ArticlePDF AvailableLiterature Review

AMPK: A key regulator of energy balance in the single cell and the whole organism

Authors:

Abstract and Figures

The AMP-activated protein kinase (AMPK) system is a key player in regulating energy balance at both the cellular and whole-body levels, placing it at centre stage in studies of obesity, diabetes and the metabolic syndrome. It is switched on in response to metabolic stresses such as muscle contraction or hypoxia, and modulated by hormones and cytokines affecting whole-body energy balance such as leptin, adiponectin, resistin, ghrelin and cannabinoids. Once activated, it switches on catabolic pathways that generate adenosine triphosphate (ATP), while switching off ATP-consuming anabolic processes. AMPK exists as heterotrimeric complexes comprising a catalytic alpha-subunit and regulatory beta- and gamma-subunits. Binding of AMP to the gamma-subunit, which is antagonized by high ATP, causes activation of the kinase by promoting phosphorylation at threonine (Thr-172) on the alpha-subunit by the upstream kinase LKB1, allowing the system to act as a sensor of cellular energy status. In certain cells, AMPK is activated in response to elevation of cytosolic Ca2+ via phosphorylation of Thr-172 by calmodulin-dependent kinase kinase-beta (CaMKKbeta). Activation of AMPK, either in response to exercise or to pharmacological agents, has considerable potential to reverse the metabolic abnormalities associated with type 2 diabetes and the metabolic syndrome. Two existing classes of antidiabetic drugs, that is, biguanides (for example, metformin) and the thiazolidinediones (for example, rosiglitazone), both act (at least in part) by activation of AMPK. Novel drugs activating AMPK may also have potential for the treatment of obesity.
Content may be subject to copyright.
REVIEW
AMPK: a key regulator of energy balance in the single
cell and the whole organism
DG Hardie
Division of Molecular Physiology, College of Life Sciences, University of Dundee, Dundee, UK
The AMP-activated protein kinase (AMPK) system is a key player in regulating energy balance at both the cellular and whole-body
levels, placing it at centre stage in studies of obesity, diabetes and the metabolic syndrome. It is switched on in response to metabolic
stresses such as muscle contraction or hypoxia, and modulated by hormones and cytokines affecting whole-body energy balance
such as leptin, adiponectin, resistin, ghrelin and cannabinoids. Once activated, it switches on catabolic pathways that generate
adenosine triphosphate (ATP), while switching off ATP-consuming anabolic processes. AMPK exists as heterotrimeric complexes
comprising a catalytic a-subunit and regulatory b-andg-subunits. Binding of AMP to the g-subunit, which is antagonized by high
ATP, causes activation of the kinase by promoting phosphorylation at threonine (Thr-172) on the a-subunit by the upstream kinase
LKB1, allowing the system to act as a sensor of cellular energy status. In certain cells, AMPK is activated in response to elevation of
cytosolic Ca
2þ
via phosphorylation of Thr-172 by calmodulin-dependent kinase kinase-b(CaMKKb). Activation of AMPK, either in
response to exercise or to pharmacological agents, has considerable potential to reverse the metabolic abnormalities associated with
type 2 diabetes and the metabolic syndrome. Two existing classes of antidiabetic drugs, that is, biguanides (for example, metformin)
and the thiazolidinediones (for example, rosiglitazone), both act (at least in part) by activation of AMPK. Novel drugs activating AMPK
may also have potential for the treatment of obesity.
International Journal of Obesity (2008) 32, S7S12; doi:10.1038/ijo.2008.116
Keywords: diabetes; metabolic syndrome; LKB1; calmodulin-dependent kinase kinase
Introduction
The AMP-activated protein kinase (AMPK) system was
discovered in mammalian cell extracts as activities that
phosphorylated and inactivated key enzymes of lipid biosynth-
esis, that is, acetyl-CoA carboxylase and 3-hydroxy-3-
methylglutaryl-CoA reductase, which regulate fatty acid and
cholesterol synthesis, respectively.
1
In 1987 the author’s
laboratory showed that these activities, which had been
presumed to be distinct, were in fact functions of the same
protein kinase. This kinase was allosterically activated by
50-AMP and was also activated by phosphorylation by upstream
kinases, and was renamed AMPK in 1988.
1
Structure and regulation of AMPK
AMP-activated protein kinase and its orthologues in lower
eukaryotes, for example, budding yeast, are now known to
exist as heterotrimeric complexes consisting of catalytic
a-subunits and regulatory b- and g-subunits. Humans and
rodents express two isoforms of aand b(a1, a2; b1, b2), and
three isoforms of g(g1, g2 and g3) encoded by distinct
genes.
2
Some of these are also subject to alternative splicing,
leading to a large and diverse array of heterotrimeric
complexes. The a-subunits have conventional serine/threo-
nine kinase domains at the N-terminus, containing a
conserved threonine residue (Thr-172) whose phosphoryla-
tion by upstream kinases is absolutely required for their
activity. The primary upstream kinase was recently identified
to be a complex between the tumour suppressor, LKB1, and
two accessory subunits, STRAD (STE-20 related adaptor
protein) and MO25.
3,4
These findings were exciting, because
LKB1 was originally identified as the gene mutated in Peutz–
Jeghers syndrome, an inherited predisposition to cancer in
humans. LKB1 is a classical tumour suppressor: subjects with
this syndrome have heterozygous loss-of-function mutations
in the LKB1 gene and develop numerous polyps (benign
tumours) in the intestine, probably due to loss of expression
of their functional gene copy. They also have a 15-fold
increased risk of developing malignant tumors at other
sites.
5
LKB1 is now known to act upstream of 12 other
members of the AMPK-related kinase family, in addition to
Correspondence: Dr DG Hardie, Division of Molecular Physiology, College of
Life Sciences, University of Dundee, Sir James Black Centre, Dow Street,
Dundee DD1 5EH, UK.
E-mail: d.g.hardie@dundee.ac.uk
International Journal of Obesity (2008) 32, S7 S12
&
2008 Macmillan Publishers Limited All rights reserved 0307-0565/08
$
30.00
www.nature.com/ijo
the two isoforms of the AMPK catalytic subunit (a1 and a2).
5
There is evidence (discussed further below) that activation of
AMPK inhibits cell growth and proliferation. This makes it
likely that the ability of LKB1 to activate AMPK explains
much of its tumour suppressor effect, although a role for the
alternate downstream kinases cannot be discounted at
present.
The LKB1 complex appears to be constitutively active and
is not regulated by AMP. Binding of AMP to AMPK promotes
net phosphorylation of Thr-172 by inhibiting its depho-
sphorylation (by making the kinase a less efficient substrate
for protein phosphatases), as well as allosterically activating
the phosphorylated form of the kinase. These two effects of
AMP effectively multiply together, so that a small increase in
AMP can produce a much larger effect on the kinase
activity.
6
Both effects are also antagonized by high concen-
trations of ATP, so that AMPK is activated in a sensitive
manner by a small rise in the AMP/ATP ratio. If the adenylate
kinase reaction is at equilibrium (as appears to be the case in
most mammalian cells), the AMP/ATP ratio will vary as the
square of the ADP/ATP ratio, making the former ratio a very
sensitive indicator of cellular energy status.
Some cells express an alternate pathway for AMPK
activation that is triggered by increases in cytoplasmic
Ca
2þ
, leading to activation of calmodulin-dependent kinase
kinase-b(CaMKKb) which, like LKB1, can phosphorylate
Thr-172 on the AMPK a-subunit.
7–9
However, unlike the
effect of LKB1, phosphorylation by CaMKKbappears to be
AMP-independent. The tissue distribution of CaMKKbis
more restricted than that of LKB1, with the former being
most abundant in neurones and cells of the endothelial/
haemopoietic lineage.
The AMPK b-subunits have two conserved regions, a
central glycogen-binding domain and a C-terminal domain
required for forming a complex with the a- and g-subunits.
10
The glycogen-binding domain causes the complex to
partially associate with glycogen particles. The function of
this interaction is not known, although glycogen synthase
(another component of glycogen particles) is a downstream
target that is known to be phosphorylated and inactivated by
AMPK.
11
One interesting possibility is that the AMPK system
can sense some aspect of glycogen structure and provide a
feedback regulation on glycogen synthesis. The g-subunits
contain four tandem repeats of a structure known as a
cystathionine-b-synthase motif: these are now known to act
in pairs to form two modules (termed Bateman domains)
each of which binds one molecule of AMP or ATP in a
mutually exclusive manner.
12
Bateman domains also occur
in a few other proteins, where they bind adenosine-contain-
ing ligands, such as AMP, ATP or S-adenosyl methionine.
12
Intriguingly, mutations in these domains lead to a variety of
human hereditary diseases that are all caused by defective
ligand binding. In the case of AMPK these mutations, which
cause heart disease of varying severity associated with
excessive glycogen storage, prevent both binding of AMP
and allosteric activation, proving that the Bateman domains
on the g-subunit form the regulatory binding sites for AMP
and ATP.
12,13
Activation of AMPK by metabolic stress
As the LKB1-AMPK signalling pathway is activated by
elevation of the AMP/ATP ratio, it is switched on by any
metabolic stress that disturbs energy balance by interfering
with ATP synthesis, such as glucose deprivation, hypoxia or
ischaemia, or metabolic poisons that inhibit glycolysis (for
example, 2-deoxyglucose), the tricarboxylic acid cycle (for
example, arsenite) or oxidative phosphorylation (for exam-
ple, oligomycin, antimycin A, rotenone).
2
With most
mammalian cells, glucose deprivation is unlikely to be a
major issue in vivo, because they express isoforms of glucose
transporter and hexokinase that are saturated by low
concentrations of glucose, such that inhibition of ATP
synthesis and activation of AMPK would only occur when
plasma glucose dropped to levels incompatible with life.
However, specialized glucose-sensing cells such as the b-cells
in the Islets of Langerhans in the pancreas, and glucose-
regulated neurones in the hypothalamus, express isoforms of
glucose transporter (GLUT2) and hexokinase (hexokinase IV,
also known as glucokinase) with a much higher K
m
for
glucose, so that AMPK in these cells is activated by decreases
in blood glucose within the more normal physiological
range. Thus, in cell lines derived from rodent pancreatic b-
cells, AMPK is activated by low glucose and inhibited by high
glucose
14
whereas, in fasted mice, intracerebroventricular
injection of glucose, or refeeding, inhibits the a2-isoform of
AMPK in regions of the hypothalamus.
15
Moreover, the
known downstream consequences of lowering external
glucose in both cell types, that is, decreased insulin secretion
(b-cells) or increased feeding behaviour (hypothalamus) can
be mimicked by the activation of AMPK at these sites, either
by pharmacological or molecular biological interventions.
14–16
Thus, the AMPK system is a key player in the regulation of
energy balance at the whole-body level, not just at the
cellular level.
Just as there are specialized glucose-sensing cells in the
pancreas and hypothalamus, so there are specialized oxygen-
sensing cells where AMPK is regulated by normal physiolo-
gical variations in oxygen tension. These include the glomus
cells in the carotid body, which sense hypoxia in blood
supplied to the brain and regulate breathing, and pulmonary
artery smooth muscle cells, which contract in response to
hypoxia (unlike most arterial smooth muscle cells, which
relax), thus diverting blood flow away from poorly oxyge-
nated regions of the lung. In collaboration with the
laboratories of Evans and Peers,
17
the author has recently
shown that activation of AMPK mimics the effects of
hypoxia in these cells: (i) in glomus cells, entry of
extracellular Ca
2þ
via voltage-gated Ca
2þ
channels and
firing of action potentials in afferent neurones leading to the
brain; and (ii) in pulmonary artery smooth muscle, release of
AMPK and energy balance
DG Hardie
S8
International Journal of Obesity
Ca
2þ
from the endoplasmic reticulum and consequent
contraction.
All of the metabolic stresses mentioned above increase
cellular AMP/ATP by inhibiting ATP production. A stress that
activates AMPK by increasing ATP consumption is contrac-
tion in skeletal muscle.
18
There is now good evidence that
many of the acute metabolic responses to exercise, including
increased glucose uptake and fatty acid oxidation,
19,20
as
well as the long-term adaptations to regular endurance
exercise, such as increased expression of the glucose
transporter GLUT4
21
and increased mitochondrial biogen-
esis,
22
are at least partly mediated by AMPK activation.
Regulation of AMPK by hormones and cytokines
Although orthologues of AMPK are present in primitive
single-celled eukaryotes, suggesting that the system existed
prior to the evolution of hormones and cytokines, the latter
nevertheless appear to have acquired the ability to regulate
the AMPK system. In particular, AMPK is modulated by
cytokines released by adipocytes (termed adipokines) that
regulate whole-body energy balance, such as leptin, adipo-
nectin and resistin (reviewed in Kahn et al.
2
). Leptin and
adiponectin both activate AMPK and increase glucose uptake
and fatty acid oxidation in skeletal muscle, thus stimulating
whole-body energy expenditure. Adiponectin also activates
AMPK in liver, stimulating fatty acid oxidation and inhibit-
ing glucose production, whereas resistin appears to have the
opposite effects. Remarkably, although leptin activates
AMPK in skeletal muscle, it causes inhibition of the a2-
isoform of the kinase in regions of the hypothalamus in
fasted mice, concomitant with repression of food intake.
Other anorexigenic agents, such as insulin, melanocortin
receptor agonists and high glucose, also inhibit AMPK in the
hypothalamus of fasted mice, whereas orexigenic agents
such as agouti-related protein, the gut hormone ghrelin, and
cannabinoids activate the kinase in the hypothalamus of fed
mice.
2,23
In addition, artificially increasing AMPK activity in
the hypothalamus by using drugs or by expressing activated
mutants stimulates food intake even in the fed state.
2
Taken
together, these findings suggest that AMPK, acting both in
the hypothalamus and in the periphery, is a key player in the
regulation of the balance between whole-body energy intake
and energy expenditure, and hence in the development of
obesity.
The mechanisms by which adipokines and other agents
affecting food intake modulate AMPK activity remain
unclear. However, in cells of the endothelial/haematopoietic
lineage the Ca
2þ
-CaMKK pathway (discussed above)
switches on AMPK in response to activation of certain
receptors. In human umbilical vein endothelial cells,
thrombin increases cytoplasmic Ca
2þ
via receptor-mediated
release of inositol-1,4,5-trisphosphate and consequent re-
lease of Ca
2þ
from the endoplasmic reticulum. This activates
AMPK, an effect that is reduced by the inhibition of CaMKKb
by pharmacological means or by interfering RNA ap-
proaches.
24
AMPK is also activated by stimulation of the
antigen receptor in T lymphocytes, an effect that can be
mimicked by Ca
2þ
ionophores and is blocked by pharma-
cological inhibitors of CaMKK.
25
The function of AMPK
activation under these circumstances is not clear, but one
can speculate that the Ca
2þ
trigger represents a feed-forward
signal that anticipates the large demand for ATP that often
follows an increase in cytoplasmic Ca
2þ
(for example, the
rapid growth and proliferation that follows activation of T
lymphocytes).
Downstream targets of AMPK
A full discussion of this topic is beyond the scope of this brief
review, and the reader is referred elsewhere for detailed
discussion.
2
In general (as summarized in Figure 1), AMPK
switches on catabolic processes that provide alternative
routes to generate ATP (for example, glucose uptake,
glycolysis, fatty acid oxidation and mitochondrial biogen-
esis), while switching off anabolic processes that consume
ATP, such as the synthesis of fatty acids, triglyceride,
cholesterol, glucose (via gluconeogenesis) and glycogen.
These effects can occur through distinct mechanisms with
different time courses: (i) acute effects on metabolism due to
direct phosphorylation of metabolic enzymes (for example,
inhibition of cholesterol synthesis due to phosphorylation of
3-hydroxy-3-methylglutaryl-CoA reductase); (ii) longer-term
effects due to changes in gene expression (for example,
upregulation of glucose and fat oxidation due to increased
expression of mitochondrial genes, downregulation of
gluconeogenic genes); and (iii) combined acute and longer-
term effects (for example, inhibition of fatty acid synthesis
via direct phosphorylation of the ACC1 isoform of acetyl-
CoA carboxylase, combined with inhibition of expression of
the ACC1 and fatty acid synthase genes). Although not
shown in Figure 1, AMPK activation also inhibits protein
synthesis, both via inhibition of the target-of-rapamycin
pathway
26
(thus inhibiting initiation of translation), and via
activation of elongation factor-2 kinase
27
(thus inhibiting
elongation of translation). The ability of AMPK to inhibit
protein synthesis and other biosynthetic pathways contri-
butes to its effects to limit hypertrophy of non-dividing cells,
whereas in proliferating cells, progress through the cell cycle
is also blocked by AMPK activation.
28
Role of AMPK in obesity, diabetes and the
metabolic syndrome
The ability of AMPK to switch cells from an anabolic to a
catabolic state suggests that activators of the kinase might be
effective agents for treatment of obesity, type 2 diabetes and
the metabolic syndrome. Type 2 diabetes is characterized by
an elevated plasma glucose primarily caused by insulin
AMPK and energy balance
DG Hardie
S9
International Journal of Obesity
resistance, and risk of developing this condition is greatly
increased by obesity. Activation of AMPK has the potential to
lower plasma glucose both by repressing expression of
enzymes of gluconeogenesis in the liver, and by increasing
glucose uptake by muscle and other tissues (Figure 1). Insulin
resistance is also often associated with elevated storage of
triglycerides in tissues other than adipose tissue such as
muscle and liver, together with a relative deficit in
mitochondrial oxidative capacity in those organs.
29
By
inhibiting fatty acid and triglyceride synthesis and stimulat-
ing fatty acid oxidation and mitochondrial biogenesis,
AMPK activation has the potential to reduce hypertriglycer-
idemia, as well as elevated storage of triglycerides in muscle
and liver. Finally, the metabolic syndrome involves a cluster
of related metabolic abnormalities that are all potentially
reversed by AMPK activation, including insulin resistance,
abdominal obesity, hypertension, and altered plasma lipids,
especially hypertriglyceridemia and low high-density lipo-
protein cholesterol. These abnormalities are all risk factors
for cardiovascular disease.
The developing epidemic of obesity and type 2 diabetes in
developed and developing countries is generally thought to
be due to increasing urbanization, with consequent de-
creased levels of exercise in the general population, coupled
with all-day and all-year availability of high-energy food-
stuffs. It is well established that regular exercise is an
effective method of treating insulin resistance and type 2
diabetes, as well as preventing their onset in susceptible
individuals, and it seems likely that the beneficial effects of
exercise are at least partly mediated by AMPK activation. The
ability to more directly test the efficacy of AMPK activation
came with the development of 5-aminoimidazole-4-carbox-
amide ribonucleoside (which is taken up into cells and
converted to the AMP mimetic ZMP) as a method to activate
AMPK in intact cells. In vivo treatment with 5-aminoimida-
zole-4-carboxamide ribonucleoside of several animal models
of insulin resistance and the metabolic syndrome, such as
genetically obese (ob/ob or fa/fa) mice and rats, and fat-fed
rats, showed that the drug was able to reverse glucose
intolerance and insulin resistance, lower plasma triglycerides
and free fatty acids, increase high-density lipoprotein
cholesterol, and even reduce hypertension (see Hardie
(2004)
30
). Further proof of the concept that AMPK activators
had potential for treatment of insulin resistance and type 2
diabetes came with findings that two existing classes of drug
used to treat these conditions, that is, the biguanides (for
example, metformin) and the thiazolidinediones (for exam-
ple, rosiglitazone) can both activate AMPK.
31,32
Both classes
of drug may activate AMPK in part by inhibiting Complex I
of the respiratory chain
33
and thus elevating cellular AMP/
ATP ratios. The thiazolidinediones are also agonists for
peroxisome proliferator-activated receptor-g(PPAR-g)in
adipocytes,
34
through which they are known to increase
the expression and release of adiponectin.
35
Release of
adiponectin explains, at least in part, the therapeutic actions
of thiazolidinediones,
36
although the downstream effects of
adiponectin are, of course, also thought to be mediated by
AMPK. In the case of metformin, there is now good evidence
fatty acid
CO2
glucose
pyruvate
glycogen
triglyceride
glucose
fatty acid fatty acid
glucose
glucose fatty acid
pyruvate CO2
SKELETAL MUSCLE
LIVER
ADIPOSE
TISSUE
Stimulated by AMPK:
Inhibited by AMPK:
acetyl
-CoA
cholesterol
glucose
fatty acid
pyruvate
CO2
glycogen
triglyceride
HEART
Figure 1 Summary of the acute and longer-term changes in carbohydrate and lipid metabolism induced by AMPK activation in the liver, heart, skeletal muscle and
adipose tissue of mammals. Processes stimulated by AMPK activation are shown with thick arrows, those inhibited by AMPK activation are shown by thin arrows with
bars across.
AMPK and energy balance
DG Hardie
S10
International Journal of Obesity
that activation of liver AMPK mediates its antihyperglycemic
effects, because they are completely ablated in a mouse with
a liver-specific knockout of the upstream kinase, LKB1, in
which AMPK can no longer be activated by the drug.
37
It remains unclear at present whether activators of AMPK
would be effective treatments for obesity per se. However,
encouraging pointers in that direction are findings that three
mouse strains that are resistant to diet-induced obesity, that
is: (i) stearoyl-CoA desaturase-1 knockouts;
38
(ii) mice
overexpressing uncoupling protein-1 in white adipocytes;
39
and (iii) mice overexpressing uncoupling protein-3 in
skeletal muscle,
40
all exhibit increased basal activity of
AMPK in the tissues affected.
Conclusions
The AMPK system is switched on either by LKB1, triggered by
increases in cellular AMP/ATP ratio or (in specific cell types)
by elevated Ca
2þ
and CaMKKb. Whatever the upstream
trigger(s), AMPK activation causes a switch from an anabolic
state promoting increased synthesis and storage of glucose,
glycogen, fatty acids, cholesterol and triglycerides, together
with increased cell growth (that is, hypertrophy) and/or
proliferation, to a catabolic state involving oxidation of
glucose, fatty acids and triglycerides, and inhibition of cell
growth and proliferation. Activation of AMPK, either by
increased levels of exercise or by pharmacological means, has
great potential to reverse the metabolic abnormalities of type
2 diabetes and the metabolic syndrome, and perhaps also
obesity.
Acknowledgements
Recent studies in the author’s laboratory have been funded
by Programme Grants from the Wellcome Trust, and by the
EXGENESIS Integrated project (LSHM-CT-2004-005272)
funded by the European Commission.
Conflict of interest
The author states no conflict of interest.
References
1 Hardie DG, Carling D, Sim ATR. The AMP-activated protein
kinaseFa multisubstrate regulator of lipid metabolism. Trends
Biochem Sci 1989; 14: 20–23.
2 Kahn BB, Alquier T, Carling D, Hardie DG. AMP-activated
protein kinase: ancient energy gauge provides clues to modern
understanding of metabolism. Cell Metab 2005; 1: 15–25.
3 Hawley SA, Boudeau J, Reid JL, Mustard KJ, Udd L, Ma
¨kela
¨TP et al.
Complexes between the LKB1 tumor suppressor, STRADa/b and
MO25a/b are upstream kinases in the AMP-activated protein
kinase cascade. J Biol 2003; 2:28.
4 Woods A, Johnstone SR, Dickerson K, Leiper FC, Fryer LG,
Neumann D et al. LKB1 is the upstream kinase in the AMP-
activated protein kinase cascade. Curr Biol 2003; 13: 2004–2008.
5 Alessi DR, Sakamoto K, Bayascas JR. Lkb1-dependent signaling
pathways. Annu Rev Biochem 2006; 75: 137–163.
6 Hardie DG, Salt IP, Hawley SA, Davies SP. AMP-activated protein
kinase: an ultrasensitive system for monitoring cellular energy
charge. Biochem J 1999; 338: 717–722.
7 Hawley SA, Pan DA, Mustard KJ, Ross L, Bain J, Edelman AM et al.
Calmodulin-dependent protein kinase kinase-beta is an alter-
native upstream kinase for AMP-activated protein kinase. Cell
Metab 2005; 2:919.
8 Woods A, Dickerson K, Heath R, Hong SP, Momcilovic M,
Johnstone SR et al. Ca2+/calmodulin-dependent protein kinase
kinase-beta acts upstream of AMP-activated protein kinase in
mammalian cells. Cell Metab 2005; 2: 21–33.
9 Hurley RL, Anderson KA, Franzone JM, Kemp BE, Means AR,
Witters LA. The Ca
2+
/calmoldulin-dependent protein kinase
kinases are AMP-activated protein kinase kinases. J Biol Chem
2005; 280: 29060–29066.
10 Hudson ER, Pan DA, James J, Lucocq JM, Hawley SA, Green KA
et al. A novel domain in AMP-activated protein kinase causes
glycogen storage bodies similar to those seen in hereditary
cardiac arrhythmias. Current Biol 2003; 13: 861–866.
11 Jørgensen SB, Nielsen JN, Birk JB, Olsen GS, Viollet B, Andreelli F
et al. The alpha2-50AMP-activated protein kinase is a site 2
glycogen synthase kinase in skeletal muscle and is responsive to
glucose loading. Diabetes 2004; 53: 3074–3081.
12 Scott JW, Hawley SA, Green KA, Anis M, Stewart G, Scullion GA
et al. CBS domains form energy-sensing modules whose binding
of adenosine ligands is disrupted by disease mutations. J Clin
Invest 2004; 113: 274–284.
13 Burwinkel B, Scott JW, Bu
¨hrer C, van Landeghem FK, Cox GF,
Wilson CJ et al. Fatal congenital heart glycogenosis caused by a
recurrent activating R531Q mutation in the g2 subunit of AMP-
activated protein kinase (PRKAG2), not by phosphorylase kinase
deficiency. Am J Hum Genet 2005; 76: 1034–1049.
14 da Silva Xavier G, Leclerc I, Varadi A, Tsuboi T, Moule SK,
Rutter GA. Role for AMP-activated protein kinase in glucose-
stimulated insulin secretion and preproinsulin gene expression.
Biochem J 2003; 371: 761–774.
15 Minokoshi Y, Alquier T, Furukawa N, Kim YB, Lee A, Xue B et al.
AMP-kinase regulates food intake by responding to hormonal
and nutrient signals in the hypothalamus. Nature 2004; 428:
569–574.
16 Andersson U, Filipsson K, Abbott CR, Woods A, Smith K, Bloom
SR et al. AMP-activated protein kinase plays a role in the control
of food intake. J Biol Chem 2004; 279: 12005–12008.
17 Evans AM, Mustard KJ, Wyatt CN, Peers C, Dipp M, Kumar P et al.
Does AMP-activated protein kinase couple inhibition of
mitochondrial oxidative phosphorylation by hypoxia to
calcium signaling in O
2
-sensing cells? J Biol Chem 2005; 280:
41504–41511.
18 Winder WW, Hardie DG. Inactivation of acetyl-CoA carboxylase
and activation of AMP-activated protein kinase in muscle during
exercise. Am J Physiol 1996; 270: E299–E304.
19 Mu J, Brozinick JT, Valladares O, Bucan M, Birnbaum MJ. A role
for AMP-activated protein kinase in contraction- and hypoxia-
regulated glucose transport in skeletal muscle. Mol Cell 2001; 7:
1085–1094.
20 Sakamoto K, McCarthy A, Smith D, Green KA, Hardie GD,
Ashworth A et al. Deficiency of LKB1 in skeletal muscle
prevents AMPK activation and glucose uptake during
contraction. EMBO J 2005; 24: 1810–1820.
21 Zheng D, MacLean PS, Pohnert SC, Knight JB, Olson AL, Winder WW
et al. Regulation of muscle GLUT-4 transcription by AMP-
activated protein kinase. J Appl Physiol 2001; 91: 1073–1083.
AMPK and energy balance
DG Hardie
S11
International Journal of Obesity
22 Zong H, Ren JM, Young LH, Pypaert M, Mu J, Birnbaum MJ et al.
AMP kinase is required for mitochondrial biogenesis in skeletal
muscle in response to chronic energy deprivation. Proc Natl Acad
Sci USA 2002; 99: 15983–15987.
23 Kola B, Hubina E, Tucci SA, Kirkham TC, Garcia EA, Mitchell SE
et al. Cannabinoids and ghrelin have both central and peripheral
metabolic and cardiac effects via AMP-activated Protein Kinase.
J Biol Chem 2005; 280: 25196–25201.
24 Stahmann N, Woods A, Carling D, Heller R. Thrombin activates
AMP-activated protein kinase in endothelial cells via a pathway
involving Ca2+/calmodulin-dependent protein kinase kinase
beta. Mol Cell Biol 2006; 26: 5933–5945.
25 Tama
´s P, Hawley SA, Clarke RG, Mustard KJ, Green K, Hardie DG
et al. Regulation of the energy sensor AMP-activated protein
kinase by antigen receptor and Ca2+ in T lymphocytes. J Exp Med
2006; 203: 1665–1670.
26 Inoki K, Zhu T, Guan KL. TSC2 mediates cellular energy response
to control cell growth and survival. Cell 2003; 115: 577–590.
27 Horman S, Browne G, Krause U, Patel J, Vertommen D, Bertrand L
et al. Activation of AMP-activated protein kinase leads to the
phosphorylation of Elongation Factor 2 and an inhibition of
protein synthesis. Current Biol 2002; 12: 1419–1423.
28 Imamura K, Ogura T, Kishimoto A, Kaminishi M, Esumi H. Cell
cycle regulation via p53 phosphorylation by a 50-AMP activated
protein kinase activator, 5-aminoimidazole-4-carboxamide-
1-beta-d-ribofuranoside, in a human hepatocellular carcinoma
cell line. Biochem Biophys Res Commun 2001; 287: 562–567.
29 Lowell BB, Shulman GI. Mitochondrial dysfunction and type 2
diabetes. Science 2005; 307: 384–387.
30 Hardie DG. AMP-activated protein kinase: a master switch in
glucose and lipid metabolism. Rev Endocr Metab Disord 2004; 5:
119–125.
31 Fryer LG, Parbu-Patel A, Carling D. The anti-diabetic drugs
rosiglitazone and metformin stimulate AMP-activated
protein kinase through distinct pathways. J Biol Chem 2002;
277: 25226–25232.
32 Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J et al. Role
of AMP-activated protein kinase in mechanism of metformin
action. J Clin Invest 2001; 108: 1167–1174.
33 Brunmair B, Staniek K, Gras F, Scharf N, Althaym A, Clara R et al.
Thiazolidinediones, like metformin, inhibit respiratory complex
I: a common mechanism contributing to their antidiabetic
actions. Diabetes 2004; 53: 1052–1059.
34 Lehmann JM, Moore LB, Smith-Oliver TA, Wilkison WO,
Willson TM, Kliewer SA. An antidiabetic thiazolidinedione
is a high affinity ligand for peroxisome proliferator-activated
receptor gamma (PPAR gamma). J Biol Chem 1995; 270:
12953–12956.
35 Maeda N, Takahashi M, Funahashi T, Kihara S, Nishizawa H,
Kishida K et al. PPARgligands increase expression and plasma
concentrations of adiponectin, an adipose-derived protein.
Diabetes 2001; 50: 2094–2099.
36 Kubota N, Terauchi Y, Kubota T, Kumagai H, Itoh S, Satoh H et al.
Pioglitazone ameliorates insulin resistance and diabetes by both
adiponectin-dependent and -independent pathways. J Biol Chem
2006; 281: 8748–8755.
37 Shaw RJ, Lamia KA, Vasquez D, Koo SH, Bardeesy N,
Depinho RA et al. The kinase LKB1 mediates glucose homeostasis
in liver and therapeutic effects of metformin. Science 2005; 310:
1642–1646.
38 Dobrzyn P, Dobrzyn A, Miyazaki M, Cohen P, Asilmaz E, Hardie
DG et al. Stearoyl-CoA desaturase 1 deficiency increases fatty acid
oxidation by activating AMP-activated protein kinase in liver.
Proc Natl Acad Sci USA 2004; 101: 6409–6414.
39 Matejkova O, Mustard KJ, Sponarova J, Flachs P, Rossmeisl M,
Miksik I et al. Possible involvement of AMP-activated protein
kinase in obesity resistance induced by respiratory uncoupling in
white fat. FEBS Lett 2004; 569: 245–248.
40 Schrauwen P, Hardie DG, Roorda B, Clapham JC, Abuin A,
Thomason-Hughes M et al. Improved glucose homeostasis in
mice overexpressing human UCP3: a role for AMP-kinase? Int J
Obes Relat Metab Disord 2004; 28: 824–828.
AMPK and energy balance
DG Hardie
S12
International Journal of Obesity
... Yu et al. [78] have posited the potential of soluble UA to elicit insulin resistance in the cardiac or cardiomyocyte context. AMPK, a pivotal molecule in the modulation of biological energy metabolism, can be activated by ATP depletion or reduced synthesis, leading to an elevation in AMPK phosphorylation levels [79]. A study demonstrated that HUA stimulates autophagy via the AMPK-ULK1 (unc-51 like kinase 1) pathway, leading to the development of cardiac hypertrophy. ...
Article
Full-text available
dial infarction, arrhythmias, and heart failure. We also combined recent findings from basic research to analyze potential mechanisms linking HUA with myocardial injury. In different pathological models (such as direct action of high uric acid on myocardial cells or combined with myocardial ischemia-reperfusion model), HUA may cause damage by activating the NOD-like receptor protein 3 inflammasome-induced inflammatory response, interfering with cardiac cell energy metabolism, affecting antioxidant defense systems, and stimulating reactive oxygen species production to enhance the oxidative stress response, ultimately resulting in decreased cardiac function. Additionally, we discuss the impact of lowering uric acid intervention therapy and potential safety issues that may arise. However, as the mechanism underlying HUA-induced myocardial injury is poorly defined, further research is warranted to aid in the development novel therapeutic strategies for HUA-related cardiovascular diseases.
... Among the pathways enriched with AKT-1, the AMPK signaling pathway is an important signaling pathway in cell homeostasis maintenance. Adenosine 5 ′ -monophosphate (AMP)activated protein kinase (AMPK) plays a key role in many cells' proliferation-related signaling pathways [47]. AMPK is a complex heterotrimer comprising an α catalytic subunit and two β and γ regulatory subunits. ...
Article
Full-text available
Under ion imbalance, water deficiency, and salt stress, the osmotic pressure of the tree sap increases, and pine wood nematodes (Bursaphelenchus xylophilus, PWN) parasitizing in the trees may be subjected to high-osmotic-pressure stress. KCl, L-malic acid, sucrose, and glycerol solutions were used as osmolytes to explore the highest osmotic concentration that PWN can tolerate. Survival analysis showed that when the treatment concentration exceeded 90%, only a few nematodes in the glycerol group survived under 6 h treatment, and most of the survivors were third-stage dispersal juveniles (DJ3). Further examination revealed that under different concentrations of glycerol-induced high osmotic pressure, the survival rate and body length change rate were the highest in the DJ3 and the lowest in the second-stage propagative juveniles. In order to explore the molecular mechanism of resistance of DJ3 to high osmotic stress, transcriptome sequencing was performed at each developmental stage of PWN and differentially expressed genes that were up-regulated or down-regulated only in DJ3 were screened. The expression of genes related to CoA in DJ3, a key enzyme in metabolism, was significantly higher than the other developmental stages. In addition, the expression of the anti-reversal signal pathway-related gene AKT-1 in DJ3 was significantly lower than in the other development stages. Therefore, the specific expression of genes in DJ3 under high osmotic pressure may help them rapidly produce and accumulate energy-related compounds and activate the adenosine 5′-monophosphate (AMP)-activated protein kinase (AMPK) pathway to respond to damage caused by high-osmotic-pressure stress in time, thus promoting survival.
... The biggest compartment, 'medium', is comprised of a Glucose reservoir (Glu_ex), which feeds into the 'cell' compartment with the rate 'k_in', regulated by the glycolysis capacity (Capacity_G), as described by Reaction 1. The glycolytic capacity aids in regulating the influx of glucose into the cell and is an overall representation of cellular glucose transport which allows regulated influx of glucose to the cell and prevents uncontrolled flooding of the cell with glucose [65][66][67][68]. This regulation was incorporated into the rate equations as 'Capacity_G -Glu', which increases the glucose inflow at low intracellular concentrations, but reduces it as the internal concentration increases, and essentially nullifies the rate when the intracellular glucose (Glu) value reaches the Capacity_G. ...
Article
Full-text available
This study aims to demonstrate the benefits of augmenting commercially available , real-time, in vitro glycolysis assays with phenomenological rate equation-based kinetic models, describing the contributions of the underpinning metabolic pathways. To this end, a commercially available glycolysis assay, sensitive to changes in extracellular acidification (extracellular pH), was used to derive the glycolysis pathway kinetics. The pathway was numerically modelled using a series of ordinary differential rate equations, to simulate the obtained experimental results. The sensitivity of the model to the key equation parameters was also explored. The cellular glycolysis pathway kinetics were determined for three different cell-lines, under nonmodulated and modulated conditions. Over the timescale studied, the assay demonstrated a two-phase metabolic response, representing the differential kinetics of glycolysis pathway rate as a function of time, and this behaviour was faithfully reproduced by the model simulations. The model enabled quantitative comparison of the pathway kinetics of three cell lines, and also the modulating effect of two known drugs. Moreover, the modelling tool allows the subtle differences between different cell lines to be better elucidated and also allows augmentation of the assay sensitivity. A simplistic numerical model can faithfully reproduce the differential pathway kinetics for three different cell lines, with and without pathway-modulating drugs, and furthermore provides insights into the cellular metabolism by elucidating the underlying mechanisms leading to the pathway end-product. This study demonstrates that augmenting a relatively simple, real-time, in vitro assay with a model of the underpinning metabolic pathway provides considerable insights into the observed differences in cellular systems.
... Each subunit has many isoforms, which combine to generate 12 different heterotrimer combinations. AMPK recognizes cellular energy status by self-activation via phosphorylation and allosteric activation, which is responsible for a variety of metabolic functions [23]. Studies showed that AMPK activation increased glucose absorption into cells while decreasing intracellular glucose synthesis which plays an important role in diabetes [24]. ...
Article
Nonalcoholic fatty liver disease (NAFLD) is a worldwide public health issue, but a widely accepted therapy is still lacking until now. Anthocyanins are natural flavonoid compounds that possess various bioactivities,...
Article
In this extensive review work, the important role of AMP-activated protein kinase (AMPK) in causing of diabetes mellitus has been highlighted. Structural feature of AMPK as well its regulations and roles are described nicely, and the association of AMPK with the diabetic complications like nephropathy, neuropathy and retinopathy are also explained along with the connection between AMPK and β-cell function, insulin resistivity, mTOR, protein metabolism, autophagy and mitophagy and effect on protein and lipid metabolism. Published journals were searched on the database like PubMed, Medline, Scopus and Web of Science by using keywords such as AMPK, diabetes mellitus, regulation of AMPK, complications of diabetes mellitus, autophagy, apoptosis etc. After extensive review, it has been found that, kinase enzyme like AMPK is having vital role in management of type II diabetes mellitus. AMPK involve in enhance the concentration of glucose transporter like GLUT 1 and GLUT 4 which result in lowering of blood glucose level in influx of blood glucose into the cells; AMPK increases the insulin sensitivity and decreases the insulin resistance and further AMPK decreases the apoptosis of β-cells which result into secretion of insulin and AMPK is also involve in declining of oxidative stress, lipotoxicity and inflammation, owing to which organ damage due to diabetes mellitus can be lowered by activation of AMPK. As AMPK activation leads to overall control of diabetes mellitus, designing and developing of small molecules or peptide that can act as AMPK agonist will be highly beneficial for control or manage diabetes mellitus.
Article
Scope Allicin, a product of enzymatic reaction when garlic is injured, plays an important role in maintaining glucose homeostasis in mammals. However, the effect of allicin on glucose homeostasis in the state of insulin resistance remains to be elucidated. This study investigates the effect of allicin on glucose metabolism using different muscle fibers in a chicken model. Methods and results Day‐old male Arbor Acres broilers are randomly divided into three groups and fed a basal diet supplemented with 0, 150, or 300 mg kg ⁻¹ allicin for 42 days. Results show that allicin improves the zootechnical performance of broilers at the finishing stage. The glucose loading test (2 g kg ⁻¹ body mass) indicates the regulatory role of allicin on glucose homeostasis. In vitro results demonstrate allicin increases glutathione (GSH) level and the expression of cystathionine γ lyase (CSE), leading to endogenous hydrogen sulfide (H 2 S) production in M. pectoralis major (PM) muscle‐derived myotubes. Allicin stimulates adenosine monophosphate‐activated protein kinase (AMPK) S‐sulfhydration and AMPK phosphorylation to promote glucose uptake, which is suppressed in the presence of d , l ‐propargylglycine (PAG, a CSE inhibitor). Conclusion This study demonstrates that allicin induces AMPK S‐sulfhydration and AMPK phosphorylation to promote glucose uptake via the CSE/H 2 S system in a muscle fiber‐dependent manner.
Article
Full-text available
Endometrial cancer (EC) is the most prevalent gynecological cancer in high-income countries. Its incidence is skyrocketing due to the increase in risk factors such as obesity, which represents a true pandemic. This study aimed to evaluate microRNA (miRNA) expression in obesity-related EC to identify potential associations between this specific cancer type and obesity. miRNA levels were analyzed in 84 EC patients stratified based on body mass index (BMI; ≥30 or <30) and nine noncancer women with obesity. The data were further tested in The Cancer Genome Atlas (TCGA) co-hort, including 384 EC patients, 235 with BMI ≥30 and 149 with BMI <30. Prediction of miRNA targets and analysis of their expression were also performed to identify the potential epigenetic networks involved in obesity modulation. In the EC cohort, BMI ≥30 was significantly associated with 11 deregulated miRNAs. The topmost de-regulated miRNAs were first analyzed in 84 EC samples by single miRNA assay and then tested in the TCGA dataset. This independent validation provided further confirmation about the significant difference of three miRNAs (miR-199a-5p, miR-449a, miR-449b-5p) in normal-weight EC patients versus EC patients with obesity, resulting significantly higher expressed in the latter. Moreover, the three miRNAs were significantly correlated with grade, histological type, and overall survival. Analysis of their target genes revealed that these miRNAs may regulate obesity-related pathways. In conclusion, we identified specific miRNAs associated with BMI that are potentially involved in modulating obesity-related pathways and that may provide novel implications for the clinical management of obese EC patients. K E Y W O R D S BMI, endometrial cancer, miRNA, obesity, personalized medicine
Article
Full-text available
Thiazolidinedione derivatives are antidiabetic agents that increase the insulin sensitivity of target tissues in animal models of non-insulin-dependent diabetes mellitus. In vitro, thiazolidinediones promote adipocyte differentiation of preadipocyte and mesenchymal stem cell lines; however, the molecular basis for this adipogenic effect has remained unclear. Here, we report that thiazolidinediones are potent and selective activators of peroxisome proliferator-activated receptor γ (PPARγ), a member of the nuclear receptor superfamily recently shown to function in adipogenesis. The most potent of these agents, BRL49653, binds to PPARγ with a Kd of approximately 40 nM. Treatment of pluripotent C3H10T1/2 stem cells with BRL49653 results in efficient differentiation to adipocytes. These data are the first demonstration of a high affinity PPAR ligand and provide strong evidence that PPARγ is a molecular target for the adipogenic effects of thiazolidinediones. Furthermore, these data raise the intriguing possibility that PPARγ is a target for the therapeutic actions of this class of compounds.
Article
Full-text available
The AMP-activated protein kinase cascade is activated by elevation of AMP and depression of ATP when cellular energy charge is compromised, leading to inhibition of anabolic pathways and activation of catabolic pathways. Here we show that the system responds in intact cells in an ultrasensitive manner over a critical range of nucleotide concentrations, in that only a 6-fold increase in activating nucleotide is required in order for the maximal activity of the kinase to progress from 10% to 90%, equivalent to a co-operative system with a Hill coefficient (h) of 2.5. Modelling suggests that this sensitivity arises from two features of the system: (i) AMP acts at multiple steps in the cascade (multistep sensitivity); and (ii) the upstream kinase is initially saturated with the downstream kinase (zero-order ultrasensitivity).
Article
Full-text available
Skeletal muscle GLUT-4 transcription in response to treatment with 5-aminoimidazole-4-carboxamide-1-beta-D-ribofuranoside (AICAR), a known activator of AMP-activated protein kinase (AMPK), was studied in rats and mice. The increase in GLUT-4 mRNA levels in response to a single subcutaneous injection of AICAR, peaked at 13 h in white and red quadriceps muscles but not in the soleus muscle. The mRNA level of chloramphenicol acyltransferase reporter gene which is driven by 1,154 or 895 bp of the human GLUT-4 proximal promoter was increased in AICAR-treated transgenic mice, demonstrating the transcriptional upregulation of the GLUT-4 gene by AICAR. However, this induction of transcription was not apparent with 730 bp of the promoter. In addition, nuclear extracts from AICAR-treated mice bound to the consensus sequence of myocyte enhancer factor-2 (from -473 to -464) to a greater extent than from saline-injected mice. Thus AMP-activated protein kinase activation by AICAR increases GLUT-4 transcription by a mechanism that requires response elements within 895 bp of human GLUT-4 proximal promoter and that may be cooperatively mediated by myocyte enhancer factor-2.
Article
Tamas et al. 2006. J. Exp. Med. doi:10.1084/jem.20052469[OpenUrl][1][Abstract/FREE Full Text][2] [1]: {openurl}?query=rft.jtitle%253DJ.%2BExp.%2BMed.%26rft_id%253Dinfo%253Adoi%252F10.1084%252Fjem.20052469%26rft_id%253Dinfo%253Apmid%252F16818670%26rft.genre%253Darticle%26rft_val_fmt%253Dinfo%
Article
The AMP-activated protein kinase is a component of a protein-kinase cascade that is activated by lipid metabolites, and which has recently been fouond to regulate several key enzymes of lipid metabolism. We propose that this system plays an important role in regulating the levels of cholesterol and fatty acids in the body.
Article
Insulin resistance and its dreaded consequence, type 2 diabetes, are major causes of atherosclerosis. Adiponectin is an adipose-specific plasma protein that possesses anti-atherogenic properties, such as the suppression of adhesion molecule expression in vascular endothelial cells and cytokine production from macrophages. Plasma adiponectin concentrations are decreased in obese and type 2 diabetic subjects with insulin resistance. A regimen that normalizes or increases the plasma adiponectin might prevent atherosclerosis in patients with insulin resistance. In this study, we demonstrate the inducing effects of thiazolidinediones (TZDs), which are synthetic PPARgamma ligands, on the expression and secretion of adiponectin in humans and rodents in vivo and in vitro. The administration of TZDs significantly increased the plasma adiponectin concentrations in insulin resistant humans and rodents without affecting their body weight. Adiponectin mRNA expression was normalized or increased by TZDs in the adipose tissues of obese mice. In cultured 3T3-L1 adipocytes, TZD derivatives enhanced the mRNA expression and secretion of adiponectin in a dose- and time-dependent manner. Furthermore, these effects were mediated through the activation of the promoter by the TZDs. On the other hand, TNF-alpha, which is produced more in an insulin-resistant condition, dose-dependently reduced the expression of adiponectin in adipocytes by suppressing its promoter activity. TZDs restored this inhibitory effect by TNF-alpha. TZDs might prevent atherosclerotic vascular disease in insulin-resistant patients by inducing the production of adiponectin through direct effect on its promoter and antagonizing the effect of TNF-alpha on the adiponectin promoter.
Article
There is currently no effective therapy for human prion diseases. However, several polyanionic glycans, including pentosan sulfate and dextran sulfate, prolong the incubation time of scrapie in rodents, and inhibit the production of the scrapie isoform of the prion protein (PrPSc), the major component of infectious prions, in cultured neuroblastoma cells. We report here that pentosan sulfate and related compounds rapidly and dramatically reduce the amount of PrPC, the non-infectious precursor of PrPSc, present on the cell surface. This effect results primarily from the ability of these agents to stimulate endocytosis of PrPC, thereby causing a redistribution of the protein from the plasma membrane to the cell interior. Pentosan sulfate also causes a change in the ultrastructural localization of PrPC, such that a portion of the protein molecules are shifted into late endosomes and/or lysosomes. In addition, we demonstrate, using PrP-containing bacterial fusion proteins, that cultured cells express saturable and specific surface binding sites for PrP, many of which are glycosaminoglycan molecules. Our results raise the possibility that sulfated glycans inhibit prion production by altering the cellular localization of PrPC precursor, and they indicate that endogenous proteoglycans are likely to play an important role in the cellular metabolism of both PrPC and PrPSc.
Article
Malonyl-CoA, an inhibitor of fatty acid oxidation in skeletal muscle mitochondria, decreases in rat skeletal muscle during exercise or in response to electrical stimulation. Regulation of rat skeletal muscle acetyl-CoA carboxylase (ACC), the enzyme that synthesizes malonyl-CoA, was studied in vitro and in vivo. Avidin-Sepharose affinity-purified ACC from hindlimb skeletal muscle was phosphorylated by purified liver AMP-activated protein kinase with a concurrent decrease in ACC activity. AMP-activated protein kinase was quantitated in resuspended ammonium sulfate precipitates of the fast-twitch red (type IIa fibers) region of the quadriceps muscle. Rats running on a treadmill at 21 m/min up a 15% grade show a 2.4-fold activation of AMP-activated protein kinase concurrently with a marked decrease in ACC activity in the resuspended ammonium sulfate precipitates at all citrate concentrations ranging from 0 to 20 mM. Malonyl-CoA decreased from a resting value of 1.85 +/- 0.29 to 0.50 +/- 0.09 nmol/g in red quadriceps muscle after 30 min of treadmill running. The activation of the AMP-activated protein kinase with consequent phosphorylation and inactivation of ACC may be one of the primary events in the control of malonyl-CoA and hence fatty acid oxidation during exercise.
Article
Eukaryotic cells possess systems for sensing nutritional stress and inducing compensatory mechanisms that minimize the consumption of ATP while utilizing alternative energy sources. Such stress can also be imposed by increased energy needs, such as in skeletal muscle of exercising animals. In these studies, we consider the role of the metabolic sensor, AMP-activated protein kinase (AMPK), in the regulation of glucose transport in skeletal muscle. Expression in mouse muscle of a dominant inhibitory mutant of AMPK completely blocked the ability of hypoxia or AICAR to activate hexose uptake, while only partially reducing contraction-stimulated hexose uptake. These data indicate that AMPK transmits a portion of the signal by which muscle contraction increases glucose uptake, but other AMPK-independent pathways also contribute to the response.