You are on page 1of 7

Gene 676 (2018) 9–15

Contents lists available at ScienceDirect

Gene
journal homepage: www.elsevier.com/locate/gene

Research paper

Osteoglycin post-transcriptional regulation by miR-155 induces cellular T


architecture changes in H9c2 cardiomyoblasts
Grasieli de Oliveiraa, Paula Paccielli Freirea, Ana Carolina Mieko Omotoa, Sarah Santiloni Curya,

Cesar Seigi Fuziwarab, Edna Teruko Kimurab, Maeli Dal-Pai-Silvaa, Robson Francisco Carvalhoa,
a
Department of Morphology, Institute of Biosciences of Botucatu, São Paulo State University, Botucatu, São Paulo, Brazil
b
Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil

A R T I C LE I N FO A B S T R A C T

Keywords: Several studies have demonstrated dysregulated cardiac microRNAs (miRNAs) following cardiac stress and
Mimecan development of cardiac hypertrophy and failure. miRNAs are also differentially expressed in the inflammation
Cardiac cell morphology that occurs in heart failure and, among these inflammatory-related miRNAs, the miR-155 has been implicated in
Cardiac pathological hypertrophy the regulation of cardiac hypertrophy. Despite these data showing the role of miRNA-155 in cardiomyocyte
hypertrophy under a hypertrophic stimulus, it is also important to understand the endogenous regulation of this
miRNA without a hypertrophic stimulus to fully appreciate its function in this cell type. The first aim of the
present study was to determine whether, without a hypertrophic stimulus, miR-155 overexpression induces
H9c2 cardiac cells hypertrophy in vitro. The second objective was to determine whether osteoglycin (Ogn), a key
regulator of heart mass in rats, mice, and humans, is post-transcriptionally regulated by miR-155 with a potential
role in inducing H9c2 cells hypertrophy. Here, we show that, without a hypertrophic stimulus, miR-155 sig-
nificantly repressed Ogn protein levels, but induce neither alteration in morphological phenotype nor in the
expression of the molecular markers that fully characterize pathological hypertrophy of H9c2 cells. However,
most importantly, Ogn silencing in H9c2 cells mimicked the effects of miR-155 overexpression in inducing
cellular architecture changes that were characterized by a transition of the cell shape from fusiform to rounded.
This is a new role of the post-transcriptional regulation of Ogn by miR-155 in the maintenance of the cardiac cell
morphology in physiological and pathological conditions.

1. Introduction nucleotides that regulate gene expression in higher eukaryotes by


binding to complementary sequences mainly within the 3′ untranslated
Cardiac remodeling involves a repertoire of pathophysiological re- region (3′UTR) of protein-coding mRNAs [reviewed in (Bartel, 2004;
sponses including molecular, cellular, and extracellular matrix changes Carrington and Ambros, 2003)].
that, in turn, promote alterations in size, shape, and function of the Several studies have demonstrated dysregulated cardiac miRNAs in
heart after injury or stress stimulation [reviewed in (Kehat and adaptive and maladaptive hypertrophy and heart failure (HF) in re-
Molkentin, 2010)]. The identification of microRNAs (miRNAs) during sponse to cardiac stress (Bagnall et al., 2012; Sucharov et al., 2008; van
cardiac remodeling has opened up new fields of investigation aiming to Rooij et al., 2008, 2006). miRNAs are also differentially expressed in
better understand gene expression regulation in cardiovascular diseases the inflammation that occurs in HF and, among these inflammatory-
[reviewed in (Liu and Olson, 2010; Small and Olson, 2011)]. miRNAs related miRNAs, the miR-155 has been implicated in the regulation of
are an evolutionarily conserved class of small noncoding RNAs of ~22 cardiac hypertrophy under a hypertrophic stimulus. Seok et al. (2014)

Abbreviations: miRNAs, microRNAs; Ogn, osteoglycin/mimecan; 3′UTR, 3′ untranslated region; HF, heart failure; Socs1, suppressor of cytokine signaling 1; FoxO3a,
Forkhead box O3a; ECM, extracellular matrix; GM, growth medium; RIN, RNA integrity number; PBS, Phosphate Buffered Saline; PMSF, phenylmethylsulfonyl
fluoride, protease inhibitor; RIPA, Radio-Immunoprecipitation Assay; SDS-PAGE, sodium dodecyl sulfate polyacrylamide gel electrophoresis; TBS, Tris buffered
saline; Gapdh, Glyceraldehyde 3-phosphate dehydrogenase; BSA, Bovine serum albumin; DAPI, (4′,6-diamidino-2-phenylindole); ANP, Atrial Natriuretic Peptide;
BNP, Brain Natriuretic Peptide; Myh6, Myosin, Heavy Chain 6, Cardiac Muscle, Alpha; Myh7, Myosin, Heavy Chain 7, Cardiac Muscle, Beta; MYH, Myosin, Heavy
Chains Isoforms; Acta1, Actin, Alpha 1, Skeletal Muscle; Ppib, Peptidylprolyl isomerase B

Corresponding author.
E-mail address: robson.carvalho@unesp.br (R.F. Carvalho).

https://doi.org/10.1016/j.gene.2018.07.020
Received 12 April 2018; Received in revised form 1 July 2018; Accepted 6 July 2018
Available online 07 July 2018
0378-1119/ © 2018 Elsevier B.V. All rights reserved.
G. de Oliveira et al. Gene 676 (2018) 9–15

uncover the miR-155 as an inducer of pathological cardiomyocyte hy- 2.2. Oligonucleotides and transfection
pertrophy by targeting Jarid2/jumonji, which is also known to critically
regulate the cardiovascular development (Seok et al., 2014); (Mysliwiec The miR-155 mimic (mirVana™ miRNA Mimic, assay ID: MC13058;
et al., 2011). The miR-155 also targets the suppressor of cytokine sig- Thermo Fisher, USA), and the small interfering RNA (Silencer® Select
naling 1 (Socs1) transcript in cardiac macrophages and promotes the siRNA, assays ID: s70945 and s70947; Thermo Fisher Scientific, USA),
inflammatory response to pressure overload, where genetic loss or against Ogn transcript, and the respective negative controls (miRNA
pharmacological inhibition of miR-155 protected against cardiac hy- Mimic Negative Control and Silencer® Select Negative Control No. 1
pertrophy and HF (Heymans et al., 2013). Furthermore, the polyphenol siRNA; Thermo Fisher Scientific, USA), were complexed with Opti-MEM
resveratrol, which has been reported to exhibit cardioprotective effects, reduced serum medium (Thermo Fisher Scientific, USA) before trans-
produced protective effects against cardiac hypertrophy by reducing fection. After the H9c2 cardiomyoblasts cultures have reached ap-
miR-155 levels (Fan et al., 2016). These authors also showed that proximately 80% confluence, the cells were transfected with RNAiMAX
Forkhead box O3a (FoxO3a) is a miR-155 target transcript involved in lipofectamine (Thermo Fisher Scientific, USA) mixed with 30 nmol of
the anti-hypertrophic effect of resveratrol in hypertrophied cardio- each oligonucleotide for 12 h. Oligonucleotide concentration and in-
myocytes. cubation time were previously standardized in our laboratory in muscle
Despite these data demonstrating the role of miRNA-155 in cardi- cells. After transfection, the medium was changed to GM, and the cells
omyocyte hypertrophy under a hypertrophic stimulus, it is also im- were analyzed after 48 h (Supplementary Fig. 1).
portant to understand the endogenous regulation of this miRNA
without a hypertrophic stimulus to fully appreciate its function in this 2.3. RNA isolation
cell type. Thus, the first aim of the present study was to determine
whether, without a hypertrophic stimulus, miR-155 overexpression H9c2 cardiomyoblasts total RNA was extracted using the TRIzol
induces H9c2 cardiac cell hypertrophy in vitro. Moreover, an important reagent (Thermo Fisher Scientific, USA), as suggested by the manu-
issue to address in the miRNA field is that a single miRNA can regulate facture. NanoVue Plus Spectrophotometer (GE Healthcare, USA) was
multiple mRNAs (Pasquinelli, 2012), challenging the identification of used to quantify RNA concentration and quality; subsequently, the ri-
new miR-155-target transcripts that may be critical for understanding bosomal RNAs were analyzed using the 2100 Bioanalyzer system
the molecular basis of cardiomyocyte biology. Recently, we demon- (Agilent, USA) to obtain the RNA integrity number (RIN) and RNA
strated that miR-155 inhibits osteoglycin/mimecan (Ogn) expression to quality. RNA preparations with A260/280 ratio of 1.8–2.0, A260/230
control myogenic differentiation of C2C12 myoblasts (Freire et al., ratio > 2.0, and RIN > 9 were used for further analysis.
2017). Interestingly, Ogn is a secreted extracellular matrix (ECM)
component that has been considered a key regulator of heart mass in 2.4. RT-qPCR
rats, mice, and humans, and a novel biomarker to predict chronic HF
outcomes and left ventricular remodeling (Motiwala et al., 2014; Total RNA samples (1 μg) were reverse transcribed into cDNA with
Petretto et al., 2008; Van Aelst et al., 2015). The second objective of the the High Capacity RNA-to-cDNA Master Mix (Thermo Fisher Scientific,
study was to determine whether the post-transcriptional regulation of USA). The qPCR reactions (15 μL) were performed with Power SYBR™
osteoglycin by miR-155 may induce H9c2 cell hypertrophy in vitro. Green Master Mix (Thermo Fisher Scientific, USA), as described by the
Considering that reactivation/upregulation of the fetal cardiomyocyte manufacturer, using specific primers (listed in Supplementary Table 1).
gene program, including the natriuretic peptide A (ANP), natriuretic The reactions were run on a QuantStudio™ 12 K Flex System (Thermo
peptide B (BNP), skeletal muscle alpha actin (Acta1), and myosin heavy Fisher Scientific, USA) using 95 °C for 10 min followed by 40 cycles of
chain beta (β-MHC or Myh7), and the downregulation of genes asso- 95 °C for 15 s and 60 °C for 1 min as cycling conditions. Finally, the raw
ciated with contractile function, such as myosin heavy chain beta (α- data was retrieved and imported into the Expression Suite Software
MHC or Myh6), are molecular hallmarks of the pathological cardiac v1.0.3 (Thermo Fisher Scientific, USA). Relative quantification of
hypertrophy (Chien et al., 1991; Depre et al., 1998; Izumo et al., 1988; mRNA expression was evaluated using the 2−ΔΔCTmethod (Livak and
Rajabi et al., 2007), we evaluated the expression of these genes as Schmittgen, 2001), and Ppib was used as reference gene to normalize
markers of H9c2 cells hypertrophy. We found that, without a hyper- the mRNA expression data. This reference gene was selected based on
trophic stimulus, miR-155 significantly repressed Ogn protein levels, geNorm calculations (Vandesompele et al., 2002), and the expression
but induce neither alteration in morphological phenotype nor in the stability was further confirmed by the comparisons of Ct values be-
expression of the molecular markers that fully characterize pathological tween control and treated samples (miR-155 mimic or siRNA-Ogn)
hypertrophy of H9c2 cells. However, most importantly, Ogn silencing samples (p > 0.80, Student t-test). Cutoffs for significant changes were
in H9c2 cells mimicked the effects of miR-155 overexpression in in- set at fold-change > 1.5 and p-value ≤ 0.05.
ducing cellular architecture changes that were characterized by a
transition of the cell shape from fusiform to rounded. This is a new role 2.5. Western blot analysis
of the post-transcriptional regulation of Ogn by miR-155 in the main-
tenance of the cardiac cell morphology in physiological and patholo- After transfections, the H9c2 cardiomyoblasts were harvested in
gical conditions. PBS (Phosphate Buffered Saline), mechanically lysated, and the cell
pellets were re-suspended and incubated in ice-cold RIPA buffer (Radio-
Immunoprecipitation Assay, Sigma, USA) containing 1% PMSF (pro-
2. Materials and methods tease inhibitor), on ice for 30 min. The lysated cells were centrifuged in
8.000 RCF for 10 min at 4 °C to collect the supernatant, and protein
2.1. Cell culture concentration was determined by Bradford protein assay kit according
to the manufacturer's instructions (Bradford3, Bio-Rad Laboratories,
H9c2 (2-1) rat BDIX cardiomyoblasts (ATCC, The Global USA). Subsequently, the samples were prepared using Lammeli buffer
Bioresource Center, USA) were cultured at 37 °C and 5% CO2 in (Sigma, USA) at 100 °C for 10 min, for SDS-PAGE protocol.
Dulbecco's modified Eagle's medium (DMEM, Thermo Fisher Scientific, Electrophoresis were performed in 10% polyacrylamide gels, and after
USA) growth medium (GM) enriched with 1% penicillin-streptomycin protein separation, proteins were electrotransferred to nitrocellulose
(Thermo Fisher Scientific, USA) and 10% fetal bovine serum (Thermo membranes (Bio-Rad, USA). Membranes were blocked for 2 h at room
Fisher Scientific, USA). All experiments were carried out using at least temperature, using 5% nonfat dry milk in TBS buffer containing 0.5%
three independent replicates per group. Tween 20 (TBST), and then incubated with specific antibodies against

10
G. de Oliveira et al. Gene 676 (2018) 9–15

Osteoglycin (Ogn, Santa Cruz Biotechnology, USA; 1:1000 dilution),


Myosin Heavy Chain (MYH, Santa Cruz Biotechnology, USA; 1:500
dilution), and Glyceraldehyde 3-phosphate dehydrogenase (Gapdh,
ABR Affinity BioReagents, USA; 1:2500 dilution), overnight at 4–8 °C.
Finally, secondary antibodies (rabbit or mouse, depending on the pro-
tein) were incubated for 2 h at room temperature to promote the
binding with primary antibody, and secondary antibody peroxidase
conjugated was detected using Enhanced Chemiluminescence
(Amersham Biosciences, USA), and autoradiography in ImageQuant™
LAS 4000 (GE Healthcare, USA). Proteins quantification of blots were
performed by ImageJ software (Version 1.49o, Wayne Rasband, NIH).
Targeted bands were normalized to the expression of GAPDH.

2.6. Immunofluorescence analysis

Firstly, 6-well plates were used to seed the H9c2 cardiomyoblasts


for immunoassay. After transfections, these cells were fixed in 4%
paraformaldehyde for 15 min at room temperature, and washed with
PBS and 0.1% TritonX-100 (Sigma, USA). The block was performed
during 1 h at room temperature using 3% BSA, 1% glycine, 8% fetal
bovine serum in PBS and 0.1% TritonX-100. After the block, the cells
were incubated overnight at 4°Cwith Ogn primary antibody, and sub-
sequently, with secondary antibody for 1 h, at room temperature.
Nucleus staining was performed with DAPI (4′,6-diamidino-2-pheny-
lindole) counter staining. All the images were acquired at room tem-
perature with a confocal microscope TCS SP5 (Leica Microsystems, UK). Fig. 1. miR-155 post transcriptionally represses Ogn protein expression in H9c2
Cellular width and length were measured by ImageJ software (Version cardiomyoblasts. RT-qPCR analysis show that miR-155 does not change Ogn
1.49o, Wayne Rasband, NIH). transcript levels (A); however, miR-155 represses Ogn protein expression, as
revealed by Western blot analysis (B). Western blot and RT-qPCR analysis
2.7. Statistical analysis confirm the efficiency of siRNA-Ogn on Ogn transcript (C) and protein (D) le-
vels in H9c2 cardiomyoblasts, respectively. RT-qPCR data were normalized by
the expression level of the reference Ppib gene, and western blot data were
All experimental data are expressed as the mean ± standard de-
normalized by Gapdh content. Results are expressed as mean ± SD of three
viation (SD). Data were analyzed using Student's t-test to establish
independent biological replicates (n = 3). Statistical difference was analyzed by
significance between the groups. p < 0.05 were considered statisti- Student's t-test. *p < 0.05 vs. Control (CT).
cally significant.

3. Results polyclonal antibody (MYH), reported to detect all muscle myosin heavy
chain isoforms as a molecular marker of protein synthesis or degrada-
3.1. miR-155 post transcriptionally represses Ogn protein expression in tion. However, the miR-155 mimetic treatment did not change MYH
H9c2 cardiomyoblasts protein levels when compared to control group (Fig. 2B).

We had previously identified and validated Ogn transcript 3.3. Ogn alters the expression levels of ANP in H9c2 cardiomyoblasts
(NM_008760.4) as a miR-155 target on its 3′UTR in fibroblasts and
myoblasts (Freire et al., 2017).To confirm the effect of miR-155 in To verify whether Ogn knockdown induce a similar pattern in the
regulating the expression of Ogn in cardiomyoblasts, we first trans- expression of cardiac stress markers as those found for the miR-155
fected H9c2 cells with miR-155 mimic and evaluated Ogn mRNA and mimic transfected cells, the transcript levels of ANP, BNP, Acta1, β-
protein expression levels. The miR-155 transfection did not affect Ogn MHC/Myh7, and α-MHC/Myh6 genes were measured in the H9c2
transcript levels (Fig. 1A); however, resulted in a reduction on protein cardiomyoblasts by RT-qPCR (Fig. 3). Ogn knockdown only increased
levels (Fig. 1B). Finally, to simulate a potential miR-155 post tran- ANP mRNA expression levels in H9c2 cardiomyoblasts when compared
scriptional regulation of Ogn in H9c2 cardiomyoblasts, we also to the control group (Fig. 3A). We also analyzed the MYH protein ex-
knocked-down Ogn with siRNA oligonucleotides and evaluated Ogn pression by western-blot in H9c2 cells treated with siRNAs for Ogn.
mRNA and protein expression levels. This Ogn knockdown induced a However, this treatment also did not alter the MYH protein levels
significant reduction in Ogn mRNA and protein expression levels in (Fig. 3B).
H9c2 cells when compared to control group (Fig. 1C and D).
3.4. miR-155 and Ogn induces cellular architecture changes in H9c2
3.2. miR-155 alters the expression of cardiac stress markers in H9c2 cardiomyoblasts
cardiomyoblasts
Although the changes in expression profile of the molecular markers
Deregulation of molecular hallmarks of the pathological cardiac of pathological hypertrophy were different among H9c2 cardiomyo-
hypertrophy (upregulation of the fetal cardiomyocyte gene program blasts treated with miR-155 mimic or siRNAs for Ogn, we clearly noted
ANP, BNP, Acta1, and Myh7; and the downregulation of Myh6) was that both treatments had a similar pattern of cardiomyoblasts pheno-
measured in the H9c2 cardiomyoblasts transfected with miR-155 by typic changes with the cells acquiring a rounded shape (Fig. 4A and B).
RT-qPCR (Fig. 2A). The miR-155 transfection did not alter the expres- These phenotypic changes in the cellular architecture was further
sion levels of the ANP and Myh6 transcripts (Fig. 2A); however, the confirmed by a morphometric analysis that showed a decreased cellular
Myh7and BNP levels were decreased, and the Acta1 levels were in- length of the H9c2 cardiomyoblasts transfected with either miR-155 or
creased when compared to the control group (Fig. 2A). We also used a siRNA-Ogn when compared to the respectively control groups (Fig. 4C

11
G. de Oliveira et al. Gene 676 (2018) 9–15

Fig. 2. miR-155 alters the expression of cardiac stress markers in H9c2 cardi-
omyoblasts. Relative expression levels of mRNAs were measured by RT-qPCR Fig. 3. Ogn alters the expression levels of ANP in H9c2 cardiomyoblasts.
analysis in H9c2 cardiomyoblasts transfected with miR-155 or control (CT) (A). Relative expression levels of mRNAs were measured by RT-qPCR analysis in
MYH protein expression levels were measured by Western blot analysis (B). RT- H9c2 cardiomyoblasts transfected with miR-155 or control (CT) (A). MYH
qPCR data were normalized by the expression level of the reference Ppib gene, protein expression levels were measured by Western blot analysis (B). RT-qPCR
and western blot data were normalized by Gapdh content. Results are expressed data were normalized by the expression level of the reference Ppib gene, and
as mean ± SD of three independent biological replicates (n = 3). Statistical western blot data were normalized by Gapdh content. Results are expressed as
difference was analyzed by Student's t-test. *p < 0.05 vs. Control (CT). ANP: mean ± SD of three independent biological replicates (n = 3). Statistical dif-
Atrial Natriuretic Peptide; BNP: Brain Natriuretic Peptide; Myh6: Myosin, ference was analyzed by Student's t-test. *p < 0.05 vs. Control (CT). ANP:
Heavy Chain 6, Cardiac Muscle, Alpha; Myh7: Myosin, Heavy Chain 7, Cardiac Atrial Natriuretic Peptide; BNP: Brain Natriuretic Peptide; Myh6: Myosin,
Muscle, Beta; MYH: Myosin, Heavy Chains Isoforms; Acta1: Actin, Alpha 1, Heavy Chain 6, Cardiac Muscle, Alpha; Myh7: Myosin, Heavy Chain 7, Cardiac
Skeletal Muscle; Ppib: Peptidylprolyl isomerase B; Gapdh: glyceraldehyde-3- Muscle, Beta; MYH: Myosin, Heavy Chain Isoforms; Acta1: Actin, Alpha 1,
phosphate dehydrogenase. Skeletal Muscle; Ppib: Peptidylprolyl isomerase B; Gapdh: glyceraldehyde-3-
phosphate dehydrogenase.

and D). Interestingly, treatment with siRNA for Ogn increases the width
of H9c2 cells, while treatment with miR-155 did not alter the width miR-155 overexpression decreased BNP and Myh7, increased skeletal
(Fig. 4C and D). α-actin, and induced no changes in the expression levels of ANP, and
Myh6. Despite these changes in the cardiomyocyte molecular markers
expression induced by miR-155 do not characterize cardiac hyper-
4. Discussion trophy, we clearly noted a change in the cell morphology, characterized
by a transition of the cell shape from fusiform to rounded.
In the present study, we identified that, without a hypertrophic Although we had not found studies showing that, without a hy-
stimulus, miR-155 significantly repressed Ogn protein levels, but in- pertrophic stimulus, in vitro miR-155 overexpression direct induces
duce neither alteration in morphological phenotype nor in the expres- cardiomyocyte hypertrophy, the role of miR-155 in signaling pathways
sion of the molecular markers that fully characterize pathological hy- and cellular responses triggered by hypertrophic stimuli is very de-
pertrophy of H9c2 cardiac cells. However, most importantly, Ogn bated. Previous study found that the mRNA expression of ANP, BNP,
silencing in cardiomyoblast mimicked the effects of miR-155 over- and Myh7 was markedly increased in cardiomyocytes from neonatal
expression in inducing cellular architecture changes that were char- mice hypertrophied with isoprenaline and transfected with miR-155
acterized by a transition of the cell shape from fusiform to rounded (Fan et al., 2016). Furthermore, cardiomyocyte hypertrophy induced by
(results are summarized in Supplementary Table 2). pressure overload or by calcineurin transgene was reduced in miR-155-
It was unclear whether changes in miR-155 expression direct induce knockout mice hearts (Seok et al., 2014). These authors also showed
cardiac hypertrophy or constitute a response to a diverse stimuli that that the hypertrophy growth induced in vitro by phenylephrine is sup-
are related to the onset of cardiac hypertrophy (Fan et al., 2016; pressed in cardiomyocytes isolated from neonatal miR-155-knockout
Heymans et al., 2013; Seok et al., 2014). Therefore, we selected a mice or in neonatal rat cardiomyocytes with endogenous inhibition of
controlled cell culture system to transfect H9c2 cardiomyoblasts with miR-155. This diverges from (Heymans et al., 2013), who showed that
miR-155, without a hypertrophic stimulus, and analyzed the gene ex- cardiomyocyte-specific miR-155 overexpression and knockdown do not
pression levels of the pathological cardiac hypertrophy markers. This

12
G. de Oliveira et al. Gene 676 (2018) 9–15

Fig. 4. miR-155 and Ogn induces cellular phenotypic changes


in H9c2 cardiomyoblasts. Ogn immunostaining (green) in
H9c2 cells treated with miR-155 (A) or siRNA-Ogn (B) and
respective controls; Nuclei were stained in blue by DAPI.
Cellular architecture was evaluated by morphometric analysis
(cellular length and width) of the H9c2 cardiomyoblasts
treated with miR-155 (C) or siRNA-Ogn (D) and respective
controls. Results are expressed as mean ± SD of three in-
dependent biological replicates (n = 3). Statistical difference
was analyzed by Student's t-test. *p < 0.05 vs. Control (CT).
Scales bars 50 μm. Ogn: Osteoglycin; DAPI: 4′,6-Diamidino-2-
phenylindole dihydrochloride. (For interpretation of the re-
ferences to color in this figure legend, the reader is referred to
the web version of this article.)

affect heart failure susceptibility. These authors further confirmed in overexpressing miR-155 remains to be determined; however, an Ogn/
vitro that miR-155 knockdown in cardiomyocytes does not affect their miR-155 pathway regulating ECM organization may have been in-
response to hypertrophic signaling. The disparities in the role of miR- volved. Beyond the involvement of miRNAs in cardiomyocytes re-
155 in cardiomyocytes hypertrophy are clearly associated with dif- modeling, these small non-coding RNAs, including miR-155, control
ferent “in vivo” and “in vitro” hypertrophic stimuli, which induce dis- extracellular matrix (ECM) production in the heart through direct ef-
tinct phenotypes in cardiomyocytes (Schaub et al., 1997). fects on fibroblasts and indirectly by interfering with cardiomyocyte
Moreover, each of these miR-155 studies described above validated paracrine biology (Duisters et al., 2009; He et al., 2015; Orenes-Piñero
a new target transcript for this miRNA, which includes Jarid2/jumonji, et al., 2013; van Rooij and Olson, 2009). Interestingly, Ogn has been
Socs1, and FoxO3a; all coding proteins that participate in the cardio- causally linked to cardiac integrity and function by maintaining a well-
myocyte hypertrophy process (Fan et al., 2016; Heymans et al., 2013; structured ECM; the absence of osteoglycin leads to an improper col-
Seok et al., 2014). These studies demonstrate the importance of the lagen assembly with consequent increased infarct rupture and wall
miRNA miR-155 in this process and challenge the fully comprehension thinning after myocardium infarction without affecting cardiac in-
of the effects of this miRNA in the hypertrophy phenotype at the flammation or cardiomyocyte hypertrophy (Van Aelst et al., 2015).
transcript level. Considering that we have recently demonstrated that Moreover, in vitro cardiomyocytes respond to patterns in ECM compo-
miR-155 inhibits Osteoglycin (Ogn) expression to regulate proliferation nents by detecting and transducing phenotypic information stored
and differentiation of C2C12 myoblast cells (Freire et al., 2017), we also within the tertiary structure of the surrounding matrix (Simpson et al.,
tested whether this new miR-155-Ogn has a role in cardiomyocyte 1994). Previous in vitro analyses also showed that Ogn knockdown in-
hypertrophy. Interestingly, Ogn is a secreted extracellular matrix (ECM) duces cellular senescence and promotes migratory activity of cardiac
component which belongs to a small leucine-rich proteoglycan family fibroblasts; these Ogn-mediated effects are mimicked by the miR-22, a
and first described to be implicated in bone formation (Bentz et al., miRNA that also targets the Ogn transcript in cardiac fibroblast
1990, 1989; Madisen et al., 1990). Ogn has also been involved in co- (Jazbutyte et al., 2013). Thus, our data demonstrate the importance of
chlear development (Williamson et al., 2008), pituitary tumor biology an Ogn/miR-155 pathway that may regulate a well-structured ECM to
(Hu et al., 2005), skin collagen fibrin abnormalities (Tasheva et al., maintain both the cellular architecture and functional integrity of the
2002), and corneal transparency (Scott and Haigh, 1988; Tasheva et al., cardiomyoblasts.
2004). Most importantly, Ogn has been considered a key regulator of The study design described here has limitations that should be
heart mass in rats, mice, and humans, and a novel biomarker to predict considered. Specifically, the biological implications of one single
chronic heart failure outcomes and left ventricular remodeling miRNA and its impact on signaling pathways cannot be determined
(Motiwala et al., 2014; Petretto et al., 2008; Van Aelst et al., 2015). with confidence using only the tools described herein. Although the
Noteworthy, Ogn silencing in cardiomyoblast mimicked the effects functionality of one single miRNAs is measurable, the regulation of
of miR-155 overexpression in the cell morphology; in both conditions, targets by miRNAs is subject to different levels of control, and recent
the cells presented similar cellular morphological changes, character- advances have presented that targets can also reciprocally control the
ized by a transition of the cell shape from fusiform to rounded. We also level and function of miRNAs (Pasquinelli, 2012). Thus, whether this
noted that cardiomyoblasts affected by RNAi-induced knockdown of miR-155-induced morphological changes reflects only a direct effect on
Ogn, except for the upregulation of ANP gene, did not change the ex- Ogn, or whether this is related to a direct or indirect modulation of
pression of the molecular markers of pathological hypertrophy. These in others transcriptional and posttranslational factors cannot be dis-
vitro results are in accordance with those observed in Ogn−/− knockout tinguished.
mice, which had no difference in left ventricular mass without per-
missive physiological perturbations required to reveal cardiovascular
5. Conclusion
phenotypes (Petretto et al., 2008). These authors also observed that
Ogn+/− and Ogn−/− mice show a significant attenuation in left ven-
In conclusion, we found that in vitro miR-155 overexpression does
tricular mass as compared to Ogn+/+ controls after subcutaneous an-
not direct induce H9c2 cardiomyoblasts hypertrophy but rather induces
giotensin II infusion as a pro-hypertrophic stimulus.
cellular architecture changes by the repression of Ogn. These results
What caused the similar pattern of the cell shape transition from a
show that the new post-transcriptional regulation of Ogn by miR-155
fusiform to rounded in cardiomyoblasts with silenced Ogn or
has a potential role in the maintenance of the cardiac cell morphology

13
G. de Oliveira et al. Gene 676 (2018) 9–15

in physiological and pathological conditions. Mayr, M., de Windt, L.J., Lutgens, E., Wouters, K., de Winther, M.P.J., Zacchigna, S.,
Supplementary data to this article can be found online at https:// Giacca, M., van Bilsen, M., Papageorgiou, A.-P., Schroen, B., 2013. Macrophage
microRNA-155 promotes cardiac hypertrophy and failure. Circulation 128,
doi.org/10.1016/j.gene.2018.07.020. 1420–1432. https://doi.org/10.1161/CIRCULATIONAHA.112.001357.
Hu, S.-M., Li, F., Yu, H.-M., Li, R.-Y., Ma, Q.-Y., Ye, T.-J., Lu, Z.-Y., Chen, J.-L., Song, H.-
Conflict of interest D., 2005. The mimecan gene expressed in human pituitary and regulated by pituitary
transcription factor-1 as a marker for diagnosing pituitary tumors. J. Clin.
Endocrinol. Metab. 90, 6657–6664. https://doi.org/10.1210/jc.2005-0322.
The authors declare that there are no conflicts of interest. Izumo, S., Nadal-Ginard, B., Mahdavi, V., 1988. Protooncogene induction and repro-
gramming of cardiac gene expression produced by pressure overload. Proc. Natl.
Acad. Sci. U. S. A. 85, 339–343.
Competing interests Jazbutyte, V., Fiedler, J., Kneitz, S., Galuppo, P., Just, A., Holzmann, A., Bauersachs, J.,
Thum, T., 2013. MicroRNA-22 increases senescence and activates cardiac fibroblasts
The authors state that they have no conflicts of interest. in the aging heart. Age (Omaha) 35, 747–762. https://doi.org/10.1007/s11357-012-
9407-9.
Kehat, I., Molkentin, J.D., 2010. Molecular pathways underlying cardiac remodeling
Author contributions during pathophysiological stimulation. Circulation 122, 2727–2735. https://doi.org/
10.1161/CIRCULATIONAHA.110.942268.
RFC, PPF and GO conceived and designed the project, with input Liu, N., Olson, E.N., 2010. MicroRNA regulatory networks in cardiovascular development.
Dev. Cell 18, 510–525. https://doi.org/10.1016/j.devcel.2010.03.010.
from MDPS, CSF, and ETK. GO conducted the cell culture experiments. Livak, K.J., Schmittgen, T.D., 2001. Analysis of relative gene expression data using real-
GO, PPF and SSC carried out immunofluorescence and morphometric time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods 25, 402–408.
analyses. PPF, SSC, ACMO, GO, and RFC performed RT-qPCR experi- https://doi.org/10.1006/meth.2001.1262.
Madisen, L., Neubauer, M., Plowman, G., Rosen, D., Segarini, P., Dasch, J., Thompson, A.,
ments and data analyses. GO, SSC and PPF carried western blots ana- Ziman, J., Bentz, H., Purchio, A.F., 1990. Molecular cloning of a novel bone-forming
lysis. GO, PPF, and RFC performed statistical analyses. GO, PPF, SSC, compound: osteoinductive factor. DNA Cell Biol. 9, 303–309.
ACMO, and RFC performed study design, data analyses and data in- Motiwala, S.R., Szymonifka, J., Belcher, A., Weiner, R.B., Baggish, A.L., Gaggin, H.K.,
Bhardwaj, A., Januzzi, J.L., 2014. Measurement of novel biomarkers to predict
terpretation. GO, RFC wrote the main manuscript, with contributions chronic heart failure outcomes and left ventricular remodeling. J. Cardiovasc. Transl.
from all authors. All authors critically discussed the results and im- Res. 7, 250–261. https://doi.org/10.1007/s12265-013-9522-8.
plications, reviewed and approved the final version of the manuscript. Mysliwiec, M.R., Bresnick, E.H., Lee, Y., 2011. Endothelial Jarid2/Jumonji is required for
normal cardiac development and proper Notch1 expression. J. Biol. Chem. 286,
17193–17204. https://doi.org/10.1074/jbc.M110.205146.
Acknowledgements Orenes-Piñero, E., Montoro-García, S., Patel, J.V., Valdés, M., Marín, F., Lip, G.Y.H.,
2013. Role of microRNAs in cardiac remodelling: new insights and future perspec-
tives. Int. J. Cardiol. 167, 1651–1659. https://doi.org/10.1016/j.ijcard.2012.09.120.
This study was supported by São Paulo Research Foundation
Pasquinelli, A.E., 2012. MicroRNAs and their targets: recognition, regulation and an
(FAPESP, grant # 2012/13961-6, RFC). GO was funded by a MSc. emerging reciprocal relationship. Nat. Rev. Genet. 13, 271–282. https://doi.org/10.
FAPESP fellowship # 2014/14340-0. 1038/nrg3162.
Petretto, E., Sarwar, R., Grieve, I., Lu, H., Kumaran, M.K., Muckett, P.J., Mangion, J.,
Schroen, B., Benson, M., Punjabi, P.P., Prasad, S.K., Pennell, D.J., Kiesewetter, C.,
References Tasheva, E.S., Corpuz, L.M., Webb, M.D., Conrad, G.W., Kurtz, T.W., Kren, V.,
Fischer, J., Hubner, N., Pinto, Y.M., Pravenec, M., Aitman, T.J., Cook, S.A., 2008.
Bagnall, R.D., Tsoutsman, T., Shephard, R.E., Ritchie, W., Semsarian, C., 2012. Global Integrated genomic approaches implicate osteoglycin (Ogn) in the regulation of left
microRNA profiling of the mouse ventricles during development of severe hyper- ventricular mass. Nat. Genet. 40, 546–552. https://doi.org/10.1038/ng.134.
trophic cardiomyopathy and heart failure. PLoS One 7, e44744. https://doi.org/10. Rajabi, M., Kassiotis, C., Razeghi, P., Taegtmeyer, H., 2007. Return to the fetal gene
1371/journal.pone.0044744. program protects the stressed heart: a strong hypothesis. Heart Fail. Rev. 12,
Bartel, D.P., 2004. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116, 331–343. https://doi.org/10.1007/s10741-007-9034-1.
281–297. Schaub, M.C., Hefti, M. a, Harder, B. a, Eppenberger, H.M., 1997. Various hypertrophic
Bentz, H., Nathan, R.M., Rosen, D.M., Armstrong, R.M., Thompson, A.Y., Segarini, P.R., stimuli induce distinct phenotypes in cardiomyocytes. J. Mol. Med. (Berl) 75,
Mathews, M.C., Dasch, J.R., Piez, K.A., Seyedin, S.M., 1989. Purification and char- 901–920. https://doi.org/10.1007/s001090050182.
acterization of a unique osteoinductive factor from bovine bone. J. Biol. Chem. 264, Scott, J.E., Haigh, M., 1988. Identification of specific binding sites for keratan sulphate
20805–20810. proteoglycans and chondroitin-dermatan sulphate proteoglycans on collagen fibrils
Bentz, H., Chang, R.J., Thompson, A.Y., Glaser, C.B., Rosen, D.M., 1990. Amino acid in cornea by the use of cupromeronic blue in “critical-electrolyte-concentration”
sequence of bovine osteoinductive factor. J. Biol. Chem. 265, 5024–5029. techniques. Biochem. J. 253, 607–610.
Carrington, J.C., Ambros, V., 2003. Role of microRNAs in plant and animal development. Seok, H.Y., Chen, J., Kataoka, M., Huang, Z.-P., Ding, J., Yan, J., Hu, X., Wang, D.-Z.,
Science 301, 336–338. https://doi.org/10.1126/science.1085242. 2014. Loss of MicroRNA-155 protects the heart from pathological cardiac hyper-
Chien, K.R., Knowlton, K.U., Zhu, H., Chien, S., 1991. Regulation of cardiac gene ex- trophy. Circ. Res. 114, 1585–1595. https://doi.org/10.1161/CIRCRESAHA.114.
pression during myocardial growth and hypertrophy: molecular studies of an adap- 303784.
tive physiologic response. FASEB J. 5, 3037–3046. Simpson, D.G., Terracio, L., Terracio, M., Price, R.L., Turner, D.C., Borg, T.K., 1994.
Depre, C., Shipley, G.L., Chen, W., Han, Q., Doenst, T., Moore, M.L., Stepkowski, S., Modulation of cardiac myocyte phenotype in-vitro by the composition and orienta-
Davies, P.J., Taegtmeyer, H., 1998. Unloaded heart in vivo replicates fetal gene ex- tion of the extracellular-matrix. J. Cell. Physiol. 161, 89–105.
pression of cardiac hypertrophy. Nat. Med. 4, 1269–1275. https://doi.org/10.1038/ Small, E.M., Olson, E.N., 2011. Pervasive roles of microRNAs in cardiovascular biology.
3253. Nature 469, 336–342. https://doi.org/10.1038/nature09783.
Duisters, R.F., Tijsen, A.J., Schroen, B., Leenders, J.J., Lentink, V., van der Made, I., Sucharov, C., Bristow, M.R., Port, J.D., 2008. miRNA expression in the failing human
Herias, V., van Leeuwen, R.E., Schellings, M.W., Barenbrug, P., Maessen, J.G., heart: functional correlates. J. Mol. Cell. Cardiol. 45, 185–192. https://doi.org/10.
Heymans, S., Pinto, Y.M., Creemers, E.E., 2009. miR-133 and miR-30 regulate con- 1016/j.yjmcc.2008.04.014.
nective tissue growth factor: implications for a role of microRNAs in myocardial Tasheva, E.S., Koester, A., Paulsen, A.Q., Garrett, A.S., Boyle, D.L., Davidson, H.J., Song,
matrix remodeling. Circ. Res. 104, 170–178. https://doi.org/10.1161/CIRCRESAHA. M., Fox, N., Conrad, G.W., 2002. Mimecan/osteoglycin-deficient mice have collagen
108.182535. (6p following 178). fibril abnormalities. Mol. Vis. 8, 407–415.
Fan, Y., Liu, L., Fang, K., Huang, T., Wan, L., Liu, Y., Zhang, S., Yan, D., Li, G., Gao, Y., Lv, Tasheva, E.S., Ke, A., Deng, Y., Jun, C., Takemoto, L.J., Koester, A., Conrad, G.W., 2004.
Y., Chen, Y., Tu, Y., 2016. Resveratrol ameliorates cardiac hypertrophy by down- Differentially expressed genes in the lens of mimecan-null mice. Mol. Vis. 10,
regulation of miR-155 through activation of breast cancer type 1 susceptibility pro- 403–416.
tein. J. Am. Heart Assoc. 5, 1–11. https://doi.org/10.1161/JAHA.115.002648. Van Aelst, L.N.L., Voss, S., Carai, P., Van Leeuwen, R., Vanhoutte, D., Sanders-van Wijk,
Freire, P.P., Cury, S.S., de Oliveira, G., Fernandez, G.J., Moraes, L.N., da Silva Duran, S., Eurlings, L., Swinnen, M., Verheyen, F.K., Verbeken, E., Nef, H., Troidl, C., Cook,
B.O., Ferreira, J.H., Fuziwara, C.S., Kimura, E.T., Dal-Pai-Silva, M., Carvalho, R.F., S.a., Brunner-La Rocca, H.-P., Möllmann, H., Papageorgiou, A.-P., Heymans, S., 2015.
2017. Osteoglycin inhibition by microRNA miR-155 impairs myogenesis. PLoS One Osteoglycin prevents cardiac dilatation and dysfunction after myocardial infarction
12, e0188464. https://doi.org/10.1371/journal.pone.0188464. through infarct collagen strengthening. Circ. Res. 116, 425–436. https://doi.org/10.
He, W., Huang, H., Xie, Q., Wang, Z., Fan, Y., Kong, B., Huang, D., Xiao, Y., 2015. MiR- 1161/CIRCRESAHA.116.304599.
155 knockout in fibroblasts improves cardiac remodeling by targeting tumor protein van Rooij, E., Olson, E.N., 2009. Searching for miR-acles in cardiac fibrosis. Circ. Res.
p53-inducible nuclear protein 1. J. Cardiovasc. Pharmacol. Ther. 21, 1–13. https:// 104, 138–140. https://doi.org/10.1161/CIRCRESAHA.108.192492.
doi.org/10.1177/1074248415616188. van Rooij, E., Sutherland, L.B., Liu, N., Williams, A.H., McAnally, J., Gerard, R.D.,
Heymans, S., Corsten, M.F., Verhesen, W., Carai, P., van Leeuwen, R.E.W., Custers, K., Richardson, J. a, Olson, E.N., 2006. A signature pattern of stress-responsive
Peters, T., Hazebroek, M., Stöger, L., Wijnands, E., Janssen, B.J., Creemers, E.E., microRNAs that can evoke cardiac hypertrophy and heart failure. Proc. Natl. Acad.
Pinto, Y.M., Grimm, D., Schürmann, N., Vigorito, E., Thum, T., Stassen, F., Yin, X., Sci. U. S. A. 103, 18255–18260. https://doi.org/10.1073/pnas.0608791103.
van Rooij, E., Sutherland, L.B., Thatcher, J.E., DiMaio, J.M., Naseem, R.H., Marshall,

14
G. de Oliveira et al. Gene 676 (2018) 9–15

W.S., Hill, J. a, Olson, E.N., 2008. Dysregulation of microRNAs after myocardial in- (RESEARCH0034).
farction reveals a role of miR-29 in cardiac fibrosis. Proc. Natl. Acad. Sci. U. S. A. 105, Williamson, R.E., Darrow, K.N., Giersch, A.B.S., Resendes, B.L., Huang, M., Conrad, G.W.,
13027–13032. https://doi.org/10.1073/pnas.0805038105. Chen, Z.-Y., Charles Liberman, M., Morton, C.C., Tasheva, E.S., 2008. Expression
Vandesompele, J., De Preter, K., Pattyn, F., Poppe, B., Van Roy, N., De Paepe, A., studies of osteoglycin/mimecan (OGN) in the cochlea and auditory phenotype of
Speleman, F., 2002. Accurate normalization of real-time quantitative RT-PCR data by Ogn-deficient mice. Hear. Res. 237, 57–65. https://doi.org/10.1016/j.heares.2007.
geometric averaging of multiple internal control genes. Genome Biol. 3 12.006.

15

You might also like