Skip to main content
Top
Published in: Cancer Cell International 1/2020

Open Access 01-12-2020 | Cancer Therapy | Review

Autophagy regulation using luteolin: new insight into its anti-tumor activity

Authors: Milad Ashrafizadeh, Zahra Ahmadi, Tahereh Farkhondeh, Saeed Samarghandian

Published in: Cancer Cell International | Issue 1/2020

Login to get access

Abstract

Application of novel methods in cancer therapy is important in terms of management and treatment of the life-threatening disorder. It appears that autophagy is a potential target in cancer therapy, as a variety of drugs targeting autophagy have shown great potential in reducing the viability and proliferation of cancer cells. Autophagy is primarily a catabolic process which provides energy during starvation. Besides, this process contributes to the degradation of aged or potentially toxic components and organelles. On the other hand, the source of a variety of naturally occurring anti-tumor drugs are flavonoids which have high anti-tumor activity. Luteolin is a polyphenolic flavone with the great pharmacological effects such as anti-diabetic, hepatoprotective, antioxidant, anti-inflammation, and anti-tumor. At the present review, we demonstrate how luteolin affects on autophagy process to induce anti-tumor activity.
Literature
1.
go back to reference Mohammadinejad R, et al. Berberine as a potential autophagy modulator. J Cell Physiol. 2019;234(9):14914–26. CrossRef Mohammadinejad R, et al. Berberine as a potential autophagy modulator. J Cell Physiol. 2019;234(9):14914–26. CrossRef
2.
go back to reference Ashrafizadeh M, Ahmadi Z. Effects of statins on gut microbiota (microbiome). Rev Clin Med. 2019;6(2):55–9. Ashrafizadeh M, Ahmadi Z. Effects of statins on gut microbiota (microbiome). Rev Clin Med. 2019;6(2):55–9.
3.
go back to reference Ashrafizadeh M, et al. Effects of newly introduced antidiabetic drugs on autophagy. Diabetes & Metabolic Syndrome: Clinical Research & Reviews; 2019. Ashrafizadeh M, et al. Effects of newly introduced antidiabetic drugs on autophagy. Diabetes & Metabolic Syndrome: Clinical Research & Reviews; 2019.
4.
go back to reference Sobhani B, et al. Histopathological analysis of testis: effects of astaxanthin treatment against nicotine toxicity. Iranian Journal of Toxicology. 2019;13(1):41–4. Sobhani B, et al. Histopathological analysis of testis: effects of astaxanthin treatment against nicotine toxicity. Iranian Journal of Toxicology. 2019;13(1):41–4.
5.
go back to reference Panche A, Diwan A, Chandra S. Flavonoids: an overview. J Nutr Sci, 2016. 5. Panche A, Diwan A, Chandra S. Flavonoids: an overview. J Nutr Sci, 2016. 5.
6.
go back to reference Swaminathan A, et al. The dietary flavonoid, luteolin, negatively affects neuronal differentiation. Front Mol Neurosci. 2019;12:41. CrossRef Swaminathan A, et al. The dietary flavonoid, luteolin, negatively affects neuronal differentiation. Front Mol Neurosci. 2019;12:41. CrossRef
7.
go back to reference Schönrich G, Raftery MJ. The PD-1/PD-L1 axis and virus infections: a delicate balance. Front Cell Infect Microbiol. 2019;9:207. CrossRef Schönrich G, Raftery MJ. The PD-1/PD-L1 axis and virus infections: a delicate balance. Front Cell Infect Microbiol. 2019;9:207. CrossRef
8.
go back to reference Imran M, et al. Luteolin, a flavonoid, as an anticancer agent: a review. Biomed Pharmacother. 2019;112:108612. CrossRef Imran M, et al. Luteolin, a flavonoid, as an anticancer agent: a review. Biomed Pharmacother. 2019;112:108612. CrossRef
9.
go back to reference Skaper SD, et al. Co-ultramicronized palmitoylethanolamide/luteolin facilitates the development of differentiating and undifferentiated rat oligodendrocyte progenitor cells. Mol Neurobiol. 2018;55(1):103–14.CrossRef Skaper SD, et al. Co-ultramicronized palmitoylethanolamide/luteolin facilitates the development of differentiating and undifferentiated rat oligodendrocyte progenitor cells. Mol Neurobiol. 2018;55(1):103–14.CrossRef
10.
go back to reference Liu L, et al. Luteolin and apigenin activate the Oct-4/Sox2 signal via NFATc1 in human periodontal ligament cells. Cell Biol Int. 2016;40(10):1094–106.CrossRef Liu L, et al. Luteolin and apigenin activate the Oct-4/Sox2 signal via NFATc1 in human periodontal ligament cells. Cell Biol Int. 2016;40(10):1094–106.CrossRef
11.
go back to reference Choi H-J, et al. Luteolin inhibits recruitment of monocytes and migration of Lewis lung carcinoma cells by suppressing chemokine (C–C motif) ligand 2 expression in tumor-associated macrophage. Biochem Biophys Res Commun. 2016;470(1):101–6.CrossRef Choi H-J, et al. Luteolin inhibits recruitment of monocytes and migration of Lewis lung carcinoma cells by suppressing chemokine (C–C motif) ligand 2 expression in tumor-associated macrophage. Biochem Biophys Res Commun. 2016;470(1):101–6.CrossRef
12.
go back to reference Hong Z, et al. Luteolin is effective in the non-small cell lung cancer model with L 858 R/T 790 M EGF receptor mutation and erlotinib resistance. Br J Pharmacol. 2014;171(11):2842–53.CrossRef Hong Z, et al. Luteolin is effective in the non-small cell lung cancer model with L 858 R/T 790 M EGF receptor mutation and erlotinib resistance. Br J Pharmacol. 2014;171(11):2842–53.CrossRef
13.
go back to reference Bai L, et al. A superoxide-mediated mitogen-activated protein kinase phosphatase-1 degradation and c-Jun NH2-terminal kinase activation pathway for luteolin-induced lung cancer cytotoxicity. Mol Pharmacol. 2012;81(4):549–55.CrossRef Bai L, et al. A superoxide-mediated mitogen-activated protein kinase phosphatase-1 degradation and c-Jun NH2-terminal kinase activation pathway for luteolin-induced lung cancer cytotoxicity. Mol Pharmacol. 2012;81(4):549–55.CrossRef
14.
go back to reference Glick D, Barth S, Macleod KF. Autophagy: cellular and molecular mechanisms. J Pathol. 2010;221(1):3–12.CrossRef Glick D, Barth S, Macleod KF. Autophagy: cellular and molecular mechanisms. J Pathol. 2010;221(1):3–12.CrossRef
15.
go back to reference Ktistakis NT. In praise of M. Anselmier who first used the term “autophagie” in 1859. Milton Park: Taylor & Francis; 2017. CrossRef Ktistakis NT. In praise of M. Anselmier who first used the term “autophagie” in 1859. Milton Park: Taylor & Francis; 2017. CrossRef
16.
go back to reference Harnett MM, et al. From Christian de Duve to Yoshinori Ohsumi: more to autophagy than just dining at home. Biomed J. 2017;40(1):9–22. CrossRef Harnett MM, et al. From Christian de Duve to Yoshinori Ohsumi: more to autophagy than just dining at home. Biomed J. 2017;40(1):9–22. CrossRef
17.
go back to reference Li W, et al. Immunofluorescence staining protocols for major autophagy proteins including LC3, P62, and ULK1 in mammalian cells in response to normoxia and hypoxia, in autophagy in differentiation and tissue maintenance. Berlin: Springer; 2018. p. 175–85. Li W, et al. Immunofluorescence staining protocols for major autophagy proteins including LC3, P62, and ULK1 in mammalian cells in response to normoxia and hypoxia, in autophagy in differentiation and tissue maintenance. Berlin: Springer; 2018. p. 175–85.
18.
go back to reference Zhang X, et al. Classical and alternative roles for autophagy in lipid metabolism. Curr Opin Lipidol. 2018;29(3):203.CrossRef Zhang X, et al. Classical and alternative roles for autophagy in lipid metabolism. Curr Opin Lipidol. 2018;29(3):203.CrossRef
19.
go back to reference Wang P, et al. Autophagy in ischemic stroke. Prog Neurobiol. 2018;163:98–117. CrossRef Wang P, et al. Autophagy in ischemic stroke. Prog Neurobiol. 2018;163:98–117. CrossRef
20.
go back to reference Kalachev AV, Yurchenko OV. Microautophagy in nutritive phagocytes of sea urchins. Protoplasma. 2017;254(1):609–14.CrossRef Kalachev AV, Yurchenko OV. Microautophagy in nutritive phagocytes of sea urchins. Protoplasma. 2017;254(1):609–14.CrossRef
21.
go back to reference Alfaro IE, et al. Chaperone mediated autophagy in the crosstalk of neurodegenerative diseases and metabolic disorders. Front Endocrinol. 2018;9:778. CrossRef Alfaro IE, et al. Chaperone mediated autophagy in the crosstalk of neurodegenerative diseases and metabolic disorders. Front Endocrinol. 2018;9:778. CrossRef
22.
go back to reference Wu D, Zhang K, Hu P. The role of autophagy in acute myocardial infarction. Front Pharmacol. 2019;10:551. CrossRef Wu D, Zhang K, Hu P. The role of autophagy in acute myocardial infarction. Front Pharmacol. 2019;10:551. CrossRef
23.
go back to reference Galluzzi L, Green DR. Autophagy-independent functions of the autophagy machinery. Cell. 2019;177(7):1682–99.CrossRef Galluzzi L, Green DR. Autophagy-independent functions of the autophagy machinery. Cell. 2019;177(7):1682–99.CrossRef
24.
go back to reference Levine B, Kroemer G. Biological functions of autophagy genes: a disease perspective. Cell. 2019;176(1–2):11–42.CrossRef Levine B, Kroemer G. Biological functions of autophagy genes: a disease perspective. Cell. 2019;176(1–2):11–42.CrossRef
25.
go back to reference Hurley JH, Young LN. Mechanisms of autophagy initiation. Annual Rev Biochem. 2017;86:225–44.CrossRef Hurley JH, Young LN. Mechanisms of autophagy initiation. Annual Rev Biochem. 2017;86:225–44.CrossRef
26.
go back to reference Bar-Yosef T, Damri O, Agam G. Dual role of autophagy in diseases of the central nervous system. Front Cell Neurosci. 2019;13:196. CrossRef Bar-Yosef T, Damri O, Agam G. Dual role of autophagy in diseases of the central nervous system. Front Cell Neurosci. 2019;13:196. CrossRef
27.
go back to reference Rajendran P, et al. Autophagy and senescence: A new insight in selected human diseases. J Cell Physiol. 2019;234(12):21485–92.CrossRef Rajendran P, et al. Autophagy and senescence: A new insight in selected human diseases. J Cell Physiol. 2019;234(12):21485–92.CrossRef
28.
go back to reference Condello M, et al. Targeting autophagy to overcome human diseases. Int J Mol Sci. 2019;20(3):725.CrossRef Condello M, et al. Targeting autophagy to overcome human diseases. Int J Mol Sci. 2019;20(3):725.CrossRef
29.
go back to reference Geisler S, et al. PINK1/Parkin-mediated mitophagy is dependent on VDAC1 and p62/SQSTM1. Nat Cell Biol. 2010;12(2):119.CrossRef Geisler S, et al. PINK1/Parkin-mediated mitophagy is dependent on VDAC1 and p62/SQSTM1. Nat Cell Biol. 2010;12(2):119.CrossRef
30.
go back to reference Xu W, Tian M, Zhou Y. The relationship between insulin resistance, adiponectin and C-reactive protein and vascular endothelial injury in diabetic patients with coronary heart disease. Exp Ther Med. 2018;16(3):2022–6. Xu W, Tian M, Zhou Y. The relationship between insulin resistance, adiponectin and C-reactive protein and vascular endothelial injury in diabetic patients with coronary heart disease. Exp Ther Med. 2018;16(3):2022–6.
31.
go back to reference Schaaf MB, et al. Autophagy in endothelial cells and tumor angiogenesis. Cell Death Differ. 2019;26:665–79.CrossRef Schaaf MB, et al. Autophagy in endothelial cells and tumor angiogenesis. Cell Death Differ. 2019;26:665–79.CrossRef
32.
go back to reference Wang Y, et al. Insights into autophagy machinery in cells related to skin diseases and strategies for therapeutic modulation. Biomed Pharmacother. 2019;113:108775. CrossRef Wang Y, et al. Insights into autophagy machinery in cells related to skin diseases and strategies for therapeutic modulation. Biomed Pharmacother. 2019;113:108775. CrossRef
33.
go back to reference Li X, et al. Beclin1 inhibition promotes autophagy and decreases gemcitabine–induced apoptosis in Miapaca2 pancreatic cancer cells. Cancer Cell Int. 2013;13(1):26.CrossRef Li X, et al. Beclin1 inhibition promotes autophagy and decreases gemcitabine–induced apoptosis in Miapaca2 pancreatic cancer cells. Cancer Cell Int. 2013;13(1):26.CrossRef
34.
go back to reference Kwon K, et al. Luteolin-induced apoptosis through activation of endoplasmic reticulum stress sensors in pheochromocytoma cells. Mol Med Rep. 2017;16(1):380–6.CrossRef Kwon K, et al. Luteolin-induced apoptosis through activation of endoplasmic reticulum stress sensors in pheochromocytoma cells. Mol Med Rep. 2017;16(1):380–6.CrossRef
35.
go back to reference Nazim UM, Park SY. Luteolin sensitizes human liver cancer cells to TRAIL induced apoptosis via autophagy and JNKmediated death receptor 5 upregulation. Int J Oncol. 2019;54(2):665–72. Nazim UM, Park SY. Luteolin sensitizes human liver cancer cells to TRAIL induced apoptosis via autophagy and JNKmediated death receptor 5 upregulation. Int J Oncol. 2019;54(2):665–72.
36.
go back to reference Chen T, et al. Effects of luteolin on proliferation and programmed cell death of human multiple myeloma cell RPMI-8226. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 2018;26(5):1425–9. Chen T, et al. Effects of luteolin on proliferation and programmed cell death of human multiple myeloma cell RPMI-8226. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 2018;26(5):1425–9.
37.
go back to reference Liu Q, et al. Luteolin promotes the sensitivity of cisplatin in ovarian cancer by decreasing PRPA1-medicated autophagy. Cell Mol Biol. 2018;64(6):17–22. CrossRef Liu Q, et al. Luteolin promotes the sensitivity of cisplatin in ovarian cancer by decreasing PRPA1-medicated autophagy. Cell Mol Biol. 2018;64(6):17–22. CrossRef
38.
go back to reference Cao Z, Zhang H, Cai X, Fang W, Chai D, Wen Y, Chen H, Chu F, Zhang Y. Luteolin promotes cell apoptosis by inducing autophagy in hepatocellular carcinoma. Cell Physiol Biochem. 2017;43(5):1803–12.CrossRef Cao Z, Zhang H, Cai X, Fang W, Chai D, Wen Y, Chen H, Chu F, Zhang Y. Luteolin promotes cell apoptosis by inducing autophagy in hepatocellular carcinoma. Cell Physiol Biochem. 2017;43(5):1803–12.CrossRef
39.
go back to reference Zhang B, Yu X, Xia H. The flavonoid luteolin enhances doxorubicin-induced autophagy in human osteosarcoma U2OS cells. Int J Clin Exp Med. 2015;8(9):15190. Zhang B, Yu X, Xia H. The flavonoid luteolin enhances doxorubicin-induced autophagy in human osteosarcoma U2OS cells. Int J Clin Exp Med. 2015;8(9):15190.
40.
go back to reference Verschooten L, et al. Autophagy inhibitor chloroquine enhanced the cell death inducing effect of the flavonoid luteolin in metastatic squamous cell carcinoma cells. PloS ONE. 2012;7(10):e48264. CrossRef Verschooten L, et al. Autophagy inhibitor chloroquine enhanced the cell death inducing effect of the flavonoid luteolin in metastatic squamous cell carcinoma cells. PloS ONE. 2012;7(10):e48264. CrossRef
41.
go back to reference Park S-H, et al. Induction of endoplasmic reticulum stress-mediated apoptosis and non-canonical autophagy by luteolin in NCI-H460 lung carcinoma cells. Food chemical toxicology. 2013;56:100–9.CrossRef Park S-H, et al. Induction of endoplasmic reticulum stress-mediated apoptosis and non-canonical autophagy by luteolin in NCI-H460 lung carcinoma cells. Food chemical toxicology. 2013;56:100–9.CrossRef
42.
go back to reference Monti E, et al., Luteolin impairs hypoxia adaptation and progression in human breast and colon cancer cells. European Journal of Pharmacology, 2020: p. 173210. Monti E, et al., Luteolin impairs hypoxia adaptation and progression in human breast and colon cancer cells. European Journal of Pharmacology, 2020: p. 173210.
43.
go back to reference Potočnjak I, et al. Antitumor activity of luteolin in human colon cancer SW620 cells is mediated by the ERK/FOXO3a signaling pathway. Toxicol In Vitro. 2020;66:104852. CrossRef Potočnjak I, et al. Antitumor activity of luteolin in human colon cancer SW620 cells is mediated by the ERK/FOXO3a signaling pathway. Toxicol In Vitro. 2020;66:104852. CrossRef
44.
go back to reference Soliman NA, Abd-Ellatif RN, ELSaadany AA, Shalaby SM, Bedeer AE. Luteolin and 5-flurouracil act synergistically to induce cellular weapons in experimentally induced Solid Ehrlich Carcinoma: realistic role of P53; a guardian fights in a cellular battle. Chem Biol Interact. 2019;310:108740. CrossRef Soliman NA, Abd-Ellatif RN, ELSaadany AA, Shalaby SM, Bedeer AE. Luteolin and 5-flurouracil act synergistically to induce cellular weapons in experimentally induced Solid Ehrlich Carcinoma: realistic role of P53; a guardian fights in a cellular battle. Chem Biol Interact. 2019;310:108740. CrossRef
45.
go back to reference Chakrabarti M, Ray SK. Anti-tumor activities of luteolin and silibinin in glioblastoma cells: overexpression of miR-7-1-3p augmented luteolin and silibinin to inhibit autophagy and induce apoptosis in glioblastoma in vivo. Apoptosis. 2016;21(3):312–28.CrossRef Chakrabarti M, Ray SK. Anti-tumor activities of luteolin and silibinin in glioblastoma cells: overexpression of miR-7-1-3p augmented luteolin and silibinin to inhibit autophagy and induce apoptosis in glioblastoma in vivo. Apoptosis. 2016;21(3):312–28.CrossRef
46.
go back to reference Wang L, Lee IM, Zhang SM, Blumberg JB, Buring JE, Sesso HD. Dietary intake of selected flavonols, flavones, and flavonoid-rich foods and risk of cancer in middle-aged and older women. Am J Clin Nutr. 2009;89(3):905–12.CrossRef Wang L, Lee IM, Zhang SM, Blumberg JB, Buring JE, Sesso HD. Dietary intake of selected flavonols, flavones, and flavonoid-rich foods and risk of cancer in middle-aged and older women. Am J Clin Nutr. 2009;89(3):905–12.CrossRef
47.
go back to reference Tarahovsky YS, Kim YA, Yagolnik EA, Muzafarov EN. Flavonoid–membrane interactions: involvement of flavonoid–metal complexes in raft signaling. Biochimica et Biophysica Acta. 2014;1838(5):1235–46. CrossRef Tarahovsky YS, Kim YA, Yagolnik EA, Muzafarov EN. Flavonoid–membrane interactions: involvement of flavonoid–metal complexes in raft signaling. Biochimica et Biophysica Acta. 2014;1838(5):1235–46. CrossRef
48.
go back to reference Böhl M, Tietze S, Sokoll A, Madathil S, Pfennig F, Apostolakis J, Fahmy K, Gutzeit HO. Flavonoids affect actin functions in cytoplasm and nucleus. Biophys J. 2007;93(8):2767–80. CrossRef Böhl M, Tietze S, Sokoll A, Madathil S, Pfennig F, Apostolakis J, Fahmy K, Gutzeit HO. Flavonoids affect actin functions in cytoplasm and nucleus. Biophys J. 2007;93(8):2767–80. CrossRef
Metadata
Title
Autophagy regulation using luteolin: new insight into its anti-tumor activity
Authors
Milad Ashrafizadeh
Zahra Ahmadi
Tahereh Farkhondeh
Saeed Samarghandian
Publication date
01-12-2020
Publisher
BioMed Central
Keyword
Cancer Therapy
Published in
Cancer Cell International / Issue 1/2020
Electronic ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-020-01634-9

Other articles of this Issue 1/2020

Cancer Cell International 1/2020 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine