Skip to main content
Log in

Oxidative Stress, DNA, Cell Cycle/Cell Cycle Associated Proteins and Multidrug Resistance Proteins: Targets of Human Amniotic Membrane in Hepatocellular Carcinoma

  • Original Article
  • Published:
Pathology & Oncology Research

Abstract

The anticancer effects of human amniotic membrane (hAM) have been studied over the last decade. However, the action mechanisms responsible for these effects are not fully understood until now. Previously results reported by our team proved that hAM is able to induce cytotoxicity and cell death in hepatocellular carcinoma (HCC), a worldwide high incident and mortal cancer. Therefore, this experimental study aimed to investigate the cellular targets of hAM protein extracts (hAMPE) in HCC through in vitro studies. Our results showed that hAMPE is able to modify oxidative stress environment in all HCC cell lines, as well as its cell cycle. hAMPE differently targets deoxyribonucleic acid (DNA), P21, P53, β-catenin and multidrug resistance (MDR) proteins in HCC cell lines. In conclusion, hAMPE has several targets in HCC, being clear that the success of this treatment depends of a personalized therapy based on the biological and genetic characteristics of the tumor.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

References

  1. Toda A, Okabe M, Yoshida T, Nikaido T (2007) The potential of amniotic membrane/amnion-derived cells for regeneration of various tissues. J Pharmacol Sci 105:215–228

    Article  CAS  PubMed  Google Scholar 

  2. Davis J (1910) Skin transplantation with a review of 550 cases at the Johns Hopkins Hospital. Johns Hopkins Med J 15:307

    Google Scholar 

  3. Gomes J, Romano A, Santos M, Dua H (2005) Amniotic membrane use in ophthalmology. Curr Opin Ophthalmol 16:233–240

    Article  PubMed  Google Scholar 

  4. Dua H, Azuara-Blanco A (1999) Amniotic membrane transplantation. Br J Ophthalmol 83:748–752

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Sawhney C (1989) Amniotic membrane as a biological dressing in the management of burns. Burns 15:339–342

    Article  CAS  PubMed  Google Scholar 

  6. Mamede A, Carvalho M, Abrantes A, et al. (2012) Amniotic membrane: from structure and functions to clinical applications. Cell Tissue Res 349:447–458

    Article  CAS  PubMed  Google Scholar 

  7. Magatti M, De Munari S, Vertua E, Parolini O (2012) Amniotic membrane-derived cells inhibit proliferation of cancer cell lines by inducing cell cycle arrest. J Cell Mol Med 16:2208–2218

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Jiao H, Guan F, Yang B, et al. (2012) Human amniotic membrane derived-mesenchymal stem cells induce C6 glioma apoptosis in vivo through the bcl-2/caspase pathways. Mol Biol Rep 39:467–473

    Article  CAS  PubMed  Google Scholar 

  9. Rolfo A, Giuffrida D, Giuffrida M, et al. (2014) New perspectives for prostate cancer treatment: in vitro inhibition of LNCaP and PC3 cell proliferation by amnion-derived mesenchymal stromal cells conditioned media. Aging Male 17:94–101

    Article  PubMed  Google Scholar 

  10. Kang N-H, Yi B-R, Lim S, et al. (2012) Human amniotic membrane-derived epithelial stem cells display anticancer activity in BALB/c female nude mice bearing disseminated breast cancer xenografts. Int J Oncol 40:2022–2028

    CAS  PubMed  Google Scholar 

  11. Niknejad H, Khayat-Khoei M, Peirovi H, Abolghasemi H (2014) Human amniotic epithelial cells induce apoptosis of cancer cells: a new anti-tumor therapeutic strategy. Cytotherapy 16:33–40

    Article  CAS  PubMed  Google Scholar 

  12. Mamede A, Laranjo M, Carvalho M, et al. (2014) Effect of amniotic membrane proteins in human cancer cell lines: an exploratory study. J Membr Biol 247:357–360

    Article  CAS  PubMed  Google Scholar 

  13. Mamede A, Guerra S, Laranjo M, et al. (2015) Selective cytotoxicity and cell death induced by human amniotic membrane in hepatocellular carcinoma. Med Oncol 32:257

    Article  CAS  PubMed  Google Scholar 

  14. Gomes M, Priolli D, Tralhão J, Botelho M (2013) Hepatocellular carcinoma: epidemiology, biology, diagnosis and therapies. Rev Assoc Med Bras 59:514–524

    Article  PubMed  Google Scholar 

  15. Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, et al. (2013) Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer 49:1374–1403

    Article  CAS  PubMed  Google Scholar 

  16. Marra M, Sordelli I, Lombardi A, et al. (2011) Molecular targets and oxidative stress biomarkers in hepatocellular carcinoma: an overview. J Transl Med 9:171

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Alves R, Alves D, Guz B, et al. (2011) Advanced hepatocellular carcinoma: review of targeted molecular drugs. Ann Hepatol 10:21–27

    PubMed  Google Scholar 

  18. Brito A, Abrantes A, Pinto-Costa C, et al. (2013) Hepatocellular carcinoma and chemotherapy: the role of p53. Chemotherapy 58:381–386

    Article  Google Scholar 

  19. Tralhão J, Abrantes A, Hoti E, et al. (2013) Hepatectomy and liver regeneration: from experimental research to clinical application. ANZ J Surg 84:665–671

    Article  PubMed  Google Scholar 

  20. Abrantes A, Serra M, Gonçalves A, et al. (2010) Hypoxia-induced redox alterations and their correlation with 99mTc-MIBI and 99mTc-HL-91 uptake in colon cancer cells. Nucl Med Biol 37:125–132

    Article  CAS  PubMed  Google Scholar 

  21. Tralhão J, Abrantes A, Gonçalves A, et al. (2013) Study of hepatocellular function in the murine model following hepatic artery selective clamping. Acta Cir Bras 28:657–663

    Article  PubMed  Google Scholar 

  22. Gomes A, Fernandes E, Lima J (2005) Fluorescence probes used for detection of reactive oxygen species. J Biochem Biophys Methods 65:45–80

    Article  CAS  PubMed  Google Scholar 

  23. Mamede A, Pires A, Abrantes A, et al. (2012) Cytotoxicity of ascorbic acid in a human colorectal adenocarcinoma cell line (WiDr): in vitro and in vivo studies. Nutr Cancer 64:1049–1057

    Article  CAS  PubMed  Google Scholar 

  24. Larrauri A, López P, Gómez-Lechón M, Castell J (1987) A cytochemical stain for glutathione in rat hepatocytes cultured on plastic. J Histochem Cytochem 35:271–274

    Article  CAS  PubMed  Google Scholar 

  25. Ioannou Y, Chen F (1996) Quantitation of DNA fragmentation in apoptosis. Nucleic Acids Res 24:992–993

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Santos K, Laranjo M, Abrantes A, et al. (2014) Targeting triple-negative breast cancer cells with 6,7-bis(hydroxymethyl)-1H,3H-pyrrolo[1,2-c]thiazoles. Eur J Med Chem 79:273–281

    Article  CAS  PubMed  Google Scholar 

  27. Serra A, Am RG, Laranjo M, et al. (2012) Synthesis of new 2-galactosylthiazolidine-4-carboxylic acid amides. antitumor evaluation against melanoma and breast cancer cells. Eur J Med Chem 53:398–402

    Article  CAS  PubMed  Google Scholar 

  28. Brito A, Abrantes A, Ribeiro M, et al. (2015) Fluorine-18 fluorodeoxyglucose uptake in hepatocellular carcinoma: correlation with glucose transporters and p53 expression. J Clin Exp Hepatol 5:183–189

    Article  PubMed  PubMed Central  Google Scholar 

  29. Gomes A, Abrantes A, Brito A, et al. (2015) P53 influence on the radiotherapy response of hepatocellular carcinoma. Clin Mol Hepatol 21:257–267

    Article  PubMed  PubMed Central  Google Scholar 

  30. Brito A, Mendes M, Abrantes A, et al. (2014) Positron emission tomography diagnostic imaging in multidrug-resistant hepatocellular carcinoma: focus on 2-deoxy-2-(18F)fluoro-d-glucose. Mol Diagn Ther 18:495–504

    Article  CAS  PubMed  Google Scholar 

  31. Casalta-Lopes J, Abrantes A, Laranjo M, et al. (2011) Efflux pumps modulation in colorectal adenocarcinoma cell lines: the role of nuclear medicine. J Cancer Ther 02:408–417

    Article  CAS  Google Scholar 

  32. Choi K, Kim J, Kim G, Choi C (2009) Oxidative stress-induced necrotic cell death via mitochondira-dependent burst of reactive oxygen species. Curr Neurovasc Res 6:213–222

    Article  CAS  PubMed  Google Scholar 

  33. Valko M, Leibfritz D, Moncol J, et al. (2007) Free radicals and antioxidants in normal physiological functions and human disease. Int J Biochem Cell Biol 39:44–84

    Article  CAS  PubMed  Google Scholar 

  34. Li Y, Schellhorn H (2007) New developments and novel therapeutic perspectives for vitamin C. J Nutr 137:2171–2184

    CAS  PubMed  Google Scholar 

  35. Georgiou C, Papapostolou I, Grintzalis K (2009) Protocol for the quantitative assessment of DNA concentration and damage (fragmentation and nicks). Nat Protoc 4:125–131

    Article  CAS  PubMed  Google Scholar 

  36. Reuter S, Gupta S, Chaturvedi M, Aggarwal B (2010) Oxidative stress, inflammation, and cancer: how are they linked? Free Radic Biol Med 49:1603–1616

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Oberhammer F, Wilson J, Dive C, et al. (1993) Apoptotic death in epithelial cells: cleavage of DNA to 300 and/or 50 kb fragments prior to or in the absence of internucleosomal fragmentation. EMBO J 12:3679–3684

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Zamai L, Falcieri E, Marhefka G, Vitale M (1996) Supravital exposure to propidium iodide identifies apoptotic cells in the absence of nucleosomal DNA fragmentation. Cytometry 23:303–311

    Article  CAS  PubMed  Google Scholar 

  39. Macleod K, Sherry N, Hannon G, et al. (1995) p53-dependent and independent expression of p21 during cell growth, differentiation, and DNA damage. Genes Dev 9:935–944

    Article  CAS  PubMed  Google Scholar 

  40. Hussain S, Schwank J, Staib F, et al. (2007) TP53 mutations and hepatocellular carcinoma: insights into the etiology and pathogenesis of liver cancer. Oncogene 26:2166–2176

    Article  CAS  PubMed  Google Scholar 

  41. Cayrol C, Knibiehler M, Ducommun B (1998) p21 binding to PCNA causes G1 and G2 cell cycle arrest in p53-deficient cells. Oncogene 16:311–320

    Article  CAS  PubMed  Google Scholar 

  42. Punchihewa C, Inoue A, Hishiki A, et al. (2012) Identification of small molecule proliferating cell nuclear antigen (PCNA) inhibitor that disrupts interactions with PIP-box proteins and inhibits DNA replication. J Biol Chem 287:14289–14300

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Stark G, Taylor W (2004) Analyzing the G2/M checkpoint. In: Schonthal AH (ed) Checkp. Control. cancer Rev. Model Syst. Humana Press, Totowa, pp. 51–82

    Google Scholar 

  44. Wu G, Xu L, Lin N, Liu B (2013) UCN-01 induces S and G2/M cell cycle arrest through the p53/p21waf1 or CHK2/CDC25C pathways and can suppress invasion in human hepatoma cell lines. BMC Cancer 13:167

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Chan K-T, Lung M (2004) Mutant p53 expression enhances drug resistance in a hepatocellular carcinoma cell line. Cancer Chemother Pharmacol 53:519–526

    Article  CAS  PubMed  Google Scholar 

  46. Tsang W-P, Chau S, Fung K-P, et al. (2003) Modulation of multidrug resistance-associated protein 1 (MRP1) by p53 mutant in saos-2 cells. Cancer Chemother Pharmacol 51:161–166

    CAS  PubMed  Google Scholar 

Download references

Acknowledgments

Ana Catarina Mamede (SFRH/BD/73649/2010) and Maria João Carvalho (SFRH/SINTD/60068/2009), wishes to thank to Portuguese Foundation for Science and Technology for their PhD Grants. The authors would also like to thank Obstetrics Service of Coimbra Hospital and University Centre for the collection of human tissues used in this work. Support: Santander Totta; Infarmed (Health Research Fund 2015); FCT, Portugal (Strategic Project PEst-C/SAU/UI3282/2013 and UID/NEU/04539/2013), COMPETE-FEDER.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to A. C. Mamede.

Ethics declarations

All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

Informed Consent

Informed consent was obtained from all individual participants included in the study.

Additional information

A. C. Mamede and S. Guerra contributed equally to this work.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mamede, A.C., Guerra, S., Laranjo, M. et al. Oxidative Stress, DNA, Cell Cycle/Cell Cycle Associated Proteins and Multidrug Resistance Proteins: Targets of Human Amniotic Membrane in Hepatocellular Carcinoma. Pathol. Oncol. Res. 22, 689–697 (2016). https://doi.org/10.1007/s12253-016-0053-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12253-016-0053-x

Keywords

Navigation