Sie sind auf Seite 1von 16

Downloaded from http://perspectivesinmedicine.cshlp.

org/ at NYU MED CTR LIBRARY on October 2, 2018 - Published by Cold


Spring Harbor Laboratory Press

Exercise Metabolism: Fuels for the Fire

Mark Hargreaves1 and Lawrence L. Spriet2


1
Department of Physiology, The University of Melbourne, Victoria 3010, Australia
2
Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario
N1G 2W1, Canada
Correspondence: m.hargreaves@unimelb.edu.au

During exercise, the supply of adenosine triphosphate (ATP) is essential for the energy-
dependent processes that underpin ongoing contractile activity. These pathways involve
both substrate-level phosphorylation, without any need for oxygen, and oxidative phosphor-
ylation that is critically dependent on oxygen delivery to contracting skeletal muscle by the
respiratory and cardiovascular systems and on the supply of reducing equivalents from the
degradation of carbohydrate, fat, and, to a limited extent, protein fuel stores. The relative
contribution of these pathways is primarily determined by exercise intensity, but also mod-
ulated by training status, preceding diet, age, gender, and environmental conditions.
Optimal substrate availability and utilization before, during, and after exercise is critical
for maintaining exercise performance. This review provides a brief overview of exercise
metabolism, with expanded discussion of the regulation of muscle glucose uptake and
fatty acid uptake and oxidation.
www.perspectivesinmedicine.org

he maintenance of contractile activity dur- quiring 300% VO2max, the values would be
T ing exercise is critically dependent on the
supply of adenosine triphosphate (ATP) to the
3.7 mmol ATP . kg21 sec21 and ,2 sec, respec-
tively. Thus, other metabolic pathways must be
myosin, Naþ-Kþ, and sarcoplasmic reticulum activated to resynthesize ATP for the energy-
Ca2þ ATPases that are essential for myofilament dependent processes in contracting skeletal
force production and the maintenance of sarco- muscle. These pathways (Table 1) involve both
lemmal excitability and sarcoplasmic reticulum substrate-level phosphorylation, without any
Ca2þ reuptake and release during excitation – need for oxygen, and oxidative phosphorylation
contraction coupling. Because the intramuscu- that is critically dependent on oxygen delivery to
lar stores of ATP are small (5 mmol . kg21 wet contracting skeletal muscle by the respiratory
muscle), sole reliance on them would only sus- and cardiovascular systems and on the supply
tain exercise for short periods. For example, dur- of reducing equivalents from the degradation
ing submaximal exercise at a power output (200 of fuel stores (Hawley et al. 2014). Although
W) requiring 75% maximal oxygen uptake amino acids can be oxidized by contracting
(VO2max), with an estimated ATP utilization skeletal muscle, they make a relatively minor
rate of 0.4 mmol ATP . kg21 sec21, exercise du- contribution to exercise metabolism, with car-
ration would be 15 sec. During “all-out,” maxi- bohydrates and fat being the primary substrates
mal exercise at a power output (900 W) re- for oxidative metabolism during exercise (Ro-

Editors: Juleen R. Zierath, Michael J. Joyner, and John A. Hawley


Additional Perspectives on The Biology of Exercise available at www.perspectivesinmedicine.org
Copyright # 2018 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a029744
Cite this article as Cold Spring Harb Perspect Med 2018;8:a029744

1
Downloaded from http://perspectivesinmedicine.cshlp.org/ at NYU MED CTR LIBRARY on October 2, 2018 - Published by Cold
Spring Harbor Laboratory Press

M. Hargreaves and L.L. Spriet

Table 1. Energy metabolism in skeletal muscle (Parolin et al. 1999). Despite the increased PDH
ATP utilization activity, the rate of pyruvate production from
ATP þ H2O ! ADP þ Pi þ Hþ þ energy glycolysis is higher than PDH activity, resulting
ATP resynthesis in significant generation of lactate that accumu-
Substrate-level phosphorylation lates in both the muscle and blood. The marked
ADP þ CP þ Hþ ! ATP þ creatine increases in ATP utilization, glycolysis, and
2ADP ! ATP þ AMP strong ion fluxes during such exercise result in
Glycogen þ 3ADP ! 2 lactate þ 2Hþ þ 3ATP metabolic acidosis.
Oxidative phosphorylation The decline in power output (fatigue) dur-
Glucose þ 6O2 þ 36ADP ! 6CO2 þ 6H2O þ ing single and repeated bouts of maximal exer-
36ATP cise is associated with creatine phosphate and
Palmitate þ 23O2 þ 130ADP ! 16CO2 þ glycogen depletion (Spriet et al. 1989; Casey
16H2O þ 130ATP
et al. 1996; Hargreaves et al. 1998), accumula-
ATP, Adenosine triphosphate; ADP, adenosine di- tion of metabolic by-products (e.g., Hþ, ADP,
phosphate.
AMP, Pi) (Spriet et al. 1989; Casey et al. 1996;
Hargreaves et al. 1998), and hyperkalemia
mijn et al. 1993; van Loon et al. 2001). The met- (Medbø and Sejersted 1990), which singly and
abolic responses to exercise have been well stud- in combination impact on excitation – contrac-
ied and described since the early 20th century. tion coupling processes within skeletal muscle
This review provides a brief overview of exercise (Allen et al. 2008).
metabolism and more in-depth summaries of During submaximal exercise, the oxidative
skeletal muscle glucose uptake and glucose metabolism of carbohydrates and lipids pro-
transporter type 4 (GLUT4) translocation and vides almost all of the ATP required for contrac-
www.perspectivesinmedicine.org

skeletal muscle fatty acid (FA) oxidation during tile activity. The major substrates for oxidation
exercise. are muscle glycogen and blood glucose derived
from liver glycogenolysis and gluconeogenesis
and the gut when carbohydrate is ingested, and
OVERVIEW OF EXERCISE METABOLISM
FAs derived from both adipose tissue and intra-
The major determinants of the relative con- muscular triglyceride (IMTG) breakdown. The
tribution of the energy-generating pathways relative contribution of these substrates is largely
(Fig. 1; Table 1) during exercise are the intensity determined by exercise intensity (Fig. 3) (Ro-
and duration of exercise. During maximal, “all- mijn et al. 1993; van Loon et al. 2001) and
out” exercise, with a peak power output duration, but is also influenced by training sta-
of 900 W, which declines over the next tus, gender, preceding diet, and environmental
30 sec, the degradation of creatine phosphate conditions, although these latter factors are be-
and of glycogen to lactate provide the majority yond the scope of the current review. At lower
of ATP, while oxidative phosphorylation ac- intensities lipid oxidation dominates, but with
counts for 25% – 30% of energy turnover increasing exercise intensity there is greater reli-
(Fig. 2). The rapid increase in muscle glycogen- ance on muscle glycogen and blood glucose.
olysis is the result of activation of glycogen phos- Maximal rates of fat oxidation, from both extra-
phorylase by increased sarcoplasmic [Ca2þ] and and intramuscular FAs, occur at 60%– 65%
inorganic phosphate (Pi), elevated cyclic AMP VO2max, declining at higher intensities as a result
secondary to increased circulating adrenaline, of reduced plasma FA delivery to contracting
and allosteric activation by ATP breakdown skeletal muscle and lower rates of mitochondrial
products (AMP, ADP, and IMP). The increase FA uptake and oxidation, secondary to increased
in Ca2þ, along with elevated muscle pyruvate glycolytic flux (Romijn et al. 1995; Spriet 2014).
levels caused by increased glycolysis, activates Increased muscle glycogenolysis at higher exer-
pyruvate dehydrogenase (PDH), the rate-limit- cise intensities is the result of enhanced glycogen
ing enzyme for muscle carbohydrate oxidation phosphorylase activity, secondary to elevated

2 Cite this article as Cold Spring Harb Perspect Med 2018;8:a029744


www.perspectivesinmedicine.org

Albumin FFA (Blood flow x concn) HbO2


Glucose O2

Blood (FABPpm; FAT/CD36; FATP) (GLUT1; GLUT4)


PM
Cytosol
Glucose Glycogen
ATG lipase
HS lipase HK Glycogen
MG lipase phosphorylase
FFA FABPc
ATPase
G-6-P G-1-P
TG Fatty acyl-CoA
ATP CK ADP + Pi
Glycerol synthase PFK
phosphate NAD+
acyltransferase Fatty Cr mtCK PCr
(GPAT-1; GPAT-2) acyl-CoA
ATP NADH
Diacylglycerol ATP ADP
acyltransferase
(DGAT) Pyruvate Lactate
LDH
CPT-I
mtOM
Spring Harbor Laboratory Press

(ACT) (FAT/CD36) (MCT) (ANT) Pi


mtIM

Cite this article as Cold Spring Harb Perspect Med 2018;8:a029744


CPT-II NAD+ PDH ADP
ATP Pi
Matrix NADH CO2 + Pi transporter

β-Oxidation H+
Fatty Acetyl-CoA F0-F1 ATPase
acyl-CoA
NAD+
NAD+ NADH TCA
cycle E
T
NADH C
ATP H+ H+
CO2
H2O O2
Downloaded from http://perspectivesinmedicine.cshlp.org/ at NYU MED CTR LIBRARY on October 2, 2018 - Published by Cold

Figure 1. (Legend continues on following page.)


Exercise Metabolism

3
Downloaded from http://perspectivesinmedicine.cshlp.org/ at NYU MED CTR LIBRARY on October 2, 2018 - Published by Cold
Spring Harbor Laboratory Press

M. Hargreaves and L.L. Spriet

Pcr hydrolysis in production of lactate that accumulates in


Glycolysis muscle and blood (Spriet et al. 2000). Although
15 Oxidative phosphorylation lactate is often considered simply a metabolic
by-product, it is an important substrate for

(mmol · kg dw–1 · s–1)


12
oxidative metabolism and gluconeogenesis,
ATP turnover rate
9 thereby providing a link between glycolytic
and oxidative metabolism (Brooks 2009).
6 During prolonged, strenuous (75%
VO2max) exercise, there is a progressive decline
3 in the rate of muscle glycogenolysis and lipoly-
sis, increased muscle glucose uptake, and a pro-
0
0 6 15 30 gressive increase in FA oxidation with elevated
Time (sec) plasma FA levels (Romijn et al. 1993) with in-
creasing exercise duration. Fatigue during such
Figure 2. Contributions of phosphocreatine (PCr),
exercise is often associated with muscle and liver
glycolysis, and oxidative phosphorylation to adeno-
sine triphosphate (ATP) turnover during 30 sec of glycogen depletion and hypoglycemia, with a
maximal isokinetic cycling exercise. (From Parolin consequent decrease in muscle carbohydrate
et al. 1999; reprinted, with permission, from the au- oxidation and neuroglucopenia. Increased die-
thors.) tary carbohydrate intake to maximize pre-exer-
cise carbohydrate stores (Hawley et al. 1997)
and carbohydrate ingestion during exercise
sarcoplasmic Ca2þ, Pi, and cyclic AMP follow-
(Cermak and van Loon 2013) are effective nu-
ing adrenaline stimulation and allosteric activa-
tritional strategies to enhance endurance exer-
www.perspectivesinmedicine.org

tion by ATP breakdown products (Howlett et al.


cise performance.
1998; Watt et al. 2001). Glucose uptake also in-
creases in relation to exercise intensity, although
glucose utilization may decrease during intense
EXERCISE-INDUCED MUSCLE GLUCOSE
exercise because of high rates of muscle glyco-
UPTAKE AND GLUT4 TRANSLOCATION
genolysis and glucose-6-phosphate-mediated
inhibition of hexokinase activity (Katz et al. Although muscle glycogen is quantitatively
1986). Accompanying the increase in skeletal more important as a carbohydrate fuel source
muscle glucose uptake is enhanced liver glucose for contracting skeletal muscle during exercise,
output, initially from liver glycogenolysis but blood glucose nevertheless makes an impor-
during more prolonged exercise from increased tant contribution to overall carbohydrate oxi-
rates of gluconeogenesis. Rates of carbohy- dation, especially during prolonged, strenuous
drate oxidation are increased with increasing ex- exercise. There has been considerable interest
ercise intensity in parallel, with PDH activation in the regulation of skeletal muscle glucose up-
(Howlett et al. 1998). Despite the increased PDH take during exercise, because it is preserved in
activity, accelerated rates of glycolysis also result states of insulin resistance such as type 2 dia-

Figure 1. (Continued) Schematic overview of skeletal muscle metabolism. Hb, Hemoglobin; FFA, free fatty acid;
FABPpm and FABPc, fatty acid binding protein –plasma membrane and cytoplasm; FAT/CD36, fatty acid trans-
locase; FATP, fatty acid transport protein; GLUT1 and 4, glucose transport proteins 1 and 4; PM, plasma
membrane; ATG, HS, and MG lipases, adipocyte glyceride, hormone-sensitive, and monoglyceride lipases;
mtOM and mtIM, outer and inner mitochondrial membranes; CPT-I and -II, carnitine palmitoyl transferase
I and II; ACT, acylcarnitine transferase; G-1-P and G-6-P, glucose 1 and 6 phosphate; HK, hexokinase; PFK,
phosphofructokinase; LDH, lactate dehydrogenase, MCT, monocarboxylate transport proteins; PDH, pyruvate
dehydrogenase; TCA, tricarboxylic acid; ANT, adenine nucleotide transport protein; Cr and PCr, creatine and
phoshocreatine; CK and mtCK, creatine kinase and mitochondrial CK.

4 Cite this article as Cold Spring Harb Perspect Med 2018;8:a029744


Downloaded from http://perspectivesinmedicine.cshlp.org/ at NYU MED CTR LIBRARY on October 2, 2018 - Published by Cold
Spring Harbor Laboratory Press

Exercise Metabolism

300 Muscle glycogen are at their highest (Katz et al. 1991). That
Muscle triglycerides said, glucose delivery remains an important
Plasma FFA determinant of muscle glucose uptake during
Plasma glucose
exercise (Zinker et al. 1993) and GLUT4 is es-

cal · kg–1 · min–1


200
sential for exercise-induced muscle glucose up-
take (Howlett et al. 2013).
Given the fundamental importance of
100 GLUT4 expression and translocation for con-
traction-stimulated muscle glucose transport,
it is not surprising that there has been con-
siderable research undertaken to identify the
0
25 65 85 underlying signals and molecular mechanisms
% of VO2max responsible for increased glucose transport dur-
ing contractions/exercise. Investigators have
Figure 3. Relative contribution of carbohydrate and used numerous in vitro, in situ, and in vivo ex-
fat fuel sources to energy metabolism during exercise
perimental models in various species, a range of
of increasing intensity. FFA, Free fatty acid. (From
Romijn et al. 1993; reprinted, with permission, pharmacological inhibitors of critical enzymes,
from the American Physiological Society # 1993.) and transgenic and knockout models. Muscle
contraction/exercise is a complex stimulus that
generates multiple signals, including increased
betes (Martin et al. 1995). It is well known that sarcoplasmic [Ca2þ], mechanical stress/force,
the effects of insulin (when increased before or metabolic perturbations (increased AMP/ATP
during exercise) and exercise on skeletal muscle and decreased creatine phosphate and glyco-
www.perspectivesinmedicine.org

glucose uptake are additive/synergistic (De- gen), changes in redox state associated with in-
Fronzo et al. 1981; Wasserman et al. 1991), creased levels of reactive oxygen species (ROS)
but mediated via different mechanisms. Thus, and increased nitric oxide (NO). In turn, such
identification of the mechanisms by which ex- signals activate various signaling pathways and
ercise increases glucose uptake may lead to op- protein kinases, notably Ca2þ/calmodulin-de-
timization of exercise interventions and to
novel therapeutic strategies to manage and pre-
vent insulin resistance. Skeletal muscle glucose GLUT4
uptake occurs by facilitated diffusion and there 43 kDa >
are three sites of regulation: (1) glucose deliv-
ery, (2) sarcolemmal glucose transport, medi- 0 min 5 min 40 min
ated by the GLUT4 glucose transporter, and
(3) glucose phosphorylation by hexokinase
6 1.5
Transport (pmol/μg/min)

and subsequent metabolism (Wasserman


GLUT4 (arbitrary units)
Transport
et al. 2011). Although glucose transport is GLUT4 ** **
thought to be rate-limiting under resting con- 4 1.0
* *
ditions, the large increases in glucose delivery,
secondary to skeletal muscle hyperemia, and 2 0.5
glucose transport caused by rapid GLUT4
translocation to surface membranes (Fig. 4) 0 0.0
(Kristiansen et al. 1997) mean that glucose 0 5 40
phosphorylation becomes an important site Time (min)
of regulation during exercise. This is perhaps Figure 4. Sarcolemmal vesicle glucose transporter
most evident during intense exercise (Katz type 4 (GLUT4) expression and glucose transport
et al. 1986) and the early stages of prolonged during exercise. (From Kristiansen et al. 1997; re-
exercise when rates of muscle glycogenolysis printed, with permission, from the authors.)

Cite this article as Cold Spring Harb Perspect Med 2018;8:a029744 5


Downloaded from http://perspectivesinmedicine.cshlp.org/ at NYU MED CTR LIBRARY on October 2, 2018 - Published by Cold
Spring Harbor Laboratory Press

M. Hargreaves and L.L. Spriet

pendent protein kinase (CaMK), protein kinase upstream kinase liver kinase B1 (LKB1) (Saka-
C (PKC), and AMP-activated protein kinase moto et al. 2005; Jeppesen et al. 2013). The latter
(AMPK). These kinases act on downstream tar- intervention could have impacted on AMPK, or
gets, notably TBC1D1 (tre-2/USP6, BUB2, on other AMPK-related kinases targeted by
cdc16 domain family member 1) and AS160 LKB1 such as sucrose nonfermenting AMPK-
(Akt substrate of 160 kDa or TBC1 domain fam- related kinase (SNARK) (Koh et al. 2010). Per-
ily member 4—TBC1D4) that are involved in haps the most convincing study is one in which
the regulation of GLUT4 vesicle trafficking. the regulatory b subunits of AMPK were ab-
Given the fundamental importance of Ca2þ lated, resulting in loss of the catalytic a subunits
in muscle contraction, there has been a long- and marked attenuation of glucose uptake dur-
standing view that it also stimulates glucose ing in vitro muscle contractions and in vivo ex-
transport. Studies utilizing caffeine, to induce ercise (O’Neill et al. 2011). Although the defin-
sarcoplasmic reticulum Ca2þ release, pharma- itive evidence confirming the essential role of
cological CaMK inhibitors, and overexpression AMPK in contraction/exercise-stimulated glu-
of a CaMKII inhibitory peptide in skeletal mus- cose uptake is perhaps lacking, it is teleologically
cle have implicated Ca2þ in the stimulation of attractive that an energy-sensing kinase would
contraction-stimulated muscle glucose trans- regulate skeletal muscle glucose uptake during
port (Wright et al. 2004; Witczak et al. 2010). exercise.
In contrast, a recent study has suggested that Other factors involved in the stimulation of
calcium effects, in the absence of force genera- glucose uptake during contractions/exercise in-
tion, are mediated via activation of energy-de- clude ROS (Merry et al. 2010; Katz 2016) and
pendent processes within muscle that, in turn, NO (Bradley et al. 1999; Merry et al. 2010). ROS
increase AMPK activity and glucose transport appear to be more important during heavy ex-
www.perspectivesinmedicine.org

(Jensen et al. 2014). These investigators con- ercise, because antioxidant treatment did not
cluded that AMPK activation and mechanical impact glucose uptake during mild to moderate
stress, not sarcoplasmic Ca2þ release, were the exercise (Katz 2016), the significance of which
primary mediators of contraction-stimulated remains to be determined. Despite good evi-
glucose uptake. Another calcium-sensitive ki- dence supporting a role for NO in stimulating
nase is PKC; however, knockout of PKCa, the glucose uptake during exercise, the source of
major isoform in skeletal muscle, did not have NO and the downstream mechanisms ultimate-
any effect on contraction-stimulated glucose ly effecting GLUT4 translocation and increased
uptake (Jensen et al. 2009). sarcolemmal glucose transport remain to be
AMPK is an important energy-sensing fully elucidated (Hong et al. 2014). Mechanical
kinase expressed in skeletal muscle that is acti- stress/force is a key element of the muscle con-
vated by exercise in an intensity-dependent traction cycle and recently it has been proposed
manner (Friedrichsen et al. 2013; Hardie et al. that the Rho family GTPase Rac1, also associ-
2014) and has been implicated in contraction- ated with the actin cytoskeleton, has an impor-
stimulated glucose uptake (Friedrichsen et al. tant role in contraction/exercise-stimulated
2013). That said, it has been difficult to conclu- muscle glucose uptake (Sylow et al. 2013) via
sively confirm a direct link between AMPK ac- effects on GLUT4 translocation (Sylow et al.
tivity and glucose uptake during exercise. The 2016).
first study to assess this question overexpressed a Important downstream targets of the
kinase-dead AMPK mutant in skeletal muscle above-mentioned signaling cascades are the Rab
and observed a reduction in muscle glucose GTPase-activating proteins TBC1D1 and AS160
transport during contractions (Mu et al. 2001). that are involved in the interactions between
Various studies utilizing knockout of the cata- Rab proteins and GLUT4 vesicles during the
lytic a subunits of AMPK have produced con- various events required for translocation of
flicting results (Friedrichsen et al. 2013), as have GLUT4 from intracellular sites to the surface
studies that have altered activity of the primary membranes. Because insulin and muscle con-

6 Cite this article as Cold Spring Harb Perspect Med 2018;8:a029744


Downloaded from http://perspectivesinmedicine.cshlp.org/ at NYU MED CTR LIBRARY on October 2, 2018 - Published by Cold
Spring Harbor Laboratory Press

Exercise Metabolism

tractions/exercise result in distinct and different that of carbohydrate, including the protein-me-
phosphorylation signatures of both these pro- diated transport of fat into muscle and the deg-
teins (Kramer et al. 2006; An et al. 2010; Treebak radation of stored IMTG (Kiens 2006; Glatz
et al. 2014; Cartee 2015), it has been suggested et al. 2010; Spriet 2012, 2014). However, other
that these proteins may be important sites of control sites are unique to fat metabolism, in-
convergence of the insulin and contraction/ex- cluding the binding of fat to a protein chaper-
ercise signaling pathways, thereby accounting one and transport in the cytoplasm, and the
for the additive effects of these two stimuli protein-mediated transport of fat into the mi-
and potentially contributing to enhanced mus- tochondria (Campbell et al. 2004; Bezaire et al.
cle insulin sensitivity in the postexercise period 2006). Later work identified the presence of fat
(Cartee 2015). transport proteins in the transverse tubule
A well-known adaptation to exercise train- membranes and translocation of transport pro-
ing is a reduction in carbohydrate oxidation teins to these membranes occurs during exercise
during exercise associated with reduced glucose (Stefanyk et al. 2012).
turnover and oxidation, at least at the same Other recent discoveries include the identi-
absolute power output (Coggan et al. 1990, fication of the enzyme adipose tissue glyceride
1995b). The training effects on glucose turnover lipase (ATGL) that works in concert with hor-
are sometimes less apparent during exercise at mone-sensitive lipase (HSL) to regulate skeletal
the same relative exercise intensity (Friedlander muscle lipolysis (Zimmermann et al. 2004; Watt
et al. 1997), but still existent in cross-sectional and Spriet 2010) and the presence of perilipin-
comparisons (Coggan et al. 1995a). The de- like proteins coating the lipid droplets in skel-
crease in glucose uptake during exercise after etal muscle (MacPherson and Peters 2015). An-
training is also associated with a lower liver other possible site of regulation that has not
www.perspectivesinmedicine.org

glucose output because of reductions in both been explored is control within the b-oxidation
hepatic glycogenolysis and gluconeogenesis pathway. Last, there is the overarching aspect of
(Coggan et al. 1995b). At maximal exercise in- the regulation of skeletal muscle fat oxidation—
tensities, there may be an increase in muscle that the mitochondrial volume (the total
glucose uptake associated with an increased ca- amount of fat transport and metabolizing pro-
pacity for glucose transport caused by increased teins) determines the overall capacity to oxidize
muscle GLUT4 protein expression following fat (Perry et al. 2008; Holloway and Spriet 2009).
training (Kristiansen et al. 2000). This increase
in GLUT4 expression is also an important factor
EXERCISE-INDUCED MUSCLE FATTY ACID
underlying enhanced muscle insulin sensitivity
UPTAKE AND FAT TRANSPORT PROTEIN
and muscle glycogen storage in the trained state.
TRANSLOCATION
The molecular mechanisms responsible for the
increase in muscle GLUT4 expression following Although a small amount of fat can diffuse
exercise training include many of the pathways through the lipid bilayer of the muscle mem-
responsible for the increases in glucose trans- brane into the muscle cell, it is now widely
port with acute exercise (for a review, see Rich- accepted that the major portion of the FAs
ter and Hargreaves 2013). that enter muscle do so via protein-mediated
mechanisms (Bonen et al. 2007; Holloway et al.
2008; Glatz et al. 2010). This involves actual
SKELETAL MUSCLE FATTY ACID
transport of FA across the muscle membrane
OXIDATION
by carrier proteins and/or facilitation of their
Our understanding of the regulation of fat me- movement across the membrane by initial
tabolism in skeletal muscle during exercise lags binding to transport proteins. The major trans-
behind that of carbohydrate metabolism. Re- port proteins include the plasma membrane
search in the past 15 years has shown that fatty acid– binding protein (FABPpm) located
many sites of control are similar in location to on the outer leaflet of the plasma membrane,

Cite this article as Cold Spring Harb Perspect Med 2018;8:a029744 7


Downloaded from http://perspectivesinmedicine.cshlp.org/ at NYU MED CTR LIBRARY on October 2, 2018 - Published by Cold
Spring Harbor Laboratory Press

M. Hargreaves and L.L. Spriet

a family of fatty acid transport proteins (mainly membrane content of FABPpm, but not the
FATP1, 4) that have many transmembrane do- content of FAT/CD36 (Fig. 5) (Bonen et al.
mains, and, most importantly, the fatty acid 1999; Talanian et al. 2010).
translocase (FAT/CD36) protein that has two A critical finding was that FAT/CD36 acute-
transmembrane domains. Much of the pio- ly translocated from an intracellular pool to the
neering research was performed in red and muscle membrane in rodent and human skele-
white rodent skeletal muscle in which the in- tal muscle during a single bout of muscle con-
vestigators showed a strong relationship be- tractions, very similar to GLUT4 (Bonen et al.
tween the expression and protein content of 2000; Bradley et al. 2012).
the putative transporters and actual FA uptake It was subsequently shown that muscle con-
(Bonen et al. 1998). The messenger RNA tractions also increased the plasma membrane
(mRNA) abundance and protein content of content of FABPpm, FATP1 and 4 proteins in
the FA transporters in the plasma membrane mouse skeletal muscle (Jain et al. 2009), and
and the FA transport capacity were several- that the membrane content of FAT/CD36,
fold higher in red oxidative rodent muscle FABPpm, and FATP4 but not FATP1 correlated
(high capacity for fat metabolism) versus white highly with the capacities for oxidative metabo-
glycolytic muscle (Bonen et al. 1998). The lism and FA oxidation in six different rat skeletal
transport of FA also appeared to be a saturable muscles (Nickerson et al. 2009). These investi-
process in sarcolemmal vesicles prepared from gators then overexpressed each of the four trans-
both red and white rat muscles. This corrobo- port proteins independently within a normal
rated earlier work in rat hind limb and human physiological range, without affecting the total
skeletal muscle exposed to high FA availability or plasma membrane content of the other three
(Turcotte et al. 1991, 1992). Chronic electrical transport proteins (Nickerson et al. 2009). All
www.perspectivesinmedicine.org

stimulation in rodents and exercise training in transport proteins increased FA transport but
humans increased total muscle FABPpm and FAT/CD36 and FATP4 were two times more ef-
FAT/CD36 protein content and the plasma fective than FABPpm, and FATP1 and all trans-

5
*
30 *
Palmitate uptake (pmol/mg prot/10 sec)

4
25
(arbitrary densitometry units)
FAT/CD36 protein

20 3

15
2

10

1
5

0 0
Control Stimulated Control Stimulated
Condition

Figure 5. Fatty acid translocase (FAT/CD36) protein expression in and palmitate uptake by giant sarcolemmal
vesicles obtained from control and chronically stimulated rat skeletal muscle. (From Bonen et al. 1999; reprinted,
with permission, from the American Physiological Society # 1999.)

8 Cite this article as Cold Spring Harb Perspect Med 2018;8:a029744


Downloaded from http://perspectivesinmedicine.cshlp.org/ at NYU MED CTR LIBRARY on October 2, 2018 - Published by Cold
Spring Harbor Laboratory Press

Exercise Metabolism

porters also increased FA oxidation but FAT/ ulated and FAT/CD36 and FABPpm contents
CD36 and FABPpm were three times more effec- were also lower (Fentz et al. 2016). These results
tive than FATP1 and 4 (Nickerson et al. 2009). suggest that AMPKa was required for normal FA
Taken together, these data showed that the metabolism during muscle contractions and ex-
movement of fat into skeletal muscle during ex- ercise (Fentz et al. 2016). Another recent report
ercise is a highly regulated process involving sev- showed that mice with a muscle-specific knock-
eral transport proteins that are responsive to out of LKB1 had decreased FA oxidation during
both the acute and chronic need for fat as a treadmill exercise and decreased expression of
fuel source. However, there has been less work genes involved in FA oxidation (Jeppesen et al.
to clarify the factors that activate the transloca- 2013). As LKB1 is upstream of AMPK, it may
tion of fat transporter proteins to the muscle also influence FA metabolism independently of
membrane during exercise. It is expected that AMPK in skeletal muscle.
Ca2þ and the factors related to the energy status As discussed earlier, Akt is a family of pro-
of the cell (e.g., free ADP, Pi, and AMPK activa- tein kinases regulating multiple anabolic path-
tion) would be involved as they play an impor- ways with three isoforms described. Akt2 is
tant role in activating the transport and docking involved in insulin signaling and contributes
of GLUT4 into the muscle membrane. However, to the regulation of lipid metabolism. However,
the time course of the changes involved in up- it is not known whether the regulation of lipid
regulating FA uptake and oxidation during exer- metabolism by Akt2 signaling extends to the
cise is generally believed to be slower than the FA transport process. In muscle from Akt2-
activation of glucose uptake during exercise. knockout mice, Akt2 was important for normal
In skeletal muscle, contraction and the activa- contraction-induced FA transport and translo-
tion of muscle AMPK by the pharmacological cation of FAT/CD36 and FATP1, but not trans-
www.perspectivesinmedicine.org

activator 5-aminoimidazole-4-carboxamide-1- location of FABPpm and FATP4 (Jain et al.


b-D-ribofuranoside (AICAR) produced translo- 2015). Instead, Akt2 mediated intracellular re-
cation of both GLUT4 and FAT/CD36 to the tention of FABPpm and FATP4. It has also been
sarcolemma, albeit with a different time-course shown that MUNC18c, which is present in skel-
(Cushman et al. 1998; Jain et al. 2009), as well as etal muscle plasma membrane and has binding
that of a number of other FA transporters, specificity for the tSNARE protein syntaxin 4
namely, FABPpm, FATP1, and FATP4 but not and which plays a role in GLUT4 trafficking, is
FATP6 (Jain et al. 2009). The working hypothesis not involved in FA trafficking in skeletal muscle
is that Ca2þ and AMPK play important roles in (Jain et al. 2012).
the translocation of FA transporters to the mus- One last point warrants mentioning. The
cle membrane. Turcotte et al. (2005) showed that long-held view in skeletal muscle has been that
inhibition of extracellular regulated kinase 1/2 the mitochondrial volume or the total amount
(ERK1/2), stimulated by Ca2þ-dependent PKC, of fat transport and metabolizing proteins
prevented the normal contraction-induced in- determines the overall capacity to oxidize fat
crease in plasma membrane FAT/CD36 content (Perry et al. 2008; Holloway and Spriet 2009).
and FA uptake in rodent muscle. A study by Jep- However, this point of view has been recently
pesen et al. (2011) using AMPKa2-null mice challenged by reports showing that up-regula-
suggested that skeletal muscle FAT/CD36 traf- tion of sarcolemmal FAT/CD36 protein in ro-
ficking and FA uptake was independent of dent skeletal muscle increases FA uptake and
AMPK as all responses to contraction and exer- oxidation and whole-body exercise perfor-
cise were normal when compared with wild-type mance independent of changes in mitochondri-
skeletal muscles. However, when AMPKa1 and al content or machinery (McFarlan et al. 2012;
a2 were specifically knocked out in mouse skel- Yoshida et al. 2013). What needs to be deter-
etal muscle, animals oxidized less fat during mined is whether these changes occur indepen-
treadmill exercise; muscle isolated from these dently in an intact whole-body situation. In
animals had lower FA oxidation rates when stim- summary, there is much work to be performed

Cite this article as Cold Spring Harb Perspect Med 2018;8:a029744 9


Downloaded from http://perspectivesinmedicine.cshlp.org/ at NYU MED CTR LIBRARY on October 2, 2018 - Published by Cold
Spring Harbor Laboratory Press

M. Hargreaves and L.L. Spriet

to elucidate the contractile-induced signals and et al. 2009; Turnbull et al. 2016). It was also
mechanisms responsible for the movement of implicated to play a significant role during ex-
FA transporters to the muscle membrane and ercise, as lipolysis was maintained when HSL
transverse tubules in skeletal muscle in response was pharmacologically inhibited or knocked
to acute and chronic exercise (Turcotte and Ab- out in rodent skeletal muscle (Alsted et al.
bott 2012). 2013). ATGL is activated by association with a
protein called comparative gene identification-
58 (CGI-58) and putatively inhibited by G(0)/
EXERCISE-INDUCED INTRAMUSCULAR
G(1) switch gene-2 protein (G0S2) (MacPher-
TRIGLYCERIDE BREAKDOWN
son et al. 2013a; Turnbull et al. 2015, 2016).
Skeletal muscle has a large amount of stored fat There was the early suggestion that the relative
in lipid droplets, especially in physically trained amount of CGI-58-to-ATGL and G0S2-to-
individuals. This IMTG or intramyocellular ATGL protein controlled the regulation of
lipid (IMCL) can be degraded and used as an ATGL. However, recent work with rodent mus-
aerobic fuel during low to moderate exercise cles of varying oxidative potentials at rest, dur-
(Fig. 3), as has been shown many times (Romijn ing contraction, and following training did not
et al. 1993; Stellingwerff et al. 2007; Loher et al. support this contention, suggesting that regula-
2016). The key enzymes involved in regulating tion must occur through more complicated
lipolysis in skeletal muscle are ATGL, HSL, and mechanisms (Turnbull et al. 2015, 2016).
monoglyceride lipase (MGL) that sequentially Perilipin 1 (PLIN1) is a member of the lipid
remove an FA from the triglyceride (TG) stored droplet – associated (PLIN) family of proteins
in lipid droplets. ATGL and HSL are highly reg- that is involved in the storage and mobilization
ulated, whereas MGL is not (Watt 2009; Watt and use of FAs in cells. PLIN1 has been impli-
www.perspectivesinmedicine.org

and Spriet 2010; Alsted et al. 2013). It is also cated in regulating lipolysis in adipose tissue by
important to note that other factors play a interacting with the TG lipases. However, a re-
role in TG breakdown, including droplet size, cent review reported that PLIN1 is not found in
droplet localization, and the fact that lipid is skeletal muscle and it is not clear whether any of
stored in droplets with a protein ( perilipins) the PLINs found in skeletal muscle (PLIN2, 3,
coating (Loher et al. 2016). The perilipin pro- and 5) are involved in regulating lipolysis in a
teins appear to separate IMTG from ATGL and manner similar to PLIN1 in adipose tissue
HSL, maintaining low rates of lipolysis at rest. (MacPherson and Peters 2015). These investi-
During moderate-intensity exercise, Ca2þ and gators cited evidence that all three skeletal mus-
adrenaline-related events phosphorylate HSL cle PLINs interact with HSL and may regulate
(Watt et al. 2003; Talanian et al. 2006) and the location of HSL or its activity on the surface
AMPK phosphorylation of perilipin is involved of the lipid droplet (MacPherson et al. 2013a,b).
in recruiting both HSL and ATGL to the lipid Interestingly, all three PLINs also interact with
droplet, collectively enhancing rates of IMTG ATGL, PLIN3 and 5 interact with CGI-58, and it
hydrolysis (Prats et al. 2006). However, at higher has been suggested that they act as locating and
power outputs, it appears that AMPK phos- scaffolding proteins for ATGL until a lipolytic
phorylates additional sites on HSL that inhibit stimulus appears (MacPherson and Peters
the phosphorylation by adrenaline and Ca2þ, 2015). However, these investigators point out
providing a potential mechanism for the lower that PLIN5 is also associated with and translo-
rates of IMTG use reported at these higher in- cates to the mitochondria suggesting that it may
tensities (Watt et al. 2003). This explanation of play a role in the regulation of FA oxidation and
events during exercise does not include a role not IMTG lipolysis.
for ATGL in skeletal muscle, but work in this In summary, although it is clear that IMTG
area is ongoing. It has been recently shown that lipolysis plays an important role in providing FA
ATGL content increased in rodent and human for oxidation during exercise (Fig. 3), under-
skeletal muscle with exercise training (Alsted standing the regulation that occurs at the level

10 Cite this article as Cold Spring Harb Perspect Med 2018;8:a029744


Downloaded from http://perspectivesinmedicine.cshlp.org/ at NYU MED CTR LIBRARY on October 2, 2018 - Published by Cold
Spring Harbor Laboratory Press

Exercise Metabolism

of the lipid droplet for FAs to be released and rates were lower in animals with no FAT/
delivered to the surface of the mitochondria CD36 (Holloway et al. 2009). In human skeletal
remains largely unknown. muscle, moderate-intensity exercise acutely in-
creased the mitochondrial membrane FAT/
CD36 protein content and mitochondrial FA
EXERCISE-INDUCED MUSCLE oxidation (Holloway et al. 2006). In other
MITOCHONDRIAL FATTY ACID UPTAKE experiments, FAT/CD36 coimmunoprecipi-
AND FAT TRANSPORTER TRANSLOCATION tated with CPTI (Schenk and Horowitz 2006)
and exercise training increased the FAT/CD36
The transport of FAs into mitochondria is a key content on the mitochondria membranes to a
step in regulating the overall rate that skeletal greater extent than the increase in mitochondri-
muscle can oxidize FAs (Holloway et al. 2008; al volume (Talanian et al. 2010). There has been
Smith et al. 2012b). At one time, the regulation little work examining the regulation of FAT/
of FA oxidation at the level of mitochondria was CD36 movement to the mitochondrial mem-
solely attributed to the relationship between car- branes during exercise, as it would be assumed
nitine palmitoyl transferase (CPT)I activity and that the system controlling movement to the
malonyl-CoA (M-CoA) (McGarry and Brown sarcolemma would also control movement to
1997). In rodent skeletal muscle at rest, M-CoA the mitochondria. In one study, Monaco et al.
levels are highest and believed to inhibit the (2015) reported that AMPKa2 was not required
transfer of FAs through the CPT complex into for skeletal muscle mitochondrial FAT/CD36
the mitochondria. On contraction, the M-CoA accumulation in AMPKa2 dead mice during
content decreased and relieved the inhibition on exercise. Smith et al. (2011, 2012b) proposed a
the CPT complex (reviewed in Spriet 2012, theory describing a dual mechanism of action
www.perspectivesinmedicine.org

2014). M-CoA has also been detected in human for skeletal muscle FAT/CD36 during exercise,
skeletal muscle, and measurements at rest and acting at both the muscle and mitochondrial
during exercise have shown that M-CoA content membranes to increase FA transport into the
is unaffected by exercise at varying power out- muscle and mitochondria. Details suggest that
puts (35% – 100% VO2max) and rates of fat oxi- FAT/CD36 is located on the outer mitochondri-
dation (Odland et al. 1998) or decreases slightly al membrane upstream of the acyl-CoA synthase
during prolonged exercise (Roepstorff et al. enzyme. This location, in some unexplained
2005). Recent work using permeabilized skeletal manner, appears to facilitate the delivery of
muscle fibers showed that M-CoA inhibition of long-chain fatty acid (LCFA) to this enzyme so
CPT1 is dependent on the palmitoyl-CoA con- it can proceed through the reaction and the CPT
tent (Smith et al. 2012a). This suggests that an complex and into the mitochondria (Smith
increase in skeletal muscle palmitoyl-CoA con- et al. 2011). This research does not downplay
tent at the onset of exercise could override any the importance of the CPT complex in mito-
inhibitory effect of M-CoA and allow FA trans- chondrial FA transport, but rather indicates a
port and oxidation to proceed. complexity in the regulation of mitochondrial
In keeping with the suggestion that the reg- FA transport not previously understood.
ulation of CPTI activity and FA transport across
the mitochondrial membranes was more com-
CONCLUDING REMARKS
plex, was the finding that FAT/CD36 also exist-
ed on the skeletal muscle mitochondrial mem- Our understanding of exercise metabolism and
branes (Campbell et al. 2004; Bezaire et al. the interplay between carbohydrate and fat me-
2006). It was also shown that, although FABPpm tabolism, including underlying regulatory
facilitated FA transport at the sarcolemma, it mechanisms, has increased over the years based
played no role at the mitochondria (Holloway on studies utilizing various methods, including
et al. 2007). FAT/CD36 appeared to regulate indirect calorimetry, metabolic tracers sam-
mitochondrial FA oxidation as FA oxidation pling, and analysis of blood and tissue samples

Cite this article as Cold Spring Harb Perspect Med 2018;8:a029744 11


Downloaded from http://perspectivesinmedicine.cshlp.org/ at NYU MED CTR LIBRARY on October 2, 2018 - Published by Cold
Spring Harbor Laboratory Press

M. Hargreaves and L.L. Spriet

from contracting skeletal muscle. In an era of Campbell, Tandon NN, Woldegiorgis G, Luiken JJFP, Glatz
JFC, Bonen A. 2004. A novel function for fatty acid trans-
expanding tools in genomics, epigenomics,
locase (FAT)/CD36. J Biol Chem 279: 36235– 36241.
transcriptomics, proteomics, and metabolo- Cartee GD. 2015. Roles of TBC1D1 and TBC1D4 in insulin-
mics, along with system and computational bi- and exercise-stimulated glucose transport of skeletal
ology approaches to data analysis, new insights muscle. Diabetologia 58: 19– 30.
should be obtained on substrate fluxes in met- Casey A, Constantin-Teodosiu D, Howell S, Hultman E,
Greenhaff PL. 1996. Metabolic responses of type I and
abolically active tissues and the molecular reg- II muscle fibres during repeated bouts of maximal exer-
ulation of exercise metabolism. cise in humans. Am J Physiol 271: E38– E43.
Cermak NM, van Loon LJ. 2013. The use of carbohydrates
during exercise as an ergogenic aid. Sports Med 43: 1139–
1155.
REFERENCES
Coggan AR, Kohrt WM, Spina RJ, Bier DM, Holloszy JO.
Allen DG, Lamb GD, Westerbald H. 2008. Skeletal 1990. Endurance training decreases plasma glucose turn-
muscle fatigue: Cellular mechanisms. Physiol Rev 88: over and oxidation during moderate-intensity exercise in
287–332. men. J Appl Physiol 68: 990–996.
Alsted TJ, Nybo L, Schweiger M, Fledelius C, Jacobsen P, Coggan AR, Raguso CA, Williams BD, Sidossis LS, Gastal-
Zimmermann R, Zechner R, Kiens B. 2009. Adipose tri- delli A. 1995a. Glucose kinetics during high-intensity
glyceride lipase in human skeletal muscle is upregulated exercise in endurance-trained and untrained humans. J
by exercise training. Am J Physiol 296: E445– E453. Appl Physiol 78: 1203– 1207.
Alsted TJ, Ploug T, Prats C, Serup AK, Høeg L, Schjerling P, Coggan AR, Swanson SC, Mendenhall LA, Habash DL, Kien
Holm C, Zimmermann R, Fledelius C, Galbo H, et al. CL. 1995b. Effect of endurance training on hepatic gly-
2013. Contraction-induced lipolysis is not impaired by cogenolysis and gluconeogenesis during prolonged exer-
inhibition of hormone-sensitive lipase in skeletal muscle. cise in men. Am J Physiol 268: E375– E383.
J Physiol 591: 5141–5155. Cushman SW, Goodyear LJ, Pilch PF, Ralston E, Galbo H,
An D, Toyoda T, Taylor EB, Yu H, Fujii N, Hirshman MF, Ploug T, Kristiansen S, Klip A. 1998. Molecular mecha-
Goodyear LJ. 2010. TBC1D1 regulates insulin- and con- nisms involved in GLUT4 translocation in muscle during
www.perspectivesinmedicine.org

traction-induced glucose transport in mouse skeletal insulin and contraction stimulation. Adv Exp Med Biol
muscle. Diabetes 59: 1358–1365. 441: 63–71.
Bezaire V, Bruce CR, Heigenhauser GJ, Tarendon NN, Glatz DeFronzo RA, Ferrannini E, Sato Y, Wahren J. 1981. Syner-
JFC, Luiken JJJF, Bonen A, Spriet LL. 2006. Identification gistic interaction between exercise and insulin on periph-
of fatty acid translocase on human skeletal muscle mito- eral glucose uptake. J Clin Invest 68: 1468– 1474.
chondrial membranes: Essential role in fatty acid oxida- Fentz J, Kjobsted R, Birk JB, Jeppesen J, Thorsen K, Schjer-
tion. Am J Physiol 290: E509–E515. ling P, Kiens B, Jessen N, Viollet B, Wojtaszewski JF. 2016.
Bonen A, Luiken JJFP, Liu S, Dyck DJ, Kiens B, Kristiansen S, AMPKa is critical for enhancing skeletal muscle fatty acid
Turcotte LP, Van Der Vusse GJ Glatz JF. 1998. Palmitate utilization during in vivo exercise in mice. FASEB J 29:
transport and fatty acid transporters in red and white 1725–1738.
muscles. Am J Physiol 275: E471– E478. Friedlander AL, Casazza GA, Horning MA, Huie MJ, Brooks
Bonen A, Dyck DJ, Ibrahimi A, Abumrad NA. 1999. Muscle GA. 1997. Training-induced alterations of glucose flux in
contractile activity increases fatty acid metabolism and men. J Appl Physiol 82: 1360– 1369.
transport and FAT/CD36. Am J Physiol 276: E642–E649.
Friedrichsen M, Mortenson B, Pehmøller C, Birk JB, Woj-
Bonen A, Luiken JJFP, Arumugam T, Glatz JF, Tandon NN. taszewski JFP. 2013. Exercise-induced AMPK activity in
2000. Acute regulation of fatty acid uptake involves the skeletal muscle: Role in glucose uptake and insulin sen-
cellular redistribution of fatty acid translocase. J Biol sitivity. Mol Cell Endocrinol 366: 204–214.
Chem 275: 14501– 14508.
Glatz JF, Luiken JJ, Bonen A. 2010. Membrane fatty acid
Bonen A, Chabowski A, Luiken JJFP, Glatz JF. 2007. Is mem- transporters as regulators of lipid metabolism: Implica-
brane transport of FFA mediated by lipid, protein, or
tions for metabolic disease. Physiol Rev 90: 367–417.
both? Mechanisms and regulation of protein-mediated
cellular fatty acid uptake: Molecular, biochemical, and Hardie DG, Ross FA, Hawley SA. 2012. AMPK: A nutrient
physiological evidence. Physiologist 22: 15– 29. and energy sensor that maintains energy homeostasis.
Nat Rev Mol Cell Biol 13: 251– 262.
Bradley SJ, Kingwell BA, McConell GK. 1999. Nitric oxide
synthase inhibition reduces leg glucose uptake but not Hargreaves M, McKenna MJ, Jenkins DG, Warmington SA,
blood flow during dynamic exercise in humans. Diabetes Li JL, Snow RJ, Febbraio MA. 1998. Muscle metabolites
48: 1815–1821. and performance during high intensity, intermittent ex-
Bradley NS, Snook LA, Jain SS, Heigenhauser, GJF, Bonen A, ercise. J Appl Physiol 84: 1687– 1691.
Spriet LL. 2012. Acute endurance exercise increases plas- Hawley JA, Schabort EJ, Noakes TD, Dennis SC. 1997. Car-
ma membrane fatty acid transport proteins in rat and bohydrate-loading and exercise performance: An update.
human skeletal muscle. Am J Physiol 302: E183– E189. Sports Med 24: 73–81.
Brooks GA. 2009. Cell– cell and intracellular lactate shuttles. Hawley JA, Hargreaves M, Joyner MJ, Zierath JR. 2014. In-
J Physiol 587: 5591–5600. tegrative biology of exercise. Cell 159: 738 –749.

12 Cite this article as Cold Spring Harb Perspect Med 2018;8:a029744


Downloaded from http://perspectivesinmedicine.cshlp.org/ at NYU MED CTR LIBRARY on October 2, 2018 - Published by Cold
Spring Harbor Laboratory Press

Exercise Metabolism

Holloway GP, Spriet LL. 2009. Skeletal muscle metabolic induced skeletal muscle FAT/CD36 trafficking and FA
adaptations to training. In The IOC textbook of science uptake is AMPK independent. J Lipid Res 52: 699– 711.
in sport, 1st ed. (ed. Maughan RJ), pp. 70–83. Wiley- Jeppesen J, Maarbjerg SJ, Jordy AB, Fritzen AM, Pehmøller
Blackwell, Oxford. C, Sylow L, Serup AK, Jessen N, Thorsen K, Prats C, et al.
Holloway GP, Bezaire V, Heigenhauser GJF, Tarendon NN, 2013. LKB1 regulates lipid oxidation during exercise in-
Glatz JFC, Luiken JJFP, Bonen A, Spriet LL. 2006. Mito- dependently of AMPK. Diabetes 62: 1490– 1499.
chondrial long chain fatty acid oxidation, fatty acid trans- Katz A. 2016. Role of reactive oxygen species in regulation of
locase/CD36 content and carnitine palmitoyltransferase glucose transport in skeletal muscle during exercise. J
I activity in human skeletal muscle during aerobic exer- Physiol 594: 2787– 2794.
cise. J Physiol 571: 201– 210.
Katz A, Broberg S, Sahlin K, Wahren J. 1986. Leg glucose
Holloway GP, Lally J, Nickerson JG, Alkhateeb H, Snook LA,
uptake during maximal dynamic exercise in humans. Am
Heigenhauser GJF, Calles-Escandon J, Glatz JFC, Luiken
J Physiol 251: E65– E70.
JJFP, Spriet LL, et al. 2007. Fatty acid binding protein
facilitates sarcolemmal fatty acid transport but not mito- Katz A, Sahlin K, Broberg S. 1991. Regulation of glucose
chondrial oxidation in rat and human skeletal muscle. J utilization in human skeletal muscle during moderate
Physiol 582: 393– 405. dynamic exercise. Am J Physiol 260: E411– E415.
Holloway GP, Luiken JJFP, Glatz JF, Spriet LL, Bonen A. Kiens B. 2006. Skeletal muscle lipid metabolism in exercise
2008. Contribution of FAT/CD36 to the regulation of and insulin resistance. Physiol Rev 86: 205–243.
skeletal muscle fatty acid oxidation: An overview. Acta Koh HJ, Toyoda T, Fujii N, Jung MM, Rathod A, Middelbeek
Physiol 194: 293– 309. RJ, Lessard SJ, Treebak JT, Tsuchihara K, Esumi H, et al.
Holloway GP, Jain SS, Bezaire V, Han XX, Glatz JF, Luiken JJ, 2010. Sucrose nonfermenting AMPK-related kinase
Harper ME, Bonen A. 2009. FAT/CD36 null mice reveal (SNARK) mediates contraction-stimulated glucose
that mitochondrial FAT/CD36 is required to up-regulate transport in mouse skeletal muscle. Proc Natl Acad Sci
mitochondrial fatty acid oxidation in contracting muscle. 107: 15541–15546.
Am J Physiol 297: R960–R967. Kramer HF, Witczak CA, Taylor EB, Fujii N, Hirshman MF,
Hong YH, Betik AC, McConell GK. 2014. Role of nitric Goodyear LJ. 2006. AS160 regulates insulin- and contrac-
oxide in skeletal muscle glucose uptake during exercise. tion-stimulated glucose uptake in mouse skeletal muscle.
Exp Physiol 99: 1569–1573. J Biol Chem 281: 31478– 31485.
Howlett RA, Parolin ML, Dyck DJ, Hultman E, Jones NL,
www.perspectivesinmedicine.org

Kristiansen S, Hargreaves M, Richter EA. 1997. Progressive


Heigenhauser GJF, Spriet LL. 1998. Regulation of skeletal increase in glucose transport and GLUT-4 in human sar-
muscle glycogen phosphorylase and PDH at varying ex- colemmal vesicles during moderate exercise. Am J Physiol
ercise power outputs. Am J Physiol 275: R418–R425. 272: E385–E389.
Howlett KF, Andrikopoulos S, Proietto J, Hargreaves M. Kristiansen S, Gade J, Wojtaszewski JF, Kiens B, Richter EA.
2013. Exercise-induced muscle glucose uptake in mice 2000. Glucose uptake is increased in trained vs. untrained
with graded, muscle-specific GLUT-4 deletion. Physiol muscle during heavy exercise. J Appl Physiol 89: 1151–
Rep 1: 1 –7. 1158.
Jain SS, Chabowski A, Snook LA, Schwenk RW, Glatz JFC, Loher H, Kreis R, Boesch C, Christ E. 2016. The flexibility of
Luiken JJFP, Bonen A. 2009. Additive effects of insulin ectopic lipids. Int J Mol Sci 17: E1554.
and muscle contraction on fatty acid transport and fatty
MacPherson RE, Peters SJ. 2015. Piecing together the puzzle
acid transporters, FAT/CD36, FABPpm, FATP1, 4 and 6.
of perilipin proteins and skeletal muscle lipolysis. Appl
FEBS Lett 583: 2294– 2300.
Physiol Nutr Metab 40: 641–651.
Jain SS, Snook LA, Glatz JF, Luiken JJ, Holloway GP, Thur-
mond DC, Bonen A. 2012. Munc18c provides stimulus- MacPherson RE, Ramos SV, Vandenboom R, Roy BD, Peters
selective regulation of GLUT4 but not fatty acid trans- SJ. 2013a. Skeletal muscle PLIN proteins, ATGL and CGI-
porter trafficking in skeletal muscle. FEBS Lett 586: 58, interactions at rest and following stimulated contrac-
2428– 2435. tion. Am J Physiol 304: R644–R650.
Jain SS, Luiken JJ, Snook LA, Han XX, Holloway GP, Glatz JF, MacPherson RE, Vandenboom R, Roy BD, Peters SJ. 2013b.
Bonen A. 2015. Fatty acid transport and transporters in Skeletal muscle PLIN3 and PLIN5 are serine phosphor-
muscle are critically regulated by Akt2. FEBS Lett 589: ylated at rest and following lipolysis during adrenergic or
2769– 2775. contractile stimulation. Physiol Rep 1: e00084.
Jensen TE, Maarbjerg SJ, Rose AJ, Leitges M, Richter EA. Martin IK, Katz A, Wahren J. 1995. Splanchnic and muscle
2009. Knockout of the predominant conventional PKC metabolism during exercise in NIDDM patients. Am J
isoform, PKCa, in mouse skeletal muscle does not affect Physiol 269: E583–E590.
contraction-stimulated glucose uptake. Am J Physiol 297: McFarlan JT, Yoshida Y, Jain SS, Snook LA, Lally J, Smith BK,
E340–E348. Glatz JF, Luiken JJ, Sayer RA, Tupling AR, et al. 2012. In
Jensen TE, Sylow L, Rose AJ, Madsen AB, Angin Y, Maarb- vivo, fatty acid translocase (CD36) critically regulates
jerg SJ, Richter EA. 2014. Contraction-stimulated glucose skeletal muscle fuel selection, exercise performance, and
transport in muscle is controlled by AMPK and mechan- training-induced adaptation of fatty acid oxidation. J Biol
ical stress but not sarcoplasmic reticulum Ca2þ release. Chem 287: 23502–23516.
Mol Metab 3: 742– 753. McGarry JD, Brown NF. 1997. The mitochondrial carnitine
Jeppesen J, Albers PH, Rose AJ, Birk JB, Schjerling P, palmitoyltransferase system. From concept to molecular
Dzamko N, Steinberg GR, Kiens B. 2011. Contraction- analysis. Eur J Biochem 244: 1 –14.

Cite this article as Cold Spring Harb Perspect Med 2018;8:a029744 13


Downloaded from http://perspectivesinmedicine.cshlp.org/ at NYU MED CTR LIBRARY on October 2, 2018 - Published by Cold
Spring Harbor Laboratory Press

M. Hargreaves and L.L. Spriet

Medbø JI, Sejersted OM. 1990. Plasma potassium changes etal muscle prevents AMPK activation and glucose up-
with high intensity exercise. J Physiol 421: 105– 122. take during contraction. EMBO J 24: 1810–1820.
Merry TL, Steinberg GR, Lynch GS, McConell GK. 2010. Schenk S, Horowitz JF. 2006. Coimmunoprecipitation of
Skeletal muscle glucose uptake during contraction is reg- FAT/CD36 and CPT I in skeletal muscle increases pro-
ulated by nitric oxide and ROS independently of AMPK. portionally with fat oxidation after endurance exercise
Am J Physiol 298: E577–E585. training. Am J Physiol 291: E254– E260.
Monaco C, Whitfield, J, Jain SS, Spriet LL, Bonen A, Hollo- Smith BK, Jain SS, Rimbaud S, Dam A, Quadrilatero J,
way GP. 2015. Acitvation of AMPKa2 is not required for Ventura-Clapier R, Bonen A, Holloway GP. 2011. FAT/
mitochondrial FAT/CD36 accumulation during exercise. CD36 is located on the outer mitochondrial membrane,
PLoS ONE 12: e0126122. upstream of long-chain acyl-CoA synthetase, and regu-
Mu J, Brozinick JT, Valladares O, Bucan M, Birnbaum MJ. lates palmitate oxidation. Biochem J 437: 125 –134.
2001. A role for AMP-activated protein kinase in contrac- Smith BK, Perry CG, Koves TR, Wright DC, Smith JC, Neu-
tion-and hypoxia-regulated glucose transport in skeletal fer PD, Muoio DM, Holloway GP. 2012a. Identification of
muscle. Mol Cell 7: 1085–1094. a novel malonyl-CoA IC50 for CPT-1: Implications for
Nickerson JG, Alkhateeb H, Benton CR, Lally J, Nickerson J, predicting in vivo fatty acid oxidation rates. Biochem J
Han XX, Wilson MH, Jain SS, Snook LA, Glatz JFC, et al. 448: 13–20.
2009. Greater transport efficiencies of the membrane fat- Smith BK, Bonen A, Holloway GP. 2012b. A dual mecha-
ty acid transporters FAT/CD36 and FATP4 compared nism of action for skeletal muscle FAT/CD36 during ex-
with FABPpm and FATP1 and differential effects on fatty ercise. Exerc Sport Sci Rev 40: 211– 217.
acid esterification and oxidation in rat skeletal muscle. J Spriet LL. 2012. The metabolic systems: Lipid metabolism.
Biol Chem 284: 16522–16530. In Advanced exercise physiology, 2nd ed. (ed. Farrell PA,
Odland LM, Howlett RA, Heigenhauser GJF, Hultman E, Joyner MJ, Caiozzo VJ), pp. 392 –407. Lippincott Wil-
Spriet LL. 1998. Skeletal muscle malonyl-CoA content liams & Wilkins, Philadelphia.
at the onset of exercise at varying power outputs in hu- Spriet LL. 2014. New insights into the interaction of carbo-
mans. Am J Physiol 274: E1080–E1085. hydrate and fat metabolism during exercise. Sports Med
O’Neill HM, Maarbjerg SJ, Crane JD, Jeppesen J, Jørgensen 44: S87– S96.
SB, Schertzer JD, Shyroka O, Kiens B, van Denderen BJ, Spriet LL, Lindinger MI, McKelvie RS, Heigenhauser GJF,
Tarnopolsky MA, et al. 2011. AMP-activated protein ki- Jones NL. 1989. Muscle glycogenolysis and Hþ concen-
nase (AMPK) b1b2 muscle null mice reveal an essential
www.perspectivesinmedicine.org

tration during maximal intermittent cycling. J Appl Phys-


role for AMPK in maintaining mitochondrial content iol 66: 8– 13.
and glucose uptake during exercise. Proc Natl Acad Sci Spriet LL, Howlett RA, Heigenhauser GJ. 2000. An enzymat-
108: 16092– 16097. ic approach to lactate production in human skeletal mus-
Parolin ML, Chesley A, Matsos MP, Spriet LL, Jones NL, cle during exercise. Med Sci Sports Exerc 32: 756–763.
Heigenhauser GJF. 1999. Regulation of glycogen phos- Stefanyk LE, Bonen A, Dyck DJ. 2012. Insulin and contrac-
phorylase and PDH during maximal intermittent exer- tion-induced movement of fatty acid transport proteins
cise. Am J Physiol 277: E890– E900.
to skeletal muscle transverse tubules is distinctly different
Perry CGR, Heigenhauser GJF, Bonen A, Spriet LL. 2008. than to the sarcolemma. Metabolism 61: 1518– 1522.
High-intensity aerobic interval training increases fat and
Stellingwerff T, Boon H, Jonkers RA, Senden JM, Spriet LL,
carbohydrate metabolic capacities in human skeletal
Koopman R, van Loon LJC. 2007. Significant intramyo-
muscle. Appl Physiol Nutr Metab 33: 1112–1123.
cellular lipid use during prolonged cycling in endurance-
Prats C, Donsmark M, Qvortrup K, Londos C, Sztalryd C, trained males as assessed by three different methodolo-
Holm C, Galbo H, Ploug T. 2006. Decrease in intramus- gies. Am J Physiol 292: E1715– E1723.
cular lipid droplets and translocation of HSL in response
Sylow L, Jensen TE, Kleinert M, Mouatt JR, Maarbjerg SJ,
to muscle contraction and epinephrine. Lipid Res 47: Jeppesen J, Prats C, Chiu TT, Boguslavsky S, Klip A, et al.
2392– 2399. 2013. Rac1 is a novel regulator of contraction-stimulated
Richter EA, Hargreaves M. 2013. Exercise, GLUT4 and skel- glucose uptake in skeletal muscle. Diabetes 62: 1139–
etal muscle glucose uptake. Physiol Rev 93: 993–1017. 1151.
Roepstorff C, Halberg N, Hillig T, Saha A, Ruderman NB, Sylow L, Nielsen IL, Kleinert M, Møller LLV, Ploug T,
Wojtaszewski JF, Richter EA, Kiens B. 2005. Malonyl-CoA Schjerling P, Bilan PJ, Klip A, Jensen TE, Richter EA.
and carnitine in regulation of fat oxidation in human 2016. Rac1 governs exercise-stimulated glucose uptake
skeletal muscle during exercise. Am J Physiol 288: in skeletal muscle through regulation of GLUT4 translo-
E133–E142. cation in mice. J Physiol 594: 4997–5008.
Romijn JA, Coyle EF, Sidossis LS, Gastaldelli A, Horowitz JF, Talanian JL, Tunstall RJ, Watt MJ, Duong M, Perry CGR,
Endert E, Wolfe RR. 1993. Regulation of endogenous fat Steinberg GR, Kemp BE, Heigenhauser GJF, Spriet LL.
and carbohydrate metabolism in relation to exercise in- 2006. b-Adrenergic regulation of human skeletal muscle
tensity and duration. Am J Physiol 265: E380–E391. hormone sensitive lipase activity during exercise onset.
Romijn JA, Coyle EF, Sidossis LS, Zhang XJ, Wolfe RR. 1995. Am J Physiol 291: R1094–R1099.
Relationship between fatty acid delivery and fatty acid Talanian JL, Holloway GP, Snook L, Heigenhauser GJF,
oxidation during strenuous exercise. J Appl Physiol 79: Bonen A, Spriet LL. 2010. Exercise training increases sar-
1939– 1945. colemmal and mitochondrial fatty acid transport pro-
Sakamoto K, McCarthy A, Smith D, Green KA, Hardie DG, teins in human skeletal muscle. Am J Physiol 299:
Ashworth A, Alessi DR. 2005. Deficiency of LKB1 in skel- E180– E188.

14 Cite this article as Cold Spring Harb Perspect Med 2018;8:a029744


Downloaded from http://perspectivesinmedicine.cshlp.org/ at NYU MED CTR LIBRARY on October 2, 2018 - Published by Cold
Spring Harbor Laboratory Press

Exercise Metabolism

Treebak JT, Pehmøller C, Kristensen JM, Kjøbsted R, Birk JB, Wasserman DH, Kang L, Ayala JE, Fueger PT, Lee-Young RS.
Schjerling P, Richter EA, Goodyear LJ, Wojtaszewski JFP. 2011. The physiological regulation of glucose flux into
2014. Acute exercise and physiological insulin induce muscle in vivo. J Exp Biol 214: 254– 262.
distinct phosphorylation signatures on TBC1D1 and Watt MJ. 2009. Triglyceride lipases alter fuel metabolism and
TBC1D4 proteins in human skeletal muscle. J Physiol mitochondrial gene expression. Appl Physiol Nutr Metab
592: 351– 375.
34: 340– 347.
Turcotte LP, Abbott MJ. 2012. Contraction-induced signal-
Watt M, Spriet LL. 2010. Triacylglycerol lipases and meta-
ing: Evidence of convergent cascades in the regulation of
bolic control: Implications for health and disease. Am J
muscle fatty acid metabolism. Can J Physiol Pharmacol
90: 1419–1433. Physiol 299: E162–E168.
Turcotte L, Kiens B, Richter EA. 1991. Saturation kinetics of Watt MJ, Howlett KF, Febbraio MA, Spriet LL, Hargreaves
palmitate uptake in perfused skeletal muscle. FEBS Lett M. 2001. Adrenaline increases skeletal muscle glycogen-
279: 327– 329. olysis, PDH activation and carbohydrate oxidation dur-
Turcotte L, Richter EA, Kiens B. 1992. Increased plasma FFA ing moderate exercise in humans. J Physiol 534: 269– 278.
uptake and oxidation during prolonged exercise in trained Watt MJ, Heigenhauser GJF, Spriet LL. 2003. Effects of dy-
vs. untrained humans. Am J Physiol 262: E791– E799. namic exercise intensity on the activation of hormone-
Turcotte LP, Raney MA, Todd MK. 2005. ERK1/2 inhibition sensitive lipase in human skeletal muscle. J Physiol 547:
prevents contraction-induced increase in plasma mem- 301 –308.
brane FAT/CD36 content and FA uptake in rodent mus- Witczak CA, Jessen N, Warro DM, Toyoda T, Fujii N, Anderson
cle. Acta Physiol Scand 184: 131–139. ME, Hirshman MF, Goodyear LJ. 2010. CaMKII regulates
Turnbull PC, Ramos SV, MacPherson REK, Roy BD, Peters contraction- but not insulin-induced glucose uptake in
SJ. 2015. Characterization of lipolytic inhibitor G(0)/ mouse skeletal muscle. Am J Physiol 298: E1150–E1160.
G(1) switch gene-2 protein (G0S2) expression in male Wright DC, Hucker KA, Holloszy JO, Han DH. 2004. Ca2þ
Sprague Dawley rat skeletal muscle compared to relative and AMPK both mediate stimulation of glucose trans-
content of adipose triglyceride lipase (ATGL) and com- port by muscle contractions. Diabetes 53: 330–335.
parative gene-idetification-58 (CGI-58). PLoS ONE 10:
e120136. Yoshida Y, Jain SS, McFarlan JT, Snook LA, Chabowski A,
Bonen A. 2013. Exercise- and training-induced upregu-
Turnbull PC, Longo AB, Ramos SV, Roy BD, Ward WE,
lation of skeletal muscle fatty acid oxidation are not solely
Peters SJ. 2016. Increases in skeletal muscle ATGL and
www.perspectivesinmedicine.org

dependent on mitochondrial machinery and biogenesis.


its inhibitor G0S2 following 8 weeks of endurance train-
ing in metabolically different rat skeletal muscles. Am J J Physiol 591: 4415–4426.
Physiol 310: R125– R133. Zimmermann R, Strauss JG, Haemmerle G, Schoiswohl G,
van Loon LJC, Greenhaff PL, Constantin-Teodosiu D, Saris Birner-Gruenberger R, Riederer M, Lass A, Neuberger G,
WHM, Wagenmakers AJM. 2001. The effects of increas- Eisenhaber F, Hermetter A, et al. 2004. Fat mobilization
ing exercise intensity on muscle fuel utilisation in hu- in adipose tissue is promoted by adipose triglyceride li-
mans. J Physiol 536: 295– 304. pase. Science 306: 1383– 1386.
Wasserman DH, Geer RJ, Rice DE, Bracy D, Flakoll PJ, Zinker BA, Lacy DB, Bracy DP, Wasserman DH. 1993. Role
Brown LL, Hill JO, Abumrad NN. 1991. Interaction of of glucose and insulin loads to exercising limb in increas-
exercise and insulin action in humans. Am J Physiol 260: ing glucose uptake and metabolism. J Appl Physiol 74:
E37– E45. 2915–2922.

Cite this article as Cold Spring Harb Perspect Med 2018;8:a029744 15


Downloaded from http://perspectivesinmedicine.cshlp.org/ at NYU MED CTR LIBRARY on October 2, 2018 - Published by Cold
Spring Harbor Laboratory Press

Exercise Metabolism: Fuels for the Fire


Mark Hargreaves and Lawrence L. Spriet

Cold Spring Harb Perspect Med 2018; doi: 10.1101/cshperspect.a029744 originally published online May
22, 2017

Subject Collection The Biology of Exercise

Molecular Basis of Exercise-Induced Skeletal Muscle-Adipose Tissue Cross Talk


Muscle Mitochondrial Biogenesis: Historical Kristin I. Stanford and Laurie J. Goodyear
Advances, Current Knowledge, and Future
Challenges
Christopher G.R. Perry and John A. Hawley
Exercise Metabolism: Fuels for the Fire Performance Fatigability: Mechanisms and Task
Mark Hargreaves and Lawrence L. Spriet Specificity
Sandra K. Hunter
Health Benefits of Exercise Adaptations to Endurance and Strength Training
Gregory N. Ruegsegger and Frank W. Booth David C. Hughes, Stian Ellefsen and Keith Baar
Molecular Regulation of Exercise-Induced Muscle The Bioenergetics of Exercise
Fiber Hypertrophy P. Darrell Neufer
Marcas M. Bamman, Brandon M. Roberts and
Gregory R. Adams
Physiological Redundancy and the Integrative Effects of Exercise on Vascular Function,
Responses to Exercise Structure, and Health in Humans
Michael J. Joyner and Jerome A. Dempsey Daniel J. Green and Kurt J. Smith
On the Run for Hippocampal Plasticity Exosomes as Mediators of the Systemic
C'iana Cooper, Hyo Youl Moon and Henriette van Adaptations to Endurance Exercise
Praag Adeel Safdar and Mark A. Tarnopolsky
Effects of Exercise and Aging on Skeletal Muscle Control of Muscle Metabolism by the Mediator
Giovanna Distefano and Bret H. Goodpaster Complex
Leonela Amoasii, Eric N. Olson and Rhonda
Bassel-Duby
Molecular Basis for Exercise-Induced Fatigue: Theoretical and Biological Evaluation of the Link
The Importance of Strictly Controlled Cellular Ca between Low Exercise Capacity and Disease Risk
2+ Handling Lauren Gerard Koch and Steven L. Britton
Arthur J. Cheng, Nicolas Place and Håkan
Westerblad

For additional articles in this collection, see http://perspectivesinmedicine.cshlp.org/cgi/collection/

Copyright © 2018 Cold Spring Harbor Laboratory Press; all rights reserved

Das könnte Ihnen auch gefallen