You are on page 1of 8

J Appl Physiol 110: 561–568, 2011.

First published November 11, 2010; doi:10.1152/japplphysiol.00941.2010. Review

HIGHLIGHTED TOPIC Signals Mediating Skeletal Muscle Remodeling by Activity

Signals mediating skeletal muscle remodeling by resistance exercise:


PI3-kinase independent activation of mTORC1
Andrew Philp,1 D. Lee Hamilton,2 and Keith Baar1
1
Department of Neurobiology, Physiology, and Behavior, University of California–Davis, Davis, California;
and 2Biomedical Research Institute, University of Dundee, Dundee, Scotland
Submitted 16 August 2010; accepted in final form 5 November 2010

Philp A, Hamilton DL, Baar K. Signals mediating skeletal muscle remod-


eling by resistance exercise: PI3-kinase independent activation of mTORC1. J
Appl Physiol 110: 561–568, 2011. First published November 11, 2010;
doi:10.1152/japplphysiol.00941.2010.—For over 10 years, we have known that
the activation of the mammalian target of rapamycin complex 1 (mTORC1) has
correlated with the increase in skeletal muscle size and strength that occurs
following resistance exercise. Initial cell culture and rodent models of muscle
growth demonstrated that the activation of mTORC1 is common to hypertrophy
induced by growth factors and increased loading. The further observation that high
loads increased the local production of growth factors led to the paradigm that
resistance exercise stimulates the autocrine production of factors that act on
membrane receptors to activate mTORC1, and this results in skeletal muscle
hypertrophy. Over the last few years, there has been a paradigm shift. From both
human and rodent studies, it has become clear that the phenotypic and molecular
responses to resistance exercise occur in a growth factor-independent manner.
Although the mechanism of load-induced mTORC1 activation remains to be
determined, it is clear that it does not require classical growth factor signaling.
mammalian target of rapamycin complex 1

SINCE THE ELEGANT EXPERIMENTS by Wong and Booth (83– 85) in ribosomal proteins and the mitogen-induced increase in protein
the late 1980s, we have known that the load across a muscle is synthesis (74). Since increased load across a muscle increased
the primary determinant of skeletal muscle hypertrophy (for the rate of protein synthesis (16, 50, 83, 84) and growth factors
review, see Ref. 3). However, how the load across a muscle is like insulin-like growth factor (IGF)-1 were known to induce
transduced into an increase in the rate of protein synthesis and muscle hypertrophy (18), the natural hypothesis was that mus-
the accretion of contractile protein has yet to be fully deter- cle hypertrophy in the adult was the result of an increase in
mined. The first molecular marker of the increase in muscle mitogen signaling following resistance exercise. In support of
mass and strength following resistance exercise was the phos- this hypothesis, treating myotubes with IGF-1 in vitro resulted
phorylation and activation of the 70-kDa ribosomal S6 protein in the activation of S6K1 and an increase in the size of the
kinase (S6K1; Refs. 4, 51, 73). Subsequent work demonstrated myotubes (60); following loading there was an increase in the
that S6K1 was under the control of the mammalian target of expression of IGF-1 mRNA in muscle that occurred even in
the absence of growth hormone (23); and stretching myotubes
rapamycin (mTOR) complex 1 (mTORC1) and that the specific
in vitro resulted in the release of a factor into the media that
mTORC1 inhibitor, rapamycin, could directly block muscle
was able to activate S6K1 (5). So the growth factor paradigm
growth (11). These initial studies have since been supported by was set: 1) resistance exercise increased the autocrine release
transgenic models of mTORC1 inactivation (59) and dissoci- of IGF-1; 2) IGF-1 bound to the IGF-1 receptor on the cell
ation (9) and global knockout of S6K1 (66). Collectively, these surface and activated S6K1 via the canonical growth factor
studies implicate mTORC1 and S6K1 as principal mediators of pathway; 3) activation of S6K1 led to an increase in protein
muscle growth in skeletal muscle. synthesis; and 4) when repeated at a sufficient frequency, this
The original observation that S6K1 correlated with muscle autocrine signaling response led to an increase in muscle mass
growth was striking since S6K1 had formally been identified as and strength (Fig. 1).
a mitogen (or growth factor) activated protein kinase (42, 55,
DECONSTRUCTING THE PARADIGM: GROWTH FACTOR-
58) whose activity was correlated with the phosphorylation of
INDEPENDENT ACTIVATION OF S6K1

Address for reprint requests and other correspondence: K. Baar, 1 Shields Over the last few years, pharmacological, genetic, and trans-
Ave., 174 Briggs Hall, Univ. of California, Davis, CA 95616 (e-mail: genic approaches have been employed by several groups to
kbaar@ucdavis.edu). systematically manipulate the IGF1-PI-3 kinase-PKB pathway
http://www.jap.org 8750-7587/11 Copyright © 2011 the American Physiological Society 561
Downloaded from www.physiology.org/journal/jappl by ${individualUser.givenNames} ${individualUser.surname} (177.184.218.005) on December 11, 2017.
Copyright © 2011 American Physiological Society. All rights reserved.
Review
562

Fig. 1. Canonical growth factor signaling path-


way from insulin/IGF1 to mTORC1 and pro-
tein synthesis. Growth factor binding to the
receptor results in auto tyrosine phosphoryla-
tion and recruitment of the insulin receptor
substrates (IRS). IRS relocalization and phos-
phorylation recruit phosphoinositol-3 kinase
(p85 and p110/PI3K) to the membrane result-
ing in the phosphorylation of PI4,5P2 into
PI3,4,5P3(PIP3). PIP3 recruits protein kinase B
(PKB) and phosphoinositol-dependent kinase
(PDK)1 to the membrane resulting in activation
of PKB. PKB phosphorylates and inactivates
the GTPase-activating tuberosclerosis complex
(TSC1/2). Inactivation of TSC results in the
activation of the ras homologous protein en-
riched in brain (Rheb) and mTORC1 (mTOR,
raptor, and G␤L). mTORC1 phosphorylates its
primary downstream targets of the eukaryotic
initiation factor 4E binding protein (4EBP) and
the 70KDa ribosomal S6 protein kinase (S6K1),
resulting in an increase in protein synthesis.

and determine its importance in load-induced muscle growth. and neither insulin nor IGF-1 could activate PKB/Akt, showing
The first suggestion that the increase in S6K1 activation fol- that IGF-1 and PI-3 kinase signaling is required for develop-
lowing loading could occur independent of canonical growth mental muscle growth. However, following 7 or 35 days of
factor signaling came in 2004 when Hornberger et al. (40) mechanical overload, both the wild-type and the MKR mice
showed that ex vivo stretch of isolated extensor digitorum showed an increase in muscle mass of ⬃50 and 90%, respec-
longus muscles in the presence of the PI3-kinase inhibitor tively. Furthermore, the phosphorylation of both PKB and
wortmannin did not prevent the stretch-induced activation in S6K1 was completely normal in the MKR mice in response to
S6K1. In these experiments, insulin activation of S6K1 was overload.
prevented by both wortmannin (PI-3 kinase inhibitor) and However, two points should be made regarding the MKR
rapamycin (an inhibitor of mTORC1), indicating that insulin mouse model. First, it should be noted that a follow-up study
activated S6K1 by the canonical growth factor ¡ PI-3 kinase by the same group (82) showed that, in response to an acute
¡ mTORC1 ¡ S6K1 pathway. However, stretch activation of bout of resistance exercise, the MKR mice did show a delayed
S6K1 occurred in a rapamycin-sensitive, wortmannin-indepen- and diminished phosphorylation of S6K1 in response to resis-
dent mechanism, demonstrating for the first time that PI-3 tance exercise, suggesting that input from PI-3 kinase maybe
kinase was not required for stretch-induced activation of required for full activation of mTORC1. Second, Heron-Mil-
mTORC1. In support of this hypothesis, protein kinase B/Akt, havet and colleagues (38) recently reported that the MKR mice
the molecular link between PI-3 kinase and mTORC1, was not demonstrate impaired muscle regeneration and myoblast dif-
required for S6K1 activation by stretch (40). However, it ferentiation following injury. Furthermore, the authors ob-
should be noted that both wortmannin and rapamycin blocked served an increase in progenitor cell number in the MKR mice
the stretch-activated increase in protein synthesis, suggesting and speculated that the normal response to overload hypertro-
that PI-3 kinase was, in the least, permissive for the actions of phy reported by Spagenburg and colleagues may be through
stretch on protein synthesis. compensatory hyperplasia (increased activation and prolifera-
The next major blow to the growth factor paradigm came in tion of progenitor cells), as has previously been reported (27).
2008 from the laboratory of Espen Spangenburg (67). Using a However, it is not clear how this would occur if myoblast
mouse with skeletal muscle that harbored an inactivating differentiation were impaired. In fact, the inability to regener-
knockin mutation in the IGF receptor (MKR) that prevented ate and a decrease in myoblast differentiation suggests a
receptor activation by insulin or IGF-1, Spangenburg and decrease in satellite cell activation and fusion in the MKR
colleagues (67) showed that, in the absence of IGF signaling, mice, bringing into question the need for these cells for
load-induced muscle hypertrophy was identical to that of hypertrophy in the mouse.
wild-type mice, even though muscles from the MKR mice did Using another genetic model (electroporation of DNA into
not grow as large developmentally. At 8 wk of age, the muscles adult muscle), Goodman et al. (33) have shown that activation
of the MKR mice were ⬃30% smaller than wild-type mice, of PI-3 kinase is not required for mTORC1-induced skeletal

J Appl Physiol • VOL 110 • FEBRUARY 2011 • www.jap.org


Downloaded from www.physiology.org/journal/jappl by ${individualUser.givenNames} ${individualUser.surname} (177.184.218.005) on December 11, 2017.
Copyright © 2011 American Physiological Society. All rights reserved.
Review
563
muscle hypertrophy. In these experiments, the small G-protein in an IRS1/2-independent manner, we generated mice with
activator of mTORC1, Rheb (ras homologous protein enriched muscles devoid of the phosphoinositol phosphatase PTEN. We
in brain), was overexpressed either in vitro or in vivo to hypothesized that if PI-3 kinase was important in the activation
increase mTORC1 activity in a PI-3 kinase-independent fash- of mTORC1 following loading, the PTEN knockout mice
ion. The authors were then able to demonstrate that augment- would show greater accumulation of PI(3,4,5)P3 and therefore
ing Rheb levels increased cap-dependent translation and re- greater PKB/mTORC1 activity and muscle hypertrophy. In
sulted in a 64% increase in the cross-sectional area of muscle contrast to this hypothesis, neither the phosphorylation of
fibers. Even though Goodman et al. (32) did not demonstrate S6K1 following resistance exercise nor the increase in muscle
Rheb association with mTORC1 following load-induced hy- mass following overload was different between the wild-type
pertrophy, their data can be interpreted to show that PI-3 and PTEN knockout mice.
kinase-independent skeletal muscle hypertrophy is genetically The activation of mTORC1 in a PI-3 kinase-independent
possible in vivo. manner is consistent with both the time course of PKB and
It is one thing to demonstrate that experimental models can S6K1 activation and the variability observed in PKB activation
be genetically created where adult muscle growth can occur in following resistance exercise (Fig. 2). We and others (54) have
a growth factor-independent manner; it is another thing entirely shown that following resistance exercise the activation of
to show this in humans. West et al. (80) have done just that by S6K1 precedes or occurs concomitant with that of PKB and, in
taking advantage of the fact that resistance exercise using a humans, the activation of PKB following resistance exercise is
large muscle mass increases circulating levels of anabolic more dependent on the feeding state rather than the intensity of
hormones (1). Using heavy resistance exercise with the legs, the exercise session (22). Since most animal studies occur in
West and colleagues were able to create a high hormone state. the fed state, whereas the feeding state is far more controlled in
The high hormone state was characterized by a 5-fold increase human studies, these findings are completely consistent with
in total testosterone, a 3-fold increase in free testosterone, and PKB being activated by an increase in nutrients resulting from
a 10-fold increase in both growth hormone and IGF-1. The
the increase in blood flow during, or immediately following,
authors then used a unilateral resistance exercise program in
exercise and not the exercise bout or the autocrine release of
the arms where one arm performed resistance exercise imme-
growth factors.
diately before heavy lifting with the legs (therefore in a high
hormone state), whereas the other arm was exercised on a
separate day without concomitant exercise of the legs (a low A NEW PARADIGM: MECHANOSENSING DRIVES
hormone state). Despite the huge differences in circulating LOAD-INDUCED SKELETAL MUSCLE HYPERTROPHY
hormone levels subsequent to each bout of exercise, there was Taken together, the above data support a new paradigm
no difference in either the increase in muscle fiber size or where mTORC1 is activated in a PI-3 kinase/PKB-independent
overall strength in the arms following 15 wk of training. manner. This shift in paradigm parallels research into the
Consistent with the absence of a difference in muscle strength activation of mTORC1 by amino acids. Like resistance exer-
or fiber size, they have also demonstrated that mTORC1
cise, amino acids activate mTORC1 in a PI-3 kinase/PKB-
signaling was not different between the high and low hormone
independent manner. Instead of working through the PI-3
states (81). This work confirms earlier animal studies that
kinase/PKB pathway, amino acids activate mTORC1 by alter-
showed that neither hypothalamic nor pancreatic hormones
ing its subcellular location (45, 46, 61, 62). In the absence of
were required for load-induced skeletal muscle hypertrophy
amino acids, mTORC1 is diffuse throughout the cell. When
(30, 31). However, the fact that circulating hormones are not
required for muscle hypertrophy does not preclude the possi- amino acids are added back, mTORC1 is shuttled to the
bility that local production of IGF-1 or another growth factor lysosomal membrane through its interaction with the Rag
may play an important role in adult skeletal muscle hypertro- family of small G-proteins and the Ragulator, a protein scaf-
phy. fold found on the lysosomal membrane (45, 61, 62). Once at
Given the data from the Spangenburg and Hornberger lab- the lysosome, mTORC1 can interact with Rheb. The interac-
oratories using transgenic approaches, growth factor-indepen- tion with Rheb increases the basal level of mTORC1 activity
dent hypertrophy is possible. However, whether this was due to and permits the activation of mTORC1 by growth factors and
compensatory changes in the muscle and whether autocrine other stimuli (61). This model fits very well with what we
signaling through tyrosine phosphorylation of the IGF-1 recep- know about skeletal muscle hypertrophy: 1) the basal level of
tor and the insulin receptor substrates (IRS) to PI-3 kinase mTORC1 activity (as determined by S6K1 phosphorylation) is
occurred normally remained an open question. We have re- extremely low; 2) the addition of amino acids increases
cently directly tested this hypothesis in rat and mouse skeletal mTORC1 activity and protein synthesis (7, 12, 13, 20); 3) the
muscle in response to high-frequency electrical stimulation ability of amino acids and loading to increase protein synthesis
(37). In support of IGF-1-independent activation of mTORC1, is dependent on a permissive level of hormonal activity (8, 26,
at no time following resistance exercise (from immediately 40); and 4) growth factors and resistance exercise can acti-
after to 48 h later) was there an increase in tyrosine phosphor- vate mTORC1 and increase protein synthesis more effi-
ylation of the IGF-1 receptor. Furthermore, we observed a ciently in the presence of amino acids (57, 72, 75, 76).
decrease in signaling through IRS1/2 to PI-3 kinase. In sharp Interestingly, we have shown that resistance exercise in-
contrast to insulin, p85 associated with either IRS1 or IRS2 creases the activity of the vesicular trafficking protein
decreased in the first 3 h after the resistance exercise before Vps34 (51). This might improve the trafficking of mTORC1
returning to control levels (37). To determine whether this was to the lysosome and permit the prolonged activation of
a measurement error or possibly that PI-3 kinase was activated mTORC1 following resistance exercise. However, whether

J Appl Physiol • VOL 110 • FEBRUARY 2011 • www.jap.org


Downloaded from www.physiology.org/journal/jappl by ${individualUser.givenNames} ${individualUser.surname} (177.184.218.005) on December 11, 2017.
Copyright © 2011 American Physiological Society. All rights reserved.
Review
564
complexes serve this purpose in muscle including the dystro-
phin-associated glycoprotein complex, integrin-associated cos-
tameric complex, and cell-cell adherins junctions. The role of
some of these protein complexes in mechanosensing has been
studied in the heart with some interesting results. In the heart,
the costameres appear to be the primary mechanical sensor.
␤-Integrins (44, 65), the integrin-linked kinase (ILK) (6),
melusin (14, 21), the muscle LIM domain protein (47), and
PKB all are found in costameres and all play an important role
in mechanosensing and cardiac hypertrophy. Of specific inter-
est is ILK. ILK is primarily a scaffolding protein but may still
harbor some kinase activity. The primary role of ILK in
hypertrophy is due to its ability to interact and bring together
␤-integrins, PKB, and the rapamycin-insensitive companion of
mTOR (rictor). Rictor coordinates the phosphorylation of PKB
at Thr473 as part of the other known mTOR complex
(mTORC2). The activation of PKB can lead to muscle hyper-
trophy in both the heart and skeletal muscle (11), suggesting
that ILK may coordinate the conversion of the mechanical
stimulus from the ␤-integrins into the chemical signal of PKB
phosphorylation. However, as discussed above, PKB␣/Akt1 is
not required for the activation of mTORC1 in response to
stretch. This means that either other isoforms can compensate
for the loss of PKB␣/Akt1 in muscle or the ␤-integrin/ILK/
PKB/mTORC1 pathway is not the mechanosensor in skeletal
muscle.
Another member of costameres and adherins junctions is
focal adhesion kinase (FAK) (15). Work by Fluck et al. (28)
demonstrated that the amount and activity of FAK increase
within 24 –36 h of the onset of stretch in the chicken ALD and
in the rat soleus after overload. Furthermore, the degree of
FAK expression and activity in muscles is related to their
loading (34). For instance, the postural soleus muscle has
greater expression and tyrosine phosphorylation of FAK than
the gastrocnemius or plantaris (34). Moreover, unloading re-
duces the phosphorylation of FAK in the soleus and the
concentration of FAK in the gastrocnemius and plantaris (34).
This suggests that load may be sensed through FAK. However,
unlike ILK, it is unclear what the downstream targets of FAK
Fig. 2. The phosphorylation of S6K1 precedes that of PKB in rodent muscle
following resistance exercise. Western blots showing the phosphorylation of
may be involved in this process; additionally, it has yet to be
PKB (A) and S6K1 (C) following resistance exercise. Notice first that S6K1 determined whether FAK is activated by acute resistance
phosphorylation occurs before that of PKB in post absorptive rats following 10 exercise with a time course consistent with it being proximal to
sets of 6 contractions induced by high-frequency electrical stimulation. Sec- mTORC1.
ond, notice that PKB phosphorylation is elevated only transiently (at 30 min), Calcium has also been proposed as a mechanosensor. Gu-
whereas S6K1 remains phosphorylated for at least 18 h. Last, notice the
magnitude of the activation. PKB phosphorylation increases 3-fold, whereas haray and Sachs (35) first identified stretch-activated ion chan-
S6K1 phosphorylation increases ⬃80-fold (36). nels that could be blocked by the antibiotic streptomycin and
the cation gadolinium. Spangenburg and McBride (68) have
shown that treating animals with streptomycin or gadolinium
resistance exercise increases mTORC1 association with Rheb before a bout of resistance exercise can decrease S6K1 activa-
at the lysosome has yet to be determined. tion without impacting muscle function, suggesting that these
Amino acid signaling through the RagGTPase proteins to channels may be important in mechanosensing. Further support
mTORC1 provides a viable signaling mechanism; namely that for this theory comes from the fact that increasing intracellular
the GTP binding state of the Rag heterodimer is altered in the calcium in muscle cells using the calcium ionophore A23187
presence of amino acids (45, 61, 62). However, it is unclear enhances both protein synthesis and degradation rates (43)
how a change in the load on a muscle would produce a PI-3 much like resistance exercise. However, stretch in combination
kinase-independent signal to mTORC1. The obvious answer is with A23187 enhances protein synthesis further than treatment
that there is a mechanosensor that can independently activate with A23187 alone (43). This means that, although calcium
mTORC1. However, the identity of such a sensor remains influx may play a role in mechanosensing, it is not the only
elusive. The ideal position for a mechanosensor is within a stretch sensor in muscle.
region that connects the internal architecture of the cell to the Goldberg and Goodman (32) were the first to observe that
extracellular matrix or to other cells (41). A number of protein amino acid transport into loaded muscles was increased in both

J Appl Physiol • VOL 110 • FEBRUARY 2011 • www.jap.org


Downloaded from www.physiology.org/journal/jappl by ${individualUser.givenNames} ${individualUser.surname} (177.184.218.005) on December 11, 2017.
Copyright © 2011 American Physiological Society. All rights reserved.
Review
565
normal and hyposectamized rats. An increase in amino acid In the absence of external amino acids, protein degradation
uptake, specifically the branched chain amino acids, is also may play an important role in supplying amino acids for the
seen 3 h after resistance exercise in humans (10) and 90 min activation of mTORC1 and protein synthesis (63, 64). Early
after resistance exercise in rats (51). As discussed above, studies in fasting humans demonstrated a direct association
amino acids, specifically leucine, are known to activate between protein synthesis and degradation following resistance
mTORC1 via the RagGTPases (45, 61, 62) and increase exercise (56). However, when essential amino acids are sup-
muscle protein synthesis, suggesting that amino acid influx plied, the increase in degradation is attenuated (75), suggesting
may function as a mechanosensor in skeletal muscle. In fact, that degradation serves to increase internal amino acids, pro-
when muscle cells are stretched in vitro, the increase in amino vide a source of essential amino acids for de novo synthesis,
acid uptake is required for stretch-induced activation of protein and possibly activate mTORC1.
synthesis (77). Since the permeability of muscle to amino acids Hornberger and his colleagues have suggested that the phos-
increases following resistance exercise and amino acids in- pholipid cleaving enzyme phospholipase D (PLD) could serve
crease mTORC1 activity and protein synthesis, it is not sur- as a mechanoreceptor. One of the products of PLD, the second
prising that protein supplementation in association with resis- messenger phosphatidic acid (PA), increases in response to
tance exercise has a synergistic effect on protein synthesis rates 15–90 min of mechanical stretch (39) or within 2 min follow-
and muscle hypertrophy (25, 48, 75). However, the influx of ing the start of electrical stimulation (17). Hornberger and his
amino acids is not seen immediately after resistance exercise group (39, 54) have used two chemically distinct inhibitors of
and rapidly returns to normal (3 h in the rat), even though PLD to demonstrate that blocking PLD prevents the production
mTORC1 activity remains high for at least another 15 h (51). of PA in response to stretch and the mechanical activation of

Fig. 3. Mechanical activation of mTORC1. In this model, the initiating signal for mTORC1 activation goes through a stretch-activated calcium channel, integrins
[␣/␤ and integrin linked kinase (ILK)], and/or amino acids. These signals activate Rheb and induce the RagGTPase-dependent recruitment of mTORC1 to the
lysosome. With mTORC1 at the lysosome, Rheb activates mTORC1 possibly through the recruitment of phospholipase D (increasing PA locally). Once active,
mTORC1 phosphorylates its downstream targets 4EBP and S6K1, upregulates miR-1 resulting in an increase in follistatin and myostatin inhibition, and increases
amino acid transporters possibly through the myc protooncogene. S6K1 in turn phosphorylates upstream binding factor (UBF) and elongation factor (EF) 2
kinase, resulting in a decrease in eEF2 phosphorylation.

J Appl Physiol • VOL 110 • FEBRUARY 2011 • www.jap.org


Downloaded from www.physiology.org/journal/jappl by ${individualUser.givenNames} ${individualUser.surname} (177.184.218.005) on December 11, 2017.
Copyright © 2011 American Physiological Society. All rights reserved.
Review
566
mTORC1 (Fig. 3). Furthermore, the same group has shown DISCLOSURES
that the increase in PA with in vivo contractions only occurs in No conflicts of interest, financial or otherwise, are declared by the author(s).
muscles that will undergo hypertrophy (54). These data suggest
that PA is important in the activation of mTORC1 by mechan- REFERENCES
ical loading. However, whether PLD activation is the mechan- 1. Ahtiainen JP, Pakarinen A, Kraemer WJ, Hakkinen K. Acute hor-
ical sensor or simply a marker of an upstream event has yet to monal and neuromuscular responses and recovery to forced vs. maximum
be determined. RNA interference experiments have revealed repetitions multiple resistance exercises. Int J Sports Med 24: 410 –418,
that the mTORC1 activator Rheb binds to, and activates, PLD 2003.
2. Amirouche A, Durieux AC, Banzet S, Koulmann N, Bonnefoy R,
in a GTP-dependent manner (70). Knockdown of PLD pre- Mouret C, Bigard X, Peinnequin A, Freyssenet D. Down-regulation of
vents Rheb-induced activation of mTORC1, whereas knock- Akt/mammalian target of rapamycin signaling pathway in response to
down of Rheb decreases the activation of PLD in response to myostatin overexpression in skeletal muscle. Endocrinology 150: 286 –
serum (70). In contrast, Rheb overexpression activates PLD in 294, 2009.
serum-starved cells. Together, these data suggest that activa- 3. Baar K. The signaling underlying FITness. Appl Physiol Nutr Metab 34:
411–419, 2009.
tion of Rheb recruits PLD to mTORC1 and enhances the local 4. Baar K, Esser K. Phosphorylation of p70(S6k) correlates with increased
production of phosphatidic acid, resulting in the activation of skeletal muscle mass following resistance exercise. Am J Physiol Cell
mTORC1. Therefore, the activation of PLD by mechanical Physiol 276: C120 –C127, 1999.
loading may simply reflect the upstream activation of Rheb and 5. Baar K, Torgan CE, Kraus WE, Esser K. Autocrine phosphorylation of
p70(S6k) in response to acute stretch in myotubes. Mol Cell Biol Res
not that PLD is a mechanosensor per se. Commun 4: 76 –80, 2000.
6. Bendig G, Grimmler M, Huttner IG, Wessels G, Dahme T, Just S,
CONCLUSIONS Trano N, Katus HA, Fishman MC, Rottbauer W. Integrin-linked
kinase, a novel component of the cardiac mechanical stretch sensor,
Even though we have focused on the role of mTORC1 and controls contractility in the zebrafish heart. Genes Dev 20: 2361–2372,
S6K1 in the development of skeletal muscle hypertrophy, this 2006.
7. Bennet WM, Connacher AA, Scrimgeour CM, Rennie MJ. The effect
is not the only mediator of adult muscle growth. Myostatin (78, of amino acid infusion on leg protein turnover assessed by L-[15N]pheny-
79), notch (19, 53), and microRNAs (24, 52) also play an lalanine and L-[1–13C]leucine exchange. Eur J Clin Invest 20: 41–50, 1990.
important role in the development of muscle hypertrophy. 8. Bennet WM, Connacher AA, Smith K, Jung RT, Rennie MJ. Inability
However, whether these are distinct pathways or part of a to stimulate skeletal muscle or whole body protein synthesis in type 1
(insulin-dependent) diabetic patients by insulin-plus-glucose during
coordinated system has yet to be determined. For instance, amino acid infusion: studies of incorporation and turnover of tracer
mTORC1 can regulate the production of miR-1, resulting in an L-[1–13C]leucine. Diabetologia 33: 43–51, 1990.
increase in the endogenous inhibitor of myostatin, follistatin 9. Bentzinger CF, Romanino K, Cloetta D, Lin S, Mascarenhas JB,
decreasing myostatin activity (71). In turn, myostatin can Oliveri F, Xia J, Casanova E, Costa CF, Brink M, Zorzato F, Hall
MN, Ruegg MA. Skeletal muscle-specific ablation of raptor, but not of
inhibit the activation of mTORC1 (2, 78). Together, these rictor, causes metabolic changes and results in muscle dystrophy. Cell
effects would produce a positive feedback loop following Metab 8: 411–424, 2008.
resistance exercise where the activation of mTORC1 would 10. Biolo G, Maggi SP, Williams BD, Tipton KD, Wolfe RR. Increased
decrease myostatin signaling, the decrease in myostatin would rates of muscle protein turnover and amino acid transport after resistance
increase mTORC1 activity, and the cycle would repeat. exercise in humans. Am J Physiol Endocrinol Metab 268: E514 –E520,
1995.
It is likely that more than one factor is responsible for 11. Bodine SC, Stitt TN, Gonzalez M, Kline WO, Stover GL, Bauerlein R,
growth signaling in response to resistance exercise. It is pos- Zlotchenko E, Scrimgeour A, Lawrence JC, Glass DJ, Yancopoulos
sible that one sensor starts the cycle and the others function to GD. Akt/mTOR pathway is a crucial regulator of skeletal muscle hyper-
maintain mTORC1 activity at various times after resistance trophy and can prevent muscle atrophy in vivo. Nat Cell Biol 3: 1014 –
1019, 2001.
exercise. Since the ability to increase protein synthesis and 12. Bohe J, Low A, Wolfe RR, Rennie MJ. Human muscle protein synthesis
accrue muscle is an important and evolutionarily conserved is modulated by extracellular, not intramuscular amino acid availability: a
process, it would not be surprising if significant redundancy dose-response study. J Physiol 552: 315–324, 2003.
existed, making it difficult to dissect out the hierarchy of these 13. Bohe J, Low JF, Wolfe RR, Rennie MJ. Latency and duration of
signals. It is important to make clear that for the sake of this stimulation of human muscle protein synthesis during continuous infusion
of amino acids. J Physiol 532: 575–579, 2001.
concise review series we decided to discuss factors involved in 14. Brancaccio M, Fratta L, Notte A, Hirsch E, Poulet R, Guazzone S, De
muscle hypertrophy following mechanical loading. As noted in Acetis M, Vecchione C, Marino G, Altruda F, Silengo L, Tarone G,
the recent Point-Counterpoint series in this Journal (29, 69), Lembo G. Melusin, a muscle-specific integrin beta1-interacting protein, is
IGF-1 has important functions in development, the remodeling required to prevent cardiac failure in response to chronic pressure over-
load. Nat Med 9: 68 –75, 2003.
of skeletal muscle, the extracellular matrix, and the activation 15. Carson JA, Wei L. Integrin signaling’s potential for mediating gene
of progenitor cells. Certainly, in humans it is likely that many expression in hypertrophying skeletal muscle. J Appl Physiol 88: 337–343,
molecular signals are required to maintain or increase muscle 2000.
mass. Therefore, future research, such as the innovative ap- 16. Chesley A, MacDougall JD, Tarnopolsky MA, Atkinson SA, Smith K.
proach employed by West et al. (80), is needed to determine Changes in human muscle protein synthesis after resistance exercise. J
Appl Physiol 73: 1383–1388, 1992.
whether murine models translate completely to humans. These 17. Cleland PJ, Appleby GJ, Rattigan S, Clark MG. Exercise-induced
studies are extremely relevant because, with the growing el- translocation of protein kinase C and production of diacylglycerol and
derly population and the anabolic resistance and muscle loss phosphatidic acid in rat skeletal muscle in vivo. Relationship to changes in
that occurs with age (49), viable clinical options to rectify this glucose transport. J Biol Chem 264: 17704 –17711, 1989.
18. Coleman ME, DeMayo F, Yin KC, Lee HM, Geske R, Montgomery C,
situation are desperately needed. These issues, as well as how Schwartz RJ. Myogenic vector expression of insulin-like growth factor I
mTORC1 is activated and what mTORC1 does, should keep us stimulates muscle cell differentiation and myofiber hypertrophy in trans-
busy for years to come. genic mice. J Biol Chem 270: 12109 –12116, 1995.

J Appl Physiol • VOL 110 • FEBRUARY 2011 • www.jap.org


Downloaded from www.physiology.org/journal/jappl by ${individualUser.givenNames} ${individualUser.surname} (177.184.218.005) on December 11, 2017.
Copyright © 2011 American Physiological Society. All rights reserved.
Review
567
19. Conboy IM, Conboy MJ, Smythe GM, Rando TA. Notch-mediated 41. Ingber DE. Tensegrity: the architectural basis of cellular mechanotrans-
restoration of regenerative potential to aged muscle. Science 302: 1575– duction. Annu Rev Physiol 59: 575–599, 1997.
1577, 2003. 42. Jeno P, Ballou LM, Novak-Hofer I, Thomas G. Identification and
20. Crozier SJ, Kimball SR, Emmert SW, Anthony JC, Jefferson LS. Oral characterization of a mitogen-activated S6 kinase. Proc Natl Acad Sci USA
leucine administration stimulates protein synthesis in rat skeletal muscle. 85: 406 –410, 1988.
J Nutr 135: 376 –382, 2005. 43. Kameyama T, Etlinger JD. Calcium-dependent regulation of protein
21. De Acetis M, Notte A, Accornero F, Selvetella G, Brancaccio M, synthesis and degradation in muscle. Nature 279: 344 –346, 1979.
Vecchione C, Sbroggio M, Collino F, Pacchioni B, Lanfranchi G, 44. Keller RS, Shai SY, Babbitt CJ, Pham CG, Solaro RJ, Valencik ML,
Aretini A, Ferretti R, Maffei A, Altruda F, Silengo L, Tarone G, Loftus JC, Ross RS. Disruption of integrin function in the murine
Lembo G. Cardiac overexpression of melusin protects from dilated myocardium leads to perinatal lethality, fibrosis, and abnormal cardiac
cardiomyopathy due to long-standing pressure overload. Circ Res 96: performance. Am J Pathol 158: 1079 –1090, 2001.
1087–1094, 2005. 45. Kim E, Goraksha-Hicks P, Li L, Neufeld TP, Guan KL. Regulation of
22. Deldicque L, Atherton P, Patel R, Theisen D, Nielens H, Rennie MJ, TORC1 by Rag GTPases in nutrient response. Nat Cell Biol 10: 935–945,
Francaux M. Decrease in Akt/PKB signalling in human skeletal muscle 2008.
by resistance exercise. Eur J Appl Physiol 104: 57–65, 2008. 46. Kim E, Guan KL. RAG GTPases in nutrient-mediated TOR signaling
23. DeVol DL, Rotwein P, Sadow JL, Novakofski J, Bechtel PJ. Activation pathway. Cell Cycle 8: 1014 –1018, 2009.
of insulin-like growth factor gene expression during work-induced skeletal 47. Knoll R, Hoshijima M, Hoffman HM, Person V, Lorenzen-Schmidt I,
muscle growth. Am J Physiol Endocrinol Metab 259: E89 –E95, 1990. Bang ML, Hayashi T, Shiga N, Yasukawa H, Schaper W, McKenna
24. Drummond MJ, McCarthy JJ, Fry CS, Esser KA, Rasmussen BB. W, Yokoyama M, Schork NJ, Omens JH, McCulloch AD, Kimura A,
Aging differentially affects human skeletal muscle microRNA expression Gregorio CC, Poller W, Schaper J, Schultheiss HP, Chien KR. The
at rest and after an anabolic stimulus of resistance exercise and essential cardiac mechanical stretch sensor machinery involves a Z disc complex
amino acids. Am J Physiol Endocrinol Metab 295: E1333–E1340, 2008. that is defective in a subset of human dilated cardiomyopathy. Cell 111:
25. Esmarck B, Andersen JL, Olsen S, Richter EA, Mizuno M, Kjaer M. 943–955, 2002.
Timing of postexercise protein intake is important for muscle hypertrophy 48. Koopman R, Wagenmakers AJ, Manders RJ, Zorenc AH, Senden JM,
with resistance training in elderly humans. J Physiol 535: 301–311, 2001. Gorselink M, Keizer HA, van Loon LJ. Combined ingestion of protein
26. Fedele MJ, Hernandez JM, Lang CH, Vary TC, Kimball SR, Jeffer- and free leucine with carbohydrate increases postexercise muscle protein
son LS, Farrell PA. Severe diabetes prohibits elevations in muscle protein synthesis in vivo in male subjects. Am J Physiol Endocrinol Metab 288:
synthesis after acute resistance exercise in rats. J Appl Physiol 88: E645–E653, 2005.
102–108, 2000. 49. Kumar V, Selby A, Rankin D, Patel R, Atherton P, Hildebrandt W,
27. Fernandez AM, Dupont J, Farrar RP, Lee S, Stannard B, Le Roith D. Williams J, Smith K, Seynnes O, Hiscock N, Rennie MJ. Age-related
Muscle-specific inactivation of the IGF-I receptor induces compensatory differences in the dose-response relationship of muscle protein synthesis to
hyperplasia in skeletal muscle. J Clin Invest 109: 347–355, 2002. resistance exercise in young and old men. J Physiol 587: 211–217, 2009.
28. Fluck M, Carson JA, Gordon SE, Ziemiecki A, Booth FW. Focal 50. Laurent GJ, Millward DJ. Protein turnover during skeletal muscle
adhesion proteins FAK and paxillin increase in hypertrophied skeletal
hypertrophy. Fed Proc 39: 42–47, 1980.
muscle. Am J Physiol Cell Physiol 277: C152–C162, 1999.
51. MacKenzie MG, Hamilton DL, Murray JT, Taylor PM, Baar K.
29. Flueck M, Goldspink G. Point:Counterpoint: IGF is/is not the major
mVps34 is activated following high-resistance contractions. J Physiol 587:
physiological regulator of muscle mass. Counterpoint: IGF is not the
253–260, 2009.
major physiological regulator of muscle mass. J Appl Physiol 108: 1821–
52. McCarthy JJ, Esser KA. MicroRNA-1 and microRNA-133a expression
1823; discussion 1823–1824; author reply 1833, 2010.
are decreased during skeletal muscle hypertrophy. J Appl Physiol 102:
30. Goldberg AL. Role of insulin in work-induced growth of skeletal muscle.
306 –313, 2007.
Endocrinology 83: 1071–1073, 1968.
53. Melov S, Tarnopolsky MA, Beckman K, Felkey K, Hubbard A.
31. Goldberg AL. Work-induced growth of skeletal muscle in normal and
hypophysectomized rats. Am J Physiol 213: 1193–1198, 1967. Resistance exercise reverses aging in human skeletal muscle. PLoS ONE
32. Goldberg AL, Goodman HM. Amino acid transport during work-in- 2: e465, 2007.
duced growth of skeletal muscle. Am J Physiol 216: 1111–1115, 1969. 54. O’Neil TK, Duffy LR, Frey JW, Hornberger TA. The role of phospho-
33. Goodman CA, Miu MH, Frey JW, Mabrey DM, Lincoln HC, Ge Y, inositide 3-kinase and phosphatidic acid in the regulation of mammalian
Chen J, Hornberger TA. A phosphatidylinositol 3-kinase/protein kinase target of rapamycin following eccentric contractions. J Physiol 587:
B-independent activation of mammalian target of rapamycin signaling is 3691–3701, 2009.
sufficient to induce skeletal muscle hypertrophy. Mol Biol Cell 21: 55. Olivier AR, Ballou LM, Thomas G. Differential regulation of S6
3258 –3268, 2010. phosphorylation by insulin and epidermal growth factor in Swiss mouse
34. Gordon SE, Fluck M, Booth FW. Selected Contribution: Skeletal muscle 3T3 cells: insulin activation of type 1 phosphatase. Proc Natl Acad Sci
focal adhesion kinase, paxillin, and serum response factor are loading USA 85: 4720 –4724, 1988.
dependent. J Appl Physiol 90: 1174 –1183; discussion 1165, 2001. 56. Phillips SM, Tipton KD, Aarsland A, Wolf SE, Wolfe RR. Mixed
35. Guharay F, Sachs F. Stretch-activated single ion channel currents in muscle protein synthesis and breakdown after resistance exercise in
tissue-cultured embryonic chick skeletal muscle. J Physiol 352: 685–701, humans. Am J Physiol Endocrinol Metab 273: E99 –E107, 1997.
1984. 57. Phillips SM, Tipton KD, Ferrando AA, Wolfe RR. Resistance training
36. Hamilton DL. PI3K/mTORC1 Signalling During Skeletal Muscle Differ- reduces the acute exercise-induced increase in muscle protein turnover.
entiation and Growth (PhD Dissertation). Dundee, Ireland: Univ. of Am J Physiol Endocrinol Metab 276: E118 –E124, 1999.
Dundee, 2009. 58. Reinhard C, Thomas G, Kozma SC. A single gene encodes two isoforms
37. Hamilton DL, Philp A, MacKenzie MG, Baar K. A limited role for of the p70 S6 kinase: activation upon mitogenic stimulation. Proc Natl
PI(3,4,5)P3 regulation in controlling skeletal muscle mass in response to Acad Sci USA 89: 4052–4056, 1992.
resistance exercise. PLoS ONE 5: e11624, 2010. 59. Risson V, Mazelin L, Roceri M, Sanchez H, Moncollin V, Corneloup
38. Heron-Milhavet L, Mamaeva D, LeRoith D, Lamb NJ, Fernandez A. C, Richard-Bulteau H, Vignaud A, Baas D, Defour A, Freyssenet D,
Impaired muscle regeneration and myoblast differentiation in mice with a Tanti JF, Le-Marchand-Brustel Y, Ferrier B, Conjard-Duplany A,
muscle-specific KO of IGF-IR. J Cell Physiol 225: 1–6, 2010. Romanino K, Bauche S, Hantai D, Mueller M, Kozma SC, Thomas G,
39. Hornberger TA, Chu WK, Mak YW, Hsiung JW, Huang SA, Chien S. Ruegg MA, Ferry A, Pende M, Bigard X, Koulmann N, Schaeffer L,
The role of phospholipase D and phosphatidic acid in the mechanical Gangloff YG. Muscle inactivation of mTOR causes metabolic and dys-
activation of mTOR signaling in skeletal muscle. Proc Natl Acad Sci USA trophin defects leading to severe myopathy. J Cell Biol 187: 859 –874,
103: 4741–4746, 2006. 2009.
40. Hornberger TA, Stuppard R, Conley KE, Fedele MJ, Fiorotto ML, 60. Rommel C, Bodine SC, Clarke BA, Rossman R, Nunez L, Stitt TN,
Chin ER, Esser KA. Mechanical stimuli regulate rapamycin-sensitive Yancopoulos GD, Glass DJ. Mediation of IGF-1-induced skeletal myo-
signalling by a phosphoinositide 3-kinase-, protein kinase B- and growth tube hypertrophy by PI(3)K/Akt/mTOR and PI(3)K/Akt/GSK3 pathways.
factor-independent mechanism. Biochem J 380: 795–804, 2004. Nat Cell Biol 3: 1009 –1013, 2001.

J Appl Physiol • VOL 110 • FEBRUARY 2011 • www.jap.org


Downloaded from www.physiology.org/journal/jappl by ${individualUser.givenNames} ${individualUser.surname} (177.184.218.005) on December 11, 2017.
Copyright © 2011 American Physiological Society. All rights reserved.
Review
568
61. Sancak Y, Bar-Peled L, Zoncu R, Markhard AL, Nada S, Sabatini 75. Tipton KD, Ferrando AA, Phillips SM, Doyle D Jr, Wolfe RR.
DM. Ragulator-Rag complex targets mTORC1 to the lysosomal surface Postexercise net protein synthesis in human muscle from orally adminis-
and is necessary for its activation by amino acids. Cell 141: 290 –303. tered amino acids. Am J Physiol Endocrinol Metab 276: E628 –E634,
62. Sancak Y, Peterson TR, Shaul YD, Lindquist RA, Thoreen CC, 1999.
Bar-Peled L, Sabatini DM. The Rag GTPases bind raptor and mediate 76. Tipton KD, Gurkin BE, Matin S, Wolfe RR. Nonessential amino acids
amino acid signaling to mTORC1. Science 320: 1496 –1501, 2008. are not necessary to stimulate net muscle protein synthesis in healthy
63. Sandri M. Autophagy in skeletal muscle. FEBS Lett 584: 1411–1416. volunteers. J Nutr Biochem 10: 89 –95, 1999.
64. Sandri M. Signaling in muscle atrophy and hypertrophy. Physiology 23: 77. Vandenburgh HH, Kaufman S. Stretch-induced growth of skeletal
160 –170, 2008. myotubes correlates with activation of the sodium pump. J Cell Physiol
65. Shai SY, Harpf AE, Ross RS. Integrins and the myocardium. Genet Eng 109: 205–214, 1981.
NY 24: 87–105, 2002. 78. Welle S, Burgess K, Mehta S. Stimulation of skeletal muscle myofibrillar
66. Shima H, Pende M, Chen Y, Fumagalli S, Thomas G, Kozma SC. protein synthesis, p70 S6 kinase phosphorylation, and ribosomal protein
Disruption of the p70(s6k)/p85(s6k) gene reveals a small mouse pheno- S6 phosphorylation by inhibition of myostatin in mature mice. Am J
type and a new functional S6 kinase. EMBO J 17: 6649 –6659, 1998. Physiol Endocrinol Metab 296: E567–E572, 2009.
67. Spangenburg EE, Le Roith D, Ward CW, Bodine SC. A functional 79. Welle S, Burgess K, Thornton CA, Tawil R. Relation between extent of
insulin-like growth factor receptor is not necessary for load-induced myostatin depletion and muscle growth in mature mice. Am J Physiol
skeletal muscle hypertrophy. J Physiol 586: 283–291, 2008. Endocrinol Metab 297: E935–E940, 2009.
68. Spangenburg EE, McBride TA. Inhibition of stretch-activated channels
80. West DW, Burd NA, Tang JE, Moore DR, Staples AW, Holwerda
during eccentric muscle contraction attenuates p70S6K activation. J Appl
AM, Baker SK, Phillips SM. Elevations in ostensibly anabolic hor-
Physiol 100: 129 –135, 2006.
mones with resistance exercise enhance neither training-induced mus-
69. Stewart CE, Pell JM. Point:Counterpoint: IGF is/is not the major phys-
cle hypertrophy nor strength of the elbow flexors. J Appl Physiol 108:
iological regulator of muscle mass. Point: IGF is the major physiological
regulator of muscle mass. J Appl Physiol 108: 1820 –1821; discussion 60 –67, 2010.
1823–1824; author reply 1832, 2010. 81. West DW, Kujbida GW, Moore DR, Atherton P, Burd NA, Padzik JP,
70. Sun Y, Fang Y, Yoon MS, Zhang C, Roccio M, Zwartkruis FJ, De Lisio M, Tang JE, Parise G, Rennie MJ, Baker SK, Phillips SM.
Armstrong M, Brown HA, Chen J. Phospholipase D1 is an effector of Resistance exercise-induced increases in putative anabolic hormones do
Rheb in the mTOR pathway. Proc Natl Acad Sci USA 105: 8286 –8291, not enhance muscle protein synthesis or intracellular signalling in young
2008. men. J Physiol 587: 5239 –5247, 2009.
71. Sun Y, Ge Y, Drnevich J, Zhao Y, Band M, Chen J. Mammalian target 82. Witkowski S, Lovering RM, Spangenburg EE. High-frequency electri-
of rapamycin regulates miRNA-1 and follistatin in skeletal myogenesis. J cally stimulated skeletal muscle contractions increase p70s6k phosphory-
Cell Biol 189: 1157–1169. lation independent of known IGF-I sensitive signaling pathways. FEBS
72. Svanberg E, Ohlsson C, Kimball SR, Lundholm K. rhIGF-I/IGFBP-3 Lett 584: 2891–2895.
complex, but not free rhIGF-I, supports muscle protein biosynthesis in rats 83. Wong TS, Booth FW. Protein metabolism in rat gastrocnemius muscle
during semistarvation. Eur J Clin Invest 30: 438 –446, 2000. after stimulated chronic concentric exercise. J Appl Physiol 69: 1709 –
73. Terzis G, Georgiadis G, Stratakos G, Vogiatzis I, Kavouras S, Manta 1717, 1990.
P, Mascher H, Blomstrand E. Resistance exercise-induced increase in 84. Wong TS, Booth FW. Protein metabolism in rat tibialis anterior muscle
muscle mass correlates with p70S6 kinase phosphorylation in human after stimulated chronic eccentric exercise. J Appl Physiol 69: 1718 –1724,
subjects. Eur J Appl Physiol 102: 145–152, 2008. 1990.
74. Thomas G. The mitogen-activated p70s6k. Biochem Soc Trans 20: 678 – 85. Wong TS, Booth FW. Skeletal muscle enlargement with weight-lifting
681, 1992. exercise by rats. J Appl Physiol 65: 950 –954, 1988.

J Appl Physiol • VOL 110 • FEBRUARY 2011 • www.jap.org


Downloaded from www.physiology.org/journal/jappl by ${individualUser.givenNames} ${individualUser.surname} (177.184.218.005) on December 11, 2017.
Copyright © 2011 American Physiological Society. All rights reserved.

You might also like