Network


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

Hotspot


Dive into the research topics where David E. Root is active.

Publication


Featured researches published by David E. Root.


Nature | 2011

lincRNAs act in the circuitry controlling pluripotency and differentiation

Mitchell Guttman; Julie Donaghey; Bryce W. Carey; Manuel Garber; Jennifer K. Grenier; Glen Munson; Geneva Young; Robert A. Ach; Laurakay Bruhn; Xiaoping Yang; Ido Amit; Alexander Meissner; Aviv Regev; John L. Rinn; David E. Root; Eric S. Lander

Although thousands of large intergenic non-coding RNAs (lincRNAs) have been identified in mammals, few have been functionally characterized, leading to debate about their biological role. To address this, we performed loss-of-function studies on most lincRNAs expressed in mouse embryonic stem (ES) cells and characterized the effects on gene expression. Here we show that knockdown of lincRNAs has major consequences on gene expression patterns, comparable to knockdown of well-known ES cell regulators. Notably, lincRNAs primarily affect gene expression in trans. Knockdown of dozens of lincRNAs causes either exit from the pluripotent state or upregulation of lineage commitment programs. We integrate lincRNAs into the molecular circuitry of ES cells and show that lincRNA genes are regulated by key transcription factors and that lincRNA transcripts bind to multiple chromatin regulatory proteins to affect shared gene expression programs. Together, the results demonstrate that lincRNAs have key roles in the circuitry controlling ES cell state.


Nature | 2009

Systematic RNA interference reveals that oncogenic KRAS -driven cancers require TBK1

David A. Barbie; Pablo Tamayo; Jesse S. Boehm; So Young Kim; Susan E. Moody; Ian F. Dunn; Anna C. Schinzel; Peter Sandy; Etienne Meylan; Claudia Scholl; Stefan Fröhling; Edmond M. Chan; Martin L. Sos; Kathrin Michel; Craig H. Mermel; Serena J. Silver; Barbara A. Weir; Jan H. Reiling; Qing Sheng; Piyush B. Gupta; Raymond C. Wadlow; Hanh Le; Ben S. Wittner; Sridhar Ramaswamy; David M. Livingston; David M. Sabatini; Matthew Meyerson; Roman K. Thomas; Eric S. Lander; Jill P. Mesirov

The proto-oncogene KRAS is mutated in a wide array of human cancers, most of which are aggressive and respond poorly to standard therapies. Although the identification of specific oncogenes has led to the development of clinically effective, molecularly targeted therapies in some cases, KRAS has remained refractory to this approach. A complementary strategy for targeting KRAS is to identify gene products that, when inhibited, result in cell death only in the presence of an oncogenic allele. Here we have used systematic RNA interference to detect synthetic lethal partners of oncogenic KRAS and found that the non-canonical IκB kinase TBK1 was selectively essential in cells that contain mutant KRAS. Suppression of TBK1 induced apoptosis specifically in human cancer cell lines that depend on oncogenic KRAS expression. In these cells, TBK1 activated NF-κB anti-apoptotic signals involving c-Rel and BCL-XL (also known as BCL2L1) that were essential for survival, providing mechanistic insights into this synthetic lethal interaction. These observations indicate that TBK1 and NF-κB signalling are essential in KRAS mutant tumours, and establish a general approach for the rational identification of co-dependent pathways in cancer.


Cell | 2009

Synthetic lethal interaction between oncogenic KRAS dependency and STK33 suppression in human cancer cells.

Claudia Scholl; Stefan Fröhling; Ian F. Dunn; Anna C. Schinzel; David A. Barbie; So Young Kim; Serena J. Silver; Pablo Tamayo; Raymond C. Wadlow; Sridhar Ramaswamy; Konstanze Döhner; Lars Bullinger; Peter Sandy; Jesse S. Boehm; David E. Root; Tyler Jacks; William C. Hahn; D. Gary Gilliland

An alternative to therapeutic targeting of oncogenes is to perform synthetic lethality screens for genes that are essential only in the context of specific cancer-causing mutations. We used high-throughput RNA interference (RNAi) to identify synthetic lethal interactions in cancer cells harboring mutant KRAS, the most commonly mutated human oncogene. We find that cells that are dependent on mutant KRAS exhibit sensitivity to suppression of the serine/threonine kinase STK33 irrespective of tissue origin, whereas STK33 is not required by KRAS-independent cells. STK33 promotes cancer cell viability in a kinase activity-dependent manner by regulating the suppression of mitochondrial apoptosis mediated through S6K1-induced inactivation of the death agonist BAD selectively in mutant KRAS-dependent cells. These observations identify STK33 as a target for treatment of mutant KRAS-driven cancers and demonstrate the potential of RNAi screens for discovering functional dependencies created by oncogenic mutations that may enable therapeutic intervention for cancers with undruggable genetic alterations.


Nature | 2008

CDK8 is a colorectal cancer oncogene that regulates β-catenin activity

Ron Firestein; Adam J. Bass; So Young Kim; Ian F. Dunn; Serena J. Silver; Isil Guney; Ellen Freed; Azra H. Ligon; Natalie Vena; Shuji Ogino; Milan G. Chheda; Pablo Tamayo; Stephen Finn; Yashaswi Shrestha; Jesse S. Boehm; Supriya K Jain; Emeric Bojarski; Craig H. Mermel; Jordi Barretina; Jennifer A. Chan; José Baselga; Josep Tabernero; David E. Root; Charles S. Fuchs; Massimo Loda; Ramesh A. Shivdasani; Matthew Meyerson; William C. Hahn

Aberrant activation of the canonical WNT/β-catenin pathway occurs in almost all colorectal cancers and contributes to their growth, invasion and survival. Although dysregulated β-catenin activity drives colon tumorigenesis, further genetic perturbations are required to elaborate full malignant transformation. To identify genes that both modulate β-catenin activity and are essential for colon cancer cell proliferation, we conducted two loss-of-function screens in human colon cancer cells and compared genes identified in these screens with an analysis of copy number alterations in colon cancer specimens. One of these genes, CDK8, which encodes a member of the mediator complex, is located at 13q12.13, a region of recurrent copy number gain in a substantial fraction of colon cancers. Here we show that the suppression of CDK8 expression inhibits proliferation in colon cancer cells characterized by high levels of CDK8 and β-catenin hyperactivity. CDK8 kinase activity was necessary for β-catenin-driven transformation and for expression of several β-catenin transcriptional targets. Together these observations suggest that therapeutic interventions targeting CDK8 may confer a clinical benefit in β-catenin-driven malignancies.


Nature Methods | 2006

Genome-scale loss-of-function screening with a lentiviral RNAi library.

David E. Root; Nir Hacohen; William C. Hahn; Eric S. Lander; David M. Sabatini

The discovery that RNA interference (RNAi) is functional in mammalian cells led us to form The RNAi Consortium (TRC) with the goal of enabling large-scale loss-of-function screens through the development of genome-scale RNAi libraries and methodologies for their use. These resources form the basis of a loss-of-function screening platform created at the Broad Institute. Our human and mouse libraries currently contain >135,000 lentiviral clones targeting 27,000 genes. Initial screening efforts have demonstrated that these libraries and methods are practical and powerful tools for high-throughput lentivirus RNAi screens.


Nature Methods | 2011

A public genome-scale lentiviral expression library of human ORFs

Xiaoping Yang; Jesse S. Boehm; Xinping Yang; Kourosh Salehi-Ashtiani; Tong Hao; Yun Shen; Rakela Lubonja; Sapana Thomas; Ozan Alkan; Tashfeen Bhimdi; Thomas M. Green; Cory M. Johannessen; Serena J. Silver; Cindy Nguyen; Ryan R. Murray; Haley Hieronymus; Dawit Balcha; Changyu Fan; Chenwei Lin; Lila Ghamsari; Marc Vidal; William C. Hahn; David E. Hill; David E. Root

Functional characterization of the human genome requires tools for systematically modulating gene expression in both loss-of-function and gain-of-function experiments. We describe the production of a sequence-confirmed, clonal collection of over 16,100 human open-reading frames (ORFs) encoded in a versatile Gateway vector system. Using this ORFeome resource, we created a genome-scale expression collection in a lentiviral vector, thereby enabling both targeted experiments and high-throughput screens in diverse cell types.


Cancer Cell | 2009

Proteomic and genetic approaches identify Syk as an AML target.

Cynthia K. Hahn; Jacob E. Berchuck; Kenneth N. Ross; Rose M. Kakoza; Karl R. Clauser; Anna C. Schinzel; Linda Ross; Ilene Galinsky; Tina N. Davis; Serena J. Silver; David E. Root; Richard Stone; Daniel J. DeAngelo; Martin Carroll; William C. Hahn; Steven A. Carr; Todd R. Golub; Andrew L. Kung; Kimberly Stegmaier

Cell-based screening can facilitate the rapid identification of compounds inducing complex cellular phenotypes. Advancing a compound toward the clinic, however, generally requires the identification of precise mechanisms of action. We previously found that epidermal growth factor receptor (EGFR) inhibitors induce acute myeloid leukemia (AML) differentiation via a non-EGFR mechanism. In this report, we integrated proteomic and RNAi-based strategies to identify their off-target, anti-AML mechanism. These orthogonal approaches identified Syk as a target in AML. Genetic and pharmacological inactivation of Syk with a drug in clinical trial for other indications promoted differentiation of AML cells and attenuated leukemia growth in vivo. These results demonstrate the power of integrating diverse chemical, proteomic, and genomic screening approaches to identify therapeutic strategies for cancer.


Science Translational Medicine | 2012

Targeted Tumor-Penetrating siRNA Nanocomplexes for Credentialing the Ovarian Cancer Oncogene ID4

Yin Ren; Hiu Wing Cheung; Amit Agrawal; Glenn S. Cowley; Barbara A. Weir; Jesse S. Boehm; Pablo Tamayo; Alison M. Karst; Joyce Liu; Michelle S. Hirsch; Jill P. Mesirov; Ronny Drapkin; David E. Root; Justin H. Lo; Valentina Fogal; Erkki Ruoslahti; William C. Hahn; Sangeeta N. Bhatia

Tumor-penetrating siRNA nanocomplexes credential ID4 as a therapeutic oncogene target in human ovarian cancer. Nanotechnology Sets Sights on Ovarian Tumors In the world of anticancer research, targeting tumor cells is one challenge; penetrating the cells to deliver therapeutics is another. The combination of specific targeting and efficient delivery is the clinical holy grail, wherein optimization of this approach could lead to highly effective cancer therapy in humans. Ren et al. have now developed a nanotechnology platform that allows for just that: targeted intracellular delivery of RNA-based therapeutics to ovarian cancer cells, which halts the oncogenic activity of a potent gene, in this case ID4. In a screen of overexpressed and essential genes in human ovarian cancer, the authors first identified a potential oncogene, ID4. They then confirmed ID4 tumorigenicity and mechanism in vitro in cell lines. After confirming that ID4 was an oncogene, Ren et al. reasoned that “silencing” the gene using small interfering RNA (siRNA) would prevent tumor growth in vivo. The trick was to make sure the siRNA could cross the cell membrane to exert its silencing effects. To accomplish this, the authors designed a tumor-penetrating nanocomplex (TPN) that could not only bind a protein overexpressed on the surface of human cancer cells but also pass through the membrane via a cell-penetrating peptide. Once inside the cells, the TPN could release the siRNA directed against ID4. Tumor homing was confirmed in mouse models of human melanoma and ovarian cancer. In mice harboring subcutaneous ovarian tumors, TPN/siRNA decreased ID4 expression by up to 90% and suppressed tumor growth by 82%. In mice bearing disseminated intra-abdominal tumors, TPN/siRNA allowed 80% of the animals to live 60 or more days. Control treatments did not prevent tumor growth in either study, and the TPN/siRNA therapy did not elicit any immunogenic side effects. Locked and loaded with siRNA, these TPNs are ready to target and kill cancer cells. The authors envision this to be a platform for credentialing oncogenes and for validating RNA interference in preclinical models before development of therapeutics. However, before moving this TPN/siRNA approach to patients, some additional preclinical optimization is necessary, including pharmacokinetics, testing in human cancer models, and increasing siRNA efficiency at lower doses. The comprehensive characterization of a large number of cancer genomes will eventually lead to a compendium of genetic alterations in specific cancers. Unfortunately, the number and complexity of identified alterations complicate endeavors to identify biologically relevant mutations critical for tumor maintenance because many of these targets are not amenable to manipulation by small molecules or antibodies. RNA interference provides a direct way to study putative cancer targets; however, specific delivery of therapeutics to the tumor parenchyma remains an intractable problem. We describe a platform for the discovery and initial validation of cancer targets, composed of a systematic effort to identify amplified and essential genes in human cancer cell lines and tumors partnered with a novel modular delivery technology. We developed a tumor-penetrating nanocomplex (TPN) that comprised small interfering RNA (siRNA) complexed with a tandem tumor-penetrating and membrane-translocating peptide, which enabled the specific delivery of siRNA deep into the tumor parenchyma. We used TPN in vivo to evaluate inhibitor of DNA binding 4 (ID4) as a novel oncogene. Treatment of ovarian tumor–bearing mice with ID4-specific TPN suppressed growth of established tumors and significantly improved survival. These observations not only credential ID4 as an oncogene in 32% of high-grade ovarian cancers but also provide a framework for the identification, validation, and understanding of potential therapeutic cancer targets.


Cancer Cell | 2013

In Vivo RNAi Screening Identifies a Leukemia-Specific Dependence on Integrin Beta 3 Signaling

Peter Miller; Fatima Al-Shahrour; Kimberly A. Hartwell; Lisa P. Chu; Marcus Järås; Rishi V. Puram; Alexandre Puissant; Kevin P. Callahan; John M. Ashton; Marie McConkey; Luke Poveromo; Glenn S. Cowley; Michael G. Kharas; Myriam Labelle; Sebastian Shterental; Joji Fujisaki; Lev Silberstein; Gabriela Alexe; Muhammad A. Al-Hajj; Christopher A. Shelton; Scott A. Armstrong; David E. Root; David T. Scadden; Richard O. Hynes; Siddhartha Mukherjee; Kimberly Stegmaier; Craig T. Jordan; Benjamin L. Ebert

We used an inxa0vivo small hairpin RNA (shRNA) screening approach to identify genes that are essential for MLL-AF9 acute myeloid leukemia (AML). We found that Integrin Beta 3 (Itgb3) is essential for murine leukemia cells inxa0vivo and for human leukemia cells in xenotransplantation studies. In leukemia cells, Itgb3 knockdown impaired homing, downregulated LSC transcriptional programs, and induced differentiation via the intracellular kinase Syk. In contrast, loss of Itgb3 in normal hematopoietic stem and progenitor cells did not affect engraftment, reconstitution, or differentiation. Finally, using an Itgb3 knockout mouse model, we confirmed that Itgb3 is dispensable for normal hematopoiesis but is required for leukemogenesis. Our results establish the significance of the Itgb3 signaling pathway as a potential therapeutic target in AML.


Genome Research | 2013

ATARiS: Computational quantification of gene suppression phenotypes from multisample RNAi screens

Diane D. Shao; Aviad Tsherniak; Shuba Gopal; Barbara A. Weir; Pablo Tamayo; Nicolas Stransky; Steven E. Schumacher; Travis I. Zack; Rameen Beroukhim; Levi A. Garraway; Adam A. Margolin; David E. Root; William C. Hahn; Jill P. Mesirov

Genome-scale RNAi libraries enable the systematic interrogation of gene function. However, the interpretation of RNAi screens is complicated by the observation that RNAi reagents designed to suppress the mRNA transcripts of the same gene often produce a spectrum of phenotypic outcomes due to differential on-target gene suppression or perturbation of off-target transcripts. Here we present a computational method, Analytic Technique for Assessment of RNAi by Similarity (ATARiS), that takes advantage of patterns in RNAi data across multiple samples in order to enrich for RNAi reagents whose phenotypic effects relate to suppression of their intended targets. By summarizing only such reagent effects for each gene, ATARiS produces quantitative, gene-level phenotype values, which provide an intuitive measure of the effect of gene suppression in each sample. This method is robust for data sets that contain as few as 10 samples and can be used to analyze screens of any number of targeted genes. We used this analytic approach to interrogate RNAi data derived from screening more than 100 human cancer cell lines and identified HNF1B as a transforming oncogene required for the survival of cancer cells that harbor HNF1B amplifications. ATARiS is publicly available at http://broadinstitute.org/ataris.

Collaboration


Dive into the David E. Root's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pablo Tamayo

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge