Network


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

Hotspot


Dive into the research topics where Zachary Steinhart is active.

Publication


Featured researches published by Zachary Steinhart.


Cell | 2015

High-Resolution CRISPR Screens Reveal Fitness Genes and Genotype-Specific Cancer Liabilities

Traver Hart; Megha Chandrashekhar; Michael Aregger; Zachary Steinhart; Kevin R. Brown; Graham MacLeod; Monika Mis; Michal Zimmermann; Amélie Fradet-Turcotte; Song Sun; Patricia Mero; Peter Dirks; Sachdev S. Sidhu; Frederick P. Roth; Olivia S. Rissland; Daniel Durocher; Stephane Angers; Jason Moffat

The ability to perturb genes in human cells is crucial for elucidating gene function and holds great potential for finding therapeutic targets for diseases such as cancer. To extend the catalog of human core and context-dependent fitness genes, we have developed a high-complexity second-generation genome-scale CRISPR-Cas9 gRNA library and applied it to fitness screens in five human cell lines. Using an improved Bayesian analytical approach, we consistently discover 5-fold more fitness genes than were previously observed. We present a list of 1,580 human core fitness genes and describe their general properties. Moreover, we demonstrate that context-dependent fitness genes accurately recapitulate pathway-specific genetic vulnerabilities induced by known oncogenes and reveal cell-type-specific dependencies for specific receptor tyrosine kinases, even in oncogenic KRAS backgrounds. Thus, rigorous identification of human cell line fitness genes using a high-complexity CRISPR-Cas9 library affords a high-resolution view of the genetic vulnerabilities of a cell.


Nature Medicine | 2017

Genome-wide CRISPR screens reveal a Wnt-FZD5 signaling circuit as a druggable vulnerability of RNF43-mutant pancreatic tumors

Zachary Steinhart; Zvezdan Pavlovic; Megha Chandrashekhar; Traver Hart; Xiaowei Wang; Xiaoyu Zhang; Mélanie Robitaille; Kevin R. Brown; Sridevi Jaksani; René M. Overmeer; Sylvia F. Boj; Jarrett J. Adams; James Pan; Hans Clevers; Sachdev S. Sidhu; Jason Moffat; Stephane Angers

Forward genetic screens with CRISPR–Cas9 genome editing enable high-resolution detection of genetic vulnerabilities in cancer cells. We conducted genome-wide CRISPR–Cas9 screens in RNF43-mutant pancreatic ductal adenocarcinoma (PDAC) cells, which rely on Wnt signaling for proliferation. Through these screens, we discovered a unique requirement for a Wnt signaling circuit: engaging FZD5, one of the ten Frizzled receptors encoded in the human genome. Our results uncover an underappreciated level of context-dependent specificity at the Wnt receptor level. We further derived a panel of recombinant antibodies that reports the expression of nine FZD proteins and confirms that FZD5 functional specificity cannot be explained by protein expression patterns. Additionally, antibodies that specifically bind FZD5 and FZD8 robustly inhibited the growth of RNF43-mutant PDAC cells grown in vitro and as xenografts in vivo, providing orthogonal support for the functional specificity observed genetically. Proliferation of a patient-derived PDAC cell line harboring an RNF43 variant was also selectively inhibited by the FZD5 antibodies, further demonstrating their use as a potential targeted therapy. Tumor organoid cultures from colorectal carcinoma patients that carried RNF43 mutations were also sensitive to the FZD5 antibodies, highlighting the potential generalizability of these findings beyond PDAC. Our results show that CRIPSR-based genetic screens can be leveraged to identify and validate cell surface targets for antibody development and therapy.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Dishevelled is a NEK2 kinase substrate controlling dynamics of centrosomal linker proteins.

Igor Cervenka; Jana Valnohova; Ondrej Bernatik; Jakub Harnoš; Matej Radsetoulal; Katerina Sedova; Katerina Hanakova; David Potesil; Alena Salašová; Zachary Steinhart; Stephane Angers; Gunnar Schulte; Aleš Hampl; Zbynek Zdrahal; Vitezslav Bryja

Significance Wnt signaling is an important orchestrator of embryonic development. We provide evidence that Wnt scaffolding protein Dishevelled (DVL) contributes to the dissolution of centrosomal linker, preceding separation of centrosomes. We show that DVL accumulates toward mitosis, and its centrosomal function is controlled by NEK2. Our data demonstrate that DVL is required for the removal of linker proteins from centrosome, an event necessary for the correct formation of a bipolar spindle and cell cycle transition. This surprising function of DVL creates a mechanistic basis for the novel crosstalk between Wnt signaling pathways and the centrosome cycle. Dishevelled (DVL) is a key scaffolding protein and a branching point in Wnt signaling pathways. Here, we present conclusive evidence that DVL regulates the centrosomal cycle. We demonstrate that DVL dishevelled and axin (DIX) domain, but not DIX domain-mediated multimerization, is essential for DVL’s centrosomal localization. DVL accumulates during the cell cycle and associates with NIMA-related kinase 2 (NEK2), which is able to phosphorylate DVL at a multitude of residues, as detected by a set of novel phospho-specific antibodies. This creates interfaces for efficient binding to CDK5 regulatory subunit-associated protein 2 (CDK5RAP2) and centrosomal Nek2-associated protein 1 (C-NAP1), two proteins of the centrosomal linker. Displacement of DVL from the centrosome and its release into the cytoplasm on NEK2 phosphorylation is coupled to the removal of linker proteins, an event necessary for centrosomal separation and proper formation of the mitotic spindle. Lack of DVL prevents NEK2-controlled dissolution of loose centrosomal linker and subsequent centrosomal separation. Increased DVL levels, in contrast, sequester centrosomal NEK2 and mimic monopolar spindle defects induced by a dominant negative version of this kinase. Our study thus uncovers molecular crosstalk between centrosome and Wnt signaling.


Journal of Cell Science | 2016

Essential role of the Dishevelled DEP domain in a Wnt-dependent human-cell-based complementation assay

Melissa Gammons; Trevor J. Rutherford; Zachary Steinhart; Stephane Angers; Mariann Bienz

ABSTRACT Dishevelled (DVL) assembles Wnt signalosomes through dynamic head-to-tail polymerisation by means of its DIX domain. It thus transduces Wnt signals to cytoplasmic effectors including β-catenin, to control cell fates during normal development, tissue homeostasis and also in cancer. To date, most functional studies of Dishevelled relied on its Wnt-independent signalling activity resulting from overexpression, which is sufficient to trigger polymerisation, bypassing the requirement for Wnt signals. Here, we generate a human cell line devoid of endogenous Dishevelled (DVL1– DVL3), which lacks Wnt signal transduction to β-catenin. However, Wnt responses can be restored by DVL2 stably re-expressed at near-endogenous levels. Using this assay to test mutant DVL2, we show that its DEP domain is essential, whereas its PDZ domain is dispensable, for signalling to β-catenin. Our results imply two mutually exclusive functions of the DEP domain in Wnt signal transduction – binding to Frizzled to recruit Dishevelled to the receptor complex, and dimerising to cross-link DIX domain polymers for signalosome assembly. Our assay avoids the caveats associated with overexpressing Dishevelled, and provides a powerful tool for rigorous functional tests of this pivotal human signalling protein. Summary: A physiological complementation assay in Dishevelled null-mutant human cells establishes an essential function of the DEP domain of Dishevelled in its binding to Frizzled for signal transduction to β-catenin.


Development | 2018

Wnt signaling in development and tissue homeostasis

Zachary Steinhart; Stephane Angers

ABSTRACT The Wnt-β-catenin signaling pathway is an evolutionarily conserved cell-cell communication system that is important for stem cell renewal, cell proliferation and cell differentiation both during embryogenesis and during adult tissue homeostasis. Genetic or epigenetic events leading to hypo- or hyper-activation of the Wnt-β-catenin signaling cascade have also been associated with human diseases such as cancer. Understanding how this pathway functions is thus integral for developing therapies to treat diseases or for regenerative medicine approaches. Here, and in the accompanying poster, we provide an overview of Wnt-β-catenin signaling and briefly highlight its key functions during development and adult tissue homeostasis. Summary: An overview of Wnt-β-catenin signaling highlighting its key functions during development and adult tissue homeostasis.


Molecular Cancer Research | 2016

Abstract IA13: Inhibiting the Wnt pathway with selective anti-Frizzled synthetic antibodies

Zachary Steinhart; Traver Hart; Sachdev S. Sidhu; Jason Moffat; Stephane Angers

Secreted Wnt glycoproteins regulate intracellular signaling pathways important for embryonic development and adult tissue homeostasis. Misregulation of Wnt signalling is frequent in human cancers. Wnt ligands are recognized at the surface of cells by various receptor complexes that are differentially expressed and determine context-dependent cellular responses. In humans, 16 Wnt ligands interact directly with 10 Frizzled seven transmembrane proteins through their extracellular cysteine rich domain (CRD). Wnts also interact with various co-receptors, such as LRP5/6, ROR1/2, RYK, PTK7, which engage different downstream signaling pathways. While genetic analysis of mouse knockouts has revealed specific roles for different Wnt receptors and co-receptors during embryonic development, relatively little is known about their respective functions during adult tissue homeostasis or in the initiation and progression of human diseases such as cancer. There is a paucity of reagents allowing the selective and temporal pharmacological inhibition of Wnt receptors. We are employing phage-display technology to identify high affinity synthetic blocking monoclonal antibodies (mAbs) against all human Wnt receptors. To date we have isolated a collection of anti-receptor antibodies exhibiting unique selectivity and functionality profiles. Our lead anti-Frizzled mAbs targeting a subset of Frizzled proteins inhibit Wnt-promoted activation of the TopFlash reporter with low nanomolar potency. These mAbs also exhibit dose dependent anti-proliferative efficacies against various pancreatic cancer cell lines grown in vitro and as xenografts in mice. These reagents will be useful to probe the function of specific Wnt receptors in tissue homeostasis and human diseases and will lead to novel therapeutic agents for their treatment. Our lab is also employing whole genome CRISPR screening approach to identify genotype specific vulnerabilities in cancer cells. These efforts have identified specific Frizzled important for cancer cells proliferation and is guiding our antibody development program. Citation Format: Zachary Steinhart, Traver Hart, Sachdev Sidhu, Jason Moffat, Stephane Angers. Inhibiting the Wnt pathway with selective anti-Frizzled synthetic antibodies. [abstract]. In: Proceedings of the AACR Special Conference: Developmental Biology and Cancer; Nov 30-Dec 3, 2015; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Res 2016;14(4_Suppl):Abstract nr IA13.


bioRxiv | 2015

Systematic discovery and classification of human cell line essential genes

Traver Hart; Megha Chandrashekhar; Michael Aregger; Zachary Steinhart; Kevin R. Brown; Stephane Angers; Jason Moffat

The study of gene essentiality in human cells is crucial for elucidating gene function and holds great potential for finding therapeutic targets for diseases such as cancer. Technological advances in genome editing using clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 systems have set the stage for identifying human cell line core and context-dependent essential genes. However, first generation negative selection screens using CRISPR technology demonstrate extreme variability across different cell lines. To advance the development of the catalogue of human core and context-dependent essential genes, we have developed an optimized, ultracomplex, genome-scale gRNA library of 176,500 guide RNAs targeting 17,661 genes and have applied it to negative and positive selection screens in a human cell line. Using an improved Bayesian analytical approach, we find CRISPR-based screens yield double to triple the number of essential genes than were previously observed using systematic RNA interference, including many genes at moderate expression levels that are largely refractory to RNAi methods. We further characterized four essential genes of unknown significance and found that they all likely exist in protein complexes with other essential genes. For example, RBM48 and ARMC7 are both essential nuclear proteins, strongly interact and are commonly amplified across major cancers. Our findings suggest the CRISPR-Cas9 system fundamentally alters the landscape for systematic reverse genetics in human cells for elucidating gene function, identifying disease genes, and uncovering therapeutic targets.


mAbs | 2018

A synthetic anti-Frizzled antibody engineered for broadened specificity exhibits enhanced anti-tumor properties

Zvezdan Pavlovic; Jarrett J. Adams; Levi L. Blazer; Amandeep Gakhal; Nick Jarvik; Zachary Steinhart; Mélanie Robitaille; Keith Mascall; James Pan; Stephane Angers; Jason Moffat; Sachdev S. Sidhu

ABSTRACT Secreted Wnt ligands play a major role in the development and progression of many cancers by modulating signaling through cell-surface Frizzled receptors (FZDs). In order to achieve maximal effect on Wnt signaling by targeting the cell surface, we developed a synthetic antibody targeting six of the 10 human FZDs. We first identified an anti-FZD antagonist antibody (F2) with a specificity profile matching that of OMP-18R5, a monoclonal antibody that inhibits growth of many cancers by targeting FZD7, FZD1, FZD2, FZD5 and FZD8. We then used combinatorial antibody engineering by phage display to develop a variant antibody F2.A with specificity broadened to include FZD4. We confirmed that F2.A blocked binding of Wnt ligands, but not binding of Norrin, a ligand that also activates FZD4. Importantly, F2.A proved to be much more efficacious than either OMP-18R5 or F2 in inhibiting the growth of multiple RNF43-mutant pancreatic ductal adenocarcinoma cell lines, including patient-derived cells.


bioRxiv | 2018

The functional genomic circuitry of human glioblastoma stem cells

Graham MacLeod; Danielle A. Bozek; Nishani Rajakulendran; Vernon Monteiro; Moloud Ahmadi; Zachary Steinhart; Michelle Kushida; Helen Yu; Fiona J. Coutinho; Ian Restall; Xiaoguang Hao; Traver Hart; H. Artee Luchman; Samuel Weiss; Peter Dirks; Stephane Angers

Summary Successful glioblastoma (GBM) therapies have remained elusive due to limitations in understanding mechanisms of growth and survival of the tumorigenic population. Using CRISPR-Cas9 approaches in patient-derived GBM stem cells to interrogate function of the coding genome, we identify diverse actionable pathways responsible for growth that reveal the gene-essential circuitry of GBM stemness. In particular, we describe the Sox developmental transcription factor family; H3K79 methylation by DOT1L; and ufmylation stress responsiveness programs as essential for GBM stemness. Additionally, we find mechanisms of temozolomide resistance and sensitivity that could lead to combination strategies with this standard of care treatment. By reaching beyond static genome analysis of bulk tumors, with a genome wide functional approach, we dive deep into a broad range of biological processes to provide new understanding of GBM growth and treatment resistance. Significance Glioblastoma (GBM) remains an incurable disease despite an increasingly thorough depth of knowledge of the genomic and epigenomic alterations of bulk tumors. Evidence from multiple approaches support that GBM reflects an aberrant developmental hierarchy, with GBM stem cells (GSCs), fueling tumor growth and invasion. The properties of this tumor subpopulation may also in part explain treatment resistance and disease recurrence. Unfortunately, we still have a limited knowledge of the molecular circuitry of these cells and progress has been slow as we have not been able, until recently, to interrogate function at the genome-wide scale. Here, using parallel genome-wide CRISPR-Cas9 screens, we identify the essential genes for GSC growth. Further, by screening in the presence of low and high dose temozolomide, we identify mechanisms of drug resistance and sensitivity. These functional screens in patient derived cells reveal new aspects of GBM biology and identify a diversity of actionable targets such as genes governing stem cell traits, epigenome regulation and the response to stress stimuli.


bioRxiv | 2016

A CRISPR screen reveals a WNT7B-FZD5 signaling circuit as a therapeutic opportunity in pancreatic cancer.

Zachary Steinhart; Traver Hart; Megha Chandrashekhar; Zvezdan Pavlovic; Mélanie Robitaille; Xiaowei Wang; Jarrett J. Adams; James Pan; Sachdev S. Sidhu; Jason Moffat; Stephane Angers

CRISPR-Cas9 genome editing enables high-resolution detection of genetic vulnerabilities of cancer cells. We conducted a genome-wide CRISPR-Cas9 screen in RNF43 mutant pancreatic ductal adenocarcinoma (PDAC) cells, which rely on Wnt signaling for proliferation, and discovered a unique requirement for a WNT7B-FZD5 signaling circuit. Our results highlight an underappreciated level of functional specificity at the ligand-receptor level. We derived a panel of recombinant antibodies that reports the expression of nine out of ten human Frizzled receptors and confirm that WNT7B-FZD5 functional specificity cannot be explained by protein expression patterns. We developed two human antibodies that target FZD5 and robustly inhibited the growth of RNF43 mutant PDAC cells grown in vitro and as xenografts, providing strong orthogonal support for the functional specificity observed genetically. Proliferation of a patient-derived PDAC cell line harboring a RNF43 variant previously associated with PDAC was also selectively inhibited by the FZD5 antibodies, further demonstrating their use as a potential targeted therapy.

Collaboration


Dive into the Zachary Steinhart's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Traver Hart

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

James Pan

University of Toronto

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge