Abstract

MicroRNAs are highly conserved non-coding small RNAs participating in almost all kinds of biological activities. MiR-181a has been reported to be involved in the differentiation of porcine primary preadipocytes, but the profound effect of miR-181a-5p on 3T3-L1 adipocyte differentiation and proliferation is still unclear. In this study, we found that supplementation of miR-181a-5p in 3T3-L1 cells significantly promoted the adipogenesis and inhibited cell proliferation with increased expression of adipogenic marker genes including peroxisome proliferator-activated receptor gamma (Pparγ), CCAAT/enhancer-binding protein alpha (C/ebpα), fatty acid-binding protein 4 (Fabp4), and Adiponectin, accompanied by an accumulation of lipid droplet, an increase of triglyceride content, and a decrease of cell proliferation. Furthermore, by using the luciferase assay, Smad7 and Tcf7l2, two important members of transforming growth factor-β (TGFβ) and Wnt signaling pathway, were proven to be the direct target genes of miR-181a-5p. Moreover, supplementation of miR-181a-5p in 3T3-L1 cells altered the expressions of proteins involved in the TGFβ signaling pathway, such as TGFBR1, p-SMAD3, SMAD4, c-MYC, and p15. Taken together, these results indicate that miR-181a-5p promotes 3T3-L1 preadipocyte differentiation and adipogenesis through regulating TGFβ/Smad and Wnt signaling pathway by directly targeting Smad7 and Tcf7l2.

Introduction

In recent years, the prevalence of overweight and obesity in China has been markedly increasing based on the China Health and Nutrition Surveys [1,2]. Studies have indicated that obesity is a complex metabolic disorder that is often associated with all kinds of diseases, such as type 2 diabetes mellitus, endometrial disorder, hypertension, coronary heart disease, and hepatic steatosis, and is also an important risk factor for cancers [36]. It has been demonstrated that in a state of obesity accompanied by the gradual expansion of adipose tissue, the whole body energy balance can be destroyed, which further increases the risk of insulin resistance, hypertension, and dyslipidemia due to adipocyte hypertrophy and hyperplasia [7]. In recent years, the mouse 3T3-L1 cells are often used as a model for studying the underlying mechanism of adipogenesis, which involves cell proliferation and differentiation [8,9]. The whole process of 3T3-L1 preadipocyte differentiation includes cell growth arrest, subsequent clonal expansion, and finally terminal differentiation into mature adipocytes [10]. So, it is beneficial for improving public health to clarify the potential molecular mechanisms of adipogenesis.

MicroRNAs are highly conserved endogenous short non-coding RNAs that work in the post-transcriptional regulation of gene expression [11,12], thereby repressing the translation of target genes and/or promoting their degradation [13]. It has been reported that miRNAs play crucial roles in many physiological and pathological processes, including energy homeostasis, tumorigenesis, development, glucose metabolism, and lipid metabolism [1419]. A recent study has indicated that miRNAs may play a functional role in regulating adipocyte differentiation and proliferation. For instance, miR-26b inhibits adipocyte differentiation and promotes proliferation in human preadipocytes [20]. The miR-375/miR-21/miR-143 cluster accelerates adipocyte differentiation in 3T3-L1 cells [2123]. On the contrary, Let-7 and miR-27 impair adipogenesis [24,25]. In addition, miR-181a has been proven to regulate adipogenesis in the porcine model [26]. Therefore, miR-181a is one of the most representative miRNAs that regulates differentiation. However, the relationships between miR-181a-5p and the potential mechanisms of 3T3-L1 cell proliferation and differentiation remain unclear.

In our previous research, with the microarray strategy we found that miR-181a-5p is possibly a positive regulator during the chicken adipose tissue development [27]. However, to date, the role of miR-181a-5p in adipogenesis is still unknown.

The 3T3-L1 cell line has widely been used as an adipogenesis model and many signaling pathways have been proven to participate in 3T3-L1 cell differentiation, while the canonical Wnt signaling is a negative regulator during adipogenesis. Tumor necrosis factor-alpha inhibits adipogenesis via activating β-catenin/TCF4 (TCF7L2)-dependent signaling pathway [28]. Meanwhile, SMAD7 is a member of transforming growth factor-β (TGFβ)/Smad signaling pathway, which antagonizes the TGFβ-mediated response. Increased SMAD7 level blocks the differentiation of 3T3-F442A cells [29].

In this study, we identified whether miR-181a-5p can regulate the differentiation and proliferation of 3T3-L1 cells. Our results indicated that miR-181a-5p is an important positive regulator of adipogenesis by targeting Tcf7l2 and Smad7. miR-181a-5p is involved in the differentiation and proliferation of 3T3-L1 cells, which may be related to the TGFβ/Smad signaling pathway. The decrease of Tcf7l2 expression may promote 3T3-L1 cell differentiation by inhibiting Wnt/β-catenin signaling pathway. This study provides more insights into the mechanism of 3T3-L1 cell differentiation.

Materials and Methods

Cell culture and differentiation

3T3-L1 preadipocytes were cultured in DMEM (Gibco, Carlsbad, USA) supplemented with 10% fetal bovine serum (FBS; Gibco) and 1% penicillin/streptomycin (Gibco) in a 5% CO2 atmosphere at 37°C. To induce the differentiation of 3T3-L1 preadipocytes, confluent preadipocytes were pre-treated for 2 days in differentiation medium: DMEM containing 10% FBS and MDI (8.61 × 10−7 M insulin, 1 × 10−6 M dexamethasone, and 5 × 10−4 M IBMX). Then, cells were washed with phosphate buffered saline (PBS) and were further cultured in DMEM containing 10% FBS and 8.61 × 10−7 M insulin (designated as Day 0).

Oil Red O staining and triglyceride content determination

According to the methods described previously [27], cells were fixed with 4% paraformaldehyde for 30 min. Then, the samples were washed twice with deionized water, and incubated with filtered Oil Red O (Sigma, St Louis, USA) working solution (60% Oil Red O stock solution and 40% deionized water) for 1 h. The stained cells were washed with isopropanol and the accumulated triglyceride was qualified by measuring the absorbance at 510 nm.

Transfection assays

The synthetic miR-181a-5p mimics (50 nM) and miRNA negative control (miR-NC) (RiboBio, Guangzhou, China) were transfected into 3T3-L1 cells by using Lipofectamine 2000 (Invitrogen, Carlsbad, USA) according to the manufacturer's instructions.

RT-PCR and quantitative real-time PCR

Total RNA was extracted from 3T3-L1 preadipocytes using Trizol reagent (Takara, Otsu, Japan) according to the manufacturer's instructions. cDNA was synthesized by using ReverTra Ace PCR RT Master Mix with gDNA Remover (FSQ-301) (Toyobo, Osaka, Japan) and ReverTra Ace PCR RT Kit (Toyobo) for RT-PCR according to the manufacturer's protocol, respectively. In RT-PCR analysis, the mRNA levels of peroxisome proliferator-activated receptor gamma (Pparγ), CCAAT/enhancer-binding protein alpha (C/ebpα), fatty acid-binding protein 4 (Fabp4), Adiponectin, Cdk4, Cdk6, CyclinD1, CyclinD2, CyclinD3, p21, and p53 were quantified using SYBR Green Supermix (Takara) with β-actin as a housekeeping gene for normalization. MiR-181a-5p level was measured by the stem-loop qPCR method [30], and U6 small nuclear RNA was used as an internal control. Data were calculated using the 2−ΔΔCT method. The sequences of all primers are listed in Table 1.

Table 1.

Sequence of primers used in PCR

GeneForward primer (5′→3′)Reverse primer (5′→3′)
PparγAAGAGCTGACCCAATGGTTGACCCTTGCATCCTTCACAAG
C/ebpαCGCAAGAGCCGAGATAAAGCCGGTCATTGTCACTGGTCAACT
AdipoqACGACCAGTATCAGGAAAAGGGTAGAGAAGAAAGCCAGTAA
Fabp4TAAAAACACCGAGATTTCCTTCACCTTTCATAACACATTCCACCA
β-actinCATCCGTAAAGACCTCTATGCCAACATGGAGCCACCGATCCACA
p21CGAGAACGGTGGAACTTTGACCAGGGCTCAGGTAGACCTTG
p53TTACCAGGGCAACTATGGCTTCCCAACTGCACAGGGCACGTCTT
CyclinD1ACCTCTGGCTCTGTGCCTTTCTATGTCCCACCTTCACCTCTTCCCT
CyclinD2GAGTGGGAACTGGTAGTGTTGGGTACATGGCAAACTTGAAGTCGGTAGC
CyclinD3AGATCAAGCCGCACATATCCAGGTAGTTCATAGCC
Cdk4CTACATACGCAACACCCGTCAAAGATTTTCCCCAACT
Cdk6CCTCTGGAGTGTCGGTTGCCTGGGAGTCCAATGATGTCC
Tcf7l2 3UTRCGAGCTCTAGCAGACTGTCCAGAAAGCATGCGTCGACTAGGGAGACATTCAAAGTATTCAC
GeneForward primer (5′→3′)Reverse primer (5′→3′)
PparγAAGAGCTGACCCAATGGTTGACCCTTGCATCCTTCACAAG
C/ebpαCGCAAGAGCCGAGATAAAGCCGGTCATTGTCACTGGTCAACT
AdipoqACGACCAGTATCAGGAAAAGGGTAGAGAAGAAAGCCAGTAA
Fabp4TAAAAACACCGAGATTTCCTTCACCTTTCATAACACATTCCACCA
β-actinCATCCGTAAAGACCTCTATGCCAACATGGAGCCACCGATCCACA
p21CGAGAACGGTGGAACTTTGACCAGGGCTCAGGTAGACCTTG
p53TTACCAGGGCAACTATGGCTTCCCAACTGCACAGGGCACGTCTT
CyclinD1ACCTCTGGCTCTGTGCCTTTCTATGTCCCACCTTCACCTCTTCCCT
CyclinD2GAGTGGGAACTGGTAGTGTTGGGTACATGGCAAACTTGAAGTCGGTAGC
CyclinD3AGATCAAGCCGCACATATCCAGGTAGTTCATAGCC
Cdk4CTACATACGCAACACCCGTCAAAGATTTTCCCCAACT
Cdk6CCTCTGGAGTGTCGGTTGCCTGGGAGTCCAATGATGTCC
Tcf7l2 3UTRCGAGCTCTAGCAGACTGTCCAGAAAGCATGCGTCGACTAGGGAGACATTCAAAGTATTCAC
Table 1.

Sequence of primers used in PCR

GeneForward primer (5′→3′)Reverse primer (5′→3′)
PparγAAGAGCTGACCCAATGGTTGACCCTTGCATCCTTCACAAG
C/ebpαCGCAAGAGCCGAGATAAAGCCGGTCATTGTCACTGGTCAACT
AdipoqACGACCAGTATCAGGAAAAGGGTAGAGAAGAAAGCCAGTAA
Fabp4TAAAAACACCGAGATTTCCTTCACCTTTCATAACACATTCCACCA
β-actinCATCCGTAAAGACCTCTATGCCAACATGGAGCCACCGATCCACA
p21CGAGAACGGTGGAACTTTGACCAGGGCTCAGGTAGACCTTG
p53TTACCAGGGCAACTATGGCTTCCCAACTGCACAGGGCACGTCTT
CyclinD1ACCTCTGGCTCTGTGCCTTTCTATGTCCCACCTTCACCTCTTCCCT
CyclinD2GAGTGGGAACTGGTAGTGTTGGGTACATGGCAAACTTGAAGTCGGTAGC
CyclinD3AGATCAAGCCGCACATATCCAGGTAGTTCATAGCC
Cdk4CTACATACGCAACACCCGTCAAAGATTTTCCCCAACT
Cdk6CCTCTGGAGTGTCGGTTGCCTGGGAGTCCAATGATGTCC
Tcf7l2 3UTRCGAGCTCTAGCAGACTGTCCAGAAAGCATGCGTCGACTAGGGAGACATTCAAAGTATTCAC
GeneForward primer (5′→3′)Reverse primer (5′→3′)
PparγAAGAGCTGACCCAATGGTTGACCCTTGCATCCTTCACAAG
C/ebpαCGCAAGAGCCGAGATAAAGCCGGTCATTGTCACTGGTCAACT
AdipoqACGACCAGTATCAGGAAAAGGGTAGAGAAGAAAGCCAGTAA
Fabp4TAAAAACACCGAGATTTCCTTCACCTTTCATAACACATTCCACCA
β-actinCATCCGTAAAGACCTCTATGCCAACATGGAGCCACCGATCCACA
p21CGAGAACGGTGGAACTTTGACCAGGGCTCAGGTAGACCTTG
p53TTACCAGGGCAACTATGGCTTCCCAACTGCACAGGGCACGTCTT
CyclinD1ACCTCTGGCTCTGTGCCTTTCTATGTCCCACCTTCACCTCTTCCCT
CyclinD2GAGTGGGAACTGGTAGTGTTGGGTACATGGCAAACTTGAAGTCGGTAGC
CyclinD3AGATCAAGCCGCACATATCCAGGTAGTTCATAGCC
Cdk4CTACATACGCAACACCCGTCAAAGATTTTCCCCAACT
Cdk6CCTCTGGAGTGTCGGTTGCCTGGGAGTCCAATGATGTCC
Tcf7l2 3UTRCGAGCTCTAGCAGACTGTCCAGAAAGCATGCGTCGACTAGGGAGACATTCAAAGTATTCAC

Western blot analysis

3T3-L1 preadipocytes were washed three times with ice-cold PBS, and then lysed with a RIPA buffer. The supernatants were collected by centrifuging at 12,000 g for 10 min at 4°C. Protein concentrations from cell lysates were measured by BCA kit (Beyotime, Haimen, China). Proteins were separated by 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and transferred onto PVDF membranes. The membranes were blocked for 1 h with 5% milk containing 0.1% Tween-20. Then, PVDF membranes were incubated with primary antibodies (all from Santa Cruz Biotech, Santa Cruz, USA) against PPARγ, C/EBPα, p-SMAD3, SMAD3, SMAD4, SMAD7, TLF7L2, c-MYC, P15, CDK4, CyclinD2, and Tubulin. Immune complexes were further incubated with specific horseradish peroxidase-conjugated secondary antibodies (Santa Cruz Biotech), and the bands were visualized with BeyoECL Plus kit (Beyotime).

Cell viability assay

Cells were seeded into 96-well plates at 5 × 103 cells/well and then were transfected with miR-181a-5p mimics or NC. At 24 and 48 h after transfection, cell proliferation was determined using the CCK-8 kit (Beyotime) according to the manufacturer's instructions.

Luciferase vector construction and reporter assays

The mouse Tcf7l2 was predicted to be the target of miR-181a-5p by using TargetScan and miRDB. The wild-type and mutant 3′UTR (RiboBio) of Tcf7l2 mRNA were amplified from mouse cDNA with the primer containing the recognition sites of XhoI and SalI listed in Table 1. The PCR product was inserted into the corresponding sites of pGL3-control (Promega, Madison, USA) to obtain the plasmid pGL3-Tcf7l2-UTR. For luciferase reporter assay, 3T3-L1 cells were co-transfected with miR-181a-5p mimics or miR-NC and the 3′UTR of pGL3-Wt-Tcf7l2 or pGL3-Mut-Tcf7l2 by using Lipofectamine 2000. After 48 h transfection, cells were collected and the luciferase activity was measured by using the Dual Luciferase Reporter Assay Kit (Promega).

Flow-cytometry analysis

Freshly prepared 3T3-L1 cells were digested with 0.25% trypsin (Gibco) and collected in PBS. Then, cells were fixed with 70% cold ethanol overnight at 4°C. Cells were washed with PBS twice and then stained with PI on ice. The cell suspensions were analyzed by flow cytometry immediately. The proportion of G0/G1, S, and G2/M phases was calculated from DNA histogram data [31].

Statistical analysis

All the experimental results were presented as the mean ± SEM. Data were analyzed using independent t-test. Differences were regarded as significant at a value of P < 0.05.

Results

The expression of miR-181a-5p during 3T3-L1 preadipocyte differentiation

To investigate the potential role of miR-181a-5p during the differentiation of 3T3-L1 preadipocytes into mature adipocytes, the expression level of miR-181a-5p mRNA was assessed by real-time PCR on Days 0, 2, 4, and 8 of differentiation. The result indicated that miR-181a-5p was gradually upregulated during 3T3-L1 preadipocyte differentiation (Fig. 1). So, we speculate that miR-181a-5p may have important function in 3T3-L1 preadipocyte differentiation.
The expression of miR-181a-5p during 3T3-L1 preadipocyte differentiation 3T3-L1 preadipocytes were induced into differentiation as described in Materials and Methods. MiR-181a-5p was detected during 3T3-L1 cell differentiation at the designated time points by using real-time qPCR. n = 3. *P < 0.05, **P < 0.01.
Figure 1.

The expression of miR-181a-5p during 3T3-L1 preadipocyte differentiation 3T3-L1 preadipocytes were induced into differentiation as described in Materials and Methods. MiR-181a-5p was detected during 3T3-L1 cell differentiation at the designated time points by using real-time qPCR. n = 3. *P < 0.05, **P < 0.01.

miR-181a-5p overexpression induces the 3T3-L1 preadipocyte differentiation

To assess whether miR-181a-5p affects 3T3-L1 preadipocyte differentiation, we transfected miR-181a-5p mimics and miR-NC into 3T3-L1 cells. At 24 h after transfection, 3T3-L1 preadipocytes were induced to adipogenic differentiation with differentiation medium. Real-time qPCR assay showed that miR-181a-5p level was markedly increased in miR-181a-5p mimics group compared with the NC group at 24 h (Fig. 2A). MiR-181a-5p overexpression significantly promoted 3T3-L1 preadipocyte differentiation, which was confirmed by Oil Red O staining and triglyceride content measurement at 510 nm (Fig. 2B,C). On Day 8, compared with NC group, the expressions of Pparγ, C/ebpα, Fabp4, and Adipoq mRNA were markedly upregulated in miR-181a-5p mimics group during 3T3-L1 cell differentiation (Fig. 2D). Furthermore, the expression levels of PPARγ and C/EBPα protein were also increased (Fig. 2E). These data indicate that miR-181a-5p has a positive effect on 3T3-L1 preadipocyte differentiation.
MiR-181a-5p promotes 3T3-L1 preadipocyte differentiation (A) MiR-181a-5p mimics or miRNA negative control (miR-NC) was transfected into 3T3-L1 cells, and at 24 h after transfection, miR-181a-5p transfection efficiency was confirmed by qPCR. (B) At 24 h post-transfection, the differentiation of 3T3-L1 preadipocytes was initiated. On Day 8 after differentiation, cells were fixed and stained with Oil Red O. (C) Triglyceride content was measured at 510 nm after extracting Oil Red O. (D) The relative mRNA expressions of adipocyte-specific molecular markers peroxisome proliferator-activated receptor gamma (Pparγ), CCAAT/enhancer-binding protein alpha (C/ebpα), fatty acid-binding protein 4 (Fabp4), and Adipoq were analyzed on Day 8 after the stimulation of differentiation. (E) The protein levels of PPARγ and C/EBPα were measured by western blotting. Scale bar, 100 μm. n = 3. *P < 0.05, **P <0.01.
Figure 2.

MiR-181a-5p promotes 3T3-L1 preadipocyte differentiation (A) MiR-181a-5p mimics or miRNA negative control (miR-NC) was transfected into 3T3-L1 cells, and at 24 h after transfection, miR-181a-5p transfection efficiency was confirmed by qPCR. (B) At 24 h post-transfection, the differentiation of 3T3-L1 preadipocytes was initiated. On Day 8 after differentiation, cells were fixed and stained with Oil Red O. (C) Triglyceride content was measured at 510 nm after extracting Oil Red O. (D) The relative mRNA expressions of adipocyte-specific molecular markers peroxisome proliferator-activated receptor gamma (Pparγ), CCAAT/enhancer-binding protein alpha (C/ebpα), fatty acid-binding protein 4 (Fabp4), and Adipoq were analyzed on Day 8 after the stimulation of differentiation. (E) The protein levels of PPARγ and C/EBPα were measured by western blotting. Scale bar, 100 μm. n = 3. *P < 0.05, **P <0.01.

miR-181a-5p overexpression inhibits proliferation of 3T3-L1 cells by inducing G1-phase cell-cycle arrest

Adipogenesis is involved in cell proliferation and differentiation. Meanwhile, the regulation of cell cycle plays an important role in cell proliferation [32]. In order to investigate the regulatory effect of miR-181a-5p on 3T3-L1 cell proliferation, miR-181a-5p mimics and miR-NC were successfully transfected into the 3T3-L1 cells (Fig. 2A). At 24 and 48 h after transfection, cell proliferation was assessed by CCK-8 assay and cell cycle was analyzed by flow cytometry. The results showed that miR-181a-5p overexpression significantly inhibited the proliferation of 3T3-L1 cells compared with NC (Fig. 3A). As shown in Fig. 3B,C, the miR-181a-5p group had a marked increase in G1-phase population compared with in the NC group. To explore the underlying mechanisms of miR-181a-5p in the cell-cycle arrest of 3T3-L1 preadipocytes, the mRNA and protein levels of G1 phase-related genes including CyclinD1, CyclinD2, CyclinD3, CDK4, CDK6, p21, and p53 were detected. Results from qRT-PCR showed that supplementation of miR-181a-5p significantly suppressed the mRNA expressions of CyclinD2, CyclinD3, and Cdk4, but there was no significant difference in CyclinD1, Cdk6, p21, and p53 (Fig. 3D). Western blot analysis showed that CyclinD2 and CDK4 expressions were downregulated and the expression of P15 was upregulated (Fig. 3E). These results indicate that miR-181a-5p inhibits cell proliferation as a result of G1-phase cell-cycle arrest.
MiR-181a-5p inhibits proliferation of 3T3-L1 cells by regulating the cell cycle (A) 3T3-L1 cells were transfected with miR-181a-5p mimics or miRNA negative control  (miR-NC). After transfection, cell proliferation was monitored at indicated time points. (B) After transfection, cells were harvested and determined the cell cycle by flow cytometry. (C) After transfection, cell cycle was analyzed. (D) At 48 h post-transfection, mRNA expression levels of cell G1 phase-related genes CyclinD1, CyclinD2, CyclinD3, Cdk4, Cdk6, p21, and p53 were analyzed by qRT-PCR. (E) Western blot analysis showed that miR-181a-5p mimics downregulated the protein expression levels of CyclinD2 and CDK4 and upregulated the protein expression level of P15. Data are presented as the mean ± SD. n = 3. *P < 0.05, **P < 0.01.
Figure 3.

MiR-181a-5p inhibits proliferation of 3T3-L1 cells by regulating the cell cycle (A) 3T3-L1 cells were transfected with miR-181a-5p mimics or miRNA negative control  (miR-NC). After transfection, cell proliferation was monitored at indicated time points. (B) After transfection, cells were harvested and determined the cell cycle by flow cytometry. (C) After transfection, cell cycle was analyzed. (D) At 48 h post-transfection, mRNA expression levels of cell G1 phase-related genes CyclinD1, CyclinD2, CyclinD3, Cdk4, Cdk6, p21, and p53 were analyzed by qRT-PCR. (E) Western blot analysis showed that miR-181a-5p mimics downregulated the protein expression levels of CyclinD2 and CDK4 and upregulated the protein expression level of P15. Data are presented as the mean ± SD. n = 3. *P < 0.05, **P < 0.01.

Tcf7l2 and Smad7 are the target genes of miR-181a-5p

In order to understand the underlying mechanism of miR-181a-5p in regulating preadipocyte differentiation, target prediction software TargetScan and miRDB were used to predict the potential target genes of miR-181a-5p. Prediction results revealed that Tcf7l2 and Smad7 may be direct target genes of miR-181a-5p. As shown in Fig. 4A, at 48 h after transfection, miR-181a-5p overexpression significantly reduced the expressions of TCF7L2 and SMAD7 protein in 3T3-L1 cells. According to a previous report, Smad7 is a direct target of miR-181a, meanwhile miR-181a increases the epithelial–mesenchymal transition of ovarian cancer through the repression of Smad7 expression [33]. To further confirm that Tcf7l2 is a target gene of miR-181a, we synthesized wild-type 3′UTR of mouse Tcf7l2-containing putative binding site for miR-181a-5p and the mutant 3′UTR with a 3-bp mutation in the seed region (Fig. 4B). The pGL3-Wt-Tcf7l2 and pGL3-Mut-Tcf7l2 luciferase reporter vectors including the binding site of miR-181a-5p were respectively co-transfected with miR-NC or miR-181a-5p mimics into 3T3-L1 cells. The results suggested that the miR-181a-5p overexpression significantly decreased the luciferase activity of the pGL3-Wt-Tcf7l2 when compared with pGL3-Mut-Tcf7l2, indicating that miR-181a-5p directly targeted Tcf7l2 gene by binding to the 3′UTR (Fig. 4C). These data indicate that miR-181a-5p promotes 3T3-L1 cell adipogenesis by targeting Smad7 [33] and Tcf7l2.
Tcf7l2 and Smad7 are direct target genes of miR-181a-5p in 3T3-L1 cells (A) Western blotting was performed to detect the protein level of TCF7L2 and SMAD7 at 48 h after transfection with miR-181a-5p mimics or miRNA negative control (miR-NC). (B) TargetScan analysis showed that Tcf7l2 has the potential binding sites of miR-181a-5p in mouse. The mutated sites of 3′UTR of Tcf7l2 were generated. (C) Wild-type or mutant Tcf7l2 3′UTR reporter constructs were transfected together with miR-181a-5p mimics or miR-NC into 3T3-L1 cells and then the relative luciferase activities were measured. Data are presented as the mean ± SD. n = 3. *P < 0.05.
Figure 4.

Tcf7l2 and Smad7 are direct target genes of miR-181a-5p in 3T3-L1 cells (A) Western blotting was performed to detect the protein level of TCF7L2 and SMAD7 at 48 h after transfection with miR-181a-5p mimics or miRNA negative control (miR-NC). (B) TargetScan analysis showed that Tcf7l2 has the potential binding sites of miR-181a-5p in mouse. The mutated sites of 3′UTR of Tcf7l2 were generated. (C) Wild-type or mutant Tcf7l2 3′UTR reporter constructs were transfected together with miR-181a-5p mimics or miR-NC into 3T3-L1 cells and then the relative luciferase activities were measured. Data are presented as the mean ± SD. n = 3. *P < 0.05.

miR-181a-5p inhibits the TGFβ/Smad signaling pathway

In the TGFβ signaling pathway, SMAD7 inhibits the SMAD2/SMAD3/SMAD4 nuclear translocation by preventing their interaction. Meanwhile, TGFβ signaling pathway may be a negative regulator of adipogenesis [34]. To test whether miR-181a-5p regulates 3T3-L1 preadipocyte adipogenesis by influencing the downstream genes of Smad7 in the TGFβ signaling pathway, western blot analysis was performed to detect the expressions of p-SMAD3, SMAD3, SMAD4, and c-MYC at the protein level in TGFβ/Smad signaling pathway at 48 h after transfection with miR-181a-5p mimics and miR-NC. The expression of p-SMAD3 protein was upregulated in 3T3-L1 cells transfected with miR-181a-5p mimics compared with miR-NC groups, while the expression of SMAD3 was not affected (Fig. 5). On the contrary, miR-181a-5p overexpression significantly reduced the protein expression of SMAD4 and c-MYC (Fig. 5). These results suggest that miR-181a-5p overexpression regulates 3T3-L1 cell differentiation by influencing the protein expressions of p-SMAD3, SMAD4, and c-MYC in TGFβ/Smad signaling pathway probably by targeting Smad7.
MiR-181a-5p regulates 3T3-L1 preadipocyte adipogenesis by influencing the downstream of target gene Smad7 in the TGFβ signaling pathway The protein expressions of p-SMAD3, SMAD3, SMAD4, and c-MYC, which are the downstream genes of target gene Smad7 in the TGFβ signaling pathway, were detected after transfection with miR-181a-5p mimics and miRNA negative control  (miR-NC) at 48 h.
Figure 5.

MiR-181a-5p regulates 3T3-L1 preadipocyte adipogenesis by influencing the downstream of target gene Smad7 in the TGFβ signaling pathway The protein expressions of p-SMAD3, SMAD3, SMAD4, and c-MYC, which are the downstream genes of target gene Smad7 in the TGFβ signaling pathway, were detected after transfection with miR-181a-5p mimics and miRNA negative control  (miR-NC) at 48 h.

Discussion

In this study, we found that miR-181a-5p was gradually upregulated during the whole process of 3T3-L1 preadipocyte adipogenesis and that supplementation of miR-181a-5p significantly upregulated the mRNA expression of the adipogenic marker genes, such as Pparγ, C/ebpα, Fabp4, and Adiponectin, as well as the protein expressions of PPARγ and C/EBPα in 3T3-L1 preadipocytes. Our results indicate that miR-181a-5p may play a crucial role in 3T3-L1 preadipocyte adipogenesis. Recently, several reports have shown that miRNAs may directly or indirectly modulate various biological processes, such as adipocyte differentiation, cell proliferation, and tissue development [35,36]. For instance, the miR-17-92 cluster, miR-378a-3p, and miR-210 accelerate adipocyte differentiation [3739]. In contrast, miR-138 and miR-155 inhibit adipogenic differentiation [40,41]. Meanwhile, it has been shown that miR-184 and miR-106 may be related to adipogenesis by regulating cell proliferation and cell-cycle arrest [42,43]. Most importantly, it has been reported that miR-181a can regulate adipogenesis in the porcine model [26]. All these findings imply that miR-181a-5p may serve as an important regulator during 3T3-L1 preadipocyte differentiation. In 3T3-L1 preadipocytes, due to the inverse correlation between differentiation and proliferation during the whole process of adipogenesis, we suppose that miR-181a-5p plays a negative role in 3T3-L1 cell proliferation. Our results showed that the transfection of miR-181a-5p mimics leads to cell-cycle arrest and significantly inhibits proliferation of 3T3-L1 cells. MiR-200a was significantly downregulated in hepatocellular carcinoma (HCC) cells, because miR-200a was found to inhibit the proliferation of HCC cells by induction of G1-phase arrest and CDK6 is a novel functional target of miR-200a [44]. Let-7c can inhibit HCC proliferation and induce cell-cycle arrest by directly targeting the cell-cycle regulator CDC25A [45]. TGFβ was found to increase the level of miR-424(322)/503 and reduce the CDC25A expression during cell-cycle arrest in mammary epithelial cells [46]. In human renal cell carcinoma, miR-99a inhibits the tumorigenicity by induction of G1-phase cell-cycle arrest [47]. These data suggest that some miRNAs may inhibit cell proliferation by inducing cell-cycle arrest.

Here, miR-181a-5p was identified as a novel miRNA that plays a role in 3T3-L1 preadipocyte differentiation and proliferation by targeting the Smad7 gene in the TGFβ/Smad signaling pathway and Tcf7l2 gene in the Wnt signaling pathway. MiRNAs have been demonstrated to regulate 3T3-L1 cell adipogenesis and related gene expression by targeting the TGFβ and Wnt signaling pathways [48].

Many previous studies indicated that miR-181a-5p played important roles in the breast cancer metastasis, the differentiation of osteoblasts, and the local immune balance by regulating the TGFβ signaling pathway [4951]. Moreover, miR-181a regulates TGFβ signaling pathway by directly targeting Smad7 gene to promote epithelial–mesenchymal transition [33], which plays a crucial role in the process of adipogenesis. This indicates that TGFβ signaling plays an important negative role in adipocyte differentiation [29]. SMAD7, a crucial member of the TGFβ signaling pathway, plays an important role in the regulation of adipocyte differentiation. TGFβ phosphorylates the SMAD2/SMAD3 by binding and activating the TGFβR1 and TGFβR2, which belong to transmembrane serine/threonine kinase receptors. The phosphorylated SMAD2/SMAD3 subsequently form complexes with SMAD4, and are finally translocated into the nucleus to regulate gene expression [52,53]. Our results demonstrated that downregulation of target gene Smad7 induces the phosphorylation of SMAD3, but suppresses the expression of SMAD4. Thereby, the downregulation of complexes composed of phosphorylated SMAD2/SMAD3 and SMAD4 inhibits TGFβ signaling pathway. Suppressed TGFβ may promote adipogenesis and induce C/EBP transactivation function to increase their transcriptional activity on the promoters of key adipogenic genes [54]. It has been reported that TGFβ promotes cell proliferation of human embryonic palatal mesenchymal through activating both Smad4 and c-myc genes during palatogenesis. SMAD4 induces c-myc gene activity through combination with Smad binding element in the c-myc gene promoter [55]. In androgen receptor (AR)-negative prostate cancer, androgen induced 1 (PMEPA1), a prostate transmembrane protein, may promote AR-negative prostate cancer cell proliferation by suppressing SMAD3 phosphorylation and nuclear translocation, blocking the formation of SMAD3/SMAD4 complex, upregulating c-MYC, and downregulating P21 [56]. In addition, in the absence of TGFβ signaling, SMAD4 cooperates with lymphoid enhancer-binding factor 1/T cell-specific factor (LEF/TCF) to activate c-MYC expression in HepG2 cells [57]. So, SMAD4 promotes the expression of c-MYC by binding to its promoter in TGFβ signaling pathway. Our results also indicated that when miR-181a-5p inhibits the activity of SMAD7, downregulated SMAD4 suppresses the expression of c-MYC. Then, downregulated c-MYC has the capacity to inhibit cell proliferation after induction of p15INK4, which can inhibit CyclinD1−CDK4 interaction and then cause cell-cycle arrest [5860]. Our results also showed that suppressed c-MYC may reduce the expression of CyclinD2/Cdk4 complexe to cause G1-phase cell-cycle arrest by promoting the expression of p15INK4 in TGF-β signaling pathway in 3T3-L1 cells. Previous studies also demonstrated that overexpression of c-myc gene could block the differentiation of mouse cells and inhibit the differentiation of 3T3-L1 cells by interrupting the entry of cells into a distinct predifferentiation stage in G0/G1 [61,62].

Tcf7l2 is a key regulator of Wnt signaling pathway whose function was demonstrated to suppress adipogenesis [63]. As a real target gene of miR-181a-5p, Tcf7l2 was suppressed by supplementation of miR-181a-5p in 3T3-L1 cells. Downregulation of Tcf7l2 can inactivate the transcription of Wnt-regulated genes by binding with β-catenin, which augments the expressions of C/EBPα and PPARγ to promote adipogenesis [64]. In the nucleus, TCF/β-catenin complexes can activate c-myc expression in the Wnt signaling pathway [65]. Therefore, downregulation of Tcf7l2 may inhibit the protein expression of c-myc in the Wnt signaling pathway, and then promote 3T3-L1 preadipocyte differentiation.

In summary, the present study suggests that miR-181a-5p regulates the differentiation and proliferation of 3T3-L1 by targeting the TGFβ and Wnt signaling pathways. Our study indicates that miR-181a-5p may be a potential target for ameliorating obesity, adipocyte hypertrophy, and hyperplasia.

Funding

This work was supported by a grant from the National Natural Science Foundation of China (No. 31272519).

References

1

Dearth-Wesley
T
,
Wang
H
,
Popkin
BM
.
Under- and overnutrition dynamics in Chinese children and adults (1991–2004)
.
Eur J Clin Nutr
2008
,
62
:
1302
1307
.

2

Xi
B
,
Liang
Y
,
He
T
,
Reilly
KH
,
Hu
Y
,
Wang
Q
,
Yan
Y
, et al. .
Secular trends in the prevalence of general and abdominal obesity among Chinese adults
.
Obes Rev
2012
,
13
:
287
296
.

3

Jelcić
J
,
Korsić
M
.
Obesity as a medical and public health problem
.
Lijec Vjesn
2009
,
131
:
279
185
.

4

Mokdad
AH
,
Ford
ES
,
Bowman
BA
,
Dietz
WH
,
Vinicor
F
,
Bales
VS
,
Marks
JS
.
Prevalence of obesity, diabetes, and obesity-related health risk factors, 2001
.
JAMA
2003
,
289
:
76
79
.

5

Huang
Z
,
Hankinson
SE
,
Colditz
GA
,
Stampfer
MJ
,
Hunter
DJ
,
Manson
JE
,
Hennekens
CH
, et al. .
Dual effects of weight and weight gain on breast cancer risk
.
JAMA
1997
,
278
:
1407
1411
.

6

Calle
EE
,
Rodriguez
C
,
Walker-Thurmond
K
,
Thun
MJ
.
Overweight, obesity, and mortality form cancer in a prospectively studied cohort of U.S. adults
.
N Engl J Med
2003
,
348
:
1625
.

7

Song
YH
,
Ka
S
,
Kim
AY
,
Kim
JB
.
Regulation of adipocyte differentiation via microRNAs
.
Endocrinol Metab
2014
,
29
:
122
135
.

8

Wang
N
,
Wang
X
,
Shi
M
,
Shi
H
,
Yan
X
,
Li
H
,
Wang
S
, et al. .
LMO4 modulates proliferation and differentiation of 3T3-L preadipocytes
.
FEBS Lett
2013
,
587
:
3032
3207
.

9

MacDougald
OA
,
Lane
MD
.
Transcriptional regulation of gene expression during adipocyte differentiation
.
Annu Rev Biochem
1995
,
64
:
345
373
.

10

Tang
QQ
,
Otto
TC
,
Lane
MD
.
Mitotic clonal expansion: a synchronous process required for adipogenesis
.
Proc Natl Acad Sci U S A
2003
,
100
:
44
49
.

11

Graves
P
,
Zeng
Y
.
Biogenesis of mammalian microRNAs: a global view
.
Genomics Proteomics Bioinformatics
2012
,
10
:
239
245
.

12

He
L
,
Hannon
GJ
.
MicroRNAs: small RNAs with a big role in gene regulation
.
Nat Rev Genet
2004
,
5
:
522
531
.

13

Eulalio
A
,
Huntzinger
E
,
Izaurralde
E
.
Getting to the root of miRNA-mediated gene silencing
.
Cell
2008
,
132
:
9
14
.

14

Teleman
AA
,
Cohen
SM
.
Drosophila lacking microRNA miR-278 are defective in energy homeostasis
.
Genes Dev
2006
,
20
:
417
422
.

15

Kloosterman
WP
,
Plasterk
RH
.
The diverse functions of microRNAs in animal development and disease
.
Dev Cell
2006
,
11
:
441
450
.

16

Stefani
G
,
Slack
F
.
Small non-coding RNAs in animal development
.
Nat Rev Mol Cell Biol
2008
,
9
:
219
230
.

17

Gauthier
BR
,
Wollheim
CB
.
MicroRNAs: ‘ribo-regulators’ of glucose homeostasis
.
Nat Med
2006
,
12
:
36
38
.

18

Poy
MN
,
Spranger
M
,
Stoffel
M
.
MicroRNAs and the regulation of glucose and lipid metabolism
.
Diabetes Obes Metab
2007
,
9
:
67
73
.

19

Krützfeldt
J
,
Stoffel
M
.
MicroRNAs: a new class of regulatory genes affecting metabolism
.
Cell Metab
2006
,
4
:
9
12
.

20

Song
G
,
Xu
G
,
Ji
C
,
Shi
C
,
Shen
Y
,
Chen
L
,
Zhu
L
, et al. .
The role of microRNA-26b in human adipocyte differentiation and proliferation
.
Gene
2014
,
533
:
481
487
.

21

Ling
HY
,
Wen
GB
,
Feng
SD
,
Tuo
QH
,
Ou
HS
,
Yao
CH
,
Zhu
BY
, et al. .
MiRNA-375 promotes 3T3-L1 adipocyte differentiation via modulation of ERK signaling
.
Clin Exp Pharmacol Physiol
2011
,
38
:
239
246
.

22

Kang
M
,
Yan
LM
,
Zhang
WY
,
Li
YM
,
Tang
AZ
,
Ou
HS
.
Role of microRNA-21 in regulating 3T3-L1 adipocyte differentiation and adiponectin expression
.
Mol Biol Rep
2013
,
40
:
5027
5034
.

23

Esau
C
,
Kang
X
,
Peralta
E
,
Hanson
E
,
Marcusson
EG
,
Ravichandran
LV
,
Sun
Y
, et al. .
MicroRNA-143 regulates adipocyte differentiation
.
J Biol Chem
2004
,
279
:
52361
52365
.

24

Sun
T
,
Fu
M
,
Bookout
AL
,
Kliewer
SA
,
Mangelsdorf
DJ
.
MicroRNA let-7 regulates 3T3-L1 adipogenesis
.
Mol Endocrinol
2009
,
23
:
925
931
.

25

Lin
Q
,
Gao
Z
,
Alarcon
RM
,
Ye
J
,
Yun
Z
.
A role of miR-27 in the regulation of adipogenesis
.
FEBS J
2009
,
276
:
2348
2358
.

26

Li
H
,
Chen
X
,
Guan
L
,
Qi
Q
,
Shu
G
,
Jiang
Q
,
Yuan
L
, et al. .
MiRNA-181a regulates adipogenesis by targeting tumor necrosis factor-α (TNF-α) in the porcine model
.
PLoS One
2013
,
8
:
e71568
.

27

Ye
Y
,
Deng
L
,
Liang
M
,
Xu
L
,
Zhang
L
,
Ma
Y
,
Li
Y
.
MicroRNA expression profiles in dispose tissues and liver from sex-linked dwarf and normal chickens
.
Acta Biochim Biophys Sin
2014
,
46
:
723
726
.

28

Chen
C
,
Peng
Y
,
Peng
Y
,
Peng
J
,
Jiang
S
.
MiR-135a-5p inhibits 3T3-L1 adipogenesis through activation of canonical Wnt/β-catenin signaling
.
J Mol Endocrinol
2014
,
52
:
311
320
.

29

Choy
L
,
Skillington
J
,
Derynck
R
.
Roles of autocrine TGF-beta receptor and Smad signaling in adipocyte differentiation
.
J Cell Biol
2000
,
149
:
667
682
.

30

Chen
C
,
Ridzon
DA
,
Broomer
AJ
,
Zhou
Z
,
Lee
DH
,
Nguyen
JT
,
Barbisin
M
, et al. .
Real-time quantification of microRNAs by stem-loop RT-PCR
.
Nucleic Acids Res
2005
,
33
:
e179
.

31

Qi
QR
,
Zhao
XY
,
Zuo
RJ
,
Wang
TS
,
Gu
XW
,
Liu
JL
,
Yang
ZM
.
Involvement of atypical transcription factor E2F8 in the polyploidization during mouse and human decidualization
.
Cell Cycle
2015
,
14
:
1842
1858
.

32

Fajas
L
.
Adipogenesis: a cross-talk between cell proliferation and cell differentiation
.
Ann Med
2003
,
35
:
79
85
.

33

Parikh
A
,
Lee
C
,
Joseph
P
,
Marchini
S
,
Baccarini
A
,
Kolev
V
,
Romualdi
C
, et al. .
MicroRNA-181a has a critical role in ovarian cancer progression through the regulation of the epithelial-mesenchymal transition
.
Nat Commun
2014
,
5
:
2977
.

34

Zhenye
L
,
Chuzhong
L
,
Youtu
W
,
Xiaolei
L
,
Lei
C
,
Lichuan
H
,
Hongyun
W
, et al. .
The expression of TGF-β1, Smad3, phospho-Smad3 and Smad7 is correlated with the development and invasion of nonfunctioning pituitary adenomas
.
J Transl Med
2014
,
12
:
71
.

35

Heneghan
HM
,
Miller
N
,
Kerin
MJ
.
Role of microRNAs in obesity and the metabolic syndrome
.
Obes Rev
2010
,
354
361
.

36

Ambros
V
.
MicroRNA pathways in flies and worms: growth, death, fat, stress, and timing
.
Cell
2003
,
113
:
673
676
.

37

Wang
Q
,
Li
YC
,
Wang
J
,
Kong
J
,
Qi
Y
,
Quigg
RJ
,
Li
X
.
MiR-17-92 cluster accelerates adipocyte differentiation by negatively regulating tumor-suppressor Rb2/p130
.
Proc Natl Acad Sci U S A
2008
,
105
:
2889
2894
.

38

Huang
N
,
Wang
J
,
Xie
W
,
Lyu
Q
,
Wu
J
,
He
J
,
Qiu
W
, et al. .
MiR-378a-3p enhances adipogenesis by targeting mitogen-activated protein kinase 1
.
Biochem Biophys Res Commun
2015
,
457
:
37
42
.

39

Liang
WC
,
Wang
Y
,
Wan
DC
,
Yeung
VS
,
Waye
MM
.
Characterization of miR-210 in 3T3-L1 adipogenesis
.
J Cell Biochem
2013
,
114
:
2699
2707
.

40

Yang
Z
,
Bian
C
,
Zhou
H
,
Huang
S
,
Wang
S
,
Liao
L
,
Zhao
RC
.
MicroRNA hsa-miR-138 inhibits adipogenic differentiation of human adipose tissue-derived mesenchymal stem cells through adenovirus EID-1
.
Stem Cells Dev
2011
,
20
:
259
267
.

41

Liu
S
,
Yang
Y
,
Wu
J
.
TNFalpha-induced up-regulation of miR-155 inhibits adipogenesis by down-regulating early adipogenic transcription factors
.
Biochem Biophys Res Commun
2011
,
414
:
618
624
.

42

Cui
QK
,
Liu
WD
,
Zhu
JX
,
Wang
YH
,
Wang
ZG
.
MiR-184 promotes proliferation ability of glioma cell by regulation FOXO3
.
Asian Pac J Trop Med
2014
,
7
:
776
779
.

43

Prasad
R
,
Katiyar
SK
.
Down-regulation of miRNA-106b inhibits growth of melanoma cells by promoting G1-phase cell cycle arrest and reactivation of p21/WAF1/Cip1 protein
.
Oncotarget
2014
,
5
:
10636
10649
.

44

Xiao
F
,
Zhang
W
,
Zhou
L
,
Xie
H
,
Xing
C
,
Ding
S
,
Chen
K
, et al. .
microRNA-200a is an independent prognostic factor of hepatocellular carcinoma and induces cell cycle arrest by targeting CDK6
.
Oncol Rep
2013
,
30
:
2203
2210
.

45

Zhu
X
,
Wu
L
,
Yao
J
,
Jiang
H
,
Wang
Q
,
Yang
Z
,
Wu
F
.
Micro-RNA let-7c inhibits cell proliferation and induces cell cycle arrest by targeting CDC25A in human hepatocellular carcinoma
.
PLoS One
2015
,
10
:
e0124266
.

46

Llobet-Navas
D
,
Rodriguez-Barrueco
R
,
de la Iglesia-Vicente
J
,
Olivan
M
,
Castro
V
,
Saucedo-Cuevas
L
,
Marshall
N
, et al. .
The microRNA 424/503 cluster reduces CDC25A expression during cell cycle arrest imposed by transforming growth factor β in mammary epithelial cells
.
Mol Cell Biol
2014
,
34
:
4216
4231
.

47

Cui
L
,
Zhou
H
,
Zhao
H
,
Zhou
Y
,
Xu
R
,
Xu
X
,
Zheng
L
, et al. .
MicroRNA-99a induces G1-phase cell cycle arrest and suppresses tumorigenicity in renal cell carcinoma
.
BMC Cancer
2012
,
12
:
546
.

48

Lu
H
,
Ward
MG
,
Adeola
O
,
Ajuwon
KM
.
Regulation of adipocyte differentiation and gene expressioncrosstalk between TGFβ and wnt signaling pathways
.
Mol Biol Rep
2013
,
40
:
5237
5245
.

49

Taylor
MA
,
Sossey-Alaoui
K
,
Thompson
CL
,
Danielpour
D
,
Schiemann
WP
.
TGFβ upregulates miR-181a expression to promote breast cancer metastasis
.
J Clin Invest
2013
,
123
:
150
163
.

50

Bhushan
R
,
Grünhagen
J
,
Becker
J
,
Robinson
PN
,
Ott
CE
,
Knaus
P
.
MiR-181a promotes osteoblastic differentiation through repression of TGFβ signaling molecules
.
Int J Biochem Cell Biol
2013
,
45
:
696
705
.

51

Liu
L
,
Wang
Y
,
Fan
H
,
Zhao
X
,
Liu
D
,
Hu
Y
,
Kidd
AR
, et al. .
MicroRNA-181a regulates local immune balance by inhibiting proliferation and immunosuppressive properties of mesenchymal stem cells
.
Stem Cells
2012
,
30
:
1756
1770
.

52

Derynck
R
,
Zhang
Y
,
Feng
XH
.
Smads: transcriptional activators of TGFβ responses
.
Cell
1998
,
95
:
737
740
.

53

Massagué
J
,
Seoane
J
,
Wotton
D
.
Smad transcription factors
.
Genes Dev
2005
,
19
:
2783
2810
.

54

Choy
L
,
Derynck
R
.
Transforming growth factor-beta inhibits adipocyte differentiation by Smad3 interacting with CCAAT/enhance -binding protein (C/EBP) and repressing C/EBP transactivation function
.
J Biol Chem
2003
,
278
:
9609
9619
.

55

Zhu
X
,
Ozturk
F
,
Liu
C
,
Oakley
GG
,
Nawshad
A
.
Transforming growth factor-β activates c-Myc to promote palatal growth
.
J Cell Biochem
2012
,
113
:
3069
3085
.

56

Liu
R
,
Zhou
Z
,
Huang
J
,
Chen
C
.
PMEPA1 promotes androgen receptor-negative prostate cell proliferation through suppressing the Smad3/4-c-Myc-p21 Cip1 signaling pathway
.
J Pathol
2011
,
223
:
683
694
.

57

Lim
SK
,
Hoffmann
FM
.
Smad4 cooperates with lymphoid enhancer-binding factor 1/T cell-specific factor to increase c-myc expression in the absence of TGF-beta signaling
.
Proc Natl Acad Sci U S A
2006
,
103
:
18580
18585
.

58

Sandhu
C
,
Garbe
J
,
Bhattacharya
N
,
Daksis
J
,
Pan
CH
,
Yaswen
P
,
Koh
J
, et al. .
Transforming growth factor beta stabilizes p15INK4B protein, increases p15INK4B–Cdk4 complexes, and inhibits cyclin D1–Cdk4 association in human mammary epithelial cells
.
Mol Cell Biol
1997
,
17
:
2458
2467
.

59

Hannon
GJ
,
Beach
D
.
p15INK4B is a potential effector of TGF-b-induced cell cycle arrest
.
Nature
1994
,
371
:
257
261
.

60

Warner
BJ
,
Blain
SW
,
Seoane
J
,
Massagué
J
.
Myc downregulation by transforming growth factor beta required for activation of the p15(INK4B) G(1) arrest pathway
.
Mol Cell Biol
1999
,
19
:
5913
5922
.

61

Dmitrovsky
E
,
Kuehl
WM
,
Hollis
GF
,
Kirsch
IR
,
Bender
TP
,
Segal
S
.
Expression of a transfected human c-myc oncogene inhibits differentiation of a mouse erythroleukemia cell line
.
Nature
1986
,
322
:
748
750
.

62

Freytag
SO
.
Enforced expression of the c-myc oncogene inhibits cell differentiation by precluding entry into a distinct predifferentiation state in G0/G1
.
Mol Cell Biol
1988
,
8
:
1614
1624
.

63

He
XC
,
Zhang
J
,
Tong
WG
,
Tawfik
O
,
Ross
J
,
Scoville
DH
,
Tian
Q
, et al. .
BMP signaling inhibits intestinal stem cell self-renewal through suppression of Wnt-beta-catenin signaling
.
Nat Genet
2004
,
36
:
1117
1121
.

64

Ross
SE
,
Hemati
N
,
Longo
KA
,
Bennett
CN
,
Lucas
PC
,
Erickson
RL
,
MacDougald
OA
.
Inhibition of adipogenesis by Wnt signaling
.
Science
2000
,
289
:
950
953
.

65

Shah
M
,
Rennoll
SA
,
Raup-Konsavage
WM
,
Yochum
GS
.
A dynamic exchange of TCF3 and TCF4 transcription factors controls myc expression in colorectal cancer cell
.
Cell Cycle
2015
,
14
:
323
332
.