Skip to main content
Top
Published in: Tumor Biology 4/2015

01-04-2015 | Research Article

Lycorine induces programmed necrosis in the multiple myeloma cell line ARH-77

Authors: Yuhao Luo, Mridul Roy, Xiaojuan Xiao, Shuming Sun, Long Liang, Huiyong Chen, Yin Fu, Yang Sun, Min Zhu, Mao Ye, Jing Liu

Published in: Tumor Biology | Issue 4/2015

Login to get access

Abstract

Lycorine, a natural alkaloid, has been widely reported to possess potential efficacy against cancer. However, the anti-multiple myeloma mechanism of lycorine is not fully understood. In this study, the results demonstrated that lycorine is effective against multiple myeloma cell line ARH-77 via inducing programmed necrosis. The mechanisms of lycorine on the multiple myeloma cell line ARH-77 are associated with G1 phase cell cycle arrest, mitochondrial dysfunction, reactive oxygen species (ROS) generation, ATP depletion, and DNA damage. Our results elucidate the new mechanism of lycorine against multiple myeloma.
Literature
1.
go back to reference Lozano R, Naghavi M, Foreman K, Lim S, Shibuya K, Aboyans V, et al. Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet. 2012;380(9859):2095–128.CrossRefPubMed Lozano R, Naghavi M, Foreman K, Lim S, Shibuya K, Aboyans V, et al. Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet. 2012;380(9859):2095–128.CrossRefPubMed
2.
go back to reference Chanan-Khan AA, San Miguel JF, Jagannath S, Ludwig H, Dimopoulos MA. Novel therapeutic agents for the management of patients with multiple myeloma and renal impairment. Clin Cancer Res. 2012;18(8):2145–63.CrossRefPubMed Chanan-Khan AA, San Miguel JF, Jagannath S, Ludwig H, Dimopoulos MA. Novel therapeutic agents for the management of patients with multiple myeloma and renal impairment. Clin Cancer Res. 2012;18(8):2145–63.CrossRefPubMed
3.
go back to reference Giralt S. Stem cell transplantation for multiple myeloma: current and future status. Hematology. 2012;17 Suppl 1:S117–20.PubMed Giralt S. Stem cell transplantation for multiple myeloma: current and future status. Hematology. 2012;17 Suppl 1:S117–20.PubMed
4.
go back to reference Mariz JM, Esteves GV. Review of therapy for relapsed/refractory multiple myeloma: focus on lenalidomide. Curr Opin Oncol. 2012;24(2):S3–11.CrossRefPubMed Mariz JM, Esteves GV. Review of therapy for relapsed/refractory multiple myeloma: focus on lenalidomide. Curr Opin Oncol. 2012;24(2):S3–11.CrossRefPubMed
6.
go back to reference Markert CL. Neoplasia: a disease of cell differentiation. Cancer Res. 1968;28(9):1908–14.PubMed Markert CL. Neoplasia: a disease of cell differentiation. Cancer Res. 1968;28(9):1908–14.PubMed
7.
go back to reference Panaretakis T, Pokrovskaja K, Shoshan MC, Grander D. Interferon-alpha-induced apoptosis in U266 cells is associated with activation of the proapoptotic Bcl-2 family members bak and bax. Oncogene. 2003;22(29):4543–56.CrossRefPubMed Panaretakis T, Pokrovskaja K, Shoshan MC, Grander D. Interferon-alpha-induced apoptosis in U266 cells is associated with activation of the proapoptotic Bcl-2 family members bak and bax. Oncogene. 2003;22(29):4543–56.CrossRefPubMed
8.
go back to reference Stromberg T, Dimberg A, Hammarberg A, Carlson K, Osterborg A, Nilsson K, et al. Rapamycin sensitizes multiple myeloma cells to apoptosis induced by dexamethasone. Blood. 2004;103(8):3138–47.CrossRefPubMed Stromberg T, Dimberg A, Hammarberg A, Carlson K, Osterborg A, Nilsson K, et al. Rapamycin sensitizes multiple myeloma cells to apoptosis induced by dexamethasone. Blood. 2004;103(8):3138–47.CrossRefPubMed
9.
go back to reference Khan SB, Maududi T, Barton K, Ayers J, Alkan S. Analysis of histone deacetylase inhibitor, depsipeptide (FR901228), effect on multiple myeloma. Br J Haematol. 2004;125(2):156–61.CrossRefPubMed Khan SB, Maududi T, Barton K, Ayers J, Alkan S. Analysis of histone deacetylase inhibitor, depsipeptide (FR901228), effect on multiple myeloma. Br J Haematol. 2004;125(2):156–61.CrossRefPubMed
10.
go back to reference Park WH, Seol JG, Kim ES, Hyun JM, Jung CW, Lee CC, et al. Arsenic trioxide-mediated growth inhibition in MC/CAR myeloma cells via cell cycle arrest in association with induction of cyclin-dependent kinase inhibitor, p21, and apoptosis. Cancer Res. 2000;60(11):3065–71.PubMed Park WH, Seol JG, Kim ES, Hyun JM, Jung CW, Lee CC, et al. Arsenic trioxide-mediated growth inhibition in MC/CAR myeloma cells via cell cycle arrest in association with induction of cyclin-dependent kinase inhibitor, p21, and apoptosis. Cancer Res. 2000;60(11):3065–71.PubMed
11.
go back to reference Liu Q, Hilsenbeck S, Gazitt Y. Arsenic trioxide-induced apoptosis in myeloma cells: p53-dependent G1 or G2/M cell cycle arrest, activation of caspase-8 or caspase-9, and synergy with APO2/TRAIL. Blood. 2003;101(10):4078–87.CrossRefPubMed Liu Q, Hilsenbeck S, Gazitt Y. Arsenic trioxide-induced apoptosis in myeloma cells: p53-dependent G1 or G2/M cell cycle arrest, activation of caspase-8 or caspase-9, and synergy with APO2/TRAIL. Blood. 2003;101(10):4078–87.CrossRefPubMed
12.
go back to reference Gottesman MM. How cancer cells evade chemotherapy: sixteenth Richard and Hinda Rosenthal Foundation award lecture. Cancer Res. 1993;53(4):747–54.PubMed Gottesman MM. How cancer cells evade chemotherapy: sixteenth Richard and Hinda Rosenthal Foundation award lecture. Cancer Res. 1993;53(4):747–54.PubMed
15.
go back to reference Wu M, Jiang Z, Duan H, Sun L, Zhang S, Chen M, et al. Deoxypodophyllotoxin triggers necroptosis in human non-small cell lung cancer NCI-H460 cells. Biomed Pharmacother. 2013;67(8):701–6.CrossRefPubMed Wu M, Jiang Z, Duan H, Sun L, Zhang S, Chen M, et al. Deoxypodophyllotoxin triggers necroptosis in human non-small cell lung cancer NCI-H460 cells. Biomed Pharmacother. 2013;67(8):701–6.CrossRefPubMed
16.
go back to reference Fu Z, Deng B, Liao Y, Shan L, Yin F, Wang Z, et al. The anti-tumor effect of shikonin on osteosarcoma by inducing RIP1 and RIP3 dependent necroptosis. BMC Cancer. 2013;13(580):1471–2407. Fu Z, Deng B, Liao Y, Shan L, Yin F, Wang Z, et al. The anti-tumor effect of shikonin on osteosarcoma by inducing RIP1 and RIP3 dependent necroptosis. BMC Cancer. 2013;13(580):1471–2407.
17.
go back to reference Basit F, Cristofanon S, Fulda S. Obatoclax (GX15-070) triggers necroptosis by promoting the assembly of the necrosome on autophagosomal membranes. Cell Death Differ. 2013;20(9):1161–73.CrossRefPubMedPubMedCentral Basit F, Cristofanon S, Fulda S. Obatoclax (GX15-070) triggers necroptosis by promoting the assembly of the necrosome on autophagosomal membranes. Cell Death Differ. 2013;20(9):1161–73.CrossRefPubMedPubMedCentral
18.
go back to reference Pasupuleti N, Leon L, Carraway 3rd KL, Gorin F. 5-Benzylglycinyl-amiloride kills proliferating and nonproliferating malignant glioma cells through caspase-independent necroptosis mediated by apoptosis-inducing factor. J Pharmacol Exp Ther. 2013;344(3):600–15.CrossRefPubMedPubMedCentral Pasupuleti N, Leon L, Carraway 3rd KL, Gorin F. 5-Benzylglycinyl-amiloride kills proliferating and nonproliferating malignant glioma cells through caspase-independent necroptosis mediated by apoptosis-inducing factor. J Pharmacol Exp Ther. 2013;344(3):600–15.CrossRefPubMedPubMedCentral
19.
go back to reference Park EJ, Min KJ, Lee TJ, Yoo YH, Kim YS. Kwon TK: beta-Lapachone induces programmed necrosis through the RIP1-PARP-AIF-dependent pathway in human hepatocellular carcinoma SK-Hep1 cells. Cell Death Dis. 2014;15(5):202. Park EJ, Min KJ, Lee TJ, Yoo YH, Kim YS. Kwon TK: beta-Lapachone induces programmed necrosis through the RIP1-PARP-AIF-dependent pathway in human hepatocellular carcinoma SK-Hep1 cells. Cell Death Dis. 2014;15(5):202.
20.
go back to reference Amaravadi RK, Thompson CB. The roles of therapy-induced autophagy and necrosis in cancer treatment. Clin Cancer Res. 2007;13(24):7271–9.CrossRefPubMed Amaravadi RK, Thompson CB. The roles of therapy-induced autophagy and necrosis in cancer treatment. Clin Cancer Res. 2007;13(24):7271–9.CrossRefPubMed
21.
go back to reference Zong WX, Ditsworth D, Bauer DE, Wang ZQ, Thompson CB. Alkylating DNA damage stimulates a regulated form of necrotic cell death. Genes Dev. 2004;18(11):1272–82.CrossRefPubMedPubMedCentral Zong WX, Ditsworth D, Bauer DE, Wang ZQ, Thompson CB. Alkylating DNA damage stimulates a regulated form of necrotic cell death. Genes Dev. 2004;18(11):1272–82.CrossRefPubMedPubMedCentral
22.
go back to reference Artal-Sanz M, Tavernarakis N. Proteolytic mechanisms in necrotic cell death and neurodegeneration. FEBS Lett. 2005;579(15):3287–96.CrossRefPubMed Artal-Sanz M, Tavernarakis N. Proteolytic mechanisms in necrotic cell death and neurodegeneration. FEBS Lett. 2005;579(15):3287–96.CrossRefPubMed
23.
24.
go back to reference Degterev A, Huang Z, Boyce M, Li Y, Jagtap P, Mizushima N, et al. Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat Chem Biol. 2005;1(2):112–9.CrossRefPubMed Degterev A, Huang Z, Boyce M, Li Y, Jagtap P, Mizushima N, et al. Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat Chem Biol. 2005;1(2):112–9.CrossRefPubMed
25.
go back to reference Degterev A, Hitomi J, Germscheid M, Ch’en IL, Korkina O, Teng X, et al. Identification of RIP1 kinase as a specific cellular target of necrostatins. Nat Chem Biol. 2008;4(5):313–21.CrossRefPubMed Degterev A, Hitomi J, Germscheid M, Ch’en IL, Korkina O, Teng X, et al. Identification of RIP1 kinase as a specific cellular target of necrostatins. Nat Chem Biol. 2008;4(5):313–21.CrossRefPubMed
26.
go back to reference He S, Wang L, Miao L, Wang T, Du F, Zhao L, et al. Receptor interacting protein kinase-3 determines cellular necrotic response to TNF-alpha. Cell. 2009;137(6):1100–11.CrossRefPubMed He S, Wang L, Miao L, Wang T, Du F, Zhao L, et al. Receptor interacting protein kinase-3 determines cellular necrotic response to TNF-alpha. Cell. 2009;137(6):1100–11.CrossRefPubMed
27.
go back to reference Zhang DW, Shao J, Lin J, Zhang N, Lu BJ, Lin SC, et al. RIP3, an energy metabolism regulator that switches TNF-induced cell death from apoptosis to necrosis. Science. 2009;325(5938):332–6.CrossRefPubMed Zhang DW, Shao J, Lin J, Zhang N, Lu BJ, Lin SC, et al. RIP3, an energy metabolism regulator that switches TNF-induced cell death from apoptosis to necrosis. Science. 2009;325(5938):332–6.CrossRefPubMed
28.
go back to reference Hu X, Han W, Li L. Targeting the weak point of cancer by induction of necroptosis. Autophagy. 2007;3(5):490–2.CrossRefPubMed Hu X, Han W, Li L. Targeting the weak point of cancer by induction of necroptosis. Autophagy. 2007;3(5):490–2.CrossRefPubMed
29.
go back to reference Mitra K, Wunder C, Roysam B, Lin G, Lippincott-Schwartz J. A hyperfused mitochondrial state achieved at G1-S regulates cyclin E buildup and entry into S phase. Proc Natl Acad Sci U S A. 2009;106(29):11960–5.CrossRefPubMedPubMedCentral Mitra K, Wunder C, Roysam B, Lin G, Lippincott-Schwartz J. A hyperfused mitochondrial state achieved at G1-S regulates cyclin E buildup and entry into S phase. Proc Natl Acad Sci U S A. 2009;106(29):11960–5.CrossRefPubMedPubMedCentral
30.
go back to reference Tennant DA, Duran RV, Boulahbel H, Gottlieb E. Metabolic transformation in cancer. Carcinogenesis. 2009;30(8):1269–80.CrossRefPubMed Tennant DA, Duran RV, Boulahbel H, Gottlieb E. Metabolic transformation in cancer. Carcinogenesis. 2009;30(8):1269–80.CrossRefPubMed
31.
go back to reference Gomes LC, Di Benedetto G, Scorrano L. During autophagy mitochondria elongate, are spared from degradation and sustain cell viability. Nat Cell Biol. 2011;13(5):589–98.CrossRefPubMedPubMedCentral Gomes LC, Di Benedetto G, Scorrano L. During autophagy mitochondria elongate, are spared from degradation and sustain cell viability. Nat Cell Biol. 2011;13(5):589–98.CrossRefPubMedPubMedCentral
32.
go back to reference Priault M, Salin B, Schaeffer J, Vallette FM, di Rago JP, Martinou JC. Impairing the bioenergetic status and the biogenesis of mitochondria triggers mitophagy in yeast. Cell Death Differ. 2005;12(12):1613–21.CrossRefPubMed Priault M, Salin B, Schaeffer J, Vallette FM, di Rago JP, Martinou JC. Impairing the bioenergetic status and the biogenesis of mitochondria triggers mitophagy in yeast. Cell Death Differ. 2005;12(12):1613–21.CrossRefPubMed
33.
go back to reference Zhang DX, Gutterman DD. Mitochondrial reactive oxygen species-mediated signaling in endothelial cells. Am J Physiol Heart Circ Physiol. 2007;292(5):19. Zhang DX, Gutterman DD. Mitochondrial reactive oxygen species-mediated signaling in endothelial cells. Am J Physiol Heart Circ Physiol. 2007;292(5):19.
34.
go back to reference Moungjaroen J, Nimmannit U, Callery PS, Wang L, Azad N, Lipipun V, et al. Reactive oxygen species mediate caspase activation and apoptosis induced by lipoic acid in human lung epithelial cancer cells through Bcl-2 down-regulation. J Pharmacol Exp Ther. 2006;319(3):1062–9.CrossRefPubMed Moungjaroen J, Nimmannit U, Callery PS, Wang L, Azad N, Lipipun V, et al. Reactive oxygen species mediate caspase activation and apoptosis induced by lipoic acid in human lung epithelial cancer cells through Bcl-2 down-regulation. J Pharmacol Exp Ther. 2006;319(3):1062–9.CrossRefPubMed
35.
go back to reference Schumacker PT. Reactive oxygen species in cancer cells: live by the sword, die by the sword. Cancer Cell. 2006;10(3):175–6.CrossRefPubMed Schumacker PT. Reactive oxygen species in cancer cells: live by the sword, die by the sword. Cancer Cell. 2006;10(3):175–6.CrossRefPubMed
36.
go back to reference Ott M, Gogvadze V, Orrenius S, Zhivotovsky B. Mitochondria, oxidative stress and cell death. Apoptosis. 2007;12(5):913–22.CrossRefPubMed Ott M, Gogvadze V, Orrenius S, Zhivotovsky B. Mitochondria, oxidative stress and cell death. Apoptosis. 2007;12(5):913–22.CrossRefPubMed
37.
go back to reference Tu HC, Ren D, Wang GX, Chen DY, Westergard TD, Kim H, et al. The p53-cathepsin axis cooperates with ROS to activate programmed necrotic death upon DNA damage. Proc Natl Acad Sci U S A. 2009;106(4):1093–8.CrossRefPubMedPubMedCentral Tu HC, Ren D, Wang GX, Chen DY, Westergard TD, Kim H, et al. The p53-cathepsin axis cooperates with ROS to activate programmed necrotic death upon DNA damage. Proc Natl Acad Sci U S A. 2009;106(4):1093–8.CrossRefPubMedPubMedCentral
38.
39.
go back to reference Cheng WH, Wu RT, Wu M, Rocourt CR, Carrillo JA, Song J, et al. Targeting Werner syndrome protein sensitizes U-2 OS osteosarcoma cells to selenium-induced DNA damage response and necrotic death. Biochem Biophys Res Commun. 2012;420(1):24–8.CrossRefPubMed Cheng WH, Wu RT, Wu M, Rocourt CR, Carrillo JA, Song J, et al. Targeting Werner syndrome protein sensitizes U-2 OS osteosarcoma cells to selenium-induced DNA damage response and necrotic death. Biochem Biophys Res Commun. 2012;420(1):24–8.CrossRefPubMed
41.
go back to reference Srivastava DK, Berg BJ, Prasad R, Molina JT, Beard WA, Tomkinson AE, et al. Mammalian abasic site base excision repair. Identification of the reaction sequence and rate-determining steps. J Biol Chem. 1998;273(33):21203–9.CrossRefPubMed Srivastava DK, Berg BJ, Prasad R, Molina JT, Beard WA, Tomkinson AE, et al. Mammalian abasic site base excision repair. Identification of the reaction sequence and rate-determining steps. J Biol Chem. 1998;273(33):21203–9.CrossRefPubMed
42.
go back to reference Liu Y, Prasad R, Beard WA, Kedar PS, Hou EW, Shock DD, et al. Coordination of steps in single-nucleotide base excision repair mediated by apurinic/apyrimidinic endonuclease 1 and DNA polymerase beta. J Biol Chem. 2007;282(18):13532–41.CrossRefPubMedPubMedCentral Liu Y, Prasad R, Beard WA, Kedar PS, Hou EW, Shock DD, et al. Coordination of steps in single-nucleotide base excision repair mediated by apurinic/apyrimidinic endonuclease 1 and DNA polymerase beta. J Biol Chem. 2007;282(18):13532–41.CrossRefPubMedPubMedCentral
43.
go back to reference Matsumoto Y, Kim K. Excision of deoxyribose phosphate residues by DNA polymerase beta during DNA repair. Science. 1995;269(5224):699–702.CrossRefPubMed Matsumoto Y, Kim K. Excision of deoxyribose phosphate residues by DNA polymerase beta during DNA repair. Science. 1995;269(5224):699–702.CrossRefPubMed
44.
go back to reference Chaitanya GV, Steven AJ, Babu PP. PARP-1 cleavage fragments: signatures of cell-death proteases in neurodegeneration. Cell Commun Signal. 2010;8(31):8–31. Chaitanya GV, Steven AJ, Babu PP. PARP-1 cleavage fragments: signatures of cell-death proteases in neurodegeneration. Cell Commun Signal. 2010;8(31):8–31.
45.
46.
go back to reference Liu XS, Jiang J, Jiao XY, Wu YE, Lin JH, Cai YM. Lycorine induces apoptosis and down-regulation of Mcl-1 in human leukemia cells. Cancer Lett. 2009;274(1):16–24.CrossRefPubMed Liu XS, Jiang J, Jiao XY, Wu YE, Lin JH, Cai YM. Lycorine induces apoptosis and down-regulation of Mcl-1 in human leukemia cells. Cancer Lett. 2009;274(1):16–24.CrossRefPubMed
47.
go back to reference Lamoral-Theys D, Andolfi A, Van Goietsenoven G, Cimmino A, Le Calve B, Wauthoz N, et al. Lycorine, the main phenanthridine amaryllidaceae alkaloid, exhibits significant antitumor activity in cancer cells that display resistance to proapoptotic stimuli: an investigation of structure-activity relationship and mechanistic insight. J Med Chem. 2009;52(20):6244–56.CrossRefPubMedPubMedCentral Lamoral-Theys D, Andolfi A, Van Goietsenoven G, Cimmino A, Le Calve B, Wauthoz N, et al. Lycorine, the main phenanthridine amaryllidaceae alkaloid, exhibits significant antitumor activity in cancer cells that display resistance to proapoptotic stimuli: an investigation of structure-activity relationship and mechanistic insight. J Med Chem. 2009;52(20):6244–56.CrossRefPubMedPubMedCentral
48.
go back to reference Li Y, Liu J, Tang LJ, Shi YW, Ren W, Hu WX. Apoptosis induced by lycorine in KM3 cells is associated with the G0/G1 cell cycle arrest. Oncol Rep. 2007;17(2):377–84.PubMed Li Y, Liu J, Tang LJ, Shi YW, Ren W, Hu WX. Apoptosis induced by lycorine in KM3 cells is associated with the G0/G1 cell cycle arrest. Oncol Rep. 2007;17(2):377–84.PubMed
49.
go back to reference Liu J, Hu JL, Shi BW, He Y, Hu WX. Up-regulation of p21 and TNF-alpha is mediated in lycorine-induced death of HL-60 cells. Cancer Cell Int. 2010;10(25):1475–2867. Liu J, Hu JL, Shi BW, He Y, Hu WX. Up-regulation of p21 and TNF-alpha is mediated in lycorine-induced death of HL-60 cells. Cancer Cell Int. 2010;10(25):1475–2867.
50.
go back to reference Li L, Dai HJ, Ye M, Wang SL, Xiao XJ, Zheng J, et al. Lycorine induces cell-cycle arrest in the G0/G1 phase in K562 cells via HDAC inhibition. Cancer Cell Int. 2012;12(1):1475–2867. Li L, Dai HJ, Ye M, Wang SL, Xiao XJ, Zheng J, et al. Lycorine induces cell-cycle arrest in the G0/G1 phase in K562 cells via HDAC inhibition. Cancer Cell Int. 2012;12(1):1475–2867.
51.
go back to reference Liu J, Li Y, Tang LJ, Zhang GP, Hu WX. Treatment of lycorine on SCID mice model with human APL cells. Biomed Pharmacother. 2007;61(4):229–34.CrossRefPubMed Liu J, Li Y, Tang LJ, Zhang GP, Hu WX. Treatment of lycorine on SCID mice model with human APL cells. Biomed Pharmacother. 2007;61(4):229–34.CrossRefPubMed
52.
go back to reference Gartel AL, Radhakrishnan SK. Lost in transcription: p21 repression, mechanisms, and consequences. Cancer Res. 2005;65(10):3980–5.CrossRefPubMed Gartel AL, Radhakrishnan SK. Lost in transcription: p21 repression, mechanisms, and consequences. Cancer Res. 2005;65(10):3980–5.CrossRefPubMed
53.
go back to reference Satyanarayana A, Kaldis P. Mammalian cell-cycle regulation: several Cdks, numerous cyclins and diverse compensatory mechanisms. Oncogene. 2009;28(33):2925–39.CrossRefPubMed Satyanarayana A, Kaldis P. Mammalian cell-cycle regulation: several Cdks, numerous cyclins and diverse compensatory mechanisms. Oncogene. 2009;28(33):2925–39.CrossRefPubMed
54.
go back to reference Liu J, Hu WX, He LF, Ye M, Li Y. Effects of lycorine on HL-60 cells via arresting cell cycle and inducing apoptosis. FEBS Lett. 2004;578(3):245–50.CrossRefPubMed Liu J, Hu WX, He LF, Ye M, Li Y. Effects of lycorine on HL-60 cells via arresting cell cycle and inducing apoptosis. FEBS Lett. 2004;578(3):245–50.CrossRefPubMed
55.
go back to reference Rasul A, Di J, Millimouno FM, Malhi M, Tsuji I, Ali M, et al. Reactive oxygen species mediate isoalantolactone-induced apoptosis in human prostate cancer cells. Molecules. 2013;18(8):9382–96.CrossRefPubMed Rasul A, Di J, Millimouno FM, Malhi M, Tsuji I, Ali M, et al. Reactive oxygen species mediate isoalantolactone-induced apoptosis in human prostate cancer cells. Molecules. 2013;18(8):9382–96.CrossRefPubMed
56.
go back to reference Tan C, Qian X, Jia R, Wu M, Liang Z. Matrine induction of reactive oxygen species activates p38 leading to caspase-dependent cell apoptosis in non-small cell lung cancer cells. Oncol Rep. 2013;30(5):2529–35.PubMed Tan C, Qian X, Jia R, Wu M, Liang Z. Matrine induction of reactive oxygen species activates p38 leading to caspase-dependent cell apoptosis in non-small cell lung cancer cells. Oncol Rep. 2013;30(5):2529–35.PubMed
57.
go back to reference Yang JT, Li ZL, Wu JY, Lu FJ, Chen CH. An oxidative stress mechanism of shikonin in human glioma cells. PLoS One. 2014;9(4). Yang JT, Li ZL, Wu JY, Lu FJ, Chen CH. An oxidative stress mechanism of shikonin in human glioma cells. PLoS One. 2014;9(4).
58.
go back to reference Trachootham D, Alexandre J, Huang P. Targeting cancer cells by ROS-mediated mechanisms: a radical therapeutic approach? Nat Rev Drug Discov. 2009;8(7):579–91.CrossRefPubMed Trachootham D, Alexandre J, Huang P. Targeting cancer cells by ROS-mediated mechanisms: a radical therapeutic approach? Nat Rev Drug Discov. 2009;8(7):579–91.CrossRefPubMed
59.
go back to reference Maillet A, Yadav S, Loo YL, Sachaphibulkij K, Pervaiz S. A novel Osmium-based compound targets the mitochondria and triggers ROS-dependent apoptosis in colon carcinoma. Cell Death Dis. 2013;6(4):185. Maillet A, Yadav S, Loo YL, Sachaphibulkij K, Pervaiz S. A novel Osmium-based compound targets the mitochondria and triggers ROS-dependent apoptosis in colon carcinoma. Cell Death Dis. 2013;6(4):185.
60.
go back to reference Bey EA, Bentle MS, Reinicke KE, Dong Y, Yang CR, Girard L, et al. An NQO1- and PARP-1-mediated cell death pathway induced in non-small-cell lung cancer cells by beta-lapachone. Proc Natl Acad Sci U S A. 2007;104(28):11832–7.CrossRefPubMedPubMedCentral Bey EA, Bentle MS, Reinicke KE, Dong Y, Yang CR, Girard L, et al. An NQO1- and PARP-1-mediated cell death pathway induced in non-small-cell lung cancer cells by beta-lapachone. Proc Natl Acad Sci U S A. 2007;104(28):11832–7.CrossRefPubMedPubMedCentral
61.
go back to reference Goldin N, Heyfets A, Reischer D, Flescher E. Mitochondria-mediated ATP depletion by anti-cancer agents of the jasmonate family. J Bioenerg Biomembr. 2007;39(1):51–7.CrossRefPubMed Goldin N, Heyfets A, Reischer D, Flescher E. Mitochondria-mediated ATP depletion by anti-cancer agents of the jasmonate family. J Bioenerg Biomembr. 2007;39(1):51–7.CrossRefPubMed
64.
go back to reference Temkin V, Huang Q, Liu H, Osada H, Pope RM. Inhibition of ADP/ATP exchange in receptor-interacting protein-mediated necrosis. Mol Cell Biol. 2006;26(6):2215–25.CrossRefPubMedPubMedCentral Temkin V, Huang Q, Liu H, Osada H, Pope RM. Inhibition of ADP/ATP exchange in receptor-interacting protein-mediated necrosis. Mol Cell Biol. 2006;26(6):2215–25.CrossRefPubMedPubMedCentral
65.
go back to reference Ouyang Z, Zhu S, Jin J, Li J, Qiu Y, Huang M, et al. Necroptosis contributes to the cyclosporin A-induced cytotoxicity in NRK-52E cells. Pharmazie. 2012;67(8):725–32.PubMed Ouyang Z, Zhu S, Jin J, Li J, Qiu Y, Huang M, et al. Necroptosis contributes to the cyclosporin A-induced cytotoxicity in NRK-52E cells. Pharmazie. 2012;67(8):725–32.PubMed
66.
go back to reference Jog NR, Caricchio R. Differential regulation of cell death programs in males and females by poly(ADP-ribose) polymerase-1 and 17beta estradiol. Cell Death Dis. 2013;8(4):251. Jog NR, Caricchio R. Differential regulation of cell death programs in males and females by poly(ADP-ribose) polymerase-1 and 17beta estradiol. Cell Death Dis. 2013;8(4):251.
Metadata
Title
Lycorine induces programmed necrosis in the multiple myeloma cell line ARH-77
Authors
Yuhao Luo
Mridul Roy
Xiaojuan Xiao
Shuming Sun
Long Liang
Huiyong Chen
Yin Fu
Yang Sun
Min Zhu
Mao Ye
Jing Liu
Publication date
01-04-2015
Publisher
Springer Netherlands
Published in
Tumor Biology / Issue 4/2015
Print ISSN: 1010-4283
Electronic ISSN: 1423-0380
DOI
https://doi.org/10.1007/s13277-014-2924-7

Other articles of this Issue 4/2015

Tumor Biology 4/2015 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