO4, 0.2 mmol/l phenylmethylsulfonyl fluoride, 10% (wt/vol.) glycerol, 1% (wt/vol.) Triton X-100, 1 µg/ml aprotinin, 1 µg/ml leupeptin and 1 μmol/l microcystin. L6 myotubes were collected into similar ice-cold lysis buffer. Homogenates were solubilised by end-over-end mixing for 60 min at 4°C and subjected to centrifugation (for 10 min at 4°C and 12,000 g). The supernatant fraction was stored at −80°C until use. Total protein was determined using a commercially available kit (Pierce, Rockford, IL, USA). Equal amounts of protein (40 µg) were diluted in Laemmli sample buffer. Proteins were separated by SDS–PAGE and transferred to membranes (Immobilon-P; Millipore). Phosphorylation and levels of several proteins were determined using the following antibodies: phospho-AMPK Thr172, phospho-Akt Ser473, phospho-Akt-substrate (PAS), phospho-glycogen synthase kinase (GSK)-α/β (Ser21/9) and phospho-CaMKII Thr286 (all from Cell Signaling Technology, MA, USA), phospho-ACCβ Ser227 and ACC (Upstate Biotechnologies, MA, USA) and GLUT4 (Chemicon, CA, USA). Anti-α1-AMPK and anti α2-AMPK antibodies used for the immunoblot analysis were kindly provided by D. G. Hardie (Division of Molecular Physiology, College of Life Sciences, University of Dundee, UK). All membranes were normalised for loading with glyceraldehyde-3-phosphate dehydrogenase (Santa Cruz Biotechnology, Santa Cruz, CA, USA) or respective total protein content. Proteins were visualised by chemiluminescence and quantified by densitometry.
Statistics
Data are reported as mean ± SE. Differences were determined using a paired t test or ANOVA as appropriate. Bonferroni test was used for post hoc analysis. Significance was established at p < 0.05.

Results

Effects of spermine NONOate on glucose transport and AMP kinase activity
We determined the effects of spermine NONOate and insulin on glucose transport in intact human skeletal muscle (Fig. 1a). Muscle strips were incubated in the absence or presence of either 5 mmol/l spermine NONOate or 120 nmol/l insulin and 3-O-methyl-glucose transport was assessed. Insulin increased glucose transport fivefold (p < 0.05) in isolated skeletal muscle strips, consistent with our earlier studies [24]. Spermine NONOate increased skeletal muscle glucose transport 2.4-fold (p < 0.05), concomitant with increased cGMP levels (p < 0.001) (Fig. 1b). AMPK activation has been implicated in the mechanism by which nitric oxide enhances glucose uptake in skeletal muscle [15]. Thus, we determined the effects of spermine NONOate exposure on α-isoform-specific AMPK signalling (Fig. 1c, d). For perspective, acute exercise increased AMPK-α1- and AMPK-α2-associated activity in skeletal muscle from healthy men by two- and 6.2-fold, respectively vs rested conditions (Fig. 1e, f). Spermine NONOate increased AMPK-α1-associated activity 1.7-fold (p < 0.05), while AMPK-α2-associated activity was unchanged in isolated skeletal muscle. Previous studies from our laboratory provide evidence that insulin does not alter AMPK-α1- or AMPK-α2-associated activity in isolated human skeletal muscle [25].
Effects of spermine NONOate on protein content and signal transduction
The effects of insulin or spermine NONOate on canonical insulin signalling were determined in isolated skeletal muscle using phospho-specific antibodies (Fig. 2a). Exposure of human skeletal muscle to insulin increased the phosphorylation of Akt (Ser473), Tre-2/BUB2/CDC16 domain-containing protein family, member 1, domain family 1/domain family 4 (pTBC1D1/D4) (detected using a PAS antibody) and GSK3α/β (Ser21/9), while spermine NONOate had no effect (Fig. 2a–d). Neither insulin nor spermine NONOate treatment altered CaMK signalling, since phosphorylation of CaMKII (Thr286) was unaltered compared with non-stimulated (basal) muscle (Fig. 2e). Abundance of the AMPK-α1 and AMPK-α2 subunits in isolated skeletal muscle was unaltered after 1 h exposure of isolated skeletal muscle to spermine NONOate or insulin (Fig. 2a). In L6 cells, nitric oxide is known to increase GLUT4 levels via an AMPK-dependent mechanism [16]. GLUT4 content was unaltered after 1 h exposure of isolated skeletal muscle to spermine NONOate or insulin (data not shown), suggesting spermine NONOate acutely increases glucose transport in muscle by recruiting existing glucose transporters to the cell surface, rather than through GLUT4 biogenesis.
Effect of spermine NONOate and insulin on glycogen synthesis and glucose uptake
To determine whether spermine NONOate and insulin have an additive effect on glucose metabolism, we measured glucose incorporation into glycogen (glycogen synthesis) (Fig. 3a) and glucose uptake (Fig. 3b) in L6 myotubes. Spermine NONOate and insulin increased glycogen synthesis 1.2- (p < 0.001) and 2.2-fold (p < 0.0001), respectively (Fig. 3a). Combined exposure to spermine NONOate and insulin had an additive effect on glycogen synthesis. Similarly, spermine NONOate and insulin independently increased glucose uptake 1.2- (p < 0.01) and 1.2-fold (p < 0.001) (Fig. 3b). Combined exposure to spermine NONOate and insulin also had an additive effect on glucose uptake. To delineate underlying mechanism for the additive effect on glucose uptake and metabolism, phosphorylation of AMPK, ACC, Akt and TBC1D1/D4 was measured (Fig. 3c–f). Spermine NONOate alone increased phosphorylation of AMPK Thr172 6.3-fold (p < 0.05) and pACC Ser227 1.7-fold (p < 0.05), while insulin alone had no effect. Interestingly, phosphorylation of AMPK Thr172 and pACC Ser227 was unaltered in the presence of spermine NONOate and insulin. Nevertheless, insulin led to a 6.5-fold increase in Akt Ser473 phosphorylation (p < 0.05) and a 3.5-fold increase in TBC1D1/D4 phosphorylation (p < 0.05), these responses being unaltered in presence of spermine NONOate. Finally, phosphorylation of Akt Ser473 and TBC1D1/D4 was unaltered in the presence of spermine NONOate.
Effect of the guanylate cyclase inhibitor LY-83583 on spermine NONOate-induced glycogen synthesis and AMPK phosphorylation
An earlier report provides evidence that LY-83583 inhibits guanylate cyclase activity [26]. Since spermine NONOate exposure increased AMPK-α1-associated activity in human skeletal muscle (Fig. 1c), as well as phosphorylation of AMPK and ACC in L6 myotubes (Fig. 3c, e), we determined whether AMPK activation lies downstream of cGMP. Treatment of L6 myotubes with spermine NONOate increased glycogen synthesis and AMPK Thr172phosphorylation, but these responses were abolished in the presence of LY-83583 (10 µmol/l) (Fig. 4a, b). However, LY-83583 (10 µmol/l) increased glycogen synthesis in L6 myotubes (p < 0.001). This dose was selected for our in vitro studies on the basis of a previous report [10]. Treatment of L6 myotubes with a lower concentration of LY-83583 (100 nmol/l) also increased glycogen synthesis (data not shown). Importantly, we provide evidence that only the α1 subunit of AMPK is present in L6 myotubes, since immunoblot analysis failed to detect the α2 subunit of AMPK (Fig. 4c).

Discussion

Regular exercise training improves glucose tolerance [27] and skeletal muscle insulin sensitivity [28] in type 2 diabetic patients, but the molecular mechanisms are incompletely resolved. Although several strategies designed to enhance compliance with physical activity regimens in patients with insulin resistance have been proposed [29], many type 2 diabetic patients rely on pharmacological treatments to improve glucose homeostasis. Yet, these current pharmacological treatments to enhance peripheral insulin sensitivity have limited efficacy [30]. Therefore, insight into novel mechanisms capable of enhancing skeletal muscle glucose uptake could lead to new pharmaceutical strategies to improve treatment and possibly prevent peripheral insulin resistance in patients with type 2 diabetes.
Nitric oxide signalling plays a key role in exercise/contraction-induced metabolic responses in skeletal muscle [1], partly via an AMPK-dependent mechanism [15]. During exercise/contraction, increased nitric oxide levels are associated with induction of glucose uptake in skeletal muscle [24]. Conversely, NOS inhibition reduces glucose uptake during exercise in type 2 diabetic patients more than in control participants [31]. Previous reports characterising the direct effects of nitric oxide on glucose uptake are limited to in vitro studies of isolated rodent skeletal muscle [911]. Here we provide evidence that pharmacological treatment of human skeletal muscle with the nitric oxide donor spermine NONOate increases glucose transport, concomitant with increased cGMP levels and AMPK-α1-associated activity. Using L6 myotubes (producing only the α1-subunit of AMPK), we provide evidence that spermine NONOate increases phosphorylation of AMPK and ACC, with a concomitant increase in glucose transport and glycogen synthesis. Our findings in human skeletal muscle are compatible with earlier studies in rodent muscle indicating that nitric oxide donors increase cGMP levels and glucose transport [10]. Similar effects on glucose transport stimulation were also observed in isolated rat skeletal muscle exposed to the cGMP analogues 8-bromo-cGMP [10] and dibutyryl cGMP [11]. The mechanism by which cGMP regulates glucose uptake may involve two enzymes, namely guanylate cyclase and phosphodiesterase, but other mechanisms are also likely to be involved. Inhibition of guanylate cyclase prevents the sodium nitroprusside-induced increase in cGMP levels and glucose transport [10]. Conversely, treatment of isolated skeletal muscle with a phosphodiesterase inhibitor (Zaprinast) increases cGMP levels, with a concomitant increase in glucose uptake [32]. Collectively, these studies provide evidence that the nitric oxide/cGMP pathway is likely to be important in the regulation of glucose transport.
Nitric oxide signalling has been linked to AMPK activation and glucose uptake [15, 16]. Here, we provide evidence that in vitro exposure to a nitric oxide donor increases glucose transport, with concomitant increase in AMPK-α1-specific activity and AMPK-α1 phosphorylation in isolated human skeletal muscle and L6 myotubes. Among the 12 possible AMPK heterotrimers, only three (α1β2γ1, α2β2γ1, α2β2γ3) have been identified in human skeletal muscle [33]. Each individual heterotrimer is activated in a manner dependent on time and intensity [34, 35], which may elicit signalling specificity in response to a distinct set of stimuli. Our results in human skeletal muscle and L6 myotubes provide evidence that nitric oxide specifically increases AMPK-α1-associated activity and AMPK-α1 phosphorylation respectively. Moreover in L6 myotubes, the guanylate cyclase enzyme inhibitor LY-83583 prevents the nitric oxide donor-induced increase in glycogen synthesis and α1-AMPK phosphorylation, providing evidence for a potential role of the AMPK-α1 subunit in the mechanism by which nitric oxide donors increase glucose transport. These observations are consistent with a previous study in rodent skeletal muscle [15], which provided evidence that sodium nitroprusside increases AMPK-α1-, but not AMPK-α2-associated activity. Furthermore, in rodent skeletal muscle, sodium nitroprusside-induced increases in AMPK-isoform-specific activity occurred independently of changes in ATP, creatinine phosphate or glycogen levels [15]. Thus, the nitric oxide–cGMP pathway may be involved in the regulation of AMPK-α1 activity and/or inhibition of the protein phosphatase responsible for AMPK regulation.
Although most Ampk (also known as Prkaa2) knockout models provide evidence against a critical role for the AMPK-α1 subunit in the regulation of skeletal muscle glucose uptake [14], AMPK-α1 activation is required for stimulation of glucose uptake in response to twitch contraction [36]. Based on our results, the promotion of glucose uptake effected by the nitric oxide donor appears to be at least partly mediated via AMPK complexes containing the α1 subunit. However, the role of AMPK in nitric oxide signalling is complex, since AMPK has also been suggested to be an upstream kinase for NOS [1719, 37]. Direct activation of AMPK, using 5′-aminoimidazole-4-carboxamide ribonucleoside (AICAR), stimulates nitric oxide production in human aortic endothelial cells [37] and increases NOS activity in H-2Kb cells [17], implicating a feedback loop between AMPK and NOS [16]. The question of whether nitric oxide pathways lie upstream or downstream of AMPK remains unresolved. For example, AICAR-induced AMPK-activated glucose transport is unaltered by NOS inhibition in isolated rat skeletal muscle [38]. The direct interaction between the nitric oxide/cGMP pathway and AMPK warrant further investigation.
To determine the intracellular mechanism by which the nitric oxide donor spermine NONOate increases glucose transport, components of the canonical insulin signalling cascade were assessed. The Akt- and Rev/Rex activation domain-binding protein (Rab) GTPase-activating proteins, TBC1D1 and TBC1D4, are the most distal signalling proteins implicated in GLUT4 translocation [39, 40]. In human skeletal muscle, insulin exposure led to an expected increase in phosphorylation of Akt, TBC1D1/D4 (detected using a PAS antibody) and GSK3, while exposure to the nitric oxide donor had no effect. These data indicate that nitric oxide-stimulated glucose transport is mediated via an insulin-independent pathway. Nitric oxide has also been linked as a positive [16] modulator of GLUT4 production. However, GLUT4 protein content was unaltered in response to spermine NONOate (data not shown), presumably because of the shorter incubation time and low concentration of the nitric oxide donor used in this study. CaMKII signalling has been implicated in the mechanism by which muscle contraction increases glucose uptake [13]. Nevertheless, CaMKII phosphorylation was unaltered in response to insulin or spermine NONOate. In contrast to our results for AMPK signalling, insulin and CaMKII signalling do not appear to play a role in nitric oxide action on skeletal muscle glucose transport. In L6 myotubes, the nitric oxide donor and insulin had an additive effect on glycogen synthesis and glucose uptake without further increase in insulin-induced Akt and TBC1D1/D4 phosphorylation. These results suggest that nitric oxide promotes glucose uptake by an insulin-independent mechanism. Furthermore, the stimulatory effect of the nitric oxide donor on AMPK and ACC phosphorylation under insulin-stimulated conditions was prevented, excluding a role for AMPK signalling in the additive effect on glycogen synthesis and glucose uptake.
In summary, the nitric oxide donor spermine NONOate increases cGMP levels and promotes glucose transport, concomitantly with AMPK-α1-isoform-specific activation in human skeletal muscle. Further study to delineate mechanisms and the therapeutic window is warranted. Spermine NONOate also increased glucose transport in L6 myotubes, concomitantly with an increase in AMPK-α1-isoform-specific phosphorylation. Moreover, these effects were prevented in presence of a guanylate cyclase inhibitor. Further studies on the mechanisms by which AMPK-α1-isoform-specific signalling is directly linked to nitric oxide action are warranted. Taken together with recent evidence showing that sodium nitroprusside increased glucose uptake in human primary myotubes derived from healthy volunteers and patients with type 2 diabetes [41], our findings have clear clinical implications, since manipulation of the nitric oxide/cGMP signalling cascade could enhance glucose uptake by an insulin-independent mechanism to potentially improve whole-body glucose homeostasis in type 2 diabetic patients.

Acknowledgements

This work was supported by grants from the European Research Council, the Swedish Research Council, the Swedish Diabetes Association, Novo Nordisk Foundation, the Foundation for Scientific Studies of Diabetology, the Swedish Centre for Sports Research, the Strategic Research Foundation, the Commission of the European Communities (Contract numbers LSHM-CT-2004-005272 EXGENESIS and LSHM-CT-2004-512013 EUGENE2). Additional support to H. A. Koistinen was provided by grants from Finnish Academy of Science and Sigrid Juselius Foundation.

Duality of interest

The authors declare that there is no duality of interest associated with this manuscript.

Open Access

This article is distributed under the terms of the Creative Commons Attribution Noncommercial License which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.
Open AccessThis is an open access article distributed under the terms of the Creative Commons Attribution Noncommercial License (https://​creativecommons.​org/​licenses/​by-nc/​2.​0), which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Jetzt bestellen und 100 € sparen!

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Jetzt bestellen und 100 € sparen!

Literatur
1.
Zurück zum Zitat McConell GK, Kingwell BA (2006) Does nitric oxide regulate skeletal muscle glucose uptake during exercise? Exerc Sport Sci Rev 34:36–41CrossRefPubMed McConell GK, Kingwell BA (2006) Does nitric oxide regulate skeletal muscle glucose uptake during exercise? Exerc Sport Sci Rev 34:36–41CrossRefPubMed
2.
Zurück zum Zitat Balon TW, Nadler JL (1994) Nitric oxide release is present from incubated skeletal muscle preparations. J Appl Physiol 77:2519–2521PubMed Balon TW, Nadler JL (1994) Nitric oxide release is present from incubated skeletal muscle preparations. J Appl Physiol 77:2519–2521PubMed
3.
Zurück zum Zitat Roberts CK, Barnard RJ, Scheck SH, Balon TW (1997) Exercise-stimulated glucose transport in skeletal muscle is nitric oxide dependent. Am J Physiol 273:E220–E225PubMed Roberts CK, Barnard RJ, Scheck SH, Balon TW (1997) Exercise-stimulated glucose transport in skeletal muscle is nitric oxide dependent. Am J Physiol 273:E220–E225PubMed
4.
Zurück zum Zitat Bradley SJ, Kingwell BA, McConell GK (1999) Nitric oxide synthase inhibition reduces leg glucose uptake but not blood flow during dynamic exercise in humans. Diabetes 48:1815–1821CrossRefPubMed Bradley SJ, Kingwell BA, McConell GK (1999) Nitric oxide synthase inhibition reduces leg glucose uptake but not blood flow during dynamic exercise in humans. Diabetes 48:1815–1821CrossRefPubMed
5.
Zurück zum Zitat Moncada S, Higgs A (1993) The l-arginine–nitric oxide pathway. N Engl J Med 329:2002–2012CrossRefPubMed Moncada S, Higgs A (1993) The l-arginine–nitric oxide pathway. N Engl J Med 329:2002–2012CrossRefPubMed
6.
Zurück zum Zitat Nakane M, Schmidt HH, Pollock JS, Förstermann U, Murad F (1993) Cloned human brain nitric oxide synthase is highly expressed in skeletal muscle. FEBS Lett 316:175–180CrossRefPubMed Nakane M, Schmidt HH, Pollock JS, Förstermann U, Murad F (1993) Cloned human brain nitric oxide synthase is highly expressed in skeletal muscle. FEBS Lett 316:175–180CrossRefPubMed
7.
Zurück zum Zitat Durham WJ, Yeckel CW, Miller SL, Gore DC, Wolfe RR (2003) Exogenous nitric oxide increases basal leg glucose uptake in humans. Metabolism 52:662–665CrossRefPubMed Durham WJ, Yeckel CW, Miller SL, Gore DC, Wolfe RR (2003) Exogenous nitric oxide increases basal leg glucose uptake in humans. Metabolism 52:662–665CrossRefPubMed
8.
Zurück zum Zitat Henstridge DC, Kingwell BA, Formosa MF et al (2005) Effects of the nitric oxide donor, sodium nitroprusside, on resting leg glucose uptake in patients with type 2 diabetes. Diabetologia 48:2602–2608CrossRefPubMed Henstridge DC, Kingwell BA, Formosa MF et al (2005) Effects of the nitric oxide donor, sodium nitroprusside, on resting leg glucose uptake in patients with type 2 diabetes. Diabetologia 48:2602–2608CrossRefPubMed
9.
Zurück zum Zitat Balon TW, Nadler JL (1997) Evidence that nitric oxide increases glucose transport in skeletal muscle. J Appl Physiol 82:359–363PubMed Balon TW, Nadler JL (1997) Evidence that nitric oxide increases glucose transport in skeletal muscle. J Appl Physiol 82:359–363PubMed
10.
Zurück zum Zitat Young ME, Radda GK, Leighton B (1997) Nitric oxide stimulates glucose transport and metabolism in rat skeletal muscle in vitro. Biochem J 322:223–228PubMed Young ME, Radda GK, Leighton B (1997) Nitric oxide stimulates glucose transport and metabolism in rat skeletal muscle in vitro. Biochem J 322:223–228PubMed
11.
Zurück zum Zitat Etgen GJ Jr, Fryburg DA, Gibbs EM (1997) Nitric oxide stimulates skeletal muscle glucose transport through a calcium/contraction- and phosphatidylinositol-3-kinase-independent pathway. Diabetes 46:1915–1919CrossRefPubMed Etgen GJ Jr, Fryburg DA, Gibbs EM (1997) Nitric oxide stimulates skeletal muscle glucose transport through a calcium/contraction- and phosphatidylinositol-3-kinase-independent pathway. Diabetes 46:1915–1919CrossRefPubMed
12.
Zurück zum Zitat Ohlstein EH, Wood KS, Ignarro LJ (1982) Purification and properties of heme-deficient hepatic soluble guanylate cyclase: effects of heme and other factors on enzyme activation by NO, NO-heme, and protoporphyrin IX. Arch Biochem Biophys 218:187–198CrossRefPubMed Ohlstein EH, Wood KS, Ignarro LJ (1982) Purification and properties of heme-deficient hepatic soluble guanylate cyclase: effects of heme and other factors on enzyme activation by NO, NO-heme, and protoporphyrin IX. Arch Biochem Biophys 218:187–198CrossRefPubMed
13.
Zurück zum Zitat Jessen N, Goodyear LJ (2005) Contraction signaling to glucose transport in skeletal muscle. J Appl Physiol 99:330–337CrossRefPubMed Jessen N, Goodyear LJ (2005) Contraction signaling to glucose transport in skeletal muscle. J Appl Physiol 99:330–337CrossRefPubMed
14.
Zurück zum Zitat Long YC, Zierath JR (2006) AMP-activated protein kinase signaling in metabolic regulation. J Clin Invest 116:1776–1783CrossRefPubMed Long YC, Zierath JR (2006) AMP-activated protein kinase signaling in metabolic regulation. J Clin Invest 116:1776–1783CrossRefPubMed
15.
Zurück zum Zitat Higaki Y, Hirshman MF, Fujii N et al (2001) Nitric oxide increases glucose uptake through a mechanism that is distinct from the insulin and contraction pathways in rat skeletal muscle. Diabetes 50:241–247CrossRefPubMed Higaki Y, Hirshman MF, Fujii N et al (2001) Nitric oxide increases glucose uptake through a mechanism that is distinct from the insulin and contraction pathways in rat skeletal muscle. Diabetes 50:241–247CrossRefPubMed
16.
Zurück zum Zitat Lira VA, Soltow QA, Long JH et al (2007) Nitric oxide increases GLUT4 expression and regulates AMPK signaling in skeletal muscle. Am J Physiol Endocrinol Metab 293:E1062–E1068CrossRefPubMed Lira VA, Soltow QA, Long JH et al (2007) Nitric oxide increases GLUT4 expression and regulates AMPK signaling in skeletal muscle. Am J Physiol Endocrinol Metab 293:E1062–E1068CrossRefPubMed
17.
Zurück zum Zitat Fryer LG, Hajduch E, Rencurel F et al (2000) Activation of glucose transport by AMP-activated protein kinase via stimulation of nitric oxide synthase. Diabetes 49:1978–1785CrossRefPubMed Fryer LG, Hajduch E, Rencurel F et al (2000) Activation of glucose transport by AMP-activated protein kinase via stimulation of nitric oxide synthase. Diabetes 49:1978–1785CrossRefPubMed
18.
Zurück zum Zitat Chen ZP, Mitchelhill KI, Michell BJ et al (1999) AMP-activated protein kinase phosphorylation of endothelial NO synthase. FEBS Lett 443:285–289CrossRefPubMed Chen ZP, Mitchelhill KI, Michell BJ et al (1999) AMP-activated protein kinase phosphorylation of endothelial NO synthase. FEBS Lett 443:285–289CrossRefPubMed
19.
Zurück zum Zitat Chen ZP, McConell GK, Michell BJ et al (2000) AMPK signaling in contracting human skeletal muscle: acetyl-CoA carboxylase and NO synthase phosphorylation. Am J Physiol Endocrinol Metab 279:E1202–E1206PubMed Chen ZP, McConell GK, Michell BJ et al (2000) AMPK signaling in contracting human skeletal muscle: acetyl-CoA carboxylase and NO synthase phosphorylation. Am J Physiol Endocrinol Metab 279:E1202–E1206PubMed
20.
Zurück zum Zitat Zierath JR (1995) In vitro studies of human skeletal muscle: hormonal and metabolic regulation of glucose transport. Acta Physiol Scand Suppl 626:1–96PubMed Zierath JR (1995) In vitro studies of human skeletal muscle: hormonal and metabolic regulation of glucose transport. Acta Physiol Scand Suppl 626:1–96PubMed
21.
Zurück zum Zitat Zierath JR, Galuska D, Engstrom A et al (1992) Human islet amyloid polypeptide at pharmacological levels inhibits insulin and phorbol ester-stimulated glucose transport in in vitro incubated human muscle strips. Diabetologia 35:26–31CrossRefPubMed Zierath JR, Galuska D, Engstrom A et al (1992) Human islet amyloid polypeptide at pharmacological levels inhibits insulin and phorbol ester-stimulated glucose transport in in vitro incubated human muscle strips. Diabetologia 35:26–31CrossRefPubMed
22.
Zurück zum Zitat Al-Khalili L, Chibalin AV, Kannisto K et al (2003) Insulin action in cultured human skeletal muscle cells during differentiation: assessment of cell surface GLUT4 and GLUT1 content. Cell Mol Life Sci 60:991–998PubMed Al-Khalili L, Chibalin AV, Kannisto K et al (2003) Insulin action in cultured human skeletal muscle cells during differentiation: assessment of cell surface GLUT4 and GLUT1 content. Cell Mol Life Sci 60:991–998PubMed
23.
Zurück zum Zitat Niu W, Huang C, Nawaz Z et al (2003) Maturation of the regulation of GLUT4 activity by p38 MAPK during L6 cell myogenesis. J Biol Chem 278:17953–17962CrossRefPubMed Niu W, Huang C, Nawaz Z et al (2003) Maturation of the regulation of GLUT4 activity by p38 MAPK during L6 cell myogenesis. J Biol Chem 278:17953–17962CrossRefPubMed
24.
Zurück zum Zitat Koistinen HA, Galuska D, Chibalin AV et al (2003) 5-amino-imidazole carboxamide riboside increases glucose transport and cell-surface GLUT4 content in skeletal muscle from subjects with type 2 diabetes. Diabetes 52:1066–1072CrossRefPubMed Koistinen HA, Galuska D, Chibalin AV et al (2003) 5-amino-imidazole carboxamide riboside increases glucose transport and cell-surface GLUT4 content in skeletal muscle from subjects with type 2 diabetes. Diabetes 52:1066–1072CrossRefPubMed
25.
Zurück zum Zitat Barnes BR, Ryder JW, Steiler TL et al (2002) Isoform-specific regulation of 5′ AMP-activated protein kinase in skeletal muscle from obese Zucker (fa/fa) rats in response to contraction. Diabetes 51:2703–2708CrossRefPubMed Barnes BR, Ryder JW, Steiler TL et al (2002) Isoform-specific regulation of 5′ AMP-activated protein kinase in skeletal muscle from obese Zucker (fa/fa) rats in response to contraction. Diabetes 51:2703–2708CrossRefPubMed
26.
Zurück zum Zitat Mulsch A, Busse R, Liebau S et al (1988) LY 83583 interferes with the release of endothelium-derived relaxing factor and inhibits soluble guanylate cyclase. J Pharmacol Exp Ther 247(1):283–288PubMed Mulsch A, Busse R, Liebau S et al (1988) LY 83583 interferes with the release of endothelium-derived relaxing factor and inhibits soluble guanylate cyclase. J Pharmacol Exp Ther 247(1):283–288PubMed
27.
Zurück zum Zitat Rogers MA, Yamamoto C, King DS et al (1988) Improvement in glucose tolerance after 1 wk of exercise in patients with mild NIDDM. Diabetes Care 11:613–618CrossRefPubMed Rogers MA, Yamamoto C, King DS et al (1988) Improvement in glucose tolerance after 1 wk of exercise in patients with mild NIDDM. Diabetes Care 11:613–618CrossRefPubMed
28.
Zurück zum Zitat Kirwan JP, Solomon TPJ, Wojta DM, Staten MA, Holloszy JO (2009) Effects of 7 days of exercise training on insulin sensitivity and responsiveness in type 2 diabetes mellitus. Am J Physiol Endocrinol Metab 297:E151–E156CrossRefPubMed Kirwan JP, Solomon TPJ, Wojta DM, Staten MA, Holloszy JO (2009) Effects of 7 days of exercise training on insulin sensitivity and responsiveness in type 2 diabetes mellitus. Am J Physiol Endocrinol Metab 297:E151–E156CrossRefPubMed
29.
Zurück zum Zitat Kirk A, de Feo P (2007) Strategies to enhance compliance to physical activity for patients with insulin resistance. Appl Physiol Nutr Metab 32:549–556CrossRefPubMed Kirk A, de Feo P (2007) Strategies to enhance compliance to physical activity for patients with insulin resistance. Appl Physiol Nutr Metab 32:549–556CrossRefPubMed
30.
Zurück zum Zitat Moller DE (2001) New drug targets for type 2 diabetes and the metabolic syndrome. Nature 414:821–827CrossRefPubMed Moller DE (2001) New drug targets for type 2 diabetes and the metabolic syndrome. Nature 414:821–827CrossRefPubMed
31.
Zurück zum Zitat Kingwell BA, Formosa M, Muhlmann M et al (2002) Nitric oxide synthase inhibition reduces glucose uptake during exercise in individuals with type 2 diabetes more than in control subjects. Diabetes 51:2572–2580CrossRefPubMed Kingwell BA, Formosa M, Muhlmann M et al (2002) Nitric oxide synthase inhibition reduces glucose uptake during exercise in individuals with type 2 diabetes more than in control subjects. Diabetes 51:2572–2580CrossRefPubMed
32.
Zurück zum Zitat Young ME, Leighton B (1998) Evidence for altered sensitivity of the nitric oxide/cGMP signalling cascade in insulin-resistant skeletal muscle. Biochem J 329:73–79PubMed Young ME, Leighton B (1998) Evidence for altered sensitivity of the nitric oxide/cGMP signalling cascade in insulin-resistant skeletal muscle. Biochem J 329:73–79PubMed
33.
Zurück zum Zitat Wojtaszewski JF, Birk JB, Frosig C et al (2005) 5′AMP activated protein kinase expression in human skeletal muscle: effects of strength training and type 2 diabetes. J Physiol 564:563–573CrossRefPubMed Wojtaszewski JF, Birk JB, Frosig C et al (2005) 5′AMP activated protein kinase expression in human skeletal muscle: effects of strength training and type 2 diabetes. J Physiol 564:563–573CrossRefPubMed
34.
Zurück zum Zitat Wojtaszewski JF, Nielsen P, Hansen BF et al (2000) Isoform-specific and exercise intensity-dependent activation of 5′-AMP-activated protein kinase in human skeletal muscle. J Physiol 528:221–226CrossRefPubMed Wojtaszewski JF, Nielsen P, Hansen BF et al (2000) Isoform-specific and exercise intensity-dependent activation of 5′-AMP-activated protein kinase in human skeletal muscle. J Physiol 528:221–226CrossRefPubMed
35.
Zurück zum Zitat Treebak JT, Birk JB, Rose AJ et al (2007) AS160 phosphorylation is associated with activation of alpha2beta2gamma1- but not alpha2beta2gamma3-AMPK trimeric complex in skeletal muscle during exercise in humans. Am J Physiol Endocrinol Metab 292:E715–E722CrossRefPubMed Treebak JT, Birk JB, Rose AJ et al (2007) AS160 phosphorylation is associated with activation of alpha2beta2gamma1- but not alpha2beta2gamma3-AMPK trimeric complex in skeletal muscle during exercise in humans. Am J Physiol Endocrinol Metab 292:E715–E722CrossRefPubMed
36.
Zurück zum Zitat Jensen TE, Schjerling P, Viollet B et al (2008) AMPK alpha1 activation is required for stimulation of glucose uptake by twitch contraction, but not by H2O2, in mouse skeletal muscle. PLoS ONE 3:e2102CrossRefPubMed Jensen TE, Schjerling P, Viollet B et al (2008) AMPK alpha1 activation is required for stimulation of glucose uptake by twitch contraction, but not by H2O2, in mouse skeletal muscle. PLoS ONE 3:e2102CrossRefPubMed
37.
Zurück zum Zitat Morrow VA, Foufelle F, Connell JM et al (2003) Direct activation of AMP-activated protein kinase stimulates nitric-oxide synthesis in human aortic endothelial cells. J Biol Chem 278:31629–31639CrossRefPubMed Morrow VA, Foufelle F, Connell JM et al (2003) Direct activation of AMP-activated protein kinase stimulates nitric-oxide synthesis in human aortic endothelial cells. J Biol Chem 278:31629–31639CrossRefPubMed
38.
Zurück zum Zitat Stephens TJ, Canny BJ, Snow RJ et al (2004) 5′-Aminoimidazole-4-carboxyamide-ribonucleoside-activated glucose transport is not prevented by nitric oxide synthase inhibition in rat isolated skeletal muscle. Clin Exp Pharmacol Physiol 31:419–423CrossRefPubMed Stephens TJ, Canny BJ, Snow RJ et al (2004) 5′-Aminoimidazole-4-carboxyamide-ribonucleoside-activated glucose transport is not prevented by nitric oxide synthase inhibition in rat isolated skeletal muscle. Clin Exp Pharmacol Physiol 31:419–423CrossRefPubMed
39.
Zurück zum Zitat Roach WG, Chavez JA, Miinea CP et al (2007) Substrate specificity and effect on GLUT4 translocation of the Rab GTPase-activating protein Tbc1d1. Biochem J 403:353–358CrossRefPubMed Roach WG, Chavez JA, Miinea CP et al (2007) Substrate specificity and effect on GLUT4 translocation of the Rab GTPase-activating protein Tbc1d1. Biochem J 403:353–358CrossRefPubMed
40.
Zurück zum Zitat Sano H, Kane S, Sano E et al (2003) Insulin-stimulated phosphorylation of a Rab GTPase-activating protein regulates GLUT4 translocation. J Biol Chem 278:14599–14602CrossRefPubMed Sano H, Kane S, Sano E et al (2003) Insulin-stimulated phosphorylation of a Rab GTPase-activating protein regulates GLUT4 translocation. J Biol Chem 278:14599–14602CrossRefPubMed
41.
Zurück zum Zitat Henstridge DC, Drew BG, Formosa MF et al (2009) The effect of the nitric oxide donor sodium nitroprusside on glucose uptake in human primary skeletal muscle cells. Nitric Oxide 21:126–131CrossRefPubMed Henstridge DC, Drew BG, Formosa MF et al (2009) The effect of the nitric oxide donor sodium nitroprusside on glucose uptake in human primary skeletal muscle cells. Nitric Oxide 21:126–131CrossRefPubMed
Metadaten
Titel
Nitric oxide increases cyclic GMP levels, AMP-activated protein kinase (AMPK)α1-specific activity and glucose transport in human skeletal muscle
verfasst von
A. S. Deshmukh
Y. C. Long
T. de Castro Barbosa
H. K. R. Karlsson
S. Glund
W. J. Zavadoski
E. M. Gibbs
H. A. Koistinen
H. Wallberg-Henriksson
J. R. Zierath
Publikationsdatum
01.06.2010
Verlag
Springer-Verlag
Erschienen in
Diabetologia / Ausgabe 6/2010
Print ISSN: 0012-186X
Elektronische ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-010-1716-x

Weitere Artikel der Ausgabe 6/2010

Diabetologia 6/2010 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

NSCLC: Progressionsfreies Überleben unter Osimertinib fast versiebenfacht

06.06.2024 ASCO 2024 Kongressbericht

Erste Ergebnisse der Phase-III-Studie LAURA etablieren Osimertinib als neuen Therapiestandard für Menschen mit nicht-resezierbarem, EGFR-mutiertem, nicht-kleinzelligem Lungenkarzinom im Stadium III, die nach definitiver Radiochemotherapie progressionsfrei sind. Auf der ASCO-Tagung wurden diese beeindruckenden Ergebnisse besprochen.

Hodgkin Lymphom: BrECADD-Regime übertrifft die Erwartungen

05.06.2024 ASCO 2024 Kongressbericht

Das Kombinationsregime BrECADD mit Brentuximab vedotin ermöglichte in der Studie HD21 beim fortgeschrittenen klassischen Hodgkin-Lymphom eine unerwartet hohe progressionsfreie Überlebensrate von 94,3% nach vier Jahren. Gleichzeitig war das Regime besser tolerabel als der bisherige Standard eBEACOPP.

Antikörper-Drug-Konjugat verdoppelt PFS bei Multiplem Myelom

05.06.2024 ASCO 2024 Nachrichten

Zwei Phase-3-Studien deuten auf erhebliche Vorteile des Antikörper-Wirkstoff-Konjugats Belantamab-Mafodotin bei vorbehandelten Personen mit Multiplem Myelom: Im Vergleich mit einer Standard-Tripeltherapie wurde das progressionsfreie Überleben teilweise mehr als verdoppelt.

Neuer TKI gegen CML: Höhere Wirksamkeit, seltener Nebenwirkungen

05.06.2024 Chronische myeloische Leukämie Nachrichten

Der Tyrosinkinasehemmer (TKI) Asciminib ist älteren Vertretern dieser Gruppe bei CML offenbar überlegen: Personen mit frisch diagnostizierter CML entwickelten damit in einer Phase-3-Studie häufiger eine gute molekulare Response, aber seltener ernste Nebenwirkungen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.