Jane L. Armstrong, David S. Hill, Christopher S

Slides:



Advertisements
Similar presentations
Inhibition of autophagy augments 5-fluorouracil chemotherapy in human colon cancer in vitro and in vivo model  Jie Li, Ni Hou, Ahmad Faried, Soichi Tsutsumi,
Advertisements

Nan-Hyung Kim, Ai-Young Lee  Journal of Investigative Dermatology 
Δ-Tocotrienol treatment is more effective against hypoxic tumor cells than normoxic cells: potential implications for cancer therapy  Akira Shibata, Kiyotaka.
The role of galectin-1 in in vitro and in vivo photodynamic therapy with a galactodendritic porphyrin  Patrícia M.R. Pereira, Sandrina Silva, José S.
The Autophagy Inhibitor Chloroquine Overcomes the Innate Resistance of Wild-Type EGFR Non-Small-Cell Lung Cancer Cells to Erlotinib  Yiyu Zou, PhD, Yi-He.
Bovine lactoferricin B induces apoptosis of human gastric cancer cell line AGS by inhibition of autophagy at a late stage  W.-R. Pan, P.-W. Chen, Y.-L.
Amanda M. Nelson, Kathryn L. Gilliland, Zhaoyuan Cong, Diane M
Cell Surface CD74–MIF Interactions Drive Melanoma Survival in Response to Interferon-γ  Keiji Tanese, Yuuri Hashimoto, Zuzana Berkova, Yuling Wang, Felipe.
Yunguang Sun, PhD, Luigi Moretti, MD, Nicholas J
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,
Vemurafenib Induces Senescence Features in Melanoma Cells
Inhibition of the Prohormone Convertase Subtilisin-Kexin Isoenzyme-1 Induces Apoptosis in Human Melanoma Cells  Nina Weiß, Agatha Stegemann, Marwa A.T.
Yongping Shao, Kaitlyn Le, Hanyin Cheng, Andrew E. Aplin 
MAGE-C2 Promotes Growth and Tumorigenicity of Melanoma Cells, Phosphorylation of KAP1, and DNA Damage Repair  Neehar Bhatia, Tony Z. Xiao, Kimberly A.
Xeroderma Pigmentosum Group A Promotes Autophagy to Facilitate Cisplatin Resistance in Melanoma Cells through the Activation of PARP1  Rui Ge, Lin Liu,
Christian Posch, Brian D
Dual Role of Apoptosis-Associated Speck-Like Protein Containing a CARD (ASC) in Tumorigenesis of Human Melanoma  Weimin Liu, Yuchun Luo, Jeffrey H. Dunn,
Volume 191, Issue 1, Pages (January 2014)
Anne L. Donato, Qian Huang, Xinjian Liu, Fang Li, Mary A
RAF Inhibition Overcomes Resistance to TRAIL-Induced Apoptosis in Melanoma Cells  Anja Berger, Sandra-Annika Quast, Michael Plötz, Nicholas-Frederik Kuhn,
Targeted Nanoparticles Deliver siRNA to Melanoma
Endocannabinoids Modulate Human Epidermal Keratinocyte Proliferation and Survival via the Sequential Engagement of Cannabinoid Receptor-1 and Transient.
Efficient TRAIL-R1/DR4-Mediated Apoptosis in Melanoma Cells by Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL)  Bahtier M. Kurbanov, Christoph.
Volume 65, Issue 2, Pages (February 2004)
The Epigenetic Regulator I-BET151 Induces BIM-Dependent Apoptosis and Cell Cycle Arrest of Human Melanoma Cells  Stuart J. Gallagher, Branka Mijatov,
RNA-Seq and ChIP-Seq Reveal SQSTM1/p62 as a Key Mediator of JunB Suppression of NF-κB-Dependent Inflammation  Xiaoling Zhang, Jane Y. Jin, Joseph Wu,
Preclinical Studies of a Specific PPARγ Modulator in the Control of Skin Inflammation  Arianna Mastrofrancesco, Daniela Kovacs, Massimiliano Sarra, Emanuela.
Combining a BCL2 Inhibitor with the Retinoid Derivative Fenretinide Targets Melanoma Cells Including Melanoma Initiating Cells  Nabanita Mukherjee, Steven.
Indomethacin Sensitizes TRAIL-Resistant Melanoma Cells to TRAIL-Induced Apoptosis through ROS-Mediated Upregulation of Death Receptor 5 and Downregulation.
The Autophagy Inhibitor Chloroquine Overcomes the Innate Resistance of Wild-Type EGFR Non-Small-Cell Lung Cancer Cells to Erlotinib  Yiyu Zou, PhD, Yi-He.
P21-Activated Kinase 4 Critically Regulates Melanogenesis via Activation of the CREB/MITF and β-Catenin/MITF Pathways  Cheong-Yong Yun, Soon-Tae You,
Protodynamic Intracellular Acidification by cis-Urocanic Acid Promotes Apoptosis of Melanoma Cells In Vitro and In Vivo  Jarmo K. Laihia, Janne P. Kallio,
Clusterin Regulates Drug-Resistance in Melanoma Cells
MiTF Regulates Cellular Response to Reactive Oxygen Species through Transcriptional Regulation of APE-1/Ref-1  Feng Liu, Yan Fu, Frank L. Meyskens  Journal.
Alexander J. Lakhter, Ravi P. Sahu, Yang Sun, William K
NF-κB and STAT3 Inhibition as a Therapeutic Strategy in Psoriasis: In Vitro and In Vivo Effects of BTH  Rosa M. Andrés, M. Carmen Montesinos, Pedro Navalón,
Inhibition of Human Melanoma Cell Growth by the Dietary Flavonoid Fisetin Is Associated with Disruption of Wnt/β-Catenin Signaling and Decreased Mitf.
Brian Poligone, Elaine S. Gilmore, Carolina V
Potent Dual Inhibitors of TORC1 and TORC2 Complexes (KU and KU ) Demonstrate In Vitro and Ex Vivo Anti-Keloid Scar Activity  Farhatullah.
Targeting Tumor-Associated Macrophages and Inhibition of MCP-1 Reduce Angiogenesis and Tumor Growth in a Human Melanoma Xenograft  Silvina Gazzaniga,
An Actin-Binding Protein Espin Is a Growth Regulator for Melanoma
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,
Inhibition of CRM1-Mediated Nucleocytoplasmic Transport: Triggering Human Melanoma Cell Apoptosis by Perturbing Multiple Cellular Pathways  Gaurav Pathria,
Combining a BCL2 Inhibitor with the Retinoid Derivative Fenretinide Targets Melanoma Cells Including Melanoma Initiating Cells  Nabanita Mukherjee, Steven.
Sema4D, the Ligand for Plexin B1, Suppresses c-Met Activation and Migration and Promotes Melanocyte Survival and Growth  Joanne Soong, Yulin Chen, Elina.
NFATc2 Is a Potential Therapeutic Target in Human Melanoma
Christiane Thallinger, Markus F
Yabin Cheng, Guangdi Chen, Magdalena Martinka, Vincent Ho, Gang Li 
Axl Promotes Cutaneous Squamous Cell Carcinoma Survival through Negative Regulation of Pro-Apoptotic Bcl-2 Family Members  Emmanouil S. Papadakis, Monika.
Select Cancer Testes Antigens of the MAGE-A, -B, and -C Families Are Expressed in Mast Cell Lines and Promote Cell Viability In Vitro and In Vivo  Bing.
Overexpression of the Transcription Factor Yin-Yang-1 Suppresses Differentiation of HaCaT Cells in Three-Dimensional Cell Culture  Shijima Taguchi, Yasuhiro.
Stat3-Targeted Therapies Overcome the Acquired Resistance to Vemurafenib in Melanomas  Fang Liu, Juxiang Cao, Jinxiang Wu, Kayleigh Sullivan, James Shen,
Resistance of Human Melanoma Cells Against the Death Ligand TRAIL Is Reversed by Ultraviolet-B Radiation via Downregulation of FLIP  Elke Zeise, Michael.
Insulin-Like Growth Factor-Binding Protein 7 Regulates Keratinocyte Proliferation, Differentiation and Apoptosis  Janna Nousbeck, Ofer Sarig, Nili Avidan,
Mark A. Rovedo, Nancy L. Krett, Steven T. Rosen 
In Vitro and In Vivo Anti-Melanoma Effects of Ciglitazone
Autophagy Has a Significant Role in Determining Skin Color by Regulating Melanosome Degradation in Keratinocytes  Daiki Murase, Akira Hachiya, Kei Takano,
Jasmine George, Minakshi Nihal, Chandra K
Collagen Synthesis Is Suppressed in Dermal Fibroblasts by the Human Antimicrobial Peptide LL-37  Hyun Jeong Park, Dae Ho Cho, Hee Jung Kim, Jun Young.
Syed M. Meeran, Thejass Punathil, Santosh K. Katiyar 
Volume 15, Issue 7, Pages (July 2007)
Macrophage Inhibitory Cytokine-1 Is Overexpressed in Malignant Melanoma and Is Associated with Tumorigenicity  Glen M. Boyle, Julie Pedley, Adam C. Martyn,
Anna Flammiger, Robert Besch, Anthony L. Cook, Tanja Maier, Richard A
Loss of Class III β-Tubulin Induced by Histone Deacetylation Is Associated with Chemosensitivity to Paclitaxel in Malignant Melanoma Cells  Kiyomi Akasaka,
Functional Modulation of IGF-Binding Protein-3 Expression in Melanoma
Volume 18, Issue 3, Pages (March 2010)
Jane L. Armstrong, David S. Hill, Christopher S
P21-Activated Kinase 4 Critically Regulates Melanogenesis via Activation of the CREB/MITF and β-Catenin/MITF Pathways  Cheong-Yong Yun, Soon-Tae You,
Dihydrotestosterone-Inducible Dickkopf 1 from Balding Dermal Papilla Cells Causes Apoptosis in Follicular Keratinocytes  Mi Hee Kwack, Young Kwan Sung,
Role and Regulation of STAT3 Phosphorylation at Ser727 in Melanocytes and Melanoma Cells  Masanobu Sakaguchi, Masahiro Oka, Tetsushi Iwasaki, Yasuo Fukami,
Presentation transcript:

Exploiting Cannabinoid-Induced Cytotoxic Autophagy to Drive Melanoma Cell Death  Jane L. Armstrong, David S. Hill, Christopher S. McKee, Sonia Hernandez-Tiedra, Mar Lorente, Israel Lopez-Valero, Maria Eleni Anagnostou, Fiyinfoluwa Babatunde, Marco Corazzari, Christopher P.F. Redfern, Guillermo Velasco, Penny E. Lovat  Journal of Investigative Dermatology  Volume 135, Issue 6, Pages 1629-1637 (June 2015) DOI: 10.1038/jid.2015.45 Copyright © 2015 The Society for Investigative Dermatology, Inc Terms and Conditions

Figure 1 The Δ9-Tetrahydrocannabinol (THC) induces autophagy in melanoma cells. (a–c) A375, SK-MEL-28, and CHL-1 cells were treated with vehicle or THC (4.5 or 5 μm) for 24 hours in the presence or absence of chloroquine (CQ; 10 μm) for the final 2 hours, and LC3 and β-actin expression was determined by western blotting. LC3-II expression was quantified and band intensity normalized to β-actin. Data are expressed as fold change relative to the mean LC3-II/β-actin value for a representative experiment and are shown above the western blot (representative data from n=3 independent experiments). (d, e) mRFP–GFP–LC3 expressing (d) A375 or (e) CHL-1 cells were treated with vehicle or THC (4.5 μm) for 18 hours. Data are representative fluorescent micrographs (bar=20 μm) of three independent experiments. (f) Total red fluorescence values were generated from ≥20 cells per treatment condition, from two independent experiments. Pixel intensities were divided by a factor of 106, and data are shown as mean±SD (**P<0.01 and ***P<0.001 vs. control for each cell line). mRFP–GFP, monomeric red fluorescent protein–green fluorescent protein. Journal of Investigative Dermatology 2015 135, 1629-1637DOI: (10.1038/jid.2015.45) Copyright © 2015 The Society for Investigative Dermatology, Inc Terms and Conditions

Figure 2 The Δ9-Tetrahydrocannabinol (THC) induces apoptosis of melanoma cells. (a) Primary melanocytes, A375, and CHL-1 cells were treated with vehicle or THC (3–10 μM) for 24 hours. (b) A375 (i) or CHL-1 (ii) cells were treated with vehicle or THC (5 μM) in the presence or absence of ZVAD (20 μM) for 24 hours. Cell viability was determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Data generated in triplicate were expressed relative to the mean of vehicle-treated cells in each experiment, for three independent experiments, and shown as mean±SD (t-test; **P<0.01 and ***P<0.001 vs. THC+ZVAD). (c) A375 cells were treated with THC (5 μM) for 24 hours. Data are representative fluorescent micrographs of cytochrome c immunostaining (bar=20 μm) of three independent experiments. Journal of Investigative Dermatology 2015 135, 1629-1637DOI: (10.1038/jid.2015.45) Copyright © 2015 The Society for Investigative Dermatology, Inc Terms and Conditions

Figure 3 The Δ9-Tetrahydrocannabinol (THC)-induced apoptosis requires autophagy. (a–c) A375 cells were transfected with small interfering RNAs (siRNAs) for Atg7 (siAtg7) or with a nonsilencing control siRNA (siCtrl) before treatment with vehicle or THC (4.5, 5 μM) for 24 hours in the presence or absence of chloroquine (CQ; 10 μM) for the (a, b) final 2 hours or for (c) 24 hours. (a, b) Atg7, LC3, cleaved caspase 3, and β-actin expression were determined by western blotting. LC3-II expression was quantified and band intensity normalized to β-actin. Data are expressed as fold change relative to the mean LC3-II/β-actin value for each experiment, for three separate experiments (mean±SD, n=3). (c) Cell viability was determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Data generated in triplicate were expressed relative to the mean of vehicle-treated siCtrl cells in each experiment, for three independent experiments, and shown as mean±SD (*P<0.05, **P<0.01, and ***P<0.001 vs. vehicle-treated siCtrl cells). Journal of Investigative Dermatology 2015 135, 1629-1637DOI: (10.1038/jid.2015.45) Copyright © 2015 The Society for Investigative Dermatology, Inc Terms and Conditions

Figure 4 The Δ9-Tetrahydrocannabinol (THC)-induced autophagy and cell death is not dependent on Beclin-1 or Ambra1. (a) A375 cells stably expressing short hairpin RNA (shRNA) for Beclin-1 (shBeclin1) or a nonsilencing control shRNA (shCtrl) were treated with vehicle or THC (4.5 and 5 μM) for 48 hours in the presence or absence of chloroquine (CQ; 10 μM) for the final 2 hours (i). Beclin-1, LC3, cleaved caspase 3, and β-actin expression were determined by western blotting (i). Cell viability (ii) was determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Data generated in triplicate were expressed relative to the mean of vehicle-treated shCtrl cells in each experiment, for three independent experiments, and shown as mean±SD (***P<0.001 vs. vehicle-treated shCtrl cells). (b) A375 cells stably expressing shRNA for Ambra1 (shAmbra1) or a nonsilencing control shRNA (shCtrl) were treated with vehicle or THC (5 μM) for 24 hours in the presence or absence of CQ (10 μM) for the final 2 hours (i). Ambra1, LC3, cleaved caspase 3, and β-actin expression were determined by western blotting (i). Cell viability (ii) was determined by the MTT assay. Data generated in triplicate were normalized to the mean of vehicle-treated cells for each shRNA in each experiment, for three independent experiments, and shown as mean±SD (***P<0.001 vs. vehicle-treated cells for each shRNA). Journal of Investigative Dermatology 2015 135, 1629-1637DOI: (10.1038/jid.2015.45) Copyright © 2015 The Society for Investigative Dermatology, Inc Terms and Conditions

Figure 5 Cannabinoids inhibit melanoma cell viability in vitro. (a–c) CHL-1 (a), A375 (b), or SK-MEL-28 (c) cells were treated with temozolomide (Temo; 10–50 μM), Δ9-Tetrahydrocannabinol (THC; 1–5 μM), or THC+CBD (0.5 μM THC+0.5 μM CBD to 2.5 μM THC+2.5 μM CBD) for 48 hours. Cell viability was determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Data generated in triplicate were expressed relative to the mean of vehicle-treated cells for each drug treatment in each experiment, for three independent experiments, and shown as mean±SD for a representative experiment. CBD, cannabidiol. Journal of Investigative Dermatology 2015 135, 1629-1637DOI: (10.1038/jid.2015.45) Copyright © 2015 The Society for Investigative Dermatology, Inc Terms and Conditions

Figure 6 The Δ9-Tetrahydrocannabinol (THC) and Sativex-like cannabinoids promote autophagy and antitumor responses in melanoma xenografts. Athymic nude mice were injected subcutaneously in the right flank with CHL-1 melanoma cells. When tumors reached a 250 mm3 size, mice were treated daily for 20 days with vehicle, temozolomide (TMZ; 5 mg kg−1; local administration), THC (15 mg kg−1; oral administration), or Sativex-like (Sat-L; 7.5 mg kg−1 THC–BDS+7.5 mg kg−1 CBD–BDS; oral administration). (a) Tumor volumes were measured daily. Each point is the mean from ≥5 tumors±SD and is expressed relative to the tumor volume on day 1 of treatment. (b–d) Immunohistochemical analysis of CHL-1 xenograft tumors treated with temozolomide, THC, or Sat-L. (b–d) Micrographs of tumor sections stained for (b) Ki67, (c) TUNEL, or (d) LC3. In each, green is staining for Ki67, TUNEL, or LC3, and red is the TO-PRO-3 counterstain. The bar graphs below each set of micrographs summarize the data analysis from tumor sections (bar=20 μm). Total fluorescence values (total fluorescence (LC3/Ki67/TUNEL)/total nuclear fluorescence) were generated in triplicate for 3 tumors in each treatment group. Data are expressed as fold change relative to the mean value obtained from control animals, from two independent staining analyses, and shown as mean±SD (**P<0.01 and ***P<0.001 vs. control animals). Journal of Investigative Dermatology 2015 135, 1629-1637DOI: (10.1038/jid.2015.45) Copyright © 2015 The Society for Investigative Dermatology, Inc Terms and Conditions