Page 1 of 36

Accepted Preprint first posted on 15 April 2015 as Manuscript JME-14-0314

1

Glucocorticoid inhibits cell proliferation in differentiating osteoblasts by microRNA-199a targeting

2

WNT signaling 1,2

Changgui Shi

3

1

1

1,2

1

1

1

*, Ping Huang *, Hui Kang , Bo Hu , Jin Qi , Min Jiang , Hanbing Zhou , Lei 1#

1

Guo , Lianfu Deng

4 1

5

Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Orthopaedics and

6

Traumatology, Shanghai Ruijin Hospital, Shanghai jiaotong University School of Medicine, China 2

7

Department of Orthopedics, Changzheng Hospital, the Second Military Medical University of China, Shanghai, China

8 9

* These authors contributed equally to this work:

10

Changgui Shi, E-mail: [email protected]

11

Ping Huang, E-mail: [email protected] #

Correspondence to:

12 13

Dr. Lei Guo

14

Professor

15

Ruijin Hospital, Shanghai jiaotong University School of Medicine

16

No.197, The Second Ruijin Road, Luwan District

17

Shanghai, 200025, P. R. of China

18

Tel: 011-86-21-643-135-34

19

Fax: 011-86-21-643-357-42

20

E-mail: [email protected]

21

Running title: MiR-199a and osteoblasts proliferation.

22

Keywords : MicroRNA-199a-5p, Glucocorticoids, WNT signaling, osteoblasts, proliferation. 1

Copyright © 2015 by the Society for Endocrinology.

Page 2 of 36

1

Word count of this manuscript: 4281.

2

Abstract

3

Inhibition of osteoblasts proliferation by glucocorticoids is very important in the etiology of

4

glucocorticoid-induced osteoporosis. The mechanisms of this process are still not fully understood.

5

Recent studies implicated an important role of microRNAs in glucocorticoid-mediated responses in

6

various cellular processes, including cell proliferation and apoptosis. Therefore, we hypothesized that

7

these regulatory molecules might be implicated in the process of glucocorticoid-decreased

8

osteoblasts proliferation. Western blot, quantitative real-time PCR, cells proliferation assay, and

9

luciferase assay were employed to investigate the role of microRNAs in glucocorticoid-inhibited

10

osteoblasts proliferation. The microRNA-199a-5p was significantly increased in osteoblasts treated

11

with dexamethasone. To delineate the role of microRNA-199a-5p, we respectively silenced and

12

overexpressed microRNA-199a-5p in osteoblasts. We found that over-expressing

13

microRNA-199a-5p remarkably enhanced the inhibition effect of dexamethasone on osteoblasts

14

proliferation and microRNA-199a-5p depletion significantly attenuated dexamethasone-inhibited

15

osteoblasts proliferation. Mechanistic studies showed that microRNA-199a-5p inhibited FZD4 and

16

WNT2 expression through a microRNA-199a-5p-binding site within the 3′- untranslational region of

17

FZD4 and WNT2. The post-transcriptional repression of FZD4 and WNT2 were further confirmed

18

by luciferase reporter assay. These results showed that microRNA-199a-5p may play a significant

19

role in the process of glucocorticoid-inhibited osteoblasts proliferation by regulating WNT signaling

20

pathway.

21 22 2

Page 3 of 36

1 2 3 4

Introduction

5

Glucocorticoids (GCs) are widely used for the treatment of inflammatory and immune diseases,

6

including asthma, rheumatoid arthritis, Crohn’s disease, etc(Shi, et al. 2014; Tait, et al. 2008).

7

Although GCs are used extensively to relieve these diseases, increased bone fragility attributed to

8

osteopenia is a serious side effect of prolonged GCs administration(Kondo, et al. 2008). Indeed,

9

GC-induced osteoporosis (GIO) is currently the third leading cause of osteoporosis following

10

sex-steroid deficiency and old age(Weinstein, et al. 1998). GC-treated patients are at a twofold

11

higher risk of suffering from a fracture, irrespective of their bone mineral density (BMD)(Weinstein

12

2001). Limited information is available on its pathogenesis, since the clinical picture of GIO mostly

13

reflects a combined effect of the underlying systemic disease and of the secondary effects induced by

14

GCs treatment.

15

The elucidation of the cellular and molecular mechanisms that lead to GIO and the development of

16

improved means of identifying those at risk remain important challenges(Hong, et al. 2008). It is

17

generally accepted that reduced bone formation is the predominant effect of GCs on bone turn

18

over(Seibel, et al. 2013; Canalis, et al. 2004). Previous studies found that induction of osteoblasts

19

apoptosis and inhibition of osteoblasts proliferation and differentiation finally lead to reduction of

20

bone formation(Hong, et al. 2011; Pereira, et al. 2001; Weinstein, et al. 1998). It has been well

21

established by both in vivo and in vitro studies that GCs regulate osteoblasts apoptosis and

22

differentiation. However, the precise molecular events underlying the effect of GCs on proliferation 3

Page 4 of 36

1

pathways in osteoblasts are not known.

2

MicroRNAs (miRNAs) are endogenous non-coding RNAs 19-25 nucleotides in length that regulate

3

various biological processes including cell proliferation, apoptosis, development, hematopoiesis,

4

organogenesis and tumorigenesis. The miRNAs bind to matched sequences in the 3'-untranslational

5

region (3'-UTR) of target mRNAs and either repress the translation or degrade their target mRNAs

6

transcript(Couzin 2007; Zamore and Haley 2005). The miRNAs may also promote translation of

7

selected mRNAs in a cell cycle phase-dependent way(Zeng 2006). Recent studies implicated that

8

some miRNAs play crucial role in bone formation, such as miR-27a, miR-29a, miR-34a, miR-125b,

9

miR-133, miR-138, miR-199a, miR-206, miR-338, miR-335, miR-378(Taipaleenmaki, et al. 2012).

10

Furthermore, previous studies confirmed that many of these miRNAs could mediate cells

11

proliferation, including miR-27a, miR-29a, miR-34a, miR-125b, miR-199a, and

12

miR-574(Chiyomaru, et al. 2013; Wu, et al. 2013; Huang, et al. 2012; Ma, et al. 2012; Wei, et al.

13

2012; Xu, et al. 2012). Moreover, these six miRNAs were reported to regulate Wnt signaling

14

pathway(Hashemi, et al. 2015; Guo, et al. 2014; Rathod, et al. 2014; Nagano, et al. 2013; Liu, et al.

15

2013; Chiyomaru, et al. 2013), a crucial regulator of glucocorticoid-mediated bone acquisition and

16

remodeling activities. Thus, we hypothesized that these molecules might be involved in the process

17

of GC-repressed osteoblasts proliferation.

18

In this study, we examined the role of miR-199a-5p in the repression of osteoblasts proliferation by

19

GCs. we detected some miRNAs expression, which have been reported to be related to bone

20

formation and mediate Wnt signaling, in osteoblasts exposed to GCs. Several miRNAs were found to

21

be altered, in which miR-199a-5p was identified as a strong candidate responsible for GC-decreased

22

osteoblasts proliferation. Further studies confirmed that miR-199a-5p regulated osteoblasts 4

Page 5 of 36

1

proliferation by targeting WNT signaling. Therefore, miRNAs and miRNA-regulated gene silencing

2

may contribute to inhibition effects of GCs on osteoblasts proliferation.

3

Materials and Methods

4

In vivo treatment of mice

5

Seven-day-old neonatal C57 female mice were used for this study. Accroding to gohel et al

6

report(Gohel, et al. 1999), stock solutions of 1 mg/ml Dex were prepared in ethanol. Dosing

7

solutions were prepared by diluting the stock solution with normal saline. After measuring their

8

weight, mice were given daily sc injections of Dex (1.0 mg/kg BW). At 72 h, mice were weighed and

9

killed. The entire calvarium was removed for quantitative real-time PCR (qRT-PCR).

10

Cell culture

11

MC3T3-E1 cell line was supplied from Shanghai Institute of Orthopaedics and Traumatology. Lines

12

< 20 passages were used in the present studies. Primary osteoblasts were obtained from neonatal

13

murine calvaria using methods previously described(Shi, et al. 2014). Cells were cultured with

14

alpha-minimal essential media (α-MEM) (Invitrogen, Paisley, UK) supplemented with 10% fetal

15

bovine serum and 100 µg/ml penicillin/streptomycin. All experiments were performed under

16

differentiation conditions, i.e. in the presence of 50 ug/ml ascorbic acid, 4 mmol/L

17

beta-glycerophosphate (Sigma-Aldrich, St Louis, USA). HEK293 were cultured in Dulbecco’s

18

Modified Eagle Medium (DMEM) (Invitrogen, Paisley, UK). The cultures were supplemented with

19

10% fetal bovine serum and 100 µg/ml penicillin/streptomycin.

20

Cell proliferation assay

21

A cell counting kit (CCK-8) assay (Dojindo Laboratories, Japan) was used to measure cell

22

proliferation according to the guidance of the manufacturer. Briefly, cells were resuspended in 200 µl 5

Page 6 of 36

1

cell culture medium and seeded at a density of 1 × 103 cells per well in 96-well microtiter plates,

2

incubated overnight for cell attachment. 10 µl CCK-8 reagents were added to each well one hour

3

before the end of incubation. The optical density value (OD) of each sample was measured at a

4

wavelength of 450 nm on a microplate reader to determine the viability of the cells. The proliferation

5

rate was normalized to the value at 0 time point.

6

5-ethynil-2'-deoxyuridine (EdU) assay

7

Logarithmic growth phase cells were seeded in 24-well plates and incubated with serum-free

8

α-MEM for 24 hours. In brief, EdU (Sigma-Aldrich, St Louis, USA) solution was added to cell

9

culture medium to a final concentration of 1:1000, and then incubated for two hours. Cell fixative

10

(containing 4% paraformaldehyde in PBS) was added before incubation at room temperature for 30

11

minutes. After washing cells with PBS for two times, click reaction buffer (Tris-HCl, pH 8.5,

12

100 mM; CuSO4, 1 mM; Apollo 550 fluorescent azide, 100 µM; ascorbic acid, 100 mM) was added

13

for 10-30 min while protecting from light. Then cells were washed with 0.5% Triton X-100 for three

14

times and stained subsequently with Hoechst (5 µg/ml) for 30 min at room temperature. Samples

15

were stored in the dark at 4°C prior to fluorescence microscope (Olympus). EdU positive cells were

16

caculated with (EdU add-in cells/Hoechst stained cells) × 100%.

17

5-(and-6)-carboxyfluorescein diacetate, succinimidyl ester (CFSE) assay

18

Cell proliferation was measured by CFSE staining and flow cytometry(Xu, et al. 2006). Cells were

19

incubated with CFSE (Invitrogen, Paisley, UK) at a concentration of 5 µmol/L in PBS for 15 minutes

20

at 37℃. The reaction was stopped by adding fetal calf serum (FCS). Cells were replated at a density

21

of 48,000/well in six-well dishes and incubated for 3 to 5 days. After preparation by trypsinization

22

and washing, fluorescence intensity was measured on flow cytometry using excitation at 488 nm at 6

Page 7 of 36

1

the FL1 detection channel and analyzed with CellQuest software.

2

MiR-199a-5p target-gene prediction

3

We used a computation and bioinformatics-based approach to predict the putative targets of

4

miR-199a-5p through TargetScan, which is hosted by the Wellcome Trust Sanger Institute. WNT

5

signaling including FZD4 and WNT2 were predicted as potential target genes of miR-199a-5p by

6

this program.

7

Western blot analysis

8

The protein samples were extracted from osteoblasts, with the procedures essentially the same as

9

described in detail elsewhere(Luo, et al. 2007; Yang, et al. 2007). Protein samples (~50 µg) were

10

fractionated by SDS-PAGE (7.5-10% polyacrylamide gels). Separated proteins were blot transferred

11

onto a nitrocellulose membrane. After blocking with 0.1% Tween 20 and 5% nonfat dry milk in

12

Tris-buffered saline at room temperature for 1 h, the membrane was incubated overnight at 4°C in

13

one of the following primary antibodies: FZD4 (Peprotech, Rocky Hill, NJ) (1:400) , WNT2 (Santa

14

Cruz, CA, USA) (1:400), B-catenin (Santa Cruz, CA, USA) (1:200), Runx2 (Santa Cruz, CA, USA)

15

(1:200), Osterix (Santa Cruz, CA, USA) (1:400) and β-actin (Santa Cruz, CA, USA) (1:1000) as an

16

internal control. The membrane was incubated with horseradish peroxidase-conjugated secondary

17

antibody (1:2000) for 1 h and detected using the Enhanced Chemiluminescence (ECL) Western blot

18

System (Amersham Biosciences).

19

Synthesis of miRNAs and sequences of miR-199a-5p inhibitors

20

MiR-199a-5p (sense: 5'-CCCAGUGUUCAGACUACCUGUUC-3' , antisense:

21

5'-ACAGGUAGUCUG AACACUGGGUU-3') and its antisense oligonucleotides (AMOs:

22

5'-GAACAGGTAGTCTGAACA CTGGG-3') were synthesized by Integrated DNA Technologies 7

Page 8 of 36

1

(IDT). Additionally, a scrambled RNA was used as negative control (NC), sense:

2

5'-UUCUCCGAACGUGUCACGUTT-3' and antisense: 5'-ACGUGACACGUUCGGAGAATT-3'.

3

DNA fragments of the 3'- UTRs of WNT2 and FZD4 mRNA containing the putative miR-199a-5p

4

binding sequence were synthesized by Invitrogen. These fragments were then respectively cloned

5

into the multiple cloning sites downstream the luciferase gene (HindIII and SpeI sites) in the

6

pMIR-REPORTTM luciferase miRNA expression reporter vector (Ambion, Inc.), as described

7

elsewhere(Yang, et al. 2007).

8

Transfection of miR-199a-5p/AMO-199a-5p in osteoblasts.

9

The MC3T3-E1 cells/primary osteoblasts were cultured with differentiation medium for 24 h.

10

Osteoblasts cultured in 6-well plates was divided into different groups. Osteoblasts in the control

11

group were cultured in differentiation culture medium. MicroRNA group were transfected with

12

miR-199a-5p or/and inhibitors for 48 h under differentiation condition, with X-tremeGENE siRNA

13

Transfection Reagent (Roche, Basel, Switzerland), as according to the manufacturer’s instructions.

14

Dexamethasone (Dex) (Sigma-Aldrich, St Louis, USA) group were treated with 10 -7M Dex for 5

15

days. MicroRNA together with Dex group cells were transfected with miR-199a-5p or/and inhibitors

16

for 48h under 10 -7M Dex and then changed with Dex for 3 days.

17

Luciferase Activity Assay

18

After 24 hours starvation in serum-free medium, HEK293 cells (1-105 per well) were transfected

19

with 1 µg miR-199a-5p/AMO-199a-5p or 1 µg PGL3-target DNA (firefly luciferase vector) and 0.1

20

µg PRL-TK (TK-driven Renilla luciferase expression vector), with Lipofectamine 2000 (Invitrogen),

21

as according to the manufacturer’s instructions. Luciferase activities were measured 48 hours after

22

transfection with a dual luciferase reporter assay kit (Promega) on a luminometer (Lumat LB9507). 8

Page 9 of 36

1

Quantification of miRNA levels

2

The mirVanaTM qRT-PCR miRNA Detection Kit (Ambion) was used in conjunction with real-time

3

PCR with SYBR Green I for quantification of miR-199a-5p transcript, as detailed elsewhere(Luo, et

4

al. 2007; Yang, et al. 2007).

5

Statistics

6

All data were analysed by using the SPSS 19.0 Software (SPSS, Inc.). The composite data are

7

expressed as means ± s.e.m. Statistical analysis was performed with one-way ANOVA followed by

8

Dunnett’s test where appropriate. Differences were considered to be significant at P≤0.05.

9

Results

10

The effect of Dex on differentiating osteoblasts proliferation.

11

To determine whether Dex regulated the proliferation of differentiating osteoblasts, we firstly used

12

CCK-8 assay to monitor cell proliferation. MC3T3-E1 cells under differentiation conditions were

13

treated with Dex at the concentrations (0, 10–9, 10–8, 10–7 M) from day 1 to day 5. Our results showed

14

that Dex decreased the proliferation of MC3T3-E1 cells in a dose- and time- dependent manner

15

(Figure 1A).

16

Furthermore, MC3T3-E1 cells were stimulated with 10–7 M Dex for 5 days prior to EdU assay.

17

EdU-stained photomicrographs and corresponding photomicrographs of total cells were shown in

18

Figure 1B. The proportion of cells with EdU-positive nuclei was shown in Figure 1C. EdU

19

incorporation was decreased in the Dex group, indicating that Dex could inhibit the MC3T3-E1 cells

20

proliferation.

21

To further examine proliferation, MC3T3-E1cells were cultured with 10–7 M Dex for 5 days and

22

stained with CFSE. CFSE irreversibly couples to cellular proteins. When cells divide, CFSE labeling 9

Page 10 of 36

1

is distributed equally between daughter cells, which are half as fluorescent as their parents. The peak

2

CFSE fluorescence intensity on flow cytometry was shifted by Dex, indicating that cells treated with

3

Dex had fewer cycles of cell replication as compared with the control group (Figure 1D).

4

Up-regulation of miR-199a-5p expression by Dex in differentiating osteoblasts.

5

To examine the role of miRNAs in the repression of osteoblasts proliferation by Dex, we firstly

6

detected some miRNAs expression, which have been reported to be related to bone formation and

7

mediate WNT signaling, in MC3T3-E1 cells exposed to 10–7 M Dex for 5 days. Several miRNAs

8

were found to be altered, in which miR-199a-5p was identified as a strong candidate responsible for

9

GC-decreased osteoblasts proliferation. We found that miR-574-3p and miR-27a were weakly

10

affected during this time frame. However, the expression of miR-199a-5p was significantly

11

upregulated by Dex (Figure 2A). Further studies demonstrated that Dex increased the expression of

12

miR-199a-5p in a time- dependent manner (Figure 2B). Similarly, the upregulation of miR-199a-5p

13

expression was also observed in primary osteoblasts treated with 10-7 M Dex for 5 days (Figure 2C).

14

Furthermore, we observed the effects of Dex on miR-199a-5p expression in vivo. We found that the

15

expression of miR-199a-5p was significantly upregulated in calvarias from mice treated with Dex

16

(Figure 2D). Thus, we hypothesized that miR-199a-5p might be involved in the process of

17

Dex-decreased osteoblasts proliferation.

18

Repression of differentiating osteoblasts proliferation by miR-199a-5p.

19

To delineate the role of miR-199a-5p in osteoblasts proliferation, we performed loss-of-function and

20

gain-of-function experiments in which we decreased and increased the quantities of miR-199a-5p

21

with miR-199a-5p inhibitor and miR-199a-5p mimic, respectively. MC3T3-E1 cells were transfected

22

with miR-199a-5p or/and AMO-199a-5p from day 1 to day 5. Then we assessed cell proliferation 10

Page 11 of 36

1

with CCK-8 assays. We found that overexpression of miR-199a-5p decreased MC3T3-E1 cells

2

proliferation. However, the depletion of miR-199a-5p with AMO-199a-5p resulted in the increased

3

MC3T3-E1 cells proliferation.

4

Furthermore, EdU assay by fluorescence microscope further demonstrated that miR-199a-5p mimic

5

clearly decreased MC3T3-E1 cells proliferation (Figure 3B,C), and miR-199a-5p inhibitor

6

significantly enhanced osteoblasts proliferation. Similar results were further confirmed by CFSE

7

fluorescence intensity assayment (Figure 3D). In addition, the effect of miR-199a-5p on osteoblasts

8

differentiation was observed by western blot. The expressions of osteogenic marker genes including

9

Runx2 and Osterix were significantly affected in MC3T3-E1 cells transfected with miR-199a-5p

10

or/and AMO-199a-5p for 48h, suggesting that miR-199a-5p was also involved in the process of

11

osteoblasts differentiation (Figure 3E).

12

Furthermore, primary osteoblasts were also used to test the effects of miR-199a-5p on cells

13

proliferation. The proliferation of primary osteoblasts transfected with miR-199a-5p or/and

14

AMO-199a-5p was measured by CCK8, EdU assay and CFSE fluorescence intensity assayment.

15

Similarly, we found that miR-199a-5p significantly decreased primary osteoblasts proliferation and

16

AMO-199a-5p enhanced it (Figure 4A-C).

17

could regulate osteoblasts proliferation. Involvement of miR-199a-5p in Dex-reduced osteoblasts

18

proliferation.

19

We consequently investigated whether miR-199a-5p was involved in Dex-decreased osteoblasts

20

proliferation. To this end, we observed the effect of Dex on proliferation in MC3T3-E1 cells

21

transfected with miR-199a-5p or/and AMO-199a-5p for 5 days. CCK-8 assay showed that

22

miR-199a-5p markedly enhanced Dex-decreased MC3T3-E1 cells proliferation. Furthermore, 11

Taken together, these results indicated that miR-199a-5p

Page 12 of 36

1

co-application of miR-199a-5p and AMO-199a-5p almost completely abolished the effect of

2

miR-199a-5p. Moreover, the inhibitory effect of Dex on MC3T3-E1 cells proliferation was

3

significantly alleviated when MC3T3-E1 cells were alone transfected with AMO-199a-5p, indicating

4

that miR-199a-5p was involved in Dex-decreased osteoblasts proliferation (Figure 5A). Similar

5

results were further confirmed by EdU assay and flow cytometric analysis of CFSE intensity (Figure

6

5B-C). All of above results were further verified in primary osteoblasts (Figure 6A-C).

7

Repression of WNT signaling by miR-199a-5p transfection.

8

Based on the above observations, miR-199a-5p was involved in osteoblasts proliferation. It is

9

possible that miR-199a-5p targets several regulatory factors related to osteoblasts proliferation. To

10

address this issue, we used a computation and bioinformatics-based approach to predict the putative

11

targets related to proliferation through TargetScan, which is hosted by the Wellcome Trust Sanger

12

Institute. These explorations lead to the identification of candidate targets of miR-199a-5p: WNT

13

signaling including FZD4 and WNT2 (Figure 7A,B). Western blot analysis of FZD4 and WNT2

14

expression in the MC3T3-E1 cells treated with 10-7 M Dex for 5 days are showed in Figure 7C,D.

15

We found that Dex decreased FZD4 and WNT2 expression in MC3T3-E1 cells. To prove that FZD4

16

and WNT2 are indeed repressed posttranscriptionally by miR-199a-5p, we determined the effect of

17

the miR-199a-5p on protein expression. Western blot analysis showed that miR-199a-5p lowered

18

markedly the levels of FZD4 and WNT2 proteins in MC3T3-E1 (Figure 7E,F). Co-application of

19

miR-199a-5p and AMO-199a-5p abolished almost completely the effect of miR-199a-5p. Moreover,

20

application of the AMO-199a-5p alone increased the levels of FZD4 and WNT2 in MC3T3-E1,

21

indicating that there is a basal level of miR-199a-5p activity in osteoblasts (Figure 7E,F). Similarly,

22

we also test the effects of miR-199a-5p on the levels of FZD4 and WNT2 proteins in primary 12

Page 13 of 36

1

osteoblasts. Our results showed that miR-199a-5p significantly decreased FZD4 and WNT2 proteins

2

expression and AMO-199a-5p increased it (Figure 8A,B). In addition, we observed the effect of

3

miR-199a-5p on the expression of B-catenin, a key mediater of WNT signaling pathway, in primary

4

osteoblasts. We found that miR-199a-5p decreased B-catenin expression, suggesting that

5

miR-199a-5p could inhibit WNT signaling pathway (Figure 8B).

6

Verification of interactions between miR-199a-5p and their target genes.

7

We constructed chimeric vectors by placing the 3'-UTRs of FZD4 and WNT2 into the 3'-UTR of a

8

luciferase reporter plasmid. We performed luciferase reporter assays in HEK293 cells that do not

9

express miR-199a-5p (Data not shown). Compared with the NC, transfection of miR-199a-5p with

10

the luciferase reporter gene linked to the 3'-UTR of FZD4 or WNT2 resulted in a significant decrease

11

of luciferase activity, and co-application of miR-199a-5p with AMO-199a-5p alleviated the decrease

12

of luciferase activity, whereas AMO-199a-5p alone had no effect (Figure 9A,B). These data suggest

13

that FZD4 and WNT2 are the targets of miR-199a-5p.

14

Successful delivery of miR-199a-5p, AMO-199a-5p and NC to the cells were further verified by

15

comparing the miR-199a-5p levels 48 hours after transfection of the constructs in MC3T3-E1 cells

16

and primary osteoblasts. Transfection resulted in significant increasing in miR-199a-5p levels

17

(Figure 9C). It is worth noting that the miR-199a-5P levels were dynamic after transfection. Our data

18

were collected at a specific time which was 48 hours after transfection, because the plateau level was

19

reached then. These results proved the feasibility of all experiments.

20

Discussion

21

Increased concentrations of GCs could cause the development of Cushing’s syndrome, with severe

22

osteoporosis. Previous studies have revealed that GCs have potent inhibitory effects on osteoblasts 13

Page 14 of 36

1

proliferation(Hong, et al. 2011; Walsh, et al. 2001). However, the molecular mechanisms involved in

2

the GC-inhibited osteoblasts proliferation are still poorly understood. In this study, we have

3

illustrated that miR-199a-5p plays a significant role in the process of GC-decreased osteoblasts

4

proliferation, by regulating WNT signaling pathway.

5

Several recent reports have suggested that GCs exert post-transcriptional control through the

6

regulation of miRNAs processing and expression(De Iudicibus, et al. 2013). Xing et al.reported that

7

GCs induced apoptosis by inhibiting miRNAs cluster miR-17~92 expression in chondrocytic

8

cells(Xing, et al. 2014). Furthermore, miR-29b and miR-29c were reported to be involved in

9

Toll-like receptor control of GC-induced apoptosis in human plasmacytoid dendritic cells(Hong, et al.

10

2013). In addition, miRNAs could also negatively regulate the GC receptor (GR) transcriptional

11

response by directly targeting the 3'-UTR of GRα mRNA(Lv, et al. 2012). It is also known that

12

osteoblasts are critical target cells of GCs. Our previous studies showed that miR-17~92a

13

downregulation by GCs leads to Bim targeting and induction of osteoblasts apoptosis(Guo, et al.

14

2013). Furthermore, GCs could also increase Receptor activator of nuclear factor B ligand (RANKL)

15

expression by down-regulation of miR-17/20a in osteoblasts, which indirectly enhances

16

osteoclastogenesis and bone resorption(Shi, et al. 2014). However, to our knowledge, there have

17

been no reports on whether miRNAs expression could be regulated by GCs in osteoblasts

18

proliferation. The present study is the first effort to observe the effect of GCs on miRNAs in

19

osteoblasts proliferation.

20

In this study, we detected some miRNAs expression in osteoblasts exposed to GCs. These miRNAs

21

have been reported to be related to cell proliferation and bone formation in previous

22

studies(Chiyomaru, et al. 2013; Wu, et al. 2013; Taipaleenmaki, et al. 2012; Huang, et al. 2012; Ma, 14

Page 15 of 36

1

et al. 2012; Wei, et al. 2012; Xu, et al. 2012). Furthermore, these miRNAs could mediate WNT

2

signaling pathway(Hashemi, et al. 2015; Guo, et al. 2014; Rathod, et al. 2014; Nagano, et al. 2013;

3

Liu, et al. 2013; Chiyomaru, et al. 2013), a curcial factor of GC-decreased bone formation. Therefore,

4

we firstly examined whether these six miRNAs were involved in the regulation of osteoblasts

5

proliferation by GC. Our results showed that only miR-199a-5p was identified as a strong candidate

6

responsible for GC-decreased osteoblasts proliferation. We speculated that miRNAs might play

7

different roles in different cells. Our study did not apply miRNAs chip to screen miRNAs expression

8

in response to GC-decreased osteoblasts proliferation. Therefore, several differentially expressed

9

miRNAs may be missed. Future studies are required to identify other miRNAs involved in

10

GC-inhibited osteoblasts proliferation.

11

Our results identified miR-199a-5p was significantly increased during Dex-decreased osteoblasts

12

proliferation. Furthermore, overexpression of miR-199a-5p decreased osteoblasts proliferation.

13

However, the depletion of miR-199a-5p with AMO-199a-5p resulted in the increased proliferation of

14

osteoblasts. Previous studies showed that miR-199a-5p was a ubiquitously expressed miRNA, whose

15

expression was modulated at key time points of development, growth, regeneration, cancers(Shi, et

16

al. 2014; Alexander, et al. 2013; Xu, et al. 2012). Shi et al. showed that miR-199a-5p affected

17

porcine preadipocyte proliferation and differentiation(Shi, et al. 2014). Alexander et al. found that

18

miR-199a-5p affected WNT signaling, cell proliferation, and myogenic differentiation(Alexander, et

19

al. 2013). Furthermore, Lentivirus-mediated overexpression of miR-199a-5p inhibited cell

20

proliferation of human hepatocellular carcinoma(Xu, et al. 2012). Although these studies have

21

reported the role of miR-199a-5p in the pathogenesis of several diseases, including breast cancer,

22

lymphoma or pulmonary hypertension, our study is the first report to uncover the role of 15

Page 16 of 36

1

miR-199a-5p in metabolic bone diseases and osteoblasts proliferation.

2

GCs, which regulate diverse physiological effects, have established both genomic and nongenomic

3

mechanisms(Stahn and Buttgereit 2008). The steroid’s nongenomic effects occur within seconds to

4

minutes and are mediated by the GR or by other means such as G protein-coupled receptors(Stahn

5

and Buttgereit 2008). It has been elucidated that GCs are unable to suppress bone formation in the

6

absence of GR expression in osteoblasts as their proliferation, differentiation, and apoptosis will

7

become immune to GCs(Rauch, et al. 2010). Therefore, GC-inhibited proliferation of osteoblasts is

8

triggered via GR-dependent transcriptional regulation. Previous study reported that GCs could

9

regulate miRNAs expression via a GR-mediated direct DNA binding mechanism(De Iudicibus, et al.

10

2013). In this study, we did not explore whether GR was involved in the regulation of miR-199a-5p

11

by GCs in osteoblasts. Therefore, further studies need to be done to uncover the progress of

12

GC-regulated miR-199a-5p in osteoblasts. WNT/ B-catenin signaling plays an important role in bone

13

development and metabolism by controlling both bone formation and resorption(Wang, et al. 2014).

14

Wnt ligands are secreted molecules that bind to cell surface receptors encoded by the Frizzled and

15

low-density lipoprotein receptor-related proteins (LRPs). Once bound, the ligands initiate a cascade

16

of intracellular events that activate the B-catenin activity and the DNA binding protein TCF, which

17

eventually result in the transcription of target genes(Wang, et al. 2014). It has been confirmed that

18

GCs could inhibit canonical WNT signaling to suppress bone formation(Guanabens, et al. 2014). In

19

this study, miR-199a-5p was involved in GC-inhibited osteoblasts proliferation through targeting

20

WNT signaling. Previous studies also showed that miR-199a-5p could target several cell

21

proliferation regulatory factors within the WNT signaling pathway, including FZD4 and

22

WNT2(Alexander, et al. 2013). 16

Page 17 of 36

1

Our data provide new evidence that miR-199a-5p plays a dominant effect in GC-inhibited

2

osteoblasts proliferation. The inhibitory effect of miR-199a-5p on osteoblasts proliferation is

3

ascribed to the disrupting of WNT signaling. This study is an effort to better understand the

4

molecular mechanism of GIO, and to provide new insights into the potential contribution of miRNAs

5

in the process of GC-mediated osteoblasts proliferation and bone formation.

6

Declaration of interest

7

The authors declare that there is no conflict of interest that could be perceived as prejudicing the

8

impartiality of the research reported.

9

Funding

10

This work was supported by the National Natural Science Foundation of China [Grant number.

11

81300713]; Innovation Program of Shanghai Municipal Education Commision [Grant number.

12

14YZ044]; and the Shanghai Jiaotong University School of Medicine Grant [Grant number.

13

13XJ10060].

14

Author contributions

15

L.G. and C.G.S. were involved in the conception and hypothesis delineation; L.G. and C.G.S.

16

designed the experiments, conducted the luciferase and quantitative real-time PCR experiments, and

17

wrote the article; J.Q., B.H., H.B.Z. and H.K. performed EdU staining and CFSE assay; P.H.,

18

modified the manuscript; M.J. and P.H. performed a part of the luciferase, western blot analysis;

19

L.F.D. designed and conducted the animal studies.

20

Acknowledgements

21

The authors thank Dr Zhaoping Qiu for expert technical assistance in carrying out Real time RT-PCR

22

and western blotting analysis. 17

Page 18 of 36

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19

References:

20

Alexander MS, Kawahara G, Motohashi N, Casar JC, Eisenberg I, Myers JA, Gasperini MJ, Estrella

21

EA, Kho AT & Mitsuhashi S, et al. 2013 MicroRNA-199a is induced in dystrophic muscle and

22

affects WNT signaling, cell proliferation, and myogenic differentiation. Cell Death And 18

Page 19 of 36

Differentiation 20 1194-1208.

1 2

Canalis E, Bilezikian JP, Angeli A & Giustina A 2004 Perspectives on glucocorticoid-induced osteoporosis. Bone 34 593-598.

3 4

Chiyomaru T, Yamamura S, Fukuhara S, Hidaka H, Majid S, Saini S, Arora S, Deng G, Shahryari V

5

& Chang I, et al. 2013 Genistein up-regulates tumor suppressor microRNA-574-3p in prostate

6

cancer. PLoS One 8 e58929.

7

Couzin J 2007 Genetics. Erasing microRNAs reveals their powerful punch. Science 316 530.

8

De Iudicibus S, Lucafo M, Martelossi S, Pierobon C, Ventura A & Decorti G 2013 MicroRNAs as tools to predict glucocorticoid response in inflammatory bowel diseases. World J Gastroenterol

9

19 7947-7954.

10 11

Gohel A, McCarthy MB & Gronowicz G 1999 Estrogen prevents glucocorticoid-induced apoptosis in osteoblasts in vivo and in vitro. Endocrinology 140 5339-5347.

12 13

Guanabens N, Gifre L & Peris P 2014 The role of Wnt signaling and sclerostin in the pathogenesis of glucocorticoid-induced osteoporosis. Curr Osteoporos Rep 12 90-97.

14 15

Guo D, Li Q, Lv Q, Wei Q, Cao S & Gu J 2014 MiR-27a targets sFRP1 in hFOB cells to regulate proliferation, apoptosis and differentiation. PLoS One 9 e91354.

16 17

Guo L, Xu J, Qi J, Zhang L, Wang J, Liang J, Qian N, Zhou H, Wei L & Deng L 2013

18

MicroRNA-17-92a upregulation by estrogen leads to Bim targeting and inhibition of osteoblast

19

apoptosis. Journal Of Cell Science 126 978-988.

20

Hashemi GA, Burkhard FC, Rehrauer H, Aquino FC & Monastyrskaya K 2015 MicroRNA

21

MiR-199a-5p Regulates Smooth Muscle Cell Proliferation and Morphology by Targeting

22

WNT2 Signaling Pathway. Journal Of Biological Chemistry 290 7067-7086. 19

Page 20 of 36

1

Hong D, Chen HX, Ge RS & Li JC 2008 The biological roles of extracellular and intracytoplasmic glucocorticoids in skeletal cells. J Steroid Biochem Mol Biol 111 164-170.

2 3

Hong D, Chen HX, Yu HQ, Wang C, Deng HT, Lian QQ & Ge RS 2011 Quantitative proteomic

4

analysis of dexamethasone-induced effects on osteoblast differentiation, proliferation, and

5

apoptosis in MC3T3-E1 cells using SILAC. Osteoporos Int 22 2175-2186.

6

Hong Y, Wu J, Zhao J, Wang H, Liu Y, Chen T, Kan X, Tao Q, Shen X & Yan K, et al. 2013

7

miR-29b and miR-29c are involved in Toll-like receptor control of glucocorticoid-induced

8

apoptosis in human plasmacytoid dendritic cells. PLoS One 8 e69926.

9

Huang Z, Chen X, Yu B, He J & Chen D 2012 MicroRNA-27a promotes myoblast proliferation by targeting myostatin. Biochem Biophys Res Commun 423 265-269.

10 11

Kondo T, Kitazawa R, Yamaguchi A & Kitazawa S 2008 Dexamethasone promotes

12

osteoclastogenesis by inhibiting osteoprotegerin through multiple levels. Journal Of Cellular

13

Biochemistry 103 335-345.

14

Liu Z, Liu H, Desai S, Schmitt DC, Zhou M, Khong HT, Klos KS, McClellan S, Fodstad O & Tan M

15

2013 miR-125b functions as a key mediator for snail-induced stem cell propagation and

16

chemoresistance. Journal Of Biological Chemistry 288 4334-4345.

17

Luo X, Xiao J, Lin H, Li B, Lu Y, Yang B & Wang Z 2007 Transcriptional activation by stimulating

18

protein 1 and post-transcriptional repression by muscle-specific microRNAs of IKs-encoding

19

genes and potential implications in regional heterogeneity of their expressions. Journal Of

20

Cellular Physiology 212 358-367.

21

Lv M, Zhang X, Jia H, Li D, Zhang B, Zhang H, Hong M, Jiang T, Jiang Q & Lu J, et al. 2012 An oncogenic role of miR-142-3p in human T-cell acute lymphoblastic leukemia (T-ALL) by

22 20

Page 21 of 36

targeting glucocorticoid receptor-alpha and cAMP/PKA pathways. Leukemia 26 769-777.

1 2

Ma W, Xie S, Ni M, Huang X, Hu S, Liu Q, Liu A, Zhang J & Zhang Y 2012 MicroRNA-29a

3

inhibited epididymal epithelial cell proliferation by targeting nuclear autoantigenic sperm

4

protein (NASP). Journal Of Biological Chemistry 287 10189-10199.

5

Nagano H, Tomimaru Y, Eguchi H, Hama N, Wada H, Kawamoto K, Kobayashi S, Mori M & Doki

6

Y 2013 MicroRNA-29a induces resistance to gemcitabine through the Wnt/Beta-catenin

7

signaling pathway in pancreatic cancer cells. International Journal Of Oncology 43 1066-1072.

8

Pereira RM, Delany AM & Canalis E 2001 Cortisol inhibits the differentiation and apoptosis of osteoblasts in culture. Bone 28 484-490.

9 10

Rathod SS, Rani SB, Khan M, Muzumdar D & Shiras A 2014 Tumor suppressive miRNA-34a

11

suppresses cell proliferation and tumor growth of glioma stem cells by targeting Akt and Wnt

12

signaling pathways. FEBS Open Bio 4 485-495.

13

Rauch A, Seitz S, Baschant U, Schilling AF, Illing A, Stride B, Kirilov M, Mandic V, Takacz A &

14

Schmidt-Ullrich R, et al. 2010 Glucocorticoids suppress bone formation by attenuating

15

osteoblast differentiation via the monomeric glucocorticoid receptor. Cell Metab 11 517-531.

16

Seibel MJ, Cooper MS & Zhou H 2013 Glucocorticoid-induced osteoporosis: mechanisms, management, and future perspectives. Lancet Diabetes Endocrinol 1 59-70.

17 18

Shi C, Qi J, Huang P, Jiang M, Zhou Q, Zhou H, Kang H, Qian N, Yang Q & Guo L, et al. 2014

19

MicroRNA-17/20a inhibits glucocorticoid-induced osteoclast differentiation and function

20

through targeting RANKL expression in osteoblast cells. Bone 68 67-75.

21

Shi XE, Li YF, Jia L, Ji HL, Song ZY, Cheng J, Wu GF, Song CC, Zhang QL & Zhu JY, et al. 2014

22

MicroRNA-199a-5p affects porcine preadipocyte proliferation and differentiation. Int J Mol Sci 21

Page 22 of 36

15 8526-8538.

1 2

Stahn C & Buttgereit F 2008 Genomic and nongenomic effects of glucocorticoids. Nat Clin Pract Rheumatol 4 525-533.

3 4

Taipaleenmaki H, Bjerre HL, Chen L, Kauppinen S & Kassem M 2012 Mechanisms in

5

endocrinology: micro-RNAs: targets for enhancing osteoblast differentiation and bone

6

formation. European Journal Of Endocrinology 166 359-371.

7

Tait AS, Butts CL & Sternberg EM 2008 The role of glucocorticoids and progestins in inflammatory, autoimmune, and infectious disease. J Leukoc Biol 84 924-931.

8 9

Walsh S, Jordan GR, Jefferiss C, Stewart K & Beresford JN 2001 High concentrations of

10

dexamethasone suppress the proliferation but not the differentiation or further maturation of

11

human osteoblast precursors in vitro: relevance to glucocorticoid-induced osteoporosis.

12

Rheumatology (Oxford) 40 74-83.

13

Wang Y, Li YP, Paulson C, Shao JZ, Zhang X, Wu M & Chen W 2014 Wnt and the Wnt signaling

14

pathway in bone development and disease. Front Biosci (Landmark Ed) 19 379-407.

15

Wei J, Shi Y, Zheng L, Zhou B, Inose H, Wang J, Guo XE, Grosschedl R & Karsenty G 2012

16

miR-34s inhibit osteoblast proliferation and differentiation in the mouse by targeting SATB2.

17

Journal Of Cell Biology 197 509-521.

18

Weinstein RS 2001 Glucocorticoid-induced osteoporosis. Rev Endocr Metab Disord 2 65-73.

19

Weinstein RS, Jilka RL, Parfitt AM & Manolagas SC 1998 Inhibition of osteoblastogenesis and

20

promotion of apoptosis of osteoblasts and osteocytes by glucocorticoids. Potential mechanisms

21

of their deleterious effects

22

on bone. Journal Of Clinical Investigation 102 274-282.

Wu D, Huang HJ, He CN & Wang KY 2013 MicroRNA-199a-3p regulates endometrial cancer cell 22

Page 23 of 36

1

proliferation by targeting mammalian target of rapamycin (mTOR). International Journal Of

2

Gynecological Cancer 23 1191-1197.

3

Xing W, Hao L, Yang X, Li F & Huo H 2014 Glucocorticoids induce apoptosis by inhibiting microRNA cluster miR-17-92 expression in chondrocytic cells. Mol Med Rep 10 881-886.

4 5

Xu N, Zhang J, Shen C, Luo Y, Xia L, Xue F & Xia Q 2012 Cisplatin-induced downregulation of

6

miR-199a-5p increases drug resistance by activating autophagy in HCC cell. Biochem Biophys

7

Res Commun 423 826-831.

8

Xu N, Zhang L, Meisgen F, Harada M, Heilborn J, Homey B, Grander D, Stahle M, Sonkoly E &

9

Pivarcsi A 2012 MicroRNA-125b down-regulates matrix metallopeptidase 13 and inhibits

10

cutaneous squamous cell carcinoma cell proliferation, migration, and invasion. Journal Of

11

Biological Chemistry 287 29899-29908.

12

Xu Z, Choudhary S, Voznesensky O, Mehrotra M, Woodard M, Hansen M, Herschman H & Pilbeam

13

C 2006 Overexpression of COX-2 in human osteosarcoma cells decreases proliferation and

14

increases apoptosis. Cancer Research 66 6657-6664.

15

Yang B, Lin H, Xiao J, Lu Y, Luo X, Li B, Zhang Y, Xu C, Bai Y & Wang H, et al. 2007 The

16

muscle-specific microRNA miR-1 regulates cardiac arrhythmogenic potential by

17

GJA1 and KCNJ2. Nature Medicine 13 486-491.

targeting

18

Zamore PD & Haley B 2005 Ribo-gnome: the big world of small RNAs. Science 309 1519-1524.

19

Zeng Y 2006 Principles of micro-RNA production and maturation. Oncogene 25 6156-6162.

20 21

23

Page 24 of 36 1

Figure legends

2

Figure 1. The inhibition of differentiating osteoblasts proliferation by Dex. A: Proliferation of MC3T3-E1

3

cells was measured by CCK8 after cells were treated with 10–9, 10–8, 10–7 M Dex from day 1 to day 5. The

4

results showed that Dex decreased the viability of MC3T3-E1 cells in a dose- and time- dependent manner.

5

n = 3, **P < 0.01. B: Representative photomicrographs of EdU staining (top panel) and corresponding total

6

cell photomicrographs (middle panel). Blue: Hoechst labeling of cell nuclei; Red: EdU labeling of nuclei of

7

proliferative cells (×400). C: Quantitative data showing the percentage of EdU-positive cells in different

8

treatment groups (number of red vs number of blue nuclei). n = 3, **P < 0.01. D: MC3T3-E1 cells were

9

stained with CFSE before plating, cultured for 5 days, and analyzed by flow cytometry as described in

10

Materials and Methods. The peak CFSE fluorescence intensity on flow cytometry was shifted by Dex. n = 3.

11

Con indicates control, Dex indicates dexamethasone.

12

Figure 2. Up-regulation of miR-199a-5p expression by Dex in differentiating osteoblasts. A: qRT-PCR

13

analysis of miR-574-3p, miR-27a, miR-29a, miR-34a, miR-125b and miR-199a-5p expression in MC3T3-

14

E1 cells treated with 10-7 M Dex for 5 days. n = 3, **P < 0.01. B: qRT-PCR analysis of miR-199a-5p

15

expression in MC3T3-E1 cells treated with 10–7 M Dex from day 1 to day 5. n = 3, **P < 0.01. C: qRT-

16

PCR analysis of miR-199a-5p expression in primary osteoblasts treated with 10–7 M Dex for 5 days. n = 3,

17

**P < 0.01. D: qRT-PCR analysis of miR-199a-5p expression in the calvarias from mice treated with Dex

18

for 3 days. n = 3, **P < 0.01. Con indicates control, Dex indicates dexamethasone, miR indicates

19

microRNA.

20

Figure 3. Repression of MC3T3-E1 cells proliferation by miR-199a-5p. A: Proliferation of MC3T3-E1 cells

21

was measured by CCK8 after cells were transfected with miR-199a-5p or/and AMO-199a-5p from day 1 to

22

day 5. n = 3, **P < 0.01. B: Representative photomicrographs of EdU staining (top panel) and

23

corresponding total cell photomicrographs (middle panel). Blue: Hochest labeling of cell nuclei; Red: EdU 1

Page 25 of 36 1

labeling of nuclei of proliferative cells (×400). C: Quantitative data showing the percentage of EdU-positive

2

cells in different treatment groups (number of red vs number of blue nuclei). n = 3, **P < 0.01. D: MC3T3-

3

E1 cells transfected with miR-199a-5p or/and AMO-199a-5p were stained with CFSE and analyzed by flow

4

cytometry as described in Materials and Methods. n = 3. E: Western blot analysis of Runx2 and Osterix

5

expression in MC3T3-E1 cells transfected with miR-199a-5p or/and AMO-199a-5p. n = 3, **P < 0.01, *P

6

< 0.05. Con indicates control, NC indicates negative control, miR indicates microRNA.

7

Figure 4. Repression of primary osteoblasts proliferation by miR-199a-5p. A: Proliferation of primary

8

osteoblasts was measured by CCK8 after cells were transfected with miR-199a-5p or/and AMO-199a-5p

9

from day 1 to day 5. n = 3, **P < 0.01. B: Quantitative data showing the percentage of EdU-positive cells in

10

different treatment groups (number of red vs number of blue nuclei). n = 3, **P < 0.01, *P < 0.05. C:

11

Primary osteoblasts transfected with miR-199a-5p or/and AMO-199a-5p were stained with CFSE and

12

analyzed by flow cytometry as described in Materials and Methods. n = 3. Con indicates control, NC

13

indicates negative control, miR indicates microRNA.

14

Figure 5. Involvement of miR-199a-5p in Dex-reduced MC3T3-E1 cells proliferation. A: Proliferation of

15

MC3T3-E1 cells was measured by CCK8 after cells were transfected with miR-199a-5p or/and AMO-199a-

16

5p under Dex from day 1 to day 5. n = 3, **P < 0.01. B: Quantitative data showing the percentage of EdU-

17

positive cells in different treatment groups (number of red vs number of blue nuclei). n = 3, **P < 0.01. C:

18

MC3T3-E1 cells transfected with miR-199a-5p or/and AMO-199a-5p under Dex were stained with CFSE

19

and analyzed by flow cytometry as described in Materials and Methods. n = 3. Con indicates control, Dex

20

indicates dexamethasone, NC indicates negative control, miR indicates microRNA.

21

Figure 6. Involvement of miR-199a-5p in Dex-reduced primary osteoblasts proliferation. A: Proliferation of

22

primary osteoblasts was measured by CCK8 after cells were transfected with miR-199a-5p or/and AMO-

23

199a-5p under Dex from day 1 to day 5. n = 3, **P < 0.01. B: Quantitative data showing the percentage of 2

Page 26 of 36 1

EdU-positive cells in different treatment groups (number of red vs number of blue nuclei). n = 3, **P

Glucocorticoid inhibits cell proliferation in differentiating osteoblasts by microRNA-199a targeting of WNT signaling.

The inhibition of osteoblast proliferation by glucocorticoids (GCs) is very important in the etiology of GC-induced osteoporosis. The mechanisms of th...
3MB Sizes 0 Downloads 5 Views