Skip to main content

Advertisement

Log in

Cordycepin enhances hyperthermia-induced apoptosis and cell cycle arrest by modulating the MAPK pathway in human lymphoma U937 cells

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

Abstract

Background

Hyperthermia induces cancer cell death. However, the cytotoxic effect of hyperthermia is not sufficient. Cordycepin can also induce apoptosis in cancer cells and enhance the antitumoral activity of irradiation. To examine cordycepin-mediated enhancement of hyperthermia-induced apoptosis, this study investigated the combined effects and apoptotic mechanisms of hyperthermia and cordycepin on human leukemia U937 cells.

Methods

Cell viability and apoptosis were measured using MTT assays, Hoechst 33342 staining and Annexin V/PI double staining. The distribution of the cell cycle and sub-G1 phase, reactive oxygen species (ROS) and mitochondrial membrane potential (MMP) were examined by flow cytometry. The expression of related proteins was analyzed by western blotting.

Results

Combined treatment with hyperthermia and cordycepin markedly augmented apoptosis by upregulating Bax and suppressing Bcl-2, Bid and activated caspase 3 and 8 expression, and apoptosis was decreased by Z-VAD-fmk (a pan caspase inhibitor). We also found that the MMP was significantly decreased and excessive ROS generation occurred. The combination treatment also induced arrest in the G2/M phase by downregulating cyclin dependent kinase 1 (CDK1) and cyclin B1 protein expression. Furthermore, it was observed that mitogen-activated protein kinase (MAPK) pathway including ERK, JNK and p38 signals was involved in the induction of apoptosis. The phosphorylated p38 and JNK were increased and ERK phosphorylation was decreased by the combined treatment. In addition, N-acetyl-l-cysteine (NAC) significantly protected the cells by restoring ROS levels and the activity of caspase-3, inactivating the MAPK pathway.

Conclusion

Cordycepin significantly enhanced hyperthermia-induced apoptosis and G2/M phase arrest in U937 cells. The combined treatment enhanced apoptosis through the MAPK pathway and mitochondrial dysfunction, and these effects could be rescued by NAC. We report for the first time that cordycepin can be used as a hyperthermia sensitizer to treat leukemia.

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. Jakobsen N, Vyas P (2018) From genomics to targeted treatment in haematological malignancies: a focus on acute myeloid leukaemia. Clin Med (Lond) 18:s47–s53

    Google Scholar 

  2. Kampen K (2012) The discovery and early understanding of leukemia. Leuk Res 36:6–13

    PubMed  Google Scholar 

  3. Shlush L, Mitchell A (2015) AML evolution from preleukemia to leukemia and relapse. Best Pract Res Clin Haematol 28:81–89

    PubMed  Google Scholar 

  4. Saliev T, Feril L, Begimbetova D et al (2017) Hyperthermia enhances bortezomib-induced apoptosis in human white blood cancer cells. J Therm Biol 67:9–14

    CAS  PubMed  Google Scholar 

  5. Zakki S, Cui Z, Sun L, Feng Q, Li M, Inadera H (2018) Baicalin augments hyperthermia-induced apoptosis in U937 cells and modulates the mapk pathway via ros generation. Cell Physiol Biochem 45:2444–2460

    CAS  PubMed  Google Scholar 

  6. Ahmed K, Tabuchi Y, Kondo T (2015) Hyperthermia: an effective strategy to induce apoptosis in cancer cells. Apoptosis 20:1411–1419

    CAS  PubMed  Google Scholar 

  7. Hwang J, Park S, Ko W et al (2017) Cordycepin induces human lung cancer cell apoptosis by inhibiting nitric oxide mediated ERK/Slug signaling pathway. Am J Cancer Res 7:417–432

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Xu J, Zhou X, Wang X et al (2019) Cordycepin induces apoptosis and G2/M phase arrest through the ERK pathways in esophageal cancer cells. J Cancer 10:2415–2424

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Li S, Ren J, Fei J, Zhang X, Du R (2019) Cordycepin induces Bax-dependent apoptosis in colorectal cancer cells. Mol Med Rep 19:901–908

    CAS  PubMed  Google Scholar 

  10. Lin Y, Liang S, Wu Y et al (2019) Cordycepin suppresses endothelial cell proliferation, migration, angiogenesis, and tumor growth by regulating focal adhesion kinase and p53. Cancers. https://doi.org/10.3390/cancers11020168

    Article  PubMed  PubMed Central  Google Scholar 

  11. Jiang Q, Lou Z, Wang H, Chen C (2019) Antimicrobial effect and proposed action mechanism of cordycepin against Escherichia coli and Bacillus subtilis. J Microbiol 57:288–297

    CAS  PubMed  Google Scholar 

  12. Han N, Moon P, Kim H, Jeong H (2018) Cordycepin ameliorates skin inflammation in a DNFB-challenged murine model of atopic dermatitis. Immunopharmacol Immunotoxicol 40:401–407

    CAS  PubMed  Google Scholar 

  13. Ashraf S, Radhi M, Gowler P et al (2019) The polyadenylation inhibitor cordycepin reduces pain, inflammation and joint pathology in rodent models of osteoarthritis. Sci Rep 9:4696

    PubMed  PubMed Central  Google Scholar 

  14. Wang J, Liu R, Liu B, Yang Y, Xie J, Zhu N (2017) Systems Pharmacology-based strategy to screen new adjuvant for hepatitis B vaccine from traditional Chinese medicine Ophiocordyceps sinensis. Sci Rep 7:44788

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Nasser MI, Masood M, Wei W et al (2017) Cordycepin induces apoptosis in SGC7901 cells through mitochondrial extrinsic phosphorylation of PI3K/Akt by generating ROS. Int J Oncol 50:911–919

    CAS  PubMed  Google Scholar 

  16. Dong J, Li Y, Xiao H et al (2019) Cordycepin sensitizes breast cancer cells toward irradiation through elevating ROS production involving Nrf2. Toxicol Appl Pharmacol 364:12–21

    CAS  PubMed  Google Scholar 

  17. Perillo B, Di Donato M, Pezone A et al (2020) ROS in cancer therapy: the bright side of the moon. Exp Mol Med 52:192–203

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Green D, Kroemer G (2004) The pathophysiology of mitochondrial cell death. Science (New York, NY) 305:626–629

    CAS  Google Scholar 

  19. Westphal D, Kluck R, Dewson G (2014) Building blocks of the apoptotic pore: how Bax and Bak are activated and oligomerize during apoptosis. Cell Death Differ 21:196–205

    CAS  PubMed  Google Scholar 

  20. Hsia C, Lin K, Lee T et al (2019) Esculetin, a Coumarin derivative, prevents thrombosis: inhibitory signaling on PLCγ2-PKC-AKT activation in human platelets. Int J Mol Sci. https://doi.org/10.3390/ijms20112731

    Article  PubMed  PubMed Central  Google Scholar 

  21. Cui ZG, Piao JL, Rehman MU et al (2014) Molecular mechanisms of hyperthermia-induced apoptosis enhanced by withaferin A. Eur J Pharmacol 723:99–107

    CAS  PubMed  Google Scholar 

  22. Tsirigotis P, Byrne M, Schmid C et al (2016) Relapse of AML after hematopoietic stem cell transplantation: methods of monitoring and preventive strategies. A review from the ALWP of the EBMT. Bone Marrow Transplant 51:1431–1438

    CAS  PubMed  Google Scholar 

  23. Huang A, Cheng H, Lin T et al (2013) Epigallocatechin gallate (EGCG), influences a murine WEHI-3 leukemia model in vivo through enhancing phagocytosis of macrophages and populations of T- and B-cells. In Vivo 27:627–634

    CAS  PubMed  Google Scholar 

  24. Chen P, Wang B, Pan B, Guo W (2016) Resveratrol-4-O-D-(2′-galloyl)-glucopyranoside exerts an anticancer effect on leukemia cells via inducing apoptosis. Mol Med Rep 13:2281–2286

    CAS  PubMed  Google Scholar 

  25. Moreno C, Greil R, Demirkan F et al (2019) Ibrutinib plus obinutuzumab versus chlorambucil plus obinutuzumab in first-line treatment of chronic lymphocytic leukaemia (iLLUMINATE): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol 20:43–56

    CAS  PubMed  Google Scholar 

  26. Melo J, Chuah C (2008) (2008) Novel agents in CML therapy: tyrosine kinase inhibitors and beyond. Hematol Am Soc Hematol Educ Progr. 1:427–435

    Google Scholar 

  27. Datta N, Puric E, Klingbiel D, Gomez S, Bodis S (2016) Hyperthermia and radiation therapy in locoregional recurrent breast cancers: a systematic review and meta-analysis. Int J Radiat Oncol Biol Phys 94:1073–1087

    PubMed  Google Scholar 

  28. Notter M, Thomsen A, Nitsche M et al (2020) Combined wIRA-hyperthermia and hypofractionated re-irradiation in the treatment of locally recurrent breast cancer: evaluation of therapeutic outcome based on a novel size classification. Cancers. https://doi.org/10.3390/cancers12030606

    Article  PubMed  PubMed Central  Google Scholar 

  29. Quintana C, Cabrera J, Perdomo J et al (2016) Melatonin enhances hyperthermia-induced apoptotic cell death in human leukemia cells. J Pineal Res 61:381–395

    CAS  PubMed  Google Scholar 

  30. Luo Z, Zheng K, Fan Q, Jiang X, Xiong D (2017) Hyperthermia exposure induces apoptosis and inhibits proliferation in HCT116 cells by upregulating miR-34a and causing transcriptional activation of p53. Exp Ther Med 14:5379–5386

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Quintana M, Saavedra E, Del Rosario H et al (2021) Ethanol enhances hyperthermia-induced cell death in human leukemia cells. Int J Mol Sci. https://doi.org/10.3390/ijms22094948

    Article  PubMed  PubMed Central  Google Scholar 

  32. Ruan Q, Ding D, Wang B et al (2021) A multi-institutional retrospective study of hyperthermic plus intravesical chemotherapy versus intravesical chemotherapy treatment alone in intermediate and high risk nonmuscle-invasive bladder cancer. Cancer Biol Med 18:308–317

    CAS  PubMed  PubMed Central  Google Scholar 

  33. González-Padilla D, González-Díaz A, Guerrero-Ramos F et al (2021) Quality of life and adverse events in patients with nonmuscle invasive bladder cancer receiving adjuvant treatment with BCG, MMC, or chemohyperthermia. Urol Oncol 39:76.e79-76.e14

    Google Scholar 

  34. Chou S, Lai W, Hong T et al (2014) Synergistic property of cordycepin in cultivated Cordyceps militaris-mediated apoptosis in human leukemia cells. Phytomedicine 21:1516–1524

    CAS  PubMed  Google Scholar 

  35. Thomadaki H, Tsiapalis C, Scorilas A (2008) The effect of the polyadenylation inhibitor cordycepin on human Molt-4 and Daudi leukaemia and lymphoma cell lines. Cancer Chemother Pharmacol 61:703–711

    CAS  PubMed  Google Scholar 

  36. Liao Y, Ling J, Zhang G et al (2015) Cordycepin induces cell cycle arrest and apoptosis by inducing DNA damage and up-regulation of p53 in Leukemia cells. Cell Cycle 14:761–771

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Tania M, Shawon J, Saif K et al (2019) Cordycepin downregulates cdk-2 to interfere with cell cycle and increases apoptosis by generating ros in cervical cancer cells: in vitro and in silico study. Curr Cancer Drug Targets 19:152–159

    CAS  PubMed  Google Scholar 

  38. Lee S, Debnath T, Kim S, Lim B (2013) Anti-cancer effect and apoptosis induction of cordycepin through DR3 pathway in the human colonic cancer cell HT-29. Food Chem Toxic 60:439–447

    CAS  Google Scholar 

  39. Liu C, Qi M, Li L, Yuan Y, Wu X, Fu J (2020) Natural cordycepin induces apoptosis and suppresses metastasis in breast cancer cells by inhibiting the Hedgehog pathway. Food Funct 11:2107–2116

    CAS  PubMed  Google Scholar 

  40. Wang C, Tsai S, Chien H et al (2020) Cordycepin inhibits human gestational choriocarcinoma cell growth by disrupting centrosome homeostasis. Drug Des Dev Ther 14:2987–3000

    CAS  Google Scholar 

  41. Zheng Q, Sun J, Li W, Li S, Zhang K (2020) Cordycepin induces apoptosis in human tongue cancer cells in vitro and has antitumor effects in vivo. Arch Oral Biol 118:104846

    CAS  PubMed  Google Scholar 

  42. Jeong J, Jin C, Park C et al (2011) Induction of apoptosis by cordycepin via reactive oxygen species generation in human leukemia cells. Toxic Vitro 25:817–824

    CAS  Google Scholar 

  43. Wang C, Shao L, Pan C et al (2019) Elevated level of mitochondrial reactive oxygen species via fatty acid β-oxidation in cancer stem cells promotes cancer metastasis by inducing epithelial-mesenchymal transition. Stem Cell Res Ther 10:175

    PubMed  PubMed Central  Google Scholar 

  44. Liou G, Storz P (2010) Reactive oxygen species in cancer. Free Radical Res 44:479–496

    CAS  Google Scholar 

  45. Kwon D, Cha H, Lee H et al (2019) Protective effect of glutathione against oxidative stress induced cytotoxicity in RAW 264.7 macrophages through activating the nuclear factor erythroid 2-related factor-2/heme oxygenase-1 pathway. Antioxidants. https://doi.org/10.3390/antiox8040082

    Article  PubMed  PubMed Central  Google Scholar 

  46. Bansal A, Simon M (2018) Glutathione metabolism in cancer progression and treatment resistance. J Cell Biol 217:2291–2298

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Choi W, Kim G, Lee W, Choi Y (2008) Sanguinarine, a benzophenanthridine alkaloid, induces apoptosis in MDA-MB-231 human breast carcinoma cells through a reactive oxygen species-mediated mitochondrial pathway. Chemotherapy 54:279–287

    CAS  PubMed  Google Scholar 

  48. Galluzzi L, Blomgren K, Kroemer G (2009) Mitochondrial membrane permeabilization in neuronal injury. Nat Rev Neurosci 10:481–494

    CAS  PubMed  Google Scholar 

  49. Aldridge D, Radford I (1998) Explaining differences in sensitivity to killing by ionizing radiation between human lymphoid cell lines. Can Res 58:2817–2824

    CAS  Google Scholar 

  50. Liu W, Chen C, Lu I et al (2014) MJ-66 induces malignant glioma cells G2/M phase arrest and mitotic catastrophe through regulation of cyclin B1/Cdk1 complex. Neuropharmacology 86:219–227

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Lee J, Kim J, Ahn J, Lee K, Baek N, Choi J (2013) Jaceosidin, isolated from dietary mugwort (Artemisia princeps), induces G2/M cell cycle arrest by inactivating cdc25C-cdc2 via ATM-Chk1/2 activation. Food Chem Toxic 55:214–221

    CAS  Google Scholar 

  52. Gao J, Zhao Y, Lv Y et al (2013) Mirk/Dyrk1B mediates G0/G1 to S phase cell cycle progression and cell survival involving MAPK/ERK signaling in human cancer cells. Cancer Cell Int 13:2

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Yung HW, Alnaes-Katjavivi P, Jones CJ et al (2016) Placental endoplasmic reticulum stress in gestational diabetes: the potential for therapeutic intervention with chemical chaperones and antioxidants. Diabetologia 59:2240–2250

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This study was funded by the Natural Science Foundation of Liaoning Province (Grant Number 2019-ZD-0564).

Author information

Authors and Affiliations

Authors

Contributions

DY designed this study. LS, HC, SF, ZJ and XL performed the experiments. LS and HC wrote the manuscript and analyzed the results. ZC and DY planned and supervised the work. DY was responsible for funding acquisition. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Zhengguo Cui or Dayong Yu.

Ethics declarations

Conflicts of interest

The authors declare no conflicts of interest.

Ethical approval

Not applicable.

Consent for publication

Consent for publication submission is approved by all authors.

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 (TIF 1851 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shi, L., Cao, H., Fu, S. et al. Cordycepin enhances hyperthermia-induced apoptosis and cell cycle arrest by modulating the MAPK pathway in human lymphoma U937 cells. Mol Biol Rep 49, 8673–8683 (2022). https://doi.org/10.1007/s11033-022-07705-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11033-022-07705-6

Keywords

Navigation