Supplementary Table S1. List of chemical inhibitors utilized in the current study. Supplementary Information.

Slides:



Advertisements
Similar presentations
Supplementary Data shLuc sh   sh  179 shLuc sh   sh  179 Asyn Noco 2.1% 6.1% 2.4% 16.5%11.4%1.1% 5.9%3.9%2.5% 52.5%10.0%4.9%
Advertisements

Supplementary Fig. 1. (a) Cells were treated with DMSO or with 1 µM of SK-7068 or LAQ824 for 18 hours. They were then fixed with 70% ethanol, stained with.
E2 GSCE2 bulkG7 GSCG7 bulk Mean SF4Gy (95% CI) 0.78 (0.72, 0.83) 0.56 (0.47, 0.64) 0.65 (0.57,0.72) 0.43 (0.34,0.51) T test of meansp = 0.001p =
Supplementary Figure 1A-B A H1299 RH LC3-I / II P-GSK-3 (Ser21/9) GSK-3α/β β-actin β-catenine P-GSK-3 (Tyr279/216) B.
Table S1. HTS positive hits.. Figure S1. Isogenic bortezomib (Btz) resistant mouse and human cell models. The indicated human (MM.1S and U266) and mouse.
Supplementary Figure 1. The effect of 17-DMAG on the growth of lung cancer cells with Met amplification Tumor cells were continuously treated with increasing.
A B Supplementary Figure 1. (A) Western blots were performed on the three cell lines to analyze the levels of p-ERK, which is a key protein involved in.
Date of download: 7/12/2016 Copyright © 2016 American Medical Association. All rights reserved. From: Inhibition of the Growth of Papillary Thyroid Carcinoma.
Date of download: 9/17/2016 Copyright © 2016 American Medical Association. All rights reserved. From: Differential Responses of Human Papillary Thyroid.
Copyright © 2007 American Medical Association. All rights reserved.
B A P-AXL P-AXL AXL AXL P-MET P-MET MET MET P-Akt-473 P-Akt-473 AKT
Copyright © 2006 American Medical Association. All rights reserved.
RanBP3 Regulates Melanoma Cell Proliferation via Selective Control of Nuclear Export  Gaurav Pathria, Bhavuk Garg, Christine Wagner, Kanika Garg, Melanie.
RhoD Inhibits RhoC-ROCK-Dependent Cell Contraction via PAK6
Aurora Kinase A Is Upregulated in Cutaneous T-Cell Lymphoma and Represents a Potential Therapeutic Target  Daniel Humme, Ahmed Haider, Markus Möbs, Hiroshi.
A B C Supplementary Figure S1. Trabectedin decreases viability of primary MPM cell cultures. A and B, dose-dependent impact of trabectedin on epithelioid.
Volume 33, Issue 2, Pages (January 2009)
RanBP3 Regulates Melanoma Cell Proliferation via Selective Control of Nuclear Export  Gaurav Pathria, Bhavuk Garg, Christine Wagner, Kanika Garg, Melanie.
Acriflavine Inhibits Acquired Drug Resistance by Blocking the Epithelial-to-Mesenchymal Transition and the Unfolded Protein Response  Jeroen Dekervel,
P90RSK Blockade Inhibits Dual BRAF and MEK Inhibitor-Resistant Melanoma by Targeting Protein Synthesis  Nicholas Theodosakis, Goran Micevic, Casey G.
Effects of Betulinic Acid Alone and in Combination with Irradiation in Human Melanoma Cells  Edgar Selzer, Emilio Pimentel, Volker Wacheck, Werner Schlegel,
NF1 silencing confers resistance of PC9 lung adenocarcinoma cells to erlotinib (erl). NF1 silencing confers resistance of PC9 lung adenocarcinoma cells.
Marissa V. Powers, Paul A. Clarke, Paul Workman  Cancer Cell 
Oncogenic BRAF Regulates Oxidative Metabolism via PGC1α and MITF
BET inhibition and depletion repress the expression of BRCA1 and RAD51
Volume 27, Issue 1, Pages (January 2015)
Volume 19, Issue 8, Pages (August 2017)
SOX10 Promotes Melanoma Cell Invasion by Regulating Melanoma Inhibitory Activity  Saskia A. Graf, Christian Busch, Anja-Katrin Bosserhoff, Robert Besch,
Marissa V. Powers, Paul A. Clarke, Paul Workman  Cancer Cell 
Meghan Bliss-Moreau, Cristian Coarfa, Preethi H
Supplementary Figure text
Volume 57, Issue 5, Pages (March 2015)
Drug network derived from the core EGFR interactome and combination strategy to overcome the resistant cells. Drug network derived from the core EGFR interactome.
Kavitha Gowrishankar, Stephanie Snoyman, Gulietta M. Pupo, Therese M
Inhibition of PI3K-AKT-mTOR Signaling Sensitizes Melanoma Cells to Cisplatin and Temozolomide  Tobias Sinnberg, Konstantinos Lasithiotakis, Heike Niessner,
Opposing Roles of JNK and p38 in Lymphangiogenesis in Melanoma
NF1 downregulation activates MAPK pathway signaling.
RasGRP3 Mediates MAPK Pathway Activation in GNAQ Mutant Uveal Melanoma
Alexander J. Lakhter, Ravi P. Sahu, Yang Sun, William K
Crosstalk between ROR1 and BCR pathways defines novel treatment strategies in mantle cell lymphoma by Hanna Karvonen, David Chiron, Wilhelmiina Niininen,
The EGF receptor confers BRAF inhibitor resistance in BRAF-mutant melanoma cells. The EGF receptor confers BRAF inhibitor resistance in BRAF-mutant melanoma.
TBP 200 LC2/ad MGH134 MGH134 CCDC6-RET MGH134 EV CCDC6-RET PC9
Inhibition of Human Melanoma Cell Growth by the Dietary Flavonoid Fisetin Is Associated with Disruption of Wnt/β-Catenin Signaling and Decreased Mitf.
Selective Inhibition of p300 HAT Blocks Cell Cycle Progression, Induces Cellular Senescence, and Inhibits the DNA Damage Response in Melanoma Cells  Gai.
Jinhua Wang, Sharon K. Huang, Diego M. Marzese, Sandy C. Hsu, Neal P
Yao Zhan, Michael S. Dahabieh, Arjuna Rajakumar, Monica C
Inhibition of CRM1-Mediated Nucleocytoplasmic Transport: Triggering Human Melanoma Cell Apoptosis by Perturbing Multiple Cellular Pathways  Gaurav Pathria,
Volume 6, Issue 5, Pages (May 2016)
Volume 20, Issue 6, Pages (December 2011)
Supplementary Figures
Plk1 negatively regulates cortical localization of dynein, NuMA, and LGN during metaphase. Plk1 negatively regulates cortical localization of dynein, NuMA,
Regulated P124SMEK1 expression cannot alter p-ERK levels or sensitivity to BRAF or MEK inhibition in V600EBRAF melanoma cell lines. Regulated P124SMEK1.
Volume 16, Issue 24, Pages (December 2006)
Oncogenic Ras-Induced Expression of Noxa and Beclin-1 Promotes Autophagic Cell Death and Limits Clonogenic Survival  Mohamed Elgendy, Clare Sheridan,
Volume 22, Issue 1, Pages (January 2015)
Opposing Roles of JNK and p38 in Lymphangiogenesis in Melanoma
Differential Regulation of Cyclooxygenase-2 Expression by Phytosphingosine Derivatives, NAPS and TAPS, and its Role in the NAPS or TAPS-Mediated Apoptosis 
Targeted Depletion of Polo-Like Kinase (Plk) 1 Through Lentiviral shRNA or a Small- Molecule Inhibitor Causes Mitotic Catastrophe and Induction of Apoptosis.
Volume 52, Issue 2, Pages (October 2013)
Macrophage Inhibitory Cytokine-1 Is Overexpressed in Malignant Melanoma and Is Associated with Tumorigenicity  Glen M. Boyle, Julie Pedley, Adam C. Martyn,
MELK Promotes Melanoma Growth by Stimulating the NF-κB Pathway
Confirmation of synergy and cytotoxicity of genotype-selective drug pairs. Confirmation of synergy and cytotoxicity of genotype-selective drug pairs. A,
Gold Nanoparticles for BCR-ABL1 Gene Silencing: Improving Tyrosine Kinase Inhibitor Efficacy in Chronic Myeloid Leukemia  Raquel Vinhas, Alexandra R.
Fig. 2. Ex vivo inducible knockout of PDCD2 in ESCs results in loss of S phase entry and increased p53.(A) Growth curve of inducible knockout and WT ESCs.
Defective Cell Cycle Checkpoint Functions in Melanoma Are Associated with Altered Patterns of Gene Expression  William K. Kaufmann, Kathleen R. Nevis,
Deon G. Uffort, Elizabeth A. Grimm, Julie A. Ellerhorst 
Targeting HR via CDK inhibition resensitizes recurrent cultures to temozolomide (TMZ). Targeting HR via CDK inhibition resensitizes recurrent cultures.
Differential effects of the RAF inhibitor PLX4032 in BRAF-mutant melanoma, thyroid, and colorectal cancer cell lines. Differential effects of the RAF inhibitor.
THZ1 in combination with targeted therapy enhances cell death and hinders the establishment of drug-resistant colonies in diverse oncogene-addicted cellular.
Volume 7, Issue 3, Pages (September 2016)
Presentation transcript:

Supplementary Table S1. List of chemical inhibitors utilized in the current study. Supplementary Information

Supplementary Table S2. iTRAQ-generated list of kinases showing differential affinities for Midostaurin (Mido) and Sunitinib (Suni).

Supplementary Table S3. (1D) LC-MS-generated list of kinases showing highest affinities for Midostaurin (Mido) and Sunitinib (Suni). pc – peptide count.

Supplementary Table S4. Mutational status of key molecules in the utilized melanoma cell lines. HD: homozygous deletion; PM: point mutation; WT: wild-type; ND: not determined; NF: non functional. Courtesy: sangar.ac.uk, cansar.icr.ac.uk, Wistar Institute, Philadelphia, PA.

Supplementary Figure S1. MITF confers sensitivity to MAPK inhibitors in melanocytes. (A) HMEL-B and HMEL-B/M cells were treated as indicated for 24 hours followed by the assessment of viability relative to DMSO control. Note: The observed increased viability in HMEL-B/M cells precluded the inclusion of this data in Fig. 1B. (B) HMEL-B and HMEL-B/M cells were treated with distinct MAPK pathway inhibitors (BRAF inhibitors: GDC-0879 (1 µM), PLX-4032 (1 µM); MEK inhibitors: CI (1 µM) PD98059 (1 µM); CRAF inhibitor: Sorafenib (5 µM) for 24 hours followed by the assessment of cell viability relative to DMSO control. (C) UACC-62 and HMEL-B/M cells were treated with DMSO control or the indicated MAPK inhibitors (BRAF inhibitor: PLX-4032 (1 µM); MEK inhibitors: CI-1040 (1 µM), PD98059 (1 µM), and U0126 (1 µM) ) for 24 hours followed by the assessment of indicated proteins by immunoblotting. Refer to Supplementary Table 1 for Drug concentration in (A) and (B). AB C

A B C D Supplementary Figure S2. Integrative selection of kinase target(s). (A) Phase-contrast images of HMEL-B/M and HMEL-B cells, treated as indicated with Midostaurin and Sunitinib for 4 weeks in a soft-agar colony formation assay. (B) HMEL-B/M cells were treated as indicated for 4 weeks in a soft-agar colony formation assay followed capturing of images, utilizing a phase- contrast microscope (n=3). (Inhibitors: SB for GSK3A; and MLN8237 for AURKA) (C) Left, HMEL-B cells were treated with GSK3A-i (SB415286) and AURKA-i (MLN8237), as indicated, for 4 weeks in a soft-agar colony formation assay followed by colony count (n=3); Right, representative images (D) Left, HMEL-B/M cells were treated with DMSO control or an increasing conc. (as indicated) of MLN8237 for 4 weeks in a soft-agar colony formation assay followed by colony count (n=3); Right, representative images. All error bars indicate ±SD; ns-non-significant; *P ≤ 0.05, **P ≤

B A Supplementary Figure S3. Role of AURKA in melanoma biology. (A) Indicated melanoma cell lines were treated with DMSO control or MLN8237 (1 µM) for 48 hours followed by AnnexinV/Propidium Iodide (PI) staining. Percentages on the bottom correspond to the early apoptotic (AnnexinV-positive)+late apoptotic (AnnexinV+PI-positive) cells. (B) Indicated melanoma cell lines were treated with DMSO control or MLN8237 (1 µM) for 48 hours followed by the immunoblotting for APAF-1. (C) BRAF inhibitor (PLX-4032)-sensitive (451Lu and WM983B) and the paired resistant (451Lu_BR and WM983B_BR) melanoma cell lines were treated as indicated for 48 hours followed by AnnexinV/Propidium Iodide (PI) staining. MLN8237 (1 µM) and PLX-4032 (1 µM) were used. Resistant melanoma cells were constantly maintained in RPMI+10% FCS containing 1 µM PLX. (D) Dermal Fibroblast cells FB2003 were treated with DMSO control and the indicated conc. of MLN8237 for 48 hours followed by AnnexinV/Propidium Iodide (PI) staining (E) Left, Dermal Fibroblast cells FB2003 were treated with MLN8237 as indicated for 24 and 48 hrs followed by the assessment of viability relative to DMSO control (n=3); Right, Scaled comparison to the relative viability of melanoma cell lines and immortalized human melanocytes upon MLN8237 treatment; the last piece of data also shown as Figure 3a. C D E

Supplementary Figure S4. Role of AURKA in melanoma biology. (A) The indicated melanoma cell lines were tested for migration (left panel) upon treatment with DMSO control or MLN8237 (40 nM) (n=3). See Methods for details. (Middle panel) corresponding cell loading, and (right panel) the corresponding changes in cell viability (B) Corresponding representative images from the migation assay. All error bars indicate ±SD; ***P ≤ B A

B D C CDKN1ACDKN1B CCND1BCL-2 MITF A Supplementary Figure S5. AURKA regulates MITF and the associated transcriptional program. (A) The indicated melanoma cells were treated with AURKA-i (MLN8237) at different concentrations followed by assessment of MITF levels by immunoblotting. (B-D) UACC-62 cells were treated as indicated for 18 hours followed by quantification of MITF (B), CDKN1A and CDKN1B (C), and CCND1 and BCL-2 (D) mRNA levels by qRT-PCR. MLN8237 (1 µM) was used. All error bars indicate ±SD.

Supplementary Figure S6. PLK inhibition overcomes MITF-mediated intrinsic resistance. HMEL-B and HMEL-B/M cells were treated for 24 hours with PLK1 inhibitor BI-2536 (20 nM) followed by the assessment of cell viability relative to DMSO control (n=3).