Skip to main content

Advertisement

Log in

Differential response of hepatocellular carcinoma glycolytic metabolism and oxidative stress markers after exposure to human amniotic membrane proteins

  • Original Article
  • Published:
Molecular Biology Reports Aims and scope Submit manuscript

Abstract

Background

The human Amniotic Membrane (hAM) has been studied as a potential therapeutic option in cancer, namely in hepatocellular carcinoma. Previously, our research group evaluated the effect of human Amniotic Membrane Protein Extracts (hAMPE) in cancer therapy, demonstrating that hAMPE inhibit the metabolic activity of human hepatocellular carcinoma cell lines: Hep3B2.1-7, HepG2 and Huh7. Therefore, and considering the close relationship between metabolic activity and oxidative stress, the aim of this study was to evaluate the effect of hAMPE treatment in glucose metabolism and its role in oxidative stress of hepatocellular carcinoma.

Methods and Results

Glucose uptake and lactate production was assessed by 1 H-NMR, and the expression of several mediators of the glycolytic pathway was evaluated by Western blot or fluorescence. Total antioxidant capacity (TAC) and biomarkers of oxidative stress effects in proteins were detected. Our results showed that hAMPE treatment increased glucose consumption on Hep3B2.1-7, HepG2, and Huh7 through the increase of GLUT1 in Hep3B2.1-7 and Huh7, and GLUT3 in HepG2 cells. It was observed an increased expression of 6-phosphofrutokinase (PFK-1L) in all cell lines though glucose was not converted to lactate on HepG2 and Huh7 cells, suggesting that hAMPE treatment may counteract the Warburg effect observed in carcinogenesis. In Hep3B2.1-7, hAMPE treatment induced an increase in expression of lactate dehydrogenase (LDH) and monocarboxylate transporter isoform 4 (MCT4). We further detected that hAMPE enhances the TAC of culture media after 2 and 8 h. This was followed by a degree of protection against proteins nitration and carbonylation.

Conclusions

Overall, this work highlights the potential usefulness of hAMPE as anticancer therapy through the modulation of the glycolytic and oxidative profile in human hepatocellular carcinoma.

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

Similar content being viewed by others

References

  1. McGlynn KA, Petrick JL, El-Serag HB (Jan. 2021) Epidemiology of Hepatocellular Carcinoma. Hepatology 73(4). no. Suppl 1doi: https://doi.org/10.1002/HEP.31288

  2. Petrick JL et al (2020) “International trends in hepatocellular carcinoma incidence, 1978–2012,” Int. J. cancer, vol. 147, no. 2, pp. 317–330, doi: https://doi.org/10.1002/IJC.32723

  3. McGlynn KA, Petrick JL, London WT (May 2015) Global epidemiology of hepatocellular carcinoma: an emphasis on demographic and regional variability. ” Clin Liver Dis 19(2):223–238. doi: https://doi.org/10.1016/j.cld.2015.01.001

  4. Alqahtani A, Khan Z, Alloghbi A, Ahmed TSS, Ashraf M, Hammouda DM (Sep. 2019) Hepatocellular Carcinoma: Molecular Mechanisms and Targeted Therapies. Med (Kaunas) 55(9). doi: https://doi.org/10.3390/MEDICINA55090526

  5. Burkhart RA, Ronnekleiv-Kelly SM, Pawlik TM (Jun. 2017) Personalized therapy in hepatocellular carcinoma: Molecular markers of prognosis and therapeutic response. Surg Oncol 26(2):138–145. doi: https://doi.org/10.1016/J.SURONC.2017.01.009

  6. WARBURG O (3191) “On the origin of cancer cells.,” Science, vol. 123, no. pp. 309–14, Feb. 1956, Accessed: Apr. 11, 2019. [Online]. Available: http://www.ncbi.nlm.nih.gov/pubmed/13298683

  7. De Matteis S et al (2018) “Aberrant Metabolism in Hepatocellular Carcinoma Provides Diagnostic and Therapeutic Opportunities,” Oxid. Med. Cell. Longev., vol. pp. 1–13, Nov. 2018, doi: https://doi.org/10.1155/2018/7512159

  8. Du D et al (2022) “Metabolic dysregulation and emerging therapeutical targets for hepatocellular carcinoma,” Acta Pharm. Sin. B, vol. 12, no. 2, pp. 558–580, doi: https://doi.org/10.1016/J.APSB.2021.09.019

  9. Kato Y, Maeda T, Suzuki A, Baba Y (Feb. 2018) Cancer metabolism: New insights into classic characteristics. Jpn Dent Sci Rev 54(1):8–21. doi: https://doi.org/10.1016/J.JDSR.2017.08.003

  10. Shokoohian B et al (2021) “Advanced therapeutic modalities in hepatocellular carcinoma: Novel insights,” J. Cell. Mol. Med., vol. 25, no. 18, pp. 8602–8614, doi: https://doi.org/10.1111/JCMM.16875

  11. Guan MC et al (2021) Early diagnosis and therapeutic strategies for hepatocellular carcinoma: From bench to bedside. World J Gastrointest Oncol 13(4):197–215. doi: https://doi.org/10.4251/WJGO.V13.I4.197

    Article  PubMed  PubMed Central  Google Scholar 

  12. Seo JH, Kim YH, Kim JS (Jan. 2008) Properties of the amniotic membrane may be applicable in cancer therapy. Med Hypotheses 70(4):812–814. doi: https://doi.org/10.1016/j.mehy.2007.08.008

  13. Mamede AC, Carvalho MJ, Abrantes AM, Laranjo M, Maia CJ, Botelho MF (2012) “Amniotic membrane: from structure and functions to clinical applications,” Cell Tissue Res., vol. 349, no. 2, pp. 447–458, Aug. doi: https://doi.org/10.1007/s00441-012-1424-6

  14. Mamede AC et al (Apr. 2014) Effect of amniotic membrane proteins in human cancer cell lines: an exploratory study. ” J Membr Biol 247(4):357–360. doi: https://doi.org/10.1007/s00232-014-9642-3

  15. Mamede AC et al (Dec. 2015) Selective cytotoxicity and cell death induced by human amniotic membrane in hepatocellular carcinoma. ” Med Oncol 32(12):257. doi: https://doi.org/10.1007/s12032-015-0702-z

  16. Brito AF et al (2012) Hepatocellular Carcinoma and Chemotherapy: The Role of p53. Chemotherapy 58(5):381–386. doi: https://doi.org/10.1159/000343656

    Article  CAS  PubMed  Google Scholar 

  17. Liang Y, Liu J, Feng Z (Feb. 2013) The regulation of cellular metabolism by tumor suppressor p53. Cell Biosci 3(1). doi: https://doi.org/10.1186/2045-3701-3-9

  18. Shao C et al (2004) “Suppression of corneal neovascularization by PEDF release from human amniotic membranes.,” Invest. Ophthalmol. Vis. Sci., vol. 45, no. 6, pp. 1758–62, Accessed: Apr. 11, 2019. [Online]. Available: http://www.ncbi.nlm.nih.gov/pubmed/15161837

  19. Rato L, Alves MG, Socorro S, Carvalho RA, Cavaco JE, Oliveira PF (Feb. 2012) Metabolic modulation induced by oestradiol and DHT in immature rat Sertoli cells cultured in vitro. Biosci Rep 32(1):61–69. doi: https://doi.org/10.1042/BSR20110030

  20. Benzie IFF, Strain JJ (1996) “The ferric reducing ability of plasma (FRAP) as a measure of ‘antioxidant power’: the FRAP assay,” Anal. Biochem., vol. 239, no. 1, pp. 70–76, Jul. doi: https://doi.org/10.1006/ABIO.1996.0292

  21. Gomes MA, Priolli DG, Tralhão JG, Botelho MF (2013) “Carcinoma hepatocelular: epidemiologia, biologia, diagnóstico e terapias,” Rev. Assoc. Med. Bras., vol. 59, no. 5, pp. 514–524, Sep. doi: https://doi.org/10.1016/j.ramb.2013.03.005

  22. Kang N-H et al (2012) “Potential antitumor therapeutic strategies of human amniotic membrane and amniotic fluid-derived stem cells,” Cancer Gene Ther., vol. 19, no. 8, pp. 517–522, doi: https://doi.org/10.1038/cgt.2012.30

  23. Magatti M, Munari S, Vertua E, Parolini O (Sep. 2012) Amniotic membrane-derived cells inhibit proliferation of cancer cell lines by inducing cell cycle arrest. J Cell Mol Med 16(9):2208–2218. doi: https://doi.org/10.1111/j.1582-4934.2012.01531.x

  24. Riedel R et al (Dec. 2019) Human amniotic membrane conditioned medium inhibits proliferation and modulates related microRNAs expression in hepatocarcinoma cells. Sci Rep 9(1). doi: https://doi.org/10.1038/S41598-019-50648-5

  25. Liu QW et al (2020) “Human amniotic mesenchymal stem cells inhibit hepatocellular carcinoma in tumour-bearing mice,” J. Cell. Mol. Med., vol. 24, no. 18, pp. 10525–10541, doi: https://doi.org/10.1111/JCMM.15668

  26. Mamede AC et al (Oct. 2016) “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(4):689–697. doi: https://doi.org/10.1007/s12253-016-0053-x

  27. Brahimi-Horn MC, Chiche J, Pouysségur J (2007) “Hypoxia and cancer,” J. Mol. Med., vol. 85, no. 12, pp. 1301–1307, Dec. doi: https://doi.org/10.1007/s00109-007-0281-3

  28. Alves AP et al (2019) “Glycolysis Inhibition as a Strategy for Hepatocellular Carcinoma Treatment?,” Curr. Cancer Drug Targets, vol. 19, no. 1, pp. 26–40, doi: https://doi.org/10.2174/1568009618666180430144441

  29. Gatenby RA, Gillies RJ (2004) “Why do cancers have high aerobic glycolysis?,” Nat. Rev. Cancer, vol. 4, no. 11, pp. 891–899, Nov. doi: https://doi.org/10.1038/nrc1478

  30. Zhao M, Zhang Z (2016) Glucose Transporter Regulation in Cancer: A Profile and the Loops. Crit Rev Eukaryot Gene Expr 26(3):223–238. doi: https://doi.org/10.1615/CritRevEukaryotGeneExpr.2016016531

    Article  PubMed  Google Scholar 

  31. Muñoz-Pinedo C, Ruiz-Ruiz C, Ruiz de Almodóvar C, Palacios C, López-Rivas A (2003) “Inhibition of Glucose Metabolism Sensitizes Tumor Cells to Death Receptor-triggered Apoptosis through Enhancement of Death-inducing Signaling Complex Formation and Apical Procaspase-8 Processing,” J. Biol. Chem., vol. 278, no. 15, pp. 12759–12768, Apr. doi: https://doi.org/10.1074/jbc.M212392200

  32. Brito AF et al (Sep. 2015) Fluorine-18 Fluorodeoxyglucose Uptake in Hepatocellular Carcinoma: Correlation with Glucose Transporters and p53 Expression. J Clin Exp Hepatol 5(3):183–189. doi: https://doi.org/10.1016/j.jceh.2015.05.003

  33. Amann T, Hellerbrand C (2009) “GLUT1 as a therapeutic target in hepatocellular carcinoma,” Expert Opin. Ther. Targets, vol. 13, no. 12, pp. 1411–1427, Dec. doi: https://doi.org/10.1517/14728220903307509

  34. Gao H et al (2020) Prognostic value of glucose transporter 3 expression in hepatocellular carcinoma. Oncol Lett 19(1):691–699. doi: https://doi.org/10.3892/OL.2019.11191

    Article  CAS  PubMed  Google Scholar 

  35. Moreno-Sánchez R et al (May 2012) Phosphofructokinase type 1 kinetics, isoform expression, and gene polymorphisms in cancer cells. ” J Cell Biochem 113(5):1692–1703. doi: https://doi.org/10.1002/jcb.24039

  36. Faloppi L et al (2016) “Lactate Dehydrogenase in Hepatocellular Carcinoma: Something Old, Something New,” Biomed Res. Int., vol. pp. 1–7, 2016, doi: https://doi.org/10.1155/2016/7196280

  37. Alves VA, Pinheiro C, Morais-Santos F, Felipe-Silva A, Longatto-Filho A, Baltazar F (Sep. 2014) Characterization of monocarboxylate transporter activity in hepatocellular carcinoma. World J Gastroenterol 20(33):11780. doi: https://doi.org/10.3748/wjg.v20.i33.11780

  38. Gao H-J et al (2015) “Monocarboxylate transporter 4 predicts poor prognosis in hepatocellular carcinoma and is associated with cell proliferation and migration,” J. Cancer Res. Clin. Oncol., vol. 141, no. 7, pp. 1151–1162, doi: https://doi.org/10.1007/s00432-014-1888-8

  39. Dalouchi F, Falak R, Bakhshesh M, Sharifiaghdam Z, Azizi Y, Aboutaleb N (2021) Human amniotic membrane mesenchymal stem cell-conditioned medium reduces inflammatory factors and fibrosis in ovalbumin-induced asthma in mice. Exp Physiol 106(2):544–554. doi: https://doi.org/10.1113/EP088911

    Article  CAS  PubMed  Google Scholar 

  40. Mirapoglu SL et al (Jul. 2021) Effects of Platelet Rich Plasma and Amniotic Cell Culture Medium on Wound Healing Following Experimental Animal Tracheal Injury Model: A Comparative Study. J Craniofac Surg 32(5):1937–1941. doi: https://doi.org/10.1097/SCS.0000000000007396

  41. Faridvand Y et al (Aug. 2020) Human Amnion Membrane Proteins Prevent Doxorubicin-Induced Oxidative Stress Injury and Apoptosis in Rat H9c2 Cardiomyocytes. Cardiovasc Toxicol 20(4):370–379. doi: https://doi.org/10.1007/S12012-020-09564-8

  42. Wu M et al (2022) “Human Amniotic Membrane Promotes Angiogenesis in an Oxidative Stress Chronic Diabetic Murine Wound Model,” Adv. wound care, doi: https://doi.org/10.1089/WOUND.2022.0005

  43. Goonetilleke M et al (Dec. 2021) Addressing the liver progenitor cell response and hepatic oxidative stress in experimental non-alcoholic fatty liver disease/non-alcoholic steatohepatitis using amniotic epithelial cells. Stem Cell Res Ther 12(1). doi: https://doi.org/10.1186/S13287-021-02476-6

  44. Shen Y-C et al (Jan. 2013) Activating oxidative phosphorylation by a pyruvate dehydrogenase kinase inhibitor overcomes sorafenib resistance of hepatocellular carcinoma. ” Br J Cancer 108(1):72–81. doi: https://doi.org/10.1038/bjc.2012.559

  45. Bonnet S et al (Jan. 2007) “A mitochondria-K + channel axis is suppressed in cancer and its normalization promotes apoptosis and inhibits cancer growth. ” Cancer Cell 11(1):37–51. doi: https://doi.org/10.1016/j.ccr.2006.10.020

Download references

Acknowledgements

Sandra Moreira Rocha (SFRH/BD/115693/2016) and Ana Catarina Mamede (SFRH/BD/73649/2010) wishes to thank to FCT 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.

Funding

This work was support by the Infarmed (Health Research found 2015) and National Funds by FCT-Foundation for Science and Technology (Project UID/Multi/00709/2019).

Author information

Authors and Affiliations

Authors

Contributions

All authors participated in the conception and design of study. APA, SMR, ACM and CJM interpret and analyzed of the data; APA, SMR, ACM, PCB and MGA conducted the experiments; ACM, MGA, PFO, MFB and CJM critically read the article; and APA and SMR wrote the manuscript. APA and SMR contributed equally to this study.

Corresponding author

Correspondence to Cláudio J Maia.

Ethics declarations

Declarations OF conflicting interests

The authors declare that they have no conflicts of interest with the contents of this article.

Ethical approval

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.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Andreia P Alves and Sandra M Rocha 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

Alves, A.P., Rocha, S.M., Mamede, A.C. et al. Differential response of hepatocellular carcinoma glycolytic metabolism and oxidative stress markers after exposure to human amniotic membrane proteins. Mol Biol Rep 49, 7731–7741 (2022). https://doi.org/10.1007/s11033-022-07598-5

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11033-022-07598-5

Keywords

Navigation