Low-intensity pulsed ultrasound suppresses proliferation and promotes apoptosis via p38 MAPK signaling in rat visceral preadipocytes

Tianhua Xu, Jia Gu, Chenghai Li, Xiasheng Guo, Juan Tu, Dong Zhang, Wei Sun, Xiangqing Kong, Tianhua Xu, Jia Gu, Chenghai Li, Xiasheng Guo, Juan Tu, Dong Zhang, Wei Sun, Xiangqing Kong

Abstract

Low-intensity pulsed ultrasound (LIPUS) has been used widely in clinical therapy for bone fracture and soft tissue injury. However, whether LIPUS regulates primary preadipocyte function and adipogenesis remains unknown. In this study, we investigated the potential role of LIPUS in regulating visceral preadipocyte function. Resuspended rat visceral preadipocytes were treated with LIPUS (0.5 MHz, 109.44 mW/cm2) for 1 min and then cultured for an additional 48 hours. Cell proliferation was examined using the CCK-8 assay, and the early apoptosis rate was determined by flow cytometry. In addition, we evaluated the related signaling pathway via examination of proliferating cell nuclear antigen (PCNA), peroxisome proliferator-activated receptor gamma (PPARγ), Bcl2, Bax, cleaved caspase 3 (C-C3), and mitogen-activated protein kinase (MAPK) member protein levels using western blot or quantitative real-time PCR (qRT-PCR). LIPUS inhibited preadipocyte proliferation and induced cell apoptosis. The protein expression of proliferation markers decreased, while expression of the apoptosis-related modulators increased following LIPUS treatment. LIPUS treatment decreased extracellular signal-regulated kinase (ERK) phosphorylation and increased p38 MAPK phosphorylation. Inhibition of p38 MAPK rescued the LIPUS-induced proliferation inhibition and apoptosis induction. Thus, treatment of rat visceral preadipocytes with 0.5 MHz LIPUS suppresses proliferation and promotes apoptosis via activation of p38 MAPK signaling.

Keywords: LIPUS; apoptosis; p38 MAPK; proliferation; rat visceral preadipocytes.

Conflict of interest statement

None.

Figures

Figure 1
Figure 1
LIPUS inhibits preadipocyte proliferation and increases apoptosis. Primary cultured preadipocytes were treated with different doses of ultrasound (500, 800, 1100, 1400, 1700, and 2000 cycles) and different duration (0.5, 1, and 1.5 min). (A) Preadipocyte proliferation was evaluated with the CCK-8 assay. Ultrasound doses reaching 1700 cycles (average sound intensity 109.44 mW/cm2) led to a remarkable reduction of preadipocyte proliferation. (B) Effects of LIPUS on preadipocytes proliferation were evaluated at different points in time and the 1-min duration was determined to be the optimal duration. (C, D) The early apoptosis rate was analyzed by flow cytometry using Annexin-V-FITC staining. LIPUS treatment increased the number of early apoptotic preadipocytes compared to control treatment. All values are expressed as the mean ± SEM of three independent trials. For (A and B), data were analyzed with one-way ANOVA analysis, for (C), data were analyzed with independent t test. *P<0.05, **P<0.01, ***P<0.001.
Figure 2
Figure 2
LIPUS affects expression of proteins associated with proliferation and apoptosis in preadipocytes. The expression levels of key proliferation- and apoptosis-related molecules (PCNA, PPARγ, Bcl2, Bax, and C-C3) were examined by western blotting in primary preadipocytes with or without LIPUS (1700 cycles, 1 min) treatment. A, B. The protein levels of PCNA protein decreased and the levels of PPARγ increased in LIPUS treated cells compared to untreated cells. C, D. LIPUS treatment downregulated the protein expression of Bcl2 and the Bcl2/Bax ratio, and upregulated the protein expression of C-C3. All values are expressed as the mean ± SEM of three independent trials. Data were analyzed with independent t test. *P<0.05, **P<0.01.
Figure 3
Figure 3
LIPUS does not promote transcription of PPAR family members during the early stage. mRNA levels of PPAR and C/EBP family members were assessed by qRT-PCR in LIPUS treated and untreated cells. β-tubulin was utilized as the internal standard for normalization. A. LIPUS did not affect the mRNA levels of PPARα, PPARγ, and PPARδ of rat visceral preadipocytes. B. LIPUS treatment downregulated the C/EBPδ mRNA level but did not affect the C/EBPα and C/EBPβ mRNA levels of rat visceral preadipocytes. All values are expressed as the mean ± SEM of three independent trials. Data were analyzed with independent t test. **P<0.01.
Figure 4
Figure 4
LIPUS induces phosphorylation of p38-MAPK. The expression and modification of MAPK members were assessed by western blotting in primary preadipocytes with or without LIPUS treatment. A. LIPUS treatment significantly promoted the phosphorylation level of p38 and inhibited the phosphorylation of ERK, and no change of the expression level of p-JNK were observed. B. Quantification of p-38, p-p38, ERK, p-ERK, JNK, and p-JNK protein levels were. Data were analyzed with independent t test. All values are expressed as the mean ± SEM of three independent trials. *P<0.05.
Figure 5
Figure 5
Inhibition of p38 phosphorylation rescues the effects of LIPUS. In order to explore whether the biological effects of LIPUS on preadipocytes are associated with p-p38, cell suspension exposed to LIPUS was treated with or without p38 MAPK inhibitor SB203580. A, B. p38 inhibition rescued the LIPUS-induced apoptosis as shown by flow cytometric analysis. The percentage of early apoptotic preadipocytes (%) was analyzed with one-way ANOVA analysis. C. CCK-8 assay showed that inhibition of p38 activation reversed the LIPUS-induced proliferation inhibition in primary preadipocytes. D. p-p38, p-ERK, C-C3, PCNA and Bcl2 protein levels were assessed by western blotting with or without p38 inhibitor treatment. Relative protein levels were analyzed with one-way ANOVA analysis. All values are expressed as the mean ± SEM of three independent trials. *P<0.05.

References

    1. Williams EP, Mesidor M, Winters K, Dubbert PM, Wyatt SB. Overweight and obesity: prevalence, consequences, and causes of a growing public health problem. Curr Obes Rep. 2015;4:363–370.
    1. Khandekar MJ, Cohen P, Spiegelman BM. Molecular mechanisms of cancer development in obesity. Nat Rev Cancer. 2011;11:886–895.
    1. Abraham TM, Pedley A, Massaro JM, Hoffmann U, Fox CS. Association between visceral and subcutaneous adipose depots and incident cardiovascular disease risk factors. Circulation. 2015;132:1639–1647.
    1. Rosen ED, Spiegelman BM. Adipocytes as regulators of energy balance and glucose homeostasis. Nature. 2006;444:847–853.
    1. Spalding KL, Arner E, Westermark PO, Bernard S, Buchholz BA, Bergmann O, Blomqvist L, Hoffstedt J, Näslund E, Britton T, Concha H, Hassan M, Rydén M, Frisén J, Arner P. Dynamics of fat cell turnover in humans. Nature. 2008;453:783–787.
    1. Fried SK, Lee MJ, Karastergiou K. Shaping fat distribution: New insights into the molecular determinants of depot- and sex-dependent adipose biology. Obesity. 2015;23:1345–1352.
    1. Claes L, Willie B. The enhancement of bone regeneration by ultrasound. Prog Biophys Mol Biol. 2007;93:384–398.
    1. Kusuyama J, Bandow K, Shamoto M, Kakimoto K, Ohnishi T, Matsuguchi T. Low intensity pulsed ultrasound (LIPUS) influences the multilineage differentiation of mesenchymal stem and progenitor cell lines through ROCK-Cot/Tpl2-MEK-ERK signaling pathway. J Biol Chem. 2014;289:10330–10344.
    1. Li YN, Zhou Q, Yang B, Hu Z, Wang JH, Li QS, Cao WW. Mechanism of rat osteosarcoma cell apoptosis induced by a combination of low-intensity ultrasound and 5-aminolevulinic acid in vitro. Genet Mol Res. 2015;14:9604–9613.
    1. Xin Z, Lin G, Lei H, Lue TF, Guo Y. Clinical applications of low-intensity pulsed ultrasound and its potential role in urology. Transl Androl Urol. 2016;5:255–266.
    1. Lopez MJ, Spencer ND. In vitro adult rat adipose tissue-derived stromal cell isolation and differentiation. Methods Mol Biol. 2011;702:37–46.
    1. Wang H, Bei Y, Shen S, Huang P, Shi J, Zhang J, Sun Q, Chen Y, Yang Y, Xu T, Kong X, Xiao J. miR-21-3p controls sepsis-associated cardiac dysfunction via regulating SORBS2. J Mol Cell Cardiol. 2016;94:43–53.
    1. Banerjee S, Bhattacharjee P, Chakraborty J, Panda AK, Bandyopadhyay A, Banik SK, Sa G. WITHDRAWN: curcumin shifts RAS-induced pro-proliferative MEK/ERK-signaling toward pro-apoptotic p38MAPK/JNK1-signaling, triggering p53 activation and apoptosis. J Biol Chem. 2017 [Epub ahead of print]
    1. Lee SG, Lee YJ, Jang MH, Kwon TR, Nam JO. Panax ginseng leaf extracts exert anti-obesity effects in high-fat diet-induced obese rats. Nutrients. 2017;9
    1. Jiao Y, Zhang J, Lu L, Xu J, Qin L. The Fto gene regulates the proliferation and differentiation of pre-adipocytes in vitro. Nutrients. 2016;8:102.
    1. Secco B, Camire E, Briere MA, Caron A, Billong A, Gelinas Y, Lemay AM, Tharp KM, Lee PL, Gobeil S, Guimond JV, Patey N, Guertin DA, Stahl A, Haddad E, Marsolais D, Bosse Y, Birsoy K, Laplante M. Amplification of adipogenic commitment by VSTM2A. Cell Rep. 2017;18:93–106.
    1. Price NL, Holtrup B, Kwei SL, Wabitsch M, Rodeheffer M, Bianchini L, Suarez Y, Fernandez-Hernando C. SREBP-1c/MicroRNA 33b genomic loci control adipocyte differentiation. Mol Cell Biol. 2016;36:1180–1193.
    1. Guo G, Ma Y, Guo Y, Zhang C, Guo X, Tu J, Yu ACH, Wu J, Zhang D. Enhanced porosity and permeability of three-dimensional alginate scaffolds via acoustic microstreaming induced by low-intensity pulsed ultrasound. Ultrason Sonochem. 2017;37:279–285.
    1. Zhang B, Zhou HS, Cheng Q, Lei L, Hu B. Low-frequency ultrasound induces apoptosis of rat aortic smooth muscle cells (A7r5) via the intrinsic apoptotic pathway. Genet Mol Res. 2014;13:3143–3153.
    1. Feng Y, Tian ZM, Wan MX, Zheng ZB. Low intensity ultrasound-induced apoptosis in human gastric carcinoma cells. World J Gastroenterol. 2008;14:4873–9.
    1. Bohari SP, Grover LM, Hukins DW. Pulsed low-intensity ultrasound increases proliferation and extracelluar matrix production by human dermal fibroblasts in three-dimensional culture. J Tissue Eng. 2015;6:204173141561577.
    1. Gao Q, Walmsley AD, Cooper PR, Scheven BA. Ultrasound stimulation of different dental stem cell populations: role of mitogen-activated protein kinase signaling. J Endod. 2016;42:425–431.
    1. Jang KW, Ding L, Seol D, Lim TH, Buckwalter JA, Martin JA. Low-intensity pulsed ultrasound promotes chondrogenic progenitor cell migration via focal adhesion kinase pathway. Ultrasound Med Biol. 2014;40:1177–1186.
    1. Huang JJ, Shi YQ, Li RL, Hu A, Lu ZY, Weng L, Wang SQ, Han YP, Zhang L, Li B, Hao CN, Duan JL. Angiogenesis effect of therapeutic ultrasound on HUVECs through activation of the PI3K-Akt-eNOS signal pathway. Am J Transl Res. 2015;7:1106–1115.
    1. Hardwick JM, Youle RJ. SnapShot: BCL-2 proteins. Cell. 2009;138:404, 404.e1.
    1. Yee C, Yang W, Hekimi S. The intrinsic apoptosis pathway mediates the pro-longevity response to mitochondrial ROS in C. elegans. Cell. 2014;157:897–909.
    1. Chandra D, Maes ME, Schlamp CL, Nickells RW. Live-cell imaging to measure BAX recruitment kinetics to mitochondria during apoptosis. PLoS One. 2017;12:e0184434.
    1. Peng Q, Deng Z, Pan H, Gu L, Liu O, Tang Z. Mitogen-activated protein kinase signaling pathway in oral cancer. Oncol Lett. 2018;15:1379–1388.

Source: PubMed

3
購読する