Skip to main content
Erschienen in: Journal of Natural Medicines 3/2020

28.03.2020 | Original Paper

Polydatin induces apoptosis and autophagy via STAT3 signaling in human osteosarcoma MG-63 cells

verfasst von: Chang-qing Jiang, Ling-ling Ma, Zhen-dong Lv, Fan Feng, Zhi Chen, Zu-De Liu

Erschienen in: Journal of Natural Medicines | Ausgabe 3/2020

Einloggen, um Zugang zu erhalten

Abstract

Polydatin, a natural product, is detected in many daily diets, such as grape juices and peanut. Autophagy regulation is recognized as a new potential strategy for cancer therapy, and previous studies demonstrated that polydatin showed remarkable anti-cancer ability. Nevertheless, the capability of polydatin to induce autophagy and its role in anti-osteosarcoma remains obscure. In this study, we investigated the anticancer effect of polydatin on human osteosarcoma cell line MG-63 and its underlying mechanism. Our results indicated that polydatin significantly inhibited proliferation of MG-63 cells in a dose- and time-dependent manner, and increased their apoptosis and autophagic flux. Further experiments showed that polydatin reduced the expression and phosphorylation (Y705) level of STAT3 (Signal transducer and activator of transcription 3), increased the expression of autophagy-related genes (Atg12, Atg14, BECN1, PIC3K3), and therewith triggered autophagic cell death in MG-63 cells. Of note, the cytotoxicity effect of polydatin was rescued by co-treatment with Colivelin (STAT3 activator), suggesting the dependency of MG-63 cells on STAT3 for survival in this process. Moreover, polydatin-triggered autophagy and apoptosis were remarkably reduced following exposure to autophagy inhibitor 3-methyladenine, while cell viability was increased. In conclusion, these data demonstrated that polydatin induced MG-63 cell death through inducing apoptosis, and autophagy which was mediated via the STAT3 signaling. Therefore, polydatin might be a potential clinical drug in the remedy of osteosarcoma.

Graphic abstract

Literatur
1.
Zurück zum Zitat Kansara M, Teng MW, Smyth MJ, Thomas DM (2014) Translational biology of osteosarcoma. Nat Rev Cancer 14:722–735CrossRef Kansara M, Teng MW, Smyth MJ, Thomas DM (2014) Translational biology of osteosarcoma. Nat Rev Cancer 14:722–735CrossRef
2.
Zurück zum Zitat Meyers PA, Healey JH, Chou AJ, Wexler LH, Merola PR, Morris CD, Laquaglia MP, Kellick MG, Abramson SJ, Gorlick R (2011) Addition of pamidronate to chemotherapy for the treatment of osteosarcoma. Cancer 117:1736–1744CrossRef Meyers PA, Healey JH, Chou AJ, Wexler LH, Merola PR, Morris CD, Laquaglia MP, Kellick MG, Abramson SJ, Gorlick R (2011) Addition of pamidronate to chemotherapy for the treatment of osteosarcoma. Cancer 117:1736–1744CrossRef
3.
Zurück zum Zitat Guo YZ, Yang XM, Li YY (2019) Effect of alkylresorcinols on autophagy, migration, and invasion of hepg2 cells. J Food Sci 84:3063–3068CrossRef Guo YZ, Yang XM, Li YY (2019) Effect of alkylresorcinols on autophagy, migration, and invasion of hepg2 cells. J Food Sci 84:3063–3068CrossRef
4.
Zurück zum Zitat Zhao XJ, Yu HW, Yang YZ, Wu WY, Chen TY, Jia KK, Kang LL, Jiao RQ, Kong LD (2018) Polydatin prevents fructose-induced liver inflammation and lipid deposition through increasing mir-200a to regulate keap1/nrf2 pathway. Redox Biol 18:124–137CrossRef Zhao XJ, Yu HW, Yang YZ, Wu WY, Chen TY, Jia KK, Kang LL, Jiao RQ, Kong LD (2018) Polydatin prevents fructose-induced liver inflammation and lipid deposition through increasing mir-200a to regulate keap1/nrf2 pathway. Redox Biol 18:124–137CrossRef
5.
Zurück zum Zitat Mele L, Paino F, Papaccio F, Regad T, Boocock D, Stiuso P, Lombardi A, Liccardo D, Aquino G, Barbieri A et al (2018) A new inhibitor of glucose-6-phosphate dehydrogenase blocks pentose phosphate pathway and suppresses malignant proliferation and metastasis in vivo. Cell Death Dis 9:572CrossRef Mele L, Paino F, Papaccio F, Regad T, Boocock D, Stiuso P, Lombardi A, Liccardo D, Aquino G, Barbieri A et al (2018) A new inhibitor of glucose-6-phosphate dehydrogenase blocks pentose phosphate pathway and suppresses malignant proliferation and metastasis in vivo. Cell Death Dis 9:572CrossRef
6.
Zurück zum Zitat Zhang T, Zhu X, Wu H, Jiang K, Zhao G, Shaukat A, Deng G, Qiu C (2019) Targeting the ros/pi3k/akt/hif-1alpha/hk2 axis of breast cancer cells: Combined administration of polydatin and 2-deoxy-d-glucose. J Cell Mol Med 23:3711–3723CrossRef Zhang T, Zhu X, Wu H, Jiang K, Zhao G, Shaukat A, Deng G, Qiu C (2019) Targeting the ros/pi3k/akt/hif-1alpha/hk2 axis of breast cancer cells: Combined administration of polydatin and 2-deoxy-d-glucose. J Cell Mol Med 23:3711–3723CrossRef
7.
Zurück zum Zitat Levine B, Kroemer G (2008) Autophagy in the pathogenesis of disease. Cell 132:27–42CrossRef Levine B, Kroemer G (2008) Autophagy in the pathogenesis of disease. Cell 132:27–42CrossRef
8.
Zurück zum Zitat Kim S, Eun HS, Jo EK (2019) Roles of autophagy-related genes in the pathogenesis of inflammatory bowel disease. Cells 8(1):77CrossRef Kim S, Eun HS, Jo EK (2019) Roles of autophagy-related genes in the pathogenesis of inflammatory bowel disease. Cells 8(1):77CrossRef
9.
Zurück zum Zitat Wu B, Tan M, Cai W, Wang B, He P, Zhang X (2018) Arsenic trioxide induces autophagic cell death in osteosarcoma cells via the ros-tfeb signaling pathway. Biochem Biophys Res Commun 496:167–175CrossRef Wu B, Tan M, Cai W, Wang B, He P, Zhang X (2018) Arsenic trioxide induces autophagic cell death in osteosarcoma cells via the ros-tfeb signaling pathway. Biochem Biophys Res Commun 496:167–175CrossRef
10.
Zurück zum Zitat Utaipan T, Athipornchai A, Suksamrarn A, Jirachotikoon C, Yuan X, Lertcanawanichakul M, Chunglok W (2017) Carbazole alkaloids from murraya koenigii trigger apoptosis and autophagic flux inhibition in human oral squamous cell carcinoma cells. J Nat Med 71:158–169CrossRef Utaipan T, Athipornchai A, Suksamrarn A, Jirachotikoon C, Yuan X, Lertcanawanichakul M, Chunglok W (2017) Carbazole alkaloids from murraya koenigii trigger apoptosis and autophagic flux inhibition in human oral squamous cell carcinoma cells. J Nat Med 71:158–169CrossRef
11.
Zurück zum Zitat Galluzzi L, Baehrecke EH, Ballabio A, Boya P, Bravo-San Pedro JM, Cecconi F, Choi AM, Chu CT, Codogno P, Colombo MI et al (2017) Molecular definitions of autophagy and related processes. EMBO J 36:1811–1836CrossRef Galluzzi L, Baehrecke EH, Ballabio A, Boya P, Bravo-San Pedro JM, Cecconi F, Choi AM, Chu CT, Codogno P, Colombo MI et al (2017) Molecular definitions of autophagy and related processes. EMBO J 36:1811–1836CrossRef
12.
Zurück zum Zitat Levy JMM, Towers CG, Thorburn A (2017) Targeting autophagy in cancer. Nat Rev Cancer 17:528–542CrossRef Levy JMM, Towers CG, Thorburn A (2017) Targeting autophagy in cancer. Nat Rev Cancer 17:528–542CrossRef
13.
Zurück zum Zitat Onorati AV, Dyczynski M, Ojha R, Amaravadi RK (2018) Targeting autophagy in cancer. Cancer 124:3307–3318CrossRef Onorati AV, Dyczynski M, Ojha R, Amaravadi RK (2018) Targeting autophagy in cancer. Cancer 124:3307–3318CrossRef
14.
Zurück zum Zitat Amaravadi RK, Kimmelman AC, Debnath J (2019) Targeting autophagy in cancer: recent advances and future directions. Cancer Discov 9:1167–1181CrossRef Amaravadi RK, Kimmelman AC, Debnath J (2019) Targeting autophagy in cancer: recent advances and future directions. Cancer Discov 9:1167–1181CrossRef
15.
Zurück zum Zitat Huynh J, Chand A, Gough D, Ernst M (2019) Therapeutically exploiting stat3 activity in cancer—using tissue repair as a road map. Nat Rev Cancer 19:82–96CrossRef Huynh J, Chand A, Gough D, Ernst M (2019) Therapeutically exploiting stat3 activity in cancer—using tissue repair as a road map. Nat Rev Cancer 19:82–96CrossRef
16.
Zurück zum Zitat Johnson DE, O'Keefe RA, Grandis JR (2018) Targeting the il-6/jak/stat3 signalling axis in cancer. Nat Rev Clin Oncol 15:234–248CrossRef Johnson DE, O'Keefe RA, Grandis JR (2018) Targeting the il-6/jak/stat3 signalling axis in cancer. Nat Rev Clin Oncol 15:234–248CrossRef
17.
Zurück zum Zitat You L, Wang Z, Li H, Shou J, Jing Z, Xie J, Sui X, Pan H, Han W (2015) The role of stat3 in autophagy. Autophagy 11:729–739CrossRef You L, Wang Z, Li H, Shou J, Jing Z, Xie J, Sui X, Pan H, Han W (2015) The role of stat3 in autophagy. Autophagy 11:729–739CrossRef
18.
Zurück zum Zitat Zhou X, Qi W, Hong T, Xiong T, Gong D, Xie M, Nie S (2018) Exopolysaccharides from lactobacillus plantarum ncu116 regulate intestinal barrier function via stat3 signaling pathway. J Agric Food Chem 66:9719–9727CrossRef Zhou X, Qi W, Hong T, Xiong T, Gong D, Xie M, Nie S (2018) Exopolysaccharides from lactobacillus plantarum ncu116 regulate intestinal barrier function via stat3 signaling pathway. J Agric Food Chem 66:9719–9727CrossRef
19.
Zurück zum Zitat Zhao C, Li H, Lin HJ, Yang S, Lin J, Liang G (2016) Feedback activation of stat3 as a cancer drug-resistance mechanism. Trends Pharmacol Sci 37:47–61CrossRef Zhao C, Li H, Lin HJ, Yang S, Lin J, Liang G (2016) Feedback activation of stat3 as a cancer drug-resistance mechanism. Trends Pharmacol Sci 37:47–61CrossRef
20.
Zurück zum Zitat Turkson J, Jove R (2000) Stat proteins: Novel molecular targets for cancer drug discovery. Oncogene 19:6613–6626CrossRef Turkson J, Jove R (2000) Stat proteins: Novel molecular targets for cancer drug discovery. Oncogene 19:6613–6626CrossRef
21.
Zurück zum Zitat Roeser JC, Leach SD, McAllister F (2015) Emerging strategies for cancer immunoprevention. Oncogene 34:6029–6039CrossRef Roeser JC, Leach SD, McAllister F (2015) Emerging strategies for cancer immunoprevention. Oncogene 34:6029–6039CrossRef
22.
Zurück zum Zitat Sun L, Hu L, Cogdell D, Lu L, Gao C, Tian W, Zhang Z, Kang Y, Fleming JB, Zhang W (2017) Mir506 induces autophagy-related cell death in pancreatic cancer cells by targeting the stat3 pathway. Autophagy 13:703–714CrossRef Sun L, Hu L, Cogdell D, Lu L, Gao C, Tian W, Zhang Z, Kang Y, Fleming JB, Zhang W (2017) Mir506 induces autophagy-related cell death in pancreatic cancer cells by targeting the stat3 pathway. Autophagy 13:703–714CrossRef
23.
Zurück zum Zitat Shen S, Niso-Santano M, Adjemian S, Takehara T, Malik SA, Minoux H, Souquere S, Marino G, Lachkar S, Senovilla L et al (2012) Cytoplasmic stat3 represses autophagy by inhibiting pkr activity. Mol Cell 48:667–680CrossRef Shen S, Niso-Santano M, Adjemian S, Takehara T, Malik SA, Minoux H, Souquere S, Marino G, Lachkar S, Senovilla L et al (2012) Cytoplasmic stat3 represses autophagy by inhibiting pkr activity. Mol Cell 48:667–680CrossRef
24.
Zurück zum Zitat Yu T, Guo F, Yu Y, Sun T, Ma D, Han J, Qian Y, Kryczek I, Sun D, Nagarsheth N et al (2017) Fusobacterium nucleatum promotes chemoresistance to colorectal cancer by modulating autophagy. Cell 170(548–563):e516 Yu T, Guo F, Yu Y, Sun T, Ma D, Han J, Qian Y, Kryczek I, Sun D, Nagarsheth N et al (2017) Fusobacterium nucleatum promotes chemoresistance to colorectal cancer by modulating autophagy. Cell 170(548–563):e516
25.
Zurück zum Zitat Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, Simonovic M, Doncheva NT, Morris JH, Bork P et al (2019) String v11: protein–protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res 47:D607–D613CrossRef Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, Simonovic M, Doncheva NT, Morris JH, Bork P et al (2019) String v11: protein–protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res 47:D607–D613CrossRef
26.
Zurück zum Zitat Laplante M, Sabatini DM (2013) Regulation of mtorc1 and its impact on gene expression at a glance. J Cell Sci 126:1713–1719CrossRef Laplante M, Sabatini DM (2013) Regulation of mtorc1 and its impact on gene expression at a glance. J Cell Sci 126:1713–1719CrossRef
27.
Zurück zum Zitat Wong RS (2011) Apoptosis in cancer: from pathogenesis to treatment. J Exp Clin Cancer Res 30:87CrossRef Wong RS (2011) Apoptosis in cancer: from pathogenesis to treatment. J Exp Clin Cancer Res 30:87CrossRef
28.
Zurück zum Zitat Pereira DM, Correia-da-Silva G, Valentao P, Teixeira N, Andrade PB (2013) Palmitic acid and ergosta-7,22-dien-3-ol contribute to the apoptotic effect and cell cycle arrest of an extract from marthasterias glacialis l neuroblastoma cells. Mar Drugs 12:54–68CrossRef Pereira DM, Correia-da-Silva G, Valentao P, Teixeira N, Andrade PB (2013) Palmitic acid and ergosta-7,22-dien-3-ol contribute to the apoptotic effect and cell cycle arrest of an extract from marthasterias glacialis l neuroblastoma cells. Mar Drugs 12:54–68CrossRef
29.
Zurück zum Zitat Van Opdenbosch N, Lamkanfi M (2019) Caspases in cell death, inflammation, and disease. Immunity 50:1352–1364CrossRef Van Opdenbosch N, Lamkanfi M (2019) Caspases in cell death, inflammation, and disease. Immunity 50:1352–1364CrossRef
30.
Zurück zum Zitat Dikic I, Elazar Z (2018) Mechanism and medical implications of mammalian autophagy. Nat Rev Mol Cell Biol 19:349–364CrossRef Dikic I, Elazar Z (2018) Mechanism and medical implications of mammalian autophagy. Nat Rev Mol Cell Biol 19:349–364CrossRef
31.
Zurück zum Zitat Larrue C, Heydt Q, Saland E, Boutzen H, Kaoma T, Sarry JE, Joffre C, Recher C (2019) Oncogenic kit mutations induce stat3-dependent autophagy to support cell proliferation in acute myeloid leukemia. Oncogenesis 8:39CrossRef Larrue C, Heydt Q, Saland E, Boutzen H, Kaoma T, Sarry JE, Joffre C, Recher C (2019) Oncogenic kit mutations induce stat3-dependent autophagy to support cell proliferation in acute myeloid leukemia. Oncogenesis 8:39CrossRef
32.
Zurück zum Zitat Yamada E, Bastie CC, Koga H, Wang Y, Cuervo AM, Pessin JE (2012) Mouse skeletal muscle fiber-type-specific macroautophagy and muscle wasting are regulated by a fyn/stat3/vps34 signaling pathway. Cell Rep 1:557–569CrossRef Yamada E, Bastie CC, Koga H, Wang Y, Cuervo AM, Pessin JE (2012) Mouse skeletal muscle fiber-type-specific macroautophagy and muscle wasting are regulated by a fyn/stat3/vps34 signaling pathway. Cell Rep 1:557–569CrossRef
Metadaten
Titel
Polydatin induces apoptosis and autophagy via STAT3 signaling in human osteosarcoma MG-63 cells
verfasst von
Chang-qing Jiang
Ling-ling Ma
Zhen-dong Lv
Fan Feng
Zhi Chen
Zu-De Liu
Publikationsdatum
28.03.2020
Verlag
Springer Singapore
Erschienen in
Journal of Natural Medicines / Ausgabe 3/2020
Print ISSN: 1340-3443
Elektronische ISSN: 1861-0293
DOI
https://doi.org/10.1007/s11418-020-01399-5

Weitere Artikel der Ausgabe 3/2020

Journal of Natural Medicines 3/2020 Zur Ausgabe