Network


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

Hotspot


Dive into the research topics where Agnès Gouble is active.

Publication


Featured researches published by Agnès Gouble.


Nature Methods | 2012

Coupling endonucleases with DNA end-processing enzymes to drive gene disruption

Michael T. Certo; Kamila S. Gwiazda; Ryan Kuhar; Blythe D. Sather; Gabrielle Curinga; Tyler Mandt; Michelle Brault; Abigail R. Lambert; Sarah K. Baxter; Kyle Jacoby; Byoung Y. Ryu; Hans Peter Kiem; Agnès Gouble; Frédéric Paques; David J. Rawlings; Andrew M. Scharenberg

Targeted DNA double-strand breaks introduced by rare-cleaving designer endonucleases can be harnessed for gene disruption applications by engaging mutagenic nonhomologous end-joining DNA repair pathways. However, endonuclease-mediated DNA breaks are often subject to precise repair, which limits the efficiency of targeted genome editing. To address this issue, we coupled designer endonucleases to DNA end–processing enzymes to drive mutagenic break resolution, achieving up to 25-fold enhancements in gene disruption rates.


Journal of Gene Medicine | 2006

Efficient in toto targeted recombination in mouse liver by meganuclease-induced double-strand break.

Agnès Gouble; Julianne Smith; Sylvia Bruneau; Christophe Perez; Valérie Guyot; Jean-Pierre Cabaniols; Sophie Leduc; Laurence Fiette; Patrick Ave; Béatrice Micheau; Philippe Duchateau

Sequence‐specific endonucleases with large recognition sites can cleave DNA in living cells, and, as a consequence, stimulate homologous recombination (HR) up to 10 000‐fold. The recent development of artificial meganucleases with chosen specificities has provided the potential to target any chromosomal locus. Thus, they may represent a universal genome engineering tool and seem to be very promising for acute gene therapy. However, in toto applications depend on the ability to target somatic tissues as well as the proficiency of somatic cells to perform double‐strand break (DSB)‐induced HR.


Nucleic Acids Research | 2012

Chromosomal context and epigenetic mechanisms control the efficacy of genome editing by rare-cutting designer endonucleases

Fayza Daboussi; Mikhail Zaslavskiy; Laurent Poirot; Mariana Loperfido; Agnès Gouble; Valérie Guyot; Sophie Leduc; Roman Galetto; Sylvestre Grizot; Danusia Oficjalska; Christophe Perez; Fabien Delacôte; Aurélie Dupuy; Isabelle Chion-Sotinel; Diane Le Clerre; Céline Lebuhotel; Olivier Danos; Frédéric Lemaire; Kahina Oussedik; Frédéric Cédrone; Jean-Charles Epinat; Julianne Smith; Rafael J. Yáñez-Muñoz; George Dickson; Linda Popplewell; Taeyoung Koo; Thierry Vandendriessche; Marinee K. Chuah; Aymeric Duclert; Philippe Duchateau

The ability to specifically engineer the genome of living cells at precise locations using rare-cutting designer endonucleases has broad implications for biotechnology and medicine, particularly for functional genomics, transgenics and gene therapy. However, the potential impact of chromosomal context and epigenetics on designer endonuclease-mediated genome editing is poorly understood. To address this question, we conducted a comprehensive analysis on the efficacy of 37 endonucleases derived from the quintessential I-CreI meganuclease that were specifically designed to cleave 39 different genomic targets. The analysis revealed that the efficiency of targeted mutagenesis at a given chromosomal locus is predictive of that of homologous gene targeting. Consequently, a strong genome-wide correlation was apparent between the efficiency of targeted mutagenesis (≤0.1% to ∼6%) with that of homologous gene targeting (≤0.1% to ∼15%). In contrast, the efficiency of targeted mutagenesis or homologous gene targeting at a given chromosomal locus does not correlate with the activity of individual endonucleases on transiently transfected substrates. Finally, we demonstrate that chromatin accessibility modulates the efficacy of rare-cutting endonucleases, accounting for strong position effects. Thus, chromosomal context and epigenetic mechanisms may play a major role in the efficiency rare-cutting endonuclease-induced genome engineering.


Human Gene Therapy | 2013

Gene Correction of a Duchenne Muscular Dystrophy Mutation by Meganuclease-Enhanced Exon Knock-In

Linda Popplewell; Taeyoung Koo; Xavier Leclerc; Aymeric Duclert; Kamel Mamchaoui; Agnès Gouble; Vincent Mouly; Thomas Voit; Frédéric Cédrone; Olga Isman; Rafael J. Yáñez-Muñoz; George Dickson

Duchenne muscular dystrophy (DMD) is a severe inherited, muscle-wasting disorder caused by mutations in the DMD gene. Gene therapy development for DMD has concentrated on vector-based DMD minigene transfer, cell-based gene therapy using genetically modified adult muscle stem cells or healthy wild-type donor cells, and antisense oligonucleotide-induced exon-skipping therapy to restore the reading frame of the mutated DMD gene. This study is an investigation into DMD gene targeting-mediated correction of deletions in human patient myoblasts using a target-specific meganuclease (MN) and a homologous recombination repair matrix. The MN was designed to cleave within DMD intron 44, upstream of a deletion hotspot, and integration-competent lentiviral vectors expressing the nuclease (LVcMN) were generated. MN western blotting and deep gene sequencing for LVcMN-induced non-homologous end-joining InDels (microdeletions or microinsertions) confirmed efficient MN expression and activity in transduced DMD myoblasts. A homologous repair matrix carrying exons 45-52 (RM45-52) was designed and packaged into integration-deficient lentiviral vectors (IDLVs; LVdRM45-52). After cotransduction of DMD myoblasts harboring a deletion of exons 45 to 52 with LVcMN and LVdRM45-52 vectors, targeted knock-in of the RM45-52 region in the correct location in DMD intron 44, and expression of full-length, correctly spliced wild-type dystrophin mRNA containing exons 45-52 were observed. This work demonstrates that genome surgery on human DMD gene mutations can be achieved by MN-induced locus-specific genome cleavage and homologous recombination knock-in of deleted exons. The feasibility of human DMD gene repair in patient myoblasts has exciting therapeutic potential.


PLOS ONE | 2013

Targeted gene therapy of xeroderma pigmentosum cells using meganuclease and TALEN

Aurélie Dupuy; Julien Valton; Sophie Leduc; Jacques Armier; Roman Galetto; Agnès Gouble; Céline Lebuhotel; Anne Stary; Frédéric Pâques; Philippe Duchateau; Alain Sarasin; Fayza Daboussi

Xeroderma pigmentosum group C (XP-C) is a rare human syndrome characterized by hypersensitivity to UV light and a dramatic predisposition to skin neoplasms. XP-C cells are deficient in the nucleotide excision repair (NER) pathway, a complex process involved in the recognition and removal of DNA lesions. Several XPC mutations have been described, including a founder mutation in North African patients involving the deletion of a TG dinucleotide (ΔTG) located in the middle of exon 9. This deletion leads to the expression of an inactive truncated XPC protein, normally involved in the first step of NER. New approaches used for gene correction are based on the ability of engineered nucleases such as Meganucleases, Zinc-Finger nucleases or TALE nucleases to accurately generate a double strand break at a specific locus and promote correction by homologous recombination through the insertion of an exogenous DNA repair matrix. Here, we describe the targeted correction of the ΔTG mutation in XP-C cells using engineered meganuclease and TALEN™. The methylated status of the XPC locus, known to inhibit both of these nuclease activities, led us to adapt our experimental design to optimize their in vivo efficacies. We show that demethylating treatment as well as the use of TALEN™ insensitive to CpG methylation enable successful correction of the ΔTG mutation. Such genetic correction leads to re-expression of the full-length XPC protein and to the recovery of NER capacity, attested by UV-C resistance of the corrected cells. Overall, we demonstrate that nuclease-based targeted approaches offer reliable and efficient strategies for gene correction.


BioTechniques | 2005

Factors affecting double-strand break-induced homologous recombination in mammalian cells

Christophe Perez; Valérie Guyot; Jean-Pierre Cabaniols; Agnès Gouble; Beatrice Micheaux; Julie Smith; Sophie Leduc; Philippe Duchateau

Double-strand break (DSB)-induced homologous recombination (HR) of direct repeats is a powerful means to achieve gene excision, a critical step in genome engineering. In this report we have used an extrachrmosomal reporter system to monitor the impact of different parameters on meganuclease-induced HR in CHO-K1 cells. We found that repeat homology length is critical. Virtually no HR could be detected with a 15-bp duplication, while, with repeats larger than 400 bp, recombination efficiency became less dependent on homology length. The presence of an intervening sequence between the duplications dramatically impairs HR, independent of the cleavage position; by 3 kb of insertion, HR is virtually undetectable. Efficient HR can be restored by positioning cleavage sites at both ends of the intervening sequence, allowing a constant level of excision with up to 10 kb of intervening sequences. Using similar constructs, 2.8-kb inserts could be efficiently removed from several chromosomal loci, illustrating the wide potential of this technology. These results fit current models of direct repeat recombination and identify DSB-induced HR as a powerful tool for gene excision.


Blood | 2015

TALEN-mediated genetic inactivation of the glucocorticoid receptor in cytomegalovirus-specific T cells

Laurie Menger; Agnès Gouble; Maria A. V. Marzolini; Annette Pachnio; Katharina Bergerhoff; Jake Y. Henry; Julianne Smith; Martin Pule; Paul Moss; Stanley R. Riddell; Sergio A. Quezada; Karl S. Peggs

Cytomegalovirus (CMV) infection is responsible for substantial morbidity and mortality after allogeneic hematopoietic stem cell transplant. T-cell immunity is critical for control of CMV infection, and correction of the immune deficiency induced by transplant is now clinically achievable by the adoptive transfer of donor-derived CMV-specific T cells. It is notable, however, that most clinical studies of adoptive T- cell therapy exclude patients with graft-versus-host disease (GVHD) from receiving systemic corticosteroid therapy, which impairs cellular immunity. This group of patients remains the highest clinical risk group for recurrent and problematic infections. Here, we address this unmet clinical need by genetic disruption of the glucocorticoid receptor (GR) gene using electroporation of transcription activator-like effector nuclease (TALEN) messenger RNA. We demonstrate efficient inactivation of the GR gene without off-target activity in Streptamer-selected CMV-specific CD8(+) T cells (HLA-A02/NLV peptide), conferring resistance to glucocorticoids. TALEN-modified CMV-specific T cells retained specific killing of target cells pulsed with the CMV peptide NLV in the presence of dexamethasone (DEX). Inactivation of the GR gene also conferred resistance to DEX in a xenogeneic GVHD model in sublethally irradiated NOD-scid IL2rγ(null) mice. This proof of concept provides the rationale for the development of clinical protocols for producing and administering high-purity genetically engineered virus-specific T cells that are resistant to the suppressive effects of corticosteroids.


Stem cell reports | 2015

Generation of a High Number of Healthy Erythroid Cells from Gene-Edited Pyruvate Kinase Deficiency Patient-Specific Induced Pluripotent Stem Cells

Zita Garate; Oscar Quintana-Bustamante; Ana M. Crane; Emmanuel Olivier; Laurent Poirot; Roman Galetto; Penelope Kosinski; Collin Hill; Charles Kung; Xabi Agirre; Israel Orman; Laura Cerrato; Omaira Alberquilla; Fatima Rodriguez-Fornes; Noemi Fusaki; Félix García-Sánchez; Tabita M. Maia; Ribeiro Ml; Julián Sevilla; Felipe Prosper; Shengfang Jin; Joanne C. Mountford; Guillermo Guenechea; Agnès Gouble; Juan A. Bueren; Brian R. Davis; José Segovia

Summary Pyruvate kinase deficiency (PKD) is a rare erythroid metabolic disease caused by mutations in the PKLR gene. Erythrocytes from PKD patients show an energetic imbalance causing chronic non-spherocytic hemolytic anemia, as pyruvate kinase defects impair ATP production in erythrocytes. We generated PKD induced pluripotent stem cells (PKDiPSCs) from peripheral blood mononuclear cells (PB-MNCs) of PKD patients by non-integrative Sendai viral vectors. PKDiPSCs were gene edited to integrate a partial codon-optimized R-type pyruvate kinase cDNA in the second intron of the PKLR gene by TALEN-mediated homologous recombination (HR). Notably, we found allele specificity of HR led by the presence of a single-nucleotide polymorphism. High numbers of erythroid cells derived from gene-edited PKDiPSCs showed correction of the energetic imbalance, providing an approach to correct metabolic erythroid diseases and demonstrating the practicality of this approach to generate the large cell numbers required for comprehensive biochemical and metabolic erythroid analyses.


Journal for ImmunoTherapy of Cancer | 2013

Treatment of B cells malignancies with anti-CD19 CAR+, TCR-, CD52- allogeneic T cells

Cécile Mannioui; Laetitia Lemaire; Laurent Poirot; Agnès Gouble; Sylvain Arnould; Roman Galetto; Julianne Smith; Andrew M. Scharenberg

Encouraging data have emerged from adoptive T-cell therapies in advanced forms of cancer. Anti-tumor immunity is found in tumor infiltrating lymphocytes as well as engineered T cells where exogenous expression of a chimeric antigen receptor (CAR) confers cancer recognition on the cells. Present adoptive immunotherapy methods are restricted to the use of autologous patient T-cells due to the limited persistence of allogeneic T cells and the potential for graft versus host disease (GvHD). The use of autologous patient T cells in cancer immunotherapy is however limited due to the fact that this approach is complex and time consuming. We propose a novel approach to treat B cell malignancies based on the use of genetically modified allogeneic T cells in conjunction with the conditioning regimen alemtuzumab. Allogeneic T cells were engineered to express an anti-CD19 CAR and to no longer express TCRalpha and CD52, responsible for GVHD and the sensitivity to alemtuzumab, respectively. The inactivation of the TCRalpha and CD52 genes in allogeneic T cells was realized by using TALEN TM ,an ovel class of sequence-specific nucleases created by the fusion of transcription activator-like effectors (TALEs) to the catalytic domain of an endonuclease. We have shown that anti-CD19 CAR+ TCR- CD52- allogeneic T cells did not respond to TCR stimulation, were resistant to alemtuzumab treatment and were able to kill target cells expressing CD19 in vitro and in vivo.


Leukemia & Lymphoma | 2018

Selection and characterization of antibody clones are critical for accurate flow cytometry-based monitoring of CD123 in acute myeloid leukemia

Nicole M. Cruz; Mayumi Sugita; Nathan Ewing-Crystal; Linda Lam; Roman Galetto; Agnès Gouble; Julianne Smith; Duane C. Hassane; Gail J. Roboz; Monica L. Guzman

Acute myelogenous leukemia (AML) is a deadly disease characterized by high relapse rates even in patients who initially achieve complete remission. Standard therapy for AML has remained largely unc...

Collaboration


Dive into the Agnès Gouble's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew M. Scharenberg

Seattle Children's Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Emmanuel Lacroix

European Bioinformatics Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge