Network


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

Hotspot


Dive into the research topics where Morgan L. Maeder is active.

Publication


Featured researches published by Morgan L. Maeder.


Nature Biotechnology | 2013

Efficient genome editing in zebrafish using a CRISPR-Cas system

Woong Y. Hwang; Yanfang Fu; Deepak Reyon; Morgan L. Maeder; Shengdar Q. Tsai; Jeffry D. Sander; Randall T. Peterson; Jing-Ruey J. Yeh; J. Keith Joung

In bacteria, foreign nucleic acids are silenced by clustered, regularly interspaced, short palindromic repeats (CRISPR)–CRISPR-associated (Cas) systems. Bacterial type II CRISPR systems have been adapted to create guide RNAs that direct site-specific DNA cleavage by the Cas9 endonuclease in cultured cells. Here we show that the CRISPR-Cas system functions in vivo to induce targeted genetic modifications in zebrafish embryos with efficiencies similar to those obtained using zinc finger nucleases and transcription activator–like effector nucleases.In bacteria, foreign nucleic acids are silenced by clustered, regularly interspaced, short palindromic repeats (CRISPR)--CRISPR-associated (Cas) systems. Bacterial type II CRISPR systems have been adapted to create guide RNAs that direct site-specific DNA cleavage by the Cas9 endonuclease in cultured cells. Here we show that the CRISPR-Cas system functions in vivo to induce targeted genetic modifications in zebrafish embryos with efficiencies similar to those obtained using zinc finger nucleases and transcription activator-like effector nucleases.


Nature Biotechnology | 2013

High-frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells

Yanfang Fu; Jennifer A. Foden; Cyd Khayter; Morgan L. Maeder; Deepak Reyon; J. Keith Joung; Jeffry D. Sander

Clustered, regularly interspaced, short palindromic repeat (CRISPR) RNA-guided nucleases (RGNs) have rapidly emerged as a facile and efficient platform for genome editing. Here, we use a human cell–based reporter assay to characterize off-target cleavage of CRISPR-associated (Cas)9-based RGNs. We find that single and double mismatches are tolerated to varying degrees depending on their position along the guide RNA (gRNA)-DNA interface. We also readily detected off-target alterations induced by four out of six RGNs targeted to endogenous loci in human cells by examination of partially mismatched sites. The off-target sites we identified harbored up to five mismatches and many were mutagenized with frequencies comparable to (or higher than) those observed at the intended on-target site. Our work demonstrates that RGNs can be highly active even with imperfectly matched RNA-DNA interfaces in human cells, a finding that might confound their use in research and therapeutic applications.


Nature Methods | 2013

CRISPR RNA-guided activation of endogenous human genes

Morgan L. Maeder; Samantha J Linder; Vincent M Cascio; Yanfang Fu; Quan H Ho; J. Keith Joung

Short guide RNAs (gRNAs) can direct catalytically inactive CRISPR-associated 9 nuclease (dCas9) to repress endogenous genes in bacteria and human cells. Here we show that single or multiple gRNAs can direct dCas9 fused to a VP64 transcriptional activation domain to increase expression of endogenous human genes. This proof-of-principle work shows that clustered regularly interspaced short palindromic repeat (CRISPR)-Cas systems can target heterologous effector domains to endogenous sites in human cells.


Nature | 2009

High frequency modification of plant genes using engineered zinc finger nucleases

Jeffrey A. Townsend; David A. Wright; Ronnie J. Winfrey; Fengli Fu; Morgan L. Maeder; J. Keith Joung; Daniel F. Voytas

An efficient method for making directed DNA sequence modifications to plant genes (gene targeting) is at present lacking, thereby frustrating efforts to dissect plant gene function and engineer crop plants that better meet the world’s burgeoning need for food, fibre and fuel. Zinc-finger nucleases (ZFNs)—enzymes engineered to create DNA double-strand breaks at specific loci—are potent stimulators of gene targeting; for example, they can be used to precisely modify engineered reporter genes in plants. Here we demonstrate high-frequency ZFN-stimulated gene targeting at endogenous plant genes, namely the tobacco acetolactate synthase genes (ALS SuRA and SuRB), for which specific mutations are known to confer resistance to imidazolinone and sulphonylurea herbicides. Herbicide-resistance mutations were introduced into SuR loci by ZFN-mediated gene targeting at frequencies exceeding 2% of transformed cells for mutations as far as 1.3 kilobases from the ZFN cleavage site. More than 40% of recombinant plants had modifications in multiple SuR alleles. The observed high frequency of gene targeting indicates that it is now possible to efficiently make targeted sequence changes in endogenous plant genes.


Nature | 2007

Discovery of functional elements in 12 Drosophila genomes using evolutionary signatures

Alexander Stark; Michael F. Lin; Pouya Kheradpour; Jakob Skou Pedersen; Leopold Parts; Joseph W. Carlson; Madeline A. Crosby; Matthew D. Rasmussen; Sushmita Roy; Ameya N. Deoras; J. Graham Ruby; Julius Brennecke; Harvard FlyBase curators; Berkeley Drosophila Genome; Emily Hodges; Angie S. Hinrichs; Anat Caspi; Benedict Paten; Seung-Won Park; Mira V. Han; Morgan L. Maeder; Benjamin J. Polansky; Bryanne E. Robson; Stein Aerts; Jacques van Helden; Bassem A. Hassan; Donald G. Gilbert; Deborah A. Eastman; Michael D. Rice; Michael Weir

Sequencing of multiple related species followed by comparative genomics analysis constitutes a powerful approach for the systematic understanding of any genome. Here, we use the genomes of 12 Drosophila species for the de novo discovery of functional elements in the fly. Each type of functional element shows characteristic patterns of change, or ‘evolutionary signatures’, dictated by its precise selective constraints. Such signatures enable recognition of new protein-coding genes and exons, spurious and incorrect gene annotations, and numerous unusual gene structures, including abundant stop-codon readthrough. Similarly, we predict non-protein-coding RNA genes and structures, and new microRNA (miRNA) genes. We provide evidence of miRNA processing and functionality from both hairpin arms and both DNA strands. We identify several classes of pre- and post-transcriptional regulatory motifs, and predict individual motif instances with high confidence. We also study how discovery power scales with the divergence and number of species compared, and we provide general guidelines for comparative studies.


Cell Stem Cell | 2009

Gene Targeting of a Disease-Related Gene in Human Induced Pluripotent Stem and Embryonic Stem Cells

Jizhong Zou; Morgan L. Maeder; Prashant Mali; Shondra M. Pruett-Miller; Stacey Thibodeau-Beganny; Bin Kuan Chou; Guibin Chen; Zhaohui Ye; In-Hyun Park; George Q. Daley; Matthew H. Porteus; J. Keith Joung; Linzhao Cheng

We report here homologous recombination (HR)-mediated gene targeting of two different genes in human iPS cells (hiPSCs) and human ES cells (hESCs). HR-mediated correction of a chromosomally integrated mutant GFP reporter gene reaches efficiencies of 0.14%-0.24% in both cell types transfected by donor DNA with plasmids expressing zinc finger nucleases (ZFNs). Engineered ZFNs that induce a sequence-specific double-strand break in the GFP gene enhanced HR-mediated correction by > 1400-fold without detectable alterations in stem cell karyotypes or pluripotency. Efficient HR-mediated insertional mutagenesis was also achieved at the endogenous PIG-A locus, with a > 200-fold enhancement by ZFNs targeted to that gene. Clonal PIG-A null hESCs and iPSCs with normal karyotypes were readily obtained. The phenotypic and biological defects were rescued by PIG-A transgene expression. Our study provides the first demonstration of HR-mediated gene targeting in hiPSCs and shows the power of ZFNs for inducing specific genetic modifications in hiPSCs, as well as hESCs.


Nature Biotechnology | 2015

Cationic lipid-mediated delivery of proteins enables efficient protein-based genome editing in vitro and in vivo

John A Zuris; David B. Thompson; Yilai Shu; John Paul Guilinger; Jeffrey L. Bessen; Johnny H. Hu; Morgan L. Maeder; J. Keith Joung; Zheng-Yi Chen; David R. Liu

Efficient intracellular delivery of proteins is needed to fully realize the potential of protein therapeutics. Current methods of protein delivery commonly suffer from low tolerance for serum, poor endosomal escape and limited in vivo efficacy. Here we report that common cationic lipid nucleic acid transfection reagents can potently deliver proteins that are fused to negatively supercharged proteins, that contain natural anionic domains or that natively bind to anionic nucleic acids. This approach mediates the potent delivery of nM concentrations of Cre recombinase, TALE- and Cas9-based transcription activators, and Cas9:sgRNA nuclease complexes into cultured human cells in media containing 10% serum. Delivery of unmodified Cas9:sgRNA complexes resulted in up to 80% genome modification with substantially higher specificity compared to DNA transfection. This approach also mediated efficient delivery of Cre recombinase and Cas9:sgRNA complexes into the mouse inner ear in vivo, achieving 90% Cre-mediated recombination and 20% Cas9-mediated genome modification in hair cells.Efficient intracellular delivery of proteins is needed to fully realize the potential of protein therapeutics. Current methods of protein delivery commonly suffer from low tolerance for serum, poor endosomal escape, and limited in vivo efficacy. Here we report that common cationic lipid nucleic acid transfection reagents can potently deliver proteins that are fused to negatively supercharged proteins, that contain natural anionic domains, or that natively bind to anionic nucleic acids. This approach mediates the potent delivery of nM concentrations of Cre recombinase, TALE- and Cas9-based transcriptional activators, and Cas9:sgRNA nuclease complexes into cultured human cells in media containing 10% serum. Delivery of Cas9:sgRNA complexes resulted in up to 80% genome modification with substantially higher specificity compared to DNA transfection. This approach also mediated efficient delivery of Cre recombinase and Cas9:sgRNA complexes into the mouse inner ear in vivo, achieving 90% Cre-mediated recombination and 20% Cas9-mediated genome modification in hair cells.


Nature Methods | 2011

Selection-free zinc-finger-nuclease engineering by context-dependent assembly (CoDA)

Jeffry D. Sander; Elizabeth J. Dahlborg; Mathew J. Goodwin; Lindsay Cade; Feng Zhang; Daniel Cifuentes; Shaun J. Curtin; Jessica S. Blackburn; Stacey Thibodeau-Beganny; Yiping Qi; Christopher J. Pierick; Ellen J. Hoffman; Morgan L. Maeder; Cyd Khayter; Deepak Reyon; Drena Dobbs; David M. Langenau; Robert M. Stupar; Antonio J. Giraldez; Daniel F. Voytas; Randall T. Peterson; Jing-Ruey J. Yeh; J. Keith Joung

Engineered zinc-finger nucleases (ZFNs) enable targeted genome modification. Here we describe context-dependent assembly (CoDA), a platform for engineering ZFNs using only standard cloning techniques or custom DNA synthesis. Using CoDA-generated ZFNs, we rapidly altered 20 genes in Danio rerio, Arabidopsis thaliana and Glycine max. The simplicity and efficacy of CoDA will enable broad adoption of ZFN technology and make possible large-scale projects focused on multigene pathways or genome-wide alterations.


Nature Biotechnology | 2013

Targeted DNA demethylation and activation of endogenous genes using programmable TALE-TET1 fusion proteins

Morgan L. Maeder; James Angstman; Marcy E. Richardson; Samantha J Linder; Vincent M Cascio; Shengdar Q. Tsai; Quan H Ho; Jeffry D. Sander; Deepak Reyon; Bradley E. Bernstein; Joseph F. Costello; Miles F. Wilkinson; J. Keith Joung

Genome-wide studies have defined cell type–specific patterns of DNA methylation that are important for regulating gene expression in both normal development and disease. However, determining the functional significance of specific methylation events remains challenging, owing to the lack of methods for removing such modifications in a targeted manner. Here we describe an approach for efficient targeted demethylation of specific CpGs in human cells using fusions of engineered transcription activator–like effector (TALE) repeat arrays and the TET1 hydroxylase catalytic domain. Using these TALE-TET1 fusions, we demonstrate that modification of critical methylated promoter CpG positions can lead to substantial increases in the expression of endogenous human genes. Our results delineate a strategy for understanding the functional significance of specific CpG methylation marks in the context of endogenous gene loci and validate programmable DNA demethylation reagents with potential utility for research and therapeutic applications.Time-course evaluation of TALE-TET1-indcued effects on HBB promoter methylation and gene expression To test whether the -266 CpG demethylation and increased HBB gene expression induced by HB-4, HB-5-, and HB-6 are stable over time, we assayed these parameters in a more extended time-course experiment. K562 cells transfected with expression plasmids encoding HB-4, HB-5, HB-6, or a control TALE-TET1 fusion targeted to the KLF4 locus were assayed for -266 CpG demethylation and HBB gene expression at 4, 7, 14, and 30 days post-transfection. HB-4, HB5, and HB-6 fusions all showed their highest levels of fold-activation at post-transfection day 4 with HBB expression steadily decreasing by day 30 to the same level observed with the control KLF4-targeted TALE-TET1 protein (Supplementary Fig. 10a). Strikingly, for all three HBBtargeted TALE-TET1 proteins, the extent of -266 CpG methylation over time in the population of transfected cells directly paralleled the fold-activation of HBB gene expression while the methylation status of another CpG at position -306 did not change significantly during the course of the experiment (Supplementary Fig. 10b).


Stem Cells | 2011

In Situ Genetic Correction of the Sickle Cell Anemia Mutation in Human Induced Pluripotent Stem Cells Using Engineered Zinc Finger Nucleases

Vittorio Sebastiano; Morgan L. Maeder; James Angstman; Bahareh Haddad; Cyd Khayter; Dana T. Yeo; Mathew J. Goodwin; John S. Hawkins; Cherie L. Ramirez; Luis F.Z. Batista; Steven E. Artandi; Marius Wernig; J. Keith Joung

The combination of induced pluripotent stem cell (iPSC) technology and targeted gene modification by homologous recombination (HR) represents a promising new approach to generate genetically corrected, patient‐derived cells that could be used for autologous transplantation therapies. This strategy has several potential advantages over conventional gene therapy including eliminating the need for immunosuppression, avoiding the risk of insertional mutagenesis by therapeutic vectors, and maintaining expression of the corrected gene by endogenous control elements rather than a constitutive promoter. However, gene targeting in human pluripotent cells has remained challenging and inefficient. Recently, engineered zinc finger nucleases (ZFNs) have been shown to substantially increase HR frequencies in human iPSCs, raising the prospect of using this technology to correct disease causing mutations. Here, we describe the generation of iPSC lines from sickle cell anemia patients and in situ correction of the disease causing mutation using three ZFN pairs made by the publicly available oligomerized pool engineering method (OPEN). Gene‐corrected cells retained full pluripotency and a normal karyotype following removal of reprogramming factor and drug‐resistance genes. By testing various conditions, we also demonstrated that HR events in human iPSCs can occur as far as 82 bps from a ZFN‐induced break. Our approach delineates a roadmap for using ZFNs made by an open‐source method to achieve efficient, transgene‐free correction of monogenic disease mutations in patient‐derived iPSCs. Our results provide an important proof of principle that ZFNs can be used to produce gene‐corrected human iPSCs that could be used for therapeutic applications. STEM CELLS 2011;29:1717–1726

Collaboration


Dive into the Morgan L. Maeder's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jeffry D. Sander

Howard Hughes Medical Institute

View shared research outputs
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
Researchain Logo
Decentralizing Knowledge