Activation of AMP-activated protein kinase (AMPK) with 5-aminoimidazole-4-carboxamide-1-β-d-ribofuranoside (AICAR) increases glucose transport in skeletal muscle via an insulin-independent pathway. To examine the effects of AMPK activation on skeletal muscle glucose transport activity and whole-body carbohydrate and lipid metabolism in an insulin-resistant rat model, awake obese Zucker fa/fa rats (n = 26) and their lean (n = 23) littermates were infused for 90 min with AICAR, insulin, or saline. The insulin infusion rate (4 mU · kg1 · min1) was selected to match the glucose requirements during AICAR (bolus, 100 mg/kg; constant, 10 mg · kg1 · min1) isoglycemic clamps in the lean rats. The effects of these identical AICAR and insulin infusion rates were then examined in the obese Zucker rats. AICAR infusion increased muscle AMPK activity more than fivefold (P < 0.01 vs. control and insulin) in both lean and obese rats. Plasma triglycerides, fatty acid concentrations, and glycerol turnover, as assessed by [2-13C]glycerol, were all decreased in both lean and obese rats infused with AICAR (P < 0.05 vs. basal), whereas insulin had no effect on these parameters in the obese rats. Endogenous glucose production rates, measured by [U-13C]glucose, were suppressed by >50% during AICAR and insulin infusions in both lean and obese rats (P < 0.05 vs. basal). In lean rats, rates of whole-body glucose disposal increased by more than twofold (P < 0.05 vs. basal) during both AICAR and insulin infusion; [3H]2-deoxy-d-glucose transport activity increased to a similar extent, by >2.2-fold (both P < 0.05 vs. control), in both soleus and red gastrocnemius muscles of lean rats infused with either AICAR or insulin. In the obese Zucker rats, neither AICAR nor insulin stimulated whole-body glucose disposal or soleus muscle glucose transport activity. However, AICAR increased glucose transport activity by ∼2.4-fold (P < 0.05 vs. control) in the red gastrocnemius from obese rats, whereas insulin had no effect. In summary, acute infusion of AICAR in an insulin-resistant rat model activates skeletal muscle AMPK and increases glucose transport activity in red gastrocnemius muscle while suppressing endogenous glucose production and lipolysis. Because type 2 diabetes is characterized by diminished rates of insulin-stimulated glucose uptake as well as increased basal rates of endogenous glucose production and lipolysis, these results suggest that AICAR-related compounds may represent a new class of antidiabetic agents.

Insulin resistance is a major factor contributing to the pathogenesis of type 2 diabetes. It is largely attributable to reduced insulin-stimulated muscle glucose uptake (1,2,3) and muscle glycogen synthesis (4), which in turn can be attributed to reduced muscle glucose transport activity (5,6). In contrast, contraction-stimulated muscle glucose transport, which is thought to be insulin independent, is normal in obese Zucker rats (7,8). In addition, exercise increases GLUT4 plasma membrane content to a similar extent in type 2 diabetic patients and normal subjects (9). Furthermore, the insulin-independent phase of glycogen resynthesis immediately after exercise is normal in the muscles of insulin-resistant offspring of type 2 diabetic patients (10). Therefore, the exercise-mediated non–insulin-dependent pathway for the stimulation of muscle glucose uptake may provide an important target for the treatment of type 2 diabetes. Recent evidence indicates that pharmacological activation of AMP-activated protein kinase (AMPK) by 5-aminoimidazole-4-carboxamide-1-β-d-ribofuranoside (AICAR) stimulates glucose transport activity in skeletal muscle (11,12,13) and heart (14) in the absence of phosphoinositide 3-kinase activation, suggesting that this action is independent of the insulin-signaling pathway. Furthermore, muscle AMPK is activated during exercise (15), and may play an important role in contraction-induced stimulation of glucose transport (12,13). However, the effect of AMPK activation on muscle glucose uptake in an insulin-resistant state has not been studied.

Increased basal rates of endogenous glucose production (EGP) and diminished insulin-induced suppression of EGP are two other characteristics of type 2 diabetes (16,17,18). In vitro incubation of rat hepatocytes with AICAR has been shown to decrease hepatic gluconeogenesis by inhibiting fructose-1,6-bisphosphatase (19). In addition, intraperitoneal injection of AICAR causes hypoglycemia in mice (20). In vivo, AICAR inhibits EGP under euglycemic clamp conditions in normal rats (13). However, the effect of in vivo AICAR infusion on EGP in an insulin-resistant state is unknown.

Increased fasting plasma fatty acid concentrations are common among patients with type 2 diabetes (21,22) because of impaired insulin-induced suppression of lipolysis (23), and may contribute to skeletal muscle (24,25,26) and liver (27,28) insulin resistance. In vivo infusion of AICAR leads to a 50% reduction in fasting plasma fatty acid concentrations in normal rats (13). Activation of AMPK stimulates the oxidation of fat in skeletal muscle (11) through the inhibition of acetyl CoA carboxylase and subsequent activation of carnitine palmitoyl-CoA transferase-1. Furthermore, isoprenaline-induced lipolysis from rat adipocytes is inhibited by AICAR, suggesting that AMPK activation exerts an antilipolytic effect by preventing the activation of hormone-sensitive lipase in rat adipocytes (29,30). Therefore, AMPK also plays an important role in the regulation of fat metabolism, a factor that has not been studied in an insulin-resistant state.

Thus, this study was performed to determine the effects of AICAR infusion on in vivo skeletal muscle glucose transport activity and whole-body glucose and glycerol turnover rates in insulin-resistant obese Zucker rats.

Infusion protocols.

We studied genetically obese Zucker fa/fa male rats ages 11–12 weeks (Charles River, Raleigh, NC) and their age-matched lean littermates. The rats were maintained on standard Rat Chow (Ralston Purina, St. Louis, MO) and housed in an environmentally controlled room with a 12:12-h light-dark cycle. Rats were chronically catheterized via the right jugular vein and carotid artery (31) and allowed to recover (5–8 days) until they regained their preoperative weight. The 12-h fasted animals were infused with [U-13C]glucose (0.4 μmol · kg1 · min1) and [2-13C]glycerol (1 μmol · kg1 · min1) (Cambridge Isotopes, Andover, MA; both 99% 13C atom percent excess) for 120 min to determine rates of basal EGP and glycerol turnover, and infusions were continued for the 90-min experimental infusions. Following this baseline period, rats were randomly infused with isotonic saline (control), AICAR (bolus, 100 mg/kg; constant, 10 mg · kg1 · min1; Sigma, St. Louis, MO), or insulin (4 mU · kg1 · min1; Humulin Regular; Eli Lilly, Indianapolis, IN). During the AICAR and insulin studies, plasma glucose concentrations were maintained at basal fasting concentrations (∼7 and ∼8 mmol/l in lean and obese rats, respectively) using a 50% (wt/vol) glucose solution enriched with [U-13C]glucose (∼0.8% isotopic enrichment). Blood was sampled every 5 min for glucose measurements and immediately before and at the end of the clamp to determine plasma insulin, fatty acid, glycerol, triglyceride, lactate, and AICAR concentrations, and every 15 min during the last 30 min before and at the end of the clamp to determine plasma isotopic enrichment of [U-13C]glucose and [2-13C]glycerol. The protocol was approved by the Yale Animal Care and Use Committee.

Glucose uptake.

Skeletal muscle glucose uptake was measured according to a previously described method (32). Briefly, 30 min into the clamp, a 37-μCi bolus of 2-deoxy-d-[1,2-3H]glucose (2-DG) was injected intravenously. Plasma samples were obtained at 0.5, 1.0, 1.5, 2, 3, 5, 7.5, 10, 15, 20, 30, 45, and 60 min after the bolus infusion to estimate the plasma tracer activity. Glucose uptake rate calculations were based on the mean plasma glucose concentration, the radiolabeled phosphorylated 2-DG tissue concentration, and the area under the plasma 2-DG curve as described by Kraegen et al. (32).

Tissue analysis.

At the end of the experiment, the rats were anesthetized intravenously with pentobarbital (50 mg/kg). The calf muscle group from the right hindlimb was quickly freeze-clamped in situ for determination of AMPK activity and nucleotide and metabolite analyses. Muscle samples were kept in liquid nitrogen until analyzed. Soleus and red gastrocnemius were dissected from the left hindlimb and individually freeze clamped for determination of glucose uptake rates.

AICAR and EGP during high plasma fatty acid concentrations.

Lean Zucker rats were infused for 5 h either with a triglyceride emulsion (8.5 mg · kg1 · min1 triglycerides; Intralipid, 20%; Abbott Laboratories, Abbott Park, IL) combined with heparin (210 mU · kg1 · min1; lipid group, n = 6) or glycerol (1.25 mg of glycerol · kg1 · min1; glycerol group, n = 5) as a control. Then 3 h into the lipid or glycerol infusion, all rats were infused with [U-13C]glucose for a period of 120 min to assess whole-body glucose metabolism. Rats then underwent a 90-min AICAR euglycemic clamp, as described above. Blood was sampled before the lipid or glycerol infusions were started, then immediately before the AICAR infusion, and finally at the end of the experiment.

Analytical procedures.

Plasma glucose and lactate concentrations were determined using an automated analyzer (YSI Instruments, Yellow Springs, OH). Immunoreactive insulin was assayed using a double-antibody immunoassay kit (Linco Research, St. Louis, MO). Plasma fatty acid (Wako, Osaka, Japan), glycerol, and triglyceride (Sigma) concentrations were assayed using colorimetric kits. AICAR plasma concentrations were determined spectrophotometrically (33). Enrichment of [U-13C]glucose and [2-13C]glycerol in plasma was determined by gas chromatography−mass spectrometry, as previously described (34). Phosphorylated muscle [3H]2-DG was separated from a perchloric acid extract using ion exchange chromatography (35). Muscle glycogen was assayed using the amyloglucosidase method (36). Muscle nucleotides and monophosphorylated AICAR (ZMP) were separated and quantified by high-performance anion-exchange liquid chromatography (37). Skeletal muscle AMPK activity was determined by following the incorporation of [32P]ATP into a synthetic peptide (15) containing the following 15–amino acid sequence: AMARAASAAALARRR (38).

Calculations.

Rates of glucose and glycerol turnover were measured under steady-state conditions and were calculated using the following formula: basal glucose or glycerol turnover = f × ([IEinfusate/IEplasma] − 1) where f is the infusion rate of either the [U-13C]glucose or the [2-13C]glycerol tracer (μmol · kg1 · min1), and IE is the isotopic enrichment (39). Clamp EGP was calculated as follows: clamp EGP = GIR × ([Einfusate/Eplasma] − 1), where GIR is the mean glucose infusion rate for the last 30 min of the clamp. The clamp glucose disposal rate (Rd) was calculated as follows: Rd = clamp EGP + GIR.

Statistical analyses.

All data are reported as means ± SE. Data from blood sampling and substrate turnover measurements were analyzed by a two-way analysis of variance (ANOVA) for repeated measures, and data obtained from tissue samples were analyzed with a two-way ANOVA. These analyses were followed by Tukey’s test for post hoc comparisons. Differences were considered statistically significant at P < 0.05.

The mean body weight of the obese rats was significantly greater when compared with their age-matched lean littermates (355 ± 10 vs. 275 ± 9 g, respectively; P < 0.01). Basal plasma concentrations of glucose, insulin, fatty acids, glycerol, and triglycerides were all significantly higher in the obese as compared with the lean rats (P < 0.01) (Table 1). The infusion of AICAR resulted in similar plasma AICAR concentrations of 2.26 ± 0.56 and 2.51 ± 0.39 mmol/l in the obese and lean rats, respectively.

During the infusion of AICAR or insulin, plasma glucose concentrations remained stable and did not differ from their respective basal concentrations. Plasma insulin concentrations were significantly increased during insulin infusion in the lean (P < 0.01) but not in the obese rats as compared with basal levels (Table 1). AICAR infusion was associated with a significant reduction in plasma insulin concentrations in the obese rats (P < 0.01), whereas concentrations did not change in the lean rats. During the AICAR and insulin clamps, plasma fatty acid and triglyceride concentrations were significantly decreased by AICAR in both lean and obese rats, whereas insulin decreased only plasma fatty acids concentrations in the lean rats (Table 1). This decrease in plasma fatty acid concentrations was greater in the AICAR-infused compared with the insulin-infused lean rats (P < 0.05). Plasma glycerol concentrations during AICAR infusion decreased in the obese rats but increased in the lean rats (both P < 0.05) (Table 1). AICAR-infused rats showed substantial increases in plasma lactate concentrations (P < 0.01) compared with insulin-infused rats; in the latter, there was just a modest increase in plasma lactate concentrations (P < 0.05) compared with the saline-infused rats (Table 1).

Tissues analyses.

Muscle glycogen concentrations were similar in lean and obese rats infused with saline (21.6 ± 2.1 vs. 21.6 ± 2.3 μmol/g wet wt, respectively), and were not altered by AICAR or insulin infusion (data not shown). Although skeletal muscle nucleotide concentrations were not significantly different between the lean and obese rats (data not shown), the infusion of AICAR significantly increased ZMP concentrations in the calf muscle group of both lean and obese rats compared with the levels in the control and insulin-infused groups (P < 0.01). Muscle ZMP content was higher in the lean rats than in the obese rats infused with AICAR (1.70 ± 0.37 vs. 0.55 ± 0.02 μmol/g wet wt, respectively; P < 0.05), although AMPK activation was similar (6.6- and 5.3-fold, respectively) when compared with that of the lean rats (P < 0.01 vs. control) (Fig. 1). Insulin infusion had no significant effect on AMPK activity.

Skeletal muscle glucose uptake.

In lean rats, both AICAR and insulin infusion increased the uptake of [3H]2-deoxy-d-glucose in the red gastrocnemius and soleus muscle of lean rats by more than twofold when compared with saline infusion (P < 0.01) (Figs. 2A and B). In obese rats, AICAR infusion stimulated glucose uptake in the red gastrocnemius by 2.4-fold (P < 0.02), but did not increase glucose uptake in the soleus muscle. In contrast, insulin failed to increase glucose uptake in either the soleus or the red gastrocnemius muscle of obese rats.

Whole-body glucose turnover.

By design, the glucose infusion rates necessary to maintain isoglycemia during the AICAR and insulin infusions were similar in the lean rats (Table 2). In the obese rats, the mean glucose infusion rates during the insulin and AICAR clamps were lower (71 and 72%, respectively) than in the lean rats (P < 0.01). During the AICAR and insulin clamps, glucose disposal rates increased two- to threefold in the lean rats compared with the control rats (both P < 0.05) (Table 2). As expected, insulin-stimulated glucose disposal was blunted in the obese rats, reflecting their insulin-resistant state (P < 0.01). Somewhat surprisingly, AICAR infusion did not increase the rate of whole-body glucose disposal in the obese rats.

Under basal conditions, EGP was 33% higher in the obese rats (P < 0.01) (Table 2). Both AICAR and insulin infusions suppressed EGP in lean and obese rats (P < 0.01). In the lean group, AICAR suppressed EGP to a greater extent than did insulin (82 ± 6 vs. 53 ± 12%, respectively; P < 0.05). However, in the obese rats, suppression of EGP during AICAR and insulin infusions was similar (52 ± 8 vs. 55 ± 11%, respectively). Although the relative suppression of EGP (expressed in percent of the basal rate) during AICAR infusion was greater in the lean rats (P < 0.05), the absolute changes in EGP were similar in lean and obese rats (29.1 ± 3.9 vs. 27.3 ± 3.9 μmol · kg1 · min1, respectively) (Table 2).

Whole-body glycerol turnover.

The basal rates of glycerol turnover were similar in the obese and lean rats (Table 2). During the AICAR infusion, glycerol turnover decreased in both obese and lean rats (P < 0.05). In contrast, insulin infusion decreased glycerol turnover in the lean, but not in the obese, rats (Table 2).

AICAR and EGP during high plasma fatty acid concentrations.

Because plasma fatty acids are potential modulators of EGP, we infused a triglyceride/heparin emulsion to eliminate the confounding effects of AICAR-induced decreases in plasma fatty acid concentrations on EGP. The infusion of lipid increased the plasma concentrations of fatty acids from 0.54 ± 0.04 to 2.36 ± 0.23 mmol/l (P < 0.005 vs. basal), whereas fatty acid concentrations remained stable (∼0.50 mmol/l) in the glycerol-infused group. AICAR infusion decreased plasma fatty acid concentrations in the glycerol-infused rats to 0.11 ± 0.01 mmol/l (P < 0.01 vs. basal), whereas plasma fatty acid concentrations remained elevated (3.08 ± 0.53 mmol/l) in the lipid group. The mean plasma insulin concentrations measured after the 5 h of lipid infusion were significantly higher than in the glycerol-infused rats (228 ± 39 vs. 90 ± 16 pmol/l, respectively; P < 0.05) and remained stable during the AICAR infusion in the lipid group, whereas plasma insulin concentrations decreased in the glycerol group to 18 ± 4 pmol/l. Glucose concentrations were clamped at similar concentrations during the AICAR infusion in both the lipid and glycerol groups (6.1 ± 0.3 and 5.8 ± 0.3 mmol/l, respectively). Glucose infusion rates during AICAR infusion were similar in both lipid and glycerol groups (89 ± 7 and 78 ± 7 μmol · kg1 · min1, respectively). AICAR infusion similarly suppressed EGP in both the lipid- and glycerol-infused groups (both P < 0.01) when compared with their respective basal rates of EGP. Thus, AICAR was able to suppress EGP independent of any changes in plasma fatty acid concentrations (Fig. 3).

In this study, the metabolic effects of AICAR, a known activator of AMPK, were compared with the effects of an equipotent dose of insulin in awake lean and obese Zucker rats. The AICAR infusion rate that was used in the present study was selected to produce a plasma AICAR concentration of ∼2 mmol/l, a concentration known to induce maximal AICAR stimulation of muscle glucose uptake in vitro (40). The insulin infusion rate of 4 mU · kg1 · min1 was chosen based on pilot studies that have shown this dose to be equally potent to AICAR with respect to glucose infusion rates required to maintain isoglycemia.

The present data extend our previous in vivo findings in normal SD rats (13) and demonstrate that under similar rates of glucose flux in lean Zucker rats, AICAR has a greater effect in suppressing EGP than insulin, indicating a potent effect of AICAR on the liver. Because plasma fatty acid concentrations can also modulate EGP (28,41), it is possible that the AICAR-induced decrease in plasma fatty acid concentrations could have indirectly decreased the rates of EGP (27). However, this possibility is not likely, as our data show that AICAR was able to suppress EGP when plasma fatty acid concentrations were maintained above 2 mmol/l by a concomitant intralipid/heparin infusion. This finding suggests that AICAR directly suppresses EGP independent of its effect on plasma fatty acid concentrations. The effect of AICAR in suppressing EGP may be mediated by activation of AMPK or by the allosteric inhibition of fructose-1,6-biphosphatase by ZMP, thereby suppressing hepatic gluconeogenesis (19). The observed increase in plasma lactate and glycerol concentrations in the lean rats infused with AICAR is consistent with the latter possibility.

AICAR significantly suppressed EGP in obese insulin-resistant Zucker rats, which have elevated fasting EGP when compared with their lean littermates, as previously reported (42). In obese Zucker rats, insulin-induced suppression of EGP was blunted during a 3.3 mU · kg1 · min1 hyperinsulinemic-euglycemic clamp that increased plasma insulin concentrations twofold (43). However, in the present study, insulin suppressed EGP to a similar extent in obese rats and lean littermates. This observation may be partly explained by the higher glucose concentration in the obese rats, as hyperglycemia per se has been shown to suppress hepatic glucose production (44,45).

Whole-body glucose disposal was increased by both insulin and AICAR infusions in the lean rats. This observation is likely attributable to the stimulation of skeletal muscle glucose uptake by both AICAR and insulin, as reflected by the soleus and red gastrocnemius muscles of lean rats. As expected, insulin-stimulated glucose disposal was impaired in the obese Zucker rats. However, somewhat surprisingly, AICAR failed to increase whole-body glucose disposal in the obese rats. Glucose uptake in the soleus was not stimulated by AICAR in the obese Zucker rats. However, AICAR increased glucose uptake in the red gastrocnemius by more than twofold. The soleus muscle in the rat is primarily composed of type I muscle fibers (I, >80%; IIa, 10%; IId/x, 0%; and IIb, 0%), whereas the red gastrocnemius is composed of mixed muscle fibers (I, 5–20%; IIa, 5–19%; IId/x, 20–40%; IIb, 1–8%) (46,47,48). The lack of an effect of AICAR on whole-body glucose disposal in obese Zucker rats might be partly explained by a high proportion (>70%) of type IIb muscle fibers, rather than type IId/x and IIa muscle fiber types, which are abundant in the red gastrocnemius (48). Although whole-body glucose disposal was not stimulated by AICAR in the obese Zucker rat, the observation that muscle glucose uptake is increased by more than twofold in the red gastrocnemius is important from a clinical standpoint, as human skeletal muscle mass is predominantly constituted of similar muscle fibers (49).

Because soleus muscle GLUT4 content appears to be similar in obese and lean rats (50,51), it is unclear why AICAR did not increase glucose uptake in the soleus muscle of obese Zucker rats. The content of monophosphorylated AICAR (ZMP) was lower in the calf muscle of obese rats compared with lean rats. The total AMPK activity also tended to be lower, although this trend was not statistically significant and AMPK activity was substantially activated in the muscle of obese rats. The lack of AICAR-induced stimulation of whole-body glucose disposal and glucose uptake in the soleus raises the possibility of impaired downstream signaling of AMPK on GLUT4 translocation in this model of insulin resistance.

Infusion of AICAR caused a decrease in lipolysis, as reflected by decreases in plasma fatty acid concentrations and glycerol turnover rates (52) in both lean and obese rats. As expected, insulin also decreased glycerol turnover in lean rats, whereas insulin failed to suppress glycerol turnover rates in the obese Zucker rats. The greater suppressive effects of AICAR infusion, as compared with the effects of insulin infusion on plasma fatty acid concentrations and glycerol turnover rates in the obese Zucker rats, suggest that AICAR has a potent antilipolytic effect in this insulin resistant rat model. In addition, AICAR infusion resulted in a decrease in plasma triglyceride concentrations in both lean and obese rats, which can likely be attributed to an inhibition of hepatic lipogenesis. The results are consistent with previous in vitro findings demonstrating AICAR-induced inhibition of mitochondrial glycerol-3-phosphate acyltransferase activity and subsequent inhibition of triacylglycerol synthesis (53).

In summary, these are the first studies to examine the effects of an acute infusion of AICAR in an insulin resistant rat model. We found that an infusion of AICAR in the awake obese Zucker rat 1) activated skeletal muscle AMPK more than fivefold, 2) increased glucose transport activity in the red gastrocnemius by ∼240%, 3) reduced EGP by ∼50%, and 4) suppressed lipolysis by ∼45%. Because type 2 diabetes is characterized by diminished rates of insulin-stimulated glucose uptake as well as increased basal rates of EGP and lipolysis, these results suggest that AICAR-related compounds may represent a new class of antidiabetic agents.

FIG. 1.

AMPK activity in calf muscle group from lean or obese Zucker rats after in vivo AICAR (bolus, 100 mg/kg body wt; constant, 10 mg · kg1 body wt · min1), insulin (4 mU · kg1 body wt · min1) or a volumetric equivalent infusion of isotonic saline (CONTROL). Values are reported as means ± SE; n = 7–8 each group. †Significantly different from both corresponding control and insulin-infused groups (P < 0.01).

FIG. 1.

AMPK activity in calf muscle group from lean or obese Zucker rats after in vivo AICAR (bolus, 100 mg/kg body wt; constant, 10 mg · kg1 body wt · min1), insulin (4 mU · kg1 body wt · min1) or a volumetric equivalent infusion of isotonic saline (CONTROL). Values are reported as means ± SE; n = 7–8 each group. †Significantly different from both corresponding control and insulin-infused groups (P < 0.01).

Close modal
FIG. 2.

Effect of an infusion of AICAR (bolus, 100 mg/kg body wt; constant, 10 mg · kg1 body wt · min1), insulin (4 mU · kg1 body wt · min1), or isotonic saline (CONTROL) in awake lean or obese Zucker rats on red gastrocnemius (A) and soleus 2-DG uptake (B). Data are reported as means ± SE; n = 7–10 each group. *Significantly different from lean littermates undergoing the same treatment (P < 0.05); †significantly different from corresponding control group (P < 0.01).

FIG. 2.

Effect of an infusion of AICAR (bolus, 100 mg/kg body wt; constant, 10 mg · kg1 body wt · min1), insulin (4 mU · kg1 body wt · min1), or isotonic saline (CONTROL) in awake lean or obese Zucker rats on red gastrocnemius (A) and soleus 2-DG uptake (B). Data are reported as means ± SE; n = 7–10 each group. *Significantly different from lean littermates undergoing the same treatment (P < 0.05); †significantly different from corresponding control group (P < 0.01).

Close modal
FIG. 3.

EGP during a 90-min AICAR clamp (bolus, 100 mg/kg body wt; constant, 10 mg · kg1 body wt · min1), estimated with the use of [U-13C]glucose in awake lean rats infused either with a triglyceride emulsion in combination with heparin (lipid) or glycerol. Data are reported as means ± SE; n = 5–6 each group. *Significantly different from lean littermates undergoing the same treatment (P < 0.05).

FIG. 3.

EGP during a 90-min AICAR clamp (bolus, 100 mg/kg body wt; constant, 10 mg · kg1 body wt · min1), estimated with the use of [U-13C]glucose in awake lean rats infused either with a triglyceride emulsion in combination with heparin (lipid) or glycerol. Data are reported as means ± SE; n = 5–6 each group. *Significantly different from lean littermates undergoing the same treatment (P < 0.05).

Close modal
TABLE 1

Plasma metabolite arterial concentrations obtained under basal state or during an isoglycemic clamp from lean and obese Zucker rats

Lean
Obese
ControlAICARInsulinControlAICARInsulin
Glucose (mmol/l) Basal 7.0 ± 0.2 6.8 ± 0.1 6.9 ± 0.2 8.1 ± 0.2* 8.2 ± 0.2* 8.6 ± 0.2* 
 Clamp 7.0 ± 0.1 6.9 ± 0.1 6.7 ± 0.1 8.3 ± 0.2* 7.6 ± 0.1* 8.4 ± 0.2* 
Insulin (pmol/l) Basal 238 ± 57 156 ± 24 167 ± 35 1189 ± 166* 895 ± 11* 1008 ± 127* 
 Clamp 290 ± 48 136 ± 48 380 ± 60 1032 ± 158* 167 ± 74 1224 ± 165* 
Fatty acids (mmol/l) Basal 0.46 ± 0.07 0.50 ± 0.08 0.59 ± 0.12 1.72 ± 0.47* 1.59 ± 0.37* 1.84 ± 0.34* 
 Clamp 0.38 ± 0.07 0.11 ± 0.02 0.34 ± 0.08 1.71 ± 0.26* 0.28 ± 0.09 1.06 ± 0.19* 
Glycerol (mmol/l) Basal 0.21 ± 0.02 0.24 ± 0.02 0.22 ± 0.03 0.56 ± 0.08* 0.55 ± 0.07* 0.65 ± 0.09* 
 Clamp 0.23 ± 0.03 0.38 ± 0.05§ 0.21 ± 0.04 0.61 ± 0.06* 0.34 ± 0.02§ 0.69 ± 0.12* 
Triglycerides (mg/dl) Basal 43.8 ± 7.3 32.7 ± 5.2 25.1 ± 4.5 97.7 ± 14.8* 117.1 ± 20.2* 89.3 ± 15.8* 
 Clamp 33.5 ± 9.1 21.1 ± 4.2 24.7 ± 3.8 96.1 ± 24.2 20.8 ± 3.8 64.6 ± 17.6 
Lactate (mmol/l) Basal 0.55 ± 0.04 0.57 ± 0.03 0.63 ± 0.04 1.10 ± 0.16* 1.21 ± 0.21* 1.30 ± 0.14* 
 Clamp 0.60 ± 0.03 13.20 ± 0.38 0.90 ± 0.06 1.03 ± 0.05 12.00 ± 0.53 1.51 ± 0.13 
Lean
Obese
ControlAICARInsulinControlAICARInsulin
Glucose (mmol/l) Basal 7.0 ± 0.2 6.8 ± 0.1 6.9 ± 0.2 8.1 ± 0.2* 8.2 ± 0.2* 8.6 ± 0.2* 
 Clamp 7.0 ± 0.1 6.9 ± 0.1 6.7 ± 0.1 8.3 ± 0.2* 7.6 ± 0.1* 8.4 ± 0.2* 
Insulin (pmol/l) Basal 238 ± 57 156 ± 24 167 ± 35 1189 ± 166* 895 ± 11* 1008 ± 127* 
 Clamp 290 ± 48 136 ± 48 380 ± 60 1032 ± 158* 167 ± 74 1224 ± 165* 
Fatty acids (mmol/l) Basal 0.46 ± 0.07 0.50 ± 0.08 0.59 ± 0.12 1.72 ± 0.47* 1.59 ± 0.37* 1.84 ± 0.34* 
 Clamp 0.38 ± 0.07 0.11 ± 0.02 0.34 ± 0.08 1.71 ± 0.26* 0.28 ± 0.09 1.06 ± 0.19* 
Glycerol (mmol/l) Basal 0.21 ± 0.02 0.24 ± 0.02 0.22 ± 0.03 0.56 ± 0.08* 0.55 ± 0.07* 0.65 ± 0.09* 
 Clamp 0.23 ± 0.03 0.38 ± 0.05§ 0.21 ± 0.04 0.61 ± 0.06* 0.34 ± 0.02§ 0.69 ± 0.12* 
Triglycerides (mg/dl) Basal 43.8 ± 7.3 32.7 ± 5.2 25.1 ± 4.5 97.7 ± 14.8* 117.1 ± 20.2* 89.3 ± 15.8* 
 Clamp 33.5 ± 9.1 21.1 ± 4.2 24.7 ± 3.8 96.1 ± 24.2 20.8 ± 3.8 64.6 ± 17.6 
Lactate (mmol/l) Basal 0.55 ± 0.04 0.57 ± 0.03 0.63 ± 0.04 1.10 ± 0.16* 1.21 ± 0.21* 1.30 ± 0.14* 
 Clamp 0.60 ± 0.03 13.20 ± 0.38 0.90 ± 0.06 1.03 ± 0.05 12.00 ± 0.53 1.51 ± 0.13 

 Data are means ± SE; n = 7–10 in each group. Rats were infused either with isotonic saline (control), AICAR (bolus, 100 mg/kg body wt; constant, 10 mg · kg−1 body wt · min−1) or insulin (4 mU · kg−1 min−1).

*

Significantly different from lean littermates undergoing the same treatment (P < 0.01);

significantly different from control and insulin-infused groups at same time interval (P < 0.01);

significantly different from control infused group at same time interval (P < 0.05);

§

clamp value is significantly different from basal (P < 0.05);

clamp value significantly different from basal (P < 0.01).

TABLE 2

Glucose infusion rate, endogenous glucose production, glucose disposal, and glycerol turnover data under basal state or during an isoglycemic clamp from lean and obese Zucker rats

Lean
Obese
ControlAICARInsulinControlAICARInsulin
Glucose infusion rates (μmol · kg−1 · min−1Clamp NA 74.3 ± 10.7 82.4 ± 3.5 NA 21.0 ± 2.2* 24.0 ± 4.4* 
Glucose disposal (μmol · kg−1 · min−1Clamp 40.5 ± 3.8 80.5 ± 5.7 103.5 ± 15.8 52.4 ± 4.2 46.4 ± 4.1* 45.7 ± 4.8* 
Endogenous glucose production (μmol · kg−1 · min−1Basal 37.3 ± 3.2 35.3 ± 2.7 45.1 ± 3.2 49.4 ± 4.4* 52.7 ± 2.8* 48.4 ± 5.2* 
Clamp 40.5 ± 3.8 6.2 ± 1.9§ 21.1 ± 5.3§ 53.3 ± 4.2 25.4 ± 2.3§ 21.7 ± 6.1§ 
Glycerol turnover (μmol · kg−1 · min−1Basal 20.6 ± 4.6 16.1 ± 2.0 17.8 ± 5.0 15.2 ± 2.2 23.1 ± 4.5 19.7 ± 2.3 
Clamp 16.4 ± 4.0 10.5 ± 1.9§ 14.0 ± 4.6§ 14.5 ± 1.4 13.2 ± 3.5§ 18.2 ± 2.0 
Lean
Obese
ControlAICARInsulinControlAICARInsulin
Glucose infusion rates (μmol · kg−1 · min−1Clamp NA 74.3 ± 10.7 82.4 ± 3.5 NA 21.0 ± 2.2* 24.0 ± 4.4* 
Glucose disposal (μmol · kg−1 · min−1Clamp 40.5 ± 3.8 80.5 ± 5.7 103.5 ± 15.8 52.4 ± 4.2 46.4 ± 4.1* 45.7 ± 4.8* 
Endogenous glucose production (μmol · kg−1 · min−1Basal 37.3 ± 3.2 35.3 ± 2.7 45.1 ± 3.2 49.4 ± 4.4* 52.7 ± 2.8* 48.4 ± 5.2* 
Clamp 40.5 ± 3.8 6.2 ± 1.9§ 21.1 ± 5.3§ 53.3 ± 4.2 25.4 ± 2.3§ 21.7 ± 6.1§ 
Glycerol turnover (μmol · kg−1 · min−1Basal 20.6 ± 4.6 16.1 ± 2.0 17.8 ± 5.0 15.2 ± 2.2 23.1 ± 4.5 19.7 ± 2.3 
Clamp 16.4 ± 4.0 10.5 ± 1.9§ 14.0 ± 4.6§ 14.5 ± 1.4 13.2 ± 3.5§ 18.2 ± 2.0 

 Data are means ± SE, n = 7–10 in each group. Rats were infused with isotonic saline (control), AICAR (bolus, 100 mg/kg body wt · constant, 10 mg · kg−1 body wt · min−1), or insulin (4 mU · kg−1 · min−1).

*

Significantly different from lean littermates undergoing the same treatment (P < 0.01);

significantly different from control group at same time interval (P < 0.01):

§

clamp value significantly different from basal (P < 0.05); NA, not applicable.

This work was supported by U.S. Public Health Service Grants R01 DK-40936 and P30-DK-45735 (G.I.S.) and HL-63811 (L.H.Y.). R.B. was supported by a postdoctoral fellowship from the Juvenile Diabetes Foundation International and a Mentor-Based Fellowship Award from the American Diabetes Association.

The authors would like to thank A. Kay, N. Barucci, S. Dufour, S. Hasan, J. Hu, D. Nuzzo, and C.L. Yu for their technical assistance.

1.
DeFronzo RA, Gunnarsson R, Bjorkman O, Olsson M, Wahren J: Effects of insulin on peripheral and splanchnic glucose metabolism in non–insulin dependent diabetes mellitus.
J Clin Invest
76
:
149
–155,
1985
2.
Campbell P, Mandarino L, Gerich J: Quantification of the relative impairment in actions of insulin on hepatic glucose production and peripheral glucose uptake in non–insulin dependent diabetes mellitus.
Metabolism
37
:
15
–22,
1988
3.
Butterfield WJH, Whichelow MJ: Peripheral glucose metabolism in control subjects and diabetic patients during glucose, glucose-insulin and insulin sensitivity tests.
Diabetologia
1
:
43
–53,
1965
4.
Shulman GI, Rothman DL, Jue T, Stein P, DeFronzo RA, Shulman RG: Quantitation of muscle glycogen synthesis in normal subjects and subjects with non–insulin dependent diabetes by 13C nuclear magnetic resonance spectroscopy.
N Engl J Med
322
:
223
–228,
1990
5.
Cline GW, Petersen KF, Krssak M, Shen J, Hundal RS, Trajanoski Z, Inzucchi S, Dresner A, Rothman DL, Shulman GI: Impaired glucose transport as a cause of decreased insulin-stimulated muscle.
N Engl J Med
341
:
240
–246,
1999
6.
Dohm GL, Tapscott EB, Pories WJ, Dabbs DJ, Flickinger EG, Meelheim D, Fushiki T, Atkinson SM, Elton CW, Caro JF: An in vitro human muscle preparation suitable for metabolic studies: decreased insulin stimulation of glucose transport in muscle from morbidly obese and diabetic subjects.
J Clin Invest
82
:
486
–494,
1988
7.
Brozinick JT Jr, Etgen GJ Jr, Yaspelkis BB III, Ivy JL: Glucose uptake and GLUT4 protein distribution in skeletal muscle of the obese Zucker rat.
Am J Physiol
267
:
R236
–R243,
1994
8.
Etgen GJ Jr, Wilson CM, Jensen J, Cushman SW, Ivy JL: Glucose transport and cell surface GLUT4 protein in skeletal muscle of the obese Zucker rat.
Am J Physiol
271: E294–E301,
1996
9.
Kennedy JW, Hirshman MF, Gervino EV, Ocel JV, Forse RA, Hoenig SJ, Aronson D, Goodyear LJ, Horton ES: Acute exercise induces GLUT4 translocation in skeletal muscle of normal human subjects and subjects with type 2 diabetes.
Diabetes
48
:
1192
–1197,
1999
10.
Price TB, Perseghin G, Duleba A, Chen W, Chase J, Rothman DL, Shulman RG, Shulman GI: NMR studies of muscle glycogen synthesis in insulin-resistant offspring of parents with non–insulin-dependent diabetes mellitus immediately after glycogen depleting exercise.
Proc Natl Acad Sci U S A
93
:
5329
–5334,
1996
11.
Merrill GF, Kurth EJ, Hardie DG, Winder WW: AICA riboside increases AMP-activated protein kinase, fatty acid oxidation, and glucose uptake in rat muscle.
Am J Physiol
273: E1107–E1112,
1997
12.
Hayashi T, Hirshman MF, Kurth EJ, Winder WW, Goodyear LJ: Evidence for 5′ AMP-activated protein kinase mediation of the effect of muscle contraction on glucose transport.
Diabetes
47
:
1369
–1373,
1998
13.
Bergeron R, Russell RR III, Young LH, Ren JM, Marccuci M, Lee A, Shulman GI: Effect of AMPK activation on muscle glucose metabolism in conscious rats.
Am J Physiol
276: E938–E944,
1999
14.
Russell RR III, Bergeron R, Shulman GI, Young LH: Translocation of myocardial GLUT4 and increased glucose uptake through activation of AMPK by AICAR.
Am J Physiol
277
:
H643
–H649,
1999
15.
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
16.
Bogardus C, Lillioja S, Howard BV, Reaven GM, Mott D: Relationships between insulin secretion, insulin action and fasting glucose concentration in non-dependent insulin subjects.
J Clin Invest
74
:
1238
–1246,
1984
17.
DeFronzo RA, Simonson D, Ferrannini E: Hepatic and peripheral insulin resistance: a common feature in non–insulin dependent and insulin-dependent diabetes.
Diabetologia
23
:
313
–319,
1982
18.
Firth R, Bell P, Rizza R: Insulin action in non–insulin dependent diabetes mellitus: the relationship between hepatic and extrahepatic insulin resistance and obesity.
Metabolism
36
:
1091
–1097,
1987
19.
Vincent MF, Marangos PJ, Gruber HE, Van den Berghe G: Inhibition by AICA riboside of gluconeogenesis in isolated rat hepatocytes.
Diabetes
40
:
1259
–1266,
1991
20.
Vincent MF, Erion MD, Gruber HE, Van den Berghe G: Hypoglycaemic effect of AICA riboside in mice.
Diabetologia
39
:
1148
–1155,
1996
21.
Golay A, Felber JP, Meyer HU, Curchod B, Maeder E, Jequier E: Study on lipid metabolism in obesity diabetes.
Metabolism
33
:
111
–116,
1984
22.
Chen Y-DI, Golay A, Swislocki ALM, Reaven GM: Resistance to insulin suppression of plasma free fatty acid concentrations and insulin stimulation of glucose uptake in non–insulin dependent diabetes mellitus.
J Clin Endocrinol Metab
64
:
7
–21,
1987
23.
DeFronzo RA: Pathogenesis of type 2 diabetes: metabolic and molecular implications for identifying diabetes genes.
Diabetes Rev
5
:
177
–269,
1987
24.
Roden M, Price TB, Perseghin G, Petersen KF, Rothman DL, Cline GW, Shulman GI: Mechanism of free fatty acid-induced insulin resistance in humans.
J Clin Invest
97
:
2859
–2865,
1996
25.
Dresner A, Laurent D, Marcucci M, Griffin ME, Dufour S, Cline GW, Slezak LA, Andersen DK, Hundal RS, Rothman DL, Petersen KF, Shulman GI: Effects of free fatty acids on glucose transport and IRS-1-associated phosphatidylinositol 3-kinase activity.
J Clin Invest
103
:
253
–259,
1999
26.
Boden G, Jadali F, White J, Liang Y, Mozzoli M, Chen X, Coleman E, Smith C: Effects of fat on insulin-stimulated carbohydrate metabolism in normal men.
J Clin Invest
88
:
960
–966,
1991
27.
Fulcher GR, Walker M, Catalano C, Agius L, Alberti KGM: Metabolic effects of suppression of nonesterified fatty acid levels with acipimox in obese NIDDM subjects.
Diabetes
41
:
1400
–1408,
1992
28.
Rebrin K, Steil GM, Mittelman SD, Bergman RN: Causal linkage between insulin suppression of lipolysis and suppression of liver glucose output in dogs.
J Clin Invest
98
:
741
–749,
1996
29.
Corton JM, Gillespie JG, Hawley SA, Hardie DG: 5-aminoimidazole-4-carboxamide ribonucleoside. A specific method for activating AMP-activated protein kinase in intact cells?
Eur J Biochem
229
:
558
–565,
1995
30.
Sullivan JE, Brocklehurst KJ, Marley AE, Carey F, Carling D, Beri RK: Inhibition of lipolysis and lipogenesis in isolated rat adipocytes with AICAR, a cell permeable activator of AMP-activated protein kinase.
FEBS Lett
353
:
33
–36,
1994
31.
Rossetti L, Smith D, Shulman GI, Papachristou D, DeFronzo RA: Correction of hyperglycemia with phlorizin normalizes tissue sensitivity to insulin in diabetic rats.
J Clin Invest
79
:
1510
–1515,
1987
32.
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
33.
Fujitaki JM, Sandoval TM, Lembach LA, Dixon R: Spectrophotometric determination of acadesine (AICA-riboside) in plasma using a diazotization coupling technique with N-(1-naphthyl)ethylenediamine.
J Biochem Biophys Methods
29
:
143
–148,
1994
34.
Previs SF, Cline GW, Shulman GI: A critical evaluation of mass isotopomer distribution analysis of gluconeogenesis in vivo.
Am J Physiol
277: E154–E160,
1999
35.
Nguyêñ VTB, Mossberg KA, Tewson T, Wong WH, Rowe RW, Coleman GC, Taegtmeyer H: Temporal analysis of myocardial glucose metabolism by 2-[18F]fluoro-2-deoxy-D-glucose.
Am J Physiol
259
:
H1022
–H1031,
1990
36.
Passonneau JV, Lauderdale VR: A comparison of three methods of glycogen measurement in tissues.
Anal Biochem
60
:
405
–412,
1974
37.
Sabina RL, Kernstine KH, Boyd RL, Holmes EW, Swain JL: Metabolism of 5-aminoimidazole-4-carboxamide riboside in cardiac and skeletal muscle.
J Biol Chem
257
:
10178
–10183,
1982
38.
Dale S, Wilson WA, Edelman AM, Hardie DG: Similar substrate recognition motifs for mammalian AMP-activated protein kinase, higher plant HMG-CoA reductase kinase-A, yeast SNF1, and mammalian calmodulin-dependent protein kinase I.
FEBS Lett
361
:
191
–195,
1995
39.
Maggs DG, Buchanan TA, Burant CF, Cline G, Gumbiner B, Hsueh WA, Inzucchi S, Kelley D, Nolan J, Olefsky JM, Polonsky KS, Silver D, Valiquett TR, Shulman GI: Metabolic effects of troglitazone monotherapy in type 2 diabetes mellitus: a randomized, double-blind, placebo-controlled trial.
Ann Intern Med
128
:
176
–185,
1998
40.
Dean DJ, Kaushik V, Lin T, Krowski T, Ruderman NR: Stimulation of muscle glucose transport by 5-aminoimidazole-4-carboxamide 1-β-ribofuranoside (AICAR) varies with fiber type (Abstract).
Diabetes
49 (Suppl. 1)
:
A193
,
2000
41.
Lee KU, Lee HK, Koh CS, Min HK: Artificial induction of intravascular lipolysis by lipid-heparin infusion leads to insulin resistance in man.
Diabetologia
31
:
285
–290,
1988
42.
Terrettaz J, Jeanrenaud B: Contribution of glycerol and alanine to basal hepatic glucose production in the genetically obese (fa/fa) rats.
Biochem J
270
:
803
–807,
1990
43.
Terrettaz J, Jeanrenaud B: In vivo hepatic and peripheral insulin resistance in genetically obese (fa/fa) rats.
Endocrinology
112
:
1346
–1351,
1983
44.
Shulman GI, Liljenquist JE, Williams PE, Lacy WW: Glucose disposal during insulinopenia in somatostatin-treated dogs: the roles of glucose and glucagon.
J Clin Invest
62
:
487
–491,
1978
45.
Rossetti L, Giaccari A, Barzilai N, Howard K, Sebel G, Hu M: Mechanism by which hyperglycemia inhibits hepatic glucose production in conscious rats: implications for the pathophysiology of fasting hyperglycemia in diabetes.
J Clin Invest
92
:
1126
–1134,
1993
46.
Staron RS, Kraemer WJ, Hikida RS, Fry AC, Murray JD, Campos GER: Fiber type composition of four hindlimb muscles of adult Fisher 344 rats.
Histochem Cell Biol
111
:
117
–123,
1999
47.
Armstrong RB, Phelps RO: Muscle fiber type composition of the rat hindlimb.
Am J Anat
171
:
259
–272,
1984
48.
Delp MD, Duan C: Composition and size of type I, IIA, IID/X, and IIB fibers and citrate synthase activity of rat muscle.
J Appl Physiol
80
:
261
–270,
1996
49.
Gardiner PF: Neuromuscular Aspects of Physical Activity. Champaign, IL, Human Kinetics,
2001
50.
Dolan PL, Boyd SG, Dohm GL: Differential effect of maturation on insulin- vs. contraction-stimulated glucose transport in obese Zucker rats.
Am J Physiol
268: E1154–E1160,
1995
51.
Brozinick JT Jr, Etgen GJ Jr, Yaspelkis BB, Ivy JL: Contraction-activated glucose uptake is normal in insulin resistant muscle of the obese Zucker rat.
J Appl Physiol
73
:
382
–387,
1992
52.
Wolfe RR: Radioactive and Stable Isotope Tracers in Biomedicine: Principles and Practice of Kinetic Analysis. New York, Wiley-Liss,
1992
53.
Muoio DM, Seefeld K, Witters LA, Coleman RA: AMP-activated kinase reciprocally regulates triacylglycerol synthesis and fatty acid oxidation in liver and muscle: evidence that sn-glycerol-3-phosphate acyltransferase is a novel target.
Biochem J
338
:
783
–791,
1999

Address correspondence and reprint requests to Dr. Gerald I. Shulman, Howard Hughes Medical Institute, Yale University School of Medicine, 295 Congress Ave., BCMM 254, New Haven, CT 06510. E-mail: gerald.shulman@yale.edu.

Received for publication 27 June 2000 and accepted in revised form 23 January 2001.

AICAR, 5-aminoimidazole-4-carboxamide-1-β-d-ribofuranoside; AMPK,AMP-activated protein kinase; ANOVA, analysis of variance; 2-DG, 2-deoxy-d-glucose; EGP, endogenous glucose production; GIR, glucose infusion rate; ZMP, monophosphorylated AICAR.