You are on page 1of 10

Am J Physiol Cell Physiol 311: C663–C672, 2016.

First published August 31, 2016; doi:10.1152/ajpcell.00144.2016.

CALL FOR PAPERS Cellular Mechanisms of Proteostasis

mTOR signaling regulates myotube hypertrophy by modulating protein


synthesis, rDNA transcription, and chromatin remodeling
Ferdinand von Walden,2 Chang Liu,2 Nicole Aurigemma,1 and Gustavo A. Nader1,2
1
Noll Laboratory, Department of Kinesiology and Huck Institutes of the Life Sciences, The Pennsylvania State University,
University Park, Pennsylvania; and 2Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden.
Submitted 19 May 2016; accepted in final form 30 August 2016

Downloaded from http://ajpcell.physiology.org/ by 10.220.32.246 on October 15, 2016


von Walden F, Liu C, Aurigemma N, Nader GA. mTOR sig- r-proteins mRNAs and accessory factors required for rRNA
naling regulates myotube hypertrophy by modulating protein synthe- processing; and Pol III transcribes 5S and tRNAs. Of these,
sis, rDNA transcription, and chromatin remodeling. Am J Physiol Cell transcription of rDNA genes by RNA Pol I is thought to be
Physiol 311: C663–C672, 2016. First published August 31, 2016; rate limiting for ribosome production and cellular growth
doi:10.1152/ajpcell.00144.2016.—Ribosome production is an early
(10, 26, 35).
event during skeletal muscle hypertrophy and precedes muscle protein
accretion. Signaling via mTOR is crucial for ribosome production and
The mechanistic target of rapamycin (mTOR) is central to
hypertrophy; however, the mechanisms by which it regulates these growth control (16, 21). In skeletal muscle, mTOR inhibition
processes remain to be identified. Herein, we investigated the activa- by rapamycin impedes hypertrophy in vivo and in vitro (2, 33)
tion of mTOR signaling in hypertrophying myotubes and determined partly by blocking rRNA synthesis (27). In addition to its role
that mTOR coordinates various aspects of gene expression important in mRNA translation, mTOR is involved in transcription by all
for ribosome production. First, inhibition of translation with cyclo- three Pols (21), and although its importance in skeletal muscle
heximide had a more potent effect on protein synthesis than rapamy- hypertrophy has recently been challenged (9), early mTOR
cin indicating that mTOR function during hypertrophy is not on activation in hypertrophying muscle is consistent with an
general, but rather on specific protein synthesis. Second, blocking Pol increase in rDNA transcription (41). Signaling to mTOR by
II transcription had a similar effect as Rapamycin and, unexpectedly, PI3K and downstream to S6K1 is important for various ana-
revealed the necessity of Pol II transcription for Pol I transcription, bolic processes (6, 15), especially Pol I transcription (4, 11,
suggesting that mTOR may regulate ribosome production also by
14). However, the functional divergence exerted by the PI3K/
controlling Class II genes at the transcriptional level. Third, Pol I
activity is essential for rDNA transcription and, surprisingly, for mTOR/S6K1 signaling cascade requires a detailed investiga-
protein synthesis as selective Pol I inhibition blunted rDNA transcrip- tion of the relative contribution each factor has in controlling
tion, protein synthesis, and the hypertrophic response of myotubes. ribosome production during muscle hypertrophy. For example,
Finally, mTOR has nuclear localization in muscle, which is not while mTOR-dependent regulation of translation is well estab-
sensitive to rapamycin. Inhibition of mTOR signaling by rapamycin lished (15), its functions in muscle gene transcription are less
disrupted mTOR-rDNA promoter interaction and resulted in altered clear. Recent studies demonstrated that mTOR can localize to
histone marks indicative of repressed transcription and formation of both cytoplasmic and nuclear compartments (13, 34, 39) indi-
higher-order chromatin structure. Thus mTOR signaling appears to cating that mTOR nuclear localization may play an important
regulate muscle hypertrophy by affecting protein synthesis, Class I role in gene regulation. Furthermore, nuclear TOR signaling
and II gene expression, and chromatin remodeling. appears to regulate histone acetylation (32) and rDNA tran-
mTOR; ribosome biogenesis; RNA Pol I; transcription; epigenetics; scription in response to nutrients (18). Thus mTOR signaling
hypertrophy may modulate muscle hypertrophy at the translational and
transcriptional levels by coordinating multiple processes im-
portant for ribosome production.
DE NOVO ribosome production is an early event during skeletal The main goal of the present study was to determine the
muscle hypertrophy. The increase in protein synthetic capacity involvement of mTOR signaling in rDNA transcription, and
is needed to sustain muscle protein accretion and is directly whether its role in protein synthesis and gene expression
proportional to muscle protein synthesis rates (25). At the onset represent two distinct and separable mTOR functions during
of muscle hypertrophy preceding any measureable protein myotube hypertrophy. We hypothesized that by having cyto-
accretion (20), increased ribosomal DNA (rDNA) transcription plasmic and nuclear localization, mTOR orchestrates ribosome
by RNA polymerase I (Pol I) leads to ribosomal RNA (rRNA) production by regulating translation and transcription of Class
accumulation, and an increase in ribosome content (41). This I and II genes. We approached these aims by first using
involves the activity of all three transcription systems: RNA rapamycin to block mTOR signaling, and then contrasting the
Polymerase (Pol) I generates a primary transcript from the 45S findings by using specific inhibitors for the downstream pro-
ribosomal (r)DNA genes to produce 18S, 5.8 S, and 28S cesses in which mTOR is known to be involved, e.g., protein
rRNA; Pol II transcription is responsible for synthesizing all synthesis and Pol I or Pol II transcription. These biochemical
dissections would reveal whether inhibition of mTOR signal-
Address for reprint requests and other correspondence: G. A. Nader, 101
ing by rapamycin has a similar impact on ribosome production
Noll Laboratory, Dept. of Kinesiology, The Pennsylvania State Univ., Univ. and hypertrophy as the specific inhibition of the downstream
Park, PA 16802 (e-mail: gan11@psu.edu). processes (i.e., protein synthesis, transcription). We provide
http://www.ajpcell.org 0363-6143/16 Copyright © 2016 the American Physiological Society C663
C664 RIBOSOMAL DNA GENE REGULATION BY mTOR DURING MUSCLE HYPERTROPHY

novel evidence indicating the necessity of protein synthesis and Leitz Fluovert microscope equipped with a digital camera. Myotube
Pol II transcription for rDNA transcription, ribosome produc- diameter was determined at the point of the widest diameter in all
tion, and hypertrophy. Additionally, we show that in myotubes, multinucleated myotubes using ImageJ (NIH, Bethesda, MD).
mTOR has nuclear localization and is involved in rDNA RNA extraction and quantification, cDNA synthesis, and qRT-PCR.
transcription in a rapamycin-sensitive manner by directly in- Total RNA was extracted with TRizol Reagent (Invitrogen, Carlsbad,
CA) and subsequently cleaned using Direct-zol columns (Zymo Re-
teracting with the rDNA gene promoter (rDNAp). We also search, Irvine, CA). RNA quantification and purity were determined
provide novel data indicating a reduction in Histone 3 Lysine on a Nanodrop ND-1000 and RNA integrity verified by agarose gel
56 acetylation (H3K56ac) and enrichment of linker histone H1 electrophoresis as previously described (27). RNA was stored at
(H1) at the rDNAp consistent with transcriptional silencing and ⫺80°C until further use. cDNA was synthesized from 1 ␮g of total
inhibition of mTOR signaling by rapamycin. RNA with the VILO cDNA (Invitrogen) synthesis kit according to the
manufacturer’s recommendations. The cDNA stock was further di-
MATERIALS AND METHODS luted for downstream Quantitative Real-Time Polymerase chain reac-
tion (qRT-PCR) depending on the target gene. qRT-PCR was per-
Cell culture and chemical inhibitors. C2C12 myoblasts (ATCC,
Manassas, VA) were grown to 90 –100% confluence in growth me- formed using a SYBR-chemistry based with GoTaq supermix (Pro-

Downloaded from http://ajpcell.physiology.org/ by 10.220.32.246 on October 15, 2016


dium (DMEM with 10% fetal bovine serum ⫹ 1% Pen-Strep) and mega, Fitchburg, WI) on a Bio-Rad touch CFX-384 system. Reactions
were induced to fuse into myotubes by switching to differentiation were run in triplicate for each sample on 384 well plates. Primers were
medium (DM ⫽ DMEM with 2% horse serum ⫹ 1% Pen-Strep) for tested on a 64-54°C heat gradient to optimize annealing temperature
4 days. To eliminate undifferentiated myoblast from the cultures, 20 and verify a primer efficiency of ⬃100%. A melt-curve analysis was
␮M AraC was added to the medium for 24 h on the third day of performed for each primer pair to ensure the amplification of a sole
differentiation. At day 4 postdifferentiation, myotubes were either PCR product and amplicon size was verified by agarose-gel elec-
maintained in DM or stimulated with high serum medium (GM ⫽ trophoresis. Relative expression levels were obtained by normal-
DMEM with 20% FBS ⫹ 1% Pen-Strep) for 1, 3, 6, 12, 24, or 48 h. ization to GADPH (or 28S for DRB and treated samples) by the
All experiments were performed in a humidified environment at 37°C comparative CT (⌬⌬CT) method in the Bio-Rad CFX Manager
and 5% CO2. For siRNA transfection experiments, myoblasts were (version 3.0) software. Primer sequences utilized in the present
seeded in 12-well plates and double transfected at day 3 and day 5 study are described in Table 1.
postdifferentiation with S6K1 siRNA (50 pM) or scrambled siRNA Cell lysis and cytosol/nuclear protein fractionation. Total protein
(50 pM) using Lipofectamine 2000 (Invitrogen, Carlsbad, CA) ac- was extracted from cells lysed in ice-cold lysis buffer (50 mM
cording to the manufacturer’s instructions. Myotubes were stimulated HEPES-KOH, pH 7.5, 140 mM NaCl, 1 mM EDTA, 1% Triton
by addition of GM for 12 h after the second transfection. Chemical X-100, 0.1% sodium deoxycholate, and 0.1% SDS) supplemented
inhibitors were dissolved in dimethyl sulfoxide (DMSO) as follows: with one tablet of Complete mini protease inhibitor cocktail and
CX-5461 {2-(4-methyl-1,4-diazepan-1-yl)-N-[(5-methylpyrazin-2-yl) PhoStop phosphatase inhibitors (Roche Diagnostics, Indianapolis,
methyl]-5-oxo[1,3]benzothiazolo[3,2a][1,8]naphthyridine-6-carboxa- IN). Cytosol/nuclear protein fractionation was carried out with NE-
mide} (1 ␮M) treatment was initiated by preincubation 2 h before PER Nuclear and Cytoplasmic extraction reagent according to the
serum stimulation. Rapamycin (25 ng/␮l) and PF-4708671 (PF) (20 ␮M) manufacturer’s recommendations (Thermo Fisher Scientific,
were added 1 h before stimulation with GM. DRB (5– 6-dichloro-1- Waltham, MA). All chemicals were purchased from Sigma unless
beta-D-ribofuranosylbenzimidazole) (75 ␮M), ␣-Amanitin (50 mg/ stated otherwise (Sigma-Aldrich, St. Louis, MO). Crude lysates were
ml), LY294002 (LY) (20 ␮M), cycloheximide (CHX) (50 ␮M) spun down at 12,000 g for 5 min and supernatants transferred to new
incubations started with the addition of GM for the experimental tubes. Protein concentration was assessed with the DC protein assay
groups and 0.1% DMSO was used for the DM and GM control (Bio-Rad, Hercules, CA) and lysates (if needed) diluted with lysis
group. All experiments were performed in 12-well plates except buffer before mixing 3:1 with 4X Laemmli buffer (Bio-Rad) contain-
48-h experiments (24-well plates), cytosolic and nuclear fraction- ing 10% ␤-mercaptoethanol. All samples were heated at 95°C for 10
ation (6-well plates), and chromatin immunoprecipitation (ChIP) min, cooled on ice, and stored at ⫺20°C until further use.
(10-cm plates). Western blotting and antibodies. Protein samples were separated by
Determination of myotube diameter. Myotube diameter was mea- SDS-page on 4 –12% polyacrylamide Criterion gradient gels and
sured manually in nine separate fields per treatment at 10 ⫻ using a transferred to PVDF membranes (Bio-Rad) activated in 100% meth-

Table 1. Primer sequences utilized in this study


Target Forward Reverse

45S Pre-rRNA (ETS) CCAAGTGTTCATGCCACGTG CGAGCGACTGCCACAAAAA


45S Pre-rRNA (ITS) FCCGGCTTGCCCGATTT GCCAGCAGGAACGAAACG
GAPDH ATGTTCCAGTATGACTCCACTCACG GAAGACACCAGTAGACTCCACGACA
c-Myc TGCGACTGACCCAACATCAG CCTGTCCTGGCTCGCAGATT
eIF4E CACCCTGCGCCCTTCA GGTCAGGGCCATCAGTCATG
TAF1C CACCCTGCGCCCTTCA GGTCAGGGCCATCAGTCATG
PAF53 TCAGAACAAGACTTTCAGGGACAA CTGCTTGGTGCTTCCAAAGG
TTF1 AAACGGAAGCATGCCTTCAG CACGGTAGTACACGAGCTTCCA
POLR1B TGGGAATCTGCGTTCTAAAACA TTCAGCTTGTCAGCCACAACA
UBF CGCGCAGCATACAAAGAATACA GTTTGGGCCTCGGAGCTT
Rrn3 ATTTTGAGCGCATTGTGTTGAGC GGGAGCATCTGGCGACTGTTC
tRNA CCTTCGATAGCTCAGCTGGTAGAGCGGAGG CGGAATTGAACCAGCGACCTAAGGATGTCC
RPS6 CATGGGGGAGTTGGACCATA TGGCCCTCTGTTCACACTA
RPS7 TGCCTCAATATCTGCGTCGG TTCTCCGGATGCCAAAGGAC
RPL11 TGCCTCAATATCTGCGTCGG TTCTCCGGATGCCAAAGGAC
45S rDNAp GACCAGTTGTTCCTTTGAGG ACCTATCTCCAGGTCCAATAG

AJP-Cell Physiol • doi:10.1152/ajpcell.00144.2016 • www.ajpcell.org


RIBOSOMAL DNA GENE REGULATION BY mTOR DURING MUSCLE HYPERTROPHY C665
anol. Western blotting was performed using standard techniques as synthesis rate ⫽ {[CPM (protein pellet)/protein in well (␮g)]/incuba-
previously described (28). In short, membranes were washed in a tion time (h)}/CPM (media).
Tris-buffered saline with 0.1% Tween (TBS-T 0.1%) and blocked in Statistical analysis. Differences between groups for rRNA, total
a protein containing buffer TBS-T 0.1%. Primary antibodies were protein, myotube diameter, gene expression, and Western blot data
diluted in a Tris-based buffer containing bovine serum albumin and were determined using the Student’s t-test or a one-way analysis of
sodium azide. Secondary antibodies were diluted in 5% nonfat dry variance (ANOVA) followed by the Newman-Keuls post hoc test (if
milk in TBS-T 0.1%. Primary antibodies towards mTOR (no. 2983), more than 2 groups were involved). Ch-IP data was analyzed inde-
PO4-mTOR Ser2448 (no. 5536), S6K1 (no. 9205), PO4-S6k1 Thr389 pendently per time point using a one-way ANOVA followed by the
(no. 9202) rpS6 (no. 2217), PO4-rpS6 Ser235/236 (no. 2211), and Newman-Keuls post hoc test. Significance was set at P ⬍ 0.05 for all
PDCD4 (no. 9535) were purchased from Cell Signaling (Beverly, statistical comparisons. Values are reported as means ⫾ SD unless
MA) and Nuclear Lamin A/C (no. sc-7293) and GAPDH (no. Sc- stated otherwise.
25778) from Santa Cruz Biotechnology (Santa Cruz, CA).
Chromatin immunoprecipitation. Myotubes were grown in 10-cm RESULTS
plates and treated on day 4 after differentiation. Cells were treated
with DM, GM, or GM ⫹ rapamycin (25 ng/␮l) for 1–12 h. Cells were Activation of mTOR signaling during myotube hypertrophy

Downloaded from http://ajpcell.physiology.org/ by 10.220.32.246 on October 15, 2016


fixed for 10 min at room temperature by addition of formaldehyde precedes transcription by all three polymerases. Serum stim-
(3.7%) directly to the culture medium for a final concentration of 1%, ulation of myotubes increased phosphorylation of mTOR/
and the reaction was quenched by the addition of glycine. Cells were S6K1/rpS6 within 3 h of stimulation (Fig. 1A). Following 48 h
collected in PBS and spun down at 1,400 rpm for 5 min at ⫹4°C.
of GM, total protein increased by ⫹50% (P ⬍ 0.001) com-
After removal of PBS, cells were lysed in ice-cold FA lysis buffer and
sonicated on a Branson 150 sonicator. Cross-linked and sonicated
pared with DM with a concomitant increase in myotube width
homogenates were stored at ⫺80°C until further use. Sample DNA (⫹28%, P ⬍ 0.001, Fig. 1B). Preceding any quantifiable
concentration was assessed using heat-reversed cross-linking over- hypertrophic response, we observed a transient increase in
night, proteinase K treatment and subsequent glycogen-assisted pre- rDNA transcriptional activity peaking at 12 h post GM (Fig.
cipitation of DNA following phenol/chloroform separation (Sigma). 1C). Total rRNA increased significantly by 12 h and continued
The air-dried DNA pellet was diluted in RNase/DNase/nucleotide- to increase throughout the 48-h stimulation period. Protein
free H2O and rehydrated for 1 h at ⫹65°C. DNA was quantified on a synthesis rates were elevated by 3 and 6 h post stimulation, and
Nanodrop 1000 at 260 nm. Ten micrograms of DNA was used per increased further concomitant with the increase in rRNA con-
immune-precipitation (IP) reaction, and samples were diluted to a tent. As ribosome biogenesis requires the coordinated function
final volume of 500 ␮l (IP dilution buffer: 16.7 mM Tris-HCl, 167 of all three polymerases, we also investigated the transcrip-
mM NaCl, 1.2 mM EDTA, 1.1% Triton-X 100, 0.01% SDS). Two to tional response of RNA Pol II and Pol III targets. Peak
three micrograms of antibody, mTOR (no. T2949, Sigma), H3K56-ac
transcription of rDNA genes by Pol I paralleled the increase in
(A-4026-050, Epigentek, Farmingdale, NY), H3 (ab1791, Abcam,
Cambridge, UK), H1 (sc-34464, Santa Cruz Biotechnology), and
Pol II transcription of several factors involved in ribosome
non-specific IgG (no. I5006, Sigma), was added to each reaction and biogenesis (c-Myc, eIF4E, Pol I regulon) and Pol III transcrip-
samples incubated overnight at ⫹4°C on a rotating wheel. Magnetic tion of tRNA (Fig. 1, D and E).
beads were blocked overnight in a blocking buffer (0.2 mg/ml glyco- Transcription of rDNA genes during myotube hypertrophy
gen, 1 mg/ml BSA, and 75 ␮g/ml salmon sperm DNA) and washed requires mTOR but not S6k1 signaling. To determine whether
before diluted in IP dilution buffer. Preblocked beads slurry (50 ␮l) signaling via PI3K/mTOR/S6K1 modulates rDNA transcrip-
was added to each reaction and samples incubated 60 min at room tion and muscle hypertrophy, we treated myotubes with PI3K
temperature. Beads were pulled down using a magnetic stand and (LY), mTOR (rapamycin), and S6K1 (PF) inhibitors. PI3K and
washed several times. Elution buffer (150 ␮l, 1% SDS, 100 mM mTOR inhibition decreased 45S pre-rRNA to the same degree
NAHCO3) was added to each tube and quickly spun down before (⫺34 – 40% from GM, P ⬍ 0.01) (Fig. 2A) but partial S6K1
heated at 30°C for 15 min. Eluted antibody-protein-DNA complexes inhibition by PF (⫺55%, P ⬍ 0.05, PO4-rpS6Ser235/236) did
were reverse crossed-linked, proteinase K digested, and the DNA not affect rDNA transcription. Furthermore, whereas rapamy-
extracted as described above for input DNA. Samples were quantified
using qPCR with primers targeting the rDNAp. The corresponding
cin significantly prevented the increase in rRNA and protein
PCR signal was normalized to a control DNA region (GAPDH accumulation following serum stimulation, inhibition of S6K1
promoter) and expressed as relative to DM treatment. by PF did not (Fig. 2, B and C). Because PF resulted in
Determination of protein synthesis. Myotubes grown in 6-well increased S6K1 Thr389 phosphorylation (Fig. 2D), we verified
plates were maintained in DM, or stimulated with GM, GM ⫹ 25 the functional S6K1 inhibition by determining the phosphory-
ng/ml rapamycin, or GM ⫹ 1 ␮M CX-5461 for 24 h after which cells lation status of its downstream target rpS6 and the abundance
were washed once with PBS and incubated in media containing of PDCD4. Serum-stimulated increase in rpS6 phosphorylation
35
S-labeled methionine (40 ␮Ci) for 90 min. Thereafter media was at Ser235/236 was partially prevented by PF and remained at DM
collected from each individual well as reference, and cells were levels, but rapamycin completely dephosphorylated it. Like-
washed three times in cold PBS and lysed in cell lysis buffer. Lysates wise, PDCD4 accumulated to control levels in myotubes
were spun down 5 min at 12,000 g to pellet insoluble debris and treated with PF although to a lesser extent than with rapamycin
supernatants transferred to new tubes. Lysates (15 ␮l) were used to (Fig. 2, E and F). To further validate the observation that
determine protein content using the DC protein assay (Bio-Rad).
reduced S6K1 activity did not compromise rDNA transcrip-
Protein was then precipitated using TCA to quantify 35S-incorpora-
tion. TCA precipitates were washed in acetone-HCl (10%) and resus- tion, we knocked down S6K1 using siRNA. Transfection with
pended in elution buffer (100 mM NaOH 1% SDS) at ⫹55°C. 35S S6K1 siRNA successfully decreased S6K1 mRNA levels by
incorporation was measured on a Wallac scintillation beta counter. ⫺60% (P ⬍ 0.001) and protein levels by ⫺90% (P ⬍ 0.001).
Counts per minute (CPM) from protein samples were normalized to Despite this marked reduction in S6K1 protein content, 45S
protein content (␮g), incubation time, and specific activity of the pre-rRNA levels were not affected (Fig. 2G). Thus, in agree-
media for each well (CPM) using the following formula: protein ment with our chemical inhibition data, knockdown of S6K1

AJP-Cell Physiol • doi:10.1152/ajpcell.00144.2016 • www.ajpcell.org


C666 RIBOSOMAL DNA GENE REGULATION BY mTOR DURING MUSCLE HYPERTROPHY

Downloaded from http://ajpcell.physiology.org/ by 10.220.32.246 on October 15, 2016

Fig. 1. Activation of mTOR signaling during myotube hypertrophy precedes transcription by all three polymerases. A, left: representative Western blots of the
mTOR pathway 1, 3, 6, 12, 24, or 48 h post serum stimulation in DM and GM groups. A, right: quantifications of PO4/total ratio of mTOR, S6K1, and rpS6
DM (n ⫽ 3) and GM (n ⫽ 3) 1 h to 48 h post stimulation. *Significantly different from DM, P ⬍ 0.05. †Significantly different from GM 3 h, P ⬍ 0.05.
**Significantly different from GM 1 h, P ⬍ 0.05. B: comparison of total protein content (left, n ⫽ 3/group) and mean diameter of myotubes (right) following
48 h serum stimulation period in DM (n ⫽ 314) and GM (n ⫽ 254). C: % increase in protein synthesis (o), rDNA transcription (䊐), and RNA content (Œ)
compared with DM control per time point, n ⫽ 3– 4 per group. D: relative gene expression of Pol II targets in DM (n ⫽ 10) and GM (n ⫽ 10) 12 h following
serum stimulation. E: tRNA 12 h following serum stimulation in DM (n ⫽ 3) and GM (n ⫽ 3) 12 h following serum stimulation. GAPDH was used for qRT-PCR
normalization in D and E. Open bars, DM; black bars, GM. *Significantly different from DM, P ⬍ 0.05. *,†,‡Significantly different from DM, P ⬍ 0.05 for
protein synthesis, rDNA transcription, and rRNA content, respectively. DM set as 100% in B, D, and E. Data are shown as means ⫾ SD.

AJP-Cell Physiol • doi:10.1152/ajpcell.00144.2016 • www.ajpcell.org


RIBOSOMAL DNA GENE REGULATION BY mTOR DURING MUSCLE HYPERTROPHY C667

Downloaded from http://ajpcell.physiology.org/ by 10.220.32.246 on October 15, 2016


Fig. 2. Transcription of rDNA genes during myotube hypertrophy requires mTOR but not S6k1 signaling. A: 45S pre-rRNA transcription (ITS) 12 h following
serum stimulation in myotubes treated with DM, GM, GM ⫹ 20 ␮M LY, GM ⫹ 25 ng/ml rapamycin, or GM ⫹ 20 ␮M PF-4708671 (DM set as 100%, n ⫽
3/group). GAPDH was used for qRT-PCR normalization. B and C: changes in rRNA and total protein content in serum stimulated myotubes at 48 h post
stimulation, n ⫽ 3– 4 per group. *Significantly different from DM, P ⬍ 0.01. †Significantly different from GM, P ⬍ 0.05. Legend indicates media and treatments
for A–C. D–F: quantifications of PO4/total ratio of S6K1 and rpS6, and PDCD4/GAPDH ratio in DM, GM, GM ⫹ 25 ng/ml rapamycin and GM ⫹ 20 ␮M
PF-4708671 (n ⫽ 3/group). Insets: representative Western blots of PO4-S6K1, PO4-rpS6, or PDCD4 (top) and total S6K1, rpS6, or GAPDH (bottom).
*Significantly different from DM, P ⬍ 0.05. †Significantly different from GM, P ⬍ 0.01. Legend indicates media and treatment for D–F. G: gene expression
of S6K1 and 45S pre-rRNA (ITS) in GM ⫹ scrambled siRNA and GM ⫹ S6K1 siRNA 12 h following serum stimulation (n ⫽ 3/group). Insets: representative
Western blots of S6K1 (top) and GAPDH (bottom) in siRNA-treated myotubes. Data are shown as means ⫾ SD. GAPDH was used for qRT-PCR normalization.

did not affect Pol I-dependent rDNA transcription in hypertro- phy with ␣-Amanitin displaying cytotoxic effects by 48 h.
phying myotubes. (data not shown).
Protein synthesis and Pol II transcription modulate rDNA Selective inhibition of RNA Pol I transcription blocks
transcription and hypertrophy. To determine the mTOR-de- protein synthesis and myotube hypertrophy. The observed
pendent effects on Pol I transcription and myotube hypertro- effect of rapamycin on rDNA transcription could, in addi-
phy, we asked whether blocking protein synthesis or Pol II tion to partially being mediated via its role in translation and
transcription could equally impact rDNA transcription. Direct Pol II transcription, be the result of a direct mTOR-mediated
inhibition of translation by CHX resulted in a more pronounced effect on RNA Pol I. To investigate whether increased RNA
suppression of protein synthesis and rDNA transcription com- Pol I activity is necessary for myotube hypertrophy, we
pared with rapamycin (Fig. 3, A and B). Both CHX and treated myotubes with the selective RNA Pol I inhibitor
rapamycin decreased mRNA levels of key proteins involved in CX-5461. Blocking Pol I transcription prevented 45S pre-
rDNA transcription in a bimodal and time-dependent manner rRNA synthesis (Fig. 4A), and the corresponding increase in
(Fig. 3C). Inhibiting Pol II transcription directly using either rRNA (Fig. 4B), without negatively affecting the expression
DRB or ␣-Amanitin caused a suppression of protein synthesis of ribosomal protein genes (Fig. 4C). Surprisingly, it also
(Fig. 3D) and a progressive decrease in mRNAs of most genes suppressed the increase in protein synthesis associated with
investigated with the exception of c-Myc (Fig. 3C). Further- myotube hypertrophy (Fig. 4D), and prevented protein ac-
more, inhibiting transcription by Pol II with either DRB or cumulation (Fig. 4E) and the increase in myotube diameter
␣-Amanitin resulted in an inconsistent expression of the 45S following serum stimulation without affecting myotube vi-
pre-rRNA transcript (Fig. 3, E and F) and prevented hypertro- ability (Fig. 4F).

AJP-Cell Physiol • doi:10.1152/ajpcell.00144.2016 • www.ajpcell.org


C668 RIBOSOMAL DNA GENE REGULATION BY mTOR DURING MUSCLE HYPERTROPHY

Downloaded from http://ajpcell.physiology.org/ by 10.220.32.246 on October 15, 2016


Fig. 3. Protein synthesis and Pol II transcription modulate rDNA transcription and hypertrophy. A: protein synthesis rates at 24 h post serum stimulation. DM
(n ⫽ 4), GM (n ⫽ 4), GM ⫹ 25 ng/ml rapamycin (n ⫽ 4), and GM ⫹ 50 ␮M CHX (n ⫽ 3). *Significantly different from DM, P ⬍ 0.01. †Significantly different
from GM, P ⬍ 0.01. ‡Significantly different from all groups, P ⬍ 0.001. B: 45S pre-rRNA (ITS) expression 12 h post serum stimulation supplemented with
50 ␮M CHX for 1 or 12 h. *Significantly different from DM, P ⬍ 0.001. †Significantly different from all other groups, P ⬍ 0.01. C: graphical illustration of
changes in mRNA levels in the following groups: DM, GM, GM ⫹ 25 ng/ml rapamycin (12 h), GM ⫹ CHX, GM ⫹ DRB, GM ⫹ ␣-Amanitin. CHX, DRB,
and ␣-Amanitin treatments were performed for 1 or 12 h. qPCR data were used for hierarchical clustering using the R script hetmap.2 from R/gplots and
ColorBrewer package. The color legend displays the transformed signal intensity range in the dataset. D: protein synthesis rates at 24 h post serum stimulation
as described in A. DM, GM, GM ⫹ 75 ␮M DRB and GM ⫹ 50 mg/ml ␣-Amanitin (n ⫽ 4/group). *Significantly different from DM, P ⬍ 0.01. †Significantly
different from GM, P ⬍ 0.01. E: 45S pre-rRNA expression (ETS and ITS) following 1 and 12 h 75 ␮M DRB. F: 45S pre-rRNA expression (ETS and ITS)
following 1 and 12 h 50 mg/ml ␣-Amanitin. *Significantly different from DM. †Significantly different from GM, P ⬍ 0.05. ‡Significantly different from all
groups, P ⬍ 0.05. Data are shown as means ⫾ SD. GAPDH or 28S was used for qRT-PCR normalization.

Rapamycin inhibits mTOR enrichment at the rDNA pro- accumulation and myotube hypertrophy and is consistent with
moter and represses active chromatin marks. While the cyto- transcription by all three polymerases (21). We have previ-
plasmic function of mTOR is well defined, its nuclear function ously demonstrated that inhibition of mTOR with rapamycin
remains elusive. During hypertrophy, rapamycin did not prevents rRNA accumulation (8) and skeletal muscle hyper-
alter mTOR compartmentalization as evidenced by its con- trophy (2, 27, 33). Thus the involvement of mTOR signaling in
tinuous nuclear localization despite interrupted rDNA tran- rDNA gene transcription appears to be necessary for ribosome
scription. Rapamycin treatment reduced mTOR enrichment production and muscle hypertrophy. To further dissect the
at the rDNAp to control levels (Fig. 5, A and B) and was hierarchical role of mTOR in muscle rDNA transcription, we
sufficient to significantly reduce H3K56ac and increase H1 at employed a biochemical approach by targeting PI3K/mTOR/
the rDNAp (Fig. 5, C and D). S6K1 signaling. Blocking PI3K or mTOR signaling resulted in
a similar level of inhibition of rDNA transcription, but pre-
DISCUSSION
venting an increase in S6K1 activity did not. We interpret this
Muscle ribosome content is a major determinant of protein as both PI3K and mTOR being involved in regulating signaling
synthesis rates (25). In the present study we investigated the leading to elevated rDNA transcription (4, 11, 14) and there-
involvement of mTOR signaling in rDNA transcription and fore the negative effect of rapamycin on Pol I transcription,
ribosome production during myotube hypertrophy. As we have ribosome production, and myotube hypertrophy is mediated by
previously shown in vivo, rDNA transcription is an early event mTOR and not S6K1 activity. Even though rapamycin mark-
during skeletal muscle hypertrophy (41). This precedes the edly suppressed rDNA transcription, our present results do not
accumulation of rRNA to support the need to increase muscle allow us to distinguish between mTORC1 and mTORC2, and
translational capacity during hypertrophy (8, 17, 27). In this because both complexes can be localized to the nucleus, further
study, we show that mTOR signaling also precedes rRNA studies should elucidate whether mTORC1 and/or mTORC2

AJP-Cell Physiol • doi:10.1152/ajpcell.00144.2016 • www.ajpcell.org


RIBOSOMAL DNA GENE REGULATION BY mTOR DURING MUSCLE HYPERTROPHY C669

Downloaded from http://ajpcell.physiology.org/ by 10.220.32.246 on October 15, 2016


Fig. 4. Selective inhibition of RNA Pol I transcription blocks protein synthesis and myotube hypertrophy. A: CX-5461 decreases 45S pre-rRNA abundance in
serum-stimulated myotubes. DM, GM, GM ⫹ 1 ␮M CX-5461 (n ⫽ 3/group). *Significantly different from DM, P ⬍ 0.05. †Significantly different from GM,
P ⬍ 0.05. B: rRNA content in myotubes serum stimulated for 48 h. DM, GM, and GM ⫹ 1 ␮M CX-5461 (n ⫽ 4/group). *Significantly different from DM, P ⬍
0.001. †Significantly different from GM, P ⬍ 0.001. C: mRNA abundance of rpS6, rpS7, and rpL11 12 h post serum stimulation in response to CX-5461
treatment. DM, GM, and GM ⫹ 1 ␮M CX-5461 (n ⫽ 3/group). *Significantly different from all groups, P ⬍ 0.05. D: effect of CX-5461 on protein synthesis
at 24 h post serum stimulation. DM, GM, and GM ⫹ 1 ␮M CX-5461 (n ⫽ 4/group). *Significantly different from DM, P ⬍ 0.01. †Significantly different from
GM, P ⬍ 0.01. E: total protein content in myotubes serum stimulated for 48 h. DM, GM, and GM ⫹ 1 ␮M CX-5461 (n ⫽ 4/group). *Significantly different
from DM, P ⬍ 0.01. †Significantly different from GM, P ⬍ 0.01. F: mean diameter of myotubes following 48 h serum stimulation. Left: comparison between
DM (n ⫽ 314), GM (n ⫽ 254), and GM ⫹ 1 ␮M CX-5461 (n ⫽ 416). ***Significantly different from DM and GM ⫹ 1 ␮M CX-5461, P ⬍ 0.001. Right:
representative light microscopic images of myotubes treated with DM, GM, or GM ⫹ 1 ␮M CX-5461. Image inset: scale bar ⫽ 100 ␮m. Data are shown as
means ⫾ SD. A and C: GAPDH was used for qRT-PCR normalization.

can regulate rDNA transcription. However, although not tested S6Ks are dispensable for cardiac growth in vivo. In our study,
in the present study, it is likely that mTORC1 and not despite evidence of a decreased S6K1 function, no effect on
mTORC2 is involved in rDNA transcription as RAPTOR has 45S pre-rRNA levels or myotube hypertrophy was observed.
been previously shown to associate with the rDNA transcrip- Thus, we interpret our findings as indicating that the repressive
tion machinery, i.e., colocalizes with fibrillarin and Pol I at effect of mTOR inhibition by rapamycin on rDNA transcrip-
nucleoli (40). Surprisingly, S6K1 inhibition did not affect tion does not involve S6K1 activity. Clearly the precise roles
rDNA transcription. It is possible that the lack of effect seen and interactions between the S6Ks signaling in muscle hyper-
with partial biochemical S6K1 inhibition was due to the re- trophy require further investigation (24).
mainder activity of this kinase, which may have sufficed to We have previously demonstrated that mTOR inhibition by
support hypertrophy. For this reason we targeted S6K1 with rapamycin prevented the increase in rRNA content and blocked
siRNA and found that S6K1 silencing also failed to prevent myotube hypertrophy (27). To distinguish between the protein
rDNA transcription. One possible explanation is that S6K2 synthesis inhibitory effects of blocking mTOR (i.e., cap-de-
may have compensated for the absence of S6K1. In S6K1⫺/⫺ pendent translation) and its transcriptional functions, we first
mice, muscle mass is smaller in the presence of intact S6K2 but used CHX to block global protein synthesis. CHX had a more
yet they are viable, which suggests that S6K2 can compensate dramatic effect on suppressing protein synthesis and rDNA
for some S6K1 functions (29, 37). In cardiomyocytes, rRNA transcription than rapamycin. This is likely due to the depletion
accumulation and hypertrophic growth in vitro have been of essential factors required for Pol I transcription (7), and
suggested to depend on S6K1 via UBF1 phosphorylation (11). suggests that mTOR may regulate rDNA transcription by a
But because both S6K1⫺/⫺ and S6K1⫺/⫺/S6K2⫺/⫺ mice are separate mechanism other than global protein synthesis. It also
able to respond to physiological, pathological, and IGF-1- indicates that Pol I transcription requires protein synthesis to
induced cardiac hypertrophy (23), it becomes apparent that the maintain basal ribosome production as CHX decreased rDNA

AJP-Cell Physiol • doi:10.1152/ajpcell.00144.2016 • www.ajpcell.org


C670 RIBOSOMAL DNA GENE REGULATION BY mTOR DURING MUSCLE HYPERTROPHY

Fig. 5. Rapamycin inhibits mTOR enrich-


ment at the rDNA promoter and represses
active chromatin. A: representative Western
blot of mTOR, GAPDH, and nuclear lamina
A/C in whole (W), cytoplasmic (C), and
nuclear (N) lysates from myotubes treated
with DM, GM, or GM ⫹ 25 ng/ml rapamy-
cin for 3 h. Bottom band in the mTOR blot
is nonspecific. B: chromatin immunoprecipi-
tation of mTOR at 1, 3, 6, and 12 h post
serum stimulation in DM, GM, and GM ⫹
25 ng/ml rapamycin (n ⫽ 3/group). qPCR
data from rDNA region normalized to
GAPDH promoter region. *Significantly dif-
ferent from DM, P ⬍ 0.05. †Significantly
different from GM, P ⬍ 0.001. C: chromatin
immunoprecipitation of H3K56ac relative to

Downloaded from http://ajpcell.physiology.org/ by 10.220.32.246 on October 15, 2016


H3 at the rDNA promoter 1 and 12 h post
serum stimulation in DM, GM, and GM ⫹
25 ng/ml rapamycin. *Significantly different
from GM, P ⬍ 0.05. D: chromatin immuno-
precipitation of H1 at 1 and 12 h in DM,
GM, and GM ⫹ 25 ng/ml rapamycin. qPCR
data from rDNA region normalized to
GAPDH promoter region. **Significantly
different from GM, P ⬍ 0.01. Data are
shown as means ⫾ SD. For B–D, data are
expressed as fold change relative to DM.

transcription below control (DM) levels. Because mTOR can similar extent. However, specific Pol I inhibition blunted
also control Pol II transcription (12, 30), we determined rDNA transcription and rRNA production without decreasing
whether specific Pol II inhibition affected rDNA gene expres- the expression of ribosomal proteins (i.e., Class II genes).
sion by Pol I in a manner similar to rapamycin. When directly Surprisingly, arresting Pol I, in addition to preventing the
arresting Pol II with either DRB or ␣-Amanitin, it became increase in protein content and myotube size, also blunted the
evident that Pol II transcription is necessary for ribosome increase in protein synthesis. This was an unexpected finding
production and hypertrophy, as ␣-Amanitin was sufficient to as the existing ribosomes should have sufficed for at least an
block Pol I transcription. Because DRB, which inhibits Pol II increase in protein synthesis (translational efficiency) before
and to a lower extent Pol I (36), had a similar effect on 45S any increase in ribosome production (translational capacity).
pre-rRNA, we consider the inhibition of Class II genes by These findings indicate that transcription of rDNA genes by
␣-Amanitin a Pol II specific effect, and an indication that Pol RNA Pol I is necessary for myotube hypertrophy, not only to
II transcription is necessary for rDNA transcription. This increase ribosome production but also to regulate protein
highlights the dependency of Pol I on Class II genes to initiate synthesis. These data also suggest the presence of a “coupling”
rDNA transcription during hypertrophy and suggests that mechanism, either direct or indirect, that links rDNA transcrip-
mTOR signaling may indirectly control Pol I by modulating tion to the translational machinery independently of ribosome
Class II gene expression. Intriguingly, arresting Pol II tran- content.
scription resulted in bidirectional changes in some mRNAs Based on the above, and in line with our previous results
important for ribosome biogenesis and other growth genes, (27), it is clear that mTOR signaling is directly involved in
again, demonstrating that blocking protein synthesis has a very ribosome production and myotube hypertrophy. While mTOR
different effect than Pol II transcription on Pol I-dependent operates at the cytoplasmic level to regulate mRNA translation
rDNA transcription. These findings provide indirect evidence (42), we demonstrate here that in skeletal muscle mTOR is also
that mTOR signaling to protein synthesis may be distinct from nuclear and binds to the rDNAp. Blocking mTOR signaling
that to Pol II transcription. Furthermore, blocking Pol II tran- with rapamycin did not result in its nuclear exclusion, but
scription with either DRB or ␣-Amanitin produced an imbal- significantly reduced mTOR-rDNAp interaction. This suggests
ance between 45S pre-rRNA ETS and ITS which suggests an that there is a fraction of the mTOR pool that is nuclear
important role of Pol II transcription on Pol I elongation more regardless of its DNA binding activity at the rDNAp. Similarly,
than initiation. Altogether, these data surface an emerging role the p110 subunit of PI3K has been reported to have nuclear
of mTOR signaling in transcriptional regulation of ribosome functions as a kinase for the Pol I factor UBF1 (4). Thus a
synthesis by partially controlling mRNA translation and Class likely function of nuclear PI3K/mTOR signaling appears to be
II gene expression. This conclusion is also supported by the the phosphorylation and activation PIC factors such as UBF
fact that ␣-Amanitin is specific for Pol II transcription and does and Rrn3 to modulate rDNA transcription (11, 22) and pro-
not interfere with Pol I (19). cessing (13). Another possible involvement of nuclear mTOR
To establish whether mTOR-dependent ribosome produc- in ribosome biogenesis appears to be the regulation of chro-
tion is directly mediated by Pol I, we specifically inhibited Pol matin. Inhibition of mTOR by rapamycin decreased H3K56ac
I transcription with CX-5461 (5). Compared with rapamycin, levels at the rDNAp, a chromatin mark consistent with tran-
CX-5461 prevented the increase in rDNA transcription to a scriptional silencing. Although the exact mechanism by which

AJP-Cell Physiol • doi:10.1152/ajpcell.00144.2016 • www.ajpcell.org


RIBOSOMAL DNA GENE REGULATION BY mTOR DURING MUSCLE HYPERTROPHY C671
mTOR signaling modulates H3K56ac in mammalian cells ACKNOWLEDGMENTS
remains unknown, this observation validates recent data from We are thankful to Dr. R. N. Laribee for sharing insight about the work
yeast implicating TORC1 signaling in the regulation of presented in Chen et al. (3).
H3K56ac and rDNA transcription. Chen et al. (3) have recently
GRANTS
shown that genetic disruption of the TORC1 subunit tco89
(tco89⌬), nutrient-dependent TORC1 activators (ego3⌬), or This work was supported by grants from the Swedish Research Council
(Vetenskapsradet VRK2008-67X-20797-01-04), Centrum för Idrottsforskning
pharmacological inhibition of TORC1 with rapamycin, nega- (P2011-01-0133), Kung Gustaf V:s 80 Årsfond (FAI2009-0065), and Reuma-
tively regulates global H3K56ac. This indicates that mTOR tikerförbundet (R-21211), Diabetesfonden (DIA2012-071), the Karolinska
signaling may be necessary for the control of this specific Institute Strategic Diabetes Program, and the Department of Kinesiology at
chromatin mark. While this connection is likely indirect, it may The Pennsylvania State University to G. A. Nader.
occur via Sirtuin deacetylases as deletions of yeast Hst3 or DISCLOSURES
Hst4, which play a critical role in maintaining H3K56 hy-
poacetylation, can restore H3K56ac levels (43). Specifically, No conflicts of interest, financial or otherwise, are declared by the author(s).
H3K56ac can regulate rDNA chromatin by controlling Hmo1 AUTHOR CONTRIBUTIONS

Downloaded from http://ajpcell.physiology.org/ by 10.220.32.246 on October 15, 2016


(a functional homolog of UBF1) (1) binding to rDNAp, as
F.v.W., C.L., and N.A. performed experiments; F.v.W., C.L., N.A., and
Hmo1 binding to rDNAp is reduced in H3K56(A) mutants. G.A.N. analyzed data; F.v.W., C.L., and G.A.N. interpreted results of exper-
These data, although from a different model system, are similar iments; F.v.W., C.L., and N.A. prepared figures; F.v.W., N.A., and G.A.N.
to our findings where inhibition of mTOR signaling results in edited and revised manuscript; F.v.W., C.L., N.A., and G.A.N. approved final
decreased H3K56ac levels and likely reduces chromatin acces- version of manuscript; G.A.N. conception and design of research; G.A.N.
drafted manuscript.
sibility to the rDNAp. Although we appreciate the limitations
of the present study in that we do not provide a mechanistic REFERENCES
link between the inhibition of mTOR signaling by rapamycin
1. Albert B, Colleran C, Leger-Silvestre I, Berger AB, Dez C, Normand
and the decrease in H3K56ac, it is safe to speculate that these C, Perez-Fernandez J, McStay B, Gadal O. Structure-function analysis
two events may be linked and both contribute to the suppres- of Hmo1 unveils an ancestral organization of HMG-Box factors involved
sion of rDNA transcription (3, 18). This points towards a in ribosomal DNA transcription from yeast to human. Nucleic Acids Res
conserved link between mTOR signaling, H3K56ac, and rDNA 41: 10135–10149, 2013.
transcription and is supported by the enrichment of histone H1 2. Bodine SC, Stitt TN, Gonzalez M, Kline WO, Stover GL, Bauerlein R,
Zlotchenko E, Scrimgeour A, Lawrence JC, Glass DJ, Yancopoulos
at the rDNAp, which reinforces the notion of increased silent GD. Akt/mTOR pathway is a crucial regulator of skeletal muscle hyper-
chromatin formation at the rDNAp upon mTOR inhibition. H1 trophy and can prevent muscle atrophy in vivo. Nat Cell Biol 3: 1014 –
linker histones repress transcription by stabilizing higher-order 1019, 2001.
chromatin structure (38), and preventing access of transcription 3. Chen H, Fan M, Pfeffer LM, Laribee RN. The histone H3 lysine 56
acetylation pathway is regulated by target of rapamycin (TOR) signaling
factors to DNA binding sites (31). To our knowledge, this is and functions directly in ribosomal RNA biogenesis. Nucleic Acids Res
the first observation linking mTOR signaling to H1 enrichment 40: 6534 –6546, 2012.
at rDNA genes, which suggests that mTOR signaling could 4. Drakas R, Tu X, Baserga R. Control of cell size through phosphorylation
regulate rDNA transcription by remodeling chromatin archi- of upstream binding factor 1 by nuclear phosphatidylinositol 3-kinase.
tecture. Collectively, a decrease in H3K56ac and increased H1 Proc Natl Acad Sci USA 101: 9272–9276, 2004.
5. Drygin D, Lin A, Bliesath J, Ho CB, O’Brien SE, Proffitt C, Omori M,
at the rDNAp supports the interpretation that, similar to yeast, Haddach M, Schwaebe MK, Siddiqui-Jain A, Streiner N, Quin JE,
rapamycin-mediated mTOR inhibition negatively affects chro- Sanij E, Bywater MJ, Hannan RD, Ryckman D, Anderes K, Rice WG.
matin remodeling, and consequently rDNA gene silencing, in Targeting RNA polymerase I with an oral small molecule CX-5461
postmitotic myotubes. inhibits ribosomal RNA synthesis and solid tumor growth. Cancer Res 71:
1418 –1430, 2011.
In summary, we provide evidence indicating a necessary and 6. Glass DJ. PI3 kinase regulation of skeletal muscle hypertrophy and
coordinating role of mTOR signaling in protein synthesis, both atrophy. Curr Topics Microbiol Immunol 346: 267–278, 2010.
Pol I and Pol II transcription, and ribosome production during 7. Gokal PK, Cavanaugh AH, Thompson EA Jr. The effects of cyclohex-
myotube hypertrophy. Our data indicate that in this setting, an imide upon transcription of rRNA, 5 S RNA, and tRNA genes. J Biol
increase in Pol I-dependent rDNA transcription is necessary for Chem 261: 2536 –2541, 1986.
8. Goldspink DF. The influence of activity on muscle size and protein
protein synthesis and enhanced ribosome production. We also turnover. J Physiol 264: 283–296, 1977.
demonstrate the necessity of Pol II transcription and proteins 9. Goodman CA, Frey JW, Mabrey DM, Jacobs BL, Lincoln HC, You
synthesis for Pol I transcription of rDNA genes. The present JS, Hornberger TA. The role of skeletal muscle mTOR in the regulation
study also provides novel findings suggesting that during of mechanical load-induced growth. J Physiol 589: 5485–5501, 2011.
10. Grummt I. Regulation of mammalian ribosomal gene transcription by
myotube hypertrophy, mTOR associates with the rDNAp and RNA polymerase I. Progr Nucleic Acid Res Mol Biol 62: 109 –154, 1999.
may modulate chromatin remodeling in a rapamycin-sensitive 11. Hannan KM, Brandenburger Y, Jenkins A, Sharkey K, Cavanaugh A,
manner. Although the precise mechanism(s) by which mTOR Rothblum L, Moss T, Poortinga G, McArthur GA, Pearson RB,
regulates chromatin structure to control transcription remains Hannan RD. mTOR-dependent regulation of ribosomal gene transcription
to be elucidated, it is clear that mTOR signaling is required for requires S6K1 and is mediated by phosphorylation of the carboxy-terminal
activation domain of the nucleolar transcription factor UBF. Mol Cell Biol
the maintenance of transcriptionally competent chromatin at 23: 8862–8877, 2003.
the rDNAp as reflected by changes in H3K56ac and histone 12. Hardwick JS, Kuruvilla FG, Tong JK, Shamji AF, Schreiber SL.
H1. Thus nuclear mTOR signaling may be responsible for Rapamycin-modulated transcription defines the subset of nutrient-sensi-
priming and/or maintaining the transcriptionally competent tive signaling pathways directly controlled by the Tor proteins. Proc Natl
Acad Sci USA 96: 14866 –14870, 1999.
state of the rDNA genes, and provides a novel possible mech- 13. Iadevaia V, Zhang Z, Jan E, Proud CG. mTOR signaling regulates the
anism for the regulation of gene expression during hypertrophy processing of pre-rRNA in human cells. Nucleic Acids Res 40: 2527–2539,
in postmitotic skeletal muscle. 2012.

AJP-Cell Physiol • doi:10.1152/ajpcell.00144.2016 • www.ajpcell.org


C672 RIBOSOMAL DNA GENE REGULATION BY mTOR DURING MUSCLE HYPERTROPHY

14. James MJ, Zomerdijk JC. Phosphatidylinositol 3-kinase and mTOR sion during human skeletal muscle hypertrophy. J Appl Physiol 116:
signaling pathways regulate RNA polymerase I transcription in response 693–702, 2014.
to IGF-1 and nutrients. J Biol Chem 279: 8911–8918, 2004. 29. Ohanna M, Sobering AK, Lapointe T, Lorenzo L, Praud C, Petrou-
15. Kubica N, Bolster DR, Farrell PA, Kimball SR, Jefferson LS. Resis- lakis E, Sonenberg N, Kelly PA, Sotiropoulos A, Pende M. Atrophy of
tance exercise increases muscle protein synthesis and translation of eu- S6K1(⫺/⫺) skeletal muscle cells reveals distinct mTOR effectors for cell
karyotic initiation factor 2Bepsilon mRNA in a mammalian target of cycle and size control. Nat Cell Biol 7: 286 –294, 2005.
rapamycin-dependent manner. J Biol Chem 280: 7570 –7580, 2005. 30. Preiss T, Baron-Benhamou J, Ansorge W, Hentze MW. Homodirec-
16. Laplante M, Sabatini DM. mTOR signaling in growth control and tional changes in transcriptome composition and mRNA translation in-
disease. Cell 149: 274 –293, 2012. duced by rapamycin and heat shock. Nat Struct Biol 10: 1039 –1047, 2003.
17. Laurent GJ, Sparrow MP, Millward DJ. Turnover of muscle protein in 31. Ramakrishnan V. Histone H1 and chromatin higher-order structure. Crit
the fowl. Changes in rates of protein synthesis and breakdown during Rev Eukaryot Gene Express 7: 215–230, 1997.
hypertrophy of the anterior and posterior latissimus dorsi muscles. 32. Rohde JR, Cardenas ME. The tor pathway regulates gene expression by
Biochem J 176: 407–417, 1978. linking nutrient sensing to histone acetylation. Mol Cell Biol 23: 629 –635,
18. Li H, Tsang CK, Watkins M, Bertram PG, Zheng XF. Nutrient 2003.
regulates Tor1 nuclear localization and association with rDNA promoter. 33. Rommel C, Bodine SC, Clarke BA, Rossman R, Nunez L, Stitt TN,
Nature 442: 1058 –1061, 2006. Yancopoulos GD, Glass DJ. Mediation of IGF-1-induced skeletal myo-
19. Lindell TJ, Weinberg F, Morris PW, Roeder RG, Rutter WJ. Specific tube hypertrophy by PI(3)K/Akt/mTOR and PI(3)K/Akt/GSK3 pathways.

Downloaded from http://ajpcell.physiology.org/ by 10.220.32.246 on October 15, 2016


inhibition of nuclear RNA polymerase II by alpha-amanitin. Science 170: Nat Cell Biol 3: 1009 –1013, 2001.
447–449, 1970. 34. Rosner M, Hengstschlager M. Cytoplasmic and nuclear distribution of
20. Marino JS, Tausch BJ, Dearth CL, Manacci MV, McLoughlin TJ, the protein complexes mTORC1 and mTORC2: rapamycin triggers de-
Rakyta SJ, Linsenmayer MP, Pizza FX. Beta2-integrins contribute to phosphorylation and delocalization of the mTORC2 components rictor and
skeletal muscle hypertrophy in mice. Am J Physiol Cell Physiol 295: sin1. Hum Mol Genet 17: 2934 –2948, 2008.
C1026 –C1036, 2008.
35. Russell J, Zomerdijk JC. RNA-polymerase-I-directed rDNA transcrip-
21. Mayer C, Grummt I. Ribosome biogenesis and cell growth: mTOR
tion, life and works. Trends Biochem Sci 30: 87–96, 2005.
coordinates transcription by all three classes of nuclear RNA polymerases.
36. Scheer U, Hugle B, Hazan R, Rose KM. Drug-induced dispersal of
Oncogene 25: 6384 –6391, 2006.
transcribed rRNA genes and transcriptional products: immunolocalization
22. Mayer C, Zhao J, Yuan X, Grummt I. mTOR-dependent activation of
and silver staining of different nucleolar components in rat cells treated
the transcription factor TIF-IA links rRNA synthesis to nutrient availabil-
with 5,6-dichloro-beta-D-ribofuranosylbenzimidazole. J Cell Biol 99: 672–
ity. Genes Dev 18: 423–434, 2004.
23. McMullen JR, Shioi T, Zhang L, Tarnavski O, Sherwood MC, Dor- 679, 1984.
fman AL, Longnus S, Pende M, Martin KA, Blenis J, Thomas G, 37. Shima H, Pende M, Chen Y, Fumagalli S, Thomas G, Kozma SC.
Izumo S. Deletion of ribosomal S6 kinases does not attenuate patholog- Disruption of the p70(s6k)/p85(s6k) gene reveals a small mouse pheno-
ical, physiological, or insulin-like growth factor 1 receptor-phosphoinosi- type and a new functional S6 kinase. EMBO J 17: 6649 –6659, 1998.
tide 3-kinase-induced cardiac hypertrophy. Mol Cell Biol 24: 6231–6240, 38. Thoma F, Koller T. Influence of histone H1 on chromatin structure. Cell
2004. 12: 101–107, 1977.
24. Mieulet V, Roceri M, Espeillac C, Sotiropoulos A, Ohanna M, Oors- 39. Tsang CK, Liu H, Zheng XF. mTOR binds to the promoters of RNA
chot V, Klumperman J, Sandri M, Pende M. S6 kinase inactivation polymerase I- and III-transcribed genes. Cell Cycle 9: 953–957, 2010.
impairs growth and translational target phosphorylation in muscle cells 40. Vazquez-Martin A, Cufi S, Oliveras-Ferraros C, Menendez JA. Rap-
maintaining proper regulation of protein turnover. Am J Physiol Cell tor, a positive regulatory subunit of mTOR complex 1, is a novel phos-
Physiol 293: C712–C722, 2007. phoprotein of the rDNA transcription machinery in nucleoli and chromo-
25. Millward DJ, Garlick PJ, James WP, Nnanyelugo DO, Ryatt JS. somal nucleolus organizer regions (NORs). Cell Cycle 10: 3140 –3152,
Relationship between protein synthesis and RNA content in skeletal 2011.
muscle. Nature 241: 204 –205, 1973. 41. von Walden F, Casagrande V, Ostlund Farrants AK, Nader GA.
26. Moss T, Langlois F, Gagnon-Kugler T, Stefanovsky V. A housekeeper Mechanical loading induces the expression of a Pol I regulon at the onset
with power of attorney: the rRNA genes in ribosome biogenesis. Cell Mol of skeletal muscle hypertrophy. Am J Physiol Cell Physiol 302: C1523–
Life Sci CMLS 64: 29 –49, 2007. C1530, 2012.
27. Nader GA, McLoughlin TJ, Esser KA. mTOR function in skeletal 42. Wang X, Proud CG. The mTOR pathway in the control of protein
muscle hypertrophy: increased ribosomal RNA via cell cycle regulators. synthesis. Physiology 21: 362–369, 2006.
Am J Physiol Cell Physiol 289: C1457–C1465, 2005. 43. Yang B, Miller A, Kirchmaier AL. HST3/HST4-dependent deacetyla-
28. Nader GA, von Walden F, Liu C, Lindvall J, Gutmann L, Pistilli EE, tion of lysine 56 of histone H3 in silent chromatin. Mol Biol Cell 19:
Gordon PM. Resistance exercise training modulates acute gene expres- 4993–5005, 2008.

AJP-Cell Physiol • doi:10.1152/ajpcell.00144.2016 • www.ajpcell.org

You might also like