You are on page 1of 11

J Appl Physiol 129: 272–282, 2020.

First published July 9, 2020; doi:10.1152/japplphysiol.00381.2020.

REVIEW Edward F. Adolph Distinguished Lecture

Edward F. Adolph Distinguished Lecture. Skeletal muscle atrophy: Multiple


pathways leading to a common outcome
Sue C. Bodine
Department of Internal Medicine/Endocrinology and Metabolism, University of Iowa Carver College of Medicine, Iowa City,
Iowa
Submitted 15 May 2020; accepted in final form 3 July 2020

Bodine SC. Edward F. Adolph Distinguished Lecture. Skeletal muscle atrophy:


multiple pathways leading to a common outcome. J Appl Physiol 129: 272–282,
2020. First published July 9, 2020; doi:10.1152/japplphysiol.00381.2020.—Skele-
tal muscle atrophy continues to be a serious consequence of many diseases and
conditions for which there is no treatment. Our understanding of the mechanisms
regulating skeletal muscle mass has improved considerably over the past two
decades. For many years it was known that skeletal muscle atrophy resulted from
an imbalance between protein synthesis and protein breakdown, with the net
balance shifting toward protein breakdown. However, the molecular and cellular
mechanisms underlying the increased breakdown of myofibrils was unknown. Over
the past two decades, numerous reports have identified novel genes and signaling
pathways that are upregulated and activated in response to stimuli such as disuse,
inflammation, metabolic stress, starvation and others that induce muscle atrophy.
This review summarizes the discovery efforts performed in the identification of
several pathways involved in the regulation of skeletal muscle mass: the mamma-
lian target of rapamycin (mTORC1) and the ubiquitin proteasome pathway and the
E3 ligases, MuRF1 and MAFbx. While muscle atrophy is a common outcome of
many diseases, it is doubtful that a single gene or pathway initiates or mediates the
breakdown of myofibrils. Interestingly, however, is the observation that upregula-
tion of the E3 ligases, MuRF1 and MAFbx, is a common feature of many divergent
atrophy conditions. The challenge for the field of muscle biology is to understand
how all of the various molecules, transcription factors, and signaling pathways
interact to produce muscle atrophy and to identify the critical factors for interven-
tion.
MAFbx; mTORC1; MuRF1; protein synthesis; ubiquitin proteasome pathway

INTRODUCTION signals from multiple systems (neural, endocrine, cardiovascu-


lar) and organs (liver, adipose, gut) within the body compli-
Skeletal muscle is a complex and dynamic tissue that com-
cates the efforts to identify the key regulators of skeletal
prises ~40% of body weight and performs critical functions
muscle mass.
related to movement (power output and sensory feedback),
Muscle size is a very plastic characteristic of limb muscles,
metabolism (substrate utilization, storage, and supply) and
changing over the life span with different signals playing
thermogenesis. Skeletal muscle mass has been shown to be
critical regulatory roles at each life stage (Fig. 1). Alterations
predictive of longevity in older adults and is a critical variable
in muscle mass can occur as a consequence of changes in both
in predicting mortality as a consequence of diseases such as
the number and/or size of individual muscle fibers depending
cancer, type II diabetes, and cardiovascular disease (75, 80).
on the life stage. The number of fibers in a muscle is estab-
The regulation of skeletal muscle mass is multifactorial inte-
lished during embryonic and fetal development and is depen-
grating signals from hormones, growth factors, cytokines,
dent on the proliferation, differentiation and fusion of myo-
nutrients, load and activity to regulate multiple intersecting
blasts to form myofibers (37, 51). The number of muscle fibers
pathways that control the balance between protein synthesis
in a muscle remains constant in healthy muscles throughout
and protein degradation (63). Furthermore, skeletal muscle is a
most of the life span, with decreases occurring during ad-
multicellular tissue that relies on an intact vascular system and
vanced aging (51). In contrast, changes in fiber size can occur
motor/sensory innervation to maintain its size and function.
throughout the life span. Increases in both the length and
The fact that skeletal muscle interacts with and integrates
cross-sectional area of individual fibers occur before and dur-
ing puberty and are dependent on satellite cell proliferation and
Correspondence: S. C. Bodine (Sue-Bodine@uiowa.edu). differentiation (67, 82). The predominant change that occurs
272 8750-7587/20 Copyright © 2020 the American Physiological Society http://www.jap.org
Downloaded from journals.physiology.org/journal/jappl (181.163.161.027) on September 4, 2021.
REGULATION OF SKELETAL MUSCLE ATROPHY 273

Fig. 1. Regulation of skeletal muscle mass


throughout the life span. This diagram illus-
trates some of the critical factors that regu-
late skeletal muscle size at different life
stages. At each life stage skeletal muscle
undergoes specific changes that are regu-
lated by different critical factors. During the
embryonic stage growth factors, hormones
and myocytes/satellite cells play a critical
role in the formation and innervation of
muscle fibers. During the postnatal stage,
external loading and increased neural activa-
tion become important factors along with
growth factors, hormones, and satellite cells
in regulating increases in the length and
cross-sectional area of muscle fibers. As
adults, maintaining muscle mass and adapt-
ing to changes in external loading and neural
activation become the dominant focus. With
advancing age, decreases in external loading
and activity, as well as, inflammation, in-
creasing levels of cytokines, oxidative stress,
and metabolic stress can lead to a decrease in
muscle mass and strength.

postpuberty is to fiber cross-sectional area, resulting in both skeletal muscle to control movement. Over the course of my
increases (i.e., hypertrophy) and decreases (i.e., atrophy) in career my research became more focused on undercovering the
size. The role of satellite cells in adaptive hypertrophy of adult mechanisms regulating skeletal muscle size, especially under
muscle is still debated, but recent evidence suggests that conditions leading to the loss of muscle mass and function. In
adaptive muscle hypertrophy can occur without satellite cells this review, I will highlight research conducted since the late
and the addition of myonuclei to individual muscle fibers (9, 1990s that has contributed to a greater understanding of the
24, 54). mechanisms underlying skeletal muscle atrophy.
Our understanding of the factors and pathways that regulate In the 1990s our understanding of the cellular and molecular
both hypertrophy and atrophy of skeletal muscle has increased mechanisms regulating skeletal muscle was relatively limited.
greatly over the past two decades. This review is a summary of It was known that loss of muscle mass occurred under a variety
the Adolph Distinguished Lecture given by me at the 2019 of diseases and conditions such as immobilization, spinal cord
Experimental Biology meeting. The work presented in this injury, aging and others as shown in Fig. 2. In animal models,
review reflects a journey that started for me as a graduate muscle atrophy could be induced by decreases in neural activ-
student interested in understanding skeletal muscle plasticity ity, decreases in external loading, increases in glucocorticoids,
and in particular how force is generated and regulated in increases in inflammatory cytokines and nutrient deficiency

Fig. 2. Skeletal muscle atrophy is prevalent


in many diseases. Skeletal muscle atrophy is
a serious consequence of the diseases and
conditions listed in this diagram.

J Appl Physiol • doi:10.1152/japplphysiol.00381.2020 • www.jap.org


Downloaded from journals.physiology.org/journal/jappl (181.163.161.027) on September 4, 2021.
274 REGULATION OF SKELETAL MUSCLE ATROPHY

(30, 52, 66). The induction of muscle atrophy was related to an provided regarding the discovery of two skeletal muscle-
imbalance between protein synthesis and degradation with the specific E3 ubiquitin ligases, MuRF1 and MAFbx/Atrogin1,
net balance being shifted toward protein breakdown. There followed by highlights of published findings related to the
were data to suggest that both decreases in protein synthesis transcriptional regulation of MuRF1 and MAFbx and their
and increases in protein degradation contributed to muscle loss, expression patterns during different forms of atrophy, as well
and that the relative contribution of each process to muscle loss as, the use of mice with global deletions of either MuRF1 or
depended on the conditions (3, 28, 29, 43, 56, 62); however, MAFbx to identify the potential mechanisms of action of these
unknown at the time was the identity of the signaling pathways E3 ligase in the regulation of skeletal muscle mass. The final
responsible for the changes in protein synthesis and degrada- sections discuss the link between MuRF1 and MAFbx expres-
tion. sion and proteasome activity, and the continuing search for
In the late 1990s, while at Regeneron Pharmaceuticals, we MuRF1 and MAFbx substrates. The review concludes by
initiated a discovery program aimed at determining the signal- highlighting some of the important questions that remain to be
ing pathways regulating the loss of skeletal muscle mass. To answered.
approach this complex problem, we developed a multi-disci-
plinary muscle research program diagrammed in Fig. 3. At the THE IDENTIFICATION OF MTORC1 AS A MAJOR
center of the program was the establishment of rodent models REGULATOR OF SKELETAL MUSCLE SIZE IN MAMMALS
of muscle atrophy and hypertrophy, from which skeletal mus-
cles were obtained at multiple time points postexperimental Skeletal muscle is highly responsive to changes in external
manipulation and then used for the identification of signaling loading, increasing in size in response to enhanced loading and
pathways and differentially regulated genes and proteins. A decreasing in size in response to reduced loading (66). It had
variety of pharmacological agents were also used to induce been shown in both human and rodent muscle, that the rate of
atrophy (e.g., dexamethasone, cytokines) and hypertrophy protein synthesis is modified in response to changes in external
(e.g., clenbuterol) or block specific enzymes and signaling loading. Following an acute bout of resistance exercise in
pathways (e.g., rapamycin). Finally, emerging technologies, humans (87) or simulated resistance exercise in rats (84),
such as mouse genetic engineering and in vivo electroporation, protein synthesis increases; whereas, the early response to
were utilized to overexpress or delete specific genes in skeletal conditions that decrease loading, such as immobilization or
muscle. These approaches allowed for the discovery of novel bedrest, is a decrease in protein synthesis (25, 28). While
pathways involved in the regulation of skeletal muscle mass. robust changes in protein synthesis in response to alterations in
In the following sections, a summary of work that led to the loading had been demonstrated, the cellular mechanisms re-
discovery of novel signaling pathways involved in the regula- sponsible for the changes were unknown. Some possible can-
tion of skeletal muscle mass is presented. It begins with a didates at the time included Insulin-like Growth Factor 1
description of the experiments that identified mTORC1 activa- (IGF-1), calcineurin, and the PI(3)K/Akt signaling pathway.
tion as a critical regulator of skeletal muscle hypertrophy in The case for IGF-1 as a major mediator of growth and protein
adult mammals; and continues with a description of the exper- synthesis in skeletal muscle was supported by reports that
imental approaches taken to identify novel signaling pathways IGF-1 induces an increase in myotube size in vitro (65, 77),
involved in the loss of skeletal muscle mass. Details are IGF-1 is elevated in vivo following acute high-load contrac-

Fig. 3. Building a multidisciplinary muscle re-


search program. A multidisciplinary research
program was developed to discover novel mech-
anisms regulating skeletal muscle size. At the
center of the program was the establishment of
animal models of skeletal muscle atrophy (de-
nervation, nerve crush-reinnervation, joint im-
mobilization, hindlimb suspension, glucocorti-
coid excess) and hypertrophy (functional over-
load, reloading following disuse). Muscle
tissues taken at different time points were used
in the identification of novel gene and protein
targets using differential gene expression and
proteomics approaches. Muscle samples were
also used to identify signaling pathways that
were altered in response to stimuli that induced
atrophy and hypertrophy. Furthermore, pharma-
cological agents, such as clenbuterol, IGF1,
rapamycin, glucocorticoids, were utilized both
in vivo and in vitro to manipulate selective
pathways to induce changes in muscle size.
Finally, technologies such as in vivo electropo-
ration and mouse genetic engineering were uti-
lized to modify gene expression in skeletal
muscle.

J Appl Physiol • doi:10.1152/japplphysiol.00381.2020 • www.jap.org


Downloaded from journals.physiology.org/journal/jappl (181.163.161.027) on September 4, 2021.
REGULATION OF SKELETAL MUSCLE ATROPHY 275
tions (1), and transgenic overexpression of IGF-1 in mice increased loading associated with resistance exercise in healthy
results in enhanced muscle size (2, 18). Another pathway that muscle. The differences between the two growth models, i.e.,
had been linked to growth in model organisms and mice was functional overload and unloading/reloading, have not been
the PI(3)K/Akt/mTOR pathway. Genetic manipulation in Dro- explored in detail. The increase in Akt/mTORC1 activation in
sophila of PI(3)K, PKB/Akt and S6K1 (p70s6k) revealed that the functional overload model was presumed to be in response
deletion of components of this pathway resulted in smaller to an increase in IGF-1 signaling since IGF-1 had been shown
cells, but not fewer cells, suggesting that the PI(3)K/Akt/ to activate the PI3k/Akt/mTORC1 pathway in myotubes and
mTOR pathway played a major role in the regulation of cell induce hypertrophy (65), and transgenic overexpression of
size (57, 81). Further support for this pathway came from the IGF1 in embryonic mice resulted in larger muscles (18).
deletion of S6K1 in mice which led to lower body weights and However, a subsequent study by Spangenburg et al. (74)
organ growth relative to wild-type (WT) littermates (72). revealed that activation of the IGF1 receptor was not required
Finally, data were emerging that the PI(3)K/Akt/mTOR path- for the phosphorylation of Akt and S6K1/p70S6K and the
way may play a role in skeletal muscle growth in response to induction of muscle hypertrophy in response to functional
resistance exercise. Baar and Esser demonstrated that follow- overload.
ing high resistance exercise training in rats, phosphorylation of In summary, many studies have now shown that activation
S6K1 (p70s6k) increased in those muscles experiencing an of mTORC1 and increases in protein synthesis are critical for
increase in loading resulting in hypertrophy (4). Given that achieving muscle hypertrophy in response to increased loading
S6K1 is downstream of mTORC1, these data suggested that in adult mammals. This increase in protein synthesis and
activation of mTORC1 and its downstream targets were im- stimulation of muscle fiber growth does not appear to be
portant for muscle growth. dependent on an increase in satellite cell proliferation and the
Given what was known at the time, we asked the following addition of myonuclei to the muscle fiber (40, 54). Further-
questions: Is the PI(3)k/Akt/mTOR pathway regulated in vivo more, suppression of mTORC1 and decreased protein synthe-
during hypertrophy or atrophy, and what happens to growth or sis can contribute to the loss of muscle mass, especially under
atrophy when this pathway is inhibited? To address the first conditions of disuse. These results might suggest that activat-
question, we used synergist ablation, a model of functional ing the mTORC1 pathway would be a good strategy for
overload, to determine whether components of the PI(3)k/Akt/ treating muscle atrophy. In fact, it has been shown that over-
mTORC1 pathways were activated in a muscle undergoing expression of Akt in muscle can lead to hypertrophy and
hypertrophy. We found that following synergist ablation, all prevent muscle atrophy (12, 49). However, chronic activation
components of the PI3k/Akt/mTORC1 were activated in the of mTORC1 has been shown to be deleterious to muscle,
plantaris muscle and that activation of this pathway was an leading to muscle atrophy (35). Closer inspection of how
early response (12). Phosphorylation and total amount of mTORC1 is activated in muscle shows that in resting adult
Akt/PKB increased early leading to activation of mTORC1 as muscle mTORC1 is at a relatively low activation state. Acti-
measured by (1) an increase in phosphorylation of mTORC1 at vation of mTORC1 occurs periodically as the result of in-
Ser2448 (64), (2) an increase in the phosphorylation and creased loading or amino acid ingestion (19, 33). In healthy
activity of S6K1/p70s6k (12), and (3) a decrease in the amount adult muscle, mTORC1 is rarely in a chronically activated
of 4EBP1/PHAS-I bound to eIF4E coupled with an increase in state, and short-term rapamycin treatment in adult animals does
the amount of eIF4E bound to eIF4G (64). These data were the not affect muscle mass (12). Furthermore, a recent study
first to show that activation of the Akt/mTORC1 pathway was showed that deletion of raptor and suppression of mTORC1
associated with skeletal muscle growth in vivo. Subsequent activity in resting adult mice for 5 mo had no effect on muscle
studies have shown that this pathway is activated in human mass (36). An exception is muscle in aged animals where
muscle in response to resistance exercise and associated with mTORC1 activation is chronically elevated under resting con-
an increase in protein synthesis (21, 22, 34, 58). The question ditions and shows reduced activation in response to anabolic
remained as to whether activation of this pathway was neces- stimuli. Chronic activation of mTORC1 is thought to contrib-
sary for skeletal muscle growth. To address this question, adult ute to the loss of mass and function with age, and a recent study
rats were given rapamycin to inhibit mTORC1 and its down- suggests that partial suppression of mTORC1 in older animals
stream targets, without affecting activation of Akt or inhibition could be beneficial (41). It should be noted that mTORC1 is
of GSK3. Rapamycin treatment for a duration of up to 14 days also activated in muscle upon denervation, a condition that
prevented 95% of hypertrophy of both slow and fast fibers in causes muscle atrophy (29). Interestingly, inhibition of
the rat plantaris muscle following functional overload (12), mTORC1 with rapamycin during denervation results in an
revealing that activation of mTORC1 was necessary to achieve attenuation of muscle atrophy (44). The mechanisms underly-
adaptive hypertrophy of adult skeletal muscle. Activation of ing the beneficial effects of rapamycin during denervation and
the Akt/mTORC1 pathways was also shown to decrease in possibly aging remain unclear and require further investiga-
response to hindlimb unloading, which induced atrophy, and to tion.
increase in response to reloading, which stimulated muscle
regrowth (12). Interestingly, rapamycin treatment given upon IDENTIFICATION OF MUSCLE ATROPHY PATHWAY
return of weight-bearing locomotion following hindlimb un-
loading significantly inhibited growth of the soleus, plantaris The loss of muscle mass occurs as the result of many
and medial gastrocnemius muscles; however, the inhibition conditions and diseases, as shown in Fig. 2. To identify novel
was not as great as that seen in the functional overload model. pathways responsible for muscle atrophy, we initially focused
These data suggest that muscle regrowth following a period of our discovery efforts on models of disuse atrophy. Disuse
atrophy is not identical to muscle growth in response to atrophy occurs as the result of decreases in external loading, as

J Appl Physiol • doi:10.1152/japplphysiol.00381.2020 • www.jap.org


Downloaded from journals.physiology.org/journal/jappl (181.163.161.027) on September 4, 2021.
276 REGULATION OF SKELETAL MUSCLE ATROPHY

well as, decreases in neural activation of muscle, and ranges in under a variety of atrophy and hypertrophy conditions, their
the degree of muscle loss; being less severe in bedrest (mod- transcriptional regulation, and the impact of their deletion on
eled as hindlimb suspension in animals) and most severe the loss of muscle mass in different atrophy conditions.
following denervation (Fig. 4). Examination of the extent of
atrophy (% change in muscle mass) of the rat medial gastroc- MuRF1 and MAFbx Are Excellent Markers of Muscle
nemius over 14 days in three models of disuse (hindlimb Atrophy
unloading, ankle joint immobilization, denervation) shows that
the rate of loss over the first 3 days was similar across the three MuRF1 and MAFbx demonstrate many characteristics that
models, whereas after 7 days the rate of loss diverges across make them excellent markers of muscle atrophy. First, both
the models (Fig. 4). In an effort to discover potential early genes are selectively expressed in muscle tissue (skeletal,
triggers of muscle atrophy, a differential expression analysis cardiac and smooth muscle), and in resting skeletal muscle
(GeneTag method) was performed on control and 3-day im- these genes are expressed at relatively low levels (11). Second,
mobilized rat medial gastrocnemius muscle (11) (Fig. 4). In the the expression of both genes rapidly increases in response to a
primary screen we identified genes that were differentially variety of stressors including unloading, decreased neural ac-
regulated by threefold in the immobilized muscle. A secondary tivity, elevated glucocorticoids, elevated cytokines, increased
screen was then performed to identify the subset of genes that oxidative stress, and malnutrition (10, 11, 32, 61). The expres-
were also differentially regulated following denervation and sion patterns of both genes vary depending on the atrophy
hindlimb suspension, in addition to immobilization. The sec- stimulus. For example, in response to unloading and denerva-
ondary screen identified two genes that were upregulated in all
tion, expression of both MuRF1 and MAFbx increases rapidly
disuse atrophy models tested, as well as, dexamethasone and
within 48 h reaching a peak around 7–10 days and then
interleukin-1 induced atrophy. Both genes were shown to be
E3 ubiquitin ligases: one gene was a RING finger protein gradually declining to baseline by 14 days. Recently, it was
previously identified in the heart as MURF1 (Trim63) and the confirmed using mass spectrometry that MuRF1 protein ex-
other gene was a novel protein that contained an F-box domain pression in mouse muscle changed over the same time course
and was shown to be a member of the Skp-Cullin1-F-box as the mRNA following denervation (50). Validation of protein
(SCF) protein family of E3 ubiquitin ligases. We named this expression, especially of MuRF1, has been difficult because of
novel protein MAFbx for Muscle Atrophy F-box (FBX032). the lack of specificity of commercially available antibodies.
This same protein was also identified by Goldberg and col- We have tested the majority of commercially available MuRF1
leagues in the mouse gastrocnemius muscle following starva- antibodies, and found positive staining with the MuRF1 anti-
tion using an Affymetrix microarray and called Atrogin-1 (32). body when used on lysates from muscles taken from MuRF1
Since their initial identification in 2001, my laboratory has KO mice. Moreover, in these KO tissues we often observe a
been studying the role of MuRF1 and MAFbx in the regulation positive band at the same molecular weight as predicted for
of skeletal muscle mass by examining their expression patterns MuRF1 (8). Our data suggest that the majority of commercial

Fig. 4. Discovery of MuRF1 and MAFbx in skeletal muscle. A: animal models of disuse atrophy range in severity from those that primarily produce decreased
external loading of the muscles, such as hindlimb unloading to those that produce both a decrease in external loading and a complete absence of neural activity
such as denervation or spinal cord injury. Joint immobilization is a model that produces decreased external loading and a reduction in neural activity depending
on the degree to which the joint is immobilized. B: loss of mass of the rat medial gastrocnemius muscle (MG) was compared in three models of disuse atrophy
(hindlimb suspension, ankle joint immobilization, and denervation). To identify a potential common trigger of muscle atrophy, 3 days postimmobilization was
chosen as the time point and model to perform a differential gene expression analysis (GeneTag, Applied Bioscience). In the primary screen, all those genes that
were differentially regulated 3-fold were identified. A secondary screen was utilized to identify those genes that were similarly regulated in three disuse models
(immobilization, hindlimb unloading, and denervation). The secondary screen consisted of using Northern blots to analyze gene expression in the medial
gastrocnemius muscle over a time course of atrophy (0, 1, 3, 7 days) for each of the disuse models. From the secondary analysis, two genes (MuRF1 and MAFbx)
were identified that were similarly upregulated in all atrophy models examined.

J Appl Physiol • doi:10.1152/japplphysiol.00381.2020 • www.jap.org


Downloaded from journals.physiology.org/journal/jappl (181.163.161.027) on September 4, 2021.
REGULATION OF SKELETAL MUSCLE ATROPHY 277
antibodies for MuRF1 are nonspecific and should not be used least in response to FOXO1. In contrast, the MuRF1 promoter
to quantify MuRF1 protein expression in muscle tissue. was similarly activated by FOXO1 and FOXO3a. Interestingly,
The expression patterns of both MuRF1 and MAFbx in we found a synergistic activation of the MuRF1 promoter only
response to elevated glucocorticoids and cytokines differ com- by FOXO1 and the activated glucocorticoid receptor (78). The
pared with disuse; rising rapidly to a peak within days and then differential response of MuRF1 and MAFbx to the various
maintaining an elevated level for as long as the signal (gluco- FOXO transcription factors is important because many studies
corticoid or cytokine levels) is present (5). Interestingly, under treat all of the FOXO transcription factors as interchangeable.
increased loading conditions (such as reloading following un- FOXO3a and FOXO1 are the predominate FOXO transcription
loading, reinnervation following nerve injury, and functional expressed in skeletal muscle. Many published reports utilize
overload) the expression of MuRF1 and MAFbx is suppressed either FOXO1 or FOXO3a in their studies, and then assume
below baseline (6, 7). While MuRF1 and MAFbx expression that the results apply to all FOXO transcriptional factors. A
are suppressed at 7 days following functional overload, the study by Milan et al. illustrates the differential gene regulation
expression of both genes is transiently increased at 1 day of MuRF1 and MAFbx in vivo by FOXO1 and FOXO3a (55).
following the synergist ablation surgery (6). This same pattern In their study, Milan et al. measured expression of MuRF1 and
of elevated expression is seen immediately following an acute MAFbx following denervation in WT mice and mice with a
bout of intensive eccentric contractions in humans (53, 86). muscle-specific deletion of FOXO1 or FOXO1/3a/4. In re-
The mechanism underlying the suppression of these genes in sponse to denervation, the expression of both FOXO1 and
response to increased loading is unknown. One possible mech- FOXO3a increases, as does the expression of MuRF1 and
anism could be a change in the redox state of the muscle upon MAFbx. Deletion of only FOXO1a in muscle significantly
loading. In summary, MuRF1 and MAFbx are excellent mark- suppressed the activation of MuRF1, but not MAFbx. In
ers of muscle atrophy due to the fact that in all atrophy models contrast, deletion of all three FOXOs in muscle resulted in a
tested to date expression of MuRF1 and MAFbx has been significant suppression of both MuRF1 and MAFbx. Interest-
shown to increase at some time point during the course of the ingly, MuRF1 and MAFbx still increased in response to de-
disease. nervation even in the absence of the FOXO transcription
factors. This activation is likely not related to an activated
MuRF1 and MAFbx Are Regulated By Multiple glucocorticoid receptor since we showed that upregulation of
Transcription Factors both MuRF1 and MAFbx following denervation was similar in
WT and muscle-specific GR knockout mice (79). These data
Examination of the expression patterns of MuRF1 and do illustrate that transcriptional control of MuRF1 and MAFbx
MAFbx under different atrophy conditions has revealed two is complex, and the literature shows that multiple transcription
interesting observations: 1) both genes are upregulated together factors can activate both genes. In addition to the glucocor-
under most atrophy conditions and 2) both genes are strongly ticoid receptor and FOXO1/FOXO3a, the following tran-
induced by the synthetic glucocorticoid, dexamethasone. Based scription factors have been shown to activate MuRF1 and
on these observations, we were interested in understanding the MAFbx: NF-␬B (15, 39, 85), KLF15 (73), C/EBP ␤ (88, 89)
transcriptional regulation of these genes under different atro- and Smad3 (13).
phy conditions. We started our investigation into the transcrip-
tional regulation of MuRF1 and MAFbx by first examining the Deletion of MuRF1, but not MAFbx, Results in Sparing of
proximal promoter regions, 5,000 bp upstream of the transcrip- Muscle Mass
tional start site, of both genes for putative transcription factor
binding sites. Interestingly, we found that the proximal region The substrates targeted for ubiquitination by MuRF1 and
of the MuRF1 promoter, but not the MAFbx promoter, con- MAFbx remain poorly defined, and thus the mechanisms by
tained a perfect palindromic glucocorticoid response element which upregulation of MuRF1 and/or MAFbx contribute to
(GRE) (78). We also found that the proximal promoter region muscle atrophy remain poorly understood. The generation of
of each gene contained several consensus Class O forkhead mice with a global deletion of either MuRF1 or MAFbx has
(FOXO) binding sites (FBE), capable of binding FOXO1, assisted us in understanding the role of these E3 ligases in the
FOXO3a, and FOXO4; all of which are expressed in skeletal regulation of skeletal muscle mass (11). The phenotype of the
muscle. This finding was particularly interesting since several knockout mice has been previously described (10). In brief,
reports had shown that activated FOXO1 (76) could increase both knockout strains develop normally and show no pheno-
MuRF1 transcription in myotubes, while activated FOXO3a type until later in life (~16 –18 mo) when the MAFbx KO mice
could increase MAFbx expression in vitro and in vivo (68). die prematurely of congestive heart failure. The MuRF1 KO
Our studies revealed for the first time that dexamethasone mice have a normal life span and have preserved muscle mass,
could directly activate the MuRF1 promoter, but not the but not function, with age (38). Muscle atrophy has been
MAFbx promoter (78). Furthermore, we provided additional examined in both knockout (KO) strains and the results have
evidence for direct activation of the MuRF1 and MAFbx revealed that, in general, deletion of MuRF1 leads to better
promoters by the FOXO transcription factors. Our studies, functional muscle sparing than deletion of MAFbx. Deletion of
however, revealed that while the promoters of both MuRF1 MuRF1 leads to functional sparing of muscle mass following
and MAFbx could be activated by all of the FOXO transcrip- denervation (31), hindlimb unloading (48), exogenous gluco-
tions factors, MAFbx was much more responsive than MuRF1. corticoid treatment (5), and acute lung injury (26). In contrast,
Moreover, the promoters, were not equally activated by all of deletion of MAFbx only spares muscle mass following dener-
the FOXO transcription factors. For example, activation of the vation, however, the sparing is not functional. Histological
MAFbx promoter was greatest in response to FOXO3a and analysis of denervated muscle from MAFbx KO mice after 14

J Appl Physiol • doi:10.1152/japplphysiol.00381.2020 • www.jap.org


Downloaded from journals.physiology.org/journal/jappl (181.163.161.027) on September 4, 2021.
278 REGULATION OF SKELETAL MUSCLE ATROPHY

days reveals an increase in vacuole-containing fibers and an The use of MuRF1 and MAFbx expression as markers of
increase in fibrosis (31). Interestingly, muscle sparing in the increased proteasome activity, and by extension protein deg-
MuRF1 KO mice following denervation occurs even though radation, is widespread in the literature. As mentioned previ-
MAFbx expression is higher than measured in WT denervated ously, MuRF1 and MAFbx expression are excellent markers of
muscle and is sustained at a higher level for a longer period of muscle atrophy; however, an increase in their expression levels
time as compared with WT mice (31). In general, our muscle does not always coincide with an increase in proteasome
atrophy studies in the KO mice have revealed that suppression activity. In the case of denervation, the increase in proteasome
of MAFbx is not required for sparing of muscle mass, however, activity occurs as the expression levels of MuRF1 and MAFbx
suppression of MuRF1 is necessary to achieve muscle sparing. are decreasing back to baseline. In another disuse model,
The one atrophy model where muscle atrophy is not affected hindlimb suspension, expression of MuRF1 and MAFbx in-
by deletion of either MuRF1 or MAFbx is nutritional depriva- creases in the soleus, medial gastrocnemius and tibialis anterior
tion (5). muscles with unloading, peaking at around 7 days and return-
ing to baseline by 14 days (8). In comparison, proteasome
Increases in MuRF1 and MAFbx Expression are not Always activity increases only in the soleus and rises continuously over
Linked to Increases in Proteasome Activity 14 days of unloading. Another example of the disconnect
between MuRF1 and MAFbx expression and proteasome ac-
MuRF1 and MAFbx are E3 ubiquitin ligases that are ex- tivity is functional overload. In response to functional over-
pressed at low levels in resting muscle and increase rapidly in load, MuRF1 and MAFbx expression significantly increases at
response to stimuli that induce muscle atrophy. Given these day 1 following the synergist ablation surgery and decreases to
facts, it has been hypothesized that increases in MuRF1 and baseline by 3 days followed by suppression below baseline (6).
MAFbx expression lead to muscle atrophy by increasing pro- In contrast, proteasome activity increases immediately follow-
teasome activity and protein degradation. In our atrophy mod- ing surgery and continues to rise until around day 7 post
els, we have examined the relationship between the expression functional overload, returning to baseline levels by 14 days.
levels of MuRF1 and MAFbx and proteasome activity. Protea- These data highlight the fact that elevated proteasome activity
some activity is measured using in vitro assays in which the is not always a sign of atrophy, but is also required for
activity of the specific proteasome subunits (␤1, ␤2, and ␤5) is remodeling and growth. These data also strongly suggest that
determined using specific proteasome inhibitors and substrates MuRF1 and MAFbx expression should not be used as substi-
in the presence (26S proteasome) or absence (20S proteasome) tute markers for proteasome activity or protein degradation.
of ATP (31). Following denervation, the expression of both
MuRF1 and MAFbx significantly increases by 3 days and then Identification of MuRF1 and MAFbx Substrates
decreases to baseline by 14 days. In comparison, at 3 days of
MuRF1 and MAFbx were first identified as atrophy-associ-
denervation, proteasome activity is elevated for only a couple
ated E3 ligases expressed selectively in muscle in 2001 (11,
of subunits (26S ␤1 and ␤2), whereas, at 14 days proteasome
32), however, much is still unknown regarding how their
activity is significantly elevated in all subunits (26S (␤1, ␤2,
upregulation contributes to the loss of skeletal muscle mass.
␤5, and 20S ␤1, ␤2, ␤5) (31). In MuRF1 KO mice, at 3 days We have compared the response of WT and MuRF1 KO mice
of denervation, proteasome activity is elevated in the 26S ␤2 to denervation and dexamethasone treatment and found that
subunit only, while at 14 days of denervation, proteasome while muscle mass is spared in the KO mice under both
activity is significantly elevated in all subunits. Interestingly, conditions, the mechanisms of action appear to be very differ-
the increase in proteasome subunit activity is significantly ent (5, 31). For example, following dexamethasone treatment,
higher in the KO mice compared with WT mice. The finding muscle sparing in the MuRF1 KO mice appears to be related to
that proteasome activity increased to a greater extent in the the suppression of FOXO1 gene expression and the mainte-
MuRF1 KO mice than the WT mice was unexpected since nance of protein synthesis (5). In comparison, following de-
higher proteasome activity has generally been linked to an nervation, muscle sparing in the MuRF1 KO mice is related to
increase in muscle atrophy (56), and a report by Cohen et al. the suppression of genes associated with inactivity (such as
suggested that proteasome activity was decreased in the HDAC4 and neuromuscular junction associated genes) and a
MuRF1 KO mice (16). paradoxical increase in the ubiquitin proteasome pathway (31).
An elevation in proteasome activity in the MuRF1 KO mice These results could suggest that different sets of substrates
was also found in response to aging. Examination of old (24 are targeted for ubiquitination by MuRF1 under these divergent
mo) WT and MuRF1 KO male mice revealed that muscle mass atrophy conditions. What mechanisms could accomplish dif-
was maintained in the MuRF1 KO mice with age, while it ferential targeting of substrates under different atrophy condi-
decreased in WT mice (38). Measurement of proteasome tions? One possibility is that MuRF1 (an E3 ligase) pairs with
activity revealed a decrease in the activity of all of the protea- different E2 ligases under different atrophy conditions. It has
some subunits as a consequence of aging in WT mice. In been shown that the specific E2-E3 pairing can alter the type of
contrast, old MuRF1 KO mice had higher proteasome subunit ubiquitin chains that are added to a substrate (42, 60). It is also
activities compared with WT. These data suggest that an possible that different E2-E3 pairings affect the specific sub-
elevated proteasome activity is protective under some condi- strates that are targeted. Furthermore, it is known that different
tions, contributing to an increase in protein quality control and proteins can bind to E3 ligases and possibly alter their local-
a decrease in cellular stress. The mechanism(s) by which a ization within the cell and substrates (83). One possibility is
deletion of MuRF1 leads to an increase in proteasome activity that in response to different stressor, MuRF1 has different
are unclear and are under study. binding partners which modify its cellular localization and

J Appl Physiol • doi:10.1152/japplphysiol.00381.2020 • www.jap.org


Downloaded from journals.physiology.org/journal/jappl (181.163.161.027) on September 4, 2021.
REGULATION OF SKELETAL MUSCLE ATROPHY 279
the cellular localization of the protein, modifying protein-
protein interactions and regulating transcriptional activity (20).
SKELETAL MUSCLE ATROPHY IS A COMPLEX PROCESS

The loss of skeletal muscle mass is the final common


outcome of many diseases and conditions. Over the past twenty
years, a number of proteins and pathways have been identified
as playing a role in the atrophy process (10, 23, 69, 71) (see
Fig. 6). The E3 ubiquitin ligases, MuRF1 and MAFbx, are
particularly interesting because they are relatively muscle spe-
cific, are expressed at low levels under resting conditions, and
are upregulated under all forms of muscle atrophy. It is note-
worthy that MuRF1 and MAFbx are upregulated under diver-
gent atrophy-inducing conditions such as disuse, inflammation,
metabolic stress, oxidative stress, and excess glucocorticoids;
which is not true for all atrophy associated genes. For example,
HDAC4 and Gadd45a are strongly upregulated in response to
denervation and disuse, but not induced in response to gluco-
corticoid treatment (14, 27). There continues to be an ongoing
search to determine whether there is a common set of genes
responsible for muscle atrophy. To answer the question, inves-
tigations should examine multiple divergent atrophy models at
multiple time points during the course of atrophy. It is possible
that different stimuli such as decreased loading, inactivity,
inflammation, metabolic stress or oxidative stress activate an
array of initial factors (e.g., activated GR, FOXOs, ATF4,
NF-␬B, HDAC4, CEBP␤, Smad1/2) that activate some com-
mon pathways (e.g., MuRF1 and MAFbx) and some divergent
pathways (e.g., increased Gadd45a and p21 expression, de-
creased mTORC1 activation, etc.) all leading to the activation
Fig. 5. E3 Ubiquitin ligases expressed in skeletal muscle. There are over 600
E3 ligases in the human genome and they can be broadly classified into two
types: RING finger E3s and the cullin-RING E3 ligase (CRL) superfamily.
MuRF1 belongs to the Ring finger E3 family and MAFbx belongs to the
cullin-RING E3 superfamily. MuRF1 (Trim63) and MAFbx (Fbxo32) repre-
sent just two of the many E3 ligases expressed in skeletal muscle. The majority
of RING finger E3s (A) and cullin-RING E3s (B) expressed in skeletal muscle
have not been studied in any detail.

potential substrates. While the literature suggests that the


primary substrates for MuRF1 are proteins associated with the
thick myofilament (16), this list is most likely incomplete and
may not be accurate since most of the data have been collected
in vitro using E2 ligases that may not associate with MuRF1 in
vivo. Clearly, more investigation is needed to identify the in
vivo substrates of MuRF1 and MAFbx under different atrophy
conditions. Identification of the substrates will provide further
understanding of the mechanisms by which MuRF1 and
MAFbx regulate skeletal muscle size.
MuRF1 and MAFbx represent just two E3 ligases that are
expressed in skeletal muscle (see Fig. 5). To date, only a
handful of E3 ligases have been investigated in skeletal muscle Fig. 6. Multiple pathways lead to muscle atrophy. This diagram illustrates that
under baseline or atrophy-inducing conditions. In addition to skeletal muscle atrophy is initiated by a variety of signals as listed in the blue
MuRF1 and MAFbx, the E3 ligases that have received the most box. These divergent signals activate a variety of transcription factors leading
attention, and been shown to play some role in atrophy, include to the upregulation of a variety of genes and alterations in signaling pathways.
The E3 ligases MuRF1 and MAFbx are highlighted since they are two genes
Fbx30 (MUSA (70),), Trim 32 (17, 46, 47), and Nedd4 (45, that are upregulated in response to divergent signals such as disuse, inflam-
59). A major challenge for the field has been identifying the mation, oxidative stress, and starvation. Other genes such as MUSA1,
substrates targeted for ubiquitination by an individual E3 ligase Gadd45a, and p21 appear to upregulate in response to specific signals.
and then understanding the effect of ubiquitination on the Suppression of the mTORC1 pathways and protein synthesis is a common
response to many atrophy conditions; an exception being denervation. All of
protein. While ubiquitination of a protein can target a protein these pathways lead to a common outcome, the loss of muscle mass, i.e.,
for degradation by the proteasome, it is can also serve other muscle atrophy. A major challenge is to understand how these various
functions such as altering the activity of the protein, altering pathways intersect to induce muscle atrophy.

J Appl Physiol • doi:10.1152/japplphysiol.00381.2020 • www.jap.org


Downloaded from journals.physiology.org/journal/jappl (181.163.161.027) on September 4, 2021.
280 REGULATION OF SKELETAL MUSCLE ATROPHY

of a common final program to increase the disassembly and REFERENCES


degradation of myofibrils leading to a decrease in muscle fiber 1. Adams GR, Haddad F. The relationships among IGF-1, DNA content,
size and muscle atrophy. If this model is correct, then a single and protein accumulation during skeletal muscle hypertrophy. J Appl
drug would likely not be able to treat all forms of atrophy. Physiol (1985) 81: 2509 –2516, 1996. doi:10.1152/jappl.1996.81.6.2509.
Furthermore, the timing of the treatment will be important. For 2. Adams GR, McCue SA. Localized infusion of IGF-I results in skeletal
example, following disuse and denervation-induced atrophy muscle hypertrophy in rats. J Appl Physiol (1985) 84: 1716 –1722, 1998.
doi:10.1152/jappl.1998.84.5.1716.
MuRF1 increases rapidly and then returns to baseline by 14 3. Arnold J, Campbell IT, Samuels TA, Devlin JC, Green CJ, Hipkin LJ,
days. Therefore, the use of a MuRF1 inhibitor after 14 days MacDonald IA, Scrimgeour CM, Smith K, Rennie MJ. Increased
may not be appropriate. However, deletion of MuRF1 contin- whole body protein breakdown predominates over increased whole body
ues to result in muscle sparing after 14 days of disuse or protein synthesis in multiple organ failure. Clin Sci (Lond) 84: 655–661,
denervation, suggesting that MuRF1 upregulation initiates spe- 1993. doi:10.1042/cs0840655.
4. Baar K, Esser K. Phosphorylation of p70(S6k) correlates with increased
cific signaling cascades that initiate and sustain muscle atro- skeletal muscle mass following resistance exercise. Am J Physiol Cell
phy. Thus, the critical missing data are the identification and Physiol 276: C120 –C127, 1999. doi:10.1152/ajpcell.1999.276.1.C120.
verification of the in vivo substrates for MuRF1 and other E3 5. Baehr LM, Furlow JD, Bodine SC. Muscle sparing in muscle RING
ligases which will provide insights into the mechanisms by finger 1 null mice: response to synthetic glucocorticoids. J Physiol 589:
which they regulate skeletal muscle mass. 4759 –4776, 2011. doi:10.1113/jphysiol.2011.212845.
6. Baehr LM, Tunzi M, Bodine SC. Muscle hypertrophy is associated with
increases in proteasome activity that is independent of MuRF1 and
CONCLUSIONS MAFbx expression. Front Physiol 5: 69, 2014. doi:10.3389/fphys.2014.
00069.
Skeletal muscle atrophy continues to be a serious conse- 7. Baehr LM, West DW, Marcotte G, Marshall AG, De Sousa LG, Baar
quence of many diseases and conditions for which there is no K, Bodine SC. Age-related deficits in skeletal muscle recovery following
treatment. The development of treatments relies on an under- disuse are associated with neuromuscular junction instability and ER
standing of the molecules and pathways involved in the atro- stress, not impaired protein synthesis. Aging (Albany NY) 8: 127–146,
2016. doi:10.18632/aging.100879.
phy process. Over the past two decades considerable progress 8. Baehr LM, West DWD, Marshall AG, Marcotte GR, Baar K, Bodine
has been made in our understanding of the molecular and SC. Muscle-specific and age-related changes in protein synthesis and
cellular mechanisms underlying the loss of muscle mass. The protein degradation in response to hindlimb unloading in rats. J Appl
data suggest that while the loss of muscle mass is a common Physiol (1985) 122: 1336 –1350, 2017. doi:10.1152/japplphysiol.00703.
outcome in many diseases, the pathways leading to the out- 2016.
9. Blaauw B, Reggiani C. The role of satellite cells in muscle hypertrophy.
come may vary depending on the initiating signal, i.e., unload- J Muscle Res Cell Motil 35: 3–10, 2014. doi:10.1007/s10974-014-9376-y.
ing, inactivity, elevated cytokines or malnutrition. Interest- 10. Bodine SC, Baehr LM. Skeletal muscle atrophy and the E3 ubiquitin
ingly, the upregulation of the E3 ligases MuRF1 and MAFbx ligases MuRF1 and MAFbx/atrogin-1. Am J Physiol Endocrinol Metab
seems to be a common early event in many divergent atrophy 307: E469 –E484, 2014. doi:10.1152/ajpendo.00204.2014.
conditions, e.g., inflammation vs disuse vs starvation. How- 11. Bodine SC, Latres E, Baumhueter S, Lai VK-M, Nunez L, Clarke BA,
Poueymirou WT, Panaro FJ, Na E, Dharmarajan K, Pan Z-Q, Va-
ever, it is clear that upregulation of MuRF1 and MAFbx is not lenzuela DM, DeChiara TM, Stitt TN, Yancopoulos GD, Glass DJ.
the complete picture since deletion of these genes does not Identification of ubiquitin ligases required for skeletal muscle atrophy.
result in the complete sparing of muscle mass. The challenge Science 294: 1704 –1708, 2001. doi:10.1126/science.1065874.
for the field of muscle biology is to determine the “network” of 12. Bodine SC, Stitt TN, Gonzalez M, Kline WO, Stover GL, Bauerlein R,
pathways involved in muscle atrophy and to understand how Zlotchenko E, Scrimgeour A, Lawrence JC, Glass DJ, Yancopoulos
GD. Akt/mTOR pathway is a crucial regulator of skeletal muscle hyper-
all of the various molecules, transcription factors and signaling trophy and can prevent muscle atrophy in vivo. Nat Cell Biol 3: 1014 –
pathways interact to produce muscle atrophy. Identification of 1019, 2001. doi:10.1038/ncb1101-1014.
the critical nodes that when suppressed result in a reduction or 13. Bollinger LM, Witczak CA, Houmard JA, Brault JJ. SMAD3 aug-
blockage of the atrophy process will be necessary for the ments FoxO3-induced MuRF-1 promoter activity in a DNA-binding-
successful development of new treatments. dependent manner. Am J Physiol Cell Physiol 307: C278 –C287, 2014.
doi:10.1152/ajpcell.00391.2013.
ACKNOWLEDGMENTS 14. Bongers KS, Fox DK, Ebert SM, Kunkel SD, Dyle MC, Bullard SA,
Dierdorff JM, Adams CM. Skeletal muscle denervation causes skeletal
Throughout my career, I have been fortunate to have worked with great muscle atrophy through a pathway that involves both Gadd45a and
colleagues at the University of California, Los Angeles; University of Cali- HDAC4. Am J Physiol Endocrinol Metab 305: E907–E915, 2013. doi:10.
fornia, San Diego; Regeneron Pharmaceuticals; University of California, 1152/ajpendo.00380.2013.
Davis; and most recently the University of Iowa. I would like to thank the 15. Cai D, Frantz JD, Tawa NE Jr, Melendez PA, Oh BC, Lidov HG,
many colleagues who have contributed to this research. Hasselgren PO, Frontera WR, Lee J, Glass DJ, Shoelson SE. IKK␤/
NF-␬B activation causes severe muscle wasting in mice. Cell 119: 285–
GRANTS 298, 2004. doi:10.1016/j.cell.2004.09.027.
This work was supported by grants awarded to S.C.B. by National 16. Cohen S, Brault JJ, Gygi SP, Glass DJ, Valenzuela DM, Gartner C,
Institutes of Health DK075801 AR070031 and the Veterans Administration Latres E, Goldberg AL. During muscle atrophy, thick, but not thin,
1I0RX000673. filament components are degraded by MuRF1-dependent ubiquitylation. J
Cell Biol 185: 1083–1095, 2009. doi:10.1083/jcb.200901052.
DISCLOSURES 17. Cohen S, Zhai B, Gygi SP, Goldberg AL. Ubiquitylation by Trim32
causes coupled loss of desmin, Z-bands, and thin filaments in muscle
Dr. Bodine holds equity in Emmyon, Inc., and serves on the Scientific atrophy. J Cell Biol 198: 575–589, 2012. doi:10.1083/jcb.201110067.
Advisory Board.
18. Coleman ME, DeMayo F, Yin KC, Lee HM, Geske R, Montgomery C,
Schwartz RJ. Myogenic vector expression of insulin-like growth factor I
AUTHOR CONTRIBUTIONS
stimulates muscle cell differentiation and myofiber hypertrophy in trans-
S.C.B. conceived and designed research, prepared figures, drafted, edited genic mice. J Biol Chem 270: 12109 –12116, 1995. doi:10.1074/jbc.270.
and revised, and approved final version of manuscript. 20.12109.

J Appl Physiol • doi:10.1152/japplphysiol.00381.2020 • www.jap.org


Downloaded from journals.physiology.org/journal/jappl (181.163.161.027) on September 4, 2021.
REGULATION OF SKELETAL MUSCLE ATROPHY 281
19. Condon KJ, Sabatini DM. Nutrient regulation of mTORC1 at a glance. 38. Hwee DT, Baehr LM, Philp A, Baar K, Bodine SC. Maintenance of
J Cell Sci 132: jcs222570, 2019. doi:10.1242/jcs.222570. muscle mass and load-induced growth in Muscle RING Finger 1 null mice
20. Deshaies RJ, Joazeiro CA. RING domain E3 ubiquitin ligases. Annu Rev with age. Aging Cell 13: 92–101, 2014. doi:10.1111/acel.12150.
Biochem 78: 399 –434, 2009. doi:10.1146/annurev.biochem.78.101807. 39. Jackman RW, Wu C-L, Kandarian SC. The ChIP-seq-defined networks
093809. of Bcl-3 gene binding support its required role in skeletal muscle atrophy.
21. Dreyer HC, Fujita S, Glynn EL, Drummond MJ, Volpi E, Rasmussen PLoS One 7: e51478, 2012. doi:10.1371/journal.pone.0051478.
BB. Resistance exercise increases leg muscle protein synthesis and mTOR 40. Jackson JR, Mula J, Kirby TJ, Fry CS, Lee JD, Ubele MF, Campbell
signalling independent of sex. Acta Physiol (Oxf) 199: 71–81, 2010. KS, McCarthy JJ, Peterson CA, Dupont-Versteegden EE. Satellite cell
doi:10.1111/j.1748-1716.2010.02074.x. depletion does not inhibit adult skeletal muscle regrowth following un-
22. Drummond MJ, Fry CS, Glynn EL, Dreyer HC, Dhanani S, Timmer- loading-induced atrophy. Am J Physiol Cell Physiol 303: C854 –C861,
man KL, Volpi E, Rasmussen BB. Rapamycin administration in humans 2012. doi:10.1152/ajpcell.00207.2012.
blocks the contraction-induced increase in skeletal muscle protein synthe- 41. Joseph GA, Wang SX, Jacobs CE, Zhou W, Kimble GC, Tse HW,
sis. J Physiol 587: 1535–1546, 2009. doi:10.1113/jphysiol.2008.163816. Eash JK, Shavlakadze T, Glass DJ. Partial inhibition of mTORC1 in
23. Ebert SM, Al-Zougbi A, Bodine SC, Adams CM. Skeletal muscle aged rats counteracts the decline in muscle mass and reverses molecular
atrophy: discovery of mechanisms and potential therapies. Physiology signaling associated with sarcopenia. Mol Cell Biol 39: e00141-19, 2019.
(Bethesda) 34: 232–239, 2019. doi:10.1152/physiol.00003.2019. doi:10.1128/MCB.00141-19.
24. Egner IM, Bruusgaard JC, Gundersen K. Satellite cell depletion 42. Kim HT, Kim KP, Lledias F, Kisselev AF, Scaglione KM, Skowyra D,
prevents fiber hypertrophy in skeletal muscle. Development 143: 2898 – Gygi SP, Goldberg AL. Certain pairs of ubiquitin-conjugating enzymes
2906, 2016. doi:10.1242/dev.134411. (E2s) and ubiquitin-protein ligases (E3s) synthesize nondegradable forked
25. Ferrando AA, Lane HW, Stuart CA, Davis-Street J, Wolfe RR. ubiquitin chains containing all possible isopeptide linkages. J Biol Chem
Prolonged bed rest decreases skeletal muscle and whole body protein 282: 17375–17386, 2007. doi:10.1074/jbc.M609659200.
synthesis. Am J Physiol Endocrinol Metab 270: E627–E633, 1996. doi: 43. Kimball SR, Jefferson LS. Mechanisms of translational control in liver
10.1152/ajpendo.1996.270.4.E627. and skeletal muscle. Biochimie 76: 729 –736, 1994. doi:10.1016/0300-
26. Files DC, D’Alessio FR, Johnston LF, Kesari P, Aggarwal NR, Garib- 9084(94)90077-9.
aldi BT, Mock JR, Simmers JL, DeGorordo A, Murdoch J, Willis MS, 44. Kline WO, Panaro FJ, Yang H, Bodine SC. Rapamycin inhibits the
Patterson C, Tankersley CG, Messi ML, Liu C, Delbono O, Furlow growth and muscle-sparing effects of clenbuterol. J Appl Physiol (1985)
JD, Bodine SC, Cohn RD, King LS, Crow MT. A critical role for 102: 740 –747, 2007. doi:10.1152/japplphysiol.00873.2006.
muscle ring finger-1 in acute lung injury-associated skeletal muscle 45. Koncarevic A, Jackman RW, Kandarian SC. The ubiquitin-protein
wasting. Am J Respir Crit Care Med 185: 825–834, 2012. doi:10.1164/ ligase Nedd4 targets Notch1 in skeletal muscle and distinguishes the
rccm.201106-1150OC. subset of atrophies caused by reduced muscle tension. FASEB J 21:
27. Furlow JD, Watson ML, Waddell DS, Neff ES, Baehr LM, Ross AP, 427–437, 2007. doi:10.1096/fj.06-6665com.
Bodine SC. Altered gene expression patterns in muscle ring finger 1 null 46. Kudryashova E, Kudryashov D, Kramerova I, Spencer MJ. Trim32 is
mice during denervation- and dexamethasone-induced muscle atrophy. a ubiquitin ligase mutated in limb girdle muscular dystrophy type 2H that
Physiol Genomics 45: 1168 –1185, 2013. doi:10.1152/physiolgenomics. binds to skeletal muscle myosin and ubiquitinates actin. J Mol Biol 354:
00022.2013. 413–424, 2005. doi:10.1016/j.jmb.2005.09.068.
28. Gibson JN, Smith K, Rennie MJ. Prevention of disuse muscle atrophy 47. Kudryashova E, Wu J, Havton LA, Spencer MJ. Deficiency of the E3
by means of electrical stimulation: maintenance of protein synthesis. ubiquitin ligase TRIM32 in mice leads to a myopathy with a neurogenic
Lancet 332: 767–770, 1988. doi:10.1016/S0140-6736(88)92417-8. component. Hum Mol Genet 18: 1353–1367, 2009. doi:10.1093/hmg/
29. Goldberg AL. Protein turnover in skeletal muscle. II. Effects of dener- ddp036.
vation and cortisone on protein catabolism in skeletal muscle. J Biol Chem 48. Labeit S, Kohl CH, Witt CC, Labeit D, Jung J, Granzier H. Modula-
244: 3223–3229, 1969. tion of muscle atrophy, fatigue and MLC phosphorylation by MuRF1 as
30. Goldberg AL, Goodman HM. Relationship between cortisone and mus- indicated by hindlimb suspension studies on MuRF1-KO mice. J Biomed
cle work in determining muscle size. J Physiol 200: 667–675, 1969. Biotechnol 2010: 693741, 2010. doi:10.1155/2010/693741.
doi:10.1113/jphysiol.1969.sp008715. 49. Lai KM, Gonzalez M, Poueymirou WT, Kline WO, Na E, Zlotchenko
31. Gomes AV, Waddell DS, Siu R, Stein M, Dewey S, Furlow JD, Bodine E, Stitt TN, Economides AN, Yancopoulos GD, Glass DJ. Conditional
SC. Upregulation of proteasome activity in muscle RING finger 1-null activation of akt in adult skeletal muscle induces rapid hypertrophy. Mol
mice following denervation. FASEB J 26: 2986 –2999, 2012. doi:10.1096/ Cell Biol 24: 9295–9304, 2004. doi:10.1128/MCB.24.21.9295-9304.2004.
fj.12-204495. 50. Lang F, Aravamudhan S, Nolte H, Türk C, Hölper S, Müller S,
32. Gomes MD, Lecker SH, Jagoe RT, Navon A, Goldberg AL. Atrogin-1, Günther S, Blaauw B, Braun T, Krüger M. Dynamic changes in the
a muscle-specific F-box protein highly expressed during muscle atrophy. mouse skeletal muscle proteome during denervation-induced atrophy. Dis
Proc Natl Acad Sci USA 98: 14440 –14445, 2001. doi:10.1073/pnas. Model Mech 10: 881–896, 2017. doi:10.1242/dmm.028910.
251541198. 51. Larsson L, Degens H, Li M, Salviati L, Lee YI, Thompson W,
33. Goodman CA. Role of mTORC1 in mechanically induced increases in Kirkland JL, Sandri M. Sarcopenia: aging-related loss of muscle mass
translation and skeletal muscle mass. J Appl Physiol (1985) 127: 581–590, and function. Physiol Rev 99: 427–511, 2019. doi:10.1152/physrev.00061.
2019. doi:10.1152/japplphysiol.01011.2018. 2017.
34. Gundermann DM, Walker DK, Reidy PT, Borack MS, Dickinson JM, 52. Li JB, Goldberg AL. Effects of food deprivation on protein synthesis and
Volpi E, Rasmussen BB. Activation of mTORC1 signaling and protein degradation in rat skeletal muscles. Am J Physiol 231: 441–448, 1976.
synthesis in human muscle following blood flow restriction exercise is doi:10.1152/ajplegacy.1976.231.2.441.
inhibited by rapamycin. Am J Physiol Endocrinol Metab 306: E1198 – 53. Louis E, Raue U, Yang Y, Jemiolo B, Trappe S. Time course of
E1204, 2014. doi:10.1152/ajpendo.00600.2013. proteolytic, cytokine, and myostatin gene expression after acute exercise
35. Guridi M, Kupr B, Romanino K, Lin S, Falcetta D, Tintignac L, in human skeletal muscle. J Appl Physiol (1985) 103: 1744 –1751, 2007.
Rüegg MA. Alterations to mTORC1 signaling in the skeletal muscle doi:10.1152/japplphysiol.00679.2007.
differentially affect whole-body metabolism. Skelet Muscle 6: 13, 2016. 54. McCarthy JJ, Mula J, Miyazaki M, Erfani R, Garrison K, Farooqui
doi:10.1186/s13395-016-0084-8. AB, Srikuea R, Lawson BA, Grimes B, Keller C, Van Zant G,
36. Ham AS, Chojnowska K, Tintignac LA, Lin S, Schmidt A, Ham DJ, Campbell KS, Esser KA, Dupont-Versteegden EE, Peterson CA.
Sinnreich M, Rüegg MA. mTORC1 signalling is not essential for the Effective fiber hypertrophy in satellite cell-depleted skeletal muscle.
maintenance of muscle mass and function in adult sedentary mice. J Development 138: 3657–3666, 2011. doi:10.1242/dev.068858.
Cachexia Sarcopenia Muscle 11: 259 –273, 2020. doi:10.1002/jcsm. 55. Milan G, Romanello V, Pescatore F, Armani A, Paik J-H, Frasson L,
12505. Seydel A, Zhao J, Abraham R, Goldberg AL, Blaauw B, DePinho RA,
37. Hughes SM, Schiaffino S. Control of muscle fibre size: a crucial factor in Sandri M. Regulation of autophagy and the ubiquitin-proteasome system
ageing. Acta Physiol Scand 167: 307–312, 1999. doi:10.1046/j.1365-201x. by the FoxO transcriptional network during muscle atrophy. Nat Commun
1999.00619.x. 6: 6670, 2015. doi:10.1038/ncomms7670.

J Appl Physiol • doi:10.1152/japplphysiol.00381.2020 • www.jap.org


Downloaded from journals.physiology.org/journal/jappl (181.163.161.027) on September 4, 2021.
282 REGULATION OF SKELETAL MUSCLE ATROPHY

56. Mitch WE, Goldberg AL. Mechanisms of muscle wasting. The role of Suematsu M, Morimoto C, Tanaka H. Crosstalk between glucocorticoid
the ubiquitin-proteasome pathway. N Engl J Med 335: 1897–1905, 1996. receptor and nutritional sensor mTOR in skeletal muscle. Cell Metab 13:
doi:10.1056/NEJM199612193352507. 170 –182, 2011. doi:10.1016/j.cmet.2011.01.001.
57. Montagne J, Stewart MJ, Stocker H, Hafen E, Kozma SC, Thomas G. 74. Spangenburg EE, Le Roith D, Ward CW, Bodine SC. A functional
Drosophila S6 kinase: a regulator of cell size. Science 285: 2126 –2129, insulin-like growth factor receptor is not necessary for load-induced
1999. doi:10.1126/science.285.5436.2126. skeletal muscle hypertrophy. J Physiol 586: 283–291, 2008. doi:10.1113/
58. Moore DR, Atherton PJ, Rennie MJ, Tarnopolsky MA, Phillips SM. jphysiol.2007.141507.
Resistance exercise enhances mTOR and MAPK signalling in human 75. Srikanthan P, Karlamangla AS. Muscle mass index as a predictor of
muscle over that seen at rest after bolus protein ingestion. Acta Physiol longevity in older adults. Am J Med 127: 547–553, 2014. doi:10.1016/j.
(Oxf) 201: 365–372, 2011. doi:10.1111/j.1748-1716.2010.02187.x. amjmed.2014.02.007.
59. Nagpal P, Plant PJ, Correa J, Bain A, Takeda M, Kawabe H, Rotin D, 76. Stitt TN, Drujan D, Clarke BA, Panaro F, Timofeyva Y, Kline WO,
Bain JR, Batt JA. The ubiquitin ligase Nedd4-1 participates in denerva- Gonzalez M, Yancopoulos GD, Glass DJ. The IGF-1/PI3K/Akt pathway
tion-induced skeletal muscle atrophy in mice. PLoS One 7: e46427, 2012. prevents expression of muscle atrophy-induced ubiquitin ligases by inhib-
doi:10.1371/journal.pone.0046427. iting FOXO transcription factors. Mol Cell 14: 395–403, 2004. doi:10.
60. Napolitano LM, Jaffray EG, Hay RT, Meroni G. Functional interac- 1016/S1097-2765(04)00211-4.
tions between ubiquitin E2 enzymes and TRIM proteins. Biochem J 434: 77. Vandenburgh HH, Karlisch P, Shansky J, Feldstein R. Insulin and
309 –319, 2011. doi:10.1042/BJ20101487. IGF-I induce pronounced hypertrophy of skeletal myofibers in tissue
61. Powers SK, Morton AB, Ahn B, Smuder AJ. Redox control of skeletal culture. Am J Physiol Cell Physiol 260: C475–C484, 1991. doi:10.1152/
muscle atrophy. Free Radic Biol Med 98: 208 –217, 2016. doi:10.1016/j. ajpcell.1991.260.3.C475.
freeradbiomed.2016.02.021. 78. Waddell DS, Baehr LM, van den Brandt J, Johnsen SA, Reichardt
62. Rennie MJ, Edwards RH, Emery PW, Halliday D, Lundholm K, HM, Furlow JD, Bodine SC. The glucocorticoid receptor and FOXO1
Millward DJ. Depressed protein synthesis is the dominant characteristic synergistically activate the skeletal muscle atrophy-associated MuRF1
of muscle wasting and cachexia. Clin Physiol 3: 387–398, 1983. doi:10. gene. Am J Physiol Endocrinol Metab 295: E785–E797, 2008. doi:10.
1111/j.1475-097X.1983.tb00847.x. 1152/ajpendo.00646.2007.
63. Rennie MJ, Wackerhage H, Spangenburg EE, Booth FW. Control of 79. Watson ML, Baehr LM, Reichardt HM, Tuckermann JP, Bodine SC,
the size of the human muscle mass. Annu Rev Physiol 66: 799 –828, 2004. Furlow JD. A cell-autonomous role for the glucocorticoid receptor in
doi:10.1146/annurev.physiol.66.052102.134444. skeletal muscle atrophy induced by systemic glucocorticoid exposure. Am
64. Reynolds TH IV, Bodine SC, Lawrence JC Jr. Control of Ser2448 J Physiol Endocrinol Metab 302: E1210 –E1220, 2012. doi:10.1152/
phosphorylation in the mammalian target of rapamycin by insulin and ajpendo.00512.2011.
skeletal muscle load. J Biol Chem 277: 17657–17662, 2002. doi:10.1074/ 80. Weijs PJ, Looijaard WG, Dekker IM, Stapel SN, Girbes AR, Oude-
jbc.M201142200. mans-van Straaten HM, Beishuizen A. Low skeletal muscle area is a
65. Rommel C, Bodine SC, Clarke BA, Rossman R, Nunez L, Stitt TN, risk factor for mortality in mechanically ventilated critically ill patients.
Yancopoulos GD, Glass DJ. Mediation of IGF-1-induced skeletal myo- Crit Care 18: R12, 2014. doi:10.1186/cc13189.
tube hypertrophy by PI(3)K/Akt/mTOR and PI(3)K/Akt/GSK3 pathways. 81. Weinkove D, Leevers SJ. The genetic control of organ growth: insights
Nat Cell Biol 3: 1009 –1013, 2001. doi:10.1038/ncb1101-1009. from Drosophila. Curr Opin Genet Dev 10: 75–80, 2000. doi:10.1016/
66. Roy RR, Baldwin KM, Edgerton VR. The plasticity of skeletal muscle: S0959-437X(99)00042-8.
effects of neuromuscular activity. Exerc Sport Sci Rev 19: 269 –312, 1991. 82. White RB, Biérinx AS, Gnocchi VF, Zammit PS. Dynamics of muscle
doi:10.1249/00003677-199101000-00008. fibre growth during postnatal mouse development. BMC Dev Biol 10: 21,
67. Rudar M, Fiorotto ML, Davis TA. Regulation of muscle growth in early 2010. doi:10.1186/1471-213X-10-21.
postnatal life in a swine model. Annu Rev Anim Biosci 7: 309 –335, 2019. 83. Williams FP, Haubrich K, Perez-Borrajero C, Hennig J. Emerging
doi:10.1146/annurev-animal-020518-115130. RNA-binding roles in the TRIM family of ubiquitin ligases. Biol Chem
68. Sandri M, Sandri C, Gilbert A, Skurk C, Calabria E, Picard A, 400: 1443–1464, 2019. doi:10.1515/hsz-2019-0158.
Walsh K, Schiaffino S, Lecker SH, Goldberg AL. Foxo transcription 84. Wong TS, Booth FW. Protein metabolism in rat tibialis anterior muscle
factors induce the atrophy-related ubiquitin ligase atrogin-1 and cause after stimulated chronic eccentric exercise. J Appl Physiol (1985) 69:
skeletal muscle atrophy. Cell 117: 399 –412, 2004. doi:10.1016/S0092- 1718 –1724, 1990. doi:10.1152/jappl.1990.69.5.1718.
8674(04)00400-3. 85. Wu CL, Kandarian SC, Jackman RW. Identification of genes that elicit
69. Sartori R, Gregorevic P, Sandri M. TGF␤ and BMP signaling in skeletal disuse muscle atrophy via the transcription factors p50 and Bcl-3. PLoS
muscle: potential significance for muscle-related disease. Trends Endocri- One 6: e16171, 2011. doi:10.1371/journal.pone.0016171.
nol Metab 25: 464 –471, 2014. https://doi.org/10.1016/j.tem.2014.06.002. 86. Yang Y, Jemiolo B, Trappe S. Proteolytic mRNA expression in response
70. Sartori R, Schirwis E, Blaauw B, Bortolanza S, Zhao J, Enzo E, to acute resistance exercise in human single skeletal muscle fibers. J Appl
Stantzou A, Mouisel E, Toniolo L, Ferry A, Stricker S, Goldberg AL, Physiol (1985) 101: 1442–1450, 2006. doi:10.1152/japplphysiol.00438.
Dupont S, Piccolo S, Amthor H, Sandri M. BMP signaling controls 2006.
muscle mass. Nat Genet 45: 1309 –1318, 2013. doi:10.1038/ng.2772. 87. Yarasheski KE, Zachwieja JJ, Bier DM. Acute effects of resistance
71. Schiaffino S, Dyar KA, Ciciliot S, Blaauw B, Sandri M. Mechanisms exercise on muscle protein synthesis rate in young and elderly men and
regulating skeletal muscle growth and atrophy. FEBS J 280: 4294 –4314, women. Am J Physiol Endocrinol Metab 265: E210 –E214, 1993. doi:10.
2013. doi:10.1111/febs.12253. 1152/ajpendo.1993.265.2.E210.
72. Shima H, Pende M, Chen Y, Fumagalli S, Thomas G, Kozma SC. 88. Zhang G, Jin B, Li YP. C/EBP␤ mediates tumour-induced ubiquitin
Disruption of the p70(s6k)/p85(s6k) gene reveals a small mouse pheno- ligase atrogin1/MAFbx upregulation and muscle wasting. EMBO J 30:
type and a new functional S6 kinase. EMBO J 17: 6649 –6659, 1998. 4323–4335, 2011. doi:10.1038/emboj.2011.292.
doi:10.1093/emboj/17.22.6649. 89. Zhang G, Li Y-P. p38␤ MAPK upregulates atrogin1/MAFbx by specific
73. Shimizu N, Yoshikawa N, Ito N, Maruyama T, Suzuki Y, Takeda S, phosphorylation of C/EBP␤. Skelet Muscle 2: 20, 2012. doi:10.1186/2044-
Nakae J, Tagata Y, Nishitani S, Takehana K, Sano M, Fukuda K, 5040-2-20.

J Appl Physiol • doi:10.1152/japplphysiol.00381.2020 • www.jap.org


Downloaded from journals.physiology.org/journal/jappl (181.163.161.027) on September 4, 2021.

You might also like