The purpose of this study was to examine the effects of AMP kinase (AMPK) activation on in vivo glucose and long-chain fatty acid (LCFA) uptake in skeletal muscle and to examine the nitric oxide (NO) dependence of any putative effects. Catheters were chronically implanted in the carotid artery and jugular vein of male Sprague-Dawley rats. After 4 days of recovery, rats were given either water or water containing 1 mg/ml nitro-l-arginine methylester (l-NAME) for 2.5 days. After an overnight fast, rats underwent one of five protocols: saline, 5-aminoimidazole-4-carboxamide-1-B-d-ribofuranoside (AICAR) (10 mg · kg−1 · min−1), l-NAME, AICAR + l-NAME, or AICAR + Intralipid (20%, 0.02 ml · kg−1 · min−1). Glucose was clamped at ∼6.5 mmol/l in all groups, and an intravenous bolus of 2-deoxy[3H]glucose and [125I]-15-(p-iodophenyl)-3-R,S-methylpentadecanoic acid was administered to obtain indexes of glucose (Kg) and LCFA (Kf) uptake and clearance. At 150 min, soleus, gastrocnemius, and superficial vastus lateralis were excised for tracer determination. Both Kg and Kf increased with AICAR in all muscles studied. Kg decreased with increasing muscle composition of type 1 slow-twitch fibers, whereas Kf increased. In addition, AICAR-induced increases in Kg but not Kf were abolished by l-NAME in the majority of muscles examined. This shows that the mechanisms by which AMPK stimulates glucose and LCFA uptake are distinct.

Type 2 diabetes and obesity are metabolic disease states characterized by impairments in glucose and fatty acid metabolism (1,2). As such, the discovery and understanding of signaling pathways involved in the uptake, oxidation, and storage of these substrates is of importance. A protein key to metabolic regulation is AMP kinase (AMPK), which functions as an intracellular fuel gauge responding to alterations in the energy status of the cell. Activation of AMPK results in glucose uptake, fatty acid oxidation, and a decline of fatty acid storage in skeletal muscle (35). Furthermore, the oral hypoglycemic agents rosiglitazone and metformin as well as hormones integral to the regulation of body adiposity, including leptin and adiponectin, are known to act in part through AMPK-mediated pathways (69).

Despite our increased knowledge of AMPK and its role in tissue-specific and whole-body metabolic regulation, the mechanism by which it exerts its actions are unclear. Recent studies indicate that its effects are in part mediated through nitric oxide (NO) or an NO-mediated pathway (10,11). NO is produced and released from skeletal muscle (12) and is necessary for both the full insulin- and exercise-stimulated skeletal muscle glucose uptake responses (1315). Elevation of NO through the NO donor sodium nitroprusside (SNP) results in increased glucose transport and plasma membrane content of GLUT-4 (13,16). Correspondingly, blockade of NO by administration of NG-nitro-l-arginine methyl ester (l-NAME) or NG-monomethyl-l-arginine (l-NMMA) during hyperinsulinemic-euglycemic clamps results in decreased blood flow and glucose disposal in vivo (14,17). To determine the dependence of AMPK on NO, a number of studies have activated AMPK in the presence or absence of NO inhibitors. In cell culture, the combination of l-NMMA and the AMPK activator 5-aminoimidazole-4-carboxamide-1-B-d-ribofuranoside (AICAR) results in the abolishment of AICAR-stimulated glucose uptake, suggesting that AMPK increases glucose uptake by an NO-dependent pathway (11). Additional evidence is found in a study by Morrow et al. (10), where activation of AMPK by AICAR in human aortic endothelial cells resulted in an increase in NO synthase (NOS) phosphorylation and NO production. When a mutant AMPK was transfected into the endothelial cells via adenovirus, AMPK-activated NO production was diminished, showing a clear mechanistic link between AMPK and NOS. Taken together, these in vitro studies provide strong evidence that AMPK acts via an NO pathway; however, in vivo data are lacking.

It is clear from the above discussion that AMPK activation stimulates muscle glucose uptake and intracellular pathways for fatty acid oxidation (3,5,18). However, it is unknown whether the increased fatty acid oxidation translates into an increased fatty acid uptake from the blood in vivo and whether such an effect is, like glucose uptake, reliant on the formation of NO. The purpose of the present study was to examine the effects of AMPK stimulation on skeletal muscle glucose and long-chain fatty acid (LCFA) uptake and clearance and to determine the dependence of any effects on NO. Skeletal muscle fiber type is a major determinant of the metabolic response to a number of stimuli. Therefore, an additional objective was to examine the magnitude and mechanism for AICAR-induced skeletal muscle glucose and LCFA uptake in muscle comprised of different fiber types. Experiments were performed in chronically catheterized conscious rats, where isotopic analogs of glucose and LCFA were administered to quantify tissue-specific substrate influx. It was hypothesized that if AMPK’s effects on NO occurred by a common pathway, then proportional reductions in tissue-specific glucose and fatty acid uptake would occur.

Male Sprague-Dawley rats (Harlan Industries, Indianapolis, IN) were housed individually and maintained at 23°C on a 0600–1800 light cycle. Rats were fed chow (Purina Nestlé, St. Louis, MO) ad libitum and given free access to water. The rats were housed under these conditions for ∼1 week, by which time their weights had reached ∼300 g. After weight gain, rats were randomly divided into each of the experimental groups (n = 7 per group). All procedures were approved by the Vanderbilt University Animal Care and Use Subcommittee and followed the National Institutes of Health guidelines for the care and use of laboratory animals.

Surgical procedures.

Surgical procedures were performed as previously described for arterial and venous catheterizations (19). Briefly, animals were anesthetized with a 50:5:1 vol/vol/vol mixture of ketamine, rompun, and acepromazine, and the left common carotid artery and right jugular vein were catheterized with PE50. Catheters were exteriorized and secured at the back of the neck, filled with heparinized saline (150 units/ml), and sealed with a stainless steel plug. Immediately postsurgery, each animal received 75 mg/kg of ampicilliin s.c. to prevent infection. After surgery, animal weights and food intake were monitored daily, and only animals in which presurgery weight was restored were used for experiments.

l-NAME administration.

After 4 days of recovery, rats were given either drinking water (n = 14) or water containing 1 mg/ml l-NAME for 2.5 days. Assessment of water consumption showed rats consumed 35–40 ml of water or ∼120–160 mg l-NAME · kg−1 · day−1. Previous studies have shown that this dose elicits a near maximal but reversible inhibition of NOS (2022). This method of administration was chosen because l-NAME administration by acute intravenous injection results in increases in body temperature (23).

Isotopic analogs.

Glucose and LCFA tracers used in the present study were 2-deoxy[3H]glucose ([2-3H]DG) and [125I]-15-(p-iodophenyl)-3-R,S-methylpentadecanoic acid ([125I]-BMIPP). [2-3H]DG was purchased from New England Nuclear (Boston, MA). BMIPP was a kind gift from Russ Knapp, Oak Ridge International Laboratories (Oak Ridge, TN). Radioiodination was performed according to the manufacturer’s suggested protocol. Briefly, BMIPP was heated in the presence of Na125I solution (740 MBq/200 μl), propionic acid, and copper (II) sulfate. Na2S2O3 was then added and the organic phase ether extracted and sequentially back extracted with saturated NaHCO3 and water. After evaporation, the [125I]BMIPP was solubilized using sonication into ursodeoxycholic acid with a final activity of 775 μCi/ml.

Experimental procedures.

Before the study, rats were fasted overnight for 10 h. On the day of the experiment, catheters were flushed with heparinized saline (10 units/ml) and connected to PE50 and silastic tubing for infusions and sampling. Throughout the experimental protocol rats were conscious and unrestrained. The experimental protocol consisted of continuous infusions of saline, AICAR (10 mg · kg−1 · min−1), or AICAR + Intralipid 20% (Intralipid; 0.02 ml · kg−1 · min−1, containing 100 units/ml heparin; Pharmacia & Upjohn, Uppsala, Sweden). All infusions lasted for 150 min. In those rats receiving AICAR, glucose (D50) was clamped to levels found in saline and l-NAME groups (∼6.5 mmol/l). Since the administration of AICAR resulted in a decline in nonesterified fatty acid (NEFA), Intralipid was infused in one protocol to maintain plasma NEFA levels and to ensure that the observed differences in LCFA uptake (Rf) were not concentration dependent. In total this resulted in five experimental groups: saline (n = 7), AICAR (n = 7), l-NAME (n = 7), AICAR + l-NAME (n = 7), and AICAR + Intralipid (n = 7). All infusions were equilibrated for 90 min in which small arterial blood samples (20 μl) were obtained every 10 min throughout the experiment for the measurement of plasma glucose. This provided feedback that was used to adjust glucose infusion rates needed to maintain glycemia. After the equilibration period, rats received an intravenous bolus of [2-3H]DG and [125I]-BMIPP for the measurement of glucose and LCFA uptakes, respectively (t = 95 min). The time period from 90 to 120 min is referred to as the “experimental period” in the results section. At time 0 and 90 min, large arterial blood samples (150 μl) were withdrawn for the measurement of insulin, glucose, and NEFA. In addition, small samples (50 μl) were obtained at 97, 100, 110, 120, 130, 140, and 150 min for the measurement of [2-3H]DG, [125I]-BMIPP, NEFA, and insulin. To prevent declines in hematocrit, the erythrocytes taken before the isotopic analog infusion were washed in saline and reinfused shortly after each sample was taken. At t = 150 min, rats were anesthetized with pentobarbital sodium, and their soleus (89% type I, 11% type IIa, 0% type IIb) (24), gastrocnemius (6% type I, 23% type IIa, 71% type IIb) (24), and superficial vastus lateralis (SVL) (0% type I, 1% type IIa, 99% type IIb) (24) were excised and rapidly freeze-clamped in liquid nitrogen.

Plasma analyses

Metabolites.

Plasma glucose concentrations were measured by the glucose oxidase method using an automated glucose analyzer (Beckman Instruments, Fullerton, CA), and immunoreactive insulin was measured in samples obtained at t = 0, 90, 120, and 150 min using a double-antibody method (25). NEFA concentrations were determined at t = 0, 90, 110, 130, and 150 min spectrophotometrically using a kit obtained from Wako Chemicals (NEFA-C, Richmond, VA) in the saline, AICAR, l-NAME, and AICAR + l-NAME groups. In the AICAR + Intralipid group, administration of heparin would have resulted in increased in vitro lipolysis due to the presence of lipoprotein lipase. As such, samples used for NEFA analysis in this group were treated with 5 mol/l NaCl as previously described (26,27) before analysis.

Isotopic analogs.

[125I]BMIPP and [2-3H]DG were measured in the same plasma samples (20 μl) as previously described (28). Briefly, 20 μl plasma was counted for [125I]BMIPP using a Beckman Gamma 5500 counter (Beckman, Fullerton, CA). After this, the plasma sample was deproteinized in 100 μl of Ba(OH)2 and 100 μl of ZnSO4 and subsequently centrifuged. Supernatant (100 μl) was then diluted in 900 μl of H2O. 3H radioactivity was counted after addition of fluor (10 ml; Ultimate Gold, Packard Bioscience, Boston, MA) using a Packard Tri-Carb 2900TR Liquid Scintillation Analyzer (PerkinElmer, Boston, MA). The relationship between γ-radioactivity and β-emissions has been established in our laboratory for that specific counter. This relationship was used to correct 3H radioactivity for β-emissions originating from [125I] radioactivity (28).

Tissue analyses

Isotopic analogs.

Tissues were analyzed for accumulation of free (2-[3H]DG) and phosphorylated deoxy-[2-3H]glucose (2-[3H]DGP) as previously described (29). Briefly, muscle [125I]BMIPP and [2-3H]DG were determined on 50 mg of soleus and 100 mg of gastrocnemius and SVL. After the determination of tissue 125I radioactivity, tissue was homogenized in 2 ml of 0.5% perchloric acid and centrifuged for 20 min. Supernatants (1.5 ml) were then neutralized using 5 mmol/l KOH, and 250 μl was counted after the addition of fluor. This fraction provided total radioactivity [2-3H]DG (free + phosphorylated ([2-3H]DGP). In addition, 500 μl of supernatant was treated with 250 μl of Ba(OH)2 and 250 μl of ZnSO4 and centrifuged. After this, 500 μl of supernatant was diluted to 1 ml in ddH2O before fluor (10 ml) was added and samples were counted. Treatment of Ba(OH)2 and ZnSO4 removed all but free [2-3H]DG (30). Therefore, [2-3H]DGP is calculated as the difference in radioactivity without (total) and with (free [2-3H]DG) Ba(OH)2 and ZnSO4. This analytical approach allows the separation of [2-3H]DGP from the fraction of [2-3H]DG that is incorporated into glycogen, inclusion of which results in the underestimation of Rg (31,32). Sample 3H radioactivity was corrected for β-emissions originating from 125I, as described for plasma samples.

Immunoblotting.

To determine the effects of AICAR on the phosphorylation of AMPK, acetyl-CoA carboxylase (ACC), and NOS, gastrocnemius muscles were homogenized in a solution containing 10% glycerol, 20 mmol/l Na-pyrophosphate, 150 mmol/l NaCl, 50 mmol/l Hepes (pH 7.5), 1% NP-40, 20 mmol/l β-glycerophosphate, 10 mmol/l NaF, 2 mmol/l EDTA (pH 8.0), 2 mmol/l phenylmethylsulfonyl fluoride, 1 mmol/l CaCl2, 1 mmol/l MgCl2, 10 μg/ml aprotinin, 10 μg/ml leupeptin, 2 mmol/l Na2VO3, and 3 mmol/l benzamide. After centrifugation (1 h at 4,500g) the pellets were discarded while supernatants retained for protein determination using a Pierce BCA protein assay kit (Rockford, IL). Proteins (30 μg) were separated on a SDS-PAGE gel and then transferred to a polyvinylidene fluoride membrane. Membranes were blotted with rabbit phospho-AMPK α (Thr172), phosphor-acetyl CoA carboxylase (Ser79), or phosho-endothelial NOS (Ser1177) (1:1,000) (Cell Signaling, Beverly, MA) and then incubated with α-rabbit-horseradish peroxidase (Pierce, Rockford, Il). Densitometry was performed using Lab Image software (Kaplan).

Calculations.

Indexes of glucose and LCFA uptake and clearance: tissue glucose clearance (Kg) and metabolic (Rg) indexes were calculated from the accumulation of [2-3H]DGP in muscle and the integral of the plasma [2-3H]DG concentration after a [2-3H]DG bolus (33,34). The relationships are defined as:

where the subscript “p” refers to mean arterial plasma from t = 90 to 120 min. The measurement of Rg has been described earlier (33,34). In an analogous manner, LCFA clearance (Kf) and metabolic (Rf) indexes were calculated after a [125I]BMIPP bolus from the accumulation of [125I]BMIPP in muscle and the integral of the plasma [125I]BMIPP concentration after the bolus.

[[125I]BMIPP]m is the [125I]BMIPP in the cell, [[125I]BMIPP]p is the [125I]BMIPP in plasma. The measurement of Rf and Kf has been described earlier (28,35)

Statistical analyses.

To compare differences, ANOVA was performed. To establish differences within the ANOVA, a Student’s Newman Keuls post hoc test was used. Significance levels of P ≤ 0.05 were used, and data are reported as means ± SEM.

Animal characteristics.

Preexperimental values for animal weight, arterial plasma glucose, insulin, and NEFA concentrations are reported in Table 1. Rats in the AICAR + l-NAME groups were lighter compared with the AICAR group with mean values of 281 ± 7 vs. 317 ± 10 g. This was not due to a treatment effect but rather lower presurgery weights. No differences were apparent between groups for plasma glucose, insulin, and NEFA. Hematocrit values were not different between groups before (44 ± 0.4%, t = 0) or after (42 ± 0.7%, t = 150 min) the experimental protocol.

Glucose, glucose infusion rates, insulin, and NEFA.

Mean arterial plasma values during the experimental period (t = 90–120 min) are reported in Table 2. No differences in plasma glucose were apparent between groups with time or treatment. In addition, no differences were seen for glucose infusion rates between AICAR, AICAR + l-NAME, and AICAR + Intralipid. Plasma insulin levels were not significantly different among the saline, AICAR, l-NAME, and AICAR + l-NAME groups during the experimental period. In AICAR + Intralipid, plasma insulin levels were higher compared with all other groups. This was not unexpected, as previous studies have shown Intralipid infusion to increase plasma insulin levels due to a decrease in insulin clearance (36). Plasma NEFAs in the AICAR and AICAR + l-NAME groups were significantly lower at all time points in the experimental period compared with baseline values (t = 0). In contrast, NEFAs were stable, and no differences were apparent with time or treatment for saline and l-NAME. In AICAR + Intralipid, baseline NEFAs (t = 0) were lower compared with 150 min; however, no differences were present among saline, l-NAME, and AICAR + Intralipid treatments. NEFA values were lower in AICAR and AICAR + l-NAME compared with saline, l-NAME, and AICAR + Intralipid in the experimental period at all time points measured.

Tissue-specific glucose kinetics.

Results of Kg and Rg are depicted in Figs. 1 and 2. Results for Rg were generally similar to those for Kg in that there were no differences in arterial plasma glucose concentrations between groups. AICAR treatment alone increased Kg and Rg in all muscles. l-NAME treatment decreased Kg in all muscles, while Rg decreased in soleus and gastrocnemius with reductions of ∼50 and ∼54%, respectively. The combination of AICAR + l-NAME resulted in no change in Rg or Kg compared with saline in all muscles. However, Rg values were significantly higher compared with l-NAME. In AICAR + Intralipid, Rg and Kg values were not different from AICAR in soleus and SVL. However, AICAR + Intralipid resulted in a greater Rg and Kg versus AICAR alone in the gastrocnemius.

Tissue-specific LCFA kinetics.

Results of Kf and Rf are depicted in Figs. 3 and 4. AICAR treatment increased Kf in all muscles. Rf was decreased in AICAR due to the decline in NEFA levels. When NEFA levels were maintained in the AICAR + Intralipid group, Kf remained elevated and Rf was increased. This indicates that the effects of AICAR on Kf are not dependent on concentration. Unlike its effect on Kg, l-NAME treatment did not lower Kf compared with saline in any of the muscles studied. The combination of AICAR + l-NAME resulted in Kf values that were greater than those for saline and l-NAME in all muscles. Although there was no statistical difference between AICAR and AICAR + l-NAME, values were slightly lower in both the soleus and gastrocnemius. In contrast, the combination of AICAR + l-NAME resulted in a Kf intermediate between AICAR and l-NAME, showing that AICAR-induced LCFA uptake in the gastrocnemius was not as dependent on NO activation as the soleus and SVL. AICAR + Intralipid had a significantly greater Kf and Rf than AICAR in gastrocnemius, but not in soleus and SVL.

Immunoblotting.

The effects of AICAR on AMPK phosphorylation (AMPK Thr179), ACC phosphorylation (ACC Ser79), and endothelial NOS phosphorylation (NOS Ser1177) in gastrocnemius muscle are shown in Fig. 5. AMPK Thr172 and ACC Ser79 phosphorylation was increased in all groups treated with AICAR, demonstrating that the dosage (10 mg · kg−1 · min−1) activated AMPK. NOS Ser1177 phosphorylation was increased in AICAR but not AICAR + NAME, l-NAME, or AICAR + Intralipid compared with saline.

Muscle specificity.

As previously mentioned, AICAR resulted in increases in Kg and Kf in all muscles compared with saline (Figs. 2 and 4). When percentage increases in AICAR-induced Kg and Kf were compared between muscle fiber types, results showed AICAR-induced glucose uptake to be greatest in fast-twitch muscles, while AICAR-induced LCFA uptake was greater in muscles comprised of slow-twitch muscle fibers.

The primary objectives of the present study were to determine the fiber type-specific effects of AMPK activation on muscle LCFA and glucose uptake in vivo and the NO dependence of the observed increases. Using isotopic analogs to measure indexes of glucose and LCFA uptake in the presence or absence of the NO synthase inhibitor l-NAME and the AMPK activator AICAR, we found that AMPK activation increased glucose and LCFA clearance. Only muscle glucose uptake was inhibited by l-NAME. This demonstrates that the mechanism by which AMPK increases glucose uptake is NO dependent, while AMPK-induced LCFA uptake occurs independent of this pathway. In addition, we demonstrate that in vivo, the magnitude of the AICAR activation of both glucose and LCFA uptake is fiber-type specific. Soleus that has the greatest percentage of slow-twitch fibers exhibits the greatest increase in LCFA uptake and the smallest increase in glucose uptake.

The finding that l-NAME impedes AMPK-induced glucose uptake is consistent with the hypothesis that NO may act as signal that couples tissue energy demands to glucose delivery. This has been clearly demonstrated for both insulin- and contraction-stimulated glucose uptake. Studies using hyperinsulinemic-euglycemic clamps have shown l-NAME to reduce whole-body glucose disposal by ∼15–30% (14,17). Similarly, moderate exercise in humans in the presence of l-NMMA reduces skeletal muscle glucose uptake by ∼30% in normal subjects and ∼75% in subjects with type 2 diabetes (15). Taken together, these data imply that a percentage of both insulin- and exercise-stimulated glucose uptake can be attributed to NO. Since l-NAME reduced basal and AICAR-stimulated glucose uptake with no effect on fatty acid uptake, the mechanism by which NO regulates glucose uptake must be to some extent distinct from that of LCFA. Indeed, there is in vitro evidence that NO directly regulates glucose transport. Isolated muscle preparations treated with SNP have elevated glucose transport and GLUT-4 translocation via a phosphatidylinositol 3-kinase-independent mechanism (13,37,38). This is thought to occur by an NO-activated guanosine 3′-5′-cyclic monophosphate (cGMP) pathway, although the exact signaling pathway of cGMP-induced glucose trafficking remains to be established (39). Alternative NO targets that could potentially influence glucose uptake include creatine kinase, cytochrome-c oxidase, sacroplasmic reticulum Ca2+ ATPase, and glyceraldehyde-3-phosphate (39).

To assess the activation of AMPK and its effects on NOS, immunoblotting for AMK Thr172, ACC Ser79, and NOS Ser1177 was performed on gastrocnemius muscle. Results demonstrated that the AICAR dose (10 mg · kg−1 · min−1) was sufficient to activate both AMK Thr172 and ACC Ser79 activity. However, results of NOS Ser1177 are not as clearly defined. Corroborating previous findings, we show that AMPK stimulation increases NOS Ser1177 compared with saline (40,41). However, no increase in NOS Ser1,177 was observed in either the AICAR + l-NAME or the AICAR + Intralipid groups. This finding reflects the complex nature of NOS regulation. Numerous kinases, including protein kinase G, protein kinase A, protein kinase B (Akt), and/or mitogen-activated protein kinases are known to act at this site (40,4246). In addition, AMPK can also phosphorylate an alternate inhibitory site on the NOS protein (Thr495) (40). Therefore, a lack of effect of AICAR on NOS Ser1177 in these groups may reflect phosphorylation of ACC Thr495 or alterations in the ability of AMPK to phosphorylate this site in the presence of l-NAME or Intralipid.

In the present study, lack of effect of l-NAME on basal and AMPK-stimulated LCFA clearance indicates that the coupling of LCFA delivery to uptake is not dependent on NO and must, therefore, be mediated by an alternative signaling intermediate. The finding that AICAR-induced increases in Kf were sustained, even in the presence of l-NAME, was unexpected considering that NO-dependent vascular perfusion was likely to be considerably reduced. To ensure that the increase in Kf was not caused by AICAR-induced declines in NEFA, AICAR was infused with Intralipid to maintain NEFA levels. Results demonstrated that the AICAR-induced increase in Kf was not dependent on NEFA concentration (Fig. 3). Given this, it is reasonable to speculate that LCFA uptake seen with AICAR was a result of LCFA protein-mediated transport. Evidence for the direct recruitment of LCFA transporters by AMPK has been demonstrated by Luiken et al. (47) who have shown that AICAR stimulation results in translocation of the CD36/FAT in cardiac myocytes. Another possibility is that fatty acids are “pulled”into the cell by AMPK-stimulated increases in LCFA oxidation. Stimulation of skeletal muscle with AICAR concurrently phosphorylates and inhibits ACC, the rate-limiting enzyme in malonyl-CoA synthesis, and also phosphorylates and activates the enzyme that deactivates malonyl-CoA, malonyl-CoA decarboxylase (48,49). Together, these targets effectively reduce malonyl-CoA levels, an allosteric inhibitor of carnitine palmitoyltransferase-1 transporter in the mitochondrial membranes allowing increased fat oxidation, which in itself may stimulate translocation of fatty acid transporters.

In addition to determining the NO dependence of AMPK’s actions, this study demonstrates that both AMPK-induced glucose and LCFA uptake are dependent on muscle fiber-type composition. Our results demonstrate that muscles comprised of a greater percentage of fast-twitch fibers are more responsive to AMPK-stimulated glucose uptake than muscles comprised of more slow-twitch fibers. This finding is consistent with previous in vitro studies showing AMPK protein content and AMPKα2 activity to be greater in fast-twitch versus slow-twitch muscle fibers (50,51). As previously mentioned, AICAR treatment also increased LCFA clearance in a muscle-specific manner, although the fiber-type dependency was opposite that seen for glucose. The effect of AICAR on LCFA uptake was actually greater in muscle with lower AMPK activity. LCFA clearance was greatest in muscle comprised of slow-twitch fibers compared with fast-twitch fibers, a finding that is likely explained by increased LCFA transporter number and LCFA oxidative capacity of slow-twitch muscle (52).

In summary, we found that AMPK activation simultaneously increased glucose and LCFA clearance in vivo. Only muscle glucose uptake, however, was inhibited by l-NAME. This demonstrates that the mechanism by which AMPK increases glucose uptake is NO dependent, while AMPK-induced LCFA uptake occurs independent of this pathway in the majority of muscles studied. In addition, the magnitude of AICAR activation of both glucose and LCFA uptake in vivo is fiber-type specific. Soleus, which has the greatest percentage of slow-twitch fibers, exhibits the greatest increase in LCFA uptake and the smallest increase in glucose uptake. Taken together, our findings provide insight into the signaling mechanisms by which AMPK results in increased glucose and LCFA uptake in skeletal muscle.

FIG. 1.

Index of glucose uptake (Rg) in the soleus (A), gastrocnemius (B), and SVL (C). AICAR + LIPID represents treatment with AICAR + Intralipid. Different letters above bars indicate significant differences between treatments (P < 0.05). All data are presented as means ± SEM.

FIG. 1.

Index of glucose uptake (Rg) in the soleus (A), gastrocnemius (B), and SVL (C). AICAR + LIPID represents treatment with AICAR + Intralipid. Different letters above bars indicate significant differences between treatments (P < 0.05). All data are presented as means ± SEM.

Close modal
FIG. 2.

Glucose clearance (Kg) for soleus (A), gastrocnemius (B), and SVL (C). AICAR + LIPID represents treatment with AICAR + Intralipid. Different letters above bars indicate significant differences between treatments (P < 0.05). All data are presented as means ± SEM.

FIG. 2.

Glucose clearance (Kg) for soleus (A), gastrocnemius (B), and SVL (C). AICAR + LIPID represents treatment with AICAR + Intralipid. Different letters above bars indicate significant differences between treatments (P < 0.05). All data are presented as means ± SEM.

Close modal
FIG. 3.

Index of LCFA uptake (Rf) in the soleus (A), gastrocnemius (B), and SVL (C). AICAR + LIPID represents treatment with AICAR + Intralipid. Different letters above bars indicate significant differences between treatments (P < 0.05). All data are presented as means ± SEM.

FIG. 3.

Index of LCFA uptake (Rf) in the soleus (A), gastrocnemius (B), and SVL (C). AICAR + LIPID represents treatment with AICAR + Intralipid. Different letters above bars indicate significant differences between treatments (P < 0.05). All data are presented as means ± SEM.

Close modal
FIG. 4.

LCFA clearance (Kf) in the soleus (A), gastrocnemius (B), and SVL (C). AICAR + LIPID represents treatment with AICAR + Intralipid. Different letters above bars indicate significant differences between treatments (P < 0.05). All data are presented as means ± SEM.

FIG. 4.

LCFA clearance (Kf) in the soleus (A), gastrocnemius (B), and SVL (C). AICAR + LIPID represents treatment with AICAR + Intralipid. Different letters above bars indicate significant differences between treatments (P < 0.05). All data are presented as means ± SEM.

Close modal
FIG. 5.

AMPK-dependent phosphorylation of gastrocnemius muscle. A: AMPK phosphorylation (AMPK Thr172). B: Acetyl-CoA carboxylase phosphorylation (ACC Ser79). C: Endothelial NOS phosphorylation (NOS Ser1177). Different letters above bars indicate significant differences between treatments (P < 0.05). All data are presented as means ± SEM.

FIG. 5.

AMPK-dependent phosphorylation of gastrocnemius muscle. A: AMPK phosphorylation (AMPK Thr172). B: Acetyl-CoA carboxylase phosphorylation (ACC Ser79). C: Endothelial NOS phosphorylation (NOS Ser1177). Different letters above bars indicate significant differences between treatments (P < 0.05). All data are presented as means ± SEM.

Close modal
TABLE 1

Baseline characteristics

nWeight (g)Glucose (mmol/l)Insulin (μU/ml)NEFA (mmol/l)
Saline 310 ± 8 6.3 ± 0.1 6.3 ± 0.7 0.83 ± 0.11 
AICAR 317 ± 10 6.0 ± 0.2 8.2 ± 0.5 0.76 ± 0.17 
l-NAME 311 ± 7 6.8 ± 0.3 6.9 ± 1.2 0.82 ± 0.09 
AICAR + l-NAME 281 ± 7* 6.4 ± 0.2 7.2 ± 0.9 0.75 ± 0.07 
AICAR + LIPID 304 ± 7 6.8 ± 0.1 9.0 ± 2.3 0.73 ± 0.06 
nWeight (g)Glucose (mmol/l)Insulin (μU/ml)NEFA (mmol/l)
Saline 310 ± 8 6.3 ± 0.1 6.3 ± 0.7 0.83 ± 0.11 
AICAR 317 ± 10 6.0 ± 0.2 8.2 ± 0.5 0.76 ± 0.17 
l-NAME 311 ± 7 6.8 ± 0.3 6.9 ± 1.2 0.82 ± 0.09 
AICAR + l-NAME 281 ± 7* 6.4 ± 0.2 7.2 ± 0.9 0.75 ± 0.07 
AICAR + LIPID 304 ± 7 6.8 ± 0.1 9.0 ± 2.3 0.73 ± 0.06 

Data are means ± SEM.

*

Significant difference (P < 0.05) from AICAR.

TABLE 2

Arterial plasma glucose, insulin, NEFA, and glucose infusion rates during the experimental period

Glucose (mmol/l)Insulin (μU/ml)NEFA (mmol/l)GIR (mg · kg−1 · min−1)
Saline 6.2 ± 0.2 7.6 ± 0.7* 0.84 ± 0.04* — 
AICAR 6.9 ± 0.2 5.8 ± 0.9* 0.21 ± 0.03† 8.1 ± 0.8 
l-NAME 6.4 ± 0.4 6.1 ± 0.7* 0.75 ± 0.02* — 
AICAR + l-NAME 6.9 ± 0.2 11.6 ± 2.1* 0.16 ± 0.02† 8.6 ± 1.2 
AICAR + LIPID 6.9 ± 0.2 24.6 ± 3.1† 0.95 ± 0.06* 10.1 ± 1.2 
Glucose (mmol/l)Insulin (μU/ml)NEFA (mmol/l)GIR (mg · kg−1 · min−1)
Saline 6.2 ± 0.2 7.6 ± 0.7* 0.84 ± 0.04* — 
AICAR 6.9 ± 0.2 5.8 ± 0.9* 0.21 ± 0.03† 8.1 ± 0.8 
l-NAME 6.4 ± 0.4 6.1 ± 0.7* 0.75 ± 0.02* — 
AICAR + l-NAME 6.9 ± 0.2 11.6 ± 2.1* 0.16 ± 0.02† 8.6 ± 1.2 
AICAR + LIPID 6.9 ± 0.2 24.6 ± 3.1† 0.95 ± 0.06* 10.1 ± 1.2 

Data are means ± SEM. Data with different symbols within a measurement indicate a significant difference (P < 0.05).

This work was supported by NIDDK DK54902 (to D.W.). J.S. is supported by a mentor-based fellowship from the American Diabetes Association.

The authors acknowledge the contributions of Paul Martin, Angela Slater, Freyja James; the scientific input of Dr. Owen McGuinness; and the laboratories of Erik Richter and Jorgen Wojtaszewski at the Copenhagen Muscle Research Centre, Copenhagen, Denmark, for their instruction and enthusiasm in AMPK research.

1.
Kahn BB, Flier JS: Obesity and insulin resistance.
J Clin Invest
106
:
473
–481,
2000
2.
Kelley D, Mandarino L: Fuel selection in human skeletal muscle in insulin resistance: a reexamination.
Diabetes
49
:
677
–683,
2000
3.
Winder WW, Hardie DG: AMP-activated protein kinase, a metabolic master switch: possible roles in type 2 diabetes.
Am J Physiol Endocrinol Metab
277
:
E1
–E10,
1999
4.
Hardie DG, Pan DA: Regulation of fatty acid synthesis and oxidation by the AMP-activated protein kinase.
Biochem Soc Trans
30
:
1064
–1070,
2002
5.
Winder WW, Hardie DG: Inactivation of acetyl-CoA carboxylase and activation of AMP-activated protein kinase in muscle during exercise.
Am J Physiol Endocrinol Metab
270
:
E299
–E304,
1996
6.
Fryer LGD, Parbu-Patel A, Carling D: Protein kinase inhibitors block the stimulation of the AMP-activated protein kinase by 5-amino-4-imidazolecarboxamide riboside.
FEBS Lett
531
:
189
–192,
2002
7.
Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J, Wu M, Ventre J, Doebber T, Fujii N, Musi N, Hirshman MF, Goodyear LJ, Moller DE: Role of AMP-activated protein kinase in mechanism of metformin action.
J Clin Invest
108
:
1167
–1174,
2001
8.
Steinberg GR, Rush JWE, Dyck DJ: AMPK expression and phosphorylation are increased in rodent muscle after chronic leptin treatment.
Am J Physiol Endocrinol Metab
284
:
E648
–E654,
2003
9.
Yamauchi T, Kamon Y, Minokoshi Y, Ito Y, Waki H, Uchida S, Yamashita S, Noda M, Kita S, Ueki K, Eto K, Akanuma Y, Froguel P, Foufelle F, Ferre P, Carling D, Kimura R, Nagai R, Kahn BB, Kadowaki T: Adiponectin stimulates glucose utilization and fatty acid oxidation by activating AMP-activated kinase.
Nat Med
8
:
1288
–1295,
2002
10.
Morrow VA, Foufelle F, Connell JMC, Petrie JR, Gould GW, Salt IP: Direct activation of AMP-activated protein kinase stimulates nitric oxide synthesis in human aortic endothelial cells.
J Biol Chem
278
:
31629
–31639,
2003
11.
Fryer L, Hajduch E, Rencurel F, Salt I, Hundal H, Hardie D, Carling D: Activation of glucose transport by AMP-activated protein kinase via stimulation of nitric oxide synthase.
Diabetes
49
:
1978
–1985,
2000
12.
Balon TW, Nadler JL: Nitric oxide release is present from incubated skeletal muscle preparations.
J Appl Physiol
77
:
2519
–2521,
1994
13.
Balon TW, Nadler JL: Evidence that nitric oxide increases glucose transport in skeletal muscle.
J Appl Physiol
82
:
359
–363,
1997
14.
Roy D, Perreault M, Marette A: Insulin stimulation of glucose uptake in skeletal muscles and adipose tissues in vivo is NO dependent.
Am J Physiol Endocrinol Metab
274
:
E692
–E699,
1998
15.
Kingwell BA, Formosa M, Muhlmann M, Bradley SJ, McConell GK: Nitric oxide synthase inhibition reduces glucose uptake during exercise in individuals with type 2 diabetes more than in control subjects.
Diabetes
51
:
2572
–2580,
2002
16.
Etgen G, Fryburg D, Gibbs E: Nitric oxide stimulates skeletal muscle glucose transport through a calcium/contraction- and phosphatidylinositol-3-kinase-independent pathway.
Diabetes
46
:
1915
–1919,
1997
17.
Baron AD, Zhu JS, Marshall S, Irsula O, Brechtel G, Keech C: Insulin resistance after hypertension induced by the nitric oxide synthesis inhibitor L-NMMA in rats.
Am J Physiol Endocrinol Metab
269
:
E709
–E715,
1995
18.
Winder WW, Hardie DG: Inactivation of acetyl-CoA carboxylase and activation of AMP-activated protein kinase in muscle during exercise.
Am J Physiol
270
:
E299
–E304,
1996
19.
Petersen HA, Fueger PT, Bracy DP, Wasserman DH, Halseth AE: Fiber type-specific determinants of Vmax for insulin-stimulated muscle glucose uptake in vivo.
Am J Physiol Endocrinol Metab
284
:
E541
–E548,
2003
20.
Zatz R, Baylis C: Chronic nitric oxide inhibition model six years on.
Hypertension
32
:
958
–964,
1998
21.
Arnal JF, Warin L, Michel JB: Determinants of aortic cyclic guanosine monophosphate in hypertension induced by chronic inhibition of nitric oxide synthase.
J Clin Invest
90
:
647
–652,
1992
22.
Kaikita K, Fogo AB, Ma L, Schoenhard JA, Brown NJ, Vaughan DE: Plasminogen activator inhibitor-1 deficiency prevents hypertension and vascular fibrosis in response to long-term nitric oxide synthase inhibition.
Circulation
104
:
839
–844,
2001
23.
Bernstein R, Ochoa F, Xu X, Forfia P, Shen W, Thompson C, Hintze T: Function and production of nitric oxide in the coronary circulation of the conscious dog during exercise.
Circ Res
79
:
840
–848,
1996
24.
Armstrong RB, Phelps RO: Muscle fiber type composition of the rat hindlimb.
Am J Anat
171
:
259
–272,
1984
25.
Morgan CR, Lazarow AL: Immunoassay of insulin: two antibody system: plasma insulin of normal, subdiabetic, and diabetic rats.
Am J Med Sci
257
:
415
–419,
1963
26.
Dyck DJ, Putman CT, Heigenhauser GJ, Hultman E, Spriet LL: Regulation of fat-carbohydrate interaction in skeletal muscle during intense aerobic cycling.
Am J Physiol Endocrinol Metab
265
:
E852
–E859,
1993
27.
Hargreaves M, Kiens B, Richter EA: Effect of increased plasma free fatty acid concentrations on muscle metabolism in exercising men.
J Appl Physiol
70
:
194
–201,
1991
28.
Rottman JN, Bracy D, Malabanan C, Yue Z, Clanton J, Wasserman DH: Contrasting effects of exercise and NOS inhibition on tissue-specific fatty acid and glucose uptake in mice.
Am J Physiol Endocrinol Metab
283
:
E116
–E123,
2002
29.
Halseth AE, Bracy DP, Wasserman DH: Limitations to basal and insulin-stimulated skeletal muscle glucose uptake in the high-fat-fed rat.
Am J Physiol Endocrinol Metab
279
:
E1064
–E1071,
2000
30.
Meszaros K, Bagby GJ, Lang CH, Spitzer JJ: Increased uptake and phosphorylation of 2-deoxyglucose by skeletal muscles in endotoxin-treated rats.
Am J Physiol Endocrinol Metab
253
:
E33
–E39,
1987
31.
Mezaros K, Lang CH, Hargrove DM, Spitzer JJ: Tissue glucose utilization during epinephrine induced hyperglycemia.
J Appl Physiol
67
:
1770
–1775,
1989
32.
Virkamaki A, Rissanen E, Hamalainen S, Utriainen T, Yki-Jarvinen H: Incorperation of [3-3H]glucose and 2-[1-14C]deoxyglucose into glycogen in heart and skeletal muscle in vivo: impliactions for the quantitation of tissue glucose uptake.
Diabetes
46
:
1106
–1110,
1997
33.
Furler SM, Jenkins AB, Kraegen EW: Effect of insulin on [3H]deoxy-D-glucose pharmacokinetics in the rat.
Am J Physiol
255
:
E806
–E811,
1988
34.
Kraegen EW, James DE, Jenkins AB, Chisholm DJ: Dose-response curves for in vivo insulin sensitivity in individual tissues in rats.
Am J Physiol
248
:
E353
–E362,
1985
35.
Coburn CT, Knapp FF Jr, Febbraio M, Beets AL, Silverstein RL, Abumrad NA: Defective uptake and utilization of long chain fatty acids in muscle and adipose tissues of CD36 knockout mice.
J Biol Chem
275
:
32523
–32529,
2000
36.
Balent B, Goswami G, Goodloe G, Rogatsky E, Rauta O, Nezami R, Mints L, Angeletti R, Stein D: Acute elevation of NEFA causes hyperinsulinemia without effect on insulin secretion rate in healthy human subjects.
Ann N Y Acad Sci
967
:
535
–543,
2002
37.
Young ME, Radda GK, Leighton B: Nitric Oxide stimulates glucose transport and metabolism in rat skeletal muscle in vitro.
Biochem J
322
:
223
–228,
1997
38.
Russell RR III, Bergeron R, Shulman GI, Young LH: Translocation of myocardial GLUT-4 and increased glucose uptake through activation of AMPK by AICAR.
Am J Physiol Heart Circ Physiol
277
:
H643
–H649,
1999
39.
Stamler JS, Meissner G: Physiology of nitric oxide in skeletal muscle.
Physiol Rev
81
:
209
–237,
2001
40.
Chen ZP, Mitchelhill KI, Michell BJ, Stapleton D, Rodriguez-Crespo I, Witters LA, Power DA, Ortiz de Montellano PR, Kemp BE: AMP-activated protein kinase phosphorylation of endothelial NO synthase.
FEBS Lett
443
:
285
–289,
1999
41.
Chen ZP, McConell GK, Michell BJ, Snow RJ, Canny BJ, Kemp BE: AMPK signaling in contracting human skeletal muscle: acetyl-CoA carboxylase and NO synthase phosphorylation.
Am J Physiol Endocrinol Metab
279
:
E1202
–E1206,
2000
42.
Michell BJ, Chen Z, Tiganis T, Stapleton D, Katsis F, Power DA, Sim AT, Kemp BE: Coordinated control of endothelial nitric-oxide synthase phosphorylation by protein kinase C and the cAMP-dependent protein kinase.
J Biol Chem
276
:
17625
–17628,
2001
43.
Harris MB, Ju H, Venema VJ, Liang H, Zou R, Michell BJ, Chen ZP, Kemp BE, Venema RC: Reciprocal phosphorylation and regulation of endothelial nitric-oxide synthase in response to bradykinin stimulation.
J Biol Chem
276
:
16587
–16591,
2001
44.
Butt E, Bernhardt M, Smolenski A, Kotsonis P, Frohlich LG, Sickmann A, Meyer HE, Lohmann SM, Schmidt HH: Endothelial nitric-oxide synthase (type III) is activated and becomes calcium independent upon phosphorylation by cyclic nucleotide-dependent protein kinases.
J Biol Chem
275
:
5179
–5187,
2000
45.
Dimmeler S, Fleming I, Fisslthaler B, Hermann C, Busse R, Zeiher AM: Activation of nitric oxide synthase in endothelial cells by Akt-dependent phosphorylation.
Nature
399
:
601
–605,
1999
46.
Fulton D, Gratton JP, McCabe TJ, Fontana J, Fujio Y, Walsh K, Franke TF, Papapetropoulos A, Sessa WC: Regulation of endothelium-derived nitric oxide production by the protein kinase Akt.
Nature
399
:
597
–601,
1999
47.
Luiken JJFP, Coort SLM, Willems J, Coumans WA, Bonen A, van der Vusse GJ, Glatz JFC: Contraction-induced fatty acid translocase/CD36 translocation in rat cardiac myocytes is mediated through AMP-activated protein kinase signaling.
Diabetes
52
:
1627
–1634,
2003
48.
Ruderman NB, Saha AK, Vavvas D, Witters LA: Malonyl-CoA, fuel sensing, and insulin resistance.
Am J Physiol
276
:
E1
–E18,
1999
49.
Saha AK, Schwarsin AJ, Roduit R, Masse F, Kaushik V, Tornheim K, Prentki M, Ruderman NB: Activation of malonyl-CoA decarboxylase in rat skeletal muscle by contraction and the AMP-activated protein kinase activator 5-aminoimidazole-4-carboxamide-1-beta-D-ribofuranoside.
J Biol Chem
275
:
24279
–24283,
2000
50.
Jessen N, Pold R, Buhl ES, Jensen LS, Schmitz O, Lund S: Effects of AICAR and exercise on insulin-stimulated glucose uptake, signaling, and GLUT-4 content in rat muscles.
J Appl Physiol
94
:
1373
–1379,
2003
51.
Ai H, Ihlemann J, Hellsten Y, Lauritzen HPMM, Hardie DG, Galbo H, Ploug T: Effect of fiber type and nutritional state on AICAR- and contraction-stimulated glucose transport in rat muscle.
Am J Physiol Endocrinol Metab
282
:
E1291
–E1300,
2002
52.
Bonen A, Dyck DJ, Luiken JJFP: Skeletal muscle fatty acid transport and transporters. In
Skeletal Muscle Metabolism in Exercise and Diabetes
. Richter EA, Kien B, Galbo H, Saltin B, Eds. New York, Plenum Press,
1998
, p.
181
–205