Early Growth Response 3 (Egr3) Contributes A Maintenance of C2C12 Myoblast Proliferation

You might also like

You are on page 1of 9

ORIGINAL RESEARCH ARTICLE

Journal of
1114
Cellular
Physiology
Early Growth Response 3 (Egr3)
Contributes a Maintenance of
C2C12 Myoblast Proliferation
MITSUTOSHI KUROSAKA, YUJI OGURA,* TOSHIYA FUNABASHI, AND TATSUO AKEMA
Department of Physiology, St. Marianna University School of Medicine, Kawasaki, Kanagawa, Japan

Satellite cell proliferation is a crucially important process for adult myogenesis. However, its regulatory mechanisms remain unknown.
Early growth response 3 (Egr3) is a zinc-finger transcription factor that regulates different cellular functions. Reportedly, Egr3 interacts
with multiple signaling molecules that are also known to regulate satellite cell proliferation. Therefore, it is possible that Egr3 is involved in
satellite cell proliferation. Results of this study have demonstrated that Egr3 transcript levels are upregulated in regenerating mouse
skeletal muscle after cardiotoxin injury. Using C2C12 myoblast culture (a model of activated satellite cells), results show that inhibition of
Egr3 by shRNA impairs the myoblast proliferation rate. Results also show reduction of NF-кB transcriptional activity in Egr3-inhibited cells.
Inhibition of Egr3 is associated with an increase in annexin Vþ cell fraction and apoptotic protein activity including caspase-3 and caspase-7,
and Poly-ADP ribose polymerase. By contrast, the reduction of cellular proliferation by inhibition of Egr3 was partially recovered by
treatment of pan-caspase inhibitor Z-VAD-FMK. Collectively, these results suggest that Egr3 is involved in myoblast proliferation by
interaction with survival signaling.
J. Cell. Physiol. 232: 1114–1122, 2017. ß 2016 Wiley Periodicals, Inc.

Skeletal muscle formation in adults principally originates from The satellite cell function is regulated by many signaling
myogenic stem cells, designated as satellite cells (Brack and molecules. Recent studies have uncovered regulatory
Rando, 2012; Yin et al., 2013; Wang et al., 2014; Dumont interactions between Egr3 and several molecules, including
et al., 2015). Upon injury, for example, the satellite cells signal transducers and activators of transcription (STAT)
become activated (i.e., myoblast); they proliferate and (Li et al., 2012; Sumitomo et al., 2013b), activator protein-1
differentiate to repair or newly form a myofiber (Brack and (Li et al., 2012; Sumitomo et al., 2013b) and nuclear factor-
Rando, 2012; Yin et al., 2013; Wang et al., 2014; Dumont kappa B (NF-кB) (Wieland et al., 2005). These latter
et al., 2015). Previous studies have demonstrated that genetic molecules are known to regulate satellite cell proliferation and
loss of satellite cells engenders a defect of muscle self-renewal (Lehtinen et al., 1996; Guttridge et al., 1999;
regeneration after injury (Lepper et al., 2011; McCarthy et al., Kurosaka and Machida, 2013; Ogura et al., 2013; Price et al.,
2011; Murphy et al., 2011; Fry et al., 2015). Therefore, proper
functioning of satellite cells is important to maintain skeletal
muscle homeostasis. Satellite cell proliferation is a crucially
important step to give a sufficient number of muscle
progenitors during adult myogenesis. Decreased satellite cell Abbreviations: ANOVA, analysis of variance; CTX, cardiotoxin; Egr,
proliferation has been demonstrated to exaggerate muscle early growth response; EdU, 5-ethylnyl-2’-deoxyuridine; GM,
regeneration after injury (Angione et al., 2011; Yoshida et al., growth medium; shRNA, short hairpin RNA; eMyHC, embryonic
2013; Ogura et al., 2015). Moreover, a decline of satellite cell myosin heavy chain; FBS, fetal bovine serum; GAPDH,
proliferation capability accounts for muscle weakness in aged glyceraldehyde-3-phosphate dehydrogenase; NF-кB, nuclear
skeletal muscle (Day et al., 2010; Bernet et al., 2014; Lacraz factor-kappa B; PBS, phosphate buffered saline; PARP, Poly ADP
et al., 2015). However, the regulation of satellite cell ribose polymerase; PPAR-g, peroxisome proliferator activated
proliferation is not yet fully understood. receptor-gamma; QRT-PCR, quantitative reverse transcription-
Early growth response 3 (Egr3) is a zinc-finger transcription polymerase chain reaction; STAT, signal transducers and activator
of transcription; TA, tibialis anterior; XIAP, X-linked inhibitor of
factor that is included in the immediately early genes group apoptosis protein.
(O’Donovan et al., 1999). Accumulated evidence has revealed
the biological role of Egr3 in mammalian cells. Examples of Conflicts of interest: The authors declare that they have no conflict
some roles are activation and cytokine production in T-cells of interest related to this study.
(Safford et al., 2005; Sumitomo et al., 2013a,b), neuronal Contract grant sponsor: The Nakatomi Foundation.
adaptation (O’Donovan et al., 1998), and transduction of Contract grant sponsor: JSPS KAKENHI;
growth signaling in endothelial cells (Suehiro et al., 2010). Contract grant numbers: #24700706, #15K01633.
Moreover, Egr3 has been implicated in the proliferation of Contract grant sponsor: St. Marianna University School of
Medicine.
mammalian cells such as hematopoietic stem cells (Cheng et al.,
2015) and T-cells (Xi et al., 2006). In skeletal muscle, Egr3 is *Correspondence to: Yuji Ogura, Department of Physiology,
reported as vitally important for the development of muscle St. Marianna University School of Medicine, 2-16-1 Sugao,
spindles as a downstream molecule of neuregulin-1/Erbb2 Miyamae-ku, Kawasaki, Kanagawa 216-8511, Japan.
E-mail: yuji_ogura@marianna-u.ac.jp
signaling (Tourtellotte and Milbrandt, 1998; Tourtellotte et al.,
2001; Hippenmeyer et al., 2002; Jacobson et al., 2004). In one Manuscript Received: 27 May 2016
study, specific deletion of ErbB2 in differentiated muscles, but Manuscript Accepted: 29 August 2016
not in satellite cells, caused a defect of muscle regeneration Accepted manuscript online in Wiley Online Library
after a crush injury in mice (Andrechek et al., 2002). However, (wileyonlinelibrary.com): 30 August 2016.
it remains unknown whether Egr3 is also necessary for satellite DOI: 10.1002/jcp.25574
cell function.

© 2 0 1 6 W I L E Y P E R I O D I C A L S , I N C .
REGULATION OF MYOBLAST PROLIFERATION BY Egr3 1115

2014; Tierney et al., 2014; Ogura et al., 2015). Therefore, we electroporation procedure routinely attains 70–80% of transfection
hypothesized that Egr3 might be involved in satellite cell efficiency at 24 h post transfection (Supplementary Fig. S1).
proliferation. This study was conducted to clarify the role of
Egr3 in satellite cell proliferation using C2C12 cells. Indirect cell number measurement

Materials and Methods Vector-transfected cells were grown for 24 and 48 h in GM. Cells at
Ethical approval each time point were counted (cell counting kit-8 #CK04; Dojindo
Laboratories, Kamimashiki-gun, Kumamoto, Japan) according to the
All animal experimental procedures were approved by the manufacturer’s description. The indirect cell number was
Institute for Animal Experimentation, St. Marianna University expressed with absolute absorbance at 450 nm (Multiskan MS; Life
School of Medicine. Experimental procedures using recombinant Technologies Inc.). The proliferation rate was calculated as the cell
DNA were approved by the St. Marianna University Gene number at 48 h divided by the cell number at 24 h.
Recombination Experiment Safety Committee.
5-Ethynyl-20 -deoxyuridine (EdU) incorporation assay
Animals
Vector-transfected myoblasts were grown in GM for 24 h. For the
From SLC Inc. (Hamamatsu, Shizuoka, Japan), 7–8-week-old male final 0.5 and 2 h, cells were labeled with EdU followed by staining
C57BL/6J mice were purchased. They were subsequently with Click-iT Plus EdU cell proliferation assay kit (Alexa fluor 488;
maintained at an institutional animal facility. All animals were given Life Technologies Inc.) (Ogura et al., 2013). Nuclei were
standard rodent chow and water ad libitum. counterstained with Hoechst 33342 in PBS for 30 min at room
temperature. Images were visualized using a fluorescence
Muscle injury microscope (Eclipse Ti-S; Nikon Instech Co., Ltd., Minato-ku,
Tokyo, Japan) attached to a digital camera (Infinity 3-6UR;
To induce injury response in skeletal muscle, 100 ml of 10 mM Lumenera Corporation, Ottawa, ON, Canada). The percentages
cardiotoxin (CTX; Latoxan, Valence, France) dissolved in saline was of EdUþ cells in mCherryþ cells were ascertained. Five images per
injected into one leg of the tibialis anterior (TA) muscle (Ogura et al., sample were analyzed.
2013), under anesthesia using an isoflurane–oxygen gas mixture.
Saline was injected into contralateral TAs as a vehicle treatment. At NF-kB promoter activity
3 and 5 days post CTX injection, TA muscles were excised under
anesthesia with inhalation of the isoflurane–oxygen mixture, after The cells were transfected with NF-kB-Luc plasmid (a gift from
which they were frozen in liquid N2 and stored at 80°C. Dr. Ahosk Kumar, University of Louisville, Louisville, KY) and pRT-TK
vector (Promega Corp., Madison, WI) with either shEgr3 or scramble
Cell culture vectors. After 24 and 48 h, the cells were harvested and processed for
NF-kB-Luc and Renilla activities using a dual luciferase assay kit
Cells of the C2C12 mouse myoblast cell line from the European (#E1910; Promega Corp.). NF-kB-Luc activity was normalized by
Collection of Cell Cultures were used as a model of activated Renilla activity and expressed relative to the control cells.
satellite cells. After purchase from Public Health England (Porton
Down, Salisbury, UK), they were maintained in growth medium Annexin V-propidium iodide (PI) staining
(GM; 10% fetal bovine serum, 1% penicillin/streptomycin, and
Dulbecco’s modified Eagle’s medium, all from Life Technologies The existence of apoptosis was estimated with annexin V-PI
Inc., Carlsbad, CA). The cell’s passage under 11 was used for the staining as described previously (Ogura et al., 2015) using a
experiment. commercially available kit (#4700; Medical and Biological
Laboratories Co. Ltd., Nagoya, Aichi, Japan). In brief, vector-
Short-hairpin RNA (shRNA) transfected cells were grown for 48 h in GM, followed by gentle
removal from culture dishes. After washing with PBS, cells were
The pLKO.1-mCherry-puro plasmid was provided by Dr. Renzhi suspended with binding buffer (85 ml) and were stained with
Han (The Ohio State University Wexner Medical Center, annexin V conjugated with fluorescein (10 ml) and PI (5 ml) for
Columbus, OH). The target siRNA sequences for mouse Egr3 20 min at room temperature (RT) in dark. The cells were then
were designed using BLOCK-iT RNAi Designer (Life diluted with addition of binding buffer and were counted using flow
Technologies Inc.). The synthesized siRNA oligonucleotides cytometry (LSR-II, BD Biosciences, Franklin Lakes, NJ). The data
(Integrated DNA Technologies, Inc., Coralville, IA) were annealed (10,000 events/each) were analyzed using software (FlowJo Ver.
and inserted into the plasmid using AgeI/EcoRI sites to contain, in 10; FlowJo LLC., Ashland, OR). Fractions of annexin V/PIþ,
order, the sense strand of sequences for mouse Egr3 (shEgr3, annexin Vþ/PIþ, annexin Vþ/PI, and annexin Vþ were calculated
GCAACAAGACCGTGACCTACT), a loop sequence respectively to determine necrotic cells, late apoptotic cells, early
(CTCGAG), the reverse complement sequences of sense strand apoptotic cells, and apoptotic cells.
for shEgr3, and a termination signal for RNA polymerase III
(TTTTT) (Ogura et al., 2015). The success of insertion was tested Caspase inhibitor treatment
using cycle DNA sequencing (ABI Prism 3100-Avant; Life
Technologies Inc.). The appropriate plasmid was amplified using a Pan-caspase inhibitor Z-VAD-FMK was purchased from Promega
standard bacterial culture. Two siRNA sequences were validated Corp. For cell treatment, Z-VAD-FMK was added to culture
for knockdown of Egr3 mRNA in preliminary experiments. One medium at a concentration of 20 mM (Ogura et al., 2015) before
was used for this study. Control cells were transfected with the incubation for 24 and 48 h. After the indicated period, the cells
backbone plasmid that harbors the scrambled sequence. were subjected to analysis of the indirect cell number.

Electroporation for plasmid delivery Western blotting


Plasmid DNA was transferred by electroporation (1,500 V, 10 ms, Samples were processed as described previously (Ogura et al.,
3 pulses) using a transfection system (Neon; Life Technologies Inc.) 2013). The primary antibodies used were the following: Egr3
as described in an earlier report (Ogura et al., 2015). Our (1:1000, #2559S; Cell Signaling Technology Inc., CST, Danvers,

JOURNAL OF CELLULAR PHYSIOLOGY


1116 K U R O S A K A E T A L.

MA), cleaved caspase 3 (1:1000; #9664P, CST), cleaved caspase 7 The mouse TA muscle was harvested after 3 and 5 days of the
(1:1000; #8438P, CST), Poly-ADP ribose polymerase (PARP, CTX-induced injury (Fig. 1A). Despite multiple attempts, we
1:2000; #9542P, CST), embryonic myosin heavy chain (eMyHC; were unable to find an effective antibody for detecting Egr3
F1.652, 1:100; Developmental Studies Hybridoma Bank, Iowa City, protein in muscle tissue. Therefore, we analyzed mRNA
IA), developmental myosin heavy chain (dMyHC; NCL-HMCd, levels of Egr3 in TA muscles after injury. Results indicated
1:100; Leica Microsystems Newcastle Ltd., Newcastle Upon Tyne, that mRNA levels of Egr3 in injured TA were increased
UK), and GAPDH (1:10000; #2118S, CST). After incubation with significantly (P < 0.05) compared to control TA at 3 and
an HRP-linked secondary antibody (1:3000, CST), the membrane 5 days (Fig. 1B). No expression of differentiation markers
was developed using enhanced chemiluminescence reagent dMyHC or eMyHC was detected at 3 days in either injured
(General Electric Co., Little Chalfont, Buckinghamshire, UK). or control muscles. However, they were evident at 5 days of
Signals were captured using an imaging system (LAS-4000; Fujifilm injury (Fig. 1C). These results suggest that Egr3 is involved in
Corp., Minato-ku, Tokyo, Japan). Densitometry analysis was muscle regeneration after injury.
conducted using Image J software.
Inhibition of Egr3 impairs C2C12 myoblast proliferation
Quantitative RT-PCR (QRT-PCR)
Muscle regeneration includes complicated processes that
Relative mRNA levels were analyzed using QRT-PCR as described require participation of different cell types. To investigate the
previously, with b-actin as an internal reference for delta–delta Ct myoblast-specific role of Egr3, we conducted experiments
analysis (Ogura et al., 2013) using a real-time PCR system (Step using C2C12 myoblast culture. C2C12 cells were transfected
One; Life Technologies Inc.) with Syber green master mix reagent with pLKO.1-mcherry vector, which harbors either Egr3
(QPS-101; Toyobo Co. Ltd., Osaka, Osaka, Japan). Following are shRNA to inhibit Egr3 or scrambled sequences. Introduction of
the primer sequences (from 50 to 30 ) used for this study: Egr3- shEgr3 significantly (P < 0.05) reduced both mRNA and protein
forward, GCTGCCTCCTTATTCCAACTGC; Egr3-reverse, levels of Egr3 at 24 and 48 h post transfection (Fig. 2A–C).
AAGATTGCTGTCCAAGGCCG; p65-forward, AGGACTC Using this approach, the indirect cell counting assay revealed
TTCTTCATGATACTCTTG; p65-reverse, GAGTTCCAGTA that the cell numbers at 24 and 48 h were significantly
CTTGCCAGAC; b-actin-forward, CATCCGTAAAGACCTC (P < 0.05) lower in shEgr3-transfected cells than in scramble-
TATGCCAAC; and b-actin-reverse, ATGGAGCCACCGATC transfected control cells (Fig. 2D and E). Moreover, the cellular
CACA. proliferation rate during 24–48 h was significantly (P < 0.05)
lower in shEgr3-transfected cells than in scramble-transfected
Statistics cells (Fig. 2F).
We examined the inhibitory effect of Egr3 on myoblast
All values are presented as the mean  SEM. Differences between proliferation further by performing the EdU incorporation
two groups were analyzed using unpaired t-tests. Results for assay in C2C12 myoblast. We inhibited Egr3 with shRNA for
caspase inhibitor experiment were analyzed using one-way analysis 24 h. The cells were then incubated with EdU for an additional
of variance (ANOVA) with a Tukey’s multiple comparison test 0.5 and 2 h, with subsequent visualization of their incorporation
(Prism 6.0; GraphPad Software, Inc., La Jolla, CA). Significance was using Click-iT chemistry (Fig. 2G). As portrayed in Fig. 2H, the
inferred for P < 0.05. percentage of EdUþ incorporated cells in mCherryþ cells
(i.e., vector-transfected cells) was significantly (P < 0.05) lower
Results in shEgr3-transfected cells than in scramble-transfected cells
Egr3 mRNA increases during muscle regeneration after without the effect of the EdU incubation period (i.e., 0.5 and
injury 2 h). Taken together, these results indicate that Egr3 is
implicated in C2C12 myoblast proliferation. These results
We first examined the effect of muscle injury on mRNA were confirmed when different shEgr3 plasmid was used
and protein levels of Egr3 in skeletal muscle of mice. (Supplementary Fig. S2).

Fig. 1. Egr3 mRNA levels increase in regenerating mouse tibialis anterior (TA) muscles. (A) Schematic representation of CTX injury
experiment. (B) mRNA levels of Egr3 after 3 and 5 days of CTX injection. N ¼ 4/each.  P < 0.05 by unpaired t-test. Error bars represent the
SEM. (C) Representative protein bands for dMyHC, eMyHC, and GAPDH in TA muscle. Both MyHC isoforms were undetected at 3 days.

JOURNAL OF CELLULAR PHYSIOLOGY


REGULATION OF MYOBLAST PROLIFERATION BY Egr3 1117

Fig. 2. Egr3 is involved in proliferation of C2C12 myoblast. (A) Egr3 mRNA level in scramble-transfected and shEgr3-transfected cells.
N ¼ 5–6/each for 24 h. N ¼ 3–4/each for 48 h.  P < 0.05 by unpaired t-test. Error bars represent the SEM. (B) Immunoblot for Egr3 in scramble
(scr)-transfected and shEgr3 (sh)-transfected cells. (C) Relative Egr3 protein levels. N ¼ 5/each.  P < 0.05 by unpaired t-test. Error bars
represent the SEM. (D) Representative image of C2C12 cells 48 h after transfection. Scale bar: 100 mm. (E) Indirect cell number in C2C12
myoblast transfected with either scramble or shEgr3. N ¼ 7–8/each.  P < 0.05 by unpaired t-test. Error bars represent the SEM. (F)
Proliferation rate from 24 to 48 h in scramble-transfected and shEgr3-transfected cells. N ¼ 8/each.  P < 0.05 by unpaired t-test. Error bars
represent the SEM. (G) EdUþ cells (green) were counted only in mCherry expressing cells (Red). Scale bar: 100 mm. Arrows indicate EdUþ/
mCherryþ cells. (H) Percentage of EdUþ/mCherryþ cells. N ¼ 4/each.  P < 0.05 by unpaired t-test. Error bars represent the SEM.

NF-кB-Luc activity and p65 protein expression were Egr3 significantly (P < 0.05) decreased the protein expression of
decreased in Egr3-inhibited myoblast p65 at 24 and 48 h compared to control cells (Fig. 3B and C).
The mRNA level of p65 significantly (P < 0.05) increased in
Results of an earlier study suggested interaction between Egr3 shEgr3-transfected cells after 24 h following transfection
and NF-кB signaling in human kidney cells (Wieland et al., 2005). (Fig. 3D). However, no significant difference was found in p65
NF-кB is also known to be important for the progression of the mRNA between Egr3-inhibited and control cells at 48 h after
cell cycle in myoblast (Guttridge et al., 1999). Therefore, we transfection (Fig. 3D). Collectively, these findings indicate that
hypothesized that decreased proliferation in Egr3-inhibited inhibition of Egr3 influences NF-кB activity in C2C12 myoblasts.
cells is associated with NF-кB activity. Figure 3A shows that
NF-кB-Luc activity was decreased significantly (P < 0.05) in
Egr3-ihibited cells compared to control cells at 24 and 48 h post Apoptosis was induced in Egr3-inhibited cells by shRNA
transfection. p65 is an important protein that assembles NF-кB
heterodimer. Therefore, we next investigated the expression of NF-кB is known to play anti-apoptotic roles in cells. Therefore,
p65 in Egr3-inhibited cells. The results indicate that inhibition of we assume that apoptotic cell death might be included in the

JOURNAL OF CELLULAR PHYSIOLOGY


1118 K U R O S A K A E T A L.

Fig. 3. Inhibition of Egr3 reduced the expression of p65 protein and NF-kB promoter activity (A) Relative NF-kB-Luc activity. N ¼ 7–8/each
for 24 h. N ¼ 4/each for 48 h.  P < 0.05 by unpaired t-test. Error bars represent the SEM. (B) Representative immunoblot for p65 and GAPDH.
Scr and sh respectively denote scramble and shEgr3. (C) Relative protein levels of p65. N ¼ 3/each.  P < 0.05 by unpaired t-test. Error bars
represent the SEM. (D) p65 mRNA level in scramble-transfected and shEgr3-transfected cells. N ¼ 7–8/each for 24 h. N ¼ 4/each for 48 h.

P < 0.05 by unpaired t-test. Error bars represent the SEM.

low number of cells in Egr3-inhibited cells. To address this shEgr3-mediated decline of the proliferation rate (Fig. 6B).
point, we transfected myoblast with shEgr3 or scramble Taken together, these results implicate apoptosis in the
vectors and grew them for 48 h. Then the cells were subjected suppression of proliferation in Egr3-inhibited C2C12 myoblast.
to annexin V-PI staining (Fig. 4A). No difference was found in
annexin V/PIþ (Fig. 4B) and annexin Vþ/PIþ (Fig. 4C) between Discussion
scramble-transfected cells and shEgr3-transfected cells.
However, annexin V þ/PI cells (Fig. 4D) and annexin Vþ cells This study was conducted to clarify the role of Egr3 in myoblast
(Fig. 4E) were significantly (P < 0.05) higher in Egr3-inhibited proliferation, a critical process for myogenesis. Our primary
cells than control cells. finding is that inhibition of Egr3 by shRNA impaired cellular
Next, we examined the activation of executor caspases and proliferation in C2C12 myoblasts. It is particularly interesting
PARP proteins in Egr3 inhibited cells by performing Western that the reduction of proliferation was associated with a
blot analysis (Fig. 5A). Results show that a cleaved form of decrease in NF-kB signaling and enhancement of apoptosis.
PARP, caspase 3, and caspase 7 were all significantly (P < 0.05) Pharmacologic caspase inhibitions partially but significantly
increased in Egr3-inhibited cells at 24 and 48 h post transection restore the shRNA-related decline of proliferation. Therefore,
than those in control cells (Fig. 5B–D). we propose that Egr3 is necessary for the maintenance of
Finally, we examined whether inhibition of caspase can myoblast proliferation.
reverse the proliferation of myoblast in Egr3-inhibited cells, or
not. The shEgr3-or scramble-vector transfected cells were Implication of apoptosis
incubated with a pan-caspase inhibitor Z-VAD-FMK or vehicle
(DMSO) for 24 and 48 h, followed by cell count analysis. As Our results suggest involvement of apoptosis in lower
depicted in Fig. 6A, no significant difference was found at 24 h proliferation in Egr3-inhibited cells (Figs. 4–6). Apoptosis is
among groups when the data were analyzed using one-way known to be an important mechanism in the multiple level of
ANOVA. In contrast, the cell number at 48 h post transfection biological events including organ development and cell size
was significantly (P < 0.05) decreased by inhibition of Egr3 with dynamics (Schwartz, 2008; Suzanne and Steller, 2013).
shRNA. Treatment of Z-VAD-FMK significantly (P < 0.05) Additionally, it is important for maintaining cellular
increased the cell number in Egr3-inhibited cells compared proliferation. Indeed, the normality of cell cycling status is well
to those treated with DMSO at 48 h (Fig. 6A). As depicted known to be monitored by check point mechanisms. Cells
in Fig. 6B, inhibition of Egr3 by shRNA significantly showing inappropriate status such as irreversible DNA damage
(P < 0.05) decreased the proliferation rate. However, the is eliminated by apoptosis (Pucci et al., 2000). Therefore, our
treatment of Z-VAD-FMK significantly (P < 0.05) increased the results of increased apoptosis in Egr3-inhibited cells might be

JOURNAL OF CELLULAR PHYSIOLOGY


REGULATION OF MYOBLAST PROLIFERATION BY Egr3 1119

Fig. 4. Annexin V-PI staining in myoblast transfected with scramble or shEgr3. (A) Typical gating results of annexin V-PI staining by flow
cytometry. Quantitative results for a fraction of annexin V/PIþ (B), annexin Vþ/PIþ (C), annexin Vþ/PI (D), and annexin Vþ (E) that
respectively indicate necrotic cells, late apoptotic cells, early apoptotic cells, and apoptotic cells. N ¼ 5/each.  P < 0.05 by unpaired t-test. Error
bars represent the SEM.

related to interference of normal cell cycling by lowered Egr3 (Tierney et al., 2014). The potential role of different signaling
availability in those myoblasts. molecules warrants examination in future studies.
It was observed that a fraction of PI positive cells or late
apoptotic cells were similar between scramble and shEgr3- Possible role of NF-kB signaling
transfected cells (Fig. 4B and C), which indicates that lower
proliferation in Egr3-inhibited cells leads to slower or ceased Our results obtained using Luc-reporter demonstrated clearly
cell cycling rather than apoptotic cellular death at the time that inhibition of Egr3 compromised NF-kB activity (Fig. 3A).
of 48 h of post transfection. This idea might be supported by One important function of NF-kB is the protection of cells
the result of a low EdU incorporated cell, which might indicate from apoptotic cell death via survival signaling (Kumar et al.,
delayed entry of DNA synthesis phase, in shEgr3-transfected 2004; Hayden and Ghosh, 2008). For example, NF-kB is known
cells (Fig. 2H). Therefore, our results might show that to stimulate the expression of X-linked inhibitor of apoptosis
induction of apoptosis process was sufficient to impair protein (XIAP) and to inhibit the activity of some caspases that
myoblast proliferation in Egr3-inhibited cells. are crucially important for apoptotic signaling (Deveraux et al.,
Our results showed that Z-VAD-FMK treatment can only 1997). Therefore, it is reasonable to expect that higher
partially reverse the compromised cell number and apoptosis in Egr3-inhibited myoblast is associated with
proliferation rate of Egr3-inhibited cells (Fig. 6). This indicates decreased NF-kB activity.
that mechanisms other than apoptosis are implicated in the In addition to this, previous studies have suggested that
proliferation defect in Egr3-inhibited cells. One possible reason NF-kB activity is important for normal proliferation of
is involvement of signal transduction mechanisms related to myoblasts during myogenesis (Kumar et al., 2004; Li et al.,
Egr3 because Egr3 can interact with multiple signaling 2008). Indeed, NF-kB activity is necessary to promote cell cycle
molecules. One example is that Egr3 inhibits STAT3 activity by progression by stimulating cyclin D1 expression in myogenic
stimulating the suppression of cytokine signaling 3 (Li et al., cells (Guttridge et al., 1999). Reduced NF-kB activity in muscle-
2012; Sumitomo et al., 2013b). STAT3 has been found to inhibit derived stem cells from haplodeletion of p65 (p65þ/) mouse is
proliferation via stimulation of MyoD expression in myoblasts associated with the proliferation defect (Lu et al., 2012).

JOURNAL OF CELLULAR PHYSIOLOGY


1120 K U R O S A K A E T A L.

Fig. 5. Activation of apoptosis-related proteins in Egr3-inhibited myoblast. (A) Representative bands for full-length and cleaved PARP,
cleaved caspase 3, cleaved caspase 7, and GAPDH in shEgr3-transfected and scramble-transfected cells. Scr and sh, respectively, denote
scramble and shEgr3. (B) Relative protein levels of cleaved PARP. N ¼ 4/each.  P < 0.05 by unpaired t-test. Error bars represent the SEM. (C)
Relative protein levels of cleaved caspase 3. N ¼ 4/each.  P < 0.05 by un-paired t-test. Error bars represent the SEM. (D) Relative protein levels
of cleaved caspase 7. N ¼ 4/each.  P < 0.05 by unpaired t-test. Error bars represent the SEM.

Therefore, these notions suggest that reduction of NF-kB can protein can protect p65 from proteasome degradation by
influence different levels of cellular function, which might suppressing E3 activity of PPAR-g. Upregulation of p65
explain the limited effect of Z-VAD-FMK on Egr3-inhibited mRNA levels at 24 h of the transfection in Egr3-inhibited
myoblast. myoblast (Fig. 3D) might be a compensation mechanism for
Our results showed that the protein level of p65 was the low level of p65 protein.
decreased in Egr3-inhibited cells to a greater extent at 48 h
post transfection (Fig. 3B and C). p65 is a necessary protein that
assembles NF-kB heterodimer with p50 protein to stimulate Interpretation of Egr3 upregulation in regenerating
canonical NF-kB signaling (Kumar et al., 2004; Hayden and skeletal muscle
Ghosh, 2008; Li et al., 2008). NF-kB activity is known to be
inhibited in muscle-derived stem cells from above p65þ/ Egr3 was upregulated by CTX-injured muscles of mice at
mouse (Lu et al., 2012). Therefore, the reduction of NF-kB least in mRNA level in this study (Fig. 1B), suggesting that
activity in Egr3-inhibited cells might be attributable to the low Egr3 is important for muscle regeneration in adult skeletal
protein level of p65 in this study. muscle. However, it is noteworthy that Egr3 is also known
In contrast to the protein level, our results showed no to be important in the function of cells other than those of
decrease in p65 mRNA levels with inhibition of Egr3 muscle cell type. Indeed, Egr3 can function in the
(Fig. 3D), which suggests that the reduction of p65 protein development of neurons (Quach et al., 2013), activation of
(Fig. 3B and C) in the present study was caused by post T-cells (Safford et al., 2005; Sumitomo et al., 2013a,b) and
translational mechanisms such as protein degradation. An vascular endothelial cells (Suehiro et al., 2010), and
earlier report described that peroxisome proliferator macrophage differentiation (Carter and Tourtellotte,
activated receptor-gamma (PPAR-g) acts as a E3-ubiquitin 2007). In this regard, previous reports have described that
ligase for p65 proteasome degradation in mouse embryonic muscle injury by CTX is neurotoxic (Harris, 2003).
fibroblast (Hou et al., 2012). Moreover, Egr1, an Egr-family Furthermore, myotoxin-induced muscle injury is known to
protein, has been found to inhibit PPAR-g protein expression be accompanied by intramuscular vessel damage (Ownby
and transcriptional activity in hepatic satellite cells (Zhou et al., 1993) and inflammation reactions (Tidball and
et al., 2009). Therefore, although the precise mechanism Villalta, 2010). Therefore, even though myogenic cells are
remains unclear, these reports lead us to infer that Egr primary components, these results of studies suggest

JOURNAL OF CELLULAR PHYSIOLOGY


REGULATION OF MYOBLAST PROLIFERATION BY Egr3 1121

Fig. 6. Caspase inhibitor partially restored proliferation in Egr3-inhibited myoblast. (A) Indirect cell number after either pan-caspase
inhibitor Z-VAD-FMK or DMSO in shEgr3-transfected and scramble-transfected cells. N ¼ 7–8/each.  P < 0.05 by one-way ANOVA with
Tukey’s multiple comparison test. Error bars represent the SEM. (B) Proliferation rate from 24 to 48 h after either Z-VAD-FMK or DMSO in
shEgr3-transfected and scramble-transfected cells. N ¼ 7–8/each.  P < 0.05 by one-way ANOVA with Tukey’s multiple comparison test. Error
bars represent the SEM.

Fig. 7. Putative role of Egr3 in myoblast proliferation. Normally, Egr3 stabilizes p65, which is necessary for basal activity of NF-kB. NF-kB
acts as a part of anti-apoptotic system leading to the preservation of myoblast proliferation (A). However, the inhibition of Egr3 results in
degradation of p65, which exaggerates NF-kB activity. In the latter case, apoptotic signaling is activated excessively (B). Pan-caspase inhibitor
only partially restores proliferation in Egr3-inhibited myoblast, indicating that mechanisms other than apoptosis exist for maintaining
myoblast proliferation by Egr3 (dashed line).

that CTX-related muscle injury and regeneration can be the physiological role of Egr3 in the function of myoblast and
associated with the activity of multiple cell types that myogenesis.
engage in restoring muscle normality. The contributions of
Egr3 in regenerating muscle cells in vivo must be confirmed
in future investigations. Authors’ Contributions
In conclusion, the results of this study constitute initial
evidence implicating Egr3 in myoblast proliferation and survival KM and YO designed and performed experiments. KM and YO
signaling. A hypothesis drawn from this study is portrayed in analyzed data. KM, YO, TF, and TA wrote the manuscript. TF
Fig. 7. An additional investigation is necessary to understand and TA supervised the study.

JOURNAL OF CELLULAR PHYSIOLOGY


1122 K U R O S A K A E T A L.

Acknowledgments Peterson CA. 2011. Effective fiber hypertrophy in satellite cell-depleted skeletal muscle.
Development 138:3657–3666.
Murphy MM, Lawson JA, Mathew SJ, Hutcheson DA, Kardon G. 2011. Satellite cells,
This work was partially supported by a grant from The Nakatomi connective tissue fibroblasts and their interactions are crucial for muscle regeneration.
Foundation to MK, JSPS KAKENHI grants (#24700706, Development 138:3625–3637.
O’Donovan KJ, Wilkens EP, Baraban JM. 1998. Sequential expression of Egr-1 and Egr-3 in
#15K01633 to MK), and by a research grant from the hippocampal granule cells following electroconvulsive stimulation. J Neurochem 70:1241–1248.
St. Marianna University School of Medicine to YO. O’Donovan KJ, Tourtellotte WG, Millbrandt J, Baraban JM. 1999. The EGR family of
transcription-regulatory factors: Progress at the interface of molecular and systems
neuroscience. Trends Neurosci 22:167–173.
Ogura Y, Mishra V, Hindi SM, Kuang S, Kumar A. 2013. Proinflammatory cytokine tumor
Literature Cited necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) suppresses satellite cell
self-renewal through inversely modulating Notch and NF-kappaB signaling pathways. J Biol
Andrechek ER, Hardy WR, Girgis-Gabardo AA, Perry RL, Butler R, Graham FL, Kahn RC, Chem 288:35159–35169.
Rudnicki MA, Muller WJ. 2002. ErbB2 is required for muscle spindle and myoblast cell Ogura Y, Hindi SM, Sato S, Xiong G, Akira S, Kumar A. 2015. TAK1 modulates satellite stem
survival. Mol Cell Biol 22:4714–4722. cell homeostasis and skeletal muscle repair. Nat Commun 6:10123.
Angione AR, Jiang C, Pan D, Wang YX, Kuang S. 2011. PPARdelta regulates satellite cell Ownby CL, Fletcher JE, Colberg TR. 1993. Cardiotoxin 1 from cobra (Naja naja atra) venom
proliferation and skeletal muscle regeneration. Skelet Muscle 1:33. causes necrosis of skeletal muscle in vivo. Toxicon 31:697–709.
Bernet JD, Doles JD, Hall JK, Kelly Tanaka K, Carter TA, Olwin BB. 2014. P38 MAPK Price FD, von Maltzahn J, Bentzinger CF, Dumont NA, Yin H, Chang NC, Wilson DH,
signaling underlies a cell-autonomous loss of stem cell self-renewal in skeletal muscle of Frenette J, Rudnicki MA. 2014. Inhibition of JAK-STAT signaling stimulates adult satellite
aged mice. Nat Med 20:265–271. cell function. Nat Med 20:1174–1181.
Brack AS, Rando TA. 2012. Tissue-specific stem cells: Lessons from the skeletal muscle Pucci B, Kasten M, Giordano A. 2000. Cell cycle and apoptosis. Neoplasia 2:291–299.
satellite cell. Cell Stem Cell 10:504–514. Quach DH, Oliveira-Fernandes M, Gruner KA, Tourtellotte WG. 2013. A sympathetic
Carter JH, Tourtellotte WG. 2007. Early growth response transcriptional regulators are neuron autonomous role for Egr3-mediated gene regulation in dendrite morphogenesis
dispensable for macrophage differentiation. J Immunol 178:3038–3047. and target tissue innervation. J Neurosci 33:4570–4583.
Cheng H, Hao S, Liu Y, Pang Y, Ma S, Dong F, Xu J, Zheng G, Li S, Yuan W, Cheng T. 2015. Safford M, Collins S, Lutz MA, Allen A, Huang CT, Kowalski J, Blackford A, Horton MR,
Leukemic marrow infiltration reveals a novel role for Egr3 as a potent inhibitor of normal Drake C, Schwartz RH, Powell JD. 2005. Egr-2 and Egr-3 are negative regulators of T cell
hematopoietic stem cell proliferation. Blood 126:1302–1313. activation. Nat Immunol 6:472–480.
Day K, Shefer G, Shearer A, Yablonka-Reuveni Z. 2010. The depletion of skeletal muscle Schwartz LM. 2008. Atrophy and programmed cell death of skeletal muscle. Cell Death
satellite cells with age is concomitant with reduced capacity of single progenitors to Differ 15:1163–1169.
produce reserve progeny. Dev Biol 340:330–343. Suehiro J, Hamakubo T, Kodama T, Aird WC, Minami T. 2010. Vascular endothelial growth
Deveraux QL, Takahashi R, Salvesen GS, Reed JC. 1997. X-linked IAP is a direct inhibitor of factor activation of endothelial cells is mediated by early growth response-3. Blood
cell-death proteases. Nature 388:300–304. 115:2520–2532.
Dumont NA, Wang YX, Rudnicki MA. 2015. Intrinsic and extrinsic mechanisms regulating
satellite cell function. Development 142:1572–1581. Sumitomo S, Fujio K, Okamura T, Morita K, Ishigaki K, Suzukawa K, Kanaya K, Kondo K,
Fry CS, Lee JD, Mula J, Kirby TJ, Jackson JR, Liu F, Yang L, Mendias CL, Dupont-Versteegden Yamasoba T, Furukawa A, Kitahara N, Shoda H, Shibuya M, Okamoto A, Yamamoto K.
EE, McCarthy JJ, Peterson CA. 2015. Inducible depletion of satellite cells in adult, 2013a. Transcription factor early growth response 3 is associated with the TGF-beta1
sedentary mice impairs muscle regenerative capacity without affecting sarcopenia. Nat expression and the regulatory activity of CD4-positive T cells in vivo. J Immunol
Med 21:76–80. 191:2351–2359.
Guttridge DC, Albanese C, Reuther JY, Pestell RG, Baldwin AS, Jr. 1999. NF-kappaB controls Sumitomo S, Fujio K, Okamura T, Yamamoto K. 2013b. Egr2 and Egr3 are the unique
cell growth and differentiation through transcriptional regulation of cyclin D1. Mol Cell regulators for systemic autoimmunity. Jak-Stat 2:e23952.
Biol 19:5785–5799. Suzanne M, Steller H. 2013. Shaping organisms with apoptosis. Cell Death Differ 20:669–675.
Harris JB. 2003. Myotoxic phospholipases A2 and the regeneration of skeletal muscles. Tidball JG, Villalta SA. 2010. Regulatory interactions between muscle and the immune
Toxicon 42:933–945. system during muscle regeneration. Am J Physiol Regul Integr Comp Physiol 298:
Hayden MS, Ghosh S. 2008. Shared principles in NF-kappaB signaling. Cell 132:344–362. R1173–R1187.
Hippenmeyer S, Shneider NA, Birchmeier C, Burden SJ, Jessell TM, Arber S. 2002. A role for Tierney MT, Aydogdu T, Sala D, Malecova B, Gatto S, Puri PL, Latella L, Sacco A. 2014.
neuregulin1 signaling in muscle spindle differentiation. Neuron 36:1035–1049. STAT3 signaling controls satellite cell expansion and skeletal muscle repair. Nat Med
Hou Y, Moreau F, Chadee K. 2012. PPARgamma is an E3 ligase that induces the degradation 20:1182–1186.
of NFkappaB/p65. Nat Commun 3:1300. Tourtellotte WG, Milbrandt J. 1998. Sensory ataxia and muscle spindle agenesis in mice
Jacobson C, Duggan D, Fischbach G. 2004. Neuregulin induces the expression of lacking the transcription factor Egr3. Nat Genet 20:87–91.
transcription factors and myosin heavy chains typical of muscle spindles in cultured human Tourtellotte WG, Keller-Peck C, Milbrandt J, Kucera J. 2001. The transcription factor Egr3
muscle. Proceedings of the National Academy of Sciences of the United States of America modulates sensory axon-myotube interactions during muscle spindle morphogenesis.
101:12218–12223. Dev Biol 232:388–399.
Kumar A, Takada Y, Boriek AM, Aggarwal BB. 2004. Nuclear factor-kappaB: Its role in health Wang YX, Dumont NA, Rudnicki MA. 2014. Muscle stem cells at a glance. J Cell Sci
and disease. J Mol Med (Berl) 82:434–448. 127:4543–4548.
Kurosaka M, Machida S. 2013. Interleukin-6-induced satellite cell proliferation is regulated by Wieland GD, Nehmann N, Muller D, Eibel H, Siebenlist U, Suhnel J, Zipfel PF, Skerka C. 2005.
induction of the JAK2/STAT3 signalling pathway through cyclin D1 targeting. Cell Prolif Early growth response proteins EGR-4 and EGR-3 interact with immune inflammatory
46:365–373. mediators NF-kappaB p50 and p65. J Cell Sci 118:3203–3212.
Lacraz G, Rouleau AJ, Couture V, Sollrald T, Drouin G, Veillette N, Grandbois M, Grenier G. Xi H, Schwartz R, Engel I, Murre C, Kersh GJ. 2006. Interplay between RORgammat, Egr3,
2015. Increased stiffness in aged skeletal muscle impairs muscle progenitor cell and E proteins controls proliferation in response to pre-TCR signals. Immunity
proliferative activity. PLoS ONE 10:e0136217. 24:813–826.
Lehtinen SK, Rahkila P, Helenius M, Korhonen P, Salminen A. 1996. Down-regulation of Yin H, Price F, Rudnicki MA. 2013. Satellite cells and the muscle stem cell niche. Physiol Rev
transcription factors AP-1, Sp-1, and NF-kappa B precedes myocyte differentiation. 93:23–67.
Biochem Biophys Res Commun 229:36–43. Yoshida T, Galvez S, Tiwari S, Rezk BM, Semprun-Prieto L, Higashi Y, Sukhanov S, Yablonka-
Lepper C, Partridge TA, Fan CM. 2011. An absolute requirement for Pax7-positive satellite Reuveni Z, Delafontaine P. 2013. Angiotensin II inhibits satellite cell proliferation and
cells in acute injury-induced skeletal muscle regeneration. Development 138:3639–3646. prevents skeletal muscle regeneration. J Biol Chem 288:23823–23832.
Li H, Malhotra S, Kumar A. 2008. Nuclear factor-kappa B signaling in skeletal muscle atrophy.
J Mol Med (Berl) 86:1113–1126. Zhou Y, Jia X, Zhou M, Liu J. 2009. Egr-1 is involved in the inhibitory effect of leptin on
Li S, Miao T, Sebastian M, Bhullar P, Ghaffari E, Liu M, Symonds AL, Wang P. 2012. The PPARgamma expression in hepatic stellate cell in vitro. Life Sci 84:544–551.
transcription factors Egr2 and Egr3 are essential for the control of inflammation and
antigen-induced proliferation of B and T cells. Immunity 37:685–696.
Lu A, Proto JD, Guo L, Tang Y, Lavasani M, Tilstra JS, Niedernhofer LJ, Wang B, Guttridge Supporting Information
DC, Robbins PD, Huard J. 2012. NF-kappaB negatively impacts the myogenic potential of
muscle-derived stem cells. Mol Ther 20:661–668.
McCarthy JJ, Mula J, Miyazaki M, Erfani R, Garrison K, Farooqui AB, Srikuea R, Lawson BA, Additional supporting information may be found in the online
Grimes B, Keller C, Van Zant G, Campbell KS, Esser KA, Dupont-Versteegden EE, version of this article at the publisher’s web-site.

JOURNAL OF CELLULAR PHYSIOLOGY

You might also like