Skip to main content
Top
Published in: Medical Oncology 7/2021

01-07-2021 | Breast Cancer | Original Paper

Melittin inhibits the expression of key genes involved in tumor microenvironment formation by suppressing HIF-1α signaling in breast cancer cells

Authors: Zabih Mir Hassani, Mohammad Nabiuni, Kazem Parivar, Somayeh Abdirad, Latifeh Karimzadeh

Published in: Medical Oncology | Issue 7/2021

Login to get access

Abstract

HIF-1α has critical roles in the formation of tumor microenvironment by regulating genes involved in angiogenesis and anaerobic respiration. TME fuels tumors' growth and metastasis and presents therapy with several challenges. Therefore, we aimed to investigate if Melittin disrupts HIF-1α signaling pathway in breast adenocarcinoma cell line MDA-MB-231. Breast adenocarcinoma cell line MDA-MB-231 was cultured in the presence of different doses of Melittin, and MTT assay was carried out to measure Melittin’s cytotoxic effects. Cells were exposed to 5% O2 to mimic hypoxic conditions and Melittin. Western blot was used to measure HIF-1α protein levels. Gene expression analysis was performed using real-time PCR to measure relative mRNA abundance of genes involved in tumor microenvironment formation. Our results revealed that Melittin effectively inhibits HIF-1α at transcriptional and translational/post-translational level. HIF-1α protein and mRNA level were significantly decreased in Melittin-treated groups. It is found that inhibition of HIF-1α by Melittin is through downregulation of NFκB gene expression. Furthermore, gene expression analysis showed a downregulation in VEGFA and LDHA expression due to inhibition of HIF-1α protein by Melittin. In addition, cell toxicity assay showed that Melittin inhibits the growth of MDA-MB-231 cell line through activation of extrinsic and intrinsic apoptotic pathways by upregulating TNFA and BAX expression. Melittin suppresses the expression of genes responsible for formation of TME physiological hallmarks by suppressing HIF-1α signaling pathway. Our results suggest that Melittin can modulate tumor microenvironment by inhibition of VEGFA and LDHA.
Literature
1.
go back to reference Höckel M, Vaupel P. Tumor hypoxia: definitions and current clinical, biologic, and molecular aspects. J Natl Cancer Inst. 2001;93(4):266–76.CrossRef Höckel M, Vaupel P. Tumor hypoxia: definitions and current clinical, biologic, and molecular aspects. J Natl Cancer Inst. 2001;93(4):266–76.CrossRef
2.
go back to reference Vaupel P, Mayer A. Hypoxia in cancer: significance and impact on clinical outcome. Cancer Metastasis Rev. 2007;26:225–39.CrossRef Vaupel P, Mayer A. Hypoxia in cancer: significance and impact on clinical outcome. Cancer Metastasis Rev. 2007;26:225–39.CrossRef
3.
go back to reference Semenza L. Hypoxia-inducible factor 1: master regulator of O2 homeostasis. Curr Opin Genet Dev. 1998;8(5):588–94.CrossRef Semenza L. Hypoxia-inducible factor 1: master regulator of O2 homeostasis. Curr Opin Genet Dev. 1998;8(5):588–94.CrossRef
4.
go back to reference Bonello S, et al. Reactive oxygen species activate the HIF-1α promoter via a functional NFκB site. Arterioscler Thromb Vasc Biol. 2007;27(4):755–61.CrossRef Bonello S, et al. Reactive oxygen species activate the HIF-1α promoter via a functional NFκB site. Arterioscler Thromb Vasc Biol. 2007;27(4):755–61.CrossRef
5.
go back to reference Niu G, et al. Signal transducer and activator of transcription 3 is required for hypoxia-inducible factor-1α RNA expression in both tumor cells and tumor-associated myeloid cells. Mol Cancer Res. 2008;6(7):1099–105.CrossRef Niu G, et al. Signal transducer and activator of transcription 3 is required for hypoxia-inducible factor-1α RNA expression in both tumor cells and tumor-associated myeloid cells. Mol Cancer Res. 2008;6(7):1099–105.CrossRef
6.
go back to reference Semenza GL (2007) Life with Oxygen. October, pp 62–64. Semenza GL (2007) Life with Oxygen. October, pp 62–64.
7.
go back to reference Semenza GL. The hypoxic tumor microenvironment: a driving force for breast cancer progression. Biochim Biophys Acta Mol Cell Res. 2015;1863(3):382–91.CrossRef Semenza GL. The hypoxic tumor microenvironment: a driving force for breast cancer progression. Biochim Biophys Acta Mol Cell Res. 2015;1863(3):382–91.CrossRef
8.
go back to reference Chen C, Pore N, Behrooz A, Ismail-Beigi F, Maity A. Regulation of glut1 mRNA by Hypoxia-inducible Factor-1. J Biol Chem. 2001;276(12):9519–25.CrossRef Chen C, Pore N, Behrooz A, Ismail-Beigi F, Maity A. Regulation of glut1 mRNA by Hypoxia-inducible Factor-1. J Biol Chem. 2001;276(12):9519–25.CrossRef
9.
go back to reference O’rourke JF, Pugh CW, Bartlett SM, Ratcliffe PJ. Identification of hypoxically inducible mRNAs in HeLa cells using differential-display PCR. Eur J Biochem. 1996;241(2):403–10.CrossRef O’rourke JF, Pugh CW, Bartlett SM, Ratcliffe PJ. Identification of hypoxically inducible mRNAs in HeLa cells using differential-display PCR. Eur J Biochem. 1996;241(2):403–10.CrossRef
10.
go back to reference Semenza GL, et al. Hypoxia response elements in the aldolase A, enolase 1, and lactate dehydrogenase A Gene promoters contain essential binding sites for hypoxia-inducible factor 1. J Biol Chem. 1996;271(51):32529–37.CrossRef Semenza GL, et al. Hypoxia response elements in the aldolase A, enolase 1, and lactate dehydrogenase A Gene promoters contain essential binding sites for hypoxia-inducible factor 1. J Biol Chem. 1996;271(51):32529–37.CrossRef
11.
go back to reference Mathupala SP, Rempel A, Pedersen PL. Glucose catabolism in cancer cells. J Biol Chem. 2001;276(46):43407–12.CrossRef Mathupala SP, Rempel A, Pedersen PL. Glucose catabolism in cancer cells. J Biol Chem. 2001;276(46):43407–12.CrossRef
12.
go back to reference Papandreou I, et al. HIF-1 mediates adaptation to hypoxia by actively downregulating mitochondrial oxygen consumption. Cell Metab. 2006;3(3):187–97.CrossRef Papandreou I, et al. HIF-1 mediates adaptation to hypoxia by actively downregulating mitochondrial oxygen consumption. Cell Metab. 2006;3(3):187–97.CrossRef
13.
go back to reference Helmlinger G, Yuan F, Dellian M, Jain RK. Interstitial pH and pO2 gradients in solid tumors in vivo: high-resolution measurements reveal a lack of correlation. Nat Med. 1997;3(2):177–82.CrossRef Helmlinger G, Yuan F, Dellian M, Jain RK. Interstitial pH and pO2 gradients in solid tumors in vivo: high-resolution measurements reveal a lack of correlation. Nat Med. 1997;3(2):177–82.CrossRef
14.
go back to reference Newell K, Franchi A, Pouysségur J, Tannock I. Studies with glycolysis-deficient cells suggest that production of lactic acid is not the only cause of tumor acidity. Proc Natl Acad Sci U S A. 1993;90(3):1127–31.CrossRef Newell K, Franchi A, Pouysségur J, Tannock I. Studies with glycolysis-deficient cells suggest that production of lactic acid is not the only cause of tumor acidity. Proc Natl Acad Sci U S A. 1993;90(3):1127–31.CrossRef
15.
go back to reference Wykoff CC, et al. Hypoxia-inducible expression of tumor-associated carbonic anhydrases. Cancer Res. 2000;60:24. Wykoff CC, et al. Hypoxia-inducible expression of tumor-associated carbonic anhydrases. Cancer Res. 2000;60:24.
16.
go back to reference Helmlinger G, Sckell A, Dellian M, Forbes NS, Jain RK. Acid production in glycolysis-impaired tumors provides new insights into tumor metabolism. Clin Cancer Res. 2002;8:4. Helmlinger G, Sckell A, Dellian M, Forbes NS, Jain RK. Acid production in glycolysis-impaired tumors provides new insights into tumor metabolism. Clin Cancer Res. 2002;8:4.
17.
go back to reference Swietach P, Vaughan-Jones RD, Harris AL. Regulation of tumor pH and the role of carbonic anhydrase 9. Cancer Metast Rev. 2007;26(2):299–310.CrossRef Swietach P, Vaughan-Jones RD, Harris AL. Regulation of tumor pH and the role of carbonic anhydrase 9. Cancer Metast Rev. 2007;26(2):299–310.CrossRef
18.
go back to reference Krock BL, Skuli N, Simon MC. Hypoxia-induced angiogenesis: good and evil. Genes Cancer. 2011;2(12):1117–33.CrossRef Krock BL, Skuli N, Simon MC. Hypoxia-induced angiogenesis: good and evil. Genes Cancer. 2011;2(12):1117–33.CrossRef
19.
go back to reference Forsythe JA, et al. Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Mol Cell Biol. 1996;16(9):4604–13.CrossRef Forsythe JA, et al. Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Mol Cell Biol. 1996;16(9):4604–13.CrossRef
20.
go back to reference Fukumura D, Jain RK. Tumor microenvironment abnormalities: causes, consequences, and strategies to normalize. J Cell Biochem. 2007;101:937–45.CrossRef Fukumura D, Jain RK. Tumor microenvironment abnormalities: causes, consequences, and strategies to normalize. J Cell Biochem. 2007;101:937–45.CrossRef
21.
go back to reference Cairns R, Papandreou I, Denko N. Overcoming physiologic barriers to cancer treatment by molecularly targeting the tumor microenvironment. Mol Cancer Rea. 2006;4:61–71.CrossRef Cairns R, Papandreou I, Denko N. Overcoming physiologic barriers to cancer treatment by molecularly targeting the tumor microenvironment. Mol Cancer Rea. 2006;4:61–71.CrossRef
22.
go back to reference Raghuraman ACH. Melittin: a membrane-active peptide with diverse functions. Biosci Rep. 2007;27:189–223.CrossRef Raghuraman ACH. Melittin: a membrane-active peptide with diverse functions. Biosci Rep. 2007;27:189–223.CrossRef
23.
go back to reference Gajski G, Garaj-Vrhovac V. Melittin: a lytic peptide with anticancer properties. Environ Toxicol Pharmacol. 2013;36(2):697–705.CrossRef Gajski G, Garaj-Vrhovac V. Melittin: a lytic peptide with anticancer properties. Environ Toxicol Pharmacol. 2013;36(2):697–705.CrossRef
24.
go back to reference Shin J-M, et al. Melittin suppresses HIF-1α/VEGF expression through Inhibition of ERK and mTOR/p70S6K pathway in human cervical carcinoma cells. PLoS ONE. 2013;8(7):e69380.CrossRef Shin J-M, et al. Melittin suppresses HIF-1α/VEGF expression through Inhibition of ERK and mTOR/p70S6K pathway in human cervical carcinoma cells. PLoS ONE. 2013;8(7):e69380.CrossRef
25.
go back to reference Bakmiwewa SM, Heng B, Guillemin GJ, Ball HJ, Hunt NH. An effective, low-cost method for achieving and maintaining hypoxia during cell culture studies. Biotechniques. 2015;59(4):223–9.CrossRef Bakmiwewa SM, Heng B, Guillemin GJ, Ball HJ, Hunt NH. An effective, low-cost method for achieving and maintaining hypoxia during cell culture studies. Biotechniques. 2015;59(4):223–9.CrossRef
26.
go back to reference Michiels C, Tellier C, Feron O. Cycling hypoxia: a key feature of the tumor microenvironment. Biochim Biophys Acta Rev Cancer. 2016;1866(1):76–86.CrossRef Michiels C, Tellier C, Feron O. Cycling hypoxia: a key feature of the tumor microenvironment. Biochim Biophys Acta Rev Cancer. 2016;1866(1):76–86.CrossRef
27.
go back to reference Delprat V, Tellier C, Demazy C, Raes M, Feron O, Michiels C. Cycling hypoxia promotes a pro-inflammatory phenotype in macrophages via JNK/p65 signaling pathway. Sci Rep. 2020;10(1):1–13.CrossRef Delprat V, Tellier C, Demazy C, Raes M, Feron O, Michiels C. Cycling hypoxia promotes a pro-inflammatory phenotype in macrophages via JNK/p65 signaling pathway. Sci Rep. 2020;10(1):1–13.CrossRef
28.
go back to reference Zhang SF, Chen Z. Melittin exerts an antitumor effect on non-small celllung cancer cells. Mol Med Rep. 2017;16(3):3581–6.CrossRef Zhang SF, Chen Z. Melittin exerts an antitumor effect on non-small celllung cancer cells. Mol Med Rep. 2017;16(3):3581–6.CrossRef
29.
go back to reference Shu Y, et al. Melittin inducing the apoptosis of renal tubule epithelial cells through upregulation of Bax/Bcl-2 expression and activation of TNF-signaling pathway. Biomed Res Int. 2019;2019:1–13. Shu Y, et al. Melittin inducing the apoptosis of renal tubule epithelial cells through upregulation of Bax/Bcl-2 expression and activation of TNF-signaling pathway. Biomed Res Int. 2019;2019:1–13.
30.
go back to reference D’Ignazio L, Batie M, Rocha S. Hypoxia and inflammation in cancer, focus on HIF and NF-κB. Biomedicines. 2017;5(2):1–10. D’Ignazio L, Batie M, Rocha S. Hypoxia and inflammation in cancer, focus on HIF and NF-κB. Biomedicines. 2017;5(2):1–10.
31.
32.
go back to reference Kim SK, et al. Melittin enhances apoptosis through suppression of IL-6/sIL-6R complex-induced NF-κB and STAT3 activation and Bcl-2 expression for human fibroblast-like synoviocytes in rheumatoid arthritis. Jt Bone Spine. 2011;78(5):471–7.CrossRef Kim SK, et al. Melittin enhances apoptosis through suppression of IL-6/sIL-6R complex-induced NF-κB and STAT3 activation and Bcl-2 expression for human fibroblast-like synoviocytes in rheumatoid arthritis. Jt Bone Spine. 2011;78(5):471–7.CrossRef
33.
go back to reference Jo M, et al. Anti-cancer effect of bee venom toxin and melittin in ovarian cancer cells through induction of death receptors and inhibition of JAK2/STAT3 pathway. Toxicol Appl Pharmacol. 2012;258(1):72–81.CrossRef Jo M, et al. Anti-cancer effect of bee venom toxin and melittin in ovarian cancer cells through induction of death receptors and inhibition of JAK2/STAT3 pathway. Toxicol Appl Pharmacol. 2012;258(1):72–81.CrossRef
34.
go back to reference Zhang Z, Zhang H, Peng T, Li D, Xu J. Melittin suppresses cathepsin S-induced invasion and angiogenesis via blocking of the VEGF-A/VEGFR-2/MEK1/ERK1/2 pathway in human hepatocellular carcinoma. Oncol Lett. 2016;11(1):610–8.CrossRef Zhang Z, Zhang H, Peng T, Li D, Xu J. Melittin suppresses cathepsin S-induced invasion and angiogenesis via blocking of the VEGF-A/VEGFR-2/MEK1/ERK1/2 pathway in human hepatocellular carcinoma. Oncol Lett. 2016;11(1):610–8.CrossRef
35.
go back to reference Jin Z, et al. Melittin constrains the expression of identified key genes associated with bladder cancer. J Immunol Res. 2018;2018:1–16. Jin Z, et al. Melittin constrains the expression of identified key genes associated with bladder cancer. J Immunol Res. 2018;2018:1–16.
36.
go back to reference Huh JE, et al. Melittin suppresses VEGF-A-induced tumor growth by blocking VEGFR-2 and the COX-2-mediated MAPK signaling pathway. J Nat Prod. 2012;75(11):1922–9.CrossRef Huh JE, et al. Melittin suppresses VEGF-A-induced tumor growth by blocking VEGFR-2 and the COX-2-mediated MAPK signaling pathway. J Nat Prod. 2012;75(11):1922–9.CrossRef
37.
go back to reference El Bakary NM, Alsharkawy AZ, Shouaib ZA, Barakat EMS. Role of bee venom and melittin on restraining angiogenesis and metastasis in γ-irradiated solid ehrlich carcinoma-bearing mice. Integr Cancer Ther. 2020;19:153473542094447.CrossRef El Bakary NM, Alsharkawy AZ, Shouaib ZA, Barakat EMS. Role of bee venom and melittin on restraining angiogenesis and metastasis in γ-irradiated solid ehrlich carcinoma-bearing mice. Integr Cancer Ther. 2020;19:153473542094447.CrossRef
38.
go back to reference Chang SN, Kim HJ, Lee KC. Melittin, a major polypeptide of bee venom, increases radiosensitivity of breast cancer in vitro and in vivo. Int J Radiat Oncol. 2020;108(3):e527.CrossRef Chang SN, Kim HJ, Lee KC. Melittin, a major polypeptide of bee venom, increases radiosensitivity of breast cancer in vitro and in vivo. Int J Radiat Oncol. 2020;108(3):e527.CrossRef
39.
go back to reference Milosevic M et al. (2001) Interstitial fluid pressure predicts survival in patients with cervix cancer independent of clinical prognostic factors and tumor. pp 6400–6405. Milosevic M et al. (2001) Interstitial fluid pressure predicts survival in patients with cervix cancer independent of clinical prognostic factors and tumor. pp 6400–6405.
Metadata
Title
Melittin inhibits the expression of key genes involved in tumor microenvironment formation by suppressing HIF-1α signaling in breast cancer cells
Authors
Zabih Mir Hassani
Mohammad Nabiuni
Kazem Parivar
Somayeh Abdirad
Latifeh Karimzadeh
Publication date
01-07-2021
Publisher
Springer US
Published in
Medical Oncology / Issue 7/2021
Print ISSN: 1357-0560
Electronic ISSN: 1559-131X
DOI
https://doi.org/10.1007/s12032-021-01526-6

Other articles of this Issue 7/2021

Medical Oncology 7/2021 Go to the issue