47 results on '"Gregory McAllister"'
Search Results
2. An iron-dependent metabolic vulnerability underlies VPS34-dependence in RKO cancer cells.
- Author
-
Marek J Kobylarz, Jonathan M Goodwin, Zhao B Kang, John W Annand, Sarah Hevi, Ellen O'Mahony, Gregory McAllister, John Reece-Hoyes, Qiong Wang, John Alford, Carsten Russ, Alicia Lindeman, Martin Beibel, Guglielmo Roma, Walter Carbone, Judith Knehr, Joseph Loureiro, Christophe Antczak, Dmitri Wiederschain, Leon O Murphy, Suchithra Menon, and Beat Nyfeler
- Subjects
Medicine ,Science - Abstract
VPS34 is a key regulator of endomembrane dynamics and cargo trafficking, and is essential in cultured cell lines and in mice. To better characterize the role of VPS34 in cell growth, we performed unbiased cell line profiling studies with the selective VPS34 inhibitor PIK-III and identified RKO as a VPS34-dependent cellular model. Pooled CRISPR screen in the presence of PIK-III revealed endolysosomal genes as genetic suppressors. Dissecting VPS34-dependent alterations with transcriptional profiling, we found the induction of hypoxia response and cholesterol biosynthesis as key signatures. Mechanistically, acute VPS34 inhibition enhanced lysosomal degradation of transferrin and low-density lipoprotein receptors leading to impaired iron and cholesterol uptake. Excess soluble iron, but not cholesterol, was sufficient to partially rescue the effects of VPS34 inhibition on mitochondrial respiration and cell growth, indicating that iron limitation is the primary driver of VPS34-dependency in RKO cells. Loss of RAB7A, an endolysosomal marker and top suppressor in our genetic screen, blocked transferrin receptor degradation, restored iron homeostasis and reversed the growth defect as well as metabolic alterations due to VPS34 inhibition. Altogether, our findings suggest that impaired iron mobilization via the VPS34-RAB7A axis drive VPS34-dependence in certain cancer cells.
- Published
- 2020
- Full Text
- View/download PDF
3. DRUG-seq for miniaturized high-throughput transcriptome profiling in drug discovery
- Author
-
Chaoyang Ye, Daniel J. Ho, Marilisa Neri, Chian Yang, Tripti Kulkarni, Ranjit Randhawa, Martin Henault, Nadezda Mostacci, Pierre Farmer, Steffen Renner, Robert Ihry, Leandra Mansur, Caroline Gubser Keller, Gregory McAllister, Marc Hild, Jeremy Jenkins, and Ajamete Kaykas
- Subjects
Science - Abstract
RNA-seq is a powerful tool to investigate how drugs affect the transcriptome but library construction can be costly. Here the authors introduce DRUG-seq, an automated platform for high-throughput transcriptome profiling.
- Published
- 2018
- Full Text
- View/download PDF
4. Autophagy-Independent Lysosomal Targeting Regulated by ULK1/2-FIP200 and ATG9
- Author
-
Jonathan M. Goodwin, William E. Dowdle, Rowena DeJesus, Zuncai Wang, Philip Bergman, Marek Kobylarz, Alicia Lindeman, Ramnik J. Xavier, Gregory McAllister, Beat Nyfeler, Gregory Hoffman, and Leon O. Murphy
- Subjects
NCOA4 ,ferritinophagy ,autophagy ,ULK1/2 ,TBK1 ,ALS ,TAX1BP1 ,ATG9A ,pooled CRISPR screen ,trafficking ,Biology (General) ,QH301-705.5 - Abstract
Iron is vital for many homeostatic processes, and its liberation from ferritin nanocages occurs in the lysosome. Studies indicate that ferritin and its binding partner nuclear receptor coactivator-4 (NCOA4) are targeted to lysosomes by a form of selective autophagy. By using genome-scale functional screening, we identify an alternative lysosomal transport pathway for ferritin that requires FIP200, ATG9A, VPS34, and TAX1BP1 but lacks involvement of the ATG8 lipidation machinery that constitutes classical macroautophagy. TAX1BP1 binds directly to NCOA4 and is required for lysosomal trafficking of ferritin under basal and iron-depleted conditions. Under basal conditions ULK1/2-FIP200 controls ferritin turnover, but its deletion leads to TAX1BP1-dependent activation of TBK1 that regulates redistribution of ATG9A to the Golgi enabling continued trafficking of ferritin. Cells expressing an amyotrophic lateral sclerosis (ALS)-associated TBK1 allele are incapable of degrading ferritin suggesting a molecular mechanism that explains the presence of iron deposits in patient brain biopsies.
- Published
- 2017
- Full Text
- View/download PDF
5. The Natural Product Cavinafungin Selectively Interferes with Zika and Dengue Virus Replication by Inhibition of the Host Signal Peptidase
- Author
-
David Estoppey, Chia Min Lee, Marco Janoschke, Boon Heng Lee, Kah Fei Wan, Hongping Dong, Philippe Mathys, Ireos Filipuzzi, Tim Schuhmann, Ralph Riedl, Thomas Aust, Olaf Galuba, Gregory McAllister, Carsten Russ, Martin Spiess, Tewis Bouwmeester, Ghislain M.C. Bonamy, and Dominic Hoepfner
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Flavivirus infections by Zika and dengue virus impose a significant global healthcare threat with no US Food and Drug Administration (FDA)-approved vaccination or specific antiviral treatment available. Here, we present the discovery of an anti-flaviviral natural product named cavinafungin. Cavinafungin is a potent and selectively active compound against Zika and all four dengue virus serotypes. Unbiased, genome-wide genomic profiling in human cells using a novel CRISPR/Cas9 protocol identified the endoplasmic-reticulum-localized signal peptidase as the efficacy target of cavinafungin. Orthogonal profiling in S. cerevisiae followed by the selection of resistant mutants pinpointed the catalytic subunit of the signal peptidase SEC11 as the evolutionary conserved target. Biochemical analysis confirmed a rapid block of signal sequence cleavage of both host and viral proteins by cavinafungin. This study provides an effective compound against the eukaryotic signal peptidase and independent confirmation of the recently identified critical role of the signal peptidase in the replicative cycle of flaviviruses. : Recent outbreaks and lack of effective treatments against dengue and Zika virus have caused public concerns. Estoppey et al. have identified cavinafungin as exerting potent and selective antiviral activity by targeting the signal-binding cleft of the catalytic subunit of the endoplasmic reticulum signal peptidase. Keywords: Zika virus, dengue virus, cavinafungin, signal peptidase, SEC11A, SEC11, CRISPR/Cas9, chemogenomic profiling
- Published
- 2017
- Full Text
- View/download PDF
6. Genome-Scale CRISPR Screens Identify Human Pluripotency-Specific Genes
- Author
-
Robert J. Ihry, Max R. Salick, Daniel J. Ho, Marie Sondey, Sravya Kommineni, Steven Paula, Joe Raymond, Beata Henry, Elizabeth Frias, Qiong Wang, Kathleen A. Worringer, Chaoyang Ye, Carsten Russ, John S. Reece-Hoyes, Robert C. Altshuler, Ranjit Randhawa, Zinger Yang, Gregory McAllister, Gregory R. Hoffman, Ricardo Dolmetsch, and Ajamete Kaykas
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Human pluripotent stem cells (hPSCs) generate a variety of disease-relevant cells that can be used to improve the translation of preclinical research. Despite the potential of hPSCs, their use for genetic screening has been limited by technical challenges. We developed a scalable and renewable Cas9 and sgRNA-hPSC library in which loss-of-function mutations can be induced at will. Our inducible mutant hPSC library can be used for multiple genome-wide CRISPR screens in a variety of hPSC-induced cell types. As proof of concept, we performed three screens for regulators of properties fundamental to hPSCs: their ability to self-renew and/or survive (fitness), their inability to survive as single-cell clones, and their capacity to differentiate. We identified the majority of known genes and pathways involved in these processes, as well as a plethora of genes with unidentified roles. This resource will increase the understanding of human development and genetics. This approach will be a powerful tool to identify disease-modifying genes and pathways. : Ihry et al. develop a CRISPR/Cas9 genetic screening platform for hPSCs that enables unbiased genome-scale genetic screening. The platform exhibits high performance and accurately detects the dropout of essential genes. Furthermore, proof-of-concept screens exploit hPSC-specific phenotypes to identify regulators of fitness, survival after single-cell dissociation, and pluripotency. Keywords: CRISPR genome-wide screening, human pluripotent stem cells, iPSC, hESC, PAWR, PMAIP1, DDR
- Published
- 2019
- Full Text
- View/download PDF
7. Functional CRISPR screening identifies the ufmylation pathway as a regulator of SQSTM1/p62
- Author
-
Rowena DeJesus, Francesca Moretti, Gregory McAllister, Zuncai Wang, Phil Bergman, Shanming Liu, Elizabeth Frias, John Alford, John S Reece-Hoyes, Alicia Lindeman, Jennifer Kelliher, Carsten Russ, Judith Knehr, Walter Carbone, Martin Beibel, Guglielmo Roma, Aylwin Ng, John A Tallarico, Jeffery A Porter, Ramnik J Xavier, Craig Mickanin, Leon O Murphy, Gregory R Hoffman, and Beat Nyfeler
- Subjects
autophagy ,SQSTM1 ,ER stress ,CRISPR ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
SQSTM1 is an adaptor protein that integrates multiple cellular signaling pathways and whose expression is tightly regulated at the transcriptional and post-translational level. Here, we describe a forward genetic screening paradigm exploiting CRISPR-mediated genome editing coupled to a cell selection step by FACS to identify regulators of SQSTM1. Through systematic comparison of pooled libraries, we show that CRISPR is superior to RNAi in identifying known SQSTM1 modulators. A genome-wide CRISPR screen exposed MTOR signalling and the entire macroautophagy machinery as key regulators of SQSTM1 and identified several novel modulators including HNRNPM, SLC39A14, SRRD, PGK1 and the ufmylation cascade. We show that ufmylation regulates SQSTM1 by eliciting a cell type-specific ER stress response which induces SQSTM1 expression and results in its accumulation in the cytosol. This study validates pooled CRISPR screening as a powerful method to map the repertoire of cellular pathways that regulate the fate of an individual target protein.
- Published
- 2016
- Full Text
- View/download PDF
8. Identification of elongation factor G as the conserved cellular target of argyrin B.
- Author
-
Beat Nyfeler, Dominic Hoepfner, Deborah Palestrant, Christina A Kirby, Lewis Whitehead, Robert Yu, Gejing Deng, Ruth E Caughlan, Angela L Woods, Adriana K Jones, S Whitney Barnes, John R Walker, Swann Gaulis, Ervan Hauy, Saskia M Brachmann, Philipp Krastel, Christian Studer, Ralph Riedl, David Estoppey, Thomas Aust, N Rao Movva, Zuncai Wang, Michael Salcius, Gregory A Michaud, Gregory McAllister, Leon O Murphy, John A Tallarico, Christopher J Wilson, and Charles R Dean
- Subjects
Medicine ,Science - Abstract
Argyrins, produced by myxobacteria and actinomycetes, are cyclic octapeptides with antibacterial and antitumor activity. Here, we identify elongation factor G (EF-G) as the cellular target of argyrin B in bacteria, via resistant mutant selection and whole genome sequencing, biophysical binding studies and crystallography. Argyrin B binds a novel allosteric pocket in EF-G, distinct from the known EF-G inhibitor antibiotic fusidic acid, revealing a new mode of protein synthesis inhibition. In eukaryotic cells, argyrin B was found to target mitochondrial elongation factor G1 (EF-G1), the closest homologue of bacterial EF-G. By blocking mitochondrial translation, argyrin B depletes electron transport components and inhibits the growth of yeast and tumor cells. Further supporting direct inhibition of EF-G1, expression of an argyrin B-binding deficient EF-G1 L693Q variant partially rescued argyrin B-sensitivity in tumor cells. In summary, we show that argyrin B is an antibacterial and cytotoxic agent that inhibits the evolutionarily conserved target EF-G, blocking protein synthesis in bacteria and mitochondrial translation in yeast and mammalian cells.
- Published
- 2012
- Full Text
- View/download PDF
9. Supplementary Table S2 from CRISPR Screens Provide a Comprehensive Assessment of Cancer Vulnerabilities but Generate False-Positive Hits for Highly Amplified Genomic Regions
- Author
-
Michael R. Schlabach, Frank Stegmeier, Tobias Schmelzle, William R. Sellers, Francesco Hofmann, Nicholas Keen, E. Robert McDonald, Audrey Kauffmann, Odile Weber, Rosalie de Beaumont, Daniel Rakiec, Sarah Y. Neshat, Matthew D. Shirley, Jessica Wan, Dorothee Abramowski, Antoine DeWeck, Gregory McAllister, Kristine Yu, David A. Ruddy, Javad Golji, Michael D. Jones, Joshua M. Korn, Eric Billy, Li Li, Pamela J. Cassiani, and Diana M. Munoz
- Abstract
Annotations for the sgRNA libraries containing sequences, target gene information, Zscore and RNAseq data for each cell line screened.
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Figure Legends, Table Legends from CRISPR Screens Provide a Comprehensive Assessment of Cancer Vulnerabilities but Generate False-Positive Hits for Highly Amplified Genomic Regions
- Author
-
Michael R. Schlabach, Frank Stegmeier, Tobias Schmelzle, William R. Sellers, Francesco Hofmann, Nicholas Keen, E. Robert McDonald, Audrey Kauffmann, Odile Weber, Rosalie de Beaumont, Daniel Rakiec, Sarah Y. Neshat, Matthew D. Shirley, Jessica Wan, Dorothee Abramowski, Antoine DeWeck, Gregory McAllister, Kristine Yu, David A. Ruddy, Javad Golji, Michael D. Jones, Joshua M. Korn, Eric Billy, Li Li, Pamela J. Cassiani, and Diana M. Munoz
- Abstract
Supplementary Figure Legends, Table Legends
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Figures S1 - S11 from CRISPR Screens Provide a Comprehensive Assessment of Cancer Vulnerabilities but Generate False-Positive Hits for Highly Amplified Genomic Regions
- Author
-
Michael R. Schlabach, Frank Stegmeier, Tobias Schmelzle, William R. Sellers, Francesco Hofmann, Nicholas Keen, E. Robert McDonald, Audrey Kauffmann, Odile Weber, Rosalie de Beaumont, Daniel Rakiec, Sarah Y. Neshat, Matthew D. Shirley, Jessica Wan, Dorothee Abramowski, Antoine DeWeck, Gregory McAllister, Kristine Yu, David A. Ruddy, Javad Golji, Michael D. Jones, Joshua M. Korn, Eric Billy, Li Li, Pamela J. Cassiani, and Diana M. Munoz
- Abstract
Supplementary Figure S1. Comparison of drop out phenotypes in MKN45, RKO, HT1080 highlighting selected pan-lethal genes. Supplementary Figure S2. The genes that scored as lethal by both RNAi and CRISPR were strongly enriched for known essential genes classes. Supplementary Figure S3. To identify likely off-target hits the lethality scores of non-expressed genes were examined, as they are expected not to be required for cell viability. Supplementary Figure S4. shRNAs directed towards CDK9 do not show robust protein depletion. Supplementary Figure S5. Additional methods measuring the proliferation effects of individual sgRNA/shRNAs to validate the impact that targeting selected genetic dependencies have on cell viability. Supplementary Figure S6. Correlation analysis displaying features that correlated most significantly with sgRNA potency. Supplementary Figure S7. Effect of relative position within a gene on sgRNA viability effects. Supplementary Figure S8. Non-scoring sgRNA in conserved Pfam domains have a reduced editing efficiency compared to guides with strong viability effects. Supplementary Figure S9. Multiple genomic cuts result in DNA damage induced G2/M cell cycle arrest. Supplementary Figure S10. Multiple genomic cuts lead to an increase in cell death. Supplementary Figure S11. Pie chart demonstrating that the overall contribution of copy number effects in determining essential genes in aneuploid lines is relatively minor.
- Published
- 2023
- Full Text
- View/download PDF
12. Data from Inhibition of Casein Kinase 1 Alpha Prevents Acquired Drug Resistance to Erlotinib in EGFR-Mutant Non–Small Cell Lung Cancer
- Author
-
Sreenath V. Sharma, Matthew J. Meyer, Frank Stegmeier, Gregory McAllister, Joshua Korn, Daniel Rakiec, David Ruddy, Elizabeth Frias, Greg Hoffman, Kaitlin McCutcheon, Dongshu Chen, and Alexandra B. Lantermann
- Abstract
Patients with lung tumors harboring activating mutations in the EGF receptor (EGFR) show good initial treatment responses to the EGFR tyrosine kinase inhibitors (TKI) erlotinib or gefitinib. However, acquired resistance invariably develops. Applying a focused shRNA screening approach to identify genes whose knockdown can prevent and/or overcome acquired resistance to erlotinib in several EGFR-mutant non–small cell lung cancer (NSCLC) cell lines, we identified casein kinase 1 α (CSNK1A1, CK1α). We found that CK1α suppression inhibits the NF-κB prosurvival signaling pathway. Furthermore, downregulation of NF-κB signaling by approaches independent of CK1α knockdown can also attenuate acquired erlotinib resistance, supporting a role for activated NF-κB signaling in conferring acquired drug resistance. Importantly, CK1α suppression prevented erlotinib resistance in an HCC827 xenograft model in vivo. Our findings suggest that patients with EGFR-mutant NSCLC might benefit from a combination of EGFR TKIs and CK1α inhibition to prevent acquired drug resistance and to prolong disease-free survival. Cancer Res; 75(22); 4937–48. ©2015 AACR.
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Figures from Inhibition of Casein Kinase 1 Alpha Prevents Acquired Drug Resistance to Erlotinib in EGFR-Mutant Non–Small Cell Lung Cancer
- Author
-
Sreenath V. Sharma, Matthew J. Meyer, Frank Stegmeier, Gregory McAllister, Joshua Korn, Daniel Rakiec, David Ruddy, Elizabeth Frias, Greg Hoffman, Kaitlin McCutcheon, Dongshu Chen, and Alexandra B. Lantermann
- Abstract
Fig. S1. Summary of EGFR status and sensitivity to erlotinib of NSCLC cell lines used in this study. Fig. S2. Knockdown of CK1α inhibits proliferation of PC9 cells more than proliferation of HCC827 or HCC4006 cells under DMSO treatment. Fig. S3. Performance of individual shRNAs against members of the CK1-family in shRNA screens. Fig. S4. Knockdown of CK1α does not sensitize NSCLC cells to doxorubicin or cisplatin. Fig. S5. Suppression of CK1α attenuates acquired resistance to erlotinib in EGFR-mutant NSCLC cells. Fig. S6. Suppression of CK1α attenuates the outgrowth of drug tolerant persisters (DTPs) to resistant clones and inhibits proliferation of resistant PC9 cells in the presence of erlotinib. Fig. S7. The CK1 inhibitor D4476 can attenuate acquired resistance to erlotinib in NSCLC cell lines. Fig. S8: Knockdown of CK1α does not influence EGFR signaling. Fig. S9: Knockdown of CK1α does not activate WNT-signaling. Fig. S10: Erlotinib treatment activates NF-κB-signaling and CK1α-knockdown decreases expression of NF-κB-target genes. Fig. S11: Inhibition of NF-κB-signaling attenuates resistance to erlotinib while only slightly affecting proliferation in the absence of erlotinib.
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Figure Legends from Inhibition of Casein Kinase 1 Alpha Prevents Acquired Drug Resistance to Erlotinib in EGFR-Mutant Non–Small Cell Lung Cancer
- Author
-
Sreenath V. Sharma, Matthew J. Meyer, Frank Stegmeier, Gregory McAllister, Joshua Korn, Daniel Rakiec, David Ruddy, Elizabeth Frias, Greg Hoffman, Kaitlin McCutcheon, Dongshu Chen, and Alexandra B. Lantermann
- Abstract
Supplementary Figure Legends 1-11 Lantermann et al.
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Table 1 from Inhibition of Casein Kinase 1 Alpha Prevents Acquired Drug Resistance to Erlotinib in EGFR-Mutant Non–Small Cell Lung Cancer
- Author
-
Sreenath V. Sharma, Matthew J. Meyer, Frank Stegmeier, Gregory McAllister, Joshua Korn, Daniel Rakiec, David Ruddy, Elizabeth Frias, Greg Hoffman, Kaitlin McCutcheon, Dongshu Chen, and Alexandra B. Lantermann
- Abstract
Supplementary Table 1: Hit list shRNA screen
- Published
- 2023
- Full Text
- View/download PDF
16. Genome-wide CRISPR screen identifies protein pathways modulating tau protein levels in neurons
- Author
-
Fiona Elwood, John Alford, Carsten Russ, Christopher Acker, Audrey Gray, John S. Reece-Hoyes, Carlos G Sanchez, Sarah J. Luchansky, Christian Doherty, Lucas Craig, Gregory McAllister, Steven Paula, Ricardo E. Dolmetsch, Shaojian An, Cheng Song, Alicia Lindeman, Malini Varadarajan, Nadire Cochran, Manuela Polydoro, Ketthsy Capre, and Gregory R. Hoffman
- Subjects
CRISPR-Cas9 genome editing ,0301 basic medicine ,Candidate gene ,QH301-705.5 ,Tau protein ,Medicine (miscellaneous) ,tau Proteins ,Molecular neuroscience ,Article ,General Biochemistry, Genetics and Molecular Biology ,Progressive supranuclear palsy ,Mice ,Neuroblastoma ,03 medical and health sciences ,0302 clinical medicine ,Ubiquitin ,CRISPR-Associated Protein 9 ,Cell Line, Tumor ,mental disorders ,medicine ,Animals ,Humans ,CRISPR ,Genetic Testing ,Biology (General) ,PI3K/AKT/mTOR pathway ,Gene Editing ,Neurons ,biology ,TOR Serine-Threonine Kinases ,Alzheimer's disease ,medicine.disease ,Cellular neuroscience ,Rats ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Genes ,biology.protein ,Neddylation ,TSC1 ,CRISPR-Cas Systems ,General Agricultural and Biological Sciences ,Metabolic Networks and Pathways ,030217 neurology & neurosurgery ,Genome-Wide Association Study - Abstract
Aggregates of hyperphosphorylated tau protein are a pathological hallmark of more than 20 distinct neurodegenerative diseases, including Alzheimer’s disease, progressive supranuclear palsy, and frontotemporal dementia. While the exact mechanism of tau aggregation is unknown, the accumulation of aggregates correlates with disease progression. Here we report a genome-wide CRISPR screen to identify modulators of endogenous tau protein for the first time. Primary screens performed in SH-SY5Y cells, identified positive and negative regulators of tau protein levels. Hit validation of the top 43 candidate genes was performed using Ngn2-induced human cortical excitatory neurons. Using this approach, genes and pathways involved in modulation of endogenous tau levels were identified, including chromatin modifying enzymes, neddylation and ubiquitin pathway members, and components of the mTOR pathway. TSC1, a critical component of the mTOR pathway, was further validated in vivo, demonstrating the relevance of this screening strategy. These findings may have implications for treating neurodegenerative diseases in the future., Using an unbiased genome-wide CRISPR screen approach, Sanchez et al. identified modulators of endogenous tau protein. This study suggests that chromatin modifiers, neddylation, ubiquitination, and the mTOR pathways regulate overall levels of tau protein in neurons, which could help in future identification of therapeutics for neurodegenerative diseases.
- Published
- 2021
17. Autophagy-Independent Lysosomal Targeting Regulated by ULK1/2-FIP200 and ATG9
- Author
-
Marek J. Kobylarz, Leon Murphy, William E. Dowdle, Rowena DeJesus, Jonathan M. Goodwin, Alicia Lindeman, Beat Nyfeler, Gregory McAllister, Zuncai Wang, Gregory R. Hoffman, Ramnik J. Xavier, and Philip Bergman
- Subjects
0301 basic medicine ,Lysosomal transport ,autophagy ,DNA, Complementary ,TAX1BP1 ,TBK1 ,ATG8 ,Vesicular Transport Proteins ,Autophagy-Related Proteins ,Protein Serine-Threonine Kinases ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,symbols.namesake ,trafficking ,Cell Line, Tumor ,Lysosome ,medicine ,Autophagy-Related Protein-1 Homolog ,Humans ,ATG9A ,lcsh:QH301-705.5 ,ferritinophagy ,biology ,Autophagy ,Intracellular Signaling Peptides and Proteins ,Membrane Proteins ,Protein-Tyrosine Kinases ,Golgi apparatus ,ULK1 ,pooled CRISPR screen ,Neoplasm Proteins ,Cell biology ,Ferritin ,HEK293 Cells ,030104 developmental biology ,medicine.anatomical_structure ,lcsh:Biology (General) ,Biochemistry ,Nuclear receptor ,Ferritins ,biology.protein ,symbols ,ALS ,Lysosomes ,NCOA4 ,ULK1/2 - Abstract
Iron is vital for many homeostatic processes and its liberation from ferritin nanocages occurs in the lysosome. Studies indicate that ferritin and its binding partner nuclear receptor coactivator-4 (NCOA4) are targeted to lysosomes by a form of selective autophagy. By using genome-scale functional screening we identify an alternative lysosomal transport pathway for ferritin that requires FIP200, ATG9A, VPS34 and TAX1BP1 but lacks involvement of the ATG8 lipidation machinery that constitutes classical macroautophagy. TAX1BP1 binds directly to NCOA4 and is required for lysosomal trafficking of ferritin under basal and iron depleted conditions. Under basal conditions ULK1/2-FIP200 controls ferritin turnover but its deletion leads to TAX1BP1-dependent activation of TBK1 which regulates redistribution of ATG9A to the Golgi enabling continued trafficking of ferritin. Cells expressing an Amyotrophic Lateral Sclerosis (ALS)-associated TBK1 allele are incapable of degrading ferritin suggesting a novel molecular mechanism that explains the presence of iron deposits in patient brain biopsies.
- Published
- 2017
- Full Text
- View/download PDF
18. The Natural Product Cavinafungin Selectively Interferes with Zika and Dengue Virus Replication by Inhibition of the Host Signal Peptidase
- Author
-
Chia Min Lee, Kah Fei Wan, Ralph Riedl, Marco Janoschke, Ghislain M. C. Bonamy, Tewis Bouwmeester, Olaf Galuba, Hongping Dong, Philippe Mathys, Tim Schuhmann, David Estoppey, Martin Spiess, Carsten Russ, Dominic Hoepfner, Ireos Filipuzzi, Boon Heng Lee, Gregory McAllister, and Thomas Aust
- Subjects
0301 basic medicine ,Signal peptide ,Protein subunit ,030106 microbiology ,Saccharomyces cerevisiae ,Dengue virus ,medicine.disease_cause ,Virus Replication ,General Biochemistry, Genetics and Molecular Biology ,Zika virus ,03 medical and health sciences ,Lipopeptides ,Viral Proteins ,medicine ,CRISPR ,Humans ,Flavivirus Infections ,lcsh:QH301-705.5 ,Signal peptidase ,Biological Products ,biology ,Cas9 ,Genome, Human ,Serine Endopeptidases ,Membrane Proteins ,Genomics ,Zika Virus ,Dengue Virus ,biology.organism_classification ,HCT116 Cells ,Virology ,Protein Subunits ,030104 developmental biology ,lcsh:Biology (General) ,Gene Knockdown Techniques ,CRISPR-Cas Systems - Abstract
Summary: Flavivirus infections by Zika and dengue virus impose a significant global healthcare threat with no US Food and Drug Administration (FDA)-approved vaccination or specific antiviral treatment available. Here, we present the discovery of an anti-flaviviral natural product named cavinafungin. Cavinafungin is a potent and selectively active compound against Zika and all four dengue virus serotypes. Unbiased, genome-wide genomic profiling in human cells using a novel CRISPR/Cas9 protocol identified the endoplasmic-reticulum-localized signal peptidase as the efficacy target of cavinafungin. Orthogonal profiling in S. cerevisiae followed by the selection of resistant mutants pinpointed the catalytic subunit of the signal peptidase SEC11 as the evolutionary conserved target. Biochemical analysis confirmed a rapid block of signal sequence cleavage of both host and viral proteins by cavinafungin. This study provides an effective compound against the eukaryotic signal peptidase and independent confirmation of the recently identified critical role of the signal peptidase in the replicative cycle of flaviviruses. : Recent outbreaks and lack of effective treatments against dengue and Zika virus have caused public concerns. Estoppey et al. have identified cavinafungin as exerting potent and selective antiviral activity by targeting the signal-binding cleft of the catalytic subunit of the endoplasmic reticulum signal peptidase. Keywords: Zika virus, dengue virus, cavinafungin, signal peptidase, SEC11A, SEC11, CRISPR/Cas9, chemogenomic profiling
- Published
- 2017
19. Determination of minimal transcriptional signatures of compounds for target prediction.
- Author
-
Florian Nigsch, Janna Hutz, Ben Cornett, Douglas W. Selinger, Gregory McAllister, Somnath Bandyopadhyay, Joseph Loureiro, and Jeremy L. Jenkins
- Published
- 2012
- Full Text
- View/download PDF
20. CRISPR Screens Provide a Comprehensive Assessment of Cancer Vulnerabilities but Generate False-Positive Hits for Highly Amplified Genomic Regions
- Author
-
Pamela J. Cassiani, Odile Weber, E. Robert McDonald, Li Li, Sarah Y. Neshat, Diana M Munoz, Matthew D. Shirley, Javad Golji, William R. Sellers, Nicholas Keen, Rosalie de Beaumont, Joshua M. Korn, Eric Billy, Dorothee Abramowski, Kristine Yu, Gregory McAllister, Michael D. Jones, Audrey Kauffmann, David A. Ruddy, Jessica Wan, Daniel P. Rakiec, Frank Stegmeier, Francesco Hofmann, Tobias Schmelzle, Antoine deWeck, and Michael R. Schlabach
- Subjects
0301 basic medicine ,Genetics ,Tiling array ,Cas9 ,Biology ,Genome ,03 medical and health sciences ,030104 developmental biology ,Oncology ,RNA interference ,Coding region ,Lethal allele ,CRISPR ,Gene - Abstract
CRISPR/Cas9 has emerged as a powerful new tool to systematically probe gene function. We compared the performance of CRISPR to RNAi-based loss-of-function screens for the identification of cancer dependencies across multiple cancer cell lines. CRISPR dropout screens consistently identified more lethal genes than RNAi, implying that the identification of many cellular dependencies may require full gene inactivation. However, in two aneuploid cancer models, we found that all genes within highly amplified regions, including nonexpressed genes, scored as lethal by CRISPR, revealing an unanticipated class of false-positive hits. In addition, using a CRISPR tiling screen, we found that sgRNAs targeting essential domains generate the strongest lethality phenotypes and thus provide a strategy to rapidly define the protein domains required for cancer dependence. Collectively, these findings not only demonstrate the utility of CRISPR screens in the identification of cancer-essential genes, but also reveal the need to carefully control for false-positive results in chromosomally unstable cancer lines. Significance: We show in this study that CRISPR-based screens have a significantly lower false-negative rate compared with RNAi-based screens, but have specific liabilities particularly in the interrogation of regions of genome amplification. Therefore, this study provides critical insights for applying CRISPR-based screens toward the systematic identification of new cancer targets. Cancer Discov; 6(8); 900–13. ©2016 AACR. See related commentary by Sheel and Xue, p. 824. See related article by Aguirre et al., p. 914. This article is highlighted in the In This Issue feature, p. 803
- Published
- 2016
- Full Text
- View/download PDF
21. Tankyrase Inhibitor Sensitizes Lung Cancer Cells to Endothelial Growth Factor Receptor (EGFR) Inhibition via Stabilizing Angiomotins and Inhibiting YAP Signaling
- Author
-
Gregory McAllister, Zinger Yang, Carsten Russ, Johnny Castillo, Greg Hoffman, Alicia Lindeman, Feng Cong, John S. Reece-Hoyes, Bo Lu, John A. Tallarico, Wenqing Xu, Markus Schirle, Yue Zhang, and Hui Wang
- Subjects
0301 basic medicine ,Lung Neoplasms ,Ubiquitin-Protein Ligases ,Down-Regulation ,Antineoplastic Agents ,Protein degradation ,Biochemistry ,Erlotinib Hydrochloride ,Gene Knockout Techniques ,03 medical and health sciences ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,medicine ,Humans ,Protein Interaction Domains and Motifs ,RNA, Small Interfering ,Molecular Biology ,Tissue homeostasis ,Adaptor Proteins, Signal Transducing ,EGFR inhibitors ,Tankyrases ,biology ,Protein Stability ,Microfilament Proteins ,Wnt signaling pathway ,Membrane Proteins ,YAP-Signaling Proteins ,Cell Biology ,Phosphoproteins ,Angiomotin ,Ubiquitin ligase ,Cell biology ,ErbB Receptors ,HEK293 Cells ,030104 developmental biology ,Angiomotins ,030220 oncology & carcinogenesis ,biology.protein ,Intercellular Signaling Peptides and Proteins ,Erlotinib ,CRISPR-Cas Systems ,Signal transduction ,Signal Transduction ,Transcription Factors ,medicine.drug - Abstract
YAP signaling pathway plays critical roles in tissue homeostasis, and aberrant activation of YAP signaling has been implicated in cancers. To identify tractable targets of YAP pathway, we have performed a pathway-based pooled CRISPR screen and identified tankyrase and its associated E3 ligase RNF146 as positive regulators of YAP signaling. Genetic ablation or pharmacological inhibition of tankyrase prominently suppresses YAP activity and YAP target gene expression. Using a proteomic approach, we have identified angiomotin family proteins, which are known negative regulators of YAP signaling, as novel tankyrase substrates. Inhibition of tankyrase or depletion of RNF146 stabilizes angiomotins. Angiomotins physically interact with tankyrase through a highly conserved motif at their N terminus, and mutation of this motif leads to their stabilization. Tankyrase inhibitor-induced stabilization of angiomotins reduces YAP nuclear translocation and decreases downstream YAP signaling. We have further shown that knock-out of YAP sensitizes non-small cell lung cancer to EGFR inhibitor Erlotinib. Tankyrase inhibitor, but not porcupine inhibitor, which blocks Wnt secretion, enhances growth inhibitory activity of Erlotinib. This activity is mediated by YAP inhibition and not Wnt/β-catenin inhibition. Our data suggest that tankyrase inhibition could serve as a novel strategy to suppress YAP signaling for combinatorial targeted therapy.
- Published
- 2016
- Full Text
- View/download PDF
22. CRISPR/Cas9-Based Chemogenomic Profiling in Mammalian Cells
- Author
-
Dominic, Hoepfner, Gregory, McAllister, and Gregory R, Hoffman
- Subjects
Gene Editing ,Dose-Response Relationship, Drug ,Cell Survival ,Gene Expression Profiling ,Genetic Vectors ,Lentivirus ,Genomics ,Haploinsufficiency ,Cell Line ,Gene Expression Regulation ,Animals ,Humans ,CRISPR-Cas Systems ,RNA, Guide, Kinetoplastida - Abstract
Chemogenomic profiling is a powerful and unbiased approach to elucidate pharmacological targets and the mechanism of bioactive compounds. It is based on identifying cellular hypersensitivity and resistance caused by individual gene modulations with genome-wide coverage. Due to the requirement of bar-coded, genome-wide deletion collections, high-resolution experiments of this nature have historically been limited to fungal systems. Pooled RNAi reagents have enabled similar attempts in mammalian cells but efforts have been hampered by significant off-target effects and experimental noise. The CRISPR/Cas9 system for the first time enables precise DNA editing at defined loci in a genome-wide fashion. Here we present the detailed protocol that leverages the CRISPR/Cas9 system for chemogenomic profiling and target identification of diverse chemical probes.
- Published
- 2018
23. CRISPR/Cas9-Based Chemogenomic Profiling in Mammalian Cells
- Author
-
Gregory McAllister, Dominic Hoepfner, and Gregory R. Hoffman
- Subjects
0301 basic medicine ,Individual gene ,010405 organic chemistry ,Cas9 ,Genomics ,Computational biology ,Biology ,01 natural sciences ,0104 chemical sciences ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,chemistry ,Genome editing ,RNA interference ,Profiling (information science) ,CRISPR ,DNA - Abstract
Chemogenomic profiling is a powerful and unbiased approach to elucidate pharmacological targets and the mechanism of bioactive compounds. It is based on identifying cellular hypersensitivity and resistance caused by individual gene modulations with genome-wide coverage. Due to the requirement of bar-coded, genome-wide deletion collections, high-resolution experiments of this nature have historically been limited to fungal systems. Pooled RNAi reagents have enabled similar attempts in mammalian cells but efforts have been hampered by significant off-target effects and experimental noise. The CRISPR/Cas9 system for the first time enables precise DNA editing at defined loci in a genome-wide fashion. Here we present the detailed protocol that leverages the CRISPR/Cas9 system for chemogenomic profiling and target identification of diverse chemical probes.
- Published
- 2018
- Full Text
- View/download PDF
24. DRUG-seq for miniaturized high-throughput transcriptome profiling in drug discovery
- Author
-
Robert J. Ihry, Nadezda Mostacci, Gregory McAllister, Martin Henault, Caroline Gubser Keller, Jeremy L. Jenkins, Steffen Renner, Daniel J. Ho, Chaoyang Ye, Ajamete Kaykas, Marilisa Neri, Chian Yang, Leandra Mansur, Marc Hild, Tripti Kulkarni, Pierre Farmer, and Ranjit Randhawa
- Subjects
0301 basic medicine ,Drug ,Computer science ,Science ,High-throughput screening ,media_common.quotation_subject ,genetic processes ,General Physics and Astronomy ,Computational biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Transcriptome ,03 medical and health sciences ,Drug Discovery ,Humans ,Transcriptome profiling ,CRISPR ,Profiling (information science) ,Clustered Regularly Interspaced Short Palindromic Repeats ,natural sciences ,lcsh:Science ,media_common ,Multidisciplinary ,Sequence Analysis, RNA ,Drug discovery ,Gene Expression Profiling ,food and beverages ,General Chemistry ,High-Throughput Screening Assays ,030104 developmental biology ,lcsh:Q - Abstract
Here we report Digital RNA with pertUrbation of Genes (DRUG-seq), a high-throughput platform for drug discovery. Pharmaceutical discovery relies on high-throughput screening, yet current platforms have limited readouts. RNA-seq is a powerful tool to investigate drug effects using transcriptome changes as a proxy, yet standard library construction is costly. DRUG-seq captures transcriptional changes detected in standard RNA-seq at 1/100th the cost. In proof-of-concept experiments profiling 433 compounds across 8 doses, transcription profiles generated from DRUG-seq successfully grouped compounds into functional clusters by mechanism of actions (MoAs) based on their intended targets. Perturbation differences reflected in transcriptome changes were detected for compounds engaging the same target, demonstrating the value of using DRUG-seq for understanding on and off-target activities. We demonstrate DRUG-seq captures common mechanisms, as well as differences between compound treatment and CRISPR on the same target. DRUG-seq provides a powerful tool for comprehensive transcriptome readout in a high-throughput screening environment., RNA-seq is a powerful tool to investigate how drugs affect the transcriptome but library construction can be costly. Here the authors introduce DRUG-seq, an automated platform for high-throughput transcriptome profiling.
- Published
- 2018
- Full Text
- View/download PDF
25. TRPS1 Is a Lineage-Specific Transcriptional Dependency in Breast Cancer
- Author
-
Anne Trinh, David Pellman, Elizabeth Frias, Nadire Ramadan, Michael D. Hogarty, Zainab Jagani, William R. Sellers, Muhammad B. Ekram, Henry W. Long, Kimberly Stegmaier, Antoine deWeck, Kristine Yu, Robert M. Witwicki, Tomas Rejtar, Ramesh A. Shivdasani, Serena J. Silver, Gregory McAllister, Javad Golji, Charles W. M. Roberts, Shaokun Shu, Ho Man Chan, Michael R. Schlabach, Myles Brown, Greg Hoffman, Robert E McDonald, David A. Ruddy, Sosathya Sovath, Xintao Qiu, Yingtian Xie, Kornelia Polyak, Michalina Janiszewska, Mijung Kwon, and Nicholas Keen
- Subjects
0301 basic medicine ,Transcription, Genetic ,Cell Survival ,Breast Neoplasms ,Triple Negative Breast Neoplasms ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Humans ,Cell Lineage ,RNA, Small Interfering ,Transcription factor ,Epigenomics ,biology ,Chromatin binding ,Cancer ,medicine.disease ,Mi-2/NuRD complex ,3. Good health ,Chromatin ,Cell biology ,DNA-Binding Proteins ,Repressor Proteins ,030104 developmental biology ,Histone ,HEK293 Cells ,biology.protein ,Female ,Carcinogenesis ,Mi-2 Nucleosome Remodeling and Deacetylase Complex ,Protein Binding ,Transcription Factors - Abstract
SUMMARY Perturbed epigenomic programs play key roles in tumorigenesis, and chromatin modulators are candidate therapeutic targets in various human cancer types. To define singular and shared dependencies on DNA and histone modifiers and transcription factors in poorly differentiated adult and pediatric cancers, we conducted a targeted shRNA screen across 59 cell lines of 6 cancer types. Here, we describe the TRPS1 transcription factor as a strong breast cancer-specific hit, owing largely to lineage-restricted expression. Knockdown of TRPS1 resulted in perturbed mitosis, apoptosis, and reduced tumor growth. Integrated analysis of TRPS1 transcriptional targets, chromatin binding, and protein interactions revealed that TRPS1 is associated with the NuRD repressor complex. These findings uncover a transcriptional network that is essential for breast cancer cell survival and propagation., Graphical Abstract, In Brief Witwicki et al. use a targeted shRNA screening strategy to identify transcriptional and epigenomic dependencies in poorly differentiated human cancers. TRPS1 is a lineage-specific transcription factor that is required for mitosis in breast cancer cells. TRPS1 is associated with the NuRD complex, and it regulates cell adhesion, cytoskeleton, and G2-M phase-related genes.
- Published
- 2018
26. Genome-Scale CRISPR Screening Identifies Novel Human Pluripotent Gene Networks
- Author
-
Gregory R. Hoffman, Kathleen A. Worringer, Ranjit Randhawa, Sravya Kommineni, Robert J. Ihry, Gregory McAllister, Marie Sondey, Steven Paula, Ricardo E. Dolmetsch, Zinger Yang, Ajamete Kaykas, Elizabeth Frias, Daniel J. Ho, Joe Raymond, Bob Altshuler, Max R. Salick, Carsten Russ, and John S. Reece-Hoyes
- Subjects
Cas9 ,Somatic cell ,Genome scale ,Gene regulatory network ,CRISPR ,Computational biology ,Stem cell ,Biology ,Induced pluripotent stem cell ,Gene - Abstract
Human pluripotent stem cells (hPSCs) generate a wide variety of disease-relevant cells that can be used to improve the translation of preclinical research. Despite the potential of hPSCs, their use for genetic screening has been limited because of technical challenges. We developed a renewable Cas9/sgRNA-hPSC library where loss-of-function mutations can be induced at will. Our inducible-mutant hPSC library can be used for an unlimited number of genome-wide screens. We screened for novel genes involved in 3 of the fundamental properties of hPSCs: Their ability to self-renew/survive, their capacity to differentiate into somatic cells, and their inability to survive as single-cell clones. We identified a plethora of novel genes with unidentified roles in hPSCs. These results are available as a resource for the community to increase the understanding of both human development and genetics. In the future, our stem cell library approach will be a powerful tool to identify disease-modifying genes.VISUAL ABSTRACT
- Published
- 2018
- Full Text
- View/download PDF
27. Genome-wide CRISPR screen for PARKIN regulators reveals transcriptional repression as a determinant of mitophagy
- Author
-
Rowena DeJesus, Andreas W. Sailer, Stephen B. Helliwell, Gregory McAllister, John S. Reece-Hoyes, Christophe Crochemore, Florian Nigsch, Carsten Russ, Christoph Potting, Walter Carbone, Matthias Müller, Judith Knehr, Gregory R. Hoffman, Alicia Lindeman, Carole Manneville, Isabel Schmidt, Guglielmo Roma, Francesca Moretti, and Rob Maher
- Subjects
0301 basic medicine ,Ubiquitin-Protein Ligases ,Induced Pluripotent Stem Cells ,Repressor ,PINK1 ,Parkin ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Mitophagy ,CRISPR ,Humans ,Phosphorylation ,Cells, Cultured ,Regulation of gene expression ,Neurons ,Multidisciplinary ,biology ,Cas9 ,Genome, Human ,Ubiquitin ,Infant, Newborn ,HCT116 Cells ,nervous system diseases ,Ubiquitin ligase ,Cell biology ,Repressor Proteins ,030104 developmental biology ,HEK293 Cells ,Gene Expression Regulation ,SI Correction ,biology.protein ,CRISPR-Cas Systems ,Protein Kinases ,030217 neurology & neurosurgery - Abstract
PARKIN, an E3 ligase mutated in familial Parkinson's disease, promotes mitophagy by ubiquitinating mitochondrial proteins for efficient engagement of the autophagy machinery. Specifically, PARKIN-synthesized ubiquitin chains represent targets for the PINK1 kinase generating phosphoS65-ubiquitin (pUb), which constitutes the mitophagy signal. Physiological regulation of PARKIN abundance, however, and the impact on pUb accumulation are poorly understood. Using cells designed to discover physiological regulators of PARKIN abundance, we performed a pooled genome-wide CRISPR/Cas9 knockout screen. Testing identified genes individually resulted in a list of 53 positive and negative regulators. A transcriptional repressor network including THAP11 was identified and negatively regulates endogenous PARKIN abundance. RNAseq analysis revealed the PARKIN-encoding locus as a prime THAP11 target, and THAP11 CRISPR knockout in multiple cell types enhanced pUb accumulation. Thus, our work demonstrates the critical role of PARKIN abundance, identifies regulating genes, and reveals a link between transcriptional repression and mitophagy, which is also apparent in human induced pluripotent stem cell-derived neurons, a disease-relevant cell type.
- Published
- 2017
28. p53 inhibits CRISPR-Cas9 engineering in human pluripotent stem cells
- Author
-
Elizabeth Frias, Carsten Russ, Sravya Kommineni, Robert J. Ihry, William C. Forrester, Marie Sondey, Kraig M. Theriault, Chaoyang Ye, Ajamete Kaykas, Daniel J. Ho, Ricardo E. Dolmetsch, Kathleen A. Worringer, Max R. Salick, Gregory R. Hoffman, Gregory McAllister, Tripti Kulkarni, Julie Chen, Zinger Yang, Ranjit Randhawa, and John S. Reece-Hoyes
- Subjects
0301 basic medicine ,Cyclin-Dependent Kinase Inhibitor p21 ,Pluripotent Stem Cells ,Transcription, Genetic ,Biology ,Genome ,General Biochemistry, Genetics and Molecular Biology ,Genome engineering ,03 medical and health sciences ,CRISPR-Associated Protein 9 ,CRISPR ,Humans ,DNA Breaks, Double-Stranded ,RNA, Messenger ,fas Receptor ,Induced pluripotent stem cell ,Cas9 ,Gene targeting ,General Medicine ,Embryonic stem cell ,Cell biology ,030104 developmental biology ,Stem cell ,CRISPR-Cas Systems ,Tumor Suppressor Protein p53 ,Genetic Engineering ,Gene Deletion ,RNA, Guide, Kinetoplastida - Abstract
CRISPR/Cas9 has revolutionized our ability to engineer genomes and conduct genome-wide screens in human cells1–3. Whereas some cell types are amenable to genome engineering, genomes of human pluripotent stem cells (hPSCs) have been difficult to engineer, with reduced efficiencies relative to tumour cell lines or mouse embryonic stem cells3–13. Here, using hPSC lines with stable integration of Cas9 or transient delivery of Cas9-ribonucleoproteins (RNPs), we achieved an average insertion or deletion (indel) efficiency greater than 80%. This high efficiency of indel generation revealed that double-strand breaks (DSBs) induced by Cas9 are toxic and kill most hPSCs. In previous studies, the toxicity of Cas9 in hPSCs was less apparent because of low transfection efficiency and subsequently low DSB induction 3 . The toxic response to DSBs was P53/TP53-dependent, such that the efficiency of precise genome engineering in hPSCs with a wild-type P53 gene was severely reduced. Our results indicate that Cas9 toxicity creates an obstacle to the high-throughput use of CRISPR/Cas9 for genome engineering and screening in hPSCs. Moreover, as hPSCs can acquire P53 mutations 14 , cell replacement therapies using CRISPR/Cas9-enginereed hPSCs should proceed with caution, and such engineered hPSCs should be monitored for P53 function.
- Published
- 2017
29. P53 toxicity is a hurdle to CRISPR/CAS9 screening and engineering in human pluripotent stem cells
- Author
-
Elizabeth Frias, Sravya Kommineni, Robert J. Ihry, William C. Forrester, Gregory R. Hoffman, Zinger Yang, Julie Chen, John S. Reece-Hoyes, Kraig M. Theriault, Ricardo E. Dolmetsch, Kathleen A. Worringer, Ranjit Randhawa, Gregory McAllister, Chaoyang Ye, Ajamete Kaykas, Marie Sondey, Daniel J. Ho, Carsten Russ, Max R. Salick, and Tripti Kulkarni
- Subjects
Genetics ,Cell type ,Cas9 ,Cell culture ,Toxicity ,CRISPR ,Biology ,Tp53 mutation ,Induced pluripotent stem cell ,Function (biology) ,Cell biology - Abstract
SUMMARYCRISPR/Cas9 has revolutionized our ability to engineer genomes and to conduct genome-wide screens in human cells. While some cell types are easily modified with Cas9, human pluripotent stem cells (hPSCs) poorly tolerate Cas9 and are difficult to engineer. Using a stable Cas9 cell line or transient delivery of ribonucleoproteins (RNPs) we achieved an average insertion or deletion efficiency greater than 80%. This high efficiency made it apparent that double strand breaks (DSBs) induced by Cas9 are toxic and kill most treated hPSCs. Cas9 toxicity creates an obstacle to the high-throughput use CRISPR/Cas9 for genome-engineering and screening in hPSCs. We demonstrated the toxic response istp53-dependent and the toxic effect oftp53severely reduces the efficiency of precise genome-engineering in hPSCs. Our results highlight that CRISPR-based therapies derived from hPSCs should proceed with caution. Following engineering, it is critical to monitor fortp53function, especially in hPSCs which spontaneously acquiretp53mutations.
- Published
- 2017
- Full Text
- View/download PDF
30. TRRAP is a central regulator of human multiciliated cell formation
- Author
-
Carsten Russ, Nicole A. Renaud, John S. Reece-Hoyes, Guglielmo Roma, Rob Maher, Gregory R. Hoffman, Zinger Yang, Angelica D. Pessotti, Walter Carbone, Nadire Cochran, Rayman Choo-Wing, Lindsey W. Plasschaert, Shivani Aryal, Aron B. Jaffe, Alicia Lindeman, Nathaniel D. Kirkpatrick, Zhao Wang, and Gregory McAllister
- Subjects
0301 basic medicine ,Cell type ,Xenopus ,Cell Cycle Proteins ,Cell Line ,Epigenesis, Genetic ,Small hairpin RNA ,03 medical and health sciences ,Report ,Humans ,Cell Lineage ,Cilia ,Receptor, Notch2 ,Progenitor cell ,RNA, Small Interfering ,Transcription factor ,Zebrafish ,Lung ,Research Articles ,Adaptor Proteins, Signal Transducing ,Regulation of gene expression ,biology ,food and beverages ,Nuclear Proteins ,Epithelial Cells ,Forkhead Transcription Factors ,Cell Biology ,biology.organism_classification ,Cell biology ,030104 developmental biology ,Gene Expression Regulation ,Motile cilium ,Signal Transduction ,Transcription Factors - Abstract
Multiciliated cells (MCCs) function to promote directional fluid flow across epithelial tissues. Wang et al. show that TRRAP, a component of multiple histone acetyltransferase complexes, is required for airway MCC formation and regulates a network of genes involved in MCC differentiation and function., The multiciliated cell (MCC) is an evolutionarily conserved cell type, which in vertebrates functions to promote directional fluid flow across epithelial tissues. In the conducting airway, MCCs are generated by basal stem/progenitor cells and act in concert with secretory cells to perform mucociliary clearance to expel pathogens from the lung. Studies in multiple systems, including Xenopus laevis epidermis, murine trachea, and zebrafish kidney, have uncovered a transcriptional network that regulates multiple steps of multiciliogenesis, ultimately leading to an MCC with hundreds of motile cilia extended from their apical surface, which beat in a coordinated fashion. Here, we used a pool-based short hairpin RNA screening approach and identified TRRAP, an essential component of multiple histone acetyltransferase complexes, as a central regulator of MCC formation. Using a combination of immunofluorescence, signaling pathway modulation, and genomic approaches, we show that (a) TRRAP acts downstream of the Notch2-mediated basal progenitor cell fate decision and upstream of Multicilin to control MCC differentiation; and (b) TRRAP binds to the promoters and regulates the expression of a network of genes involved in MCC differentiation and function, including several genes associated with human ciliopathies.
- Published
- 2017
31. Identification of a novel NAMPT inhibitor by CRISPR/Cas9 chemogenomic profiling in mammalian cells
- Author
-
Chantale T. Guy, Markus Schirle, Bertran Gerrits, David Estoppey, Xuewen Pan, Jason R. Thomas, Malini Varadarajan, Annick Waldt, Gregory McAllister, Jeffrey Hewett, Kevin Xie, Nadire Ramadan, Sven Schuierer, Guglielmo Roma, Judith Knehr, Elizabeth Frias, Qiong Wang, Zinger Yang, Tewis Bouwmeester, Alicia Lindeman, John S. Reece-Hoyes, Walter Carbone, Edmund Harrington, Dominic Hoepfner, Carsten Russ, Xin Chen, Rachel Cuttat, and Gregory R. Hoffman
- Subjects
0301 basic medicine ,Induced Pluripotent Stem Cells ,Nicotinamide phosphoribosyltransferase ,Computational biology ,Biology ,Chemical genetics ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Target identification ,Drug Discovery ,medicine ,Humans ,CRISPR ,Enzyme Inhibitors ,Nicotinamide Phosphoribosyltransferase ,Induced pluripotent stem cell ,Cells, Cultured ,Genetics ,Multidisciplinary ,Drug discovery ,Cas9 ,Gene deletion ,Pharmacogenomic Testing ,030104 developmental biology ,Mechanism of action ,chemistry ,030220 oncology & carcinogenesis ,CRISPR-Cas Systems ,medicine.symptom ,Gene Deletion - Abstract
Scientific Reports, 7, ISSN:2045-2322
- Published
- 2017
- Full Text
- View/download PDF
32. Functional epigenetics approach identifies BRM/SMARCA2 as a critical synthetic lethal target in BRG1-deficient cancers
- Author
-
Craig Mickanin, Linda Bagdasarian, Gregory R. Hoffman, Jeffery A. Porter, Kay Xiang, Huili Zhai, Zainab Jagani, Matthew J. Meyer, Frank Buxton, Vic E. Myer, Nadire Ramadan, Rami Rahal, Elizabeth Frias, Charles W. M. Roberts, Janina Huber, Frank Stegmeier, Kristy Haas, Gregory McAllister, Tanushree Phadke, Mariela Jaskelioff, Rodrigo Romero, Veronica Saenz-Vash, Alicia Lindeman, Dongshu Chen, Boris G. Wilson, Margaret E. McLaughlin, and Nicholas Keen
- Subjects
animal structures ,Blotting, Western ,Synthetic lethality ,Chromatin remodeling ,Epigenesis, Genetic ,Histones ,Cell Line, Tumor ,Neoplasms ,Humans ,Immunoprecipitation ,Epigenetics ,RNA, Small Interfering ,Cellular Senescence ,Gene Library ,Genetics ,Multidisciplinary ,biology ,DNA Helicases ,Nuclear Proteins ,Cell Cycle Checkpoints ,Biological Sciences ,Chromatin ,Histone ,Gene Knockdown Techniques ,Multiprotein Complexes ,Cancer cell ,Cancer research ,biology.protein ,SMARCA4 ,Cell aging ,Transcription Factors - Abstract
Defects in epigenetic regulation play a fundamental role in the development of cancer, and epigenetic regulators have recently emerged as promising therapeutic candidates. We therefore set out to systematically interrogate epigenetic cancer dependencies by screening an epigenome-focused deep-coverage design shRNA (DECODER) library across 58 cancer cell lines. This screen identified BRM/SMARCA2, a DNA-dependent ATPase of the mammalian SWI/SNF (mSWI/SNF) chromatin remodeling complex, as being essential for the growth of tumor cells that harbor loss of function mutations in BRG1/SMARCA4. Depletion of BRM in BRG1-deficient cancer cells leads to a cell cycle arrest, induction of senescence, and increased levels of global H3K9me3. We further demonstrate the selective dependency of BRG1-mutant tumors on BRM in vivo. Genetic alterations of the mSWI/SNF chromatin remodeling complexes are the most frequent among chromatin regulators in cancers, with BRG1/SMARCA4 mutations occurring in ∼10-15% of lung adenocarcinomas. Our findings position BRM as an attractive therapeutic target for BRG1 mutated cancers. Because BRG1 and BRM function as mutually exclusive catalytic subunits of the mSWI/SNF complex, we propose that such synthetic lethality may be explained by paralog insufficiency, in which loss of one family member unveils critical dependence on paralogous subunits. This concept of "cancer-selective paralog dependency" may provide a more general strategy for targeting other tumor suppressor lesions/complexes with paralogous subunits.
- Published
- 2014
- Full Text
- View/download PDF
33. YAP, but Not RSPO-LGR4/5, Signaling in Biliary Epithelial Cells Promotes a Ductular Reaction in Response to Liver Injury
- Author
-
Florian Nigsch, Lapo Morelli, Zinger Yang, Tianliang Sun, Maryam Syed, Linda E. Greenbaum, Guglielmo Roma, John S. Reece-Hoyes, Jan S. Tchorz, Luigi Terracciano, Thomas B. Nicholson, Andreas W. Sailer, Caroline Gubser Keller, Lara Planas-Paz, Nadire Cochran, Sven Schuierer, Tewis Bouwmeester, Yi Yang, Carlos M. Cobos, Jesse J. Lugus, Annick Waldt, Vanessa Orsini, John Alford, Jasna Jetzer, Xiaohong Mao, Philipp S. Hoppe, Frederic Sigoillot, Wibke Schwarzer, Monika Pikiolek, Nicole Carballido-Perrig, Gregory McAllister, Carsten Russ, Feng Cong, Sebastian Bergling, Le Zhang, Gregory R. Hoffman, Marilisa Neri, Rachel Cuttat, and Bernd Kinzel
- Subjects
Liver injury ,0303 health sciences ,LGR5 ,Wnt signaling pathway ,Liver Stem Cell ,Cell Biology ,Biology ,medicine.disease ,Liver regeneration ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Genetics ,medicine ,AXIN2 ,Molecular Medicine ,Signal transduction ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Summary Biliary epithelial cells (BECs) form bile ducts in the liver and are facultative liver stem cells that establish a ductular reaction (DR) to support liver regeneration following injury. Liver damage induces periportal LGR5+ putative liver stem cells that can form BEC-like organoids, suggesting that RSPO-LGR4/5-mediated WNT/β-catenin activity is important for a DR. We addressed the roles of this and other signaling pathways in a DR by performing a focused CRISPR-based loss-of-function screen in BEC-like organoids, followed by in vivo validation and single-cell RNA sequencing. We found that BECs lack and do not require LGR4/5-mediated WNT/β-catenin signaling during a DR, whereas YAP and mTORC1 signaling are required for this process. Upregulation of AXIN2 and LGR5 is required in hepatocytes to enable their regenerative capacity in response to injury. Together, these data highlight heterogeneity within the BEC pool, delineate signaling pathways involved in a DR, and clarify the identity and roles of injury-induced periportal LGR5+ cells.
- Published
- 2019
- Full Text
- View/download PDF
34. Genome-Scale CRISPR Screens Identify Human Pluripotency-Specific Genes
- Author
-
Max R. Salick, Carsten Russ, Sravya Kommineni, Ranjit Randhawa, Robert J. Ihry, Gregory R. Hoffman, John S. Reece-Hoyes, Marie Sondey, Elizabeth Frias, Kathleen A. Worringer, Beata Henry, Robert C. Altshuler, Gregory McAllister, Qiong Wang, Joe Raymond, Zinger Yang, Daniel J. Ho, Chaoyang Ye, Ajamete Kaykas, Steven Paula, and Ricardo E. Dolmetsch
- Subjects
Pluripotent Stem Cells ,0301 basic medicine ,Cell type ,Genome ,Cas9 ,Mutant ,Genome scale ,Computational biology ,Biology ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Preclinical research ,030104 developmental biology ,0302 clinical medicine ,lcsh:Biology (General) ,Humans ,CRISPR ,Genetic Testing ,CRISPR-Cas Systems ,Induced pluripotent stem cell ,lcsh:QH301-705.5 ,Gene ,030217 neurology & neurosurgery - Abstract
Summary: Human pluripotent stem cells (hPSCs) generate a variety of disease-relevant cells that can be used to improve the translation of preclinical research. Despite the potential of hPSCs, their use for genetic screening has been limited by technical challenges. We developed a scalable and renewable Cas9 and sgRNA-hPSC library in which loss-of-function mutations can be induced at will. Our inducible mutant hPSC library can be used for multiple genome-wide CRISPR screens in a variety of hPSC-induced cell types. As proof of concept, we performed three screens for regulators of properties fundamental to hPSCs: their ability to self-renew and/or survive (fitness), their inability to survive as single-cell clones, and their capacity to differentiate. We identified the majority of known genes and pathways involved in these processes, as well as a plethora of genes with unidentified roles. This resource will increase the understanding of human development and genetics. This approach will be a powerful tool to identify disease-modifying genes and pathways. : Ihry et al. develop a CRISPR/Cas9 genetic screening platform for hPSCs that enables unbiased genome-scale genetic screening. The platform exhibits high performance and accurately detects the dropout of essential genes. Furthermore, proof-of-concept screens exploit hPSC-specific phenotypes to identify regulators of fitness, survival after single-cell dissociation, and pluripotency. Keywords: CRISPR genome-wide screening, human pluripotent stem cells, iPSC, hESC, PAWR, PMAIP1, DDR
- Published
- 2019
- Full Text
- View/download PDF
35. The Multidimensional Perturbation Value: A Single Metric to Measure Similarity and Activity of Treatments in High-Throughput Multidimensional Screens
- Author
-
Douglas W. Selinger, Savina Jaeger, Ben Cornett, Somnath Bandyopadhyay, Hua Wu, Florian Nigsch, Ioannis Moutsatsos, Janna Hutz, Gregory McAllister, Jeremy L. Jenkins, and Thomas Nelson
- Subjects
Principal Component Analysis ,Computer science ,Statistics as Topic ,Multidimensional data ,Computational biology ,Hydroxamic Acids ,Bioinformatics ,Biochemistry ,High-Throughput Screening Assays ,Analytical Chemistry ,High-content screening ,MCF-7 Cells ,Humans ,Molecular Medicine ,Computer Simulation ,DNA microarray ,Biotechnology - Abstract
Screens using high-throughput, information-rich technologies such as microarrays, high-content screening (HCS), and next-generation sequencing (NGS) have become increasingly widespread. Compared with single-readout assays, these methods produce a more comprehensive picture of the effects of screened treatments. However, interpreting such multidimensional readouts is challenging. Univariate statistics such as t-tests and Z-factors cannot easily be applied to multidimensional profiles, leaving no obvious way to answer common screening questions such as "Is treatment X active in this assay?" and "Is treatment X different from (or equivalent to) treatment Y?" We have developed a simple, straightforward metric, the multidimensional perturbation value (mp-value), which can be used to answer these questions. Here, we demonstrate application of the mp-value to three data sets: a multiplexed gene expression screen of compounds and genomic reagents, a microarray-based gene expression screen of compounds, and an HCS compound screen. In all data sets, active treatments were successfully identified using the mp-value, and simulations and follow-up analyses supported the mp-value's statistical and biological validity. We believe the mp-value represents a promising way to simplify the analysis of multidimensional data while taking full advantage of its richness.
- Published
- 2013
- Full Text
- View/download PDF
36. Author response: Functional CRISPR screening identifies the ufmylation pathway as a regulator of SQSTM1/p62
- Author
-
Zuncai Wang, Walter Carbone, Jennifer Kelliher, Rowena DeJesus, Guglielmo Roma, John S. Reece-Hoyes, Aylwin Ng, Francesca Moretti, Craig Mickanin, Phil Bergman, Alicia Lindeman, Shanming Liu, Gregory R. Hoffman, John Alford, Martin Beibel, Beat Nyfeler, Leon Murphy, Carsten Russ, John A. Tallarico, Gregory McAllister, Judith Knehr, Ramnik J. Xavier, Elizabeth Frias, and Jeffery A. Porter
- Subjects
Regulator ,CRISPR ,Computational biology ,Biology - Published
- 2016
- Full Text
- View/download PDF
37. Functional CRISPR screening identifies the ufmylation pathway as a regulator of SQSTM1/p62
- Author
-
Craig Mickanin, Gregory R. Hoffman, Jeffery A. Porter, Guglielmo Roma, Carsten Russ, Gregory McAllister, Walter Carbone, John A. Tallarico, Beat Nyfeler, Zuncai Wang, Jennifer Kelliher, Francesca Moretti, Ramnik J. Xavier, Leon Murphy, John S. Reece-Hoyes, John Alford, Phil Bergman, Alicia Lindeman, Elizabeth Frias, Shanming Liu, Rowena DeJesus, Judith Knehr, Martin Beibel, and Aylwin Ng
- Subjects
0301 basic medicine ,Cell signaling ,autophagy ,QH301-705.5 ,Science ,Regulator ,Computational biology ,Biology ,Biochemistry ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,Genome editing ,RNA interference ,Sequestosome-1 Protein ,CRISPR ,Humans ,Clustered Regularly Interspaced Short Palindromic Repeats ,SQSTM1 ,Genetic Testing ,Biology (General) ,Genetics ,CRISPR interference ,General Immunology and Microbiology ,General Neuroscience ,TOR Serine-Threonine Kinases ,Signal transducing adaptor protein ,Proteins ,General Medicine ,Cell Biology ,Flow Cytometry ,Tools and Resources ,030104 developmental biology ,Gene Expression Regulation ,Gene Targeting ,Unfolded protein response ,Medicine ,ER stress ,Protein Processing, Post-Translational ,Signal Transduction ,Human - Abstract
SQSTM1 is an adaptor protein that integrates multiple cellular signaling pathways and whose expression is tightly regulated at the transcriptional and post-translational level. Here, we describe a forward genetic screening paradigm exploiting CRISPR-mediated genome editing coupled to a cell selection step by FACS to identify regulators of SQSTM1. Through systematic comparison of pooled libraries, we show that CRISPR is superior to RNAi in identifying known SQSTM1 modulators. A genome-wide CRISPR screen exposed MTOR signalling and the entire macroautophagy machinery as key regulators of SQSTM1 and identified several novel modulators including HNRNPM, SLC39A14, SRRD, PGK1 and the ufmylation cascade. We show that ufmylation regulates SQSTM1 by eliciting a cell type-specific ER stress response which induces SQSTM1 expression and results in its accumulation in the cytosol. This study validates pooled CRISPR screening as a powerful method to map the repertoire of cellular pathways that regulate the fate of an individual target protein. DOI: http://dx.doi.org/10.7554/eLife.17290.001
- Published
- 2016
38. Comparative expression analysis of four breast cancer subtypes versus matched normal tissue from the same patients
- Author
-
Paolo Martini, Robert K. Campbell, Jadwiga Bienkowska, Jennifer Jackson, Gregory McAllister, Deanne Taylor, and Heike Keilhack
- Subjects
Pathology ,medicine.medical_specialty ,Microarray ,Receptor, ErbB-2 ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,Estrogen receptor ,Breast Neoplasms ,Biology ,Biochemistry ,Endocrinology ,Breast cancer ,medicine ,Humans ,skin and connective tissue diseases ,Molecular Biology ,Gene ,Cell Proliferation ,Regulation of gene expression ,Gene Expression Profiling ,Cancer ,Cell Biology ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Gene expression profiling ,Receptors, Estrogen ,Health ,Disease Progression ,Cancer research ,Molecular Medicine ,DNA microarray - Abstract
Gene expression studies have been widely used in an effort to identify signatures that can predict clinical progression of cancer. In this study we focused instead on identifying gene expression differences between breast tumors and adjacent normal tissue, and between different subtypes of tumor classified by clinical marker status. We have collected a set of 20 breast cancer tissues, matched with the adjacent pathologically normal tissue from the same patient. The cancer samples representing each subtype of breast cancer identified by estrogen receptor ER(+/-) and Her2(+/-) status and divided into four subgroups (ER+/Her2+, ER+/Her2-, ER-/Her2+, and ER-/Her2-) were hybridized on Affymetrix HG-133 Plus 2.0 microarrays. By comparing cancer samples with their matched normal controls we have identified 3537 overall differentially expressed genes using data analysis methods from Bioconductor. When we looked at the genes in common of the four subgroups, we found 151 regulated genes, some of them encoding known targets for breast cancer treatment. Unique genes in the four subgroups instead suggested gene regulation dependent on the ER/Her2 markers selection. In conclusion, the results indicate that microarray studies using robust analysis of matched tumor and normal samples from the same patients can be used to identify genes differentially expressed in breast cancer tumor subtypes even when small numbers of samples are considered and can further elucidate molecular features of breast cancer.
- Published
- 2008
- Full Text
- View/download PDF
39. Macroautophagy is dispensable for growth of KRAS mutant tumors and chloroquine efficacy
- Author
-
Christina H. Eng, Lourdes Toral-Barza, Judy Lucas, Elizabeth George, Leon Murphy, Stephanie Fitzgerald, Zuncai Wang, Kevin Bray, Valeria Fantin, Rowena De Jesus, Gregory McAllister, Gregory R. Hoffman, Diane Tkach, Beat Nyfeler, Shanming Liu, Luanna Lemon, Robert T. Abraham, Elizabeth Frias, Nadire Cochran, and Savuth Ugwonali
- Subjects
0301 basic medicine ,Indoles ,Antineoplastic Agents ,Ubiquitin-Activating Enzymes ,Biology ,medicine.disease_cause ,Autophagy-Related Protein 7 ,Radiation Tolerance ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,chemistry.chemical_compound ,Erlotinib Hydrochloride ,Gene Knockout Techniques ,Chloroquine ,Cell Line, Tumor ,medicine ,Autophagy ,Sunitinib ,Humans ,Pyrroles ,Protein Kinase Inhibitors ,Cell Proliferation ,Multidisciplinary ,Cell growth ,Biological Sciences ,030104 developmental biology ,Cell Transformation, Neoplastic ,chemistry ,Drug Resistance, Neoplasm ,Mutation ,Cancer research ,Erlotinib ,KRAS ,Growth inhibition ,Carcinogenesis ,Tyrosine kinase ,medicine.drug - Abstract
Macroautophagy is a key stress-response pathway that can suppress or promote tumorigenesis depending on the cellular context. Notably, Kirsten rat sarcoma (KRAS)-driven tumors have been reported to rely on macroautophagy for growth and survival, suggesting a potential therapeutic approach of using autophagy inhibitors based on genetic stratification. In this study, we evaluated whether KRAS mutation status can predict the efficacy to macroautophagy inhibition. By profiling 47 cell lines with pharmacological and genetic loss-of-function tools, we were unable to confirm that KRAS-driven tumor lines require macroautophagy for growth. Deletion of autophagy-related 7 (ATG7) by genome editing completely blocked macroautophagy in several tumor lines with oncogenic mutations in KRAS but did not inhibit cell proliferation in vitro or tumorigenesis in vivo. Furthermore, ATG7 knockout did not sensitize cells to irradiation or to several anticancer agents tested. Interestingly, ATG7-deficient and -proficient cells were equally sensitive to the antiproliferative effect of chloroquine, a lysosomotropic agent often used as a pharmacological tool to evaluate the response to macroautophagy inhibition. Moreover, both cell types manifested synergistic growth inhibition when treated with chloroquine plus the tyrosine kinase inhibitors erlotinib or sunitinib, suggesting that the antiproliferative effects of chloroquine are independent of its suppressive actions on autophagy.
- Published
- 2015
40. Evidence-Based and Quantitative Prioritization of Tool Compounds in Phenotypic Drug Discovery
- Author
-
Allen Cornett, Jeremy L. Jenkins, Yuan Wang, Florian Nigsch, Yi Mao, C. Gregory Paris, Gregory McAllister, and Frederick J. King
- Subjects
0301 basic medicine ,Prioritization ,Evidence-based practice ,Phenotypic screening ,Clinical Biochemistry ,Computational biology ,Biology ,Bioinformatics ,01 natural sciences ,Biochemistry ,Cell Line ,03 medical and health sciences ,Automation ,High-Throughput Screening Assays ,Drug Discovery ,Humans ,Polypharmacology ,Molecular Biology ,Pharmacology ,Molecular Structure ,010405 organic chemistry ,Drug discovery ,0104 chemical sciences ,030104 developmental biology ,Phenotype ,Molecular Probes ,Molecular Medicine ,Identification (biology) ,Classical pharmacology ,Databases, Chemical - Abstract
The use of potent and selective chemical tools with well-defined targets can help elucidate biological processes driving phenotypes in phenotypic screens. However, identification of selective compounds en masse to create targeted screening sets is non-trivial. A systematic approach is needed to prioritize probes, which prevents the repeated use of published but unselective compounds. Here we performed a meta-analysis of integrated large-scale, heterogeneous bioactivity data to create an evidence-based, quantitative metric to systematically rank tool compounds for targets. Our tool score (TS) was then tested on hundreds of compounds by assessing their activity profiles in a panel of 41 cell-based pathway assays. We demonstrate that high-TS tools show more reliably selective phenotypic profiles than lower-TS compounds. Additionally we highlight frequently tested compounds that are non-selective tools and distinguish target family polypharmacology from cross-family promiscuity. TS can therefore be used to prioritize compounds from heterogeneous databases for phenotypic screening.
- Published
- 2015
41. Inhibition of Casein Kinase 1 Alpha Prevents Acquired Drug Resistance to Erlotinib in EGFR-Mutant Non-Small Cell Lung Cancer
- Author
-
Kaitlin J. McCutcheon, Elizabeth Frias, David A. Ruddy, Dongshu Chen, Matthew J. Meyer, Gregory McAllister, Daniel P. Rakiec, Sreenath V. Sharma, Gregory R. Hoffman, Joshua M. Korn, Alexandra Lantermann, and Frank Stegmeier
- Subjects
Cancer Research ,Lung Neoplasms ,Immunoblotting ,Mice, Nude ,Antineoplastic Agents ,Drug resistance ,Pharmacology ,Real-Time Polymerase Chain Reaction ,Erlotinib Hydrochloride ,Mice ,Gefitinib ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Medicine ,Animals ,Humans ,RNA, Small Interfering ,Lung cancer ,neoplasms ,Oligonucleotide Array Sequence Analysis ,Gene knockdown ,business.industry ,Casein Kinase I ,Genes, erbB-1 ,medicine.disease ,Xenograft Model Antitumor Assays ,respiratory tract diseases ,Oncology ,Drug Resistance, Neoplasm ,Gene Knockdown Techniques ,Cancer research ,Female ,Casein kinase 1 ,Erlotinib ,Signal transduction ,business ,medicine.drug - Abstract
Patients with lung tumors harboring activating mutations in the EGF receptor (EGFR) show good initial treatment responses to the EGFR tyrosine kinase inhibitors (TKI) erlotinib or gefitinib. However, acquired resistance invariably develops. Applying a focused shRNA screening approach to identify genes whose knockdown can prevent and/or overcome acquired resistance to erlotinib in several EGFR-mutant non–small cell lung cancer (NSCLC) cell lines, we identified casein kinase 1 α (CSNK1A1, CK1α). We found that CK1α suppression inhibits the NF-κB prosurvival signaling pathway. Furthermore, downregulation of NF-κB signaling by approaches independent of CK1α knockdown can also attenuate acquired erlotinib resistance, supporting a role for activated NF-κB signaling in conferring acquired drug resistance. Importantly, CK1α suppression prevented erlotinib resistance in an HCC827 xenograft model in vivo. Our findings suggest that patients with EGFR-mutant NSCLC might benefit from a combination of EGFR TKIs and CK1α inhibition to prevent acquired drug resistance and to prolong disease-free survival. Cancer Res; 75(22); 4937–48. ©2015 AACR.
- Published
- 2015
42. Project DRIVE: A Compendium of Cancer Dependencies and Synthetic Lethal Relationships Uncovered by Large-Scale, Deep RNAi Screening
- Author
-
Christine Stephan, William R. Sellers, Deborah Castelletti, Jeffery A. Porter, Julie L. Bernard, Sandra Mollé, Mark Stump, Tami Hood, Joshua M. Korn, Audrey Kauffmann, Giorgio G. Galli, Kristine Yu, Li Li, Marc Hattenberger, Javad Golji, Zainab Jagani, Marco Wallroth, Tobias Schmelzle, Philippe Megel, Raymond Pagliarini, Rosemary Barrett, Yingzi Yue, Richard S. Eldridge, Jan Weiler, Alberto C. Vitari, Konstantinos J. Mavrakis, Kalyani Gampa, Elizabeth Ackley, Rosalie deBeaumont, Qiong Shen, Joel Berger, Tanja Schouwey, Franklin Chung, E. Robert McDonald, Gregory McAllister, Christelle Stamm, Frances Shanahan, Aurore Desplat, Iris Kao, Thomas A. Perkins, Antoine de Weck, Kavitha Venkatesan, Albert Lai, Jennifer Johnson, Roland Widmer, David A. Ruddy, Avnish Kapoor, Brian Repko, François Gauter, Nicholas Keen, Tanushree Phadke, Eric Billy, Sosathya Sovath, Typhaine Martin, Elizabeth Frias, Justina X. Caushi, Vic E. Myer, Malini Varadarajan, William C. Forrester, Fei Feng, Hans Bitter, Ralph Tiedt, Yue Liu, Jing Zhang, Dorothee Abramowski, Dhiren Belur, Volker M. Stucke, Odile Weber, Mathias Jenal, Ali Farsidjani, Jianjun Yu, Rebecca Billig, JiaJia Feng, A. B. Meyer, Kristen Hurov, Veronica Gibaja, Michael D. Jones, Daisy Flemming, Donald A. Dwoske, Jilin Liu, Clara Delaunay, William Duong, Frank Buxton, Kaitlin J. Macchi, Saskia M. Brachmann, Alice T. Loo, Craig Mickanin, Francesco Hofmann, Frank Stegmeier, Kristy Haas, Gregory R. Hoffman, Marta Cortes-Cros, Roger Caothien, Shumei Liu, Serena J. Silver, Michael R. Schlabach, Emma Lees, Nadire Ramadan, Qiumei Liu, and Zhenhai Gao
- Subjects
0301 basic medicine ,Lineage (genetic) ,Tumor suppressor gene ,Mutant ,Biology ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,RNA interference ,Cell Line, Tumor ,Neoplasms ,medicine ,Humans ,Gene Regulatory Networks ,RNA, Small Interfering ,Gene ,Gene Library ,Genetics ,Gene knockdown ,Cancer ,Translation (biology) ,Oncogenes ,medicine.disease ,030104 developmental biology ,Multiprotein Complexes ,RNA Interference ,Signal Transduction ,Transcription Factors - Abstract
Elucidation of the mutational landscape of human cancer has progressed rapidly and been accompanied by the development of therapeutics targeting mutant oncogenes. However, a comprehensive mapping of cancer dependencies has lagged behind and the discovery of therapeutic targets for counteracting tumor suppressor gene loss is needed. To identify vulnerabilities relevant to specific cancer subtypes, we conducted a large-scale RNAi screen in which viability effects of mRNA knockdown were assessed for 7,837 genes using an average of 20 shRNAs per gene in 398 cancer cell lines. We describe findings of this screen, outlining the classes of cancer dependency genes and their relationships to genetic, expression, and lineage features. In addition, we describe robust gene-interaction networks recapitulating both protein complexes and functional cooperation among complexes and pathways. This dataset along with a web portal is provided to the community to assist in the discovery and translation of new therapeutic approaches for cancer.
- Published
- 2017
- Full Text
- View/download PDF
43. Identification of Elongation Factor G as the Conserved Cellular Target of Argyrin B
- Author
-
Christian Studer, John A. Tallarico, Christine D. Wilson, David Estoppey, Lewis Whitehead, Zuncai Wang, Ralph Riedl, Adriana K. Jones, S. Whitney Barnes, Swann Gaulis, Ruth E. Caughlan, Philipp Krastel, Leon Murphy, Thomas Aust, Gregory McAllister, N. Rao Movva, Gejing Deng, Michael Salcius, Dominic Hoepfner, Deborah Palestrant, Ervan Hauy, Christina A. Kirby, Gregory A. Michaud, Beat Nyfeler, Saskia M. Brachmann, Robert Yu, Angela L. Woods, Charles R. Dean, and John R. Walker
- Subjects
Macromolecular Assemblies ,Mitochondrial translation ,Mutant ,lcsh:Medicine ,Crystallography, X-Ray ,Ribosome ,Biochemistry ,Conserved sequence ,Drug Discovery ,Molecular Cell Biology ,Protein biosynthesis ,Peptide Elongation Factor G ,Biomacromolecule-Ligand Interactions ,lcsh:Science ,Conserved Sequence ,Cellular Stress Responses ,Mammals ,Multidisciplinary ,biology ,Cell Death ,Pseudomonas aeruginosa ,Structural Proteins ,Oligopeptides ,Allosteric Site ,Research Article ,Protein Binding ,Burkholderia ,Saccharomyces cerevisiae ,Molecular Sequence Data ,Biophysics ,Microbial Sensitivity Tests ,Cell Growth ,Molecular Genetics ,Mitochondrial Proteins ,Genetic Mutation ,Cell Line, Tumor ,Chemical Biology ,Genetics ,Animals ,Humans ,Amino Acid Sequence ,Biology ,Sequence Homology, Amino Acid ,lcsh:R ,Proteins ,biology.organism_classification ,Elongation factor ,lcsh:Q ,Mutant Proteins - Abstract
Argyrins, produced by myxobacteria and actinomycetes, are cyclic octapeptides with antibacterial and antitumor activity. Here, we identify elongation factor G (EF-G) as the cellular target of argyrin B in bacteria, via resistant mutant selection and whole genome sequencing, biophysical binding studies and crystallography. Argyrin B binds a novel allosteric pocket in EF-G, distinct from the known EF-G inhibitor antibiotic fusidic acid, revealing a new mode of protein synthesis inhibition. In eukaryotic cells, argyrin B was found to target mitochondrial elongation factor G1 (EF-G1), the closest homologue of bacterial EF-G. By blocking mitochondrial translation, argyrin B depletes electron transport components and inhibits the growth of yeast and tumor cells. Further supporting direct inhibition of EF-G1, expression of an argyrin B-binding deficient EF-G1 L693Q variant partially rescued argyrin B-sensitivity in tumor cells. In summary, we show that argyrin B is an antibacterial and cytotoxic agent that inhibits the evolutionarily conserved target EF-G, blocking protein synthesis in bacteria and mitochondrial translation in yeast and mammalian cells.
- Published
- 2012
44. Pharmacological Validation of Trypanosoma brucei Phosphodiesterases B1 and B2 as Druggable Targets for African Sleeping Sickness
- Author
-
Robert K. Campbell, Lara Gechijian, Michael P. Pollastri, Alden E. Gustafson, Stefan O. Ochiana, Zhouxi Wang, Trent D. Ashton, Nicholas D. Bland, Gregory McAllister, Cuihua Wang, Kristina Cotter, Rajiv Gangurde, Anna P. Fang, Mary Jo Ondrechen, Craig Tallman, Ron Ortenberg, and Norman Garceau
- Subjects
Models, Molecular ,Pyridines ,Trypanosoma brucei brucei ,Druggability ,Computational biology ,Trypanosoma brucei ,Pharmacology ,Article ,chemistry.chemical_compound ,Structure-Activity Relationship ,Catalytic Domain ,Drug Discovery ,parasitic diseases ,medicine ,Humans ,African trypanosomiasis ,Repurposing ,biology ,Molecular Structure ,Drug discovery ,Phosphodiesterase ,biology.organism_classification ,medicine.disease ,Trypanocidal Agents ,Recombinant Proteins ,Trypanosomiasis, African ,chemistry ,3',5'-Cyclic-AMP Phosphodiesterases ,Benzamides ,Molecular Medicine ,Piclamilast ,Trypanosomiasis - Abstract
Neglected tropical disease drug discovery requires application of pragmatic and efficient methods for development of new therapeutic agents. In this report, we describe our target repurposing efforts for the essential phosphodiesterase (PDE) enzymes TbrPDEB1 and TbrPDEB2 of Trypanosoma brucei , the causative agent for human African trypanosomiasis (HAT). We describe protein expression and purification, assay development, and benchmark screening of a collection of 20 established human PDE inhibitors. We disclose that the human PDE4 inhibitor piclamilast, and some of its analogues, show modest inhibition of TbrPDEB1 and B2 and quickly kill the bloodstream form of the subspecies T. brucei brucei . We also report the development of a homology model of TbrPDEB1 that is useful for understanding the compound-enzyme interactions and for comparing the parasitic and human enzymes. Our profiling and early medicinal chemistry results strongly suggest that human PDE4 chemotypes represent a better starting point for optimization of TbrPDEB inhibitors than those that target any other human PDEs.
- Published
- 2011
45. Abstract PR06: A functional screen of the epigenome identifies BRM/SMARCA2 as a critical synthetic lethal target in BRG1-deficient cancers
- Author
-
Margaret E. McLaughlin, Tanushree Phadke, Charles W. M. Roberts, Boris G. Wilson, Kay Xiang, Linda Bagdasarian, Rami Rahal, Janina Huber, Zainab Jagani, Nicholas Keen, Craig Mickanin, Matthew J. Meyer, Rodrigo Romero, Frank Stegmeier, Nadire Ramadan, Gregory R. Hoffman, Gregory McAllister, Elizabeth Frias, Mariela Jaskelioff, Vic E. Myer, Jeffrey A. Porter, Dongshu Chen, Frank Buxton, Alicia Lindeman, and Kristy Haas
- Subjects
Genetics ,Cancer Research ,Cancer ,Epigenome ,Biology ,medicine.disease ,medicine.disease_cause ,Chromatin ,Oncology ,Cancer cell ,medicine ,Gene silencing ,Epigenetics ,Carcinogenesis ,Epigenomics - Abstract
Epigenetic dysregulation is an emerging hallmark of cancers, and the identification of recurrent somatic mutations in chromatin regulators implies a causal role for altered chromatin states in tumorigenesis. As the majority of epigenetic mutations are inactivating and thus do not present directly druggable targets, we reasoned that these mutations may alter the epigenomic state of cancer cells and thereby expose novel epigenetic vulnerabilities. To systematically search for epigenetic synthetic lethal interactions, we performed a deep coverage pooled shRNA screen across a large collection of cancer cell lines using a library targeting a diverse set of epigenetic regulators. Strikingly, this unbiased screen revealed that silencing of the SWI/SNF ATPase subunit BRM/SMARCA2, selectively inhibits the proliferation of BRG1-deficient cancer cells. The mammalian SWI/SNF complexes (mSWI/SNF) regulate chromatin structure through ATP-dependent nucleosome remodeling. Recent cancer genome studies have revealed a significant frequency of mutations in several components of the mSWI/SNF complexes including loss of the catalytic subunit BRG1 in non-small cell lung cancers. Our studies reveal that BRM knockdown selectively induced cell cycle arrest in BRG1-mutant cancer cells and significantly impaired the growth of BRG1-mutant lung tumor xenografts. BRM is the paralog of BRG1, suggesting a model in which mSWI/SNF mutations lead to a hypomorphic complex that promotes tumorigenesis but cannot tolerate complete inactivation. Therefore, our studies present BRM as an attractive therapeutic target in BRG1-mutant cancers. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):PR06. Citation Format: Zainab Jagani, Gregory Hoffman, Rami Rahal, Frank Buxton, Gregory McAllister, Kay Xiang, Elizabeth Frias, Janina Huber, Alicia Lindeman, Dongshu Chen, Linda Bagdasarian, Rodrigo Romero, Nadire Ramadan, Tanushree Phadke, Kristy Haas, Mariela Jaskelioff, Boris Wilson, Matthew Meyer, Margaret E. McLaughlin, Charles WM Roberts, Vic Myer, Jeff Porter, Nicholas Keen, Craig Mickanin, Frank Stegmeier. A functional screen of the epigenome identifies BRM/SMARCA2 as a critical synthetic lethal target in BRG1-deficient cancers. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2013 Oct 19-23; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(11 Suppl):Abstract nr PR06.
- Published
- 2013
- Full Text
- View/download PDF
46. Pharmacological Validationof TrypanosomabruceiPhosphodiesterases B1 and B2 as Druggable Targetsfor African Sleeping Sickness.
- Author
-
NicholasD. Bland, Cuihua Wang, Craig Tallman, Alden E. Gustafson, Zhouxi Wang, Trent D. Ashton, Stefan O. Ochiana, Gregory McAllister, Kristina Cotter, Anna P. Fang, Lara Gechijian, Norman Garceau, Rajiv Gangurde, Ron Ortenberg, Mary Jo Ondrechen, Robert K. Campbell, and Michael P. Pollastri
- Published
- 2011
- Full Text
- View/download PDF
47. Determination of minimal transcriptional signatures of compounds for target prediction
- Author
-
Jeremy L. Jenkins, Somnath Bandyopadhyay, Janna Hutz, Joseph Loureiro, Ben Cornett, Gregory McAllister, Douglas W. Selinger, and Florian Nigsch
- Subjects
compute unified device architecture (CUDA) ,Drug discovery ,Computer science ,Systems biology ,Research ,graphics processing unit (GPU) programming ,target prediction ,Computational biology ,computer.software_genre ,General Biochemistry, Genetics and Molecular Biology ,Computer Science Applications ,Gene expression profiling ,Computational Mathematics ,Mechanism of action ,Molecular targets ,medicine ,genetic algorithm ,Data mining ,medicine.symptom ,Mode of action ,Gene ,computer ,transcriptional profiling ,Biological variability - Abstract
The identification of molecular target and mechanism of action of compounds is a key hurdle in drug discovery. Multiplexed techniques for bead-based expression profiling allow the measurement of transcriptional signatures of compound-treated cells in high-throughput mode. Such profiles can be used to gain insight into compounds' mode of action and the protein targets they are modulating. Through the proxy of target prediction from such gene signatures we explored important aspects of the use of transcriptional profiles to capture biological variability of perturbed cellular assays. We found that signatures derived from expression data and signatures derived from biological interaction networks performed equally well, and we showed that gene signatures can be optimised using a genetic algorithm. Gene signatures of approximately 128 genes seemed to be most generic, capturing a maximum of the perturbation inflicted on cells through compound treatment. Moreover, we found evidence for oxidative phosphorylation to be one of the most general ways to capture compound perturbation.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.