scholarly journals Styrene Oxide Caused Cell Cycle Arrest and Abolished Myogenic Differentiation of C2C12 Myoblasts

2020 ◽  
Vol 2020 ◽  
pp. 1-11
Author(s):  
Piyaporn Surinlert ◽  
Nitchamon Kongthong ◽  
Mariam Watthanard ◽  
Thannicha Sae-lao ◽  
Piyawat Sookbangnop ◽  
...  

Contaminations of chemicals in foods and drinks are raising public concerns. Among these, styrene, a monomer for plastic production, receives increasing interest due to its ability to leach from the packaging and contaminate in foods and drinks causing many health problems. The present study was designed to investigate the effects of styrene monomer (STR) and its metabolite styrene oxide (STO) on C2C12 myoblast proliferation and differentiation. Based on an MTT assay, both STR and STO showed no cytotoxic effect at 10–100 μM. However, at 50–100 μM STO, but not STR, significantly inhibited cell proliferation. The STO-treated cells were accumulated in S-phase of cell cycles as revealed by flow cytometry. The antioxidant enzyme (catalase and superoxide dismutase) activities and the gene expressing these enzymes of the arrested cells were decreased and ultimately led to nuclear condensation and expression of apoptotic markers such as cleaved caspase-3 and-9, but not cleaved caspase-8. In addition, STO significantly suppressed myogenic differentiation by decreasing both the number and size of differentiated myotubes. Biochemical analysis showed attenuations of total protein synthesis and myosin heavy chain (MHC) protein expression. In conclusion, a metabolite of styrene, STO, leached from plastic packaging of foods and beverages suppressed both myoblast proliferation and differentiation, which would affect skeletal muscle development and regeneration.

Author(s):  
Hyunju Liu ◽  
Su-Mi Lee ◽  
Hosouk Joung

AbstractSUMOylation is one of the post-translational modifications that involves the covalent attachment of the small ubiquitin-like modifier (SUMO) to the substrate. SUMOylation regulates multiple biological processes, including myoblast proliferation, differentiation, and apoptosis. 2-D08 is a synthetically available flavone, which acts as a potent cell-permeable SUMOylation inhibitor. Its mechanism of action involves preventing the transfer of SUMO from the E2 thioester to the substrate without influencing SUMO-activating enzyme E1 (SAE-1/2) or E2 Ubc9-SUMO thioester formation. However, both the effects and mechanisms of 2-D08 on C2C12 myoblast cells remain unclear. In the present study, we found that treatment with 2-D08 inhibits C2C12 cell proliferation and differentiation. We confirmed that 2-D08 significantly hampers the viability of C2C12 cells. Additionally, it inhibited myogenic differentiation, decreasing myosin heavy chain (MHC), MyoD, and myogenin expression. Furthermore, we confirmed that 2-D08-mediated anti-myogenic effects impair myoblast differentiation and myotube formation, reducing the number of MHC-positive C2C12 cells. In addition, we found that 2-D08 induces the activation of ErK1/2 and the degradation of MyoD and myogenin in C2C12 cells. Taken together, these results indicated that 2-D08 treatment results in the deregulated proliferation and differentiation of myoblasts. However, further research is needed to investigate the long-term effects of 2-D08 on skeletal muscles.


2021 ◽  
Vol 12 ◽  
Author(s):  
Lingzhi Yang ◽  
Qi Qi ◽  
Jian Wang ◽  
Chengchuang Song ◽  
Yanhong Wang ◽  
...  

microRNAs are a kind of endogenous, non-coding, single-strand small RNA. They have been reported as an important regulatory factor in skeletal myogenesis. In this study, miR-452 was selected from RNA high-throughput sequencing data to explore its regulatory role in myogenesis. Functionally, miR-452 overexpression could promote C2C12 myoblast proliferation while inhibiting myogenic differentiation. On the contrary, inhibition of miR-452 could suppress C2C12 myoblast proliferation but accelerate myogenic differentiation. Bioinformatics analysis and dual luciferase report assays showed that Angiopoietin 1 (ANGPT1), RB1, and CACNB4 were the potential target genes of miR-452. To further confirm the target relationship between ANGPT1, RB1, and CACNB4 with miR-452, the mRNA level and protein level of these genes were detected by using RT-qPCR and Western blot, respectively. Result analysis indicated that ANGPT1 was a target gene of miR-452. In addition, knockdown of ANGPT1 could obviously promote C2C12 myoblast proliferation but block their differentiation. In summary, these results demonstrated that miR-452 promoted C2C12 myoblast proliferation and inhibited their differentiation via targeting ANGPT1.


2021 ◽  
Vol 12 (2) ◽  
Author(s):  
Xuemei Shen ◽  
Jia Tang ◽  
Rui Jiang ◽  
Xiaogang Wang ◽  
Zhaoxin Yang ◽  
...  

AbstractMany novel non-coding RNAs, such as microRNAs (miRNAs) and circular RNAs (circRNAs), are involved in various physiological and pathological processes. The PI3K/AKT signaling pathway is important for its role in regulating skeletal muscle development. In this study, molecular and biochemical assays were used to confirm the role of miRNA-145 (miR-145) in myoblast proliferation and apoptosis. Based on sequencing data and bioinformatics analysis, we identified a new circRILPL1, which acts as a sponge for miR-145. The interactions between circRILPL1 and miR-145 were examined by bioinformatics, a luciferase assay, and RNA immunoprecipitation. Mechanistically, knockdown or exogenous expression of circRILPL1 in the primary myoblasts was performed to prove the functional significance of circRILPL1. We investigated the inhibitory effect of miR-145 on myoblast proliferation by targeting IGF1R to regulate the PI3K/AKT signaling pathway. A novel circRILPL1 was identified that could sponge miR-145 and is related to AKT activation. In addition, circRILPL1 was positively correlated with muscle proliferation and differentiation in vitro and could inhibit cell apoptosis. The newly identified circRILPL1 functions as a miR-145 sponge to regulate the IGF1R gene and rescue the inhibitory effect of miR-145 on the PI3K/AKT signaling pathway, thereby promoting myoblast growth.


2018 ◽  
Vol 97 (4) ◽  
pp. 257-268 ◽  
Author(s):  
Han Wang ◽  
Qian Zhang ◽  
BinBin Wang ◽  
WangJun Wu ◽  
Julong Wei ◽  
...  

2007 ◽  
Vol 179 (1) ◽  
pp. 129-138 ◽  
Author(s):  
Luguo Sun ◽  
Kewei Ma ◽  
Haixia Wang ◽  
Fang Xiao ◽  
Yan Gao ◽  
...  

Skeletal muscle stem cell–derived myoblasts are mainly responsible for postnatal muscle growth and injury-induced muscle regeneration. However, the cellular signaling pathways controlling the proliferation and differentiation of myoblasts are not fully understood. We demonstrate that Janus kinase 1 (JAK1) is required for myoblast proliferation and that it also functions as a checkpoint to prevent myoblasts from premature differentiation. Deliberate knockdown of JAK1 in both primary and immortalized myoblasts induces precocious myogenic differentiation with a concomitant reduction in cell proliferation. This is caused, in part, by an accelerated induction of MyoD, myocyte enhancer–binding factor 2 (MEF2), p21Cip1, and p27Kip1, a faster down-regulation of Id1, and an increase in MEF2-dependent gene transcription. Downstream of JAK1, of all the signal transducer and activator of transcriptions (STATs) present in myoblasts, we find that only STAT1 knockdown promotes myogenic differentiation in both primary and immortalized myoblasts. Leukemia inhibitory factor stimulates myoblast proliferation and represses differentiation via JAK1–STAT1–STAT3. Thus, JAK1–STAT1–STAT3 constitutes a signaling pathway that promotes myoblast proliferation and prevents premature myoblast differentiation.


2020 ◽  
Author(s):  
Li Li ◽  
Xiao Zhang ◽  
Hailong Yang ◽  
Xiaoli Xu ◽  
Yuan Chen ◽  
...  

Abstract BackgroundAs a well-known cancer-related miRNA, miR-193b-3p is enriched in skeletal muscle but dysregulated in muscle disease. However, mechanism underpinning has not been addressed so far. MethodsHere, we probed the impact of miR-193b-3p on myogenesis by mainly using goat tissues and skeletal muscle satellite cells (MuSCs), with combined methods including RNA-seq to profile the transcriptome affected by miR-193b-3p, cell-counting kit-8 (CCK-8) and 5-ethynyl-2'-deoxyuridine (EdU) for cell proliferation assay, and RNA-RNA dual-labeled fluorescence in situ hybridization (FISH) for RNA colocalization. ResultsmiR-193b-3p is highly enriched in goat skeletal muscles, and ectopic miR-193b-3p promotes MuSCs proliferation and differentiation. Moreover, insulin-like growth factor-2 mRNA-binding protein 1 (IGF2BP1) is the most activated insulin signaling genes when overexpression miR-193b-3p and the miRNA recognition element (MRE) within IGF1BP1 3ʹ untranslated region (UTR) is indispensable for its activation caused by miR-193b-3p. Consistently, expression patterns and function of IGF2BP1 were similar to those of miR-193b-3p in tissues and MuSCs. While the overexpression of miR-193b-3p failed to induce pax7 expression and myoblast proliferation when IGF2BP1 knockdown. Furthermore, miR-193b-3p destabilized IGF2BP1 mRNA but unexpectedly promoted levels of IGF2BP1 heteronuclear RNA (hnRNA) dramatically. Moreover, miR-193b-3p could enhance fly luciferase activity when inserted upstream of its promoter, and induce neighboring genes of itself. However, miR-193b-3p inversely regulated IGF2BP1 and myoblast proliferation in mouse C2C12 myoblast. These data unveil that goat miR-193b-3p promotes myoblast proliferation via activating IGF2BP1 by binding on its 3ʹ UTR.ConclusionsOur novel findings highlight the positive regulation between miRNA and its target genes in muscle development, which further extends the repertoire of miRNA functions.


Author(s):  
Tomoya Kitakaze ◽  
Meiku Oshimo ◽  
Yasuyuki Kobayashi ◽  
Mizuyuki Ryu ◽  
Yasushi Suzuki ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document