Low‑intensity pulsed ultrasound promotes apoptosis and inhibits angiogenesis via p38 signaling‑mediated endoplasmic reticulum stress in human endothelial cells

Zhongping Su, Tianhua Xu, Yaqing Wang, Xiasheng Guo, Juan Tu, Dong Zhang, Xiangqing Kong, Yanhui Sheng, Wei Sun, Zhongping Su, Tianhua Xu, Yaqing Wang, Xiasheng Guo, Juan Tu, Dong Zhang, Xiangqing Kong, Yanhui Sheng, Wei Sun

Abstract

Aberrant increase in angiogenesis contributes to the progression of malignant solid tumors. An alternative anti‑angiogenesis therapy is critical for cancer, since the current anti‑angiogenesis drugs lack specificity for tumor cells. In the present study, the effects and mechanisms of low‑intensity pulsed ultrasound (LIPUS) on human umbilical vein endothelial cells (HUVECs) and human microvascular endothelial cells (HMECs) were investigated, and the therapeutic potential of this technology was assessed. HUVECs and HMECs were treated with LIPUS (0.5 MHz; 210 mW/cm2) for 1 min and cultured for 24 h. Flow cytometry and Cell Counting Kit‑8 assays demonstrated that LIPUS treatment at a dose of 210 mW/cm2 promoted apoptosis and decreased the viability in HUVECs and HMECs. Real‑time cell analysis also revealed that LIPUS did not affect the proliferation or migration of HUVECs. An endothelial cell tube formation assay indicated that LIPUS treatment inhibited the angiogenic ability of HUVECs and HMECs. Furthermore, LIPUS increased the protein levels of the apoptosis‑associated cleaved Caspase‑3 and decreased the B‑cell lymphoma‑2 levels. LIPUS increased the phosphorylation of p38 mitogen‑activated protein kinase (MAPK), and the levels of endoplasmic reticulum (ER) stress‑associated markers, including activating transcription factor‑4 (ATF‑4) and phosphorylated eukaryotic initiation factor 2α (eIF2α). The p38 inhibitor SB203580 reversed the pro‑apoptotic and anti‑angiogenic effects of LIPUS in cells. Finally, inhibition of p38 decreased the LIPUS‑induced elevation of p‑eIF2α and ATF‑4 levels. Taken together, these results suggested that LIPUS promoted apoptosis and inhibited angiogenesis in human endothelial cells via the activation of p38 MAPK‑mediated ER stress signaling.

Figures

Figure 1.
Figure 1.
LIPUS promoted apoptosis and inhibited cell viability in HUVECs and HMECs. Flow cytometry analysis of apoptosis rate in (A) HUVECs and (B) HMECs treated with different dose of LIPUS (70–280 mW/cm2). Quantification of early apoptosis in (C) HUVECs and (D) HMECs. Viability of (E) HUVECs and (F) HMECs treated with different doses of LIPUS (70–280 mW/cm2) was assessed by CCK-8 assay. Real-time monitoring of the (G) proliferation and (H) migration of HUVECs by real-time cell analysis. All values are expressed as the mean ± standard error of three independent experiments. *P<0.05, **P<0.01 and ***P<0.001. LIPUS, low-intensity pulsed ultrasound; HUVECs, human umbilical vein endothelial cells; HMECs, human microvascular endothelial cells; CCK-8, Cell Counting Kit-8; Ctrl, control; NS, not significant.
Figure 2.
Figure 2.
Effects of LIPUS on HUVEC and HMEC tube formation. Following incubation for 8 h, images (magnification, 10×) of tube-like structures in the Ctrl and LIPUS-treated cells were obtained using a microscope camera, and quantification of branches was performed with ImageJ software. (A) Images of HUVEC tube formation and (B) quantified results. (C) Images of HMEC tube formation and (D) quantified results. All values are expressed as the mean ± standard error of three independent experiments (scale bars=100 µm). **P

Figure 3.

Low-intensity pulsed ultrasound regulated the…

Figure 3.

Low-intensity pulsed ultrasound regulated the expression of apoptosis-associated proteins. (A) Cleaved Caspase-3, Bcl-2,…

Figure 3.
Low-intensity pulsed ultrasound regulated the expression of apoptosis-associated proteins. (A) Cleaved Caspase-3, Bcl-2, Bax and LC3B levels, measured by western blotting. (B) Quantification of the relative levels of cleaved Caspase-3, Bax and LC3-II, which were respectively normalized to β-tubulin, Bcl-2 and LC3-I levels. (C) Phosphorylation and total protein levels of p38, ERK and JNK were assessed by western blotting. (D) Quantification of p-p38, p-ERK and p-JNK protein levels, normalized to their total protein levels. (E) Protein levels of ATF-4 and p-eIF2α were assessed by western blotting. (F) Quantification of ATF-4 and p-eIF2α protein levels, normalized to β-tubulin. All values are expressed as the mean ± standard error of three independent experiments. **P

Figure 4.

Inhibition of p38 phosphorylation rescued…

Figure 4.

Inhibition of p38 phosphorylation rescued the pro-apoptotic and anti-angiogenic effects of low-intensity pulsed…

Figure 4.
Inhibition of p38 phosphorylation rescued the pro-apoptotic and anti-angiogenic effects of low-intensity pulsed ultrasound. (A) Flow cytometry analysis of apoptosis rates in HUVECs treated with the p38 inhibitor SB203580. (B) Quantification of the percentage of early apoptotic HUVECs. (C) Western blots and (D) quantified protein levels of cleaved Caspase-3, Bcl-2 and Bax in HUVECs treated with S203580. (E) Images (magnification, 10×) of tube-like structures and (F) quantified results in HUVECs. (G) Images of tube-like structures and (H) quantified results in HMECs. Quantification of branches was conducted with ImageJ software. All values are expressed as the mean ± standard error of three independent experiments (scale bars=100 µm). **P##P<0.01 and ###P<0.001, vs. ultrasound group. HUVECs, human umbilical vein endothelial cells; HMECs, human microvascular endothelial cells; Bcl-2, B-cell lymphoma-2; Bax, Bcl-2-associated X protein; Ctrl, control.

Figure 5.

Low-intensity pulsed ultrasound-induced apoptosis was…

Figure 5.

Low-intensity pulsed ultrasound-induced apoptosis was dependent on p38 phosphorylation. (A) Western blots and…

Figure 5.
Low-intensity pulsed ultrasound-induced apoptosis was dependent on p38 phosphorylation. (A) Western blots and (B) quantified protein levels of p-p38 and p-ERK in HUVECs treated with S203580, normalized to the corresponding total protein levels. (C) Western blots and (D) quantified protein levels of p-eIF2α and ATF-4 in HUVECs treated with S203580, normalized to β-tubulin. All values are expressed as the mean ± standard error of three independent experiments. ***P###P<0.001 vs. ultrasound group. HUVECs, human umbilical vein endothelial cells; ERK, extracellular signal-regulated kinase; ATF-4, activating transcription factor-4; eIF2α, eukaryotic initiation factor 2α; p-, phosphorylated; Ctrl, control.
Similar articles
Cited by
References
    1. Stylianopoulos T. The solid mechanics of cancer and strategies for improved therapy. J Biomech Eng. 2017;139 doi: 10.1115/1.4034991. - DOI - PMC - PubMed
    1. Fukumura D, Jain RK. Tumor microenvironment abnormalities: Causes, consequences, and strategies to normalize. J Cell Biochem. 2007;101:937–949. doi: 10.1002/jcb.21187. - DOI - PubMed
    1. Goel S, Duda DG, Xu L, Munn LL, Boucher Y, Fukumura D, Jain RK. Normalization of the vasculature for treatment of cancer and other diseases. Physiol Rev. 2011;91:1071–1121. doi: 10.1152/physrev.00038.2010. - DOI - PMC - PubMed
    1. Martin JD, Fukumura D, Duda DG, Boucher Y, Jain RK. Reengineering the tumor microenvironment to alleviate hypoxia and overcome cancer heterogeneity. Cold Spring Harb Perspect Med. 2016;6 doi: 10.1101/cshperspect.a027094. - DOI - PMC - PubMed
    1. Jain RK. Normalizing tumor microenvironment to treat cancer: Bench to bedside to biomarkers. J Clin Oncol. 2013;31:2205–2218. doi: 10.1200/JCO.2012.46.3653. - DOI - PMC - PubMed
Show all 44 references
MeSH terms
Related information
[x]
Cite
Copy Download .nbib .nbib
Format: AMA APA MLA NLM
Figure 3.
Figure 3.
Low-intensity pulsed ultrasound regulated the expression of apoptosis-associated proteins. (A) Cleaved Caspase-3, Bcl-2, Bax and LC3B levels, measured by western blotting. (B) Quantification of the relative levels of cleaved Caspase-3, Bax and LC3-II, which were respectively normalized to β-tubulin, Bcl-2 and LC3-I levels. (C) Phosphorylation and total protein levels of p38, ERK and JNK were assessed by western blotting. (D) Quantification of p-p38, p-ERK and p-JNK protein levels, normalized to their total protein levels. (E) Protein levels of ATF-4 and p-eIF2α were assessed by western blotting. (F) Quantification of ATF-4 and p-eIF2α protein levels, normalized to β-tubulin. All values are expressed as the mean ± standard error of three independent experiments. **P

Figure 4.

Inhibition of p38 phosphorylation rescued…

Figure 4.

Inhibition of p38 phosphorylation rescued the pro-apoptotic and anti-angiogenic effects of low-intensity pulsed…

Figure 4.
Inhibition of p38 phosphorylation rescued the pro-apoptotic and anti-angiogenic effects of low-intensity pulsed ultrasound. (A) Flow cytometry analysis of apoptosis rates in HUVECs treated with the p38 inhibitor SB203580. (B) Quantification of the percentage of early apoptotic HUVECs. (C) Western blots and (D) quantified protein levels of cleaved Caspase-3, Bcl-2 and Bax in HUVECs treated with S203580. (E) Images (magnification, 10×) of tube-like structures and (F) quantified results in HUVECs. (G) Images of tube-like structures and (H) quantified results in HMECs. Quantification of branches was conducted with ImageJ software. All values are expressed as the mean ± standard error of three independent experiments (scale bars=100 µm). **P##P<0.01 and ###P<0.001, vs. ultrasound group. HUVECs, human umbilical vein endothelial cells; HMECs, human microvascular endothelial cells; Bcl-2, B-cell lymphoma-2; Bax, Bcl-2-associated X protein; Ctrl, control.

Figure 5.

Low-intensity pulsed ultrasound-induced apoptosis was…

Figure 5.

Low-intensity pulsed ultrasound-induced apoptosis was dependent on p38 phosphorylation. (A) Western blots and…

Figure 5.
Low-intensity pulsed ultrasound-induced apoptosis was dependent on p38 phosphorylation. (A) Western blots and (B) quantified protein levels of p-p38 and p-ERK in HUVECs treated with S203580, normalized to the corresponding total protein levels. (C) Western blots and (D) quantified protein levels of p-eIF2α and ATF-4 in HUVECs treated with S203580, normalized to β-tubulin. All values are expressed as the mean ± standard error of three independent experiments. ***P###P<0.001 vs. ultrasound group. HUVECs, human umbilical vein endothelial cells; ERK, extracellular signal-regulated kinase; ATF-4, activating transcription factor-4; eIF2α, eukaryotic initiation factor 2α; p-, phosphorylated; Ctrl, control.
Figure 4.
Figure 4.
Inhibition of p38 phosphorylation rescued the pro-apoptotic and anti-angiogenic effects of low-intensity pulsed ultrasound. (A) Flow cytometry analysis of apoptosis rates in HUVECs treated with the p38 inhibitor SB203580. (B) Quantification of the percentage of early apoptotic HUVECs. (C) Western blots and (D) quantified protein levels of cleaved Caspase-3, Bcl-2 and Bax in HUVECs treated with S203580. (E) Images (magnification, 10×) of tube-like structures and (F) quantified results in HUVECs. (G) Images of tube-like structures and (H) quantified results in HMECs. Quantification of branches was conducted with ImageJ software. All values are expressed as the mean ± standard error of three independent experiments (scale bars=100 µm). **P##P<0.01 and ###P<0.001, vs. ultrasound group. HUVECs, human umbilical vein endothelial cells; HMECs, human microvascular endothelial cells; Bcl-2, B-cell lymphoma-2; Bax, Bcl-2-associated X protein; Ctrl, control.
Figure 5.
Figure 5.
Low-intensity pulsed ultrasound-induced apoptosis was dependent on p38 phosphorylation. (A) Western blots and (B) quantified protein levels of p-p38 and p-ERK in HUVECs treated with S203580, normalized to the corresponding total protein levels. (C) Western blots and (D) quantified protein levels of p-eIF2α and ATF-4 in HUVECs treated with S203580, normalized to β-tubulin. All values are expressed as the mean ± standard error of three independent experiments. ***P###P<0.001 vs. ultrasound group. HUVECs, human umbilical vein endothelial cells; ERK, extracellular signal-regulated kinase; ATF-4, activating transcription factor-4; eIF2α, eukaryotic initiation factor 2α; p-, phosphorylated; Ctrl, control.

References

    1. Stylianopoulos T. The solid mechanics of cancer and strategies for improved therapy. J Biomech Eng. 2017;139 doi: 10.1115/1.4034991.
    1. Fukumura D, Jain RK. Tumor microenvironment abnormalities: Causes, consequences, and strategies to normalize. J Cell Biochem. 2007;101:937–949. doi: 10.1002/jcb.21187.
    1. Goel S, Duda DG, Xu L, Munn LL, Boucher Y, Fukumura D, Jain RK. Normalization of the vasculature for treatment of cancer and other diseases. Physiol Rev. 2011;91:1071–1121. doi: 10.1152/physrev.00038.2010.
    1. Martin JD, Fukumura D, Duda DG, Boucher Y, Jain RK. Reengineering the tumor microenvironment to alleviate hypoxia and overcome cancer heterogeneity. Cold Spring Harb Perspect Med. 2016;6 doi: 10.1101/cshperspect.a027094.
    1. Jain RK. Normalizing tumor microenvironment to treat cancer: Bench to bedside to biomarkers. J Clin Oncol. 2013;31:2205–2218. doi: 10.1200/JCO.2012.46.3653.
    1. Vassilakopoulou M, Psyrri A, Argiris A. Targeting angiogenesis in head and neck cancer. Oral Oncol. 2015;51:409–415. doi: 10.1016/j.oraloncology.2015.01.006.
    1. Tas SW, Maracle CX, Balogh E, Szekanecz Z. Targeting of proangiogenic signalling pathways in chronic inflammation. Nat Rev Rheumatol. 2016;12:111–122. doi: 10.1038/nrrheum.2015.164.
    1. Simon T, Gagliano T, Giamas G. Direct effects of anti-angiogenic therapies on tumor cells: VEGF signaling. Trends Mol Med. 2017;23:282–292. doi: 10.1016/j.molmed.2017.01.002.
    1. Ye W. The complexity of translating anti-angiogenesis therapy from basic science to the clinic. Dev Cell. 2016;37:114–125. doi: 10.1016/j.devcel.2016.03.015.
    1. Al-Bataineh O, Jenne J, Huber P. Clinical and future applications of high intensity focused ultrasound in cancer. Cancer Treat Rev. 2012;38:346–353. doi: 10.1016/j.ctrv.2011.08.004.
    1. Kennedy JE. High-intensity focused ultrasound in the treatment of solid tumours. Nat Rev Cancer. 2005;5:321–327. doi: 10.1038/nrc1591.
    1. Brown MR, Farquhar-Smith P, Williams JE, ter Haar G, deSouza NM. The use of high-intensity focused ultrasound as a novel treatment for painful conditions-a description and narrative review of the literature. Br J Anaesth. 2015;115:520–530. doi: 10.1093/bja/aev302.
    1. Pan H, Zhou W, Wang S. Pulsed focused ultrasound stimulates the release of tumor biomarkers into the blood circulation. Radiology. 2017;285:1058–1060. doi: 10.1148/radiol.2017171654.
    1. Hariharan P, Myers MR, Banerjee RK. HIFU procedures at moderate intensities-effect of large blood vessels. Phys Med Biol. 2007;52:3493–3513. doi: 10.1088/0031-9155/52/12/011.
    1. Qiao Y, Yin H, Li Z, Wan M. Cavitation distribution within large phantom vessel and mechanical damage formed on surrounding vessel wall. Ultrason Sonochem. 2013;20:1376–1383. doi: 10.1016/j.ultsonch.2013.03.007.
    1. Mizrahi N, Zhou EH, Lenormand G, Krishnan R, Weihs D, Butler JP, Weitz DA, Fredberg JJ, Kimmel E. Low intensity ultrasound perturbs cytoskeleton dynamics. Soft matter. 2012;8:2438–2443. doi: 10.1039/c2sm07246g.
    1. Rutten S, Nolte PA, Korstjens CM, Klein-Nulend J. Low-intensity pulsed ultrasound affects RUNX2 immunopositive osteogenic cells in delayed clinical fracture healing. Bone. 2009;45:862–869. doi: 10.1016/j.bone.2009.07.012.
    1. Hitchcock KE, Holland CK. Ultrasound-assisted thrombolysis for stroke therapy: Better thrombus break-up with bubbles. Stroke. 2010;41:S50–S53. doi: 10.1161/STROKEAHA.110.595348.
    1. Zhang Z, Chen J, Chen L, Yang X, Zhong H, Qi X, Bi Y, Xu K. Low frequency and intensity ultrasound induces apoptosis of brain glioma in rats mediated by caspase-3, Bcl-2, and survivin. Brain Res. 2012;1473:25–34. doi: 10.1016/j.brainres.2012.06.047.
    1. Zhou XY, Wu SY, Zhang ZC, Wang F, Yang YL, Li M, Wei XZ. Low-intensity pulsed ultrasound promotes endothelial cell-mediated osteogenesis in a conditioned medium coculture system with osteoblasts. Medicine (Baltimore) 2017;96:e8397. doi: 10.1097/MD.0000000000008397.
    1. Marquez RT, Xu L. Bcl-2: Beclin 1 complex: Multiple, mechanisms regulating autophagy/apoptosis toggle switch. Am J Cancer Res. 2012;2:214–221.
    1. Rovetta F, Stacchiotti A, Consiglio A, Cadei M, Grigolato PG, Lavazza A, Rezzani R, Aleo MF. ER signaling regulation drives the switch between autophagy and apoptosis in NRK-52E cells exposed to cisplatin. Exp Cell Res. 2012;318:238–250. doi: 10.1016/j.yexcr.2011.11.008.
    1. Furusawa Y, Zhao QL, Hassan MA, Tabuchi Y, Takasaki I, Wada S, Kondo T. Ultrasound-induced apoptosis in the presence of Sonazoid and associated alterations in gene expression levels: A possible therapeutic application. Cancer Lett. 2010;288:107–115. doi: 10.1016/j.canlet.2009.06.029.
    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. doi: 10.1016/j.joen.2015.12.019.
    1. Lee GS, Park JH, Shin US, Kim HW. Direct deposited porous scaffolds of calcium phosphate cement with alginate for drug delivery and bone tissue engineering. Acta Biomater. 2011;7:3178–3186. doi: 10.1016/j.actbio.2011.04.008.
    1. Mason TJ. Therapeutic ultrasound an overview. Ultrason Sonochem. 2011;18:847–852. doi: 10.1016/j.ultsonch.2011.01.004.
    1. Hou R, Xu Y, Lu Q, Zhang Y, Hu B. Effect of low-frequency low-intensity ultrasound with microbubbles on prostate cancer hypoxia. Tumour Biol. 2017;39:1010428317719275. doi: 10.1177/1010428317719275.
    1. Chen H, Brayman AA, Bailey MR, Matula TJ. Blood vessel rupture by cavitation. Urol Res. 2010;38:321–326. doi: 10.1007/s00240-010-0302-5.
    1. Elmore S. Apoptosis: A review of programmed cell death. Toxicol Pathol. 2007;35:495–516. doi: 10.1080/01926230701320337.
    1. Chen Y, Brandizzi F. IRE1: ER stress sensor and cell fate executor. Trends Cell Biol. 2013;23:547–555. doi: 10.1016/j.tcb.2013.06.005.
    1. Bhutia SK, Dash R, Das SK, Azab B, Su ZZ, Lee SG, Grant S, Yacoub A, Dent P, Curiel DT, et al. Mechanism of autophagy to apoptosis switch triggered in prostate cancer cells by antitumor cytokine melanoma differentiation-associated gene 7/interleukin-24. Cancer Res. 2010;70:3667–3676. doi: 10.1158/0008-5472.CAN-09-3647.
    1. Wu H, Che X, Zheng Q, Wu A, Pan K, Shao A, Wu Q, Zhang J, Hong Y. Caspases: A molecular switch node in the crosstalk between autophagy and apoptosis. Int J Biol Sci. 2014;10:1072–1083. doi: 10.7150/ijbs.9719.
    1. Obata T, Brown GE, Yaffe MB. MAP kinase pathways activated by stress: The p38 MAPK pathway. Crit Care Med. 2000;28:N67–N77. doi: 10.1097/00003246-200004001-00008.
    1. Whitmarsh AJ. A central role for p38 MAPK in the early transcriptional response to stress. BMC Biol. 2010;8:47. doi: 10.1186/1741-7007-8-47.
    1. Trempolec N, Dave-Coll N, Nebreda AR. SnapShot: p38 MAPK substrates. Cell. 2013;152:924–924.e1. doi: 10.1016/j.cell.2013.01.047.
    1. Grossi V, Peserico A, Tezil T, Simone C. p38alpha MAPK pathway: A key factor in colorectal cancer therapy and chemoresistance. World J Gastroenterol. 2014;20:9744–9758. doi: 10.3748/wjg.v20.i29.9744.
    1. Olson JM, Hallahan AR. p38 MAP kinase: A convergence point in cancer therapy. Trends Mol Med. 2004;10:125–129. doi: 10.1016/j.molmed.2004.01.007.
    1. Pang H, Cai L, Yang Y, Chen X, Sui G, Zhao C. Knockdown of osteopontin chemosensitizes MDA-MB-231 cells to cyclophosphamide by enhancing apoptosis through activating p38 MAPK pathway. Cancer Biother Radiopharm. 2011;26:165–173. doi: 10.1089/cbr.2010.0838.
    1. Sui X, Kong N, Ye L, Han W, Zhou J, Zhang Q, He C, Pan H. p38 and JNK MAPK pathways control the balance of apoptosis and autophagy in response to chemotherapeutic agents. Cancer Lett. 2014;344:174–179. doi: 10.1016/j.canlet.2013.11.019.
    1. Matsui H, Fukuno N, Kanda Y, Kantoh Y, Chida T, Nagaura Y, Suzuki O, Nishitoh H, Takeda K, Ichijo H, et al. The expression of Fn14 via mechanical stress-activated JNK contributes to apoptosis induction in osteoblasts. J Biol Chem. 2014;289:6438–6450. doi: 10.1074/jbc.M113.536300.
    1. Mu C, Lv T, Wang Z, Ma S, Ma J, Liu J, Yu J, Mu J. Mechanical stress stimulates the osteo/odontoblastic differentiation of human stem cells from apical papilla via erk 1/2 and JNK MAPK pathways. Biomed Res Int. 2014;2014:494378. doi: 10.1155/2014/494378.
    1. Xu T, Gu J, Li C, Guo X, Tu J, Zhang D, Sun W, Kong X. Low-intensity pulsed ultrasound suppresses proliferation and promotes apoptosis via p38 MAPK signaling in rat visceral preadipocytes. Am J Transl Res. 2018;10:948–956.
    1. Jiang Q, Li F, Shi K, Wu P, An J, Yang Y, Xu C. Involvement of p38 in signal switching from autophagy to apoptosis via the PERK/eIF2α/ATF-4 axis in selenite-treated NB4 cells. Cell Death Dis. 2014;5:e1270. doi: 10.1038/cddis.2014.200.
    1. Rozpedek W, Pytel D, Mucha B, Leszczynska H, Diehl JA, Majsterek I. The role of the PERK/eIF2α/ATF-4/CHOP signaling pathway in tumor progression during endoplasmic reticulum stress. Curr Mol Med. 2016;16:533–544. doi: 10.2174/1566524016666160523143937.

Source: PubMed

3
Předplatit