You are on page 1of 12

J Appl Physiol

91: 1017–1028, 2001.

invited review
Energy-sensing and signaling by AMP-activated protein
kinase in skeletal muscle

W. W. WINDER
Department of Zoology, Brigham Young University, Provo, Utah 84602

Winder, W. W. Energy-sensing and signaling by AMP-activated pro-


tein kinase in skeletal muscle. J Appl Physiol 91: 1017–1028, 2001.—
AMP-activated protein kinase (AMPK) is emerging as an important
energy-sensing/signaling system in skeletal muscle. This kinase is acti-
vated allosterically by 5⬘-AMP and inhibited allosterically by creatine
phosphate. Phosphorylation of AMPK by an upstream kinase, AMPK
kinase (also activated allosterically by 5⬘-AMP), results in activation. It
is activated in both rat and human muscle in response to muscle con-
traction, the extent of activation depending on work rate and muscle
glycogen concentration. AMPK can also be activated chemically in rest-
ing muscle with 5-aminoimidazole-4-carboxamide-riboside, which enters
the muscle and is phosphorylated to form ZMP, a nucleotide that mimics
the effect of 5⬘-AMP. Once activated, AMPK is hypothesized to phosphor-
ylate proteins involved in triggering fatty acid oxidation and glucose
uptake. Evidence is also accumulating for a role of AMPK in inducing
some of the adaptations to endurance training, including the increase in
muscle GLUT-4, hexokinase, uncoupling protein 3, and some of the
mitochondrial oxidative enzymes. It thus appears that AMPK has the
capability of monitoring intramuscular energy charge and then acutely
stimulating fat oxidation and glucose uptake to counteract the increased
rates of ATP utilization during muscle contraction. In addition, this
system may have the capability of enhancing capacity for ATP produc-
tion when the muscle is exposed to endurance training.
fatty acid oxidation; GLUT-4; glucose transport; malonyl-CoA; muscle
mitochondria

ONE OF THE FASCINATING PURSUITS in the field of exercise that trigger accumulation of increased quantities of
physiology involves elucidation of the intramuscular mitochondrial oxidative enzymes responsible for oxida-
signals that allow matching of the rate of ATP produc- tion of carbohydrate and fat? Recent studies have pro-
tion with the increased energy requirements during vided initial evidence that at least one of the energy-
exercise. What is the intramuscular change that trig- sensing/signaling proteins of the muscle is AMP-
gers an enhancement of glucose uptake and fatty acid activated protein kinase (AMPK) (16, 34–36, 99). This
oxidation during a single bout of exercise? How is the protein appears to be designed to monitor the energy
availability of energy stores monitored? When a muscle charge of the muscle fiber and to initiate responses
is exposed repeatedly to endurance exercise, an adap- that prevent high-energy phosphate depletion. This
tation occurs that allows enhancement of the rate of signaling system is currently being characterized with
ATP production, thus allowing increased endurance at respect to muscle phosphorylation targets. These pro-
higher intensities. During the course of an endurance tein targets of AMPK appear to be involved in both
training program, what are the intramuscular signals regulation of short-term metabolic responses and
chronic adaptation to exercise. In this review, the
AMPK system will be described in terms of its occur-
Address for reprint requests and other correspondence: W. W. rence in skeletal muscle, the mechanisms of activation,
Winder, Dept. of Zoology, Brigham Young Univ., Provo, UT 84602 processes influenced by AMPK, recently described tar-
(E-mail: william_winder@byu.edu). gets of the kinase, possible roles in Type 2 diabetes and
http://www.jap.org 8750-7587/01 $5.00 Copyright © 2001 the American Physiological Society 1017
1018 INVITED REVIEW

high relative abundance of the ␥3-isoform mRNA in


muscle (15). Because Western blots were performed
only on postnuclear supernatants, it is possible that
AMPK containing the ␥3-isoform sedimented with the
cellular particulate fraction. The ␥-subunits appear to
play a role in determining sensitivity to AMP (15).
Activity of AMPK can be measured on ammonium
sulfate precipitates, polyethylene glycol precipitates,
or immunoprecipitates of muscle homogenates (34, 35,
91, 98). The assay measures incorporation of 32P-la-
beled phosphate from [32P]ATP into an artificial pep-
tide substrate resembling an AMPK phosphorylation
target site on acetyl-CoA carboxylase (ACC). Most in-
vestigators have used a 15-amino acid peptide called
“SAMS” peptide as substrate. It should be remembered
that an increase in AMPK activity measured in these
assays represents increases due to phosphorylation of
the enzyme. As will be discussed in detail later, 5⬘-
AMP (AMP) can allosterically activate AMPK and cre-
atine phosphate (CP) can allosterically inhibit AMPK.
Fig. 1. Model of subunit association of AMP-activated protein kinase The phosphorylated fraction of the enzyme could be
(AMPK) in the absence and presence of 5⬘-AMP (see Ref. 15). Both activated allosterically in the intact muscle, and this
the ␣- and the ␥-subunits participate in the allosteric activation of increased AMPK activity would not show up in the
AMPK by AMP. Both the catalytic domain of this kinase and the
phosphorylation site [threonine 172 (T172)] for AMPK kinase assay, since AMP and CP would for the most part be
(AMPKK) are located on the ␣-subunit. discarded in the supernatant after collection of the
precipitated fraction. In the assay mix, AMP is added
in maximally effective activating concentrations.
obesity, and feasibility of manipulation of the kinase in MECHANISMS OF MUSCLE AMPK ACTIVATION
treatment of obesity and diabetes.
In addition to the glycolytic pathway and oxidative
STRUCTURE, DISTRIBUTION, AND QUANTITATION phosphorylation, two enzymes are responsible for
OF AMPK maintaining relatively stable ATP concentrations in
the muscle as muscle begins to contract and utilize
AMPK is a protein consisting of three subunits des- ATP at increased rates (Fig. 2). Myokinase (adenylate
ignated ␣, ␤, and ␥ (34, 35) (Fig. 1). The approximate kinase) transfers a phosphate from one ADP to an-
molecular masses are 63 and 38 kDa for the ␣- and other, resulting in the products AMP and ATP. An
␤-subunits, respectively (34, 35, 99). The ␥-subunit increase in free ADP concentration resulting from in-
molecular mass varies considerably among isoforms
(15). The ␣-subunit is the catalytic subunit containing
the kinase domain, which transfers a phosphate from
ATP to the target protein (34, 35). The ␤- and ␥-sub-
units are considered regulatory components (15, 34,
35). All three subunits are required for expression of
full activity (Fig. 1) (15, 24, 104). Each subunit has two
or three isoforms, designated ␣l, ␣2, ␤1, ␤2, ␥1, ␥2, and
␥3 (9, 15, 35). Information is available on tissue distri-
bution based on immunoprecipitation studies (15,000 g
supernatant) using antibodies to all three subunits and
in combination with AMPK activity measurements
(15). Both ␣1- and ␣2-isoforms are found in skeletal
muscle, but the ␣3-isoform is more abundant, repre-
senting 80% of the total AMPK activity (15, 82, 83, 88,
91). The ␣2-isoform is more sensitive to AMP (81). Both
␤1 and ␤2 are found in extensor digitorum longus (EDL;
predominantly type II fibers), but only ␤1 appears in
the soleus (type I fibers) (13). Data on distribution of all
subunits in the different muscle types are not currently
available. On the basis of immunoprecipitation stud-
ies, the ␥1-isoform predominates in skeletal muscle Fig. 2. Creatine phosphate (CP) decreases, creatine (C) increases,
(muscle type unspecified), but these data are not con- and AMP increases in muscle during exercise as a result of actions of
sistent with Northern blot analyses, which indicate creatine phosphokinase (CPK) and myokinase (adenylate kinase).

J Appl Physiol • VOL 91 • SEPTEMBER 2001 • www.jap.org


INVITED REVIEW 1019

creased rates of ATP utilization allows this reaction to AICAR has since been determined to be effective in
proceed. This reaction contributes to buffering of ex- activating AMPK in perfused hindlimb muscle, incu-
pected contraction-induced ATP concentration changes bated muscle of the rat, cultured mouse muscle cells,
in the muscle. As muscle contracts, estimated AMP and muscle of injected rats in vivo (29, 38, 50, 60, 101).
concentration increases as a function of work rate (23).
Another enzyme responsible for buffering ATP changes CONTRACTION-INDUCED ACTIVATION OF AMPK
is creatine phosphokinase. During the course of con- IN MUSCLE
traction, phosphate may be transferred from CP to
ADP, forming ATP. Thus CP decreases and AMP in- The first indication that AMPK could be involved in
creases in the muscle fiber during contraction. regulation of muscle metabolism during exercise came
AMPK could be considered a protein that monitors from a study reported in 1996 (98). Rats were run on a
the energy state of the muscle cell and triggers meta- treadmill at 21 m/min up a 15% grade for periods up to
bolic processes, which when activated are designed to 30 min. The deep red region of the quadriceps muscle
restore high-energy phosphate concentrations. This was quickly removed and frozen for AMPK isolation
enzyme is regulated by both allosteric and covalent and analyses. AMPK activity was found to increase
mechanisms (Figs. 1 and 3). AMPK is activated allos- two- to threefold within 5 min of the beginning of
terically by an increase in AMP concentration (12, 16, exercise and remained elevated for as long as the rat
37). In addition, it is inhibited by CP and ATP (16, 19, continued to run. In rats running on the treadmill, the
37, 73). The increase in AMP and the decrease in CP as increase in AMPK activity was found to be dependent
muscle contracts result in allosteric activation of on work rate (75), and the activity remained elevated
AMPK. AMP binds to AMPK, making it a better sub- for several minutes following 5- and 30-min exercise
strate for the upstream kinase, AMPK kinase bouts (74). When rat gastrocnemius muscle was stim-
(AMPKK) (37). AMP also directly activates AMPKK, ulated in situ at a frequency of once per second, 10-ms
which phosphorylates the ␣-subunit of AMPK at thre- duration, the estimated free AMP concentration in-
onine 172, resulting in activation (37). AMP binding creased, CP decreased, and AMPK activity increased
also makes AMPK a poor substrate for protein phos- (53). Interestingly, the decline in ACC activity (phos-
phatase 2C, the phosphatase that appears to be re- phorylation target for AMPK, a reporter for AMPK
sponsible for removal of the phosphate at threonine activity) correlated more closely with the decline in CP
172 and inactivation of the enzyme (19). CP apparently than with the rise in free AMP or with the increase in
has no effect on the phosphorylation of AMPK; its measurable AMPK activity. This illustrates the impor-
inhibitory effects are entirely allosteric (see Ref. 99). tance of the allosteric mechanisms. AMPK activity also
AMPKK and AMPK can also be activated artificially increased in incubated epitrochlearis muscles in re-
by use of the chemical 5-aminoimidazole-4-carboxam- sponse to contraction and AICAR treatment (38)
ide-riboside (AICAR) (17, 41, 42, 60, 79, 87). AICAR Vavvas et al. (91) published the first information on
was reported to be taken up by the cells and phosphor- the activation of specific isoforms of AMPK in response
ylated to form an AMP analog, termed ZMP, which to muscle contraction. The gastrocnemius muscle was
then activated AMPKK and AMPK similarly to AMP. stimulated via the sciatic nerve (5 pulses/s, 100-ms
trains, 50 Hz, 10-ms duration) to contract in situ for
periods up to 5 min. The ␣1- and ␣2-isoforms were
precipitated from muscle homogenates using specific
antisera. The ␣2-isoform was maximally activated (3-
to 4-fold) within 30 s of the beginning of stimulation,
but no significant increase occurred in the ␣1-isoform
(91). In isolated epitrochlearis muscle, both the ␣1- and
␣2-isoforms were reported to be activated in response
to a number of stimuli that decreased CP and glycogen,
including contraction, hypoxia, dinitrophenol, rote-
none, and sorbitol (hyperosmotic stress) (29, 39). Only
the ␣2-isoform was activated in epitrochlearis of rats
running on the treadmill (65).
Studies published during the last year indicate that
the glycogen content of muscle may modulate the
AMPK response to contraction (21, 55). Derave et al.
(21) subjected rats to 2 h of swimming, followed by
feeding with a 100% fat diet overnight or with normal
chow ⫹ 20% glucose in drinking water. The two proto-
Fig. 3. Activation of AMPK in muscle in response to muscle contrac- cols produced rats with low and high muscle glycogen,
tion. AMPK is activated as a result of allosteric stimulatory effects of respectively. Hindlimbs of these rats were perfused
AMP, the removal of allosteric inhibitory effects by CP, and the
phosphorylation by AMPK kinase in the T172 position. ⫺, CP allo-
with cell-free medium. After a 5-min washout period,
sterically inhibits AMPK; ⫹, AMP allosterically activates both one sciatic nerve was stimulated for 10 min with
AMPKK and AMPK. 100-ms trains (100 Hz) at 2-s intervals. In fast-twitch
J Appl Physiol • VOL 91 • SEPTEMBER 2001 • www.jap.org
1020 INVITED REVIEW

white fibers from the gastrocnemius, AMPK activity of the ␣2-isoform increases. In 30-s maximal sprints,
measured in postnuclear supernatants of stimulated activities of both isoforms increase. The presence of
muscles was increased 1.8-fold over resting muscles in high glycogen in muscle appears to attenuate the in-
the high-glycogen group compared with 4.6-fold over crease in AMPK during prolonged exercise.
resting muscles in the low-glycogen group. In slow-
REGULATION OF MUSCLE FATTY ACID OXIDATION
twitch soleus muscle, a contraction-stimulated activa-
BY AMPK
tion of AMPK occurred in the muscles with low glyco-
gen but not in those with high glycogen. The activity of The rate of fatty acid oxidation in muscle is con-
ACC declined during contraction, however, indicating trolled in part at the level of carnitine palmitoyltrans-
that the AMPK may have been activated allosterically. ferase-1 (CPT-1) (Fig. 4). This enzyme is subject to
This would not have been detected in the final AMPK inhibition by malonyl-CoA (59, 78). Malonyl-CoA de-
assay procedure. Studies using the incubated epitroch- clines rapidly in the red region of the quadriceps mus-
learis muscle (fast twitch) have also demonstrated an cle of rats during exercise, presumably relieving inhi-
attenuation of the AMPK response in muscles with bition and allowing fatty acid oxidation to increase as
high glycogen (55). It is unclear whether there is a long-chain acyl-CoA becomes available (74, 75, 97).
direct effect of glycogen on AMPK activity or whether Malonyl-CoA is synthesized by the muscle isoform of
the attenuation is due to blunting of changes in ATP, ACC, which has a molecular mass of ⬃272 kDa (89).
AMP, and CP. ATP and CP responses to stimulation The ACC isoform isolated from rat hindlimb muscle
did not appear to be significantly altered in the soleus can be phosphorylated in vitro by both cAMP-depen-
regardless of glycogen content, although the time dent protein kinase (PKA) and by AMPK, but only
course of changes were not studied (21). phosphorylation by AMPK inactivates the enzyme,
Recent studies have clearly demonstrated AMPK to thus differing from principal isoforms in both liver and
be activated in human muscle during exercise. heart, which respond to phosphorylation by PKA with
Wojtaszewski et al. (103) exercised (cycle ergometer) a decline in activity (98, 102).
human subjects at 50% of maximal oxygen consump- The muscle isoform of ACC is activated by citrate
tion for 90 min or at 55 min at 75% followed by 5 min over a range of 0–20 mM (89, 91, 98). The shape of this
at 90% of maximal oxygen consumption. They reported activation curve is dependent on the phosphorylation
no change in AMPK activity in needle biopsies from the state of the enzyme (91, 98). Phosphorylation by AMPK
vastus lateralis in response to the lower intensity work causes the curve to shift to the right, and the maximal
bout but a three- to fourfold increase in activity of the activation at high-citrate concentrations (Vmax) is re-
␣2-isoform in response to the higher intensity work duced. The activation constant (Ka) for citrate in-
bout. The activity of the ␣1-isoform was not changed in creases as a consequence of phosphorylation. Although
response to either work rate. The activity of the ␣2- the determination of the citrate activation curve (ac-
isoform had returned to baseline by 3 h postexercise. tivity of ACC with citrate varied between 0 and 20 mM)
Fugii et al. (30) earlier the same year reported no is important in characterizing ACC and its phosphor-
change in either isoform of AMPK in the vastus late- ylation state, muscle levels of citrate are in the range of
ralis of human subjects in response to 20 min of work 0.2 mM. The phosphorylated ACC is essentially inac-
at 50% of maximal oxygen consumption, whereas ac- tive at physiological citrate concentrations. Neverthe-
tivity of the ␣2-isoform increased after 20 and 60 min of less, measurement of the citrate activation curve for
work at 70% of maximal oxygen consumption on the ACC isolated from muscle is a convenient way to assess
cycle ergometer. Muscle CP and glycogen were signif-
icantly decreased in response to subjects working at
70% but not at 50% of maximal oxygen consumption.
One recent report indicates that the activity of both ␣1-
and ␣2-isoforms may increase with very high-intensity
work. Chen et al. (14) had subjects do a 30-s sprint
(peak power ⫽ 834 W) on a cycle ergometer and found
the activity of both isoforms to be increased. Richter et
al. (76a) recently obtained data from human subjects
indicating that elevated glycogen prevents the increase
in AMPK activity in response to working at 70% of
maximal oxygen consumption for 60 min, confirming
the relationship between glycogen content and AMPK
activity first demonstrated in animal studies (personal
communication).
In summary, from the currently available data, it
appears that the muscle AMPK increases in response
to muscle contraction. The increase is dependent on Fig. 4. Malonyl-CoA inhibits fatty acid oxidation by inhibiting car-
work rate. In prolonged exercise at work rates exceed- nitine palmitoyltransferase-1 (CPT-1), the enzyme necessary for
ing 70% of maximal oxygen consumption, only activity entrance of long-chain fatty acids into mitochondria for oxidation.

J Appl Physiol • VOL 91 • SEPTEMBER 2001 • www.jap.org


INVITED REVIEW 1021

whether AMPK is activated (either allosterically or by EDL by incubation with AICAR. Phosphorylation of
phosphorylation). ACC activity thus serves as a re- purified MCD with purified AMPK in vitro has not yet
porter for in vivo activation of AMPK. been reported. The AICAR data provide initial evi-
ACC is inactivated concurrently with AMPK activa- dence that AMPK regulates not only synthesis but also
tion in muscle of rats running on the treadmill (75, 98) degradation of malonyl-CoA (Fig. 5).
and in muscle stimulated electrically (53, 91). It is also The role of the AMPK system in control of fatty acid
clear that ACC can be inactivated early during the oxidation in human muscle is less well defined. All
time course of prolonged submaximal muscle contrac- components of the signaling system are present in
tion with no detectable increase in AMPK activity (53), human muscle, including AMPK, ACC, and malonyl-
implying that the AMPK may be activated allosteri- CoA (14, 20). Evidence has been presented above for
cally or that other mechanisms exist for inactivation of activation of AMPK in muscle of human subjects dur-
ACC. When AMPK is activated using AICAR in resting ing exercise. Data indicate that ACC is phosphorylated
perfused muscle of rats, ACC activity declines, malo- (14) and inactivated (14, 20) in muscle during exercise.
nyl-CoA declines, and the rate of radiolabeled palmi- A small decline in malonyl-CoA has been reported by
tate oxidation increases (60, 61). These effects are also one group (20) but not so in the majority of studies
observed in incubated soleus (3) and in cultured C2C12 (68–70). The concentration of malonyl-CoA is much
myocytes and myotubes (63). The rate of palmitate lower (⬃0.1) in human muscle than in rat muscle,
oxidation is dependent on palmitate concentration, but making it more difficult to measure, particularly in
inclusion of AICAR in the medium increased the rate of small muscle biopsy samples (7, 20, 68–70, 77). It is
oxidation regardless of palmitate concentration (61). also more difficult to obtain muscle samples that are
The processes regulating the substrate oxidation mix homogeneous with respect to fiber type, as can be done
(carbohydrate vs. fat) in muscle are poorly understood. in rat studies. In one of the first rat studies, malonyl-
Although a number of factors may be important in CoA was found to decline rapidly in type IIA fibers but
determining this mix (including availability of glucose, not in IIB fibers (97). If a mixture of contracting and
concentration of insulin, muscle glycogen, availability noncontracting fibers was sampled in the biopsy of
of fatty acids, availability of carnitine and CoA, and so human muscle, any change in contracting fibers would
forth; see Ref. 94), it is likely that AMPK plays a role. be diluted. One possibility is that ACC in human mus-
Addition of insulin to medium during rat hindlimb cle is arranged in close proximity to CPT-1 so that local
perfusion increases glucose uptake and tends to reduce concentration of malonyl-CoA is responsible for regu-
the rate of palmitate oxidation in perfused muscles lation. In this case, total tissue malonyl-CoA may not
(100). If AICAR is also included in the medium to change significantly, yet the concentration near CPT-1
activate AMPK, inactivate ACC, and reduce malonyl- could vary markedly depending on the ACC phosphor-
CoA, the presence of insulin, even at very high concen- ylation state. This is entirely speculative, and it is
trations, does not prevent an acceleration of the rate of possible that malonyl-CoA-independent mechanisms
palmitate oxidation. With the use of this method, a (see Refs. 68–70, 84, 94) are more important in control
wide range of malonyl-CoA concentrations were gener- of fatty acid oxidation in human muscle. The fact
ated in perfused rat hindlimbs. A curvilinear relation- remains, however, that ACC is present in the muscle.
ship between malonyl-CoA and palmitate oxidation It is phosphorylated at the AMPK target site, and its
was observed, with a half-maximal effect inhibition activity decreases during exercise.
seen at ⬃0.6 nmol malonyl-CoA/g of muscle. This is
much higher than would have been predicted from data
generated from malonyl-CoA inhibition curves in iso-
lated mitochondria (59, 78).
Although a reduction of ACC activity would be ex-
pected to reduce the rate of malonyl-CoA synthesis
during muscle contraction, there remained the ques-
tion of what actually caused the reduction in malonyl-
CoA concentration. This question was approached by
Saha et al. (80), who studied muscle responses of ma-
lonyl-CoA decarboxylase (MCD) activity to electrical
stimulation and AICAR treatment. MCD is thought to
catalyze the major pathway for degradation of malo-
nyl-CoA in muscle, removing one carbon and producing
Fig. 5. Malonyl-CoA content of muscle is controlled by the relative
acetyl-CoA. MCD activity increased approximately rate of synthesis by acetyl-CoA carboxylase (ACC) and the rate of
twofold in the gastrocnemius muscle in response to degradation by malonyl-CoA decarboxylase (MCD). AMPK phos-
electrical stimulation. Treatment of muscle extracts phorylates (P) and inactivates ACC. On the basis of effects of 5-ami-
noimidazole-4-carboxamide-riboside (AICAR) on activation of MCD
with protein phosphatase 2A reversed this stimulation in incubated extensor digitorum longus, AMPK is hypothesized to
of activity, providing evidence that phosphorylation is phosphorylate and activate MCD (Ref. 80). These changes could
involved. In addition, MCD could be activated in the result in a decline in malonyl-CoA (rat muscle studies).

J Appl Physiol • VOL 91 • SEPTEMBER 2001 • www.jap.org


1022 INVITED REVIEW

ROLE IN GLUCOSE UPTAKE contraction effect (38). Wortmannin, an inhibitor of


phosphatidylinositol 3-kinase, inhibited insulin-stimu-
During the first hindlimb perfusion studies on the
role of AMPK in regulation of fatty acid oxidation in lation of 3-MG transport but not that caused by con-
muscle, it was noted that AICAR not only activated traction or AICAR. Stimulatory effects of AICAR were
AMPK and increased palmitate oxidation but also it additive with those of insulin but not with those of
stimulated an increase in glucose uptake (60). It was contraction. In perfused hindlimbs, AICAR was found
hypothesized at the time that AMPK activated by mus- to stimulate glucose uptake in the absence of insulin
cle contraction stimulated both an increase in fatty and to cause GLUT-4 translocation to the sarcolemmal
acid oxidation and an increase in glucose uptake to fraction from the microvesicle fraction (56). Infusion of
meet the energy demands of working muscle (60). AICAR into live rats resulted in increased labeled
The insulin-like effect of muscle contraction has been hexose uptake into all three muscle fiber types (8).
extensively studied (31, 32, 40, 47). The insulin-sensi- More recent studies have demonstrated that several
tive glucose transporter, GLUT-4, resides in two loca- factors that decrease CP and glycogen in incubated
tions in the muscle (31, 32, 40, 47). In the basal state epitrochlearis (contraction, hypoxia, dinitrophenol, ro-
(low insulin), the GLUT-4 transporters reside in mi- tenone, and sorbitol) also activate AMPK and stimu-
crovesicles beneath the sarcolemma and T-tubule late glucose transport (29, 39). These data provide
membranes. In response to insulin or contraction, evidence that AMPK activation mediates the effect of
these transporters are translocated and inserted into contraction on stimulation of glucose transport.
membranes of the T tubules and sarcolemma, thus Two studies (29, 39) have now demonstrated that
increasing the capacity for moving glucose into the both the contraction-induced increase in AMPK activ-
muscle fiber (Fig. 6). The signaling pathways for the ity and the contraction stimulation of glucose uptake
two systems differ. The insulin pathway involves occu- are decreased in muscles composed of type II fibers
pied receptor-catalyzed phosphorylation of insulin re- when glycogen content of the muscle is elevated.
ceptor substrate 1 followed by activation of phosphati- Kawanaka et al. (55) hypothesized that the smaller
dylinositol 3-kinase, whereas the contraction pathway degree of AMPK activation was responsible for the
does not involve either of these signaling proteins (31, decreased contraction stimulated translocation of
32, 40, 47, 105). The maximal effect of contraction on GLUT-4 in glycogen supercompensated epitrochlearis
translocation of GLUT-4 is additive with the effects of muscles. Derave et al. (21) reported a complete absence
high insulin. Although careful and extensive efforts
of AMPK activation in perfused supercompensated so-
have been expended by many laboratories in attempts
leus muscles (composed of type I fibers) in response to
to elucidate initial steps in the contraction-mediated
electrical stimulation, but glucose transport was not
GLUT-4 translocation, the signaling steps have been
elusive. The possibility that AMPK activation may be diminished. On the basis of this apparent dissociation
involved has therefore generated considerable interest. between the two processes in the soleus, the authors
Using the incubated epitrochlearis, it was deter- concluded that AMPK activation is not an obligatory
mined that AICAR would increase 3-methylglucose signaling step in contraction activation of glucose
(3-MG) transport with characteristics similar to the transport. There was some evidence of allosteric acti-
vation of AMPK in the soleus, however, because ACC
activity at 0.2 mM citrate was lower after stimulation.
Nevertheless, it is entirely possible that redundant
pathways are present for the insulin-like effect of con-
traction and that the AMPK-independent pathway
predominates in type I fibers.
Part of the difficulty in identifying AMPK-dependent
processes in the muscle has been due to the lack of
specific inhibitors of AMPK. In isolated epitrochlearis,
iodotubercidin, Ara-A, and 8-bromo-AMP inhibited ef-
fects of AICAR on 3-MG uptake and AMPK ␣2 activa-
tion but had no or little effect on contraction-stimu-
lated AMPK activity and 3-MG uptake (65). It will be
important to identify more specific inhibitors of AMPK
to clearly link control of cellular processes to the AMPK
signaling pathway.
Another approach to linking AMPK activation with
Fig. 6. Both insulin and muscle contraction trigger translocation of contraction-stimulated glucose uptake is that of pro-
GLUT-4 to the membranes of the sarcolemma and T tubules, result- duction of transgenic mice lacking functional forms of
ing in enhancement of glucose uptake. These processes occur by AMPK. This approach is complicated by the occurrence
different signaling pathways. In the case of the contraction-initiated
pathway, other signals besides AMPK activation are possible. PI3K,
of multiple isoforms of the three subunits of AMPK. Mu
phosphatidylinositol 3-kinase; IR, insulin receptor; IRS-1, insulin and Birnbaum (62) found that muscle from mice ex-
receptor substrate 1. pressing a kinase-inactive ␣2-catalytic subunit of
J Appl Physiol • VOL 91 • SEPTEMBER 2001 • www.jap.org
INVITED REVIEW 1023

AMPK resulted in a depression of endogenous AMPK ROLE IN CONTROL OF GLUT-4 AND HEXOKINASE
activity in both the resting and contracting muscle.
One of the benefits of performing regular endurance
Stimulation of hexose transport was completely
exercise is that muscles tend to become more sensitive
blocked in AICAR-treated and hypoxic muscles but
to the action of insulin (4, 22, 26, 47–49, 54, 67, 76, 90,
was only partially blocked in electrically stimulated
93). This is thought to be due in part to the increased
contracting muscles. The authors concluded that a
amounts of GLUT-4 that accumulate in muscle in re-
redundant AMPK-independent pathway must in part sponse to repetitive bouts of exercise over a several-day
mediate the effect of contraction on glucose uptake in period (26, 52, 54, 76). With daily exposure of the
muscle. It is unclear whether residual endogenous muscle to exercise, there is an increase in the amount
AMPK could have been activated allosterically to me- of GLUT-4 mRNA that accumulates in muscle after
diate contraction-stimulated glucose transport in these each bout (66, 76). The GLUT-4 gene has a regulatory
transgenic mice. No mention was made of measure- region that appears to have what is termed an “exer-
ment of ACC activity as a reporter of possible allosteric cise response element” that mediates the effect of reg-
activation. ular bouts of contraction on GLUT-4 transcription (27,
Not all effects of AMPK on glucose transport may be 66). Although the presence of this regulatory element
due to effects on translocation of GLUT-4. Abbud et al. has been demonstrated, it is unclear what transcrip-
(1) studied effects of AICAR on glucose transport in tion factor(s) binds to this region and how this factor
C2C12 myoblasts, which rely on GLUT-1 to transport (or factors) is influenced by the contractile process.
glucose. In these cells, a two- to threefold increase in AMPK appears to be a candidate for controlling the
glucose transport was observed in response to incuba- putative transcription factors, thereby mediating
tion with AICAR. Thus enhancement of the activity of adaptive responses to exercise. AMPK would be ex-
other isoforms of glucose transporters may also help pected to be activated with each bout of exercise. Could
explain the effect of AICAR on glucose transport in it then phosphorylate a target transcription factor,
muscle. which would then be more effective in turning on the
Some attention has been given to identifying the GLUT-4 gene? Another possibility is that activated
specific target protein(s) for mediating the effect of AMPK could trigger synthesis of relevant transcription
AMPK on GLUT-4 translocation and glucose trans- factors? AMPK has previously been reported to be
port. Previous studies have demonstrated a possible responsible for negative regulation of several genes
role of nitric oxide (NO) in contraction stimulation of involved in gluconeogenesis and fatty acid metabolism
glucose transport (5, 6). Fryer et al. (29) recently re- in liver and in pancreatic islet ␤-cell metabolism (18,
ported nitric oxide synthase (NOS) activity to be in- 28, 57, 58).
creased along with glucose transport in H-2K myo- If AMPK activation is involved in inducing an in-
tubes in response to activation of AMPK with AICAR. crease in GLUT-4 transcription in response to training,
NOS inhibitors blocked the increase in glucose trans- it should be possible to mimic the effect of training with
port after AMPK activation. Both the neuronal NOS chronic chemical activation of AMPK in resting mus-
and endothelial NOS isoforms can be phosphorylated cle. Injection of a high dose (1 mg/g body wt) of AICAR
by AMPK in vitro. NO activates guanylate cyclase, into resting rats was found to produce an increase in
which then catalyzes synthesis of cGMP. Treatment of muscle ZMP, induce activation of AMPK, and induce a
these cells with an inhibitor of guanylate cyclase pre- decrease in ACC activity and in malonyl-CoA content
vents the increase in glucose transport wrought by (50). The AMPK was activated in muscle for at least 2 h
AICAR. In soleus and EDL muscle strips, the stimula- in response to a single injection. When muscle AMPK
tory effect of AICAR on glucose transport was also was activated daily with AICAR in resting rats for a
blocked by the NOS inhibitor, NG-nitro-L-arginine 5-day period, the total amount of GLUT-4 in epitroch-
methyl ester. These results provide evidence that an learis and gastrocnemius muscle was increased by
obligatory target of AMPK in inducing an increase in 100% and 60%, respectively. Hexokinase activity was
glucose transport is NOS. also significantly increased (⬃2.5-fold) in both muscles.
A more recent study using isolated EDL and soleus The AICAR-treated rats exhibited glycogen supercom-
muscles provides evidence against NO being a media- pensation the morning after injection, similarly to
tor of the contraction-induced activation of glucose trained rats after an overnight resting period. This
uptake (43). Incubation of muscles with sodium nitro- increase in GLUT-4 was maintained for at least 4 wk
prusside (SNP ⫽ NO donor) resulted in stimulation of with daily treatment of rats with AICAR (101). The
2-deoxy-D-glucose uptake. SNP was also reported to increase in muscle GLUT-4 induced by AICAR injec-
activate ␣1- but not the ␣2-isoform of AMPK. Inclusion tion into rats has recently been confirmed and ex-
of a NOS inhibitor in the incubation medium failed to tended (11, 107). The increase in GLUT-4 protein is
block the effect of contraction on 3-MG transport. The accompanied by increases in mRNA (11, 107) and in-
authors proposed that NO activates glucose uptake by creases in glucose uptake of isolated epitrochlearis in
a pathway separate from the insulin- and contraction- response to insulin (11). Ojuka et al. (71) reported that
induced signaling pathways. The reason for the appar- incubation of epitrochlearis muscles with 0.5 mM
ently contradicting results in these two studies is un- AICAR for 18 h induced an increase in both GLUT-4
clear at this time. protein and hexokinase activity, implying that the in-
J Appl Physiol • VOL 91 • SEPTEMBER 2001 • www.jap.org
1024 INVITED REVIEW

crease in these proteins in muscle was due to direct genes has also been reported for human muscle in the
effects of AMPK activation and not to some indirect or postexercise period (72).
humoral change (92, 107). It should be clear that AICAR injection does not
Transgenic mice have been produced with regions of mimic all changes in high-energy phosphate concentra-
the 5⬘-flanking region of the human GLUT-4 gene tions that occur during muscle contraction. ATP and
controlling expression of the bacterial chloramphenicol CP are not markedly influenced. ZMP increases to
acetyltransferase (CAT) reporter gene. These trans- mimic the effects of AMP. ZTP was also found to
genics have been utilized to determine which region of increase, and this change may interfere allosterically
the promoter is responsible for the AICAR induction of with full AMPK activation (101). These differences
mRNA synthesis (107). The transgenic mice with ei- may explain why changes in mitochondrial enzymes
ther 1,154 or 895 bp of the promoter responded to were not of the magnitude seen with daily 2-h bouts of
AICAR with an increase in CAT mRNA expression, but exercise (44, 101). The decline in CP may be essential
the transgenic with 730 bp was not responsive. This for full manifestation of the effect of contraction on
implies that the region between ⫺730 and ⫺895 bp these adaptations. Nevertheless, these data suggest
from the GLUT-4 transcription start site contains an that the regular activation of AMPK during daily bouts
element essential for induction of GLUT-4 mRNA syn- of exercise may be responsible for inducing increases in
thesis by AMPK activation. Evidence was also ob- some, but not all, mitochondrial oxidative enzymes.
tained from gel-shift assays for possible roles of The mitochondrial uncoupling protein 3 (UCP3) has
MEF-2A and/or MEF-2D transcription factors in this also recently been discovered to be under the influence
enhancement of transcription (107). A recent prelimi- of AMPK (108). Treatment of incubated muscles with
nary report indicates hexokinase II transcription (us- AICAR resulted in an increased UCP3 mRNA expres-
ing nuclear run-on analysis) can be enhanced 6.5- and sion within 30 min. These findings are supported by
10-fold, respectively, in red and white gastrocnemius recent work indicating a marked stimulation of skele-
locally infused with AICAR (86). tal muscle UCP3 gene transcription after in vivo infu-
sion of AICAR into muscle of conscious rats (86). Al-
ROLE IN CONTROL OF MITOCHONDRIAL PROTEINS though the function of UCP3 in muscle is not entirely
clear, if it is indeed functioning as an uncoupler of
When muscle is exposed to repeated bouts of exercise oxidative phosphorylation, more energy substrate
over several days and weeks, adaptations occur that would be consumed for producing a given amount of
result in an increase in the maximal capacity to gen- ATP when mitochondrial UCP3 is increased. Is it pos-
erate ATP. These adaptations include increases in mi- sible that transient increases in UCP3 after exercise
tochondrial enzymes of the citric acid cycle and fatty bouts during training could contribute to an increase in
acid oxidation, proteins of the electron transport chain, oxygen consumption and caloric expenditure during
and myoglobin (10, 25, 44–46, 51). Although consider- each postexercise period? Teleologically speaking, it is
able work has been done in characterizing these adap- advantageous for those involved in regular endurance
tations, the intramuscular signaling mechanisms re- exercise to have a lower body mass. Could a transient
sponsible for the accumulation of these proteins are not increase in UCP3 after daily bouts of exercise contrib-
clearly defined (see Ref. 51). Recently reported data ute to the weight loss and low-fat content of those who
provide evidence of AMPK involvement in inducing train by running, swimming, or cycling many hours per
these adaptations (101). When muscle AMPK is acti- week?
vated by AICAR injection into nonexercising rats for
periods up to 4 wk, citrate synthase, succinate dehy- CLINICAL CONSIDERATIONS
drogenase, malate dehydrogenase, cytochrome c,
␦-aminolevulinate synthetase, and hexokinase in- Two very exciting reports this year emphasize the
crease in a fiber-type-specific manner. Significant in- importance of continued research into this exciting
creases (in the range of 40–50%) were seen in the white field and possible application of the basic research
quadriceps (type IIb fibers) and soleus (type I fibers) findings to treatment or prevention of metabolic dis-
but not in the red region of the quadriceps (type IIa eases. Musi et al. (64) reported that the ␣2-isoform of
fibers). However, the muscle AMPK response to AICAR AMPK is activated in muscle of patients with Type 2
injection was less at the end of 4 wk than at the diabetes in response to 20 and 45 min of exercise. They
beginning. When rats were given intermittent injec- also demonstrated with Western blots that both ␣-iso-
tions of AICAR rather than daily injections (to mini- forms are expressed normally in muscles of these pa-
mize the downregulation), a significant increase in tients. It has been previously suggested that some
citrate synthase in the red quadriceps was observed forms of Type 2 diabetes could possibly be due to
after 2-wk treatment (101). Enzymes of fatty acid oxi- deficiencies in the AMPK signaling pathway (98). This
dation were not significantly influenced by AICAR may still prove to be the case, since subjects used in the
treatment. A more recent study, however, provides above-mentioned study were not obese and were capa-
evidence of a twofold increase in the rate of CPT-1 gene ble of exercising at work rates required to activate
transcription in response to local infusion of AICAR AMPK. A recent preliminary report indicates that
into muscle of conscious rats in vivo (86). An increase transgenic mice deficient in muscle AMPK activity
in transcription of CPT-1 along with other metabolic have reduced exercise tolerance (62). When the numer-
J Appl Physiol • VOL 91 • SEPTEMBER 2001 • www.jap.org
INVITED REVIEW 1025

ous actions of AMPK are considered (activation of glu-


cose transport in muscle, control of GLUT-4 expression
in muscle, activation of muscle fatty acid oxidation,
inhibition of hepatic cholesterol synthesis and lipogen-
esis, stimulation of hepatic fatty acid oxidation, and
ketogenesis), it is not difficult to imagine that a defi-
ciency in AMPK could lead to insulin resistance and
obesity. Nevertheless, in those individuals with intact
AMPK signaling, exercise of sufficient duration and
intensity will most likely be beneficial in prevention
and possibly treatment of Type 2 diabetes.
Another major finding this year was from a study in
mice lacking the skeletal muscle isoform of ACC (2).
The consequent low malonyl-CoA in skeletal muscle
was postulated to result in chronically increased fatty
acid oxidation in muscle. Labeled palmitate oxidation
was 30% higher in incubated soleus muscle from these
mice compared with that of wild-type mice. In addition, Fig. 7. Postulated consequences of acute activation of AMPK on
insulin suppression of fatty acid oxidation was not metabolism during single bouts of exercise (fatty acid oxidation and
glucose transport) and in response to repetitive activation of AMPK
observed in soleus muscles from the ACC-deficient as would be the case during endurance training [effects on GLUT-4,
mice. These mice accumulated less fat in epididymal hexokinase (HK), uncoupling protein 3 (UCP3), and oxidative en-
fat pads while eating normally. This confirms the stud- zymes of the citric acid cycle and electron transport chain]. Possible
ies in rats that demonstrated that daily AICAR injec- phosphorylation target proteins ACC, MCD, and nitric oxide syn-
thetase (NOS) are indicated. ?, Phosphorylation targets in these
tions (which results in activation of AMPK, inactiva- actions are uncertain or completely unknown.
tion of ACC, and decreased malonyl-CoA in muscle)
reduced fat pad size compared with pair-fed controls
(101).
It has been suggested previously that development of On a broader scale, evidence is accumulating for a
pharmaceutical activators of AMPK may be a feasible role of chronic AMPK activation in increasing the max-
approach for treatment of obesity and Type 2 diabetes imal capacity for ATP production. This includes en-
(50, 96, 99–101). AICAR probably will not be the drug hancement of the rate of transcription of the GLUT-4
of choice, since large quantities are required for induc- gene, allowing increased amounts of GLUT-4 to accu-
tion of AMPK activation in vivo. Liver hypertrophy mulate in the muscle in response to training. This
develops when large doses of AICAR are given for a would allow increases in maximal glucose uptake dur-
prolonged period (101). Much more potent and specific ing very heavy exercise. The increase in mitochondrial
AMPK activators must be developed. oxidative enzymes, hypothesized to be due in part to
the chronic activation of AMPK, would allow increased
SUMMARY AND FUTURE DIRECTIONS
maximal rates of pyruvate oxidation and ATP produc-
tion as the glucose is metabolized. Thus the AMPK
Evidence has been presented to support the hypoth- energy-sensing and signaling system appears to be
esis that AMPK, activated in response to muscle con- involved in counteracting high-energy phosphate de-
traction, is involved in regulation of fatty acid oxida- pletion during single bouts of exercise and, in the long
tion and stimulation of glucose uptake during exercise term, in inducing adaptations that counteract high-
(Fig. 7). The phosphorylation targets for AMPK include energy phosphate depletion during prolonged heavy
ACC and possibly MCD, which then induce a decline in exercise. The phosphorylation targets of AMPK re-
malonyl-CoA in rat muscle (but not in human muscle) sponsible for mediating increases in GLUT-4 and mi-
and allow an increased rate of entrance of long-chain tochondrial proteins are yet to be identified.
fatty acyl-CoA into the mitochondria for oxidation. In The basic research on the AMPK signaling pathway
AMPK stimulation of glucose uptake, NOS has been provides additional strong rationale for the use of ex-
hypothesized to be a phosphorylation target, but data ercise in prevention and treatment of Type 2 diabetes
have been presented that are inconsistent with the and obesity (96). Regular activation of this system
hypothesis that NOS activation mediates the effect of appears to increase fatty acid oxidation, glucose up-
muscle contraction. Certainly, it will be important to take, and responsiveness of muscle to insulin (by in-
focus research efforts on this action of AMPK to discern creasing GLUT-4). It may be also feasible to develop
the downstream signaling mechanisms responsible for suitable pharmaceutical AMPK activators, which
GLUT-4 translocation and stimulation of glucose up- would mimic many of the effects of exercise. Obviously,
take. Development of specific inhibitors of AMPK and exercise will always be the most economical and most
the use of transgenic models will be important. Both of effective way to activate this kinase and produce the
these rapid actions in muscle would have the effects of beneficial effects, but, for those who are unable to
enhancing the rate of ATP production during acute exercise, the development of AMPK activators may
bouts of exercise. have clinical application (96).
J Appl Physiol • VOL 91 • SEPTEMBER 2001 • www.jap.org
1026 INVITED REVIEW

Work in the author’s laboratory was supported by National Insti- 19. Davies SP, Helps NR, Cohen PTW, and Hardie DG. 5⬘-
tute of Arthritis and Musculoskeletal and Skin Diseases Grant AMP inhibits dephosphorylation, as well as promoting phos-
AR-41438. phorylation, of the AMP-activated protein kinase. Studies using
bacterially expressed human protein phosphatase-2C␣ and na-
tive bovine protein phosphatase-2C␣. FEBS Lett 377: 421–425,
REFERENCES 1995.
1. Abbud W, Habinowski S, Zhang JZ, Kendrew J, Elkairi 20. Dean D, Daugaard JR, Young ME, Saha A, Vavvas D, Asp
FS, Kemp BE, Witters LA, and Ismail-Beigi F. Stimulation S, Kiens B, Kim KH, Witters L, Richter EA, and Ruder-
of AMP-activated protein kinase (AMPK) is associated with man N. Exercise diminishes the activity of acetyl-CoA carbox-
enhancement of GLUT1-mediated glucose transport. Arch Bio- ylase in human muscle. Diabetes 49: 1295–1300, 2000.
chem Biophys 380: 347–352, 2000. 21. Derave W, Ai H, Ihlemann J, Witters LA, Kristiansen S,
2. Abu-Etheiga L, Matzuk MM, Abo-Hashema KAH, and Richter EA, and Ploug T. Dissociation of AMP-activated
Wakil SJ. Continuous fatty acid oxidation and reduced fat protein kinase activation and glucose transport in contracting
storage in mice lacking acetyl-CoA carboxylase 2. Science 291: slow-twitch muscle. Diabetes 49: 1281–1287, 2000.
2613–2616, 2001. 22. Devlin JT, Hirshman M, Horton ED, and Horton ES.
3. Alam N and Saggerson ED. Malonyl-CoA and the regulation Enhanced peripheral and splanchnic insulin sensitivity in
of fatty acid oxidation in soleus muscle. Biochem J 334: 233– NIDDM men after a single bout of exercise. Diabetes 36: 434–
241, 1998. 439, 1987.
4. American Diabetes Association. Diabetes mellitus and ex- 23. Dudley GA, Tullson PC, and Terjung RL. Influence of
ercise. Diabetes Care 21: S40–S44, 1998. mitochondrial content on sensitivity of respiratory control.
5. Balon TW. Integrative biology of nitric oxide and exercise. J Biol Chem 262: 9109–9114, 1987.
Exerc Sport Sci Rev 27: 219–253, 1999. 24. Dyck JRB, Gao G, Widmer J, Stapleton D, Fernandez CS,
6. Balon TW. Role of nitric oxide in contraction induced glucose Kemp BE, and Witters LA. Regulation of the 5⬘-activated
transport. Adv Exp Med Biol 441: 87–95, 1998. protein kinase activity by the non-catalytic ␤ and ␥ subunits.
7. Bavenholm PN, Pigon J, Saha AK, Ruderman NB, and J Biol Chem 271: 17798–17803, 1996.
Efendic S. Fatty acid oxidation and the regulation of malonyl- 25. Essig DA. Contractile activity-induced mitochondrial biogene-
CoA in human muscle. Diabetes 49: 1078–1083, 2000. sis in skeletal muscle. Exerc Sport Sci Rev 24: 289–319, 1996.
8. Bergeron R, Russell RR, Young LH, Ren JM, Marchucci 26. Etgen GJ, Jensen J, Wilson CM, Hunt DG, Cushman SW,
M, Lee A, and Shulman GI. Effect of AMPK activation on and Ivy JL. Exercise training reverses insulin resistance in
muscle glucose metabolism in conscious rats. Am J Physiol muscle by enhanced recruitment of GLUT-4 to the cell surface.
Endocrinol Metab 276: E938–E944, 1999. Am J Physiol Endocrinol Metab 272: E864–E869, 1997.
9. Beri RK, Marley AE, See CG, Sopwith WF, Aguan K, 27. Ezaki O. Regulatory elements in the insulin-responsive glu-
Carling D, and Carey F. Molecular cloning, expression and cose transporter (GLUT4) gene. Biochem Biophys Res Commun
chromosomal localisation of human AMP-activated protein ki- 241: 1–6, 1997.
nase. FEBS Lett 356: 117–121, 1994. 28. Foretz M, Carling D, Guichard C, Ferr P, and Foufelle F.
10. Booth FW and Baldwin KM. Muscle plasticity: energy de- AMP-activated protein kinase inhibits the glucose-activated
mand and supply processes. In: Handbook of Physiology. Exer- expression of fatty acid synthase gene in rat hepatocytes. J Biol
cise: Regulation and Integration of Multiple Systems. Bethesda, Chem 273: 14767–14771, 1998.
MD: Am. Physiol. Soc., 1996, sect. 12, chapt. 24, p. 1075–1123. 29. Fryer LGD, Hajduch E, Rencurel F, Salt IP, Hundal HS,
11. Buhl ES, Jessen N, Schmitz O, Pedersen SB, Pedersen O, Hardie DG, and Carling D. Activation of glucose transport by
Holman GD, and Lund S. Chronic treatment with 5-amino- AMP-activated protein kinase via stimulation of nitric oxide
imidazole-4-carboxamide-1-␤-D-ribofuranoside increases insu-
synthetase. Diabetes 49: 1978–1985, 2000.
lin-stimulated glucose uptake and GLUT4 translocation in rat
30. Fugii N, Hayashi T, Hirshman MF, Smith JT, Habinowski
skeletal muscles in fiber type-specific manner. Diabetes 50:
SA, Kaijser L, Mu J, Ljungqvist O, Birnbaum MJ, Witters
12–17, 2001.
LA, Thorell A, and Goodyear LJ. Exercise induces isoform-
12. Carling D, Clarke PR, Zammit VA, and Hardie DG. Puri-
specific increase in 5⬘AMP-activated protein kinase activity in
fication and characterization of the AMP-activated protein ki-
human skeletal muscle. Biochem Biophys Res Commun 273:
nase. Copurification of acetyl-CoA carboxylase kinase and 3-hy-
droxy-3-methylglutaryl-CoA reductase kinase activities. Eur 1150–1155, 2000.
J Biochem 186: 129–136, 1989. 31. Goodyear LJ. AMP-activated protein kinase: a critical signal-
13. Chen ZP, Heierhorst J, Mann RJ, Mitchelhill KI, Michell ing intermediary for exercise-stimulated glucose transport? Ex-
BJ, Witters LA, Lynch GS, Kemp BE, and Stapleton D. erc Sport Sci Rev 28: 113–116, 2000.
Expression of the AMP-activated protein kinase ␤1 and ␤2 32. Goodyear LJ and Kahn BB. Exercise, glucose transport, and
subunits in skeletal muscle. FEBS Lett 460: 343–348, 1999. insulin sensitivity. Annu Rev Med 49: 235–261, 1998.
14. Chen ZP, McConnell GK, Michell BJ, Snow RJ, Canny 33. Gulve EA and Spina RJ. Effect of 7–10 days of cycle ergome-
BJ, and Kemp BE. AMPK Signaling in contracting human ter exercise on skeletal muscle GLUT-4 protein content. J Appl
skeletal muscle: acetyl-CoA carboxylase and NO synthase phos- Physiol 79: 1562–1566, 1995.
phorylation. Am J Physiol Endocrinol Metab 279: E1202– 34. Hardie DG and Carling D. The AMP-activated protein ki-
E1206, 2000. nase. Fuel gauge of the mammalian cell? Eur J Biochem 246:
15. Cheung PCF, Salt IP, Davies SP, Hardie DG, and Carling 259–273, 1997.
D. Characterization of AMP-activated protein kinase ␥-subunit 35. Hardie DG, Carling D, and Carlson M. The AMP-activated/
isoforms and their role in AMP binding. Biochem J 346: 659– SNF1 protein kinase subfamily: metabolic sensors of the eu-
669, 2000. karyotic cell? Annu Rev Biochem 67: 821–855, 1998.
16. Corton JM, Gillespie JG, and Hardie DG. Role of the 36. Hardie DG, Salt IP, Hawley SA, and Davies SP. AMP-
AMP-activated protein kinase in the cellular stress response. activated protein kinase: an ultrasensitive system for monitor-
Curr Biol 4: 315–324, 1994. ing cellular energy charge. Biochem J 338: 717–722, 1999.
17. Corton JM, Gillespie JG, Hawley SA, and Hardie DG. 37. Hawley SA, Davison Woods A, Davies SP, Beri RK,
5-Aminoimidazole-4-carboxamide ribonucleoside: a specific Carling D, and Hardie DG. Characterization of the AMP-
method for activating AMP-activated protein kinase in intact activated protein kinase kinase from rat liver, and identifica-
cells? Eur J Biochem 229: 558–565, 1995. tion of threonine-172 as the major site at which it phosphory-
18. Da Silva XG, Leclerc I, Salt IP, Doiron B, Hardie DG, lates and activates AMP-activated protein kinase. J Biol Chem
Kahn A, and Rutter GA. Role of AMP-activated protein ki- 271: 27879–27887, 1996.
nase in regulation by glucose of islet beta cell gene expression. 38. Hayashi T, Hirshman MF, Kurth EJ, Winder WW, and
Proc Natl Acad Sci USA 97: 4023–4028, 2000. Goodyear LJ. Evidence for 5⬘-AMP-activated protein kinase

J Appl Physiol • VOL 91 • SEPTEMBER 2001 • www.jap.org


INVITED REVIEW 1027

mediation of the effect of muscle contraction on glucose trans- 59. McGarry JD and Brown NF. The mitochondrial carnitine
port. Diabetes 47: 1369–1373, 1998. palmitoyltransferase system from concept to molecular analy-
39. Hayashi T, Hirshman MF, Fugii N, Habinowski SA, Wit- sis. Eur J Biochem 244: 1–14, 1997.
ters LA, and Goodyear LJ. Metabolic stress and altered 60. Merrill GF, Kurth EJ, Hardie DG, and Winder WW. AICA
glucose transport. Activation of AMP-activated protein kinase riboside increases AMP-activated protein kinase, fatty acid
as a unifying coupling mechanism. Diabetes 49: 527–531, 2000. oxidation, and glucose uptake in rat muscle. Am J Physiol
40. Hayashi T, Wojtaszewski JF, and Goodyear LJ. Exercise Endocrinol Metab 273: E1107–E1112, 1997.
regulation of glucose transport in skeletal muscle. Am J Physiol 61. Merrill GF, Kurth EJ, Rasmussen BB, and Winder WW.
Endocrinol Metab 273: E1039–E1051, 1997. Influence of malonyl-CoA and palmitate concentration on rate
41. Henin N, Vincent MF, Gruber HE, and Van den Berghe G. of palmitate oxidation in rat muscle. J Appl Physiol 85: 1909–
Inhibition of fatty acid and cholesterol synthesis by stimulation 1914, 1998.
of AMP-activated protein kinase. FASEB J 9: 541–546, 1995. 62. Mu J and Birnbaum MJ. A role for AMP-activated protein
42. Henin N, Vincent MF, and Van den Berghe G. Stimulation kinase in contraction and hypoxia regulated glucose transport
of rat liver AMP-activated protein kinase by AMP analogues. in skeletal muscle (Abstract). FASEB J 15: A1164, 2001.
Biochim Biophys Acta 1290: 197–203, 1996. 63. Muoio DM, Seefeld K, Witters LA, and Coleman RA. AMP-
43. Higaki Y, Hirshman MF, Fujii N, and Goodyear LJ. Nitric activated kinase reciprocally regulates triacylglycerol synthesis
oxide increases glucose uptake through a mechanism that is and fatty acid oxidation in liver and muscle: evidence that
distinct from the insulin and contraction pathways in rat skel- sn-glycerol-3-phosphate acyltransferase is a novel target. Bio-
etal muscle. Diabetes 50: 241–247, 2001. chem J 338: 783–791, 1999.
44. Holloszy JO. Biochemical adaptations in muscle. Effects of 64. Musi N, Fugii N, Hirshman MF, Ekberg I, Froberg S,
exercise on mitochondrial oxygen uptake and respiratory en- Ljungqvist O, Thorell A, and Goodyear LJ. AMP-activated
zyme activity in skeletal muscle. J Biol Chem 242: 2278–2282, protein kinase (AMPK) is activated in muscle of subjects with
1967. type 2 diabetes during exercise. Diabetes 50: 921–927, 2001.
45. Holloszy JO and Booth FW. Biochemical adaptations to 65. Musi N, Hayashi T, Fugii N, Hirshman MF, Witters LA,
endurance exercise in muscle. Annu Rev Physiol 38: 273–291, and Goodyear LJ. AMP-activated protein kinase activity and
1976. glucose uptake in rat skeletal muscle. Am J Physiol Endocrinol
46. Holloszy JO and Coyle EF. Adaptations of skeletal muscle to Metab 280: E677–E684, 2001.
endurance exercise and their metabolic consequences. J Appl 66. Neufer PD and Dohm GL. Exercise induces a transient
Physiol 56: 831–838, 1984. increase in transcription of the GLUT-4 gene in skeletal mus-
47. Holloszy JO and Hansen PA. Regulation of glucose transport cle. Am J Physiol Cell Physiol 265: C1597–C1603, 1993.
into skeletal muscle. Rev Physiol Biochem Pharmacol 128: 67. Oakes ND, Bell KS, Furler SM, Camilleri S, Saha AK,
99–193, 1996. Ruderman NB, Chisholm DJ, and Kraegen EW. Diet-in-
48. Holloszy JO and Kohrt WM. Regulation of carbohydrate and duced muscle insulin resistance in rats is ameliorated by acute
fat metabolism during and after exercise. Annu Rev Nutr 16: dietary lipid withdrawal or a single bout of exercise. Diabetes
121–138, 1996. 46: 2022–2028, 1997.
49. Holloszy JO, Schultz J, Kusnierkiewicz J, Hagberg JM, 68. Odland LM, Heigenhauser GJ, and Spriet LL. Effects of
and Ehsani AA. Effects of exercise on glucose tolerance and high fat provision on muscle PDH activation and malonyl-CoA
insulin resistance. Acta Med Scand Suppl 711: 55–65, 1986. content in moderate exercise. J Appl Physiol 89: 2352–2358,
50. Holmes BF, Kurth-Kraczek EJ, and Winder WW. Chronic 2000.
activation of 5⬘-AMP-activated protein kinase increases 69. Odland LM, Heigenhauser GJ, Lopaschuk GD, and
GLUT-4, hexokinase, and glycogen in muscle. J Appl Physiol Spriet LL. Human skeletal muscle malonyl-CoA at rest and
87: 1990–1995, 1999. during prolonged submaximal exercise. Am J Physiol Endocri-
51. Hood DA. Contractile activity-induced mitochondrial biogene- nol Metab 270: E541–E544, 1996.
sis in skeletal muscle. J Appl Physiol 90: 1137–1157, 2001. 70. Odland LM, Howlett RA, Heigenhauser GJ, Hultman E,
52. Houmard JA, Egan PC, Neufer PD, Friedman JE, and Spriet LL. Skeletal muscle malonyl-CoA content at the
Wheeler WS, Israel RG, and Dohm GL. Elevated skeletal onset of exercise at varying power outputs in humans. Am J
muscle glucose transporter levels in exercise-trained, middle- Physiol Endocrinol Metab 274: E1080–E1085, 1998.
aged men. Am J Physiol Endocrinol Metab 261: E437–E443, 71. Ojuka EO, Nolte LA, and Holloszy JO. Increased expression
1991. of GLUT-4 and hexokinase in rat epitrochlearis muscles ex-
53. Hutber CA, Hardie DG, and Winder WW. Electrical stimu- posed to AICAR in vitro. J Appl Physiol 88: 1072–1075, 2000.
lation inactivates muscle acetyl-CoA carboxylase and increases 72. Pilegaard H, Ordway GA, Saltin B, and Neufer PD. Tran-
AMP-activated protein kinase. Am J Physiol Endocrinol Metab scriptional regulation of gene expression in human skeletal
272: E262–E266, 1997. muscle during recovery from exercise. Am J Physiol Endocrinol
54. Ivy JL. Role of exercise training in the prevention and treat- Metab 279: E806–E814, 2000.
ment of insulin resistance and non-insulin-dependent diabetes 73. Ponticos M, Lu QL, Morgan JE, Hardie DG, Partridge TA,
mellitus. Sports Med 24: 321–336, 1997. and Carling D. Dual regulation of the AMP-activated protein
55. Kawanaka K, Nolte LA, Han DH, Hansen PA, and Hol- kinase provides a novel mechanism for the control of creatine
loszy JO. Mechanisms underlying impaired GLUT-4 translo- kinase in skeletal muscle. EMBO J 17: 1688–1699, 1998.
cation in glycogen supercompensated muscles of exercised rats. 74. Rasmussen BB, Hancock CR, and Winder WW. Postexer-
Am J Physiol Endocrinol Metab 279: E1311–E1318, 2000. cise recovery of skeletal muscle malonyl-CoA, acetyl-CoA car-
56. Kurth-Kraczek EJ, Hirshman MF, Goodyear LJ, and boxylase, and AMP-activated protein kinase. J Appl Physiol 85:
Winder WW. 5⬘-AMP activated protein kinase activation 1629–1634, 1998.
causes GLUT4 translocation in skeletal muscle. Diabetes 48: 75. Rasmussen BB and Winder WW. Effect of exercise intensity
1667–1671, 1999. on skeletal muscle malonyl-CoA and acetyl-CoA carboxylase.
57. Leclerc I, Kahn A, and Doiron B. The 5⬘-AMP-activated J Appl Physiol 83: 1104–1109, 1997.
protein kinase inhibits the transcriptional stimulation by glu- 76. Ren JM, Semenkovich CF, Gulve EA, Gao J, and Holloszy
cose in liver cells, acting through the glucose response complex. JO. Exercise induces rapid increases in GLUT4 expression,
FEBS Lett 431: 180–4, 1998. glucose transport capacity, and insulin-stimulated glycogen
58. Lochhead PA, Salt IP, Walker KS, Hardie DG, and Suth- storage in muscle. J Biol Chem 269: 14396–14401, 1994.
erland C. 5-Amino-4-carboxamide riboside mimics the effects 76a.Richter EA, MacDonald C, Kı̀ens B, Hardie G, and
of insulin on the expression of the 2 key gluconeogenic genes Wojtaszewski JFP. Dissociation of 5⬘-AMP-activated protein
PEPCK and glucose-6-phosphatase. Diabetes 49: 896–903, kinase activity and glucose clearance in human skeletal muscle
2000. during exercise (Abstract). Diabetes 50, Suppl 2: A62, 2001.

J Appl Physiol • VOL 91 • SEPTEMBER 2001 • www.jap.org


1028 INVITED REVIEW

77. Ruderman NB, Saha AK, Vavvas D, Kurowski T, Laybutt 92. Vincent MF, Marangos P, Gruber HE, and Van den
DR, Schmitz-Peiffer C, Biden T, and Kraegen EW. Malo- Berghe G. AICA riboside inhibits gluconeogenesis in isolated
nyl-CoA as a metabolic switch and regulator of insulin sensi- rat hepatocytes. Adv Exp Med Biol 309B: 1991–1995, 1991.
tivity. Adv Exp Med Biol 441: 263–270, 1998. 93. Wallberg-Henriksson H, Rincon J, and Zierath JR. Exer-
78. Ruderman NB, Saha AK, Vavvas D, and Witters LA. Ma- cise in the management of non-insulin-dependent diabetes mel-
lonyl-CoA, fuel sensing, and insulin resistance. Am J Physiol litus. Sports Med 25: 25–35, 1998.
Endocrinol Metab 276: E1–E18, 1999. 94. Winder WW. Intramuscular mechanisms regulating fatty acid
79. Sabina RL, Patterson D, and Holmes EW. 5-Amino-4-imi- oxidation during exercise. Adv Exp Med Biol 441: 239–248,
dazole carboxamide riboside (Z-riboside) metabolism in eukary- 1998.
otic cells. J Biol Chem 260: 6107–6114, 1991. 95. Winder WW. Malonyl-CoA—regulator of fatty acid oxidation in
80. Saha AK, Schwarsin AJ, Roduit R, Massé F, Kaushik V, muscle during exercise. Exerc Sport Sci Rev 26: 117–132, 1998.
Tornheim K, Prentki M, and Ruderman NB. Activation of 96. Winder WW. AMP-activated protein kinase: possible target for
malonyl-CoA decarboxylase in rat skeletal muscle by contrac- treatment of type 2 diabetes. Diabetes Technol Therapeut 2:
tion and the AMP-activated protein kinase activator aminoimi- 441–447, 2000.
dazole-4-carboxamide-1-D-ribofuranoside. J Biol Chem 275:
97. Winder WW, Arogyasami J, Elayan IM, and Cartmill D.
24279–24283, 2000.
Time course of the exercise-induced decline in malonyl-CoA in
81. Salt IP, Celler JW, Hawley SA, Prescott A, Woods A,
different muscle types. Am J Physiol Endocrinol Metab 259:
Carling D, and Hardie DG. AMP-activated protein kinase—
greater AMP dependence, and preferential nuclear localization, E266–E271, 1990.
of complexes containing the ␣2 isoform. Biochem J 334: 177– 98. Winder WW and Hardie DG. Inactivation of acetyl-CoA car-
187, 1998. boxylase and activation of AMP-activated protein kinase in
82. Stapleton D, Mitchelhill KI, Gao G, Widmer G, Mitchell muscle during exercise. Am J Physiol Endocrinol Metab 270:
BJ, The T, House CM, Fernandez CS, Cox T, Witters LA, E299–E304, 1996.
and Kemp BE. Mammalian AMP-activated protein kinase 99. Winder WW and Hardie DG. AMP-activated protein kinase,
subfamily. J Biol Chem 271: 611–614, 1996. a metabolic master switch: possible roles in Type 2 diabetes.
83. Stapleton D, Woollatt E, Mitchelhill KI, Nicholl JK, Fer- Am J Physiol Endocrinol Metab 277: E1–E10, 1999.
nandez CS, Michell BJ, Witters LA, Power DA, Suther- 100. Winder WW and Holmes BF. Insulin stimulation of glucose
land GR, and Kemp BE. AMP-activated protein kinase isoen- uptake fails to decrease palmitate oxidation in muscle if AMPK
zyme family: subunit structure and chromosomal location. is activated. J Appl Physiol 89: 2430–2437, 2000.
FEBS Lett 409: 452–456, 1997. 101. Winder WW, Holmes BF, Rubink DS, Jensen EB, Chen M,
84. Starritt EC, Howlett RA, Heigenhauser GJ, and Spriet and Holloszy JO. Activation of AMP-activated protein kinase
LL. Sensitivity of CPT I to malonyl-CoA in trained and un- increases mitochondrial enzymes in skeletal muscle. J Appl
trained human skeletal muscle. Am J Physiol Endocrinol Physiol 88: 2219–2226, 2000.
Metab 278: E462–E468, 2000. 102. Winder WW, Wilson HA, Hardie DG, Rasmussen BB, Hut-
85. Stein SC, Woods A, Jones NA, Davison MD, and Carling ber CA, Call GB, Clayton RD, Conley LM, Yoon S, and
D. The regulation of AMP-activated protein kinase by phos- Zhou B. Phosphorylation of rat muscle acetyl-CoA carboxylase
phorylation. Biochem J 345: 437–443, 2000. by AMP-activated protein kinase and protein kinase A. J Appl
86. Stoppani J, Hildebrandt AL, and Neufer PD. Activation of Physiol 82: 219–225, 1997.
AMP-activated protein kinase increases transcription of meta- 103. Wojtaszewski JF, Nielsen P, Hansen BF, Richter EA, and
bolic genes in rat gastrocnemius muscle (Abstract). FASEB J Kiens B. Isoform-specific and exercise intensity-dependent ac-
15: A415, 2001. tivation of 5⬘-AMP-activated protein kinase in human skeletal
87. Sullivan JE, Brocklehurst KJ, Marley AE, Carey F, muscle. J Physiol (Lond) 528: 221–226, 2000.
Carling D, and Beri RK. Inhibition of lipolysis and lipogene- 104. Woods A, Salt I, Scott J, Hardie DG, and Carling D. The ␣1
sis in isolated rat hepatocytes with AICAR, a cell permeable and ␣2 isoforms of the AMP-activated protein kinase have
activator of AMP-activated protein kinase. FEBS Lett 353: similar activities in rat liver but exhibit differences in substrate
33–36, 1994.
specificity in vitro. FEBS Lett 397: 347–351, 1996.
88. Thornton C, Snowden MA, and Carling D. Identification of
105. Yeh JI, Gulve EA, Remeh L, and Birnbaum MJ. The effects
a novel AMP-activated protein kinase subunit isoform that is
of wortmannin on rat skeletal muscle. Dissociation of signaling
highly expressed in skeletal muscle. J Biol Chem 273: 12443–
12450, 1998. pathways for insulin- and contraction-activated hexose trans-
89. Trumble GE, Smith MA, and Winder WW. Purification and port. J Biol Chem 270: 2107–2111, 1995.
characterization of rat skeletal muscle acetyl-CoA carboxylase. 106. Young ME, Radda GK, and Leighton B. Activation of gly-
Eur J Biochem 231: 192–198, 1995. cogen phosphorylase and glycogenolysis in rat skeletal muscle
90. US Department of Health and Human Services. The ef- by AICAR: an activator of AMP-activated protein kinase. FEBS
fects of physical activity on health and disease. In: Physical Lett 382: 43–47, 1996.
Activity and Health: A Report of the Surgeon General. Centers 107. Zheng D, MacLean PS, Pohnert SC, Knight JB, Olson AL,
for Disease Prevention and Control, National Center for Winder WW, and Dohm GL. Regulation of muscle GLUT-4
Chronic Disease Prevention and Health Promotion, Washing- transcription by AMP-activated protein kinase. J Appl Physiol
ton, DC: GPO, 1996. In press.
91. Vavvas D, Apazidis A, Saha AK, Gamble J, Patel A, Kemp 108. Zhou M, Lin BZ, Coughlin S, Valega G, and Pilch PF.
BE, Witters LA, and Ruderman NB. Contraction-induced UCP-3 expression in skeletal muscle: effects of exercise, hyp-
changes in acetyl-CoA carboxylase and 5⬘-AMP-activated ki- oxia, and AMP-activated protein kinase. Am J Physiol Endo-
nase in skeletal muscle. J Biol Chem 272: 13256–13261, 1997. crinol Metab 279: E622–E629, 2000.

J Appl Physiol • VOL 91 • SEPTEMBER 2001 • www.jap.org

You might also like