Network


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

Hotspot


Dive into the research topics where Michael R. Schlabach is active.

Publication


Featured researches published by Michael R. Schlabach.


Cell | 2009

A Genome-wide RNAi Screen Identifies Multiple Synthetic Lethal Interactions with the Ras Oncogene

Ji Luo; Michael J. Emanuele; Danan Li; Chad J. Creighton; Michael R. Schlabach; Thomas F. Westbrook; Kwok-Kin Wong; Stephen J. Elledge

Oncogenic mutations in the small GTPase Ras are highly prevalent in cancer, but an understanding of the vulnerabilities of these cancers is lacking. We undertook a genome-wide RNAi screen to identify synthetic lethal interactions with the KRAS oncogene. We discovered a diverse set of proteins whose depletion selectively impaired the viability of Ras mutant cells. Among these we observed a strong enrichment for genes with mitotic functions. We describe a pathway involving the mitotic kinase PLK1, the anaphase-promoting complex/cyclosome, and the proteasome that, when inhibited, results in prometaphase accumulation and the subsequent death of Ras mutant cells. Gene expression analysis indicates that reduced expression of genes in this pathway correlates with increased survival of patients bearing tumors with a Ras transcriptional signature. Our results suggest a previously underappreciated role for Ras in mitotic progression and demonstrate a pharmacologically tractable pathway for the potential treatment of cancers harboring Ras mutations.


Nature Genetics | 2005

Second-generation shRNA libraries covering the mouse and human genomes

Jose M. Silva; Mamie Z. Li; Ken Chang; Wei Ge; Michael C. Golding; Richard J. Rickles; Despina Siolas; Guang Hu; Patrick J. Paddison; Michael R. Schlabach; Nihar Sheth; Jeff Bradshaw; Julia Burchard; Amit A. Kulkarni; Guy Cavet; Ravi Sachidanandam; W. Richard McCombie; Michele A. Cleary; Stephen J. Elledge; Gregory J. Hannon

Loss-of-function phenotypes often hold the key to understanding the connections and biological functions of biochemical pathways. We and others previously constructed libraries of short hairpin RNAs that allow systematic analysis of RNA interference–induced phenotypes in mammalian cells. Here we report the construction and validation of second-generation short hairpin RNA expression libraries designed using an increased knowledge of RNA interference biochemistry. These constructs include silencing triggers designed to mimic a natural microRNA primary transcript, and each target sequence was selected on the basis of thermodynamic criteria for optimal small RNA performance. Biochemical and phenotypic assays indicate that the new libraries are substantially improved over first-generation reagents. We generated large-scale-arrayed, sequence-verified libraries comprising more than 140,000 second-generation short hairpin RNA expression plasmids, covering a substantial fraction of all predicted genes in the human and mouse genomes. These libraries are available to the scientific community.


Cell | 2005

A Genetic Screen for Candidate Tumor Suppressors Identifies REST

Thomas F. Westbrook; Eric Martin; Michael R. Schlabach; Yumei Leng; Anthony C. Liang; Bin Feng; Jean Zhao; Thomas M. Roberts; Gail Mandel; Gregory J. Hannon; Ronald A. DePinho; Lynda Chin; Stephen J. Elledge

Tumorigenesis is a multistep process characterized by a myriad of genetic and epigenetic alterations. Identifying the causal perturbations that confer malignant transformation is a central goal in cancer biology. Here we report an RNAi-based genetic screen for genes that suppress transformation of human mammary epithelial cells. We identified genes previously implicated in proliferative control and epithelial cell function including two established tumor suppressors, TGFBR2 and PTEN. In addition, we uncovered a previously unrecognized tumor suppressor role for REST/NRSF, a transcriptional repressor of neuronal gene expression. Array-CGH analysis identified REST as a frequent target of deletion in colorectal cancer. Furthermore, we detect a frameshift mutation of the REST gene in colorectal cancer cells that encodes a dominantly acting truncation capable of transforming epithelial cells. Cells lacking REST exhibit increased PI(3)K signaling and are dependent upon this pathway for their transformed phenotype. These results implicate REST as a human tumor suppressor and provide a novel approach to identifying candidate genes that suppress the development of human cancer.


Science | 2008

Cancer Proliferation Gene Discovery Through Functional Genomics

Michael R. Schlabach; Ji Luo; Nicole L. Solimini; Guang Hu; Qikai Xu; Mamie Z. Li; Zhenming Zhao; Agata Smogorzewska; Mathew E. Sowa; Xiaolu L. Ang; Thomas F. Westbrook; Anthony C. Liang; Kenneth Chang; Jennifer A. Hackett; J. Wade Harper; Gregory J. Hannon; Stephen J. Elledge

Retroviral short hairpin RNA (shRNA)–mediated genetic screens in mammalian cells are powerful tools for discovering loss-of-function phenotypes. We describe a highly parallel multiplex methodology for screening large pools of shRNAs using half-hairpin barcodes for microarray deconvolution. We carried out dropout screens for shRNAs that affect cell proliferation and viability in cancer cells and normal cells. We identified many shRNAs to be antiproliferative that target core cellular processes, such as the cell cycle and protein translation, in all cells examined. Moreover, we identified genes that are selectively required for proliferation and survival in different cell lines. Our platform enables rapid and cost-effective genome-wide screens to identify cancer proliferation and survival genes for target discovery. Such efforts are complementary to the Cancer Genome Atlas and provide an alternative functional view of cancer cells.


Science | 2008

Profiling essential genes in human mammary cells by multiplex RNAi screening

Jose M. Silva; Krista Marran; Joel S. Parker; Javier Silva; Michael C. Golding; Michael R. Schlabach; Stephen J. Elledge; Gregory J. Hannon; Kenneth Chang

By virtue of their accumulated genetic alterations, tumor cells may acquire vulnerabilities that create opportunities for therapeutic intervention. We have devised a massively parallel strategy for screening short hairpin RNA (shRNA) collections for stable loss-of-function phenotypes. We assayed from 6000 to 20,000 shRNAs simultaneously to identify genes important for the proliferation and survival of five cell lines derived from human mammary tissue. Lethal shRNAs common to these cell lines targeted many known cell-cycle regulatory networks. Cell line–specific sensitivities to suppression of protein complexes and biological pathways also emerged, and these could be validated by RNA interference (RNAi) and pharmacologically. These studies establish a practical platform for genome-scale screening of complex phenotypes in mammalian cells and demonstrate that RNAi can be used to expose genotype-specific sensitivities.


Science | 2012

A SUMOylation-Dependent Transcriptional Subprogram Is Required for Myc-Driven Tumorigenesis

Jessica D. Kessler; Kristopher T. Kahle; Tingting Sun; Kristen L. Meerbrey; Michael R. Schlabach; Earlene M. Schmitt; Samuel O. Skinner; Qikai Xu; Mamie Z. Li; Zachary C. Hartman; Mitchell Rao; Peng Yu; Rocio Dominguez-Vidana; Anthony C. Liang; Nicole L. Solimini; Ronald J. Bernardi; Bing Yu; Tiffany Hsu; Ido Golding; Ji Luo; C. Kent Osborne; Chad J. Creighton; Susan G. Hilsenbeck; Rachel Schiff; Chad A. Shaw; Stephen J. Elledge; Thomas F. Westbrook

Taking the Myc Despite nearly 30 years of research into the mechanisms by which Myc oncogene dysregulation contributes to tumorigenesis, there are still no effective therapies that inhibit Myc activity. Kessler et al. (p. 348, published online 8 December; see the Perspective by Evan) searched for gene products that support Myc-driven tumorigenesis. One pharmacologically tractable target that emerged from the screen was the SUMO-activating enzyme complex SAE1/2, which catalyzes a posttranslational modification (SUMOylation) that alters protein behavior and function. SUMOylation was found to control the Myc transcriptional response, and its inhibition caused mitotic defects and apoptosis in Myc-dependent breast cancer cells. An RNA interference screen identifies a “druggable” enzyme whose inhibition halts tumor cell growth. Myc is an oncogenic transcription factor frequently dysregulated in human cancer. To identify pathways supporting the Myc oncogenic program, we used a genome-wide RNA interference screen to search for Myc–synthetic lethal genes and uncovered a role for the SUMO-activating enzyme (SAE1/2). Loss of SAE1/2 enzymatic activity drives synthetic lethality with Myc. Inactivation of SAE2 leads to mitotic catastrophe and cell death upon Myc hyperactivation. Mechanistically, SAE2 inhibition switches a transcriptional subprogram of Myc from activated to repressed. A subset of these SUMOylation-dependent Myc switchers (SMS genes) is required for mitotic spindle function and to support the Myc oncogenic program. SAE2 is required for growth of Myc-dependent tumors in mice, and gene expression analyses of Myc-high human breast cancers suggest that low SAE1 and SAE2 abundance in the tumors correlates with longer metastasis-free survival of the patients. Thus, inhibition of SUMOylation may merit investigation as a possible therapy for Myc-driven human cancers.


Science | 2012

Recurrent Hemizygous Deletions in Cancers May Optimize Proliferative Potential

Nicole L. Solimini; Qikai Xu; Craig H. Mermel; Anthony C. Liang; Michael R. Schlabach; Ji Luo; Anna E. Burrows; Anthony N. Anselmo; Andrea L. Bredemeyer; Mamie Z. Li; Rameen Beroukhim; Matthew Meyerson; Stephen J. Elledge

Cancer Gene Islands Human tumors are riddled with genomic alterations that rearrange, remove, amplify, or otherwise disrupt a wide spectrum of genes, and a key challenge is identifying which of these alterations are causally involved in tumorigenesis. The role of recurrent hemizygous focal deletions is especially puzzling because these deletions preferentially affect certain chromosomal regions and result in the loss of one copy of a whole cluster of adjacent genes. Solimini et al. (p. 104, published online 24 May; see the Perspective by Greenman) found that these deletions span genomic regions that are enriched in genes that negatively regulate cell proliferation. The cumulative reduction in dosage and tumor suppressive function of the genes within these “cancer gene islands” may represent a critical factor driving tumor growth. The genomes of cancer cells have preferentially lost genes that inhibit cell growth. Tumors exhibit numerous recurrent hemizygous focal deletions that contain no known tumor suppressors and are poorly understood. To investigate whether these regions contribute to tumorigenesis, we searched genetically for genes with cancer-relevant properties within these hemizygous deletions. We identified STOP and GO genes, which negatively and positively regulate proliferation, respectively. STOP genes include many known tumor suppressors, whereas GO genes are enriched for essential genes. Analysis of their chromosomal distribution revealed that recurring deletions preferentially overrepresent STOP genes and underrepresent GO genes. We propose a hypothesis called the cancer gene island model, whereby gene islands encompassing high densities of STOP genes and low densities of GO genes are hemizygously deleted to maximize proliferative fitness through cumulative haploinsufficiencies. Because hundreds to thousands of genes are hemizygously deleted per tumor, this mechanism may help to drive tumorigenesis across many cancer types.


Nature Medicine | 2015

Studying clonal dynamics in response to cancer therapy using high-complexity barcoding

Hyo-eun C. Bhang; David A. Ruddy; Viveksagar Krishnamurthy Radhakrishna; Justina X. Caushi; Rui Zhao; Matthew M Hims; Angad P. Singh; Iris Kao; Daniel Rakiec; Pamela Shaw; Marissa Balak; Alina Raza; Elizabeth Ackley; Nicholas Keen; Michael R. Schlabach; Michael Palmer; Rebecca J. Leary; Derek Y. Chiang; William R. Sellers; Franziska Michor; Vesselina G. Cooke; Joshua Korn; Frank Stegmeier

Resistance to cancer therapies presents a significant clinical challenge. Recent studies have revealed intratumoral heterogeneity as a source of therapeutic resistance. However, it is unclear whether resistance is driven predominantly by pre-existing or de novo alterations, in part because of the resolution limits of next-generation sequencing. To address this, we developed a high-complexity barcode library, ClonTracer, which enables the high-resolution tracking of more than 1 million cancer cells under drug treatment. In two clinically relevant models, ClonTracer studies showed that the majority of resistant clones were part of small, pre-existing subpopulations that selectively escaped under therapeutic challenge. Moreover, the ClonTracer approach enabled quantitative assessment of the ability of combination treatments to suppress resistant clones. These findings suggest that resistant clones are present before treatment, which would make up-front therapeutic combinations that target non-overlapping resistance a preferred approach. Thus, ClonTracer barcoding may be a valuable tool for optimizing therapeutic regimens with the goal of curative combination therapies for cancer.


Cancer Discovery | 2016

CRISPR Screens Provide a Comprehensive Assessment of Cancer Vulnerabilities but Generate False-Positive Hits for Highly Amplified Genomic Regions

Diana M Munoz; Pamela J. Cassiani; Li Li; Eric Billy; Joshua Korn; Michael D. Jones; Javad Golji; David A. Ruddy; Kristine Yu; Gregory McAllister; Antoine deWeck; Dorothee Abramowski; Jessica Wan; Matthew D. Shirley; Sarah Y. Neshat; Daniel Rakiec; Rosalie de Beaumont; Odile Weber; Audrey Kauffmann; E. Robert McDonald; Nicholas Keen; Francesco Hofmann; William R. Sellers; Tobias Schmelzle; Frank Stegmeier; Michael R. Schlabach

UNLABELLED 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. 824See related article by Aguirre et al., p. 914This article is highlighted in the In This Issue feature, p. 803.


Science | 2016

Disordered methionine metabolism in MTAP/CDKN2A-deleted cancers leads to dependence on PRMT5.

Konstantinos Mavrakis; E. Robert McDonald; Michael R. Schlabach; Eric Billy; Gregory R. Hoffman; Antoine deWeck; David A. Ruddy; Kavitha Venkatesan; Jianjun Yu; Gregg McAllister; Mark Stump; Rosalie deBeaumont; Samuel Ho; Yingzi Yue; Yue Liu; Yan Yan-Neale; Guizhi Yang; Fallon Lin; Hong Yin; Hui Gao; D. Randal Kipp; Songping Zhao; Joshua T. McNamara; Elizabeth R. Sprague; Bing Zheng; Ying Lin; Young Shin Cho; Justin Gu; Kenneth Crawford; David N. Ciccone

Tumors put in a vulnerable position Cancer cells often display alterations in metabolism that help fuel their growth. Such metabolic “rewiring” may also work against the cancer cells, however, by creating new vulnerabilities that can be exploited therapeutically. A variety of human tumors show changes in methionine metabolism caused by loss of the gene coding for 5-methylthioadenosine phosphorylase (MTAP). Mavrakis et al. and Kryukov et al. found that the loss of MTAP renders cancer cell lines sensitive to growth inhibition by compounds that suppress the activity of a specific arginine methyltransferase called PRMT5. Conceivably, drugs that inhibit PRMT5 activity could be developed into a tailored therapy for MTAP-deficient tumors. Science, this issue pp. 1208 and 1214 Tumors cope with a genomic change by rewiring their metabolism, but this makes them more susceptible to certain drugs. 5-Methylthioadenosine phosphorylase (MTAP) is a key enzyme in the methionine salvage pathway. The MTAP gene is frequently deleted in human cancers because of its chromosomal proximity to the tumor suppressor gene CDKN2A. By interrogating data from a large-scale short hairpin RNA–mediated screen across 390 cancer cell line models, we found that the viability of MTAP-deficient cancer cells is impaired by depletion of the protein arginine methyltransferase PRMT5. MTAP-deleted cells accumulate the metabolite methylthioadenosine (MTA), which we found to inhibit PRMT5 methyltransferase activity. Deletion of MTAP in MTAP-proficient cells rendered them sensitive to PRMT5 depletion. Conversely, reconstitution of MTAP in an MTAP-deficient cell line rescued PRMT5 dependence. Thus, MTA accumulation in MTAP–deleted cancers creates a hypomorphic PRMT5 state that is selectively sensitized toward further PRMT5 inhibition. Inhibitors of PRMT5 that leverage this dysregulated metabolic state merit further investigation as a potential therapy for MTAP/CDKN2A-deleted tumors.

Collaboration


Dive into the Michael R. Schlabach's collaboration.

Top Co-Authors

Avatar

Stephen J. Elledge

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Ji Luo

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Qikai Xu

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Anthony C. Liang

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Mamie Z. Li

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Nicole L. Solimini

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge