Yao Zhan, Michael S. Dahabieh, Arjuna Rajakumar, Monica C

Slides:



Advertisements
Similar presentations
IL-18 Downregulates Collagen Production in Human Dermal Fibroblasts via the ERK Pathway  Hee Jung Kim, Seok Bean Song, Jung Min Choi, Kyung Moon Kim,
Advertisements

Volume 145, Issue 4, Pages e2 (October 2013)
Juewon Kim, Hyunjung Choi, Eun-Gyung Cho, Tae R. Lee 
A Signal Transduction Pathway from TGF-β1 to SKP2 via Akt1 and c-Myc and its Correlation with Progression in Human Melanoma  Xuan Qu, Liangliang Shen,
Jasmine George, Minakshi Nihal, Chandra K
Cdc42 Inhibits ERK-Mediated Collagenase-1 (MMP-1) Expression in Collagen-Activated Human Keratinocytes  Maryam G. Rohani, Brian K. Pilcher, Peter Chen,
Yongping Shao, Kaitlyn Le, Hanyin Cheng, Andrew E. Aplin 
Translational Repression Protects Human Keratinocytes from UVB-Induced Apoptosis through a Discordant eIF2 Kinase Stress Response  Ann E. Collier, Ronald.
RAF Inhibition Overcomes Resistance to TRAIL-Induced Apoptosis in Melanoma Cells  Anja Berger, Sandra-Annika Quast, Michael Plötz, Nicholas-Frederik Kuhn,
Identification of miR-145 as a Key Regulator of the Pigmentary Process
BAP1 Has a Survival Role in Cutaneous Melanoma
Plakophilin 2 Affects Cell Migration by Modulating Focal Adhesion Dynamics and Integrin Protein Expression  Jennifer L. Koetsier, Evangeline V. Amargo,
Volume 145, Issue 4, Pages e2 (October 2013)
Amanda Croft, Kwang H. Tay, Suzanah C. Boyd, Su T. Guo, Chen C
SOX10 Promotes Melanoma Cell Invasion by Regulating Melanoma Inhibitory Activity  Saskia A. Graf, Christian Busch, Anja-Katrin Bosserhoff, Robert Besch,
Volume 120, Issue 6, Pages (March 2005)
Tie2-R849W Mutant in Venous Malformations Chronically Activates a Functional STAT1 to Modulate Gene Expression  Hsiao-Tang Hu, Yi-Hsien Huang, Yi-Ann.
Cadherin 11, a miR-675 Target, Induces N-Cadherin Expression and Epithelial– Mesenchymal Transition in Melasma  Nan-Hyung Kim, Soo-Hyun Choi, Tae Ryong.
Andreea M. Bujor, Jaspreet Pannu, Shizhong Bu, Edwin A. Smith, Robin C
Arsenite-Mediated Promotion of Anchorage-Independent Growth of HaCaT Cells through Placental Growth Factor  Ichiro Yajima, Mayuko Y. Kumasaka, Shoko Ohnuma,
Histamine Contributes to Tissue Remodeling via Periostin Expression
Stefan W. Stoll, Jessica L. Johnson, Yong Li, Laure Rittié, James T
Substance P Stimulates Endothelin 1 Secretion via Endothelin-Converting Enzyme 1 and Promotes Melanogenesis in Human Melanocytes  Phil June Park, Tae.
Osteopontin Expression Correlates with Melanoma Invasion
Kavitha Gowrishankar, Stephanie Snoyman, Gulietta M. Pupo, Therese M
Synchronized Targeting of Notch and ERBB Signaling Suppresses Melanoma Tumor Growth through Inhibition of Notch1 and ERBB3  Keman Zhang, Poki Wong, Christine.
Inhibition of PI3K-AKT-mTOR Signaling Sensitizes Melanoma Cells to Cisplatin and Temozolomide  Tobias Sinnberg, Konstantinos Lasithiotakis, Heike Niessner,
SOX4 Promotes Proliferative Signals by Regulating Glycolysis through AKT Activation in Melanoma Cells  Wei Dai, Xinyuan Xu, Shuli Li, Jingjing Ma, Qiong.
Pro-Invasive Activity of the Hippo Pathway Effectors YAP and TAZ in Cutaneous Melanoma  Flore Nallet-Staub, Véronique Marsaud, Ling Li, Cristèle Gilbert,
PDZK1 Upregulation in Estrogen-Related Hyperpigmentation in Melasma
Brian Poligone, Elaine S. Gilmore, Carolina V
Regulation of Fibulin-2 Gene Expression by Integrin α3β1 Contributes to the Invasive Phenotype of Transformed Keratinocytes  Dara S. Missan, Sridar V.
Min Qin, Aslan Pirouz, Myung-Hwa Kim, Stephan R. Krutzik, Hermes J
C/EBPγ Regulates Wound Repair and EGF Receptor Signaling
Plexin B1 Suppresses c-Met in Melanoma: A Role for Plexin B1 as a Tumor-Suppressor Protein through Regulation of c-Met  Laurel Stevens, Lindy McClelland,
Vitiligo-Inducing Phenols Activate the Unfolded Protein Response in Melanocytes Resulting in Upregulation of IL6 and IL8  Siavash Toosi, Seth J. Orlow,
Inhibition of CRM1-Mediated Nucleocytoplasmic Transport: Triggering Human Melanoma Cell Apoptosis by Perturbing Multiple Cellular Pathways  Gaurav Pathria,
Sema4D, the Ligand for Plexin B1, Suppresses c-Met Activation and Migration and Promotes Melanocyte Survival and Growth  Joanne Soong, Yulin Chen, Elina.
Microtubule-Targeted Drugs Inhibit VEGF Receptor-2 Expression by both Transcriptional and Post-Transcriptional Mechanisms  Markus Meissner, Andreas Pinter,
Human Mitochondrial NAD(P)+–Dependent Malic Enzyme Participates in Cutaneous Melanoma Progression and Invasion  Yung-Lung Chang, Hong-Wei Gao, Chien-Ping.
Anne T. Funding, Claus Johansen, Matthias Gaestel, Bo M
Wnt5A Activates the Calpain-Mediated Cleavage of Filamin A
SiRNA Knockdown of Ribonucleotide Reductase Inhibits Melanoma Cell Line Proliferation Alone or Synergistically with Temozolomide  Jonathan E. Zuckerman,
Somatic Mutation of GRIN2A in Malignant Melanoma Results in Loss of Tumor Suppressor Activity via Aberrant NMDAR Complex Formation  Todd D. Prickett,
GSK3β Inhibition Blocks Melanoma Cell/Host Interactions by Downregulating N- Cadherin Expression and Decreasing FAK Phosphorylation  Jobin K. John, Kim.
Min Qin, Aslan Pirouz, Myung-Hwa Kim, Stephan R. Krutzik, Hermes J
Yabin Cheng, Guangdi Chen, Magdalena Martinka, Vincent Ho, Gang Li 
Volume 68, Issue 2, Pages (August 2005)
All-Trans-Retinoic Acid Induces Interleukin-8 via the Nuclear Factor-κB and p38 Mitogen-Activated Protein Kinase Pathways in Normal Human Keratinocytes 
Oncogenic B-RAFV600E Signaling Induces the T-Box3 Transcriptional Repressor to Repress E-Cadherin and Enhance Melanoma Cell Invasion  Suzanah C. Boyd,
Kellie J. White, Vincent J. Maffei, Marvin Newton-West, Robert A
Chi-Hyun Park, Youngji Moon, Chung Min Shin, Jin Ho Chung 
The IL-6 Trans-Signaling-STAT3 Pathway Mediates ECM and Cellular Proliferation in Fibroblasts from Hypertrophic Scar  Sutapa Ray, Xiaoxi Ju, Hong Sun,
Stat3-Targeted Therapies Overcome the Acquired Resistance to Vemurafenib in Melanomas  Fang Liu, Juxiang Cao, Jinxiang Wu, Kayleigh Sullivan, James Shen,
Critical Role of Paxillin in Aging of Human Skin
Insulin-Like Growth Factor-Binding Protein 7 Regulates Keratinocyte Proliferation, Differentiation and Apoptosis  Janna Nousbeck, Ofer Sarig, Nili Avidan,
Green Tea Polyphenol Epigallocatechin-3-Gallate Suppresses Collagen Production and Proliferation in Keloid Fibroblasts via Inhibition of the STAT3-Signaling.
IL-18 Downregulates Collagen Production in Human Dermal Fibroblasts via the ERK Pathway  Hee Jung Kim, Seok Bean Song, Jung Min Choi, Kyung Moon Kim,
Cornulin Is Induced in Psoriasis Lesions and Promotes Keratinocyte Proliferation via Phosphoinositide 3-Kinase/Akt Pathways  Changji Li, Lei Xiao, Jinjing.
Roland Houben, Claudia S. Vetter-Kauczok, Sonja Ortmann, Ulf R
Jasmine George, Minakshi Nihal, Chandra K
Targeted Depletion of Polo-Like Kinase (Plk) 1 Through Lentiviral shRNA or a Small- Molecule Inhibitor Causes Mitotic Catastrophe and Induction of Apoptosis.
MELK Promotes Melanoma Growth by Stimulating the NF-κB Pathway
Keratinocyte G2/M Growth Arrest by 1,25-Dihydroxyvitamin D3 Is Caused by Cdc2 Phosphorylation Through Wee1 and Myt1 Regulation  Xiuju Dai, Kenshi Yamasaki,
Dimethylfumarate Specifically Inhibits the Mitogen and Stress-Activated Kinases 1 and 2 (MSK1/2): Possible Role for its Anti-Psoriatic Effect  Borbala.
Suppression of VEGFR2 Expression in Human Endothelial Cells by Dimethylfumarate Treatment: Evidence for Anti-Angiogenic Action  Markus Meissner, Monika.
John M. Lamar, Vandana Iyer, C. Michael DiPersio 
Deon G. Uffort, Elizabeth A. Grimm, Julie A. Ellerhorst 
Sphingosine 1-Phosphate Restrains Insulin-Mediated Keratinocyte Proliferation via Inhibition of Akt through the S1P2 Receptor Subtype  Melanie Schüppel,
Role and Regulation of STAT3 Phosphorylation at Ser727 in Melanocytes and Melanoma Cells  Masanobu Sakaguchi, Masahiro Oka, Tetsushi Iwasaki, Yasuo Fukami,
Presentation transcript:

The Role of eIF4E in Response and Acquired Resistance to Vemurafenib in Melanoma  Yao Zhan, Michael S. Dahabieh, Arjuna Rajakumar, Monica C. Dobocan, Marie-Noël M’Boutchou, Christophe Goncalves, Shiru L. Lucy, Filippa Pettersson, Ivan Topisirovic, Léon van Kempen, Sonia V. del Rincón, Wilson H. Miller  Journal of Investigative Dermatology  Volume 135, Issue 5, Pages 1368-1376 (May 2015) DOI: 10.1038/jid.2015.11 Copyright © 2015 The Society for Investigative Dermatology, Inc Terms and Conditions

Figure 1 eIF4E knockdown inhibits proliferation in melanoma cell lines that highly express eIF4E, p-AKT, and hyperphosphorylated 4E-BP1. (a) Western blottings displaying eIF4E and 4E-BP1 protein expression in a panel of melanoma cell lines compared with immortalized melanocytes (MelST). Underlined are the cell lines that possess a BRAF V600E mutation. The schematic (right) shows the relationship between the phosphorylation status of 4E-BP and its ability to bind and inhibit eIF4E. (b) Western blotting of p-AKT, AKT, phospho-extracellular signal–regulated kinase (p-ERK), ERK2, and p-4E-BP1 in melanoma cell lines versus immortalized melanocytes (MelST). Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is a loading control in all immunoblots. Four bands can be seen on the p-4E-BP1 blot: the upper band represents the hyperphosphorylated form, whereas the lower band represents the hypophosphorylated form. The lowest band identified with an asterisk shows the phosphorylation of 4E-BP2. Correlation between the expression of p-AKT and p-4E-BP1 in the melanoma cell lines is shown (right). Pearson’s correlation statistics have been used. (c) Cell proliferation assay plot following 4-day eIF4E siRNA treatment, assessed by sulforhodamine B (SRB) staining. Error bars are defined as mean±SD, n=3. (d) Correlation between the levels of proliferation inhibition upon eIF4E siRNA silencing versus p-4E-BP1 expression. Pearson’s correlation statistics have been used. Note: p-4E-BP1 and p-AKT protein levels were normalized to corresponding GAPDH levels. Journal of Investigative Dermatology 2015 135, 1368-1376DOI: (10.1038/jid.2015.11) Copyright © 2015 The Society for Investigative Dermatology, Inc Terms and Conditions

Figure 2 Vemurafenib drastically inhibits the phosphorylation of the eIF4E inhibitory protein 4E-BP1 in BRAFV600E melanomas. (a) Western blotting of eIF4E, p-4E-BP1, 4E-BP1, and p-ERK and ERK2 in A375, SkMel28, and A375M upon 2.5μM vemurafenib treatment at the times indicated. A concentration of 1μM PP242 was used as a positive control to promote 4E-BP1 hypophosphorylation. (b) Western blotting of p-S6, S6, p-P70S6K, P70S6K, and p-AKT and AKT in A375, SkMel28, and A375M upon 2.5μM vemurafenib treatment at the times indicated. (c) Cap-binding assay in A375, SkMel28, and A375M upon 2.5μM vemurafenib treatment. (d) Polysome profile (% sucrose gradient vs. 254nm rRNA absorbance) in A375 cells in the absence and presence of 2.5μM vemurafenib for 24hours (left panel). Western blotting analysis of cyclin D3, vascular endothelial growth factor (VEGF), c-Myc, and Bcl-2 following treatment with 2.5μM vemurafenib for 24hours (right panel). (e) Quantitative real-time reverse-transcriptase–PCR (qRT–PCR) was used to determine the distribution of cyclin D3 and β-actin mRNAs in polysome fractions (heavy vs. light polysomes) isolated from A375 cells treated with DMSO or 2.5μM vemurafenib for 24hours. Journal of Investigative Dermatology 2015 135, 1368-1376DOI: (10.1038/jid.2015.11) Copyright © 2015 The Society for Investigative Dermatology, Inc Terms and Conditions

Figure 3 eIF4E has a key role in vemurafenib resistance in A375 BRAFV600E human melanoma cell lines. (a) Cell proliferation assessment by sulforhodamine B (SRB) staining 4 days after si4E transfection with either vehicle (DMSO) or 2.5μM vemurafenib cotreatment in A375, AR1WO (wash-off), AR2WO, AR1, and AR2. Note: AR1 and AR2 are continuously maintained in 2.5μM vemurafenib. Error bars are defined as mean±SD, n=3. Statistical significance was determined by the Student’s t-test. (b) Cap-binding assay in parental A375 and resistant lines AR1 and AR2 (maintained in 2.5μM vemurafenib), and corresponding vemurafenib WO lines AR1WO and AR2WO. PP242 is a positive control, for 4E-BP1 hypophosphorylation. (c) Western blotting of cyclin D3, vascular endothelial growth factor (VEGF), and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) in A375 parental, resistant lines AR1 and AR2, and corresponding WO lines, AR1WO and AR2WO, respectively. *P<0.05. Journal of Investigative Dermatology 2015 135, 1368-1376DOI: (10.1038/jid.2015.11) Copyright © 2015 The Society for Investigative Dermatology, Inc Terms and Conditions

Figure 4 Stable knockdown of 4E-BP1/2 contributes to the development of vemurafenib resistance in A375 cells. (a) Western blotting of c-Myc, 4E-BP1, 4E-BP2, and glyceraldehyde-3-phosphate dehydrogenase (GAPDH; loading control) in A375 shCTL and shBP1/2 cell lines. (b) Cell proliferation assay after 4 days with or without 0.5μM vemurafenib treatment in A375 shCTL and shBP1/2 stable cell lines. Error bars are defined as mean±SD, n=3. Statistical significance was determined by one-way analysis of variance followed by the Newman–Keuls post-hoc test using Prism version 3.0 (GraphPad Software, San Diego, CA). (c) Clonogenic assay after 14 days with or without 0.5μM vemurafenib treatment in A375 shCTL and shBP1/2 stable cell lines. The number of colonies was counted manually and graphed. Error bars are defined as mean±SD, n=3. Statistical significance was determined by one-way analysis of variance followed by the Newman–Keuls post-hoc test using Prism version 3.0 (GraphPad Software). *P<0.05, **P<0.01. Representative pictures are shown on the right. NS, not significant. Journal of Investigative Dermatology 2015 135, 1368-1376DOI: (10.1038/jid.2015.11) Copyright © 2015 The Society for Investigative Dermatology, Inc Terms and Conditions