You are on page 1of 22

J Physiol 0.

0 (2021) pp 1–22 1

Genetic and epigenetic regulation of skeletal muscle


ribosome biogenesis with exercise
Vandré C. Figueiredo1,2 , Yuan Wen2,3 , Björn Alkner4 , Rodrigo Fernandez-Gonzalo5 ,
Jessica Norrbom6 , Ivan J. Vechetti Jr2,7 , Taylor Valentino2,3 , C. Brooks Mobley2,3 , Gabriel E. Zentner8 ,
Charlotte A. Peterson1,2,3 , John J. McCarthy2,3 , Kevin A. Murach1,2,# and Ferdinand von Walden2,3,9,#
1
Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY, USA
2
The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
3
Department of Physiology, University of Kentucky, Lexington, KY, USA
4
Department of Orthopaedics, Eksjö, Region Jönköping County and Department of Biomedical and Clinical Sciences, Linköping University, Linköping,
Sweden
5
Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, and Unit of Clinical Physiology, Karolinska University
Hospital, Stockholm, Sweden
6
Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
7
The Journal of Physiology

Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE, USA
8
Department of Biology, Indiana University, Bloomington, IN, USA
9
Division of Pediatric Neurology, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden

Edited by: Scott Powers & Troy Hornberger


Linked articles: This article is highlighted in a Perspectives article by Goodman. To read this article, visit
https://doi.org/10.1113/281835.

Key points
r Ribosome biogenesis and MYC transcription are associated with acute resistance exercise (RE)
and are distinct from endurance exercise in human skeletal muscle throughout a 24 h time course
of recovery.
r A PCR-based method for relative ribosomal DNA (rDNA) copy number estimation was validated
by whole genome sequencing and revealed that rDNA dosage is positively correlated with
ribosome biogenesis in response to RE.
r Acute RE modifies rDNA methylation patterns in enhancer, intergenic spacer and non-canonical
MYC-associated regions, but not the promoter.
r Myonuclear-specific rDNA methylation patterns with acute mechanical overload in mice
corroborate and expand on rDNA findings with RE in humans.
r A genetic predisposition for hypertrophic responsiveness may exist based on rDNA gene dosage.

Vandré C. Figueiredo completed his PhD in Biomedical Sciences at the Liggins Institute, The University of Auckland, under the
supervision of Dr David Cameron-Smith before joining the laboratory of Dr John McCarthy at the University of Kentucky for his
postdoctoral training in 2016. His research is focused on the role of ribosome biogenesis and the cellular signalling regulating skeletal
muscle size. He seeks to understand the mechanism of muscle size determination hoping to apply this knowledge to ameliorate muscle
atrophy conditions, with particular interest in ageing, cachexia and muscle disuse.

#
These authors contributed equally to this work.
This article was first published as a preprint. Figueiredo VC, Wen Y, Alkner B, Fernandez-Gonzalo R, Norrbom J, Vechetti Jr. IJ, Valentino T,
Mobley CB, Zentner GE, Peterson CA, McCarthy JJ, Murach KA, von Walden F. 2020. Genetic and epigenetic regulation of skeletal muscle ribosome
biogenesis with exercise. bioRxiv. https://doi.org/10.1101/2020.12.14.422642.

© 2021 The Authors. The Journal of Physiology © 2021 The Physiological Society DOI: 10.1113/JP281244

Licenciado para - Anderson Sanches Gonçalves - 06263970901 - Protegido por Eduzz.com


2 V. C. Figueiredo and others J Physiol 0.0

Abstract Ribosomes are the macromolecular engines of protein synthesis. Skeletal muscle ribosome
biogenesis is stimulated by exercise, although the contribution of ribosomal DNA (rDNA) copy
number and methylation to exercise-induced rDNA transcription is unclear. To investigate the
genetic and epigenetic regulation of ribosome biogenesis with exercise, a time course of skeletal
muscle biopsies was obtained from 30 participants (18 men and 12 women; 31 ± 8 years,
25 ± 4 kg m–2 ) at rest and 30 min, 3 h, 8 h and 24 h after acute endurance (n = 10, 45 min cycling,
70% V̇O2 max ) or resistance exercise (n = 10, 4 × 7 × 2 exercises); 10 control participants underwent
biopsies without exercise. rDNA transcription and dosage were assessed using quantitative PCR
and whole genome sequencing. rDNA promoter methylation was investigated using massARRAY
EpiTYPER and global rDNA CpG methylation was assessed using reduced-representation bisulphite
sequencing. Ribosome biogenesis and MYC transcription were associated primarily with resistance
but not endurance exercise, indicating preferential up-regulation during hypertrophic processes.
With resistance exercise, ribosome biogenesis was associated with rDNA gene dosage, as well as
epigenetic changes in enhancer and non-canonical MYC-associated areas in rDNA, but not the
promoter. A mouse model of in vivo metabolic RNA labelling and genetic myonuclear fluorescence
labelling validated the effects of an acute hypertrophic stimulus on ribosome biogenesis and Myc
transcription, and also corroborated rDNA enhancer and Myc-associated methylation alterations
specifically in myonuclei. The present study provides the first information on skeletal muscle genetic
and rDNA gene-wide epigenetic regulation of ribosome biogenesis in response to exercise, revealing
novel roles for rDNA dosage and CpG methylation.
(Received 14 December 2020; accepted after revision 20 April 2021; first published online 29 April 2021)
Corresponding authors F. von Walden: Department of Women’s and Children’s health, Karolinska Institute, ALB Q2:07,
Karolinska University Hospital, 17176 Stockholm, Sweden. Email: ferdinand.von.walden@ki.se
K. A. Murach: Department of Physical Therapy and The Center for Muscle Biology, 900 South Limestone CTW 445,
Lexington, KY 40536, USA. Email: kmu236@g.uky.edu

Introduction transcribes 45S pre-rRNA, nothing is known about how


rDNA copy number (dosage) affects ribosome biogenesis
Ribosomes are the molecular factories responsible for in muscle. Furthermore, although ribosome biogenesis is
protein synthesis, which is considered to be a key implicated in the muscle hypertrophic process, little is
process in the long-term adaptive response to exercise known about its contribution to the endurance exercise
in skeletal muscle (Figueiredo, 2019a; McCarthy & response.
Murach, 2019). Ribosome biogenesis is stimulated by We recently showed that an acute hypertrophic stimulus
muscle loading (Figueiredo & McCarthy, 2019b; Kim results in widespread CpG hypomethylation in promoter
et al. 2019; von Walden, 2019b) and the magnitude sites of genes related to growth [i.e. mechanistic target
of de novo synthesis of ribosomes is proportional to of rapamycin (mTOR) pathway and Myc] specifically
the amount of load-induced adult muscle hypertrophy within muscle fibre nuclei (myonuclei) (Von Walden
in both rodents (Nakada et al. 2016) and humans et al. 2020a). These data concur with earlier findings
(Figueiredo et al. 2015; Stec et al. 2016; Hammarstrom showing epigenetic and signalling-related regulation of
et al. 2020). Mechanical loading rapidly induces RNA rDNA promoter regions in response to hypertrophic
polymerase I (Pol I) transcription of ribosomal DNA stimuli both in vivo and in vitro (von Walden et al.
(rDNA), as assessed by 45S pre-rRNA transcription levels 2012; von Walden et al. 2016). Thus, early dynamic
and accumulation of rRNA (von Walden et al. 2012b; epigenetic events associate with robust transcriptional
Kirby et al. 2016; Figueiredo et al. 2019b). The production responses required for successful adaptation to exercise
of the long 45S pre-rRNA transcript, which is processed (Jozsi et al. 2000; Pilegaard et al. 2000). rDNA is highly
into the mature 18S, 5.8S and 28S rRNAs, is considered regulated at the epigenetic level in general (Grummt, 2007;
to be the rate limiting step of ribosome biogenesis Murayama et al. 2008), although it is currently unknown
(Moss 2004). By contrast to protein-coding genes that whether rDNA promoter hypomethylation contributes to
commonly occur in the human genome as two copies, rRNA accumulation in response to exercise in muscle.
rDNA genes number in the hundreds and vary widely Furthermore, epigenetic patterning and regulation of
across individuals (Gibbons et al. 2015; Malinovskaya rDNA transcription is unique partly as a result of its
et al. 2018; Parks et al. 2018). Although each rDNA tandem repeat organization and large intergenic spacer
locus can participate in the formation of the nucleolus, (IGS) (Baldridge et al. 1992; Mougey et al. 1996; Zentner
comprising the subnuclear compartment where Pol I et al. 2011a; Audas et al. 2012; Shiue et al. 2014),

© 2021 The Authors. The Journal of Physiology © 2021 The Physiological Society

Licenciado para - Anderson Sanches Gonçalves - 06263970901 - Protegido por Eduzz.com


J Physiol 0.0 rDNA regulation with exercise 3

Table 1. Characteristics of the research participants

Control (CON) Endurance exercise (EE) Resistance exercise (RE)

Age (years) 30.5 ± 8.3 27.8 ± 6.8 33.3 ± 7.2


Sex (male/female) 6/4 6/4 6/4
Height (cm) 178 ± 8.2 176 ± 10.5 180 ± 8.9
Weight (kg) 81.4 ± 15.5 78.7 ± 17.0 81.2 ± 9.9
Body mass index (kg m–2 ) 25,5 ± 4.0 25.2 ± 4.7 24.9 ± 2.0
AbsoluteV̇O2 max (L min–1 ) 3.57 ± 0.8 3.62 ± 0.9 3.58 ± 0.9
RelativeV̇O2 max (mL min–1 kg–1 ) 44.7 ± 8.8 47.1 ± 11.2 44.1 ± 9.2
Knee extension (kg) 64.0 ± 17.0 72.5 ± 23.2 68.5 ± 16.7
Leg press (kg) 142 ± 40.3 152.6 ± 55.8 149.5 ± 42.7
VO2max, estimated maximal oxygen uptake. No significant differences were observed between groups. Data are presented as the
mean ± SD.

although there is no information on whether methylation Research participants


of alternative regulatory sites in the rDNA repeat, such as
Thirty healthy male and female participants (18 males
enhancer regions and non-canonical transcription factor
and 12 females) were recruited and randomized to either
binding areas, are affected by mechanical loading and
a control (CON, n = 10), an endurance exercise (EE,
associate with ribosome biogenesis in muscle.
n = 10) or a resistance exercise group (RE, n = 10). All
In the present study, we report the first time-course
groups included six males and four females. Participant
of ribosome biogenesis and rRNA transcription
characteristics are presented in Table 1. All participants
regulatory factor responses to acute endurance and
were recreationally active (i.e. involved in EE one to
resistance exercise (EE and RE, respectively) in
three times per week and/or RE one to two times per
human skeletal muscle, as well as comprehensively
week). Inclusion criterion was 18–50 years of age, and
evaluate how rDNA gene dosage and methylation
exclusion criteria were cardiovascular disease, neuro-
relates to rDNA transcription. To accomplish this,
muscular disease or severe knee problems.
we employed whole-genome sequencing, targeted
mass spectrometry-based rDNA promoter methylation
analysis, reduced-representation bisulphite sequencing Human study design
(RRBS), and mRNA- and protein-level measures. We
complemented the human analyses with an analogous At least 5 days prior to the intervention, participants
murine model of acute muscle loading, in vivo metabolic were familiarized with the experimental set-up. All sub-
RNA labelling and myonuclear-specific RRBS. With jects performed a submaximal test on a cycle ergometer
these parallel approaches, we reveal novel information on (Monark 828 E; Monark Exercise AB, Vansbro, Sweden)
genetic and epigenetic transcriptional regulation of the to estimate V̇O2 max (Ekblom-Bak et al. 2014; Bjorkman
translational apparatus following exercise. Our findings et al. 2016) and seven repetition maximum for knee
may have implications for individual heterogeneity in extension and leg press was titrated. These data were
exercise responsiveness to training (Ahtiainen et al. 2016; used to determine the load for the acute exercise bouts
Sparks, 2017; Lavin et al. 2019), as well as the epigenetic and to characterize the three groups with respect to
mechanisms of potentiated exercise adaptability related to physical status. Subjects were instructed not to perform
long-term cellular ‘muscle memory’ (Dungan et al. 2019, any strenuous resistance for the legs 3 days prior to
Murach et al. 2019, 2020; Seaborne et al. 2018; Snijders the intervention and no training the day prior. A liquid
et al. 2020). formula [1.05 g carbohydrates kg–1 body weight (bw), 0.28
g protein kg–1 bw and 0.25 g fat kg–1 bw] was provided
Methods as breakfast 1 h prior to collection of the pre-exercise
sample and as lunch (2.10 g carbohydrates kg–1 bw, 0.56 g
Ethical approval
protein kg–1 bw and 0.50 g fat kg–1 bw) immediately after
The study protocol was approved by the Regional Ethical the 3 h biopsy and at breakfast on day 2, 2 h before the final
Review board in Linköping (2017/183-31) and conformed biopsy. The liquid formulas contained 5.6 g of protein, 21 g
to the Declaration of Helsinki, except for registration in of carbohydrates and 5.0 g of fat per 100 mL (Resource
a database. After receiving written and oral information komplett Näring; Nestlé Health Science, Stockholm,
about the study, the participants provided their informed Sweden). This dietary formula was selected in order
consent to participate. Animal experiments were approved to remain consistent with previously published exercise
by the IACUC at the University of Kentucky (2020-3525). studies from members of our research team where a

© 2021 The Authors. The Journal of Physiology © 2021 The Physiological Society

Licenciado para - Anderson Sanches Gonçalves - 06263970901 - Protegido por Eduzz.com


4 V. C. Figueiredo and others J Physiol 0.0

similar diet was utilized (Fernandez-Gonzalo et al. 2013, evening on the day of the experiment (Camelon et al.
Lundberg et al. 2012, 2013, 2014, 2014ab, 2016). The 1998). Skeletal muscle biopsies were collected before the
subjects were instructed to eat a standard dinner (plate intervention (rest, or Pre) and at 30 min, 3 h, 8 h and
model) in the evening before the experiment and in the 24 h post exercise (Fig. 1A). Biopsies for the RE and CON

Figure 1. Exercise-related signalling in


response to acute EE and RE over a 24 h time
course in human skeletal muscle
A, experimental study design timeline illustrating
exercise and muscle biopsy collection in EE, RE
and control (CON) participants at rest (Pre),
30 min, 3 h, 8 h and 24 h after exercise. B,
western blot images for exercise-responsive
protein targets in CON, EE and RE. C,
quantification of phosphorylated AMPK at
Thr172 in CON (CON; Pre/30 min/3 h/8 h/24 h,
n = 10/10/10/9/10), EE (EE;
Pre/30 min/3 h/8 h/24 h, n = 10/10/10/8/9) and
RE (RE; Pre/30 min/3 h/8 h/24 h, n = 10/10/9/8/8).
ANOVA results: interaction, P = 0.209; main
effect for group, P = 0.762; main effect of time,
P = 0.010. †Significantly different from CON
within the same time point (P = 0.05). D,
quantification of phosphorylated p70S6K at
Thr389 in CON (CON; Pre/30 min/3 h/8 h/24 h,
n = 10/10/10/9/10), EE (EE;
Pre/30 min/3 h/8 h/24 h, n = 10/10/10/8/9) and
RE (RE; Pre/30 min/3 h/8 h/24 h, n = 10/10/9/8/8).
ANOVA results: interaction, P = 0.022; main
effect of group, P = 0.113; main effect for time,
P < 0.001. E, quantification of phosphorylated
RPS6 at Ser240/244 in CON (CON;
Pre/30 min/3 h/8 h/24 h, n = 10/10/10/9/10), EE
(EE; Pre/30 min/3 h/8 h/24 h, n = 10/10/10/8/9)
and RE (RE; Pre/30 min/3 h/8 h/24 h,
n = 10/10/9/8/8). ANOVA results: interaction,
P = 0.254; main effect of group, P = 0.016; main
effect of time, P = 0.005. ∗ Significantly different
from respective Pre biopsy (P < 0.05).
†Significantly different from CON within the
same time point (P < 0.05). Data are shown as
the mean ± SD.

© 2021 The Authors. The Journal of Physiology © 2021 The Physiological Society

Licenciado para - Anderson Sanches Gonçalves - 06263970901 - Protegido por Eduzz.com


J Physiol 0.0 rDNA regulation with exercise 5

groups were synchronized with the completion of the were incubated overnight at 4°C with a primary anti-
45 minute EE bout, to ensure comparable sampling time body (Cell Signaling Technology, Inc., Danvers, MA,
points between exercise modalities. Eight subjects had one USA) in blocking solution. The antibodies used were:
or two fewer biopsies taken for various reasons (e.g. failed phospho-p70S6 Kinase (Thr389, 108D2, #9234; dilution
biopsy, adverse event), which affected the sample size for 1:2000), pan-p70S6 Kinase (#9202; dilution 1:2000),
some analyses, and, in some cases, there was only sufficient phospho-S6 Ribosomal Protein (Ser240/244, D68F8,
material for a specific analysis. Muscle biopsies were #5364; dilution 1:2,000), pan-S6 Ribosomal Protein
obtained from the vastus lateralis muscle percutaneously (5G10, #2217; dilution 1:3000), phospho-AMPKα
after injection of local anaesthetic (carbocain 10 mg mL–1 ) (Thr172, 40H9, #2535; dilution 1:1000) and pan-AMPKα
using a Bergström biopsy needle with a diameter of 5 mm (#2532; dilution 1:2000). The MYC antibody was obtained
(Stille AB, Torshälla, Sweden). Skeletal muscle tissue was from DSHB (9E10 concentrate, dilution 1:1000). The next
blotted for excess blood, cleaned of non-skeletal muscle day, membranes were incubated with a goat anti-rabbit
tissue and snap-frozen in liquid nitrogen. All samples were (#G-21234; Thermo Fisher Scientific) secondary antibody
stored at −80°C until further analysis. (dilution 1:10,000 in blocking solution) for 1 h at room
temperature. Membranes were incubated with enhanced
chemiluminescence (ECL) reagent (Clarity Western ECL
Human exercise protocols substrate, #170-5060; Bio-Rad) before exposure to a
ChemiDoc MP Imaging System (Bio-Rad). Coomassie
RE consisted of two lower limb exercises; leg press
blue staining was utilized to confirm equal loading.
(Nordic Gym AB, Bollnäs, Sweden) and knee extension
For MYC and total RPS6, total protein was visualized
(Nordic Gym AB). After a short warm-up on submaximal
and quantified using Revert 700 (Licor, Lincoln, NE,
loads, the participants performed four sets per exercise
USA) for the purpose of normalization, and two samples
at seven repetition maximum load with 2 min of rest
were excluded as a result of protein degradation. An
between sets and 5 min between exercises. EE consisted
isotype-specific mouse IgG1 secondary antibody for MYC
of 45 min of cycling (Monark 828 E; Monark Exercise
was employed at 1:1000 for 2 h. Bands were quantified
AB) at 70% of estimated V̇O2 max . Heart rate was monitored
using ImageJ software (NIH, Bethesda, MD, USA).
continuously (Garmin Edge 25; Garmin, Lenexa, KS,
USA) and participants were asked to rate their level of
perceived exertion every 5 min using the Borg RPE scale
(Borg, 1970).
RNA extraction, cDNA synthesis and gene expression
analysis
Western blotting
Muscle tissue (∼25 mg) was used to extract RNA
Protein was extracted from the organic phase of TRI using TRI Reagent (Sigma-Aldrich, St Louis, MO, USA).
Reagent following RNA extraction using the optimized Tissue was homogenized using beads and the Bullet
protocol (Wen et al. 2020). Briefly, following the final Blender Tissue Homogenizer (Next Advance, Troy, NY,
step of ethanol addition, samples were centrifuged and USA). Following homogenization, RNA was isolated via
the resulting protein pellet solubilized using SDS-urea phase separation by addition of bromochloropropane and
buffer (100 mm Tris, pH 6.8, 12% glycerol, 4% SDS, centrifugation. The supernatant was then transferred to
0.008% bromophenol blue, 2% β-mercaptoethanol, 5 m a new tube and further processed on columns using
urea) supplemented with Halt Protease (#78438; Thermo the Direct-zol Kit (Zymo Research, Irvine, CA, USA).
Fisher Scientific, Waltham, MA, USA) and phosphatase RNA was treated in-column with DNAse and eluted
(#78426; Thermo Fisher Scientific) inhibitor cocktails. in nuclease-free water before being stored at −80°C.
The RC DC Protein Assay (Bio-Rad, Hercules, CA, For quantitative reverse transcription PCR (qRT-PCR),
USA) was used to determine protein concentration. 750 ng of total RNA was reverse transcribed using
Twenty micrograms of protein per sample was loaded SuperScript IV VILO Master Mix (Invitrogen, Carlsbad,
on a gradient gel (Criterion Precast Gels; Bio-Rad) CA, USA). SsoAdvanced Universal SYBR Green Super-
and electrophoretically transferred to a polyvinylidene mix (Bio-Rad) was used for qRT-PCR on the CFX384
fluoride membrane (Bio-Rad). Pool control samples Thermocycler (Bio-Rad). qRT-PCR data were normalized
were loaded on all gels. Membranes were blocked in by the geometric mean of three stable reference genes
5% BSA (#A-420-1; Gold Biotechnology, St Louis, MO, (EMC7, VCP, C1ORF43). Primer sequences are available
USA), for phospho-specific antibodies, or 5% Non-Fat upon request. Melting curves were performed for every
dry milk (#170-6404; Bio-Rad), for pan-antibodies, primer pair to confirm a single-product amplification.
in Tris-buffered saline with 0.1% Tween 20 for 2 h at qRT-PCR data were analysed using the 2– CT method.
room temperature. Following blocking, membranes A missed biopsy, insufficient tissue or a technical issue

© 2021 The Authors. The Journal of Physiology © 2021 The Physiological Society

Licenciado para - Anderson Sanches Gonçalves - 06263970901 - Protegido por Eduzz.com


6 V. C. Figueiredo and others J Physiol 0.0

with sample processing resulted in n < 10 for qRT-PCR to same concentration. Next, 2.5 ng of DNA was
data in some instances (see Results). loaded per well in triplicate. qPCR was run using
Fast SYBR Green Master Mix (Applied Biosystems,
Foster City, CA, USA) in a QuantStudio 3 Real-Time
Targeted human rDNA promoter methylation analysis PCR System (Thermo Fisher Scientific). The sequence
of the primers (18S, 5.8S 28S and 5S, and TP53 as a
Genomic DNA was isolated from muscle samples using
reference gene), utilized in the present study to assess
the QIAamp DNA Mini Kit (Qiagen, Hilden, Germany)
rDNA dosage, are from Gibbons et al. (2015): TP53
in accordance with the manufacturer’s instructions. In
forward (F) 5 -TGTCCTTCCTGGAGCGATCT-3 and
brief, ∼25 mg of frozen muscle samples were incubated
reverse (R) 5 -CAAACCCCTGGTTTAGCACTTC-3 ;
and homogenized by enzyme digestion and mechanical
5S rDNA F 5 -TCGTCTGATCTCGGAAGCTAA-3 and
disruption. After tissue had been completely dissolved,
R 5 -AAGCCTACAGCACCCGGTAT-3 ; 5.8S rDNA
the mixture was added to mini spin-columns and a series
F 5 -CGACTCTTAGCGGTGGATCA-3 and R 5 -GAT
of wash steps was performed. Each sample was diluted
CAATGTGTCCTGCAATTC-3 ; 18S rDNA F 5 -GAC
in 70 μL of distilled water. Immediately after DNA had
TCAACACGGGAAACCTC-3 and R 5 -AGACAAATC
been extracted, quantity and quality were determined in a
GCTCCACCAAC-3 ; and 28S rDNA F 5 -GCGGGTGGT
NanoPhotometer NP80 (Implen, München, Germany).
AAACTCCATCT-3 and R 5 -CACGCCCTCTTGAACT
Quantitative methylation analysis was performed
CTCT-3 . Data were normalized to TP53 and expressed
using the EpiTYPER methodology (Ehrich et al. 2005)
in arbitrary units (AU). Data were not compared with
and the MassARRAY system (Agena Biosciences, San
a standard curve with known rDNA quantity and are
Diego, CA, USA) in accordance with the manufacturer’s
therefore referred to as the relative rDNA dosage. The
recommendations and protocols, and as described pre-
sample number is smaller (n = 7) when comparing rDNA
viously (von Walden et al. 2020b). In this method,
gene dosage with rDNA transcription at 24 h post exercise
a targeted amplification of bisulphite converted DNA is
for the technical reasons outlined above, as well as DNA
followed by in vitro transcription, RNase cleavage and sub-
for one sample not being suitable for analysis.
sequent fragment mass analysis by matrix-assisted laser
desorption/ionization time of flight mass spectrometry
to quantify CpG sites. PCR primers were adapted
from D’Aquila et al. (2017). EpiTect methylated and Whole genome sequencing (WGS) and bioinformatics
non-methylated bisulphite-treated control DNA (Qiagen)
Based on the qPCR results, DNA from the same samples
was used to evaluate the quantitative recapture of
from nine participants spread across the full range of
methylation ratios of the amplicons. The amplicon
rDNA gene dosage (n = 3, low; n = 3, middle; n = 3,
used in the present study met the quality criteria of
high) was selected for WGS. At least 1.5 μg of skeletal
methylated and non-methylated data points measured
muscle DNA was used for analysis. To avoid potential
at >79% and <5% methylation ratios, respectively,
amplification bias, a PCR-free protocol was used for
as well as SD percentages <5%. Samples were run in
library preparation. We calculated the rDNA copy number
duplicate and SD percentages >20% were removed from
using an approach similar to that previously described
the study. The remaining data points correlated with
by Gibbons et al. (2014) by calculating relative depth
r2 = 0.72. Bisulphite conversion efficiency was evaluated
differences between rRNA sequences and the back-
by analysing one non-CpG C in a subset of the study
ground (whole genome). Reads were trimmed for adapter
samples. All data were checked manually and visually
sequences and low quality (minimum phred score of 20)
inspecting the mass spectra.
before aligning to the GRCh37 (hg19) human reference
genome assembly using Bowtie2, version 2.3.4.3, with
Estimation of rDNA copy number via quantitative PCR the ‘-end-to-end’ option (Langmead & Salzberg, 2012;
Langmead et al. 2019). Alignment results, produced in
(qPCR)
random order, were sorted with respect to their genomic
Relative ribosomal DNA copy number was estimated by positions using the samtools sort function and read
qPCR (rDNA dosage). Genomic (g)DNA was extracted depth at each position was computed using samtools
from muscle biopsies (from n = 27 participants) and depth function. We took advantage of the 45S rRNA
isolated using the Monarch Kit for DNA isolation (New sequence on the supercontig GL000220.1 in the GRCh37
England Biolabs, Ipswich, MA, USA) in accordance (hg19) reference assembly and used this as a surrogate
with the manufacturer’s instructions. Proteinase K for the consensus rDNA repeat sequence (U13369), as
digestion was performed overnight, and all samples described by Gibbons et al. (2014). We found that read
were RNAse treated before being purified on column. depths computed using the supercontig rRNA regions
gDNA was eluted in nuclease-free H2 O and diluted were highly correlated with those computed from using

© 2021 The Authors. The Journal of Physiology © 2021 The Physiological Society

Licenciado para - Anderson Sanches Gonçalves - 06263970901 - Protegido por Eduzz.com


J Physiol 0.0 rDNA regulation with exercise 7

U13369 (r2 > 0.96, data not shown). This approach columns with on column DNAse treatment (Zymo
circumvented the need to generate a custom reference Research). RNA was resuspended in molecular-grade
assembly to combine the rDNA sequence with the rest H2 O and quantified by measuring the optical density
of the genome and ensured assembly version consistency, (230, 260 and 280 nm) with a Nanodrop 1000
thereby limiting the confounding effects of the genome Spectrophotometer (Thermo Fisher Scientific). Nascent
assembly version differences on the variability in back- (EU-labelled) RNA was purified from 1 μg of RNA per
ground read depths among participants. Maximum read sample using the commercially available Click-iT Nascent
depth corresponding to each rRNA coding region (18S, RNA Capture Kit (Life Technologies, Carlsbad, CA, USA).
5.8S and 28S) were divided by the average read depth for cDNA was generated on 500 ng of total RNA and all EU
the whole genome to obtain rDNA component dosage, affinity-purified RNA using the SuperScript VILO cDNA
which is an estimate of the number of copies of rDNA in Synthesis Kit (Life Technologies). We assessed relative
a haploid genome. gene expression in the total RNA and nascent RNA
fractions by normalization to EMC7 via the comparative
Ct (2–࢞࢞Ct ) method.
Mouse study design
To specifically label myonuclei via genetic means, we
generated female HSA+/– -GFP+/– mice by crossing homo- RRBS analysis of human skeletal muscle and mouse
zygous human skeletal actin reverse tetracycline trans-
myonuclear DNA
activator (HSA-rtTA) mice developed by our laboratory
(Iwata et al. 2018) with homozygous tetracycline response DNA isolation from human biopsy samples (≤5 mg) and
element histone 2B green fluorescent protein mice mouse myonuclear samples was carried out as described
(TetO-H2B-GFP) obtained from the Jackson Laboratory in detail by Begue et al. (2017) and Von Walden et al.
(005104; Jackson Laboratory, Bar Harbor, ME, USA). GFP (2020a), respectively. Briefly, using the QIAamp DNA
labelling is >90%, is highly specific to myonuclei, and does Micro Kit (Qiagen), muscle samples or myonuclear pellets
not result in the labelling of satellite cell-derived myo- were re-suspended in ‘buffer ATL’ supplemented with
nuclei during the experimental period (Iwata et al. 2018), proteinase K overnight at 56°C. DNA binding to the
thus making the results specific to resident myonuclei. column was conducted using 1 μg of carrier RNA (only
Mice were treated with doxycycline in drinking water for myonuclear samples), and washes and centrifugations
(0.5 mg mL–1 with 2% sucrose) for 1 week. Mice were were carried out in accordance with the manufacturer’s
allowed food and water ad libitum. Following doxycycline instructions. DNA was eluted in 20 μL of molecular grade
treatment and a 6-day washout, mice underwent bilateral H2 O and was stored at −80°C until the time of analysis.
sham surgery (biological duplicate) or synergist ablation DNA quality assessment and RRBS was conducted
mechanical overload (OV) of the plantaris (biological in collaboration with Zymo Research. Quality and
triplicate) as described previously (von Walden et al. concentration were assessed using a Fragment Analyzer
2020a). Post-surgery, mice recovered on a heating pad (Advanced Analytical Technologies, Inc., Ames, IA, USA).
and were monitored until fully ambulatory, and then were ‘Classic’ RRBS library preparation was performed by
checked daily thereafter. Mice were killed via a lethal digesting 5 ng (mouse) and 25-50 ng (human) of genomic
injection of sodium pentobarbital and cervical dislocation DNA with 30 units of MspI enzyme (New England
72 h later. The mice in these experiments were ∼3 months BioLabs, Ipswich, MA, USA) and fragments were ligated
of age at the time of surgery, and immunohistochemistry to pre-annealed adapters containing 5’-methyl-cyotosine.
and single fibre imaging for representative images is Adapter-ligated fragments ≥50 bp were recovered using
described in von Walden et al. (2020a). For the metabolic the DNA Clean & Concentrator Kit (D4003; Zymo
RNA labelling experiments, age-matched C57BL/6J mice Research) and bisulphite-treated using the EZ DNA
were subjected to sham and OV by the same surgeon as Methylation-Lightning Kit (D5030; Zymo Research).
the HSA-rtTA mice (biological duplicate for sham and Preparative-scale PCR was performed, and the products
triplicate for OV). were purified again using the Clean & Concentrator
Kit. Paired-end sequencing was performed using HiSeq
(Illumina, San Diego, CA, USA) and sequenced reads
Mouse in vivo metabolic RNA labelling
from bisulphite-treated libraries were identified using the
Five hours prior to tissue harvesting after sham and OV, standard Illumina base calling software.
all mice were pulsed with 2 mg of 5-ethynyl-uridine Raw FASTQ files were adapter, filled-in nucleotides
(EU) dissolved in 200 μL of PBS via an i.p. injection, as and were quality-trimmed using TrimGalore 0.6.4
described previously (Kirby et al., 2016). Muscles were (Babraham Bioinformatics, Cambridge, UK) using
snap frozen in liquid nitrogen upon collection. RNA was options for ‘–rrbs’ and ‘–non_directional’ mode retaining
extracted using Trizol reagent (Invitrogen) and DirectZol reads with minimum quality above phred score of 30.

© 2021 The Authors. The Journal of Physiology © 2021 The Physiological Society

Licenciado para - Anderson Sanches Gonçalves - 06263970901 - Protegido por Eduzz.com


8 V. C. Figueiredo and others J Physiol 0.0

FastQC 0.11.8 (Babraham Bioinformatics) was used to background fluorescence, and myonuclei were classified
assess the effect of trimming and overall quality of the as GFP+/PI+ after elimination of doublets via forward
data. A custom genome assembly to interrogate rDNA scatter area vs. height (biological duplicate for sham and
methylation was generated by adding the consensus triplicate for OV) and collected in 15 mL conical tubes.
rDNA repeat sequences, GenBank U13369.1 (Gonzales Myonuclear suspensions were pelleted at 500 g for 5 min
and Sylvester, 1995) and BK000964.3 (Grozdanov using a swinging-bucket rotor prior to DNA isolation.
et al. 2003), as a separate chromosome to the human
(GRCh38p13) and mouse (GRCm39) reference genome
assemblies, respectively. As a result of mapping inter- Statistical analysis
ference from highly similar sequences within the
reference assemblies, these sequences were found using Data were analysed using two-way ANOVA with exercise
Blast (https://blast.ncbi.nlm.nih.gov) by comparing modality as one factor (EE vs. RE vs. CON) and time
the respective rDNA sequences with the reference of biopsy as another (Pre, 30 min, 3 h, 8 h and 24 h).
assembly followed by masking of these sequences in the Gene expression data and western blot data were analysed
genome using ‘N’s. Alignment to the custom human and using two-way ANOVA with repeated measures. rDNA
mouse reference genomes was performed using Bismark methylation was analysed as two-way ANOVA, and not
0.19.0 (Babraham Bioinformatics). Methylated and repeated measures as a result of missing two subject’s pre
unmethylated read totals for each CpG site were collected biopsies in each group. When significant interactions or
using the methylation extractor tool. Methylation levels main effects were found, post hoc tests were performed
of each sampled cytosine was estimated as the number of using the Holm–Sidak method. Potential relationships
reads reporting a ‘C’, divided by the total number of reads among the variables assessed were investigated using
reporting a ‘C’ or ‘T’. Differential methylation analyses one-tailed Pearson’s correlation analysis. The level of
were performed using R Bioconductor package, methylSig significance was set at 5% (P < 0.05) and Prism, version
v1.0.0 (Park et al. 2014), which accounts for both read 9.0 (GraphPad Software Inc., San Diego, CA, USA) and
coverage (minimum set to 5×, as previously described SigmaPlot, version 14.0 (Systat Software Inc., San Jose,
by Begue et al. 2017 and von Walden et al. 2020a) and CA, USA) were used for the analysis. Pre vs. 24 h western
biological variation. Differentially methylated regions blot data were analysed using one-tailed dependent t
were determined with tiling using 100 bp segments and tests, with P < 0.05 considered statistically significant. For
no minimum cut-off for CpG sites. The mouse data were methylation data, a general linear model was employed,
analysed using a beta-binomial distribution and, for and P < 0.05 accounting for read depth was utilized, as
individual CpG data, sites where a CpG was present in well as FDR (calculated as Benjamini–Hochberg adjusted
every sample were included for analysis (unless otherwise P values) (Benjamini & Hochberg 1995). Data are pre-
noted). The human data were analysed using a generalized sented as the mean ± SD.
linear model accounting for the interaction between
exercise status and time after the last bout and included
three control participants in the model as a covariate (11 Results
total samples), making for a robust statistical approach. RE promotes mTORC1, whereas EE promotes AMPK
CpG coverage in the human samples at individual sites can signalling over the 24 h time course of recovery in
vary using RRBS, but, in all instances where significant P
muscle
values and low false discovery rate (FDR) (reported as the
Benjamini-Hochberg adjusted P value in the Results) was To verify that our chosen exercise modalities caused an
reported, CpGs in ≥4 exercised participants were present increase in canonical signalling associated with exercise
in the dataset. adaptation (for study design, see Fig. 1A), we analysed the
Murine myonuclear isolations were conducted pre- phosphorylation of key proteins involved with EE and RE
viously, as described (Von Walden et al. 2020a). Briefly, (namely AMPK and mTORC1 signalling). As expected,
after death via lethal CO2 asphyxiation and cervical EE and RE promoted different signalling patterns. Only
dislocation, plantaris muscles were harvested and EE induced phosphorylation of AMPK (at Thr172) 30 min
processed for myonuclear isolation via fluorescence post exercise (P = 0.002 compared to Pre; P = 0.050
activated cell sorting (FACS). Muscle was Dounce homo- compared to CON) (Fig. 1B & 1C). RE promoted
genized by hand in a sucrose-based physiological buffer, signalling through mTORC1 as assessed by 70S6K at
and then the crude nuclear suspension was filtered Thr389, specifically at 30 min (P = 0.044 compared to
through a 40 μm strainer and spiked with 4 μL of Pre) and 8 h (P = 0.036 compared to Pre; P = 0.013
propidium iodide (PI) (1 mg mL–1 stock), then sub- compared to CON) (Fig. 1B & 1D), whereas EE did not
jected to FACS analysis. Muscle from a control mouse activate p70S6K (Fig. 1D). In the control participants,
(sucrose-only treated HSA-GFP) was used to determine p-p70S6K was induced at 24 h after the pre biopsy

© 2021 The Authors. The Journal of Physiology © 2021 The Physiological Society

Licenciado para - Anderson Sanches Gonçalves - 06263970901 - Protegido por Eduzz.com


J Physiol 0.0 rDNA regulation with exercise 9

following breakfast, indicating that food intake stimulates (P = 0.031). TIF-IA was different relative to Pre at 8 h and
p-p70S6K to a similar extent as food intake combined with 24 h in the EE group (both P < 0.001). RE was different
exercise 24 h prior, or that there was a potential lingering than CON at 3 h (P = 0.042) and 24 h (P = 0.014), and EE
effect of inflammation from the biopsy (P = 0.004 at 8 h (P < 0.001) and 24 h (P < 0.001) for TIF-IA. NOP56
compared to Pre). Phosphorylation of RPS6 at Ser240/244 was elevated relative to Pre in EE at 8 h (P = 0.009) and
increased mainly following RE, and no significant change was also different than CON at 8 h (P = 0.001), as was RE
was observed in CON or EE. At 30 min (P = 0.008 (P = 0.031). PES1 was lower at 30 min (P = 0.049) and
compared to Pre) and 8 h, p-RPS6 was increased in the 3 h (P = 0.028) in RE relative to CON. Few differences
RE group only (P = 0.007 compared to Pre; P = 0.005 in gene expression were found in the CON group across
compared to CON). Globally, EE stimulated AMPK, the time course and are provided in the Supporting
whereas RE stimulated mTORC1 signalling, consistent information.
with the literature (Vissing et al. 2013). C-MYC (referred to as MYC for human and Myc for
mice), which is upstream of Pol I regulon formation
(von Walden et al. 2012), was >15 fold higher after
RE preferentially stimulates ribosome biogenesis and 3 h (P < 0.001) and ∼10-fold up-regulated in response
MYC gene expression independent from transcription to RE after 8 h relative to Pre (P = 0.041) (Fig. 2C);
MYC was also up-regulated with EE relative to Pre at
of RNA Pol I-associated factors
8 h (P = 0.025) (Fig. 2C). Relative to CON and EE,
We utilized 45S pre-rRNA abundance as a readout of MYC was elevated 3 h after RE (P < 0.001). Relative to
Pol I activity and ribosome biogenesis (Leary & Huang, CON, EE (P = 0.019) and RE (P = 0.027) were both
2001). EE caused an acute reduction of Pol I activity higher at 8 h. Marked responsiveness of MYC within
that was evident by 30 min post-exercise (63% ± 30% 24 h of acute RE in vastus lateralis muscle is supported
decrease, P = 0.039 compared to CON at the same by meta-analytical data from RE transcriptome studies
timepoint) and EE was significantly different than RE at (Zierath and Pillon Laboratories; https://www.metamex.
both 3 h (P = 0.020) and 24 h (P = 0.004) (Fig. 2A). eu) spanning 110 healthy young and middle-aged men
Ribosome biogenesis was significantly induced by RE at and women (log fold change = 1.48, FDR = 0.007 ×
3 h (126 ± 47%, P = 0.005, compared to CON; P = 0.020 10−4 ) (Pillon et al. 2020); for reference, log fold change for
compared to EE) and 24 h post-exercise (127% ± 32%, MSTN in the same individuals was −0.81 (FDR = 0.001
P = 0.065 compared to CON; P = 0.004 compared to EE), × 10−4 ). At the protein level, MYC may peak ∼48 h
indicating a divergent ‘early’ and ‘late’ ribosome biogenesis after RE (Figueiredo et al. 2016). We probed for MYC at
response. 24 h and found that it was elevated in the majority of
Genes that are responsive to exercise, MSTN, our samples relative to Pre, but two subjects demonstrated
ATROGIN1 and MURF1 (Louis et al. 2007), were generally a sharp decrease, so the difference was not significant
affected in our time-course, but to a greater extent with RE (P = 0.28) (Fig. 3). We also measured total RPS6 as
(Fig. 2B). MSTN was reduced relative to CON 3 h and 8 h a readout of ribosome biogenesis at the protein level
after EE (P < 0.001 and P = 0.025, respectively) and RE because recent studies in rodents and humans report
(P < 0.001 and P = 0.069, respectively). ATROGIN1 was increased total RPS6 after acute RE (Roberts et al. 2015,
elevated at 3 h (P = 0.019) and lower at 8 h (P = 0.046) Ato et al. 2020, Sase et al. 2020, Takegaki et al. 2020) and
with EE relative to Pre, but was lower at all timepoints in similarly found a significant induction by 24 h (P = 0.04)
RE compared to Pre (all P < 0.001, with the exception (Fig. 3). Summary data and statistical analyses for all
of 30 min, P = 0.007). ATROGIN1 was also lower with gene and protein measures are provided in the Supporting
RE relative to CON and EE at 3 h (P < 0.001 for both), information.
8 h (P < 0.001 and P = 0.002, respectively) and 24 h
(P < 0.001 for both). In the RE group, MURF1 was
elevated at 8 h compared to Pre (P < 0.001). Relative rDNA gene dosage is related to rDNA
In general, gene expression of factors associated with
transcription following a bout of RE
Pol I regulon formation/transcription (UBTF, TIF1A,
PAF53) and 45S pre-rRNA processing factors (BOP1, Because rDNA copy number can vary widely across
NOP56, PES1, NUCLEOLIN) were modestly affected by people (Gibbons et al., 2014), we hypothesized that
exercise (Fig. 2B). UBTF only changed in the RE group relative rDNA dosage would correlate with the ribosome
relative to Pre; it was reduced at 3 h (P = 0.007), 8 h biogenesis response to RE. First, to accurately assess
(P < 0.001) and 24 h (P = 0.004). UBTF was also elevated relative rDNA gene dosage in our participants, we ranked
at 8 h and 24 h with RE relative to CON (P < 0.001 and all 30 individuals according to their normalized 18S
P = 0.002, respectively) and EE (P = 0.006 and P = 0.021, rDNA gene content using a qPCR approach for rDNA
respectively); EE was also different from CON at 8 h quantification (Gibbons et al., 2014). The range of relative

© 2021 The Authors. The Journal of Physiology © 2021 The Physiological Society

Licenciado para - Anderson Sanches Gonçalves - 06263970901 - Protegido por Eduzz.com


10 V. C. Figueiredo and others J Physiol 0.0

Figure 2. Transcriptional responses to acute EE and RE over a 24 h time course in human skeletal muscle
A, RNA Polymerase I (Pol I) transcription of ribosomal rDNA, as assessed by 45S pre-rRNA transcription levels,
in control (CON; Pre/30 min/3 h/8 h/24 h, n = 10/8/10/7/8), EE (Pre/30 min/3 h/8 h/24 h, n = 9/7/7/6/7) and RE
(Pre/30 min/3 h/8 h/24 h, n = 9/9/7/6/7) participants, normalized to Pre levels. ANOVA results: interaction, P = 0.005;
main effect for group, P = 0.034; main effect for time, P = 0.798. B, heatmap illustrating gene expression for
exercise-responsive genes (MSTN, ATROGIN1, MURF1), Pol I transcription factors (UBFT, TIF-1A, PAF53) and 45S
pre-rRNA processing factors (BOP1, NOP56, PES1, NUCLEOLIN) in CON (Pre/30 min/3 h/8 h/24 h, n = 10/8/10/7/8),
EE (Pre/30 min/3 h/8 h/24 h, n = 9/7/7/6/7) and RE (Pre/30 min/3 h/8 h/24 h, n = 9/9/7/6/7) participants. Interactions
and main effects for group and time were, respectively: MSTN, P = 0.061, P < 0.001, P = 0.158; ATROGIN1, all
P < 0.001; MURF1, P = 0.560, P = 0.948, P ≤ 0.001; UBTF, P < 0.001, P < 0.001, P = 0.098; TIF-IA, P = 0.002,
P = 0.001, P < 0.001; PAF53, P = 0.831, P = 0.618, P = 0.161; BOP1, P = 0.299, P = 0.962, P = 0.015; NOP56,
P = 0.044, P = 0.106, P = 0.025; PES1, P = 0.170, P = 0.175, P < 0.001; NUCLEOLIN, P = 0.324, P = 0.288,
P = 0.250. C, MYC gene expression in CON (Pre/30 min/3 h/8 h/24 h, n = 10/8/10/7/8), EE (Pre/30 min/3 h/8 h/24 h,
n = 9/7/7/6/7) and RE (Pre/30 min/3 h/8 h/24 h, n = 9/9/7/6/7) participants. ANOVA results: interaction, P = 0.003;
main effect for group, P < 0.001; main effect for time, P < 0.001, normalized to Pre levels. ∗ P < 0.05 compared
to Pre. †P < 0.05 compared to CON at the same time point. #P < 0.05 compared to EE at the same time point.
Data are shown as the mean ± SD.

© 2021 The Authors. The Journal of Physiology © 2021 The Physiological Society

Licenciado para - Anderson Sanches Gonçalves - 06263970901 - Protegido por Eduzz.com


J Physiol 0.0 rDNA regulation with exercise 11

rDNA gene dosage among our 30 participants displayed Promoter methylation is not associated with
a ∼3-fold difference when comparing the lowest vs. the ribosome biogenesis at rest
highest values (Fig. 4A). Based on this ranking, we selected
the top three, bottom three and three individuals in the The human rDNA promoter encompasses a 174 bp long
middle of the distribution and performed WGS to directly section of the 45S RNA gene that include two important
assess rDNA copy number. From the WGS data, we elements; the core promotor spanning −45 to +18 and
calculated rDNA copy number and observed a significant the upstream core element −156 to −107, both relative
positive correlation between this value and our qPCR to the transcription start site (TSS) (Haltiner et al. 1986)
quantification of 18S (r = 0.793, P = 0.005) (Fig. 4B) and (Fig. 5A). We first used the Agena EpiTYPER, a sensitive
28S (r = 0.590, P = 0.047) (Fig. 4C) rDNA. Moreover, and targeted mass spectrometry-based array method to
the qPCR quantification of the different coding regions compare the degree of methylation of the rDNA promoter
of the rDNA gene showed strong inter-correlation (28S in resting skeletal muscle at five sites within the promoter
vs. 18S, r = 0.846, P < 0.0001, 28S vs. 5.8S, r = 0.975, region. We calculated an average skeletal muscle rDNA
P < 0.0001 and 18S vs. 5.8S, r = 0.862, P < 0.0001) promoter methylation of 21% ± 8% in resting skeletal
(Fig. 4D–4F), whereas quantification of the 5S rDNA gene muscle (Fig. 5B), in agreement with our previous work
(not part of 45S RNA gene) was less correlated with the (Von Walden et al. 2020b) and consistent with levels
other rRNA regions (5S vs. 28S, r = 0.297, P = 0.066, reported in other non-muscle tissues in humans (Pietrzak
5S vs. 18S, r = 0.386, P = 0.023, 5S vs. 5.8S, r = 0.382, et al. 2011; Uemura et al. 2012). Ribosomal DNA promoter
P = 0.042) (Fig. 4G–4I). Relative rDNA dosage did not methylation varied from 4% to 41% depending on the
correlate to ribosome biogenesis at rest (18S, r = 0.026, site, but site-specific methylation on a person-by-person
P = 0.45; 28S, r = −0.028, P = 0.446; 5.8S, r = −0.0005, basis tracked well (coloured dots), so the average value
P = 0.499) (data not shown). Finally, the increase in is reflective of methylation along the promoter (Fig. 5C).
45S pre-rRNA 24 h after RE was positively correlated Average methylation was neither related to 45S pre-rRNA
with relative rDNA dosage using all coding regions (18S, levels (r = 0.24, P = 0.15) (Fig. 5D), nor total RNA content
r = 0.652, P = 0.056; 28S, r = 0.729, P = 0.0313; 5.8S, (r = 0.1, P = 0.33) (Fig. 5E) at rest. These data collectively
r = 0.696, P = 0.041) (Fig. 4J–4L); this relationship was suggest that the regulation of ribosome biogenesis in
not apparent at 3 h (18S, r = 0.203, P = 0.330; 28S, skeletal muscle under resting conditions is not influenced
r = 0.253, P = 0.292; 5.8S, r = −0.004, P = 0.496) (data by rDNA promoter methylation, nor gene dosage.
not shown). Interestingly, there was a negative correlation
between p70S6K phosphorylation at Thr389 with 45S RE acutely modifies methylation of the rDNA gene
pre-rRNA at 3 h (r = −0.7269, P = 0.032), but no
without affecting the promoter
association at 24 h was observed (r = −0.285, P = 0.291)
(data not shown). To explore whether the acute modulation of 45S
pre-rRNA abundance following EE (down-regulation)
and RE (up-regulation) was associated with epigenetic
modifications at the rDNA promoter, we analysed targeted
rDNA promoter methylation at all time points in all
participants via EpiTYPER. Although there was a group
A main effect (P < 0.001), no post hoc test showed any
changes in average rDNA promoter methylation (Fig. 6A).
B Pre Although rDNA promoter methylation was not altered
4000
24h RE by exercise, the robust induction of ribosome biogenesis
Normalized Density Units

and MYC throughout the 24 h time course following RE


3000
provided rationale to characterize methylation of other
regulatory regions along the rDNA repeat in greater
2000 detail.
We conducted RRBS on DNA from a subset of RE
1000 participants at pre exercise, as well as 30 min post and
24 h post exercise (n = 8) and included data from
0 three non-exercise participants at the corresponding
MYC Total RPS6 time points in the analysis to account for the effects of
biopsy and time. There is a strong inter-relationship
Figure 3. Myc and RPS6 protein levels 24 h after acute RE
A, representative western blots for MYC and RPS6 Pre and 24 h after between chromatin modifications and DNA methylation
RE. B, MYC and RPS6 protein levels Pre and 24 h after RE (n = 8). (Eden et al. 1998; Fuks, 2005; Nemeth et al. 2008;
∗ P < 0.05. Cedar & Bergman, 2009), so published chromatin

© 2021 The Authors. The Journal of Physiology © 2021 The Physiological Society

Licenciado para - Anderson Sanches Gonçalves - 06263970901 - Protegido por Eduzz.com


12 V. C. Figueiredo and others J Physiol 0.0

Figure 4. rDNA dosage for all participants at rest and its association to ribosome biogenesis in response
to acute RE
A, relative rDNA dosage determined via quantitative PCR (qPCR) for all participants (n = 27). Comparison of rDNA
copy number determined by whole genome sequencing (WGS) relative to rDNA copy number estimated by qPCR
using 18S rDNA (B) and 28S rDNA (C) (n = 9). Using the qPCR approach for relative DNA dosage determination,
relationships between different locations in the 45S gene: 28S vs. 18S (D), 5.8S vs. 28S (E) and 5.8S vs. 18S (F),
as well as relationships between 5S (not part of 45S) and 18S (G) 5S and 28S (H) and 5S and 5.8S (I) (n = 27).
Relationships between ribosome biogenesis (measured as 45S pre-rRNA) and estimated relative rDNA dosage using
18S (J), 28S (K) and 5.8S (L) determined using qPCR (n = 7).

© 2021 The Authors. The Journal of Physiology © 2021 The Physiological Society

Licenciado para - Anderson Sanches Gonçalves - 06263970901 - Protegido por Eduzz.com


J Physiol 0.0 rDNA regulation with exercise 13

immunoprecipitation and rDNA mapping data (Grandori et al. 2011a; Agrawal & Ganley, 2018), which can be
et al. 2005; Zentner et al. 2011a, 2014; Shiue et al. 2014; defined as H3K4me1/2 enriched (Zentner et al. 2011b).
Agrawal & Ganley, 2018) were used to guide our analysis Because Myc was robustly up-regulated with RE, we
and infer implications of methylation patterns revealed looked for methylation differences in rDNA regions
by RRBS. Of the few CpG sites altered at both time where MYC protein may associate. Thirty minutes after
points after RE, site 42,523 relative to the TSS had high RE, an IGS CpG site in an area with MYC binding affinity
coverage across individuals and was hypomethylated (Grandori et al. 2005; Agrawal & Ganley, 2018) was
relative to Pre (55% ± 22%) at 30 min (34% ± 11%, modestly hypomethylated (site 17,711, 95% ± 6%,
P = 0.0002, q value FDR = 0.03) and 24 h (19% ± 10%, P = 0.0005, FDR = 0.08) relative to Pre (100% ± 0%)
P = 0.00003, FDR = 0.01) (Fig. 6B); this site is in a (Fig. 6C). Upstream of a region where MYC is highly
region characteristic of the rDNA enhancer (Zentner enriched on human rDNA (∼13 kb from the TSS)

Figure 5. rDNA promoter methylation at rest measured via massARRAY EpiTYPER


A, illustration of specific CpG sites in the promoter region of rDNA that were measured in this study. Sites 2 and 4
were combined as a result of them having the same molecular weight. B, average rDNA promoter CpG methylation
levels across all sites measured. Different colours represent the same participants across sites (n = 24). C, rDNA CpG
methylation levels at individual sites in the promoter region (n = 24). D, relationship between ribosome biogenesis
and percent promoter methylation (n = 21, Pearson r = 0.24, P = 0.15). E, relationship between RNA concentration
(ng per mg of tissue) and percent average CpG promoter methylation (n = 23, Pearson r = 0.10, P = 0.33). Data
are shown as the mean ± SD.

© 2021 The Authors. The Journal of Physiology © 2021 The Physiological Society

Licenciado para - Anderson Sanches Gonçalves - 06263970901 - Protegido por Eduzz.com


14 V. C. Figueiredo and others J Physiol 0.0

(Grandori et al. 2005), a CpG site was hypomethylated 33,806 and 34,428 were hypermethylated (FDR < 0.05,
30 min after RE (site 12,054, 82% ± 15%, P = 0.0007, data not shown). Finally, a cluster of hypermethylated
FDR = 0.09) relative to Pre (98% ± 4%). Human rDNA CpG sites 30 min after RE emerged 4–5 kb downstream of
has a unique enhancer-like region in the IGS (Zentner the TSS in the 18S coding region (4359, 4367, 4369, 4374
et al. 2011a) that contains MYC occupancy sites (Agrawal and 4382) (FDR < 0.05, data not shown).
& Ganley, 2018) and codes for a stress-responsive IGS
transcript (IGS28 RNA) (Audas et al. 2012). Immediately MYC transcription and rDNA methylation changes
upstream of this region, two highly methylated CpG sites observed in human biopsy samples with RE were
in close proximity (sites 27,603 and 27,614) were both generally conserved with an acute hypertrophic
∼30% hypomethylated 30 min after RE (35% ± 22%, stimulus in mice
P = 0.0003, FDR = 0.05 and 34% ± 24%, P = 0.002,
FDR = 0.21, respectively) relative to Pre (68% ± 22% Because skeletal muscle is a mixed tissue containing nuclei
and 73% ± 23%, respectively) (Fig. 6D). Other hypo- from various cell types, any epigenetic changes in rDNA
methylated CpG sites 30 min after RE were 4159 genes in myonuclei could be obscured by non-muscle
and 34,987, and hypermethylated sites were 11,938, nuclei. Using a novel murine in vivo genetic myonuclear
20,819, 33,349 and 33,624 (FDR < 0.05, data not shown). labelling strategy (HSA-GFP) to investigate global DNA
Twenty-four hours after RE, CpG sites 37,512 and 39,163 methylation changes in response to an acute hypertrophic
were hypomethylated, and 765, 11,938, 20,819, 33,349, stimulus, we previously reported that the responses are

A 50
(average for promoter)
% rDNA methylation

40

30

20

10

0
Pre 30min 3h 8h 24h Pre 30min 3h 8h 24h Pre 30min 3h 8h 24h
CON EE RE

B Enhancer C MYC-associated IGS D MYC/IGS Transcript Region


Pre Pre
120 Pre 120 30 min 120 30 min
30 min
100 24h 100
* 24h
100
24h
% Methylation

% Methylation

% Methylation

80 80 80 * *
60 * 60 60

40 * 40 40

20 20 20

0 0 0
42523 17711 27603 27614
rDNA CpG Site Position

Figure 6. rDNA methylation in response to acute RE


A, average promoter CpG methylation measured via massARRAY EpiTYPER in control (CON;
Pre/30 min/3 h/8 h/24 h, n = 8/10/9/9/10), EE (Pre/30 min/3 h/8 h/24 h, n = 8/9/9/9/9) and RE
(Pre/30 min/3 h/8 h/24 h, n = 8/9/9/5/7) participants over a 24 h time course in human skeletal muscle.
ANOVA results: interaction, P = 0.784; main effect for group, P < 0.001; main effect for time, P = 0.661. B,
CpG methylation at a site in the rDNA enhancer region Pre (n = 8), 30 min (n = 8) and 24 h (n = 7) after RE
(30 min, P = 0.0002, q value FDR = 0.03; 24 h, P = 0.00003, FDR = 0.01). C, CpG methylation at a site in the
rDNA associated with intergenic spacer (IGS) MYC occupancy area Pre (n = 5), 30 min (n = 6) and 24 h (n = 6)
after RE (P = 0.0005, FDR = 0.08). D, CpG methylation at sites in the IGS that is enhancer-like and immediately
upstream of an IGS-derived transcript Pre (27,603, n = 6, 27,614 n = 5), 30 min (27,603, n = 5, 27,614, n = 4)
and 24 h (27,603, n = 6, 27,614, n = 5) after RE (site 27,603, 30 min, P = 0.0003, FDR = 0.05; site 27,614, 24 h,
P = 0.002, FDR = 0.21). ∗ P < 0.01 relative to Pre. Data are shown as the mean ± SD.

© 2021 The Authors. The Journal of Physiology © 2021 The Physiological Society

Licenciado para - Anderson Sanches Gonçalves - 06263970901 - Protegido por Eduzz.com


J Physiol 0.0 rDNA regulation with exercise 15

distinct between purified myonuclei and interstitial nuclei OV (P < 0.05) (Fig. 8C). There are three major occupancy
(von Walden et al. 2020a). Here, we confirm that 72 h regions for Myc protein in mouse rDNA according to
of mechanical OV of the plantaris muscle via synergist chromatin immunoprecipitation sequencing: canonical
ablation in mice was associated with significantly higher binding in the upstream core element/promoter, a site
abundance of the 45S pre-rRNA (P = 0.047) (Fig. 7A). within 1 kb downstream of the TSS, and in a ∼7–13
In vivo metabolic labelling of nascent RNA revealed that kb downstream region (Zentner et al. 2014); differential
greater levels of rRNA was a result of de novo synthesis, methylation during OV coincided with the latter two
as illustrated by the elevated abundance of this transcript regions (Fig. 8A). The 201–300 kb (53 ± 10%) and
in the EU-labelled fraction following OV (P = 0.021) 401–500 kb (8 ± 7%) regions downstream of the TSS
(Fig. 7A). Myc was also highly enriched in the EU fraction were hypomethylated with OV (P = 0.01, FDR = 0.12
after 72 h of OV (P = 0.0085) (Fig. 7B). These findings and P = 0.02 and FDR = 0.14, respectively) relative
verified that rRNA and Myc are primarily regulated at to sham (73% ± 2% and 24% ± 6%, respectively),
the transcriptional level in response to OV, and not by within which specific CpG sites were significantly hypo-
post-transcriptional mechanisms, such as enhanced RNA methylated (56% ± 9% at CpG site 273 and 28 ± 8%
stability. at site 466, one OV data point missing for site 466,
To complement the human RRBS data, we gathered P < 0.05) relative to sham (75% ± 1% at site 273 and
detailed information on the epigenetic regulation of rDNA 92% ± 11% at site 466). The 11,601–11,700 downstream
specifically within myonuclei in response to an acute region was hypermethylated during OV (30% ± 10%,
hypertrophic stimulus in mice. Fluorescence labelled P = 0.02, FDR = 0.14) relative to sham (9% ± 1%)
myonuclei from HSA-GFP mice were isolated via FACS with an individual significantly hypermethylated CpG site
(Fig. 8A) and myonuclear rDNA methylation in sham and therein (2.5% ± 3.5% sham vs. 30% ± 2% OV at site
72 h OV plantaris muscle was analysed (Fig. 8B). DNA 11,674, one site missing in OV, P = 0.047). On average,
methylation patterns were altered in regions throughout methylation in the 7–13 kb region was low and slightly
the rDNA repeat. Similar to humans, we observed hypermethylated with OV (∼23% vs. 27%), although we
differential methylation patterns with OV relative to observed that, in individual differentially methylated CpG
sham in an rDNA enhancer region ∼43–45 kb down- sites where methylation was above 50% at either time point
stream of the TSS (Zentner et al. 2014), with hyper- in this region, hypomethylation with OV predominated
methylation in the 44,501–44,600 kb region (29% ± 9%, (P < 0.05) (Fig. 8D). Finally, in the IGS, the 39,401–39,500
P = 0.02, FDR = 0.14) relative to sham (8% ± 4%) region was hypomethylated after OV (85% ± 5%, P = 0.03,
and hypomethylation in the 44,701–48,000 kb region FDR = 0.14) relative to sham (95% ± 1%) with one specific
(20% ± 3%, P = 0.002, FDR = 0.08) relative to sham CpG site therein hypomethylated with OV (93% ± 2%
(32% ± 4%) (Fig. 8B). In this enhancer region, differential sham vs. 86 ± 6% OV at CpG site 39,484, one site missing
methylation generally occurred at sites with higher levels in OV, P = 0.06).
of methylation (>50%) and, where methylation was >50%
at either time point, hypomethylation predominated with
Discussion
80 The present study reveals that rDNA transcription in
A 30 B
response EE or RE is not associated with changes
* in promoter methylation, but may be influenced by
60
45S pre-rRNA (AU)

Myc mRNA (AU)

20 methylation within enhancer, IGS and MYC binding


40
regions specifically with RE. The human findings are
supported by myonuclear-specific rDNA methylation
10
* after acute OV in mice, indicating a conserved response in
20
murine muscle fibres. rDNA copy number correlates to the
ribosome biogenesis response to RE, whereas the skeletal
0 0
Sham OV Sham OV Sham OV muscle response to EE appears to be less reliant on genetic
Total RNA Nascent RNA Nascent RNA and epigenetic factors regulating rDNA transcription.
Ribosome biogenesis is suppressed in response to EE,
Figure 7. Ribosome biogenesis and Myc levels in acutely indicating that rDNA transcription is a feature specific
(72 h) OV plantaris muscle with metabolic RNA labelling to hypertrophic adaptations, at least within the 24 h time
A, total (sham vs. OV P = 0.047) and metabolically labelled (nascent,
window after an acute bout.
sham vs. OV P = 0.021) rRNA in sham (n = 2) and 72 h OV muscle
(n = 3). B, Myc levels in the nascent RNA fraction in sham (n = 2) vs. To characterize our stimuli at the molecular level,
72 h OV muscle (n = 3), P = 0.009. ∗ P < 0.05. Data are shown as the we measured ribosome biogenesis and phosphorylation
mean ± SD. status of key proteins in signalling transduction pathways

© 2021 The Authors. The Journal of Physiology © 2021 The Physiological Society

Licenciado para - Anderson Sanches Gonçalves - 06263970901 - Protegido por Eduzz.com


16 V. C. Figueiredo and others J Physiol 0.0

Figure 8. Myonuclear-specific rDNA methylation in acutely OV mice measured via RRBS


A, experimental study design illustrating the treatment and OV of HSA-GFP mice, myonuclear-specific GFP labelling
and isolation of highly-purified GFP+ myonuclei via fluorescence activated cell sorting. B, myonuclear rDNA CpG
methylation in sham and 72-h OV plantaris muscle: 201–300, P = 0.01, FDR = 0.12; 401–500, P = 0.02, FDR = 0.14;
11,601–11,700, P = 0.02, FDR = 0.14; 39,401, P = 0.03, FDR = 0.15; 45,501–44,600, P = 0.02, FDR = 0.14;
44,701–44,800, P = 0.002, FDR = 0.08, n ≥ 1 sham and ≥ 2 OV. rDNA repeat is depicted above the graph based
on CpG coverage. C, CpG methylation in the murine rDNA enhancer region (sites with >50% methylation). CpG
site 43,509, P = 0.038; 43,926, P = 0.009; 43,993, P = 0.003; 44,161, P = 0.02; 44,839, P = 0.04; 44,974, P = 0.04,
biological duplicate for sham and triplicate for OV. D, CpG methylation in murine rDNA Myc occupancy region (sites
with >50% methylation). CpG site 8598, P = 0.03; 8610, P = 0.02; 8712, P = 0.049; 9753, P = 0.017; 12 543,
P = 0.004, biological duplicate for sham and triplicate for OV. ∗ P < 0.05. Data are shown as the mean ± SD.

© 2021 The Authors. The Journal of Physiology © 2021 The Physiological Society

Licenciado para - Anderson Sanches Gonçalves - 06263970901 - Protegido por Eduzz.com


J Physiol 0.0 rDNA regulation with exercise 17

known to be responsive to exercise, specifically AMPK available rDNA template (i.e. number of rDNA copies).
and mTORC1. Acute EE may repress ribosome biogenesis The sustained (late) ribosome biogenesis response after
(Hansson et al. 2019) and ribosome biogenesis throughout RE may therefore have a genetic predisposition, which
our time course (pre-45S rRNA) was generally repressed could translate to hypertrophic potential.
by EE but was induced at 3 h and 24 h after RE. In muscle undergoing hypertrophy, histone
AMPK signalling was more prominent with EE vs. RE, modifications at the rDNA promoter coincide with
broadly consistent with the literature (Vissing et al. 2013; increased rDNA transcription (von Walden et al. 2012).
Murach & Bagley, 2016). mTORC1 is involved in muscle In light of this epigenetic regulation of rDNA during
ribosome biogenesis (von Walden et al. 2016) and p70S6K muscle cell growth, and in context with prior studies
Thr389 is a direct target of mTORC1, known to be highly reporting genome-wide promoter methylation changes
phosphorylated in response to RE. Phosphorylation of with acute exercise (Barres et al. 2012; Sharples et al. 2016;
this site was significantly increased in the RE group Seaborne et al. 2018; Sharples & Seaborne, 2019; Turner
throughout the time course, but was also stimulated et al. 2019; Maasar et al. 2020), we hypothesized that CpG
by feeding alone and endurance exercise with feeding methylation changes to the rDNA promoter region may
at the 24 h time point, uncoupled from ribosome associate with the response to RE in muscle; however,
biogenesis. Our ribosome biogenesis findings generally this was not the case. rDNA promoter methylation in our
provide human in vivo support for a hypothesis where adult muscle samples was low at rest (∼20% on average),
mTOR promotes Pol I transcription, whereas AMPK consistent with our previous report in children (von
inhibits it (von Walden, 2019), but also suggest that Walden et al. 2020b), suggesting the promoter is generally
mechanisms beyond signalling play a role in ribosome available for transcription in a CpG methylation context,
biogenesis with acute exercise. Furthermore, changes in and this does not change with acute EE or RE.
transcriptional regulators involved in ribosome biogenesis In addition to promoters, various regulatory elements
and rRNA processing were unrelated to changes in 45S such as enhancer regions and non-canonical transcription
pre-rRNA at any time point, whereas the early induction factor binding regions may also influence rDNA
of MYC did coincide with ribosome biogenesis. Although transcription (Zentner et al. 2011a; Shiue et al. 2014;
robust up-regulation of MYC is consistent with its role Zentner et al. 2014). Hypermethylation of rDNA enhancer
in hypertrophic muscle adaptation (Chaillou et al. 2014; regions generally signifies an inactive gene (Brock & Bird,
Wen et al. 2016; Figueiredo & McCarthy, 2019b; von 1997; Stancheva et al. 1997). Hypomethylation of rDNA
Walden, 2019), the overall signalling and transcriptional enhancer sites in humans and mice after a hypertrophic
findings motivated us to explore alternative regulatory stimulus, as well as near a presumed IGS enhancer
mechanisms for ribosome biogenesis. site/transcript coding region (Zentner et al. 2011a; Audas
Skeletal muscle mass regulation has long been hypo- et al. 2012), suggests that epigenetic remodelling of
thesized to possess a strong genetic component (Seeman the rDNA repeat may support rDNA transcription in
et al. 1996). Although the search for genetic explanations muscle fibres in response to exercise. In our human
of muscle hypertrophy have largely focused on normal data, a CpG in the enhancer region showed stepwise
protein-coding genes and gene polymorphisms (Ivey et al. hypomethylation 30 min and 24 h after RE, suggesting
2000; Riechman et al. 2004; Charbonneau et al. 2008; Li a protracted role in rDNA regulation. MYC, a universal
et al. 2014), rDNA copy number has been overlooked. amplifier of expressed genes (Nie et al. 2012, 2020), is an
We first utilized WGS to quantify rDNA copy number in rDNA-associated transcription factor that is central to
nine selected individuals, then correlated the results with ribosome biogenesis (Arabi et al. 2005), protein synthesis
a qPCR method using validated primers from a previous (Van Riggelen et al. 2010; West et al. 2016) and growth
study (Gibbons et al. 2015) to confirm the veracity of this (Kim et al. 2000; Xiao et al. 2001; Zhong et al. 2006).
approach in our samples. The qPCR method correlated MYC localizes in myonuclei during development and
well with the WGS data, supporting qPCR based relative hypertrophy (Alway, 1997; Veal & Jackson, 1998) and its
rDNA gene dosage estimation. In our cohort, there was DNA binding extends beyond canonical E-box motifs
a 3-fold difference between individuals with the lowest (Nie et al. 2012, 2020; Guo et al. 2014; Allevato et al.
and the highest relative rDNA dosage, aligning with what 2017 ) and is inhibited by CpG methylation (Prendergast
has been reported previously (Gibbons et al., 2014b). The et al. 1991; Perini et al. 2005). A site in the promoter of
available evidence indicates that 45S pre-rRNA levels may Myc is hypomethylated after 72 h of OV in mouse myo-
peak at 24h after RE (Stec et al. 2015; Figueiredo et al. nuclei (von Walden et al. 2020a), which corresponds with
2016). We observed inductions at 3 h and 24 h that were nascent Myc transcription in our murine experiments,
divergent from the EE responses, where 45S pre-rRNA as well as Myc translation (Chen et al. 2002) and protein
was repressed relative to RE. Our data suggest rDNA accumulation after RE in human and rodent muscle
transcription after acute exercise is mode-dependent, and (Apró et al. 2013, Figueiredo et al. 2016, Ogasawara
that the late RE response is positively related to the et al. 2016). We speculate that altered methylation of

© 2021 The Authors. The Journal of Physiology © 2021 The Physiological Society

Licenciado para - Anderson Sanches Gonçalves - 06263970901 - Protegido por Eduzz.com


18 V. C. Figueiredo and others J Physiol 0.0

MYC-associated areas in rDNA is one component of a Alway SE (1997). Overload-induced C-Myc oncoprotein
co-ordinated epigenetic regulatory network involving is reduced in aged skeletal muscle. J Gerentol Ser A 52,
MYC up-regulation, ribosome biogenesis and hyper- B203–B211.
trophic adaptation in skeletal muscle. There is also a Allevato M, Bolotin E, Grossman M, Mane-Padros D, Sladek
functional link between dynamic MYC binding to IGS FM, & Martinez E (2017). Sequence specific binding by
MYC/MAX to low-affinity non-E-box motifs. PloS One 12,
rDNA, rearrangement of nuclear architecture and rDNA
e0180147.
transcription specifically during growth in human cells Apró W, Wang L, Pontén M, Blomstrand E & Sahlin K (2013)
(Shiue et al. 2014), although more work is needed to Resistance exercise induced mTORC1 signaling is not
uncover the temporal and spatial aspects of MYC inter- impaired by subsequent endurance exercise in human
action with rDNA during muscle hypertrophy. In light skeletal muscle. Am J Physiol Endo Metab 305, E22–E32.
of the unique functional roles for transcripts originating Arabi A, Wu S, Ridderstråle K, Bierhoff H, Shiue C, Fatyol K,
from the rDNA IGS (Mayer et al. 2006; Santoro et al. Fahlén S, Hydbring P, Söderberg O & Grummt I (2005).
2010; Audas et al. 2012; Vacík et al. 2019), further inquiry c-Myc associates with ribosomal DNA and activates RNA
into the consequences and stability of repeat-wide rDNA polymerase I transcription. Nat Cell Biol 7, 303–310.
methylation with exercise will broaden the emerging area Ato S, Maruyama Y, Yoshizato H & Ogasawara R (2020).
of epigenetic regulation of rDNA transcription in skeletal Habitual high-protein diet does not influence muscle
protein synthesis in response to acute resistance exercise
muscle.
in rats. Nutrition 78, 110795.
Although the current investigation represents a Audas TE, Jacob MD & Lee S (2012). Immobilization of
challenging study design and provides new directions proteins in the nucleolus by ribosomal intergenic spacer
in understanding the regulation of ribosome biogenesis noncoding RNA. Mol Cell 45, 147–157.
with exercise, our sample sizes were relatively small for a Baldridge GD, Dalton MW & Fallon AM (1992). Is
randomized trial, and the mouse study was limited in size higher-order structure conserved in eukaryotic ribosomal
as a result of the tedious nature and high cost of low-input DNA intergenic spacers? J Mol Evo 35, 514–523.
myonucleus-specific RRBS. As such, the results should be Barres R, Yan J, Egan B, Treebak JT, Rasmussen M, Fritz T,
subject to replication and confirmation in a larger study. Caidahl K, Krook A, O’Gorman DJ & Zierath JR (2012).
The induction of rDNA transcription induced by RE is Acute exercise remodels promoter methylation in human
positively associated with rDNA gene dosage in humans, skeletal muscle. Cell Metab 15, 405–411.
Begue G, Raue U, Jemiolo B & Trappe S (2017). DNA
at least after 24 h. Given the large variability of rDNA
methylation assessment from human slow-and fast-twitch
copy number across individuals, and that the muscle skeletal muscle fibers. J Appl Physiol 122, 952–967.
hypertrophy response to exercise is highly heterogeneous Benjamini Y & Hochberg H (1995). Controlling the false
(Churchward-Venne et al. 2015; Ahtiainen et al. 2016; discovery rate: a practical and powerful approach to
Lavin et al. 2019), we propose that rDNA dosage is a multiple testing. J Royal Soc B 57, 289–300.
potential genetic factor that relates to skeletal muscle Bjorkman F, Ekblom-Bak E, Ekblom O & Ekblom B (2016).
hypertrophy induced by resistance training. Acute RE Validity of the revised Ekblom Bak cycle ergometer test in
also elicits reorganization of rDNA methylation patterns adults. Eur J Appl Physiol 116, 1627–1638.
along the repeat. The specific consequences and lasting Borg G (1970). Perceived exertion as an indicator of somatic
effects of this acute epigenetic remodelling of rDNA stress. Scand J Rehabil Med 2, 92–98.
deserve further investigation, but, if persistent over time, Brock GJ & Bird A (1997). Mosaic methylation of the repeat
unit of the human ribosomal RNA genes. Human Molec
they could contribute to a previously observed epigenetic
Gene 6, 451–456.
‘epi-memory’ of prior exercise exposure that may facilitate Camelon KM, Hadell K, Jamsen PT, Ketonen KJ, Kohtamaki
future muscle adaptability (Sharples et al. 2016; Seaborne HM, Makimatilla S, Tormala ML & Valve RH (1998). The
et al. 2018; Moberg et al. 2020; Murach et al. 2020; Snijders Plate Model: a visual method of teaching meal planning.
et al. 2020). DAIS Project Group. Diabetes atherosclerosis intervention
study. J Am Diet Assoc 98, 1155–1158.
Cedar H & Bergman Y (2009). Linking DNA methylation and
histone modification: patterns and paradigms. Nat Rev Gene
References
10, 295–304.
Agrawal S & Ganley AR (2018). The conservation landscape Chaillou T, Kirby TJ & McCarthy JJ (2014). Ribosome
of the human ribosomal RNA gene repeats. PloS One 13, biogenesis: emerging evidence for a central role in the
e0207531. regulation of skeletal muscle mass. J Cell Physiol 229,
Ahtiainen JP, Walker S, Peltonen H, Holviala J, Sillanpää E, 1584–1594.
Karavirta L, Sallinen J, Mikkola J, Valkeinen H & Mero A Charbonneau DE, Hanson ED, Ludlow AT, Delmonico MJ,
(2016). Heterogeneity in resistance training-induced muscle Hurley BF & Roth SM (2008). ACE genotype and the
strength and mass responses in men and women of different muscle hypertrophic and strength responses to strength
ages. Age 38, 10. training. Med Sci Sports Exerc 40, 677–683.

© 2021 The Authors. The Journal of Physiology © 2021 The Physiological Society

Licenciado para - Anderson Sanches Gonçalves - 06263970901 - Protegido por Eduzz.com


J Physiol 0.0 rDNA regulation with exercise 19

Chen YW, Nader GA, Baar KR, Fedele MJ, Hoffman EP & Gibbons JG, Branco AT, Yu S & Lemos B (2014a). Ribosomal
Esser KA (2002). Response of rat muscle to acute resistance DNA copy number is coupled with gene expression
exercise defined by transcriptional and translational variation and mitochondrial abundance in humans. Nat
profiling. J Physiol 545, 27–41. Comm 5, 1–12.
Churchward-Venne TA, Tieland M, Verdijk LB, Leenders M, Gonzalez IL & Sylverster JE (1995). Complete sequence of
Dirks ML, de Groot LCPGM & van Loon LJC (2015). There the 43-kb human ribosomal DNA repeat: analysis of the
are no nonresponders to resistance-type exercise training intergenic spacer. Genomics, 27, 320–328.
inolder men and women. J Am Med Dir Assoc 16, 400–411. Grandori C, Gomez-Roman N, Felton-Edkins ZA, Ngouenet
D’Aquila P, Montesanto A, Mandala M, Garasto S, Mari C, Galloway DA, Eisenman RN & White RJ (2005).
V, Corsonello A, Bellizzi D & Passarino G (2017). c-Myc binds to human ribosomal DNA and stimulates
Methylation of the ribosomal RNA gene promoter is transcription of rRNA genes by RNA polymerase I. Nat
associated with aging and age-related decline. Aging Cell Cell Biol 7, 311–318.
16, 966–975. Grozdanov P, Georgiev O & Karagyozov L (2003). Complete
Dungan CM, Murach KA, Frick KK, Jones SR, Crow SE, sequence of the 45-kb mouse ribosomal DNA repeat:
Englund DA, Vechetti Jr. IJ, Figueiredo VC, Levitan BM, analysis of the intergenic spacer. Genomics 82, 637–643.
Satin J, McCarthy JJ, & Peterson CA (2019). Elevated myo- Grummt I (2007). Different epigenetic layers engage in
nuclear density during skeletal muscle hypertrophy in complex crosstalk to define the epigenetic state of
response to training is reversed during detraining. Am J mammalian rRNA genes. Hum Mol Gene 16, R21–R27.
Physiol Cell Physiol 316, C649-654. Guo J, Li T, Schipper J, Nilson KA, Fordjour FK, Cooper
Eden S, Hashimshony T, Keshet I, Cedar H & Thorne A JJ, Gordan R & Price DH (2014). Sequence specificity
(1998). DNA methylation models histone acetylation. incompletely defines the genome-wide occupancy of Myc.
Nature 394, 842–842. Genome Biol 15, 482.
Ekblom-Bak E, Bjorkman F, Hellenius ML & Ekblom B Haltiner MM, Smale ST & Tjian R (1986). Two distinct
(2014). A new submaximal cycle ergometer test for promoter elements in the human rRNA gene identified
prediction of VO2 max. Scand J Med Sci Sports 24, by linker scanning mutagenesis. Mol Cell Biol 6,
319–326. 227–235.
Fernandez-Gonzalo R, Lundberg TR & Tesch PA (2013). Acute Hammarstrom D, Ofsteng S, Koll L, Hanestadhaugen M,
molecular responses in untrained and trained muscle sub- Hollan I, Apro W, Whist JE, Blomstrand E, Ronnestad BR
jected to aerobic and resistance exercise training versus & Ellefsen S (2020). Benefits of higher resistance-training
resistance training alone. Acta Physiol 209, 283–94. volume are related to ribosome biogenesis. J Physiol 598,
Figueiredo VC (2019a). Revisiting the roles of protein 543–565.
synthesis during skeletal muscle hypertrophy induced Hansson B, Olsen LA, Nicoll JX, von Walden F, Melin
by exercise. Am J Physiol Reg Integr Comp Physiol 317, M, Strömberg A, Rullman E, Gustafsson T, Fry AC,
R709–R718. Fernandez-Gonzalo R & Lundberg TR (2019) Skeletal
Figueiredo VC, Caldow MK, Massie V, Markworth JF, muscle signaling responses to resistance exercise of the
Cameron-Smith D & Blazevich AJ (2015). Ribosome elbow extensors are not compromised by a preceding bout
biogenesis adaptation in resistance training-induced human of aerobic exercise. Am J Physiol Reg Comp Integr Physiol.
skeletal muscle hypertrophy. Am J Physiol Endo Metab 30, 317, R83–R92.
E72–E83. Ivey FM, Roth SM, Ferrell RE, Tracy BL, Lemmer JT, Hurlbut
Figueiredo VC, Englund DA, Vechetti IJ Jr., Alimov A, DE, Martel GF, Siegel EL, Fozard JL, Jeffrey Metter E, Fleg
Peterson CA & McCarthy JJ (2019). Phosphorylation JL & Hurley BF (2000). Effects of age, gender, and myostatin
of eukaryotic initiation factor 4E is dispensable for genotype on the hypertrophic response to heavy resistance
skeletal muscle hypertrophy. Am J Physio Cell Physiol 317, strength training. J Gerentol Ser A 55, M641–M648.
C1247–C1255. Iwata M, Englund DA, Wen Y, Dungan CM, Murach KA,
Figueiredo VC & McCarthy JJ (2019b). Regulation of Vechetti IJ, Mobley CB, Peterson CA & McCarthy JJ (2018).
ribosome biogenesis in skeletal muscle hypertrophy. Physio- A novel tetracycline-responsive transgenic mouse strain for
logy 34, 30–42. skeletal muscle-specific gene expression. Skelet Muscle 8, 33.
Figueiredo VC, Roberts LA, Markworth JF, Barnett MPG, Jozsi AC, Dupont-Versteegden EE, Taylor-Jones JM, Evans WJ,
Coombes JS, Raastad T, Peake JM & Cameron-Smith Trappe TA, Campbell WW & Peterson CA (2000). Aged
D (2016). Impact of resistance exercise on ribosome human muscle demonstrates an altered gene expression
biogenesis is acutely regulated by post-exercise recovery profile consistent with an impaired response to exercise.
strategies. Physiol Rep 4, e12670. Mech Age Dev 120, 45–56.
Fuks F (2005). DNA methylation and histone modifications: Kim HG, Guo B & Nader GA (2019). Regulation of ribosome
teaming up to silence genes. Curr Opin Gene Devel 15, biogenesis during skeletal muscle hypertrophy. Exerc Sport
490–495. Sci Rev 47, 91–97.
Gibbons JG, Branco AT, Godinho SA, Yu S & Lemos B (2015). Kim S, Li Q, Dang CV & Lee LA (2000). Induction
Concerted copy number variation balances ribosomal DNA of ribosomal genes and hepatocyte hypertrophy by
dosage in human and mouse genomes. Proc Nat Acad Sci adenovirus-mediated expression of c-Myc in vivo. Proc
U S A 112, 2485–2490. Nat Acad Sci U S A 97, 11198–11202.

© 2021 The Authors. The Journal of Physiology © 2021 The Physiological Society

Licenciado para - Anderson Sanches Gonçalves - 06263970901 - Protegido por Eduzz.com


20 V. C. Figueiredo and others J Physiol 0.0

Kirby TJ, Patel RM, McClintock TS, Dupont-Versteegden Mayer C, Schmitz K-M, Li J, Grummt I & Santoro R (2006).
EE, Peterson CA & McCarthy JJ (2016a). Myonuclear Intergenic transcripts regulate the epigenetic state of rRNA
transcription is responsive to mechanical load and DNA genes. Mol Cell 22, 351–361.
content but uncoupled from cell size during hypertrophy. McCarthy JJ & Murach KA (2019). Anabolic and catabolic
Mol Biol Cell 27, 788–798. signaling pathways that regulate skeletal muscle
Langmead B & Salzberg SL (2012). Fast gapped-read mass. In Nutrition and Enhanced Sports Performance,
alignment with Bowtie 2. Nat Meth 9, 357. pp. 275–290.Elsevier, Amsterdam.
Langmead B, Wilks C, Antonescu V & Charles R (2019). Moberg M, Lindholm ME, Reitzner SM, Ekblom B, Sundberg
Scaling read aligners to hundreds of threads on C-J & Psilander N (2020). Exercise induces different
general-purpose processors. Bioinformatics 35, 421–432. molecular responses in trained and untrained human
Lavin KM, Roberts BM, Fry CS, Moro T, Rasmussen BB & muscle. Med Sci Sport Exerc 52, 1679–1690.
Bamman MM (2019). The importance of resistance exercise Moss T (2004). At the crossroads of growth control; making
training to combat neuromuscular aging. Physiology 34, ribosomal RNA. Curr Op Gene Dev 14, 210–217.
112–122. Mougey EB, Pape LK & Sollner-Webb B (1996). Virtually the
Leary DJ & Huang S (2001). Regulation of ribosome entire Xenopus laevis rDNA multikilobase intergenic spacer
biogenesis within the nucleolus. FEBS Lett 509, 145–150. serves to stimulate polymerase I transcription. J Biol Chem
Li X, Wang SJ, Tan SC, Chew PL, Liu L, Wang L, Wen L & Ma 271, 27138–27145.
L (2014). The A55T and K153R polymorphisms of MSTN Murach KA & Bagley JR (2016). Skeletal muscle hypertrophy
gene are associated with the strength training-induced with concurrent exercise training: contrary evidence for an
muscle hypertrophy among Han Chinese men. J Sports Sci interference effect. Sport Med 46, 1029–1039.
32, 883–891. Murach KA, Dungan CM, Dupont-Versteegden EE, McCarthy
Louis E, Raue U, Yang Y, Jemiolo B & Trappe S (2007). JJ & Peterson CA (2019). “Muscle memory” not mediated
Time course of proteolytic, cytokine, and myostatin gene by myonuclear number? Secondary analysis of human
expression after acute exercise in human skeletal muscle. J detraining data. J Appl Physiol 127, 1814–1816.
Appl Physiol 103, 1744–1751. Murach KA, Mobley CB, Zdunek CJ, Frick KK, Jones SR,
Lundberg TR, Fernandez-Gonzalo R, Gustafsson T & Tesch McCarthy JJ, Peterson CA & Dungan CM (2020). Muscle
PA (2012). Aerobic exercise alters skeletal muscle molecular memory: myonuclear accretion, maintenance, morphology,
responses to resistance exercise. Med Sci Sports Exerc 44, and miRNA levels with training and detraining in adult
1680–1688. mice. J Cachex Sarc Muscle 11, 1705–1722. doi: https:
Lundberg TR, Fernandez-Gonzalo R, Gustafsson T & Tesch //doi.org/10.1002/jcsm.12617.
PA (2013). Aerobic exercise does not compromise muscle Murayama A, Ohmori K, Fujimura A, Minami H,
hypertrophy response to short-term resistance training. J Yasuzawa-Tanaka K, Kuroda T, Oie S, Daitoku H, Okuwaki
Appl Physiol 114, 81–89. M & Nagata K (2008). Epigenetic control of rDNA
Lundberg TR, Fernandez-Gonzalo R & Tesch PA (2014). loci in response to intracellular energy status. Cell 133,
Exercise-induced AMPK activation does not interfere with 627–639.
muscle hypertrophy in response to resistance training in Nakada S, Ogasawara R, Kawada S, Maekawa T & Ishii N
men. J Appl Physiol 116, 611–620. (2016). Correlation between ribosome biogenesis and the
Lundberg TR, Fernandez-Gonzalo R, Norrbom J, Fischer H, magnitude of hypertrophy in overloaded skeletal muscle.
Tesch PA & Gustafsson T (2014). Truncated splice variant PloS One 11, e0147284–e0147284.
PGC-1α4 is not associated with exercise-induced human Nemeth A, Guibert S, Tiwari VK, Ohlsson R & Längst
muscle hypertrophy. Acta Physiol 212, 142–151. G (2008). Epigenetic regulation of TTF-I-mediated
Lundberg TR, Fernandez-Gonzalo R, Tesch PA, Rullman E promoter–terminator interactions of rRNA genes. EMBO
& Gustafsson T (2016) Aerobic exercise augments muscle J 27, 1255–1265.
transcriptome profile of resistance exercise. Am J Physiol Nie Z, Guo C, Das SK, Chow CC, Batchelor E, Jnr SSS &
Regul Integr Comp Physiol 310, R1279–R1287. Levens D (2020). Dissecting transcriptional amplification
Maasar M-F, Turner DC, Gorski PP, Seaborne RA, Strauss JA, by MYC. eLife 9, e52483.
Shepherd SO, Cocks M, Pillon NJ, Zierath JR, Hulton AT, Nie Z, Hu G, Wei G, Cui K, Yamane A, Resch W, Wang R,
Drust B, Sharples AP (2021). The Comparative Methylome Green DR, Tessarollo L & Casellas R (2012). c-Myc is a
and Transcriptome After Change of Direction Compared universal amplifier of expressed genes in lymphocytes and
to Straight Line Running Exercise in Human Skeletal embryonic stem cells. Cell 151, 68–79.
Muscle. Front Physiol, 12. https://doi.org/10.3389/fphys. Ogasawara R, Fujita S, Hornberger TA, Kitaoka Y, Makanae
2021.619447. Y, Nakazato K & Naokata I (2016). The role of mTOR
Malinovskaya EM, Ershova ES, Golimbet VE, Porokhovnik signalling in the regulation of skeletal muscle mass in a
LN, Lyapunova NA, Kutsev SI, Veiko NN & Kostyuk SV rodent model of resistance exercise. Sci Rep. 6, 31142.
(2018). Copy number of human ribosomal genes with Park Y, Figueroa ME, Rozek LS & Sartor MA (2014).
aging: unchanged mean, but narrowed range and decreased MethylSig: a whole genome DNA methylation analysis
variance in elderly group. Front Gene 9, 306. pipeline. Bioinformatics 30, 2414–2422.

© 2021 The Authors. The Journal of Physiology © 2021 The Physiological Society

Licenciado para - Anderson Sanches Gonçalves - 06263970901 - Protegido por Eduzz.com


J Physiol 0.0 rDNA regulation with exercise 21

Parks MM, Kurylo CM, Dass RA, Bojmar L, Lyden D, Vincent Shiue C-N, Nematollahi-Mahani A & Wright AP (2014).
CT & Blanchard SC (2018). Variant ribosomal RNA alleles Myc-induced anchorage of the rDNA IGS region to
are conserved and exhibit tissue-specific expression. Sci Adv nucleolar matrix modulates growth-stimulated changes
4, eaao0665. in higher-order rDNA architecture. Nucleic Acids Res 42,
Perini G, Diolaiti D, Porro A & Della Valle G (2005). In 5505–5517.
vivo transcriptional regulation of N-Myc target genes is Snijders T, Aussieker T, Holwerda A, Parise G, van Loon L &
controlled by E-box methylation. Proc Nat Acad Sci U S A Verdijk LB (2020). The concept of skeletal muscle memory:
102, 12117–12122. evidence from animal and human studies. Acta Physiol 229,
Pietrzak M, Rempala G, Nelson PT, Zheng J-J & Hetman M e13465.
(2011). Epigenetic silencing of nucleolar rRNA genes in Sparks LM (2017). Exercise training response heterogeneity:
Alzheimer’s disease. PloS One 6, e22585. physiological and molecular insights. Diabetologia 60,
Pilegaard H, Ordway GA, Saltin B & Neufer PD (2000). Trans- 2329–2336.
criptional regulation of gene expression in human skeletal Stancheva I, Lucchini R, Koller T & Sogo JM (1997).
muscle during recovery from exercise. Am J Physiol Endo Chromatin structure and methylation of rat rRNA
Metab 279, E806–E814. genes studied by formaldehyde fixation and psoralen
Pillon NJ, Gabriel BM, Dollet L, Smith JAB, Puig LS, Botella cross-linking. Nucleic Acids Res 25, 1727–1735.
J, Bishop DJ, Krook A & Zierath JR (2020). Transcriptional Stec MJ, Kelly NA, Many GM, Windham ST, Tuggle SC &
profoiling of skeletal muscle adaptations to exericise and Bamman MM (2016). Ribosome biogenesis may augment
inactivity. Nat Comm 11, 1–15. resistance training-induced myofiber hypertrophy and is
Prendergast GC, Lawe D & Ziff EB (1991). Association required for myotube growth in vitro. Am J Physiol Endo
of Myn, the murine homolog of max, with c-Myc Metab 8, E652–E661.
stimulates methylation-sensitive DNA binding and ras Stec MJ, Mayhew DL & Bamman MM (2015). The effects of
cotransformation. Cell 65, 395–407. age and resistance loading on skeletal muscle ribosome
Riechman SE, Balasekaran G, Roth SM & Ferrell biogenesis. J Appl Physiol 119, 851–857.
RE (2004). Association of interleukin-15 protein Takegaki J, Ogasawara R, Kouzaki K, Fujita S, Nakazato
and interleukin-15 receptor genetic variation with K & Ishii N (2020). The distribution of euraryotic
resistance exercise training responses. J Appl Physiol 97, initiation factor 4E after bouts of resistance exericse is
2214–2219. altered by shortening of recovery periods. J Physiol Sci
Roberts RA, Raastad T, Markworth JF, Figueiredo VC, 70, 54.
Egner IM, Shield A, Cameron-Smith D, Coombes JS & Turner DC, Seaborne RA & Sharples AP (2019). Comparative
Peake JM (2015). Post-exercise cold water immersion transcriptome and methylome analysis in human skeletal
attenuates acute anabolic signalling and long-term muscle anabolism, hypertrophy and epigenetic memory. Sci
adaptations in muscle to strength training. J Physiol 593, Rep 9, 1–12.
4285–4301. Uemura M, Zheng Q, Koh CM, Nelson WG,
Santoro R, Schmitz KM, Sandoval J & Grummt I (2010). Inter- Yegnasubramanian S & De Marzo AM (2012). Over-
genic transcripts originating from a subclass of ribosomal expression of ribosomal RNA in prostate cancer is common
DNA repeats silence ribosomal RNA genes in trans. EMBO but not linked to rDNA promoter hypomethylation.
Rep 11, 52–58. Oncogene 31, 1254–1263.
Sase K, Kido K, Ato S, & Fujita S (2020). Effect of resistance Vacík T, Kereïche S, Raška I, Cmarko D & Smirnov E (2019).
training on rat skeletal muscle during severe food Life time of some RNA products of rDNA intergenic spacer
restriction. Transl Sport Med 00, 1–8. in HeLa cells. Histochem Cell Biol 152, 271–280.
Seaborne RA, Strauss J, Cocks M, Shepherd S, O’Brien TD, Van Riggelen J, Yetil A & Felsher DW (2010). MYC as a
van Someren KA, Bell PG, Murgatroyd C, Morton JP, regulator of ribosome biogenesis and protein synthesis.
Stewart CE & Sharples AP (2018). Human skeletal muscle Nat Rev Cancer 10, 301.
possesses an epigenetic memory of hypertrophy. Sci Rep 8, Veal E & Jackson M (1998). C-myc is expressed in mouse
1898. skeletal muscle nuclei during post-natal maturation. Int J
Seeman E, Hopper JL, Young NR, Formica C, Goss P Biochem Cell Biol 30, 811–821.
& Tsalamandris C (1996). Do genetic factors explain Vissing K, McGee SL, Farup J, Kjølhede T, Vendelbo
associations between muscle strength, lean mass, and MH & Jessen N (2013). Differentiated mTOR but not
bone density? A twin study. Am J Physiol 270, E320– AMPK signaling after strength vs endurance exercise in
E327. training-accustomed individuals. Scand J Med Sci Sport 23,
Sharples AP & Seaborne RA (2019). Exercise and DNA 355–66.
methylation in skeletal muscle. In Sports, exercise, von Walden F, Rea M, Mobley CB, Fondufe-Mittendorf Y,
and nutritional genomics, pp. 211–229.Elsevier, McCarthy JJ, Peterson CA & Murach KA (2020a). The
Amsterdam. myonuclear DNA methylome in response to an acute hyper-
Sharples AP, Stewart CE & Seaborne RA (2016). Does skeletal trophic stimulus. Epigenetics 15, 1151–1162.
muscle have an ‘epi’-memory? The role of epigenetics in von Walden F (2019). Ribosome biogenesis in skeletal muscle:
nutritional programming, metabolic disease, aging and coordination of transcription and translation. J Appl Physiol
exercise. Aging Cell 15, 603–616. 127, 591–598.

© 2021 The Authors. The Journal of Physiology © 2021 The Physiological Society

Licenciado para - Anderson Sanches Gonçalves - 06263970901 - Protegido por Eduzz.com


22 V. C. Figueiredo and others J Physiol 0.0

von Walden F, Casagrande V, Östlund Farrants A-K & Nader (GSE144774 and GSE162392). The BioProject ID for the WGS
GA (2012). Mechanical loading induces the expression of data is PRJNA705275.
a Pol I regulon at the onset of skeletal muscle hypertrophy.
Am J Physiol Cell Physiol 302, C1523–C1530.
von Walden F, Fernandez-Gonzalo R, Pingel J, McCarthy Competing interests
J, Stål P & Pontén E (2020b). Epigenetic marks at the The authors declare that they have no competing interests.
ribosomal DNA promoter in skeletal muscle are negatively
associated with degree of impairment in cerebral palsy.
Front Ped 8, 236. Author contributions
von Walden F, Liu C, Aurigemma N & Nader GA (2016).
mTOR signaling regulates myotube hypertrophy by VCF, YW, IJV, TV, CBM, KAM and FvW conducted
modulating protein synthesis, rDNA transcription experiments. VCF, YW, IJV, GEZ, KAM and FvW conducted
and chromatin remodeling. Am J Physiol Cell Physiol data analysis. VCF, YW, GEZ, KAM and FvW generated figures.
311, C663–C672, ajpcell.00144.02016-ajpcell.00144. VCF, KAM and FvW drafted the manuscript. YW and GEZ
02016. assisted with manuscript preparation. BA, RF-G and JN assisted
Wen Y, Alimov AP & McCarthy JJ (2016). Ribosome with data collection. BA, RF-G, JN, CAP, JJM, KAM and FvW
biogenesis is necessary for skeletal muscle hypertrophy. provided resources. KA, FvW and BA co-ordinated the study.
Exerc Sport Sci Rev 44, 110. KAM generated the graphical abstract. All authors read, edited
Wen Y, Vechetti Jr IJ, Valentino TR & McCarthy JJ (2020). and approved the final version of the manuscript submitted for
High-yield skeletal muscle protein recovery from TRIzol publication.
after RNA and DNA extraction. BioTechniques 69,
264–269. Funding
West DW, Baehr LM, Marcotte GR, Chason CM, Tolento L,
Gomes AV, Bodine SC & Baar K (2016). Acute resistance The study was funded by Futurum – the Academy for Health and
exercise activates rapamycin-sensitive and-insensitive Care, Region Jönköping County, Sweden to BA and the Swedish
mechanisms that control translational activity and capacity Kidney Foundation and Swedish Research Council for Sports
in skeletal muscle. J Physiol 594, 453–468. to FvW, and a National Institutes of Health grant (NIH K99
Xiao G, Mao S, Baumgarten G, Serrano J, Jordan MC, Roos AG063994) to KAM.
KP, Fishbein MC & MacLellan WR (2001). Inducible
activation of c-Myc in adult myocardium in vivo provokes
Acknowledgements
cardiac myocyte hypertrophy and reactivation of DNA
synthesis. Circ Res 89, 1122–1129. We thank the subjects for their extraordinary effort and
Zentner GE, Balow SA & Scacheri PC (2014). Genomic participation. We further greatly acknowledge the help from
characterization of the mouse ribosomal DNA locus. G3 staff at Höglandssjukhuset District Hospital in Eksjö where
4, 243–254. the human study took place, especially Annica Eriksson, Lena
Zentner GE, Saiakhova A, Manaenkov P, Adams MD & Norrbrand, Björn Otto and the personnel at the Orthopaedic
Scacheri PC (2011a). Integrative genomic analysis of human Department. We wish to thank Jennifer Strange of the University
ribosomal DNA. Nucleic Acids Res 39, 4949–4960. of Kentucky Flow Cytometry Core, Dr Ann-Charlotte Rönn of
Zentner GE, Tesar PJ & Scacheri PC (2011b). Epigenetic the Karolinska Institutet Mutation Analysis Facility (EpiTyper
signatures distinguish multiple classes of enhancers
analysis) and Dr Keith Booher at Zymo Research. The graphical
with distinct cellular functions. Genome Res 21, 1273–
abstract was generated using BioRender (https://biorender.
1283.
com).
Zhong W, Mao S, Tobis S, Angelis E, Jordan MC, Roos KP,
Fishbein MC, de Alborán IM & MacLellan WR (2006).
Hypertrophic growth in cardiac myocytes is mediated by Keywords
Myc through a cyclin D2-dependent pathway. EMBO J 25,
3869–3879. CpG methylation, enhancer, intergenic spacer, Myc, rDNA copy
number, rDNA

Additional information Supporting information


Additional supporting information may be found online in the
Data availability statement
Supporting Information section at the end of the article.
The raw RRBS data for human and mice experiments have
been deposited in the NCBI Gene Expression Omnibus database Statistical Summary Document

© 2021 The Authors. The Journal of Physiology © 2021 The Physiological Society

Licenciado para - Anderson Sanches Gonçalves - 06263970901 - Protegido por Eduzz.com

You might also like