Network


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

Hotspot


Dive into the research topics where Yannick Doyon is active.

Publication


Featured researches published by Yannick Doyon.


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.


Molecular and Cellular Biology | 2004

Structural and functional conservation of the NuA4 histone acetyltransferase complex from yeast to humans

Yannick Doyon; William Selleck; William S. Lane; Song Tan; Jacques Côté

ABSTRACT The NuA4 histone acetyltransferase (HAT) multisubunit complex is responsible for acetylation of histone H4 and H2A N-terminal tails in yeast. Its catalytic component, Esa1, is essential for cell cycle progression, gene-specific regulation and has been implicated in DNA repair. Almost all NuA4 subunits have clear homologues in higher eukaryotes, suggesting that the complex is conserved throughout evolution to metazoans. We demonstrate here that NuA4 complexes are indeed present in human cells. Tip60 and its splice variant Tip60b/PLIP were purified as stable HAT complexes associated with identical polypeptides, with 11 of the 12 proteins being homologs of yeast NuA4 subunits. This indicates a highly conserved subunit composition and the identified human proteins underline the role of NuA4 in the control of mammalian cell proliferation. ING3, a member of the ING family of growth regulators, links NuA4 to p53 function which we confirmed in vivo. Proteins specific to the human NuA4 complexes include ruvB-like helicases and a bromodomain-containing subunit linked to ligand-dependent transcription activation by the thyroid hormone receptor. We also demonstrate that subunits MRG15 and DMAP1 are present in distinct protein complexes harboring histone deacetylase and SWI2-related ATPase activities, respectively. Finally, analogous to yeast, a recombinant trimeric complex formed by Tip60, EPC1, and ING3 is sufficient to reconstitute robust nucleosomal HAT activity in vitro. In conclusion, the NuA4 HAT complex is highly conserved in eukaryotes, in which it plays primary roles in transcription, cellular response to DNA damage, and cell cycle control.


Nature | 2011

In vivo genome editing restores haemostasis in a mouse model of haemophilia

Hojun Li; Virginia Haurigot; Yannick Doyon; Tianjian Li; Sunnie Y. Wong; Anand S. Bhagwat; Nirav Malani; Xavier M. Anguela; Rajiv Sharma; Lacramiora Ivanciu; Samuel L. Murphy; Jonathan D. Finn; Fayaz R. Khazi; Shangzhen Zhou; David Paschon; Edward J. Rebar; Frederic D. Bushman; Philip D. Gregory; Michael C. Holmes; Katherine A. High

Editing of the human genome to correct disease-causing mutations is a promising approach for the treatment of genetic disorders. Genome editing improves on simple gene-replacement strategies by effecting in situ correction of a mutant gene, thus restoring normal gene function under the control of endogenous regulatory elements and reducing risks associated with random insertion into the genome. Gene-specific targeting has historically been limited to mouse embryonic stem cells. The development of zinc finger nucleases (ZFNs) has permitted efficient genome editing in transformed and primary cells that were previously thought to be intractable to such genetic manipulation. In vitro, ZFNs have been shown to promote efficient genome editing via homology-directed repair by inducing a site-specific double-strand break (DSB) at a target locus, but it is unclear whether ZFNs can induce DSBs and stimulate genome editing at a clinically meaningful level in vivo. Here we show that ZFNs are able to induce DSBs efficiently when delivered directly to mouse liver and that, when co-delivered with an appropriately designed gene-targeting vector, they can stimulate gene replacement through both homology-directed and homology-independent targeted gene insertion at the ZFN-specified locus. The level of gene targeting achieved was sufficient to correct the prolonged clotting times in a mouse model of haemophilia B, and remained persistent after induced liver regeneration. Thus, ZFN-driven gene correction can be achieved in vivo, raising the possibility of genome editing as a viable strategy for the treatment of genetic disease.


Nature Methods | 2011

Enhancing zinc-finger-nuclease activity with improved obligate heterodimeric architectures

Yannick Doyon; Thuy D Vo; Matthew C. Mendel; Shon G Greenberg; Jianbin Wang; Danny F Xia; Jeffrey C. Miller; Fyodor D. Urnov; Philip D. Gregory; Michael C. Holmes

Zinc-finger nucleases (ZFNs) drive efficient genome editing by introducing a double-strand break into the targeted gene. Cleavage is induced when two custom-designed ZFNs heterodimerize upon binding DNA to form a catalytically active nuclease complex. The importance of this dimerization event for subsequent cleavage activity has stimulated efforts to engineer the nuclease interface to prevent undesired homodimerization. Here we report the development and application of a yeast-based selection system designed to functionally interrogate the ZFN dimer interface. We identified critical residues involved in dimerization through the isolation of cold-sensitive nuclease domains. We used these residues to engineer ZFNs that have superior cleavage activity while suppressing homodimerization. The improvements were portable to orthogonal domains, allowing the concomitant and independent cleavage of two loci using two different ZFN pairs. These ZFN architectures provide a general means for obtaining highly efficient and specific genome modification.


Plant Molecular Biology | 2009

Targeted transgene integration in plant cells using designed zinc finger nucleases

Charles Q. Cai; Yannick Doyon; W. Michael Ainley; Jeffrey C. Miller; Russell DeKelver; Erica A. Moehle; Jeremy M. Rock; Ya-Li Lee; Robbi Janette Garrison; Lisa Lynn Schulenberg; Ryan C. Blue; Andrew Worden; Lisa W. Baker; Farhoud Faraji; Lei Zhang; Michael C. Holmes; Edward J. Rebar; Trevor Collingwood; Beth Rubin-Wilson; Philip D. Gregory; Fyodor D. Urnov; Joseph F. Petolino

Targeted transgene integration in plants remains a significant technical challenge for both basic and applied research. Here it is reported that designed zinc finger nucleases (ZFNs) can drive site-directed DNA integration into transgenic and native gene loci. A dimer of designed 4-finger ZFNs enabled intra-chromosomal reconstitution of a disabled gfp reporter gene and site-specific transgene integration into chromosomal reporter loci following co-transformation of tobacco cell cultures with a donor construct comprised of sequences necessary to complement a non-functional pat herbicide resistance gene. In addition, a yeast-based assay was used to identify ZFNs capable of cleaving a native endochitinase gene. Agrobacterium delivery of a Ti plasmid harboring both the ZFNs and a donor DNA construct comprising a pat herbicide resistance gene cassette flanked by short stretches of homology to the endochitinase locus yielded up to 10% targeted, homology-directed transgene integration precisely into the ZFN cleavage site. Given that ZFNs can be designed to recognize a wide range of target sequences, these data point toward a novel approach for targeted gene addition, replacement and trait stacking in plants.


Nature Cell Biology | 2011

Rapid and efficient clathrin-mediated endocytosis revealed in genome-edited mammalian cells

Jeffrey B. Doyon; Bryan Zeitler; Jackie Cheng; Aaron T. Cheng; Jennifer M. Cherone; Yolanda Santiago; Andrew H. Lee; Thuy D Vo; Yannick Doyon; Jeffrey C. Miller; David Paschon; Lei Zhang; Edward J. Rebar; Philip D. Gregory; Fyodor D. Urnov; David G. Drubin

Clathrin-mediated endocytosis (CME) is the best-studied pathway by which cells selectively internalize molecules from the plasma membrane and surrounding environment. Previous live-cell imaging studies using ectopically overexpressed fluorescent fusions of endocytic proteins indicated that mammalian CME is a highly dynamic but inefficient and heterogeneous process. In contrast, studies of endocytosis in budding yeast using fluorescent protein fusions expressed at physiological levels from native genomic loci have revealed a process that is very regular and efficient. To analyse endocytic dynamics in mammalian cells in which endogenous protein stoichiometry is preserved, we targeted zinc finger nucleases (ZFNs) to the clathrin light chain A and dynamin-2 genomic loci and generated cell lines expressing fluorescent protein fusions from each locus. The genome-edited cells exhibited enhanced endocytic function, dynamics and efficiency when compared with previously studied cells, indicating that CME is highly sensitive to the levels of its protein components. Our study establishes that ZFN-mediated genome editing is a robust tool for expressing protein fusions at endogenous levels to faithfully report subcellular localization and dynamics.


Nature Methods | 2010

Transient cold shock enhances zinc-finger nuclease–mediated gene disruption

Yannick Doyon; Vivian M. Choi; Danny F Xia; Thuy D Vo; Philip D. Gregory; Michael C. Holmes

Zinc-finger nucleases (ZFNs) are powerful tools for editing the genomes of cell lines and model organisms. Given the breadth of their potential application, simple methods that increase ZFN activity, thus ensuring genome modification, are highly attractive. Here we show that transient hypothermia generally and robustly increased the level of stable, ZFN-induced gene disruption, thereby providing a simple technique to enhance the experimental efficacy of ZFNs.


Molecular Cell | 2009

HBO1 HAT complexes target chromatin throughout gene coding regions via multiple PHD finger interactions with histone H3 tail.

Nehmé Saksouk; Nikita Avvakumov; Karen S. Champagne; Tiffany Hung; Yannick Doyon; Christelle Cayrou; Eric Paquet; Mukta Ullah; Anne-Julie Landry; Valérie Côté; Xiang-Jiao Yang; Or Gozani; Tatiana G. Kutateladze; Jacques Côté

The HBO1 HAT protein is the major source of histone H4 acetylation in vivo and has been shown to play critical roles in gene regulation and DNA replication. A distinctive characteristic of HBO1 HAT complexes is the presence of three PHD finger domains in two different subunits: tumor suppressor proteins ING4/5 and JADE1/2/3. Biochemical and functional analyses indicate that these domains interact with histone H3 N-terminal tail region, but with a different specificity toward its methylation status. Their combinatorial action is essential in regulating chromatin binding and substrate specificity of HBO1 complexes, as well as cell growth. Importantly, localization analyses on the human genome indicate that HBO1 complexes are enriched throughout the coding regions of genes, supporting a role in transcription elongation. These results underline the importance and versatility of PHD finger domains in regulating chromatin association and histone modification crosstalk within a single protein complex.


Molecular and Cellular Biology | 2008

Molecular Architecture of Quartet MOZ/MORF Histone Acetyltransferase Complexes.

Mukta Ullah; Nadine Pelletier; Lin Xiao; Song Ping Zhao; Kainan Wang; Cindy Degerny; Soroush Tahmasebi; Christelle Cayrou; Yannick Doyon; Siew-Lee Goh; Nathalie Champagne; Jacques Côté; Xiang-Jiao Yang

ABSTRACT The monocytic leukemia zinc finger protein MOZ and the related factor MORF form tetrameric complexes with ING5 (inhibitor of growth 5), EAF6 (Esa1-associated factor 6 ortholog), and the bromodomain-PHD finger protein BRPF1, -2, or -3. To gain new insights into the structure, function, and regulation of these complexes, we reconstituted them and performed various molecular analyses. We found that BRPF proteins bridge the association of MOZ and MORF with ING5 and EAF6. An N-terminal region of BRPF1 interacts with the acetyltransferases; the enhancer of polycomb (EPc) homology domain in the middle part binds to ING5 and EAF6. The association of BRPF1 with EAF6 is weak, but ING5 increases the affinity. These three proteins form a trimeric core that is conserved from Drosophila melanogaster to humans, although authentic orthologs of MOZ and MORF are absent in invertebrates. Deletion mapping studies revealed that the acetyltransferase domain of MOZ/MORF is sufficient for BRPF1 interaction. At the functional level, complex formation with BRPF1 and ING5 drastically stimulates the activity of the acetyltransferase domain in acetylation of nucleosomal histone H3 and free histones H3 and H4. An unstructured 18-residue region at the C-terminal end of the catalytic domain is required for BRPF1 interaction and may function as an “activation lid.” Furthermore, BRPF1 enhances the transcriptional potential of MOZ and a leukemic MOZ-TIF2 fusion protein. These findings thus indicate that BRPF proteins play a key role in assembling and activating MOZ/MORF acetyltransferase complexes.

Collaboration


Dive into the Yannick Doyon'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

Katherine A. High

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Rajiv Sharma

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Shangzhen Zhou

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Xavier M. Anguela

Children's Hospital of Philadelphia

View shared research outputs
Researchain Logo
Decentralizing Knowledge