29 results on '"Darshan S. Sappal"'
Search Results
2. Supplementary Figure 3 from Nedd8-Activating Enzyme Inhibitor MLN4924 Provides Synergy with Mitomycin C through Interactions with ATR, BRCA1/BRCA2, and Chromatin Dynamics Pathways
- Author
-
Eric S. Lightcap, Peter G. Smith, Syamala Bandi, Ray Liu, Michael D. Pickard, Mike Kuranda, Michael P. Thomas, Katherine Cosmopoulos, Greg Hather, Darshan S. Sappal, Xiaozhen J. Liu, David C. Bouck, Jonathan L. Blank, and Khristofer Garcia
- Abstract
PDF file - 339KB, Comparison of Isobolograms Calculated for Drug Response Surface Plots for 10 Different Agents.
- Published
- 2023
- Full Text
- View/download PDF
3. Data from Nedd8-Activating Enzyme Inhibitor MLN4924 Provides Synergy with Mitomycin C through Interactions with ATR, BRCA1/BRCA2, and Chromatin Dynamics Pathways
- Author
-
Eric S. Lightcap, Peter G. Smith, Syamala Bandi, Ray Liu, Michael D. Pickard, Mike Kuranda, Michael P. Thomas, Katherine Cosmopoulos, Greg Hather, Darshan S. Sappal, Xiaozhen J. Liu, David C. Bouck, Jonathan L. Blank, and Khristofer Garcia
- Abstract
MLN4924 is an investigational small-molecule inhibitor of the Nedd8-activating enzyme currently in phase I clinical trials. MLN4924 induces DNA damage via rereplication in most cell lines. This distinct mechanism of DNA damage may affect its ability to combine with standard-of-care agents and may affect the clinical development of MLN4924. As such, we studied its interaction with other DNA-damaging agents. Mitomycin C, cisplatin, cytarabine, UV radiation, SN-38, and gemcitabine demonstrated synergy in combination with MLN4924 in vitro. The combination of mitomycin C and MLN4924 was shown to be synergistic in a mouse xenograft model. Importantly, depletion of genes within the ataxia telangiectasia and Rad3 related (ATR) and BRCA1/BRCA2 pathways, chromatin modification, and transcription-coupled repair reduced the synergy between mitomycin C and MLN4924. In addition, comet assay demonstrated increased DNA strand breaks with the combination of MLN4924 and mitomycin C. Our data suggest that mitomycin C causes stalled replication forks, which when combined with rereplication induced by MLN4924 results in frequent replication fork collisions, leading to cell death. This study provides a straightforward approach to understand the mechanism of synergy, which may provide useful information for the clinical development of these combinations. Mol Cancer Ther; 13(6); 1625–35. ©2014 AACR.
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Figure 4 from Nedd8-Activating Enzyme Inhibitor MLN4924 Provides Synergy with Mitomycin C through Interactions with ATR, BRCA1/BRCA2, and Chromatin Dynamics Pathways
- Author
-
Eric S. Lightcap, Peter G. Smith, Syamala Bandi, Ray Liu, Michael D. Pickard, Mike Kuranda, Michael P. Thomas, Katherine Cosmopoulos, Greg Hather, Darshan S. Sappal, Xiaozhen J. Liu, David C. Bouck, Jonathan L. Blank, and Khristofer Garcia
- Abstract
PDF file - 145KB, Comparison of the Degree of Interaction between MLN4924 and 14 Different Agents as Measured by Nonlinear Blending Values.
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Figure 1 from Nedd8-Activating Enzyme Inhibitor MLN4924 Provides Synergy with Mitomycin C through Interactions with ATR, BRCA1/BRCA2, and Chromatin Dynamics Pathways
- Author
-
Eric S. Lightcap, Peter G. Smith, Syamala Bandi, Ray Liu, Michael D. Pickard, Mike Kuranda, Michael P. Thomas, Katherine Cosmopoulos, Greg Hather, Darshan S. Sappal, Xiaozhen J. Liu, David C. Bouck, Jonathan L. Blank, and Khristofer Garcia
- Abstract
PDF file - 1215KB, Full Length Western Blots.
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Figure 2 from Nedd8-Activating Enzyme Inhibitor MLN4924 Provides Synergy with Mitomycin C through Interactions with ATR, BRCA1/BRCA2, and Chromatin Dynamics Pathways
- Author
-
Eric S. Lightcap, Peter G. Smith, Syamala Bandi, Ray Liu, Michael D. Pickard, Mike Kuranda, Michael P. Thomas, Katherine Cosmopoulos, Greg Hather, Darshan S. Sappal, Xiaozhen J. Liu, David C. Bouck, Jonathan L. Blank, and Khristofer Garcia
- Abstract
PDF file - 664KB, Sensitivity of Cancer Cell Lines to DNA Damaging Agents.
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Materials and Methods, Figure Legends from Nedd8-Activating Enzyme Inhibitor MLN4924 Provides Synergy with Mitomycin C through Interactions with ATR, BRCA1/BRCA2, and Chromatin Dynamics Pathways
- Author
-
Eric S. Lightcap, Peter G. Smith, Syamala Bandi, Ray Liu, Michael D. Pickard, Mike Kuranda, Michael P. Thomas, Katherine Cosmopoulos, Greg Hather, Darshan S. Sappal, Xiaozhen J. Liu, David C. Bouck, Jonathan L. Blank, and Khristofer Garcia
- Abstract
PDF file - 68KB
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Figure 5 from Nedd8-Activating Enzyme Inhibitor MLN4924 Provides Synergy with Mitomycin C through Interactions with ATR, BRCA1/BRCA2, and Chromatin Dynamics Pathways
- Author
-
Eric S. Lightcap, Peter G. Smith, Syamala Bandi, Ray Liu, Michael D. Pickard, Mike Kuranda, Michael P. Thomas, Katherine Cosmopoulos, Greg Hather, Darshan S. Sappal, Xiaozhen J. Liu, David C. Bouck, Jonathan L. Blank, and Khristofer Garcia
- Abstract
PDF file - 159KB, The Combination of MLN4924 and Mitomycin C Results in a BRCA1-dependent Activation of ATR following Induction of DNA Strand Breaks.
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Table 1 from Nedd8-Activating Enzyme Inhibitor MLN4924 Provides Synergy with Mitomycin C through Interactions with ATR, BRCA1/BRCA2, and Chromatin Dynamics Pathways
- Author
-
Eric S. Lightcap, Peter G. Smith, Syamala Bandi, Ray Liu, Michael D. Pickard, Mike Kuranda, Michael P. Thomas, Katherine Cosmopoulos, Greg Hather, Darshan S. Sappal, Xiaozhen J. Liu, David C. Bouck, Jonathan L. Blank, and Khristofer Garcia
- Abstract
XLS file - 1161KB, Data Supporting Figures within Paper.
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Figures 5-7 from Genome-Wide siRNA Screen for Modulators of Cell Death Induced by Proteasome Inhibitor Bortezomib
- Author
-
Eric S. Lightcap, Jay P. Morgenstern, James J. Spelman, Claudia Rabino, John Ringeling, Patrick J. Leroy, Paul Hales, Jesse Gray, Darshan S. Sappal, Khristofer Garcia, Wei Chen, Anne L. Burkhardt, Trupti Lingaraj, Denise L. Driscoll, Jie Yu, Saurabh Menon, Michael D. Pickard, David M. Rappoli, Xiaozhen J. Liu, Theodore Peters, Jonathan L. Blank, and Siquan Chen
- Abstract
Supplementary Figures 5-7 from Genome-Wide siRNA Screen for Modulators of Cell Death Induced by Proteasome Inhibitor Bortezomib
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Figure 8 from Genome-Wide siRNA Screen for Modulators of Cell Death Induced by Proteasome Inhibitor Bortezomib
- Author
-
Eric S. Lightcap, Jay P. Morgenstern, James J. Spelman, Claudia Rabino, John Ringeling, Patrick J. Leroy, Paul Hales, Jesse Gray, Darshan S. Sappal, Khristofer Garcia, Wei Chen, Anne L. Burkhardt, Trupti Lingaraj, Denise L. Driscoll, Jie Yu, Saurabh Menon, Michael D. Pickard, David M. Rappoli, Xiaozhen J. Liu, Theodore Peters, Jonathan L. Blank, and Siquan Chen
- Abstract
Supplementary Figure 8 from Genome-Wide siRNA Screen for Modulators of Cell Death Induced by Proteasome Inhibitor Bortezomib
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Methods, Figure and Table Legends from Genome-Wide siRNA Screen for Modulators of Cell Death Induced by Proteasome Inhibitor Bortezomib
- Author
-
Eric S. Lightcap, Jay P. Morgenstern, James J. Spelman, Claudia Rabino, John Ringeling, Patrick J. Leroy, Paul Hales, Jesse Gray, Darshan S. Sappal, Khristofer Garcia, Wei Chen, Anne L. Burkhardt, Trupti Lingaraj, Denise L. Driscoll, Jie Yu, Saurabh Menon, Michael D. Pickard, David M. Rappoli, Xiaozhen J. Liu, Theodore Peters, Jonathan L. Blank, and Siquan Chen
- Abstract
Supplementary Methods, Figure and Table Legends from Genome-Wide siRNA Screen for Modulators of Cell Death Induced by Proteasome Inhibitor Bortezomib
- Published
- 2023
- Full Text
- View/download PDF
13. Data from Genome-Wide siRNA Screen for Modulators of Cell Death Induced by Proteasome Inhibitor Bortezomib
- Author
-
Eric S. Lightcap, Jay P. Morgenstern, James J. Spelman, Claudia Rabino, John Ringeling, Patrick J. Leroy, Paul Hales, Jesse Gray, Darshan S. Sappal, Khristofer Garcia, Wei Chen, Anne L. Burkhardt, Trupti Lingaraj, Denise L. Driscoll, Jie Yu, Saurabh Menon, Michael D. Pickard, David M. Rappoli, Xiaozhen J. Liu, Theodore Peters, Jonathan L. Blank, and Siquan Chen
- Abstract
Multiple pathways have been proposed to explain how proteasome inhibition induces cell death, but mechanisms remain unclear. To approach this issue, we performed a genome-wide siRNA screen to evaluate the genetic determinants that confer sensitivity to bortezomib (Velcade (R); PS-341). This screen identified 100 genes whose knockdown affected lethality to bortezomib and to a structurally diverse set of other proteasome inhibitors. A comparison of three cell lines revealed that 39 of 100 genes were commonly linked to cell death. We causally linked bortezomib-induced cell death to the accumulation of ASF1B, Myc, ODC1, Noxa, BNIP3, Gadd45α, p-SMC1A, SREBF1, and p53. Our results suggest that proteasome inhibition promotes cell death primarily by dysregulating Myc and polyamines, interfering with protein translation, and disrupting essential DNA damage repair pathways, leading to programmed cell death. Cancer Res; 70(11); 4318–26. ©2010 AACR.
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Figures 1-4 from Genome-Wide siRNA Screen for Modulators of Cell Death Induced by Proteasome Inhibitor Bortezomib
- Author
-
Eric S. Lightcap, Jay P. Morgenstern, James J. Spelman, Claudia Rabino, John Ringeling, Patrick J. Leroy, Paul Hales, Jesse Gray, Darshan S. Sappal, Khristofer Garcia, Wei Chen, Anne L. Burkhardt, Trupti Lingaraj, Denise L. Driscoll, Jie Yu, Saurabh Menon, Michael D. Pickard, David M. Rappoli, Xiaozhen J. Liu, Theodore Peters, Jonathan L. Blank, and Siquan Chen
- Abstract
Supplementary Figures 1-4 from Genome-Wide siRNA Screen for Modulators of Cell Death Induced by Proteasome Inhibitor Bortezomib
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Table 3A-F from Genome-Wide siRNA Screen for Modulators of Cell Death Induced by Proteasome Inhibitor Bortezomib
- Author
-
Eric S. Lightcap, Jay P. Morgenstern, James J. Spelman, Claudia Rabino, John Ringeling, Patrick J. Leroy, Paul Hales, Jesse Gray, Darshan S. Sappal, Khristofer Garcia, Wei Chen, Anne L. Burkhardt, Trupti Lingaraj, Denise L. Driscoll, Jie Yu, Saurabh Menon, Michael D. Pickard, David M. Rappoli, Xiaozhen J. Liu, Theodore Peters, Jonathan L. Blank, and Siquan Chen
- Abstract
Supplementary Table 3A-F from Genome-Wide siRNA Screen for Modulators of Cell Death Induced by Proteasome Inhibitor Bortezomib
- Published
- 2023
- Full Text
- View/download PDF
16. Nedd8-Activating Enzyme Inhibitor MLN4924 Provides Synergy with Mitomycin C through Interactions with ATR, BRCA1/BRCA2, and Chromatin Dynamics Pathways
- Author
-
David C. Bouck, Greg Hather, Xiaozhen J. Liu, Darshan S. Sappal, Jonathan L. Blank, Michael D. Pickard, Eric S. Lightcap, Ray Liu, Syamala Bandi, Katherine Cosmopoulos, Khristofer Garcia, Michael P. Thomas, Peter G. Smith, and Mike Kuranda
- Subjects
Cancer Research ,Programmed cell death ,Ultraviolet Rays ,DNA damage ,Mitomycin ,Apoptosis ,Ataxia Telangiectasia Mutated Proteins ,Cyclopentanes ,Ubiquitin-Activating Enzymes ,Biology ,Mice ,chemistry.chemical_compound ,Cell Line, Tumor ,medicine ,Animals ,Humans ,BRCA2 Protein ,Cisplatin ,BRCA1 Protein ,Mitomycin C ,Drug Synergism ,Xenograft Model Antitumor Assays ,Molecular biology ,Chromatin ,Comet assay ,Pyrimidines ,Oncology ,chemistry ,Cancer research ,Ataxia telangiectasia and Rad3 related ,DNA ,DNA Damage ,medicine.drug - Abstract
MLN4924 is an investigational small-molecule inhibitor of the Nedd8-activating enzyme currently in phase I clinical trials. MLN4924 induces DNA damage via rereplication in most cell lines. This distinct mechanism of DNA damage may affect its ability to combine with standard-of-care agents and may affect the clinical development of MLN4924. As such, we studied its interaction with other DNA-damaging agents. Mitomycin C, cisplatin, cytarabine, UV radiation, SN-38, and gemcitabine demonstrated synergy in combination with MLN4924 in vitro. The combination of mitomycin C and MLN4924 was shown to be synergistic in a mouse xenograft model. Importantly, depletion of genes within the ataxia telangiectasia and Rad3 related (ATR) and BRCA1/BRCA2 pathways, chromatin modification, and transcription-coupled repair reduced the synergy between mitomycin C and MLN4924. In addition, comet assay demonstrated increased DNA strand breaks with the combination of MLN4924 and mitomycin C. Our data suggest that mitomycin C causes stalled replication forks, which when combined with rereplication induced by MLN4924 results in frequent replication fork collisions, leading to cell death. This study provides a straightforward approach to understand the mechanism of synergy, which may provide useful information for the clinical development of these combinations. Mol Cancer Ther; 13(6); 1625–35. ©2014 AACR.
- Published
- 2014
- Full Text
- View/download PDF
17. Abstract B127: Evaluating PRKACA as a therapeutic target for Fibrolamellar Carcinoma
- Author
-
Darshan S. Sappal, Vivek Kadambi, Riadh Lobbardi, Joseph L. Kim, Adam Shutes, Michael Palmer, Nicolas Stransky, Robert M. Campbell, Richard Woessner, Timothy LaBranche, Klaus P. Hoeflich, Neil Bifulco, Andrew P. Garner, Grace O. Silva, Erin O’Hearn, Kevin J. Wilson, Timothy J. Guzi, Stefanie Schalm, Marion Dorsch, Lucian DiPietro, Stephen D. Miller, and Christoph Lengauer
- Subjects
Cancer Research ,business.industry ,Kinase ,Cancer ,Malignancy ,medicine.disease ,Chronic liver disease ,PRKACA ,Small hairpin RNA ,Therapeutic approach ,Oncology ,hemic and lymphatic diseases ,Cancer research ,Medicine ,business ,Fibrolamellar Carcinoma - Abstract
Introduction: Fibrolamellar Carcinoma (FLC) is a rare primary liver malignancy, affecting children and young adults without chronic liver disease. FLC tumors are largely resistant to chemotherapy, making the identification of effective treatment options urgently needed. Recent genomic data strongly suggest that DNAJB1-PRKACA kinase fusions are the drivers of the vast majority of FLC cases. However, it has not been assessed whether FLC tumors remain dependent on DNAJB1-PRKACA expression and whether PRKACA inhibition could be a therapeutic approach for FLC. Here we summarize the preclinical evaluation of PRKACA as a potential therapeutic target for FLC. Methods: We established a xenograft model from a FLC- patient and then developed inducible PRKACA shRNA cell lines from this model. We also designed potent tool compounds that selectively inhibit the PRKACA protein to assess PRKACA as a potential therapeutic target for FLC. Results: We characterized a patient-derived xenograft (PDX) model of FLC (LI5132) and confirmed DNAJB1-PRKACA fusion expression and constitutive PRKACA pathway activation measured by phospho-VASP. The model also shows fibrolamellar type histology by H&E staining and expression of typical FLC markers like cytokeratin 7 and CD68 by IHC. Using inducible PRKACA-specific shRNA cell lines from this PDX model we demonstrated that the FLC transcriptional gene signature correlates strongly with expression of the DNAJB1-PRKACAfusion protein. Importantly, we demonstrated for three inducible PRKACA shRNA cell-line-derived xenograft models that the in vivo tumor growth remained dependent on DNAJB1-PRKACA fusion expression (TGI-72%-78%, day 22). PRKACA knockdown tumors displayed reduced Ki67 index (6.4 %) when compared to non-induced controls (37.1 %) further confirming that proliferation of the tumors depends on the fusion expression. To investigate the PRKACA catalytic dependency of the FLC model, we designed potent and selective PRKACA inhibitors based on starting points from our proprietary kinase inhibitor library. These investigational compounds are the first selective and potent PRKACA inhibitors and provide excellent tools to assess in vitro and in vivo PRKACA dependency. These compounds achieved potent PRKACA pathway inhibition and dose-dependent inhibition of FLC-specific gene expression, including genes such as carbamoyl phosphate synthetase (CPS1) and forkhead box C1 (FoxC1). We established a pharmacokinetic/ pharmacodynamic relationship and demonstrated in vivo PRKACA pathway inhibition in PDX tumors, as measured by phospho-VASP. Importantly, oral delivery of a potent and selective PRKACA inhibitor achieved up to 80% PRKACA kinase inhibition and led to statistically significant FLC tumor growth inhibition (54%, day 34) on a tolerated schedule. These data demonstrate that FLC depends on PRKACA kinase activity. Conclusion: This study is the first evaluation of PRKACA kinase inhibition as a therapeutic approach for FLC. The results from these preclinical experiments provide strong evidence that FLC depends on PRKACA catalytic activity and that novel PRKACA inhibitors may significantly decrease tumor growth in vivo. Citation Format: Stefanie S Schalm, Erin O’Hearn, Kevin Wilson, Timothy LaBranche, Grace Silva, Lucian DiPietro, Neil Bifulco, Richard Woessner, Nicolas Stransky, Darshan Sappal, Adam Shutes, Robert Campbell, Riadh Lobbardi, Michael Palmer, Joseph Kim, Stephen Miller, Marion Dorsch, Christoph Lengauer, Timothy Guzi, Vivek Kadambi, Andrew Garner, Klaus P Hoeflich. Evaluating PRKACA as a therapeutic target for Fibrolamellar Carcinoma [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics; 2019 Oct 26-30; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2019;18(12 Suppl):Abstract nr B127. doi:10.1158/1535-7163.TARG-19-B127
- Published
- 2019
- Full Text
- View/download PDF
18. A small-molecule inhibitor of the ubiquitin activating enzyme for cancer treatment
- Author
-
John Newcomb, James M. Gavin, Nancy J. Bump, Stephen Tirrell, Lawrence R. Dick, Saurabh Menon, Jessica Huck, Petter Veiby, Benjamin S. Amidon, Yu Yang, Marc L. Hyer, Paul D. Greenspan, Fleming Paul E, Teresa A. Soucy, Jim Brownell, Michael Sintchak, Josh Powe, Steve Langston, Mark Manfredi, Judy Shi, Jeff Ciavarri, Darshan S. Sappal, Frank Bruzzese, Mike Kuranda, Katherine Galvin, Michael Milhollen, Ping Li, Neil F. Bence, Jing Tao Wu, Claudia Rabino, Chris Claiborne, Tary Traore, Jennifer Duffy, Jessica Riceberg, Robert J. Griffin, Kara Hoar, Anya Lublinsky, Bradley Stringer, and Sai M Pulukuri
- Subjects
0301 basic medicine ,Proteasome Endopeptidase Complex ,DNA Repair ,Ubiquitin-activating enzyme ,Plasma protein binding ,Ubiquitin-Activating Enzymes ,Sulfides ,Imides ,General Biochemistry, Genetics and Molecular Biology ,Small Molecule Libraries ,03 medical and health sciences ,Mice ,Ubiquitin ,Cell Line, Tumor ,Neoplasms ,Animals ,Humans ,chemistry.chemical_classification ,Sulfonamides ,biology ,Chemistry ,Nucleosides ,General Medicine ,Small molecule ,Xenograft Model Antitumor Assays ,030104 developmental biology ,Enzyme ,Pyrimidines ,Proteasome ,Cell culture ,Cancer cell ,Cancer research ,biology.protein ,Pyrazoles ,DNA Damage ,Protein Binding - Abstract
The ubiquitin-proteasome system (UPS) comprises a network of enzymes that is responsible for maintaining cellular protein homeostasis. The therapeutic potential of this pathway has been validated by the clinical successes of a number of UPS modulators, including proteasome inhibitors and immunomodulatory imide drugs (IMiDs). Here we identified TAK-243 (formerly known as MLN7243) as a potent, mechanism-based small-molecule inhibitor of the ubiquitin activating enzyme (UAE), the primary mammalian E1 enzyme that regulates the ubiquitin conjugation cascade. TAK-243 treatment caused depletion of cellular ubiquitin conjugates, resulting in disruption of signaling events, induction of proteotoxic stress, and impairment of cell cycle progression and DNA damage repair pathways. TAK-243 treatment caused death of cancer cells and, in primary human xenograft studies, demonstrated antitumor activity at tolerated doses. Due to its specificity and potency, TAK-243 allows for interrogation of ubiquitin biology and for assessment of UAE inhibition as a new approach for cancer treatment.
- Published
- 2016
19. Characterization of a new series of non-covalent proteasome inhibitors with exquisite potency and selectivity for the 20S β5-subunit
- Author
-
Edward J. Olhava, Paul Hales, Cynthia Barrett, Jane X. Liu, Matthew Jones, Frank J. Bruzzese, Kenneth M. Gigstad, Khristofer Garcia, Christopher Tsu, Darshan S. Sappal, Teresa A. Soucy, Paul E. Fleming, Christopher Blackburn, Michael D. Sintchak, Jonathan L. Blank, Nancy Bump, and Lawrence R. Dick
- Subjects
Models, Molecular ,IκB, inhibitory protein of NFκB ,Plasma protein binding ,PA, proteasomal activator ,Crystallography, X-Ray ,β5-subunit ,TNF-α, tumour necrosis factor-α ,Biochemistry ,Bortezomib ,Ubiquitin ,Correction Article ,Luciferases ,26S proteasome ,ubiquitin–proteasome system (UPS) ,Molecular Structure ,biology ,NF-kappa B ,Boronic Acids ,Boc, t-butoxycarbonyl ,RNAi, RNA interference ,Pyrazines ,RNA Interference ,HT29 Cells ,Oligopeptides ,Proteasome Inhibitors ,Protein Binding ,Research Article ,medicine.drug ,Proteasome Endopeptidase Complex ,UPS, ubiquitin–proteasome system ,LC50, half-maximal lethal concentration ,Molecular Sequence Data ,Cell Line ,TEV, tobacco etch virus ,immunoproteasome ,HEK, human embryonic kidney ,PDL, poly-D-lysine ,Cell Line, Tumor ,medicine ,Humans ,Protease Inhibitors ,Amino Acid Sequence ,AMC, 7-amino-4-methylcoumarin ,Suc, succinyl ,Binding site ,Z, benzyloxycarbonyl ,Molecular Biology ,Cell Proliferation ,Ac, acetyl ,Binding Sites ,Sequence Homology, Amino Acid ,proteasome inhibitor ,HBTU, O-benzotriazole-N,N,N′,N′-tetramethyluronium hexafluorophosphate ,chymotrypsin-like ,MPD, 2-methyl-2,4-pentanediol ,Cell Biology ,HCT116 Cells ,NFKB1 ,In vitro ,Protein Structure, Tertiary ,NFκB-Luc, NFκB–luciferase ,Kinetics ,Protein Subunits ,NFκB, nuclear factor κB ,4xUb-Luc, tetra-ubiquitin–luciferase ,Proteasome ,siRNA, small interfering RNA ,Cancer cell ,biology.protein ,Proteasome inhibitor - Abstract
The mammalian 26S proteasome is a 2500 kDa multi-catalytic complex involved in intracellular protein degradation. We describe the synthesis and properties of a novel series of non-covalent di-peptide inhibitors of the proteasome based [corrected] on a capped tri-peptide that was first identified by high-throughput screening of a library of approx. 350000 compounds for inhibitors of the ubiquitin-proteasome system in cells. We show that these compounds are entirely selective for the beta5 (chymotrypsin-like) site over the beta1 (caspase-like) and beta2 (trypsin-like) sites of the 20S core particle of the proteasome, and over a panel of less closely related proteases. Compound optimization, guided by X-ray crystallography of the liganded 20S core particle, confirmed their non-covalent binding mode and provided a structural basis for their enhanced in vitro and cellular potencies. We demonstrate that such compounds show low nanomolar IC50 values for the human 20S beta5 site in vitro, and that pharmacological inhibition of this site in cells is sufficient to potently inhibit the degradation of a tetra-ubiquitin-luciferase reporter, activation of NFkappaB (nuclear factor kappaB) in response to TNF-alpha (tumour necrosis factor-alpha) and the proliferation of cancer cells. Finally, we identified capped di-peptides that show differential selectivity for the beta5 site of the constitutively expressed proteasome and immunoproteasome in vitro and in B-cell lymphomas. Collectively, these studies describe the synthesis, activity and binding mode of a new series of non-covalent proteasome inhibitors with unprecedented potency and selectivity for the beta5 site, and which can discriminate between the constitutive proteasome and immunoproteasome in vitro and in cells.
- Published
- 2010
- Full Text
- View/download PDF
20. Genome-Wide siRNA Screen for Modulators of Cell Death Induced by Proteasome Inhibitor Bortezomib
- Author
-
Paul Hales, Saurabh Menon, Theodore Peters, Claudia Rabino, Khristofer Garcia, Jesse Gray, John Ringeling, Anne L. Burkhardt, Patrick J. LeRoy, Siquan Chen, Jay P. Morgenstern, Jonathan L. Blank, Wei Chen, Trupti Lingaraj, Darshan S. Sappal, Michael D. Pickard, Jie Yu, David M. Rappoli, Eric S. Lightcap, Xiaozhen J. Liu, Denise L. Driscoll, and James J. Spelman
- Subjects
Cancer Research ,Programmed cell death ,DNA damage ,Antineoplastic Agents ,Protein Serine-Threonine Kinases ,Biology ,Transfection ,Bortezomib ,Proto-Oncogene Proteins c-myc ,medicine ,Humans ,Protease Inhibitors ,RNA, Small Interfering ,Melanoma ,Gene knockdown ,Cell Death ,TOR Serine-Threonine Kinases ,Intracellular Signaling Peptides and Proteins ,HCT116 Cells ,Boronic Acids ,Oncology ,Proteasome ,Cell culture ,Gene Knockdown Techniques ,Pyrazines ,Colonic Neoplasms ,Proteasome inhibitor ,Cancer research ,Proteasome Inhibitors ,Ribosomes ,DNA Damage ,HeLa Cells ,medicine.drug - Abstract
Multiple pathways have been proposed to explain how proteasome inhibition induces cell death, but mechanisms remain unclear. To approach this issue, we performed a genome-wide siRNA screen to evaluate the genetic determinants that confer sensitivity to bortezomib (Velcade (R); PS-341). This screen identified 100 genes whose knockdown affected lethality to bortezomib and to a structurally diverse set of other proteasome inhibitors. A comparison of three cell lines revealed that 39 of 100 genes were commonly linked to cell death. We causally linked bortezomib-induced cell death to the accumulation of ASF1B, Myc, ODC1, Noxa, BNIP3, Gadd45α, p-SMC1A, SREBF1, and p53. Our results suggest that proteasome inhibition promotes cell death primarily by dysregulating Myc and polyamines, interfering with protein translation, and disrupting essential DNA damage repair pathways, leading to programmed cell death. Cancer Res; 70(11); 4318–26. ©2010 AACR.
- Published
- 2010
- Full Text
- View/download PDF
21. The antiproliferative agent MLN944 preferentially inhibits transcription
- Author
-
David H. Price, Jeffery Brown, Sarah A. Byers, Darshan S. Sappal, and Blanca Schafer
- Subjects
Cancer Research ,Transcription, Genetic ,Antineoplastic Agents ,RNA polymerase II ,HeLa ,chemistry.chemical_compound ,Transcription (biology) ,Tumor Cells, Cultured ,Humans ,Polymerase ,Cell Proliferation ,Nucleic Acid Synthesis Inhibitors ,biology ,Topoisomerase ,RNA ,biology.organism_classification ,Molecular biology ,Growth Inhibitors ,Oncology ,chemistry ,Dactinomycin ,biology.protein ,Phenazines ,RNA Polymerase II ,Transcription factor II B ,DNA - Abstract
MLN944 is a novel compound currently being codeveloped by Millennium Pharmaceuticals and Xenova Ltd. as a cancer therapeutic and is in a phase I clinical trial for solid tumors. Although MLN944 was originally proposed to function as a topoisomerase I and II inhibitor, more recent data has shown that it is a DNA-intercalating agent that does not inhibit the catalytic activity of topoisomerase I or II. We show here that MLN944 inhibits incorporation of radiolabeled precursors into RNA preferentially over incorporation into DNA and protein in HCT116 and H460 cells. To determine if MLN944 inhibits transcription, a human RNA polymerase II in vitro transcription system was used. MLN944 inhibited initiation when added before or after the formation of preinitiation complexes and inhibited elongation at higher concentrations. The preferential inhibition of initiation differentiates MLN944 from actinomycin D, which more strongly inhibits elongation. Transcription of all RNA polymerases was inhibited in nuclei isolated from HeLa cells treated with low concentrations of MLN944. Our data are consistent with transcription as the target of the potent cytotoxic effects of MLN944.
- Published
- 2005
- Full Text
- View/download PDF
22. Biological characterization of MLN944: A potent DNA binding agent
- Author
-
Darshan S. Sappal, A. Kathleen McClendon, James A. Fleming, Vala Thoroddsen, Kelly Connolly, Corinne Reimer, Ronald K. Blackman, Christine E. Bulawa, Neil Osheroff, Peter Charlton, and Laura A. Rudolph-Owen
- Subjects
Cancer Research ,Oncology - Abstract
MLN944 (XR5944) is a novel bis-phenazine that has demonstrated exceptional efficacy against a number of murine and human tumor models. The drug was reported originally as a dual topoisomerase I/II poison, but a precise mechanism of action for this compound remains to be determined. Several lines of evidence, including the marginal ability of MLN944 to stabilize topoisomerase-dependent cleavage, and the sustained potency of MLN944 in mammalian cells with reduced levels of both topoisomerases, suggest that other activities of the drug exist. In this study, we show that MLN944 intercalates into DNA, but has no effect on the catalytic activity of either topoisomerase I or II. MLN944 displays no significant ability to stimulate DNA scission mediated by either topoisomerase I or II compared with camptothecin or etoposide, respectively. In addition, yeast genetic models also point toward a topoisomerase-independent mechanism of action. To examine cell cycle effects, synchronized human HCT116 cells were treated with MLN944, doxorubicin, camptothecin, or a combination of the latter two to mimic a dual topoisomerase poison. MLN944 treatment was found to induce a G1 and G2 arrest in cells that is unlike the typical G2-M arrest noted with known topoisomerase poisons. Finally, transcriptional profiling analysis of xenograft tumors treated with MLN944 revealed clusters of regulated genes distinct from those observed in irinotecan hydrochloride (CPT-11)-treated tumors. Taken together, these findings suggest that the primary mechanism of action of MLN944 likely involves DNA binding and intercalation, but does not appear to involve topoisomerase inhibition.
- Published
- 2004
- Full Text
- View/download PDF
23. Abstract 3719: TAK-243, a small molecule inhibitor of the ubiquitin activating enzyme (UAE), disrupts DNA damage repair and sensitizes tumor cells and xenografts to ionizing radiation
- Author
-
Allison Berger, Darshan S. Sappal, Kenichi Iwai, Yuko Ishii, Neil F. Bence, Jennifer Duffy, Eric S. Lightcap, Jeffrey Ciavarri, Marc L. Hyer, Akihiro Ohashi, Jessica Huck, Judy Qiuju Shi, Tary Traore, Michael Milhollen, and Claudia Rabino
- Subjects
Cancer Research ,biology ,DNA damage ,DNA repair ,Ubiquitin-activating enzyme ,Cancer ,medicine.disease ,Molecular biology ,Proliferating cell nuclear antigen ,Oncology ,Ubiquitin ,FANCD2 ,biology.protein ,medicine ,Homologous recombination - Abstract
Radiation therapy, as a primary therapy or as a combination partner, is used in half of all worldwide cancer treatments. Research is ongoing to identify agents which potentiate the effects of ionizing radiation (IR) in tumor cells. Because IR causes DNA double strand breaks (DSBs), inhibition of DNA damage repair mechanisms could enhance the effects of radiation. DNA repair at DSBs is mediated by the non-homologous end-joining (NHEJ) and homologous recombination (HR) pathways, both of which rely on the post-translational modification of proteins by ubiquitin (Ub). A phosphorylation and ubiqutination cascasde at DSBs results in Ub-dependent recruitment of 53BP1 and BRCA1 complexes. We have identified a first in class investigational drug, TAK-243 (MLN7243), which targets the ubiquitin activating enzyme, UAE (UBA1), the enzyme responsible for activating > 99% of all cellular Ub. Previously, TAK-243 was shown to inhibit mono-Ub of PCNA and FANCD2, key proteins within the translesion synthesis (TLS) and Fanconi Anemia (FA) DNA repair pathways, and also to inhibit Ub transfer to UBC13, an E2 ubiquitin-conjugating enzyme utilized in DSB repair. We hypothesized that TAK-243 would prevent repair of DSBs and thereby potentiate IR-induced cell death. Here we show that TAK-243 pre-treatment potentiates the effect of IR on HCT-116 cells in a colony formation assay in vitro. To link this combination benefit to the disruption of DNA damage repair, we demonstrate that TAK-243 pre-treatment blocks the IR-induced recruitment of 53BP1 to sites of DNA damage both in vitro and in vivo. In a patient-derived xenograft (PDX) model of non-small cell lung cancer, formation of IR-induced 53BP1 foci is inhibited when TAK-243 is dosed 1 hour before beam-focused radiation exposure. In contrast, levels of IR-induced pH2Ax are not significantly changed by TAK-243 treatment, suggesting that TAK-243 does not prevent formation or detection of DSBs, but rather acts downstream to prevent DNA damage repair. Additive-to-synergistic effects on tumor growth inhibition were observed in several xenograft models treated with the combination of TAK-243 and beam-focused IR, with persistent tumor regressions noted in some NSCLC and breast cancer models. The results of our experiments provide a mechanistic rationale for combining radiation with TAK-243 in the clinical setting. Currently, TAK-243 is being evaluated in a solid tumor phase I clinical trial evaluating safety, tolerability, pharmacokinetics, pharmacodynamics and anti-tumor activity (NCT02045095). Citation Format: Michael A. Milhollen, Judy Qiuju Shi, Tary Traore, Jessica Huck, Darshan Sappal, Kenichi Iwai, Akihiro Ohashi, Claudia Rabino, Jennifer A. Duffy, Eric Lightcap, Yuko Ishii, Jeffrey Ciavarri, Neil Bence, Allison J. Berger, Marc L. Hyer. TAK-243, a small molecule inhibitor of the ubiquitin activating enzyme (UAE), disrupts DNA damage repair and sensitizes tumor cells and xenografts to ionizing radiation. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3719.
- Published
- 2016
- Full Text
- View/download PDF
24. Optimization of a series of dipeptides with a P3 threonine residue as non-covalent inhibitors of the chymotrypsin-like activity of the human 20S proteasome
- Author
-
Kenneth M. Gigstad, Khristofer Garcia, Paul Hales, Nancy Bump, Frank J. Bruzzese, Paul E. Fleming, Lawrence R. Dick, Zhigen Hu, Christopher Blackburn, Matthew Jones, Christopher Tsu, Michael D. Sintchak, Cynthia Barrett, Jane X. Liu, Jonathan L. Blank, and Darshan S. Sappal
- Subjects
Models, Molecular ,Threonine ,Proteasome Endopeptidase Complex ,Stereochemistry ,Clinical Biochemistry ,Pharmaceutical Science ,Tripeptide ,Biochemistry ,20s proteasome ,Residue (chemistry) ,chemistry.chemical_compound ,Drug Discovery ,Chymotrypsin ,Humans ,Chymotrypsin like ,Molecular Biology ,Dipeptide ,biology ,Organic Chemistry ,Active site ,Dipeptides ,Proteasome ,chemistry ,biology.protein ,Molecular Medicine - Abstract
Starting from a tripeptide screening hit, a series of dipeptide inhibitors of the proteasome with Thr as the P3 residue has been optimized with the aid of crystal structures in complex with the β-5/6 active site of y20S. Derivative 25, (β5 IC(50)=7.4 nM) inhibits only the chymotryptic activity of the proteasome, shows cellular activity against targets in the UPS, and inhibits proliferation.
- Published
- 2010
25. Abstract A164: The small molecule UAE inhibitor TAK-243 (MLN7243) prevents DNA damage repair and reduces cell viability/tumor growth when combined with radiation, carboplatin and docetaxel
- Author
-
Michael Milhollen, Tary Traore, Marc L. Hyer, Jennifer Duffy, Yuko Ishii, Paul E. Fleming, Jeff Ciavarri, Judi Shi, Darshan S. Sappal, Eric S. Lightcap, Neil F. Bence, and Jessica Huck
- Subjects
UAE Inhibitor TAK-243 ,Cancer Research ,biology ,Cell growth ,DNA damage ,Ubiquitin-activating enzyme ,Carboplatin ,Proliferating cell nuclear antigen ,Comet assay ,chemistry.chemical_compound ,Oncology ,chemistry ,Ubiquitin ,Immunology ,biology.protein ,Cancer research - Abstract
Clinical results of VELCADE® (bortezomib) For Injection have prompted evaluation of other enzymes within the ubiquitin proteasome system (UPS) as druggable targets for human cancer. We have identified a first in class investigational drug, TAK-243 (MLN7243), which targets the ubiquitin activating enzyme, UAE (UBA1), an essential cellular enzyme responsible for activating > 99% of all cellular ubiquitin. Ubiquitin is involved in multiple cellular processes including ubiquitin-dependent protein turnover, cell cycle progression, regulation of apoptosis, protein localization and response to DNA damage. Experiments combining targeted siRNA knockdown with TAK-243 identified DNA damage repair genes necessary for UAE inhibitor-induced cell death. A more focused approach revealed TAK-243 treatment blocked essential monoubiquitination events within the Translesion synthesis (TLS), Fanconi Anemia (FA) and Homologous recombination (HR) pathways. Inhibition of UAE prevented mono-ubiquitin signaling of key mediators within these pathways, including PCNA and FANCD2, by blocking formation of their specific E2-ubiquitin thioesters. In vitro cell-based assays combining TAK-243 with ultraviolet (UV) and radiation, both known to induce DNA damage, yielded inhibition of cell growth and enhanced DNA damage as observed through colony formation assays and Comet assay detection, respectively. Xenograft tumor bearing mice were treated with carboplatin or docetaxel, combined with TAK-243, to evaluate combination benefits in vivo. Synergistic and additive anti-tumor combination benefits were observed in animals treated with TAK-243 + carboplatin and TAK-243 + docetaxel. These important mechanistic in vitro and in vivo studies indicate the dependency of ubiquitination signaling in DNA damage repair and provide a mechanistic rationale for combining radiation, carboplatin or docetaxel with TAK-243 in the clinical setting. Currently, TAK-243 is being evaluated in a solid tumor phase I clinical trial evaluating safety, tolerability, pharmacokinetics, pharmacodynamics and anti-tumor activity (ClinicalTrials.gov identifier: NCT02045095). Citation Format: Michael A. Milhollen, Judi Shi, Tary Traore, Jessica Huck, Darshan Sappal, Jennifer Duffy, Eric Lightcap, Yuko Ishii, Jeff Ciavarri, Paul Fleming, Neil Bence, Marc L. Hyer. The small molecule UAE inhibitor TAK-243 (MLN7243) prevents DNA damage repair and reduces cell viability/tumor growth when combined with radiation, carboplatin and docetaxel. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr A164.
- Published
- 2015
- Full Text
- View/download PDF
26. Optimization of a series of dipeptides with a P3 β-neopentyl asparagine residue as non-covalent inhibitors of the chymotrypsin-like activity of human 20S proteasome
- Author
-
Nancy Bump, Cindy Q. Xia, Lawrence R. Dick, Paul Hales, Frank J. Bruzzese, Cynthia Barrett, Darshan S. Sappal, Kenneth M. Gigstad, Khristofer Garcia, Jane X. Liu, Christopher Blackburn, Jonathan L. Blank, Paul E. Fleming, Masayuki Nagayoshi, Michael D. Sintchak, Christopher Tsu, Matthew Jones, and Xiansi Zhou
- Subjects
Pharmacology ,Chemistry ,Stereochemistry ,Organic Chemistry ,Pharmaceutical Science ,Biochemistry ,20s proteasome ,Yeast ,In vitro ,Residue (chemistry) ,chemistry.chemical_compound ,Proteasome ,Covalent bond ,Drug Discovery ,Molecular Medicine ,Asparagine ,Boronic acid - Abstract
Inhibition of the proteasome by covalent inhibitors is a clinically proven anti-cancer therapy. We report here that dipeptides with a P3 neopentyl Asn residue are potent, reversible, non-covalent inhibitors selective for the chymotryptic activity of the 20S proteasome in vitro and in cells. The X-ray structure of compound 20 in complex with yeast 20S reveals the importance of hydrophobic bonding interactions of the neopentyl group within the S3 binding pocket of the 20S β5 sub-unit. Four compounds show comparable potencies to boronic acid inhibitors in a panel of assays.
- Published
- 2012
- Full Text
- View/download PDF
27. Abstract C82: Identification and preclinical characterization of inhibitors of the ubiquitin-activating enzymes UBA1 and UBA6
- Author
-
Paul E. Fleming, James M. Gavin, Yu Yang, Marc Hyer, Petter Veiby, Michael Milhollen, Peter G. Smith, Tary Traore, Sai M. Pulukuri, Jeff Ciavarri, Mark Manfredi, Darshan S. Sappal, Neil Bence, James E. Brownell, Kara Hoar, Jessica Huck, Christopher F. Claiborne, Lawrence Dick, and Derek Liqiang Tou
- Subjects
Cancer Research ,biology ,DNA damage ,Ubiquitin-activating enzyme ,UBA1 ,Cell cycle ,Ubiquitin ligase ,Proliferating cell nuclear antigen ,Cell biology ,Oncology ,Biochemistry ,Ubiquitin ,Proteasome ,biology.protein - Abstract
Millennium Pharmaceuticals, Inc. is dedicated to the discovery and development of novel oncology therapeutics in the area of protein homeostasis. Here we report the identification and characterization of compounds that target the ubiquitin activating enzymes, UBA1 and UBA6. These compounds are mechanism based inhibitors that inactivate the ubiquitin E1 enzymes by forming a ubiquitin compound adduct that remains tightly associated with the E1 adenylate binding site. Treatment of cells with these inhibitors results in cellular effects consistent with known Uba1 biology including rapid loss of E2 ubiquitin thioesters, loss of total ubiquitin conjugates, and accumulation of many ubiquitin proteasome system substrates. Following prolonged treatment, cells primarily arrest in the G2 phase of the cell cycle and ultimately undergo apoptosis. Reflecting the extensive cellular roles of ubiquitin, the compounds also impact global protein turnover, ER stress and DNA damage repair. UBA1 inhibition impairs ubiquitination of PCNA and the Fanconia Anemia protein FANCD2 leading to defective repair of UV induced DNA damage. UBA1 inhibition impacts numerous biological pathways relevant to cancer, results in apoptosis in vitro and is capable of inhibiting tumor growth in mouse xenografts in vivo. These data implicate UBA1 as a target for the treatment of cancer. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2011 Nov 12-16; San Francisco, CA. Philadelphia (PA): AACR; Mol Cancer Ther 2011;10(11 Suppl):Abstract nr C82.
- Published
- 2011
- Full Text
- View/download PDF
28. Abstract A70: A genome-wide siRNA screen for modulators of cell death induced by the proteasome inhibitor bortezomib
- Author
-
Trupti Lingaraj, Darshan S. Sappal, Jie Yu, Claudia Rabino, Wei Chen, Jonathan L. Blank, Paul Hales, Jay P. Morgenstern, Eric S. Lightcap, Jesse Gray, Saurabh Menon, Anne L. Burkhardt, Patrick J. LeRoy, David M. Rappoli, Michael D. Pickard, Jane Liu, Denise L. Driscoll, James J. Spelman, Theodore Peters, Khristofer Garcia, John Ringeling, and Siquan Chen
- Subjects
Cancer Research ,Gene knockdown ,Programmed cell death ,Bortezomib ,Cancer ,Biology ,medicine.disease ,biology.organism_classification ,Cell biology ,HeLa ,Oncology ,Proteasome ,Cell culture ,Proteasome inhibitor ,medicine ,medicine.drug - Abstract
Multiple pathways have been proposed as the mechanism by which proteasome inhibition induces cell death. To clarify their relative importance, we performed a genome-wide siRNA screen to evaluate the genetic determinants that confer sensitivity of the HCT-116 colon cancer cell line to bortezomib (VELCADE®, PS-341). The screen identified 100 genes whose knock-down affects the lethality of bortezomib. From this list, the accumulation of the proteins ASF1B, Myc, ODC1, PMAIP1 (Noxa), BNIP3, Gadd45α, p-SMC1A, SREBF1, and p53 by proteasome inhibition was linked to the induction of cell death. Fifty-nine genes in the A375 melanoma cell line and 56 genes in the HeLa cervical cancer cell line showed similar interactions with bortezomib to those seen in HCT-116 and a subset of 39 genes were common to all three cell lines. Finally, knockdown of these 100 genes in HCT-116 cells similarly affected their responsiveness to a structurally diverse set of proteasome inhibitors. Our results suggest that proteasome inhibition promotes cell death primarily by dysregulating Myc and polyamines, interfering with protein translation, and disrupting essential DNA damage repair pathways, leading to programmed cell death. Citation Information: Mol Cancer Ther 2009;8(12 Suppl):A70.
- Published
- 2009
- Full Text
- View/download PDF
29. Characterization of a new series of non-covalent proteasome inhibitors with exquisite potency and selectivity for the 20S β5-subunit.
- Author
-
Christopher Blackburn, Kenneth M. Gigstad, Paul Hales, Khristofer Garcia, Matthew Jones, Frank J. Bruzzese, Cynthia Barrett, Jane X. Liu, Teresa A. Soucy, Darshan S. Sappal, Nancy Bump, Edward J. Olhava, Paul Fleming, Lawrence R. Dick, Christopher Tsu, Michael D. Sintchak, and Jonathan L. Blank
- Subjects
PROTEASE inhibitors ,PEPTIDES ,CANCER cells ,X-ray crystallography ,TUMOR necrosis factors ,B cells ,LYMPHOMAS - Abstract
The mammalian 26S proteasome is a 2500 kDa multi-catalytic complex involved in intracellular protein degradation. We describe the synthesis and properties of a novel series of non-covalent di-peptide inhibitors of the proteasome used on a capped tri-peptide that was first identified by high-throughput screening of a library of approx. 350000 compounds for inhibitors of the ubiquitin–proteasome system in cells. We show that these compounds are entirely selective for the β5 (chymotrypsin-like) site over the β1 (caspase-like) and β2 (trypsin-like) sites of the 20S core particle of the proteasome, and over a panel of less closely related proteases. Compound optimization, guided by X-ray crystallography of the liganded 20S core particle, confirmed their non-covalent binding mode and provided a structural basis for their enhanced in vitro and cellular potencies. We demonstrate that such compounds show low nanomolar IC50 values for the human 20S β5 site in vitro, and that pharmacological inhibition of this site in cells is sufficient to potently inhibit the degradation of a tetra-ubiquitin–luciferase reporter, activation of NFκB (nuclear factor κB) in response to TNF-α (tumour necrosis factor-α) and the proliferation of cancer cells. Finally, we identified capped di-peptides that show differential selectivity for the β5 site of the constitutively expressed proteasome and immunoproteasome in vitro and in B-cell lymphomas. Collectively, these studies describe the synthesis, activity and binding mode of a new series of non-covalent proteasome inhibitors with unprecedented potency and selectivity for the β5 site, and which can discriminate between the constitutive proteasome and immunoproteasome in vitro and in cells. [ABSTRACT FROM AUTHOR]
- Published
- 2010
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.