Network


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

Hotspot


Dive into the research topics where Edward J. Rebar is active.

Publication


Featured researches published by Edward J. Rebar.


Nature Biotechnology | 2011

A TALE nuclease architecture for efficient genome editing

Jeffrey C. Miller; Siyuan Tan; Guijuan Qiao; Kyle A Barlow; Jianbin Wang; Danny F Xia; Xiangdong Meng; David Paschon; Elo Leung; Sarah J. Hinkley; Gladys P Dulay; Kevin L. Hua; Irina Ankoudinova; Gregory J. Cost; Fyodor D. Urnov; H. Steve Zhang; Michael C. Holmes; Lei Zhang; Philip D. Gregory; Edward J. Rebar

Nucleases that cleave unique genomic sequences in living cells can be used for targeted gene editing and mutagenesis. Here we develop a strategy for generating such reagents based on transcription activator–like effector (TALE) proteins from Xanthomonas. We identify TALE truncation variants that efficiently cleave DNA when linked to the catalytic domain of FokI and use these nucleases to generate discrete edits or small deletions within endogenous human NTF3 and CCR5 genes at efficiencies of up to 25%. We further show that designed TALEs can regulate endogenous mammalian genes. These studies demonstrate the effective application of designed TALE transcription factors and nucleases for the targeted regulation and modification of endogenous genes.


Nature Biotechnology | 2011

Genetic engineering of human pluripotent cells using TALE nucleases

Dirk Hockemeyer; Haoyi Wang; Samira Kiani; Christine S. Lai; Qing Gao; John P. Cassady; Gregory J. Cost; Lei Zhang; Yolanda Santiago; Jeffrey C. Miller; Bryan Zeitler; Jennifer M. Cherone; Xiangdong Meng; Sarah J. Hinkley; Edward J. Rebar; Philip D. Gregory; Fyodor D. Urnov; Rudolf Jaenisch

Targeted genetic engineering of human pluripotent cells is a prerequisite for exploiting their full potential. Such genetic manipulations can be achieved using site-specific nucleases. Here we engineered transcription activator–like effector nucleases (TALENs) for five distinct genomic loci. At all loci tested we obtained human embryonic stem cell (ESC) and induced pluripotent stem cell (iPSC) clones carrying transgenic cassettes solely at the TALEN-specified location. Our data suggest that TALENs employing the specific architectures described here mediate site-specific genome modification in human pluripotent cells with similar efficiency and precision as do zinc-finger nucleases (ZFNs).


Nature Biotechnology | 2008

Establishment of HIV-1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases

Elena E. Perez; Jianbin Wang; Jeffrey C. Miller; Yann Jouvenot; Kenneth Kim; Olga Liu; Nathaniel Wang; Gary Lee; Victor Bartsevich; Ya-Li Lee; Dmitry Guschin; Igor Rupniewski; Adam Waite; Carmine Carpenito; Richard G. Carroll; Jordan S. Orange; Fyodor D. Urnov; Edward J. Rebar; Dale Ando; Philip D. Gregory; James L. Riley; Michael C. Holmes; Carl H. June

Homozygosity for the naturally occurring Δ32 deletion in the HIV co-receptor CCR5 confers resistance to HIV-1 infection. We generated an HIV-resistant genotype de novo using engineered zinc-finger nucleases (ZFNs) to disrupt endogenous CCR5. Transient expression of CCR5 ZFNs permanently and specifically disrupted ∼50% of CCR5 alleles in a pool of primary human CD4+ T cells. Genetic disruption of CCR5 provided robust, stable and heritable protection against HIV-1 infection in vitro and in vivo in a NOG model of HIV infection. HIV-1-infected mice engrafted with ZFN-modified CD4+ T cells had lower viral loads and higher CD4+ T-cell counts than mice engrafted with wild-type CD4+ T cells, consistent with the potential to reconstitute immune function in individuals with HIV/AIDS by maintenance of an HIV-resistant CD4+ T-cell population. Thus adoptive transfer of ex vivo expanded CCR5 ZFN–modified autologous CD4+ T cells in HIV patients is an attractive approach for the treatment of HIV-1 infection.


Nature Biotechnology | 2009

Efficient targeting of expressed and silent genes in human ESCs and iPSCs using zinc-finger nucleases

Dirk Hockemeyer; Frank Soldner; Caroline Beard; Qing Gao; Maisam Mitalipova; Russell DeKelver; George E. Katibah; Ranier Amora; Elizabeth A. Boydston; Bryan Zeitler; Xiangdong Meng; Jeffrey C. Miller; Lei Zhang; Edward J. Rebar; Philip D. Gregory; Fyodor D. Urnov; Rudolf Jaenisch

Realizing the full potential of human embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs) requires efficient methods for genetic modification. However, techniques to generate cell type–specific lineage reporters, as well as reliable tools to disrupt, repair or overexpress genes by gene targeting, are inefficient at best and thus are not routinely used. Here we report the highly efficient targeting of three genes in human pluripotent cells using zinc-finger nuclease (ZFN)–mediated genome editing. First, using ZFNs specific for the OCT4 (POU5F1) locus, we generated OCT4-eGFP reporter cells to monitor the pluripotent state of hESCs. Second, we inserted a transgene into the AAVS1 locus to generate a robust drug-inducible overexpression system in hESCs. Finally, we targeted the PITX3 gene, demonstrating that ZFNs can be used to generate reporter cells by targeting non-expressed genes in hESCs and hiPSCs.


Nature Biotechnology | 2007

An improved zinc-finger nuclease architecture for highly specific genome editing

Jeffrey C. Miller; Michael C. Holmes; Jianbin Wang; Dmitry Guschin; Ya-Li Lee; Igor Rupniewski; Christian Beausejour; Adam Waite; Nathaniel Wang; Kenneth Kim; Philip D. Gregory; Carl O. Pabo; Edward J. Rebar

Genome editing driven by zinc-finger nucleases (ZFNs) yields high gene-modification efficiencies (>10%) by introducing a recombinogenic double-strand break into the targeted gene. The cleavage event is induced using two custom-designed ZFNs that heterodimerize upon binding DNA to form a catalytically active nuclease complex. Using the current ZFN architecture, however, cleavage-competent homodimers may also form that can limit safety or efficacy via off-target cleavage. Here we develop an improved ZFN architecture that eliminates this problem. Using structure-based design, we engineer two variant ZFNs that efficiently cleave DNA only when paired as a heterodimer. These ZFNs modify a native endogenous locus as efficiently as the parental architecture, but with a >40-fold reduction in homodimer function and much lower levels of genome-wide cleavage. This architecture provides a general means for improving the specificity of ZFNs as gene modification reagents.


Science | 2009

Knockout Rats via Embryo Microinjection of Zinc-Finger Nucleases

Aron M. Geurts; Gregory J. Cost; Yevgeniy Freyvert; Bryan Zeitler; Jeffrey C. Miller; Vivian M. Choi; Shirin S. Jenkins; Adam Wood; Xiaoxia Cui; Xiangdong Meng; Anna I Vincent; Stephen Lam; Mieczyslaw Michalkiewicz; Rebecca Schilling; Jamie Foeckler; Shawn Kalloway; Hartmut Weiler; Séverine Ménoret; Ignacio Anegon; Gregory D. Davis; Lei Zhang; Edward J. Rebar; Philip D. Gregory; Fyodor D. Urnov; Howard J. Jacob; Roland Buelow

Targeted gene disruption in rats paves the way for new human disease models. The toolbox of rat genetics currently lacks the ability to introduce site-directed, heritable mutations into the genome to create knockout animals. By using engineered zinc-finger nucleases (ZFNs) designed to target an integrated reporter and two endogenous rat genes, Immunoglobulin M (IgM) and Rab38, we demonstrate that a single injection of DNA or messenger RNA encoding ZFNs into the one-cell rat embryo leads to a high frequency of animals carrying 25 to 100% disruption at the target locus. These mutations are faithfully and efficiently transmitted through the germline. Our data demonstrate the feasibility of targeted gene disruption in multiple rat strains within 4 months time, paving the way to a humanized monoclonal antibody platform and additional human disease models.


Nature Biotechnology | 2008

Heritable Targeted Gene Disruption in Zebrafish Using Designed Zinc Finger Nucleases

Yannick Doyon; Jasmine M. McCammon; Jeffrey C. Miller; Farhoud Faraji; Catherine Ngo; George E. Katibah; Rainier Amora; Toby D Hocking; Lei Zhang; Edward J. Rebar; Philip D. Gregory; Fyodor Urnov; Sharon L. Amacher

We describe the use of zinc-finger nucleases (ZFNs) for somatic and germline disruption of genes in zebrafish (Danio rerio), in which targeted mutagenesis was previously intractable. ZFNs induce a targeted double-strand break in the genome that is repaired to generate small insertions and deletions. We designed ZFNs targeting the zebrafish golden and no tail/Brachyury (ntl) genes and developed a budding yeast–based assay to identify the most active ZFNs for use in vivo. Injection of ZFN-encoding mRNA into one-cell embryos yielded a high percentage of animals carrying distinct mutations at the ZFN-specified position and exhibiting expected loss-of-function phenotypes. Over half the ZFN mRNA-injected founder animals transmitted disrupted ntl alleles at frequencies averaging 20%. The frequency and precision of gene-disruption events observed suggest that this approach should be applicable to any loci in zebrafish or in other organisms that allow mRNA delivery into the fertilized egg.


Nature | 2009

Precise genome modification in the crop species Zea mays using zinc-finger nucleases

Vipula K. Shukla; Yannick Doyon; Jeffrey C. Miller; Russell DeKelver; Erica A. Moehle; Sarah E. Worden; Jon C. Mitchell; Nicole L. Arnold; Sunita Gopalan; Xiangdong Meng; Vivian M. Choi; Jeremy M. Rock; Ying-Ying Wu; George E. Katibah; Gao Zhifang; David McCaskill; Matthew Simpson; Beth Blakeslee; Scott A. Greenwalt; Holly Butler; Sarah J. Hinkley; Lei Zhang; Edward J. Rebar; Philip D. Gregory; Fyodor Urnov

Agricultural biotechnology is limited by the inefficiencies of conventional random mutagenesis and transgenesis. Because targeted genome modification in plants has been intractable, plant trait engineering remains a laborious, time-consuming and unpredictable undertaking. Here we report a broadly applicable, versatile solution to this problem: the use of designed zinc-finger nucleases (ZFNs) that induce a double-stranded break at their target locus. We describe the use of ZFNs to modify endogenous loci in plants of the crop species Zea mays. We show that simultaneous expression of ZFNs and delivery of a simple heterologous donor molecule leads to precise targeted addition of an herbicide-tolerance gene at the intended locus in a significant number of isolated events. ZFN-modified maize plants faithfully transmit these genetic changes to the next generation. Insertional disruption of one target locus, IPK1, results in both herbicide tolerance and the expected alteration of the inositol phosphate profile in developing seeds. ZFNs can be used in any plant species amenable to DNA delivery; our results therefore establish a new strategy for plant genetic manipulation in basic science and agricultural applications.


Cell | 2011

Generation of isogenic pluripotent stem cells differing exclusively at two early onset Parkinson point mutations

Frank Soldner; Josee Laganiere; Albert W. Cheng; Dirk Hockemeyer; Qing Gao; Raaji K. Alagappan; Vikram Khurana; Lawrence I. Golbe; Richard H. Myers; Susan Lindquist; Lei Zhang; Dmitry Guschin; Lauren K. Fong; B. Joseph Vu; Xiangdong Meng; Fyodor D. Urnov; Edward J. Rebar; Philip D. Gregory; H. Steve Zhang; Rudolf Jaenisch

Patient-specific induced pluripotent stem cells (iPSCs) derived from somatic cells provide a unique tool for the study of human disease, as well as a promising source for cell replacement therapies. One crucial limitation has been the inability to perform experiments under genetically defined conditions. This is particularly relevant for late age onset disorders in which in vitro phenotypes are predicted to be subtle and susceptible to significant effects of genetic background variations. By combining zinc finger nuclease (ZFN)-mediated genome editing and iPSC technology, we provide a generally applicable solution to this problem, generating sets of isogenic disease and control human pluripotent stem cells that differ exclusively at either of two susceptibility variants for Parkinsons disease by modifying the underlying point mutations in the α-synuclein gene. The robust capability to genetically correct disease-causing point mutations in patient-derived hiPSCs represents significant progress for basic biomedical research and an advance toward hiPSC-based cell replacement therapies.


Science | 2011

Targeted Genome Editing Across Species Using ZFNs and TALENs

Andrew J. Wood; Te-Wen Lo; Bryan Zeitler; Catherine S. Pickle; Edward J. Ralston; Andrew H. Lee; Rainier Amora; Jeffrey C. Miller; Elo Leung; Xiangdong Meng; Lei Zhang; Edward J. Rebar; Philip D. Gregory; Fyodor D. Urnov; Barbara J Meyer

Engineered nucleases target specific DNA sequences for gene disruption in nonmodel organisms. Evolutionary studies necessary to dissect diverse biological processes have been limited by the lack of reverse genetic approaches in most organisms with sequenced genomes. We established a broadly applicable strategy using zinc finger nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs) for targeted disruption of endogenous genes and cis-acting regulatory elements in diverged nematode species.

Collaboration


Dive into the Edward J. Rebar'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge