You are on page 1of 7

Adv Physiol Educ 38: 308–314, 2014;

Refresher Course doi:10.1152/advan.00080.2014.

Exercise and type 2 diabetes: molecular mechanisms regulating glucose


uptake in skeletal muscle
Kristin I. Stanford and Laurie J. Goodyear
Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Department of Medicine, Brigham and Women’s
Hospital, Harvard Medical School, Boston, Massachusetts
Submitted 2 July 2014; accepted in final form 2 October 2014

Stanford KI, Goodyear LJ. Exercise and type 2 diabetes: molec- Exercise Training Improves Metabolic Health in People
ular mechanisms regulating glucose uptake in skeletal muscle. Adv With Type 2 Diabetes
Physiol Educ 38: 308 –314, 2014; doi:10.1152/advan.00080.2014.—
Exercise is a well-established tool to prevent and combat type 2 While rates of diabetes are on the rise, it has long been
diabetes. Exercise improves whole body metabolic health in people recognized that exercise has important health benefits for
with type 2 diabetes, and adaptations to skeletal muscle are essential people with type 2 diabetes. One of the environmental factors
for this improvement. An acute bout of exercise increases skeletal considered a risk for the development of insulin resistance and
muscle glucose uptake, while chronic exercise training improves
type 2 diabetes is a lack of physical activity, and regular
mitochondrial function, increases mitochondrial biogenesis, and in-
creases the expression of glucose transporter proteins and numerous physical exercise can delay or prevent the onset of this disease
metabolic genes. This review focuses on the molecular mechanisms (18, 58, 78).
that mediate the effects of exercise to increase glucose uptake in Randomized trials have found that lifestyle interventions
skeletal muscle. including ⬃150 min/wk of physical activity, combined with
diet-induced weight loss, reduced the risk of type 2 diabetes by
exercise; type 2 diabetes
58% in an at-risk population (58, 78). Interventions of exercise
alone have proved to be just as effective in terms of prevention
DIABETES IS A COMPLEX DISEASE that affects millions of people of the progression of type 2 diabetes as programs of diet alone
worldwide. There are 23.6 million people in the United States or diet and exercise combined (9). Exercise training in type 2
or ⬃8% of the population that have diabetes, a number that has diabetic patients improves management of blood glucose lev-
doubled over the last 15 yr and is continuing to increase at els, body weight, lipids, blood pressure, cardiovascular disease,
epidemic rates (56). It is predicted that 1 in 3 adults in the mortality, and overall quality of life (13, 19, 38, 47, 48, 62, 82).
United States will have diabetes by 2050, yielding enormous The more recent Look AHEAD study has shown that combined
tolls at individual, public health, and economic levels (56). weight loss and physical activity in people with type 2 diabetes
Type 2 diabetes arises from a combination of genetic sus- resulted in modest weight loss of ⬃6%, improved glycolated
ceptibility and environmental factors including physical inac- hemoglobin, improved mobility, and improved kidney func-
tivity and poor nutrition (25). Obesity plays a role in the tion, but no improvement in cardiovascular disease over a
majority of cases of type 2 diabetes (7a). The progression of 10-yr period (13, 47, 48, 62). However, since the level of
obesity increases the risk for the development of type 2 diabetes; fitness was only assessed through year 4 of the study, conclu-
as a person becomes more obese, they enter a more insulin- sions on the effects of fitness level on cardiovascular disease
resistant state, leading to impaired glucose tolerance, which can cannot be made (13, 47, 48, 62). Another recent study (23) has
potentially lead to the onset of type 2 diabetes. As the disease shown that 6 mo of moderate-intensity exercise training de-
progresses, the risk of complications, such as retinopathy, ne- creased visceral fat mass and decreased hepatic triglyceride
phropathy, and neuropathy, increases. In fact, type 2 diabetes is content in people with type 2 diabetes and that this program of
the leading cause of blindness, kidney failure, and amputations exercise alone was more effective than programs of diet alone.
and a major cause of heart attacks and strokes (56). Another recent study (19) showed that increasing physical
In people with type 2 diabetes, insulin levels are normal or activity in adults with type 2 diabetes resulted in partial or
high, but tissues such as liver, skeletal muscle, and adipose complete remission of type 2 diabetes in 11.5% of subjects
tissue become resistant to insulin. The pancreas compensates within the first year of intervention and an additional 7% had
by producing large amounts of insulin, and this overproduction partial or complete remission of type 2 diabetes after 4 yr of
of insulin can eventually fail. This increase in circulating exercise intervention. Complete remission was defined by
insulin can result in impaired glucose transport into liver, glucose normalization without need for drugs, and partial
skeletal muscle, and adipose tissue (11). While type 2 diabetes remission was defined as a transition to prediabetic or normal
is usually adult onset, the number of children and adolescents glucose levels without drug treatment (19). Taken together,
afflicted by this disease is increasing, likely as a result of these data demonstrate the beneficial effects of exercise train-
increased rates of obesity and poor nutrition and an increas- ing to combat type 2 diabetes.
ingly sedentary lifestyle. One of the most well-established mechanisms through which
type 2 diabetics improve metabolic health with exercise is
through adaptations to skeletal muscle, which, in turn, de-
Address for reprint requests and other correspondence: K. I. Stanford,
Section on Integrative Physiology and Metabolism, Joslin Diabetes Center,
creases skeletal muscle insulin resistance. Here, we will dis-
Dept. of Medicine, Brigham and Women’s Hospital, Harvard Medical School, cuss the effects of exercise on skeletal muscle, because skeletal
Boston, MA 02215 (e-mail: kristin.stanford@joslin.harvard.edu). muscle is responsible for the majority of glucose uptake in the
308 1043-4046/14 Copyright © 2014 The American Physiological Society
Downloaded from journals.physiology.org/journal/advances (114.005.241.243) on February 23, 2021.
Refresher Course
MOLECULAR MECHANISMS REGULATING GLUCOSE UPTAKE IN MUSCLE 309

postprandial state (6, 11). In the following sections, we will activation of phosphatidylinositol 3-kinase (14, 17). Exercise,
discuss specific adaptations of skeletal muscle to both acute however, has no effect on insulin receptor and insulin receptor
and exercise training on skeletal muscle glucose uptake and substrate-1/2 tyrosine phosphorylation or on phosphatidylino-
metabolism. sitol 3-kinase activity (17, 75). In fact, mice that lack insulin
receptors in skeletal muscle [muscle-specific insulin receptor
Effects of Acute Exercise on Skeletal Muscle Glucose Uptake knockout (KO) mice] have normal exercise-stimulated glucose
uptake (85). These data clearly demonstrate that insulin and
It is well established that insulin is a potent simulator of exercise mediate GLUT4 translocation in skeletal muscle
glucose transport in skeletal muscle. In people with type 2 through distinct proximal signaling mechanisms.
diabetes, insulin-stimulated glucose uptake in skeletal muscle Acute exercise activates multiple signaling pathways, but
is impaired. However, exercise-stimulated glucose uptake in the activated signaling pathways necessary for increased glu-
people with type 2 diabetes is normal or at near normal levels cose uptake and GLUT4 translocation are not well understood.
(51). Because exercise-stimulated glucose uptake is normal in Muscle contraction involves changes in energy status (i.e.,
people with type 2 diabetes, defining insulin-independent increased AMP/ATP), increases in intracellular Ca2⫹ concen-
mechanisms in the control of exercise-stimulated skeletal mus- tration, increased ROS, and PKC. These changes activate
cle glucose uptake is of critical importance as a potential means various signaling cascades, some of which likely work to
to treat diabetes. During the last several years, researchers have phosphorylate Tre-2/USP6, BUB2, cdc16 domain family
learned much about the signaling mechanisms that regulate member 1 (TBC1D1) and Akt substrate of 160 kDa (AS160)
exercise-induced glucose transport. There are many lines of and activate GLUT4 translocation. Here, we will discuss some
evidence that show that exercise activates molecular signals of the various signaling cascades that have been implicated in
that bypass defects in insulin action in skeletal muscle. exercise-stimulated glucose uptake (Fig. 1) (63, 66).
Both insulin and exercise increase skeletal muscle glucose
uptake by translocation of glucose transporter 4 (GLUT4), the AMP-Activated Protein Kinase
predominant GLUT in muscle, from an intracellular location to
the plasma membrane. Insulin and exercise stimulate GLUT4 AMP-activated protein kinase (AMPK) and its primary up-
translocation through distinct signaling mechanisms. Insulin stream kinase, liver kinase B1 (LKB1), are the most widely
signaling involves rapid phosphorylation of the insulin recep- studied proteins implicated in skeletal muscle glucose transport
tor, insulin receptor substrate-1/2 on tyrosine residues, and the in response to exercise. AMPK is a heterotrimeric protein

Insulin Glucose
Exercise
Insulin
Receptor

P
P Rac1 Ca2+ AMP/ATP ROS Mechanical
P
P
P
LKB1
Rac1
P IRS-1 P p85- p110 PAK1
calmodulin NRG
PI 3-Kinase SNARK AMPK
aPKCs
CaMKs

PAK1
Akt GLUT4 Translocation

P PP P P P P PP P P P
RAB RAB
CBD GAP CBD GAP
domain domain

TBC1D1 AS160
Fig. 1. Exercise and insulin regulation of glucose transport. A proposed model for the signaling pathways mediating exercise- and insulin-induced skeletal muscle
glucose transport is shown. IRS-1, insulin receptor substrate-1; PAK, p21 protein (Cdc42/Rac)-activated kinase 1; LKB1, liver kinase B1; PI3K, phosphati-
dylinositol 3-kinase; CaMK, Ca2⫹/calmodulin-dependent protein kinase; SNARK, sucrose nonfermenting AMP-dependent protein kinase (AMPK)-related
kinase; NRG, neuroglian; aPKCs, atypical PKCs; GLUT, glucose transporter; TBC1D1, Tre-2/USP6, BUB2, cdc16 domain family member 1; AS160, Akt
substrate of 160 kDa; CBD, calmodulin-binding domain. [Adapted from Ref. 66.]

Advances in Physiology Education • doi:10.1152/advan.00080.2014 • http://advan.physiology.org


Downloaded from journals.physiology.org/journal/advances (114.005.241.243) on February 23, 2021.
Refresher Course
310 MOLECULAR MECHANISMS REGULATING GLUCOSE UPTAKE IN MUSCLE

composed of a catalytic ␣-subunit and regulatory ␤- and etal muscle showed that inhibition of CaMK signaling with
␥-subunits. The ␣- and ␤-subunits each exist in two isoforms KN-93 inhibited contraction-induced skeletal muscle glucose
(␣1, ␣2, ␤1, and ␤2), and the ␥-subunit exists in three isoforms uptake through an AMPK-dependent signaling pathway. These
(␥1, ␥2, and ␥3). AMPK is activated by phosphorylation by one data suggest that CaMKs play important roles in the regulation
or more upstream kinases, including LKB1 (20, 39). A previ- of contraction-induced skeletal muscle glucose uptake, but the
ous study (53) involving incubation with the AMP-analog role of AMPK in the regulation of Ca2⫹/calmodulin-mediated
5-aminoimidazole-4-carboxamide ribonucleoside (AICAR) increases in skeletal muscle glucose uptake is unclear. Re-
have shown that activation of AMPK is positively correlated cently, electroporation of a specific CaMKII inhibitor into
with increased skeletal muscle glucose uptake. This increase in mouse tibialis anterior muscle reduced exercise-stimulated
glucose uptake by AICAR stimulation is lost in mouse models glucose uptake by 30% (84). However, a separate study (29)
deficient in AMPK ␣2- or ␥3-subunits (10, 33). Some studies found that increases in Ca2⫹ concentration in muscle caused
(1, 28, 30, 33, 57, 69) have shown that mice overexpressing very little increase in glucose uptake when the contractile
a dominant negative AMPK-␣2 construct in muscle or response of the muscle was impaired. These data point to an
AMPK-␣1 and -␤ KO mice have impaired exercise-stimulated indirect effect of Ca2⫹ on muscle glucose uptake.
glucose uptake. However, other studies using mouse models
with ablated AMPK activity have demonstrated that inhibition Downstream Targets of Insulin- and Exercise-Stimulated
of AMPK has little or no effect on exercise-induced glucose Contraction (AS160, TBC1D1, and Rac1)
uptake (15, 33, 55) or exercise-stimulated glucose uptake in The downstream signaling pathways for insulin and exercise
vivo (50). Thus, whether AMPK is necessary for exercise- have revealed a converging signal for insulin- and the exercise-
stimulated glucose uptake is not fully understood. In a mouse stimulated glucose uptake in skeletal muscle. Some of the
muscle-specific LKB1 KO model, AMPK-␣2 activation was molecules involved in this point of convergence are AS160 and
completely inhibited and exercise-stimulated glucose uptake TBC1D1. The link between AS160 and TBC1D1 as well as
was severely blunted (41, 67). This impairment could be due to GLUT4 translocation involves Rab proteins. Rab proteins are
decreased activation of AMPK and one or more of the AMPK- members of the Ras small GTPase superfamily (81). These
related kinases that are substrates of LKB1, for example, proteins are involved in many membrane trafficking events,
sucrose nonfermenting AMPK-related kinase, which is in- and active Rabs recruit various effectors that are involved in
volved in exercise-induced glucose uptake (42). However, the vesicle budding, tethering, and fusion and therefore also in
role of LKB-1 in exercise-stimulated glucose uptake is debat- GLUT4 translocation (34, 63, 81). In addition to the well-
able (31). Whereas previous studies (41, 67) have shown that established roles of Rab proteins, there is evidence that the Rho
exercise-stimulated glucose uptake was impaired in LKB-1 KO family GTPase Rac1 is involved in both insulin- and exercise-
mice, another recent study (31) showed that glucose uptake stimulated GLUT4 translocation (71, 72). Mice deficient in
during treadmill running was similar, if not higher, in LKB-1 Rac1 (Rac1 KO mice) have decreased insulin-stimulated
KO mice compared with wild-type control mice. An additional
study (23) has also shown that muscle-specific deletion of
LKB1 only partially inhibits exercise-stimulated glucose trans- #
port. These data suggest that while AMPK and LKB1 are 0.06 * *
important in the regulation of exercise-stimulated glucose up-
PCr recovery rate constant (s-1)

take, there are several potential mechanisms involved in the 0.05


control of exercise-stimulated glucose transport in skeletal
muscle.
0.04

Ca2⫹/Calmodulin-Dependent Protein Kinases


0.03
A fundamental part of skeletal muscle contraction is the
increase in intracellular Ca2⫹ concentration. More recently,
0.02
studies have indicated Ca2⫹/calmodulin signaling and Ca2⫹/
calmodulin-dependent protein kinases (CaMKs) as critical
components of Ca2⫹- and exercise-stimulated skeletal muscle 0.01
glucose uptake. Incubation of rat skeletal muscle with the
Ca2⫹/calmodulin inhibitor KN-93 decreased skeletal muscle 0.00
glucose transport in response to contraction (86). Incubation C T2D

with KN-93 also significantly inhibited exercise-induced CaM- Fig. 2. Exercise restores mitochondrial function in type 2 diabetic (T2D)
KII phosphorylation in the absence of AMPK inhibition. This subjects. In vivo mitochondrial function was measured in vastus lateralis
suggests that CaMKs regulate glucose uptake independently of muscle and expressed as the rate constant (in s⫺1) before (solid bars) and after
training (open bars). A high rate constant reflects high in vivo mitochondrial
AMPK signaling (83, 86). These studies also showed that function. Pre- and posttraining leg extension exercise was performed at 0.5 Hz
overexpression of constitutively active CaMKK␣ in mouse to an acoustic cue on a magnetic resonance-compatible ergometer and a weight
skeletal muscle increased AMPK Thr172 phosphorylation and corresponding to 60% of the predetermined maximum. Postexercise phospho-
skeletal muscle glucose uptake (83). However, this increase in creatine (PCr) resynthesis is driven almost purely oxidatively, and the resyn-
thesis rate reflects in vivo mitochondrial function. Data are expressed as means ⫾
glucose uptake was also observed in muscle overexpressing SE. #Data for T2D subjects were significantly different from those of the
dead AMPK-␣2 and thus is likely independent of AMPK control (C) group. *Posttraining was significantly different from pretraining.
activation. In contrast, another study (30) using isolated skel- [Adapted from Ref. 52.]

Advances in Physiology Education • doi:10.1152/advan.00080.2014 • http://advan.physiology.org


Downloaded from journals.physiology.org/journal/advances (114.005.241.243) on February 23, 2021.
Refresher Course
MOLECULAR MECHANISMS REGULATING GLUCOSE UPTAKE IN MUSCLE 311

GLUT4 translocation (71, 83), and Rac1 inhibition decreased increases TBC1D1 PAS phosphorylation in skeletal muscle (2,
contraction-stimulated glucose uptake in mouse skeletal mus- 73, 80), but, unlike AS160, TBC1D1 can regulate insulin-
cle (72). stimulated glucose transport through a PAS-independent mech-
AS160. AS160 regulates insulin-stimulated GLUT4 translo- anism (2). Studies (2, 80) have shown that distinct mutations of
cation (5, 68, 74). It is phosphorylated on six different phos- TBC1D1 differentially regulate insulin- and exercise-stimu-
pho-Akt-substrate (PAS) sites in response to both insulin and lated glucose transport in skeletal muscle. These data suggest
exercise in skeletal muscle (5, 43, 76). Studies have shown that that TBC1D1 regulates both insulin- and exercise-stimulated
AS160 PAS phosphorylation is increased after prolonged ex- glucose uptake through distinct phosphorylation sites. Taken
ercise in both humans (12, 70, 76) and rats (6, 14). Studies have together, these data suggest that TBC1D1, along with AS160,
shown that AMPK phosphorylates AS160 (PAS site) in re- may also serve as a point of convergence for the regulation of
sponse to AICAR and exercise in skeletal muscle (43) and that GLUT4 translocation via both insulin- and exercise-stimulated
mutation of four PAS sites significantly inhibits both insulin glucose uptake in skeletal muscle.
and exercise-induced glucose uptake (44). Additional data
have demonstrated that mutation of the calmodulin-binding Effects of Chronic Exercise Whole Body Metabolic Health
domain on AS160 significantly inhibits exercise-stimulated
glucose uptake but not insulin-stimulated glucose uptake (45). Exercise training has important therapeutic implications for
Interestingly, whole body KO of AS160 does not result in a people with type 2 diabetes. Here, we will discuss several
significant increase in glucose transport in skeletal muscle (8). adaptations of skeletal muscle to chronic exercise and how
These data clearly show that both phosphorylation and calmod- these adaptations can improve metabolic health in people with
ulin binding on AS160 are involved in the regulation of type 2 diabetes, specifically by improving mitochondrial func-
exercise-stimulated glucose uptake. These data also suggest tion and increasing GLUT4 protein expression.
that while AS160 may serve as a point of convergence for both
insulin- and exercise-dependent signaling in the regulation of Effects of Chronic Exercise on Skeletal Muscle Mitochondria
glucose uptake, other proteins may be involved in this regula-
tion of glucose uptake. People with type 2 diabetes have been reported to have
TBC1D1. Recent studies (5, 36, 37, 43, 64, 73, 77) have smaller, damaged, or dysfunctional mitochondria (59) and
identified TBC1D1, the paralog of AS160, as another potential decreased expression of peroxisome proliferator-activated re-
molecular link among signaling pathways converging on ceptor-␥ coactivator-1␣, a marker of mitochondrial biogenesis
GLUT4 translocation in skeletal muscle. TBC1D1 and AS160 (47, 55). Skeletal muscle mitochondrial dysfunction has also
are 47% identical overall and have several comparable struc- been linked with insulin resistance and type 2 diabetes (59),
tural features. TBC1D1 was first identified in adipocytes in although more studies are needed to directly connect mitochon-
culture but is highly expressed in skeletal muscle (64). Insulin drial deficits with impaired muscle glucose metabolism. It is

Pre Post Pre Post


A B
Glucose Concentration (mmol)

9
GLUT4 protein content (a.u.)

*
24 hr Average Blood

8
4
7
*
3
6
2 Fig. 3. GLUT4 and mitochondrial markers
5
increased after 2 wk of low-volume high-
1 4
intensity interval training. A–D: 2 wk of
high-intensity training increased GLUT4
0 protein content in skeletal muscle (A), glyce-
Pre Post Pre Post mic control (average blood glucose) as mea-
sured by continuous glucose monitoring (B),
citrate synthase (CS) activity (C), and mito-
C D chondrial markers in skeletal muscle (D).
Pre
3.0 Pre, before training; Post, after training;
Protein content (a.u.)

*
(mmol•kg protein-1•hr-1)

p=0.06 Post
4 2.5 NDUFA9, NADH dehydrogenase (ubiqui-
* none) 1␣ subcomplex subunit 9. Values are
2.0 * * p=0.12 means ⫾ SD; n ⫽ 7. *P ⬍ 0.05. [Adapted
CS activity

3
1.5 from Ref. 46.]
2
1.0

1 0.5

0.0
0
9

α
ni
FA

ei

tI

se
bu

Pre Post
ot

ni
U

ha
pr
su

bu
D

nt
IN

su
a

sy
kD

e
ex

or

IV

TP
pl

70

ex

A
om

III
II

pl
ex
ex

om
C

pl
pl

C
om
om

C
C

Advances in Physiology Education • doi:10.1152/advan.00080.2014 • http://advan.physiology.org


Downloaded from journals.physiology.org/journal/advances (114.005.241.243) on February 23, 2021.
Refresher Course
312 MOLECULAR MECHANISMS REGULATING GLUCOSE UPTAKE IN MUSCLE

also unclear if the mitochondrial defect precedes the develop- Summary


ment of type 2 diabetes or vice versa.
Endurance exercise training has been shown to increase In summary, exercise is critical in the treatment and preven-
tion of type 2 diabetes. Acute exercise activates alternative
mitochondrial content and activity (22, 52). A recent study (52)
molecular signals that can bypass defects in insulin signaling in
examined the effects of exercise training in people with type 2
skeletal muscle, resulting in an insulin-independent increase in
diabetes and determined if exercise could restore mitochondrial glucose uptake. Exercise training results in increased skeletal
content and function and insulin sensitivity in a patient popu- muscle mitochondria and GLUT4 protein expression, which
lation. Eighteen male subjects underwent 12 wk of exercise are associated with improved skeletal muscle insulin sensitivity
training. Subjects exercised 3 times/wk, 2 times/wk on a cycle and whole body metabolic health. Exercise-induced adapta-
ergometer (55% of their maximal O2 consumption), and 1 tions to skeletal muscle are essential to prevent and combat
time/wk with resistance exercise (at 75% maximal voluntary type 2 diabetes.
contraction). After 12 wk of exercise, the defects previously
observed in skeletal muscle mitochondrial activity in people GRANTS
with type 2 diabetes were completely negated. Interestingly, This work was supported by an American College of Sports Medicine
this restored mitochondrial function (Fig. 2) was correlated to Research Endowment Grant (to K. I. Stanford), Mary K. Iacocca Fellowship
improved insulin-stimulated glucose disposal (52). These data (to K. I. Stanford), and National Institutes of Health Grants R01-AR-42238 (to
L. J. Goodyear), R01-DK-101043 (to L. J. Goodyear), and 5-P30-DK-36836
indicate that the restoration of mitochondrial function in type 2 (Diabetes Research Center, Joslin Diabetes Center).
diabetic patients accompanies the improved skeletal muscle
insulin sensitivity. DISCLOSURES
No conflicts of interest, financial or otherwise, are declared by the author(s).
Effects of Chronic Exercise on Skeletal Muscle GLUT4
AUTHOR CONTRIBUTIONS
Expression
Author contributions: K.I.S. drafted manuscript; K.I.S. and L.J.G. edited
It is well established that endurance exercise training and revised manuscript; K.I.S. and L.J.G. approved final version of manuscript.
increases skeletal muscle GLUT4 (22, 63). The increase in REFERENCES
skeletal muscle GLUT4 causes an increase in skeletal mus-
cle glucose uptake (24, 26, 47, 63). Most studies that have 1. Abbott MJ, Bogachus LD, Turcotte L. AMPK␣2 deficiency uncovers
time-dependency in the regulation of contraction-induced palmitate and
examined the therapeutic effects of exercise in people with glucose uptake in mouse muscle. J Appl Physiol 111: 125–134, 2011.
type 2 diabetes have focused on low- to moderate-intensity 2. An D, Toyoda T, Taylor EB, Yu H, Fujii N, Hirshman MF, Goodyear
aerobic exercise, which has been established as an effective LJ. TBC1D1 regulates insulin- and contraction-induced glucose transport
in mouse skeletal muscle. Diabetes 59: 1358 –1365, 2010.
strategy to improve the metabolic health of people with type 3. Bain J, Plater L, Elliott M, Shpiro N, Hastie CJ, McLauchlan H,
2 diabetes (9). The recommended amount of exercise for Klevernic I, Arthur JS, Alessi DR, Cohen P. The selectivity of protein
people with type 2 diabetes is 150 min of moderate intensity kinase inhibitors: a further update. Biochem J 408: 297–315, 2007.
exercise per week (9). However, the amount of exercise 4. Bonadonna RC, Del Prato S, Saccomani MP, Bonora E, Gulli G,
Ferrannini E, Bier D, Cobelli C, DeFronzo RA. Transmembrane glu-
required for improvements in whole body and skeletal cose transport in skeletal muscle of patients with non-insulindependent
muscle metabolic health is debatable. Recent studies have diabetes. J Clin Invest 92: 486 – 494, 2004.
examined if the effects of low-volume high-intensity inter- 5. Bruss MD, Arias EB, Lienhard GE, Cartee GD. Increased phosphor-
val training (HIT), a time-efficient strategy to improve ylation of Akt substrate of 160 kDa (AS160) in rat skeletal muscle in
response to insulin or contractile activity. Diabetes 54: 41–50, 2005.
health and fitness, could be applied to people with type 2 6. Capaldo B, Gastaldelli A, Antoniello S, Auletta M, Pardo F, Ciociaro
diabetes as an intervention to improve glucose regulation D, Guida R, Ferrannini E, Sacca L. Splanchnic and leg substrate
and skeletal muscle metabolism (82). This study involved exchange after ingestion of a natural mixed meal in humans. Diabetes 48:
14 people with type 2 diabetes (45). Eight of these subjects 958 –966, 1999.
7. Cartee GD, Funai K. Exercise and insulin: convergence or divergence at
underwent HIT for 2 wk, where they trained on a cycle AS160 and TBC1D1? Exerc Sport Sci Rev 37: 188 –195, 2009.
ergometer a total of six times over the 2-wk period. Each 7a.Centers for Disease Control and Prevention. Prevalence of overweight
session consisted of a 5-min warmup, 10 ⫻ 60 s of cycling and obesity among adults with diagnosed diabetes–United States, 1988 –
1994 and 1999 –2000. Morbid Mortal Week Rep 53: 1066 –1068, 2004.
at 90% maximal effort with 60 s of recovery, and a 5-min 8. Chavez JA, Roach WG, Keller SR, Lane WS, Lienhard GE. Inhibition
cooldown. After 2 wk of HIT, people with type 2 diabetes of GLUT4 translocation by Tbc1d1, a Rab GTPase-activating protein
had a marked increase in skeletal muscle GLUT4 expression abundant in skeletal muscle, is partially relieved by AMP-activated protein
and a significant decrease in blood glucose concentrations. kinase activation. J Biol Chem 283: 9187–9195, 2008.
9. Colberg SR, Sigal RJ, Fernahll B, Regensteiner JG, Blissmer BJ,
Markers of mitochondrial activity, such as citrate synthase Rubin RR, Chasan-Taber L, Albright AL, Braun B. Exercise and type
activity and expression of complex I, II, and III, were also 2 diabetes: the American College of Sports Medicine and the American
significantly increased after 2 wk of HIT (Fig. 3). These data Diabetes Association: joint position statement executive summary. Dia-
indicate that only 2 wk of HIT, where the weekly training betes Care 33: 2692–2696, 2010.
10. Daugaard JR, Richter EA. Muscle- and fibre type-specific expression of
time was 50% lower than the recommended guidelines for glucose transporter 4, glycogen synthase and glycogen phosphorylase
exercise in type 2 diabetic patients (9), increased glucose proteins in human skeletal muscle. Pflügers Arch 447: 452– 456, 2004.
uptake and improved mitochondrial function in people with 11. DeFronzo RA, Tripathy D. Skeletal muscle insulin resistance is the
primary defect in type 2 diabetes. Diabetes Care 32: S157–S163, 2009.
type 2 diabetes and improved whole body metabolism. This 12. Deshmukh A, Coffey VG, Zhong Z, Chibalin AV, Hawley JA, Zierath
study indicated that HIT is a potential time-efficient exercise JR. Exercise-induced phosphorylation of the novel Akt substrates AS160
strategy for the treatment of type 2 diabetes. and filamin A in human skeletal muscle. Diabetes 55: 1776 –1782, 2006.

Advances in Physiology Education • doi:10.1152/advan.00080.2014 • http://advan.physiology.org


Downloaded from journals.physiology.org/journal/advances (114.005.241.243) on February 23, 2021.
Refresher Course
MOLECULAR MECHANISMS REGULATING GLUCOSE UPTAKE IN MUSCLE 313

13. Espeland MA, Glick HA, Bertoni A, Brancati FL, Bray GA, Clark 31. Jeppesen J, Maarbjerg SJ, Jordy AB, Fritzen AM, Pehmoller C,
JM, Curtis JM, Egan C, Evans M, Foreyt JP, Ghazarian S, Gregg Sylow L, Serup AK, Jessen N, Thorsen K, Prats C, Qvortrup K, Dyck
EW, Hazuda HP, Hill JO, Hire D, Horton ES, Hubbard VS, Jakicic DJR, Hunter RW, Sakamoto K, Thomson D, Schjerling P, Wojtasze-
JM, Jeffery RW, Johnson KC, Kahn SE, Killean T, Kitabchi AE, wski JF, Richter EA, Kiens B. LKB-1 regulates lipid oxidation during
Knowler WC, Kriska A, Lewis CE, Miller M, Montez MG, Murillo A, exercise independently of AMPK. Diabetes 62: 1490 –1499, 2013.
Nathan DM, Nyenwe E, Patricio J, Peters AL, Pi-Sunyer X, Pownall 32. Jonker JT, de Mol P, de Vries ST, Widya RL, Hammer S, van
H, Redmon JB, Rushing J, Ryan DH, Safford M, Tsai AG, Wadden Schinkel LD, van der Meer RW, Gans RO, Webb AG, Kan HE, de
TA, Wing RR, Yanovski SZ, Zhang P; Look AHEAD Research Koning EJ, Bilo HJ, Lamb HJ. Exercise and type 2 diabetes mellitus:
Group. Impact of an intensive lifestyle intervention on use and cost of changes in tissue-specific fat distribution and cardiac function. Radiology
medical services among overweight and obese adults with type 2 diabetes: 269: 434 – 442, 2013.
the action for health in diabetes. Diabetes Care 37: 2548 –56, 2014. 33. Jorgensen SB, Viollet B, Andreelli F, Frosig C, Birk JB, Schjerling P,
14. Folli F, Saad MJA, Backer JM, Kahn CR, Saad MJ. Insulin stimulation Vaulont S, Richter EA, Wojtaszewski JF. Knockout of the ␣2 but not
of phosphatidylinositol 3-kinase activity and association with insulin ␣1 5=-AMP-activated protein kinase isoform abolishes 5-aminoimidazole-
receptor substrate 1 in liver and muscle of the intact rat. J Biol Chem 267: 4-carboxamide-1-␤-4-ribofuranosidebut not contraction-induced glucose
22171–22177, 1992. uptake in skeletal muscle. J Biol Chem 279: 1070 –1079, 2004.
15. Fujii N, Hirshman MF, Kane EM, Ho RC, Peter LE, Seifert MM, 34. Kaddai V, Le Marchand-Brustel Y, Cormont M. Rab proteins in
Goodyear LJ. AMP-activated protein kinase ␣2 activity is not essential endocytosis and Glut4 trafficking. Acta Physiol 192: 75– 88, 2008.
for contraction- and hyperosmolarity-induced glucose transport in skeletal 35. Kahn CR. Banting Lecture. Insulin action, diabetogenes, and the cause of
muscle. J Biol Chem 280: 39033–39041, 2005. type II diabetes. Diabetes 43: 1066 –1084, 1994.
16. Funai K, Cartee GD. Inhibition of contraction-stimulated AMP-activated 36. Kane S, Sano H, Liu SC, Asara JM, Lane WS, Garner CC, Lienhard
protein kinase inhibits contraction-stimulated increases in PAS-TBC1D1 GE. A method to identify serine kinase substrates. Akt phosphorylates a
and glucose transport without altering PAS-AS160 in rat skeletal muscle. novel adipocyte protein with a Rab GTPase-activating protein (GAP)
Diabetes 58: 1096 –1104, 2009. domain. J Biol Chem 277: 22115–22118, 2002.
17. Goodyear LJ, Giorgino F, Balon TW, Condorelli G, Smith RJ. Effects 37. Karlsson HK, Zierath JR, Kane S, Krook A, Lienhard GE, Wallberg-
of contractile activity on tyrosine phosphoproteins and phosphatidylino- Henriksson H. Insulin-stimulated phosphorylation of the Akt substrate
sitol 3-kinase activity in rat skeletal muscle. Am J Physiol Endocrinol AS160 is impaired in skeletal muscle of type 2 diabetic subjects. Diabetes
Metab 268: E987–E995, 1995. 54: 1692–1697, 2005.
18. Goodyear LJ, Kahn BB. Exercise, glucose transport, and insulin sensi- 38. Kelley DE, He J, Menshikova EV, Ritov VB. Dysfunction of mitochon-
tivity. Annu Rev Med 49: 235–261, 1998. dria in human skeletal muscle in type 2 diabetes. Diabetes 51: 2944 –2950,
19. Gregg EW, Chen H, Wagenknecht LE, Clark JM, Delahanty LM, 2002.
Bantle J, Pownall HJ, Johnson KC, Safford MM, Kitabchi AE, 39. Kemp BE, Mitchelhill KI, Stapleton D, Michell BJ, Chen ZP, Witters
Pi-Sunyer FX, Wing RR, Bertoni AG; Look AHEAD Research LA. Dealing with energy demand: the AMP-activated protein kinase.
Group. Association of an intensive lifestyle intervention with remission of
Trends Biochem Sci 24: 22–25, 1999.
type 2 diabetes. JAMA 308: 2489 –96, 2012.
40. Kennedy JW, Hirshman MF, Gervino EV, Ocel JV, Forse RA, Hoenig
20. Hardie DG, Carling D, Carlson M. The AMP-activated/SNF1 protein
SJ, Aronson D, Goodyear LJ, Horton ES. Acute exercise induces
kinase subfamily: metabolic sensors of the eukaryotic cell? Annu Rev
GLUT4 translocation in skeletal muscle of normal human subjects and
Biochem 67: 821– 855, 1998.
subjects with type 2 diabetes. Diabetes 48: 1192–1197, 1999.
21. Harris MI, Flegal KM, Cowie CC, Eberhardt MS, Goldstein DE,
41. Koh HJ, Arnolds DE, Fujii N, Tran TT, Rogers MJ, Jessen N, Li Y,
Little RR, Wiedmeyer HM, Byrd-Holt DD. Prevalence of diabetes,
Liew CW, Ho RC, Hirshman MF, Kulkarni RN, Kahn CR, Goodyear
impaired fasting glucose, and impaired glucose tolerance in U.S. adults.
LJ. Skeletal muscle-selective knockout of LKB1 increases insulin sensi-
The Third National Health and Nutrition Examination Survey, 1988 –
tivity, improves glucose homeostasis, and decreases TRB3. Mol Cell Biol
1994. Diabetes Care 21: 518 –524, 1998.
22. Higashida K, Kim SH, Higuchi M, Holloszy JO, Han DH. Normal 26: 8217– 8227, 2006.
adaptations to exercise despite protection against oxidative stress. Am J 42. Koh HJ, Toyoda T, Fujii N, Jung MM, Rathod A, Middelbeek RJ,
Physiol Endocrinol Metab 301: E779 –E784, 2011. Lessard SJ, Treebak JT, Tsuchihara K, Esumi H, Richter EA, Woj-
23. Hirshman MF, Koh HJ, Goodyear LJ. LKB1 in muscle is critical for taszewski JF, Hirshman MF, Goodyear LJ. Sucrose nonfermenting
exercise capacity and partially regulates glucose transport. Diabetes 55, AMPK-related kinase (SNARK) mediates contraction-stimulated glucose
Suppl 1: A13, 2006. transport in mouse skeletal muscle. Proc Natl Acad Sci USA 107: 15541–
24. Howlett KF, Andrikopoulos S, Proietto J, Hargreaves M. Exercise- 15546, 2010.
induced muscle glucose uptake in mice with graded, muscle-specific 43. Kramer HF, Witczak CA, Fujii N, Jessen N, Taylor EB, Arnolds DE,
GLUT4 deletion. Physiol Rep 1: e00065, 2013. Sakamoto K, Hirshman MF, Goodyear LJ. Distinct signals regulate
25. Hu FB, Manson JE, Stampfer MJ, Colditz G, Liu S, Solomon CG, AS160 phosphorylation in response to insulin, AICAR, and contraction in
Willett WC. Diet, lifestyle, and the risk of type 2 diabetes mellitus in mouse skeletal muscle. Diabetes 55: 2067–2076, 2006.
women. N Engl J Med 345: 790 –797, 2001. 44. Kramer HF, Witczak CA, Taylor EB, Fujii N, Hirshman MF, Good-
26. Hussey SE, McGee SL, Garnham A, McConell GK, Hargreaves M. year LJ. AS160 regulates insulin- and contraction-stimulated glucose
Exercise increases skeletal muscle GLUT4 gene expression in patients uptake in mouse skeletal muscle. J Biol Chem 281: 31478 –31485, 2006.
with type 2 diabetes. Diabetes Obes Metab 14: 768 –771, 2012. 45. Kramer HF, Taylor EB, Witczak CA, Fujii N, Hirshman MF, Good-
27. Jebailey L, Rudich A, Huang X, DiCiano-Oliveira C, Kapus A, Klip A. year LJ. The calmodulin-binding domain of AS160 regulates contraction-
Skeletal muscle cells and adipocytes differ in their reliance on TC10 and but not insulin-stimluated glucose uptake in skeletal muscle. Diabetes 56:
Rac for insulin-induced actin remodeling. Mol Endocrinol 18: 359 –372, 2854 –2058, 2006.
2004. 46. Little JP, Gillen JB, Percival ME, Safdar A, Tarnopolsky MA,
28. Jensen TE, Rose AJ, Jorgensen SB, Brandt N, Schjerling P, Wojtasze- Punthakee Z, Jung ME, Gibala MJ. Low-volume high-intensity interval
wski JF, Richter EA. Possible CaMKK-dependent regulation of AMPK training reduces hyperglycemia and increases skeletal muscle mitochon-
phosphorylation and glucose uptake at the onset of mild tetanic skeletal drial capacity in patients with type 2 diabetes. J Appl Physiol 111:
muscle contraction. Am J Physiol Endocrinol Metab 292: E1308 –E1317, 1554 –1560, 2011.
2007. 47. Look AHEAD Research Group. Effect of a long-term behavioral weight
29. Jensen TE, Sylow L, Rose AJ, Madsen AB, Angin YM, Stine J, loss intervention on nephropathy in overweight or obese adults with type
Richter EA. Contraction-stimulated glucose transport in muscle is con- 2 diabetes: a secondary analysis of the Look AHEAD randomized clinical
trolled by AMPK and mechanical stress but not sarcoplasmic reticulum trial. Lancet Diabetes Endocrinol 2: 801– 809, 2014.
Ca2⫹ release. Molecular Metabolism. In press. 48. Look AHEAD Research Group. Cardiovascular effects of intensive
30. Jensen TE, Schjerling P, Viollet B, Wojtaszewski JF, Richter EA. lifestyle intervention in type 2 diabetes. N Engl J Med 369: 145–154,
AMPK ␣1 activation is required for stimulation of glucose uptake by 2013.
twitch contraction, but not by H2O2, in mouse skeletal muscle. PLOS ONE 49. Lowell BB, Shulman GI. Mitochondrial dysfunction and type 2 diabetes.
3: e2102, 2008. Science 307: 384 –387, 2005.

Advances in Physiology Education • doi:10.1152/advan.00080.2014 • http://advan.physiology.org


Downloaded from journals.physiology.org/journal/advances (114.005.241.243) on February 23, 2021.
Refresher Course
314 MOLECULAR MECHANISMS REGULATING GLUCOSE UPTAKE IN MUSCLE

50. Maarbjerg SJ, Jørgensen SB, Rose AJ, Jeppesen J, Jensen TE, 69. Simard-Lefort N, St-Amand E, Morasse S, Cote CH, Marette A. The
Treebak JT, Birk JB, Schjerling P, Wojtaszewski JF, Richter EA. ␣ subunit of AMPK is essential for submaximal contraction-mediated
Genetic impairment of AMPK␣2 signaling does not reduce muscle glu- glucose transport in skeletal muscle in vitro. Am J Physiol Endocrinol
cose uptake during treadmill exercise in mice. Am J Physiol Endocrinol Metab 295: E1447–E1454, 2008.
Metab 297: E924 –E934, 2009. 70. Sriwijitkamol A, Coletta DK, Wajcberg E, Balbontin GB, Reyna SM,
51. Martin IK, Katz A, Wahren J. Splanchnic and muscle metabolism Barrientes J, Eagan PA, Jenkinson CP, Cersosimo E, DeFronzo RA,
during exercise in NIDDM patients. Am J Physiol Endocrinol Metab 269: Sakamoto K, Musi N. Effect of acute exercise on AMPK signaling in
E583–E590, 1995. skeletal muscle of subjects with type 2 diabetes: a time-course and
52. Meex RC, Schrauwen-Hinderling VB, Moonen-Kornips E, Schaart G, dose-response study. Diabetes 56: 836 – 848, 2007.
Mensink M, Phielix E, van de Weijer T, Sels JP, Schrauwen P, 71. Sylow L, Jensen TE, Kleinert M, Hojlund K, Kiens B, Wojtaszewski
Hesselink MK. Restoration of muscle mitochondrial function and meta- J, Prats C, Schjerling P, Richter EA. Rac1 signaling is required for
bolic flexibility in type 2 diabetes by exercise training is paralleled by insulin-stimulated glucose uptake and is dysregulated in insulin-resistant
increased myocellular fat storage and improved insulin sensitivity. Dia- murine and human skeletal muscle. Diabetes 62: 1865–1875, 2013.
betes 59: 572–579, 2010. 72. Sylow L, Jensen TE, Kleinert M, Mouatt JR, Maarbjerg SJ, Jeppesen
53. Merrill GF, Kurth EJ, Hardie DG, Winder WW. AICA riboside J, Prats C, Chiu TT, Boguslavsky S, Klip A, Schjerling P, Richter EA.
increases AMP-activated protein kinase, fatty acid oxidation, and glucose Rac1 is a novel regulator of contraction-stimulated glucose uptake in
uptake in rat muscle. Am J Physiol Endocrinol Metab 273: E1107–E1112, skeletal muscle. Diabetes 62: 1139 –1151, 2013.
1997. 73. Taylor EB, An D, Kramer HF, Yu H, Fujii NL, Roeckl KS, Bowles N,
54. Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihaq S, Hirshman MF, Xie J, Feener EP, Goodyear LJ. Discovery of TBC1D1
Lehar J, Puigserver P, Carlsson E, Ridderstrale M, Laurila E, Houstis as an insulin-, AICAR-, and contraction-stimulated signaling nexus in
N, Daly MJ, Patterson N, Mesirov JP, Golub TR, Tamayo P, Spiegel- mouse skeletal muscle. J Biol Chem 283: 9787–9796, 2008.
man B, Lander ES, Hirschhorn JN, Altshuler D, Groop LC. PCG-1␣- 74. Thong FS, Dugani CB, Klip A. Turning signals on and off: GLUT4
responsive genes involved in oxidative phosphorylation are coordinately traffic in the insulin-signaling highway. Physiology (Bethesda) 20: 271–
down regulated in human diabetes. Nat Genet 34: 267–273, 2003. 284, 2005.
55. Mu J, Brozinick JT Jr, Valladares O, Bucan M, Birnbaum MJ. A role 75. Treadway JL, James DE, Burcel E, Ruderman NB. Effect of exercise
for AMP-activated protein kinase in contraction- and hypoxia-regulated on insulin receptor binding and kinase activity in skeletal muscle. Am J
glucose transport in skeletal muscle. Mol Cell 7: 1085–1094, 2001. Physiol Endocrinol Metab 256: E138 –E144, 1989.
56. National Task Force on the Prevention and Treatment of Obesity. 76. Treebak JT, Birk JB, Rose AJ, Kiens B, Richter EA, Wojtaszewski
Overweight, obesity, and health risk. National Task Force on the Preven- JF. AS160 phosphorylation is associated with activation of ␣2␤2␥1- but
tion and Treatment of Obesity. Arch Intern Med 160: 898 –904, 2000. not ␣2␤2␥3-AMPK trimeric complex in skeletal muscle during exercise in
57. O’Neill HM, Maarbjerg SJ, Crane JD, Jeppesen J, Jorgensen SB, humans. Am J Physiol Endocrinol Metab 292: E715–E722, 2007.
Schertzer JD, Shyroka O, Kiens B, Van Denderen BJ, Tarnopolsky 77. Treebak JT, Glund S, Deshmukh A, Klein DK, Long YC, Jensen TE,
MA, Kemp BE, Richter EA, Steinberg GR. AMP-activated protein Jorgensen SB, Viollet B, Andersson L, Neumann D, Wallimann T,
Richter EA, Chibalin AV, Zierath JR, Wojtaszewski JF. AMPK-
kinase (AMPK) ␤1␤2 muscle null mice reveal an essential role for AMPK
mediated AS160 phosphorylation in skeletal muscle is dependent on
in maintaining mitochondrial content and glucose uptake during exercise.
AMPK catalytic and regulatory subunits. Diabetes 55: 2051–2058, 2006.
Proc Natl Acad Sci USA 108: 16092–16097, 2011.
78. Tuomilehto j Lindstrom J, Eriksson JG, Valle TT, Hamalainen H,
58. Pan XR, Li GW, Hu YH, Wang JX, Yang WY, An ZX, Hu ZX, Lin J,
Ilanne-Parikka P, Keinanen-Kiukaanniemi S, Laakso M, Louheranta
Xiao JZ, Cao HB, Liu PA, Jiang XG, Jiang YY, Wang JP, Zheng H,
A, Rastas M, Salminen V, Uusitupa M. Prevention of type 2 diabetes
Zhang H, Bennet PH, Howard PV. Effects of diet and exercise in
mellitus by changes in lifestyle among subjects with impaired glucose
preventing NIDDM in people with impaired glucose tolerance. The Da
tolerance. N Engl J Med 344: 1343–1350, 2001.
Qing IGT and Diabetes Study. Diabetes Care 20: 537–544, 1997.
79. Ueda S, Kitazaw S, Ishida K, Nishikawa Y, Matsui M, Matsumoto H,
59. Patti ME, Butte AJ, Crunkhorn S, Cusi K, Berria R, Kashyap S, Aoki T, Nozaki S, Takeda T, Tamori Y, Aiba A, Kahn CR, Kataoka
Miyazaki Y, Kohane I, Costello M, Saccone R, Landaker EJ, Goldfine T, Satoh T. Crucial role of the small GTPase Rac1 in insulin-stimulated
AB, Mun E, DeFronzo R, Finlayson J, Kahn CR, Mandarino LJ. translocation of glucose transporter 4 to the mouse skeletal muscle
Coordinated reduction of genes of oxidative metabolism in humans with sarcolemma. FASEB J 24: 2254 –2261, 2010.
insulin resistance and diabetes: potential role of PGC1 and NRF1. Proc 80. Vichaiwong K, Purohit S, An D, Toyoda T, Jessen N, Hirshman MF,
Natl Acad Sci USA 100: 8466 – 8471, 2003. Goodyear LJ. Contraction regulates site-specific phosphorylation of
60. Pette D, Staron RS. Transitions of muscle fiber phenotypic profiles. TBC1D1 in skeletal muscle. Biochem J 431: 311–320, 2010.
Histochem Cell Biol 115: 359 –372, 2001. 81. Wennerberg K, Rossman KL, Der CJ. The Ras superfamily at a glance.
62. Rejeski WJ, Bray GA, Chen SH, Clark JM, Evans M, Hill JO, Jakicic J Cell Sci 118: 843– 846, 2005.
JM, Johnson KC, Neiberg R, Ip EH; Look AHEAD Research Group. 82. Wing RR, Lang W, Wadden TA, Safford M, Knowler WC, Bertoni
Aging and physical function in type 2 diabetes: 8 years of an intensive AG, Hill JO, Brancati FL, Peters A, Wagenknecht L; Look AHEAD
lifestyle intervention. J Gerontol A Biol Sci Med Sci; doi:10.1093/gerona/ Research Group. Benefits of modest weight loss in improving cardio-
glu083. vascular risk factors in overweight and obese individuals with type 2
63. Richter EA, Hargreaves M. Exercise, GLUT4, and skeletal muscle diabetes. Diabetes Care 34: 1481–1486, 2011.
glucose uptake. Physiol Rev 93: 993–1017, 2013. 83. Witczak CA, Fujii N, Hirshman MF, Goodyear LJ. Ca2⫹/calmodulin-
64. Roach WG, Chavez JA, Mîinea CP, Lienhard GE:. Substrate specific- dependent protein kinase kinase-alpha regulates skeletal muscle glucose
ity and effect on GLUT4 translocation of the Rab GTPase-activating uptake independent of AMP-activated protein kinase and Akt activation.
protein Tbc1d1. Biochem J 403: 353–358, 2007. Diabetes 56: 1403–1409, 2007.
65. Robertson RP. Antagonist: diabetes and insulin resistance-philosophy, 84. Witczak CA, Jessen N, Warro DM, Toyoda T, Fujii N, Anderson ME,
science, and the multiplier hypothesis. J Lab Clin Med 125: 560 –564, Hirshman MF, Goodyear LJ. CaMKII regulates contraction- but not
1995. insulin-induced glucose uptake in mouse skeletal muscle. Am J Physiol
66. Rockl KS, Witczak CA, Goodyear LJ. Signaling mechanisms in skeletal Endocrinol Metab 298: E1150 –E1160, 2010.
muscle: acute responses and chronic adaptations to exercise. IUBMB Life 85. Wojtaszewski JF, Higaki Y, Hirshman MF, Michael MD, Dufresne
60: 145–53, 2008. SD, Kahn CR, Goodyear LJ. Exercise modulates postreceptor insulin
67. Sakamoto K, McCarthy A, Smith D, Green KA, Grahame HD, signaling and glucose transport in muscle-specific insulin receptor knock-
Ashworth A, Alessi DR. Deficiency of LKB1 in skeletal muscle prevents out mice. J Clin Invest 104: 1257–1264, 1999.
AMPK activation and glucose uptake during contraction. EMBO J 24: 86. Wright DC, Hucker KA, Holloszy JO, Han DH. Ca2⫹ and AMPK both
1810 –1820, 2005. mediate stimulation of glucose transport by muscle contractions. Diabetes
68. Sano H, Kane S, Sano E, Miinea CP, Asara JM, Lane WS, Garner 53: 330 –335, 2004.
CW, Lienhard GE. Insulin-stimulated phosphorylation of a Rab GTPase- 87. Zierath JR, Krook A, Wallberg-Henriksson H. Insulin action in skeletal
activating protein regulates GLUT4 translocation. J Biol Chem 278: muscle from patients with NIDDM. Mol Cell Biochem 182: 153–160,
14599 –14602, 2003. 1998.

Advances in Physiology Education • doi:10.1152/advan.00080.2014 • http://advan.physiology.org


Downloaded from journals.physiology.org/journal/advances (114.005.241.243) on February 23, 2021.

You might also like