Skip to main content
Drug Design, Development and Therapy logoLink to Drug Design, Development and Therapy
. 2015 Oct 5;9:5511–5551. doi: 10.2147/DDDT.S89621

Plumbagin suppresses epithelial to mesenchymal transition and stemness via inhibiting Nrf2-mediated signaling pathway in human tongue squamous cell carcinoma cells

Shu-Ting Pan 1, Yiru Qin 2, Zhi-Wei Zhou 2,3, Zhi-Xu He 3, Xueji Zhang 4, Tianxin Yang 5, Yin-Xue Yang 6, Dong Wang 7, Shu-Feng Zhou 2,, Jia-Xuan Qiu 1,
PMCID: PMC4599573  PMID: 26491260

Abstract

Tongue squamous cell carcinoma (TSCC) is the most common malignancy in oral and maxillofacial tumors with highly metastatic characteristics. Plumbagin (5-hydroxy-2-methyl-1, 4-naphthoquinone; PLB), a natural naphthoquinone derived from the roots of Plumbaginaceae plants, exhibits various bioactivities, including anticancer effects. However, the potential molecular targets and underlying mechanisms of PLB in the treatment of TSCC remain elusive. This study employed stable isotope labeling by amino acids in cell culture (SILAC)-based quantitative proteomic approach to investigate the molecular interactome of PLB in human TSCC cell line SCC25 and elucidate the molecular mechanisms. The proteomic data indicated that PLB inhibited cell proliferation, activated death receptor-mediated apoptotic pathway, remodeled epithelial adherens junctions pathway, and manipulated nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated oxidative stress response signaling pathway in SCC25 cells with the involvement of a number of key functional proteins. Furthermore, we verified these protein targets using Western blotting assay. The verification results showed that PLB markedly induced cell cycle arrest at G2/M phase and extrinsic apoptosis, and inhibited epithelial to mesenchymal transition (EMT) and stemness in SCC25 cells. Of note, N-acetyl-l-cysteine (NAC) and l-glutathione (GSH) abolished the effects of PLB on cell cycle arrest, apoptosis induction, EMT inhibition, and stemness attenuation in SCC25 cells. Importantly, PLB suppressed the translocation of Nrf2 from cytosol to nucleus, resulting in an inhibition in the expression of downstream targets. Taken together, these results suggest that PLB may act as a promising anticancer compound via inhibiting Nrf2-mediated oxidative stress signaling pathway in SCC25 cells. This study provides a clue to fully identify the molecular targets and decipher the underlying mechanisms of PLB in the treatment of TSCC.

Keywords: PLB, SILAC, EMT, stemness, Nrf2, tongue squamous cell carcinoma

Introduction

Tongue squamous cell carcinoma (TSCC) is the most prevalent type of oral and maxillofacial tumor, with an estimated 14,320 new cases and 2,190 deaths in the US in 2015.1 The most important etiological factors are tobacco, excessive consumption of alcohol, and betel quid usage, which act separately or synergistically.2 It has been reported that TSCC is more commonly found in males, with a percentage of 72.0% of all TSCC cases, compared with a percentage of 28.0% in females.1 Of note, the incidence of TSCC in young white women is significantly increasing, according to the Surveillance, Epidemiology, and End Results program data from 1973 to 2010 in the US.3 Because of the mobility and masticatory function of the tongue, TSCC is inclined to spread locally, involving perioral structures, and metastasize to local regional lymph nodes. For this reason, TSCC always demonstrates a much more aggressive behavior than other kinds of oral and maxillofacial tumors.4 Although there has been advancement in the sequential therapies, including radiation, surgery, and chemotherapy, the patients still suffer from serious relapse and the 5-year survival rate shows no inspiring progress.5 It requires the development of novel therapeutics with improved therapeutic effect and reduced side effect for TSCC treatment.

It has been revealed that acquisition of epithelial to mesenchymal transition (EMT) and induction of cancer stem cell (CSC)-like properties are closely involved in the initiation, development, progression, metastasis, and relapse of solid tumors.6,7 Normal epithelial cells show apical–basal polarity maintained by apical tight junctions and basolateral adherens junctions. The loss of epithelial property and acquisition of mesenchymal features enables cancer cells to metastasize easily and quickly. The reverse process of mesenchymal to epithelial transition (MET) can also occur. Cancer progression is affected by the balance between EMT and MET.8,9 Thus, interfering with the EMT process may help regress cancer metastasis. On the other hand, the proliferation of tumors is driven by a bulk of dedicated stem cells, the CSCs. CSCs, also known as cancer-initiating cells, are involved in cancer cell renewal and differentiation.10 CSCs can display EMT characteristics such as loss of adhesion protein E-cadherin.11 It is reported that CSCs play an important role in chemotherapy resistance due to the self-renewal ability. Multiple oncogenes are involved in the maintenance of stemness and tumorigenicity of CSCs, such as Octamer-4 (Oct-4), Bmi-1, Nanog, and sex-determing region Y-box 2 (Sox-2).1215 Hence, targeting the CSCs shows great therapeutic potential in cancer therapy.16

Plumbagin (5-hydroxy-2-methyl-1, 4-naphthoquinone; PLB) is isolated from the root of Plumbago zeylanica L, Juglans regia, Juglans cinerea, and Juglans nigra, with a variety of pharmacological activities including anti-inflammatory, antiatherosclerotic, antibacterial, antifungal, and anticancer activities in in vitro and in vivo models.17 Notably, PLB shows a potent ability in killing cancer cells with minimal side effects.18 Previous studies from our and other groups have shown that PLB regulates various cellular processes such as cell cycle, apoptosis, autophagy, and cellular redox status.1921 PLB also induces cancer cell apoptosis and autophagy by inhibition of nuclear factor kappa B (NF-κB) activation and phosphatidylinositide 3-kinase (PI3K)/protein kinase B (Akt)/mTOR signaling pathway.2125 Besides, PLB can efficiently facilitate reactive oxygen species (ROS) generation, which also contributes to the cancer cell killing effect.2628 However, the full spectrum of the molecular targets and therapeutic effects of PLB in TSCC are not clear.

Accumulating evidence shows that stable isotope labeling by amino acids in cell culture (SILAC)-based quantitative proteomic approach has the capability of revealing the potential targets of a given compound or drug.29,30 In this study, we aimed at elucidating the possible mechanisms for PLB’s anticancer effect in the treatment of TSCC using a SILAC-based quantitative proteomic approach to take a panoramic view of PLB in a TSCC cell line (SCC25). The corresponding verifications were also performed. In addition, we also carried out separate experiments to investigate the relationship between PLB-induced ROS generation and PLB-mediated cell cycle arrest, apoptosis induction, EMT inhibition, and stemness attenuation.

Materials and methods

Chemicals and reagents

Dulbecco’s Modified Eagle’s Medium (DMEM) and Ham’s F12 medium were obtained from Corning Cellgro Inc. (Herndon, VA, USA). Fetal bovine serum (FBS), PLB, dimethyl sulfoxide (DMSO), hydrocortisone, N-acetyl-l-cysteine (NAC, a ROS scavenger), l-glutathione (GSH, a ROS scavenger), ammonium persulfate, d-glucose, propidium iodide (PI), ribonuclease, protease and phosphatase inhibitor cocktails, radioimmunoprecipitation assay buffer (RIPA), bovine serum albumin (BSA), Tris base, sodium dodecyl sulfate (SDS), ethylenediaminetetraacetic acid, Dulbecco’s phosphate-buffered saline (PBS), dithiothreitol (DTT), 13C6-l-lysine, l-lysine, 13C6 15N4-l-arginine, and l-arginine were purchased from Sigma-Aldrich (St Louis, MO, USA). FASP™ protein digestion kit was bought from Protein Discovery Inc. (Knoxville, TN, USA). The Annexin V:PE apoptosis detection kit was purchased from BD Pharmingen Biosciences (San Jose, CA, USA). Ionic Detergent Compatibility Reagent (IDCR) kit, nuclear and cytoplasmic extraction kit, Pierce bicinchoninic acid (BCA) protein assay kit, skimmed milk, and Western blotting substrate were bought from Thermo Fisher Scientific (Waltham, MA, USA). The polyvinylidene difluoride (PVDF) membrane was purchased from Bio-Rad (Hercules, CA, USA). Primary antibodies against human CDK1/cdc2, Cyclin B1, cdc25, Fas (TNFRSF6)-associated via death domain (FADD), TNF1 receptor-associated death domain (TRADD), TRAIL-R2 (DR5), cleaved caspase-3 (CC3), E-cadherin, N-cadherin, Snail, Slug, zinc finger E-box-binding homeobox 1 (TCF8/ZEB1), vimentin, β-Catenin, zona occludens protein 1 (ZO-1), claudin-1, Oct-4, Bmi-1, Nanog, Sox-2, and glutathione S-transferase (GST) were purchased from Cell Signaling Technology Inc. (Beverly, MA, USA). Primary antibodies against nuclear factor erythroid 2-related factor 2 (Nrf2), NAD(P)H quinone oxidoreductase 1 (NQO1), and heat shock protein 90 (HSP90) were purchased from Santa Cruz Biotechnology Inc. (Santa Cruz, CA, USA). The antibodies against human β-actin and Histone H3 were obtained from Santa Cruz Biotechnology Inc.

Cell line and cell culture

The TSCC cell line SCC25 was obtained from the American Type Culture Collection (Manassas, VA, USA) and cultured in a 1:1 mixture of DMEM and Ham’s F12 medium containing 1.2 g/L sodium bicarbonate, 2.5 mM l-glutamine, 15 mM HEPES, and 0.5 mM sodium pyruvate and was supplemented with 400 ng/mL hydrocortisone and 10% heat-inactivated FBS. The cells were maintained at 37°C in a 5% CO2/95% air humidified incubator. PLB was dissolved in DMSO with a stock concentration of 100 mM and was freshly diluted to the desired concentrations with the culture medium. The final concentration of DMSO was at 0.05% (v/v, volume per volume). The control cells received only the vehicle. No ethics approval was required for the use of this cell line this paper.

SILAC quantitative proteomics

Quantitative proteomic experiments were performed using SILAC as described previously.3133 Briefly, SCC25 cells were cultured in the medium with or without stable isotope-labeled amino acids (13C6 l-lysine and 13C6 15N4 l-arginine). SCC25 cells were passaged for five times by changing medium or splitting cells. Then, cells with stable isotope-labeled amino acids were treated with 5 μM PLB for 24 hours. Following that, the cell samples were harvested and lysed with hot lysis buffer (100 mM Tris base, 4% SDS, and 100 mM DTT). The proteins were denatured at 95°C for 5 minutes and sonicated at 20% amplitude (AMPL) for 3 seconds with 6 pulses. Later, the samples were centrifuged at 15,000× g for 20 minutes and the supernatant was collected in clean tubes. The protein concentration was determined using the IDCR kits. Then, equal amounts of heavy and light protein sample were combined to reach a total volume of 30–60 μL containing 300–600 μg proteins. The combined protein sample was digested using FASP™ protein digestion kit. After proteins were digested, the resultant sample was acidified to pH of 3 and desalted using a C18 solid-phase extraction column. The samples were then concentrated using vacuum concentrator at 45°C for 120 minutes and the peptide mixtures (5 μL) were subject to the hybrid linear ion trap-Orbitrap (LTQ Orbitrap XL, Thermo Scientific Inc.). Liquid chromatography-tandem mass spectrometry (LC-MS/MS) was performed using a 10 cm long 75 μm (inner diameter) reversed-phase column packed with 5 μm diameter C18 material with 300 Å pore size (New Objective, Woburn, MA, USA), with a gradient mobile phase of 2%–40% acetonitrile in 0.1% formic acid at 200 μL/minute for 125 minutes. The Orbitrap full MS scanning was performed at a mass (m/z) resolving power of 60,000, with positive polarity in profile mode (M+H+). Peptide SILAC ratio was calculated using MaxQuant version 1.2.0.13. The SILAC ratio was determined by averaging all peptide SILAC ratios from peptides identified of the same protein. The protein IDs were identified using Scaffold 4.3.2 from Proteome Software Inc. (Portland, OR, USA) and the pathway was analyzed using Ingenuity Pathway Analysis (IPA) from QIAGEN (Redwood City, CA, USA).

Cell cycle distribution analysis

The effect of PLB on cell cycle distribution of SCC25 cells was determined by flow cytometry using PI as the DNA stain as described previously.20 Briefly, SCC25 cells were treated with PLB at concentrations of 0.1, 1, and 5 μM for 24 hours. In separate experiments, SCC25 cells were treated with 5 μM PLB for 6, 24, and 48 hours. In addition, the effect of ROS scavengers (GSH and NAC)34 on PLB-induced G2/M arrest was also examined. Cells were trypsinized and resuspended in 1 mL serum-free medium. Subsequently, the cells were fixed with 3 mL 70% ethanol at −20°C overnight. The cells were stained using 50 μg/mL PI. A total number of 1×104 cells was subject to cell cycle analysis using a flow cytometer (Becton Dickinson Immunocytometry Systems, San Jose, CA, USA). Finally, the raw data were analyzed by ModFitLT software (version 3.2.1) (Verity Software House, Topsham, MA, USA).

Quantification of cellular apoptosis

We used Annexin V:PE apoptosis detection kit to measure apoptotic cells after the cells were treated with 0.1, 1, and 5 μM PLB for 24 hours. In separate experiments, SCC25 cells were treated with 5 μM PLB for 6, 24, and 48 hours. In addition, the effect of GSH and NAC on PLB-induced apoptosis was also examined. Briefly, cells were trypsinized and washed twice with cold PBS, and then resuspended in 1× binding buffer with 5 μL of PE Annexin V and 5 μL of 7-amino-actinomycin D at a concentration of 1×105/mL cells in a total volume of 100 μL. The cells were gently mixed and incubated in the dark for 15 minutes at room temperature. Following that, a quota of 1× binding buffer (400 μL) was added to each test tube and the number of apoptotic cells was quantified by flow cytometry (BD LSR II Analyzer) within 1 hour.

Western blotting assay

The levels of various cellular proteins were determined using Western blotting assays. The SCC25 cells were incubated with PLB at 0.1, 1, and 5 μM for 24 hours. In separate experiments, SCC25 cells were treated with 5 μM PLB for 6, 24, and 48 hours. After PLB treatment, cells were washed twice with precold PBS and lysed with the RIPA buffer containing the protease inhibitor and phosphatase inhibitor cocktails. In addition, we extracted the nuclear proteins according to the instruction given in the nuclear and cytoplasmic extraction kit. Protein concentrations were measured using the Pierce BCA protein assay kit. Equal amounts of protein samples at 20 μg were electrophoresed on 7%−12% sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS-PAGE) minigel after thermal denaturation for 5 minutes at 95°C. Proteins were transferred onto immobilon PVDF membrane at 80 V for 3 hours at 4°C. Subsequently, membranes were blocked with 5% BSA and probed with indicated primary antibody overnight at 4°C and then blotted with respective secondary antibody. Visualization was performed using the Bio-Rad system. Cytosolic protein level was normalized to the matching densitometric value of β-actin, and nuclear protein level was normalized to the matching densitometric value of Histone H3.

Statistical analysis

Data are presented as the mean ± standard deviation (SD). Multiple comparisons were evaluated by one-way analysis of variance (ANOVA), followed by Tukey’s multiple comparison. A value of P<0.05 was considered statistically significant. All the assays were performed in triplicate.

Results

Summary of proteomic response to PLB treatment in SCC25 cells

We first performed SILAC-based proteomics to evaluate the potential molecular targets of PLB in SCC25 cells. PLB increased the expression level of 143 protein molecules, but decreased the expression level of 255 protein molecules in SCC25 cells (Tables 1 and 2). Subsequently, these proteins were subject to IPA. The results showed that 101 signaling pathways were potentially regulated by PLB in SCC25 cells (Table 3 and Figure 1). The top ten targeted signaling pathways were EIF2 signaling pathway, regulation of eIF4 and p70S6K signaling, remodeling of epithelial adherens junctions pathway, mTOR signaling pathway, protein ubiquitination pathway, Nrf2-mediated oxidative stress response signaling pathway, epithelial adherens junction signaling pathway, caveolar-mediated endocytosis signaling pathway, RhoA signaling pathway, and oxidative phosphorylation pathway (Table 3). Notably, a number of molecules were involved in cell survival, cell proliferation, redox homeostasis, cell metabolism, cell migration, and cell death, such as p53, CDK1/cdc2, FADD, Nrf2, MAPK, mTOR, p70S6K, E-cadherin, and vimentin.

Table 1.

Proteins upregulated by PLB in SCC25 cells

Fold change ID Symbol Entrez gene name Location Type(s)
16.699 Q9Y4L1 HYOU1 Hypoxia upregulated 1 Cytoplasm Other
6.339 Q86UP2 KTN1 Kinectin 1 (kinesin receptor) Plasma membrane Transmembrane receptor
5.171 P07996 THBS1 Thrombospondin 1 Extracellular space Other
4.993 Q9H3K6 BOLA2/BC BolA family member 2 Cytoplasm Other
4.304 Q9NY33 DPP3 Dipeptidyl-peptidase 3 Cytoplasm Peptidase
3.861 P12277 CKB Creatine kinase, brain Cytoplasm Kinase
3.794 P14866 HNRNPL Heterogeneous nuclear ribonucleoprotein L Nucleus Other
3.14 H0Y4R1 IMPDH2 IMP (inosine 5′-monophosphate) dehydrogenase 2 Cytoplasm Enzyme
3.022 Q9NZB2 FAM120A Family with sequence similarity 120A Cytoplasm Other
2.667 Q9P2E9 RRBP1 Ribosome binding protein 1 Cytoplasm Other
2.61 Q9BQE3 TUBA1C Tubulin, alpha 1c Cytoplasm Other
2.289 P62136 PPP1CA Protein phosphatase 1, catalytic subunit, alpha isozyme Cytoplasm Phosphatase
2.164 P23284 PPIB Peptidylprolyl isomerase B (cyclophilin B) Cytoplasm Enzyme
2.142 P14923 JUP Junction plakoglobin Plasma membrane Other
2.136 P38646 HSPA9 Heat shock 70 kDa protein 9 (mortalin) Cytoplasm Other
2.052 C9JZR2 CTNND1 Catenin (cadherin-associated protein), delta 1 Nucleus Other
1.978 O43852 CALU Calumenin Cytoplasm Other
1.871 Q01082 SPTBN1 Spectrin, beta, non-erythrocytic 1 Plasma membrane Other
1.864 P50454 SERPINH1 Serpin peptidase inhibitor, clade H (heat shock protein 47), member 1, (collagen binding protein 1) Extracellular space Other
1.753 Q13751 LAMB3 Laminin, beta 3 Extracellular space Transporter
1.742 Q13158 FADD Fas (TNFRSF6)-associated via death domain Cytoplasm Other
1.732 Q9UHX1 PUF60 Poly-U binding splicing factor 60 kDa Nucleus Other
1.718 P32320 CDA Cytidine deaminase Nucleus Enzyme
1.695 P05556 ITGB1 Integrin, beta 1 (fibronectin receptor, beta polypeptide, antigen CD29 includes MDF2, MSK12) Plasma membrane Transmembrane receptor
1.682 P04844 RPN2 Ribophorin II Cytoplasm Enzyme
1.615 P17844 DDX5 DEAD (Asp-Glu-Ala-Asp) box helicase 5 Nucleus Enzyme
1.612 O14579 COPE Coatomer protein complex, subunit epsilon Cytoplasm Transporter
1.581 Q13409 DYNC1/2 Dynein, cytoplasmic 1, intermediate chain 2 Cytoplasm Other
1.576 Q13753 LAMC2 Laminin, gamma 2 Extracellular space Other
1.556 Q96QK1 VPS35 Vacuolar protein sorting 35 (S. cerevisiae) Cytoplasm Transporter
1.536 P04181 OAT Ornithine aminotransferase Cytoplasm Enzyme
1.535 E7EPN9 PRRC2C Proline-rich coiled-coil 2C Other Other
1.534 P22102 GART Phosphoribosylglycinamide formyltransferase, phosphoribosylglycinamide synthetase, phosphoribosylaminoimidazole synthetase Cytoplasm Enzyme
1.518 P45974 USP5 Ubiquitin specific peptidase 5 (isopeptidase T) Cytoplasm Peptidase
1.508 P62263 RPS14 Ribosomal protein S14 Cytoplasm Translation regulator
1.474 P08779 KRT16 Keratin 16 Cytoplasm Other
1.463 P62701 RPS4X Ribosomal protein S4, X-linked Cytoplasm Other
1.454 P31939 ATIC 5-Aminoimidazole-4-carboxamide ribonucleotide formyltransferase/IMP cyclohydrolase Cytoplasm Enzyme
1.442 Q6NZI2 PTRF Polymerase I and transcript release factor Nucleus Transcription regulator
1.439 B1AH77 RAC2 Ras-related C3 botulinum toxin substrate 2 (rho family, small GTP binding protein Rac2) Cytoplasm Enzyme
1.412 P05787 KRT8 Keratin 8 Cytoplasm Other
1.41 P60228 EIF3E Eukaryotic translation initiation factor 3, subunit E Cytoplasm Other
1.408 A1A4Z1 IQUB IQ motif and ubiquitin domain containing Cytoplasm Other
1.405 Q15459 SF3A1 Splicing factor 3a, subunit 1, 120 kDa Nucleus Other
1.404 O95817 BAG3 BCL2-associated athanogene 3 Cytoplasm Other
1.398 F5GWP8 KRT17 Keratin 17 Cytoplasm Other
1.396 O00303 EIF3F Eukaryotic translation initiation factor 3, subunit F Cytoplasm Translation regulator
1.388 P11021 HSPA5 Heat shock 70 kDa protein 5 (glucose-regulated protein, 78 kDa) Cytoplasm Enzyme
1.374 Q07065 CKAP4 Cytoskeleton-associated protein 4 Cytoplasm Other
1.367 P49411 TUFM Tu translation elongation factor, mitochondrial Cytoplasm Translation regulator
1.363 K7EK07 H3F3A/H3F3B H3 histone, family 3A Nucleus Other
1.36 P15924 DSP Desmoplakin Plasma membrane Other
1.357 P20700 LMNB1 Lamin B1 Nucleus Other
1.324 P14625 HSP90B1 Heat shock protein 90 kDa beta (Grp94), member 1 Cytoplasm Other
1.315 F8VY35 NAP1L1 Nucleosome assembly protein 1-like 1 Nucleus Other
1.314 F8VZX2 PCBP2 Poly(rC) binding protein 2 Nucleus Other
1.302 P42224 STAT1 Signal transducer and activator of transcription 1, 91 kDa Nucleus Transcription regulator
1.286 P35613 BSG Basigin (Ok blood group) Plasma membrane Transporter
1.268 Q9UQ80 PA2G4 Proliferation-associated 2G4, 38 kDa Nucleus Transcription regulator
1.258 K7EJ78 RPS15 Ribosomal protein S15 Cytoplasm Other
1.254 F8VPF3 PDE6H Phosphodiesterase 6H, cGMP-specific, cone, gamma Cytoplasm Enzyme
1.252 P32969 RPL9 Ribosomal protein L9 Cytoplasm Other
1.243 P09972 ALDOC Aldolase C, fructose-bisphosphate Cytoplasm Enzyme
1.243 P26038 MSN Moesin Plasma membrane Other
1.234 P68366 TUBA4A Tubulin, alpha 4a Cytoplasm Other
1.232 P23526 AHCY Adenosylhomocysteinase Cytoplasm Enzyme
1.224 P31949 S100A11 S100 calcium binding protein A11 Cytoplasm Other
1.22 R4GNH3 PSMC3 Proteasome (prosome, macropain) 26S subunit, ATPase, 3 Nucleus Transcription regulator
1.215 Q12906 ILF3 Interleukin enhancer binding factor 3, 90 kDa Nucleus Transcription regulator
1.214 P46777 RPL5 Ribosomal protein L5 Cytoplasm Other
1.21 H0YA96 HNRNPD Heterogeneous nuclear ribonucleoprotein D (AU-rich element RNA binding protein 1, 37kDa) Nucleus Transcription regulator
1.209 P22314 UBA1 Ubiquitin-like modifier activating enzyme 1 Cytoplasm Enzyme
1.199 P02786 TFRC Transferrin receptor Plasma membrane Transporter
1.198 P02545 LMNA Lamin A/C Nucleus Other
1.17 Q08211 DHX9 DEAH (Asp-Glu-Ala-His) box helicase 9 Nucleus Enzyme
1.163 P31153 MAT2A Methionine adenosyltransferase II, alpha Cytoplasm Enzyme
1.162 P55072 VCP Valosin containing protein Cytoplasm Enzyme
1.16 P62979 RPS27A Ribosomal protein S27a Cytoplasm Other
1.16 P31947 SFN Stratifin Cytoplasm Other
1.159 P23381 WARS Tryptophanyl-tRNA synthetase Cytoplasm Enzyme
1.154 P46940 IQGAP1 IQ motif containing GTPase activating protein 1 Cytoplasm Other
1.153 Q13177 PAK2 p21 protein (Cdc42/Rac)-activated kinase 2 Cytoplasm Kinase
1.148 Q9Y446 PKP3 Plakophilin 3 Plasma membrane Other
1.147 H0YLC2 PSMA4 Proteasome (prosome, macropain) subunit, alpha type, 4 Cytoplasm Peptidase
1.144 P25705 ATP5A1 ATP synthase, H+ transporting, mitochondrial F1 complex, alpha subunit 1, cardiac muscle Cytoplasm Transporter
1.143 Q14697 GANAB Glucosidase, alpha; neutral AB Cytoplasm Enzyme
1.137 P06576 ATP5B ATP synthase, H+ transporting, mitochondrial F1 complex, beta polypeptide Cytoplasm Transporter
1.136 P07355 ANXA2 Annexin A2 Plasma membrane Other
1.136 Q99460 PSMD1 Proteasome (prosome, macropain) 26S subunit, non-ATPase, 1 Cytoplasm Other
1.133 P07858 CTSB Cathepsin B Cytoplasm Peptidase
1.129 Q9UMS4 PRPF19 Pre-mRNA processing factor 19 Nucleus Enzyme
1.127 P31948 STIP1 Stress-induced phosphoprotein 1 Cytoplasm Other
1.126 P05783 KRT18 Keratin 18 Cytoplasm Other
1.125 P20618 PSMB1 Proteasome (prosome, macropain) subunit, beta type, 1 Cytoplasm Peptidase
1.123 P30101 PDIA3 Protein disulfide isomerase family A, member 3 Cytoplasm Peptidase
1.122 P04083 ANXA1 Annexin A1 Plasma membrane Enzyme
1.115 Q96FW1 OTUB1 OTU deubiquitinase, ubiquitin aldehyde binding 1 Cytoplasm Enzyme
1.115 P55735 SEC13 SEC13 homolog (S. cerevisiae) Cytoplasm Transporter
1.111 P09936 UCHL1 Ubiquitin carboxyl-terminal esterase L1 (ubiquitin thiolesterase) Cytoplasm Peptidase
1.108 O60506 SYNCRIP Synaptotagmin binding, cytoplasmic RNA interacting protein Nucleus Other
1.104 P02533 KRT14 Keratin 14 Cytoplasm Other
1.103 P36952 SERPINB5 Serpin peptidase inhibitor, clade B (ovalbumin), member 5 Extracellular space Other
1.103 O43399 TPD52L2 Tumor protein D52-like 2 Cytoplasm Other
1.099 P50991 CCT4 Chaperonin containing TCP1, subunit 4 (delta) Cytoplasm Other
1.096 P11142 HSPA8 Heat shock 70 kDa protein 8 Cytoplasm Enzyme
1.096 P40926 MDH2 Malate dehydrogenase 2, NAD (mitochondrial) Cytoplasm Enzyme
1.096 P31946 YWHAB Tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, beta Cytoplasm Transcription regulator
1.094 Q92597 NDRG1 N-myc downstream regulated 1 Nucleus Kinase
1.086 H7C5W9 ATP2A2 ATPase, Ca2+ transporting, cardiac muscle, slow twitch 2 Cytoplasm Transporter
1.086 P13489 RNH1 Ribonuclease/angiogenin inhibitor 1 Cytoplasm Other
1.083 P31930 UQCRC1 Ubiquinol-cytochrome c reductase core protein I Cytoplasm Enzyme
1.08 P17655 CAPN2 Calpain 2, (m/II) large subunit Cytoplasm Peptidase
1.08 P34932 HSPA4 Heat shock 70 kDa protein 4 Cytoplasm Other
1.076 P62241 RPS8 Ribosomal protein S8 Cytoplasm Other
1.071 P13647 KRT5 Keratin 5 Cytoplasm Other
1.07 P02538 KRT6A Keratin 6A Other Other
1.058 P61981 YWHAG Tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, gamma Cytoplasm Other
1.057 P04075 ALDOA Aldolase A, fructose-bisphosphate Cytoplasm Enzyme
1.053 Q96AG4 LRRC59 Leucine rich repeat containing 59 Cytoplasm Other
1.05 P23219 PTGS1 Prostaglandin-endoperoxide synthase 1 (prostaglandin G/H synthase and cyclooxygenase) Cytoplasm Enzyme
1.05 P27348 YWHAQ Tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, theta Cytoplasm Other
1.046 P00387 CYB5R3 Cytochrome b5 reductase 3 Cytoplasm Enzyme
1.046 Q9UL46 PSME2 Proteasome (prosome, macropain) activator subunit 2 (PA28 beta) Cytoplasm Peptidase
1.037 P06748 NPM1 Nucleophosmin (nucleolar phosphoprotein B23, numatrin) Nucleus Transcription regulator
1.036 P36578 RPL4 Ribosomal protein L4 Cytoplasm Enzyme
1.033 P28066 PSMA5 Proteasome (prosome, macropain) subunit, alpha type, 5 Cytoplasm Peptidase
1.032 P08238 HSP90AB1 Heat shock protein 90 kDa alpha (cytosolic), class B member 1 Cytoplasm Enzyme
1.03 P52907 CAPZA1 Capping protein (actin filament) muscle Z-line, alpha 1 Cytoplasm Other
1.03 P16152 CAR1 Carbonyl reductase 1 Cytoplasm Enzyme
1.028 Q14019 COTL1 Coactosin-like F-actin binding protein 1 Cytoplasm Other
1.027 P04632 CAPNS1 Calpain, small subunit 1 Cytoplasm Peptidase
1.026 P19105 MYL12A Myosin, light chain 12A, regulatory, nonsarcomeric Cytoplasm Other
1.024 P12814 ACTN1 Actinin, alpha 1 Cytoplasm Other
1.021 P50395 GD12 GDP dissociation inhibitor 2 Cytoplasm Other
1.02 Q01518 CAP1 CAP, adenylate cyclase-associated protein 1 (yeast) Plasma membrane Other
1.018 J3KPE3 GNB2L1 Guanine nucleotide binding protein (G protein), beta polypeptide 2-like 1 Cytoplasm Enzyme
1.014 Q99880 HIST1H2BL Histone cluster 1, H2bl Nucleus Other
1.012 P27695 APEX1 APEX nuclease (multifunctional DNA repair enzyme) 1 Nucleus Enzyme
1.007 P68371 TUBB4B Tubulin, beta 4B class IVb Cytoplasm Other
1.006 F5GZS6 SLC3A2 Solute carrier family 3 (amino acid transporter heavy chain), member 2 Plasma membrane Transporter
1.004 O15371 EIF3D Eukaryotic translation initiation factor 3, subunit D Cytoplasm Other
1.002 D6RG13 RPS3A Ribosomal protein S3A Nucleus Other
1.001 M0R2L9 RPS19 Ribosomal protein S19 Cytoplasm Other

Abbreviation: PLB, plumbagin.

Table 2.

Proteins downregulated by PLB in SCC25 cells

Fold change ID Symbol Entrez gene name Location Type(s)
−3.32 P52306 RAP1GDS1 RAP1, GTP-GDP dissociation stimulator 1 Cytoplasm Other
−2.964 P29966 MARCKS Myristoylated alanine-rich protein kinase C substrate Plasma membrane Other
−2.937 P04264 KRT1 Keratin 1 Cytoplasm Other
−2.79 Q15942 ZYX Zyxin Plasma membrane Other
−2.644 P35527 KRT9 Keratin 9 Other Other
−2.642 P80723 BASP1 Brain abundant, membrane attached signal protein 1 Nucleus Transcription regulator
−2.561 B8ZZQ6 PTMA Prothymosin, alpha Nucleus Other
−2.542 P25786 PSMA1 Proteasome (prosome, macropain) subunit, alpha type, 1 Cytoplasm Peptidase
−2.476 P35637 FUS FUS RNA binding protein Nucleus Transcription regulator
−2.439 P16989 YBX3 Y box binding protein 3 Nucleus Transcription regulator
−2.394 K7EL20 EIF3G Eukaryotic translation initiation factor 3, subunit G Cytoplasm Other
−2.346 P13726 F3 Coagulation factor III (thromboplastin, tissue factor) Plasma membrane Transmembrane receptor
−2.3 Q08J23 NSUN2 NOP2/Sun RNA methyltransferase family, member 2 Nucleus Enzyme
−2.244 P47895 ALDH1A3 Aldehyde dehydrogenase 1 family, member A3 Cytoplasm Enzyme
−2.128 P26358 DNMT1 DNA (cytosine-5-)-methyltransferase 1 Nucleus Enzyme
−2.111 Q15582 TGFBI Transforming growth factor, beta-induced, 68 kDa Extracellular space Other
−2.081 P30153 PPP2R1A Protein phosphatase 2, regulatory subunit A, alpha Cytoplasm Phosphatase
−2.052 P06744 GPI Glucose-6-phosphate isomerase Extracellular space Enzyme
−2.018 Q5JXT2 NOP56 NOP56 ribonucleoprotein Nucleus Other
−2.013 P53621 COPA Coatomer protein complex, subunit alpha Cytoplasm Transporter
−1.946 Q16777 HIST2H2AC Histone cluster 2, H2ac Nucleus Other
−1.921 C9JNR4 RHOA Ras family member A Cytoplasm Enzyme
−1.921 F8VWS0 RPLP0 Ribosomal protein, large, P0 Cytoplasm Other
−1.915 P09211 GSTP1 Glutathione S-transferase pi 1 Cytoplasm Enzyme
−1.88 Q06830 PRDX1 Peroxiredoxin 1 Cytoplasm Enzyme
−1.87 P11498 PC Pyruvate carboxylase Cytoplasm Enzyme
−1.86 P33176 KIF5B Kinesin family member 5B Cytoplasm Other
−1.858 I7HJJ0 SLC25A6 Solute carrier family 25 (mitochondrial carrier; adenine nucleotide translocator), member 6 Cytoplasm Transporter
−1.856 P52566 ARHGDIB Rho GDP dissociation inhibitor (GDI) beta Cytoplasm Other
−1.844 P67809 YBX1 Y box binding protein 1 Nucleus Transcription regulator
−1.764 Q9UKY7 CDV3 CDV3 homolog (mouse) Cytoplasm Other
−1.764 B7Z972 PCMT1 Protein-l-isoaspartate (d-aspartate) O-methyltransferase Cytoplasm Enzyme
−1.737 E9PBS1 PAICS Phosphoribosylaminoimidazole carboxylase, phosphoribo-sylaminoimidazole succinocarboxamide synthetase Cytoplasm Enzyme
−1.727 P13010 XRCC5 X-ray repair complementing defective repair in Chinese hamster cells 5 (double-strand-break rejoining) Nucleus Enzyme
−1.723 P26368 U2AF2 U2 small nuclear RNA auxiliary factor 2 Nucleus Other
−1.722 P28074 PSMB5 Proteasome (prosome, macropain) subunit, beta type, 5 Cytoplasm Peptidase
−1.715 P15374 UCHL3 Ubiquitin carboxyl-terminal esterase L3 (ubiquitin thiolesterase) Cytoplasm Peptidase
−1.709 P68104 EEF1A1 Eukaryotic translation elongation factor 1 alpha 1 Cytoplasm Translation regulator
−1.698 J3KTF8 ARHGDIA Rho GDP dissociation inhibitor (GDI) alpha Cytoplasm Other
−1.694 O75367 H2AFY H2A histone family, member Y Nucleus Other
−1.663 P63010 AP2B1 Adaptor-related protein complex 2, beta 1 subunit Plasma membrane Transporter
−1.655 B4DUR8 CCT3 Chaperonin containing TCP1, subunit 3 (gamma) Cytoplasm Other
−1.646 P13667 PDIA4 Protein disulfide isomerase family A, member 4 Cytoplasm Enzyme
−1.638 P23229 ITGA6 Integrin, alpha 6 Plasma membrane Transmembrane receptor
−1.632 F8VZ29 UBE2N Ubiquitin-conjugating enzyme E2N Cytoplasm Enzyme
−1.631 Q9UHI5 SLC7A8 Solute carrier family 7 (amino acid transporter light chain, L system), member 8 Plasma membrane Transporter
−1.629 F8W726 UBAP2L Ubiquitin associated protein 2-like Other Other
−1.617 Q9HB71 CACYBP Calcyclin binding protein Nucleus Other
−1.614 Q12905 ILF2 Interleukin enhancer binding factor 2 Nucleus Transcription regulator
−1.599 O43390 HNRNPR Heterogeneous nuclear ribonucleoprotein R Nucleus Other
−1.599 E9PK47 PYGL Phosphorylase, glycogen, liver Cytoplasm Enzyme
−1.579 Q6NYC8 PPP1R18 Protein phosphatase 1, regulatory subunit 18 Other Other
−1.574 O60884 DNAJA2 DNAJ (Hsp40) homolog, subfamily A, member 2 Nucleus Enzyme
−1.566 E9PDF6 MYO1B Myosin IB Cytoplasm Other
−1.565 Q02878 RPL6 Ribosomal protein L6 Cytoplasm Other
−1.562 P05386 RPLP1 Ribosomal protein, large, P1 Cytoplasm Other
−1.56 E9PLD0 RAB1B RAB1B, member RAS oncogene family Cytoplasm Other
−1.556 Q14247 CTTN Cortactin Plasma membrane Other
−1.554 P42704 LRPPRC Leucine-rich pentatricopeptide repeat containing Cytoplasm Other
−1.549 Q9H4M9 EHD1 EH-domain containing 1 Cytoplasm Other
−1.498 P30084 ECHS1 Enoyl CoA hydratase, short chain, 1, mitochondrial Cytoplasm Enzyme
−1.498 P49915 GMPS Guanine monphosphate synthase Nucleus Enzyme
−1.496 Q86VP6 CAND1 Cullin-associated and neddylation-dissociated 1 Cytoplasm Transcription regulator
−1.49 P49588 AARS Alanyl-tRNA synthetase Cytoplasm Enzyme
−1.482 P62314 SNRPD1 Small nuclear ribonucleoprotein D1 polypeptide 16 kDa Nucleus Other
−1.474 P20290 BTF3 Basic transcription factor 3 Nucleus Transcription regulator
−1.473 Q9ULV4 CORO1C Coronin, actin binding protein, 1C Cytoplasm Other
−1.468 P51858 HDGF Hepatoma-derived growth factor Extracellular space Growth factor
−1.468 G3V1A1 RPL8 Ribosomal protein L8 Other Other
−1.466 Q9UHD8 SEPT9 Septin 9 Cytoplasm Enzyme
−1.461 P55060 CSE1L CSE1 chromosome segregation 1-like (yeast) Nucleus Transporter
−1.46 P61978 HNRNPK Heterogeneous nuclear ribonucleoprotein K Nucleus Other
−1.457 P84077 ARF1 ADP-ribosylation factor 1 Cytoplasm Enzyme
−1.452 P62081 RPS7 Ribosomal protein S7 Cytoplasm Other
−1.447 K7EJ57 TOMM40 Translocase of outer mitochondrial membrane 40 homolog (yeast) Cytoplasm Ion channel
−1.443 Q9Y5B9 SUPT16H Suppressor of Ty 16 homolog (S. cerevisiae) Nucleus Transcription regulator
−1.434 P49327 FASN Fatty acid synthase Cytoplasm Enzyme
−1.432 P30086 PEBP1 Phosphatidylethanolamine binding protein 1 Cytoplasm Other
−1.421 C9J9K3 RPSA Ribosomal protein SA Cytoplasm Translation regulator
−1.421 Q12874 SF3A3 Splicing factor 3a, subunit 3, 60 kDa Nucleus Other
−1.42 P43490 NAMPT Nicotinamide phosphoribosyltransferase Extracellular space Cytokine
−1.41 Q14444 CAPRIN1 Cell cycle associated protein 1 Plasma membrane Other
−1.409 P13928 ANXA8/ANXA8L1 Annexin A8-like 1 Plasma membrane Other
−1.408 Q9UK76 HN1 Hematological and neurological expressed 1 Nucleus Other
−1.404 P00367 GLUD1 Glutamate dehydrogenase 1 Cytoplasm Enzyme
−1.402 Q14764 MVP Major vault protein Nucleus Other
−1.391 Q9NTK5 OLA1 Obg-like ATPase 1 Cytoplasm Other
−1.391 P08670 VIM Vimentin Cytoplasm Other
−1.387 P07195 LDHB Lactate dehydrogenase B Cytoplasm Enzyme
−1.384 P07384 CAPN1 Calpain 1, (mu/I) large subunit Cytoplasm Peptidase
−1.378 Q5T7C4 HMGB1 High mobility group box 1 Nucleus Transcription regulator
−1.374 P55884 EIF3B Eukaryotic translation initiation factor 3, subunit B Cytoplasm Translation regulator
−1.367 Q15417 CNN3 Calponin 3, acidic Cytoplasm Other
−1.367 Q96AE4 FUBP1 Far upstream element (FUSE) binding protein 1 Nucleus Transcription regulator
−1.366 F8W1N5 NACA Nascent polypeptide-associated complex alpha subunit Cytoplasm Transcription regulator
−1.364 Q32Q12 NME1-NME2 NME1-NME2 readthrough Cytoplasm Other
−1.361 P21796 VDAC1 Voltage-dependent anion channel 1 Cytoplasm Ion channel
−1.359 P54136 RARS Arginyl-tRNA synthetase Cytoplasm Enzyme
−1.359 Q9H2G2 SLK STE20-like kinase Nucleus Kinase
−1.359 Q12792 TWF1 Twinfilin actin-binding protein 1 Cytoplasm Kinase
−1.354 Q7KZF4 SND1 Staphylococcal nuclease and tudor domain containing 1 Nucleus Enzyme
−1.347 P20042 EIF2S2 Eukaryotic translation initiation factor 2, subunit 2 beta, 38 kDa Cytoplasm Translation regulator
−1.344 Q14204 DYNC1H1 Dynein, cytoplasmic 1, heavy chain 1 Cytoplasm Peptidase
−1.34 K7EIP4 LAMA3 Laminin, alpha 3 Extracellular space Other
−1.337 P24534 EEF1B2 Eukaryotic translation elongation factor 1 beta 2 Cytoplasm Translation regulator
−1.331 P61353 RPL27 Ribosomal protein L27 Cytoplasm Other
−1.33 P41250 GARS Glycyl-tRNA synthetase Cytoplasm Enzyme
−1.326 P04080 CSTB Cystatin B (stefin B) Cytoplasm Peptidase
−1.326 G8JLD5 DNM1L Dynamin 1-like Cytoplasm Enzyme
−1.317 P04792 HSPB1 Heat shock 27 kDa protein 1 Cytoplasm Other
−1.315 P12956 XRCC6 X-ray repair complementing defective repair in Chinese hamster cells 6 Nucleus Enzyme
−1.308 P0CW22 RPS17 Ribosomal protein S17 Cytoplasm Other
−1.306 Q13283 G3BP1 GTPase activating protein (SH3 domain) binding protein 1 Nucleus Enzyme
−1.306 Q15181 PPA1 Pyrophosphatase (inorganic) 1 Cytoplasm Enzyme
−1.302 P14618 PKM Pyruvate kinase, muscle Cytoplasm Kinase
−1.297 P27797 CALR Calreticulin Cytoplasm Transcription regulator
−1.287 P48643 CCT5 Chaperonin containing TCP1, subunit 5 (epsilon) Cytoplasm Other
−1.286 P61158 ACTR3 ARP3 actin-related protein 3 homolog (yeast) Plasma membrane Other
−1.286 Q07021 C1QBP Complement component 1, q subcomponent binding protein Cytoplasm Transcription regulator
−1.283 P41091 EIF2S3 Eukaryotic translation initiation factor 2, subunit 3 gamma, 52 kDa Cytoplasm Translation regulator
−1.28 P26639 TARS Threonyl-tRNA synthetase Nucleus Enzyme
−1.279 P43243 MATR3 Matrin 3 Nucleus Other
−1.278 B4DS13 EIF4B Eukaryotic translation initiation factor 4B Cytoplasm Translation regulator
−1.277 P09914 IFIT1 Interferon-induced protein with tetratricopeptide repeats 1 Cytoplasm Other
−1.277 Q15084 PDIA6 Protein disulfide isomerase family A, member 6 Cytoplasm Enzyme
−1.276 P52292 KPNA2 Karyopherin alpha 2 (RAG cohort 1, importin alpha 1) Nucleus Transporter
−1.271 P12004 PCNA Proliferating cell nuclear antigen Nucleus Enzyme
−1.269 P13639 EEF2 Eukaryotic translation elongation factor 2 Cytoplasm Translation regulator
−1.267 F5H018 RAN RAN, member RAS oncogene family Nucleus Enzyme
−1.265 P40121 CAPG Capping protein (actin filament), gelsolin-like Nucleus Other
−1.264 E7EQR4 EZR Ezrin Plasma membrane Other
−1.264 P43686 PSMC4 Proteasome (prosome, macropain) 26S subunit, ATPase, 4 Nucleus Peptidase
−1.263 P63241 EIF5A Eukaryotic translation initiation factor 5A Cytoplasm Translation regulator
−1.261 B1AK85 CAPZB Capping protein (actin filament) muscle Z-line, beta Cytoplasm Other
−1.254 Q14974 KPNB1 Karyopherin (importin) beta 1 Nucleus Transporter
−1.253 P07237 P4HB Prolyl 4-hydroxylase, beta polypeptide Cytoplasm Enzyme
−1.251 B4DLR8 NQO1 NAD(P)H dehydrogenase, quinone 1 Cytoplasm Enzyme
−1.251 O60664 PLIN3 Perilipin 3 Cytoplasm Other
−1.251 P54725 RAD23A RAD23 homolog A (S. cerevisiae) Nucleus Other
−1.25 P18669 PGAM1 Phosphoglycerate mutase 1 (brain) Cytoplasm Phosphatase
−1.248 P05141 SLC25A5 Solute carrier family 25 (mitochondrial carrier; adenine nucleotide translocator), member 5 Cytoplasm Transporter
−1.247 P55263 ADK Adenosine kinase Nucleus Kinase
−1.245 P49321 NASP Nuclear autoantigenic sperm protein (histone-binding) Nucleus Other
−1.244 P22626 HNRNPA2B1 Heterogeneous nuclear ribonucleoprotein A2/B1 Nucleus Other
−1.243 P01892 HLA-A Major histocompatibility complex, class I, A Plasma membrane Other
−1.242 P54727 RAD23B RAD23 homolog B (S. cerevisiae) Nucleus Other
−1.239 P07737 PFN1 Profilin 1 Cytoplasm Other
−1.237 P21333 FLNA Filamin A, alpha Cytoplasm Other
−1.236 P06733 ENO1 Enolase 1, (alpha) Cytoplasm Enzyme
−1.234 P09382 LGALS1 Lectin, galactoside-binding, soluble, 1 Extracellular space Other
−1.232 P35998 PSMC2 Proteasome (prosome, macropain) 26S subunit, ATPase, 2 Nucleus Peptidase
−1.23 P52272 HNRNPM Heterogeneous nuclear ribonucleoprotein M Nucleus Other
−1.229 P06396 GSN Gelsolin Extracellular space Other
−1.229 Q99714 HSD17B10 Hydroxysteroid (17-beta) dehydrogenase 10 Cytoplasm Enzyme
−1.227 P30044 PRDX5 Peroxiredoxin 5 Cytoplasm Enzyme
−1.224 P00491 PNP Purine nucleoside phosphorylase Nucleus Enzyme
−1.223 P39023 RPL3 Ribosomal protein L3 Cytoplasm Other
−1.221 P37802 TAGLN2 Transgelin 2 Cytoplasm Other
−1.22 Q7L2H7 EIF3M Eukaryotic translation initiation factor 3, subunit M Other Other
−1.22 P62906 RPL10A Ribosomal protein L10a Nucleus Other
−1.219 P62937 PPIA Peptidylprolyl isomerase A (cyclophilin A) Cytoplasm Enzyme
−1.215 P11766 ADH5 Alcohol dehydrogenase 5 (class III), chi polypeptide Cytoplasm Enzyme
−1.213 Q09666 AHNAK AHNAK nucleoprotein Nucleus Other
−1.207 E7ETK0 RPS24 Ribosomal protein S24 Cytoplasm Other
−1.205 Q03135 CAV1 Caveolin 1, caveolae protein, 22 kDa Plasma membrane Transmembrane receptor
−1.203 P35606 COPB2 Coatomer protein complex, subunit beta 2 (beta prime) Cytoplasm Transporter
−1.203 P52209 PGD Phosphogluconate dehydrogenase Cytoplasm Enzyme
−1.203 Q13200 PSMD2 Proteasome (prosome, macropain) 26S subunit, non-ATPase, 2 Cytoplasm Other
−1.198 P60842 EIF4A1 Eukaryotic translation initiation factor 4A1 Cytoplasm Translation regulator
−1.198 P18124 RPL7 Ribosomal protein L7 Nucleus Transcription regulator
−1.193 P05161 ISG15 ISG15 ubiquitin-like modifier Extracellular space Other
−1.184 A0A075B730 EPPK1 Epiplakin 1 Cytoplasm Other
−1.184 Q02790 FKBP4 FK506 binding protein 4, 59 kDa Nucleus Enzyme
−1.184 M0R0F0 RPS5 Ribosomal protein S5 Cytoplasm Other
−1.183 P35221 CTNNA1 Catenin (cadherin-associated protein), alpha 1, 102 kDa Plasma membrane Other
−1.179 Q01105 SET SET nuclear proto-oncogene Nucleus Phosphatase
−1.175 P05387 RPLP2 Ribosomal protein, large, P2 Cytoplasm Other
−1.174 Q00839 HNRNPU Heterogeneous nuclear ribonucleoprotein U (scaffold attachment factor A) Nucleus Transporter
−1.173 O15143 ARPC1B Actin related protein 2/3 complex, subunit 1B, 41 kDa Cytoplasm Other
−1.173 P23528 CFL1 Cofilin 1 (non-muscle) Nucleus Other
−1.173 A2A2Y8 COL17A1 Collagen, type XVII, alpha 1 Extracellular space Other
−1.171 E7EQV3 PABPC1 Poly(A) binding protein, cytoplasmic 1 Cytoplasm Translation regulator
−1.171 F8W7C6 RPL10 Ribosomal protein L10 Cytoplasm Other
−1.158 Q14980 NUMA1 Nuclear mitotic apparatus protein 1 Nucleus Other
−1.157 P27824 CANX Calnexin Cytoplasm Other
−1.155 Q14134 TRIM29 Tripartite motif containing 29 Cytoplasm Transcription regulator
−1.154 E9PCY7 HNRNPH1 Heterogeneous nuclear ribonucleoprotein H1 (H) Nucleus Other
−1.153 O00299 CLIC1 Chloride intracellular channel 1 Nucleus Ion channel
−1.145 Q13813 SPTAN1 Spectrin, alpha, nonerythrocytic 1 Plasma membrane Other
−1.144 P07900 HSP90AA1 Heat shock protein 90 kDa alpha (cytosolic), class A member 1 Cytoplasm Enzyme
−1.143 P68363 TUBA1B Tubulin, alpha 1b Cytoplasm Other
−1.14 Q5JP53 TUBB Tubulin, beta class I Cytoplasm Other
−1.137 P60174 TPI1 Triosephosphate isomerase 1 Cytoplasm Enzyme
−1.136 P27482 CALML3 Calmodulin-like 3 Cytoplasm Other
−1.135 P19338 NCL Nucleolin Nucleus Other
−1.124 P18206 VCL Vinculin Plasma membrane Enzyme
−1.117 P10809 HSPD1 Heat shock 60 kDa protein 1 (chaperonin) Cytoplasm Enzyme
−1.117 Q92598 HSPH1 Heat shock 105 kDa/110 kDa protein 1 Cytoplasm Other
−1.114 K7ELL7 PRKCSH Protein kinase C substrate 80K-H Cytoplasm Enzyme
−1.112 P05198 EIF2S1 Eukaryotic translation initiation factor 2, subunit 1 alpha, 35 kDa Cytoplasm Translation regulator
−1.111 P30050 RPL12 Ribosomal protein L12 Nucleus Other
−1.108 F8W6I7 HNRNPA1 Heterogeneous nuclear ribonucleoprotein A1 Nucleus Other
−1.108 P00338 LDHA Lactate dehydrogenase A Cytoplasm Enzyme
−1.106 Q16658 FSCN1 Fascin actin-bundling protein 1 Cytoplasm Other
−1.103 P78371 CCT2 Chaperonin containing TCP1, subunit 2 (beta) Cytoplasm Kinase
−1.102 P68133 ACTA1 Actin, alpha 1, skeletal muscle Cytoplasm Other
−1.095 P29728 OAS2 2′–5′-oligoadenylate synthetase 2, 69/71 kDa Cytoplasm Enzyme
−1.092 P13797 PLS3 Plastin 3 Cytoplasm Other
−1.09 D6RFM5 SDHA Succinate dehydrogenase complex, subunit A, flavoprotein (Fp) Cytoplasm Enzyme
−1.082 B4E022 TKT Transketolase Cytoplasm Enzyme
−1.081 O95433 AHSA1 AHA1, activator of heat shock 90 kDa protein ATPase homologue 1 (yeast) Cytoplasm Other
−1.077 E7EX73 EIF4G1 Eukaryotic translation initiation factor 4 gamma, 1 Cytoplasm Translation regulator
−1.076 P04406 GAPDH Glyceraldehyde-3-phosphate dehydrogenase Cytoplasm Enzyme
−1.076 P26599 PTBP1 Polypyrimidine tract binding protein 1 Nucleus Enzyme
−1.076 P11216 PYGB Phosphorylase, glycogen; brain Cytoplasm Enzyme
−1.072 Q13838 DDX39B DEAD (Asp-Glu-Ala-Asp) box polypeptide 39B Nucleus Enzyme
−1.072 Q99733 NAP1L4 Nucleosome assembly protein 1-like 4 Cytoplasm Other
−1.068 P29692 EEF1D Eukaryotic translation elongation factor 1 delta (guanine nucleotide exchange protein) Cytoplasm Translation regulator
−1.067 O00151 PDLIM1 PDZ and LIM domain 1 Cytoplasm Transcription regulator
−1.065 O75369 FLNB Filamin B, beta Cytoplasm Other
−1.063 P62191 PSMC1 Proteasome (prosome, macropain) 26S subunit, ATPase, 1 Nucleus Peptidase
−1.059 P00558 PGK1 Phosphoglycerate kinase 1 Cytoplasm Kinase
−1.055 P50990 CCT8 Chaperonin containing TCP1, subunit 8 (theta) Cytoplasm Enzyme
−1.052 F5H7V9 TNC Tenascin C Extracellular space Other
−1.049 Q99613 EIF3C Eukaryotic translation initiation factor 3, subunit C Other Translation regulator
−1.048 P45880 VDAC2 Voltage-dependent anion channel 2 Cytoplasm Ion channel
−1.043 P26641 EEF1G Eukaryotic translation elongation factor 1 gamma Cytoplasm Translation regulator
−1.043 P00441 SOD1 Superoxide dismutase 1, soluble Cytoplasm Enzyme
−1.039 E9PFD7 EGFR Epidermal growth factor receptor Plasma membrane Kinase
−1.039 Q9NQC3 RTN4 Reticulon 4 Cytoplasm Other
−1.037 Q9NUQ9 FAM49B Family with sequence similarity 49, member B Extracellular space Other
−1.037 P08729 KRT7 Keratin 7 Cytoplasm Other
−1.037 P25398 RPS12 Ribosomal protein S12 Cytoplasm Other
−1.034 Q13347 EIF3I Eukaryotic translation initiation factor 3, subunit I Cytoplasm Translation regulator
−1.034 F8VQE1 TRMT1 tRNA methyltransferase 1 homolog (S. cerevisiae) Extracellular space Enzyme
−1.031 P63261 ACTG1 Actin, gamma 1 Cytoplasm Other
−1.027 O43707 ACTN4 Actinin, alpha 4 Cytoplasm Other
−1.027 P13796 LCP1 Lymphocyte cytosolic protein 1 (L-plastin) Cytoplasm Other
−1.027 E7EUY0 PRKDC Protein kinase, DNA-activated, catalytic polypeptide Nucleus Kinase
−1.025 Q8NC51 SERBP1 SERPINE1 mRNA binding protein 1 Cytoplasm Other
−1.025 Q13263 TRIM28 Tripartite motif containing 28 Nucleus Transcription regulator
−1.023 Q00610 CLTC Clathrin, heavy chain (Hc) Plasma membrane Other
−1.022 P40227 CCT6A Chaperonin containing TCP1, subunit 6A (zeta 1) Cytoplasm Other
−1.021 P63104 YWHAZ Tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, zeta Cytoplasm Enzyme
−1.02 P51149 RAB7A RAB7A, member RAS oncogene family Cytoplasm Enzyme
−1.018 P25787 PSMA2 Proteasome (prosome, macropain) subunit, alpha type, 2 Cytoplasm Peptidase
−1.016 P35268 RPL22 Ribosomal protein L22 Nucleus Other
−1.012 Q04828 AKR1C1/AKR1C2 Aldo-keto reductase family 1, member C2 Cytoplasm Enzyme
−1.01 P08758 ANXA5 Annexin A5 Plasma membrane Other
−1.01 Q5VU59 TPM3 Tropomyosin 3 Cytoplasm Other
−1.009 Q15233 NONO Non-POU domain containing, octamer-binding Nucleus Other
−1.009 P62258 YWHAE Tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, epsilon Cytoplasm Other
−1.008 P35579 MYH9 Myosin, heavy chain 9, nonmuscle Cytoplasm Enzyme
−1.005 P16144 ITGB4 Integrin, beta 4 Plasma membrane Transmembrane receptor
−1.001 Q15149 PLEC Plectin Cytoplasm Other

Abbreviation: PLB, plumbagin.

Table 3.

Potential signaling pathways regulated by PLB in SCC25 cells

Ingenuity canonical pathways −logP Protein molecules
EIF2 signaling 3.31E01 EIF2S1, EIF2S2, EIF2S3, EIF3B, EIF3C, EIF3D, EIF3E, EIF3F, EIF3G, EIF3I, EIF3M, EIF4A1, EIF4G1, PABPC1, PPP1CA, RPL3, RPL4, RPL5, RPL6, RPL7, RPL8, RPL9, RPL10, RPL12, RPL22, RPL27, RPL10A, RPLP0, RPLP1, RPLP1, RPLP2, RPS5, RPS7, RPS8, RPS12, RPS14, RPS15, RPS17, RPS19, RPS24, RPS27A, RPS3A, RPS4X, RPSA
Regulation of eIF4 and p70S6k signaling 2.03E01 EIF2S1, EIF2S2, EIF2S3, EIF3B, EIF3C, EIF3D, EIF3E, EIF3F, EIF3G, EIF3I, EIF3M, EIF4A1, EIF4G1, ITGB1, PABPC1, PPP2R1A, RPS5, RPS7, RPS8, RPS12, RPS14, RPS15, RPS17, RPS19, RPS24, RPS27A, RPS3A, RPS4X, RPSA
Remodeling of epithelial adherens junctions 1.52E01 ACTA1, ACTG1, ACTN1, ACTN4, ACTR3, ARPC1B, CTNNA1, CTNND1, DNM1L, IQGAP1, RAB7A, TUBA1B, TUBA1C, TUBA4A, TUBB, TUBB4B, VCL, ZYX
mTOR signaling 1.43E01 EIF3B, EIF3C, EIF3D, EIF3E, EIF3F, EIF3G, EIF3I, EIF3M, EIF4A1, EIF4B, EIF4G1, PPPAR1A, RHOA, RPS5, RPS7, RPS8, RPS12, RPS14, RPS15, RPS17, RPS19, RPS24, RPS27A, RPS3A, RPS4X, RPSA
Protein ubiquitination pathway 1.36E01 HLA-A, HSP90AA1, HSP90AB1, HSP90B1, HSPA4, HSPA5, HSPA8, HSPA9, HSPB1, HSPD1, HSPH1, PSMA1, PSMA2, PSMA4, PSMA5, PSMB1, PSMB5, PSMC1, PSMC2, PSMC3, PSMC4, PSMD1, PSMD2, PSME2, UBA1, UBE2N, UCHL1, UCHL3, USP5
Nrf2-mediated oxidative stress response 5.42E00 ACTA1, ACTG1, CBR1, DNAJA2, GSTP1, NQO1, HSP90AA1, PPIB, PRDX1, SOD1, STIP1, VCP
Epithelial adherens junction signaling 5.21E00 ACTA1, ACTG1, ACTN1, ACTN4, ACTR3, ARPC1B, CTNNA1, CTNND1, EGFR, IQGAP1, JUP, MYH9, RHO1, TUBA1B, TUBA1C, TUBA4A, TUBB, TUBB4B, VAL, ZYX
Caveolar-mediated endocytosis signaling 5.18E00 ACTA1, ACTG1, CAV1, COPA, COPB2, COPE, EGFR, FLNA, FLNB, HLA-A, ITGA6, ITGB1, ITGB4, PTRF
RhoA signaling 5.10E00 ACTA1, ACTG1, ACTR3, ARPC1B, CFL1, EZR, KTN1, MAN, MYL12A, PFN1, RHOA, SEPT9
Oxidative phosphorylation 5.08E00 UQCRH, ATP5D, ATP5L, UQCRB, MT-CO2, ATP5H, NDUFA5, NDUFAB1, NDUFB6, ATP5F1, COX4I1, SDHA, ATP5J, COX7A2, COX6B1, COX17, ATP5O, ATP5A1, NDUFS3, ATP5C1, MT-ND1, NDUFB11, ATP5B, NDUFS8, UQCR10, CYC1, UQCRC2, COX5A, CYCS, UQCRC1, COX5B
TCA cycle II (eukaryotic) 4.85E00 SDHA, SUCLA2, CS, SUCLG1, DLST, ACO2, DLD, IDH3A, OGDH, MDH2, FH, MDH1, IDH3B
Germ cell–Sertoli cell junction signaling 4.76E00 ACTA1, ACTG1, ACTN1, ACTN4, CFL1, CTNNA1, CTNND1, GSN, IQGAP1, ITGA6, ITGB1, JUP, PAK2, PAC2, RHOA, TUBA1B, TUBA1C, TUBA4A, TUBB, TUBB4B, ZYX
Actin nucleation by ARP-WASP complex 4.64E00 RHOA, ARPC3
Aspartate degradation II 4.11E00 GOT2
Superpathway of methionine degradation 4.08E00 GOT2
Tight junction signaling 4.08E00 MYL6, PPP2CA, HSF1, ACTA2, VAPA, PRKAR2A, RAC1, YBX3, CDC42, ACTG1, CPSF6, PPP2R1A, CLDN4, MYH9, SAFB, VCL, SPTAN1, CTNNA1, CSTF3, VASP, RHOA
2-Ketoglutarate dehydrogenase complex 3.89E00 DLST, DLD, OGDH
Integrin signaling 3.87E00 ACTA1, ACTG1, ACTN1, ACTN4, ACTR3, ARF1, ARPC1B, CAPN1, CAPN2, CAPNS1, CAV1, CTTN, ITGA6, ITGB1, ITGB4, MYL12A, PAK2, RAC2, RHOA, VCL, ZYX
Clathrin-mediated endocytosis signaling 3.80E00 ACTA1, ACTG1, ACTR3, AP2B1, ARPC1B, CLTC, CTTN, DNM1L, HSPA8, ITGB1, ITGB4, RAB7A, TFRC
Cell cycle: G2/M DNA damage checkpoint regulation 3.78E00 YWHAQ, PRKDC, YWHAG, YWHAE, YWHAH, YWHAB, YWHAZ, SFN, SKP1, CDK1
Glycolysis I 3.75E00 ALDOA, ALD0C, ENO1, GAPDH, GPI, PGAM1, PGAM1, PKG1, PKM, TPI1
Ephrin receptor signaling 3.73E00 RHOA, AKT2, ARPC3, RAC2, ACTR3, MAPK1
Unfolded protein response 3.66E00 CALR, CANX, DNAJA2, HSP90B1, HSPA4, HSPA5, HSPA8, HSPA9, HSPH1, P4HB, VCP
Actin cytoskeleton signaling 3.64E00 ACTA1, ACTG1, ACTN1, ACTN4, ACTR3, ARPC1B, CFL1, FLNA, GSN, IQGAP1, ITGB1, MSN, MYH9, MYL12A, PAK2, PFN1, RAC2, RHOA, VCL
Semaphorin signaling in neurons 3.47E00 RHOA, DPYSL2, PAK2, CFL2, MAPK1, CFL1, RHOC, RAC1
Gluconeogenesis I 3.46E00 ALDOA, ALDOC, ENO1, GAPDH, GPI, MDH2, PGAM1, PGK1
Virus entry via endocytic pathways 3.32E00 ACTA1, ACTG1, AP2B1 CAV1, CLTC, FLNA, FLNB, HLA-A, ITGA6, ITGB1, ITGB4, RAC2, TFRC
Regulation of actin-based motility by rho 3.31E00 ACTA1, ACTR3, ARHGDIA, ARPC1B, CFL1, GSN, ITGB1, MYL12A, PAK2, PFN1, RAC2, RHOA
Mechanisms of viral exit from host cells 3.27E00 CHMP4B, ACTA2, XPO1, LMNB2, PDCD6IP, ACTG1, LMNB1
14-3-3-Mediated signaling 2.95E00 PDIA3, SFN, TUBA1B, TUBA1C, TUBA4A, TUBB, TUBB4B, VIM, YWHAB, YWHAE, YWHAG, YWHAQ, YWHAZ
Superoxide radicals degradation 2.87E00 SOD1, SOD2, NQO1
Spliceosomal cycle 2.86E00 U2AF2, U2AF1
ILK signaling 2.84E00 ACTA1, ACTG1, ACTN1, ACTN4, CDL1, DSP, FLNA, FLNB, ITGB1, ITGB4, KRT18, MYH9, NACA, PPP2R1A, RHOA, VIM
Aryl hydrocarbon receptor signaling 2.76E00 NQO1, GSTP1, ALDH, HSP27
Ethanol degradation II 2.75E00 ADH5, HSD17B10, AKR1A1, ACSL3, DHRS9, ALDH1A3, ALDH3A2, ALDH9A1
Noradrenaline and adrenaline degradation 2.74E00 ADH5, HSD17B10, AKR1A1, DHRS9, ALDH1A3, ALDH3A2, ALDH9A1
Glycogen degradation III 2.72E00 PGM3, PGM1, PYGB, PYGL
Palmitate biosynthesis I (animals) 2.71E00 FASN
Granzyme B signaling 2.64E00 NUMA1, LMNB2, CYCS, LMNB1, PARP1
Regulation of cellular mechanics by caplain protease 2.55E00 ACTN1, ACTN4, CAPN1, CAPN2, CAPNS1, EGFR, EZR, ITGB1, VCL
Pentose phosphate pathway 2.51E00 PGD, TKT, PGLS, TALDO1
BER pathway 2.44E00 PCNA, PARP1, APEX1
RhoGDI signaling 2.43E00 ACTA1, ACTG1, ACTR3, ARHGDIA, ARHGDIB, ARPC1B, CFL1, EZR, GDI2, GNB2L1, ITGB1, MSN, MYL12A, PAK2, RHOA
IGF-1 signaling 2.42E00 SFN, STAT3
Erk/MAPK signaling 2.41E00 RAP1B, ITGB1, PPP1CC, PXN, YWHAG, PAK2, YWHAH, MAPK1, YWHAB, PPP2CA, RRAS, ITGA2, YWHAZ, RAC1, PRKAR2A, TLN1, PPP1R14B, YWHAQ, PPP2R1A, HSPB1, PRKAR1A
Adenine and adenosine salvage I 2.39E00 PNP
Purine nucleotides de novo biosynthesis II 2.36E00 ATIC, GART, GMPS, IMPDH2, PAICS
Hypoxia signaling in the cardiovascular system 2.34E00 HSP90AA1, HSP90AB1, HSP90B1, LDHA, NQO1, P4HB, UBE2N
Glycogen degradation II 2.31E00 PGM3, PGM1, PYGB, PYGL
Sertoli cell–Sertoli cell junction signaling 2.26E00 ACTA1, ACTG1, ACTN1, ACTN4, CTNNA1, ITGB1, JUP, SPTAN1, SPTBN1, TUBA1B, TUBA1C, TUBA4A, TUBB, TUBB4B, YBX3
Lipid antigen presentation by CD1 2.22E00 CALR, AP2A1, PDIA3, CANX
Guanine and guanosine salvage I 2.18E00 PNP
Myc mediated apoptosis signaling 2.17E00 FADD, SFN, YWHAB, YWHAE, YWHAG, YWHAQ, YWHAZ
Ephrin B signaling 2.14E00 CDC42, GNB1, CFL2, RAC2, RHOA
DNA double-strand break repair by nonhomologous end joining 2.13E00 XRCC1, XRCC4, PARP1
Urate biosynthesis/inosine 5′- phosphate degradation 2.12E00 PRKDC, XRCC6, XRCC5, PARP1
Telomere extension by Telomerase 2.11E00 HNRNPA1, HNRNPA2B1, XRCC5, XRCC6
Rac signaling 2.09E00 PAK2, CFL1, MAPK1, ARPC1B, RRAS, ITGA2, RAC1, IQGAP1, CDC42, CFL2, CD44, ARPC3, ARPC4, RHOA
VEGF signaling 2.08E00 ACTA1, ACTG1, ACTN1, ACTN4, EIF2S1, EIF2S2, EIF2S3, SFN, VCL, YWHAE
Formaldehyde oxidation II (glutathione-dependent) 2.05E00 ADH5, ESD
p53 signaling 2.03E00 PCNA, MAPK1
Apoptosis signaling 1.77E00 ACIN1, CAPNS1, MAPK1, RRAS, LMNA, CAPN2, SPTAN1, CYCS, CDK1, PARP1, AIFM1
Inosine-5′-phosphate biosynthesis II 1.73E00 PAICS, ATIC
Gap junction signaling 1.67E00 ACTA1, ACTG1, CAV1, EGFR, PDIA3, TUBA1B, TUBA1C, TUBA4A, TUBB, TUBB4B
Antigen presentation pathway 1.64E00 CALR, CANX, HLA-A, PDIA3, PSMB5
RAN signaling 1.62E00 CSE1L, KPNA2, KPNB1, RAN
eNOS signaling 1.51E00 HSP90AA1, CAV1
Pyruvate fermentation to lactate 1.45E00 LDHA, LDHB
Breast cancer regulation by Stathmin1 1.38E00 RHOA, PPP2R1A, TUBB6, RHOA, TUBA4A, PPP1CA
ERK5 signaling 1.35E00 EGFR, SFN, YWHAB, YWHAE, YWHAG, YWHAQ, YWHAZ
tRNA charging 1.26E00 AARS, GARS, RARS, TARS, WARS
Arginine biosynthesis IV 1.26E00 OAT, GLUD1
Mitotic roles of polo-like kinase 1.25E00 SLK, HSP90B1, PPP2R1A, HSP90AB1, PPP2CA, HSP90AA1, CAPN1
Death receptor signaling 1.23E00 ACIN1, CYCS, ACTB, FADD
Sucrose degradation V (Mammalian) 1.09E00 ALDOA, ALDOC, TPI1
Cdc42 signaling 1.08E00 ITGB1, ACTR2, PAK2, MYL6, ARPC1B, MAPK1, CFL1, HLA-A, ITGA2, IQGAP1, CDC42, ACTR3, CFL2, MYL12B, ARPC3, ARPC4
Xanthine and xanthosine salvage 1.02E00 PNP
Glutamate biosynthesis II 8.15E-01 GLUD1
Mitochondrial dysfunction 7.5E-01 CYTB, VDAC2
PTEN signaling 7.32E-01 MAPK1, YWHAH, RRAS, CSNK2A1, RAC1, CSNK2B, CDC42
Glutamate degradation X 7.01E-01 GLUD1
Fatty acid biosynthesis initiation II 6.5E-01 FASN
Neuregulin signaling 6.39E-01 EGFR, RPS6, HSP90B1, MAPK1, HSP90AB1, RRAS, ITGA2, HSP90AA1
Endoplasmic reticulum stress pathway 5.98E-01 CALR, EIF2S1, HSP90B1, HSPA5
Telomerase signaling 5.78E-01 EGFR, HSP90AA1, PPP2R1A
CDK5 signaling 5.46E-01 PPP2R1A, PPP1CA
Role of tissue factor in cancer 5.15E-01 EGFR, AKT2, CFL2, MAPK1
Isoleucine degradation I 5.06E-01 IMPDH2, PNP
P70S6K signaling 4.74E-01 EEF2, EGFR, PDIA3, PPP2R1A, SFN, YWHAB, YWHAE, YWHAG, YWHAQ, YWHAZ
Axonal guidance signaling 4.51E-01 DPYSL2, RAC2, AKT2, MYL6, PDIA3, TUBA4A, ACTR3, TUBB6, CFL2, RHOA, RTN4, ARPC3, PFN2, PFN1, PSMD14
Role of PKR in interferon induction and antiviral response 4.16E-01 CYCS
Protein kinase A signaling 3.54E-01 APEX1, FLNA, FLNB, GNB2L1, HSF3A/H3F3B, MYL12A, PDE6H, PDIA3, PPP1CA, PYGB, PYGL, RHOA, SFN, YWHAB, YWHAE, YWHAG, YWHAQ, YWHAZ
Macropinocytosis signaling 3.53E-01 RHOA, ITGB1, RRAS, RAC1, ACTN4
HIPPO signaling 3.27E-01 PPP1CA, PPP2R1A, SFN, YWHAB, YWHAE, YWHAG, YWHAQ, YWHAZ
Nitric oxide signaling in the cardiovascular system 2.86E-01 CAV1, HSP90AB1
Glucocorticoid receptor signaling 2.82E-01 HMGB1, AKT2, HSP90AA1, STAT1
Fcy receptor-mediated phagocytosis in macrophages and monocytes 2.5E-01 RAC, CDC42
Tec kinase signaling 2.49E-01 RHO, STAT, FADD
Activation of IRF by cytosolic pattern recognition receptors 2.45E-01 PPIB, MAVS, ADAR, ISG15, STAT2, CYPB
Prostate cancer signaling 2.22E-01 HSP90B1, MAPK1, PA2G4, HSP90AB1, RRAS, HSP90AA1, CTNNB1, GSTP1
HGF signaling 1.78E-01 AKT2

Abbreviations: ACT, actin; ACTN, actinin; ACTR3, ACTR3 actin-related 3 homolog; Akt, protein kinase B; ALDH, aldehyde dehydrogenase; ARPC, actin related protein 2/3 complex; CAV1, caveolin 1; CDC, cell division cycle; CDK, cyclin-dependent kinase; COPA, coatomer protein complex subunit alpha; CTNN, cadherin-associated protein; DNM1L, dynamin 1-like; EGFR, epidermal growth factor receptor; EIF, eukaryotic initiation factor; eNOS, endothelial nitric oxide synthase; FADD, Fas (TNFRSF6)-associated via death domain; FLN, filamin; GSTP1, glutathione S-transferase pi 1; HLA-A, major histocompatibility complex class I; HGF, hepatocyte growth factor; HMGB1, high mobility group protein B1; HSP, heat shock protein; IQGAP1, IQ motif containing GTPase activating protein 1; ITGB, intergrin beta; NQO1, NAD(P)H: quinone oxidoreductase 1; MAPK, mitogen-activated protein kinase; mTOR, mammalian target of rapamycin; Nrf2, Nuclear factor erythroid 2-related factor 2; PA2G4, proliferation-associated 2G4; PAK, p21-activated kinase; PABPC1, poly(A) binding protein cytoplasmic 1; PI3K, phosphoinositide 3-kinase; PLB, plumbagin; PPIB, peptidylprolyl isomerase B; PSM, proteasome subunit; PTEN, phosphatase and tensin-like protein; RAR, retinoic acid receptor; RHO, Ras homolog gene family; RhoGDI, Rho GDP-dissociation inhibitor; RPS, ribosomal protein S; RPL, ribosomal protein L; S6K, S6 kinase; SOD, superoxide dismutase; STAT, signal transducer and activator of transcription; TUBA, tubulin alpha; VEGF, vascular endothelial growth factor; VIM, vimentin.

Figure 1.

Figure 1

Proteomic analysis reveals a network of signaling pathways regulated by PLB in SCC25 cells.

Notes: A network of signaling pathways was analyzed by IPA according to the 398 molecules and 101 related pathways that were regulated by PLB in SCC25 cells.

Abbreviations: IPA, Ingenuity Pathway Analysis; PLB, plumbagin.

PLB regulates cell cycle regulators of SCC25 cells

The cell cycle arresting effect of PLB is considered as a critical contributor to its anticancer activities. We treated SCC25 cells with 5 μM PLB for 24 hours, and then, cell samples were subject to quantitative proteomic analysis. The results showed that PLB regulated cell cycle at G2/M DNA damage checkpoint in SCC25 cells with the involvement of multiple functional proteins (Table 3). These included YWHAQ, PRKDC, YWHAG, YWHAE, YWHAH, YWHAB, YWHAZ, SFN, SKP1, and CDK1 at G2/M checkpoint (Figure 2).

Figure 2.

Figure 2

PLB regulates cell cycle at G2/M checkpoint in SCC25 cells.

Notes: SCC25 cells were treated with 5 μM PLB for 24 hours and the protein samples were subject to quantitative proteomic analysis. Red indicates upregulation; green indicates a downregulation. The intensity of green and red molecule colors indicates the degree of down- or upregulation, respectively. Solid arrows indicate direct interaction.

Abbreviations: PLB, plumbagin; UV, ultraviolet.

PLB regulates apoptosis in SCC25 cells

Apoptosis is the type I programmed cell death pathway and has been considered as a promising target for the treatment of cancer either via intrinsic (mitochondrial-mediated) or extrinsic (death receptor-mediated) apoptosis pathways. As listed in Table 3, PLB regulated apoptotic signaling pathway and death receptor signaling pathway involving a number of functional proteins. These included ACIN1, CAPNS1, MAPK1, RRAS, LMNA, CAPN2, SPTAN1, CYCS, CDK1, PARP1, AIFM1, FADD, and ACTB. Moreover, the IPA results showed that mTOR signaling pathway played a central role in the regulation of cell metabolism, growth, proliferation, and survival through the integration of both intracellular and extracellular signals (Table 3). We subsequently investigated extrinsic apoptosis mediated by FADD in SCC25 cells with the treatment of PLB.

PLB regulates EMT pathways in SCC25 cells

EMT has a close association with cell migration, invasion, and stemness. Suppressing the progress of EMT is thought to be clinically helpful for cancer therapy. We analyzed the effect of PLB on EMT-related proteins and signaling pathways using SILAC-based proteomic approach. The proteomic data showed that PLB regulated epithelial adherens junction signaling pathway in SCC25 cells involving a number of functional proteins, including ACTA1, ACTG1, ACTN1, ACTN4, ACTR3, ARPC1B, CTNNA1, CTNND1, DNM1L, EGFR, IQGAP1, JUP, MYH9, RAB7A, RHO1, TUBA1B, TUBA1C, TUBA4A, TUBB, TUBB4B, VAL, VCL, and ZYX.

PLB regulates redox homeostasis involving Nrf2-mediated signaling pathways in SCC25 cells

Induction of ROS generation plays a critical role in the cytokine production, contributing to the cancer cell killing effect of PLB. However, the regulatory effect of PLB on ROS generation-related molecules and signaling pathways has not been fully understood. In this study, we observed that PLB regulated several critical signaling pathways related to ROS generation and redox homeostasis in SCC25 cells. Our quantitative proteomic study showed that PLB treatment regulated Nrf2-mediated oxidative stress response and oxidative phosphorylation in SCC25 cells (Table 2). A number of functional proteins were found to be involved in these pathways, including ACTA1, ACTG1, CBR, DNAJA2, GSTP1, NQO1, HSP90AA1, PPIB, SOD1, STIP1, and VCP (Table 2). Of note, Nrf2-mediated signaling pathways have critical roles in the maintenance of intracellular redox homeostasis in response to various stimuli via regulating antioxidant responsive elements. The quantitative proteomic data suggest that modulation of the expression of functional proteins involved in Nrf2-mediated signaling pathways may contribute to the anticancer effect of PLB in the treatment of TSCC. The results showed that PLB can efficiently induce ROS generation and that this can be abolished by NAC and GSH. Hence, we subsequently studied the relationship of ROS-generation-inducing effect of PLB with other cellular biological functions, including cell cycle arrest, cell apoptosis, and EMT.

Verification of molecular targets of PLB in SCC25 cells by Western blotting assay

Our aforementioned quantitative proteomic studies have predicted and shown that PLB can modulate a number of signaling pathways and functional proteins related to cell proliferation, cell migration, cell death, and cell survival. On the basis of our previous experimental and present proteomic data in SCC25 cells, we further examined the effect of PLB with a focus on cell cycle, apoptosis, EMT, and redox homeostasis and its related signaling pathways to delineate the underlying mechanisms.

PLB induces G2/M arrest in SCC25 cells via downregulation of cyclin B1, CDK1/cdc2, and cdc25

First, we examined the effect of PLB on cell cycle distribution of SCC25 cells using a flow cytometer. PLB markedly induced a G2/M phase arrest (P<0.05, 0.01, or 0.001; Figure 3). Compared with the control cells (2.4%), the percentage of SCC25 cells in G2/M phase was increased in a concentration-dependent manner after PLB treatment (Figure 3A and B). The percentage of cells in G2/M phase was 5.8%, 9.1%, and 13.1% when treated with PLB at 0.1, 1, and 5 μM, respectively. On the other hand, PLB significantly decreased the percentage of SCC25 cells in G1 phase when treated with 5 μM in comparison to the control cells (P<0.001; Figure 3B). In a separate experiment, the effect of 5 μM PLB on cell cycle distribution was examined in SCC25 cells over 48 hours (Figure 3C and D). Compared to the control cells, the percentage of SCC25 cells in the G2/M phase was increased from 2.7% at basal level to 8.1%, 9.4%, and 11.1% after 6-, 24-, and 48-hour treatment with 5 μM PLB, respectively (P<0.001; Figure 3D), whereas 5 μM PLB treatment decreased the percentage of SCC25 cells in G1 phase from 48.8% at basal level to 41.3% after 48 hours of treatment (P<0.05; Figure 3D).

Figure 3.

Figure 3

PLB induces G2/M arrest in SCC25 cells.

Notes: Cell cycle distribution of SCC25 cells after the treatment of PLB in the concentration and time course experiments. (A) Representative flow cytometric plots of cell cycle distribution of SCC25 cells and (B) bar graphs showing the percentage of SCC25 cells in G1, S, and G2/M phases after the treatment of 0.1, 1, and 5 μM PLB for 24 hours. (C) Representative flow cytometric plots of cell cycle distribution of SCC25 cells and (D) bar graphs showing the percentage of SCC25 cells in G1, S and G2/M phases after the treatment of 5 μM PLB for 6, 24, and 48 hours. Data are the mean ± SD of three independent experiments. *P<0.05; **P<0.01; and ***P<0.001 by one-way ANOVA.

Abbreviations: PLB, plumbagin; ANOVA, analysis of variance; SD, standard deviation; PI, propidium iodide.

To explore the mechanisms for PLB-induced cell cycle arrest in SCC25 cells, the expression level of key regulators responsible for G2/M checkpoint was examined using Western blotting assay. CDK1/cdc2, cyclin B1, and cdc25 are important regulators for G2 to M phase transition and thus their expression level was determined in SCC25 cells. The expression level of cdc2 was marked suppressed in SCC25 cells after treatment with PLB at concentrations of 0.1, 1, and 5 μM for 24 hours (P<0.001; Figure 4A and C). Compared with the control cells, the expression level of cyclin B1 was decreased by 18.3%, 46.0%, and 62.3% when SCC25 cells were treated with 0.1, 1, and 5 μM PLB for 24 hours, respectively (P<0.01 or 0.001; Figure 4A and C). There was a 10.7%, 14.0%, and 35.3% reduction in the expression level of cdc25 in SCC25 cells when treated with PLB at 0.1, 1, and 5 μM for 24 hours, respectively (P<0.05, 0.01, or 0.001; Figure 4A and C). Next, we conducted separate experiment over 48 hours. In comparison to the control cells, the expression level of cdc2 was decreased by 18.3% and 19.0% when SCC25 cells were treated with 5 μM PLB for 24 and 48 hours, respectively (P<0.001; Figure 4B and D); there was a 34.3% and 49.3% reduction in the expression level of cyclin B1 when SCC25 cells were incubated with 5 μM PLB for 24 and 28 hours, respectively (P<0.001; Figure 4B and D); and the expression level of cdc25 was also markedly suppressed (P<0.01 or 0.001; Figure 4B and D). These results demonstrate that PLB downregulates cyclin B1, CDK1/cdc2, and cdc25 in SCC25 cells. Importantly, these results have confirmed the regulatory effect of PLB on cell proliferation-related signaling pathways, which was predicted by our proteomic studies.

Figure 4.

Figure 4

Figure 4

PLB regulates the expression of CDK1/cdc2, cyclin B1, and cdc25 in SCC25 cells.

Notes: SCC25 cells were treated with PLB in the concentration and time course experiments and the protein samples were subject to Western blotting assay. (A) Representative blots of CDK1/cdc2, cyclin B1, and cdc25 in SCC25 cells after the treatment of 0.1, 1, and 5 μM PLB for 24 hours, and (B) representative blots of CDK1/cdc2, cyclin B1, and cdc25 in SCC25 cells after the treatment of 5 μM PLB for 6, 24, and 48 hours. (C) Bar graphs showing the relative levels of CDK1/cdc2, cyclin B1, and cdc25 in SCC25 cells after the treatment of 0.1, 1, and 5 μM PLB for 24 hours, and (D) bar graphs showing the relative level of CDK1/cdc2, cyclin B1, and cdc25 in SCC25 cells after the treatment of 5 μM PLB for 6, 24, and 48 hours. Data are the mean ± SD of three independent experiments. *P<0.05; **P<0.01; and ***P<0.001 by one-way ANOVA.

Abbreviations: PLB, plumbagin; ANOVA, analysis of variance; SD, standard deviation.

PLB induces apoptosis via FADD-mediated extrinsic signaling pathway

Apoptosis is a typical type of programmed cell death that plays an important role in PLB-induced cancer cell death. Previously, we observed that PLB significantly induced intrinsic apoptosis in SCC25 cells in a concentration- and time-dependent manner.20 On the basis of the results hinted at by the proteomic results (Figure 5), we further tested the apoptosis inducing effect of PLB with a focus on FADD-mediated signaling pathway. First, we employed flow cytometry to analyze the apoptosis-inducing effect of PLB in SCC25 cells. Subsequently, the modulation effect of PLB on the expression of FADD, TRADD, DR5, and cleaved caspase 3 in SCC25 cells was measured with a consideration to fully understand the apoptosis-inducing effect of PLB via FADD-mediated extrinsic signaling pathway. Cells were treated with PLB at concentrations of 0.1, 1, and 5 μM for 24 hours. The apoptosis level was increased from 3.0% to 6.1%, 8.2%, and 20.3% when SCC25 cells were treated with 0.1, 1, and 5 μM for 24 hours, respectively (P<0.05, 0.01, or 0.001; Figure 6A and B). Next, we conducted the time course experiment, and the results showed that the apoptosis level was increased from 3.5% at basal level to 11.4%, 21.2%, and 35.7% when SCC25 cells were treated with 5 μM PLB for 6, 24, and 48 hours, respectively (P<0.001; Figure 6C and D). Furthermore, There was a 1.3- and 1.6-fold increase in the expression level of FADD after SCC25 cells were treated with 1 and 5 μM PLB for 24 hours, respectively (P<0.01 or 0.001; Figure 7A and C). The expression level of TRADD and DR5 was also increased when cells were treated with PLB (P<0.01 or 0.001; Figure 7A and C). The cleavage of caspase 3 is the determinant process in both intrinsic and extrinsic apoptosis. We found that PLB markedly increased the level of cleaved caspase 3 to 1.2- and 1.3-fold when SCC25 cells were treated with 1 and 5 μM for 24 hours, respectively (P<0.01 or 0.001; Figure 7A and C). In a separate experiment, we examined the apoptosis-inducing effect of PLB over 48 hours. The expression level of FADD was increased 1.5-, 1.5-, and 1.6-fold when SCC25 cells were treated with 5 μM for 6, 24, and 48 hours, respectively (P<0.001; Figure 7B and D). The expression level of DR5 was also markedly increased in comparison with the control cells (P<0.05 or 0.001; Figure 7B and D). Meanwhile, there was a 1.1-, 1.3-, and 1–2 fold rise in the expression level of cleaved caspase 3, when SCC25 cells were treated with 5 μM PLB for 6, 24, and 48 hours, respectively (P<0.01 or 0.001; Figure 7B and D). The expression level of TRADD was also remarkably increased when SCC25 cells were treated with 5 μM PLB for 48 hours. These results clearly show that PLB induces apoptosis by involving the FADD-mediated extrinsic pathway in SCC25 cells, and these data are in agreement with our proteomic findings.

Figure 5.

Figure 5

PLB regulates death receptor signaling in SCC25 cells.

Notes: SCC25 cells were treated with 5 μM PLB for 24 hours and the protein samples were subject to quantitative proteomic analysis. Red indicates upregulation; green indicates downregulation. The intensity of green and red molecule colors indicates the degree of down- or upregulation, respectively. Solid arrows indicate direct interaction and dashed arrows indicate indirect interaction. The arrow with white head indicates translocation. The arrow with gray head indicates activation, causation, expression, localization, membership, modification, molecular cleavage, phosphorylation, protein-DNA interactions, protein-RNA interactions, regulation of binding, transcription.

Abbreviation: PLB, plumbagin.

Figure 6.

Figure 6

PLB induces apoptosis in SCC25 cells.

Notes: Apoptosis induction effect of PLB in SCC25 cells was examined. (A) Representative flow cytometric plots of apoptosis in SCC25 cells and (B) bar graphs showing the percentage of total apoptosis in SCC25 cells after the treatment of 0.1, 1, and 5 μM PLB for 24 hours. (C) Representative flow cytometric plots of apoptosis in SCC25 cells and (D) bar graphs showing the total apoptosis in SCC25 cells after the treatment of 5 μM PLB for 6, 24, and 48 hours. Data are the mean ± SD of three independent experiments. *P<0.05; **P<0.01; and ***P<0.001 by one-way ANOVA.

Abbreviations: PLB, plumbagin; ANOVA, analysis of variance; SD, standard deviation.

Figure 7.

Figure 7

Figure 7

PLB regulates the expression of FADD, TRADD, and DR5 in SCC25 cells.

Notes: SCC25 cells were treated with PLB in the concentration and time course experiments and protein samples were subject to Western blotting assay. (A) Representative blots of FADD, TRADD, and DR5 in SCC25 cells after the treatment of 0.1, 1, and 5 μM PLB for 24 hours, and (B) representative blots of FADD, TRADD, and DR5 in SCC25 cells after the treatment of 5 μM PLB for 6, 24, and 48 hours. (C) Bar graphs showing the relative level of FADD, TRADD, and DR5 in SCC25 cells after the treatment of 0.1, 1, and 5 μM PLB for 24 hours, and (D) bar graphs showing the relative level of FADD, TRADD, and DR5 in SCC25 cells after the treatment of 5 μM PLB for 6, 24, and 48 hours. Data are the mean ± SD of three independent experiments. *P<0.05; **P<0.01; and ***P<0.001 by one-way ANOVA.

Abbreviations: PLB, plumbagin; ANOVA, analysis of variance; SD, standard deviation; FADD, Fas (TNFRSF6)-associated via death domain; TRADD, TNF1 receptor-associated death domain.

PLB inhibits EMT and stemness in SCC25 cells

EMT is a critical process involved in the invasion, metastasis, and stemness of cancer.35 EMT depends on a reduction in expression of cell adhesion molecules. Tight junctions function as complete barriers between epithelium and endothelium and contribute to the maintenance of cell polarity. Claudin and occludin proteins are integral structural and functional components of tight junctions.36,37 ZO-1, 2, and 3 are peripheral membrane adaptor proteins that link junctional transmem-brane proteins to the actin cytoskeleton.3840 Cadherins are a superfamily of transmembrane glycoproteins, which include N-, P-, R-, B-, and E-cadherins.41 E-cadherin is considered an active suppressor of invasion in many epithelial cancers.42 Cancer cells often have upregulated N-cadherin in addition to loss of E-cadherin.43 Furthermore, the cytoplasmic domain of classical cadherins interacts with β-catenin, γ-catenin, and p120 catenin.44,45 It is reported that snail can interact with β-catenin via Wnt signaling pathway.46 Herein, on the basis of the proteomic results (Figure 8), we verified the effect of PLB treatment on EMT-associated markers in SCC25 cells using Western blotting assay. Incubation of SCC25 cells with PLB resulted in a concentration- and time-dependent increase in the expression level of E-cadherin and a decrease in the expression level of N-cadherin (Figure 9A–D). There was a 1.2- and 1.8-fold increase in the expression of E-cadherin when treated with 1 and 5 μM PLB for 24 hours, respectively, whereas 5 μM PLB suppressed expression level of N-cadherin 23% (P<0.05 or 0.001; Figure 9A and C). When SCC25 cells were treated with 5 μM PLB over 48 hours, the expression level of E-cadherin was increased 1.2-, 1.4-, and 1.8-fold after the treatment of 5 μM PLB for 6, 24, and 48 hours, respectively (P<0.01 or 0.001; Figure 9B and D). The expression level of N-cadherin was decreased by 23.3% and 45.0% when SCC 25 cells were treated with 5 μM PLB for 24 and 48 hours, respectively (P<0.05 or 0.001; Figure 9B and D).

Figure 8.

Figure 8

PLB regulates remodeling of epithelial adherens junctions pathway in SCC25 cells.

Notes: SCC25 cells were treated with 5 μM PLB for 24 hours and the protein samples were subject to quantitative proteomic analysis. Red indicates upregulation; green indicates downregulation. The intensity of green and red molecule colors indicates the degree of down or upregulation, respectively. Solid arrows indicate direct interaction. The arrow with white head indicates translocation. The arrow with gray head indicates activation, causation, expression, localization, membership, modification, molecular cleavage, phosphorylation, protein-DNA interactions, protein-RNA interactions, regulation of binding, transcription.

Abbreviation: PLB, plumbagin.

Figure 9.

Figure 9

Figure 9

PLB regulates the expression of EMT-related markers in SCC25 cells.

Notes: SCC25 cells were treated with PLB in the concentration and time course experiments and protein samples were subject to Western blotting assay. (A) Representative blots of E-cadherin, N-cadherin, snail, slug, TCF8/ZEB1, vimentin, β-catenin, ZO-1, and claudin-1 in SCC25 cells after the treatment of 0.1, 1, and 5 μM PLB for 24 hours; (B) representative blots of E-cadherin, N-cadherin, snail, slug, TCF8/ZEB1, vimentin, β-catenin, ZO-1, and claudin-1 in SCC25 cells after the treatment of 5 μM PLB for 6, 24, and 48 hours. (C) Bar graphs showing the relative level of E-cadherin, N-cadherin, snail, slug, TCF8/ZEB1, vimentin, β-catenin, ZO-1, and claudin-1 in SCC25 cells after the treatment of 0.1, 1, and 5 μM PLB for 24 hours, and (D) bar graphs showing the relative level of E-cadherin, N-cadherin, snail, slug, TCF8/ZEB1, vimentin, β-catenin, ZO-1, and claudin-1 in SCC25 cells after the treatment of 5 μM PLB for 6, 24, and 48 hours. Data are the mean ± SD of three independent experiments. *P<0.05; **P<0.01; and ***P<0.001 by one-way ANOVA.

Abbreviations: PLB, plumbagin; ANOVA, analysis of variance; SD, standard deviation.

In order to further examine the effect of PLB on EMT in SCC25 cells, we measured the expression level of several key regulators of E-cadherin. Snail and slug (both zinc finger transcriptional factors) together with TCF8/ZEB1 are suppressors of E-cadherin in EMT. PLB significantly reduced the expression level of snail and slug in SCC25 (Figure 9A and C). When SCC25 cells were treated with 5 μM PLB for 24 hours, the expression level of snail and slug was decreased by 46% and 41%, respectively (Figure 9A and C). Furthermore, PLB induced a time-dependent reduction in the expression level of TCF-8/ZEB1 in SCC25 cells. The expression level of TCF-8/ZEB1 was decreased by 22.0%, 27.0%, and 70.3% when SCC25 cells were treated with 5 μM PLB for 6, 24, and 48 hours, respectively (Figure 9B and D).

Vimentin is a type III intermediate filament protein in mesenchymal cells. β-Catenin can act as an integral component of a protein complex in adherens junctions that helps cells maintain epithelial layers, and β-catenin participates in the Wnt signaling pathway as a downstream target. In SCC25 cells, PLB inhibited the expression of vimentin in a concentration- and time-dependent manner. The expression level of vimentin was decreased by 19.3% and 24.7% when cells were treated with 1 and 5 μM PLB for 24 hours, respectively (P<0.01; Figure 9A and C). Consistently, the expression level of vimentin was decreased by 21.0% and 51.3% when SCC25 cells were incubated with 5 μM PLB for 24 and 48 hours, respectively (P<0.01 or 0.001; Figure 9B and D). There was also a significant reduction in the expression level of β-catenin in both the concentration and time course experiment when SCC25 cells were treated with 5 μM PLB. PLB at 5 μM markedly decreased the expression level of β-catenin by 24.7% with a 24-hour incubation period (P<0.01; Figure 9A and C). Finally, the expression of tight junction proteins ZO-1 and claudin-1 were examined in SCC25 cells after the treatment of PLB. ZO-1 and -2 are required for tight junction formation and function. The effect of PLB on the expression of ZO-1 and claudin-1 in SCC25 cells increased significantly in a concentration- and time-dependent manner (Figure 9A and B). Treatment of cells with 5 μM PLB for 24 hours led to a 1.8- and 3.5-fold rise in the expression level of ZO-1 and claudin-1, compared to the control cells, respectively (P<0.001; Figure 9A and C). These results from Western blotting assay verified our proteomic data.

In addition, there is increasing evidence showing that CSCs can display EMT characteristics such as loss of the adhesion protein E-cadherin. This relationship drives us to think about the stemness-attenuating effect of PLB in SCC25 cells. Therefore, we primarily conducted the Western blotting assay to examine the key stemness markers, including Oct-4, Bmi-1, Nanog, and Sox-2. Oct-4, also known as Oct-3, belongs to the POU (Pit-Oct-Unc) transcription factor family and plays an important role during early embryogenesis.47 Sox-2 is a member of the Sox gene family that encodes transcription factors and plays an important role in the maintenance of stemness.48 Nanog transcription factor cooperates with Oct-4 and Sox-2 and is identified as a key CSCs marker.49 Bmi-1 is a transcriptional repressor that belongs to the polycomb-group family of proteins that determine the proliferation and senescence of normal and CSCs.50 The Western blotting results showed that PLB significantly decreased the expression level of Oct-4, Sox-2, Nanog, and Bmi-1. Incubation of SCC25 cells with 5 μM PLB remarkably decreased the expression level of Oct-4, Sox-2, Nanog, and Bni-1 by 35.7%, 27.0%, 70.7%, and 38.3%, respectively, compared with the control cells (P<0.001; Figure 10A and C). In a separate experiment, we evaluated the effect of different incubation times with 5 μM PLB. Totally, the expression levels of Bmi-1, Nanog, and Sox-2 were decreased significantly after 6 hours of incubation, while the Oct-4 expression level was decreased significantly after 24 hours of incubation (Figure 10B and D). These results indicate that PLB suppresses the stemness of human TSCC cells.

Figure 10.

Figure 10

PLB regulates the expression of stemness markers in SCC25 cells.

Notes: SCC25 cells were treated with PLB in the concentration and time course experiments and protein samples were subject to Western blotting assay. (A) Representative blots of Oct-4, Bmi-1, Nanog, and Sox-2 in SCC25 cells after the treatment of 0.1, 1, and 5 μM PLB for 24 hours, and (B) representative blots of Oct-4, Bmi-1, Nanog, and Sox-2 in SCC25 cells after the treatment of 5 μM PLB for 6, 24, and 48 hours. (C) Bar graphs showing the relative levels of Oct-4, Bmi-1, Nanog, and Sox-2 in SCC25 cells after the treatment of 0.1, 1, and 5 μM PLB for 24 hours, and (D) bar graphs showing the relative levels of Oct-4, Bmi-1, Nanog, and Sox-2 in SCC25 cells after the treatment of 5 μM PLB for 6, 24, and 48 hours. Data are the mean ± SD of three independent experiments. *P<0.05; **P<0.01; and ***P<0.001 by one-way ANOVA.

Abbreviations: PLB, plumbagin; ANOVA, analysis of variance; SD, standard deviation.

PLB induces intracellular ROS generation and regulates redox homeostasis via suppressing Nrf2-mediated oxidative signaling pathway in SCC25 cells

As shown in the proteomic results, PLB exhibited a regulating effect on intracellular redox homeostasis in SCC25 cells, which may possibly contribute to the cell cycle arresting, apoptosis inducing, EMT inhibiting, and stemness-attenuating effects of PLB. Herein, we examined the role of Nrf2-mediated signaling pathway in PLB’s beneficial action in SCC25 cells. Nrf2, also known as Nfe2I2, is a nuclear factor that controls the expression of various detoxifying enzymes, ROS elimination proteins, drug transporters, and antiapoptotic proteins. Normally, Nrf2 is suppressed in the cytoplasm by the interaction with Kelch-like ECH-associated protein 1 (Keap1) and Cullin3. Once exposed to the antioxidant response element (ARE)-mediated inducers, Nrf2 will translocate to the nucleus where it forms a heterodimer with a small Maf protein and binds to other nuclear factors and initiates the transcription of antioxidative genes.51,52 As noted earlier, the proteomic results indicated that Nrf2-oxidative signaling pathway responded to the PLB treatment in SCC25 cells (Figure 11). Therefore, we analyzed this pathway using Western blotting assay. First, SCC25 cells were treated with 0.1, 1, and 5 μM PLB for 24 hours and their nuclear proteins were extracted. The results showed that the expression levels of nuclear (n)-Nrf2, NQO1, GST, and HSP90 were all decreased, while the expression level of cytosolic (c)-Nrf2 was increased in SCC25 cells, compared with the control cells (Figure 12A). The ratio of n-Nrf2 to c-Nrf2 was decreased by 13.3% and 37.0% when SCC25 cells were incubated with 1 and 5 μM PLB for 24 hours, respectively (P<0.01 or 0.001; Figure 12C). In addition, the expression levels of c-Nrf2, n-Nrf2, NQO1, GST, and HSP90 were also examined with the treatment of 5 μM PLB over a 48-hour treatment period. The ratio of n-Nrf2 to c-Nrf2 was decreased by 28.3% and 39.0% when SCC25 cells were incubated with 5 μM PLB for 24 and 48 hours, respectively (P<0.001; Figure 12D). The expression levels of NQO1, GST, and HSP90 were all markedly decreased (P<0.05 or 0.001; Figure 12D). Taken together, the results show that Nrf2-oxidative signaling pathway is involved in the ROS-generation-inducing effect of PLB, contributing to the regulatory activities of PLB on intracellular redox homeostasis in SCC25 cells (Figure 13).

Figure 11.

Figure 11

PLB regulates Nrf2 oxidative signaling pathway in SCC25 cells.

Notes: SCC25 cells were treated with 5 μM PLB for 24 hours and the protein samples were subject to quantitative proteomic analysis. Red indicates upregulation; green indicates downregulation. The intensity of green and red molecule colors indicates the degree of down or upregulation, respectively. Solid arrows indicate direct interaction and dashed arrows indicate indirect interaction.

Abbreviations: PLB, plumbagin; UV, ultraviolet.

Figure 12.

Figure 12

PLB regulates the expression of Nrf2, NQO1, GST, and HSP90 in SCC25 cells.

Notes: SCC25 cells were treated with PLB in the concentration and time course experiments and protein samples were subject to Western blotting assay. (A) Representative blots of c-Nrf2, n-Nrf2, NQO1, GST, and HSP90 in SCC25 cells after the treatment of 0.1, 1, and 5 μM PLB for 24 hours, and (B) representative blots of Nrf2, NQO1, GST, and HSP90 in SCC25 cells after the treatment of 5 μM PLB for 6, 24, and 48 hours. (C) Bar graphs showing the relative level of Nrf2, NQO1, GST, and HSP90 in SCC25 cells after the treatment of 0.1, 1, and 5 μM PLB for 24 hours, and (D) bar graphs showing the relative levels of Nrf2, NQO1, GST, and HSP90 in SCC25 cells after the treatment of 5 μM PLB for 6, 24, and 48 hours. Data are the mean ± SD of three independent experiments. *P<0.05; **P<0.01; and ***P<0.001 by one-way ANOVA.

Abbreviations: PLB, plumbagin; ANOVA, analysis of variance; SD, standard deviation.

Figure 13.

Figure 13

Schematic diagram shows that PLB regulates the Nrf2-mediated oxidative signaling pathway and its downstream proteins NQO1, GST, and HSP90 in SCC25 cells.

Abbreviations: ROS, reactive oxygen species; GST, glutathione S-transferase.

Relationship between PLB-induced ROS generation and PLB-mediated cell cycle arrest, apoptosis induction, EMT inhibition, and stemness attenuation

The relationship between PLB-induced ROS generation and PLB-mediated cell cycle arrest, apoptosis induction, EMT inhibition, and stemness attenuation was further examined in SCC25 cells with the application of ROS scavengers (NAC and GSH). SCC25 cells were treated with 5 μM PLB, 100 μM NAC, 1 mM GSH, 5 μM PLB plus 100 μM NAC, and 5 μM PLB plus 1 mM GSH, respectively. As shown in Figures 1416, the cell cycle arresting, apoptosis inducing, EMT inhibiting, and stemness-attenuating effects of PLB were abolished by NAC and GSH. The percentage of cells in G2/M phase was decreased by 53.2% and 61.4% in SCC25 cells when cells were coincubated with NAC and PLB or GSH and PLB, compared to PLB-treated cells, respectively (P<0.01 or 0.001; Figure 14B). In addition, in comparison to PLB-treated cells, NAC and GSH ablated PLB-induced apoptosis 53.3% and 55.6%, respectively (P<0.001; Figure 15B). Furthermore, the expression of EMT and stemness representative markers were measured (Figure 16). Compared to PLB-treated cells, the expression level of E-cadherin was decreased by 30.4% and 36.2% when cells were cotreated with NAC and PLB or GSH and PLB, respectively (P<0.001; Figure 16A and B). On the contrary, the expression level of N-cadherin, Oct-4, Bmi-1, and Nanog was increased when cells were coincubated with NAC and PLB or GSH and PLB (Figure 16A and B). However, the expression level of Sox-2 did not show statistical significance (Figure 16A and B). This may be partially ascribed to PLB-mediated other pathways that downregulate the Sox-2 level. Taken together, PLB-induced ROS generation may interact with the actions of PLB-mediated cell cycle arrest, apoptosis induction, EMT inhibition, and stemness attenuation.

Figure 14.

Figure 14

The effect of ROS scavengers NAC and GSH in PLB-induced G2/M arrest in SCC25 cells.

Notes: (A) Representative flow cytometric plots of cell cycle distribution of SCC25 and (B) bar graphs showing the percentage of SCC25 cells in G1, S and G2/M phases after the treatment of 5 μM PLB, 100 μM NAC, 1 mM GSH, 5 μM PLB plus 100 μM NAC, and 5 μM PLB plus 1 mM GSH for 24 hours. Data are the mean ± SD of three independent experiments. **P<0.01; and ***P<0.001 by one-way ANOVA.

Abbreviations: ROS, reactive oxygen species; NAC, N-acetyl-l-cysteine; GSH, l-glutathione; PLB, plumbagin; ANOVA, analysis of variance; SD, standard deviation.

Figure 15.

Figure 15

The effect of ROS scavengers NAC and GSH in PLB-induced apoptosis in SCC25 cells.

Notes: (A) Representative flow cytometric plots of apoptosis in SCC25 and (B) bar graphs showing the apoptotic percentage of SCC25 cells after the treatment of 5 μM PLB, 100 μM NAC, 1 mM GSH, 5 μM PLB plus 100 μM NAC, and 5 μM PLB plus 1 mM GSH for 24 hours. Data are the mean ± SD of three independent experiments. ***P<0.001 by one-way ANOVA.

Abbreviations: ROS, reactive oxygen species; NAC, N-acetyl-l-cysteine; GSH, l-glutathione; PLB, plumbagin; ANOVA, analysis of variance; SD, standard deviation.

Figure 16.

Figure 16

Figure 16

The effect of ROS scavengers NAC and GSH in PLB-mediated EMT inhibition and stemness attenuation in SCC25 cells.

Notes: (A) Representative blots of E-cadherin, N-cadherin, Oct-4, Bmi-1, Nanog, and Sox-2 in SCC25 cells and (B) bar graphs showing the relative level of E-cadherin, N-cadherin, Oct-4, Bmi-1, Nanog, and Sox-2 in SCC25 cells after the treatment of 5 μM PLB, 100 μM NAC, 1 mM GSH, 5 μM PLB plus 100 μM NAC, and 5 μM PLB plus 1 mM GSH for 24 hours. Data are the mean ± SD of three independent experiments. ***P<0.001 by one-way ANOVA.

Abbreviations: ROS, reactive oxygen species; NAC, N-acetyl-l-cysteine; GSH, l-glutathione; EMT, epithelial to mesenchymal transition; PLB, plumbagin; ANOVA, analysis of variance; SD, standard deviation.

Discussion

TSCC remains one of the devastating malignancies in oral and maxillofacial tumors. TSCC is notorious for its lymphatic metastasis and relapse. Although sequential treatments are available, including radiotherapy, surgery, and chemotherapy, the therapeutic efficacy is not so optimistic.1 This is partially due to hyperactive cell survival pathways and radiotherapy/chemotherapy resistance.53,54 It is urgent to probe into the corresponding molecular alterations and seek novel effective drugs for TSCC treatment. PLB is an active naphthoquinone constituent isolated from the roots of Plumbaginaceae plants.17 It has been reported that PLB exhibits anticancer activities with minimal side effect in vitro and in vivo, which is greatly ascribed to its effects on multiple signaling pathways related to ROS generation, apoptosis, and autophagy.23,55,56 In this study, we employed a SILAC-based quantitative proteomic study to obtain a comprehensive view of the proteomic response to PLB treatment in TSCC cell line SCC25, and the findings have shown that PLB regulates a variety of functional protein molecules and signaling pathways involved in critical cellular processes. Further validation results have shown that PLB induces G2/M arrest and extrinsic apoptosis, but inhibits EMT and stemness via ROS generation through Nrf2-mediated oxidative signaling pathway in TSCC cell line SCC25 cells.

The SILAC-based proteomic approach can provide a system-level analysis to tackle the challenges in cancer treatment, such as chemoresistance. One study applied SILAC-based quantitative proteomic approach to analyze differences in protein expression level between parental hepatocellular carcinoma cell line HuH-7 and sorafenib-acquired resistance HuH-7 (HuH-7R) cells. Results indicated that galectin-1 is a predictive marker of sorafenib resistance and a downstream target of the Akt/mTOR/HIF-1a signaling pathway.57 The SILAC-based proteomic approach can also quantitatively evaluate the effect of a given compound or drug and identify its potential molecular targets and related signaling pathways.5860 For example, the SILAC-based proteomic approach was used to screen the therapeutic targets of histone deacetylases inhibitor vorinostat in human breast cancer MDA-MB-231 cell line, and the results found that 61 proteins were lysine acetylated by vironostat.30 This study demonstrated that PLB modulated a plethora of protein molecules, of which the expression levels of 143 protein molecules were increased while the levels of 255 protein molecules were decreased. Furthermore, 101 signaling pathways were potentially regulated by PLB in SCC25 cells. The following proteins are widely involved in cell survival, cell proliferation, redox homeostasis, cell metabolism, cell migration, and cell death: YWHAQ, PRKDC, YWHAG, YWHAE, YWHAH, YWHAB, YWHAZ, SFN, SKP1, CDK1, ACIN1, CAPNS1, MAPK1, RRAS, LMNA, CAPN2, SPTAN1, CYCS, PARP1, AIFM1, FADD, ACTB, ACTA1, ACTG1, ACTN1, ACTN4, ACTR3, ARPC1B, CTNNA1, CTNND1, DNM1L, EGFR, IQGAP1, JUP, MYH9, RAB7A, RHO1, TUBA1B, TUBA1C, TUBA4A, TUBB, TUBB4B, VAL, VCL, ZYX, CBR, DNAJA2, GSTP1, NQO1, HSP90AA1, PPIB, SOD1, STIP1, and VCP. The network of signaling pathways was mainly related to cell cycle distribution, cell migration, redox hemostasis, and cell death. The top ten targeted signaling pathways were EIF2 signaling pathway, regulation of eIF4 and p70S6K signaling, remodeling of epithelial adherens junctions pathway, mTOR signaling pathway, protein ubiquitination pathway, Nrf2-mediated oxidative stress response signaling pathway, epithelial adherens junction signaling pathway, caveolar-mediated endocytosis signaling pathway, RhoA signaling pathway, and oxidative phosphorylation pathway. The proteomic results indicate that PLB may target these molecules and related signaling pathways to elicit its anticancer effects in the treatment of TSCC. Notably, we have observed a differential effect of PLB on turmoral and nontumoral cells, with a higher half-maximal inhibitory concentration toward to nontumoral cells than that to the corresponding tumoral cells,20 which renders PLB a promising anticancer drug candidate.

Subsequently, we further validated the proteomic responses to PLB in SCC25 cells. We found that PLB induced G2/M arrest in SCC25 cells in a concentration- and time-dependent manner. Meanwhile, the expression level of key regulators of G2/M phase, such as cdc2, cyclin B1, and cdc25, were decreased after the treatment of PLB. It has been reported that cell cycle progression is tightly regulated by cyclins and CDKs.61 The complex formed by the association of CDK1/cdc2 and Cyclin B1 plays a major role in the entry of cells into mitosis. Phosphorylation of CDK1/cdc2 at Thr161 by CDK-activating kinase is essential for CDK1/cdc2 kinase activity. The process of the CDK1/cdc2 phosphorylation and dephosphorylation are mediated by the nuclear kinase Wee1 and the dual-specificity phosphatase cdc25. Wee1 can stop mitosis by inhibiting the CDK1/cdc2 phosphorylation, while cdc25 can help entry into mitosis by eliminating the inhibitory phosphorylation.61 Thus, taking the proteomic, flow cytometric, and Western blotting results into consideration, PLB-induced cell cycle arrest may be mediated through the regulation of key modulators controlling the G2/M check point in SCC25 cells.

Apoptosis is a conserved physiological mechanism that is important during embryogenesis and homeostasis of tissue. Dysregulated apoptosis has been implicated in many diseases including cancer.62 The successful execution of apoptosis is crucial for many chemotherapy drugs.63 It is widely accepted that there are two types of apoptosis: intrinsic apoptosis and extrinsic apoptosis. Mitochondrial disruption and the subsequent cytochrome c release can initiate the caspase-dependent apoptosis. Bcl-2 family plays an important role during this process.64,65 Previously, our findings showed that PLB triggered the mitochondrial-mediated apoptosis in SCC25 cells.20 In this study, the proteomic study showed that PLB regulated mitochondrial function and death receptor signaling pathway. We found that PLB could induce FADD-mediated extrinsic apoptosis in SCC25 cells. The proteomic results hinted that PLB regulated the death receptor signaling and that the protein molecule FADD was upregulated by PLB. FADD is a key adaptor protein for death receptor-mediated apoptosis. The death domain of FADD binds to the death domain of death receptor. Subsequently, procaspase-8 is recruited and caspase 3, 6, and 7 are cleaved. Finally, apoptosis is induced; and both intrinsic and extrinsic apoptosis converge into the cleavage of caspase 3. TRADD is a tumor necrosis factor receptor 1 (TNFR1) associated signal transducer that enhances association of FADD with TNFR1.66,67 DR5, also known as TRAIL-R2, can bind to adaptor molecules FADD and TRADD.68 In this study, we found that PLB increased the level of FADD, TRADD, and DR5. This indicated the extrinsic apoptosis-inducing effect of PLB in SCC25. Together with the results from our previous paper,20 it seems that PLB can efficiently induce both intrinsic and extrinsic apoptosis in SCC25 cells.

TSCC is notorious for its metastasis and relapse, and acquisition of EMT and induction of CSC-like properties are inevitably responsible for the metastasis and relapse.69 EMT is a highly conserved biological process that converts epithelial cells into mesenchymal cells via the modulation of various transcription factors.7073 EMT has been classified into three types according to specific physiological context. Type I EMT plays an important role in embryogenesis and organ development. Type II EMT functions in wound healing and tissue regeneration. Type III EMT, the one we focus on, is involved in cancer metastasis and CSC formation.35,74 The EMT process is characterized by loss of epithelial markers such as E-cadherin, claudin, and occludin, and acquisition of mesenchymal markers such as N-cadherin, vimentin, snail, slug, and ZO-1.75,76 Many signaling pathways, such as Wnt, TGF-β, and STAT3, can mediate the EMT process via activating a plethora of transcriptional regulators of mesenchymal markers.75,76 Furthermore, CSCs can display EMT characteristics such as loss of adhesion protein E-cadherin.11 Static CSCs that have moved to distant sites might be responsible for metastases and relapse, especially after curative surgical treatment of a primary tumor.77 Seminal findings demonstrate that EMT activators, such as Twist1 and Prrx1, can serve as a direct molecular link between EMT and stemness.78,79 Our proteomic study showed that PLB could regulate the epithelial adherens junctions pathway in SCC25 cells. The Western blotting assay validated that PLB could increase the expression level of E-cadherin and decrease the level of N-cadherin in SCC25 cells. Furthermore, we examined other key regulators of EMT. We found that PLB significantly reduced the expression level of snail, slug, TCF-8/ZEB1, β-catenin, and vimentin, while increasing the expression level of claudin-1 and ZO-1. All these implied the EMT-inhibiting role of PLB in SCC25 cells. Subsequently, CSCs markers were also measured, and the expression levels of Oct-4, Bmi-1, Nanog, and Sox-2 were all found to be decreased by PLB in a concentration- and time-dependent manner in SCC25 cells. Taken together, PLB can inhibit EMT and attenuate stemness in SCC25 cells. We can speculate that there are interconnections between EMT and stemness, such as the loss of E-cadherin. Given that PLB can regulate the key factors in EMT, the stemness property may more or less be affected. However, the exact network between EMT and stemness needs further investigation. Importantly, it has been reported that ROS may link the EMT and stemness via glucose metabolism.80 In our previous paper,20 we found that PLB can efficiently induce ROS generation, which can be abolished by ROS scavengers NAC and GSH. On the basis of the results of the present proteomic analysis, PLB can efficiently regulate the Nrf2-mediated oxidative stress response signaling pathway. Nrf2, also known as Nfe2I2, is a nuclear factor that controls the expression of various detoxifying enzymes, ROS elimination proteins, drug transporters, and antiapoptotic proteins. Normally, Nrf2 is suppressed in the cytoplasm by combination with Keap1 and Cullin3. Once exposed to the ARE-mediated inducers, Nrf2 will translocate to the nucleus where it forms a heterodimer with a small Maf protein and binds to other nuclear factors and initiates the transcription of antioxidative genes.51,52 The Western blotting results showed that the ratio of n-Nrf2 to c-Nrf2 was significantly decreased. Furthermore, the downstream effectors NQO1, GST, and HSP90 were examined. These effectors are involved in ROS elimination and detoxification.8183 Results showed that PLB can decrease the expression level of NQO1, GST, and HSP90 in SCC25 cells. We can carefully draw a conclusion that PLB can kill TSCC cells via ROS generation and attenuation of ROS elimination factors. Finally, we employed the ROS scavengers NAC and GSH to further examine the effect of PLB in cell cycle arrest, apoptosis induction, EMT inhibition, and stemness attenuation. Interestingly, results showed that the cell cycle arresting, apoptosis inducing, EMT inhibiting, and stemness-attenuating effect of PLB can be efficiently abolished by NAC and GSH. This highlights the ROS-generation-inducing effect of PLB as an upstream effector to regulate other downstream biological activities such as cell cycle arrest and apoptosis.

In summary, the quantitative SILAC-based proteomic approach showed that PLB inhibited cell proliferation, activated death receptor-mediated apoptotic pathway, remodeled epithelial adherens junctions pathway, and increased intracellular level of ROS via Nrf2-mediated oxidative stress response signaling pathway in human SCC25 cells involving a number of key functional proteins. This study may provide a clue to fully identify the molecular targets and elucidate the underlying mechanisms of PLB in the treatment of TSCC.

Acknowledgments

The authors appreciate the financial support by the National Natural Science Foundation of China (grant number: 81560440) and the Key Project of Chinese Medicine Research Plan of Jiangxi province (grant number: 2013Z010).

Footnotes

Disclosure

The authors report no conflicts of interest in the work.

References

  • 1.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 2015;65(1):5–29. doi: 10.3322/caac.21254. [DOI] [PubMed] [Google Scholar]
  • 2.Li R, Koch WM, Fakhry C, Gourin CG. Distinct epidemiologic characteristics of oral tongue cancer patients. Otolaryngol Head Neck Surg. 2013;148(5):792–796. doi: 10.1177/0194599813477992. [DOI] [PubMed] [Google Scholar]
  • 3.Joseph LJ, Goodman M, Higgins K, et al. Racial disparities in squamous cell carcinoma of the oral tongue among women: a SEER data analysis. Oral Oncol. 2015;51(6):586–592. doi: 10.1016/j.oraloncology.2015.03.010. [DOI] [PubMed] [Google Scholar]
  • 4.Markopoulos AK. Current aspects on oral squamous cell carcinoma. Open Dent J. 2012;6:126–130. doi: 10.2174/1874210601206010126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Dogan E, Cetinayak HO, Sarioglu S, Erdag TK, Ikiz AO. Patterns of cervical lymph node metastases in oral tongue squamous cell carcinoma: implications for elective and therapeutic neck dissection. J Laryngol Otol. 2014;128(3):268–273. doi: 10.1017/S0022215114000267. [DOI] [PubMed] [Google Scholar]
  • 6.Liu F, Kong X, Lv L, Gao J. TGF-beta1 acts through miR-155 to down-regulate TP53INP1 in promoting epithelial-mesenchymal transition and cancer stem cell phenotypes. Cancer Lett. 2015;359(2):288–298. doi: 10.1016/j.canlet.2015.01.030. [DOI] [PubMed] [Google Scholar]
  • 7.Donnenberg VS, Donnenberg AD. Stem cell state and the epithelial-to-mesenchymal transition: implications for cancer therapy. J Clin Pharmacol. 2015;55(6):603–619. doi: 10.1002/jcph.486. [DOI] [PubMed] [Google Scholar]
  • 8.Perdigao-Henriques R, Petrocca F, Altschuler G, et al. miR-200 promotes the mesenchymal to epithelial transition by suppressing multiple members of the Zeb2 and Snail1 transcriptional repressor complexes. Oncogene. 2015 doi: 10.1038/onc.2015.69. Epub. [DOI] [PubMed] [Google Scholar]
  • 9.Brabletz T. EMT and MET in metastasis: where are the cancer stem cells? Cancer Cell. 2012;22(6):699–701. doi: 10.1016/j.ccr.2012.11.009. [DOI] [PubMed] [Google Scholar]
  • 10.Morrison SJ, Kimble J. Asymmetric and symmetric stem-cell divisions in development and cancer. Nature. 2006;441(7097):1068–1074. doi: 10.1038/nature04956. [DOI] [PubMed] [Google Scholar]
  • 11.Martin-Belmonte F, Perez-Moreno M. Epithelial cell polarity, stem cells and cancer. Nat Rev Cancer. 2012;12(1):23–38. doi: 10.1038/nrc3169. [DOI] [PubMed] [Google Scholar]
  • 12.Boumahdi S, Driessens G, Lapouge G, et al. SOX2 controls tumour initiation and cancer stem-cell functions in squamous-cell carcinoma. Nature. 2014;511(7508):246–250. doi: 10.1038/nature13305. [DOI] [PubMed] [Google Scholar]
  • 13.Chiou SH, Yu CC, Huang CY, et al. Positive correlations of Oct-4 and Nanog in oral cancer stem-like cells and high-grade oral squamous cell carcinoma. Clin Cancer Res. 2008;14(13):4085–4095. doi: 10.1158/1078-0432.CCR-07-4404. [DOI] [PubMed] [Google Scholar]
  • 14.Molofsky AV, Pardal R, Iwashita T, et al. Bmi-1 dependence distinguishes neural stem cell self-renewal from progenitor proliferation. Nature. 2003;425(6961):962–967. doi: 10.1038/nature02060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Kagias K, Ahier A, Fischer N, Jarriault S. Members of the NODE (Nanog and Oct4-associated deacetylase) complex and SOX-2 promote the initiation of a natural cellular reprogramming event in vivo. Proc Natl Acad Sci U S A. 2012;109(17):6596–6601. doi: 10.1073/pnas.1117031109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Stuckey DW, Shah K. Stem cell-based therapies for cancer treatment: separating hope from hype. Nat Rev Cancer. 2014;14(10):683–691. doi: 10.1038/nrc3798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Padhye S, Dandawate P, Yusufi M, Ahmad A, Sarkar FH. Perspectives on medicinal properties of plumbagin and its analogs. Med Res Rev. 2012;32(6):1131–1158. doi: 10.1002/med.20235. [DOI] [PubMed] [Google Scholar]
  • 18.Sung B, Oyajobi B, Aggarwal BB. Plumbagin inhibits osteoclastogenesis and reduces human breast cancer-induced osteolytic bone metastasis in mice through suppression of RANKL signaling. Mol Cancer Ther. 2012;11(2):350–359. doi: 10.1158/1535-7163.MCT-11-0731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Sinha S, Pal K, Elkhanany A, et al. Plumbagin inhibits tumorigenesis and angiogenesis of ovarian cancer cells in vivo. Int J Cancer. 2013;132(5):1201–1212. doi: 10.1002/ijc.27724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Pan ST, Qin Y, Zhou ZW, et al. Plumbagin induces G2/M arrest, apoptosis, and autophagy via p38 MAPK- and PI3K/Akt/mTOR-mediated pathways in human tongue squamous cell carcinoma cells. Drug Des Devel Ther. 2015;9:1601–1626. doi: 10.2147/DDDT.S76057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Li YC, He SM, He ZX, et al. Plumbagin induces apoptotic and autophagic cell death through inhibition of the PI3K/Akt/mTOR pathway in human non-small cell lung cancer cells. Cancer Lett. 2014;344(2):239–259. doi: 10.1016/j.canlet.2013.11.001. [DOI] [PubMed] [Google Scholar]
  • 22.Sandur SK, Ichikawa H, Sethi G, Ahn KS, Aggarwal BB. Plumbagin (5-hydroxy-2-methyl-1,4-naphthoquinone) suppresses NF-kappaB activation and NF-kappaB-regulated gene products through modulation of p65 and IkappaBalpha kinase activation, leading to potentiation of apoptosis induced by cytokine and chemotherapeutic agents. J Biol Chem. 2006;281(25):17023–17033. doi: 10.1074/jbc.M601595200. [DOI] [PubMed] [Google Scholar]
  • 23.Kuo PL, Hsu YL, Cho CY. Plumbagin induces G2-M arrest and autophagy by inhibiting the AKT/mammalian target of rapamycin pathway in breast cancer cells. Mol Cancer Ther. 2006;5(12):3209–3221. doi: 10.1158/1535-7163.MCT-06-0478. [DOI] [PubMed] [Google Scholar]
  • 24.Wang F, Wang Q, Zhou ZW, et al. Plumbagin induces cell cycle arrest and autophagy and suppresses epithelial to mesenchymal transition involving PI3K/Akt/mTOR-mediated pathway in human pancreatic cancer cells. Drug Des Devel Ther. 2015;9:537–560. doi: 10.2147/DDDT.S73689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Zhou ZW, Li XX, He ZX, et al. Induction of apoptosis and autophagy via sirtuin1- and PI3K/Akt/mTOR-mediated pathways by plumbagin in human prostate cancer cells. Drug Des Devel Ther. 2015;9:1511–1554. doi: 10.2147/DDDT.S75976. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 26.Sun J, McKallip RJ. Plumbagin treatment leads to apoptosis in human K562 leukemia cells through increased ROS and elevated TRAIL receptor expression. Leuk Res. 2011;35(10):1402–1408. doi: 10.1016/j.leukres.2011.06.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Xu KH, Lu DP. Plumbagin induces ROS-mediated apoptosis in human promyelocytic leukemia cells in vivo. Leuk Res. 2010;34(5):658–665. doi: 10.1016/j.leukres.2009.08.017. [DOI] [PubMed] [Google Scholar]
  • 28.Lee JH, Yeon JH, Kim H, et al. The natural anticancer agent plumbagin induces potent cytotoxicity in MCF-7 human breast cancer cells by inhibiting a PI-5 kinase for ROS generation. PLoS One. 2012;7(9):e45023. doi: 10.1371/journal.pone.0045023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Edelmann MJ, Shack LA, Naske CD, Walters KB, Nanduri B. SILAC-based quantitative proteomic analysis of human lung cell response to copper oxide nanoparticles. PLoS One. 2014;9(12):e114390. doi: 10.1371/journal.pone.0114390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Zhou Q, Chaerkady R, Shaw PG, et al. Screening for therapeutic targets of vorinostat by SILAC-based proteomic analysis in human breast cancer cells. Proteomics. 2010;10(5):1029–1039. doi: 10.1002/pmic.200900602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Ong SE, Mann M. A practical recipe for stable isotope labeling by amino acids in cell culture (SILAC) Nat Protoc. 2006;1(6):2650–2660. doi: 10.1038/nprot.2006.427. [DOI] [PubMed] [Google Scholar]
  • 32.Pan ST, Zhou ZW, He ZX, et al. Proteomic response to 5,6-dimethylxanthenone 4-acetic acid (DMXAA, vadimezan) in human non-small cell lung cancer A549 cells determined by the stable-isotope labeling by amino acids in cell culture (SILAC) approach. Drug Des Devel Ther. 2015;9:937–968. doi: 10.2147/DDDT.S76021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Qiu JX, Zhou ZW, He ZX, et al. Plumbagin elicits differential proteomic responses mainly involving cell cycle, apoptosis, autophagy, and epithelial-to-mesenchymal transition pathways in human prostate cancer PC-3 and DU145 cells. Drug Des Devel Ther. 2015;9:349–417. doi: 10.2147/DDDT.S71677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Qin Y, Zhou ZW, Pan ST, et al. Graphene quantum dots induce apoptosis, autophagy, and inflammatory response via p38 mitogen-activated protein kinase and nuclear factor-kappaB mediated signaling pathways in activated THP-1 macrophages. Toxicology. 2015;327:62–76. doi: 10.1016/j.tox.2014.10.011. [DOI] [PubMed] [Google Scholar]
  • 35.Thiery JP, Lim CT. Tumor dissemination: an EMT affair. Cancer Cell. 2013;23(3):272–273. doi: 10.1016/j.ccr.2013.03.004. [DOI] [PubMed] [Google Scholar]
  • 36.Stebbing J, Filipovic A, Giamas G. Claudin-1 as a promoter of EMT in hepatocellular carcinoma. Oncogene. 2013;32(41):4871–4872. doi: 10.1038/onc.2012.591. [DOI] [PubMed] [Google Scholar]
  • 37.Furuse M, Hirase T, Itoh M, et al. Occludin: a novel integral membrane protein localizing at tight junctions. J Cell Biol. 1993;123(6 Pt 2):1777–1788. doi: 10.1083/jcb.123.6.1777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Furuse M, Itoh M, Hirase T, et al. Direct association of occludin with ZO-1 and its possible involvement in the localization of occludin at tight junctions. J Cell Biol. 1994;127(6 Pt 1):1617–1626. doi: 10.1083/jcb.127.6.1617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Haskins J, Gu L, Wittchen ES, Hibbard J, Stevenson BR. ZO-3, a novel member of the MAGUK protein family found at the tight junction, interacts with ZO-1 and occludin. J Cell Biol. 1998;141(1):199–208. doi: 10.1083/jcb.141.1.199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Umeda K, Ikenouchi J, Katahira-Tayama S, et al. ZO-1 and ZO-2 independently determine where claudins are polymerized in tight-junction strand formation. Cell. 2006;126(4):741–754. doi: 10.1016/j.cell.2006.06.043. [DOI] [PubMed] [Google Scholar]
  • 41.Gheldof A, Berx G. Cadherins and epithelial-to-mesenchymal transition. Prog Mol Biol Transl Sci. 2013;116:317–336. doi: 10.1016/B978-0-12-394311-8.00014-5. [DOI] [PubMed] [Google Scholar]
  • 42.Janda E, Nevolo M, Lehmann K, et al. Raf plus TGFbeta-dependent EMT is initiated by endocytosis and lysosomal degradation of E-cadherin. Oncogene. 2006;25(54):7117–7130. doi: 10.1038/sj.onc.1209701. [DOI] [PubMed] [Google Scholar]
  • 43.Zhang X, Liu G, Kang Y, et al. N-cadherin expression is associated with acquisition of EMT phenotype and with enhanced invasion in erlotinib-resistant lung cancer cell lines. PLoS One. 2013;8(3):e57692. doi: 10.1371/journal.pone.0057692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Polakis P. More than one way to skin a catenin. Cell. 2001;105(5):563–566. doi: 10.1016/s0092-8674(01)00379-8. [DOI] [PubMed] [Google Scholar]
  • 45.Eger A, Stockinger A, Park J, et al. Beta-Catenin and TGFbeta signalling cooperate to maintain a mesenchymal phenotype after FosER-induced epithelial to mesenchymal transition. Oncogene. 2004;23(15):2672–2680. doi: 10.1038/sj.onc.1207416. [DOI] [PubMed] [Google Scholar]
  • 46.Stemmer V, de Craene B, Berx G, Behrens J. Snail promotes Wnt target gene expression and interacts with beta-catenin. Oncogene. 2008;27(37):5075–5080. doi: 10.1038/onc.2008.140. [DOI] [PubMed] [Google Scholar]
  • 47.Koo BS, Lee SH, Kim JM, et al. Oct4 is a critical regulator of stemness in head and neck squamous carcinoma cells. Oncogene. 2015;34(18):2317–2324. doi: 10.1038/onc.2014.174. [DOI] [PubMed] [Google Scholar]
  • 48.Sarkar A, Hochedlinger K. The sox family of transcription factors: versatile regulators of stem and progenitor cell fate. Cell Stem Cell. 2013;12(1):15–30. doi: 10.1016/j.stem.2012.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Hayashi Y, Caboni L, Das D, et al. Structure-based discovery of NANOG variant with enhanced properties to promote self-renewal and reprogramming of pluripotent stem cells. Proc Natl Acad Sci U S A. 2015;112(15):4666–4671. doi: 10.1073/pnas.1502855112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Lessard J, Sauvageau G. Bmi-1 determines the proliferative capacity of normal and leukaemic stem cells. Nature. 2003;423(6937):255–260. doi: 10.1038/nature01572. [DOI] [PubMed] [Google Scholar]
  • 51.DeNicola GM, Karreth FA, Humpton TJ, et al. Oncogene-induced Nrf2 transcription promotes ROS detoxification and tumorigenesis. Nature. 2011;475(7354):106–109. doi: 10.1038/nature10189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Zhang Y, Gordon GB. A strategy for cancer prevention: stimulation of the Nrf2-ARE signaling pathway. Mol Cancer Ther. 2004;3(7):885–893. [PubMed] [Google Scholar]
  • 53.Tsurutani J, West KA, Sayyah J, Gills JJ, Dennis PA. Inhibition of the phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin pathway but not the MEK/ERK pathway attenuates laminin-mediated small cell lung cancer cellular survival and resistance to imatinib mesylate or chemotherapy. Cancer Res. 2005;65(18):8423–8432. doi: 10.1158/0008-5472.CAN-05-0058. [DOI] [PubMed] [Google Scholar]
  • 54.Zhao G, Cai C, Yang T, et al. MicroRNA-221 induces cell survival and cisplatin resistance through PI3K/Akt pathway in human osteosarcoma. PLoS One. 2013;8(1):e53906. doi: 10.1371/journal.pone.0053906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Aziz MH, Dreckschmidt NE, Verma AK. Plumbagin, a medicinal plant-derived naphthoquinone, is a novel inhibitor of the growth and invasion of hormone-refractory prostate cancer. Cancer Res. 2008;68(21):9024–9032. doi: 10.1158/0008-5472.CAN-08-2494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Wang CC, Chiang YM, Sung SC, et al. Plumbagin induces cell cycle arrest and apoptosis through reactive oxygen species/c-Jun N-terminal kinase pathways in human melanoma A375.S2 cells. Cancer Lett. 2008;259(1):82–98. doi: 10.1016/j.canlet.2007.10.005. [DOI] [PubMed] [Google Scholar]
  • 57.Yeh CC, Hsu CH, Shao YY, et al. Integrated SILAC and iTRAQ quantitative proteomic analysis identifies galectin-1 as a potential biomarker for predicting sorafenib resistance in liver cancer. Mol Cell Proteomics. 2015;14(6):1527–1545. doi: 10.1074/mcp.M114.046417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Dolai S, Xu Q, Liu F, Molloy MP. Quantitative chemical proteomics in small-scale culture of phorbol ester stimulated basal breast cancer cells. Proteomics. 2011;11(13):2683–2692. doi: 10.1002/pmic.201000801. [DOI] [PubMed] [Google Scholar]
  • 59.Geiger T, Cox J, Ostasiewicz P, Wisniewski JR, Mann M. Super-SILAC mix for quantitative proteomics of human tumor tissue. Nat Methods. 2010;7(5):383–385. doi: 10.1038/nmeth.1446. [DOI] [PubMed] [Google Scholar]
  • 60.Everley PA, Krijgsveld J, Zetter BR, Gygi SP. Quantitative cancer proteomics: stable isotope labeling with amino acids in cell culture (SILAC) as a tool for prostate cancer research. Mol Cell Proteomics. 2004;3(7):729–735. doi: 10.1074/mcp.M400021-MCP200. [DOI] [PubMed] [Google Scholar]
  • 61.Hunter T. Protein kinases and phosphatases: the yin and yang of protein phosphorylation and signaling. Cell. 1995;80(2):225–236. doi: 10.1016/0092-8674(95)90405-0. [DOI] [PubMed] [Google Scholar]
  • 62.Evan GI, Vousden KH. Proliferation, cell cycle and apoptosis in cancer. Nature. 2001;411(6835):342–348. doi: 10.1038/35077213. [DOI] [PubMed] [Google Scholar]
  • 63.Reed JC. Apoptosis-based therapies. Nat Rev Drug Discov. 2002;1(2):111–121. doi: 10.1038/nrd726. [DOI] [PubMed] [Google Scholar]
  • 64.Boehning D, Patterson RL, Sedaghat L, et al. Cytochrome c binds to inositol (1,4,5) trisphosphate receptors, amplifying calcium-dependent apoptosis. Nat Cell Biol. 2003;5(12):1051–1061. doi: 10.1038/ncb1063. [DOI] [PubMed] [Google Scholar]
  • 65.Yang J, Liu X, Bhalla K, et al. Prevention of apoptosis by Bcl-2: release of cytochrome c from mitochondria blocked. Science. 1997;275(5303):1129–1132. doi: 10.1126/science.275.5303.1129. [DOI] [PubMed] [Google Scholar]
  • 66.Pobezinskaya YL, Liu Z. The role of TRADD in death receptor signaling. Cell Cycle. 2012;11(5):871–876. doi: 10.4161/cc.11.5.19300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Park YC, Ye H, Hsia C, et al. A novel mechanism of TRAF signaling revealed by structural and functional analyses of the TRADD-TRAF2 interaction. Cell. 2000;101(7):777–787. doi: 10.1016/s0092-8674(00)80889-2. [DOI] [PubMed] [Google Scholar]
  • 68.Zlotorynski E. Apoptosis. DR5 unfolds ER stress. Nat Rev Mol Cell Biol. 2014;15(8):498–499. doi: 10.1038/nrm3843. [DOI] [PubMed] [Google Scholar]
  • 69.Biddle A, Mackenzie IC. Cancer stem cells and EMT in carcinoma. Cancer Metastasis Rev. 2012;31(1–2):285–293. doi: 10.1007/s10555-012-9345-0. [DOI] [PubMed] [Google Scholar]
  • 70.Okada T, Sinha S, Esposito I, et al. The Rho GTPase Rnd1 suppresses mammary tumorigenesis and EMT by restraining Ras-MAPK signalling. Nat Cell Biol. 2015;17(1):81–94. doi: 10.1038/ncb3082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Kudo-Saito C, Shirako H, Takeuchi T, Kawakami Y. Cancer metastasis is accelerated through immunosuppression during Snail-induced EMT of cancer cells. Cancer Cell. 2009;15(3):195–206. doi: 10.1016/j.ccr.2009.01.023. [DOI] [PubMed] [Google Scholar]
  • 72.Ansieau S, Bastid J, Doreau A, et al. Induction of EMT by twist proteins as a collateral effect of tumor-promoting inactivation of premature senescence. Cancer Cell. 2008;14(1):79–89. doi: 10.1016/j.ccr.2008.06.005. [DOI] [PubMed] [Google Scholar]
  • 73.Waerner T, Alacakaptan M, Tamir I, et al. ILEI: a cytokine essential for EMT, tumor formation, and late events in metastasis in epithelial cells. Cancer Cell. 2006;10(3):227–239. doi: 10.1016/j.ccr.2006.07.020. [DOI] [PubMed] [Google Scholar]
  • 74.Qureshi R, Arora H, Rizvi MA. EMT in cervical cancer: its role in tumour progression and response to therapy. Cancer Lett. 2015;356(2 Pt B):321–331. doi: 10.1016/j.canlet.2014.09.021. [DOI] [PubMed] [Google Scholar]
  • 75.De Craene B, Berx G. Regulatory networks defining EMT during cancer initiation and progression. Nat Rev Cancer. 2013;13(2):97–110. doi: 10.1038/nrc3447. [DOI] [PubMed] [Google Scholar]
  • 76.Rhim AD, Mirek ET, Aiello NM, et al. EMT and dissemination precede pancreatic tumor formation. Cell. 2012;148(1–2):349–361. doi: 10.1016/j.cell.2011.11.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Clevers H. The cancer stem cell: premises, promises and challenges. Nat Med. 2011;17(3):313–319. doi: 10.1038/nm.2304. [DOI] [PubMed] [Google Scholar]
  • 78.Mani SA, Guo W, Liao MJ, et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133(4):704–715. doi: 10.1016/j.cell.2008.03.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Ocana OH, Corcoles R, Fabra A, et al. Metastatic colonization requires the repression of the epithelial-mesenchymal transition inducer Prrx1. Cancer Cell. 2012;22(6):709–724. doi: 10.1016/j.ccr.2012.10.012. [DOI] [PubMed] [Google Scholar]
  • 80.Schieber MS, Chandel NS. ROS links glucose metabolism to breast cancer stem cell and EMT phenotype. Cancer Cell. 2013;23(3):265–267. doi: 10.1016/j.ccr.2013.02.021. [DOI] [PubMed] [Google Scholar]
  • 81.Matsui A, Ikeda T, Enomoto K, et al. Increased formation of oxidative DNA damage, 8-hydroxy-2′-deoxyguanosine, in human breast cancer tissue and its relationship to GSTP1 and COMT genotypes. Cancer Lett. 2000;151(1):87–95. doi: 10.1016/s0304-3835(99)00424-3. [DOI] [PubMed] [Google Scholar]
  • 82.Beck R, Dejeans N, Glorieux C, et al. Hsp90 is cleaved by reactive oxygen species at a highly conserved N-terminal amino acid motif. PLoS One. 2012;7(7):e40795. doi: 10.1371/journal.pone.0040795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Li L, Dong H, Song E, et al. Nrf2/ARE pathway activation, HO-1 and NQO1 induction by polychlorinated biphenyl quinone is associated with reactive oxygen species and PI3K/AKT signaling. Chem Biol Interact. 2014;209:56–67. doi: 10.1016/j.cbi.2013.12.005. [DOI] [PubMed] [Google Scholar]

Articles from Drug Design, Development and Therapy are provided here courtesy of Dove Press

RESOURCES