Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Hum Mol Genet ; 31(1): 41-56, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34312665

ABSTRACT

Alternative splicing has emerged as a fundamental mechanism for the spatiotemporal control of development. A better understanding of how this mechanism is regulated has the potential not only to elucidate fundamental biological principles, but also to decipher pathological mechanisms implicated in diseases where normal splicing networks are misregulated. Here, we took advantage of human pluripotent stem cells to decipher during human myogenesis the role of muscleblind-like (MBNL) proteins, a family of tissue-specific splicing regulators whose loss of function is associated with myotonic dystrophy type 1 (DM1), an inherited neuromuscular disease. Thanks to the CRISPR/Cas9 technology, we generated human-induced pluripotent stem cells (hiPSCs) depleted in MBNL proteins and evaluated the consequences of their losses on the generation of skeletal muscle cells. Our results suggested that MBNL proteins are required for the late myogenic maturation. In addition, loss of MBNL1 and MBNL2 recapitulated the main features of DM1 observed in hiPSC-derived skeletal muscle cells. Comparative transcriptomic analyses also revealed the muscle-related processes regulated by these proteins that are commonly misregulated in DM1. Together, our study reveals the temporal requirement of MBNL proteins in human myogenesis and should facilitate the identification of new therapeutic strategies capable to cope with the loss of function of these MBNL proteins.


Subject(s)
Induced Pluripotent Stem Cells , Myotonic Dystrophy , Alternative Splicing , Gene Editing , Humans , Induced Pluripotent Stem Cells/metabolism , Muscle Development/genetics , Myotonic Dystrophy/pathology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
2.
Mol Ther ; 30(1): 145-163, 2022 01 05.
Article in English | MEDLINE | ID: mdl-34418541

ABSTRACT

Sickle cell disease (SCD) is caused by a mutation in the ß-globin gene leading to polymerization of the sickle hemoglobin (HbS) and deformation of red blood cells. Autologous transplantation of hematopoietic stem/progenitor cells (HSPCs) genetically modified using lentiviral vectors (LVs) to express an anti-sickling ß-globin leads to some clinical benefit in SCD patients, but it requires high-level transgene expression (i.e., high vector copy number [VCN]) to counteract HbS polymerization. Here, we developed therapeutic approaches combining LV-based gene addition and CRISPR-Cas9 strategies aimed to either knock down the sickle ß-globin and increase the incorporation of an anti-sickling globin (AS3) in hemoglobin tetramers, or to induce the expression of anti-sickling fetal γ-globins. HSPCs from SCD patients were transduced with LVs expressing AS3 and a guide RNA either targeting the endogenous ß-globin gene or regions involved in fetal hemoglobin silencing. Transfection of transduced cells with Cas9 protein resulted in high editing efficiency, elevated levels of anti-sickling hemoglobins, and rescue of the SCD phenotype at a significantly lower VCN compared to the conventional LV-based approach. This versatile platform can improve the efficacy of current gene addition approaches by combining different therapeutic strategies, thus reducing the vector amount required to achieve a therapeutic VCN and the associated genotoxicity risk.


Subject(s)
Anemia, Sickle Cell , Gene Editing , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/therapy , CRISPR-Associated Protein 9/genetics , Fetal Hemoglobin/genetics , Gene Editing/methods , Humans , beta-Globins/genetics
3.
J Cell Sci ; 133(10)2020 05 27.
Article in English | MEDLINE | ID: mdl-32461338

ABSTRACT

Detection of the apoptosis signature becomes central in understanding cell death modes. We present here a whole-cell biosensor that detects Apaf-1 association and apoptosome formation using a split-luciferase complementary assay. Fusion of N-terminal (Nluc) and C-terminal (Cluc)-fragments of firefly luciferase to the N-terminus of human Apaf-1 was performed in HEK293 cells by using CRISPR-Cas9 technology. This resulted in a luminescent form of the apoptosome that we named 'Lumiptosome'. During Apaf-1 gene editing, a high number of knock-in events were observed without selection, suggesting that the Apaf-1 locus is important for the integration of exogenous transgenes. Since activation of caspase-9 is directly dependent on the apoptosome formation, measured reconstitution of luciferase activity should result from the cooperative association of Nluc-Apaf-1 and Cluc-Apaf-1. Time-response measurements also confirmed that formation of the apoptosome occurs prior to activation of caspase-3. Additionally, overexpression of the Bcl2 apoptosis regulator in transgenic and normal HEK293 cells confirmed that formation of the Lumiptosome depends on release of cytochrome c Thus, HEK293 cells that stably express the Lumiptosome can be utilized to screen pro- and anti-apoptotic drugs, and to examine Apaf-1-dependent cellular pathways.


Subject(s)
Apoptosis , Apoptosomes , Apoptosis/genetics , Apoptosomes/metabolism , Apoptotic Protease-Activating Factor 1/genetics , Apoptotic Protease-Activating Factor 1/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cell Death , Cytochromes c/genetics , Cytochromes c/metabolism , HEK293 Cells , Humans
4.
PLoS Genet ; 15(10): e1008355, 2019 10.
Article in English | MEDLINE | ID: mdl-31584931

ABSTRACT

Deficiency in several of the classical human RAD51 paralogs [RAD51B, RAD51C, RAD51D, XRCC2 and XRCC3] is associated with cancer predisposition and Fanconi anemia. To investigate their functions, isogenic disruption mutants for each were generated in non-transformed MCF10A mammary epithelial cells and in transformed U2OS and HEK293 cells. In U2OS and HEK293 cells, viable ablated clones were readily isolated for each RAD51 paralog; in contrast, with the exception of RAD51B, RAD51 paralogs are cell-essential in MCF10A cells. Underlining their importance for genomic stability, mutant cell lines display variable growth defects, impaired sister chromatid recombination, reduced levels of stable RAD51 nuclear foci, and hyper-sensitivity to mitomycin C and olaparib, with the weakest phenotypes observed in RAD51B-deficient cells. Altogether these observations underscore the contributions of RAD51 paralogs in diverse DNA repair processes, and demonstrate essential differences in different cell types. Finally, this study will provide useful reagents to analyze patient-derived mutations and to investigate mechanisms of chemotherapeutic resistance deployed by cancers.


Subject(s)
DNA Repair/genetics , DNA-Binding Proteins/genetics , Homologous Recombination/genetics , Rad51 Recombinase/genetics , Cell Nucleus/genetics , Chromatids/genetics , DNA Damage/genetics , Genome, Human/genetics , HEK293 Cells , Humans , Mutation
5.
PLoS Genet ; 15(8): e1008013, 2019 08.
Article in English | MEDLINE | ID: mdl-31437150

ABSTRACT

Teleost fishes, thanks to their rapid evolution of sex determination mechanisms, provide remarkable opportunities to study the formation of sex chromosomes and the mechanisms driving the birth of new master sex determining (MSD) genes. However, the evolutionary interplay between the sex chromosomes and the MSD genes they harbor is rather unexplored. We characterized a male-specific duplicate of the anti-Müllerian hormone (amh) as the MSD gene in Northern Pike (Esox lucius), using genomic and expression evidence as well as by loss-of-function and gain-of-function experiments. Using RAD-Sequencing from a family panel, we identified Linkage Group (LG) 24 as the sex chromosome and positioned the sex locus in its sub-telomeric region. Furthermore, we demonstrated that this MSD originated from an ancient duplication of the autosomal amh gene, which was subsequently translocated to LG24. Using sex-specific pooled genome sequencing and a new male genome sequence assembled using Nanopore long reads, we also characterized the differentiation of the X and Y chromosomes, revealing a small male-specific insertion containing the MSD gene and a limited region with reduced recombination. Our study reveals an unexpectedly low level of differentiation between a pair of sex chromosomes harboring an old MSD gene in a wild teleost fish population, and highlights both the pivotal role of genes from the amh pathway in sex determination, as well as the importance of gene duplication as a mechanism driving the turnover of sex chromosomes in this clade.


Subject(s)
Anti-Mullerian Hormone/genetics , Esocidae/physiology , Sex Chromosomes/genetics , Sex Determination Processes/genetics , Animals , Animals, Genetically Modified , Chromosome Mapping , Female , Gene Duplication , Gene Knockdown Techniques , Male , Phylogeny , Synteny
6.
PLoS Genet ; 14(8): e1007581, 2018 08.
Article in English | MEDLINE | ID: mdl-30080860

ABSTRACT

Cis-regulation plays an essential role in the control of gene expression, and is particularly complex and poorly understood for developmental genes, which are subject to multiple levels of modulation. In this study, we performed a global analysis of the cis-acting elements involved in the control of the zebrafish developmental gene krox20. krox20 encodes a transcription factor required for hindbrain segmentation and patterning, a morphogenetic process highly conserved during vertebrate evolution. Chromatin accessibility analysis reveals a cis-regulatory landscape that includes 6 elements participating in the control of initiation and autoregulatory aspects of krox20 hindbrain expression. Combining transgenic reporter analyses and CRISPR/Cas9-mediated mutagenesis, we assign precise functions to each of these 6 elements and provide a comprehensive view of krox20 cis-regulation. Three important features emerged. First, cooperation between multiple cis-elements plays a major role in the regulation. Cooperation can surprisingly combine synergy and redundancy, and is not restricted to transcriptional enhancer activity (for example, 4 distinct elements cooperate through different modes to maintain autoregulation). Second, several elements are unexpectedly versatile, which allows them to be involved in different aspects of control of gene expression. Third, comparative analysis of the elements and their activities in several vertebrate species reveals that this versatility is underlain by major plasticity across evolution, despite the high conservation of the gene expression pattern. These characteristics are likely to be of broad significance for developmental genes.


Subject(s)
Early Growth Response Protein 2/genetics , Gene Expression Regulation, Developmental , Rhombencephalon/metabolism , Zebrafish Proteins/genetics , Zebrafish/genetics , Amino Acid Sequence , Animals , CRISPR-Cas Systems , Chromatin/metabolism , Early Growth Response Protein 2/physiology , Enhancer Elements, Genetic , Evolution, Molecular , Genetic Loci , Morphogenesis/genetics , Transcriptional Activation , Zebrafish/embryology
7.
Nucleic Acids Res ; 46(W1): W242-W245, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29762716

ABSTRACT

CRISPOR.org is a web tool for genome editing experiments with the CRISPR-Cas9 system. It finds guide RNAs in an input sequence and ranks them according to different scores that evaluate potential off-targets in the genome of interest and predict on-target activity. The list of genomes is continuously expanded, with more 150 genomes added in the last two years. CRISPOR tries to provide a comprehensive solution from selection, cloning and expression of guide RNA as well as providing primers needed for testing guide activity and potential off-targets. Recent developments include batch design for genome-wide CRISPR and saturation screens, creating custom oligonucleotides for guide cloning and the design of next generation sequencing primers to test for off-target mutations. CRISPOR is available from http://crispor.org, including the full source code of the website and a stand-alone, command-line version.


Subject(s)
CRISPR-Cas Systems , Gene Editing , RNA/chemistry , Software , Base Sequence , CRISPR-Associated Protein 9 , Gene Knockout Techniques , Genome , Internet , Mutagenesis , Oligonucleotides , Workflow
8.
EMBO J ; 34(11): 1572-88, 2015 Jun 03.
Article in English | MEDLINE | ID: mdl-25908840

ABSTRACT

Transcriptional networks defining stemness in adult neural stem cells (NSCs) are largely unknown. We used the proximal cis-regulatory element (pCRE) of the retina-specific homeobox gene 2 (rx2) to address such a network. Lineage analysis in the fish retina identified rx2 as marker for multipotent NSCs. rx2-positive cells located in the peripheral ciliary marginal zone behave as stem cells for the neuroretina, or the retinal pigmented epithelium. We identified upstream regulators of rx2 interrogating the rx2 pCRE in a trans-regulation screen and focused on four TFs (Sox2, Tlx, Gli3, and Her9) activating or repressing rx2 expression. We demonstrated direct interaction of the rx2 pCRE with the four factors in vitro and in vivo. By conditional mosaic gain- and loss-of-function analyses, we validated the activity of those factors on regulating rx2 transcription and consequently modulating neuroretinal and RPE stem cell features. This becomes obvious by the rx2-mutant phenotypes that together with the data presented above identify rx2 as a transcriptional hub balancing stemness of neuroretinal and RPE stem cells in the adult fish retina.


Subject(s)
Adult Stem Cells/metabolism , Fish Proteins/metabolism , Homeodomain Proteins/metabolism , Neural Stem Cells/metabolism , Oryzias/metabolism , Retina/metabolism , Transcription Factors/metabolism , Adult Stem Cells/cytology , Animals , Fish Proteins/genetics , Gene Expression Regulation/physiology , Homeodomain Proteins/genetics , Neural Stem Cells/cytology , Oryzias/genetics , Retina/cytology , Transcription Factors/genetics
9.
Genome Res ; 26(5): 681-92, 2016 05.
Article in English | MEDLINE | ID: mdl-26957310

ABSTRACT

CRISPR/Cas9-mediated targeted mutagenesis allows efficient generation of loss-of-function alleles in zebrafish. To date, this technology has been primarily used to generate genetic knockout animals. Nevertheless, the study of the function of certain loci might require tight spatiotemporal control of gene inactivation. Here, we show that tissue-specific gene disruption can be achieved by driving Cas9 expression with the Gal4/UAS system. Furthermore, by combining the Gal4/UAS and Cre/loxP systems, we establish a versatile tool to genetically label mutant cell clones, enabling their phenotypic analysis. Our technique has the potential to be applied to diverse model organisms, enabling tissue-specific loss-of-function and phenotypic characterization of live and fixed tissues.


Subject(s)
CRISPR-Cas Systems , Gene Silencing , Organisms, Genetically Modified , Zebrafish , Animals , Organisms, Genetically Modified/genetics , Organisms, Genetically Modified/metabolism , Zebrafish/genetics , Zebrafish/metabolism
10.
Stem Cells ; 36(9): 1421-1429, 2018 09.
Article in English | MEDLINE | ID: mdl-29808941

ABSTRACT

Heterozygous PAX6 gene mutations leading to haploinsufficiency are the main cause of congenital aniridia, a rare and progressive panocular disease characterized by reduced visual acuity. Up to 90% of patients suffer from aniridia-related keratopathy (ARK), caused by a combination of factors including limbal epithelial stem cell (LSC) deficiency, impaired healing response and abnormal differentiation of the corneal epithelium. It usually begins in the first decade of life, resulting in recurrent corneal erosions, sub-epithelial fibrosis, and corneal opacification. Unfortunately, there are currently no efficient treatments available for these patients and no in vitro model for this pathology. We used CRISPR/Cas9 technology to introduce into the PAX6 gene of LSCs a heterozygous nonsense mutation found in ARK patients. Nine clones carrying a p.E109X mutation on one allele were obtained with no off-target mutations. Compared with the parental LSCs, heterozygous mutant LSCs displayed reduced expression of PAX6 and marked slow-down of cell proliferation, migration and detachment. Moreover, addition to the culture medium of recombinant PAX6 protein fused to a cell penetrating peptide was able to activate the endogenous PAX6 gene and to rescue phenotypic defects of mutant LSCs, suggesting that administration of such recombinant PAX6 protein could be a promising therapeutic approach for aniridia-related keratopathy. More generally, our results demonstrate that introduction of disease mutations into LSCs by CRISPR/Cas9 genome editing allows the creation of relevant cellular models of ocular disease that should greatly facilitate screening of novel therapeutic approaches. Stem Cells 2018;36:1421-1429.


Subject(s)
Aniridia/genetics , CRISPR-Cas Systems/physiology , Epithelium, Corneal/metabolism , Gene Editing/methods , PAX6 Transcription Factor/genetics , Aniridia/pathology , Humans
11.
Nucleic Acids Res ; 45(7): 4158-4173, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28003477

ABSTRACT

Argonaute (Ago) proteins associate with microRNAs (miRNAs) to form the core of the RNA-induced silencing complex (RISC) that mediates post-transcriptional gene silencing of target mRNAs. As key players in anti-viral defense, Ago proteins are thought to have the ability to interact with human immunodeficiency virus type 1 (HIV-1) RNA. However, the role of this interaction in regulating HIV-1 replication has been debated. Here, we used high throughput sequencing of RNA isolated by cross-linking immunoprecipitation (HITS-CLIP) to explore the interaction between Ago2 and HIV-1 RNA in infected cells. By only considering reads of 50 nucleotides length in our analysis, we identified more than 30 distinct binding sites for Ago2 along the viral RNA genome. Using reporter assays, we found four binding sites, located near splice donor sites, capable of repressing Luciferase gene expression in an Ago-dependent manner. Furthermore, inhibition of Ago1 and Ago2 levels in cells expressing HIV-1 led to an increase of viral multiply spliced transcripts and to a strong reduction in the extracellular CAp24 level. Depletion of Dicer did not affect these activities. Our results highlight a new role of Ago proteins in the control of multiply spliced HIV-1 transcript levels and viral production, independently of the miRNA pathway.


Subject(s)
Alternative Splicing , Argonaute Proteins/metabolism , HIV-1/genetics , RNA, Messenger/metabolism , RNA, Viral/metabolism , Binding Sites , DEAD-box RNA Helicases/metabolism , Eukaryotic Initiation Factors/metabolism , Genome, Viral , HEK293 Cells , HIV-1/physiology , HeLa Cells , High-Throughput Nucleotide Sequencing , Humans , Immunoprecipitation , Jurkat Cells , RNA Precursors/metabolism , RNA Splice Sites , RNA, Viral/chemistry , Ribonuclease III/metabolism , Sequence Analysis, RNA , Virion/physiology
12.
Nucleic Acids Res ; 44(18): 8621-8640, 2016 Oct 14.
Article in English | MEDLINE | ID: mdl-27302134

ABSTRACT

Myogenic regulatory factors of the MyoD family have the ability to reprogram differentiated cells toward a myogenic fate. In this study, we demonstrate that Six1 or Six4 are required for the reprogramming by MyoD of mouse embryonic fibroblasts (MEFs). Using microarray experiments, we found 761 genes under the control of both Six and MyoD. Using MyoD ChIPseq data and a genome-wide search for Six1/4 MEF3 binding sites, we found significant co-localization of binding sites for MyoD and Six proteins on over a thousand mouse genomic DNA regions. The combination of both datasets yielded 82 genes which are synergistically activated by Six and MyoD, with 96 associated MyoD+MEF3 putative cis-regulatory modules (CRMs). Fourteen out of 19 of the CRMs that we tested demonstrated in Luciferase assays a synergistic action also observed for their cognate gene. We searched putative binding sites on these CRMs using available databases and de novo search of conserved motifs and demonstrated that the Six/MyoD synergistic activation takes place in a feedforward way. It involves the recruitment of these two families of transcription factors to their targets, together with partner transcription factors, encoded by genes that are themselves activated by Six and MyoD, including Mef2, Pbx-Meis and EBF.


Subject(s)
Cellular Reprogramming/genetics , Genome , Homeodomain Proteins/metabolism , MyoD Protein/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Trans-Activators/metabolism , Animals , Base Sequence , Binding Sites/genetics , Cells, Cultured , Chromatin Immunoprecipitation , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Homeodomain Proteins/genetics , Humans , Luciferases/metabolism , Mice, Knockout , Muscle Development/genetics , Mutation/genetics , Nuclear Proteins/metabolism , Nucleotide Motifs/genetics , Reproducibility of Results , Trans-Activators/genetics , Transcription Factors/metabolism
13.
Nucleic Acids Res ; 44(16): 7804-16, 2016 09 19.
Article in English | MEDLINE | ID: mdl-27466392

ABSTRACT

Mitochondrial diseases are frequently associated with mutations in mitochondrial DNA (mtDNA). In most cases, mutant and wild-type mtDNAs coexist, resulting in heteroplasmy. The selective elimination of mutant mtDNA, and consequent enrichment of wild-type mtDNA, can rescue pathological phenotypes in heteroplasmic cells. Use of the mitochondrially targeted zinc finger-nuclease (mtZFN) results in degradation of mutant mtDNA through site-specific DNA cleavage. Here, we describe a substantial enhancement of our previous mtZFN-based approaches to targeting mtDNA, allowing near-complete directional shifts of mtDNA heteroplasmy, either by iterative treatment or through finely controlled expression of mtZFN, which limits off-target catalysis and undesired mtDNA copy number depletion. To demonstrate the utility of this improved approach, we generated an isogenic distribution of heteroplasmic cells with variable mtDNA mutant level from the same parental source without clonal selection. Analysis of these populations demonstrated an altered metabolic signature in cells harbouring decreased levels of mutant m.8993T>G mtDNA, associated with neuropathy, ataxia, and retinitis pigmentosa (NARP). We conclude that mtZFN-based approaches offer means for mtDNA heteroplasmy manipulation in basic research, and may provide a strategy for therapeutic intervention in selected mitochondrial diseases.


Subject(s)
DNA, Mitochondrial/genetics , Endonucleases/metabolism , Mitochondria/metabolism , Mutation/genetics , Zinc Fingers , Cell Line, Tumor , Flow Cytometry , Gene Dosage , Humans , RNA, Catalytic/metabolism
14.
Genome Res ; 24(1): 142-53, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24179142

ABSTRACT

Sequence-specific nucleases like TALENs and the CRISPR/Cas9 system have greatly expanded the genome editing possibilities in model organisms such as zebrafish. Both systems have recently been used to create knock-out alleles with great efficiency, and TALENs have also been successfully employed in knock-in of DNA cassettes at defined loci via homologous recombination (HR). Here we report CRISPR/Cas9-mediated knock-in of DNA cassettes into the zebrafish genome at a very high rate by homology-independent double-strand break (DSB) repair pathways. After co-injection of a donor plasmid with a short guide RNA (sgRNA) and Cas9 nuclease mRNA, concurrent cleavage of donor plasmid DNA and the selected chromosomal integration site resulted in efficient targeted integration of donor DNA. We successfully employed this approach to convert eGFP into Gal4 transgenic lines, and the same plasmids and sgRNAs can be applied in any species where eGFP lines were generated as part of enhancer and gene trap screens. In addition, we show the possibility of easily targeting DNA integration at endogenous loci, thus greatly facilitating the creation of reporter and loss-of-function alleles. Due to its simplicity, flexibility, and very high efficiency, our method greatly expands the repertoire for genome editing in zebrafish and can be readily adapted to many other organisms.


Subject(s)
CRISPR-Associated Proteins/metabolism , DNA Repair , Gene Knock-In Techniques , Genetic Engineering/methods , Zebrafish/genetics , Animals , CRISPR-Associated Proteins/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , DNA Breaks, Double-Stranded , Genome , Mutagenesis , Recombinational DNA Repair , Zebrafish/embryology , RNA, Small Untranslated
15.
Genome Res ; 24(8): 1371-83, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24989021

ABSTRACT

The generation of genetically modified animals is important for both research and commercial purposes. The rat is an important model organism that until recently lacked efficient genetic engineering tools. Sequence-specific nucleases, such as ZFNs, TALE nucleases, and CRISPR/Cas9 have allowed the creation of rat knockout models. Genetic engineering by homology-directed repair (HDR) is utilized to create animals expressing transgenes in a controlled way and to introduce precise genetic modifications. We applied TALE nucleases and donor DNA microinjection into zygotes to generate HDR-modified rats with large new sequences introduced into three different loci with high efficiency (0.62%-5.13% of microinjected zygotes). Two of these loci (Rosa26 and Hprt1) are known to allow robust and reproducible transgene expression and were targeted for integration of a GFP expression cassette driven by the CAG promoter. GFP-expressing embryos and four Rosa26 GFP rat lines analyzed showed strong and widespread GFP expression in most cells of all analyzed tissues. The third targeted locus was Ighm, where we performed successful exon exchange of rat exon 2 for the human one. At all three loci we observed HDR only when using linear and not circular donor DNA. Mild hypothermic (30°C) culture of zygotes after microinjection increased HDR efficiency for some loci. Our study demonstrates that TALE nuclease and donor DNA microinjection into rat zygotes results in efficient and reproducible targeted donor integration by HDR. This allowed creation of genetically modified rats in a work-, cost-, and time-effective manner.


Subject(s)
Gene Targeting , Genetic Engineering , Animals , Base Sequence , Cells, Cultured , DNA Restriction Enzymes/biosynthesis , DNA Restriction Enzymes/genetics , Female , Hypoxanthine Phosphoribosyltransferase/genetics , Male , Microinjections , Rats, Sprague-Dawley , Rats, Transgenic , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinational DNA Repair , Zygote
16.
Genome Res ; 23(7): 1182-93, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23568838

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.


Subject(s)
Endonucleases/metabolism , Neoplasms/enzymology , Neoplasms/genetics , Translocation, Genetic , Zinc Fingers , Cell Line , Chromosome Breakpoints , Humans , Nucleophosmin , Protein-Tyrosine Kinases/genetics , Sarcoma, Ewing/genetics , Sarcoma, Ewing/metabolism
17.
Methods ; 69(1): 102-7, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24583114

ABSTRACT

The rat is a model of choice to understanding gene function and modeling human diseases. Since recent years, successful engineering technologies using gene-specific nucleases have been developed to gene edit the genome of different species, including the rat. This development has become important for the creation of new rat animals models of human diseases, analyze the role of genes and express recombinant proteins. Transcription activator-like (TALE) nucleases are designed nucleases consist of a DNA binding domain fused to a nuclease domain capable of cleaving the targeted DNA. We describe a detailed protocol for generating knockout rats via microinjection of TALE nucleases into fertilized eggs. This technology is an efficient, cost- and time-effective method for creating new rat models.


Subject(s)
Gene Knockout Techniques , Mutagenesis, Site-Directed/methods , Animals , DNA End-Joining Repair , Deoxyribonucleases/chemistry , Deoxyribonucleases/genetics , Embryo Transfer , Embryo, Mammalian , Female , Homologous Recombination , Microinjections , Rats , Rats, Sprague-Dawley
18.
Hum Mol Genet ; 21(9): 1968-78, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22246292

ABSTRACT

SERPINA3 (Serpin peptidase inhibitor clade A member 3), also known as a1-antichymotrypsin, is a serine protease inhibitor involved in a wide range of biological processes. Recently, it has been shown to be up-regulated in human placental diseases in association with a hypomethylation of the 5' region of the gene. In the present study, we show that the promoter of SERPINA3 is transcriptionally activated by three transcription factors (TFs) (SP1, MZF1 and ZBTB7B), the level of induction being dependent on the rs1884082 single nucleotide polymorphism (SNP) located inside the promoter, the T allele being consistently induced to a higher level than the G, with or without added TFs. When the promoter was methylated, the response to ZBTB7B was allele specific (the G allele was strongly induced, while the T allele was strongly down-regulated). We propose an adaptive model to explain the interest of such a regulation for placental function and homeostasis. Overexpression of SERPINA3 in JEG-3 cells, a trophoblast cell model, decreased cell adhesion to the extracellular matrix and to neighboring cells, but protects them from apoptosis, suggesting a way by which this factor could be deleterious at high doses. In addition, we show in different human populations that the T allele appears to predispose to Intra Uterine Growth Restriction (IUGR), while a G allele at a second SNP located in the second exon (rs4634) increases the risk of preeclampsia. Our results provide mechanistic views inside the involvement of SERPINA3 in placental diseases, through its regulation by a combination of epigenetic, genetic and TF-mediated regulations.


Subject(s)
Placenta Diseases/genetics , Serpins/genetics , Alleles , Apoptosis , Base Sequence , Case-Control Studies , Cell Adhesion , Cell Line , DNA Methylation , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Female , Fetal Growth Retardation/genetics , Fetal Growth Retardation/metabolism , Gene Expression Regulation , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Hypoxia/genetics , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kruppel-Like Transcription Factors/metabolism , Models, Biological , Placenta Diseases/metabolism , Polymorphism, Single Nucleotide , Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , Pregnancy , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Trophoblasts/cytology , Trophoblasts/metabolism , Zinc Fingers
19.
Stem Cell Res Ther ; 15(1): 10, 2024 01 02.
Article in English | MEDLINE | ID: mdl-38167524

ABSTRACT

BACKGROUND: Beyond the observed alterations in cellular structure and mitochondria, the mechanisms linking rare genetic mutations to the development of heart failure in patients affected by desmin mutations remain unclear due in part, to the lack of relevant human cardiomyocyte models. METHODS: To shed light on the role of mitochondria in these mechanisms, we investigated cardiomyocytes derived from human induced pluripotent stem cells carrying the heterozygous DESE439K mutation that were either isolated from a patient or generated by gene editing. To increase physiological relevance, cardiomyocytes were either cultured on an anisotropic micropatterned surface to obtain elongated and aligned cardiomyocytes, or as a cardiac spheroid to create a micro-tissue. Moreover, when applicable, results from cardiomyocytes were confirmed with heart biopsies of suddenly died patient of the same family harboring DESE439K mutation, and post-mortem heart samples from five control healthy donors. RESULTS: The heterozygous DESE439K mutation leads to dramatic changes in the overall cytoarchitecture of cardiomyocytes, including cell size and morphology. Most importantly, mutant cardiomyocytes display altered mitochondrial architecture, mitochondrial respiratory capacity and metabolic activity reminiscent of defects observed in patient's heart tissue. Finally, to challenge the pathological mechanism, we transferred normal mitochondria inside the mutant cardiomyocytes and demonstrated that this treatment was able to restore mitochondrial and contractile functions of cardiomyocytes. CONCLUSIONS: This work highlights the deleterious effects of DESE439K mutation, demonstrates the crucial role of mitochondrial abnormalities in the pathophysiology of desmin-related cardiomyopathy, and opens up new potential therapeutic perspectives for this disease.


Subject(s)
Cardiomyopathies , Induced Pluripotent Stem Cells , Humans , Desmin/genetics , Desmin/metabolism , Induced Pluripotent Stem Cells/metabolism , Cardiomyopathies/metabolism , Mutation/genetics , Myocytes, Cardiac/metabolism , Mitochondria/genetics , Mitochondria/metabolism
20.
Elife ; 132024 Jul 09.
Article in English | MEDLINE | ID: mdl-38980300

ABSTRACT

Tardigrades are microscopic animals renowned for their ability to withstand extreme conditions, including high doses of ionizing radiation (IR). To better understand their radio-resistance, we first characterized induction and repair of DNA double- and single-strand breaks after exposure to IR in the model species Hypsibius exemplaris. Importantly, we found that the rate of single-strand breaks induced was roughly equivalent to that in human cells, suggesting that DNA repair plays a predominant role in tardigrades' radio-resistance. To identify novel tardigrade-specific genes involved, we next conducted a comparative transcriptomics analysis across three different species. In all three species, many DNA repair genes were among the most strongly overexpressed genes alongside a novel tardigrade-specific gene, which we named Tardigrade DNA damage Response 1 (TDR1). We found that TDR1 protein interacts with DNA and forms aggregates at high concentration suggesting it may condensate DNA and preserve chromosome organization until DNA repair is accomplished. Remarkably, when expressed in human cells, TDR1 improved resistance to Bleomycin, a radiomimetic drug. Based on these findings, we propose that TDR1 is a novel tardigrade-specific gene conferring resistance to IR. Our study sheds light on mechanisms of DNA repair helping cope with high levels of DNA damage inflicted by IR.


Subject(s)
DNA Repair , DNA-Binding Proteins , Radiation, Ionizing , Tardigrada , Transcriptome , Tardigrada/genetics , Tardigrada/metabolism , Animals , Humans , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Gene Expression Profiling , DNA Damage , Radiation Tolerance/genetics
SELECTION OF CITATIONS
SEARCH DETAIL