Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Plant Biotechnol J ; 21(5): 918-930, 2023 05.
Article in English | MEDLINE | ID: mdl-36715107

ABSTRACT

Resistance to potyviruses in plants has been largely provided by the selection of natural variant alleles of eukaryotic translation initiation factors (eIF) 4E in many crops. However, the sources of such variability for breeding can be limited for certain crop species, while new virus isolates continue to emerge. Different methods of mutagenesis have been applied to inactivate the eIF4E genes to generate virus resistance, but with limited success due to the physiological importance of translation factors and their redundancy. Here, we employed genome editing approaches at the base level to induce non-synonymous mutations in the eIF4E1 gene and create genetic diversity in cherry tomato (Solanum lycopersicum var. cerasiforme). We sequentially edited the genomic sequences coding for two regions of eIF4E1 protein, located around the cap-binding pocket and known to be important for susceptibility to potyviruses. We show that the editing of only one of the two regions, by gene knock-in and base editing, respectively, is not sufficient to provide resistance. However, combining amino acid mutations in both regions resulted in resistance to multiple potyviruses without affecting the functionality in translation initiation. Meanwhile, we report that extensive base editing in exonic region can alter RNA splicing pattern, resulting in gene knockout. Altogether our work demonstrates that precision editing allows to design plant factors based on the knowledge on evolutionarily selected alleles and enlarge the gene pool to potentially provide advantageous phenotypes such as pathogen resistance.


Subject(s)
Potyvirus , Solanum lycopersicum , Gene Editing , Solanum lycopersicum/genetics , Eukaryotic Initiation Factor-4E/genetics , Potyvirus/genetics , Plant Proteins/genetics , Plant Breeding , Mutation , Plant Diseases/genetics
2.
Oncotarget ; 8(6): 9974-9985, 2017 Feb 07.
Article in English | MEDLINE | ID: mdl-28039489

ABSTRACT

TRAIL induces selective tumor cell death through TRAIL-R1 and TRAIL-R2. Despite the fact that these receptors share high structural homologies, induction of apoptosis upon ER stress, cell autonomous motility and invasion have solely been described to occur through TRAIL-R2. Using the TALEN gene-editing approach, we show that TRAIL-R1 can also induce apoptosis during unresolved unfolded protein response (UPR). Likewise, TRAIL-R1 was found to co-immunoprecipitate with FADD and caspase-8 during ER stress. Its deficiency conferred resistance to apoptosis induced by thaspigargin, tunicamycin or brefeldin A. Our data also demonstrate that tumor cell motility and invasion-induced by TRAIL-R2 is not cell autonomous but induced in a TRAIL-dependant manner. TRAIL-R1, on the other hand, is unable to trigger cell migration owing to its inability to induce an increase in calcium flux. Importantly, all the isogenic cell lines generated in this study revealed that apoptosis induced TRAIL is preferentially induced by TRAIL-R1. Taken together, our results provide novel insights into the physiological functions of TRAIL-R1 and TRAIL-R2 and suggest that targeting TRAIL-R1 for anticancer therapy is likely to be more appropriate owing to its lack of pro-motile signaling capability.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/drug therapy , Colonic Neoplasms/drug therapy , Endoplasmic Reticulum Stress/drug effects , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CHO Cells , Calcium Signaling/drug effects , Caspase 8/metabolism , Cell Movement/drug effects , Chick Embryo , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cricetulus , Dose-Response Relationship, Drug , Fas-Associated Death Domain Protein/metabolism , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Signal Transduction/drug effects , Time Factors , Transfection
3.
Methods ; 69(2): 151-70, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25047178

ABSTRACT

TALEN is one of the most widely used tools in the field of genome editing. It enables gene integration and gene inactivation in a highly efficient and specific fashion. Although very attractive, the apparent simplicity and high success rate of TALEN could be misleading for novices in the field of gene editing. Depending on the application, specific TALEN designs, activity assessments and screening strategies need to be adopted. Here we report different methods to efficiently perform TALEN-mediated gene integration and inactivation in different mammalian cell systems including induced pluripotent stem cells and delineate experimental examples associated with these approaches.


Subject(s)
Gene Targeting/methods , Genome/genetics , Transcriptional Activation/genetics , Transfection/methods , Animals , Base Sequence , Cell Line , DNA-Binding Proteins/genetics , HCT116 Cells , Humans , Molecular Sequence Data
4.
PLoS One ; 8(1): e53217, 2013.
Article in English | MEDLINE | ID: mdl-23359797

ABSTRACT

Targeting DNA double-strand breaks is a powerful strategy for gene inactivation applications. Without the use of a repair plasmid, targeted mutagenesis can be achieved through Non-Homologous End joining (NHEJ) pathways. However, many of the DNA breaks produced by engineered nucleases may be subject to precise re-ligation without loss of genetic information and thus are likely to be unproductive. In this study, we combined engineered endonucleases and DNA-end processing enzymes to increase the efficiency of targeted mutagenesis, providing a robust and efficient method to (i) greatly improve targeted mutagenesis frequency up to 30-fold, and; (ii) control the nature of mutagenic events using meganucleases in conjunction with DNA-end processing enzymes in human primary cells.


Subject(s)
DNA End-Joining Repair , DNA/metabolism , Endonucleases/metabolism , Mutagenesis , Animals , Base Sequence , CHO Cells , Cricetinae , Cricetulus , DNA/genetics , DNA Primers , HEK293 Cells , Humans
5.
Nucleic Acids Res ; 40(13): 6367-79, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22467209

ABSTRACT

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.


Subject(s)
Chromosomal Position Effects , DNA Restriction Enzymes/metabolism , Animals , CHO Cells , Cell Line , Cricetinae , Cricetulus , DNA Restriction Enzymes/chemistry , Gene Targeting , Genetic Engineering , Genome, Human , Humans , Mutagenesis
6.
J Nucleic Acids ; 2011: 947212, 2011.
Article in English | MEDLINE | ID: mdl-21716659

ABSTRACT

Homologous gene targeting (HGT) is a precise but inefficient process for genome engineering. Several methods for increasing its efficiency have been developed, including the use of rare cutting endonucleases. However, there is still room for improvement, as even nuclease-induced HGT may vary in efficiency as a function of the nuclease, target site, and cell type considered. We have developed a high-throughput screening assay for the identification of factors stimulating meganuclease-induced HGT. We used this assay to explore a collection of siRNAs targeting 19,121 human genes. At the end of secondary screening, we had identified 64 genes for which knockdown affected nuclease-induced HGT. Two of the strongest candidates were characterized further. We showed that siRNAs directed against the ATF7IP gene, encoding a protein involved in chromatin remodeling, stimulated HGT by a factor of three to eight, at various loci and in different cell types. This method thus led to the identification of a number of genes, the manipulation of which might increase rates of targeted recombination.

7.
DNA Repair (Amst) ; 7(9): 1500-16, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18603484

ABSTRACT

Ultraviolet A (UVA) radiation represents more than 90% of the UV spectrum reaching Earth's surface. Exposure to UV light, especially the UVA part, induces the formation of photoexcited states of cellular photosensitizers with subsequent generation of reactive oxygen species (ROS) leading to damages to membrane lipids, proteins and nucleic acids. Although UVA, unlike UVC and UVB, is poorly absorbed by DNA, it inhibits cell cycle progression, especially during S-phase. In the present study, we examined the role of the DNA damage checkpoint response in UVA-induced inhibition of DNA replication. We provide evidence that UVA delays S-phase in a dose dependent manner and that UVA-irradiated S-phase cells accumulate in G2/M. We show that upon UVA irradiation ATM-, ATR- and p38-dependent signalling pathways are activated, and that Chk1 phosphorylation is ATR/Hus1 dependent while Chk2 phosphorylation is ATM dependent. To assess for a role of these pathways in UVA-induced inhibition of DNA replication, we investigated (i) cell cycle progression of BrdU labelled S-phase cells by flow cytometry and (ii) incorporation of [methyl-(3)H]thymidine, as a marker of DNA replication, in ATM, ATR and p38 proficient and deficient cells. We demonstrate that none of these pathways is required to delay DNA replication in response to UVA, thus ruling out a role of the canonical S-phase checkpoint response in this process. On the contrary, scavenging of UVA-induced reactive oxygen species (ROS) by the antioxidant N-acetyl-L-cystein or depletion of vitamins during UVA exposure significantly restores DNA synthesis. We propose that inhibition of DNA replication is due to impaired replication fork progression, rather as a consequence of UVA-induced oxidative damage to protein than to DNA.


Subject(s)
DNA Damage , Reactive Oxygen Species/pharmacology , S Phase/drug effects , Ultraviolet Rays , Ataxia Telangiectasia Mutated Proteins , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Cycle Proteins/metabolism , Cell Line, Transformed , DNA/biosynthesis , DNA Replication/drug effects , DNA Replication/radiation effects , DNA-Binding Proteins/metabolism , Humans , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , S Phase/radiation effects , Signal Transduction , Tumor Suppressor Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Cell Cycle ; 7(1): 33-8, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18196958

ABSTRACT

A DNA double-strand break (DSB) is a highly harmful lesion that can lead to genome rearrangements. Two main pathways compete for DSB repair: homologous recombination (HR) and nonhomologous end-joining (NHEJ). Depending on the cell cycle phase, the choice of one DSB repair pathway over the other will secure genome stability maintenance or in contrast will increase the risk of genetic instability. HR with the sister chromatid is an efficient way to maintain genome stability, for damage occurring at a post-replication stage. However, in G(1) checkpoint-defective cells, DSBs produced in the G(1) phase and not repaired by NHEJ, can progress through S phase and be processed by HR in late S/G(2) phase. We propose the "trans-S DSB repair" model to account for these data. In this situation HR cannot use the sister chromatid (which is also broken at the same locus) and is thus forced to use ectopic homologous sequences dispersed through the genome, increasing the risk of genetic instability. This shows that the two DSB repair pathways can compete through the cell cycle and underlines the importance of the association between the cell cycle checkpoint and the appropriate DNA repair pathway for genome stability maintenance.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair/physiology , Genomic Instability/physiology , Models, Genetic , S Phase/genetics , Signal Transduction/genetics , Animals , Cell Cycle/genetics , Humans , Recombination, Genetic/physiology
9.
Mutat Res ; 615(1-2): 125-33, 2007 Feb 03.
Article in English | MEDLINE | ID: mdl-17174359

ABSTRACT

DNA double-strand breaks (DSBs) are highly toxic lesions leading to genome variability/instability. The balance between homologous recombination (HR) and non-homologous end-joining (NHEJ), two alternative DSB repair systems, is essential to ensure genome maintenance in mammalian cells. Here, we transfected CHO hamster cells with the pcDNA3.1/Zeo plasmid, and selected transfectants with Zeocin, a bleomycin analog which produces DSBs. Despite the presence of a Zeocin resistance gene in pcDNA3.1/Zeo, Zeocin induced 8-10 gamma-H2AX foci per cell. This shows that the Zeocin resistance gene failed to fully detoxify cells treated with Zeocin, and that during selection cells were submitted to a chronic sublethal DSB stress. Selected clones show decreases in both spontaneous and induced intrachromosomal HR. In contrast, in an in vitro assay, these clones show an increase in NHEJ products specific to the KU86 pathway. We selected cells, in the absence of pcDNA3.1/Zeo, with low and sublethal doses of Zeocin, producing a mean 8-10 gamma-H2AX foci per cell. Newly selected clones exhibited similar phenotypes: HR decrease accompanied by an increase in KU86-dependent NHEJ efficiency. Thus chronic exposure to sublethal numbers of DSBs selects cells whose HR versus NHEJ balance is altered. This may well have implications for radio- and chemotherapy, and for management of environmental hazards.


Subject(s)
Bleomycin/toxicity , Recombination, Genetic , Animals , Bleomycin/administration & dosage , CHO Cells , Clone Cells , Cricetinae , Cricetulus , DNA Breaks, Double-Stranded , DNA Repair , In Vitro Techniques , Transfection
10.
Cell Signal ; 14(12): 969-75, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12359302

ABSTRACT

DNA double-strand breaks (DSBs) are the major lethal lesion induced by ionizing radiation or by replication block. However, cells can take advantage of DSB-induced recombination in order to generate genetic diversity in physiological processes such as meiosis and V(D)J recombination. Two main alternative pathways compete for DSB repair: homologous recombination (HR) and non-homologous end-joining (NHEJ). This review will briefly present the mechanisms and the enzymatic complex for HR and NHEJ. The signalling of the DSB through the ATM pathway will be presented. Then, we will focus on the case of the RAD51 protein, which plays a pivotal role in HR and is conserved from bacteria to humans. Post-translational regulation of RAD51 is presented. Two contrasting situations are discussed: one with up-regulation (expression of the oncogene BCR/ABL) and one with a down-regulation (expression of the oncogene BCL-2) of RAD51, associated with apoptosis inhibition and tumour predisposition.


Subject(s)
DNA Repair , DNA-Binding Proteins/metabolism , Signal Transduction , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins , DNA Damage , Fusion Proteins, bcr-abl/metabolism , Genetic Predisposition to Disease , Models, Genetic , Neoplasms/genetics , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rad51 Recombinase , Recombination, Genetic , Tumor Suppressor Proteins
11.
Nucleic Acids Res ; 30(15): 3454-63, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12140331

ABSTRACT

Non-homologous end joining (NHEJ) and homologous recombination (HR) are two alternative/competitor pathways for the repair of DNA double-strand breaks (DSBs). To gain further insights into the regulation of DSB repair, we detail here the different HR pathways affected by (i) the inactivation of DNA-PK activity, by treatment with Wortmannin, and (ii) a mutation in the xrcc4 gene, involved in a late NHEJ step, using the XR-1 cell line. Here we have analyzed not only the impact of NHEJ inactivation on recombination induced by a single DSB targeted to the recombination substrate (using I-SceI endonuclease) but also on gamma-ray- and UV-C-induced and spontaneous recombination and finally on Rad51 foci formation, i.e. on the assembly of the homologous recombination complex, at the molecular level. The results presented here show that in contrast to embryonic stem cells, the xrcc4 mutation strongly stimulates I-SceI-induced HR in adult hamster cells. More precisely, we show here that both single strand annealing and gene conversion are stimulated. In contrast, Wortmannin does not affect I-SceI-induced HR. In addition, gamma-ray-induced recombination is stimulated by both xrcc4 mutation and Wortmannin treatment in an epistatic-like manner. In contrast, neither spontaneous nor UV-C-induced recombination was affected by xrcc4 mutation, showing that the channeling from NHEJ to HR is specific to DSBs. Finally, we show here that xrcc4 mutation or Wortmannin treatment results in a stimulation of Rad51 foci assembly, thus that a late NHEJ step is able to affect Rad51 recombination complex assembly. The present data suggest a model according to which NHEJ and HR do not simply compete for DSB repair but can act sequentially: a defect in a late NHEJ step is not a dead end and can make DSB available for subsequent Rad51 recombination complex assembly.


Subject(s)
Androstadienes/pharmacology , DNA Repair , DNA-Binding Proteins/genetics , Enzyme Inhibitors/pharmacology , Recombination, Genetic , Animals , CHO Cells , Cell Line , Cricetinae , DNA Damage , DNA-Activated Protein Kinase , DNA-Binding Proteins/metabolism , Gamma Rays , Models, Genetic , Mutation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Rad51 Recombinase , Radiation Tolerance , Signal Transduction , Ultraviolet Rays , Wortmannin
SELECTION OF CITATIONS
SEARCH DETAIL
...