Volume 19, Issue 12, Pages (December 2017)

Slides:



Advertisements
Similar presentations
* Supplementary Figure 1: Treatment of siramesine with various chemotherapeutic drugs in MDA MB 231 cells. MDA MB 231 cells were treated with 10μM siramesine.
Advertisements

Nicotinamide Phosphoribosyltransferase: A Potent Therapeutic Target in Non-small Cell Lung Cancer with Epidermal Growth Factor Receptor-Gene Mutation 
Aldehyde Dehydrogenase 1A1 Possesses Stem-Like Properties and Predicts Lung Cancer Patient Outcome  Xiao Li, MD, Liyan Wan, MD, Jian Geng, MD, Chin-Lee.
MicroRNA-101 Inhibits Growth, Proliferation and Migration and Induces Apoptosis of Breast Cancer Cells by Targeting Sex-Determining Region Y-Box 2 Cell.
Upregulation of miR-142-3p Improves Drug Sensitivity of Acute Myelogenous Leukemia through Reducing P-Glycoprotein and Repressing Autophagy by Targeting.
Volume 144, Issue 3, Pages e4 (March 2013)
A Signal Transduction Pathway from TGF-β1 to SKP2 via Akt1 and c-Myc and its Correlation with Progression in Human Melanoma  Xuan Qu, Liangliang Shen,
Volume 67, Issue 2, Pages (August 2017)
Integrin αvβ6 Promotes Lung Cancer Proliferation and Metastasis through Upregulation of IL-8–Mediated MAPK/ERK Signaling  Pengwei Yan, Huanfeng Zhu, Li.
DNMT3B Overexpression by Deregulation of FOXO3a-Mediated Transcription Repression and MDM2 Overexpression in Lung Cancer  Yi-Chieh Yang, MS, Yen-An Tang,
Deregulation of SLIT2-Mediated Cdc42 Activity Is Associated with Esophageal Cancer Metastasis and Poor Prognosis  Ruo-Chia Tseng, PhD, Jia-Ming Chang,
Sp1 Suppresses miR-3178 to Promote the Metastasis Invasion Cascade via Upregulation of TRIOBP  Hui Wang, Kai Li, Yu Mei, Xuemei Huang, Zhenglin Li, Qingzhu.
Volume 85, Issue 2, Pages (January 2014)
Volume 19, Issue 7, Pages (July 2017)
SDHB deficiency promotes TGFβ-mediated invasion and metastasis of colorectal cancer through transcriptional repression complex SNAIL1-SMAD3/4  Haiyu Wang,
Volume 133, Issue 1, Pages (July 2007)
Overexpression of ornithine decarboxylase enhances endothelial proliferation by suppressing endostatin expression by Takahiro Nemoto, Hisae Hori, Masataka.
IFN-γ Induces Gastric Cancer Cell Proliferation and Metastasis Through Upregulation of Integrin β3-Mediated NF-κB Signaling  Yuan-Hua Xu, Zheng-Li Li,
MicroRNA-489 Plays an Anti-Metastatic Role in Human Hepatocellular Carcinoma by Targeting Matrix Metalloproteinase-7  Yixiong Lin, Jianjun Liu, Yuqi Huang,
Therapeutic Suppression of miR-4261 Attenuates Colorectal Cancer by Targeting MCC  Guanming Jiao, Qi Huang, Muren Hu, Xuchun Liang, Fuchen Li, Chunling.
Hydrogen Sulfide Demonstrates Promising Antitumor Efficacy in Gastric Carcinoma by Targeting MGAT5  Rui Wang, Qilin Fan, Junjie Zhang, Xunan Zhang, Yuqi.
Volume 132, Issue 3, Pages (March 2007)
Volume 85, Issue 2, Pages (January 2014)
Volume 21, Issue 5, Pages (October 2017)
Volume 138, Issue 2, Pages (February 2010)
Volume 11, Issue 2, Pages (February 2007)
Aldehyde Dehydrogenase 1A1 Possesses Stem-Like Properties and Predicts Lung Cancer Patient Outcome  Xiao Li, MD, Liyan Wan, MD, Jian Geng, MD, Chin-Lee.
Antisense Oligonucleotides Targeting Y-Box Binding Protein-1 Inhibit Tumor Angiogenesis by Downregulating Bcl-xL-VEGFR2/-Tie Axes  Kiyoko Setoguchi, Lin.
Volume 28, Issue 4, Pages (October 2015)
Nicastrin/miR-30a-3p/RAB31 Axis Regulates Keratinocyte Differentiation by Impairing EGFR Signaling in Familial Acne Inversa  Yanyan He, Haoxiang Xu, Chengrang.
Volume 12, Issue 3, Pages (July 2015)
Volume 19, Issue 3, Pages (March 2017)
Volume 20, Issue 2, Pages (February 2018)
Volume 19, Issue 2, Pages (February 2011)
Uc.454 Inhibited Growth by Targeting Heat Shock Protein Family A Member 12B in Non- Small-Cell Lung Cancer  Jun Zhou, Chenghai Wang, Weijuan Gong, Yandan.
Molecular Therapy - Nucleic Acids
Volume 19, Issue 4, Pages (April 2017)
Molecular Therapy - Nucleic Acids
Volume 25, Issue 3, Pages (March 2017)
MicroRNA-101 Exerts Tumor-Suppressive Functions in Non-small Cell Lung Cancer through Directly Targeting Enhancer of Zeste Homolog 2  Ji-guang Zhang,
Volume 142, Issue 7, Pages e2 (June 2012)
Inhibition of KLF4 by Statins Reverses Adriamycin-Induced Metastasis and Cancer Stemness in Osteosarcoma Cells  Yangling Li, Miao Xian, Bo Yang, Meidan.
Volume 26, Issue 1, Pages (January 2018)
Volume 21, Issue 12, Pages (December 2017)
MiR-135b Stimulates Osteosarcoma Recurrence and Lung Metastasis via Notch and Wnt/β-Catenin Signaling  Hua Jin, Song Luo, Yun Wang, Chang Liu, Zhenghao.
miR-124 Inhibits Lung Tumorigenesis Induced by K-ras Mutation and NNK
Bo-Kuan Wu, Charles Brenner  Cell Reports 
Molecular Therapy - Nucleic Acids
Kun-Peng Zhu, Xiao-Long Ma, Chun-Lin Zhang  Molecular Therapy 
Volume 19, Issue 8, Pages (August 2011)
Volume 20, Issue 3, Pages (July 2017)
Volume 50, Issue 2, Pages (April 2013)
Volume 17, Issue 2, Pages (February 2009)
Volume 25, Issue 4, Pages (April 2017)
Shrimp miR-34 from Shrimp Stress Response to Virus Infection Suppresses Tumorigenesis of Breast Cancer  Yalei Cui, Xiaoyuan Yang, Xiaobo Zhang  Molecular.
An Epigenetic Switch Involving NF-κB, Lin28, Let-7 MicroRNA, and IL6 Links Inflammation to Cell Transformation  Dimitrios Iliopoulos, Heather A. Hirsch,
Volume 25, Issue 3, Pages (March 2017)
MELK Promotes Melanoma Growth by Stimulating the NF-κB Pathway
Negative Regulation of Tumor Suppressor p53 by MicroRNA miR-504
Volume 18, Issue 3, Pages (March 2010)
Molecular Therapy - Nucleic Acids
The Tumor Suppressor p53 Limits Ferroptosis by Blocking DPP4 Activity
MicroRNA-125b Promotes Hepatic Stellate Cell Activation and Liver Fibrosis by Activating RhoA Signaling  Kai You, Song-Yang Li, Jiao Gong, Jian-Hong Fang,
Volume 26, Issue 3, Pages (March 2018)
The Expression of MicroRNA-598 Inhibits Ovarian Cancer Cell Proliferation and Metastasis by Targeting URI  Feng Xing, Shuo Wang, Jianhong Zhou  Molecular.
Volume 26, Issue 10, Pages (October 2018)
Molecular Therapy - Nucleic Acids
Volume 23, Issue 4, Pages (April 2015)
Molecular Therapy - Nucleic Acids
Anticancer Activity of the Cholesterol Exporter ABCA1 Gene
Presentation transcript:

Volume 19, Issue 12, Pages 1022-1032 (December 2017) Cysteine Dioxygenase 1 Mediates Erastin-Induced Ferroptosis in Human Gastric Cancer Cells  Shihui Hao, Jiang Yu, Wanming He, Qiong Huang, Yang Zhao, Bishan Liang, Shuyi Zhang, Zhaowei Wen, Shumin Dong, Jinjun Rao, Wangjun Liao, Min Shi  Neoplasia  Volume 19, Issue 12, Pages 1022-1032 (December 2017) DOI: 10.1016/j.neo.2017.10.005 Copyright © 2017 The Authors Terms and Conditions

Figure 1 CDO1 suppression contributes to ferroptosis resistance. (A) GC cells were treated with 0, 2.5, 5, 10, 15, 20, or 40 μg/ml erastin for 24 hours. Cell viabilities were assayed by the MTT assay. (B) GPX4, solute carrier family 7 member 11(Slc7a11), and CDO1 expression were assessed by the qRT-PCR in GC cells. (C) GC cells were treated with erastin (E, 10 μg/ml) and with or without Z-VAD-FMK (apoptosis inhibitor), necrostatin (necroptosis inhibitor), 3-methyladenine (3-MA, autophagy inhibitor), and ferrostatin-1 or liproxstatin-1 (ferroptosis inhibitors) for 24 hours. Cell viability was assayed by the MTT assay. (D) GC cells were treated with or without erastin (10 μg/ml). LDH release assay was examined. (E) The siRNA sequences [CDO1 siRNA (2) and (3)] were designed for silencing CDO1. The mRNA levels of CDO1 in GC cells were examined by qRT-PCR. (F) The siRNA sequences [CDO1 siRNA (2) and (3)] were designed for silencing CDO1. The protein levels of CDO1 in GC cells were examined by Western blotting. (G) CDO1 siRNA (2)– and CDO1 siRNA (3)–silenced GC cells were treated with erastin (0, 2.5, 5, 10, 15, and 20 μg/ml) for 24 hours, and cell viabilities were assayed by the MTT assay. Quantitative data are presented as means ± SD from three independent experiments. ⁎P<.05, ⁎⁎P<.01, ⁎⁎⁎P<.001. Neoplasia 2017 19, 1022-1032DOI: (10.1016/j.neo.2017.10.005) Copyright © 2017 The Authors Terms and Conditions

Figure 2 CDO1 regulates ferroptosis biomarkers. CDO1-silenced GC cells were treated with 10 μg/ml erastin for 24 hours. (A) GSH, (B) ROS, and (C) MDA levels were assayed. CDO1 siRNA-silenced GC cells were treated with 10 μg/ml erastin for 24 hours. GPX4 expression was assessed by the qRT-PCR (D), Western blotting (E), and immunofluorescence (F). (G) Transmission electron microscopic images of CDO1 siRNA (2)– and CDO1 siRNA(3)–silenced AGS cells treated with 10 μg/ml erastin for 24 hours. Left, 1×104 magnification, scale bar=2 μm; right, 5×104 magnification, scale bar=500 nm. Quantitative data are presented as means ± SD from three independent experiments. ⁎P<.05, ⁎⁎P<.01, ⁎⁎⁎P<.001. Neoplasia 2017 19, 1022-1032DOI: (10.1016/j.neo.2017.10.005) Copyright © 2017 The Authors Terms and Conditions

Figure 3 C-Myb regulates CDO1 expression during ferroptosis. (A) GC cells were treated with erastin (10 or 30 μg/ml) for 24 hours. Expression levels of CDO1, c-Myb, and GPX4 proteins were detected by Western blotting. (B) The siRNA sequences [c-Myb siRNA (1), (2), and (3)] were designed for silencing c-Myb. The mRNA levels of c-Myb in GC cells were examined by qRT-PCR. (C) The siRNA sequences [c-Myb siRNA (1), (2), and (3)] were designed for silencing c-Myb. The protein levels of c-Myb in GC cells were examined by Western blotting. (D) Cell viabilities were determined by the MTT assay after transfection with three different c-Myb siRNA sequences to silence c-Myb expression. (E) c-Myb–silenced GC cells were treated with erastin (5, 10, or 20 μg/ml) for 24 hours, and cell viabilities were detected by the MTT assay. C-Myb was overexpressed, and (F) mRNA and (G) protein expression levels of CDO1 and GPX4 were detected by the qRT-PCR and Western blotting, respectively (V, vector; M, overexpression). (H) Immunofluorescence staining of GPX4 was determined when c-Myb was overexpressed. (I) Cell viabilities were determined by the MTT assay after overexpression of c-Myb or inhibited expression of CDO1 in GC cells treated with erastin (10 μg/ml). (J) The interaction between CDO1 and c-Myb was verified by a luciferase reporter assay. (K) The chromatin immunoprecipitation assay shows that c-Myb has three positive binding sites with the CDO1 promoter. Quantitative data are presented as means ± SD from three independent experiments. ⁎P<.05, ⁎⁎P<.01, ⁎⁎⁎P<.001. Neoplasia 2017 19, 1022-1032DOI: (10.1016/j.neo.2017.10.005) Copyright © 2017 The Authors Terms and Conditions

Figure 4 Suppression of CDO1 inhibits ferroptosis in vivo. (A) Nude mice were injected subcutaneously with BGC823 cells (1×106 cells/mouse) and treated with erastin (20 mg/kg, intraperitoneally, twice every other day) starting at day 10 for 4 weeks. The tumor volume was calculated every 5 days (n=5). (B, C) Lung, liver, spleen, kidney, heart, and grafted subcutaneous tumors of nude mice were removed and stained with hematoxylin-eosin. Magnification: 100× (left panel), scale bar=100 μm; 400× (right panel), scale bar=25 μm. Expression of CDO1, GPX4, and PTGS2 (E) in subcutaneous tumors of nude mice using immunohistochemistry. Magnification: 100× (left panel); scale bar=100 μm; 200× (right panel), scale bar=50 μm; 400×(right panel), scale bar=25 μm. Neoplasia 2017 19, 1022-1032DOI: (10.1016/j.neo.2017.10.005) Copyright © 2017 The Authors Terms and Conditions

Figure 5 Diagram summarizing the role of CDO1 during erastin-induced ferroptosis. Erastin inhibits cellular cysteine uptake by suppressing system XC− and depleting GSH. This leads to inactivation of GPX4, increased ROS, and subsequently ferroptosis. CDO1, transcriptionally regulated by c-Myb, can transform cysteine to taurine and thereby competitively deprive cells of the cysteine used to synthesize GSH. When CDO1 expression is inhibited, the conversion of cysteine to taurine is decreased, enhancing GSH generation. Therefore, GPX4 activity is promoted, inhibiting ROS accumulation and lipid peroxidation and ultimately reducing the risk of ferroptosis. Neoplasia 2017 19, 1022-1032DOI: (10.1016/j.neo.2017.10.005) Copyright © 2017 The Authors Terms and Conditions

Supplementary Figure 1 Expression of CDO1 in GC and normal gastric tissues. (A) CDO1 mRNA expression levels of 16 paired patient specimens (C, cancer; N, normal). (B) CDO1 protein expression levels of four paired patient specimens (C, cancer; N, normal). (C) Viabilities of GC cells overexpressing CDO1 following treatment with 5, 10, or 20 μg/ml erastin for 24 hours (V, vector; M, overexpression). Quantitative data are presented as means ± SD from three independent experiments. ⁎P<.05, ⁎⁎P<.01, ⁎⁎⁎P<.001. Neoplasia 2017 19, 1022-1032DOI: (10.1016/j.neo.2017.10.005) Copyright © 2017 The Authors Terms and Conditions

Supplementary Figure 2 GPX4 mRNA levels by alteration of CDO1. Overexpressing CDO1-GC cells were treated with 10 μg/ml erastin for 24 hours (V, vector; M, overexpression). Quantitative data are presented as means ± SD from three independent experiments. ⁎P<.05, ⁎⁎P<.01, ⁎⁎⁎P<.001. Neoplasia 2017 19, 1022-1032DOI: (10.1016/j.neo.2017.10.005) Copyright © 2017 The Authors Terms and Conditions

Supplementary Figure 3 Immunohistochemistry of Ki-67 in tumors. Expression of Ki-67 in subcutaneous tumors of nude mice using immunohistochemistry. Magnification: 100× (left panel); scale bar=100 μm; 200× (right panel), scale bar=50 μm; 400×(right panel), scale bar=25 μm. Neoplasia 2017 19, 1022-1032DOI: (10.1016/j.neo.2017.10.005) Copyright © 2017 The Authors Terms and Conditions