Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Glenn S. Cowley is active.

Publication


Featured researches published by Glenn S. Cowley.


Nature | 2011

Functional genomics reveal that the serine synthesis pathway is essential in breast cancer

Richard Possemato; Kevin Marks; Yoav D. Shaul; Michael E. Pacold; Dohoon Kim; Kivanc Birsoy; Shalini Sethumadhavan; Hin-Koon Woo; Hyun Gyung Jang; Abhishek K. Jha; Walter W. Chen; Francesca G. Barrett; Nicolas Stransky; Zhi-Yang Tsun; Glenn S. Cowley; Jordi Barretina; Nada Y. Kalaany; Peggy P. Hsu; Kathleen Ottina; Albert M. Chan; Bingbing Yuan; Levi A. Garraway; David E. Root; Mari Mino-Kenudson; Elena F. Brachtel; Edward M. Driggers; David M. Sabatini

Cancer cells adapt their metabolic processes to drive macromolecular biosynthesis for rapid cell growth and proliferation (1,2). RNAi-based loss of function screening has proven powerful for the identification of novel and interesting cancer targets, and recent studies have used this technology in vivo to identify novel tumor suppressor genes (3). Here, we developed a method for identifying novel cancer targets via negative selection RNAi screening in solid tumours. Using this method, we screened a set of metabolic genes associated with aggressive breast cancer and stemness to identify those required for in vivo tumourigenesis. Among the genes identified, phosphoglycerate dehydrogenase (PHGDH) is in a genomic region of recurrent copy number gain in breast cancer and PHGDH protein levels are elevated in 70% of ER-negative breast cancers. PHGDH catalyzes the first step in the serine biosynthesis pathway, and breast cancer cells with high PHGDH expression have elevations in serine synthesis flux. Suppression of PHGDH in cell lines with elevated PHGDH expression, but not those without, causes a strong decrease in cell proliferation and a reduction in serine synthesis. We find that PHGDH suppression does not affect intracellular serine levels, but causes a drop in the levels of alpha-ketoglutarate, another output of the pathway and a TCA cycle intermediate. In cells with high PHGDH expression, the serine synthesis pathway contributes approximately 50% of the total anaplerotic flux of glutamine into the TCA cycle. These results reveal that certain breast cancers are dependent upon increased serine pathway flux caused by PHGDH over-expression and demonstrate the utility of in vivo negative selection RNAi screens for finding potential anticancer targets.


Neuron | 1992

Transgenic mice with a rhodopsin mutation (Pro23His): A mouse model of autosomal dominant retinitis pigmentosa

Jane E. Olsson; Jon W. Gordon; Basil S. Pawlyk; Dorothy Roof; Annmarie Hayes; Robert S. Molday; Shizuo Mukai; Glenn S. Cowley; Eliot L. Berson; Thaddeus P. Dryja

We inserted into the germline of mice either a mutant or wild-type allele from a patient with retinitis pigmentosa and a missense mutation (P23H) in the rhodopsin gene. All three lines of transgenic mice with the mutant allele developed photoreceptor degeneration; the one with the least severe retinal photoreceptor degeneration had the lowest transgene expression, which was one-sixth the level of endogenous murine rod opsin. Of two lines of mice with the wild-type allele, one expressed approximately equal amounts of transgenic and murine opsin and maintained normal retinal function and structure. The other expressed approximately 5 times more transgenic than murine opsin and developed a retinal degeneration similar to that found in mice carrying a mutant allele, presumably due to the overexpression of this protein. Our findings help to establish the pathogenicity of mutant human P23H rod opsin and suggest that overexpression of wild-type human rod opsin leads to a remarkably similar photoreceptor degeneration.


Science | 2013

(R)-2-Hydroxyglutarate Is Sufficient to Promote Leukemogenesis and Its Effects Are Reversible

Julie-Aurore Losman; Ryan E. Looper; Peppi Koivunen; Sungwoo Lee; Rebekka K. Schneider; Christine McMahon; Glenn S. Cowley; David E. Root; Benjamin L. Ebert; William G. Kaelin

Focusing on the Right Metabolite A variety of human cancers, including acute leukemias and brain tumors, have mutations in the genes encoding isocitrate dehydrogenase 1 or 2 (IDH1, IDH2), which cause overproduction of a metabolite called 2-hydroxyglutarate (2HG). Losman et al. (p. 1621, published online 7 February) show that the R- but not the S-enantiomer of 2HG can transform cells and that R-2HG mediates transformation at least in part through effects on protein modifying EglN prolyl hydroxylases. Importantly, the transforming activity of R-2HG was reversible, suggesting that therapeutic strategies focusing on inhibition of R-2HG production or inhibition of EglN prolyl hydroxylases merit further investigation. A metabolite specific to certain cancers, and of therapeutic interest, exists in two forms, only one of which is oncogenic. Mutations in IDH1 and IDH2, the genes coding for isocitrate dehydrogenases 1 and 2, are common in several human cancers, including leukemias, and result in overproduction of the (R)-enantiomer of 2-hydroxyglutarate [(R)-2HG]. Elucidation of the role of IDH mutations and (R)-2HG in leukemogenesis has been hampered by a lack of appropriate cell-based models. Here, we show that a canonical IDH1 mutant, IDH1 R132H, promotes cytokine independence and blocks differentiation in hematopoietic cells. These effects can be recapitulated by (R)-2HG, but not (S)-2HG, despite the fact that (S)-2HG more potently inhibits enzymes, such as the 5′-methylcytosine hydroxylase TET2, that have previously been linked to the pathogenesis of IDH mutant tumors. We provide evidence that this paradox relates to the ability of (S)-2HG, but not (R)-2HG, to inhibit the EglN prolyl hydroxylases. Additionally, we show that transformation by (R)-2HG is reversible.


The New England Journal of Medicine | 1990

Mutations within the Rhodopsin Gene in Patients with Autosomal Dominant Retinitis Pigmentosa

Thaddeus P. Dryja; Terri L. McGee; Lauri B. Hahn; Glenn S. Cowley; Jane E. Olsson; Elias Reichel; Michael A. Sandberg; Eliot L. Berson

BACKGROUND Night blindness is an early symptom of retinitis pigmentosa. The rod photoreceptors are responsible for night vision and use rhodopsin as the photosensitive pigment. METHODS AND RESULTS We found three mutations in the human rhodopsin gene; each occurred exclusively in the affected members of some families with autosomal dominant retinitis pigmentosa. Two mutations were C-to-T transitions involving separate nucleotides of codon 347; the third was a C-to-G transversion in codon 58. Each mutation corresponded to a change in one amino acid residue in the rhodopsin molecule. None of these mutations were found in 106 unrelated normal subjects who served as controls. When the incidence of these three mutations was added to that of a previously reported mutation involving codon 23, 27 of 150 unrelated patients with autosomal dominant retinitis pigmentosa (18 percent) were found to carry one of these four defects in the rhodopsin gene. All 27 patients had abnormal rod function on monitoring of their electroretinograms. It appears that patients with the mutation involving codon 23 probably descend from a single ancestor. CONCLUSIONS In some patients with autosomal dominant retinitis pigmentosa, the disease is caused by one of a variety of mutations of the rhodopsin gene.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Systematic investigation of genetic vulnerabilities across cancer cell lines reveals lineage-specific dependencies in ovarian cancer

Hiu Wing Cheung; Glenn S. Cowley; Barbara A. Weir; Jesse S. Boehm; Scott F. Rusin; Justine A. Scott; Alexandra East; Levi D. Ali; Patrick H. Lizotte; Terence C. Wong; Guozhi Jiang; Jessica Hsiao; Craig H. Mermel; Gad Getz; Jordi Barretina; Shuba Gopal; Pablo Tamayo; Joshua Gould; Aviad Tsherniak; Nicolas Stransky; Biao Luo; Yin Ren; Ronny Drapkin; Sangeeta N. Bhatia; Jill P. Mesirov; Levi A. Garraway; Matthew Meyerson; Eric S. Lander; David E. Root; William C. Hahn

A comprehensive understanding of the molecular vulnerabilities of every type of cancer will provide a powerful roadmap to guide therapeutic approaches. Efforts such as The Cancer Genome Atlas Project will identify genes with aberrant copy number, sequence, or expression in various cancer types, providing a survey of the genes that may have a causal role in cancer. A complementary approach is to perform systematic loss-of-function studies to identify essential genes in particular cancer cell types. We have begun a systematic effort, termed Project Achilles, aimed at identifying genetic vulnerabilities across large numbers of cancer cell lines. Here, we report the assessment of the essentiality of 11,194 genes in 102 human cancer cell lines. We show that the integration of these functional data with information derived from surveying cancer genomes pinpoints known and previously undescribed lineage-specific dependencies across a wide spectrum of cancers. In particular, we found 54 genes that are specifically essential for the proliferation and viability of ovarian cancer cells and also amplified in primary tumors or differentially overexpressed in ovarian cancer cell lines. One such gene, PAX8, is focally amplified in 16% of high-grade serous ovarian cancers and expressed at higher levels in ovarian tumors. Suppression of PAX8 selectively induces apoptotic cell death of ovarian cancer cells. These results identify PAX8 as an ovarian lineage-specific dependency. More generally, these observations demonstrate that the integration of genome-scale functional and structural studies provides an efficient path to identify dependencies of specific cancer types on particular genes and pathways.


Cancer Discovery | 2013

A Genome-Scale RNA Interference Screen Implicates NF1 Loss in Resistance to RAF Inhibition

Steven Whittaker; Jean-Philippe Theurillat; Eliezer M. Van Allen; Nikhil Wagle; Jessica Hsiao; Glenn S. Cowley; Dirk Schadendorf; David E. Root; Levi A. Garraway

RAF inhibitors such as vemurafenib and dabrafenib block BRAF-mediated cell proliferation and achieve meaningful clinical benefit in the vast majority of patients with BRAF(V600E)-mutant melanoma. However, some patients do not respond to this regimen, and nearly all progress to therapeutic resistance. We used a pooled RNA interference screen targeting more than 16,500 genes to discover loss-of-function events that could drive resistance to RAF inhibition. The highest ranking gene was NF1, which encodes neurofibromin, a tumor suppressor that inhibits RAS activity. NF1 loss mediates resistance to RAF and mitogen-activated protein kinase (MAPK) kinase kinase (MEK) inhibitors through sustained MAPK pathway activation. However, cells lacking NF1 retained sensitivity to the irreversible RAF inhibitor AZ628 and an ERK inhibitor. NF1 mutations were observed in BRAF-mutant tumor cells that are intrinsically resistant to RAF inhibition and in melanoma tumors obtained from patients exhibiting resistance to vemurafenib, thus showing the clinical potential for NF1-driven resistance to RAF/MEK-targeted therapies.


Scientific Data | 2014

Parallel genome-scale loss of function screens in 216 cancer cell lines for the identification of context-specific genetic dependencies

Glenn S. Cowley; Barbara A. Weir; Francisca Vazquez; Pablo Tamayo; Justine A. Scott; Scott F. Rusin; Alexandra East-Seletsky; Levi D. Ali; William F.J. Gerath; Sarah E. Pantel; Patrick H. Lizotte; Guozhi Jiang; Jessica Hsiao; Aviad Tsherniak; Elizabeth Dwinell; Simon Aoyama; Michael Okamoto; William F. Harrington; Ellen Gelfand; Thomas M. Green; Mark J. Tomko; Shuba Gopal; Terrence C. Wong; Hubo Li; Sara Howell; Nicolas Stransky; Ted Liefeld; Dongkeun Jang; Jonathan Bistline; Barbara Hill Meyers

Using a genome-scale, lentivirally delivered shRNA library, we performed massively parallel pooled shRNA screens in 216 cancer cell lines to identify genes that are required for cell proliferation and/or viability. Cell line dependencies on 11,000 genes were interrogated by 5 shRNAs per gene. The proliferation effect of each shRNA in each cell line was assessed by transducing a population of 11M cells with one shRNA-virus per cell and determining the relative enrichment or depletion of each of the 54,000 shRNAs after 16 population doublings using Next Generation Sequencing. All the cell lines were screened using standardized conditions to best assess differential genetic dependencies across cell lines. When combined with genomic characterization of these cell lines, this dataset facilitates the linkage of genetic dependencies with specific cellular contexts (e.g., gene mutations or cell lineage). To enable such comparisons, we developed and provided a bioinformatics tool to identify linear and nonlinear correlations between these features.


Cancer Discovery | 2016

Genomic Copy Number Dictates a Gene-Independent Cell Response to CRISPR/Cas9 Targeting

Andrew J. Aguirre; Robin M. Meyers; Barbara A. Weir; Francisca Vazquez; Cheng-Zhong Zhang; Uri Ben-David; April Cook; Gavin Ha; William F. Harrington; Mihir Doshi; Maria Kost-Alimova; Stanley Gill; Han Xu; Levi D. Ali; Guozhi Jiang; Sasha Pantel; Yenarae Lee; Amy Goodale; Andrew D. Cherniack; Coyin Oh; Gregory V. Kryukov; Glenn S. Cowley; Levi A. Garraway; Kimberly Stegmaier; Charles W. M. Roberts; Todd R. Golub; Matthew Meyerson; David E. Root; Aviad Tsherniak; William C. Hahn

UNLABELLED The CRISPR/Cas9 system enables genome editing and somatic cell genetic screens in mammalian cells. We performed genome-scale loss-of-function screens in 33 cancer cell lines to identify genes essential for proliferation/survival and found a strong correlation between increased gene copy number and decreased cell viability after genome editing. Within regions of copy-number gain, CRISPR/Cas9 targeting of both expressed and unexpressed genes, as well as intergenic loci, led to significantly decreased cell proliferation through induction of a G2 cell-cycle arrest. By examining single-guide RNAs that map to multiple genomic sites, we found that this cell response to CRISPR/Cas9 editing correlated strongly with the number of target loci. These observations indicate that genome targeting by CRISPR/Cas9 elicits a gene-independent antiproliferative cell response. This effect has important practical implications for the interpretation of CRISPR/Cas9 screening data and confounds the use of this technology for the identification of essential genes in amplified regions. SIGNIFICANCE We found that the number of CRISPR/Cas9-induced DNA breaks dictates a gene-independent antiproliferative response in cells. These observations have practical implications for using CRISPR/Cas9 to interrogate cancer gene function and illustrate that cancer cells are highly sensitive to site-specific DNA damage, which may provide a path to novel therapeutic strategies. Cancer Discov; 6(8); 914-29. ©2016 AACR.See related commentary by Sheel and Xue, p. 824See related article by Munoz et al., p. 900This article is highlighted in the In This Issue feature, p. 803.


Cell | 2012

Cancer vulnerabilities unveiled by genomic loss

Deepak Nijhawan; Travis I. Zack; Yin Ren; Matthew R. Strickland; Rebecca Lamothe; Steven E. Schumacher; Aviad Tsherniak; Henrike C. Besche; Joseph Rosenbluh; Shyemaa Shehata; Glenn S. Cowley; Barbara A. Weir; Alfred L. Goldberg; Jill P. Mesirov; David E. Root; Sangeeta N. Bhatia; Rameen Beroukhim; William C. Hahn

Due to genome instability, most cancers exhibit loss of regions containing tumor suppressor genes and collateral loss of other genes. To identify cancer-specific vulnerabilities that are the result of copy number losses, we performed integrated analyses of genome-wide copy number and RNAi profiles and identified 56 genes for which gene suppression specifically inhibited the proliferation of cells harboring partial copy number loss of that gene. These CYCLOPS (copy number alterations yielding cancer liabilities owing to partial loss) genes are enriched for spliceosome, proteasome, and ribosome components. One CYCLOPS gene, PSMC2, encodes an essential member of the 19S proteasome. Normal cells express excess PSMC2, which resides in a complex with PSMC1, PSMD2, and PSMD5 and acts as a reservoir protecting cells from PSMC2 suppression. Cells harboring partial PSMC2 copy number loss lack this complex and die after PSMC2 suppression. These observations define a distinct class of cancer-specific liabilities resulting from genome instability.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Complementary genomic approaches highlight the PI3K/mTOR pathway as a common vulnerability in osteosarcoma

Jennifer A. Perry; Adam Kiezun; Peter Tonzi; Eliezer M. Van Allen; Scott L. Carter; Sylvan C. Baca; Glenn S. Cowley; Ami S. Bhatt; Esther Rheinbay; Chandra Sekhar Pedamallu; Elena Helman; Amaro Taylor-Weiner; Aaron McKenna; David S. DeLuca; Michael S. Lawrence; Lauren Ambrogio; Carrie Sougnez; Andrey Sivachenko; Loren D. Walensky; Nikhil Wagle; Jaume Mora; Carmen Torres; Cinzia Lavarino; Simone dos Santos Aguiar; José Andrés Yunes; Silvia Regina Brandalise; Gabriela Elisa Mercado-Celis; Jorge Melendez-Zajgla; Rocio Cardenas-Cardos; Liliana Velasco-Hidalgo

Significance We present, to our knowledge, the first comprehensive next-generation sequencing of osteosarcoma in combination with a functional genomic screen in a genetically defined mouse model of osteosarcoma. Our data provide a strong rationale for targeting the phosphatidylinositol 3-kinase/mammalian target of rapamycin pathway in osteosarcoma and a foundation for rational clinical trial design. These findings present an immediate clinical opportunity because multiple inhibitors of this pathway are currently in clinical trials. Osteosarcoma is the most common primary bone tumor, yet there have been no substantial advances in treatment or survival in three decades. We examined 59 tumor/normal pairs by whole-exome, whole-genome, and RNA-sequencing. Only the TP53 gene was mutated at significant frequency across all samples. The mean nonsilent somatic mutation rate was 1.2 mutations per megabase, and there was a median of 230 somatic rearrangements per tumor. Complex chains of rearrangements and localized hypermutation were detected in almost all cases. Given the intertumor heterogeneity, the extent of genomic instability, and the difficulty in acquiring a large sample size in a rare tumor, we used several methods to identify genomic events contributing to osteosarcoma survival. Pathway analysis, a heuristic analytic algorithm, a comparative oncology approach, and an shRNA screen converged on the phosphatidylinositol 3-kinase/mammalian target of rapamycin (PI3K/mTOR) pathway as a central vulnerability for therapeutic exploitation in osteosarcoma. Osteosarcoma cell lines are responsive to pharmacologic and genetic inhibition of the PI3K/mTOR pathway both in vitro and in vivo.

Collaboration


Dive into the Glenn S. Cowley's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Benjamin L. Ebert

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge