Volume 13, Issue 9, Pages (December 2015)

Slides:



Advertisements
Similar presentations
A Novel Cinnamide YLT26 Induces Breast Cancer Cells Apoptosis via ROS-Mitochondrial Apoptotic Pathway in Vitro and Inhibits.
Advertisements

Volume 114, Issue 5, Pages (May 1998)
Cantharidin Inhibits the Growth of Triple-Negative Breast Cancer Cells by Suppressing Autophagy and Inducing Apoptosis in Vitro and in.
Volume 21, Issue 12, Pages (December 2017)
S Braun, N Gaza, R Werdehausen, H Hermanns, I Bauer, M. E. Durieux, M
Volume 117, Issue 6, Pages (June 2004)
Volume 129, Issue 1, Pages (July 2005)
Droxinostat, a Histone Deacetylase Inhibitor, Induces Apoptosis in Hepatocellular Carcinoma Cell Lines via Activation of the Mitochondrial Pathway and.
Volume 18, Issue 5, Pages (May 2010)
RAF Inhibition Overcomes Resistance to TRAIL-Induced Apoptosis in Melanoma Cells  Anja Berger, Sandra-Annika Quast, Michael Plötz, Nicholas-Frederik Kuhn,
How cells die: Apoptosis pathways
Volume 9, Issue 2, Pages (August 2017)
The Zinc Ionophore PCI-5002 Radiosensitizes Non-small Cell Lung Cancer Cells by Enhancing Autophagic Cell Death  Kwang Woon Kim, PhD, Christina K. Speirs,
Marissa V. Powers, Paul A. Clarke, Paul Workman  Cancer Cell 
H.T. Lee, M. Kim, M. Jan, R.B. Penn, C.W. Emala  Kidney International 
Volume 10, Issue 7, Pages (July 2003)
Volume 96, Issue 10, Pages (May 2009)
Enhancement of depsipeptide-mediated apoptosis of lung or esophageal cancer cells by flavopiridol: Activation of the mitochondria-dependent death-signaling.
PKC-θ is a negative regulator of TRAIL-induced and FADD-mediated apoptotic spectrin aggregation DOI: /FHC.a Spectrin phosphorylation upon.
Antisense Oligonucleotides Targeting Y-Box Binding Protein-1 Inhibit Tumor Angiogenesis by Downregulating Bcl-xL-VEGFR2/-Tie Axes  Kiyoko Setoguchi, Lin.
Volume 19, Issue 8, Pages (August 2017)
Combining a BCL2 Inhibitor with the Retinoid Derivative Fenretinide Targets Melanoma Cells Including Melanoma Initiating Cells  Nabanita Mukherjee, Steven.
Volume 6, Issue 1, Pages (January 2014)
Marissa V. Powers, Paul A. Clarke, Paul Workman  Cancer Cell 
A Stapled p53 Helix Overcomes HDMX-Mediated Suppression of p53
Volume 26, Issue 2, Pages (August 2014)
Volume 18, Issue 11, Pages (March 2017)
Protodynamic Intracellular Acidification by cis-Urocanic Acid Promotes Apoptosis of Melanoma Cells In Vitro and In Vivo  Jarmo K. Laihia, Janne P. Kallio,
Volume 18, Issue 23, Pages (December 2008)
Volume 41, Issue 2, Pages (January 2011)
Volume 7, Issue 1, Pages (April 2014)
Volume 91, Issue 5, Pages (November 1997)
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.
Volume 26, Issue 1, Pages (July 2013)
Bax Activation and Induction of Apoptosis in Human Keratinocytes by the Protein Kinase C δ Catalytic Domain  Leonid A. Sitailo, Shalini S. Tibudan, Mitchell.
Volume 12, Issue 1, Pages (July 2015)
A JNK-Dependent Pathway Is Required for TNFα-Induced Apoptosis
Volume 4, Issue 3, Pages (March 2015)
Volume 23, Issue 3, Pages (March 2013)
Molecular Therapy - Nucleic Acids
Volume 6, Issue 1, Pages (January 2014)
PARP Determines the Mode of Cell Death in Skin Fibroblasts, but not Keratinocytes, Exposed to Sulfur Mustard  Dana Anderson, Betty Benton, Zhao-Qi Wang,
Volume 47, Issue 4, Pages e9 (November 2018)
Volume 14, Issue 5, Pages (February 2016)
Volume 19, Issue 1, Pages (April 2017)
Volume 10, Issue 2, Pages (January 2015)
Volume 12, Issue 1, Pages (July 2007)
Volume 19, Issue 7, Pages (May 2017)
Volume 8, Issue 4, Pages (October 2005)
Volume 17, Issue 12, Pages (December 2016)
Volume 10, Issue 7, Pages (July 2003)
MELK Promotes Melanoma Growth by Stimulating the NF-κB Pathway
Targeted Cleavage of Signaling Proteins by Caspase 3 Inhibits T Cell Receptor Signaling in Anergic T Cells  Irene Puga, Anjana Rao, Fernando Macian  Immunity 
Volume 18, Issue 3, Pages (March 2010)
Volume 19, Issue 1, Pages (April 2017)
Gold Nanoparticles for BCR-ABL1 Gene Silencing: Improving Tyrosine Kinase Inhibitor Efficacy in Chronic Myeloid Leukemia  Raquel Vinhas, Alexandra R.
Volume 114, Issue 5, Pages (May 1998)
The Tumor Suppressor p53 Limits Ferroptosis by Blocking DPP4 Activity
Volume 25, Issue 9, Pages e5 (November 2018)
Jerry E. Chipuk, Ulrich Maurer, Douglas R. Green, Martin Schuler 
Marijn T.M. van Jaarsveld, Difan Deng, Erik A.C. Wiemer, Zhike Zi 
Volume 18, Issue 3, Pages (March 2003)
Volume 26, Issue 12, Pages e4 (March 2019)
DHA induces apoptosis independently from caspase-8 and FADD in Jurkat T-lymphoma cells. DHA induces apoptosis independently from caspase-8 and FADD in.
Volume 26, Issue 10, Pages (October 2018)
Volume 7, Issue 2, Pages (August 2016)
Volume 11, Issue 2, Pages (August 2018)
Volume 14, Issue 4, Pages (May 2004)
Differential Induction of Immunogenic Cell Death and Interferon Expression in Cancer Cells by Structured ssRNAs  Jaewoo Lee, Youngju Lee, Li Xu, Rebekah.
Presentation transcript:

Volume 13, Issue 9, Pages 2027-2036 (December 2015) A Small Molecule that Induces Intrinsic Pathway Apoptosis with Unparalleled Speed  Rahul Palchaudhuri, Michael J. Lambrecht, Rachel C. Botham, Kathryn C. Partlow, Tjakko J. van Ham, Karson S. Putt, Laurie T. Nguyen, Seok-Ho Kim, Randall T. Peterson, Timothy M. Fan, Paul J. Hergenrother  Cell Reports  Volume 13, Issue 9, Pages 2027-2036 (December 2015) DOI: 10.1016/j.celrep.2015.10.042 Copyright © 2015 The Authors Terms and Conditions

Cell Reports 2015 13, 2027-2036DOI: (10.1016/j.celrep.2015.10.042) Copyright © 2015 The Authors Terms and Conditions

Figure 1 Raptinal Rapidly Induces Apoptosis (A) Structure of Raptinal. (B) Scanning electron micrographs of U-937 cells show pronounced apoptotic blebbing after 1 hr of treatment with 10 μM Raptinal (right) versus vehicle control-treated cells (left). (C) AV/PI graphs of U-937 cells treated with 10 μM Raptinal for 2 hr show transition of cells through the early apoptotic AV+/PI– quadrant that is prevented by the pan-caspase inhibitor Q-VD-OPh. (D) Immunoblots of U-937 cells treated with various toxins for 1 hr show more prominent activation of procaspase-3 (PC-3) to caspase-3 (C-3) and cleavage of PARP-1 (to cPARP-1) by Raptinal (10 μM) versus 25 other toxins (all tested at 10 μM). (E) Cell viability of U-937 cells assessed by AV/PI after 2 hr treatment with 10 μM Raptinal and 25 other small molecules (all tested at 10 μM). Data represent the mean ± SD from three independent experiments. (F) Time-course analysis of U-937 cell viability upon treatment with 10 μM of various anticancer agents and biological tool molecules. Cell viability was assessed by AV/PI analysis. (G) Time-course analysis of adherent cell viability upon treatment with Raptinal, 1541B, and staurosporine (all tested at 10 μM). Cell viability was assessed by AV/PI analysis. See also Figure S1. Cell Reports 2015 13, 2027-2036DOI: (10.1016/j.celrep.2015.10.042) Copyright © 2015 The Authors Terms and Conditions

Figure 2 Raptinal Activates the Intrinsic Pathway and Requires Functional Mitochondria for Apoptosis Induction (A) Time-course immunoblots of mitochondrial and cytosolic fractions of U-937 cells treated with 10 μM Raptinal show cytochrome c release and subsequent caspase-9 activation occur after 20 min of treatment. (B) Time-course immunoblot analysis of caspase-9, -3, and -8 activation in U-937 cells treated with 10 μM Raptinal. (C) Relative percent caspase-3/-7 activity of MIA PaCa-2 cells treated with 10 μM Raptinal for 1 hr upon siRNA knockdown of apoptosis genes. (D) Jurkat C8−/− are equally susceptible and Jurkat FADD−/− are more susceptible, while Jurkat Bcl-2 overexpressing cells are less susceptible than wild-type cells to 10 μM Raptinal as assessed by AV/PI assay after 2 hr. Data represent the mean ± SD from three independent experiments. ∗∗p values <0.02. (E) Transmission electron micrographs of U-937 cells treated with vehicle or 10 μM Raptinal for 5, 30, and 60 min. The images show rapid changes in mitochondrial morphology (arrows) after 5 min of treatment with Raptinal. At 30 min, mitochondria are largely devoid of cristae, and at 60 min peripheral nuclear condensation is apparent. Scale bars, 1 μm. (F) Raptinal does not induce cytochrome c release from the mitochondrial pellet into the supernatant of isolated mitochondria treated with Raptinal in vitro under non-respiring conditions. The positive control, pro-apoptotic Bid protein is able to induce cytochrome c release. (G) U-937 cells pre-treated with various potential cytoprotective agents and inhibitors of various cellular pathways were treated with 10 μM Raptinal for 2 hr, and protection from the effects of Raptinal was assessed by AV/PI. Data represent the mean % protection ± SD from three independent experiments. ∗p values <0.05. (H) Protective mitochondrial agents retard cytochrome c release and caspase-9 activation in U-937 cells. See also Figure S2. Cell Reports 2015 13, 2027-2036DOI: (10.1016/j.celrep.2015.10.042) Copyright © 2015 The Authors Terms and Conditions

Figure 3 Raptinal Exhibits Activity In Vivo (A) Zebrafish embryos expressing secreted annexin V-YFP exhibit pronounced punctate YFP signal indicating phosphatidylserine externalization following 1.5 hr of treatment with 10 μM Raptinal. (B) Quantification of apoptotic cells in Raptinal- versus DMSO-treated zebrafish under the conditions in (A). Data represent the mean ± SD (n = 5 and n = 7 embryos for Raptinal and DMSO, respectively; ∗∗∗p value <0.001). (C and D) Raptinal inhibits subcutaneous B16-F10 melanoma tumor growth in vivo as measured by tumor volume (∗∗p value < 0.005) in (C) and tumor mass after tumor excision in (D) (∗p value <0.05). (E and F) Raptinal inhibits subcutaneous 4T1 breast cancer tumor growth in vivo as measured by tumor volume (∗p value <0.05) in (E) and tumor mass after tumor excision (∗p value <0.05) in (F). Arrows in (C) and (E) indicate intraperitoneal Raptinal administration at 20 mg/kg once a day. Tumor images in (D) and (F) are representative of tumor size at the conclusion of the studies. Data in (C)–(F) represent the mean ± SEM (n = 7 mice/group). See also Figure S3. Cell Reports 2015 13, 2027-2036DOI: (10.1016/j.celrep.2015.10.042) Copyright © 2015 The Authors Terms and Conditions