Amniotic membrane-derived cells inhibit proliferation of cancer cell lines by inducing cell cycle arrest

Marta Magatti, Silvia De Munari, Elsa Vertua, Ornella Parolini, Marta Magatti, Silvia De Munari, Elsa Vertua, Ornella Parolini

Abstract

Cells derived from the amniotic foetal membrane of human term placenta have drawn particular attention mainly for their plasticity and immunological properties, which render them interesting for stem-cell research and cell-based therapeutic applications. In particular, we have previously demonstrated that amniotic mesenchymal tissue cells (AMTC) inhibit lymphocyte proliferation in vitro and suppress the generation and maturation of monocyte-derived dendritic cells. Here, we show that AMTC also significantly reduce the proliferation of cancer cell lines of haematopoietic and non-haematopoietic origin, in both cell-cell contact and transwell co-cultures, therefore suggesting the involvement of yet-unknown inhibitory soluble factor(s) in this 'cell growth restraint'. Importantly, we provide evidence that the anti-proliferative effect of AMTC is associated with induction of cell cycle arrest in G0/G1 phase. Gene expression analyses demonstrate that AMTC can down-regulate cancer cells' mRNA expression of genes associated with cell cycle progression, such as cyclins (cyclin D2, cyclin E1, cyclin H) and cyclin-dependent kinase (CDK4, CDK6 and CDK2), whilst they up-regulate cell cycle negative regulator such as p15 and p21, consistent with a block in G0/G1 phase with no progression to S phase. Taken together, these findings warrant further studies to investigate the applicability of these cells for controlling cancer cell proliferation in vivo.

© 2012 The Authors Journal of Cellular and Molecular Medicine © 2012 Foundation for Cellular and Molecular Medicine/Blackwell Publishing Ltd.

Figures

Fig 1
Fig 1
AMTC reduce the proliferation of cancer cells of both haematopoietic and non-haematopoietic origin. Haematopoietic and non-haematopoietic cancer cell lines were cultured alone (control), or with AMTC either in direct contact (upper panel) or with physical separation (transwell setting, lower panel). Different cancer cell: AMTC ratios were used. After 3 days of culture, cancer cell proliferation was assessed by [3H]-thymidine incorporation and reported as a percentage of cell proliferation in comparison with control cancer cell proliferation. Data are expressed as mean ± S.D. of at least four independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001 versus corresponding control sample.
Fig 2
Fig 2
Alteration in cancer cell proliferation by AMTC and other cell lines. Jurkat and U937 cells were cultured alone or in the presence of different γ-irradiated cell types (AMTC or BM-MSC, dermal fibroblast cells, KG1a, U937 and Jurkat). Co-cultures were performed in both contact and transwell settings, at cancer cell: different cell types ratios of 1:2. After 1, 2 and 3 days of culture, Jurkat and U937 cell proliferation was assessed by [3H]-thymidine incorporation. Data are expressed as mean of more than four independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001 versus corresponding control sample.
Fig 3
Fig 3
AMTC arrest the cell cycle of cancer cells. Jurkat (upper panel) and U937 (lower panel) cells were cultured alone or in the presence of AMTC, in either contact or transwell settings, at a cancer cell: AMTC ratio of 1:2. At 16, 24 and 48 hrs, cultures were pulsed with BrdU for 1h, and cancer cells were then collected and stained with anti-BrdU APC and 7-AAD. The percentage of cells in the G0/G1, S or G2/M phase of the cell cycle is indicated. The FACS profiles shown are representative of three independent experiments.

References

    1. Garcia-Gomez I, Elvira G, Zapata AG, et al. Mesenchymal stem cells: biological properties and clinical applications. Expert Opin Biol Ther. 2010;10:1453–68.
    1. Parolini O, Alviano F, Bergwerf I, et al. Toward cell therapy using placenta-derived cells: disease mechanisms, cell biology, preclinical studies, and regulatory aspects at the round table. Stem Cells Dev. 2010;19:143–54.
    1. Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat Rev Immunol. 2008;8:726–36.
    1. Pistoia V, Raffaghello L. Potential of mesenchymal stem cells for the therapy of autoimmune diseases. Expert Rev Clin Immunol. 2010;6:211–8.
    1. Bernardo ME, Locatelli F, Fibbe WE. Mesenchymal stromal cells. Ann N Y Acad Sci. 2009;1176:101–17.
    1. Ramasamy R, Lam EW, Soeiro I, et al. Mesenchymal stem cells inhibit proliferation and apoptosis of tumor cells: impact on in vivo tumor growth. Leukemia. 2007;21:304–10.
    1. Sarmadi VH, Heng FS, Ramasamy R. The effect of human mesenchymal stem cells on tumour cell proliferation. Med J Malaysia. 2008;63:63–4.
    1. Zhang HM, Zhang LS. Influence of human bone marrow mesenchymal stem cells on proliferation of chronic myeloid leukemia cells. Chin J Cancer. 2009;28:29–32.
    1. Motaln H, Schichor C, Lah TT. Human mesenchymal stem cells and their use in cell-based therapies. Cancer. 2010;116:2519–30.
    1. Mousavi Niri N, Jaberipour M, Razmkhah M, et al. Mesenchymal stem cells do not suppress lymphoblastic leukemic cell line proliferation. Iran J Immunol. 2009;6:186–94.
    1. Djouad F, Bony C, Apparailly F, et al. Earlier onset of syngeneic tumors in the presence of mesenchymal stem cells. Transplantation. 2006;82:1060–6.
    1. Kidd S, Spaeth E, Klopp A, et al. The (in) auspicious role of mesenchymal stromal cells in cancer: be it friend or foe. Cytotherapy. 2008;10:657–67.
    1. Feng B, Chen L. Review of mesenchymal stem cells and tumors: executioner or coconspirator? Cancer Biother Radiopharm. 2009;24:717–21.
    1. Qiao L, Xu Z, Zhao T, et al. Suppression of tumorigenesis by human mesenchymal stem cells in a hepatoma model. Cell Res. 2008;18:500–7.
    1. Lu YR, Yuan Y, Wang XJ, et al. The growth inhibitory effect of mesenchymal stem cells on tumor cells in vitro and in vivo. Cancer Biol Ther. 2008;7:245–51.
    1. Xu G, Zhang Y, Zhang L, et al. Bone marrow stromal cells induce apoptosis of lymphoma cells in the presence of IFNgamma and TNF by producing nitric oxide. Biochem Biophys Res Commun. 2008;375:666–70.
    1. Loebinger MR, Janes SM. Stem cells as vectors for antitumour therapy. Thorax. 2010;65:362–9.
    1. Hung SC, Deng WP, Yang WK, et al. Mesenchymal stem cell targeting of microscopic tumors and tumor stroma development monitored by noninvasive in vivo positron emission tomography imaging. Clin Cancer Res. 2005;11:7749–56.
    1. Nakamizo A, Marini F, Amano T, et al. Human bone marrow-derived mesenchymal stem cells in the treatment of gliomas. Cancer Res. 2005;65:3307–18.
    1. Khakoo AY, Pati S, Anderson SA, et al. Human mesenchymal stem cells exert potent antitumorigenic effects in a model of Kaposi's sarcoma. J Exp Med. 2006;203:1235–47.
    1. Dwyer RM, Khan S, Barry FP, et al. Advances in mesenchymal stem cell-mediated gene therapy for cancer. Stem Cell Res Ther. 2010;1:25.
    1. Klopp AH, Gupta A, Spaeth E, et al. Concise review: dissecting a discrepancy in the literature: do mesenchymal stem cells support or suppress tumor growth? Stem Cells. 2011;29:11–9.
    1. Bailo M, Soncini M, Vertua E, et al. Engraftment potential of human amnion and chorion cells derived from term placenta. Transplantation. 2004;78:1439–48.
    1. Magatti M, De Munari S, Vertua E, et al. Human amnion mesenchyme harbors cells with allogeneic T-cell suppression and stimulation capabilities. Stem Cells. 2008;26:182–92.
    1. Magatti M, De Munari S, Vertua E, et al. Amniotic mesenchymal tissue cells inhibit dendritic cell differentiation of peripheral blood and amnion resident monocytes. Cell Transplant. 2009;18:899–914.
    1. Parolini O, Caruso M. Review: Preclinical studies on placenta-derived cells and amniotic membrane: an update. Placenta. 2011;32:S186–95.
    1. Soncini M, Vertua E, Gibelli L, et al. Isolation and characterization of mesenchymal cells from human foetal membranes. J Tissue Eng Regen Med. 2007;1:296–305.
    1. Koeffler HP, Billing R, Lusis AJ, et al. An undifferentiated variant derived from the human acute myelogenous leukemia cell line (KG-1) Blood. 1980;56:265–73.
    1. Jost JP, Oakeley EJ, Zhu B, et al. 5-Methylcytosine DNA glycosylase participates in the genome-wide loss of DNA methylation occurring during mouse myoblast differentiation. Nucleic Acids Res. 2001;29:4452–61.
    1. Yu ZK, Gervais JL, Zhang H. Human CUL-1 associates with the SKP1/SKP2 complex and regulates p21(CIP1/WAF1) and cyclin D proteins. Proc Natl Acad Sci USA. 1998;95:11324–9.
    1. Guo W, Shang F, Liu Q, et al. Differential regulation of components of the ubiquitin-proteasome pathway during lens cell differentiation. Invest Ophthalmol Vis Sci. 2004;45:1194–201.
    1. Parolini O, Alviano F, Bagnara GP, et al. Concise review: isolation and characterization of cells from human term placenta: outcome of the first international Workshop on Placenta Derived Stem Cells. Stem Cells. 2008;26:300–11.
    1. Zhu Y, Sun Z, Han Q, et al. Human mesenchymal stem cells inhibit cancer cell proliferation by secreting DKK-1. Leukemia. 2009;23:925–33.
    1. Kronsteiner B, Wolbank S, Peterbauer A, et al. Human mesenchymal stem cells from adipose tissue and amnion influence T-cells depending on stimulation method and presence of other immune cells. Stem Cells Dev. 2011;20:2115–26.
    1. Eisenthal A, Kashtan H, Rabau M, et al. Antitumor effects of recombinant interleukin-6 expressed in eukaryotic cells. Cancer Immunol Immunother. 1993;36:101–7.
    1. Kojiro S, Yano H, Ogasawara S, et al. Antiproliferative effects of 5-fluorouracil and interferon-alpha in combination on a hepatocellular carcinoma cell line in vitro and in vivo. J Gastroenterol Hepatol. 2006;21:129–37.
    1. Driessens G, Nuttin L, Gras A, et al. Development of a successful antitumor therapeutic model combining in vivo dendritic cell vaccination with tumor irradiation and intratumoral GM-CSF delivery. Cancer Immunol Immunother. 2011;60:273–81.
    1. Maestroni GJ, Hertens E, Galli P. Factor(s) from nonmacrophage bone marrow stromal cells inhibit Lewis lung carcinoma and B16 melanoma growth in mice. Cell Mol Life Sci. 1999;55:663–7.
    1. Potian JA, Aviv H, Ponzio NM, et al. Veto-like activity of mesenchymal stem cells: functional discrimination between cellular responses to alloantigens and recall antigens. J Immunol. 2003;171:3426–34.
    1. Krampera M, Glennie S, Dyson J, et al. Bone marrow mesenchymal stem cells inhibit the response of naive and memory antigen-specific T cells to their cognate peptide. Blood. 2003;101:3722–9.
    1. Haniffa MA, Wang XN, Holtick U, et al. Adult human fibroblasts are potent immunoregulatory cells and functionally equivalent to mesenchymal stem cells. J Immunol. 2007;179:1595–604.
    1. Haniffa MA, Collin MP, Buckley CD, et al. Mesenchymal stem cells: the fibroblasts' new clothes? Haematologica. 2009;94:258–63.
    1. Li H, Niederkorn JY, Neelam S, et al. Immunosuppressive factors secreted by human amniotic epithelial cells. Invest Ophthalmol Vis Sci. 2005;46:900–7.
    1. Li X, Ling W, Pennisi A, et al. Human placenta-derived adherent cells prevent bone loss, stimulate bone formation, and suppress growth of multiple myeloma in bone. Stem Cells. 2011;29:263–73.
    1. Malumbres M, Barbacid M. Cell cycle, CDKs and cancer: a changing paradigm. Nat Rev Cancer. 2009;9:153–66.
    1. Hengst L, Reed SI. Inhibitors of the Cip/Kip family. Curr Top Microbiol Immunol. 1998;227:25–41.
    1. Harper JW, Adami GR, Wei N, et al. The p21 Cdk-interacting protein Cip1 is a potent inhibitor of G1 cyclin-dependent kinases. Cell. 1993;75:805–16.
    1. Horne MC, Donaldson KL, Goolsby GL, et al. Cyclin G2 is up-regulated during growth inhibition and B cell antigen receptor-mediated cell cycle arrest. J Biol Chem. 1997;272:12650–61.
    1. Bennin DA, Don AS, Brake T, et al. Cyclin G2 associates with protein phosphatase 2A catalytic and regulatory B' subunits in active complexes and induces nuclear aberrations and a G1/S phase cell cycle arrest. J Biol Chem. 2002;277:27449–67.
    1. Martinez-Gac L, Marques M, Garcia Z, et al. Control of cyclin G2 mRNA expression by forkhead transcription factors: novel mechanism for cell cycle control by phosphoinositide 3-kinase and forkhead. Mol Cell Biol. 2004;24:2181–9.
    1. Goodrich DW, Lee WH. Abrogation by c-myc of G1 phase arrest induced by RB protein but not by p53. Nature. 1992;360:177–9.
    1. Wirt SE, Sage J. p107 in the public eye: an Rb understudy and more. Cell Div. 2010;5:9.

Source: PubMed

3
Abonnere