Volume 10, Issue 4, Pages (October 2006)

Slides:



Advertisements
Similar presentations
MiR-128b is a potent glucocorticoid sensitizer in MLL-AF4 acute lymphocytic leukemia cells and exerts cooperative effects with miR-221 by Ai Kotani, Daon.
Advertisements

Volume 15, Issue 6, Pages (June 2009)
Volume 15, Issue 6, Pages (June 2012)
Volume 9, Issue 5, Pages (May 2006)
Synergistic action of dual IGF1/R and MEK inhibition sensitizes childhood acute lymphoblastic leukemia (ALL) cells to cytotoxic agents and involves downregulation.
Merkel Cell Carcinoma Dependence on Bcl-2 Family Members for Survival
Modulation of K-Ras-Dependent Lung Tumorigenesis by MicroRNA-21
Volume 19, Issue 2, Pages (February 2017)
Marissa V. Powers, Paul A. Clarke, Paul Workman  Cancer Cell 
Short Telomeres Limit Tumor Progression In Vivo by Inducing Senescence
Volume 28, Issue 4, Pages (October 2015)
Marissa V. Powers, Paul A. Clarke, Paul Workman  Cancer Cell 
Volume 12, Issue 2, Pages (August 2007)
Volume 120, Issue 6, Pages (March 2005)
Correlating Notch Signaling with Thymocyte Maturation
Volume 21, Issue 4, Pages (April 2012)
Volume 23, Issue 6, Pages (June 2013)
Volume 136, Issue 5, Pages (March 2009)
Kavitha Gowrishankar, Stephanie Snoyman, Gulietta M. Pupo, Therese M
Volume 23, Issue 1, Pages (January 2013)
Oliver I. Fregoso, Shipra Das, Martin Akerman, Adrian R. Krainer 
AT-101, a Pan-Bcl-2 Inhibitor, Leads to Radiosensitization of Non-small Cell Lung Cancer  Luigi Moretti, MD, Bo Li, MD, Kwang Woon Kim, PhD, Heidi Chen,
Volume 11, Issue 2, Pages (February 2007)
Volume 24, Issue 6, Pages (June 2006)
Volume 130, Issue 4, Pages (August 2007)
Volume 137, Issue 5, Pages (May 2009)
Volume 24, Issue 3, Pages (March 2006)
Volume 14, Issue 6, Pages (December 2008)
mTOR Regulates Cellular Iron Homeostasis through Tristetraprolin
Ryang Hwa Lee, Nara Yoon, John C. Reneau, Darwin J. Prockop 
Volume 23, Issue 10, Pages (October 2016)
Volume 25, Issue 3, Pages (March 2014)
Volume 20, Issue 5, Pages (November 2016)
Volume 14, Issue 8, Pages (March 2016)
Jungmook Lyu, Vicky Yamamoto, Wange Lu  Developmental Cell 
14-3-3σ Regulates Keratinocyte Proliferation and Differentiation by Modulating Yap1 Cellular Localization  Sumitha A.T. Sambandam, Ramesh B. Kasetti,
Volume 9, Issue 5, Pages (May 2009)
HDAC5, a Key Component in Temporal Regulation of p53-Mediated Transactivation in Response to Genotoxic Stress  Nirmalya Sen, Rajni Kumari, Manika Indrajit.
Volume 9, Issue 1, Pages (January 2006)
Volume 38, Issue 1, Pages (April 2010)
Volume 13, Issue 3, Pages (March 2006)
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 13, Issue 4, Pages (April 2013)
Synthetic Lethal Screening Identifies Compounds Activating Iron-Dependent, Nonapoptotic Cell Death in Oncogenic-RAS-Harboring Cancer Cells  Wan Seok Yang,
Volume 21, Issue 6, Pages (March 2006)
Volume 10, Issue 3, Pages (September 2006)
Volume 50, Issue 2, Pages (April 2013)
Volume 10, Issue 3, Pages (September 2006)
Resistance of Human Melanoma Cells Against the Death Ligand TRAIL Is Reversed by Ultraviolet-B Radiation via Downregulation of FLIP  Elke Zeise, Michael.
Volume 9, Issue 5, Pages (May 2006)
MYC Is a Major Determinant of Mitotic Cell Fate
Inhibition of PAX3 by TGF-β Modulates Melanocyte Viability
Volume 12, Issue 1, Pages (July 2007)
Volume 13, Issue 4, Pages (April 2008)
Volume 25, Issue 4, Pages (April 2014)
Volume 12, Issue 1, Pages (July 2007)
Volume 8, Issue 4, Pages (October 2005)
Volume 12, Issue 4, Pages (July 2015)
Volume 16, Issue 10, Pages (May 2006)
MELK Promotes Melanoma Growth by Stimulating the NF-κB Pathway
Volume 15, Issue 3, Pages (March 2009)
Volume 7, Issue 3, Pages (March 2005)
Volume 39, Issue 3, Pages (August 2010)
Volume 8, Issue 5, Pages (September 2014)
A dual PI3 kinase/mTOR inhibitor reveals emergent efficacy in glioma
Shipra Das, Olga Anczuków, Martin Akerman, Adrian R. Krainer 
Oliver I. Fregoso, Shipra Das, Martin Akerman, Adrian R. Krainer 
Proapoptotic Function of the Retinoblastoma Tumor Suppressor Protein
Volume 7, Issue 6, Pages (June 2014)
Presentation transcript:

Volume 10, Issue 4, Pages 331-342 (October 2006) Gene expression-based chemical genomics identifies rapamycin as a modulator of MCL1 and glucocorticoid resistance  Guo Wei, David Twomey, Justin Lamb, Krysta Schlis, Jyoti Agarwal, Ronald W. Stam, Joseph T. Opferman, Stephen E. Sallan, Monique L. den Boer, Rob Pieters, Todd R. Golub, Scott A. Armstrong  Cancer Cell  Volume 10, Issue 4, Pages 331-342 (October 2006) DOI: 10.1016/j.ccr.2006.09.006 Copyright © 2006 Elsevier Inc. Terms and Conditions

Figure 1 Gene expression-based screening identifies rapamycin as a potential sensitizing agent to GC-induced apoptosis A: The top 50 probe sets that are correlated with either GC sensitivity or resistance are shown. A detailed list of genes can be found at http://www.broad.mit.edu/cgi-bin/cancer/datasets.cgi/. Permutation analysis identified 157 probes correlated with this distinction (p < 0.0005). B: GSEA analysis was performed using 163 pathway-associated gene sets curated by Biocarta to assess for enrichment of specific pathways in either resistant or sensitive samples. All pathways that demonstrated enrichment with p < 0.005 are demonstrated and ordered based on the p value. Note that all gene sets with enrichment were more highly expressed in the resistant ALL samples. C: Sirolimus (rapamycin) is positively connected with the GC resistance/sensitivity signature. The “barview” is constructed from 453 horizontal lines, each representing an individual treatment instance in the Connectivity Map data set, ordered by their corresponding connectivity score with the query signature. Each of the ten rapamycin instances in the data set is colored in black. Colors applied to the remaining instances reflect the sign of their scores (green, positive; gray, null; red, negative). D: CEM-c1 cells were treated with either DMSO or 10 nM rapamycin for 3 and 24 hr after which the gene expression profile was determined. The 157 probe sets used in 1C were assessed for overlap with those genes more highly expressed in the cells treated with DMSO (resistant) as compared to those treated with rapamycin (potentially sensitive). The blue lines demonstrate where the GC probe sets fall within the 22,000 probe sets ordered from left (1) to right (22,000) based on the DMSO/rapamycin distinction with gene #1 most highly expressed in DMSO-treated cells. The red line represents the running ES score that becomes more positive as probe sets are identified toward the top of the list before they would be expected if randomly distributed. Note that at 24 hr more probe sets (blue lines) are identified toward the top (left) of the ordered list of genes. Cancer Cell 2006 10, 331-342DOI: (10.1016/j.ccr.2006.09.006) Copyright © 2006 Elsevier Inc. Terms and Conditions

Figure 2 Rapamycin sensitizes lymphoid malignancy cells to GC-induced apoptosis A: Mouse T cell hybridoma 2B4 cells were engineered to express a constitutively active form of Akt by retroviral transduction. The GFP-positive Akt-transduced or control-transduced cells were treated with an increasing concentration of dexamethasone for 14 hr with or without 10 nM rapamycin, followed by FACS analysis. GFP-positive cells were assessed for viability by Annexin V staining. B: A panel of lymphoid malignancy-derived cell lines were treated with vehicle control, 10 nM rapamycin, 1 μM dexamethasone, or the combination of dexamethasone and rapamycin, and the percentage of viable cells was determined at 48 hr. C: Viable cells were assessed in T-ALL CEM-c1 cells after treatment with increasing concentrations of rapamycin at 24, 48, and 72 hr. D: The percentage of viable cells was determined in CEM-c1 cells 24 hr after treatment with an increasing concentration of dexamethasone ± pretreatment with 10 nM rapamycin. E: The percentage of viable cells was determined in 697 cells 24 hr after treatment with an increasing concentration of dexamethasone ± 10 nM rapamycin. F: The percentage of viable cells was determined in Ramos cells 48 hr after treatment with an increasing concentration of dexamethasone ± 10 nM rapamycin. Error bars represent the mean ± standard deviation. Cancer Cell 2006 10, 331-342DOI: (10.1016/j.ccr.2006.09.006) Copyright © 2006 Elsevier Inc. Terms and Conditions

Figure 3 Rapamycin specifically downregulates MCL1 among BCL2 family proteins A: Dexamethasone-induced expression of BH3-only Bim and Puma was not affected by rapamycin in CEM-c1 cells as determined by quantitative real-time PCR analysis. Error bars represent the mean ± standard deviation of triplicates. B: CEM-c1 cells were pretreated with or without Rapamycin for 16 hr, followed by 1 μM dexamethasone for the indicated time, and Bim expression was assessed by immunoblot. C: CEM-c1 cells were treated with rapamycin at the indicated concentration for 16 hr, and antiapoptotic protein expression was assessed by immunoblot. D–F: 697 cells (D), NAMALWA cells (E), or Ramos cells (F) were treated with rapamycin for 16 hr, and MCL1 levels were assessed by Western blotting. All cell lines demonstrated > 90% viability when they were assessed for MCL1 expression. G: MCL1 expression was assessed in 2B4 cells expressing constitutively active Akt as compared to vector control. Cancer Cell 2006 10, 331-342DOI: (10.1016/j.ccr.2006.09.006) Copyright © 2006 Elsevier Inc. Terms and Conditions

Figure 4 Expression of MCL1 confers GC resistance and sequesters BIM 2B4 cells overexpressing MCL1 are resistant to dexamethasone treatment. A: MCL1 overexpression in 2B4 cells by retroviral-mediated expression. B: 2B4 cells transduced with either an MCL1 retrovirus or vector control were treated with increasing concentrations of dexamethasone for 16 hr, followed by FACS analysis for Annexin V staining. C: 2B4 cells expressing FLAG-MCL1 or control were treated with increasing concentrations of dexamethasone and either 0 nM (0R), 1 nM (1R), or 10 nM (10R) rapamycin for 16 hr, followed by FACS analysis. D: 2B4 cells expressing FLAG-MCL1 were treated with 100 nM dexamethasone for the indicated time, and immunoprecipitation with an anti-FLAG M2 antibody was performed. Immunoprecipitated BIM and MCL1 were assessed by immunoblot. E: Bim expression was suppressed by lentivirus-mediated shRNA. Extra-long (EL), long (L), and short (S) isoforms of BIM were assessed by immunoblot. F: 2B4 cells transduced with the Bim-directed shRNA or a control luciferase-directed shRNA were treated with dexamethasone for 16 hr, followed by FACS analysis. Luc, control luciferase knockdown. BimKD, BIM knockdown. Error bars represent the mean ± standard deviation. Cancer Cell 2006 10, 331-342DOI: (10.1016/j.ccr.2006.09.006) Copyright © 2006 Elsevier Inc. Terms and Conditions

Figure 5 MCL1 confers GC resistance in primary lymphoid cells A: Illustration of H2K-Mcl1 transgene construct. B: An immunoblot for MCL1 expression in thymocytes from H2K-Mcl1 transgenic and wild-type mice. C: Immunoblot for MCL1 expression in splenocytes from H2K-Mcl1 transgenic and wild-type mice. D: Thymocytes from H2K-Mcl1 transgenic mice and littermate controls were treated with increasing concentrations of dexamethasone for 24 hr followed by FACS analysis for Annexin V−/PI− thymocytes. E: B220+, Annexin V−/PI− splenic B cells were counted as viable cells and standardized against cells without dexamethasone treatment. Four mice for each genotype were tested. Representative results are shown. Error bars represent the mean ± standard deviation. Cancer Cell 2006 10, 331-342DOI: (10.1016/j.ccr.2006.09.006) Copyright © 2006 Elsevier Inc. Terms and Conditions

Figure 6 MCL1, but not BCL2, is required for GC resistance in lymphoid malignancy cells A: MCL1 expression was assessed in CEM-c1 cells transduced with a lentivirus expressing either a luciferase-directed shRNA or an MCL1-directed shRNA, or after treatment with either 10 nM rapamycin or DMSO. B: CEM-c1 cells treated with either shRNA or rapamycin/DMSO as in A were treated with increasing concentrations of dexamethasone. Note that the CEM-c1 cells transduced with the MCL1 shRNA and the rapamycin-treated CEM-c1 cells are equally sensitive to dexamethasone. C: CEM-c1 cells treated with either control or MCL1-directed shRNAs were treated with increasing concentrations of doxorubicin. D: CEM-c1 cells treated with either control or MCL1-directed shRNAs were treated with increasing concentrations of etoposide. E: 697 cells treated with either control or MCL1-directed shRNAs were treated with increasing concentrations of dexamethasone. F: NAMALWA cells treated with either control or MCL1-directed shRNAs were treated with increasing concentrations of dexamethasone. G: BCL2 suppression in CEM-c1 cells transduced with a BCL2-directed shRNA. H: CEM-c1 cells with decreased BCL2 expression are still resistant to dexamethasone-induced apoptosis. Cells were subjected to increasing concentrations of dexamethasone for 24 hr followed by FACS analysis. Error bars represent the mean ± standard deviation. Cancer Cell 2006 10, 331-342DOI: (10.1016/j.ccr.2006.09.006) Copyright © 2006 Elsevier Inc. Terms and Conditions