• Title/Summary/Keyword: C2C12 skeletal muscle cell

Search Result 38, Processing Time 0.026 seconds

Potential role of exercise-induced glucose-6-phosphate isomerase in skeletal muscle function

  • Kwak, Seong Eun;Shin, Hyung Eun;Zhang, Di Di;Lee, Jihyun;Yoon, Kyung Jin;Bae, Jun Hyun;Moon, Hyo Youl;Song, Wook
    • Korean Journal of Exercise Nutrition
    • /
    • v.23 no.2
    • /
    • pp.28-33
    • /
    • 2019
  • [Purpose] Recent studies have shown that glucose-6-phosphate isomerase (GPI)-which is a glycolysis interconversion enzyme-reduces oxidative stress. However, these studies are limited to tumors such as fibrosarcoma, and there are no studies that have examined the effects of exercise on GPI expression in mice skeletal muscle. Furthermore, GPI acts in an autocrine manner thorough its receptor, autocrine motility factor receptor (AMFR); therefore, we investigated expression level changes of secreted GPI from skeletal muscle in in vitro study to examine the potential role of GPI on skeletal muscle. [Methods] First, we performed an in vitro study, to identify the condition that upregulates GPI levels in skeletal muscle cells; we treated C2C12 muscle cells with an exercise-mimicking chemical, AICAR. AICAR treatment upregulated GPI expression level in C2C12 cell and its secretomes. To confirm the direct effect of GPI on skeletal muscle cells, we treated C2C12 cells with GPI recombinant protein. [Results] We found that GPI improved the viability of C2C12 cells. In the in vivo study, the exercise-treated mice group showed upregulated GPI expression in skeletal muscle. Based on the in vitro study results, we speculated that expression level of GPI in skeletal muscle might be associated with muscle function. We analyzed the association between GPI expression level and the grip strength of the all mice group. The mice group's grip strengths were upregulated after 2 weeks of treadmill exercise, and GPI expression level positively correlated with the grip strength. [Conclusion] These results suggested that the exercise-induced GPI expression in skeletal muscle might have a positive effect on skeletal muscle function.

Insulin-like Growth Factor-I Modulates BDNF Expression by Inhibition of Histone Deacetylase in C2C12 Skeletal Muscle Cells (C2C12 골격근 세포에서 히스톤 탈 아세틸 효소의 억제가 인슐린 유사성장인자(IGF-I)에 의한 BDNF 발현 조절에 미치는 영향)

  • Kim, Hye Jin;Lee, Won Jun
    • Journal of Life Science
    • /
    • v.27 no.8
    • /
    • pp.879-887
    • /
    • 2017
  • It is well established that brain-derived neurotrophic factor (BDNF) is expressed not only in the brain but also in skeletal muscle, and is required for normal neuromuscular system function. Histone deacetylases (HDACs) and insulin-like growth factor-I (IGF-I) are potent regulators of skeletal muscle myogenesis and muscle gene expression, but the mechanisms of HDAC and IGF-I in skeletal muscle-derived BDNF expression have not been examined. In this study, we examined the effect of IGF-I and suberoylanilide hydroxamic acid (SAHA), a pan-HDAC inhibitor, on BDNF induction. Proliferating or differentiating C2C12 skeletal muscle cells were treated with increasing concentrations (0-50 ng/ml) of IGF-I in the absence or presence of $5{\mu}M$ SAHA for various time periods (3-24 hr). Treatment of C2C12 cells with IGF-I resulted in a dose- and time-dependent decrease in BDNF mRNA expression. However, inhibition of HDAC led to a significant increase in the expression of BDNF mRNA levels. In addition, immunocytochemistry revealed high BDNF protein levels in undifferentiated C2C12 skeletal muscle cells, whether untreated, IGF-I-treated, or exposed to SAHA. These results represent the first evidence that IGF-I can suppress the mRNA and protein expression of BDNF; conversely, SAHA attenuates the effects of IGF-I. Consequently, SAHA upregulates BDNF expression in C2C12 skeletal muscle cells.

Effects of Chaenomelis Fructus Extract on the regulation of myoblasts differentiation and the expression of biogenetic factors in C2C12 myotubes (모과추출물의 C2C12 근육세포에서 근분화 및 에너지대사조절인자 발현 증진 효과 연구)

  • Kang, Seok Yong;Hyun, Sun Young;Kwon, Yedam;Park, Yong-Ki;Jung, Hyo Won
    • The Korea Journal of Herbology
    • /
    • v.34 no.6
    • /
    • pp.99-107
    • /
    • 2019
  • Objective : The present study was conducted to investigate the effects of Chaenomelis Fructus (CF) on the regulation of biogenesis in C2C12 mouse skeletal muscle cells. Methods : C2C12 myoblasts were differentiated into myotubes in 2% horse serum-containing medium for 5 days, and then treated with CF extract at different concentrations for 48 hr. The expression of muscle differentiation markers, myogenin and myosin heavy chain (MHC) and mitochondrial biogenesis-regulating factors, peroxisome proliferator-activated receptor gamma coactivator 1alpha (PGC1α), sirtuin1 (Sirt1), nuclear respiratory factor1 (NRF1) and transcription factor A, mitochondrial (TFAM), and the phosphorylation of AMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase (ACC) were determined in C2C12 myotubes by reverse transcriptase (RT)-polymerase chain reaction (RT-PCR) and western blot, respectively. The cellular glucose levels and total ATP contents were measured by cellular glucose uptake and ATP assays, respectively. Results : Treatment with CF extract (0.01, 0.02, and 0.05 mg/㎖) significantly increased the expression of MHC protein in C2C12 myotubes compared with non-treated cells. CF extract significantly increased the expression of PGC1α and TFAM in the myotubes. Also, CF extract significantly increased glucose uptake levels and ATP contents in the myotubes. Conclusion : CF extract can stimulate C2C12 myoblasts differentiation into myotubes and increase energy production through upregulation of the expression of mitochondrial biogenetic factors in C2C12 mouse skeletal muscle cell. This suggests that CF can help to improve skeletal muscle function with stimulation of the energy metabolism.

Inhibition of mitochondrial activity induces muscle fiber type change from slow to fast in C2C12 myotubes

  • Park, Su Hyun;Kim, Young Hwa;Lee, Hyun Jeong;Baek, Youl Chang;Kim, Min Seok;Jeong, Jin Young;Oh, Young Kyun;Park, Sung Kwon
    • Korean Journal of Agricultural Science
    • /
    • v.44 no.4
    • /
    • pp.586-594
    • /
    • 2017
  • Mitochondrial activity affects skeletal muscle energy metabolism and phenotype. To address whether mitochondrial activity can modulate muscle phenotype in vitro, protein expression of myosin heavy chain (MyHC) in C2C12 muscle cell lines was investigated after treated with antimycin A, an inhibitor of oxidative phosphorylation in mitochondria. Fully differentiated C2C12 myotubes were administrated with different concentration of antimycin A including 0, 100, 200, 500, 700, and 1000 ng/mL. After 72 h treatment, myosin heavy chain isoform expression and related enzyme activity (lactate dehydrogenase; LDH and creatine kinase) were analyzed. Administration of antimycin A changed expression of MyHC in C2C12 myotubes showing a shift from slow to fast twitching muscle type. Protein expression of MyHC type 2b (fast twitching muscle type) was decreased (P < 0.05) by antimycin A treatment (500, 700, and 1000 ng/mL) when compared with control group. Administration of antimycin A (1000 ng/mL), however, decreased (P < 0.05) MyHC type I (slow twitching muscle type). Interestingly, LDH activity was increased (P < 0.05) by antimycin A treatment. Results from our current study proposed a possibility that skeletal muscle phenotype, including MyHC and LDH activity, can be shifted from slow to fast twitching type by inhibiting the mitochondrial activity in C2C12 myotubes.

Insulin-like Growth Factor-I Induces Plectin and MACF1 Expression in C2C12 Myotubes (C2C12 myotube에서 insulin-like growth factor-I이 plectin과 MACF1 발현에 미치는 영향)

  • Kim, Hye Jin;Hwang, Ji Sun;Kwak, Yi-Sub;Lee, Won Jun
    • Journal of Life Science
    • /
    • v.22 no.12
    • /
    • pp.1651-1657
    • /
    • 2012
  • Plectin and microtubule actin cross-linking factor 1 (MACF1) are architectural proteins that contribute to the function of skeletal muscle as generators of mechanical force. However, the influence of insulin- like growth factor-I (IGF-I), a master regulator of skeletal muscle cells, on plectin and MACF1 in skeletal muscle cells has not been demonstrated. The effect of IGF-I on plectin and MACF1 gene expression was investigated by treating differentiated C2C12 murine skeletal muscle cells with 20 ng/ml of IGF-I at different time points. The IGF-I treatment increased plectin protein expression in a dose-dependent manner. The mRNA level of plectin was measured by real-time quantitative PCR to determine if plectin induction was regulated pretranslationally. IGF-I treatment resulted in a very rapid induction of plectin mRNA transcript in C2C12 myotubes. Plectin mRNA increased by 140 and 180% after 24 and 48 hours of IGF-I treatment, respectively, and returned to the control level after 72 hours of IGF-I treatment. MACF1 mRNA increased 86 and 90% after 24 and 48 hours of IGF-I treat-ment, respectively, and returned to the control level after 72 hours of IGF-I treatment. These results suggested that the plectin gene is regulated pretranslationally by IGF-I in skeletal muscle cells. In conclusion, IGF-I induces a rapid transcriptional modification of the plectin and MACF1 genes in C2C12 skeletal muscle cells and has modulating effects on a cytolinker protein as well as on contractile proteins.

A Molecular Study of Sopungsungi-won(Shufengshunqiyuan) about Regulation of PPARs in Mouse NMu2Li Liver Cells and C2C12 Skeletal Muscle Myogenic Progenital Cells (소풍순기원(疏風順氣元)이 mouse의 NMu2Li 간세포와 C2C12 골격근세포에서 PPARs 조절의 분자기전에 미치는 영향)

  • Oh, Young-Jin;Shin, Soon-Shik;Yoon, Mi-Chung;Kim, Bo-Kyung
    • Journal of Oriental Neuropsychiatry
    • /
    • v.20 no.1
    • /
    • pp.147-164
    • /
    • 2009
  • Objectives : We investigated the effects of Sopungsungi-won(Shu!engshunqiyuan) (SSEx1, SSEx2) to treat the metabolic syndrome by the molecular mechanism of regulation of PPAR and modulation of mitochondrial MCAD, VLCAD mRNA expression. Methods : Mouse NMu2Li liver cells and C2C12 skeletal muscle myogenic progenital cells were transiently transfected with expression plasmids for PPAR(PPAR${\alpha}$, PPAR${\delta}$), a luciferase reporter gene construct containing 3 copies of the PPRE from the rat acyl-CoA oxidase gene and ${\beta}$-galactosidase gene. Cells were treated with several concentrated kinds of SSEx1, SSEx2 at the initial time of culture and analyzed PPAR${\alpha}$, PPAR${\delta}$ reporter gene activity using spectrophotometer (405 nm). Total RNA was extracted from SSEx1, SSEx2 and measured mRNA levels of mitochondrial MCAD, VLCAD. Representative RT-PCR bands are shown. Results : 1. SSEx1 increased the expression of PPAR${\alpha}$ reporter gene activities at 0.1 ${\mu}$g/ml (p${\mu}$g/ml (p<0.05), SSEx2 at 0.1 ${\mu}$g/ml (p${\mu}$g/ml (p<0.05) significantly in NMu2Li liver cell lines. 2. SSEx1 increased the expression of PPAR${\alpha}$ reporter gene activities at 1 ${\mu}$g/ml (p${\mu}$g/ml (p${\alpha}$ reporter gene activities in C2C12 skeletal muscle cells. 4. SSEx1 increased the modulation of mitochondrial MCAD mRNA expression (p<0.05) significantly in NMu2Li liver cell lines. 5. SSEx1, SSEx2 both increased the modulation of mitochondrial MCAD mRNA expression (p<0.05) significantly in C2C12 skeletal muscle cells. Conclusions : These results show the SSEx1, SSEx2 can be used as therapeutic agent for metabolic syndrome and it's molecular mechanisms of PPAR more contribute to the activation of PPAR${\alpha}$ then PPAR${\delta}$ reporter gene activities and it's total RNA more contribute to the modulation of mitochondrial MCAD then VLCAD mRNA expression.

  • PDF

Effect of trichostatin A on NF-κB DNA binding activity and myogenesis in C2Cl2 skeletal muscle Precursor cell (C2C12 근육아세포에서 trichostatin A에 의한 NF-κB DNA 결합 활성과 근육발생에 미치는 영향)

  • 임운기;김경창;신혜자
    • Journal of Life Science
    • /
    • v.12 no.1
    • /
    • pp.55-60
    • /
    • 2002
  • The differentiation of skeletal muscle precursor cells in culture is marked by the transcriptional activation of muscle-specific genes and the morphological differentiation of myoblast into multinucleate myotube. In this study, we examined the effect of TSA (Trichostatin A) on WF-kB DNA binding activity and muscle cell fusion in the process of myogenesis. Under TSA treatment, C2C12 myoblast could not fuse to myotube and its NF-kB DNA binding activity was also blocked. To investigate whether these phenomenons were affected by TSA in direct or not, differentiation media (DM) used to differentiate cells without TSA was concentrated and added to C2C12 myoblast with TSA simultaneously. C2C12 myoblast was fused to myotube and NF-kB DNA binding activity was recovered. These results suggest that TSA affects on the differentiation of myoblast, perhaps through several factors, by inhibiting myoblst fusion and blocking NF-kB DNA binding activity.

Effect of Low-Energy Laser Irradiation on the Proliferation and Gene Expression of Myoblast Cells (저출력 레이져 자극이 근육세포의 증식 및 유전자 발현에 미치는 효과)

  • Kwag, J.H.;Jeon, O.H.;Kang, D.Y.;Ryu, H.H.;Kim, K.H.;Jung, B.J.;Kim, C.H.
    • Journal of Biomedical Engineering Research
    • /
    • v.31 no.1
    • /
    • pp.81-86
    • /
    • 2010
  • Laser irradiation is known to affect various tissues such as skin, bone, nerve, and skeletal muscle. Laser irradiation promotes ATP synthesis, facilitates wound healing, and stimulates cell proliferation and angiogenesis. In skeletal muscle, laser irradiation is related to the proliferation of skeletal muscle satellite cells. Normal skeletal muscle contains remodeling capacity from myogenic cells that are derived from mononuclear satellite cells. Their processes are activated by the expression of genes related with myogenesis such as muscle-specific transcription factors (MyoD and Myf5) and VEGF (vascular endothelial growth factor). In this study, we hypothesized that laser irradiation would enhance and regulate muscle cell proliferation and regeneration through modulation of the gene expressions related with the differentiation of skeletal muscle satellite cells. $C_2C_{12}$ myoblastic cells were exposed to continuous/non-continuous laser irradiation (660nm/808nm) for 10 minutes daily for either 1 day or 5 days. After laser irradiation, cell proliferation and gene expression (MyoD, Myf5, VEGF) were quantified. Continuous 660nm laser irradiation significantly increased cell proliferation and gene expression compared to control, continuous 808nm laser irradiation, and non-continuous 660nm laser irradiation groups. These results indicate that continuous 660nm laser irradiation can be applied to the treatment and regeneration of skeletal muscle tissue.

Ginsenoside Rg1 augments oxidative metabolism and anabolic response of skeletal muscle in mice

  • Jeong, Hyeon-Ju;So, Hyun-Kyung;Jo, Ayoung;Kim, Hye-Been;Lee, Sang-Jin;Bae, Gyu-Un;Kang, Jong-Sun
    • Journal of Ginseng Research
    • /
    • v.43 no.3
    • /
    • pp.475-481
    • /
    • 2019
  • Background: The ginsenoside Rg1 has been shown to exert various pharmacological activities with health benefits. Previously, we have reported that Rg1 promoted myogenic differentiation and myotube growth in C2C12 myoblasts. In this study, the in vivo effect of Rg1 on fiber-type composition and oxidative metabolism in skeletal muscle was examined. Methods: To examine the effect of Rg1 on skeletal muscle, 3-month-old mice were treated with Rg1 for 5 weeks. To assess muscle strength, grip strength tests were performed, and the lower hind limb muscles were harvested, followed by various detailed analysis, such as histological staining, immunoblotting, immunostaining, and real-time quantitative reverse transcription polymerase chain reaction. In addition, to verify the in vivo data, primary myoblasts isolated from mice were treated with Rg1, and the Rg1 effect on myotube growth was examined by immunoblotting and immunostaining analysis. Results: Rg1 treatment increased the expression of myosin heavy chain isoforms characteristic for both oxidative and glycolytic muscle fibers; increased myofiber sizes were accompanied by enhanced muscle strength. Rg1 treatment also enhanced oxidative muscle metabolism with elevated oxidative phosphorylation proteins. Furthermore, Rg1-treated muscles exhibited increased levels of anabolic S6 kinase signaling. Conclusion: Rg1 improves muscle functionality via enhancing muscle gene expression and oxidative muscle metabolism in mice.

Effects of polysaccharide (polycan) derived from black yeast in dexamethasone-induced muscle atrophy cell model (Dexamethasone으로 유도한 근위축 세포모델에서 흑효모 배양물 유래 polycan의 근위축 개선에 대한 효과)

  • Hwang, Su-Jin;Lim, Jong-Min;Ku, Bon-Hwa;Cheon, Da-Mi;Jung, Yu Jin;Kim, Young-Suk;Oh, Tae Woo
    • Herbal Formula Science
    • /
    • v.29 no.1
    • /
    • pp.45-55
    • /
    • 2021
  • Objectives : This study was conducted to evaluate the anti-atrophic effect of polycan in dexamethasone-induced skeletal muscle atrophy in vitro model. Methods : C2C12 myoblast were differentiated into myotube by 2% horese serum medium for 6 days, and then treated polycan extract at different concentrations for 24h. The effect of dexamethasone on the induction of muscle atrophy and expression of atrophy-related genes in differentiated C2C12 myotubes using a GSH, ROS, real-time PCR, western blots analysis. Results : The results showed that Treatment with polycan (100 and 200 ㎍/㎖) noncytotoxic levels on both myoblast and myotube. Polycan decreased the ROS level overproduced with dexamethasone and improved the depletion of GSH level. Dexamethasone showed a decrease in myotube diameter, which was associated with up-regulation muscle-specific ubiquitin ligases markers, such as atrogin-1, FoxO3, myostatin and muscle RING finger-1 (MuRF1), and down-regulation of myogenin, MEF2, Myogenic regulatory factor 5, 6 and MyoD. The results showed that polycan treatment significantly dose-dependently inhibited it. Furthermore, decreased expressions of PI3K/Akt signal pathway by dexamethasone were reversed by treatment with polycan. Conclusions : Thus, polycan suppresses dexamethasone induced muscle atrophy in C2C12 myotube in vitro model through activation of PI3K/Akt pathway and protective effect of improve skeletal muscle function.