44 results on '"Doyon Y"'
Search Results
2. CRISPR-driven modeling of clinically relevant genetic variants in hematopoietic stem and progenitor-derived erythroid cells
- Author
-
Boccacci, Y., primary, Dumont, N., additional, Doyon, Y., additional, and Laganiere, J., additional
- Published
- 2018
- Full Text
- View/download PDF
3. 369 - CRISPR-driven modeling of clinically relevant genetic variants in hematopoietic stem and progenitor-derived erythroid cells
- Author
-
Boccacci, Y., Dumont, N., Doyon, Y., and Laganiere, J.
- Published
- 2018
- Full Text
- View/download PDF
4. IL-1 Gene Deletion Protects Oligodendrocytes after Spinal Cord Injury through Upregulation of the Survival Factor Tox3
- Author
-
Bastien, D., primary, Bellver Landete, V., additional, Lessard, M., additional, Vallieres, N., additional, Champagne, M., additional, Takashima, A., additional, Tremblay, M.-E., additional, Doyon, Y., additional, and Lacroix, S., additional
- Published
- 2015
- Full Text
- View/download PDF
5. In vivo dissection of the mouse tyrosine catabolic pathway with CRISPR-Cas9 identifies modifier genes affecting hereditary tyrosinemia type 1.
- Author
-
Rivest JF, Carter S, Goupil C, Antérieux P, Cyr D, Ung RV, Dal Soglio D, Mac-Way F, Waters PJ, Paganelli M, and Doyon Y
- Subjects
- Animals, Mice, Genes, Modifier, Liver metabolism, Hydrolases genetics, Disease Models, Animal, Mutation, Tyrosinemias genetics, CRISPR-Cas Systems, Gene Editing methods, Tyrosine metabolism
- Abstract
Hereditary tyrosinemia type 1 is an autosomal recessive disorder caused by mutations (pathogenic variants) in fumarylacetoacetate hydrolase, an enzyme involved in tyrosine degradation. Its loss results in the accumulation of toxic metabolites that mainly affect the liver and kidneys and can lead to severe liver disease and liver cancer. Tyrosinemia type 1 has a global prevalence of approximately 1 in 100,000 births but can reach up to 1 in 1,500 births in some regions of Québec, Canada. Mutating functionally related "modifier' genes (i.e. genes that, when mutated, affect the phenotypic impacts of mutations in other genes) is an emerging strategy for treating human genetic diseases. In vivo somatic genome editing in animal models of these diseases is a powerful means to identify modifier genes and fuel treatment development. In this study, we demonstrate that mutating additional enzymes in the tyrosine catabolic pathway through liver-specific genome editing can relieve or worsen the phenotypic severity of a murine model of tyrosinemia type 1. Neonatal gene delivery using recombinant adeno-associated viral vectors expressing Staphylococcus aureus Cas9 under the control of a liver-specific promoter led to efficient gene disruption and metabolic rewiring of the pathway, with systemic effects that were distinct from the phenotypes observed in whole-body knockout models. Our work illustrates the value of using in vivo genome editing in model organisms to study the direct effects of combining pathological mutations with modifier gene mutations in isogenic settings., Competing Interests: Conflicts of interest The authors declare that they have no competing interests., (© The Author(s) 2024. Published by Oxford University Press on behalf of The Genetics Society of America.)
- Published
- 2024
- Full Text
- View/download PDF
6. Dairy phages escape CRISPR defence of Streptococcus thermophilus via the anti-CRISPR AcrIIA3.
- Author
-
Pastuszka A, Rousseau GM, Somerville V, Levesque S, Fiset JP, Goulet A, Doyon Y, and Moineau S
- Subjects
- Humans, Streptococcus thermophilus genetics, Streptococcus thermophilus metabolism, CRISPR-Cas Systems genetics, Gene Editing, Bacteriophages genetics, Bacteriophages metabolism, Streptococcus Phages genetics
- Abstract
Bacterial community collapse due to phage infection is a major risk in cheese making processes. As virulent phages are ubiquitous and diverse in milk fermentation factories, the use of phage-resistant lactic acid bacteria (LAB) is essential to obtain high-quality fermented dairy products. The LAB species Streptococcus thermophilus contains two type II-A CRISPR-Cas systems (CRISPR1 and CRISPR3) that can effectively protect against phage infection. However, virulent streptococcal phages carrying anti-CRISPR proteins (ACR) that block the activity of CRISPR-Cas systems have emerged in yogurt and cheese environments. For example, phages carrying AcrIIA5 can impede both CRISPR1 and CRISPR3 systems, while AcrIIA6 stops only CRISPR1. Here, we explore the activity and diversity of a third streptococcal phage anti-CRISPR protein, namely AcrIIA3. We were able to demonstrate that AcrIIA3 is efficiently active against the CRISPR3-Cas system of S. thermophilus. We used AlphaFold2 to infer the structure of AcrIIA3 and we predicted that this new family of functional ACR in virulent streptococcal phages has a new α-helical fold, with no previously identified structural homologs. Because ACR proteins are being explored as modulators in genome editing applications, we also tested AcrIIA3 against SpCas9. We found that AcrIIA3 could block SpCas9 in bacteria but not in human cells. Understanding the diversity and functioning of anti-defence mechanisms will be of importance in the design of long-term stable starter cultures., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
7. Accessory-cell-free differentiation of hematopoietic stem and progenitor cells into mature red blood cells.
- Author
-
Boccacci Y, Dumont N, Doyon Y, and Laganière J
- Subjects
- Animals, Adult, Humans, Cell Differentiation genetics, Erythropoiesis, Erythrocytes, Hematopoietic Stem Cells
- Abstract
Background Aims: The culture and ex vivo engineering of red blood cells (RBCs) can help characterize genetic variants, model diseases, and may eventually spur the development of applications in transfusion medicine. In the last decade, improvements to the in vitro production of RBCs have enabled efficient erythroid progenitor proliferation and high enucleation levels from several sources of hematopoietic stem and progenitor cells (HSPCs). Despite these advances, there remains a need for refining the terminal step of in vitro human erythropoiesis, i.e., the terminal maturation of reticulocytes into erythrocytes, so that it can occur without feeder or accessory cells and animal-derived components., Methods: Here, we describe the near-complete erythroid differentiation of cultured RBCs (cRBCs) from adult HSPCs in accessory-cell-free and xeno-free conditions., Results: The approach improves post-enucleation cell integrity and cell survival, and it enables subsequent storage of cRBCs for up to 42 days in classical additive solution conditions without any specialized equipment., Conclusions: We foresee that these improvements will facilitate the characterization of RBCs derived from gene-edited HSPCs., Competing Interests: Declaration of Competing Interest The authors have no commercial, proprietary or financial interest in the products or companies described in this article., (Copyright © 2023 International Society for Cell & Gene Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
8. Marker-free co-selection for successive rounds of prime editing in human cells.
- Author
-
Levesque S, Mayorga D, Fiset JP, Goupil C, Duringer A, Loiselle A, Bouchard E, Agudelo D, and Doyon Y
- Subjects
- DNA genetics, Gene Editing, Humans, Sodium, CRISPR-Cas Systems, Sodium-Potassium-Exchanging ATPase genetics
- Abstract
Prime editing enables the introduction of precise point mutations, small insertions, or short deletions without requiring donor DNA templates. However, efficiency remains a key challenge in a broad range of human cell types. In this work, we design a robust co-selection strategy through coediting of the ubiquitous and essential sodium/potassium pump (Na
+ /K+ ATPase). We readily engineer highly modified pools of cells and clones with homozygous modifications for functional studies with minimal pegRNA optimization. This process reveals that nicking the non-edited strand stimulates multiallelic editing but often generates tandem duplications and large deletions at the target site, an outcome dictated by the relative orientation of the protospacer adjacent motifs. Our approach streamlines the production of cell lines with multiple genetic modifications to create cellular models for biological research and lays the foundation for the development of cell-type specific co-selection strategies., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
9. Recurrent chromosomal translocations in sarcomas create a megacomplex that mislocalizes NuA4/TIP60 to Polycomb target loci.
- Author
-
Sudarshan D, Avvakumov N, Lalonde ME, Alerasool N, Joly-Beauparlant C, Jacquet K, Mameri A, Lambert JP, Rousseau J, Lachance C, Paquet E, Herrmann L, Thonta Setty S, Loehr J, Bernardini MQ, Rouzbahman M, Gingras AC, Coulombe B, Droit A, Taipale M, Doyon Y, and Côté J
- Subjects
- Chromatin, DNA-Binding Proteins metabolism, Female, Histones metabolism, Humans, Polycomb Repressive Complex 2 genetics, Polycomb Repressive Complex 2 metabolism, Polycomb-Group Proteins genetics, Polycomb-Group Proteins metabolism, Translocation, Genetic genetics, Endometrial Neoplasms genetics, Endometrial Neoplasms metabolism, Endometrial Neoplasms pathology, Sarcoma genetics, Sarcoma, Endometrial Stromal genetics, Sarcoma, Endometrial Stromal metabolism, Sarcoma, Endometrial Stromal pathology
- Abstract
Chromosomal translocations frequently promote carcinogenesis by producing gain-of-function fusion proteins. Recent studies have identified highly recurrent chromosomal translocations in patients with endometrial stromal sarcomas (ESSs) and ossifying fibromyxoid tumors (OFMTs), leading to an in-frame fusion of PHF1 (PCL1) to six different subunits of the NuA4/TIP60 complex. While NuA4/TIP60 is a coactivator that acetylates chromatin and loads the H2A.Z histone variant, PHF1 is part of the Polycomb repressive complex 2 (PRC2) linked to transcriptional repression of key developmental genes through methylation of histone H3 on lysine 27. In this study, we characterize the fusion protein produced by the EPC1 - PHF1 translocation. The chimeric protein assembles a megacomplex harboring both NuA4/TIP60 and PRC2 activities and leads to mislocalization of chromatin marks in the genome, in particular over an entire topologically associating domain including part of the HOXD cluster. This is linked to aberrant gene expression-most notably increased expression of PRC2 target genes. Furthermore, we show that JAZF1-implicated with a PRC2 component in the most frequent translocation in ESSs, JAZF1-SUZ12 -is a potent transcription activator that physically associates with NuA4/TIP60, its fusion creating outcomes similar to those of EPC1-PHF1 Importantly, the specific increased expression of PRC2 targets/ HOX genes was also confirmed with ESS patient samples. Altogether, these results indicate that most chromosomal translocations linked to these sarcomas use the same molecular oncogenic mechanism through a physical merge of NuA4/TIP60 and PRC2 complexes, leading to mislocalization of histone marks and aberrant Polycomb target gene expression., (© 2022 Sudarshan et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2022
- Full Text
- View/download PDF
10. Rewired Cas9s with Minimal Sequence Constraints.
- Author
-
Levesque S, Agudelo D, and Doyon Y
- Subjects
- Gene Editing, CRISPR-Cas Systems genetics, Clustered Regularly Interspaced Short Palindromic Repeats
- Abstract
The genome editing toolkit is ever expanding. Although CRISPR-Cas systems can target virtually any gene, single-nucleotide resolution is yet to be achieved. Walton and colleagues engineered nucleases and base editors compatible with every protospacer adjacent motif (PAM) to achieve high-precision targeting. Their findings revealed the striking plasticity of Cas9., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
11. Versatile and robust genome editing with Streptococcus thermophilus CRISPR1-Cas9.
- Author
-
Agudelo D, Carter S, Velimirovic M, Duringer A, Rivest JF, Levesque S, Loehr J, Mouchiroud M, Cyr D, Waters PJ, Laplante M, Moineau S, Goulet A, and Doyon Y
- Subjects
- Animals, CRISPR-Associated Protein 9 chemistry, Cell Line, Cells, Cultured, DNA Cleavage, Humans, Mammals, Mice, Mice, Knockout, Structure-Activity Relationship, Substrate Specificity, CRISPR-Associated Protein 9 metabolism, CRISPR-Cas Systems, Clustered Regularly Interspaced Short Palindromic Repeats, Gene Editing, Streptococcus thermophilus enzymology, Streptococcus thermophilus genetics
- Abstract
Targeting definite genomic locations using CRISPR-Cas systems requires a set of enzymes with unique protospacer adjacent motif (PAM) compatibilities. To expand this repertoire, we engineered nucleases, cytosine base editors, and adenine base editors from the archetypal Streptococcus thermophilus CRISPR1-Cas9 (St1Cas9) system. We found that St1Cas9 strain variants enable targeting to five distinct A-rich PAMs and provide a structural basis for their specificities. The small size of this ortholog enables expression of the holoenzyme from a single adeno-associated viral vector for in vivo editing applications. Delivery of St1Cas9 to the neonatal liver efficiently rewired metabolic pathways, leading to phenotypic rescue in a mouse model of hereditary tyrosinemia. These robust enzymes expand and complement current editing platforms available for tailoring mammalian genomes., (© 2020 Agudelo et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2020
- Full Text
- View/download PDF
12. Cas9 Allosteric Inhibition by the Anti-CRISPR Protein AcrIIA6.
- Author
-
Fuchsbauer O, Swuec P, Zimberger C, Amigues B, Levesque S, Agudelo D, Duringer A, Chaves-Sanjuan A, Spinelli S, Rousseau GM, Velimirovic M, Bolognesi M, Roussel A, Cambillau C, Moineau S, Doyon Y, and Goulet A
- Subjects
- Allosteric Regulation, Bacteriophages genetics, Binding Sites, CRISPR-Associated Protein 9 genetics, CRISPR-Associated Protein 9 ultrastructure, DNA genetics, DNA ultrastructure, Escherichia coli enzymology, Escherichia coli genetics, Humans, K562 Cells, Kinetics, Mutation, Protein Binding, Protein Conformation, Streptococcus thermophilus genetics, Structure-Activity Relationship, Viral Proteins genetics, Viral Proteins ultrastructure, Bacteriophages metabolism, CRISPR-Associated Protein 9 metabolism, CRISPR-Cas Systems, Clustered Regularly Interspaced Short Palindromic Repeats, DNA metabolism, Streptococcus thermophilus enzymology, Viral Proteins metabolism
- Abstract
In the arms race against bacteria, bacteriophages have evolved diverse anti-CRISPR proteins (Acrs) that block CRISPR-Cas immunity. Acrs play key roles in the molecular coevolution of bacteria with their predators, use a variety of mechanisms of action, and provide tools to regulate Cas-based genome manipulation. Here, we present structural and functional analyses of AcrIIA6, an Acr from virulent phages, exploring its unique anti-CRISPR action. Our cryo-EM structures and functional data of AcrIIA6 binding to Streptococcus thermophilus Cas9 (St1Cas9) show that AcrIIA6 acts as an allosteric inhibitor and induces St1Cas9 dimerization. AcrIIA6 reduces St1Cas9 binding affinity for DNA and prevents DNA binding within cells. The PAM and AcrIIA6 recognition sites are structurally close and allosterically linked. Mechanistically, AcrIIA6 affects the St1Cas9 conformational dynamics associated with PAM binding. Finally, we identify a natural St1Cas9 variant resistant to AcrIIA6 illustrating Acr-driven mutational escape and molecular diversification of Cas9 proteins., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
13. Widespread anti-CRISPR proteins in virulent bacteriophages inhibit a range of Cas9 proteins.
- Author
-
Hynes AP, Rousseau GM, Agudelo D, Goulet A, Amigues B, Loehr J, Romero DA, Fremaux C, Horvath P, Doyon Y, Cambillau C, and Moineau S
- Subjects
- Bacteriophages genetics, CRISPR-Associated Protein 9 genetics, CRISPR-Cas Systems genetics, CRISPR-Cas Systems physiology, Gene Editing, Humans, Virulence genetics, Virulence physiology, CRISPR-Associated Protein 9 physiology, Clustered Regularly Interspaced Short Palindromic Repeats genetics
- Abstract
CRISPR-Cas systems are bacterial anti-viral systems, and bacterial viruses (bacteriophages, phages) can carry anti-CRISPR (Acr) proteins to evade that immunity. Acrs can also fine-tune the activity of CRISPR-based genome-editing tools. While Acrs are prevalent in phages capable of lying dormant in a CRISPR-carrying host, their orthologs have been observed only infrequently in virulent phages. Here we identify AcrIIA6, an Acr encoded in 33% of virulent Streptococcus thermophilus phage genomes. The X-ray structure of AcrIIA6 displays some features unique to this Acr family. We compare the activity of AcrIIA6 to those of other Acrs, including AcrIIA5 (also from S. thermophilus phages), and characterize their effectiveness against a range of CRISPR-Cas systems. Finally, we demonstrate that both Acr families from S. thermophilus phages inhibit Cas9-mediated genome editing of human cells.
- Published
- 2018
- Full Text
- View/download PDF
14. Marker-free coselection for CRISPR-driven genome editing in human cells.
- Author
-
Agudelo D, Duringer A, Bozoyan L, Huard CC, Carter S, Loehr J, Synodinou D, Drouin M, Salsman J, Dellaire G, Laganière J, and Doyon Y
- Subjects
- Genetic Markers genetics, Humans, CRISPR-Cas Systems genetics, Cells, Cultured physiology, DNA Repair genetics, Gene Editing methods, Mutagenesis, Site-Directed
- Abstract
Targeted genome editing enables the creation of bona fide cellular models for biological research and may be applied to human cell-based therapies. Therefore, broadly applicable and versatile methods for increasing its efficacy in cell populations are highly desirable. We designed a simple and robust coselection strategy for enrichment of cells with either nuclease-driven nonhomologous end joining (NHEJ) or homology-directed repair (HDR) events by harnessing the multiplexing capabilities of CRISPR-Cas9 and Cpf1 systems. Selection for dominant alleles of the ubiquitous sodium/potassium pump (Na
+ /K+ ATPase) that rendered cells resistant to ouabain was used to enrich for custom genetic modifications at another unlinked locus of interest, thereby effectively increasing the recovery of engineered cells. The process is readily adaptable to transformed and primary cells, including hematopoietic stem and progenitor cells. The use of universal CRISPR reagents and a commercially available small-molecule inhibitor streamlines the incorporation of marker-free genetic changes in human cells.- Published
- 2017
- Full Text
- View/download PDF
15. Gene Therapy in Tyrosinemia: Potential and Pitfalls.
- Author
-
Carter S and Doyon Y
- Subjects
- Animals, Clustered Regularly Interspaced Short Palindromic Repeats genetics, Gene Transfer Techniques, Genetic Therapy methods, Humans, Liver metabolism, Tyrosinemias genetics, Tyrosinemias therapy
- Abstract
In this chapter, we intend to review gene therapy concepts applied to the potential treatment of tyrosinemia for parents and pediatricians. Therefore, our main objective is to give general informations in a comprehensible manner. Considering the nature of tyrosinemia and the current state of technology, a particular focus will be put on strategies using viral delivery of DNA to the liver. In light of the recent development of the CRISPR technology and the revival of promises for previously unavailable therapeutical tools, the present chapter aims at presenting up to date facts and potential pitfalls towards an application for metabolic diseases, in particular tyrosinemia.
- Published
- 2017
- Full Text
- View/download PDF
16. The TIP60 Complex Regulates Bivalent Chromatin Recognition by 53BP1 through Direct H4K20me Binding and H2AK15 Acetylation.
- Author
-
Jacquet K, Fradet-Turcotte A, Avvakumov N, Lambert JP, Roques C, Pandita RK, Paquet E, Herst P, Gingras AC, Pandita TK, Legube G, Doyon Y, Durocher D, and Côté J
- Subjects
- Acetylation, Binding Sites, Binding, Competitive, CRISPR-Cas Systems, Chromosomal Proteins, Non-Histone genetics, DNA Breaks, Double-Stranded, DNA Repair, Histone Acetyltransferases genetics, Histones genetics, Humans, K562 Cells, Lysine Acetyltransferase 5, Promoter Regions, Genetic, Protein Binding, RNA Interference, Repressor Proteins genetics, Repressor Proteins metabolism, Signal Transduction, Time Factors, Transcription, Genetic, Transfection, Tumor Suppressor p53-Binding Protein 1 genetics, Ubiquitination, Chromatin Assembly and Disassembly, Chromosomal Proteins, Non-Histone metabolism, Histone Acetyltransferases metabolism, Histones metabolism, Protein Processing, Post-Translational, Tumor Suppressor p53-Binding Protein 1 metabolism
- Abstract
The NuA4/TIP60 acetyltransferase complex is a key regulator of genome expression and stability. Here we identified MBTD1 as a stable subunit of the complex, and we reveal that, via a histone reader domain for H4K20me1/2, MBTD1 allows TIP60 to associate with specific gene promoters and to promote the repair of DNA double-strand breaks by homologous recombination. It was previously suggested that TIP60-dependent acetylation of H4 regulates binding of the non-homologous end joining factor 53BP1, which engages chromatin through simultaneous binding of H4K20me2 and H2AK15ub. We find that the TIP60 complex regulates association of 53BP1 partly by competing for H4K20me2 and by regulating H2AK15ub. Ubiquitylation of H2AK15 by RNF168 inhibits chromatin acetylation by TIP60, while this residue can be acetylated by TIP60 in vivo, blocking its ubiquitylation. Altogether, these results uncover an intricate mechanism orchestrated by the TIP60 complex to regulate 53BP1-dependent repair through competitive bivalent binding and modification of chromatin., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
17. Preparation and Analysis of Native Chromatin-Modifying Complexes.
- Author
-
Doyon Y and Côté J
- Subjects
- Cell Line, Chromatin metabolism, Chromosomal Proteins, Non-Histone chemistry, Chromosomal Proteins, Non-Histone genetics, Chromosomal Proteins, Non-Histone metabolism, Endonucleases metabolism, Enhancer of Zeste Homolog 2 Protein chemistry, Enhancer of Zeste Homolog 2 Protein genetics, Enhancer of Zeste Homolog 2 Protein metabolism, Gene Targeting methods, Histone Acetyltransferases chemistry, Histone Acetyltransferases metabolism, Histone Methyltransferases, Histone-Lysine N-Methyltransferase chemistry, Histone-Lysine N-Methyltransferase metabolism, Humans, Repressor Proteins chemistry, Repressor Proteins genetics, Repressor Proteins metabolism, Transcription Activator-Like Effector Nucleases metabolism, Zinc Fingers, CRISPR-Cas Systems, Gene Editing methods, Genetic Engineering methods, Histone Acetyltransferases genetics, Histone-Lysine N-Methyltransferase genetics
- Abstract
Nucleosomes, the basic units of chromatin, are decorated with a myriad of posttranslational modifications (PTMs) by the action of chromatin modifiers. These enzymes function almost exclusively as part of stable protein complexes that assist their recruitment to specific genomic loci, specify their substrate, and provide allosteric control. By altering the interactions within nucleosomes or with neighboring nucleosomes and serving as a platform to engage effector proteins, PTMs deposited by histone-modifying complexes influence virtually every nuclear process and are at the heart of the epigenetic mechanisms. Hence, it is critical to identify their components, define their structures, and characterize their biochemical activities. Here we describe protocols for tandem affinity purification (TAP) of native histone acetyltransferase (HAT) and methyltransferase (HMT) complexes from human cells engineered to express bait proteins from a genomic safe harbor or their endogenous chromosomal genes, using zinc-finger nucleases (ZFNs), TAL effector nucleases (TALENs), and clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 systems. The approaches presented aim to preserve natural transcriptional and posttranscriptional regulation and minimize biochemical artifacts due to ectopic expression. Near homogenous preparations of native complexes are obtained in sufficient amounts to perform biochemical assays and characterize their components., (© 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
18. A Scalable Genome-Editing-Based Approach for Mapping Multiprotein Complexes in Human Cells.
- Author
-
Dalvai M, Loehr J, Jacquet K, Huard CC, Roques C, Herst P, Côté J, and Doyon Y
- Subjects
- Cell Line, Tumor, Genome, Human, Humans, Protein Binding, CRISPR-Cas Systems, Gene Targeting methods, Minichromosome Maintenance Proteins chemistry, Proteomics methods
- Abstract
Conventional affinity purification followed by mass spectrometry (AP-MS) analysis is a broadly applicable method used to decipher molecular interaction networks and infer protein function. However, it is sensitive to perturbations induced by ectopically overexpressed target proteins and does not reflect multilevel physiological regulation in response to diverse stimuli. Here, we developed an interface between genome editing and proteomics to isolate native protein complexes produced from their natural genomic contexts. We used CRISPR/Cas9 and TAL effector nucleases (TALENs) to tag endogenous genes and purified several DNA repair and chromatin-modifying holoenzymes to near homogeneity. We uncovered subunits and interactions among well-characterized complexes and report the isolation of MCM8/9, highlighting the efficiency and robustness of the approach. These methods improve and simplify both small- and large-scale explorations of protein interactions as well as the study of biochemical activities and structure-function relationships., (Copyright © 2015 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
19. In vivo genome editing of the albumin locus as a platform for protein replacement therapy.
- Author
-
Sharma R, Anguela XM, Doyon Y, Wechsler T, DeKelver RC, Sproul S, Paschon DE, Miller JC, Davidson RJ, Shivak D, Zhou S, Rieders J, Gregory PD, Holmes MC, Rebar EJ, and High KA
- Subjects
- Albumins metabolism, Animals, Dependovirus genetics, Endonucleases, Fabry Disease genetics, Fabry Disease therapy, Factor IX genetics, Factor VIII genetics, Gaucher Disease genetics, Gaucher Disease therapy, Genetic Vectors administration & dosage, Hemophilia A genetics, Hemophilia A therapy, Hemophilia B genetics, Hemophilia B therapy, High-Throughput Nucleotide Sequencing, Humans, Lysosomes enzymology, Mice, Mice, Inbred C57BL, Mucopolysaccharidosis I genetics, Mucopolysaccharidosis I therapy, Mucopolysaccharidosis II genetics, Mucopolysaccharidosis II therapy, Promoter Regions, Genetic genetics, RNA Editing, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Zinc Fingers, Albumins genetics, Enzyme Replacement Therapy, Genetic Therapy, Genome, Liver metabolism, Transgenes physiology
- Abstract
Site-specific genome editing provides a promising approach for achieving long-term, stable therapeutic gene expression. Genome editing has been successfully applied in a variety of preclinical models, generally focused on targeting the diseased locus itself; however, limited targeting efficiency or insufficient expression from the endogenous promoter may impede the translation of these approaches, particularly if the desired editing event does not confer a selective growth advantage. Here we report a general strategy for liver-directed protein replacement therapies that addresses these issues: zinc finger nuclease (ZFN) -mediated site-specific integration of therapeutic transgenes within the albumin gene. By using adeno-associated viral (AAV) vector delivery in vivo, we achieved long-term expression of human factors VIII and IX (hFVIII and hFIX) in mouse models of hemophilia A and B at therapeutic levels. By using the same targeting reagents in wild-type mice, lysosomal enzymes were expressed that are deficient in Fabry and Gaucher diseases and in Hurler and Hunter syndromes. The establishment of a universal nuclease-based platform for secreted protein production would represent a critical advance in the development of safe, permanent, and functional cures for diverse genetic and nongenetic diseases., (© 2015 by The American Society of Hematology.)
- Published
- 2015
- Full Text
- View/download PDF
20. Robust ZFN-mediated genome editing in adult hemophilic mice.
- Author
-
Anguela XM, Sharma R, Doyon Y, Miller JC, Li H, Haurigot V, Rohde ME, Wong SY, Davidson RJ, Zhou S, Gregory PD, Holmes MC, and High KA
- Subjects
- Animals, Dependovirus genetics, Disease Models, Animal, Endonucleases metabolism, Factor IX genetics, Factor IX metabolism, Hemophilia B genetics, Hemophilia B pathology, Liver metabolism, Male, Mice, Mice, Transgenic, Protein Multimerization, Endonucleases genetics, Factor IX biosynthesis, Genetic Therapy methods, Genetic Vectors, Genome, Hemophilia B therapy, Zinc Fingers genetics
- Abstract
Monogenic diseases, including hemophilia, represent ideal targets for genome-editing approaches aimed at correcting a defective gene. Here we report that systemic adeno-associated virus (AAV) vector delivery of zinc finger nucleases (ZFNs) and corrective donor template to the predominantly quiescent livers of adult mice enables production of high levels of human factor IX in a murine model of hemophilia B. Further, we show that off-target cleavage can be substantially reduced while maintaining robust editing by using obligate heterodimeric ZFNs engineered to minimize unwanted cleavage attributable to homodimerization of the ZFNs. These results broaden the therapeutic potential of AAV/ZFN-mediated genome editing in the liver and could expand this strategy to other nonreplicating cell types.
- Published
- 2013
- Full Text
- View/download PDF
21. Cancer translocations in human cells induced by zinc finger and TALE nucleases.
- Author
-
Piganeau M, Ghezraoui H, De Cian A, Guittat L, Tomishima M, Perrouault L, René O, Katibah GE, Zhang L, Holmes MC, Doyon Y, Concordet JP, Giovannangeli C, Jasin M, and Brunet E
- Subjects
- Cell Line, Chromosome Breakpoints, Humans, Nucleophosmin, Protein-Tyrosine Kinases genetics, Sarcoma, Ewing genetics, Sarcoma, Ewing metabolism, Endonucleases metabolism, Neoplasms enzymology, Neoplasms genetics, Translocation, Genetic, Zinc Fingers
- Abstract
Chromosomal translocations are signatures of numerous cancers and lead to expression of fusion genes that act as oncogenes. The wealth of genomic aberrations found in cancer, however, makes it challenging to assign a specific phenotypic change to a specific aberration. In this study, we set out to use genome editing with zinc finger (ZFN) and transcription activator-like effector (TALEN) nucleases to engineer, de novo, translocation-associated oncogenes at cognate endogenous loci in human cells. Using ZFNs and TALENs designed to cut precisely at relevant translocation breakpoints, we induced cancer-relevant t(11;22)(q24;q12) and t(2;5)(p23;q35) translocations found in Ewing sarcoma and anaplastic large cell lymphoma (ALCL), respectively. We recovered both translocations with high efficiency, resulting in the expression of the EWSR1-FLI1 and NPM1-ALK fusions. Breakpoint junctions recovered after ZFN cleavage in human embryonic stem (ES) cell-derived mesenchymal precursor cells fully recapitulated the genomic characteristics found in tumor cells from Ewing sarcoma patients. This approach with tailored nucleases demonstrates that expression of fusion genes found in cancer cells can be induced from the native promoter, allowing interrogation of both the underlying mechanisms and oncogenic consequences of tumor-related translocations in human cells. With an analogous strategy, the ALCL translocation was reverted in a patient cell line to restore the integrity of the two participating chromosomes, further expanding the repertoire of genomic rearrangements that can be engineered by tailored nucleases.
- Published
- 2013
- Full Text
- View/download PDF
22. Targeted gene addition to a predetermined site in the human genome using a ZFN-based nicking enzyme.
- Author
-
Wang J, Friedman G, Doyon Y, Wang NS, Li CJ, Miller JC, Hua KL, Yan JJ, Babiarz JE, Gregory PD, and Holmes MC
- Subjects
- Amino Acid Sequence, Catalytic Domain, Cell Line, Transformed, Cell Line, Tumor, Cloning, Molecular, DNA Breaks, Double-Stranded, DNA Breaks, Single-Stranded, DNA End-Joining Repair, Deoxyribonucleases, Type II Site-Specific genetics, Fibroblasts cytology, Fibroblasts metabolism, Genetic Vectors, Histones metabolism, Humans, INDEL Mutation, Molecular Sequence Data, Protein Engineering methods, Receptors, CCR5 genetics, Recombinant Fusion Proteins genetics, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae metabolism, Transformation, Genetic, Deoxyribonucleases, Type II Site-Specific metabolism, Gene Targeting methods, Genome, Human, Recombinant Fusion Proteins metabolism, Zinc Fingers
- Abstract
Zinc-finger nucleases (ZFNs) drive highly efficient genome editing by generating a site-specific DNA double-strand break (DSB) at a predetermined site in the genome. Subsequent repair of this break via the nonhomologous end-joining (NHEJ) or homology-directed repair (HDR) pathways results in targeted gene disruption or gene addition, respectively. Here, we report that ZFNs can be engineered to induce a site-specific DNA single-strand break (SSB) or nick. Using the CCR5-specific ZFNs as a model system, we show that introduction of a nick at this target site stimulates gene addition using a homologous donor template but fails to induce significant levels of the small insertions and deletions (indels) characteristic of repair via NHEJ. Gene addition by these CCR5-targeted zinc finger nickases (ZFNickases) occurs in both transformed and primary human cells at efficiencies of up to ∼1%-8%. Interestingly, ZFNickases targeting the AAVS1 "safe harbor" locus revealed similar in vitro nicking activity, a marked reduction of indels characteristic of NHEJ, but stimulated far lower levels of gene addition-suggesting that other, yet to be identified mediators of nick-induced gene targeting exist. Introduction of site-specific nicks at distinct endogenous loci provide an important tool for the study of DNA repair. Moreover, the potential for a SSB to direct repair pathway choice (i.e., HDR but not NHEJ) may prove advantageous for certain therapeutic applications such as the targeted correction of human disease-causing mutations.
- Published
- 2012
- Full Text
- View/download PDF
23. Conserved molecular interactions within the HBO1 acetyltransferase complexes regulate cell proliferation.
- Author
-
Avvakumov N, Lalonde ME, Saksouk N, Paquet E, Glass KC, Landry AJ, Doyon Y, Cayrou C, Robitaille GA, Richard DE, Yang XJ, Kutateladze TG, and Côté J
- Subjects
- Cell Line, Histone Acetyltransferases chemistry, Histones chemistry, Histones metabolism, Humans, Protein Structure, Tertiary, Tumor Suppressor Proteins chemistry, Tumor Suppressor Proteins metabolism, p21-Activated Kinases chemistry, p21-Activated Kinases metabolism, Cell Proliferation, Histone Acetyltransferases metabolism
- Abstract
Acetyltransferase complexes of the MYST family with distinct substrate specificities and functions maintain a conserved association with different ING tumor suppressor proteins. ING complexes containing the HBO1 acetylase are a major source of histone H3 and H4 acetylation in vivo and play critical roles in gene regulation and DNA replication. Here, our molecular dissection of HBO1/ING complexes unravels the protein domains required for their assembly and function. Multiple PHD finger domains present in different subunits bind the histone H3 N-terminal tail with a distinct specificity toward lysine 4 methylation status. We show that natively regulated association of the ING4/5 PHD domain with HBO1-JADE determines the growth inhibitory function of the complex, linked to its tumor suppressor activity. Functional genomic analyses indicate that the p53 pathway is a main target of the complex, at least in part through direct transcription regulation at the initiation site of p21/CDKN1A. These results demonstrate the importance of ING association with MYST acetyltransferases in controlling cell proliferation, a regulated link that accounts for the reported tumor suppressor activities of these complexes.
- Published
- 2012
- Full Text
- View/download PDF
24. In vivo genome editing restores haemostasis in a mouse model of haemophilia.
- Author
-
Li H, Haurigot V, Doyon Y, Li T, Wong SY, Bhagwat AS, Malani N, Anguela XM, Sharma R, Ivanciu L, Murphy SL, Finn JD, Khazi FR, Zhou S, Paschon DE, Rebar EJ, Bushman FD, Gregory PD, Holmes MC, and High KA
- Subjects
- Animals, Base Sequence, Cell Line, Tumor, DNA Breaks, Double-Stranded, Endonucleases chemistry, Endonucleases genetics, Endonucleases metabolism, Exons genetics, Factor IX analysis, Factor IX genetics, Genetic Vectors genetics, HEK293 Cells, Hemophilia B physiopathology, Humans, Introns genetics, Liver metabolism, Liver Regeneration, Mice, Mice, Inbred C57BL, Mutation genetics, Phenotype, Sequence Homology, Zinc Fingers, DNA Repair genetics, Disease Models, Animal, Gene Targeting methods, Genetic Therapy methods, Genome genetics, Hemophilia B genetics, Hemostasis
- Abstract
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., (©2011 Macmillan Publishers Limited. All rights reserved)
- Published
- 2011
- Full Text
- View/download PDF
25. Efficient targeted gene disruption in the soma and germ line of the frog Xenopus tropicalis using engineered zinc-finger nucleases.
- Author
-
Young JJ, Cherone JM, Doyon Y, Ankoudinova I, Faraji FM, Lee AH, Ngo C, Guschin DY, Paschon DE, Miller JC, Zhang L, Rebar EJ, Gregory PD, Urnov FD, Harland RM, and Zeitler B
- Subjects
- Animals, Animals, Genetically Modified, Carrier Proteins metabolism, Deoxyribonucleases metabolism, Xenopus, Xenopus Proteins metabolism, Zinc Fingers, Alleles, Carrier Proteins genetics, Deoxyribonucleases genetics, Gene Targeting methods, Xenopus Proteins genetics
- Abstract
The frog Xenopus, an important research organism in cell and developmental biology, currently lacks tools for targeted mutagenesis. Here, we address this problem by genome editing with zinc-finger nucleases (ZFNs). ZFNs directed against an eGFP transgene in Xenopus tropicalis induced mutations consistent with nonhomologous end joining at the target site, resulting in mosaic loss of the fluorescence phenotype at high frequencies. ZFNs directed against the noggin gene produced tadpoles and adult animals carrying up to 47% disrupted alleles, and founder animals yielded progeny carrying insertions and deletions in the noggin gene with no indication of off-target effects. Furthermore, functional tests demonstrated an allelic series of activity between three germ-line mutant alleles. Because ZFNs can be designed against any locus, our data provide a generally applicable protocol for gene disruption in Xenopus.
- Published
- 2011
- Full Text
- View/download PDF
26. Rapid and efficient clathrin-mediated endocytosis revealed in genome-edited mammalian cells.
- Author
-
Doyon JB, Zeitler B, Cheng J, Cheng AT, Cherone JM, Santiago Y, Lee AH, Vo TD, Doyon Y, Miller JC, Paschon DE, Zhang L, Rebar EJ, Gregory PD, Urnov FD, and Drubin DG
- Subjects
- Animals, Base Sequence, Cell Lineage, Cell Membrane metabolism, Dynamin II metabolism, Genome, Green Fluorescent Proteins metabolism, Humans, Microscopy, Fluorescence methods, Models, Genetic, Molecular Sequence Data, Polymerase Chain Reaction, Clathrin metabolism, Endocytosis
- Abstract
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., (© 2011 Macmillan Publishers Limited. All rights reserved)
- Published
- 2011
- Full Text
- View/download PDF
27. Enhancing zinc-finger-nuclease activity with improved obligate heterodimeric architectures.
- Author
-
Doyon Y, Vo TD, Mendel MC, Greenberg SG, Wang J, Xia DF, Miller JC, Urnov FD, Gregory PD, and Holmes MC
- Subjects
- DNA metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Dimerization, Endonucleases genetics, Genome, Saccharomyces cerevisiae enzymology, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae metabolism, Zinc Fingers genetics, Endonucleases metabolism, Zinc Fingers physiology
- Abstract
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.
- Published
- 2011
- Full Text
- View/download PDF
28. Inducing high rates of targeted mutagenesis in zebrafish using zinc finger nucleases (ZFNs).
- Author
-
McCammon JM, Doyon Y, and Amacher SL
- Subjects
- Animal Husbandry, Animals, Embryo, Nonmammalian metabolism, Female, Genes, Reporter genetics, Genetic Loci genetics, Genotype, Germ Cells cytology, Germ Cells metabolism, Male, Microinjections, Phenotype, RNA biosynthesis, Saccharomyces cerevisiae genetics, Zebrafish embryology, Deoxyribonucleases chemistry, Deoxyribonucleases metabolism, Genetic Techniques, Mutagenesis, Zebrafish genetics, Zinc Fingers
- Abstract
Animal models, including the zebrafish, without a reliable embryonic stem cell system are not easily amenable to targeted mutagenesis for studying gene function. Three recent publications have shown that zinc finger nucleases (ZFNs) have circumvented this shortcoming in zebrafish. Similar to restriction enzymes, ZFNs can introduce site-specific double-strand breaks (DSBs); moreover, they can be designed to recognize virtually any target sequence. Because the preferred DSB repair pathway in zebrafish embryos, non-homologous end joining, is error-prone, ZFNs can be used to create mutations in a gene of interest. Here we review the protocols for a yeast-based assay to detect effective ZFNs. Additionally, we detail the procedures for synthesis and injection of ZFN-encoding mRNA into zebrafish embryos, screening of injected embryos for induced mutations in the soma, and recovery of germline mutations.
- Published
- 2011
- Full Text
- View/download PDF
29. Transient cold shock enhances zinc-finger nuclease-mediated gene disruption.
- Author
-
Doyon Y, Choi VM, Xia DF, Vo TD, Gregory PD, and Holmes MC
- Subjects
- Cold Temperature, HeLa Cells, Humans, K562 Cells, Deoxyribonucleases metabolism, Gene Targeting methods, Zinc Fingers
- Abstract
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.
- Published
- 2010
- Full Text
- View/download PDF
30. Precise genome modification in the crop species Zea mays using zinc-finger nucleases.
- Author
-
Shukla VK, Doyon Y, Miller JC, DeKelver RC, Moehle EA, Worden SE, Mitchell JC, Arnold NL, Gopalan S, Meng X, Choi VM, Rock JM, Wu YY, Katibah GE, Zhifang G, McCaskill D, Simpson MA, Blakeslee B, Greenwalt SA, Butler HJ, Hinkley SJ, Zhang L, Rebar EJ, Gregory PD, and Urnov FD
- Subjects
- Deoxyribonucleases genetics, Food, Genetically Modified, Genes, Plant genetics, Herbicide Resistance genetics, Herbicides pharmacology, Heredity, Inositol Phosphates metabolism, Mutagenesis, Site-Directed methods, Plants, Genetically Modified, Recombination, Genetic genetics, Reproducibility of Results, Biotechnology methods, Deoxyribonucleases chemistry, Deoxyribonucleases metabolism, Gene Targeting methods, Genome, Plant genetics, Zea mays genetics, Zinc Fingers
- Abstract
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.
- Published
- 2009
- Full Text
- View/download PDF
31. Targeted transgene integration in plant cells using designed zinc finger nucleases.
- Author
-
Cai CQ, Doyon Y, Ainley WM, Miller JC, Dekelver RC, Moehle EA, Rock JM, Lee YL, Garrison R, Schulenberg L, Blue R, Worden A, Baker L, Faraji F, Zhang L, Holmes MC, Rebar EJ, Collingwood TN, Rubin-Wilson B, Gregory PD, Urnov FD, and Petolino JF
- Subjects
- Amino Acid Sequence, Base Sequence, Binding Sites, Cells, Cultured, Chitinases genetics, Endonucleases genetics, Glucuronidase genetics, Glucuronidase metabolism, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Molecular Sequence Data, Polymerase Chain Reaction, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Recombination, Genetic, Nicotiana cytology, Nicotiana genetics, Nicotiana metabolism, Transfection methods, Endonucleases metabolism, Transgenes genetics, Zinc Fingers genetics
- Abstract
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.
- Published
- 2009
- Full Text
- View/download PDF
32. HBO1 HAT complexes target chromatin throughout gene coding regions via multiple PHD finger interactions with histone H3 tail.
- Author
-
Saksouk N, Avvakumov N, Champagne KS, Hung T, Doyon Y, Cayrou C, Paquet E, Ullah M, Landry AJ, Côté V, Yang XJ, Gozani O, Kutateladze TG, and Côté J
- Subjects
- Acetylation, Binding Sites, Cells, Cultured, DNA-Binding Proteins genetics, HeLa Cells, Histone Acetyltransferases genetics, Histones genetics, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, Humans, Methylation, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae metabolism, Chromatin metabolism, DNA-Binding Proteins metabolism, Histone Acetyltransferases metabolism, Histones metabolism
- Abstract
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.
- Published
- 2009
- Full Text
- View/download PDF
33. Molecular architecture of quartet MOZ/MORF histone acetyltransferase complexes.
- Author
-
Ullah M, Pelletier N, Xiao L, Zhao SP, Wang K, Degerny C, Tahmasebi S, Cayrou C, Doyon Y, Goh SL, Champagne N, Côté J, and Yang XJ
- Subjects
- Adaptor Proteins, Signal Transducing, Animals, Binding Sites, Cell Line, DNA-Binding Proteins, Histone Acetyltransferases genetics, Humans, Mutation, Nuclear Proteins genetics, Nuclear Proteins metabolism, Protein Structure, Tertiary, Protein Subunits genetics, Protein Subunits metabolism, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Transcription Factors genetics, Transcription Factors metabolism, Transcription, Genetic, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism, Histone Acetyltransferases metabolism, Multiprotein Complexes chemistry, Multiprotein Complexes metabolism
- 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.
- Published
- 2008
- Full Text
- View/download PDF
34. Heritable targeted gene disruption in zebrafish using designed zinc-finger nucleases.
- Author
-
Doyon Y, McCammon JM, Miller JC, Faraji F, Ngo C, Katibah GE, Amora R, Hocking TD, Zhang L, Rebar EJ, Gregory PD, Urnov FD, and Amacher SL
- Subjects
- Animals, Deoxyribonucleases genetics, Protein Engineering methods, Animals, Genetically Modified physiology, Gene Targeting methods, Genetic Engineering methods, Mutagenesis, Site-Directed methods, Zebrafish genetics, Zebrafish Proteins genetics, Zinc Fingers genetics
- Abstract
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.
- Published
- 2008
- Full Text
- View/download PDF
35. Eaf1 is the platform for NuA4 molecular assembly that evolutionarily links chromatin acetylation to ATP-dependent exchange of histone H2A variants.
- Author
-
Auger A, Galarneau L, Altaf M, Nourani A, Doyon Y, Utley RT, Cronier D, Allard S, and Côté J
- Subjects
- Acetylation, Acetyltransferases chemistry, Acid Phosphatase, Adenosine Triphosphatases chemistry, Adenosine Triphosphatases physiology, Eukaryotic Cells metabolism, Evolution, Molecular, Histone Acetyltransferases chemistry, Humans, Lysine Acetyltransferase 5, Promoter Regions, Genetic, Protein Interaction Mapping, Protein Structure, Tertiary, Saccharomyces cerevisiae Proteins chemistry, Saccharomyces cerevisiae Proteins genetics, Saccharomyces cerevisiae Proteins metabolism, Species Specificity, Structure-Activity Relationship, Acetyltransferases physiology, Adenosine Triphosphate metabolism, Chromatin metabolism, Histone Acetyltransferases physiology, Histones metabolism, Protein Processing, Post-Translational physiology, Saccharomyces cerevisiae Proteins physiology
- Abstract
Eaf1 (for Esa1-associated factor 1) and Eaf2 have been identified as stable subunits of NuA4, a yeast histone H4/H2A acetyltransferase complex implicated in gene regulation and DNA repair. While both SWI3-ADA2-N-CoR-TF IIIB domain-containing proteins are required for normal cell cycle progression, their depletion does not affect the global Esa1-dependent acetylation of histones. In contrast to all other subunits, Eaf1 is found exclusively associated with the NuA4 complex in vivo. It serves as a platform that coordinates the assembly of functional groups of subunits into the native NuA4 complex. Eaf1 shows structural similarities with human p400/Domino, a subunit of the NuA4-related TIP60 complex. On the other hand, p400 also possesses an SWI2/SNF2 family ATPase domain that is absent from the yeast NuA4 complex. This domain is highly related to the yeast Swr1 protein, which is responsible for the incorporation of histone variant H2AZ in chromatin. Since all of the components of the TIP60 complex are homologous to SWR1 or NuA4 subunits, we proposed that the human complex corresponds to a physical merge of two yeast complexes. p400 function in TIP60 then would be accomplished in yeast by cooperation between SWR1 and NuA4. In agreement with such a model, NuA4 and SWR1 mutants show strong genetic interactions, NuA4 affects histone H2AZ incorporation/acetylation in vivo, and both preset the PHO5 promoter for activation. Interestingly, the expression of a chimeric Eaf1-Swr1 protein recreates a single human-like complex in yeast cells. Our results identified the key central subunit for the structure and functions of the NuA4 histone acetyltransferase complex and functionally linked this activity with the histone variant H2AZ from yeast to human cells.
- Published
- 2008
- Full Text
- View/download PDF
36. [Mystification and cleverness of tumor suppressors in nuclear functions].
- Author
-
Cayrou C, Doyon Y, Landry AJ, Côté V, and Côté J
- Subjects
- DNA genetics, Homeodomain Proteins genetics, Humans, Receptors, Cytoplasmic and Nuclear genetics, Tumor Suppressor Proteins genetics, Cell Nucleus physiology, Genes, Tumor Suppressor, Neoplasms genetics, Neoplasms prevention & control
- Published
- 2006
- Full Text
- View/download PDF
37. ING tumor suppressor proteins are critical regulators of chromatin acetylation required for genome expression and perpetuation.
- Author
-
Doyon Y, Cayrou C, Ullah M, Landry AJ, Côté V, Selleck W, Lane WS, Tan S, Yang XJ, and Côté J
- Subjects
- Acetylation, Acetyltransferases genetics, Acetyltransferases metabolism, Amino Acid Sequence, Cell Cycle physiology, Cell Cycle Proteins classification, Cell Cycle Proteins genetics, DNA Replication, Histone Acetyltransferases genetics, Histone Acetyltransferases metabolism, Histones metabolism, Homeodomain Proteins classification, Homeodomain Proteins genetics, Humans, Inhibitor of Growth Protein 1, Intracellular Signaling Peptides and Proteins classification, Intracellular Signaling Peptides and Proteins genetics, Lysine Acetyltransferase 5, Molecular Sequence Data, Multiprotein Complexes, Nuclear Proteins classification, Nuclear Proteins genetics, Protein Subunits genetics, Protein Subunits metabolism, RNA Interference, Receptors, Cytoplasmic and Nuclear classification, Receptors, Cytoplasmic and Nuclear genetics, Repressor Proteins genetics, Repressor Proteins metabolism, Sequence Alignment, Sin3 Histone Deacetylase and Corepressor Complex, Trans-Activators classification, Trans-Activators genetics, Transcription Factors genetics, Tumor Suppressor Proteins classification, Tumor Suppressor Proteins genetics, Cell Cycle Proteins metabolism, Chromatin metabolism, Gene Expression, Genes, Tumor Suppressor, Homeodomain Proteins metabolism, Intracellular Signaling Peptides and Proteins metabolism, Nuclear Proteins metabolism, Receptors, Cytoplasmic and Nuclear metabolism, Trans-Activators metabolism, Transcription Factors metabolism, Tumor Suppressor Proteins metabolism
- Abstract
Members of the ING family of tumor suppressors regulate cell cycle progression, apoptosis, and DNA repair as important cofactors of p53. ING1 and ING3 are stable components of the mSin3A HDAC and Tip60/NuA4 HAT complexes, respectively. We now report the purification of the three remaining human ING proteins. While ING2 is in an HDAC complex similar to ING1, ING4 associates with the HBO1 HAT required for normal progression through S phase and the majority of histone H4 acetylation in vivo. ING5 fractionates with two distinct complexes containing HBO1 or nucleosomal H3-specific MOZ/MORF HATs. These ING5 HAT complexes interact with the MCM helicase and are essential for DNA replication to occur during S phase. Our data also indicate that ING subunits are crucial for acetylation of chromatin substrates. Since INGs, HBO1, and MOZ/MORF contribute to oncogenic transformation, the multisubunit assemblies characterized here underscore the critical role of epigenetic regulation in cancer development.
- Published
- 2006
- Full Text
- View/download PDF
38. The highly conserved and multifunctional NuA4 HAT complex.
- Author
-
Doyon Y and Côté J
- Subjects
- DNA Repair physiology, Histone Acetyltransferases, Humans, Lysine Acetyltransferase 5, Saccharomyces cerevisiae enzymology, Transcription, Genetic physiology, Acetyltransferases physiology, Multienzyme Complexes physiology
- Abstract
Histone acetyltransferase complexes have been shown to be key regulators of gene expression. Among these, the NuA4 complex, first characterized in yeast, stands out as it controls multiple key nuclear functions in eukaryotic cells. Many subunits of this protein assembly have been directly linked to global and targeted acetylation of histone H4 tails in vivo, regulation of transcription, cell-cycle progression as well as to the process of DNA repair. Recent studies presented here have established its remarkable structural conservation from yeast to human cells and contributed to the understanding of its diverse functions.
- Published
- 2004
- Full Text
- View/download PDF
39. Structural and functional conservation of the NuA4 histone acetyltransferase complex from yeast to humans.
- Author
-
Doyon Y, Selleck W, Lane WS, Tan S, and Côté J
- Subjects
- Acetylation, Acetyltransferases genetics, Amino Acid Sequence, Animals, Carrier Proteins genetics, Carrier Proteins metabolism, Cell Cycle physiology, Cell Line, DNA Helicases genetics, DNA Helicases metabolism, Genes, Tumor Suppressor, Histone Acetyltransferases, Histones metabolism, Homeodomain Proteins, Humans, Lysine Acetyltransferase 5, Molecular Sequence Data, Multienzyme Complexes, Protein Subunits genetics, Proteins genetics, Proteins metabolism, Repressor Proteins genetics, Repressor Proteins metabolism, Saccharomyces cerevisiae Proteins genetics, Sequence Alignment, Trans-Activators, Transcription Factors genetics, Transcription Factors metabolism, Tumor Suppressor Proteins, Acetyltransferases metabolism, Protein Subunits metabolism, Saccharomyces cerevisiae Proteins metabolism
- 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.
- Published
- 2004
- Full Text
- View/download PDF
40. Identification and analysis of native HAT complexes.
- Author
-
McMahon SJ, Doyon Y, Côté J, and Grant PA
- Subjects
- Acetyltransferases genetics, Electrophoresis, Polyacrylamide Gel methods, Histone Acetyltransferases, Humans, Indicators and Reagents, Nucleosomes ultrastructure, Protein Denaturation, Protein Renaturation, Protein Subunits isolation & purification, Saccharomyces cerevisiae enzymology, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae Proteins genetics, Saccharomyces cerevisiae Proteins isolation & purification, Saccharomyces cerevisiae Proteins metabolism, Acetyltransferases isolation & purification, Acetyltransferases metabolism
- Published
- 2004
- Full Text
- View/download PDF
41. Effect of C-domain N-glycosylation and deletion on rat pancreatic alpha-amylase secretion and activity.
- Author
-
Doyon Y, Home W, Daull P, and LeBel D
- Subjects
- Animals, Asparagine metabolism, Cell Line, Glycosylation, Mutagenesis, Site-Directed, Pancreas metabolism, Protein Biosynthesis, Protein Folding, Protein Transport, Rats, Sequence Deletion, Transcription, Genetic, alpha-Amylases chemistry, alpha-Amylases genetics, Pancreas enzymology, alpha-Amylases metabolism
- Abstract
Even though all animal alpha-amylases include glycosylation sequons (Asn-Xaa-Thr/Ser) in their sequences, amylases purified from natural sources are not quantitatively glycosylated. When wild-type rat pancreatic alpha-amylase, which contains two glycosylation sequons, was expressed in animal cell lines the protein displayed a very low rate of glycosylation (approx. 2%), even after Brefeldin A treatment to increase the contact with the glycosylation machinery. Site-directed mutagenesis of the first glycosylation sequon (Asn(410)-->Gln) resulted in 90% of the protein being glycosylated at the second glycosylation sequon (Asn(459)). Mutation of the second sequon completely inhibited glycosylation. In order to ascertain if the interference in the glycosylation of Asn(459) that was eliminated by the Asn(410)-->Gln mutation could be due to the position of the asparagine residue in the Cys(448)-Cys(460) disulphide bridge, these cysteine residues were mutated to serine residues. The resulting mutant was found to be 100% glycosylated. All mutants with mutations in the C-domain had specific activities identical to that of the wild-type enzyme, indicating that enzymic activity is independent of the structure and modification of the C-terminal domain. To further test the independence of the C-domain with respect to the two N-terminal domains of the protein, which harbour the catalytic site, the last seven of the ten beta\beta-strands that make up the beta-sandwich configuration of the domain were deleted. The truncated protein was not secreted from cells and all enzyme activity was destroyed. These observations show that Asn(459) is the only site that can be glycosylated in wild-type amylase, and confirm the relative independence of the C-terminal domain of alpha-amylase with respect to enzyme activity. In addition, they also establish that the C-terminal domain is absolutely essential for the correct post-translational folding of the enzyme that is responsible for its activity and allows for its secretion.
- Published
- 2002
- Full Text
- View/download PDF
42. Role of an ING1 growth regulator in transcriptional activation and targeted histone acetylation by the NuA4 complex.
- Author
-
Nourani A, Doyon Y, Utley RT, Allard S, Lane WS, and Côté J
- Subjects
- Acetylation, Amino Acid Sequence, Cell Cycle Proteins, Cell Division, Cyclin-Dependent Kinase Inhibitor p21, Cyclins genetics, DNA-Binding Proteins, Genes, Tumor Suppressor, Histone Acetyltransferases, Homeodomain Proteins metabolism, Humans, Inhibitor of Growth Protein 1, Intracellular Signaling Peptides and Proteins, Molecular Sequence Data, Nuclear Proteins, Plant Proteins genetics, Plant Proteins metabolism, Promoter Regions, Genetic, Proteins, Saccharomyces cerevisiae enzymology, Saccharomyces cerevisiae genetics, Trans-Activators genetics, Trans-Activators metabolism, Trans-Activators physiology, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Tumor Suppressor Protein p53 physiology, Acetyltransferases metabolism, Histones metabolism, Homeodomain Proteins genetics, Homeodomain Proteins physiology, Plant Proteins physiology, Saccharomyces cerevisiae Proteins, Transcriptional Activation, Tumor Suppressor Proteins
- Abstract
The yeast NuA4 complex is a histone H4 and H2A acetyltransferase involved in transcription regulation and essential for cell cycle progression. We identify here a novel subunit of the complex, Yng2p, a plant homeodomain (PHD)-finger protein homologous to human p33/ING1, which has tumor suppressor activity and is essential for p53 function. Mass spectrometry, immunoblotting, and immunoprecipitation experiments confirm the stable stoichiometric association of this protein with purified NuA4. Yeast cells harboring a deletion of the YNG2 gene show severe growth phenotype and have gene-specific transcription defects. NuA4 complex purified from the mutant strain is low in abundance and shows weak histone acetyltransferase activity. We demonstrate conservation of function by the requirement of Yng2p for p53 to function as a transcriptional activator in yeast. Accordingly, p53 interacts with NuA4 in vitro and in vivo, an interaction reminiscent of the p53-ING1 physical link in human cells. The growth defect of Delta yng2 cells can be rescued by the N-terminal part of the protein, lacking the PHD-finger. While Yng2 PHD-finger is not required for p53 interaction, it is necessary for full expression of the p53-responsive gene and other NuA4 target genes. Transcriptional activation by p53 in vivo is associated with targeted NuA4-dependent histone H4 hyperacetylation, while histone H3 acetylation levels remain unchanged. These results emphasize the essential role of the NuA4 complex in the control of cell proliferation through gene-specific transcription regulation. They also suggest that regulation of mammalian cell proliferation by p53-dependent transcriptional activation functions through recruitment of an ING1-containing histone acetyltransferase complex.
- Published
- 2001
- Full Text
- View/download PDF
43. Compressed work-week: psychophysiological and physiological repercussions.
- Author
-
Volle M, Brisson GR, Pérusse M, Tanaka M, and Doyon Y
- Subjects
- Adult, Aged, Flicker Fusion physiology, Hand physiology, Humans, Male, Middle Aged, Psychophysiology, Task Performance and Analysis, Fatigue etiology, Work
- Published
- 1979
- Full Text
- View/download PDF
44. [Group psychotherapy of preadolescents presenting behavior problems of neurotic origin].
- Author
-
Chabot F and Doyon Y
- Subjects
- Adolescent, Child, Child Behavior Disorders etiology, Humans, Male, Neurotic Disorders complications, Child Behavior Disorders therapy, Neurotic Disorders therapy, Psychotherapy, Group
- Published
- 1973
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.