Skip to main content

Advertisement

Log in

Myricetin loaded in solid lipid nanoparticles induces apoptosis in the HT-29 colorectal cancer cells via mitochondrial dysfunction

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

Abstract

Background

Among the flavonoids, Myricetin (MCN) has negligible side effects and anti-cancer properties. However, the therapeutic potential of MCN has been limited mainly by its low bioavailability. Nanocarriers improve the bioavailability and stability of flavonoids. The toxic effects of MCN loaded in solid lipid nanoparticles (MCN-SLNs) on the HT-29 human colorectal cancer cells were investigated in this study.

Methods and results

HT-29 cells were exposed to the 30 µmol MCN or MCN-SLNs for 24 h. Colony formation, cell viability, apoptosis, and expression of the Bax, Bcl-2, and AIF (apoptosis-inducing factor) have been investigated. Mitochondrial membrane potential (MMP) and reactive oxygen species (ROS) generation were also measured. The MCN-SLNs with appropriate characteristics and a slow sustained MCN release until 48 h made. MCN-SLNs could diminish colony numbers and survival of the HT-29 cells. The apoptosis index of MCN-SLNs-treated cells significantly increased compared to the free MCN (p < 0.001). The expression of Bax and AIF were elevated (p < 0.01 and p < 0.001, respectively) while Bcl-2 expression was decreased in MCN-SLNs treatment (p < 0.05). Moreover, MCN-SLNs significantly enhanced the ROS formation and reduced MMP compared to the free MCN-treated cells (p < 0.01).

Conclusions

The SLN formulation of MCN can effectively induce colon cancer cell death by raising ROS formation and activating the apoptosis process.

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

Data availability

The authors confirm that the data supporting the findings of this study are available in the supplemental file.

References

  1. Siegel RL, Miller KD, Fuchs HE, Jemal A (2022) Cancer statistics. CA Cancer J Clin 72:7–33. https://doi.org/10.3322/caac.21708

    Article  PubMed  Google Scholar 

  2. Patel SG, Karlitz JJ, Yen T, Lieu CH, Boland CR (2022) The rising tide of early-onset colorectal cancer: a comprehensive review of epidemiology, clinical features, biology, risk factors, prevention, and early detection. Lancet Gastroentrol Hepatol 7:262–274. https://doi.org/10.1016/S2468-1253(21)00426-X

    Article  Google Scholar 

  3. Rawla P, Sunkara T, Barsouk A (2019) Epidemiology of colorectal cancer: incidence, mortality, survival, and risk factors. Prz Gastroentero 14:89–103. https://doi.org/10.5114/pg.2018.81072

    Article  CAS  Google Scholar 

  4. Taheri Y, Suleria HAR, Martins N, Sytar O, Beyatli A, Yeskaliyeva B, Seitimova G, Salehi B, Semwal B, Painuli S, Anuj Kumar A, Azzini E, Martorell M, Setzer WN, Maroyi A, Sharifi-Rad J (2020) Myricetin bioactive effects: moving from preclinical evidence to potential clinical applications. BMC Complement Med Ther 20:241. https://doi.org/10.1186/s12906-020-03033-z

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Khorsandi L, Mansouri E, Rashno M, Karami MA, Ashtari A (2020) Myricetin loaded solid lipid nanoparticles upregulate MLKL and RIPK3 in human lung adenocarcinoma. Int J Pept Res Ther. https://doi.org/10.1007/s10989-019-09895-3

    Article  Google Scholar 

  6. Ye C, Zhang C, Huang H, Yang B, Xiao G, Kong D, Tian Q, Song Q, Song Y, Tan H, Wang Y, Zhou T, Zi X, Sun Y (2018) The natural compound myricetin effectively represses the malignant progression of prostate cancer by inhibiting PIM1 and disrupting the PIM1/CXCR4 interaction. Cell Physiol Biochem 48:1230–1244. https://doi.org/10.1159/000492009

    Article  CAS  PubMed  Google Scholar 

  7. Sajedi S, Homayoun M, Mohammadi F, Soleimani M (2020) Myricetin exerts its apoptotic effects on MCF-7 breast cancer cells through evoking the BRCA1-GADD45 pathway. Asian Pac J Cancer Prev 21:3461–3468. https://doi.org/10.31557/APJCP.2020.21.12.3461

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Zhu ML, Zhang PM, Jiang M, Yu SW, Wang L (2020) Myricetin induces apoptosis and autophagy by inhibiting PI3K/Akt/mTOR signalling in human colon cancer cells. BMC Complement Med Ther 20:209. https://doi.org/10.1186/s12906-020-02965-w

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Knickle A, Fernando W, Greenshields AL, Rupasinghe HPV, Hoskin DW (2018) Myricetin-induced apoptosis of triple-negative breast cancer cells is mediated by the iron-dependent generation of reactive oxygen species from hydrogen peroxide. Food Chem Toxicol 118:154–167. https://doi.org/10.1016/j.fct.2018.05.005

    Article  CAS  PubMed  Google Scholar 

  10. Lee JH, Choi YL, Park SH, Nam MJ (2018) Potential role of nucleoside diphosphate kinase in myricetin-induced selective apoptosis in colon cancer HCT-15 cells. Food Chem Toxicol 116:315–322. https://doi.org/10.1016/j.fct.2018.04.053

    Article  CAS  PubMed  Google Scholar 

  11. Kim ME, Ha TK, Yoon JH, Lee JS (2014) Myricetin induces cell death of human colon cancer cells via BAX/BCL2-dependent pathway. Anticancer Res 34:701–706

    CAS  PubMed  Google Scholar 

  12. Musika J, Chudapongse N (2020) Development of lipid-based nanocarriers for increasing gastrointestinal absorption of lupinifolin. Planta Med 86:364–372. https://doi.org/10.1055/a-1095-1129

    Article  CAS  PubMed  Google Scholar 

  13. Montoto SS, Muraca G, Ruiz ME (2020) Solid lipid nanoparticles for drug delivery: pharmacological and biopharmaceutical aspects. Front Mol Biosci 7:587997. https://doi.org/10.3389/fmolb.2020.587997

    Article  CAS  Google Scholar 

  14. Bayón-Cordero L, Alkorta I, Arana L (2019) Application of solid lipid nanoparticles to improve the efficiency of anticancer drugs. Nanomaterials 9:474. https://doi.org/10.3390/nano9030474

    Article  CAS  PubMed Central  Google Scholar 

  15. Ban C, Park JB, Cho S, Kim HR, Kim YJ, Choi YJ, Chung WJ, Kweon DH (2020) Reduction of focal sweating by lipid nanoparticle-delivered myricetin. Sci Rep 10:13132. https://doi.org/10.1038/s41598-020-69985-x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Gaber DM, Nafee N, Abdallah OY (2017) Myricetin solid lipid nanoparticles: Stability assurance from system preparation to site of action. Eur J Pharm Sci 109:569–580. https://doi.org/10.1016/j.ejps.2017.08

    Article  CAS  PubMed  Google Scholar 

  17. Ha K, Jung I, Kim ME, Bae SK, Lee JS (2017) Anti-cancer activity of myricetin against human papillary thyroid cancer cells involves mitochondrial dysfunction-mediated apoptosis. Biomed Pharmacother 91:378–384. https://doi.org/10.1016/j.biopha.2017.04.100

    Article  CAS  PubMed  Google Scholar 

  18. Osman AMM, Al-Johani HS, Kamel FO, Ahmed OA, Huwait EA, Sayed-Ahmed MA (2020) 5-Fluorouracil and simvastatin loaded solid lipid nanoparticles for effective treatment of colorectal cancer cells. Int J Pharmacol 16:205–213. https://doi.org/10.3923/ijp.2020.205.21

    Article  CAS  Google Scholar 

  19. Chandran SP, Nachinmuthu KP, Natarajan SB, Inamdar MG, Shahimi MSBM (2018) Papain loaded solid lipid nanoparticles for colorectal cancer therapy. Curr Cancer Ther 14:75–87. https://doi.org/10.2174/1573394713666170929160933

    Article  CAS  Google Scholar 

  20. Wang G, Wang JJ, Tang XJ, Du L, Li F (2016) In vitro and in vivo evaluation of functionalized chitosan–pluronic micelles loaded with myricetin on glioblastoma cancer. Nanotechnol Biol Med 12:1263–1278. https://doi.org/10.1016/j.nano.2016.02.004

    Article  CAS  Google Scholar 

  21. Serini S, Cassano R, Corsetto P, Rizzo A, Calviello G, Trombino S (2018) Omega-3 PUFA loaded in resveratrol-based solid lipid nanoparticles: physicochemical properties and antineoplastic activities in human colorectal cancer cells in vitro. Int J Mol Sci 19:586. https://doi.org/10.3390/ijms19020586

    Article  CAS  PubMed Central  Google Scholar 

  22. Watkins R, Wu L, Zhang C, Davis RM, Xu B (2015) Natural product-based nanomedicine: recent advances and issues. Int J Nanomed 10:6055–6074. https://doi.org/10.2147/IJN.S92162

    Article  CAS  Google Scholar 

  23. Pistritto G, Trisciuoglio D, Ceci C, Garufi A, D’Orazi G (2016) Apoptosis as anticancer mechanism: function and dysfunction of its modulators and targeted therapeutic strategies. Aging (Albany NY) 8:603–619

    Article  CAS  Google Scholar 

  24. Li HG, Chen JX, Xiong JH, Zhu JW (2016) Myricetin exhibits anti-glioma potential by inducing mitochondrial-mediated apoptosis, cell cycle arrest, inhibition of cell migration and ROS generation. J BUON 21(2016):182–190

    PubMed  Google Scholar 

  25. Zhang XH, Chen SY, Tang L, Shen YZ, Luo L, Xu CW, Liu Q, Li D (2013) Myricetin induces apoptosis in HepG2 cells through Akt/p70S6K/Bad signaling and mitochondrial apoptotic pathway. Anticancer Agents Med Chem 13:1575–1581. https://doi.org/10.2174/1871520613666131125123059

    Article  CAS  PubMed  Google Scholar 

  26. Arfin S, Jha NK, Jha SK, Kesari KK, Ruokolainen J, Roychoudhury S, Rathi B, Kumar D (2021) Oxidative stress in cancer cell metabolism. Antioxidants (Basel) 10:642. https://doi.org/10.3390/antiox10050642

    Article  CAS  Google Scholar 

  27. Park H, Park S, Bazer FW, Lim W, Song G (2018) Myricetin treatment induces apoptosis in canine osteosarcoma cells by inducing DNA fragmentation, disrupting redoxhomeostasis, and mediating loss of mitochondrial membrane potential. J Cell Physiol 233:7457–7466. https://doi.org/10.1002/jcp.26598

    Article  CAS  PubMed  Google Scholar 

  28. Yang F, Chen WD, Deng R, Zhang H, Tang J, Wu KW, Li DD, Feng GK, Lan WJ, Li HJ, Zhu XF (2013) Hirsutanol A, a novel sesquiterpene compound from fungus Chondrostereum sp., induces apoptosis and inhibits tumor growth through mitochondrial-independent ROS production: hirsutanol A inhibits tumor growth through ROS production. J Transl Med 11:1479–5876. https://doi.org/10.1186/1479-5876-11-32

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work has received a Grant from the Ahvaz Jundishapur University of Medical Sciences of Iran. We thank our colleagues from the Cellular and Molecular Research Center.

Funding

This work has received a Grant from the Ahvaz Jundishapur University of Medical Sciences of Iran (No: CMRC-9640).

Author information

Authors and Affiliations

Authors

Contributions

LK: supervised the study, statistical data analysis, and critical reviewing of the manuscript. HA: performed sampling, collection of data, experiments, and data analysis. AA: designing the study, searching the literature, and manuscript preparation. AS: performed sampling. MAK: designing the study and manuscript editing.

Corresponding author

Correspondence to Layasadat Khorsandi.

Ethics declarations

Conflict of interest

The authors declared no conflict of interest.

Ethical approval

This study was performed based on ethical principles approved by the Ethics Committee of Ahvaz Jundishapur University of Medical Sciences, Iran (No: IR.AJUMS.REC.1396.1120).

Consent to participate

In this study, we used human cell lines. Thus, there is not any consent to participate in this research study.

Consent for publication

In this study, we used human cell lines. Thus, there is not any consent for publication.

Additional information

Publisher's Note

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

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (DOCX 17 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Alidadi, H., Ashtari, A., Samimi, A. et al. Myricetin loaded in solid lipid nanoparticles induces apoptosis in the HT-29 colorectal cancer cells via mitochondrial dysfunction. Mol Biol Rep 49, 8537–8545 (2022). https://doi.org/10.1007/s11033-022-07683-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11033-022-07683-9

Keywords

Navigation