Electroacupuncture promotes chondrocyte proliferation via accelerated G1/S transition in the cell cycle

  • Authors:
    • Yali Huang
    • Guangwen Wu
    • Huailing Fan
    • Jinxia Ye
    • Xianxiang Liu
  • View Affiliations

  • Published online on: April 5, 2013     https://doi.org/10.3892/ijmm.2013.1336
  • Pages: 1443-1448
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of the present study was to investigate the effects of electroacupuncture (EA) on the proliferation of chondrocytes and the molecular mechanism(s) involved. Passage 2 chondrocytes were randomly divided into four groups and treated with EA or nocodazole. After treatment, cell proliferation was determined using an MTT assay and DNA staining followed by FACS. The mRNA expression levels of cyclin D1, cyclin-dependent kinase (CDK)4, CDK6, phosphorylated retinoblastoma (pRb) and P16 were detected by RT-PCR, and the protein levels of cyclin D1, CDK4, CDK6, pRb and P16 were detected by western blotting. EA treatment significantly increased cell viability in a time-dependent manner and decreased the number of G0/G1 and G2/M phase chondrocytes and increased the number of S phase cells. The mRNA and protein levels of cyclin D1, CDK4, CDK6, (p)Rb and P16 consistently demonstrated a reverse trend with the levels in the chondrocytes treated with nocodazole. The expression levels of cyclin D1, CDK4, CDK6 and Rb were higher in chondrocytes receiving EA treatment when compared to levels in the untreated cells while expression of P16 was lower. In conclusion, EA treatment promotes chondrocyte proliferation via promotion of G1/S checkpoint transition in the cell cycle dependent on the activity of the P16-cyclin D1-CDK4/6-pRb pathway and this may, in part, explain its clinical effect in the treatment of osteoarthritis.

Introduction

Osteoarthritis (OA) is characterized by a basic pathology of cartilage degeneration caused by the mutual influence of mechanical and biological factors. The balance of catabolism and anabolism within chondrocytes helps to maintain the structural and functional integrity of the extracellular cartilage matrix (ECM). In the early stages of OA development, cartilage tissues show self-repair activity, the volume of chondrocytes increases and the synthesis of proteoglycan accelerates. In the late stage of OA, the balance is broken, and the damaging effect of inflammation becomes more dramatic. Chondrocytes can rapidly respond to changes in the microenvironment of the joint and regulate the dynamic equilibrium between the degradation and synthesis of the ECM (14). Therefore, the functional changes in chondrocytes play important roles in the degeneration of joint cartilage, and chondrocyte proliferation is one of the important factors contributing to the maintenance of cellular function.

The cell cycle is composed of four different phases, G1, S, G2 and M, which regulate cell proliferation, differentiation and apoptosis, essential to cell life. The G1/S checkpoint that exists at the end of G1 stage is the key point of intracellular and extracellular signaling which integrates in the nucleus, then stimulates S phase cells to begin a new round of proliferation, differentiation, death or exit the cell cycle into the G0 phase (5,6). Cell cycle progression involves the activity of cyclin-dependent kinases (CDKs), which are complexes of cyclin and CDK proteins. The G1/S transition of the cell cycle is dependent on the activity of cyclin D1-CDK4/6. Once activated, these CDK complexes phosphorylate retinoblastoma (Rb) and related family members and release sequestered E2F transcription factors, thereby promoting the transcription of genes required for progression through the cell cycle. Such progression is under the control of CDK inhibitors (CDKIs), such as P16, which cause transient or permanent cell cycle arrest in cells carrying DNA damage. Cell cycle transitions are tightly regulated, and changes in the expression of CDKs or CDKIs may lead to exacerbated cell proliferation (7,8).

Acupuncture, which has been used for the treatment of various types of diseases in Eastern countries for thousands of years, is currently gaining acceptance as an alternative medicine in Western countries (9,10). Electroacupuncture (EA) is a modified acupuncture technique that utilizes electrical stimulation. Previous studies have demonstrated that EA has therapeutic effects on chondral defects including knee osteoarthritis (1113) and produces cytokines with multiple biological activities in various types of diseases (14,15). Our previous study showed that EA can be employed as a novel non-drug-inducing method for the differentiation of BMSCs into chondrocytes (16). However, the effects of EA on the proliferation of chondrocytes, have not yet been reported. To further elucidate the precise mechanism of the potential treatment of OA, in the present study we investigated the effect of EA on the proliferation of chondrocytes and investigated the underlying molecular mechanism.

Materials and methods

Materials and reagents

Fetal bovine serum (FBS), Dulbecco’s modified Eagle’s medium (DMEM), trypsin and toluidine blue stain were purchased from Hyclone Inc. (Carlsbad, CA, USA); MTT, type-II collagenase, and nocodazole were obtained from Sigma. Cell cycle test kit was obtained from Becton-Dickinson (San Jose, CA, USA). A total protein extraction kit and an ECL kit were purchased from Beyotime Biotech (Nanjing, Jiangsu, China). Cyclin D1, CDK4, CDK6, phospho-Rb, p16INK4a and β-actin antibodies and horseradish peroxidase (HRP)-conjugated secondary antibodies were obtained from Cell Signaling Technology, Inc. (Danvers, MA, USA). DNA primers were synthesized by Sangon Biotech (Shanghai, China).

Animals

Healthy and clean, 4-week-old Sprague Dawley rats (90–110 g weight) of either gender (n=42) were purchased from SLAC Laboratory Animal Inc. (Shanghai, China) [Laboratory Animal Use Certificate no. SCXK(SH)2007-0005] and raised in a sterile environment. All experiments involving the animals complied with Guidance Suggestions for the Care and Use of Laboratory Animals 2006 administered by the Ministry of Science and Technology, China.

Isolation and culture of rat chondrocytes

Rat chondrocytes were isolated and cultured as previously described (17). The cells used in these experiments were counted by a hemocytometer and adjusted to 104–106 cells/ml.

EA stimulation

The EA stimulation method was in accordance with a previously described method (16). The acupuncture stimulation was applied daily for 0, 15, 30, 60 or 120 min.

Cell treatment and grouping

The passage 2 chondrocytes were seeded at 1×105/ml in a T-25 flask, cultured for 48 h and starved in DMEM medium without FBS for 24 h. The cells were then randomly divided into four groups: control group (normal culture without treatment); experimental group 1 (treated with 50 nM nocodazole for 24 h and receiving no EA treatment); experimental group 2 (receiving no nocodazole treatment and treated with EA stimulator for 60 min) and experimental group 3 (treated with 50 nM nocodazole for 24 h and receiving EA stimulator for 60 min). After treatment, cell proliferation was detected using an MTT assay and DNA staining followed by FACS analysis. Cells were also processed to measure the mRNA levels of cyclin D1, CDK4, CDK6, Rb and P16 by RT-PCR and the protein levels of cyclin D1, CDK4, CDK6, pRb and P16 by western blotting.

MTT assay

The passage 2 chondrocytes were seeded in a 6-well plate at 1×105/ml (2 ml/well) and treated with nocodazole and EA stimulator. The chondrocytes were then washed with PBS once, and 1 ml of a 0.5% MTT solution was added to each well. After incubation at 37°C for 4 h, wells were emptied, supplied with 1 ml of DMSO and shaken for 10 min. The absorbance was measured at 490 nm using an EL×808 absorbance microplate reader (BioTek Instruments, Inc., Winooski, VT, USA).

Detection of cell cycle distribution by flow cytometric analysis with propidium iodide (PI) staining

Chondrocytes were digested with 0.25% trypsin and incubated in 25-ml culture flasks at a density of 1×105 cells/ml in 4 ml of medium for 24 h and starved for 24 h in serum-free DMEM medium and were then treated with or without nocodazole and EA stimulator. After treatment, the cell cycle distribution of the chondrocytes was determined by flow cytometric analysis using a fluorescence-activated cell sorting FACSCalibur cytometer and a cell cycle assay kit. PI staining was performed according to the manufacturer’s instructions. The percentage of cells in the different phases was calculated by ModFit LT version 3.0 software, and the numbers of cell in the G0/G1, S and G2/M phases were determined.

RNA extraction and RT-PCR analysis

Total RNA from the treated cells was isolated with TRIzol reagent (Invitrogen). Oligo(dT)-primed RNA (2 μg) was reverse-transcribed with SuperScript II reverse transcriptase (Promega) according to the manufacturer’s instructions. The obtained cDNA was used to determine the mRNA levels of cyclin D1, CDK4, CDK6, Rb and P16 by PCR with TaqDNA polymerase (Fermentas). β-actin was used as an internal control. The primers and the annealing temperature (°C) used for amplification of cyclin D1, CDK4, CDK6, Rb, P16 and β-actin transcripts were as follows: cyclin D1, sense, 5′-GAC ACC AAT CTC CTC AAC GAC-3′ and antisense, 5′-AGA CAA GAA ACG GTC CAG GTA G-3′ (216 bp, 55°C); CDK4, sense, 5′-CCT ACG GAC ATA CCT GGA CAA-3′ and antisense, 5′-GAG GCA ATC CAA TGA GAT CAA-3′ (404 bp, 55°C); CDK6, sense, 5′-GTT TCA GCT TCT CCG AGG TCT-3′ and anti-sense, 5′-CGT CAA GCA TTT CAG AAG GAG-3′ (469 bp, 55°C); Rb, sense, 5′-CTT TAT TGG CCT GTG CTC TTG-3′ and antisense, 5′-ATT CCA TGA TTC GAT GCT CAC-3′ (225 bp, 53°C); P16, sense, 5′-GCT CTC CTG CTC TCC TAT GGT-3′, and antisense, 5′-AGA AGT TAT GCC TGT CGG TGA-3′ (268 bp, 54°C); β-actin, sense, 5′-GGG AAG TGC TGG ATA G-3′ and antisense, 5′-GTG ATG TTT CGG ATG G-3′ (385 bp, 55°C).

Western blot analysis

After treatment, cells were lysed, and protein concentrations were determined by BCA assay using bovine serum albumin as a standard. Samples were loaded with 20 μg of protein and separated by electrophoresis on 12% SDS polyacrylamide gels. After electrophoresis, proteins were transferred to PVDF membranes in 5% w/v nonfat dry milk using a semidry blotting system, and detected with antibodies against cyclin D1, CDK4, CDK6, pRb, P16 and β-actin and developed with ECL. The intensity of each band was quantified utilizing the Image Lab gel analyzing system and normalized to the band intensity of β-actin.

Statistical analysis

Statistical data are expressed as means ± SD. Statistical analysis of the data was performed with the Student’s t-test and one-way analysis of variance (ANOVA). Differences were considered statistically significant at P<0.05.

Results

Optimization of EA treatment time

Without treatment, the passage 2 chondrocytes proliferated normally, and there was no significant difference between the four groups. After treatment with EA, the optical densities (ODs) of cells receiving 60- and 120-min treatments were significantly higher than the ODs of cells receiving 0-min (P=0.000) and 15-min treatments (P=0.001). The OD of cells receiving a 60-min treatment was significantly higher than the OD of cells receiving a 30-min treatment (P= 0.000), but no significant difference was noted between the OD of cells receiving a 30-min treatment and the OD of cells receiving a 120-min treatment (P=0.242) (Table I).

Table I

Proliferation of chondrocytes as detected by MTT assay.

Table I

Proliferation of chondrocytes as detected by MTT assay.

GroupOD before treatmentOD after treatment
EA treatment for 0 min0.328±0.006 0.363±0.005d,e
EA treatment for 15 min0.327±0.009 0.369±0.003b,f
EA treatment for 30 min0.333±0.010 0.376±0.003a,d
EA treatment for 60 min0.332±0.004 0.393±0.006a,c,e
EA treatment for 120 min0.335±0.009 0.379±0.005a,c

a P<0.01,

b P<0.05 compared with 0-min EA treatment group;

c P<0.01,

d P<0.05 compared with 15-min EA treatment group;

e P<0.01,

f P<0.05 compared with 30-min EA treatment group. EA, electroacupuncture. OD, optical density. Values are expressed as means ± SD.

Effect of the EA treatment on nocodazole-induced proliferation

The effect of the EA treatment on the nocodazole-induced proliferation was assessed by MTT assay. Prior to treatment, there was no difference between the different groups. After treatment, the OD value of experimental group 1 was significantly lower than the OD values of the control group, experimental group 2 and experimental group 3 (P=0.000). Notably, there were significantly more cells in the experimental group 2 compared with the control group (P=0.001) (Table II).

Table II

Effect of nocodazole and EA on the proliferation of chondrocytes detected by MTT assay.

Table II

Effect of nocodazole and EA on the proliferation of chondrocytes detected by MTT assay.

GroupNocodazole (nM)EA treatment (min)OD before treatmentOD after treatment
Control000.335±0.0100.364±0.005b
Experimental 15000.337±0.0080.348±0.006a
Experimental 20600.329±0.005 0.391±0.007ac
Experimental 350600.336±0.0090.370±0.006b

a P<0.01 compared with control group;

b P<0.01 compared with experimental group 1;

c P<0.01 compared with experimental group 3. EA, electroacupuncture. OD, optical density. Values are expressed as means ± SD.

Cell cycle distribution of the treated chondrocytes as determined by FACS

Before treatment, all cells were starved for 24 h to synchronize the cell cycle stage (Fig. 1A–D). The FACS results showed that the cell cycle distribution of the cells in the different groups was similar. The G0/G1 ratio of cells in experimental group 1 was significantly higher than the ratio in the cells of experimental group 2 (P=0.000) following treatment, while the G0/G1 ratios of cells in experimental group 2 and 3 were significantly lower than that of the control group (P=0.000, P=0.003). The S ratio of cells in experimental group 1 was significantly lower than the ratios of cells of other groups (P=0.000), and the S ratio of cells in experimental group 2 was significantly higher than the ratios of the control group (P=0.000) and experimental group 3 (P=0.001). The G2/M ratio of cells in experimental group 1 was significantly higher than those of the control group (P=0.000) and experimental group 2 (P=0.002). The G2/M ratio of cells in the experimental group 2 was higher than that of the control group (P=0.002) (Fig. 1E–H and Table III).

Table III

Cell cycle distribution as detected by FACS (%).

Table III

Cell cycle distribution as detected by FACS (%).

Group G0/G1S G2/M
Before treatment
  Control98.273±0.1291.533±0.0730.193±0.119
  Experimental 198.390±0.3471.372±0.3780.238±0.552
  Experimental 298.332±0.0821.370±0.1120.298±0.097
  Experimental 398.362±0.3811.327±0.2790.313±0.198
After treatment
  Control 44.338±0.038b 31.627±0.075b 24.035±0.109b
  Experimental 1 41.627±0.054a 28.885±0.038a 29.488±0.065a
  Experimental 2 36.548±0.048a,b 36.673±0.056a,b 26.778±0.053a,b
  Experimental 3 41.902±0.072a 33.393±0.032a,b 24.705±0.091b

a P<0.01 compared with the control group after treatment;

b P<0.01 compared with experimental group 1 after treatment. Values are expressed as means ± SD.

The mRNA expression of cyclin D1, CDK4, CDK6, Rb and P16 in chondrocytes following EA treatment

To further explore the mechanism of the EA treatment, we analyzed the mRNA expression levels of cyclin D1, CDK4, CDK6, Rb and P16. The amplified products of cyclin D1, CDK4, CDK6, Rb and P16 were clearly visible on the agarose gel (Fig. 2). Quantification of the PCR products indicated that the levels of cyclin D1, CDK4, CDK6, Rb were significantly higher in control group than in experimental group 2 (P=0.002, P=0.001, P=0.001, P=0.001, respectively). However, the P16 mRNA level in experimental group 2 was significantly lower than that in control group (P=0.005) (Table IV).

Table IV

mRNA expression of cyclin D1, CDK4, CDK6, Rb and P16 in the chondrocytes.

Table IV

mRNA expression of cyclin D1, CDK4, CDK6, Rb and P16 in the chondrocytes.

Groupcyclin D1CDK4CDK6RbP16
Control0.750±0.1110.310±0.036d0.385±0.034d0.335±0.034c0.315±0.038
Experimental 10.643±0.0640.248±0.034a0.327±0.044b0.277±0.028a0.353±0.053
Experimental 2 0.940±0.116a,c,e 0.387±0.040a,c 0.473±0.039a,c 0.412±0.040a,c,e 0.238±0.029b,c
Experimental 30.792±0.076d 0.353±0.025b,c 0.437±0.031b,c 0.367±0.037b,c0.282±0.045c

a P<0.01,

b P<0.05 compared with the control group;

c P<0.01,

d P<0.05 compared with experimental group 1;

e P<0.05 compared with experimental group 3. Rb, phosphorylate retinoblastoma; CDK, cyclin-dependent kinase.

Protein expression of cyclin D1, CDK4, CDK6, pRb and P16 in chondrocytes following EA treatment

The protein expression of cyclin D1, CDK4, CDK6, pRb and P16 was also detected using western blotting. Quantification of the western blotting bands showed that levels of cyclin D1, CDK4, CDK6 and pRb proteins in experimental group 2 were significantly higher than levels in the control group (P=0.000, P=0.000, P=0.001, P=0.000, respectively). In contrast, the P16 protein level in experimental group 2 was significantly lower than that in the control group (P=0.000) (Fig. 3 and Table V).

Table V

Protein expression of cyclin D1, CDK4, CDK6, pRb and P16 in the chondrocytes.

Table V

Protein expression of cyclin D1, CDK4, CDK6, pRb and P16 in the chondrocytes.

GroupCyclin D1CDK4CDK6pRbP16
Control0.078±0.012d0.378±0.025d0.425±0.024c0.065±0.0050.167±0.008d
Experimental 10.067±0.008b0.305±0.021b0.317±0.029a0.053±0.0100.177±0.005b
Experimental 2 0.107±0.010a,c 0.647±0.054a,c,e 0.628±0.050a,c,e 0.095±0.015a,e 0.112±0.008a,c,e
Experimental 3 0.097±0.008a,c 0.540±0.072a,c 0.482±0.022a,c0.071±0.009d 0.135±0.010a,c

a P<0.01,

b P<0.05 compared with the control group;

c P<0.01,

d P<0.05 compared with experimental group 1;

e P<0.01 compared with experimental group 3. Rb, phosphorylate retinoblastoma; CDK, cyclin-dependent kinase.

Discussion

Osteoarthritis (OA), the most common age-related cartilage and joint disorder (18), is a slowly progressive degenerative disease characterized by degradation of the extracellular matrix and cell death resulting in a gradual loss of articular cartilage integrity (19,20). The only cell type present in mature cartilage is the chondrocyte. This cell type is responsible for repairing damaged cartilage tissue. We previously demonstrated that chondrocytes can be obtained by mechanical and chemical isolation methods and that the purity of isolated chondrocytes is high (17,21). This provided us with the ability to investigate the effect of EA on chondrocyte proliferation.

The working mode of EA is a disant wave and dense wave alternating waveform. The frequency of the distant wave is one fifth of the frequency of the dense wave; the duration time of the distant wave is 5 sec and the duration time of the dense wave is 10 sec. The function uses low-frequency electric current stimulation to change the distribution of hydronium between intracellular and extracellular regions, to interfere with the signal transmission of the organism, thus playing an effective role in cell proliferation. Nocodazole is a drug, like colchicines, that specifically binds to microtubules and interferes with spindle formation, thereby blocking the cell cycle at the G2/M transition (2224). In our study, we showed that EA treatment efficiently promoted chondrocyte proliferation, and the degree of the proliferative effect was dependent on the time of EA treatment in a certain range. After nocodazole treatment, chondrocyte proliferation was obviously inhibited in a dose-dependent manner, but once the cells were treated with EA, the inhibitory effect of nocodazole was weakened, suggesting that EA treatment may function by promoting cartilage cells to pass through the G1/S transition of the cell cycle.

There are four stages in the cell cycle: G1, preparation for DNA synthesis; S, DNA synthesis; G2, preparation for mitosis; and M, mitosis (25). Repetition of this cell cycle carries out cell proliferation. The amount of DNA in a cell changes during the cell cycle, allowing the different stages of the cycle to be identified by analyzing DNA content. The DNA content is 2N in G0 and G1 cells. After DNA synthesis in S phase, the DNA content becomes 4N in the G2 and M phases (26,27). FACS analysis, which measures the DNA content of cells, is more sensitive to the changes during the cell cycle than the MTT method. In the present study, we found that after EA treatment, the G0/G1 and G2/M ratios dropped, and the S ratio increased, indicating that EA treatment promotes cell proliferation by accelerating the G1/S transition. This conclusion is also supported by our result that after nocodazole treatment, the S phase cell ratio dramatically decreased and the G2/M cell ratio increased.

Upstream signals are usually transduced by signaling pathways and ultimately function on genes encoding cell cycle regulating factors, leading to cell cycle changes by altering the expression of these factors. The G1/S checkpoint that exists at the end of G1 stage is the key point of intracellular and extracellular signaling which integrates in the nucleus, which stimulates cells to begin a new round of proliferation, differentiation, death or exit the cell cycle into the G0 phase. The G1/S transition is dependent on the activity of cyclin D1-CDK4/6. Once activated, these CDK complexes phosphorylate retinoblastoma (Rb) and related family members and release sequestered E2F transcription factors, thereby promoting the transcription of genes required for progression through the cell cycle. Such progression is under the control of P16, which causes transient or permanent cell cycle arrest in cells carrying DNA damage. Our results showed that EA treatment effectively enhanced the mRNA and protein levels of cyclin D1, CDK4, CDK6 and (p)Rb and inhibited P16.

In summary, EA treatment effectively promotes chondrocyte proliferation. The molecular mechanism of EA may act by inducing the expression of cyclin D1, CDK4, CDK6 and Rb and inhibiting P16, thereby accelerating G1/S transition and promoting chondrocyte cell cycle progression. This may, in part, explain its clinical effect in the treatment of osteoarthritis.

References

1 

Yang X, Chen L, Xu X, Li C, Huang C and Deng CX: TGF-β/Smad3 signals repress chondrocyte hypertrophic differentiation and are required for maintaining articular cartilage. J Cell Biol. 153:35–46. 2001.

2 

Iannone F, De Bari C, Scioscia C, Patella V and Lapadula G: Increased Bcl-2/p53 ratio in human osteoarthritic cartilage: a possible role in regulation of chondrocyte metabolism. Ann Rheum Dis. 64:217–221. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Zhou HW, Lou SQ and Zhang K: Recovery of function in osteoarthritic chondrocytes induced by p16INK4a-siRNA in vitro. Rheumatology. 43:555–568. 2004. View Article : Google Scholar : PubMed/NCBI

4 

Tetlow LC and Woolley DE: Histamine stimulates the proliferation of human articular chondrocytes in vitro and is expressed by chondrocytes in osteoarthritic cartilage. Ann Rheum Dis. 62:991–994. 2003. View Article : Google Scholar : PubMed/NCBI

5 

Hulleman E, Bijvelt JJ, Verkleij AJ, Verrips CT and Boonstra J: Nuclear translocation of mitogen-activated protein kinase p42MAPKduring the ongoing cell cycle. J Cell Physiol. 180:325–333. 1999. View Article : Google Scholar : PubMed/NCBI

6 

Ekholm SV, Zickert P, Reed SI and Zetterberg A: Accumulation of cyclin E is not a prerequisite for passage through the restriction point. Mol Cell Biol. 21:3256–3265. 2001. View Article : Google Scholar : PubMed/NCBI

7 

Clurman BE and Roberts JM: Cell cycle and cancer. J Natl Cancer Inst. 87:1499–1501. 1995. View Article : Google Scholar : PubMed/NCBI

8 

Hirama T and Koeffler HP: Role of the cyclin-dependent kinase inhibitors in the development of cancer. Blood. 86:841–854. 1995.PubMed/NCBI

9 

Kaptchuk TJ: Acupuncture: theory, efficacy, and practice. Ann Intern Med. 136:374–383. 2002. View Article : Google Scholar : PubMed/NCBI

10 

No authors listed:. NIH Concensus Conference. Acupuncture. JAMA. 280:1518–1524. 1998.PubMed/NCBI

11 

Jubb RW, Tukmachi ES, Jones PW, Dempsey E, Waterhouse L and Brailsford S: A blinded randomised trial of acupuncture (manual and electroacupuncture) compared with a non-penetrating sham for the symptoms of osteoarthritis of the knee. Acupunct Med. 26:69–78. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Tukmachi E, Jubb R, Dempsey E and Jones P: The effect of acupuncture on the symptoms of knee osteoarthritis - an open randomised controlled study. Acupunct Med. 22:14–22. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Vas J and White A: Evidence from RCTs on optimal acupuncture treatment for knee osteoarthritis - an exploratory review. Acupunct Med. 25:29–35. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Aoki E, Kasahara T, Hagiwara H, Sunaga M, Hisamitsu N and Hisamitsu T: Electroacupuncture and moxibustion influence the lipopolysaccharide-induced TNF-alpha production by macrophages. In Vivo. 19:495–500. 2005.PubMed/NCBI

15 

Takaoka Y, Ohta M, Ito A, et al: Electroacupuncture suppresses myostatin gene expression: cell proliferative reaction in mouse skeletal muscle. Physiol Genomics. 30:102–110. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Wu G, Peng J, Wu M, Li Y, Huang Y, Lin R, Cai Q and Liu X: Experimental study of low-frequency electroacupuncture-induced differentiation of bone marrow mesenchymal stem cells into chondrocytes. Int J Mol Med. 27:79–86. 2011.PubMed/NCBI

17 

Li X, Du M, Liu X, Wu M, Ye H, Lin J, Chen W and Wu G: Millimeter wave treatment inhibits NO-induced apoptosis of chondrocytes through the p38MAPK pathway. Int J Mol Med. 25:393–399. 2010.PubMed/NCBI

18 

Heinegard D, Bayliss M and Lorenzo P: Biochemistry and metabolism of normal and osteoarthritic cartilage. Osteoarthritis. Brandt KD, Doherty M and Lohmander LS: Oxford University Press; New York: pp. 74–84. 1998

19 

Pritzker K: Pathology of osteoarthritis. Osteoarthritis. Brandt KD, Doherty M and Lohmander LS: Oxford University Press; New York: pp. 50–61. 1998

20 

Kim HA and Blanco FJ: Cell death and apoptosis in osteoarthritic cartilage. Curr Drug Targets. 8:333–345. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Li X, Du M, Liu X, Chen W, Wu M, Lin J and Wu G: Millimeter wave treatment promotes chondrocyte proliferation by upregulating the expression of cyclin-dependent kinase 2 and cyclin A. Int J Mol Med. 26:77–84. 2010.PubMed/NCBI

22 

Hsu SL, Yu CT, Yin SC, Tang MJ, Tien AC, Wu YM and Huang CY: Caspase 3, periodically expressed and activated at G2/M transition, is required for nocodazole-induced mitotic checkpoint. Apoptosis. 11:765–771. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Song H, Kim SI, Ko MS, et al: Overexpression of DRG2 increases G2/M phase cells and decreases sensitivity to nocodazole-induced apoptosis. J Biochem. 135:331–335. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Ho YS, Duh JS, Jeng JH, et al: Griseofulvin potentiates anti-tumorigenesis effects of nocodazole through induction of apoptosis and G2/M cell cycle arrest in human colorectal cancer cells. Int J Cancer. 91:393–401. 2001.PubMed/NCBI

25 

Zhang M, Xie R, Hou W, et al: PTHrP prevents chondrocyte premature hypertrophy by inducing cyclin-D1-dependent Run×2 and Run×3 phosphorylation, ubiquitylation and proteasomal degradation. J Cell Sci. 122:1382–1389. 2009.PubMed/NCBI

26 

Hwang SG, Song SM, Kim JR, Park CS, Song WK and Chun JS: Regulation of type II collagen expression by cyclin-dependent kinase 6, cyclin D1, and p21 in articular chondrocytes. IUBMB Life. 59:90–98. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Li TF, Chen D, Wu Q, et al: Transforming growth factor-beta stimulates cyclin D1 expression through activation of beta-catenin signaling in chondrocytes. J Biol Chem. 281:21296–21304. 2006. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June 2013
Volume 31 Issue 6

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Huang Y, Wu G, Fan H, Ye J and Liu X: Electroacupuncture promotes chondrocyte proliferation via accelerated G1/S transition in the cell cycle. Int J Mol Med 31: 1443-1448, 2013
APA
Huang, Y., Wu, G., Fan, H., Ye, J., & Liu, X. (2013). Electroacupuncture promotes chondrocyte proliferation via accelerated G1/S transition in the cell cycle. International Journal of Molecular Medicine, 31, 1443-1448. https://doi.org/10.3892/ijmm.2013.1336
MLA
Huang, Y., Wu, G., Fan, H., Ye, J., Liu, X."Electroacupuncture promotes chondrocyte proliferation via accelerated G1/S transition in the cell cycle". International Journal of Molecular Medicine 31.6 (2013): 1443-1448.
Chicago
Huang, Y., Wu, G., Fan, H., Ye, J., Liu, X."Electroacupuncture promotes chondrocyte proliferation via accelerated G1/S transition in the cell cycle". International Journal of Molecular Medicine 31, no. 6 (2013): 1443-1448. https://doi.org/10.3892/ijmm.2013.1336