Till minne av min älskade morfar

Till minne av min älskade morfar
All our dreams can come true,
if we have the courage to pursue them
Walt Disney (1901-1966)
POPULÄRVETENSKAPLIG
SAMMANFATTNING
Av en människas totala vikt utgör skelettmuskulaturen ca 40 %.
Skelettmusklerna är viljestyrda och behövs för att vi skall kunna hålla oss
upprätta och röra oss som vi vill. Muskler är uppbyggda av muskelfibrer och är
den största proteinreservoaren i kroppen. Proteiner är uppbyggda av
aminosyror, vissa av dessa aminosyror är essentiella och kan inte syntetiseras i
kroppen utan dessa kan kroppen endast få via födan. Vid t.ex. svält, cancer,
sepsis etc. bryts proteinerna ner till aminosyror som sedan omvandlas i levern
till glukos för att ge kroppen ny energi. Ett organ i kroppen som är speciellt
beroende av glukos är hjärnan. Förlust av muskelmassa som följd av
proteinnedbrytning kan leda till försvagning av kroppen och orsaka
följdsjukdomar. Därför är det viktigt att hålla musklerna friska. Forskningen i
denna avhandling är grundforskning och i avhandlingen har olika proteiner
involverade i muskelförtvining, atrofi, och uppbyggnad, hypertrofi, studerats
ingående. Atrofi och hypertrofi regleras med hjälp av proteinnedbrytning samt
proteinsyntes. Normalt balanserar dessa varandra men om den ena alternativt
den andra överväger kan atrofi alternativt hypertrofi förekomma. Vid atrofi
minskar muskelmassan, detta beror på att cellerna förlorar organeller,
proteiner och cytoplasma. För att muskler skall kunna byggas upp alternativt
brytas ned krävs att det skickas signaler via olika signaleringsvägar. En
signaleringsväg är som en lång väg med påfarter, avfarter samt trafikljus och
beroende på vilken riktning signalen går får man olika resultat.
För att kunna studera atrofi/hypertrofi i muskler används olika djurmodeller,
oftast mus och råtta men även djur som går in i dvala, så som björn och ekorre,
studeras flitigt eftersom det har visat sig att de inte förlorar så mycket
muskelmassa trots deras inaktivitet i flera månader. Detta är något som man
skulle vilja kunna överföra på människan och i vården då människor ligger till
sängs länge vid sjukdom vilket ger en försvagning och minskning i
muskelmassa. Djurmodellen som har använts i de olika studierna till denna
avhandling är en 6 dagars denerverad musmodell. Där man antingen har
denerverat musens ben alternativt musens diafragma genom att klippa bort en
bit av den nerv som styr den specifika muskelgruppen. Arbetena i denna
avhandling har utgått från proteinet Akt och studerat olika faktorer som
påverkas av Akt direkt, genom fosforylering och inhibering, och indirekt men
även proteiner och faktorer som finns runt omkring och påverkar proteinsyntes
och proteinnedbrytning. I paper I studerade vi Akt ingående och dess
nedströmsfaktorer som indikerade en ökad proteinsyntes vilket fortsatte i
paper IV där vi tittade på eIF4G som också påverkar proteinsyntesen. Paper II
handlade om FoxO vars aktivitet kan regleras via fosforylering och acetylering
och hur det påverkar faktorer av betydelse för proteinnedbrytningen. I paper
III studerades hypotesen att skillnaden mellan denerverad atrofisk och
hypertrofisk muskel beror på var p38 och MK2 är lokaliserade, om det är i
cellens kärna eller i cellens cytoplasma. Slutsatsen som kan dras från dessa fyra
arbeten är att det är inte bara en faktor som påverkar proteinnedbrytning och
proteinsyntes utan flera och de verkar tillsammans i ett komplicerat system där
man ännu inte är riktigt säker på vad alla faktorer gör och hur de påverkar
varandra.
LIST OF PUBLICATIONS
This thesis is based on the following publications, referred to in the text by
their roman numerals.
Paper I:
Akt (protein kinase B) isoform phosphorylation and signaling
downstream of mTOR (mammalian target of rapamycin) in
denervated atrophic and hypertrophic mouse skeletal muscle.
Marlene Norrby, Kim Evertsson*, Ann-Kristin Fjällström*, Anna
Svensson and Sven Tågerud
Journal of Molecular Signaling 2012, 7:7.
*equal contributors
Paper II:
Forkhead box O1 and Muscle RING Finger 1 protein
expression in atrophic and hypertrophic denervated mouse
skeletal muscle
Ann-Kristin Fjällström, Kim Evertsson, Marlene Norrby and
Sven Tågerud
Journal of Molecular Signaling 2014, 9:9.
Paper III:
p38 mitogen-activated protein kinase and mitogen-activated
protein kinase-activated protein kinase 2 (MK2) signaling in
atrophic and hypertrophic denervated mouse skeletal muscle
Kim Evertsson, Ann-Kristin Fjällström, Marlene Norrby and
Sven Tågerud
Journal of Molecular Signaling 2014, 7:7.
Paper IV:
Expression and phosphorylation of eukaryotic translation
initiation factor 4-gamma (eIF4G) in denervated atrophic and
hypertrophic mouse skeletal muscle
Ann-Kristin Fjällström, Marlene Norrby and Sven Tågerud
Submitted
All published papers are reproduced with permission from the respective
publisher.
Additional work outside the scope of this thesis
Effects of adjunct galantamine to risperidone, or haloperidol, in animal
models of antipsychotic activity and extrapyramidal side-effect liability:
involvement of the cholinergic muscarinic receptor.
Marie-Louise G Wadenberg, Ann-Kristin Fjällström, Malin Federley,
Pernilla Persson and Pia Stenqvist.
Int J Neuropsychopharmacol. 2011, Jun;14(5):644-54.
ABBREVIATIONS
4EBP1
Eukaryotic initiation factor 4E
binding protein 1
AKT
Protein kinase B
CBP
CREB-binding protein
eIF
Eukaryotic Initiation Factor
Fn14
Factor-inducible 14
FoxO
Forkhead Box O
GR
Glucocorticoid receptors
GSK
Glycogen synthase kinase
HAT
Histone acetyltransferase
Hsp
Heat shock protein
IGF
Insulin-like growth factor
MAFbx/Atrogin-1
F-box protein atrogin 1/MAFbx/FBXO32
MK2
Mitogen-activated protein kinase-activated
protein kinase 2 (MAPKAPK-2)
Mnks
Mitogen activated protein kinaseinteracting kinases
mTOR
Mammalian Target Of Rapamycin
MuRF1
Muscle specific ring finger protein 1/ TRIM63
MyHC
Myosin Heavy Chain
NFκB
Nuclear factor kappa B
p38 MAPK
p38 Mitogen Activated Protein Kinase
p70S6K1
70 ribosomal protein S6 kinase
PDK1
3’-phosphoinositide-dependent kinase 1
PIKK
Phosphatidylinositol kinase related
protein kinase
PI3K
Phosphatidylinositol 3-kinase
rpS6
Ribosomal protein S6
S
Serine
T
Threonine
TNFα
Tumor necrosis factor α
TWEAK
TNF-like weak inducer of apoptosis
Y
Tyrosine
TABLE OF CONTENTS
1. INTRODUCTION ................................................................................ 13
2. SKELETAL MUSCLE .......................................................................... 15
Skeletal muscle anatomy ..................................................................... 15
Fiber types .......................................................................................... 16
Models for studying atrophy and hypertrophy ................................... 17
Atrophy models .................................................................................. 17
Disease models ................................................................................... 17
Disuse models ..................................................................................... 18
Hibernation ........................................................................................ 20
Hypertrophy models ........................................................................... 21
Denervation ........................................................................................ 21
3. ATROPHY and HYPERTROPHY SIGNALING PATHWAYS ..... 22
Protein degradation ............................................................................ 25
MuRF1 and MAFbx/atrogin-1.......................................................... 25
FoxO................................................................................................... 27
NFκB and TNFα ............................................................................... 31
p38, MK2 and heat shock proteins .................................................... 32
Glucocorticoids................................................................................... 36
Akt-Protein kinase B.......................................................................... 37
Protein synthesis ................................................................................. 37
p70S6K1 and rpS6 ............................................................................. 37
The eIF4 family .................................................................................. 37
eIF4G ................................................................................................. 39
4EBP1 and eIF4E .............................................................................. 40
eIF4F, sepsis and leucine .................................................................... 41
Mammalian Target of Rapamycin ..................................................... 42
GSK3β and eIF2B.............................................................................. 44
Akt-Protein kinase B.......................................................................... 44
4. AIM and OBJECTIVES ........................................................................ 47
Paper I ................................................................................................ 47
Paper II ............................................................................................... 47
Paper III ............................................................................................. 48
Paper IV ............................................................................................. 48
5. METHODOLOGICAL CONSIDERATIONS ................................. 49
Animals .............................................................................................. 49
Surgery and muscles ........................................................................... 49
Standard protein extraction Papers I-IV ............................................ 51
Nuclear and cytosolic fractions Papers II and III ............................... 51
Western blotting................................................................................. 52
Data analysis and statistics....................................................................... 54
6. RESULTS and DISCUSSION.................................................................... 56
Paper I ..................................................................................................... 57
Paper II .................................................................................................... 59
Paper III .................................................................................................. 62
Paper IV................................................................................................... 63
7. CONCLUSIONS......................................................................................... 66
8. ACKNOWLEDGEMENTS ...................................................................... 68
9. REFERENCES............................................................................................ 73
1. INTRODUCTION
The human body is built up of cells, each cell has a purpose for example
skeletal muscle cells build up muscle fibers to big muscles which control our
movements and the ability to stand. Approximately 40 % of the human body
weight consists of skeletal muscles. This muscle type is involved in many
functions that are vital for maintaining a healthy life such as force generation
and locomotion, heat production and glucose homeostasis. Muscles are also
the body’s protein reservoir and regulating protein synthesis is important for
cell metabolism and growth.
Skeletal muscle mass and growth is controlled by protein synthesis and protein
degradation which function together to maintain a balance [1] between
increased or decreased muscle mass [2]. This balance is changed when atrophy
occurs, a decrease in muscle mass that can be triggered by disuse, chronic
diseases, immobilization, cancer [3] elevated glucocorticoids, inflammation or
nutritional deprivation [4], or when hypertrophy occurs, a state of increased
muscle mass due to increased mechanical load, high usage and/or anabolic
stimulation [2, 5]. Plasticity in muscle mass and muscle function results from a
number of changes in intracellular activities and signal pathways in response to
various signals. These signals and responses occur both in the nucleus and the
cytoplasm [1]. The main cellular degradation pathways include the ubiquitinproteasome system and the autophagy-lysosome pathway [6].
13
Atrophy and hypertrophy can be studied using different animal models. In
this thesis a denervation model has been used. Other models include e.g.
sepsis/starvation as atrophy models and models based on alterations in muscle
use such as hind limb suspension or mechanical overload resulting in atrophy
and hypertrophy respectively.
14
2. SKELETAL MUSCLE
Skeletal muscles consist of a large number of muscle fibers that are very large
and multinucleated cells in contrast to most other cells that have only one
nucleus. Each muscle fiber is surrounded by a basal membrane, a tube-like
structure consisting of glycoproteins. Inside the basal membrane there are
mononucleated satellite cells that can divide to form a new muscle fiber within
the tube of basal membrane following damage to the original muscle fiber [7].
Skeletal muscle anatomy
Skeletal muscles can be described at different levels e.g. whole muscles, motor
units and muscle fiber types. A motor unit consists of a motor neuron and all
the muscle fibers innervated by this motor neuron. The size of the motor unit
i.e. the number of muscle fibers that a motor neuron controls varies between
different muscles. Muscles requiring a high degree of fine motor control only
have a few muscle fibers innervated by each motor neuron whereas e.g.
postural muscles have larger numbers of muscle fibers in each motor unit. All
muscle fibers in a given motor unit are of the same fiber type. The activity
pattern in the motor neuron determines the fiber type of the innervated
muscle fibers. There are four different skeletal muscle fiber types, type 1, 2A,
2X and 2B. For type 1 fibers the motor neurons show the highest degree of
activity [8]. Most muscles consist of a mixture of fiber types.
15
Fiber types
The four different fiber types in skeletal muscles express different specific
myosin heavy chain (MyHC) isoforms [9].
Type 1 – MyHC-1/slow, coded by the MYH7 gene
Type 2A – MyHC-2A, coded by the MYH2 gene
Type 2X – MyHC-2X, coded by the MYH1 gene
Type 2B – MyHC-2B, coded by the MYH4 gene [9]
Type 1 and type 2A fibers use oxidative metabolism and are fibers that can
endure a lot, while type 2B fibers use glycolytic metabolism and are fibers that
do not endure for long and produce lactic acid. Type 2X fibers are
intermediate and have properties intermediate between 2A and 2B fibers. All
four fiber types can be found in most mammals but type 1, 2A and 2X are the
fiber types that are present in most human muscles [9, 10]. When a muscle
fiber only expresses a single MyHC it is a pure fiber type and when expressing
two or more MyHCs it is called a hybrid fiber type [11]. The number of
hybrid fibers (1 and 2A, 2A and 2X, or 2X and 2B) increases when fiber type
shifts take place in response to muscle atrophy, induced by denervation or
other causes, but also in response to exercise and electrical stimulation [12].
16
Models for studying atrophy and hypertrophy
Skeletal muscle is a very plastic tissue and a number of different conditions can
cause alterations in muscle mass. Such changes in muscle mass likely occur as a
result of changes in the rates of muscle protein synthesis and protein
degradation.
Atrophy models
Disease models
Sepsis, a systemic inflammatory response, may be caused by microbial
infection or by administration of endotoxin, lipopolysaccharide (LPS), found
in the cell wall of Gramnegative bacteria [13]. LPS administration inhibits
protein synthesis and activates proteolysis in muscles through the ubiquitinproteasome pathway [13]. In rats made septic by injection of E. coli an acute
septic phase (2 days post injection) was associated with increased protein
degradation and a later, chronic septic phase (6 days post injection), was
associated with both increased protein synthesis and increased proteolysis in
epitrochlearis muscle incubated in vitro [14]. Caecal ligation and puncture
(CLP) is another model of sepsis that has been associated with decreased
protein synthesis and increased protein degradation in skeletal muscle [13].
Half of all patients with cancer experience cachexia and muscle wasting and
approximately one fifth of these patients will die as a direct consequence of
cachexia. A decrease in protein synthesis and an increase in protein
degradation through activation of the ubiquitin-proteasome system results in
loss of skeletal muscle mass. Studies of cachexia and cancer in animal models
are done by transplanting tumour cells to the animal or by administering large
amounts of potent carcinogens [13]. Increased protein degradation and
17
decreased protein synthesis appears to be common factors in many disease
models of skeletal muscle atrophy.
When muscles become atrophic in pathological states such as sepsis,
starvation, cachexia etc. an increase in circulating glucocorticoid levels can be
detected which indicates that glucocorticoids could trigger atrophy in these
conditions [15]. Glucocorticoids increases the rate of protein degradation and
decreases the rate of protein synthesis in skeletal muscle [15].
Disuse models
Disuse muscle atrophy occurs as a consequence of bed rest, spinal cord injury,
joint immobilization and neurodegeneration. During disuse, loss of muscle
mass and cross-sectional area occurs in the muscles that are used in normal
standing and locomotion such as the lower limb extensors and flexors [4]. The
muscle loss is rapid during the first 14-30 days of unloading but a nadir is
eventually reached after which no more muscle loss occurs despite the fact that
the muscles remain in a state of unloading and inactivity [4]. In unloading
conditions there is an immediate suppression of basal protein synthesis which
remains suppressed for the whole unloading period. Animal and human
studies appear to have a discrepancy regarding the rate of muscle loss in
response to disuse where animals show a larger muscle loss than humans [4].
The degree of unloading and inactivity and the muscle type are factors that
influence the rate and amount of muscle mass loss [16].
A number of different models of hind-limb immobilization have been used for
studies of muscle atrophy. A significant atrophy and loss of muscle strength in
hind-limb muscles was observed after two weeks of cast immobilization.
There were no significant differences between atrophy in slow-twitch soleus
following immobilization with plantarflexion and fast-twitch EDL muscles
following immobilization with dorsiflexion [17]. The medial gastrocnemius
18
muscle in immobilization and hind-limb unloading disuse models has a
muscle mass loss of 20-40% after 7-14 days depending on both a decrease in
protein synthesis but also an increase in protein degradation [16].
Hind-limb suspension is a tail suspension model based on the absence of
weight bearing, which is necessary for maintaining skeletal muscle mass and is
used to study microgravity-induced atrophy. Rodents are elevated by their tail
with an angle of 30° head-down tilt which avoids weight bearing by the
hindquarters but provides normal weight bearing on the forelimbs [18].
Muscle mass reduction was maximal after 14 days of hind-limb suspension
(quadriceps, anterior tibial, extensor digitorum longus, soleus, plantaris, left
calf complex and gastrocnemius muscle). Both the functional strength and the
isolated muscle strength were reduced [19].
Up to 30% of intensive care unit (ICU) patients experience muscle wasting.
This is added to the primary disease of the patient and has negative effects
both on the recovery time and increases the mortality [13]. This condition is
called acute quadriplegic myopathy, AQM, and occurs in patients as a
consequence of anesthesiology and intensive care. It causes severe muscle
weakness and atrophy. The condition is studied in rodents in a model of
corticosteroids combined with peripheral denervation of distal hind-limb
muscles or in pharmacologically paralyzed and mechanically ventilated animals
[20].
Protein synthesis is decreased in this disuse model and protein
degradation is increased [13].
Spinal cord isolation is another disuse model in which a portion of the spinal
cord is isolated via complete spinal cord transections. The muscles associated
with the affected motor neurons become inactivated [21].
19
Hibernation
Hibernation is a physiological state that many animals go through during the
winter as a period of inactivity with unloading and starvation and with lower
core body temperature and suppressed metabolic rate [22]. Hibernation is
unique in the sense that the skeletal muscles are largely protected from loss of
muscle mass despite unloading, inactivity and nutritional deprivation during
this period [4].
A frequently studied mammalian hibernator is the thirteen-lined ground
squirrel which has a hibernation period of 6-7 months. During this period the
squirrel has deep torpor but also short periods of arousal that last less than 24
hours. A reason for these arousal periods could be to decrease the disuseinduced muscle atrophy by having regular neural activation [4]. When present,
skeletal muscle atrophy largely occurs in the first few months and does not
progress in the final 3 months of hibernation [23].
The brown bear hibernates 5-6 months each year and skeletal muscle atrophy
is minimal during this time period. Unilateral transection of the common
peroneal nerve in summer active and winter hibernating bears showed that
after 11 weeks of denervation the loss of muscle mass and fiber cross-sectional
area in cranial tibial muscle (equivalent to anterior tibial in human) and the
long digital extensor (EDL) were significantly less in hibernating animals
compared to active animals [24].
20
Hypertrophy models
Functional overload (compensatory overload) [25] by synergist ablation or
hind-limb reloading following hind-limb suspension are two models of
mechanical loading of muscles [26]. A chronic increase in loading and
activation and subsequent increase in muscle mass and strength in rodents
occurs after functional overload (FO) [27, 28] caused by removal of the major
synergistic muscle to the investigated muscle [29]. Chronic stretch is a
nonsurgical hypertrophy model that gives a very fast and large hypertrophic
response. The advantage of this model is that it can be executed in a similar
way in both animals and humans. The animal has a cast that forces the muscle
in question to be in a lengthened position [25].
Denervation
Denervation is a model that leads to inactivity in muscles after a complete
breakoff in the communication between muscle and nerve. This breakoff
results in many changes in the properties of muscles, such as increased
expression of acetylcholine receptors and expression of the embryonic
acetylcholine receptor gamma-subunit [30]. Other changes include an increase
in the expression of tetrodotoxin-resistant sodium channels [31, 32]. Hindlimb muscles such as the anterior tibial, gastrocnemius and soleus muscles
undergo a continuous atrophy after denervation, like most skeletal muscles
[33]. The hemidiaphragm muscle, on the other hand, goes through a transient
hypertrophic state after denervation which may be as a result of passive
stretching. This passive stretch is the result of the continued contractions in
the
contralateral
innervated
hemidiaphragm
[34-36].
The
transient
hypertrophic state in the hemidiaphragm lasts somewhere between 6-10 days,
after this time period the muscle loses muscle mass and becomes atrophic
instead [34, 36].
21
3. ATROPHY AND
HYPERTROPHY SIGNALING
PATHWAYS
Skeletal muscle mass and fiber size changes depend on the balance between
protein degradation and protein synthesis. Physical activity, metabolism and
hormones are factors that induce adaptive changes in skeletal muscle mass.
When the rate of protein degradation exceeds that of protein synthesis a state
of muscle wasting (atrophy) occurs. Loss of muscle mass is a process that is
regulated by catabolic conditions or inactivity induced signaling pathways. The
ubiquitin-proteasome and the autophagy-lysosome system are major protein
degradation pathways that are activated during muscle atrophy. A
transcription dependent program that modulates the expression of ratelimiting factors in these proteolytic systems controls this response [6].
Atrophy is to some extent controlled by atrogenes which are controlled by
transcription factors, and these transcription factors are controlled by signaling
factors such as insulin-like growth factor 1 (IGF-1), amino acids, the protein
kinase
B
(Akt)
etc.
Atrogenes
include
Muscle
RING
finger
1
(MuRF1/TRIM63) and muscle atrophy F-box protein (MAFbx/atrogin-1)
which are atrophy-related E3 ubiquitin ligases. Forkhead box O (“others”)
22
(FoxO) transcription factors activate gene transcription of the atrogenes
(Figure 1) [6, 37-41].
When phosphorylated by e.g. Akt, FoxO is transported out of the nucleus to
the cytoplasm and will become transcriptionally inactive. This leads to less
transcription of factors such as MuRF1 and MAFbx/atrogin-1, and decreased
protein degradation. When dephosphorylated in the cytoplasm FoxO will be
transported back into the nucleus. This increases the transcription of target
genes and leads to increased muscle protein degradation [42].
Activation of Akt, thus, reduces protein degradation but also increases protein
synthesis largely through activation of mammalian target of rapamycin
(mTOR) and downstream factors [43] although Akt also influences protein
synthesis through inhibition of glycogen synthase kinase 3-β (GSK-3β) [44].
mTOR is also regulated by factors other than Akt and will in the presence of
glucose and nutrients increase protein synthesis through a rapamycin sensitive
signaling pathway [45] and promote phosphorylation of eukaryotic translation
initiation factor 4E-binding protein 1 (4EBP1) and 70 kDa ribosomal protein
S6 kinase 1 (p70S6K1) [46] both of which are important for the regulation of
protein synthesis.
23
Figure 1. Simplified schematic image over signaling factors that affect protein synthesis and
protein degradation.
Eukaryotic translation factors are required for protein synthesis and include
eukaryotic initiation-, elongation- and termination factors [5]. p38 Mitogen
Activated Protein Kinase, p38 MAPK has also been suggested to play a role in
the balance between protein synthesis and protein degradation [47, 48]. p38
phosphorylates serine/threonine residues in substrates [49] such as mitogenactivated protein kinase-activated protein kinase 2 (MAPKAPK-2/MK2)
[50]. MK2 may regulate nuclear export of p38 [51] and also phosphorylates
the heat shock protein (Hsp) 25/27 in the cytoplasm. Heat shock proteins are
a group of proteins believed to have a major role in the maintenance of muscle
homeostasis, especially during adaptation to various stressors [52-54].
24
Protein degradation
MuRF1 and MAFbx/atrogin-1
MuRF1 and MAFbx/atrogin-1 are atrophy-related E3 ubiquitin ligases [6,
38-40, 55]. Proteins that are degraded by the 26S proteasome are targeted by
ubiquitin (Figure 2) [55]. E1 is an ubiquitin activating enzyme, E2 is an
ubiquitin conjugating enzyme and E3 is an ubiquitin-ligase that moves
ubiquitin from E2 to the protein substrate [47, 55]. The ubiquitinated protein
becomes docked at the proteasome and degraded. Only a few E3s are known
to be involved in the regulation of atrophy and are transcriptionally induced in
atrophying muscle [6].
Figure 2. Proteins that are to be degraded by the 26S proteasome will be targeted by ubiquitin.
The enzymes E1 (ubiquitin activating enzyme), E2 (ubiquitin conjugating enzyme) and E3
(ubiquitin-ligase) move ubiquitin residues between each other until the substrate has several
ubiquitin residues attached and is ready to enter the proteasome where the protein is degraded to
peptides.
25
MuRF1 and MAFbx/atrogin-1 expression in resting muscles are low but as a
response to e.g. unloading, inactivity, denervation, nutritional deprivation,
elevated glucocorticoids, inflammation and oxidative stress they become
transcriptionally up-regulated [4, 56]. MuRF1 and MAFbx/atrogin-1 have
been used as markers of muscle atrophy and null deletions of each gene in
mice result in attenuation of muscle mass loss following disuse, including
denervation [57-59]. After denervation gastrocnemius muscles of MuRF1-/mice had less muscle mass loss compared to wildtype mice. A significant
difference was observed at day 14 but not at day 7 after denervation.
MAFbx/atrogin-1–/– mice had significant muscle sparing at both 7 and 14
days after denervation [59].
Immobilization of anterior tibial muscle induced muscle atrophy with a 36 %
reduction in myofiber size compared with the untreated contralateral muscle
within a few days after immobilization. MuRF1 and MAFbx/atrogin-1 were
used as molecular markers and mRNA expressions were significantly upregulated by 5.9 and 1.9 fold respectively in mice [60]. MuRF1 inactivation
(MuRF1-/- mice) prevented atrophy of the soleus muscle after 10 days of hindlimb suspension [58]. A significant decrease in muscle mass and fiber crosssectional area with age occurs in wildtype but not in MuRF1-/- mice studied
up to the age of 24 months. The plantaris muscles of old wildtype mice (18
months) had significantly less growth relative to young mice (6 months) after
functional overload whereas old MuRF1-/-
mice had a normal growth
response [61].
After 14 days of dexamethasone treatment MuRF1-/- mice had a major
sparing of fibre cross-sectional area in gastrocnemius muscles and had
attenuated muscle weight loss in anterior tibial and triceps surae muscles.
26
MAFbx/atrogin-1–/– and wildtype mice, however, showed similar extents of
muscle weight loss after dexamethasone treatment [57].
FoxO
FoxO is a superfamily of proteins involved in for example stress resistance and
metabolism by regulating the expression of target genes. There are four
different types of FoxO in the FoxO family in mammalians, FoxO1 (FKHL),
FoxO3 (FKHRL1), FoxO4 (AFX) and FoxO6 [62-64]. FoxO1 and FoxO3
are thought to be involved in the regulation of muscle mass since
overexpression of these transcription factors has been shown to lead to reduced
skeletal muscle mass [40, 65]. There is only one FoxO gene in invertebrates
compared to the mammalian four and it seems that this transcription factor in
invertebrates extends longevity [42]. FoxO proteins bind to the conserved
consensus core recognition motif TTGTTTAC and act as transcriptional
activators [66]. A wide range of external stimuli e.g. insulin, IGF-1, other
growth factors, neurotrophins, nutrients, cytokines and oxidative stress
regulate the FoxO transcription factors by influencing FoxO protein levels,
subcellular localization, DNA-binding and transcriptional activity [42].
Phosphorylation, acetylation, ubiquitination (mono and poly) and possibly
other modifications regulate FoxO functions (Figure 3) [42]. FoxO proteins
are to a high extent located in the cytoplasm in growing cells [67] since
nuclear export is a response to growth signals and nuclear import is a response
to stress signals such as oxidative stress [67, 68]. Phosphorylations of FoxO
occur at serine and threonine sites [69], and are sequential [67]. There are
three phosphorylation sites for Akt that are essential for the translocation
27
Figure 3. Phosphorylation of FoxO by Akt and acetylation forces FoxO out of the nucleus and into
the cytoplasm where it becomes inactivated. Dephosphorylation and deacetylation in the cytoplasm
makes it possible for FoxO to be transported back to the nucleus and become active again to
stimulate gene expression.
of FoxO into the cytoplasm (cytoplasmic translocation), the location of these
three sites are: one in the forkhead domain, one at the N-terminal and the last
one at the C-terminal [67]. In contrast to FoxO1, 3 and 4, FoxO6 is not
regulated by nucleo-cytoplasmic shuttling. A reason for this could be that
FoxO6 becomes phosphorylated at only two of three sites [70] compared to
FoxO1, FoxO3 and FoxO4 [42].
Akt, serum- and glucocorticoid-inducible kinases (SGKs) phosphorylate
FoxO1 at the first (T24) and second (S256 in human FoxO1, S253 in mouse
FoxO1) phosphorylation sites which leads to creation of a binding site for
chaperone protein 14-3-3 [71-73]. 14-3-3 proteins belong to a family of
regulatory proteins that are involved in e.g. cell cycle control, apoptosis etc.
[74, 75]. They bind to other proteins in a phosphorylation-dependent way and
function as molecular scaffolds controlling the conformation of their binding
28
partners [76, 77]. This chaperone protein modifies the DNA binding domain
of FoxO and reduces the DNA-binding activity [78]. 14-3-3 binds to the
FoxO factors inside the nucleus and induces an active export of FoxO out to
the cytosol. This probably occurs by exposing FoxOs nuclear export sequence
[79]. The nuclear localization signal of FoxO is also affected by the binding of
14-3-3 [71] leading to reduced re-entry into the nucleus. A negative charge in
the basic stretch of residues that forms the nuclear localization signal is
introduced when the second site (S256 in FoxO1) is phosphorylated which
prevents FoxO from returning to the nucleus [80]. This leads to increased
FoxO levels in the cytoplasm due to nuclear export and a decrease in the
amount of FoxO returning to the nucleus. Mutations of T32 and S253 in
FoxO3 (correspond to T24 and S256 in FoxO1 human sequences), the two
sites that 14-3-3 proteins bind to, promote nuclear localization of FoxO3 and
an increase in transcriptional activity [73]. In a FoxO1 mutant where the
nuclear export sequence had been disrupted FoxO1 was still inhibited by the
PI3K-AKT/SGK/ pathway but resided inside the nucleus instead of in the
cytoplasm [81].
When Akt phosphorylates FoxO1 at S256 a binding site for S-phase kinaseassociated protein 2 (Skp2) [67], an oncogenic subunit of the Skp1/Cul1/Fbox protein ubiquitin complex [82], is also created [67]. Skp2 decreases the
level of FoxO1 through degradation [82]. FoxO1 can also be phosphorylated
by SGKs at S319, SGK becomes activated by the PI3K pathway the same as
for Akt [68].
The response to acetylation of transcription factors depends on which
functional domain gets acetylated. FoxO1 has three different acetylation sites
which are positioned within the wing 2 region on the C terminus of the
forkhead domain. This region partakes in DNA recognition and stabilization
29
of the FoxO DNA complex and acetylation of positively charged lysine
residues in wing 2 could inhibit FoxO binding to DNA [64]. Acetylation of
FoxO1 appears to decrease the DNA binding capability but also seems to
induce increased phosphorylation on S253. It appears that acetylation and
phosphorylation influence each other to control FoxO1s function [64, 83].
p300 and CREB-binding protein (CBP) are histone acetyltransferase (HAT)
proteins which have an intrinsic acetyltransferase activity that transfers an
acetyl group to specific lysine residues on target proteins such as FoxO [8385]. Transfection of rat soleus muscle with a dominant-negative p300 (lacks
HAT activity and inhibits endogenous p300 HAT activity) leads to an
increase in FoxO activity and increased transcription of MAFbx/atrogin-1.
HAT activity increased after transfection of wild-type p300 or wild-type CBP
which leads to a decrease in FoxO activation in vivo as a response to muscle
disuse [86]. An increase in HAT results in a decrease in FoxO3 nuclear
localization whereas p300 appears to give an increase in FoxO1 nuclear
localization.
Evidence for FoxOs being involved in the regulation of muscle mass include
an observed decrease in the nuclear content of FoxO1 in human quadriceps
muscle after resistance training associated with muscle growth and then during
a de-training period the amount of FoxO1 increases in the nucleus [87]. After
unilateral denervation of rat diaphragm, the amount of nuclear FoxO1 was
significantly increased after 1 day but decreased with time [88]. Removal of
growth factors and starvation of C2C12 myotubes for 6h led to myotube
atrophy, reduced phosphorylation of FoxO1 and FoxO3, an increase in FoxO
binding to DNA and increased expression of MAFbx/atrogin-1 [40].
Constitutively active FoxO1 did not, however, increase the expression of
MAFbx/atrogin-1 or MuRF1 in myotubes [39]. A decrease in muscle mass
occurs when human FoxO1 is overexpressed in mouse skeletal muscle [65] but
30
transgenic mice overexpressing FoxO1 do not have consistent alterations in
MAFbx/atrogin-1 or MuRF1 levels [65]. In the presence of FoxO1 the
glucocorticoid concentration needed for activation of MuRF1 transcription
was, however, strongly reduced [37].
Transfection of constitutively active FoxO3 increases MAFbx transcription in
C2C12 myotubes and induces muscle fiber atrophy in mouse anterior tibial
muscle [40]. A dominant negative FoxO3 prevents immobilization induced
increases in MAFbx/atrogin-1 and MuRF1 promoter activities in rat soleus
muscle as well as muscle fiber atrophy [89, 90].
Hibernating mammals suppress protein synthesis for energy conservation.
Skeletal muscles require activation of the mTORC1 complex for growth [4]
and during torpor, Akt phosphorylation is suppressed relative to summer
active animals [91]. No evidence suggest an increase in transcription or a
decrease in phosphorylation of FoxO1 or FoxO3 [91] whereas in mice and
rats with disuse-induced atrophy the FoxO transcription factors are activated
and seem to be important mediators of the atrophy response [40, 89]. The
expression levels of MuRF1 and MAFbx/atrogin-1 in quadriceps muscles
were decreased in hibernating compared to summer active thirteen-lined
squirrels [91].
NFκB and TNFα
Nuclear factor kappa B, NFκB, is a transcription factor and is suggested to
mediate muscle wasting and cachexia caused by tumor necrosis factor α,
TNFα, and other inflammatory cytokines [92]. NFκB exists in an active and
an inactive state. In the inactive state NFκB is located in the cytoplasm bound
to inhibitory proteins IκB. IκB will in response to TNFα become
phosphorylated which results in ubiquitination and proteasomal degradation
31
which frees NFκB for nuclear translocation and activation of gene
transcription. Treatment with TNFα causes up-regulation of MuRF1 and
MAFbx/atrogin-1 [92].
A member of the TNF superfamily is also TNF-like weak inducer of
apoptosis, TWEAK that has also been found to induce muscle atrophy.
TWEAK binds to fibroblast growth factor-inducible 14, Fn14, which is a
cell-surface receptor that is up-regulated in denervated muscle which leads to
NFκB activation and MuRF1 expression. The level of Fn14 does not increase
in all types of muscle atrophy such as dexamethasone treatment [92].
p38, MK2 and heat shock proteins
p38 is a mitogen-activated protein kinase (MAPK) and has four different
isoforms p38α,β,γ and δ [93]. p38α (MAPK14 also called CSAIDs binding
protein (CSBP) and SAPK2a) has an essential role in myogenesis [94]. p38β
(MAPK11 also called SAPK2b and p38-2) overexpression up-regulates the
E3 ubiquitin ligase MAFbx/atrogin-1 to cause loss of muscle mass [95]. p38γ
(MAPK12 also called SAPK3 and ERK6) is involved in mitochondrial
biogenesis and angiogenesis in response to endurance exercise [96]. p38δ,
(MAPK13 also called SAPK4) and p38γ have low kinase activity towards the
substrate MK2 [97-100]. The p38 α and β isoforms are also involved in
muscle differentiation [49]. After activation by phosphorylation the p38
isoforms phosphorylate serine/threonine residues in their substrates [49].
MK2 is a target of activated p38 (mainly α and possibly β) and has been
suggested to be involved in regulating mRNA stability, chromatin remodeling,
cell cycle regulation, cell migration etc. [50]. MK2 may also be the main
protein mediating nuclear export of p38 [51]. MK2 is phosphorylated by p38
at two sites, T205 and T317 in mouse (T222 and T334 in human) [101, 102].
Both phosphorylation sites are believed to be important for activation of
32
MK2, it is believed that phosphorylation on T317 might serve as a switch for
MK2s nuclear import and export [103]. An auto-inhibitory helix is released
from the core of the kinase domain on MK2 when it becomes phosphorylated
at T317 thereby exposing the nuclear export signal which allows MK2 to leave
the nucleus in a complex with p38 [104, 105]. However, it has also been
proposed that MK2 can leave the nucleus without p38 but still reliant on its
phosphorylation [106].
p38 phosphorylation (activation) is associated with muscle growth as a
response to functional overload and mechanical stimuli such as stretch and
exercise [107-112]. In fast twitch muscles of rats (epitrochlearis and extensor
digitotium longus) an increase in phosphorylation of p38 MAPK has also
been shown to occur after intermittent tetanic stimulation [113]. p38
activation (phosphorylation) has, however, also been found in different
atrophy models such as cast immobilization [114], denervation [115] and
hind-limb unloading [116]. Gastrocnemius muscle displays increased MuRF1
expression and elevated phosphorylation of p38 after cast immobilization in
rats. L6 rat skeletal myoblasts had an increase in MuRF1 expression after
serum starvation and this response decreased after inhibition of p38. Serum
starvation also induced size changes of L6 myoblasts and this was reversed
after transfection of MuRF1 siRNA or treatment with a p38 inhibitor [114].
TNF-α has also been suggested to increase MAFbx/atrogin-1 gene expression
in skeletal muscle by acting via p38 [117].
MK2 might have a role in the regulation of muscle mass together with its upstream activator p38 [48]. Hsp25/27 is an established substrate of MK2 in the
cytoplasm [118]. Murine Hsp25 is the homolog to Hsp27 in human, both are
functionally similar and share >80% homology at the amino acid level [119].
33
Heat shock proteins are a group of proteins believed to have a major role in
the maintenance of muscle homeostasis, especially during adaptation to
various stressors They help cells to survive stressful situations such as oxidative
stress, inflammation and muscle damage [52-54]. This group of proteins
includes both large, e.g. Hsp70, and small, e.g. Hsp25/27 proteins. In skeletal
muscle Hsp25/27 and Hsp70 are heavily expressed [52, 120]. Increased
Hsp25/27 phosphorylation is associated with skeletal muscle hypertrophy
whereas decreased phosphorylation occurs in atrophy [111, 121, 122].
Spinal cord isolation, an inactivity model, was used in rats to study Hsp25
total protein levels in soleus, plantaris, adductor longus and anterior tibial
muscles. After 7 days the level of Hsp25 was unchanged in anterior tibial
muscle. In soleus, plantaris and adductor muscles the levels of Hsp25 were
lower compared to control animals [120]. Skeletal muscle disuse atrophy in
rats has been shown to decrease after overexpression of Hsp27 [119].
Functional overload of soleus and plantaris muscles in rats is associated with
an increase in Hsp 25 expression and phosphorylation [111, 122]. After
functional overload in rats Hsp25 mRNA showed a time dependent increase
in both soleus and plantaris muscles. At three and seven days of functional
overload the expression of Hsp25 protein and phosphorylated Hsp25 in the
soluble fraction was increased in both muscle types but higher in plantaris.
Hsp25 was increased after three and seven days in the insoluble fraction in
both muscles but phosphorylated Hsp25 increased in the plantaris muscle at
day seven. The plantaris muscle had an increase in both p38 and
phosphorylated p38 at both time points whereas in the soleus muscle only
phosphorylated p38 increased at day seven [111]. Expression level and
phosphorylation of Hsp25 increased in mouse plantaris muscle 3 and 7 days
34
after functional overload and in soleus muscle 7 days after functional overload
compared to controls. [123].
Hsp70 is one of the proteins that has been studied the most in skeletal muscle,
both in human and animal models, it responds to acute and chronic changes in
muscle activity and loading [124, 125]. Mice and rats with functional overload
of plantaris and soleus muscles showed high levels of Hsp70 after 3 and 7 days
compared to control animals [123].
The soleus muscle and the plantaris muscle in the rat consist of predominantly
slow fibers respectively fast fibers and the soleus muscle has higher levels of
Hsp70 compared to plantaris [126]. Hsp25 also seem to be highly expressed in
rat skeletal muscles containing mainly slow type 1 muscle fibers [120]. This
difference was not observed in mice [123]. Hsp70 is expressed in rat soleus
and plantaris muscles at embryonic day 22 in slow type I fibers. The level of
Hsp70 in soleus muscles increased from embryonic day 22 to postnatal day 56
in parallel with the increase in the type I MyHC isoform. In the plantaris
muscle, only a small increase could be detected [52]. In plantaris muscles of
rats exposed to functional overload the size of all fiber types increase, the levels
of Hsp70 increase, and, the relative percentage of slower fiber types and
MyHC isoforms also increase [127].
Several models of muscle atrophy such as hind-limb unloading and tail
suspension show a significant down regulation of Hsp70 [128-131] and this
down regulations seems to last for up to nine weeks in a disuse model in rats
[131]. Hsp70 seems to be able to inhibit FoxO3 and NFκB signaling which
can help to prevent atrophy. This inhibition of FoxO3 resulted in a decrease in
the promotor activity for MAFbx/atrogin-1 and MuRF1 in rats in an
immobilization model [90].
35
Glucocorticoids
Glucocorticoids are steroid hormones and include cortisol, corticosterone and
cortisone which are secreted from the adrenal cortex. Glucocorticoids
influence metabolism in different tissues and skeletal muscle is a major target
tissue where glucocorticoids mainly regulate protein and glucose metabolism.
Glucocorticoids cause increased protein degradation and decreased protein
synthesis mainly by acting on the intracellular glucocorticoid receptors, GR
[56]. Amino acid transport into the muscle is inhibited by glucocorticoids,
which decreases protein synthesis. Glucocorticoids can also inhibit the
stimulatory effect of insulin, IGF-1 and amino acids on the phosphorylation
of 4EBP1 and p70S6K1, two factors involved in the control of protein
synthesis [15]. An increased muscle proteolysis through the activation of the
ubiquitin proteasome and lysosomal systems is thought to play a major part in
the catabolic action of glucocorticoids. The expression levels of atrogenes such
as MuRF1 and MAFbx/atrogin1 are increased following glucocorticoid
stimulation [15]. The expression of FoxO1 and FoxO3 in skeletal muscle is
also increased by glucocorticoids [37, 132]. Several GR binding regions are
present in or near the FoxO3 genomic region indicating that FoxO3 is a
primary glucocorticoid target [133, 134].
Younger adult rats have a faster recovery time than older after glucocorticoid
induced atrophy, a reason behind this could be that glucocorticoids cause
mainly an increase in protein breakdown in younger adult animals but also a
depressed protein synthesis in aged animals [135]. Fast-twitch, glycolytic
muscle fiber atrophy, demonstrated by decreased fiber cross-sectional area and
reduced myofibrillar protein content is characteristic for glucocorticoidinduced muscle atrophy [15].
36
Akt-Protein kinase B
Akt is suggested to block the up-regulation of MAFbx/atrogin-1 and MuRF1
through negative regulation of FoxO transcription factors [39, 40]. FoxO
proteins are phosphorylated by Akt which leads to FoxO being exported out
of the nucleus to the cytoplasm. Akt is also involved in protein synthesis (see
below).
Protein synthesis
p70S6K1 and rpS6
p70S6K1 is primarily located in the cytoplasm [136] and has three rapamycin
sensitive phosphorylation sites T220, T389 and S404 where T389 seems to be
critical for kinase activity [137-139]. rpS6 is a substrate of p70S6K1 and is
phosphorylated in the specific order of S236, S235, S240, S244 and S247
[140]. Hind-limb unloading in rats causes atrophy and a decrease in
phosphorylation of rpS6 [121] while synergist ablation, which causes
hypertrophy, increases phosphorylation of rpS6 in rats [121, 141, 142]. Mice
deficient in rpS6 phosphorylation have decreased muscle mass and decreased
abundance of contractile proteins [143]. Skeletal muscle atrophy occurs in
mice that lack the gene for p70S6K1 [144]. Interestingly, the life span of
female mice was increased with 19 % in p70S6K1
-/-
mice but for male mice
there was no significant difference in life span [143].
The eIF4 family
Eukaryotic initiation factor 4, eIF4, consists of several subunits; eIF4A, eIF4E
and eIF4G which together make up the eIF4F translation initiation complex
with the task to catalyze recognition, unwinding and binding of mRNA to the
43S preinitiation complex (Figure 4) [145]. The number of eIF4F complexes
formed depends on the availability of free eIF4E and on phosphorylated
37
eIF4G. In nutrient-poor conditions 4EBP1 is unphosphorylated and binds to
eIF4E thereby decreasing the amount of eIF4F complexes formed. In growth
promoting conditions 4EBP1 becomes phosphorylated by mTOR which
releases eIF4E to participate in the formation of eIF4F complexes [5] leading
to increased protein synthesis [146, 147]. eIF4B helps in the binding of the
ribosome to the mRNA. Without eIF4B being present, the 48S initiation
complex can still be formed which suggests that eIF4B only has an assisting
role [148]. Formation of the 43S preinitiation complex seems not to increase
at feeding [145] but assembly of active eIF4F increases with acute provision of
nutrients [145, 149]. eIF4F is necessary for cap dependent translation [150,
151].
The control of mRNA translation plays a critical role in cell growth,
proliferation and differentiation. mRNA is translated in a cap-dependent
manner in most eukaryotes. The cap structure consists of m7GpppN, where N
can be any nucleotide, and is located at the 5’ terminus [46]. In addition to
being a rate-limiting factor for the formation of the eIF4F complex eIF4E is a
mRNA 5’ cap-binding protein [46]. eIF4A, an ATP-dependent helicase, and
eIF4G, a large scaffolding protein, are also part of the eIF4F complex. eIF4G
is the docking site for other proteins [46].
38
Figure 4. The 43S preinitiation complex is formed by the 40S ribosomal subunit and Met-tRNA.
Formation of the 48S initiation complex occurs with the help of eIF4 factors. The 60S ribosomal
subunit is then attached to form the 80S complex for protein synthesis.
eIF4G
eIF4G is important for translation initiation where it functions as a scaffold
protein [152] for eIF4E, eIF4A and the mRNA. Binding of eIF4G to eIF4E
appears to accelerate the mRNA translation initiation [153]. eIF4G has
binding sites for eIF4E, eIF4A and eIF3 making eIF4G appear as a nucleus,
around which the initiation complex will form which is vital for promoting
ribosome recruitment to the mRNA [153]. eIF4G is a phosphoprotein and
delivers the 43S preinitiation complex to the 5’ cap of RNA molecules by
interacting with eIF4E, eIF4A, poly(A)-binding protein (PABP), mitogen
activated protein kinase-interacting kinases (Mnks) and eIF3 [145, 154, 155].
eIF4G has 30 identified serine or threonine phosphorylation sites.
Phosphorylation of eIF4G at S1108 is associated with increased protein
translation and the phosphorylation is promoted by IGF-1, insulin and serum.
mTOR is suggested to control the phosphorylation of eIF4G at S1108 since
39
rapamycin and starvation decrease the phosphorylation [5]. Sepsis also affects
protein synthesis in a negative way and reduces the phosphorylation of S1108.
S1108 is located in the C-terminal of eIF4G and Mnks bind near S1108 [5].
Mnk2 has recently been shown to inhibit eIF4G [5]. Knockdown of Mnk2 in
cultured myotubes increases the phosphorylation of eIF4G and overrules the
inhibitory effect of rapamycin on eIF4G. Mice lacking Mnk2 had an increase
in phosphorylation of eIF4G at S1108 in gastrocnemius muscle, the increase
was stable during atrophy conditions and upon starvation in the null-Mnk2
mice. Increased Mnk2 mRNA expression was also reported in gastrocnemius
muscle 4 days following denervation and this was associated with a decrease in
S1108 phosphorylation of eIF4G. The effect of Mnk2 on eIF4G was
suggested to be indirect and mediated through a pathway involving serinearginine-rich protein kinase (SRPK) [5].
4EBP1 and eIF4E
4EBPs are small heat-stable proteins which inhibit cap-dependent translation.
There are three 4EBPs; 4EBP1, 4EBP2 and 4EBP3 [46]. 4EBP1, has several
phosphorylation sites but only four (T37, T46, S65 and T70) are considered
to be important for release of eIF4E from the complex it forms with 4EBP1
[156]. 4EBP1 reduces translation initiation by binding to eIF4E and this is a
rate limiting step for translation [146, 148]. In the hypophosphorylated state
4EBP1 binds strongly to eIF4E but the binding is weakened when 4EBP1 is
hyperphosphorylated (Figure 5). Free eIF4E, that binds to eIF4G, increases
by extracellular stimuli such as amino acids, hormones, growth factors etc.
which increase phosphorylation of 4EBP1 [148]. C2C12 myotubes that are
nutrient-deprived and are given insulin or IGF-1 will have an increase in
protein synthesis and a dose-dependent phosphorylation of 4EBP1 [157].
40
Figure 5. 4EBP1 forms a complex with eIF4E, the complex dissociates after 4EBP1 gets
phosphorylated at several sites which makes it possible for eIF4G to bind to and form a complex
with eIF4E which will lead to increased protein synthesis.
The interaction between 4EBP1 and eIF4E has been studied in hibernating
Golden-mantled ground squirrels using Western blot. The regulation of
eIF4E by 4EBP1 differed with the different seasons. 4EPB1 was
hyperphosphorylated during the summer, which promotes initiation and the
activity of eIF4E was controlled through direct phosphorylation. In the
winter, when the squirrel was in torpor, 4EBP1 was hypophosphorylated
which lead to restricted translation through regulation of the availability of
eIF4E [158].
eIF4F, sepsis and leucine
Leucine is a branched-chain amino acid that can by itself account for most of
the stimulation of protein synthesis caused by a mixture of amino acids.
Leucine is involved in postprandial stimulation of muscle protein synthesis
and functions as an important nutritional signal. After short term starvation
followed by amino acid refeeding an increase in muscle protein syntheses will
41
occur [150]. Leucine-induced phosphorylation of 4EBP1 and p70S6K1 gives
an increase in eIF4F-complexes [150] but also an increase in rpS6
phosphorylation which leads to an increase in protein synthesis [159].
A diet of high leucine and protein concentration was introduced to rats and
accessible for 3 hours. After 0.5, 1, 3, 6 and 9 hours post meal introduction the
gastrocnemius muscle was investigated and an increase in eIF4G-eIF4E
complex was found which returned to normal 3 hours post meal introduction
[145].
The normal protein synthesis response to leucine is impaired by sepsis and
endotoxins. Sepsis results in increased secretion of TNFα and glucocorticoids,
these will also impair protein synthesis. Sepsis gives an increase in muscle
protein degradation and a decrease in muscle protein synthesis which leads to
muscle catabolism [150, 159].
Mammalian Target of Rapamycin
mTOR is a member of the family phosphatidylinositol kinase-related protein
kinases (PIKK) with a carboxy-terminal region that has a sequence similar to
the catalytic domains of PI3-kinases. Despite this homology to lipid kinases it
acts as a serine/threonine protein kinase [45, 46]. Rapamycin inhibits mTOR
signaling and reduces muscle hypertrophy [160]. Rapamycin, also known as
Sirolimus, is used clinically as an immunosuppressive drug and is an inhibitor
of the mTORC1 complex [161, 162]. Rapamycin causes G1-phase arrest in
yeast cells and in mammalian lymphocytes but in other cell types rapamycin
rather decreases cell cycle progression instead of blocking it [162]. There are
two TOR proteins, TOR1 and TOR2, which form complexes TORC1
(associated with raptor) and TORC2 (associated with rictor). These two
proteins were first discovered in yeast but later one was cloned in mammals
42
and is known as mTOR or FKBP12-rapamycin-associated protein (FRAP,
RAFT or RAPT) [46, 162]. Nutrient availability controls the activity of TOR
in yeast; in mammals both growth factors and nutrients control TOR [45,
151]. mTOR becomes active in nutrient-rich conditions and inactive in
nutrient poor conditions [163]. A stress response program is triggered in yeast
by TOR gene depletion or rapamycin exposure, this response is similar to the
nutrient starvation phenotype in yeast [45, 151].
Growth factors, nutrients and certain hormones induce anabolic cellular
processes, such as ribosomal gene transcription, protein synthesis, cell growth
and cell proliferation, when they signal through mTORC1 [161]. mTORC1
and mTORC2 share some proteins but also contain unique ones, that results
in unique cellular functions but also different sensitivity to rapamycin where
mTORC1 is the more sensitive complex compared to mTORC2 [151].
Raptor, regulatory associated protein of mTOR, functions as a scaffolding
protein between mTOR kinase and mTORC1 substrates to promote
mTORC1 signaling. The binding of raptor and mTOR is weakened by
rapamycin [151, 161].
Akt activates mTOR indirectly by phosphorylation of TSC2 in the
TSC1/TSC2 heterodimer, which otherwise inhibits mTOR [164]. This starts
a chain reaction where downstream factors of mTOR become phosphorylated
[156, 165]. The phosphorylation of 4EBP1 and p70S6K1 leads to release of
eIF4E and phosphorylation of rpS6 which correlates with an increase in
translation of 5’ terminal oligopyrimidine tract, 5’TOP, containing mRNAs
that encode poly (A) binding proteins, ribosomal proteins and elongation
factors [46]. 4EBP1 and p70S6K1 contain TOR signaling (TOS) motifs,
mammalian cell size is controlled through these two substrates and
downstream pathways [45, 162, 166]. These two substrates are bound directly
43
to raptor which links them with mTOR kinase [151]. 4EBP1 and p70S6K1
become rapidly dephosphorylated when cultured mammalian cells are
transferred to a medium without amino acids and glucose. By nutrient
restoration mTOR multiphosphorylates 4EBP1 resulting in release of eIF4E
and increased protein synthesis. Nutrients will also rephosphorylate p70S6K1
by mTOR and increase translational capacity by stimulating ribosome
biogenesis [167]. Rapamycin inhibits overall protein synthesis, translation of
mRNA and 4EBP1 phosphorylation independent of p70S6K1 activity [168].
The rapamycin-sensitive and mTOR-regulated pathway has been studied with
regard to p70S6K1 and 4EBP1, it was found that ribosomal biogenesis was
regulated through p70S6K1 and the overall translation initiation rate through
4EBP1 [168].
GSK3β and eIF2B
GSK-3β is a downstream target of Akt [169] and inactivates eukaryotic
initiation factor 2B, elF2B, by phosphorylation. eIF2B is both important for
increased protein synthesis and ribosome recycling. Akt phosphorylates S9 on
GSK-3β and thereby inhibits its functions, eIF2B becomes activated when
GSK-3β is phosphorylated and an increase in protein synthesis occurs [44,
47]. Response from growth factors and insulin will inhibit GSK-3β through
serine phosphorylation [169]. Y216 is a tyrosine site where GSK-3β can also
be phosphorylated by an unidentified tyrosine kinase in mammals; it has been
identified in slime molds as Zak1 [2, 44, 47, 170].
Akt-Protein kinase B
Akt has three isoforms; Akt1, Akt2 and Akt3 which are encoded by the three
genes PKBα, PKBβ and PKBγ [171]. The three genes have a homology of
more than 85% sequence identity, and share the same structural organization
44
[172]. Akt2 and Akt3 have 40 respectively 25 amino acids more than Akt1 in
the C-terminal domain [172, 173]. To a certain extent each Akt isoform has
distinctive functions which have been evaluated with the help of murine gene
disruption [171]. Akt1 seems to be involved in growth compared to Akt2
which has been suggested to have a major role in glucose metabolism.
Evidence to support this has been shown in studies on mice lacking Akt1 or
Akt2 [174, 175]. Akt1 and Akt2 are both involved in skeletal muscle protein
synthesis and degradation. Skeletal muscles become hypertrophic when Akt1
is overexpressed [176] but both Akt1 and Akt2 are suggested to be involved in
muscle growth and size [177] and are expressed in most tissues [178]. Akt3 is
primarily found in the brain and testes but can also be found to a lesser extent
in mammary gland, lung and fat [178].
IGF-1 and insulin trigger the activation of the Akt pathway by activating
phosphatidylinositol 3-kinase, PI(3)K. PI(3)K generates phosphatidylinositol
triphosphate, PIP3, in the cell membrane which binds to Akt and translocate
Akt to the plasma membrane where Akt needs to be phosphorylated at both
S473 and T308 for full activation [179, 180].
Akt is a serine/threonine kinase and is thought to play an important role in
controlling apoptosis and cell survival through phosphorylation and inhibition
of factors such as FoxO [171]. Akt phosphorylates FoxO1, FoxO3 and FoxO4
which results in nuclear export and transcriptional inactivity of FoxO [39].
In resistance exercise and weight-bearing locomotion following a period of
disuse an increase in mechanical loading occurs. This leads to increased
mTORC1 complex formation and activation with an increase in protein
translation and ribosome biogenesis that result in adaptive muscle hypertrophy
[181]. A decrease in activation of Akt and mTORC1 occurs as a result of
45
disuse induced by immobilization or hind-limb unloading in young adult
rodents [160, 182].
In models of skeletal muscle hypertrophy, such as functional overload of the
rat or mouse plantaris muscle, the levels of S473 phosphorylated Akt is
increased [160]. In atrophy models based on skeletal muscle inactivity, such as
10 days of hind-limb immobilization or 10–14 days of hind-limb suspension,
Akt S473 phosphorylation has been reported to be decreased in rat medial
gastrocnemius muscle [160] and soleus muscle [183-186] but not in rat
extensor digitorum longus muscle [184]. Constitutively active Akt has been
shown to inhibit atrophy of denervated anterior tibial muscle in mice and
denervated soleus muscle in rats [160, 187].
46
4. AIM AND OBJECTIVES
The overall aim of this thesis was to investigate changes in expression and post
translation modifications of some factors involved in the regulation of protein
synthesis and protein degradation in 6-days denervated atrophic and
hypertrophic mouse skeletal muscle.
Paper I
Paper I examines the hypothesis that Akt/mTOR signaling is increased in
hypertrophic and decreased in atrophic denervated muscle. Protein expression
and phosphorylation of Akt1, Akt2, GSK-3β, 4EBP1, p70S6K1 and rpS6
were examined in six-days denervated mouse anterior tibial (atrophic) and
hemidiaphragm (hypertrophic) muscles.
Paper II
The purpose of paper II was to investigate FoxO1 protein expression,
phosphorylation and acetylation as well as MuRF1 protein expression in
atrophic (anterior tibial and pooled gastrocnemius and soleus) and
hypertrophic (hemidiaphragm) 6-days denervated mouse skeletal muscle.
47
Paper III
Paper III examines the hypothesis that the differential response of
hemidiaphragm (hypertrophy) and hind-limb muscles (atrophy) to
denervation is related to a differential nuclear/cytosolic localization of p38
and/or MK2. The expression and phosphorylation of p38, MK2 and related
proteins were studied in cytosolic and nuclear fractions from 6-days
denervated atrophic anterior tibial muscle and hypertrophic hemidiaphragm
muscle. Similar studies were also performed on unfractionated homogenates of
pooled gastrocnemius and soleus muscles.
Paper IV
The aim of paper IV was to test the hypothesis that S1108 phosphorylation of
eIF4G is differentially affected in 6-days denervated atrophic hind-limb
muscles (pooled gastrocnemius and soleus muscles) and 6-days denervated
hypertrophic (hemidiaphragm) muscle.
48
5. METHODOLOGICAL
CONSIDERATIONS
Animals
The animals used in this thesis were adult male NMRI mice with an
approximate weight of 30g. The reason for using males rather than females is
that females have a hormonal cycle that may affect the results. Animals were
housed with environment enrichment and with free access to a standard
laboratory diet and tap water for a week before experiments so they became
familiar with the surrounding. The studies on protein expression were
performed 6-days after denervation. The reason for choosing this time point is
because of the hypertrophic peak in denervated hemidiaphragm before the
hemidiaphragm becomes atrophic [34-36]. All the experiments have been
approved by the Ethical Committee for Animal Experiments, Linköping,
Sweden.
Surgery and muscles
The animals were anaesthetized by inhalation of isoflurane and received a
subcutaneous injection of buprenorphine (50 μg/kg) for post-operative
analgesia. The atrophic muscles studied in this thesis were anterior tibial and
pooled gastrocnemius and soleus muscles. These muscles were denervated by
49
sectioning and removing a few mm of the sciatic nerve and the wound was
closed with a metallic clip [188]. Lidocain was applied onto the sciatic nerve
before removing a part of it to prevent neural activity and autotomy [189]. To
obtain a hypertrophic muscle the hemidiaphragm was denervated by a
unilateral thoracotomy and then a few mm of the left phrenic nerve was
removed and the wound was closed with metallic clips (see Table I). Eight
randomly selected animals were used in each group (denervated
hemidiaphragm muscle, denervated anterior tibial muscle, denervated pooled
gastrocnemius and soleus muscles). For hemidiaphragm studies eight animals
went through sham surgery. These animals were anaesthetized by inhalation
of isoflurane, had a subcutaneous injection of buprenorphine and a unilateral
thoracotomy without touching the phrenic nerve. The mice were killed six
days after denervation or sham surgery by cervical dislocation, muscles were
dissected (hemidiaphragm, anterior tibial and gastrocnemius together with
soleus), weighed, frozen on dry ice and stored at -80o C. Innervated left
control hemidiaphragms were collected from eight separate animals that had
received no surgery. Innervated control hind-limb muscles (anterior tibial and
pooled gastrocnemius and soleus muscles) were collected from the
contralateral (right) leg of animals that were denervated by sectioning the left
sciatic nerve. An additional eight animals for each of denervated and
innervated hemidiaphragm muscles and innervated/denervated anterior tibial
muscles were used for studies on separated nuclear and cytosolic fractions.
50
Table I. Different muscle types that were investigated in this thesis with the corresponding nerve
that has been sectioned and which type of response that occurs in the different muscles.
Muscle
Nerve
Left/right
Fiber
side of
types
Atrophy/hypertrophy
animal
Anterior tibial
Sciatic
Left
II
Atrophy
Gastrocnemius and
Sciatic
Left
I,II
Atrophy
Phrenic
Left
I,II
Hypertrophy
soleus (pooled)
Hemidiaphragm
Standard protein extraction Papers I-IV
Muscles were homogenized using an Ultra-Turrax homogenizer (Janke and
Kunkel, Staufen, Germany) in 1 ml (hemidiaphragm and anterior tibial
muscles) or 2 ml (pooled gastrocnemius and soleus muscles) buffer containing
100 mM Tris-HCl, pH 7.6, 150 mM NaCl, 1 mM EDTA, 1% NP-40, 0.1%
sodium deoxycholate and 1% HaltTM Protease and Phosphatase Inhibitor
Cocktail (Thermo Scientific, Rockford, IL). The supernatant was recovered
and the pellet was resuspended in 0.5 ml (anterior tibial and hemidiaphragm
muscles) or 1.0 ml (pooled gastrocnemius and soleus muscles) of
homogenization buffer and recentrifuged. The supernatants were combined
and the protein concentration was determined using the Bradford assay [190].
Nuclear and cytosolic fractions Papers II and III
Hemidiaphragm and anterior tibial muscles were used for cytosolic and
nuclear protein extraction. The method used was slightly modified from [191].
The muscles were homogenized using an Ultra-Turrax homogenizer (Janke
and Kunkel, Staufen, Germany) in 1 ml low salt lysis buffer (10 mM HEPES,
10 mM KCl, 1.5 mM MgCl2, 0.1 mM EDTA, 0.1 mM EGTA, 1 mM
51
dithiothreitol (DTT); pH 7.9 with 1% HaltTM Protease and Phosphatase
Inhibitor Cocktail from Thermo Scientific, Rockford, IL). The homogenized
tissue was vortexed for 15 s, put on ice for 10 min, vortexed again for 15 s and
centrifuged at 16.000 g for 15 s. The supernatant cytosolic extract was
immediately frozen (-80o C) for subsequent analyses. The nuclear pellet was
resuspended on ice in a high salt nuclear extraction buffer (20 mM HEPES,
420 mM NaCl, 1 mM EDTA, 1 mM EGTA, 1 mM DTT, 25% glycerol; pH
7.9 with 1% HaltTM Protease and Phosphatase Inhibitor Cocktail from
Thermo Scientific, Rockford, IL) at a ratio of 4 μl of nuclear extraction buffer
per mg muscle wet weight. Preparations were incubated on ice for 30 min and
vortexed for 10 s every 5 min before being centrifuged at 16.000 g for 6 min.
The supernatant nuclear extract was then removed and frozen (-80o C) for
subsequent analyses. Protein concentrations for each fraction were obtained
using the Bradford assay [190]. The author of the present thesis did not
actively participate in this part of papers II and III.
Western blotting
Western blot is a widely used method in research which makes it possible to
fractionate
proteins
by
sodium
dodecyl
sulfate
polyacrylamide
gel
electrophoresis (SDS-PAGE), transfer onto a polyvinylidene difluoride
(PVDF) membrane, detect with antibodies and semi-quantify the amount of
protein in a sample with chemiluminescence. Even proteins in low abundance
can be detected with this method. The different antibodies used in the present
thesis are shown in Table II.
52
Table II. Antibodies used in the present thesis.
Antibody
Site
Dilution
Paper
Ab Supplier
Akt1
-
1:2000
I
2967+
pAkt1
S473
1:2000
I
07-310**
Akt2
-
1:2000
I
2962+
pAkt2
S474
1:2000
I
38513++
GSK-3β
-
1:2000
I
610202***
pGSK-3β
S9
1:2000
I
9336+
4EBP1
-
1:1000
I
9452+
p4EBP1
S65
1:1000
I
9451+
p70S6K1
-
1:500
I
9202+
pp70S6K1
T389
1:500
I
9205+
rpS6
-
1:2000
I
2317+
prpS6
S235/236
1:2000
I
2211+
FoxO1
-
1:800
II
2880+
pFoxO1
S256
1:800
II
9461+
Ac-FoxO1
-
1:2000
II
sc-49437*
FoxO3
-
1:800
unpublished
2497+
pFoxO3
S253
1:800
unpublished
9466+
MuRF1
-
1:1000
II
AF5366+++
Lamin A/C
-
1:1000
III
2032+
p38 MAPK
-
1:1000
III
9212+
pp38 MAPK
T180/Y182
1:1000
III
4511+
MK2
-
1:2000
III
3042+
pMK2
T317
1:1000
III
3041+
pMK2
T205
1:1000
III
3316+
Hsp27
-
1:1000
III
2442+
pHsp27
S82
1:1000
III
2401+
Hsp70
-
1:1000
III
4876+
SOD-1
-
1:10000
III
Sc-8637*
53
eIF4G
-
1:800
IV
2858+
peIF4G
S1108
1:800
IV
2441+
β-actin
-
1:10 000
unpublished
8227++
+
Cell signaling Technology, *Santa Cruz Biotechnology,
Solutions,
***
++
Abcam, **Upstate Cell Signaling
BD Transduction Laboratories, +++R&D Systems
Data analysis and statistics
The expression levels of total, phosphorylated and acetylated proteins were
studied semi-quantitatively using data from Western blots. Equal amounts of
total, cytosolic or nuclear proteins from innervated, sham operated or
denervated muscles were loaded on the gels. Measured levels of total,
phosphorylated or acetylated proteins are expressed without normalization to
any specific protein (loading control). Any differences in protein
quantifications, pipetting steps, protein transfers etc. are, thus, included in the
variations of the data sets.
Image analysis was performed using the gel plotting macro of the program
ImageJ (Rasband, W.S., ImageJ, US National Institutes of Health, Bethesda,
MD, http://rsb.info.nih.gov/ij/, 1997–2007). Results were obtained in
uncalibrated optical density units.
For quantification of protein expression in the different whole muscle
homogenates studied one of the innervated muscle samples was used as a
reference sample and was included in all gels. All other samples were measured
relative to this reference, the signal of which was set to 100.0. In the final
analysis all signals were again normalized in such a way that the average signal
from innervated muscles became 100.0.
54
For quantification of protein expression in separated cytosolic and nuclear
fractions one of the cytosolic fractions from an innervated muscle was used as
a reference sample and was included in all gels. All other samples were
measured relative to this reference, the signal of which was set to 100.0. From
the amount of protein loaded on gels in relation to the total amount of protein
extracted in the nuclear and cytosolic fractions a total cytosolic and a total
nuclear signal was calculated for whole muscles. In the final analysis total
cytosolic and total nuclear signals were again normalized in such a way that
the sum of the average nuclear and cytosolic signals became 100.0 in
innervated muscle.
Data are presented as mean values ± standard error of the mean (SEM). For
statistical comparisons of unfractionated hemidiaphragm muscles (innervated,
sham operated and denervated) one-way ANOVA was used followed by the
Tukey’s multiple comparisons test, for normally distributed data (according to
D’Agostino-Pearson omnibus K2 normality test). Statistical significance for
data not being normally distributed was determined using the Kruskal-Wallis
test with Dunn’s multiple comparisons test. For other comparisons Student’s
t-test (paired observations for hind-limb muscles, unpaired observations for
hemidiaphragm muscles) was used for normally distributed data (according to
D’Agostino-Pearson omnibus K2 normality test). Statistical significance for
data not being normally distributed was determined using the Mann-Whitney
test (hemidiaphragm muscles) and Wilcoxon matched-pairs signed rank test
(anterior tibial and pooled gastrocnemius and soleus muscles).
55
6. RESULTS AND DISCUSSION
Most skeletal muscles become atrophic after denervation but the
hemidiaphragm is unique since it undergoes a transient hypertrophic state
after denervation which might be a as a result of passive stretch because of the
continued contractions in the contralateral innervated hemidiaphragm [3436]. All the muscles used in papers I-IV were weighed before freezing and the
results are compiled in Table III.
Table III. Muscle wet weights for muscles used in papers I-IV. n=8 except in *n=9
Muscle
Anterior
Tibial
Hemidiaphragm
Gastrocnemius
and soleus
56
Innervated
Sham
Denervated
Paper
55.1+1.7 mg
-
44.1+1.8 mg
I
65.9+1.8 mg
60.8+1.9mg
28.8+1.4 mg
28.2+0.8 mg
29.6+0.6 mg
198.3+5.7 mg
29.7+0.9 mg
-
51.6+1.8 mg
44.7+1.8mg
44.4+0.7 mg*
43.3+0.7 mg
40.3+1.5 mg
149.1+4.4 mg
II,III,IV
II,III
I
II,III,IV
II,III
II,III,IV
Paper I
It has been proposed that the Akt/mTOR signaling pathway plays a major
role in the regulation of skeletal muscle mass. Total protein expression and
phosphorylation levels of Akt1 and Akt2 as well as the Akt substrate GSK-3β
and the downstream factors of mTOR, 4EBP1, p70S6K1 and rpS6 were
studied in 6-days denervated atrophic and hypertrophic muscles.
The results obtained in paper I are summarized in Table IV. In hypertrophic
hemidiaphragm expression of all proteins and phosphorylations were increased
except 4EBP1 total protein. No decreases in total protein expressions or
phosphorylation levels were observed in denervated atrophic anterior tibial
muscle. On the contrary total Akt1, total Akt2 and total rpS6 as well as
phosphorylated levels of Akt2, 4EBP1, p70S6K1 and rpS6 were increased in
denervated atrophic muscle. The results obtained agree with several other
studies of hypertrophic skeletal muscle which indicate increased signaling
through the Akt/mTOR pathway [87, 108, 160]. p70S6K1 and 4EBP1 with
increased phosphorylation levels have been reported previously [88] in
denervated rat hemidiaphragm muscles, increased rpS6 [192] and GSK-3β
[193] phosphorylations have also been reported previously.
In the present study no decrease was observed in signaling through the
Akt/mTOR pathway in atrophic denervated anterior tibial muscle. GSK-3β
(S9) showed no decrease in phosphorylation in atrophic muscle which has also
been reported previously [193]. Another study has also reported increased Akt
protein expression and phosphorylation levels in atrophic mouse muscle two
weeks after denervation [194].
57
The results from this paper suggest increased rather than decreased signaling
through mTOR, which indicates increased protein synthesis, in denervated
atrophic muscles as well as in denervated hypertrophic muscles. It is therefore
likely that increased protein degradation is responsible for the loss of muscle
mass in denervated atrophic muscles rather than decreased protein synthesis.
Previous studies and the present results are in line with each other indicating
increased protein synthesis in hypertrophic hemidiaphragm muscle [195] as
well as in atrophic hind-limb muscles of adult mice [33] denervated for similar
time periods as in the present study.
Akt S473 phosphorylation has been reported to be decreased in medial
gastrocnemius muscle [160] and soleus muscle [183-186] from rat but not in
extensor digitorum longus muscle [184] in atrophy models based on skeletal
muscle inactivity such as 10 days of hind-limb immobilization or 10– 14 days
of hind-limb suspension. Increased protein degradation may be more
important after denervation compared to hind-limb suspension where a
suppression of protein synthesis has been suggested to be more important for
muscle atrophy [196]. The difference in the response of GSK-3β to
denervation in atrophic anterior tibial and hypertrophic hemidiaphragm
muscles could be further studied in denervated pooled gastrocnemius and
soleus muscles to investigate whether or not the response depends on
differences in muscle fiber type composition.
58
Table IV. Changes in total protein expression and phosphorylations in 6-days denervated atrophic
anterior tibial muscle and in 6-days denervated hypertrophic hemidiaphragm. ~ No change.
Anterior tibial (atrophic)
Hemidiaphragm
(hypertrophic)
Akt1
↑
↑
pAkt1 (S473)
~
↑
Akt2
↑
↑
pAkt2 (S474)
↑
↑
GSK-3β
~
↑
pGSK-3β (S9)
~
↑
4EBP1
~
~
p4EBP1 (S65)
↑
↑
p70S6K1
~
↑
pp70S6K1 (T389)
↑
↑
rpS6
↑
↑
prpS6 (S235/236)
↑
↑
Paper II
FoxO1 total expression, phosphorylation level and acetylation as well as
MuRF1 protein expression were examined in this paper in both atrophic and
hypertrophic denervated muscle. Since anterior tibial and hemidiaphragm
muscles differ in fiber type compositions pooled gastrocnemius and soleus
muscles were also included in paper II. In mouse the hemidiaphragm muscle
consists primarily of type II fibers but also has around 12% of type I fibers
[197] whereas the anterior tibial muscle contains no type I fibers [198].
Pooled gastrocnemius and soleus muscles contain both type I and type II
59
fibers [198, 199]. Separated nuclear and cytosolic fractions of innervated and
denervated anterior tibial and hemidiaphragm muscles were also included in
paper II.
The results obtained in paper II are summarized in Table V. FoxO1 total
protein expression was increased in both atrophic and hypertrophic muscles
after 6 days of denervation. The level of phosphorylated FoxO1 was upregulated both in denervated hemidiaphragm (hypertrophic) and pooled
denervated gastrocnemius and soleus muscles (atrophic) but not in denervated
anterior tibial muscle. Acetylated FoxO1 was up-regulated in denervated
atrophic anterior tibial muscles but not in denervated hemidiaphragm muscles
(hypertrophic) nor in atrophic denervated pooled gastrocnemius and soleus
muscles. Total MuRF1 protein expression was increased in all denervated
muscles studied both in atrophic anterior tibial and pooled gastrocnemius and
soleus muscles and in hypertrophic hemidiaphragm muscle.
In denervated atrophic anterior tibial muscle total FoxO1 increased only in the
cytoplasmic fraction. A small but statistically significant increase in
phosphorylated FoxO1 was also observed in cytosolic fractions of denervated
anterior tibial muscle. In denervated hypertrophic hemidiaphragm total
FoxO1 as well as phosphorylated FoxO1 increased in both cytosolic and
nuclear fractions.
This study confirms an increased MuRF1 protein expression in denervated
atrophic hind-limb muscle and also shows that MuRF1 protein expression is
increased in denervated hemidiaphragm muscle at a time point when the
muscle is in a hypertrophic state relative to innervated control muscles.
Despite the hypertrophic state previous studies on denervated rat
hemidiaphragm indicate that from 5 days following denervation protein
60
degradation, as well as protein synthesis, is increased in this muscle [200]. The
present study shows that the expression of FoxO1 protein is increased in all 6days denervated skeletal muscles studied, atrophic as well as hypertrophic.
This may suggest that FoxO1 plays a role in denervation changes other than
those leading to alterations in muscle mass. Increased FoxO1 expression in
denervated atrophic hind-limb muscle was recently also published by others
[201]. Differences in fiber type composition of the muscles studied may be
related to the different effects of denervation on the level of phosphorylated
and acetylated FoxO1 in the different muscles studied.
FoxO3 expression has previously been reported to increase in denervated
atrophic skeletal muscle [202-204]. In the present study the ambition was to
also compare FoxO3 total protein expression and phosphorylation (at S253) in
denervated atrophic and hypertrophic muscles. Despite several attempts with
different antibodies, different dilutions of antibodies and different
concentrations of milk and bovine serum albumin in the blocking buffer no
detectable bands of correct size that could be analyzed were obtained.
Table V. Changes in total protein expression, phosphorylation and acetylation in 6-days
denervated atrophic and hypertrophic muscle. Whole muscle homogenates. ~ No change.
Muscles
Fiber
FoxO1
types
Anterior tibial
Gastrocnemius and
pFoxO1
Ac-
(S256)
FoxO1
MuRF1
I and II
↑
↑
~
↑
↑
~
↑
↑
I and II
↑
↑
~
↑
II
soleus (pooled)
Hemidiaphragm
61
Paper III
A previous study has shown that MK2 phosphorylation on T317 correlates
with muscle weight in denervated atrophic anterior tibial muscle, denervated
hypertrophic hemidiaphragm muscle and innervated control muscles [48].
Paper III examines the hypothesis that the differential response of
hemidiaphragm and hind-limb muscles to denervation is related to a
differential nuclear/cytosolic localization of p38 and/or MK2. The protein
expression and phosphorylation status of p38, MK2 and the MK2 substrate
Hsp25 as well as Hsp70 protein expression were examined in separated
cytosolic and nuclear fractions. The proteins and phosphorylations were also
examined in unfractionated homogenates of pooled gastrocnemius and soleus
muscles in order to examine hind-limb muscles that, like the hemidiaphragm,
contain type I muscle fibers in addition to type II fibers [197, 199] but like the
anterior tibial muscle become atrophic following denervation.
The results obtained in paper III are summarized in Table VI. No support was
obtained for a differential nuclear/cytosolic localization of p38 or MK2 in
denervated atrophic and hypertrophic muscle. A differential response of MK2
T317 phosphorylation was confirmed in denervated atrophic and hypertrophic
muscle and decreased T317 phosphorylation was also observed in denervated
atrophic pooled gastrocnemius and soleus muscles. Hsp70 increased
substantially only in denervated hypertrophic muscle which might suggest that
Hsp70 could be important for the differential effect of denervation on T317
phosphorylation of MK2 in denervated hypertrophic and atrophic muscle.
Hsp70 has been reported to play a role in the phosphorylation of MK2 by p38
[205]. A significant increase of Hsp25 phosphorylation in all denervated
62
muscles was observed suggesting that other factors in addition to MK2 are
important for the regulation of this phosphorylation.
Table VI. Changes in total protein expression and phosphorylations in 6-days denervated atrophic
and hypertrophic muscle. Whole muscle homogenates or separated nuclear and cytosolic fractions
+
( ).~ No change.
Anterior tibial
(atrophic)
↓+
↑cytosol+
p38
pp38
(T180/Y182)
MK2
pMK2 (T205)
pMK2 (T317)
Hsp25
pHsp25 (S82)
Hsp70
Gastrocnemius and
soleus (atrophic)
↓+
↑cytosol+
↓+
↑+
↑cytosol+
↑nuclear+
Hemidiaphragm
(hypertrophic)
~
↑
~+
↓nuclear+
↓
↑
↓
↑
↑
~
↑+
↑cytosol+
↑+
↑+
↑+
↑+
Paper IV
The eukaryotic translation initiation factor 4-gamma (eIF4G) is important for
initiation of protein synthesis and phosphorylation on S1108 correlates with
increased protein synthesis [206]. A decrease in S1108 phosphorylation of
eIF4G has been reported by Hu et al. 2012 [5] in denervated gastrocnemius
muscle. The decrease in S1108 phosphorylation was associated with an
increase in Mnk2 mRNA expression. A previous study from our lab has also
found increased Mnk2 mRNA expression in denervated hind-limb muscle but
this was not observed in denervated hemidiaphragm (unpublished results).
The present study tests the hypothesis that S1108 phosphorylation of eIF4G
is differentially affected in 6-days denervated atrophic hind-limb muscles
(pooled
gastrocnemius
and
soleus
muscles) and 6-days denervated
hypertrophic (hemidiaphragm) muscle.
63
The results obtained are shown in Table VII. The results from denervated
atrophic pooled gastrocnemius and soleus muscle show a decrease in the
phosphorylation but no change in total eIF4G expression, as also reported by
Hu et al. 2012. In denervated hypertrophic hemidiaphragm a significant
increase was found in both total eIF4G expression and S1108 phosphorylated
eIF4G, with no significant change in the ratio of phosphorylated/total eIF4G.
Table VII. Changes in eIF4G total protein expression and S1108 phosphorylation in 6-days
denervated atrophic and hypertrophic muscle. Whole muscle homogenates. ~ No change.
Gastrocnemius and
soleus (atrophic)
eIF4G
peIF4G (S1108)
peIF4G (S1108) / eIF4G
~
↓
↓
Hemidiaphragm
(hypertrophic)
↑
↑
~
The different alterations in muscle mass of hind-limb and hemidiaphragm
muscles following denervation should be possible to relate to alterations in the
regulation of muscle protein synthesis and/or degradation. The assembly of
the eIF4F-complex is an important step for the control of the initiation of
protein synthesis. This complex contains eIF4A, an RNA helicase, eIF4E
and eIF4G. eIF4G acts as a scaffold for the other components in this
complex. Phosphorylation of eIF4G S1108 is associated with increased
formation of eIF4G-eIF4E complex which correlates with increased protein
synthesis [206]. The amount of free eIF4E that can bind to eIF4G is
regulated by 4EBP1 which competes with eIF4G to bind to eIF4E [207,
208]. The results reported in paper I indicate that phosphorylation of 4EBP1
increases in denervated hypertrophic hemidiaphragm and denervated atrophic
hind-limb muscle which suggests increased availability of eIF4E in both cases
therefore allowing increased formation of eIF4F-complexes and protein
synthesis. The increased or maintained phosphorylation of eIF4G S1108 in
hypertrophic hemidiaphragm versus decreased phosphorylation in atrophic
64
hind-limb muscle may represent one regulatory mechanism that helps explain
the different responses of the hemidiaphragm muscle compared to hind-limb
muscles following denervation.
65
7. CONCLUSIONS
In this thesis different cell signaling pathways and factors involved in atrophy
and hypertrophy have been investigated in denervated skeletal muscle.
In paper I Akt and downstream factors of mTOR were investigated and the
results suggest an increase in protein synthesis in both denervated atrophic
anterior tibial muscle and hypertrophic hemidiaphragm muscle. This implies
that increased protein degradation is more likely to be responsible for the loss
of muscle mass in denervated atrophic muscle than a decrease in protein
synthesis.
In paper II FoxO1 and MuRF1 expression were shown to increase in both
denervated atrophic and hypertrophic muscles which suggest that these two
proteins participate in the tissue remodeling that occurs after denervation.
Acetylation and phosphorylation of FoxO1 after denervation differed in the
different muscles studied which could be related to differences in fiber type
compositions of the muscles.
In paper III p38 and related factors were investigated. A differential response
of MK2 T317 phosphorylation in denervated hypertrophic and atrophic
muscles was confirmed and Hsp70 was suggested to possibly be important for
66
this differential response. In all denervated muscles studied an increased
phosphorylation of Hsp25 was observed which suggests that other factors than
MK2 are important for regulating this phosphorylation.
In paper IV the S1108 phosphorylation of eIF4G was investigated in atrophic
pooled gastrocnemius and soleus muscles and hypertrophic hemidiaphragm
muscle. A decrease in S1108 phosphorylation of eIF4G was confirmed in
denervated atrophic muscles, but was not observed in denervated hypertrophic
muscle. In denervated hypertrophic hemidiaphragm muscle there was an
increase in expression of both total and S1108 phosphorylated eIF4G. The
differential effect of denervation on S1108 phosphorylation of eIF4G in
hemidiaphragm and hind-limb muscles may represent a mechanism that could
clarify the differential responses of these muscles following denervation.
All the research reported in this thesis is basic research which is an important
first step in science, but it then needs to be taken a step further so it can help
people that suffer from different conditions in which loss of muscle mass can
be devastating.
67
8. ACKNOWLEDGEMENTS
The writing of this thesis has taken more time then I could ever have
imagined. Despite the frustrations and the feeling I would never get here, I
finally made the finish line and it feels so good. There are so many people that
have made such a difference for me during this journey and I hope I haven’t
forgotten to mention anyone.
This thesis could not have been completed without the help of my Supervisor
Sven Tågerud. Sven taught me that science is at least 2 steps forward and 1.5
step backwards and that we learn more from our mistakes then our successes.
And you are right; discoveries can’t be made without mistakes. Thank you for
giving me the opportunity to research and the willingness to teach me all
about atrophy and hypertrophy; and for wanting your PhD students to be as
prepared as possible for when we stand on our own two feet.
I also would like to thank Marlene Norrby, who was a part of this research
group as a senior PhD student when I joined the group, and who today are my
co-supervisor. All the help you have given me over these years and the proof
reading has been wonderful. You are the best at “fulfixa” thank you .
A huge thanks to Marie-Louise Wadenberg, I am forever grateful to you for
the inspiration to move to Kalmar. Your knowledge, your enthusiasm for
68
research and your willingness to teach has been crucial for my journey
forwards this goal.
I am so grateful to Jonas Burén for opening my eyes to research all those years
ago in Umeå. Thank you for showing me how much fun research is, and
giving me a special opportunity and an internship in a lab. I am forever
grateful too you!
I want to especially thank Kim, present member of the research group. Thank
you for all the fun on the conferences and helping out whenever needed, but I
know you shake your head at me in the lab some days . Anna, my dear
colleague and friend you always listen and have the best advice. You have a
magical gift in getting me to say what I really think about things. THANK
YOU!
Norrgård is filled with wonderful people but there are some that needs an
extra mentioning such as Johanna, who is the best optician I know and always
helpful with my eye sight. Thanks to Camilla for being a wonderful travel
companion on your conferences, it has been great. A special thanks to Anneli
for being such a wonderful lunch companion, I am so glad that it is not just
me that is hungry before 11 am . How Norrgård would survive without
Stefan, Bosse and Mattias is for me a mystery, you always help out when we
have problems and for that I am very grateful. I also would like to thank the
Ann, Biggan and Ing-marie for making my mornings feel less lonely; it is
always fun talking with you. Nina and Annette for helping and making sure
that we have the material we need to make everything in the student lab run
smoothly. Lena, Berit, Lilita, Ymke, Per and Anna-Lena, what would I have
done without the great and prompt service you have provided that have made
my life so much easier as a PhD student.
69
A big thanks to a little boy named Max for being so nice and lending me his
mummy when I need someone to talk to. There is no one like you Solveig, and
you make my problems disappear with your solutions. And thank you Scott for
not stopping to try to teach me how electronic devices such as computers and
phones work .
Kalle and Karin, my wonderful friends, we have so much fun, great dinners,
evenings with wine, long talks and board games. Twiggy has a very special
place in this family and we are all so happy when she comes and visit and
thank you for taking so good care of Malou, she loves it.
To Susanne my wonderful “sister”, your love and enthusiasm for bearded
collies is what made us friends but today that friendship is so much more. To
have you, your husband, Torbjörn, and your three bearded collies in our life,
fills it with more love and laughs and the children love spending time with
their godparents. Thank you both for being there for us.
When I was at a crossroad and did not know which way to go, Gun and Plurre
talked some sense into me and I am very grateful for that today. I followed
your advice and it turned out great, thank you for letting me stay with you
whenever I am in Stockholm. Britt and Rickard, you are the best godparents
anyone could ever wish for and I am so happy that you are still part of my life.
70
Till min älskade farmor, en krutgumma på 95 år. Jag är väldigt övertygad om
varifrån jag har fått min envishet och egensinnighet. Jag hoppas att jag är lika
pigg som du när jag blir gammal, du är min idol. Kära mormor, tack för allt
stöd och underbara minnen som du och morfar har givit mig, jag önskar att
morfar hade fått se mig idag, men jag är övertygad om att han tittar ner och är
stolt. Jag hoppas att jag i framtiden kan bli en lika bra mormor som du är till
mig. Stora Gun, min extra mormor, du har alltid funnits där och det har varit så
roligt att få hälsa på dig på loven. Vi har varit på många äventyr och haft
massor av skoj och har många minnen. Jag älskar er alla tre så oerhört mycket,
tack för att ni finns i mitt liv.
Not everyone can say that they have an extra set of parents but I can. Hans and
Monica, life is more interesting with you two in my life. I know that you can’t
choose your in-laws but I have the best ones there is. Thank you both for
always being there when I need you and for all the help and love you give.
Thanks to Camilla and Kristoffer for your support. Camilla you are always
kind and listen to me and give me good advice. Kristoffer, you are always fun
to be around and you “get me”. I love you all.
I have two loving parents, mum and dad, that read my articles and try to
understand what I am working on, I know it is not easy but I love that you
keep trying . You both have always supported me in everything I do. All that
support and love goes a long way and look at me now, I made it! Thank you
both from the bottom of my heart, and I love you both so very much!
71
Björn, thank you for all the help you have given us with the children, dogs and
the fun movie nights. You are a big part of our family. Me, Joakim and the
children, we all love “björnen”. You are the best little (big) brother there is, I
love you!
I don’t know what my life would be without my two wonderful children,
Anton and Isabelle. Your smiles and laughs make the sun come out even on the
darkest and rainiest day. One kiss or hug melts me to a big puddle of love. I
love you both so very much.
To my husband Joakim, the love of my life. You are always there for me, even
when the world around me seems to be crumbling down. Your unconditional
love and support means the world to me. I love you all the way to eternity and
back, and some more…
72
9. REFERENCES
1.
Sandri M: Signaling in muscle atrophy and hypertrophy. Physiology 2008,
23:160-170.
2.
Nader GA: Molecular determinants of skeletal muscle mass: getting the
"AKT" together. International Journal of Biochemistry & Cell Biology 2005,
37:1985-1996.
3.
Fanzani A, Conraads VM, Penna F, Martinet W: Molecular and cellular
mechanisms of skeletal muscle atrophy: an update. Journal of cachexia,
sarcopenia and muscle 2012, 3:163-179.
4.
Bodine SC: Hibernation: The search for treatments to prevent disuseinduced skeletal muscle atrophy. Exp Neurol 2013, 248:129-135.
5.
Hu SI, Katz M, Chin S, Qi X, Cruz J, Ibebunjo C, Zhao S, Chen A,
Glass DJ: MNK2 inhibits eIF4G activation through a pathway involving
serine-arginine-rich protein kinase in skeletal muscle. Science signaling
2012, 5:ra14.
6.
Sandri M: Protein breakdown in muscle wasting: Role of autophagylysosome and ubiquitin-proteasome. Int J Biochem Cell Biol 2013,
45:2121-2129.
7.
Price HM, Van de Velde RL: Ultra structure of the skeletal muscle fibre.
In Disorders of voluntary muscle. fourth edition. Edited by Walton SJ.
Edinburgh: Churchill Linvingstone; 1981: 1070
73
8.
Gundersen K: Excitation-transcription coupling in skeletal muscle: the
molecular pathways of exercise. Biol Rev 2011, 86:564-600.
9.
Schiaffino S, Reggiani C: Fiber types in mammalian skeletal muscles.
Physiological reviews 2011, 91:1447-1531.
10.
Smerdu V, Karsch-Mizrachi I, Campione M, Leinwand L, Schiaffino S:
Type IIx myosin heavy chain transcripts are expressed in type IIb fibers
of human skeletal muscle. The American journal of physiology 1994,
267:C1723-1728.
11.
Pette D, Staron RS: Myosin isoforms, muscle fiber types, and
transitions. Microscopy research and technique 2000, 50:500-509.
12.
Ciciliot S, Rossi AC, Dyar KA, Blaauw B, Schiaffino S: Muscle type and
fiber type specificity in muscle wasting. Int J Biochem Cell Biol 2013,
45:2191-2199.
13.
Holecek M: Muscle wasting in animal models of severe illness.
International journal of experimental pathology 2012, 93:157-171.
14.
Voisin L, Breuille D, Combaret L, Pouyet C, Taillandier D, Aurousseau
E, Obled C, Attaix D: Muscle wasting in a rat model of long-lasting
sepsis results from the activation of lysosomal, Ca2+ -activated, and
ubiquitin-proteasome proteolytic pathways. The Journal of clinical
investigation 1996, 97:1610-1617.
15.
Schakman O, Kalista S, Barbe C, Loumaye A, Thissen JP:
Glucocorticoid-induced skeletal muscle atrophy. Int J Biochem Cell Biol
2013, 45:2163-2172.
16.
Bodine SC: Disuse-induced muscle wasting. Int J Biochem Cell Biol 2013,
45:2200-2208.
17.
Frimel TN, Kapadia F, Gaidosh GS, Li Y, Walter GA, Vandenborne K:
A model of muscle atrophy using cast immobilization in mice. Muscle
Nerve 2005, 32:672-674.
18.
Morey-Holton ER, Globus RK: Hindlimb unloading rodent model:
technical aspects. J Appl Physiol (1985) 2002, 92:1367-1377.
74
19.
Hanson AM, Harrison BC, Young MH, Stodieck LS, Ferguson VL:
Longitudinal characterization of functional, morphologic, and
biochemical adaptations in mouse skeletal muscle with hindlimb
suspension. Muscle Nerve 2013, 48:393-402.
20.
Larsson L: Acute quadriplegic myopathy: an acquired "myosinopathy".
Advances in experimental medicine and biology 2008, 642:92-98.
21.
Roy RR, Zhong H, Khalili N, Kim SJ, Higuchi N, Monti RJ, Grossman
E, Hodgson JA, Edgerton VR: Is spinal cord isolation a good model of
muscle disuse? Muscle Nerve 2007, 35:312-321.
22.
Staples JF, Brown JC: Mitochondrial metabolism in hibernation and
daily torpor: a review. Journal of comparative physiology B, Biochemical,
systemic, and environmental physiology 2008, 178:811-827.
23.
Nowell MM, Choi H, Rourke BC: Muscle plasticity in hibernating
ground squirrels (Spermophilus lateralis) is induced by seasonal, but not
low-temperature, mechanisms. Journal of comparative physiology B,
Biochemical, systemic, and environmental physiology 2011, 181:147-164.
24.
Lin DC, Hershey JD, Mattoon JS, Robbins CT: Skeletal muscles of
hibernating brown bears are unusually resistant to effects of denervation.
The Journal of experimental biology 2012, 215:2081-2087.
25.
Lowe DA, Alway SE: Animal models for inducing muscle hypertrophy:
are they relevant for clinical applications in humans? The Journal of
orthopaedic and sports physical therapy 2002, 32:36-43.
26.
Hwee DT, Bodine SC: Age-related deficit in load-induced skeletal
muscle growth. The journals of gerontology Series A, Biological sciences and
medical sciences 2009, 64:618-628.
27.
Armstrong RB, Marum P, Tullson P, Saubert CWt: Acute hypertrophic
response of skeletal muscle to removal of synergists. Journal of applied
physiology: respiratory, environmental and exercise physiology 1979, 46:835842.
75
28.
Roy RR, Edgerton VR: Response of mouse plantaris muscle to
functional overload: comparison with rat and cat. Comparative
biochemistry and physiology Part A, Physiology 1995, 111:569-575.
29.
Baldwin KM, Valdez V, Herrick RE, MacIntosh AM, Roy RR:
Biochemical properties of overloaded fast-twitch skeletal muscle. Journal
of applied physiology: respiratory, environmental and exercise physiology 1982,
52:467-472.
30.
Gu Y, Hall ZW: Characterization of acetylcholine receptor subunits in
developing and in denervated mammalian muscle. J Biol Chem 1988,
263:12878-12885.
31.
Lupa MT, Krzemien DM, Schaller KL, Caldwell JH: Expression and
distribution of sodium channels in short- and long-term denervated
rodent skeletal muscles. J Physiol 1995, 483 ( Pt 1):109-118.
32.
Thesleff S: Botulinal neurotoxins as tools in studies of synaptic
mechanisms. Q J Exp Physiol 1989, 74:1003-1017.
33.
Goldspink DF: The effects of denervation on protein turnover of the
soleus and extensor digitorum longus muscles of adult mice. Comp
Biochem Physiol B 1978, 61:37-41.
34.
Gutmann E, Hanikova M, Hajek I, Klicpera M, Syrovy I: The
postdenervation hypertrophy of the diaphragm. Physiologia
Bohemoslovaca 1966, 15:508-524.
35.
Feng TP, Lu DX: New lights on the phenomenon of transient
hypertrophy in the denervated hemidiaphragm of the rat. Scientia Sinica
1965, 14:1772-1784.
36.
Sola OM, Martin AW: Denervation hypertrophy and atrophy of the
hemidiaphragm of the rat. The American journal of physiology 1953,
172:324-332.
37.
Waddell DS, Baehr LM, van den Brandt J, Johnsen SA, Reichardt HM,
Furlow JD, Bodine SC: The glucocorticoid receptor and FOXO1
synergistically activate the skeletal muscle atrophy-associated MuRF1
gene. Am J Physiol Endocrinol Metab 2008, 295:E785-797.
76
38.
Adams V, Mangner N, Gasch A, Krohne C, Gielen S, Hirner S, Thierse
HJ, Witt CC, Linke A, Schuler G, Labeit S: Induction of MuRF1 is
essential for TNF-alpha-induced loss of muscle function in mice. J Mol
Biol 2008, 384:48-59.
39.
Stitt TN, Drujan D, Clarke BA, Panaro F, Timofeyva Y, Kline WO,
Gonzalez M, Yancopoulos GD, Glass DJ: The IGF-1/PI3K/Akt
pathway prevents short article expression of muscle atrophy-induced
ubiquitin ligases by inhibiting FOXO transcription factors. Molecular
Cell 2004, 14:395-403.
40.
Sandri M, Sandri C, Gilbert A, Skurk C, Calabria E, Picard A, Walsh
K, Schiaffino S, Lecker SH, Goldberg AL: Foxo transcription factors
induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal
muscle atrophy. Cell 2004, 117:399-412.
41.
Shimizu N, Yoshikawa N, Ito N, Maruyama T, Suzuki Y, Takeda S,
Nakae J, Tagata Y, Nishitani S, Takehana K, Sano M, Fukuda K,
Suematsu M, Morimoto C, Tanaka H: Crosstalk between glucocorticoid
receptor and nutritional sensor mTOR in skeletal muscle. Cell Metab
2011, 13:170-182.
42.
Calnan DR, Brunet A: The FoxO code. Oncogene 2008, 27:2276-2288.
43.
Smith LR, Meyer G, Lieber RL: Systems analysis of biological networks
in skeletal muscle function. Wiley interdisciplinary reviews Systems biology
and medicine 2013, 5:55-71.
44.
Frame S, Cohen P: GSK3 takes centre stage more than 20 years after its
discovery. Biochem J 2001, 359:1-16.
45.
Abraham RT: Identification of TOR signaling complexes: more TORC
for the cell growth engine. Cell 2002, 111:9-12.
46.
Mamane Y, Petroulakis E, Rong L, Yoshida K, Ler LW, Sonenberg N:
eIF4E--from translation to transformation. Oncogene 2004, 23:31723179.
47.
Glass DJ: Skeletal muscle hypertrophy and atrophy signaling pathways.
International Journal of Biochemistry & Cell Biology 2005, 37:1974-1984.
77
48.
Norrby M, Tagerud S: Mitogen-Activated Protein Kinase-Activated
Protein Kinase 2 (MK2) in Skeletal Muscle Atrophy and Hypertrophy.
Journal of Cellular Physiology 2010, 223:194-201.
49.
Keren A, Tamir Y, Bengal E: The p38 MAPK signaling pathway: a
major regulator of skeletal muscle development. Molecular and cellular
endocrinology 2006, 252:224-230.
50.
Gaestel M: MAPKAP kinases - MKs - two's company, three's a crowd.
Nature reviews Molecular cell biology 2006, 7:120-130.
51.
Gorog DA, Jabr RI, Tanno M, Sarafraz N, Clark JE, Fisher SG, Cao
XB, Bellahcene M, Dighe K, Kabir AM, Quinlan RA, Kato K, Gaestel
M, Marber MS, Heads RJ: MAPKAPK-2 modulates p38-MAPK
localization and small heat shock protein phosphorylation but does not
mediate the injury associated with p38-MAPK activation during
myocardial ischemia. Cell stress & chaperones 2009, 14:477-489.
52.
Ogata T, Oishi Y, Roy RR, Ohmori H: Endogenous expression and
developmental changes of HSP72 in rat skeletal muscles. J Appl Physiol
(1985) 2003, 95:1279-1286.
53.
Moseley PL: Heat shock proteins and the inflammatory response. Annals
of the New York Academy of Sciences 1998, 856:206-213.
54.
Welch WJ: Mammalian stress response: cell physiology,
structure/function of stress proteins, and implications for medicine and
disease. Physiological reviews 1992, 72:1063-1081.
55.
Bodine SC, Baehr LM: Skeletal muscle atrophy and the E3 ubiquitin
ligases MuRF1 and MAFbx/atrogin-1. Am J Physiol Endocrinol Metab
2014, 307:E469-E484.
56.
Kuo T, Harris CA, Wang JC: Metabolic functions of glucocorticoid
receptor in skeletal muscle. Molecular and cellular endocrinology 2013,
380:79-88.
57.
Baehr LM, Furlow JD, Bodine SC: Muscle sparing in muscle RING
finger 1 null mice: response to synthetic glucocorticoids. J Physiol 2011,
589:4759-4776.
78
58.
Labeit S, Kohl CH, Witt CC, Labeit D, Jung J, Granzier H:
Modulation of muscle atrophy, fatigue and MLC phosphorylation by
MuRF1 as indicated by hindlimb suspension studies on MuRF1-KO
mice. Journal of biomedicine & biotechnology 2010, 2010:693741.
59.
Bodine SC, Latres E, Baumhueter S, Lai VKM, Nunez L, Clarke BA,
Poueymirou WT, Panaro FJ, Na EQ, Dharmarajan K, Pan ZQ,
Valenzuela DM, DeChiara TM, Stitt TN, Yancopoulos GD, Glass DJ:
Identification of ubiquitin ligases required for skeletal muscle atrophy.
Science 2001, 294:1704-1708.
60.
Caron AZ, Drouin G, Desrosiers J, Trensz F, Grenier G: A novel
hindlimb immobilization procedure for studying skeletal muscle atrophy
and recovery in mouse. J Appl Physiol (1985) 2009, 106:2049-2059.
61.
Hwee DT, Baehr LM, Philp A, Baar K, Bodine SC: Maintenance of
muscle mass and load-induced growth in Muscle RING Finger 1 null
mice with age. Aging cell 2013.
62.
Obsil T, Obsilova V: Structure/function relationships underlying
regulation of FOXO transcription factors. Oncogene 2008, 27:22632275.
63.
Zhao Y, Wang Y, Zhu WG: Applications of post-translational
modifications of FoxO family proteins in biological functions. Journal of
molecular cell biology 2011, 3:276-282.
64.
Daitoku H, Sakamaki J, Fukamizu A: Regulation of FoxO transcription
factors by acetylation and protein-protein interactions. Biochim Biophys
Acta 2011, 1813:1954-1960.
65.
Kamei Y, Miura S, Suzuki M, Kai Y, Mizukami J, Taniguchi T,
Mochida K, Hata T, Matsuda J, Aburatani H, Nishino I, Ezaki O:
Skeletal muscle FOXO1 (FKHR) transgenic mice have less skeletal
muscle mass, down-regulated Type I (slow twitch/red muscle) fiber
genes, and impaired glycemic control. J Biol Chem 2004, 279:4111441123.
79
66.
Furuyama T, Nakazawa T, Nakano I, Mori N: Identification of the
differential distribution patterns of mRNAs and consensus binding
sequences for mouse DAF-16 homologues. Biochem J 2000, 349:629634.
67.
Vogt PK, Jiang H, Aoki M: Triple layer control: phosphorylation,
acetylation and ubiquitination of FOXO proteins. Cell Cycle 2005,
4:908-913.
68.
Huang HJ, Tindall DJ: Dynamic FoxO transcription factors. Journal of
Cell Science 2007, 120:2479-2487.
69.
Hu MC, Lee DF, Xia W, Golfman LS, Ou-Yang F, Yang JY, Zou Y,
Bao S, Hanada N, Saso H, Kobayashi R, Hung MC: IkappaB kinase
promotes tumorigenesis through inhibition of forkhead FOXO3a. Cell
2004, 117:225-237.
70.
Jacobs FM, van der Heide LP, Wijchers PJ, Burbach JP, Hoekman MF,
Smidt MP: FoxO6, a novel member of the FoxO class of transcription
factors with distinct shuttling dynamics. J Biol Chem 2003, 278:3595935967.
71.
Obsilova V, Vecer J, Herman P, Pabianova A, Sulc M, Teisinger J,
Boura E, Obsil T: 14-3-3 Protein interacts with nuclear localization
sequence of forkhead transcription factor FoxO4. Biochemistry 2005,
44:11608-11617.
72.
Li J, Tewari M, Vidal M, Lee SS: The 14-3-3 protein FTT-2 regulates
DAF-16 in Caenorhabditis elegans. Developmental biology 2007, 301:8291.
73.
Brunet A, Bonni A, Zigmond MJ, Lin MZ, Juo P, Hu LS, Anderson
MJ, Arden KC, Blenis J, Greenberg ME: Akt promotes cell survival by
phosphorylating and inhibiting a forkhead transcription factor. Cell
1999, 96:857-868.
74.
Fu H, Subramanian RR, Masters SC: 14-3-3 proteins: structure,
function, and regulation. Annu Rev Pharmacol Toxicol 2000, 40:617-647.
80
75.
Muslin AJ, Xing H: 14-3-3 proteins: regulation of subcellular
localization by molecular interference. Cellular signalling 2000, 12:703709.
76.
Banik U, Wang GA, Wagner PD, Kaufman S: Interaction of
phosphorylated tryptophan hydroxylase with 14-3-3 proteins. J Biol
Chem 1997, 272:26219-26225.
77.
Obsil T, Ghirlando R, Klein DC, Ganguly S, Dyda F: Crystal structure
of the 14-3-3zeta:serotonin N-acetyltransferase complex. a role for
scaffolding in enzyme regulation. Cell 2001, 105:257-267.
78.
Boura E, Silhan J, Herman P, Vecer J, Sulc M, Teisinger J, Obsilova V,
Obsil T: Both the N-terminal loop and wing W2 of the forkhead
domain of transcription factor Foxo4 are important for DNA binding. J
Biol Chem 2007, 282:8265-8275.
79.
Brunet A, Kanai F, Stehn J, Xu J, Sarbassova D, Frangioni JV, Dalal SN,
DeCaprio JA, Greenberg ME, Yaffe MB: 14-3-3 transits to the nucleus
and participates in dynamic nucleocytoplasmic transport. J Cell Biol
2002, 156:817-828.
80.
Rena G, Prescott AR, Guo S, Cohen P, Unterman TG: Roles of the
forkhead in rhabdomyosarcoma (FKHR) phosphorylation sites in
regulating 14-3-3 binding, transactivation and nuclear targetting.
Biochem J 2001, 354:605-612.
81.
Tsai WC, Bhattacharyya N, Han LY, Hanover JA, Rechler MM: Insulin
inhibition of transcription stimulated by the forkhead protein Foxo1 is
not solely due to nuclear exclusion. Endocrinology 2003, 144:5615-5622.
82.
Huang H, Regan KM, Wang F, Wang D, Smith DI, van Deursen JM,
Tindall DJ: Skp2 inhibits FOXO1 in tumor suppression through
ubiquitin-mediated degradation. Proc Natl Acad Sci U S A 2005,
102:1649-1654.
81
83.
Matsuzaki H, Daitoku H, Hatta M, Aoyama H, Yoshimochi K,
Fukamizu A: Acetylation of Foxo1 alters its DNA-binding ability and
sensitivity to phosphorylation. Proc Natl Acad Sci U S A 2005, 102:1127811283.
84.
Daitoku H, Hatta M, Matsuzaki H, Aratani S, Ohshima T, Miyagishi
M, Nakajima T, Fukamizu A: Silent information regulator 2 potentiates
Foxo1-mediated transcription through its deacetylase activity. Proc Natl
Acad Sci U S A 2004, 101:10042-10047.
85.
Dansen TB, Smits LM, van Triest MH, de Keizer PL, van Leenen D,
Koerkamp MG, Szypowska A, Meppelink A, Brenkman AB, Yodoi J,
Holstege FC, Burgering BM: Redox-sensitive cysteines bridge
p300/CBP-mediated acetylation and FoxO4 activity. Nature chemical
biology 2009, 5:664-672.
86.
Senf SM, Sandesara PB, Reed SA, Judge AR: p300 Acetyltransferase
activity differentially regulates the localization and activity of the FOXO
homologues in skeletal muscle. Am J Physiol Cell Physiol 2011,
300:C1490-1501.
87.
Leger B, Cartoni R, Praz M, Lamon S, Deriaz O, Crettenand A,
Gobelet C, Rohmer P, Konzelmann M, Luthi F, Russell AP: Akt
signalling through GSK-3 beta, mTOR and Foxo1 is involved in human
skeletal muscle hypertrophy and atrophy. Journal of Physiology-London
2006, 576:923-933.
88.
Argadine HM, Mantilla CB, Zhan WZ, Sieck GC: Intracellular
signaling pathways regulating net protein balance following diaphragm
muscle denervation. Am J Physiol Cell Physiol 2011, 300:C318-327.
89.
Senf SM, Dodd SL, Judge AR: FOXO signaling is required for disuse
muscle atrophy and is directly regulated by Hsp70. Am J Physiol Cell
Physiol 2010, 298:C38-45.
90.
Senf SM, Dodd SL, McClung JM, Judge AR: Hsp70 overexpression
inhibits NF-kappaB and Foxo3a transcriptional activities and prevents
skeletal muscle atrophy. FASEB J 2008, 22:3836-3845.
82
91.
Andres-Mateos E, Brinkmeier H, Burks TN, Mejias R, Files DC,
Steinberger M, Soleimani A, Marx R, Simmers JL, Lin B, Finanger
Hedderick E, Marr TG, Lin BM, Hourde C, Leinwand LA, Kuhl D,
Foller M, Vogelsang S, Hernandez-Diaz I, Vaughan DK, Alvarez de la
Rosa D, Lang F, Cohn RD: Activation of serum/glucocorticoid-induced
kinase 1 (SGK1) is important to maintain skeletal muscle homeostasis
and prevent atrophy. EMBO molecular medicine 2013, 5:80-91.
92.
Bonaldo P, Sandri M: Cellular and molecular mechanisms of muscle
atrophy. Disease models & mechanisms 2013, 6:25-39.
93.
Kyriakis JM, Avruch J: Mammalian mitogen-activated protein kinase
signal transduction pathways activated by stress and inflammation.
Physiological reviews 2001, 81:807-869.
94.
Perdiguero E, Ruiz-Bonilla V, Serrano AL, Munoz-Canoves P: Genetic
deficiency of p38alpha reveals its critical role in myoblast cell cycle exit:
the p38alpha-JNK connection. Cell Cycle 2007, 6:1298-1303.
95.
Zhang G, Li YP: p38beta MAPK upregulates atrogin1/MAFbx by
specific phosphorylation of C/EBPbeta. Skeletal muscle 2012, 2:20.
96.
Pogozelski AR, Geng T, Li P, Yin X, Lira VA, Zhang M, Chi JT, Yan
Z: p38gamma mitogen-activated protein kinase is a key regulator in
skeletal muscle metabolic adaptation in mice. PLoS One 2009, 4:e7934.
97.
Li Z, Jiang Y, Ulevitch RJ, Han J: The primary structure of p38 gamma:
a new member of p38 group of MAP kinases. Biochem Biophys Res
Commun 1996, 228:334-340.
98.
Kumar S, McDonnell PC, Gum RJ, Hand AT, Lee JC, Young PR:
Novel homologues of CSBP/p38 MAP kinase: activation, substrate
specificity and sensitivity to inhibition by pyridinyl imidazoles. Biochem
Biophys Res Commun 1997, 235:533-538.
83
99.
Wang XS, Diener K, Manthey CL, Wang S, Rosenzweig B, Bray J,
Delaney J, Cole CN, Chan-Hui PY, Mantlo N, Lichenstein HS,
Zukowski M, Yao Z: Molecular cloning and characterization of a novel
p38 mitogen-activated protein kinase. J Biol Chem 1997, 272:2366823674.
100.
Cuenda A, Cohen P, Buee-Scherrer V, Goedert M: Activation of stressactivated protein kinase-3 (SAPK3) by cytokines and cellular stresses is
mediated via SAPKK3 (MKK6); comparison of the specificities of
SAPK3 and SAPK2 (RK/p38). EMBO J 1997, 16:295-305.
101.
Engel K, Schultz H, Martin F, Kotlyarov A, Plath K, Hahn M,
Heinemann U, Gaestel M: Constitutive activation of mitogen-activated
protein kinase-activated protein kinase 2 by mutation of
phosphorylation sites and an A-helix motif. J Biol Chem 1995,
270:27213-27221.
102.
Ben-Levy R, Leighton IA, Doza YN, Attwood P, Morrice N, Marshall
CJ, Cohen P: Identification of novel phosphorylation sites required for
activation of MAPKAP kinase-2. EMBO J 1995, 14:5920-5930.
103.
Meng W, Swenson LL, Fitzgibbon MJ, Hayakawa K, Ter Haar E,
Behrens AE, Fulghum JR, Lippke JA: Structure of mitogen-activated
protein kinase-activated protein (MAPKAP) kinase 2 suggests a
bifunctional switch that couples kinase activation with nuclear export. J
Biol Chem 2002, 277:37401-37405.
104.
Engel K, Kotlyarov A, Gaestel M: Leptomycin B-sensitive nuclear
export of MAPKAP kinase 2 is regulated by phosphorylation. EMBO J
1998, 17:3363-3371.
105.
Ben-Levy R, Hooper S, Wilson R, Paterson HF, Marshall CJ: Nuclear
export of the stress-activated protein kinase p38 mediated by its
substrate MAPKAP kinase-2. Current biology : CB 1998, 8:1049-1057.
106.
Ronkina N, Kotlyarov A, Gaestel M: MK2 and MK3--a pair of
isoenzymes? Frontiers in bioscience : a journal and virtual library 2008,
13:5511-5521.
84
107.
Boppart MD, Hirshman MF, Sakamoto K, Fielding RA, Goodyear LJ:
Static stretch increases c-Jun NH2-terminal kinase activity and p38
phosphorylation in rat skeletal muscle. Am J Physiol Cell Physiol 2001,
280:C352-358.
108.
Sakamoto K, Aschenbach WG, Hirshman MF, Goodyear LJ: Akt
signaling in skeletal muscle: regulation by exercise and passive stretch.
Am J Physiol Endocrinol Metab 2003, 285:E1081-1088.
109.
Hornberger TA, Chien S: Mechanical stimuli and nutrients regulate
rapamycin-sensitive signaling through distinct mechanisms in skeletal
muscle. Journal of cellular biochemistry 2006, 97:1207-1216.
110.
Ito Y, Obara K, Ikeda R, Ishii M, Tanabe Y, Ishikawa T, Nakayama K:
Passive stretching produces Akt- and MAPK-dependent augmentations
of GLUT4 translocation and glucose uptake in skeletal muscles of mice.
Pflugers Arch 2006, 451:803-813.
111.
Huey KA: Regulation of HSP25 expression and phosphorylation in
functionally overloaded rat plantaris and soleus muscles. J Appl Physiol
(1985) 2006, 100:451-456.
112.
Carlson CJ, Fan Z, Gordon SE, Booth FW: Time course of the MAPK
and PI3-kinase response within 24 h of skeletal muscle overload. J Appl
Physiol (1985) 2001, 91:2079-2087.
113.
Wretman C, Widegren U, Lionikas A, Westerblad H, Henriksson J:
Differential activation of mitogen-activated protein kinase signalling
pathways by isometric contractions in isolated slow- and fast-twitch rat
skeletal muscle. Acta physiologica Scandinavica 2000, 170:45-49.
114.
Kim J, Won KJ, Lee HM, Hwang BY, Bae YM, Choi WS, Song H,
Lim KW, Lee CK, Kim B: p38 MAPK Participates in Muscle-Specific
RING Finger 1-Mediated Atrophy in Cast-Immobilized Rat
Gastrocnemius Muscle. Korean Journal of Physiology & Pharmacology
2009, 13:491-496.
85
115.
Paul PK, Gupta SK, Bhatnagar S, Panguluri SK, Darnay BG, Choi Y,
Kumar A: Targeted ablation of TRAF6 inhibits skeletal muscle wasting
in mice. J Cell Biol 2010, 191:1395-1411.
116.
Derbre F, Ferrando B, Gomez-Cabrera MC, Sanchis-Gomar F,
Martinez-Bello VE, Olaso-Gonzalez G, Diaz A, Gratas-Delamarche A,
Cerda M, Vina J: Inhibition of xanthine oxidase by allopurinol prevents
skeletal muscle atrophy: role of p38 MAPKinase and E3 ubiquitin
ligases. PLoS One 2012, 7:e46668.
117.
Li YP, Chen YL, John J, Moylan J, Jin BW, Mann DL, Reid MB:
TNF-alpha acts via p38 MAPK to stimulate expression of the ubiquitin
ligase atrogin1/MAFbx in skeletal muscle. Faseb Journal 2005, 19:362370.
118.
Stokoe D, Engel K, Campbell DG, Cohen P, Gaestel M: Identification
of MAPKAP kinase 2 as a major enzyme responsible for the
phosphorylation of the small mammalian heat shock proteins. FEBS Lett
1992, 313:307-313.
119.
Dodd SL, Hain B, Senf SM, Judge AR: Hsp27 inhibits IKK betainduced NF-kappa B activity and skeletal muscle atrophy. Faseb Journal
2009, 23:3415-3423.
120.
Huey KA, Thresher JS, Brophy CM, Roy RR: Inactivity-induced
modulation of Hsp20 and Hsp25 content in rat hindlimb muscles.
Muscle Nerve 2004, 30:95-101.
121.
Kawano F, Matsuoka Y, Oke Y, Higo Y, Terada M, Wang XD, Nakai
N, Fukuda H, Imajoh-Ohmi S, Ohira Y: Role(s) of nucleoli and
phosphorylation of ribosomal protein S6 and/or HSP27 in the regulation
of muscle mass. American Journal of Physiology-Cell Physiology 2007,
293:C35-C44.
122.
Huey KA, McCall GE, Zhong H, Roy RR: Modulation of HSP25 and
TNF-alpha during the early stages of functional overload of a rat slow
and fast muscle. J Appl Physiol (1985) 2007, 102:2307-2314.
86
123.
Huey KA, Burdette S, Zhong H, Roy RR: Early response of heat shock
proteins to functional overload of the soleus and plantaris in rats and
mice. Experimental physiology 2010, 95:1145-1155.
124.
Noble EG, Milne KJ, Melling CW: Heat shock proteins and exercise: a
primer. Applied physiology, nutrition, and metabolism = Physiologie
appliquee, nutrition et metabolisme 2008, 33:1050-1065.
125.
Liu Y, Gampert L, Nething K, Steinacker JM: Response and function of
skeletal muscle heat shock protein 70. Frontiers in bioscience : a journal
and virtual library 2006, 11:2802-2827.
126.
Locke M, Noble EG, Atkinson BG: Inducible isoform of HSP70 is
constitutively expressed in a muscle fiber type specific pattern. The
American journal of physiology 1991, 261:C774-779.
127.
Oishi Y, Ogata T, Ohira Y, Taniguchi K, Roy RR: Calcineurin and heat
shock protein 72 in functionally overloaded rat plantaris muscle. Biochem
Biophys Res Commun 2005, 330:706-713.
128.
Naito H, Powers SK, Demirel HA, Sugiura T, Dodd SL, Aoki J: Heat
stress attenuates skeletal muscle atrophy in hindlimb-unweighted rats. J
Appl Physiol (1985) 2000, 88:359-363.
129.
Lawler JM, Song W, Kwak HB: Differential response of heat shock
proteins to hindlimb unloading and reloading in the soleus. Muscle Nerve
2006, 33:200-207.
130.
Lecker SH, Jagoe RT, Gilbert A, Gomes M, Baracos V, Bailey J, Price
SR, Mitch WE, Goldberg AL: Multiple types of skeletal muscle atrophy
involve a common program of changes in gene expression. FASEB J
2004, 18:39-51.
131.
Oishi Y, Taniguchi K, Matsumoto H, Kawano F, Ishihara A, Ohira Y:
Upregulation of HSP72 in reloading rat soleus muscle after prolonged
hindlimb unloading. The Japanese journal of physiology 2003, 53:281-286.
87
132.
Nishimura M, Mikura M, Hirasaka K, Okumura Y, Nikawa T, Kawano
Y, Nakayama M, Ikeda M: Effects of dimethyl sulphoxide and
dexamethasone on mRNA expression of myogenesis- and muscle
proteolytic system-related genes in mouse myoblastic C2C12 cells.
Journal of biochemistry 2008, 144:717-724.
133.
Yu CY, Mayba O, Lee JV, Tran J, Harris C, Speed TP, Wang JC:
Genome-wide analysis of glucocorticoid receptor binding regions in
adipocytes reveal gene network involved in triglyceride homeostasis.
PLoS One 2010, 5:e15188.
134.
Kuo T, Lew MJ, Mayba O, Harris CA, Speed TP, Wang JC: Genomewide analysis of glucocorticoid receptor-binding sites in myotubes
identifies gene networks modulating insulin signaling. Proc Natl Acad Sci
U S A 2012, 109:11160-11165.
135.
Dardevet D, Sornet C, Savary I, Debras E, Patureau-Mirand P, Grizard
J: Glucocorticoid effects on insulin- and IGF-I-regulated muscle protein
metabolism during aging. J Endocrinol 1998, 156:83-89.
136.
Ruvinsky I, Meyuhas O: Ribosomal protein S6 phosphorylation: from
protein synthesis to cell size. Trends BiochemSci 2006, 31:342-348.
137.
Laser M, Kasi VS, Hamawaki M, Cooper G, Kerr CM, Kuppuswamy D:
Differential activation of p70 and p85 S6 kinase isoforms during cardiac
hypertrophy in the adult mammal. Journal of Biological Chemistry 1998,
273:24610-24619.
138.
Pullen N, Thomas G: The modular phosphorylation and activation of
p70s6k. FEBS Lett 1997, 410:78-82.
139.
Pearson RB, Dennis PB, Han JW, Williamson NA, Kozma SC,
Wettenhall RE, Thomas G: The principal target of rapamycin-induced
p70s6k inactivation is a novel phosphorylation site within a conserved
hydrophobic domain. EMBO J 1995, 14:5279-5287.
140.
Fumagalli S, Thomas G: S6 Phosphorylation and Signal Transduction.
In: Translational Control of Gene Expression (Sonenberg, N et al, eds),
Woodbury, NY, USA: Cold Spring Harbor Laboratory Press 2000:695-717.
88
141.
Chale-Rush A, Morris EP, Kendall TL, Brooks NE, Fielding RA:
Effects of Chronic Overload on Muscle Hypertrophy and mTOR
Signaling in Young Adult and Aged Rats. Journals of Gerontology Series aBiological Sciences and Medical Sciences 2009, 64:1232-1239.
142.
Thomson DM, Gordon SE: Impaired overload-induced muscle growth
is associated with diminished translational signalling in aged rat fasttwitch skeletal muscle. Journal of Physiology-London 2006, 574:291-305.
143.
Ruvinsky I, Katz M, Dreazen A, Gielchinsky Y, Saada A, Freedman N,
Mishani E, Zimmerman G, Kasir J, Meyuhas O: Mice Deficient in
Ribosomal Protein S6 Phosphorylation Suffer from Muscle Weakness
that Reflects a Growth Defect and Energy Deficit. PLoS One 2009, 4:11.
144.
Ohanna M, Sobering AK, Lapointe T, Lorenzo L, Praud C, Petroulakis
E, Sonenberg N, Kelly PA, Sotiropoulos A, Pende M: Atrophy of
S6K1(-/-) skeletal muscle cells reveals distinct mTOR effectors for cell
cycle and size control. Nature Cell Biology 2005, 7:286-294.
145.
Vary TC, Lynch CJ: Meal feeding enhances formation of eIF4F in
skeletal muscle: role of increased eIF4E availability and eIF4G
phosphorylation. Am J Physiol Endocrinol Metab 2006, 290:E631-642.
146.
Raught B, Gingras AC, Sonenberg N: The target of rapamycin (TOR)
proteins. Proceedings of the National Academy of Sciences of the United States
of America 2001, 98:7037-7044.
147.
Khaleghpour K, Pyronnet S, Gingras AC, Sonenberg N: Translational
homeostasis: eukaryotic translation initiation factor 4E control of 4Ebinding protein 1 and p70 S6 kinase activities. Mol Cell Biol 1999,
19:4302-4310.
148.
Gingras AC, Raught B, Sonenberg N: eIF4 initiation factors: effectors
of mRNA recruitment to ribosomes and regulators of translation.
Annual review of biochemistry 1999, 68:913-963.
89
149.
Balage M, Sinaud S, Prod'homme M, Dardevet D, Vary TC, Kimball
SR, Jefferson LS, Grizard J: Amino acids and insulin are both required
to regulate assembly of the eIF4E. eIF4G complex in rat skeletal muscle.
Am J Physiol Endocrinol Metab 2001, 281:E565-574.
150.
Lang CH, Frost RA: Glucocorticoids and TNFalpha interact
cooperatively to mediate sepsis-induced leucine resistance in skeletal
muscle. Mol Med 2006, 12:291-299.
151.
Foster KG, Fingar DC: Mammalian target of rapamycin (mTOR):
conducting the cellular signaling symphony. J Biol Chem 2010,
285:14071-14077.
152.
Hinton TM, Coldwell MJ, Carpenter GA, Morley SJ, Pain VM:
Functional analysis of individual binding activities of the scaffold protein
eIF4G. J Biol Chem 2007, 282:1695-1708.
153.
Lamphear BJ, Kirchweger R, Skern T, Rhoads RE: Mapping of
functional domains in eukaryotic protein synthesis initiation factor 4G
(eIF4G) with picornaviral proteases. Implications for cap-dependent
and cap-independent translational initiation. J Biol Chem 1995,
270:21975-21983.
154.
Prevot D, Darlix JL, Ohlmann T: Conducting the initiation of protein
synthesis: the role of eIF4G. Biology of the cell / under the auspices of the
European Cell Biology Organization 2003, 95:141-156.
155.
Pyronnet S, Imataka H, Gingras AC, Fukunaga R, Hunter T, Sonenberg
N: Human eukaryotic translation initiation factor 4G (eIF4G) recruits
mnk1 to phosphorylate eIF4E. EMBO J 1999, 18:270-279.
156.
Proud CG: Signalling to translation: how signal transduction pathways
control the protein synthetic machinery. Biochemical Journal 2007,
403:217-234.
157.
Shen WH, Boyle DW, Wisniowski P, Bade A, Liechty EA: Insulin and
IGF-I stimulate the formation of the eukaryotic initiation factor 4F
complex and protein synthesis in C2C12 myotubes independent of
availability of external amino acids. J Endocrinol 2005, 185:275-289.
90
158.
van Breukelen F, Sonenberg N, Martin SL: Seasonal and statedependent changes of eIF4E and 4E-BP1 during mammalian
hibernation: implications for the control of translation during torpor.
American journal of physiology Regulatory, integrative and comparative
physiology 2004, 287:R349-353.
159.
Lang CH, Frost RA: Differential effect of sepsis on ability of leucine and
IGF-I to stimulate muscle translation initiation. Am J Physiol Endocrinol
Metab 2004, 287:E721-730.
160.
Bodine SC, Stitt TN, Gonzalez M, Kline WO, Stover GL, Bauerlein R,
Zlotchenko E, Scrimgeour A, Lawrence JC, Glass DJ, Yancopoulos GD:
Akt/mTOR pathway is a crucial regulator of skeletal muscle hypertrophy
and can prevent muscle atrophy in vivo. Nature Cell Biology 2001,
3:1014-1019.
161.
Soliman GA, Acosta-Jaquez HA, Fingar DC: mTORC1 inhibition via
rapamycin promotes triacylglycerol lipolysis and release of free fatty acids
in 3T3-L1 adipocytes. Lipids 2010, 45:1089-1100.
162.
Fingar DC, Richardson CJ, Tee AR, Cheatham L, Tsou C, Blenis J:
mTOR controls cell cycle progression through its cell growth effectors
S6K1 and 4E-BP1/eukaryotic translation initiation factor 4E. Molecular
and Cellular Biology 2004, 24:200-216.
163.
Schiaffino S, Dyar KA, Ciciliot S, Blaauw B, Sandri M: Mechanisms
regulating skeletal muscle growth and atrophy. The FEBS journal 2013,
280:4294-4314.
164.
Dunlop EA, Tee AR: Mammalian target of rapamycin complex 1:
signalling inputs, substrates and feedback mechanisms. Cellular
signalling 2009, 21:827-835.
165.
Matheny RW, Jr., Adamo ML: Effects of PI3K catalytic subunit and
Akt isoform deficiency on mTOR and p70S6K activation in myoblasts.
Biochem Biophys Res Commun 2009, 390:252-257.
91
166.
Fingar DC, Salama S, Tsou C, Harlow E, Blenis J: Mammalian cell size
is controlled by mTOR and its downstream targets S6K1 and
4EBP1/eIF4E. Genes & Development 2002, 16:1472-1487.
167.
Gingras AC, Raught B, Sonenberg N: Regulation of translation
initiation by FRAP/mTOR. Genes Dev 2001, 15:807-826.
168.
Kawasome H, Papst P, Webb S, Keller GM, Johnson GL, Gelfand EW,
Terada N: Targeted disruption of p70(s6k) defines its role in protein
synthesis and rapamycin sensitivity. Proc Natl Acad Sci U S A 1998,
95:5033-5038.
169.
Cross DA, Alessi DR, Cohen P, Andjelkovich M, Hemmings BA:
Inhibition of glycogen synthase kinase-3 by insulin mediated by protein
kinase B. Nature 1995, 378:785-789.
170.
Doble BW, Woodgett JR: GSK-3: tricks of the trade for a multi-tasking
kinase. J Cell Sci 2003, 116:1175-1186.
171.
Matheny RW, Jr., Adamo ML: Role of Akt isoforms in IGF-I-mediated
signaling and survival in myoblasts. Biochem Biophys Res Commun 2009,
389:117-121.
172.
Kandel ES, Hay N: The regulation and activities of the multifunctional
serine/threonine kinase Akt/PKB. Exp Cell Res 1999, 253:210-229.
173.
Matheny RW, Jr., Adamo ML: Current perspectives on Akt Akt-ivation
and Akt-ions. Exp Biol Med (Maywood) 2009, 234:1264-1270.
174.
Cho H, Thorvaldsen JL, Chu Q, Feng F, Birnbaum MJ:
Akt1/PKBalpha is required for normal growth but dispensable for
maintenance of glucose homeostasis in mice. J Biol Chem 2001,
276:38349-38352.
175.
Cho H, Mu J, Kim JK, Thorvaldsen JL, Chu Q, Crenshaw EB, 3rd,
Kaestner KH, Bartolomei MS, Shulman GI, Birnbaum MJ: Insulin
resistance and a diabetes mellitus-like syndrome in mice lacking the
protein kinase Akt2 (PKB beta). Science 2001, 292:1728-1731.
92
176.
Lai KM, Gonzalez M, Poueymirou WT, Kline WO, Na E, Zlotchenko
E, Stitt TN, Economides AN, Yancopoulos GD, Glass DJ: Conditional
activation of akt in adult skeletal muscle induces rapid hypertrophy. Mol
Cell Biol 2004, 24:9295-9304.
177.
Peng XD, Xu PZ, Chen ML, Hahn-Windgassen A, Skeen J, Jacobs J,
Sundararajan D, Chen WS, Crawford SE, Coleman KG, Hay N:
Dwarfism, impaired skin development, skeletal muscle atrophy, delayed
bone development, and impeded adipogenesis in mice lacking Akt1 and
Akt2. Genes Dev 2003, 17:1352-1365.
178.
Yang ZZ, Tschopp O, Hemmings-Mieszczak M, Feng J, Brodbeck D,
Perentes E, Hemmings BA: Protein kinase B alpha/Akt1 regulates
placental development and fetal growth. J Biol Chem 2003, 278:3212432131.
179.
Hay N, Sonenberg N: Upstream and downstream of mTOR. Genes Dev
2004, 18:1926-1945.
180.
Datta SR, Brunet A, Greenberg ME: Cellular survival: a play in three
Akts. Genes Dev 1999, 13:2905-2927.
181.
Hornberger TA: Mechanotransduction and the regulation of mTORC1
signaling in skeletal muscle. Int J Biochem Cell Biol 2011, 43:1267-1276.
182.
Hornberger TA, Hunter RB, Kandarian SC, Esser KA: Regulation of
translation factors during hindlimb unloading and denervation of
skeletal muscle in rats. Am J Physiol Cell Physiol 2001, 281:C179-187.
183.
Sugiura T, Abe N, Nagano M, Goto K, Sakuma K, Naito H, Yoshioka
T, Powers SK: Changes in PKB/Akt and calcineurin signaling during
recovery in atrophied soleus muscle induced by unloading. American
journal of physiology Regulatory, integrative and comparative physiology
2005, 288:R1273-1278.
93
184.
Han B, Zhu MJ, Ma C, Du M: Rat hindlimb unloading down-regulates
insulin like growth factor-1 signaling and AMP-activated protein
kinase, and leads to severe atrophy of the soleus muscle. Applied
physiology, nutrition, and metabolism = Physiologie appliquee, nutrition et
metabolisme 2007, 32:1115-1123.
185.
Gwag T, Lee K, Ju H, Shin H, Lee JW, Choi I: Stress and signaling
responses of rat skeletal muscle to brief endurance exercise during
hindlimb unloading: a catch-up process for atrophied muscle. Cellular
physiology and biochemistry : international journal of experimental cellular
physiology, biochemistry, and pharmacology 2009, 24:537-546.
186.
Childs TE, Spangenburg EE, Vyas DR, Booth FW: Temporal
alterations in protein signaling cascades during recovery from muscle
atrophy. Am J Physiol Cell Physiol 2003, 285:C391-398.
187.
Sartori R, Milan G, Patron M, Mammucari C, Blaauw B, Abraham R,
Sandri M: Smad2 and 3 transcription factors control muscle mass in
adulthood. Am J Physiol Cell Physiol 2009, 296:C1248-1257.
188.
Magnusson C, Hogklint L, Libelius R, Tagerud S: Expression of mRNA
for plasminogen activators and protease nexin-1 in innervated and
denervated mouse skeletal muscle. Journal of Neuroscience Research 2001,
66:457-463.
189.
Gonzalez-Darder JM, Barbera J, Abellan MJ: Effects of prior
anaesthesia on autotomy following sciatic transection in rats. Pain 1986,
24:87-91.
190.
Bradford MM: A rapid and sensitive method for the quantitation of
microgram quantities of protein utilizing the principle of protein-dye
binding. Anal Biochem 1976, 72:248-254.
191.
Siegel AL, Bledsoe C, Lavin J, Gatti F, Berge J, Millman G, Turin E,
Winders WT, Rutter J, Palmeiri B, Carlson CG: Treatment with
inhibitors of the NF-kappaB pathway improves whole body tension
development in the mdx mouse. Neuromuscular disorders : NMD 2009,
19:131-139.
94
192.
Nielsen PJ, Manchester KL, Towbin H, Gordon J, Thomas G: The
phosphorylation of ribosomal protein-S6 in rat-tissues following
cycloheximide injection, in diabetes, and after denervation of diaphragm
- a simple immunological determination of the extent of S6phosphorylation on protein blots. Journal of Biological Chemistry 1982,
257:2316-2321.
193.
Svensson A, Norrby M, Libelius R, Tagerud S: Secreted frizzled related
protein 1 (Sfrp1) and Wnt signaling in innervated and denervated
skeletal muscle. Journal of molecular histology 2008, 39:329-337.
194.
Machida M, Takeda K, Yokono H, Ikemune S, Taniguchi Y, Kiyosawa
H, Takemasa T: Reduction of ribosome biogenesis with activation of the
mTOR pathway in denervated atrophic muscle. J Cell Physiol 2012,
227:1569-1576.
195.
Turner LV, Garlick PJ: The effect of unilateral phrenicectomy on the
rate of protein synthesis in rat diaphragm in vivo. Biochim Biophys Acta
1974, 349:109-113.
196.
Kline WO, Panaro FJ, Yang H, Bodine SC: Rapamycin inhibits the
growth and muscle-sparing effects of clenbuterol. J Appl Physiol (1985)
2007, 102:740-747.
197.
Green HJ, Reichmann H, Pette D: Inter- and intraspecies comparisons
of fibre type distribution and of succinate dehydrogenase activity in type
I, IIA and IIB fibres of mammalian diaphragms. Histochemistry 1984,
81:67-73.
198.
Carlson CJ, Booth FW, Gordon SE: Skeletal muscle myostatin mRNA
expression is fiber-type specific and increases during hindlimb
unloading. The American journal of physiology 1999, 277:R601-606.
199.
Sher J, Cardasis C: Skeletal muscle fiber types in the adult mouse. Acta
neurologica Scandinavica 1976, 54:45-56.
200.
Argadine HM, Hellyer NJ, Mantilla CB, Zhan WZ, Sieck GC: The
effect of denervation on protein synthesis and degradation in adult rat
diaphragm muscle. Journal of Applied Physiology 2009, 107:438-444.
95
201.
Kang J, Jeong MG, Oh S, Jang EJ, Kim HK, Hwang ES: A FoxO1dependent, but NRF2-independent induction of heme oxygenase-1
during muscle atrophy. FEBS Lett 2014, 588:79-85.
202.
Bertaggia E, Coletto L, Sandri M: Posttranslational modifications
control FoxO3 activity during denervation. Am J Physiol Cell Physiol
2012, 302:C587-596.
203.
Lokireddy S, Wijesoma IW, Teng S, Bonala S, Gluckman PD,
McFarlane C, Sharma M, Kambadur R: The ubiquitin ligase Mul1
induces mitophagy in skeletal muscle in response to muscle-wasting
stimuli. Cell Metab 2012, 16:613-624.
204.
Wei B, Dui W, Liu D, Xing Y, Yuan Z, Ji G: MST1, a key player, in
enhancing fast skeletal muscle atrophy. BMC biology 2013, 11:12.
205.
Gong X, Luo T, Deng P, Liu Z, Xiu J, Shi H, Jiang Y: Stress-induced
interaction between p38 MAPK and HSP70. Biochem Biophys Res
Commun 2012, 425:357-362.
206.
Vary TC, Lynch CJ: Nutrient signaling components controlling protein
synthesis in striated muscle. The Journal of nutrition 2007, 137:18351843.
207.
Haghighat A, Sonenberg N: eIF4G dramatically enhances the binding
of eIF4E to the mRNA 5'-cap structure. J Biol Chem 1997, 272:2167721680.
208.
Haghighat A, Mader S, Pause A, Sonenberg N: Repression of capdependent translation by 4E-binding protein 1: competition with p220
for binding to eukaryotic initiation factor-4E. Embo Journal 1995,
14:5701-5709.
96