Human amniotic epithelial cells inhibit growth of epithelial ovarian cancer cells via TGF‑β1-mediated cell cycle arrest

Shixia Bu, Qiuwan Zhang, Qian Wang, Dongmei Lai, Shixia Bu, Qiuwan Zhang, Qian Wang, Dongmei Lai

Abstract

It is reported that human amniotic epithelial cells (hAECs) endow intrinsic antitumor effects on certain kinds of cancer. This research was designed to evaluate whether hAECs endowed potential anticancer properties on epithelial ovarian cancer (EOC) cells in vivo and in vitro, which has not been reported before. In this study, we established a xenografted BALB/c nude mouse model by subcutaneously co-injecting ovarian cancer cell line, SK-OV-3, and hAECs for 28 days. In ex vivo experiments, CCK‑8 cell viability assay, real-time PCR, cell counting assay, cell cycle analysis and immunohistochemistry (IHC) assay were used to detect the effects of hAEC‑secreted factors on the proliferation and cell cycle progression of EOC cells. A cytokine array was conducted to detect anticancer-related cytokines released from hAECs. Human recombinant TGF‑β1 and TGF‑β1 antibody were used to treat EOC cells and analyzed whether TGF‑β1 contributed to the cell cycle arrest. Results from in vivo and ex vivo experiments showed that hAEC-secreted factors and rhTGF‑β1 decreased proliferation of EOC cells and induced G0/G1 cell cycle arrest in cancer cells, which could be partially reversed by excess TGF‑β1 antibody. These data indicate that hAECs endow potential anticancer properties on epithelial ovarian cancer in vivo and in vitro which is partially mediated by hAEC‑secreted TGF‑β1-induced cell cycle arrest. This study suggests a potential application of hAEC‑based therapy against epithelial ovarian cancer.

Figures

Figure 1
Figure 1
hAECs inhibit proliferation of EOC cells in vitro in a paracrine manner. (A) hAECs was negative for mesenchymal marker vimentin (green) and positive for epithelial marker CK-7 (red) by using immunofluorescence. DAPI (blue) staining showed the nuclei (scale bar, 100 µm). (B) CCK-8 cell viability assay used to test the effects of hAEC-CM in different concentrations on the viability of EOC cells at 48 h (n=6; performed in triplicate). Ordinary ANOVA was used for statistical analysis. (C) Real-time PCR were used to test the effects of hAECs on the expression levels of PCNA and Ki-67 in EOC cells cultured in Transwell system for 48 h (n=3; performed in triplicate). Data are represented as means ± SEM. *p<0.05 and ***p<0.001.
Figure 2
Figure 2
hAECs inhibit growth of SK-OV-3 cells in vivo. (A) Gross observation of subcutaneous xenografts obtained from SK-OV-3 injected group and SK-OV-3/hAECs co-injected group (scale bar, l cm). (B) The average tumor weight of SK-OV-3 injected group was significantly higher than that of SK-OV-3/hAECs co-injected group at day 28 (n=10). (C) The average tumor volume of SK-OV-3 injected group was significantly bigger than that of SK-OV-3/hAECs co-injected group at day 14 and day 28 (n=10). (D) (a) Representative image of GFP-transfected hAECs (scale bar, 200 µm). (b) We did not find positive signal of hAECsGFP+ in the negative control (scale bar, 100 µm). (c) The positive signals were found in the xenografted tumor tissues obtained from SK-OV-3/hAECsGFP+ injected group (scale bar, 100 µm). (E and F) Proliferation of cancer cells were tested by IHC using antibodies against PCNA and Ki-67 in SK-OV-3 and SK-OV-3/hAECs tumor tissues (scale bar, 100 µm; n=5). Data were analyzed by Mann-Whitney U test and were represented as means ± SEM. *p<0.05, **p<0.01 and ***p<0.001.
Figure 3
Figure 3
hAECs induce G0/G1 cell cycle arrest in EOC cells in a paracrine manner. (A) The effects of hAEC-CM on EOC cell division were tested by cell counting assay (n=6; performed in triplicate). (B) Cell cycle analysis revealed that hAEC-secreted factors induced G0/G1 cell cycle arrest in EOC cells in Transwell system (n=3; performed in triplicate). Data are represented as means ± SEM. *p<0.05, **p<0.01 and ***p<0.001.
Figure 4
Figure 4
hAECs regulate the expression of proteins related to G0/G1 cell cycle arrest determined by IHC. hAECs upregulated p16INK4A, p21 and phospho-JNK expression and downregulated phospho-pRB (Ser807) expression in EOC cells (n=5). Scale bar of (A), (B) and (C) were 100 µm and that of (D) was 50 µm. H-score system was used for semi-quantification of p16INK4A, p21 and phospho-JNK between two groups. Data were analyzed by Mann-Whitney U test and presented as means ± SEM. **p<0.01.
Figure 5
Figure 5
TGF-β1 was enriched in hAEC-released cell cycle-regulatory cytokines and induced G0/G1 cell cycle arrest in EOC cells. (A) (a) Selective map of the human antibody array 1000. hAECs released multiple cell cycle-regulatory cytokines, including TGF-β1, GM-CSF, IL-8, IFN-γ, IL-6, IL-1, TNF-α, TGF-β2 and Smad 7. (b) The intensities of the signals were quantified by densitometry and the expression of Smad 7 was regarded as control. (B) CCK-8 cell viability assay used to test the effects of rhTGF-β1 in different concentrations on the viability of EOC cells at 48 h (n=6; performed in triplicate). Data were analyzed by Mann-Whitney U test. (C) After being treated with rhTGF-β1, cell cycle of EOC cells was analyzed by flow cytometry. (D) ELISA was used to test the efficiency of TGF-β1 antibody added to neutralize the function of hAEC-secreted TGF-β1. Data were analyzed by Kruskal-Wallis test. (E) After being treated with TGF-β1 antibody in the Transwell system, cell cycle of EOC cells was analyzed by flow cytometry. Data are presented as means ± SEM. *p<0.05, **p<0.01 and ***p<0.001.

References

    1. Barbati A, Grazia Mameli M, Sidoni A, Di Renzo GC. Amniotic membrane: Separation of amniotic mesoderm from amniotic epithelium and isolation of their respective mesenchymal stromal and epithelial cells. Curr Protoc Stem Cell Biol Chapter. 2012;1:8.
    1. Ilancheran S, Michalska A, Peh G, Wallace EM, Pera M, Manuelpillai U. Stem cells derived from human fetal membranes display multilineage differentiation potential. Biol Reprod. 2007;77:577–588. doi: 10.1095/biolreprod.106.055244.
    1. Miki T, Lehmann T, Cai H, Stolz DB, Strom SC. Stem cell characteristics of amniotic epithelial cells. Stem Cells. 2005;23:1549–1559. doi: 10.1634/stemcells.2004-0357.
    1. Yao X, Guo Y, Wang Q, Xu M, Zhang Q, Li T, Lai D. The paracrine effect of transplanted human amniotic epithelial cells on ovarian function improvement in a mouse model of chemotherapy-induced primary ovarian insufficiency. Stem Cells Int. 2016;2016:4148923. doi: 10.1155/2016/4148923.
    1. Si JW, Zhang JJ, Dai JW, Yu DD, Yu HB, Shi J, Wang XD, Shen SGF, Guo LH. Osteogenic differentiation of human amniotic epithelial cells and its application in alveolar defect restoration. Stem Cells Transl Med. 2014;3:1504–1513. doi: 10.5966/sctm.2014-0118.
    1. Jiang LW, Chen H, Lu H. Using human epithelial amnion cells in human de-epidermized dermis for skin regeneration. J Dermatol Sci. 2016;81:26–34. doi: 10.1016/j.jdermsci.2015.10.018.
    1. Niknejad H, Paeini-Vayghan G, Tehrani FA, Khayat-Khoei M, Peirovi H. Side dependent effects of the human amnion on angiogenesis. Placenta. 2013;34:340–345. doi: 10.1016/j.placenta.2013.02.001.
    1. Niknejad H, Khayat-Khoei M, Peirovi H, Abolghasemi H. Human amniotic epithelial cells induce apoptosis of cancer cells: A new anti-tumor therapeutic strategy. Cytotherapy. 2014;16:33–40. doi: 10.1016/j.jcyt.2013.07.005.
    1. Kang NH, Yi BR, Lim SY, Hwang KA, Baek YS, Kang KS, Choi KC. Human amniotic membrane-derived epithelial stem cells display anticancer activity in BALB/c female nude mice bearing disseminated breast cancer xenografts. Int J Oncol. 2012;40:2022–2028.
    1. Mamede AC, Laranjo M, Carvalho MJ, Abrantes AM, Pires AS, Brito AF, Moura P, Maia CJ, Botelho MF. Effect of amniotic membrane proteins in human cancer cell lines: An exploratory study. J Membr Biol. 2014;247:357–360. doi: 10.1007/s00232-014-9642-3.
    1. Zhang Q, Xu M, Yao X, Li T, Wang Q, Lai D. Human amniotic epithelial cells inhibit granulosa cell apoptosis induced by chemotherapy and restore the fertility. Stem Cell Res Ther. 2015;6:152. doi: 10.1186/s13287-015-0148-4.
    1. Liu T, Zhao L, Zhang Y, Chen W, Liu D, Hou H, Ding L, Li X. Ginsenoside 20(S)-Rg3 targets HIF-1α to block hypoxia-induced epithelial-mesenchymal transition in ovarian cancer cells. PLoS One. 2014;9:e103887. doi: 10.1371/journal.pone.0103887.
    1. Budwit-Novotny DA, McCarty KS, Cox EB, Soper JT, Mutch DG, Creasman WT, Flowers JL, McCarty KS., Jr Immunohistochemical analyses of estrogen receptor in endometrial adenocarcinoma using a monoclonal antibody. Cancer Res. 1986;46:5419–5425.
    1. Jin G, Cao Z, Sun X, Wang K, Huang T, Shen B. Protein O-glucosyltransferase 1 overexpression downregulates p16 in BT474 human breast cancer cells. Oncol Lett. 2014;8:594–600.
    1. Datto MB, Yu Y, Wang XF. Functional analysis of the transforming growth factor beta responsive elements in the WAF1/Cip1/p21 promoter. J Biol Chem. 1995;270:28623–28628. doi: 10.1074/jbc.270.48.28623.
    1. Kim GY, Mercer SE, Ewton DZ, Yan Z, Jin K, Friedman E. The stress-activated protein kinases p38 alpha and JNK1 stabilize p21 (Cip1) by phosphorylation. J Biol Chem. 2002;277:29792–29802. doi: 10.1074/jbc.M201299200.
    1. Wang F, Wang L, Yao X, Lai D, Guo L. Human amniotic epithelial cells can differentiate into granulosa cells and restore folliculogenesis in a mouse model of chemotherapy-induced premature ovarian failure. Stem Cell Res Ther. 2013;4:124. doi: 10.1186/scrt335.
    1. Jayson GC, Kohn EC, Kitchener HC, Ledermann JA. Ovarian cancer. Lancet. 2014;384:1376–1388. doi: 10.1016/S0140-6736(13)62146-7.
    1. Di Germanio C, Bernier M, Petr M, Mattioli M, Barboni B, de Cabo R. Conditioned medium derived from rat amniotic epithelial cells confers protection against inflammation, cancer, and senescence. Oncotarget. 2016;7:39051–39064. doi: 10.18632/oncotarget.9694.
    1. Dickson MA. Molecular pathways: CDK4 inhibitors for cancer therapy. Clin Cancer Res. 2014;20:3379–3383. doi: 10.1158/1078-0432.CCR-13-1551.
    1. Magatti M, De Munari S, Vertua E, Parolini O. Amniotic membrane-derived cells inhibit proliferation of cancer cell lines by inducing cell cycle arrest. J Cell Mol Med. 2012;16:2208–2218. doi: 10.1111/j.1582-4934.2012.01531.x.
    1. Chen X, Zheng S, Gao Y, Dai H, Mou H, Yang H. Growth regulation of ovarian cancer cell line HO-8910 by transforming growth factor beta 1 in vitro. Chin Med J (Engl) 1998;111:546–550.
    1. Ketley NJ, Allen PD, Kelsey SM, Newland AC. Modulation of idarubicin-induced apoptosis in human acute myeloid leukemia blasts by all-trans retinoic acid, 1,25(OH)2 vitamin D3, and granulocyte-macrophage colony-stimulating factor. Blood. 1997;90:4578–4587.
    1. Schinke C, Giricz O, Li W, Shastri A, Gordon S, Barreyro L, Bhagat T, Bhattacharyya S, Ramachandra N, Bartenstein M, et al. IL8-CXCR2 pathway inhibition as a therapeutic strategy against MDS and AML stem cells. Blood. 2015;125:3144–3152. doi: 10.1182/blood-2015-01-621631.
    1. Resnitzky D, Tiefenbrun N, Berissi H, Kimchi A. Interferons and interleukin 6 suppress phosphorylation of the retinoblastoma protein in growth-sensitive hematopoietic cells. Proc Natl Acad Sci USA. 1992;89:402–406. doi: 10.1073/pnas.89.1.402.
    1. Muthukkumar S, Sells SF, Crist SA, Rangnekar VM. Interleukin-1 induces growth arrest by hypophosphorylation of the retinoblastoma susceptibility gene product RB. J Biol Chem. 1996;271:5733–5740. doi: 10.1074/jbc.271.10.5733.
    1. Lillibridge CD, O'Connell BC. In human salivary gland cells, overexpression of E2F1 overcomes an interferon-gamma- and tumor necrosis factor-alpha-induced growth arrest but does not result in complete mitosis. J Cell Physiol. 1997;172:343–350. doi: 10.1002/(SICI)1097-4652(199709)172:3<343::AID-JCP8>;2-O.
    1. Wu DT, Bitzer M, Ju W, Mundel P, Böttinger EP. TGF-beta concentration specifies differential signaling profiles of growth arrest/differentiation and apoptosis in podocytes. J Am Soc Nephrol. 2005;16:3211–3221. doi: 10.1681/ASN.2004121055.
    1. Kitamura K, Aota S, Sakamoto R, Emori T, Okazaki K. Smad7 induces G0/G1 cell cycle arrest in mesenchymal cells by inhibiting the expression of G1 cyclins. Dev Growth Differ. 2005;47:537–552. doi: 10.1111/j.1440-169X.2005.00829.x.
    1. Malumbres M, Barbacid M. Cell cycle, CDKs and cancer: A changing paradigm. Nat Rev Cancer. 2009;9:153–166. doi: 10.1038/nrc2602.
    1. Schlosshauer PW, Deligdisch L, Penault-Llorca F, Fatemi D, Qiao R, Yao S, Pearl M, Yang Z, Sheng T, Dong J. Loss of p16I NK4A expression in low-grade ovarian serous carcinomas. Int J Gynecol Pathol. 2011;30:22–29. doi: 10.1097/PGP.0b013e3181ed89b3.
    1. Kudoh K, Ichikawa Y, Yoshida S, Hirai M, Kikuchi Y, Nagata I, Miwa M, Uchida K. Inactivation of p16/CDKN2 and p15/MTS2 is associated with prognosis and response to chemotherapy in ovarian cancer. Int J Cancer. 2002;99:579–582. doi: 10.1002/ijc.10331.
    1. Buchynska LG, Nesina IP, Yurchenko NP, Bilyk OO, Grinkevych VN, Svintitsky VS. Expression of p53, p21WAF1/CIP1, p16INK4A and Ki-67 proteins in serous ovarian tumors. Exp Oncol. 2007;29:49–53.
    1. Bali A, O'Brien PM, Edwards LS, Sutherland RL, Hacker NF, Henshall SM. Cyclin D1, p53, and p21Waf1/Cip1 expression is predictive of poor clinical outcome in serous epithelial ovarian cancer. Clin Cancer Res. 2004;10:5168–5177. doi: 10.1158/1078-0432.CCR-03-0751.
    1. Khalil N, Xu YD, O'Connor R, Duronio V. Proliferation of pulmonary interstitial fibroblasts is mediated by transforming growth factor-beta1-induced release of extracellular fibroblast growth factor-2 and phosphorylation of p38 MAPK and JNK. J Biol Chem. 2005;280:43000–43009. doi: 10.1074/jbc.M510441200.
    1. Henley SA, Dick FA. The retinoblastoma family of proteins and their regulatory functions in the mammalian cell division cycle. Cell Div. 2012;7:10. doi: 10.1186/1747-1028-7-10.
    1. Francis SM, Bergsied J, Isaac CE, Coschi CH, Martens AL, Hojilla CV, Chakrabarti S, Dimattia GE, Khoka R, Wang JY, et al. A functional connection between pRB and transforming growth factor beta in growth inhibition and mammary gland development. Mol Cell Biol. 2009;29:4455–4466. doi: 10.1128/MCB.00473-09.

Source: PubMed

3
订阅