MicroRNAs (miRNAs) play important roles in the development of skeletal muscle. In our previous study, expression of miR-195 and miR-497 were shown to be upregulated during muscle development in pigs. In this study, we investigated the roles of these two miRNAs in myogenesis and analyzed their transcriptional regulation. Our results showed that miR-195 and miR-497 were upregulated during muscle development and myoblast differentiation. Moreover, miR-195 and miR-497 inhibited proliferation but not differentiation in C2C12 cells. Further investigation revealed that Igf1r, Insr, Ccnd2 and Ccne1 were directly targeted by miR-195 and miR-497 in myoblasts. In addition, we confirmed that miR-195 and miR-497, which shared the similar expression profiling, were negatively regulated by nuclear factor κB (NF-κB) in both myoblasts and skeletal muscle tissue. Our data illustrate that the signaling pathway NF-κB–miR-195/497–Igf1r/Insr-Ccnd2/Ccne1 plays important roles in myogenesis. Our study provides novel evidence for the roles of miR-195 and miR-497 in muscle development.

MicroRNAs (miRNAs; miR) were first discovered in 1993 (Lee et al., 1993; Wightman et al., 1993) and have since attracted global attention to their various functions in a broad range of biological processes. Skeletal-muscle-specific miRNAs, including miR-1, miR-133, miR-206 and miR-208b/499 (Chen et al., 2006; Kim et al., 2006; McCarthy et al., 2009; Yang et al., 2015), muscle-enriched miRNAs including miR-486 (Xu et al., 2012), and ubiquitous miRNAs including miR-21 (Bai et al., 2015) have all been shown to play important roles in myogenesis.

miR-195 and miR-497 are closely linked in the genome and have been shown to share the same seed sequences. Both belong to the miR-15 family, which contains two more members, namely miR-15 and miR-16. In this family, miR-15 and miR-16 have been reported to target the well-known anti-apoptotic gene Bcl2 (Cimmino et al., 2005). Therefore, members of the miR-15 family are thought to play some roles in tumorigenesis. Consistent with this notion, miR-195 and miR-497 have been shown to be associated with various types of cancers. For example, miR-195 has been shown to act as a tumor suppressor in gastric cancer (Deng et al., 2013), hepatocellular carcinoma (Ding et al., 2013; Xu et al., 2009), bladder cancer (Lin et al., 2012), breast cancer (Luo et al., 2014; Singh and Saini, 2012), leukemia (Diaz-Beya et al., 2012) and other cancer types. miR-497 and miR-195 play similar roles in most physiological processes and miR-497 has also been found to be involved in tumorigenesis (Waters et al., 2012). The CCND1, CCND3, CCNE1, CDC25A and CDK4 genes are usually shown as being the targets of miR-195 and miR-497 in some cancer cells (Furuta et al., 2013; Li et al., 2011). Therefore, miR-195 and miR-497 are potential therapeutic targets in many types of cancers (Furuta et al., 2013; Guo et al., 2012; Itesako et al., 2014; Li et al., 2011; Ozata et al., 2011). Additionally, miR-195 has been implicated in neurodevelopment (Ai et al., 2013; Liu et al., 2013; Zhu et al., 2012), placental pathogenesis (Bai et al., 2012; Hu et al., 2009) and glucose metabolism (Joglekar et al., 2007; Ortega et al., 2014), suggesting its importance in a range of biological processes.

Previous studies have suggested that miR-195 and miR-497 participate in muscle development. In a previous work from our laboratory, we found that both miR-195 and miR-497 were significantly upregulated in longissimus muscle tissue during muscle development in pigs (Huang et al., 2008). Moreover, miR-195 exhibited higher expression in the gluteus medius in horses suffering from polysaccharide storage myopathy (PSSM) or recurrent exertional rhabdomyolysis (RER) (Barrey et al., 2011). Furthermore, miR-195 might regulate the proliferation and apoptosis of cardiomyocytes, and it is also associated with heart development and heart disease; for example, hypertrophy, heart failure and lipotoxic cardiomyopathy (Porrello et al., 2011; van Rooij et al., 2006; Zhu et al., 2011). Finally, miR-195 and miR-497 have been reported to induce quiescence of satellite cells at the postnatal stage in mice (Sato et al., 2014). However, the direct roles of miR-195 and miR-497 in skeletal muscle development remain largely undefined.

In this study, we examined the role of the miR-195 and miR-497 cluster in skeletal muscle development. Our data provide insight into the targets of these miRNAs and demonstrate the involvement of nuclear factor κB (NF-κB) in the regulation of this miRNA cluster. Furthermore, our results support the idea that miRNAs play important roles in skeletal muscle growth and development.

MiR-195 and miR-497 were upregulated during skeletal muscle development and differentiation in C2C12 cells

First, we used quantitative real-time polymerase chain reaction (qPCR) to analyze miR-195 and miR-497 expression patterns during myogenesis in C2C12 cells that were stimulated to differentiate into myotubes (Fig. 1A), and observed an upregulation of miR-195 (Fig. 1B). Consistent with this finding, miR-195 was also gradually upregulated during muscle development from 2 days to 6 weeks of age in the thigh muscles of mice (Fig. 1C). miR-497 exhibited a similar expression profile, but the fold change of miR-497 was lower than that of miR-195 in muscle tissue (Fig. 1B,C). Thus, expression of miR-195 and miR-497 was upregulated during skeletal muscle growth and myoblast differentiation.

Fig. 1.

Upregulation of miR-195 and miR-497 during C2C12 myoblast differentiation and skeletal muscle development. (A) C2C12 myoblasts (left) were induced to differentiate into myotubes (right) using differentiation medium, and immunofluorescence staining was performed to detect cells expressing α-actin (green). Cell nuclei were stained blue by DAPI. Scale bars: 100 µm. (B,C) qPCR was performed to determine the fold change of miR-195 and miR-497 expression in C2C12 cells that were differentiated for 0 and 4 days (B), and in the thigh muscle of mice at 2 days, 2 weeks, 4 weeks and 6 weeks of age (C). The expression was normalized to that of the small nuclear RNA U6, and the relative fold change was determined relative to the expression at day 0 for B or day 2 for C. Three independent individuals or replicates were used per time point, and the results are presented as the mean±s.e.m. **P<0.01 (Student's t-test).

Fig. 1.

Upregulation of miR-195 and miR-497 during C2C12 myoblast differentiation and skeletal muscle development. (A) C2C12 myoblasts (left) were induced to differentiate into myotubes (right) using differentiation medium, and immunofluorescence staining was performed to detect cells expressing α-actin (green). Cell nuclei were stained blue by DAPI. Scale bars: 100 µm. (B,C) qPCR was performed to determine the fold change of miR-195 and miR-497 expression in C2C12 cells that were differentiated for 0 and 4 days (B), and in the thigh muscle of mice at 2 days, 2 weeks, 4 weeks and 6 weeks of age (C). The expression was normalized to that of the small nuclear RNA U6, and the relative fold change was determined relative to the expression at day 0 for B or day 2 for C. Three independent individuals or replicates were used per time point, and the results are presented as the mean±s.e.m. **P<0.01 (Student's t-test).

miR-195 and miR-497 inhibited the proliferation of C2C12 cells

To determine the roles of miR-195 and miR-497 in myoblasts, miR-195 and miR-497 were overexpressed by transfecting synthetic miRNA mimics in C2C12 cells, and the cell cycle distribution was determined by flow cytometry. The flow cytometry results showed that the proportion of cells arrested in the G1 or G0 phase was significantly higher in cells transfected with miR-195 or miR-497 mimics than in cells transfected with the negative control (NC) mimics, and the proliferation index was also decreased following the transfection of miR-195 or miR-497 mimics (P<0.01; Fig. 2A). These results indicate that both miR-195 and miR-497 inhibit the proliferation of myoblasts by blocking the G1-S transition.

Fig. 2.

Inhibition of C2C12 myoblast proliferation by miR-195 and miR-497. (A) After C2C12 cells were transfected with miR-195 and miR-497 or scrambled negative control (NC), flow cytometry was used to analyze the cell cycle phase (left), and the proliferation index was then analyzed (right). (B) After transfection with miR-195/497-C1 or pEGFP-C1, cells were collected, and the fold change of miR-195 and miR-497 expression was analyzed by qPCR. The expression was normalized to that of the small nuclear RNA U6, and the relative fold change was determined relative to the expression in pEGFP-C1-transfected cells. (C) After transfection with miR-195/497-C1 or pEGFP-C1 (green), cells were stained with EdU (red). Scale bars: 200 µm. (D) After treatment as described in C, cells were trypsinized and analyzed by flow cytometry, and the percentage of GFP+ and Edu+ cells out of the total number of GFP+ cells was determined. Each experiment was replicated using at least three samples, and the results are presented as the mean±s.e.m. *P<0.05; **P<0.01 (Student's t-test).

Fig. 2.

Inhibition of C2C12 myoblast proliferation by miR-195 and miR-497. (A) After C2C12 cells were transfected with miR-195 and miR-497 or scrambled negative control (NC), flow cytometry was used to analyze the cell cycle phase (left), and the proliferation index was then analyzed (right). (B) After transfection with miR-195/497-C1 or pEGFP-C1, cells were collected, and the fold change of miR-195 and miR-497 expression was analyzed by qPCR. The expression was normalized to that of the small nuclear RNA U6, and the relative fold change was determined relative to the expression in pEGFP-C1-transfected cells. (C) After transfection with miR-195/497-C1 or pEGFP-C1 (green), cells were stained with EdU (red). Scale bars: 200 µm. (D) After treatment as described in C, cells were trypsinized and analyzed by flow cytometry, and the percentage of GFP+ and Edu+ cells out of the total number of GFP+ cells was determined. Each experiment was replicated using at least three samples, and the results are presented as the mean±s.e.m. *P<0.05; **P<0.01 (Student's t-test).

Additionally, we confirmed the effects of miR-195 and miR-497 on myoblast proliferation with 5-ethynyl-2′-deoxyuridine (EdU) assays. First, a miR-195 and miR-497 overexpression vector, named miR-195/497-C1, was constructed by inserting a 738-bp DNA fragment, which contained the miR-195 and miR-497 primary transcript cluster, into the pEGFP-C1 vector. qPCR results indicated a significant overexpression of both miR-195 and miR-497 following the transfection of C2C12 cells with the miR-195/497-C1 construct (P<0.01; Fig. 2B). Moreover, EdU staining showed that the proportion of EdU-positive cells was significantly lower following transfection with miR-195/497-C1 than with pEGFP-C1 (P<0.01; Fig. 2C,D). These results indicate that DNA synthesis was inhibited in myoblasts when miR-195 and miR-497 were overexpressed. Taken together, the above results confirmed that miR-195 and miR-497 could inhibit the proliferation of myoblasts.

Genes encoding cyclin E1 and cyclin D2 were directly targeted by miR-195 and miR-497 in C2C12 cells

To elucidate the mechanisms through which miR-195 and miR-497 affected cell proliferation, the potential targets were predicted by TargetScan (http://www.targetscan.org/). Genes encoding cyclin E1 and cyclin D2 (Ccne1 and Ccnd2, respectively), which were found to contain multiple binding sites for miR-195 and miR-497 at their 3′ untranslated regions (UTRs), were primarily selected as potential candidates (Fig. 3A).

Fig. 3.

Identification of Ccne1 and Ccnd2 as targets of miR-195 and miR-497 in C2C12 myoblasts. (A) Binding sites for miR-195 and miR-497 in the 3′UTR of Ccne1 and Ccnd2 are shown. (B,C) Luciferase activity was analyzed after cells were co-transfected with miR-195, miR-497 or negative control (NC), and psiCHECK-2 constructs that contain binding sites in the 3′UTR of Ccne1 (B) and Ccnd2 (C). BS, binding site. (D) After cells were transfected with miR-195, miR-497 or NC, the levels of the cyclin E1 and cyclin D2 proteins were visualized by western blotting. Triplicate samples were analyzed for each treatment, and the results are presented as the mean±s.e.m. **P<0.01 (Student's t-test).

Fig. 3.

Identification of Ccne1 and Ccnd2 as targets of miR-195 and miR-497 in C2C12 myoblasts. (A) Binding sites for miR-195 and miR-497 in the 3′UTR of Ccne1 and Ccnd2 are shown. (B,C) Luciferase activity was analyzed after cells were co-transfected with miR-195, miR-497 or negative control (NC), and psiCHECK-2 constructs that contain binding sites in the 3′UTR of Ccne1 (B) and Ccnd2 (C). BS, binding site. (D) After cells were transfected with miR-195, miR-497 or NC, the levels of the cyclin E1 and cyclin D2 proteins were visualized by western blotting. Triplicate samples were analyzed for each treatment, and the results are presented as the mean±s.e.m. **P<0.01 (Student's t-test).

The dual luciferase reporter system was first used to analyze the interaction between miR-195 and/or miR-497 and these two candidate genes. Fragments of 3′UTR containing the binding sites were inserted into the psiCHECK-2 vector and then the constructs were transfected into C2C12 cells together with miRNA mimics. Overexpression of miR-195 and miR-497 resulted in decreased relative luciferase activity when the proximal binding site of Ccne1 3′UTR was inserted. However, no significant difference was detected when the distal binding site was inserted (Fig. 3B). Moreover, overexpression of miR-195 and miR-497 decreased the relative luciferase activity when the proximal or distal binding sites of Ccnd2 3′UTR were inserted (Fig. 3C). These results indicate that miR-195 and miR-497 can interact with the Ccne1 and Ccnd2 3′UTR. Furthermore, western blot analysis demonstrated that cyclin E1 and cyclin D2 proteins were decreased when miR-195 or miR-497 was overexpressed (Fig. 3D). Therefore, miR-195 and miR-497 could directly target the Ccne1 and Ccnd2 cell cycle genes in myoblasts.

miR-195 and miR-497 directly target the Igf1r and Insr genes in C2C12 cells

In addition to cell-cycle-related genes, we also predicted that other genes related to cell growth might be targeted by these miRNAs. TargetScan prediction results showed that the 3′UTR of insulin-like growth factor 1 receptor (Igf1r or IGF-1R) contained one binding site for miR-195 and miR-497, whereas that for insulin receptor (Insr) contained two binding sites for miR-195 and miR-497 (Fig. 4A).

Fig. 4.

Identification of Igf1r and Insr as direct targets of miR-195 and miR-497 in C2C12 myoblasts. (A) Binding sites for miR-195 and miR-497 in the 3′UTR of Igf1r and Insr according to TargetScan prediction results are shown. (B) Luciferase activity was analyzed after cells were co-transfected with psiCHECK-2, which has binding sites in the Igf1r or Insr 3′UTR, and miR-195, miR-195 mut, miR-497, miR-497 mut or negative control (NC). (C) After transfection with miR-195, miR-497 or negative control, cell lysates were subjected to western blot to detect endogenous Igf1r and Insr protein. Triplicate samples were analyzed for each treatment, and the results are presented as the mean±s.e.m. *P<0.05; **P<0.01 (Student's t-test).

Fig. 4.

Identification of Igf1r and Insr as direct targets of miR-195 and miR-497 in C2C12 myoblasts. (A) Binding sites for miR-195 and miR-497 in the 3′UTR of Igf1r and Insr according to TargetScan prediction results are shown. (B) Luciferase activity was analyzed after cells were co-transfected with psiCHECK-2, which has binding sites in the Igf1r or Insr 3′UTR, and miR-195, miR-195 mut, miR-497, miR-497 mut or negative control (NC). (C) After transfection with miR-195, miR-497 or negative control, cell lysates were subjected to western blot to detect endogenous Igf1r and Insr protein. Triplicate samples were analyzed for each treatment, and the results are presented as the mean±s.e.m. *P<0.05; **P<0.01 (Student's t-test).

Overexpression of miR-195 or miR-497 significantly decreased the luciferase activity of the construct containing the binding site from the Igf1r gene as compared to that of the NC control (P<0.01; Fig. 4B). Mutant miR-195 or miR-497 generated through mutation of the seed sequence did not inhibit the luciferase activity (Fig. 4B). This result indicates that miR-195 and miR-497 can specifically interact with the Igf1r gene. For the Insr gene, miR-195 significantly inhibited both binding sites (P<0.01, P<0.05), whereas miR-497 significantly inhibited the first binding site (P<0.05), but not the second binding site (Fig. 4B). This result indicates that miR-195 and miR-497 target the Insr gene. Furthermore, the western blotting results confirmed that miR-195 and miR-497 inhibited the expression of Igf1r and Insr protein in C2C12 cells (Fig. 4C). Taken together, these results demonstrate that miR-195 and miR-497 directly target the Igf1r and Insr 3′UTRs in C2C12 cells.

Igf1r and Insr genes play important roles in the proliferation of C2C12 cells

Several studies indicate that alterations in Igf1r and/or Insr genes in modified L6 myoblasts or muscle satellite cells affect the sensitivity of these cells to growth-related hormone, thereby altering cell proliferation (Arabkhari et al., 2010; Bonnesen et al., 2010; Dong et al., 2013; Kim et al., 2005; Quinn and Roh, 1993). Here, we also studied the effects of Igf1r and Insr on the proliferation of C2C12 myoblasts. First, we used flow cytometry to analyze the effects of Igf1r or Insr knockdown on the cell cycle distribution in C2C12 cells. More cells were arrested in the G1-G0 phase in cells transfected with small interfering RNA (siRNA) against Igf1r1 (si-Igfr-1) and Insr1 (si-Insr-1) than in control-transfected cells (Fig. 5A,B; P<0.05). Moreover, the proliferation index was also decreased following knockdown of Igf1r or Insr (Fig. 5C, P<0.05). These results indicate that downregulation of Igf1r and Insr blocks the G1-to-S phase transition in C2C12 cells. Additionally, the interaction between miR-195/497 and the IGF-1/insulin signaling pathway was also studied. An EdU incorporation assay indicated that the proportion of EdU-positive cells was significantly decreased in cells transfected with miR-195/497-C1 as compared with control cells. Moreover, following stimulation with IGF-1, the proportion of EdU-positive cells was slightly increased in cells transfected with miR-195/497-C1, but the level was still lower than that in control cells (Fig. 5D). Taken together, these results imply that Igf1r and Insr promotes C2C12 myoblast proliferation, and that miR-195 and miR-497 inhibit myoblast proliferation through targeting the Igf1r and Insr genes.

Fig. 5.

Effects of Igf1r and Insr on C2C12 myoblast proliferation. (A) Cells were transfected with siRNAs targeting Igf1r (si-Igf1r-1, -2 and -3), siRNAs targeting Insr (si-Insr-1, -2, -3 and -4), scrambled negative control (NC) or nothing (mock), and endogenous Igf1r and Insr proteins levels were visualized by western blotting. (B,C) C2C12 cells were transfected with si-Igf1r-1, si-Insr-1 or scrambled negative control, and the cell cycle distribution was analyzed by propidium iodine staining followed by flow cytometry (B). The proliferation index is shown (C). (D) After transfection with miR-195/497-C1 or pEGFP-C1, followed by stimulation with IGF-1 or its vehicle acetic acid, cells were analyzed by flow cytometry, and the percentage of GFP+ and Edu+ cells out of the total number of GFP+ cells was determined. Triplicate samples were analyzed for each treatment, and the results are presented as the mean±s.e.m. *P<0.05; **P<0.01 (Student's t-test).

Fig. 5.

Effects of Igf1r and Insr on C2C12 myoblast proliferation. (A) Cells were transfected with siRNAs targeting Igf1r (si-Igf1r-1, -2 and -3), siRNAs targeting Insr (si-Insr-1, -2, -3 and -4), scrambled negative control (NC) or nothing (mock), and endogenous Igf1r and Insr proteins levels were visualized by western blotting. (B,C) C2C12 cells were transfected with si-Igf1r-1, si-Insr-1 or scrambled negative control, and the cell cycle distribution was analyzed by propidium iodine staining followed by flow cytometry (B). The proliferation index is shown (C). (D) After transfection with miR-195/497-C1 or pEGFP-C1, followed by stimulation with IGF-1 or its vehicle acetic acid, cells were analyzed by flow cytometry, and the percentage of GFP+ and Edu+ cells out of the total number of GFP+ cells was determined. Triplicate samples were analyzed for each treatment, and the results are presented as the mean±s.e.m. *P<0.05; **P<0.01 (Student's t-test).

MiR-195 and miR-497 does not affect the differentiation of C2C12 cells

Next, we evaluated the roles of miR-195 and miR-497 during myoblast differentiation. Following transfection of miR-195, miR-497, or negative control mimics, differentiation was induced in C2C12 cells for 72 h, and the fused myotubes were stained for the myogenic marker myosin (Fig. 6A). The number of fused myotubes in each treatment was analyzed (Fig. 6B). There were no obvious differences in differentiation between cells transfected with miR-195 or miR-497 mimics and cells transfected with negative control mimics. Moreover, qPCR showed that the expression of the myogenic marker genes myogenin, MyHC (also known as myosin heavy chain 1, MYH1) and MCK (also known as CKM) was not significantly different among groups (Fig. 6C). These results indicate that myogenic differentiation was not affected by miR-195 and miR-497 in C2C12 cells.

Fig. 6.

Effects of miR-195 and miR-497 on myoblast differentiation. (A) After transfection with miR-195, miR-497 or negative control (NC), C2C12 cells were induced to differentiate into myotubes and immunofluorescence staining was performed to detect myotubes using antibodies against fast skeletal myosin. Scale bars: 200 µm. (B) Six photos for each group were randomly selected for quantitative analysis of the number of myotubes. (C) Cells treated as in A were collected, and qPCR was performed to analyze the fold change in expression of the myogenic markers genes myogenin, MyHC and MCK. The expression was normalized to that of tubulin, and the relative fold change was compared to expression in the NC group. Triplicate samples were analyzed for each treatment, and the results are presented as the mean±s.e.m.

Fig. 6.

Effects of miR-195 and miR-497 on myoblast differentiation. (A) After transfection with miR-195, miR-497 or negative control (NC), C2C12 cells were induced to differentiate into myotubes and immunofluorescence staining was performed to detect myotubes using antibodies against fast skeletal myosin. Scale bars: 200 µm. (B) Six photos for each group were randomly selected for quantitative analysis of the number of myotubes. (C) Cells treated as in A were collected, and qPCR was performed to analyze the fold change in expression of the myogenic markers genes myogenin, MyHC and MCK. The expression was normalized to that of tubulin, and the relative fold change was compared to expression in the NC group. Triplicate samples were analyzed for each treatment, and the results are presented as the mean±s.e.m.

NF-κB binds directly to the promoter region of the miR-195 and miR-497 cluster

Because miR-195 and miR-497 play roles in regulating myoblast proliferation, we next investigated their transcriptional mechanism. Based on their genomic structure and expression pattern, we hypothesized that miR-195 and miR-497 were transcribed from the same primary transcript and share the same regulatory mechanism during myogenesis. To test this hypothesis, we first predicted the transcription start site (TSS) of the Mus musculus miR-195 and miR-497 cluster according to the gene annotation database of humans. Then, the potential binding sites for transcription factors in a 3-kb promoter region of the miR-195 and miR-497 cluster were predicted using TFSEARCH software. According to these results, three potential binding sites for p65 (also known as RelA, the active subunit of NF-κB), located at −604, −337 and +106 bp, were found in the promoter region (Fig. 7A). These binding sites share highly conserved nucleotides with the consensus sequence (GGGRNNYYC) for p65 (Fig. 7B). Furthermore, a 3090-bp fragment harboring these three p65-binding sites was cloned into the pGL3 luciferase reporter vector. Deletion mutations showed that the relative luciferase activity of the miR-195 and miR-497 promoter was significantly elevated when the first p65-binding site (−604 bp) was deleted. However, no significant variation was observed when the second (−337 bp) or third (+106 bp) binding sites were deleted (Fig. 7B). As expected, a strong gel-shift signal was detected when the first p65-binding site probe was added in the C2C12 nuclear extracts in electrophoretic mobility shift assays (EMSAs). However, there was almost no gel-shift signal for the probes of the second or third p65-binding sites (Fig. 7C). The shift band signal generated using the probe for the first p65-binding site was decreased when excess unlabeled competitor probe was added, but was not affected by the same amount of mutant competitor probe (Fig. 7D). Moreover, this gel-shift band was intensified in a concentration-dependent manner when nuclear extract was added, and was dramatically reduced when the 10-fold to 100-fold excess of unlabeled competitor probe was added (Fig. 7E). Chromatin immunoprecipitation (ChIP)-PCR was then performed to detect the binding of p65 with these three sites in C2C12 cells. Similar to the results of the luciferase and gel-shift assays, ChIP-PCR showed that the DNA fragments containing the first binding site were pulled down by anti-p65 antibodies (Fig. 7F). Therefore, these results confirm that NF-κB can directly bind to the specific site of the promoter of the miR-195 and miR-497 cluster in C2C12 cells.

Fig. 7.

Identification of the NF-κB binding site in the promoter regions of miR-195 and miR-497 cluster. (A) Three binding sites (BS) were predicted near the TSS of the miR-195 and miR-497 transcript. (B) Luciferase activity was detected following transfection with fragments containing all the wild-type binding sites (WT), or after deletion of the first binding site (−604 to −594 bp) (WTΔ1), the second binding site (−377∼−367 bp) (WTΔ2) or the third binding site (+106∼+116 bp) (WTΔ3). (C) Gel-shift signals of three different NF-κB probes incubated with nuclear extracts of C2C12 cells. (D) Gel-shift signals for probes for the first binding site, wild-type compete probe (WT-CP) or mutant compete probe (Mut-CP) incubated with nuclear extracts (NE) or cytoplasm extracts (CE) of C2C12 cells. (E) Gel-shift signals for the first binding site probe incubated with different amounts of nuclear extracts (0, 0.1, 1, 5, and 10 μg) or with a constant amount of NE (5 μg) together with different amounts of WT-CP (100×, 50×, 20× or 10×). (F) Chromatin immunoprecipitation PCR results. The DNA fragments interacting with p65 protein were first pulled down by anti-p65 antibodies, and PCR was then performed to test the binding of p65 with the three binding sites using the pulled-down DNA as the template. Triplicate samples were analyzed for each treatment, and the results are presented as the mean±s.e.m. **P<0.01 (Student's t-test).

Fig. 7.

Identification of the NF-κB binding site in the promoter regions of miR-195 and miR-497 cluster. (A) Three binding sites (BS) were predicted near the TSS of the miR-195 and miR-497 transcript. (B) Luciferase activity was detected following transfection with fragments containing all the wild-type binding sites (WT), or after deletion of the first binding site (−604 to −594 bp) (WTΔ1), the second binding site (−377∼−367 bp) (WTΔ2) or the third binding site (+106∼+116 bp) (WTΔ3). (C) Gel-shift signals of three different NF-κB probes incubated with nuclear extracts of C2C12 cells. (D) Gel-shift signals for probes for the first binding site, wild-type compete probe (WT-CP) or mutant compete probe (Mut-CP) incubated with nuclear extracts (NE) or cytoplasm extracts (CE) of C2C12 cells. (E) Gel-shift signals for the first binding site probe incubated with different amounts of nuclear extracts (0, 0.1, 1, 5, and 10 μg) or with a constant amount of NE (5 μg) together with different amounts of WT-CP (100×, 50×, 20× or 10×). (F) Chromatin immunoprecipitation PCR results. The DNA fragments interacting with p65 protein were first pulled down by anti-p65 antibodies, and PCR was then performed to test the binding of p65 with the three binding sites using the pulled-down DNA as the template. Triplicate samples were analyzed for each treatment, and the results are presented as the mean±s.e.m. **P<0.01 (Student's t-test).

The expression of miR-195 and miR-497 are negatively regulated by NF-κB both in vitro and in vivo

After confirming the binding of NF-κB to the promoter of the miR-195 and miR-497 cluster, we studied the relationship between NF-κB and the expression of miR-195 and miR-497. Immunofluorescence staining showed that p65 was highly expressed in the nucleus of myoblasts, but weakly expressed in the nucleus and predominantly expressed in the cytoplasm of myotubes (Fig. 8A). These results indicate that NF-κB is deactivated during myoblast differentiation. By contrast, the expression of miR-195 and miR-497 was opposite to the activity of NF-κB during myogenesis. Taken together with the above results showing that NF-κB can directly bind to their promoter region, we hypothesized that NF-κB could directly regulate the expression of miR-195 and miR-497. To test this hypothesis, we studied the relationship between the activity of NF-κB and the promoter activity of the miR-195 and miR-497 cluster in vitro and in vivo. Overexpression of p65 significantly increased NF-κB activity but significantly reduced the promoter activity of miR-195 and miR-497 cluster in C2C12 cells (Fig. 8B,C). In contrast, knockdown of p65 decreased NF-κB activity but enhanced the promoter activity of miR-195 and miR-497 cluster (Fig. 8B,D). Furthermore, qPCR showed that miR-195 and miR-497 expression was decreased when p65 was overexpressed and increased when p65 was knocked down (Fig. 8E).

Fig. 8.

miR-195 and miR-497 expression was inhibited by NF-κB. (A) Immunofluorescence staining for p65 protein. The myoblasts and myotubes [cultured in differentiation medium (DM) for 4 days] were fixed and stained using anti-p65 antibodies (green). The nuclei were stained blue with DAPI. Scale bars: 100 µm. (B) Western blot analysis of p65 expression. C2C12 cells were transfected with pcDNA-p65, pcDNA-3.1 or nothing (mock), and western blotting was then performed to determine p65 protein expression (left). C2C12 cells were transfected with siRNAs against p65 [sip65_1, sip65_2, sip65_3, sip65_pool (sip65_1, sip65_2 and sip65_3 were pooled equally)], scrambled negative control (NC) or mock (without any treatment). Western blotting was then performed to determine p65 protein expression (right). (C) NF-κB activity was detected after overexpression of p65 (left). The transcriptional activity of the miR-195 and miR-497 promoter was determined using a luciferase reporter vector following overexpression of p65 (right). (D) NF-κB activity was analyzed following knockdown of p65. C2C12 cells were transfected with sip65_1, sip65_2 or scrambled NC, and the relative activity of miR-195 and miR-497 promoter was then analyzed. (E) qPCR analysis of the fold change of miR-195 and miR-497 expression. C2C12 cells were transfected with pcDNA-p65 or pcDNA-3.1 and with sip65-1 or negative control, and then the fold change of miR-195 and miR-497 expression was analyzed by qPCR. (F) Mice were treated with LPS or PBS by intraperitoneal injection, and then the fold change of miR-195 (left) and miR-497 (right) expression in the thigh muscle of the mice was analyzed after 24 h or 48 h by qPCR. (G) After LPS or PBS treatment for 24 h, the p65, Igf1r and Insr protein levels in the muscle tissue were analyzed by western blotting. (H) Schematic diagram of the NF-κB–miR-195/497–Igf1r/Insr–Ccnd2/Ccne1 pathway and its role in myoblast proliferation. The results shown for the expression were normalized to that of the small nuclear RNA U6, and the fold change was determined relative to the expression in pcDNA-3.1 and NC transfected cells, or PBS-injected mice. Triplicate samples were analyzed for each treatment, and the results are presented as the mean±s.e.m. *P<0.05; **P<0.01 (Student's t-test).

Fig. 8.

miR-195 and miR-497 expression was inhibited by NF-κB. (A) Immunofluorescence staining for p65 protein. The myoblasts and myotubes [cultured in differentiation medium (DM) for 4 days] were fixed and stained using anti-p65 antibodies (green). The nuclei were stained blue with DAPI. Scale bars: 100 µm. (B) Western blot analysis of p65 expression. C2C12 cells were transfected with pcDNA-p65, pcDNA-3.1 or nothing (mock), and western blotting was then performed to determine p65 protein expression (left). C2C12 cells were transfected with siRNAs against p65 [sip65_1, sip65_2, sip65_3, sip65_pool (sip65_1, sip65_2 and sip65_3 were pooled equally)], scrambled negative control (NC) or mock (without any treatment). Western blotting was then performed to determine p65 protein expression (right). (C) NF-κB activity was detected after overexpression of p65 (left). The transcriptional activity of the miR-195 and miR-497 promoter was determined using a luciferase reporter vector following overexpression of p65 (right). (D) NF-κB activity was analyzed following knockdown of p65. C2C12 cells were transfected with sip65_1, sip65_2 or scrambled NC, and the relative activity of miR-195 and miR-497 promoter was then analyzed. (E) qPCR analysis of the fold change of miR-195 and miR-497 expression. C2C12 cells were transfected with pcDNA-p65 or pcDNA-3.1 and with sip65-1 or negative control, and then the fold change of miR-195 and miR-497 expression was analyzed by qPCR. (F) Mice were treated with LPS or PBS by intraperitoneal injection, and then the fold change of miR-195 (left) and miR-497 (right) expression in the thigh muscle of the mice was analyzed after 24 h or 48 h by qPCR. (G) After LPS or PBS treatment for 24 h, the p65, Igf1r and Insr protein levels in the muscle tissue were analyzed by western blotting. (H) Schematic diagram of the NF-κB–miR-195/497–Igf1r/Insr–Ccnd2/Ccne1 pathway and its role in myoblast proliferation. The results shown for the expression were normalized to that of the small nuclear RNA U6, and the fold change was determined relative to the expression in pcDNA-3.1 and NC transfected cells, or PBS-injected mice. Triplicate samples were analyzed for each treatment, and the results are presented as the mean±s.e.m. *P<0.05; **P<0.01 (Student's t-test).

For the in vivo study, lipopolysaccharides (LPS) was used to activate NF-κB. Expression of miR-195 and miR-497 in the skeletal muscle tissue was significantly lower than that in the phosphate-buffered saline (PBS) control group 24 h after LPS treatment, whereas a decreasing tendency that did not reach significance was observed at 48 h after LPS treatment (Fig. 8F). Western blot results showed that, along with upregulation of p65, the protein levels of Igf1r and Insr, targets of miR-195 and miR-497, were also increased at 24 h after LPS treatment (Fig. 8G). These results indicate that transcription of miR-195 and miR-497 is also negatively regulated by NF-κB in the physiological context.

Taken together, these results demonstrate that NF-κB can directly downregulate miR-195 and miR-497 in both myoblasts and skeletal muscle tissue.

Previous studies have demonstrated that miR-195 and miR-497 play important roles in tumorigenesis, and miR-195 and miR-497 have also been shown to be involved in muscle development. In this study, we observed that miR-195 and miR-497 were upregulated during myogenesis in vivo and in vitro, consistent with our previous study in pigs (Huang et al., 2008). Moreover, we found that overexpression of miR-195 and miR-497 could prohibit myoblast proliferation. These results indicate that miR-195 and miR-497 play important roles in myogenesis.

Our present study identified Ccnd2 as a new target of miR-195 and miR-497, besides Ccne1, in C2C12 cells. These target genes are important for cell cycle progression, which might at least partially provide a basis for the proliferation inhibition mediated by miR-195 and miR-497 in myoblasts. Consistent with our results, recent studies have shown that miR-195 and miR-497 act as tumor suppressors by inhibiting multiple cell cycle regulators. miR-195 has been shown to directly target CCND1 and CCNE1 in human glioma and breast cancer (Hui et al., 2013; Luo et al., 2014), and both miR-195 and miR-497 have been shown to target CCND1, CCND3, CCNE1, CDC25A and CDK4 in human breast cancer and hepatocellular carcinoma (Furuta et al., 2013; Li et al., 2011). These results indicate that miR-195 and miR-497 can target many cell cycle genes and inhibit cell proliferation in both pathological and physiological conditions.

More importantly, the present study identified Igf1r and Insr as new targets of miR-195 and miR-497 in C2C12 cells. We confirmed that miR-195 and miR-497 could inhibit proliferation of C2C12 by targeting IGF1 signaling. Many studies have confirmed that the IGF-1 and insulin signaling pathways play important roles in the proliferation of myoblasts and the progression of myogenesis. IGF-1 and insulin have been shown to stimulate the proliferation and differentiation of muscle cells in a time- and concentration-dependent manner (Allen et al., 1985; Cassar-Malek et al., 1999; Cen et al., 2008; Engert et al., 1996; Grabiec et al., 2014); these important mediators also modulate protein metabolism (Castillo et al., 2004; Crown et al., 2000; Fryburg, 1994). Moreover, the myogenesis program can be influenced when the expression of IGF-1 and insulin receptors are altered (Arabkhari et al., 2010; Bonnesen et al., 2010). In rat and bovine myoblasts, overexpression of Igfr1 has been shown to enhance cell sensitivity to IGF-1 and to increase the proliferation and differentiation of the cells (Quinn and Haugk, 1996; Quinn and Roh, 1993). In contrast, satellite cells isolated from Igf1r+/− mouse muscles showed reduced proliferation and differentiation (Dong et al., 2013). It has also been shown that MKR mice, which have impaired Igf1r and Insr function, did not respond to growth hormone treatment and fail to exhibit increased muscle mass, proliferation and differentiation of satellite cells (Kim et al., 2005). Therefore, this study provides a new mechanism underlying the inhibition of myoblast proliferation by miR-195 and miR-497 through targeting Igf1r and Insr genes, which might explain the recent finding that miR-195 and miR-497 induce quiescence of satellite cells (Sato et al., 2014).

In this study, we also found that transcription of the miR-195 and miR-497 cluster was negatively regulated by NF-κB in both myoblasts and skeletal muscle tissue. Previous studies have indicated that NF-κB is constitutively activated in myoblasts, and its expression is decreased following the differentiation of C2C12 cells (Bakkar et al., 2008; Guttridge et al., 1999). In this study, our immunofluorescence results showed that p65 translocated from the nucleus to the cytoplasm during myoblast differentiation; thus, the reduced activation of NF-κB was mainly caused by translocation alteration during myogenic differentiation. Furthermore, NF-κB has been shown to play an important role in myogenesis by regulating cell cycle genes and myogenic genes. Many studies have indicated that NF-κB can increase the proliferation of myoblasts and inhibit myogenic differentiation (Bakkar et al., 2008; Guttridge et al., 1999; Langen et al., 2001). In a previous study, NF-κB was shown to enhance Ccnd1 transcription and promote retinoblastoma protein hyperphosphorylation, both of which are crucial for myoblast proliferation (Guttridge et al., 1999). Moreover, NF-κB inhibits MyoD expression at the post-transcription level and can promote the degradation of myoD mRNA (Guttridge et al., 2000).

NF-κB could also affect myogenesis through regulating crucial miRNAs. In this study, we demonstrated that NF-κB could negatively regulate the miR-195 and miR-497 cluster, which could inhibit the proliferation of myoblasts by targeting cell cycle and growth factor receptor genes. Thus, our results identified a new pathway mediating the proliferation of myoblasts, as shown in Fig. 8H. In this pathway, NF-κB regulated the transcription of the miR-195 and miR-497 cluster, which subsequently mediated the expression of Igf1r, Insr, Ccne1 and Ccnd2 to affect myoblast proliferation. Similarly, another myoblast proliferation inhibitor, miR-29, is also negatively regulated by NF-κB (Mott et al., 2010; Wang et al., 2008). These results confirm that NF-κB regulates the myogenesis program through modulating not only functional genes but also miRNAs.

In summary, our study provides direct evidence that miR-195 and miR-497 participate in skeletal muscle development by regulating myoblast proliferation. The potential mechanism underlying the inhibition of myoblast proliferation by these miRNAs involves direct targeting of Ccnd2, Ccne1, Igf1r and Insr genes. Additionally, miR-195 and miR-497 were negatively regulated by NF-κB. Finally, our data indicate a novel signaling pathway that involves NF-κB–miR-195/497–Igf1r/Insr–Ccnd2/Ccne1 in the modulation of the proliferation of myoblasts in this study. These findings provide important insight into the mechanisms of muscle development and might have great significance on animal breeding and health care.

Animals and cells

Balb/c mice were purchased from the Hubei Centre for Disease Control and Prevention (Wuhan, China). Thigh muscle samples were collected from male Balb/c mice at different ages (postnatal 2 days, 2 weeks, 4 weeks and 6 weeks). Samples were frozen in liquid nitrogen and stored at −80°C until RNA isolation. All experiments were performed according to the Guide for the Care and Use of Laboratory Animals (Institute of Laboratory Animal Resources, Commission on Life Sciences, National Research Council, 1996).

The growth medium for C2C12 cells was high-glucose Dulbecco's modified Eagle medium (DMEM; Hyclone) plus 10% (v/v) fetal bovine serum (Gibco). The differentiation medium was DMEM plus 2% (v/v) horse serum (Gibco).

LPS treatment

Balb/c mice at postnatal 4 weeks were purchased from the Hubei Centre for Disease Control and Prevention (Wuhan, China). Mice were randomly divided into four groups (three mice for every group). Two groups were treated with LPS (1 mg/kg body weight) or PBS for 24 h by intraperitoneal injection. The other two groups of mice were treated with LPS (1 mg/kg body weight) or PBS for 48 h by intraperitoneal injection. The injection was repeated every 12 h. After treatment, thigh muscle samples were collected from every mouse, and samples were frozen in liquid nitrogen and stored at −80°C until RNA and protein isolation.

qPCR

TRIzol reagent (Invitrogen) was used for total RNA isolation. Before reverse transcription, DNase I (Fermentas) was used to remove the residual DNA. A RevertAid First Strand cDNA Synthesis Kit (Fermentas) was used for reverse transcription, and oligo(dT), random primers, or miRNA-specific reverse transcription primers were added as appropriate. THUNDERBIRD SYBR qPCR Mix (TOYOBO) was used for qPCR, and the results were monitored using a LightCycler 480 II (Roche) system or CFX96 Real-Time PCR Detection System (Bio-Rad). The sequences for PCR primers were as follows: myogenin-F, 5′-CAATGCACTGGAGTTCGGT-3′, myogenin-R, 5′-CTGGGAAGGCAACAGACAT-3′; MyHC-2d-F, 5′-CGCAAGAATGTTCTCAGGCT-3′, MyHC-2d-R, 5′-GCCAGGTTGACATTGGATTG-3′; MCK-F, 5′-GCTTATGGTGGAGATGGAGA-3′, MCK-R, 5′-GGCCATCACGGACTTTTATT-3′; and tubulin-F, 5′-GACTATGGACTCCGTTCGCTC-3′, tubulin-R, 5′-TATTCTTCCCGGATCTTGCTG-3′. The miRNA-specific reverse transcription primers and PCR detection primers for miR-195 and miR-497 were provided by RiboBio.

Immunofluorescence

Fixed cells were incubated with monoclonal anti-myosin (skeletal, fast) antibodies (M4276; Sigma), anti-actin-α1 skeletal muscle antibodies (NB120-7799; Novus Biologicals) or anti-p65 antibodies (sc-372; Santa Cruz Biotechnology) as primary antibodies. Cells were then incubated with Alexa-Fluor-488-conjugated goat anti-mouse-IgG, -IgM (H+L) or Alexa-Fluor-488-conjugated goat anti-rabbit-IgG (H+L) (Invitrogen) secondary antibodies. The cell nuclei were stained with 4′,6-diamidino-2-phenylindole (DAPI). Images were captured using a Nikon ECLIPSE TE2000-S system.

Luciferase activity assay

The following primers were used to amplify the 3′UTR fragments containing predicted miR-195- and miR-497-binding sites: Ccne1 proximal, 5′-CCGCTCGAGGCAGGAGACAGAATGAC-3′ and 5′-ATAAGAATGCGGCCGCGCAACCTACAACACCC-3′, Ccne1 distal, 5′-CCGCTCGAGTGCTCGGGTGTTGTAGGT-3′ and 5′-ATAAGAATGCGGCCGCGGCGGATTTCTGAGTTTG-3′, Ccnd2 proximal, 5′-CCGCTCGAGGCCAGGACCGCATGAGTAGT-3′ and 5′-ATAAGAATGCGGCCGCTGAGACTTAGGAGCCGTTCACC-3′, Ccnd2 distal, 5′-CCGCTCGAGGAACAGGGCTGCTTTGAAGAGT-3′ and 5′-ATAAGAATGCGGCCGCCAGAAGGACAGGGACAGAGGAA-3′, Igf1r 3′UTR-BS, 5′-CCGCTCGAGGGACTTCTTCATGGGTCTCTCA-3′ and 5′-ATAAGAATGCGGCCGCGAAGCATCAGTTGCCGGAGAA-3′, Insr 3′UTR-BS1, 5′-CCGCTCGAGCTGTGGAGGGCTAACTGTGAA-3′ and 5′-ATAAGAATGCGGCCGCGAAGAAGACACCTGAGCAAGAC-3′, and Insr 3′UTR-BS2, 5′-CCGCTCGAGCGGGCAATGGGAATAACAGAA-3′ and 5′-ATAAGAATGCGGCCGCTAAACTATATGATGCTACTTGGCTCT-3′. The restriction enzymes XhoI (Fermentas) and NotI (Fermentas) were used to insert these fragments into the psiCHECK-2 vector (Promega). Additionally, P195-F, 5′-tcaACGCGTGATGCTCCCATGTGAGACTGTC-3′ and P195-R, 5′-tcaGCTAGCGGTCGAGAGGAAGACAGCACAG-3′ were used to amplify the fragment containing predicted p65-binding sites (lowercase letters represent protective bases for the restriction endounclease site), and the restriction enzymes MluI (Fermentas) and NheI (Fermentas) were used to insert this fragment into the PGL3 vector (Promega). pNFκB-TA-luc (Beyotime) was used to reflect the activity variation of NF-κB. A dual luciferase reporter assay system (Promega) was used to analyze the relative luciferase activity.

Transfection

Cells were transfected using Lipofectamine 2000 (Invitrogen) according to the manufacturer's recommendations. miRNA mimics, mutants, and scrambled negative control were provided by RiboBio. The sequences of miRNAs used in this study were as follows: miR-195, 5′-UAGCAGCACAGAAAUAUUGGC-3′; miR-195 mut, 5′-UUCGUCGUCAGAAAUAUUGGC-3′; miR-497, 5′-CAGCAGCACACUGUGGUUUGUA-3′; and miR-497 mut, 5′-CUCGUCGUCACUGUGGUUUGUA-3′. The sequences of siRNAs used in this study were as follows: si-Igf1r-1, 5′-CAAUGAGUACAACUACCGCTT-3′; si-Igf1r-2, 5′-CGACUAUCAGCAGCUGAAGTT-3′; si-Igf1r-3, 5′-CAACGAAGCUUCUGUGAUGUUTT-3′; si-Insr-1, 5′-AACAAGACAGCUGGUACCAGGTT-3′; si-Insr-2, 5′-ACACAACCUCACCAUCACUTT-3′; si-Insr-3, 5′-ACUGCAUGGUUGCCCAUGATT-3′; si-Insr-4, 5′-AGAUGACAACGAGGAAUGUTT-3′; and scrambled negative control, 5′-UUCUCCGAACGUGUCACGUTT-3′. siRNA targeting p65 (sip65) was purchased from Invitrogen. p65-F, 5′-GTCaagcttCCCTGACCATGGACGATCTG-3′ and p65-R, 5′-CATgcggccgCACCTTAGGAGCTGATCTGAC-3′ were used to amplify the coding sequence (CDS) of p65 (lowercase letters represent restriction endounclease site), and the restriction enzymes HindIII (Fermentas) and NotI (Fermentas) were used to insert p65 CDS into the pCDNA3.1(+) vector.

Cell proliferation analysis

Cell cycle flow cytometry

Treated cells were fixed and incubated in propidium iodide solution. The cell cycle phase was then analyzed using a FACSCalibur instrument (Becton Dickinson) and ModFit software (Verity Software House). The proliferation index indicates the proportion of mitotic cells (S-phase cells and G2-phase cells) to total cells.

EdU assay

miR-195/497-F, 5′-cccaagcttaaGGCCATGTTTGCCATTCACAC-3′, and miR-195/497-R, 5′-aaactgcagTGGATCACCTGGGACTCTAAG-3′ (lowercase letters represent the restriction endounclease site and its protective bases), were used to amplify the fragment containing the primary miR-195 and miR-497 cluster transcript, and the restriction enzymes HindIII (Fermentas) and PstI (Fermentas) were used to insert this fragment into the pEGFP-C1 vector. Cells were transfected with the construct miR-195/497-C1 or pEGFP-C1 and incubated in medium containing EdU (Invitrogen). The proportions of EdU- and GFP-positive cells were then determined using flow cytometry.

For experiments using IGF-1 stimulation, cells were transfected with miR-195/497-C1 or pEGFP-C1 for 24 h, stimulated with IGF-1 (100 ng/ml, Sigma) or vehicle (acetic acid) for an additional 12 h, and then subjected to EdU staining and flow cytometry analysis.

Western blotting

Cells protein lysates were obtained using M-PER Mammalian Protein Extraction Reagent (Pierce). SDS-PAGE was used to separate the proteins, and a Mini Trans-Blot Cell (Bio-Rad) was used to transfer protein onto polyvinylidene fluoride (PVDF) membranes (Millipore). Primary antibodies specific for cyclin E1 (Cell Signaling Technology), cyclin D2 (Cell Signaling Technology), IGF1 receptor β (Cell Signaling Technology), insulin receptor β (Cell Signaling Technology), tubulin (Beyotime), and NF-κB p65 (Santa Cruz Biotechnology), as well as horseradish peroxidase (HRP)-labeled anti-rabbit-IgG and anti-mouse-IgG secondary antibodies (Beyotime), were used for immunoblotting. An Image Quant LAS4000 mini (GE Healthcare Bio-Sciences) was used to detect the signal produced by Immobilon Western Chemiluminescent HRP Substrate (Millipore).

EMSAs

Nuclear and cytoplasmic extracts were isolated using an NE-PER Nuclear Protein Extraction Kit (Pierce). The probes were synthesized and double-labeled with FITC (Invitrogen). The sequences of the probes were as follows: probe1, 5′-GCGGGGGAGGGGTTTCCAACGG-3′ and 5′-CCGTTGGAAACCCCTCCCCCGC-3′; probe2, 5′-GGAGGATCGGGCTTTCCTGCTT-3′ and 5′-AAGCAGGAAAGCCCGATCCTCC-3′; probe3, 5′-TCTTGTGGGGGTCCCCCACCCC-3′ and 5′-GGGGTGGGGGACCCCCACAAGA-3′. The reagents and methods used for this experiment were from the EMSA/Gel-Shift kit (Beyotime).

ChIP-PCR

A VCX130 system (Sonics & Materials, Inc.) was used for cell sonication. Anti-NF-κB p65 antibodies (sc-372; Santa Cruz Biotechnology) and normal anti-mouse-IgG agarose-conjugated antibodies (sc-2343; Santa Cruz Biotechnology) were used for the immunoprecipitation reaction. The ChIP Assay kit (P2078) was purchased from Beyotime, and the protocol was performed according to the manufacturer's instructions. The sequences for the following PCR detection primers were as follows: first BS-F, 5′-GTGCTGCTGAAACTGGCAAGGA-3′, first BS-R, 5′-TCTGGCCTGGCTTACCCTCTAG-3′; second BS-F, 5′-GCAGATTCCAGTCCTCCAGGCT-3′, second BS-R, 5′-GCCAAGAGGTCAAGCTCATTGC-3′; third BS-F, 5′-TCTGACTGGGAGTGGAGGAACC-3′, third BS-R: 5′-GGGTGCATCCCTCACATTTGGG-3′.

Statistical analysis

All results are shown as the mean±s.e.m., and at least three independent individuals or replicates were used per group. Unpaired Student's t-tests were used to determine statistical significance, and P<0.05 was considered significantly different.

The authors would like to thank both Dori Miller and Dr Juming Zhong at Auburn University College of Veterinary Medicine, USA for the language proof of this article.

Author contributions

W.W., W.-Y.Z., J.-B.B., H.-X.Z. and Y.-Y.Z. performed the experiments and prepared the materials involved in this study. X.-Y.L. conceived this study. S.-H.Z., X.-Y.L., W.W. and J.-B.B. participated in its design and coordination. X.-Y.L. and W.W. contributed to analysis and interpretation of data. W.W. drafted the manuscript. S.-H.Z. and X.-Y.L. helped to revise the manuscript. All authors read and approved the final manuscript.

Funding

This work was supported by the grants from the NSFC-CGIAR Cooperation project [grant number 31361140365], the National foundation of NSFC [grant number 31372291], the Chinese National Major Special Project on New Varieties Cultivation for Transgenic Organisms [grant number 2014ZX0800950B] and the Fundamental Research Funds for the Central Universities of China [grant number 2013PY050].

Ai
,
J.
,
Sun
,
L.-H.
,
Che
,
H.
,
Zhang
,
R.
,
Zhang
,
T.-Z.
,
Wu
,
W.-C.
,
Su
,
X.-L.
,
Chen
,
X.
,
Yang
,
G.
,
Li
,
K.
, et al. 
(
2013
).
MicroRNA-195 protects against dementia induced by chronic brain hypoperfusion via its anti-amyloidogenic effect in rats
.
J. Neurosci.
33
,
3989
-
4001
.
Allen
,
R. E.
,
Luiten
,
L. S.
and
Dodson
,
M. V.
(
1985
).
Effect of insulin and linoleic acid on satellite cell differentiation
.
J. Anim. Sci.
60
,
1571
-
1579
.
Arabkhari
,
M.
,
Bunda
,
S.
,
Wang
,
Y.
,
Wang
,
A.
,
Pshezhetsky
,
A. V.
and
Hinek
,
A.
(
2010
).
Desialylation of insulin receptors and IGF-1 receptors by neuraminidase-1 controls the net proliferative response of L6 myoblasts to insulin
.
Glycobiology
20
,
603
-
616
.
Bai
,
Y.
,
Yang
,
W.
,
Yang
,
H.-X.
,
Liao
,
Q.
,
Ye
,
G.
,
Fu
,
G.
,
Ji
,
L.
,
Xu
,
P.
,
Wang
,
H.
,
Li
,
Y.-X.
, et al. 
(
2012
).
Downregulated miR-195 detected in preeclamptic placenta affects trophoblast cell invasion via modulating ActRIIA expression
.
PLoS ONE
7
,
e38875
.
Bai
,
L.
,
Liang
,
R.
,
Yang
,
Y.
,
Hou
,
X.
,
Wang
,
Z.
,
Zhu
,
S.
,
Wang
,
C.
,
Tang
,
Z.
and
Li
,
K.
(
2015
).
MicroRNA-21 regulates PI3K/Akt/mTOR signaling by targeting TGFbetaI during skeletal muscle development in pigs
.
PLoS ONE
10
,
e0119396
.
Bakkar
,
N.
,
Wang
,
J.
,
Ladner
,
K. J.
,
Wang
,
H.
,
Dahlman
,
J. M.
,
Carathers
,
M.
,
Acharyya
,
S.
,
Rudnicki
,
M. A.
,
Hollenbach
,
A. D.
and
Guttridge
,
D. C.
(
2008
).
IKK/NF-kappaB regulates skeletal myogenesis via a signaling switch to inhibit differentiation and promote mitochondrial biogenesis
.
J. Cell Biol.
180
,
787
-
802
.
Barrey
,
E.
,
Bonnamy
,
B.
,
Barrey
,
E. J.
,
Mata
,
X.
,
Chaffaux
,
S.
and
Guerin
,
G.
(
2011
).
Muscular microRNA expressions in healthy and myopathic horses suffering from polysaccharide storage myopathy or recurrent exertional rhabdomyolysis
.
Equine Vet. J.
42 (suppl. 38),
303
-
310
.
Bonnesen
,
C.
,
Nelander
,
G.-M.
,
Hansen
,
B. F.
,
Jensen
,
P.
,
Krabbe
,
J. S.
,
Jensen
,
M. B.
,
Hegelund
,
A. C.
,
Svendsen
,
J. E.
and
Oleksiewicz
,
M. B.
(
2010
).
Synchronization in G0/G1 enhances the mitogenic response of cells overexpressing the human insulin receptor A isoform to insulin
.
Cell Biol. Toxicol.
26
,
293
-
307
.
Cassar-Malek
,
I.
,
Langlois
,
N.
,
Picard
,
B.
and
Geay
,
Y.
(
1999
).
Regulation of bovine satellite cell proliferation and differentiation by insulin and triiodothyronine
.
Domestic Anim. Endocrinol.
17
,
373
-
388
.
Castillo
,
J.
,
Codina
,
M.
,
Martinez
,
M. L.
,
Navarro
,
I.
and
Gutierrez
,
J.
(
2004
).
Metabolic and mitogenic effects of IGF-I and insulin on muscle cells of rainbow trout
.
Am. J. Physiol. Regul. Integr. Comp. Physiol.
286
,
R935
-
R941
.
Cen
,
S.
,
Zhang
,
J.
,
Huang
,
F.
,
Yang
,
Z.
and
Xie
,
H.
(
2008
).
[Effect of IGF-1 on proliferation and differentiation of primary human embryonic myoblasts]
.
Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi
22
,
84
-
87
.
Chen
,
J.-F.
,
Mandel
,
E. M.
,
Thomson
,
J. M.
,
Wu
,
Q.
,
Callis
,
T. E.
,
Hammond
,
S. M.
,
Conlon
,
F. L.
and
Wang
,
D.-Z.
(
2006
).
The role of microRNA-1 and microRNA-133 in skeletal muscle proliferation and differentiation
.
Nat. Genet.
38
,
228
-
233
.
Cimmino
,
A.
,
Calin
,
G. A.
,
Fabbri
,
M.
,
Iorio
,
M. V.
,
Ferracin
,
M.
,
Shimizu
,
M.
,
Wojcik
,
S. E.
,
Aqeilan
,
R. I.
,
Zupo
,
S.
,
Dono
,
M.
, et al. 
(
2005
).
miR-15 and miR-16 induce apoptosis by targeting BCL2
.
Proc. Natl. Acad. Sci. USA
102
,
13944
-
13949
.
Crown
,
A. L.
,
He
,
X. L.
,
Holly
,
J. M.
,
Lightman
,
S. L.
and
Stewart
,
C. E.
(
2000
).
Characterisation of the IGF system in a primary adult human skeletal muscle cell model, and comparison of the effects of insulin and IGF-I on protein metabolism
.
J. Endocrinol.
167
,
403
-
415
.
Deng
,
H.
,
Guo
,
Y.
,
Song
,
H.
,
Xiao
,
B.
,
Sun
,
W.
,
Liu
,
Z.
,
Yu
,
X.
,
Xia
,
T.
,
Cui
,
L.
and
Guo
,
J.
(
2013
).
MicroRNA-195 and microRNA-378 mediate tumor growth suppression by epigenetical regulation in gastric cancer
.
Gene
518
,
351
-
359
.
Diaz-Beya
,
M.
,
Navarro
,
A.
,
Ferrer
,
G.
,
Diaz
,
T.
,
Gel
,
B.
,
Camos
,
M.
,
Pratcorona
,
M.
,
Torrebadell
,
M.
,
Rozman
,
M.
,
Colomer
,
D.
, et al. 
(
2012
).
Acute myeloid leukemia with translocation (8;16)(p11;p13) and MYST3-CREBBP rearrangement harbors a distinctive microRNA signature targeting RET proto-oncogene
.
Leukemia
27
,
595
-
603
.
Ding
,
J.
,
Huang
,
S.
,
Wang
,
Y.
,
Tian
,
Q.
,
Zha
,
R.
,
Shi
,
H.
,
Wang
,
Q.
,
Ge
,
C.
,
Chen
,
T.
,
Zhao
,
Y.
, et al. 
(
2013
).
Genome-wide screening reveals that miR-195 targets the TNF-alpha/NF-kappaB pathway by down-regulating IkappaB kinase alpha and TAB3 in hepatocellular carcinoma
.
Hepatology
58
,
654
-
666
.
Dong
,
Y.
,
Lakhia
,
R.
,
Thomas
,
S. S.
,
Dong
Y.
,
Wang
,
X. H.
,
Silva
,
K. A. S.
and
Zhang
,
L.
(
2013
).
Interactions between p-Akt and Smad3 in injured muscles initiate myogenesis or fibrogenesis
.
Am. J. Physiol. Endocrinol. Metab.
305
,
E367
-
E375
.
Engert
,
J. C.
,
Berglund
,
E. B.
and
Rosenthal
,
N.
(
1996
).
Proliferation precedes differentiation in IGF-I-stimulated myogenesis
.
J. Cell Biol.
135
,
431
-
440
.
Fryburg
,
D. A.
(
1994
).
Insulin-like growth factor I exerts growth hormone- and insulin-like actions on human muscle protein metabolism
.
Am. J. Physiol.
267
,
E331
-
E336
.
Furuta
,
M.
,
Kozaki
,
K.-I.
,
Tanimoto
,
K.
,
Tanaka
,
S.
,
Arii
,
S.
,
Shimamura
,
T.
,
Niida
,
A.
,
Miyano
,
S.
and
Inazawa
,
J.
(
2013
).
The tumor-suppressive miR-497–195 cluster targets multiple cell-cycle regulators in hepatocellular carcinoma
.
PLoS ONE
8
,
e60155
.
Grabiec
,
K.
,
Gajewska
,
M.
,
Milewska
,
M.
,
Blaszczyk
,
M.
and
Grzelkowska-Kowalczyk
,
K.
(
2014
).
The influence of high glucose and high insulin on mechanisms controlling cell cycle progression and arrest in mouse C2C12 myoblasts: the comparison with IGF-I effect
.
J. Endocrinol. Invest.
37
,
233
-
245
.
Guo
,
S. T.
,
Jiang
,
C. C.
,
Wang
,
G. P.
,
Li
,
Y. P.
,
Wang
,
C. Y.
,
Guo
,
X. Y.
,
Yang
,
R. H.
,
Feng
,
Y.
,
Wang
,
F. H.
,
Tseng
,
H.-Y.
, et al. 
(
2012
).
MicroRNA-497 targets insulin-like growth factor 1 receptor and has a tumour suppressive role in human colorectal cancer
.
Oncogene
32
,
1910
-
1920
.
Guttridge
,
D. C.
,
Albanese
,
C.
,
Reuther
,
J. Y.
,
Pestell
,
R. G.
and
Baldwin
,
A. S.
 Jr
. (
1999
).
NF-kappaB controls cell growth and differentiation through transcriptional regulation of cyclin D1
.
Mol. Cell. Biol.
19
,
5785
-
5799
.
Guttridge
,
D. C.
,
Mayo
,
M. W.
,
Madrid
,
L. V.
,
Wang
,
C.-Y.
and
Baldwin
,
A. S.
 Jr
. (
2000
).
NF-kappaB-induced loss of MyoD messenger RNA: possible role in muscle decay and cachexia
.
Science
289
,
2363
-
2366
.
Hu
,
Y.
,
Li
,
P.
,
Hao
,
S.
,
Liu
,
L.
,
Zhao
,
J.
and
Hou
,
Y.
(
2009
).
Differential expression of microRNAs in the placentae of Chinese patients with severe pre-eclampsia
.
Clin. Chem. Lab. Med.
47
,
923
-
929
.
Huang
,
T.-H.
,
Zhu
,
M.-J.
,
Li
,
X.-Y.
and
Zhao
,
S.-H.
(
2008
).
Discovery of porcine microRNAs and profiling from skeletal muscle tissues during development
.
PLoS ONE
3
,
e3225
.
Hui
,
W.
,
Yuntao
,
L.
,
Lun
,
L.
,
WenSheng
,
L.
,
ChaoFeng
,
L.
,
HaiYong
,
H.
and
Yueyang
,
B.
(
2013
).
MicroRNA-195 inhibits the proliferation of human glioma cells by directly targeting cyclin D1 and cyclin E1
.
PLoS ONE
8
,
e54932
.
Itesako
,
T.
,
Seki
,
N.
,
Yoshino
,
H.
,
Chiyomaru
,
T.
,
Yamasaki
,
T.
,
Hidaka
,
H.
,
Yonezawa
,
T.
,
Nohata
,
N.
,
Kinoshita
,
T.
,
Nakagawa
,
M.
, et al. 
(
2014
).
The microRNA expression signature of bladder cancer by deep sequencing: the functional significance of the miR-195/497 cluster
.
PLoS ONE
9
,
e84311
.
Joglekar
,
M. V.
,
Parekh
,
V. S.
,
Mehta
,
S.
,
Bhonde
,
R. R.
and
Hardikar
,
A. A.
(
2007
).
MicroRNA profiling of developing and regenerating pancreas reveal post-transcriptional regulation of neurogenin3
.
Dev. Biol.
311
,
603
-
612
.
Kim
,
H.
,
Barton
,
E.
,
Muja
,
N.
,
Yakar
,
S.
,
Pennisi
,
P.
and
LeRoith
,
D.
(
2005
).
Intact insulin and insulin-like growth factor-I receptor signaling is required for growth hormone effects on skeletal muscle growth and function in vivo
.
Endocrinology
146
,
1772
-
1779
.
Kim
,
H. K.
,
Lee
,
Y. S.
,
Sivaprasad
,
U.
,
Malhotra
,
A.
and
Dutta
,
A.
(
2006
).
Muscle-specific microRNA miR-206 promotes muscle differentiation
.
J. Cell Biol.
174
,
677
-
687
.
Langen
,
R. C. J.
,
Schols
,
A. M. W. J.
,
Kelders
,
M. C. J. M.
,
Wouters
,
E. F. M.
and
Janssen-Heininger
,
Y. M. W.
(
2001
).
Inflammatory cytokines inhibit myogenic differentiation through activation of nuclear factor-kappaB
.
FASEB J.
15
,
1169
-
1180
.
Lee
,
R. C.
,
Feinbaum
,
R. L.
and
Ambros
,
V.
(
1993
).
The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14
.
Cell
75
,
843
-
854
.
Li
,
D.
,
Zhao
,
Y.
,
Liu
,
C.
,
Chen
,
X.
,
Qi
,
Y.
,
Jiang
,
Y.
,
Zou
,
C.
,
Zhang
,
X.
,
Liu
,
S.
,
Wang
,
X.
, et al. 
(
2011
).
Analysis of MiR-195 and MiR-497 expression, regulation and role in breast cancer
.
Clin. Cancer Res.
17
,
1722
-
1730
.
Lin
,
Y.
,
Wu
,
J.
,
Chen
,
H.
,
Mao
,
Y.
,
Liu
,
Y.
,
Mao
,
Q.
,
Yang
,
K.
,
Zheng
,
X.
and
Xie
,
L.
(
2012
).
Cyclin-dependent kinase 4 is a novel target in micoRNA-195-mediated cell cycle arrest in bladder cancer cells
.
FEBS Lett.
586
,
442
-
447
.
Liu
,
C.
,
Teng
,
Z.-Q.
,
McQuate
,
A. L.
,
Jobe
,
E. M.
,
Christ
,
C. C.
,
von Hoyningen-Huene
,
S. J.
,
Reyes
,
M. D.
,
Polich
,
E. D.
,
Xing
,
Y.
,
Li
,
Y.
, et al. 
(
2013
).
An epigenetic feedback regulatory loop involving microRNA-195 and MBD1 governs neural stem cell differentiation
.
PLoS ONE
8
,
e51436
.
Luo
,
Q.
,
Wei
,
C.
,
Li
,
X.
,
Li
,
J.
,
Chen
,
L.
,
Huang
,
Y.
,
Song
,
H.
,
Li
,
D.
and
Fang
,
L.
(
2014
).
MicroRNA-195-5p is a potential diagnostic and therapeutic target for breast cancer
.
Oncol. Rep.
31
,
1096
-
1102
.
McCarthy
,
J. J.
,
Esser
,
K. A.
,
Peterson
,
C. A.
and
Dupont-Versteegden
,
E. E.
(
2009
).
Evidence of MyomiR network regulation of beta-myosin heavy chain gene expression during skeletal muscle atrophy
.
Physiol. Genomics
39
,
219
-
226
.
Mott
,
J. L.
,
Kurita
,
S.
,
Cazanave
,
S. C.
,
Bronk
,
S. F.
,
Werneburg
,
N. W.
and
Fernandez-Zapico
,
M. E.
(
2010
).
Transcriptional suppression of mir-29b-1/mir-29a promoter by c-Myc, hedgehog, and NF-kappaB
.
J. Cell. Biochem.
110
,
1155
-
1164
.
Ortega
,
F. J.
,
Mercader
,
J. M.
,
Moreno-Navarrete
,
J. M.
,
Rovira
,
O.
,
Guerra
,
E.
,
Esteve
,
E.
,
Xifra
,
G.
,
Martinez
,
C.
,
Ricart
,
W.
,
Rieusset
,
J.
, et al. 
(
2014
).
Profiling of circulating microRNAs reveals common microRNAs linked to type 2 diabetes that change with insulin sensitization
.
Diabetes Care
37
,
1375
-
1383
.
Ozata
,
D. M.
,
Caramuta
,
S.
,
Velazquez-Fernandez
,
D.
,
Akcakaya
,
P.
,
Xie
,
H.
,
Hoog
,
A.
,
Zedenius
,
J.
,
Backdahl
,
M.
,
Larsson
,
C.
and
Lui
,
W.-O.
(
2011
).
The role of microRNA deregulation in the pathogenesis of adrenocortical carcinoma
.
Endocr. Relat. Cancer
18
,
643
-
655
.
Porrello
,
E. R.
,
Johnson
,
B. A.
,
Aurora
,
A. B.
,
Simpson
,
E.
,
Nam
,
Y.-J.
,
Matkovich
,
S. J.
,
Dorn
,
G. W.
, II
,
van Rooij
,
E.
and
Olson
,
E. N.
(
2011
).
MiR-15 family regulates postnatal mitotic arrest of cardiomyocytes
.
Circ. Res.
109
,
670
-
679
.
Quinn
,
L. S.
and
Haugk
,
K. L.
(
1996
).
Overexpression of the type-1 insulin-like growth factor receptor increases ligand-dependent proliferation and differentiation in bovine skeletal myogenic cultures
.
J. Cell. Physiol.
168
,
34
-
41
.
Quinn
,
L. S.
and
Roh
,
J. S.
(
1993
).
Overexpression of the human type-1 insulin-like growth factor receptor in rat L6 myoblasts induces ligand-dependent cell proliferation and inhibition of differentiation
.
Exp. Cell Res.
208
,
504
-
508
.
Sato
,
T.
,
Yamamoto
,
T.
and
Sehara-Fujisawa
,
A.
(
2014
).
miR-195/497 induce postnatal quiescence of skeletal muscle stem cells
.
Nat. Commun.
5
,
4597
.
Singh
,
R.
and
Saini
,
N.
(
2012
).
Downregulation of BCL2 by miRNAs augments drug-induced apoptosis--a combined computational and experimental approach
.
J. Cell Sci.
125
,
1568
-
1578
.
van Rooij
,
E.
,
Sutherland
,
L. B.
,
Liu
,
N.
,
Williams
,
A. H.
,
McAnally
,
J.
,
Gerard
,
R. D.
,
Richardson
,
J. A.
and
Olson
,
E. N.
(
2006
).
A signature pattern of stress-responsive microRNAs that can evoke cardiac hypertrophy and heart failure
.
Proc. Natl. Acad. Sci. USA
103
,
18255
-
18260
.
Wang
,
H.
,
Garzon
,
R.
,
Sun
,
H.
,
Ladner
,
K. J.
,
Singh
,
R.
,
Dahlman
,
J.
,
Cheng
,
A.
,
Hall
,
B. M.
,
Qualman
,
S. J.
,
Chandler
,
D. S.
, et al. 
(
2008
).
NF-kappaB-YY1-miR-29 regulatory circuitry in skeletal myogenesis and rhabdomyosarcoma
.
Cancer Cell
14
,
369
-
381
.
Waters
,
P. S.
,
McDermott
,
A. M.
,
Wall
,
D.
,
Heneghan
,
H. M.
,
Miller
,
N.
,
Newell
,
J.
,
Kerin
,
M. J.
and
Dwyer
,
R. M.
(
2012
).
Relationship between circulating and tissue microRNAs in a murine model of breast cancer
.
PLoS ONE
7
,
e50459
.
Wightman
,
B.
,
Ha
,
I.
and
Ruvkun
,
G.
(
1993
).
Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans
.
Cell
75
,
855
-
862
.
Xu
,
T.
,
Zhu
,
Y.
,
Xiong
,
Y.
,
Ge
,
Y.-Y.
,
Yun
,
J.-P.
and
Zhuang
,
S.-M.
(
2009
).
MicroRNA-195 suppresses tumorigenicity and regulates G1/S transition of human hepatocellular carcinoma cells
.
Hepatology
50
,
113
-
121
.
Xu
,
J.
,
Li
,
R.
,
Workeneh
,
B.
,
Dong
,
Y.
,
Wang
,
X.
and
Hu
,
Z.
(
2012
).
Transcription factor FoxO1, the dominant mediator of muscle wasting in chronic kidney disease, is inhibited by microRNA-486
.
Kidney Int.
82
,
401
-
411
.
Yang
,
Y.
,
Sun
,
W.
,
Wang
,
R.
,
Lei
,
C.
,
Zhou
,
R.
,
Tang
,
Z.
and
Li
,
K.
(
2015
).
Wnt antagonist, secreted frizzled-related protein 1, is involved in prenatal skeletal muscle development and is a target of miRNA-1/206 in pigs
.
BMC Mol. Biol.
16
,
4
.
Zhu
,
H.
,
Yang
,
Y.
,
Wang
,
Y.
,
Li
,
J.
,
Schiller
,
P. W.
and
Peng
,
T.
(
2011
).
MicroRNA-195 promotes palmitate-induced apoptosis in cardiomyocytes by down-regulating Sirt1
.
Cardiovasc. Res.
92
,
75
-
84
.
Zhu
,
H.-C.
,
Wang
,
L.-M.
,
Wang
,
M.
,
Song
,
B.
,
Tan
,
S.
,
Teng
,
J.-F.
and
Duan
,
D.-X.
(
2012
).
MicroRNA-195 downregulates Alzheimer's disease amyloid-beta production by targeting BACE1
.
Brain Res. Bull.
88
,
596
-
601
.

Competing interests

The authors declare no competing or financial interests.