Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Stem Cell Res ; 67: 103012, 2023 03.
Article in English | MEDLINE | ID: mdl-36610307

ABSTRACT

C-terminal Binding Protein 1 (CTBP1) is a ubiquitously expressed transcriptional co-repressor and membrane trafficking regulator. A recurrent de novo c.991C>T mutation in CTBP1 leads to expression of p.R331W CTBP1 and causes hypotonia, ataxia, developmental delay, and tooth enamel defects syndrome (HADDTS), a rare early onset neurodevelopmental disorder. We generated hESCs lines with heterozygote and homozygote c.991C>T in CTBP1 using CRISPR/Cas9 genome editing and validated them for genetic integrity, off-target mutations, and pluripotency. They will be useful for investigation of HADDTS pathophysiology and for screening for potential therapeutics.


Subject(s)
Human Embryonic Stem Cells , Humans , Ataxia/genetics , CRISPR-Cas Systems , Heterozygote , Homozygote , Muscle Hypotonia/genetics , Mutation , Transcription Factors/genetics
2.
Stem Cell Res ; 65: 102952, 2022 12.
Article in English | MEDLINE | ID: mdl-36283273

ABSTRACT

Aggregation of alpha-synuclein (aSyn) is closely linked to Parkinson's disease, probably due to the loss of physiological functions and/or gain of toxic functions of aggregated aSyn. Significant efforts have been made elucidating the physiological structure and function of aSyn, however, with limited success thus far in human-derived cells, partly because of restricted resources. Here, we developed two human-induced pluripotent stem cell lines using CRISPR/Cas9-mediated allele-specific frame-shift deletion of the aSyn encoding gene SNCA, resulting in homo- and heterozygous SNCA knockout. The generated cell lines are promising cellular tools for studying aSyn dosage-dependent functions and structural alterations in human neural cells.


Subject(s)
Induced Pluripotent Stem Cells , alpha-Synuclein , Humans , alpha-Synuclein/genetics , Gene Knockout Techniques , CRISPR-Cas Systems/genetics
3.
Brain ; 145(9): 3131-3146, 2022 09 14.
Article in English | MEDLINE | ID: mdl-36103408

ABSTRACT

Pathogenic variants in SPAST, the gene coding for spastin, are the single most common cause of hereditary spastic paraplegia, a progressive motor neuron disease. Spastin regulates key cellular functions, including microtubule-severing and endoplasmic reticulum-morphogenesis. However, it remains unclear how alterations in these cellular functions due to SPAST pathogenic variants result in motor neuron dysfunction. Since spastin influences both microtubule network and endoplasmic reticulum structure, we hypothesized that spastin is necessary for the regulation of Ca2+ homeostasis via store-operated calcium entry. Here, we show that the lack of spastin enlarges the endoplasmic reticulum and reduces store-operated calcium entry. In addition, elevated levels of different spastin variants induced clustering of STIM1 within the endoplasmic reticulum, altered the transport of STIM1 to the plasma membrane and reduced store-operated calcium entry, which could be rescued by exogenous expression of STIM1. Importantly, store-operated calcium entry was strongly reduced in induced pluripotent stem cell-derived neurons from hereditary spastic paraplegia patients with pathogenic variants in SPAST resulting in spastin haploinsufficiency. These neurons developed axonal swellings in response to lack of spastin. We were able to rescue both store-operated calcium entry and axonal swellings in SPAST patient neurons by restoring spastin levels, using CRISPR/Cas9 to correct the pathogenic variants in SPAST. These findings demonstrate that proper amounts of spastin are a key regulatory component for store-operated calcium entry mediated Ca2+ homeostasis and suggest store-operated calcium entry as a disease relevant mechanism of spastin-linked motor neuron disease.


Subject(s)
Spastic Paraplegia, Hereditary , Calcium/metabolism , Humans , Microtubules , Motor Neurons/metabolism , Spastin/genetics
4.
Stem Cell Res ; 56: 102520, 2021 10.
Article in English | MEDLINE | ID: mdl-34479069

ABSTRACT

Pathogenic bi-allelic variants in the SPG11 gene result in rare motor neuron disorders such as Hereditary Spastic Paraplegia type 11, Charcot-Marie Tooth, and Juvenile Amyotrophic Lateral Sclerosis-5. The main challenge in SPG11-linked disease research is the lack of antibodies against SPG11 encoded spatacsin. Here, we describe the CRISPR/Cas9 mediated generation and validation of an endogenously tagged SPG11- human iPSC line that contains an HA tag at the C-terminus of SPG11. The line exhibits multi-lineage differentiation potential and holds promise for studying the role of spatacsin and for the elucidation of SPG11-associated pathogenesis. Resource Table.


Subject(s)
Induced Pluripotent Stem Cells , Spastic Paraplegia, Hereditary , CRISPR-Cas Systems/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Mutation , Proteins/genetics , Spastic Paraplegia, Hereditary/genetics
5.
Stem Cell Res ; 47: 101889, 2020 Jun 29.
Article in English | MEDLINE | ID: mdl-32682288

ABSTRACT

ARID1B haploinsufficiency induced by missense or nonsense mutations of ARID1B is a cause of Coffin-Siris syndrome (CSS), a neurodevelopmental disorder associated with intellectual disability. At present, no appropriate human stem cell model for ARID1B-associated CSS has been reported. Here, we describe the generation and validation of ARID1B+/- hESCs by introducing out of frame deletions into exon 5 or 6 of ARID1B with CRISPR/Cas9 genome editing. These ARID1B+/- hESC lines allow to study the pathophysiology of ARID1B-associated CSS in 2D and 3D models of human neurodevelopment.

6.
Hum Mol Genet ; 28(15): 2589-2599, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31035284

ABSTRACT

The SOXC transcription factors Sox4, Sox11 and Sox12, are critical neurodevelopmental regulators that are thought to function in a highly redundant fashion. Surprisingly, heterozygous missense mutations or deletions of SOX11 were recently detected in patients with Coffin-Siris syndrome-like syndrome (CSSLS), a neurodevelopmental disorder associated with intellectual disability, demonstrating that in humans SOX11 haploinsufficiency cannot be compensated and raising the question of the function of SOX11 in human neurodevelopment. Here, we describe the generation of SOX11+/- heterozygous human embryonic stem cell (hESC) lines by CRISPR/Cas9 genome engineering. SOX11 haploinsufficiency impaired the generation of neurons and resulted in a proliferation/differentiation imbalance of neural precursor cells and enhanced neuronal cell death. Using the SOX11+/- hESC model we provide for the first time experimental evidence that SOX11 haploinsufficiency is sufficient to impair key processes of human neurodevelopment, giving a first insight into the pathophysiology of CSSLS and SOX11 function in human neurodevelopment.


Subject(s)
Cell Line , Gene Dosage , Human Embryonic Stem Cells/physiology , Models, Biological , Neurodevelopmental Disorders/metabolism , SOXC Transcription Factors/genetics , Abnormalities, Multiple/genetics , Abnormalities, Multiple/metabolism , CRISPR-Cas Systems , Cell Differentiation , Cell Proliferation , Face/abnormalities , Gene Editing , Gene Expression Regulation , Hand Deformities, Congenital/genetics , Hand Deformities, Congenital/metabolism , Haploinsufficiency , Human Embryonic Stem Cells/metabolism , Humans , Intellectual Disability/genetics , Intellectual Disability/metabolism , Micrognathism/genetics , Micrognathism/metabolism , Neck/abnormalities , Neural Stem Cells , Neurodevelopmental Disorders/genetics
7.
Front Neurosci ; 12: 914, 2018.
Article in English | MEDLINE | ID: mdl-30574063

ABSTRACT

Mutations in SPG11 cause a complicated autosomal recessive form of hereditary spastic paraplegia (HSP). Mechanistically, there are indications for the dysregulation of the GSK3ß/ßCat signaling pathway in SPG11. In this study, we tested the therapeutic potential of the GSK3ß inhibitor, tideglusib, to rescue neurodegeneration associated characteristics in an induced pluripotent stem cells (iPSCs) derived neuronal model from SPG11 patients and matched healthy controls as well as a CRISPR-Cas9 mediated SPG11 knock-out line and respective control. SPG11-iPSC derived cortical neurons, as well as the genome edited neurons exhibited shorter and less complex neurites than controls. Administration of tideglusib to these lines led to the rescue of neuritic impairments. Moreover, the treatment restored increased cell death and ameliorated the membranous inclusions in iPSC derived SPG11 neurons. Our results provide a first evidence for the rescue of neurite pathology in SPG11-HSP by tideglusib. The current lack of disease-modifying treatments for SPG11 and related types of complicated HSP renders tideglusib a candidate compound for future clinical application.

8.
Cell Tissue Res ; 371(1): 91-103, 2018 01.
Article in English | MEDLINE | ID: mdl-29079881

ABSTRACT

During development, generation of neurons is coordinated by the sequential activation of gene expression programs by stage- and subtype-specific transcription factor networks. The SoxC group transcription factors, Sox4 and Sox11, have recently emerged as critical components of this network. Initially identified as survival and differentiation factors for neural precursors, SoxC factors have now been linked to a broader array of developmental processes including neuronal subtype specification, migration, dendritogenesis and establishment of neuronal projections, and are now being employed in experimental strategies for neuronal replacement and axonal regeneration in the diseased central nervous system. This review summarizes the current knowledge regarding SoxC factor function in CNS development and disease and their promise for regeneration.


Subject(s)
Brain/embryology , Neurogenesis/physiology , Neurons/physiology , SOXC Transcription Factors/metabolism , Animals , Cellular Reprogramming , Gene Expression Regulation, Developmental , Humans , Mice , Models, Animal , Regeneration
9.
Mol Ther ; 24(4): 685-96, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26916285

ABSTRACT

Limb girdle muscular dystrophies types 2B (LGMD2B) and 2D (LGMD2D) are degenerative muscle diseases caused by mutations in the dysferlin and alpha-sarcoglycan genes, respectively. Using patient-derived induced pluripotent stem cells (iPSC), we corrected the dysferlin nonsense mutation c.5713C>T; p.R1905X and the most common alpha-sarcoglycan mutation, missense c.229C>T; p.R77C, by single-stranded oligonucleotide-mediated gene editing, using the CRISPR/Cas9 gene-editing system to enhance the frequency of homology-directed repair. We demonstrated seamless, allele-specific correction at efficiencies of 0.7-1.5%. As an alternative, we also carried out precise gene addition strategies for correction of the LGMD2B iPSC by integration of wild-type dysferlin cDNA into the H11 safe harbor locus on chromosome 22, using dual integrase cassette exchange (DICE) or TALEN-assisted homologous recombination for insertion precise (THRIP). These methods employed TALENs and homologous recombination, and DICE also utilized site-specific recombinases. With DICE and THRIP, we obtained targeting efficiencies after selection of ~20%. We purified iPSC corrected by all methods and verified rescue of appropriate levels of dysferlin and alpha-sarcoglycan protein expression and correct localization, as shown by immunoblot and immunocytochemistry. In summary, we demonstrate for the first time precise correction of LGMD iPSC and validation of expression, opening the possibility of cell therapy utilizing these corrected iPSC.


Subject(s)
Gene Editing/methods , Induced Pluripotent Stem Cells/metabolism , Membrane Proteins/genetics , Muscle Proteins/genetics , Muscular Dystrophies, Limb-Girdle/therapy , Mutation , Alleles , CRISPR-Cas Systems , Cell Line , Cell- and Tissue-Based Therapy/methods , Dysferlin , Female , Genetic Therapy , Homologous Recombination , Humans , Membrane Proteins/metabolism , Muscle Proteins/metabolism , Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies, Limb-Girdle/pathology , Sarcoglycans/genetics
10.
Appl Microbiol Biotechnol ; 98(23): 9723-33, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25158835

ABSTRACT

Over the years, Chinese hamster ovary (CHO) cells have emerged as the major host for expressing biotherapeutic proteins. Traditional methods to generate high-producer cell lines rely on random integration(s) of the gene of interest but have thereby left the identification of bottlenecks as a challenging task. For comparison of different producer cell lines derived from various transfections, a system that provides control over transgene expression behavior is highly needed. This motivated us to develop a novel "DUKX-B11 F3/F" cell line to target different single-chain antibody fragments into the same chromosomal target site by recombinase-mediated cassette exchange (RMCE) using the flippase (FLP)/FLP recognition target (FRT) system. The RMCE-competent cell line contains a gfp reporter fused to a positive/negative selection system flanked by heterospecific FRT (F) variants under control of an external CMV promoter, constructed as "promoter trap". The expression stability and FLP accessibility of the tagged locus was demonstrated by successive rounds of RMCE. As a proof of concept, we performed RMCE using cassettes encoding two different anti-HIV single-chain Fc fragments, 3D6scFv-Fc and 2F5scFv-Fc. Both targeted integrations yielded homogenous cell populations with comparable intracellular product contents and messenger RNA (mRNA) levels but product related differences in specific productivities. These studies confirm the potential of the newly available "DUKX-B11 F3/F" cell line to guide different transgenes into identical transcriptional control regions by RMCE and thereby generate clones with comparable amounts of transgene mRNA. This new host is a prerequisite for cell biology studies of independent transfections and transgenes.


Subject(s)
Gene Expression Profiling , Single-Chain Antibodies/biosynthesis , Animals , CHO Cells , Cricetulus , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Single-Chain Antibodies/genetics , Transgenes
11.
Gene ; 546(2): 135-44, 2014 Aug 10.
Article in English | MEDLINE | ID: mdl-24905650

ABSTRACT

Where possible, developments enabling the establishment of cell lines with predictable, long-term stable expression capacity are based on single-copy integrations at safe genomic loci with predictable properties. Robust performance could be assigned to lentiviral transduction systems anchoring single LV-units at sites with adequate transcription potential. In the case of gene therapeutic vectors it is essential that the expression interval can be safely terminated following individual requirements, which has mostly been achieved by lox-mediated excision ("floxing"). To extend the spectrum of possible applications we replaced the common, phage-derived Cre/loxP-setup by modules derived from the yeast "Flp/FRT" site-specific recombination system. This change enables a variety of additional options, for instance by "multiplexing" strategies, which rely on a variety of heterospecific FRT-site variants (F'). If we provide lentiviral LTRs with a "twin-site", here an FF3 fusion, the presence of Flp-recombinase will effectively excise the expression cassette, leaving behind a single neutral, genomically anchored FF3 unit. This tag serves to identify the integration locus and to apply sequence- and structural (SIDD-) analyses to predict its functions. Candidate loci are then used to accommodate, at the given site, other genes of interest by "Recombinase-Mediated Twin Site Targeting" (RMTT), a contemporary extension of existing cassette exchange (RMCE-) routines. Supported by the fact that FF3 twins remain accessible within the host genome, RMTT provides access to certified cell lines as it complies with recently defined stringent genomic safe harbor criteria. Our discussion- and outlook-sections will cover lentiviral targeting strategies and current possibilities to enable their fine-tuning.


Subject(s)
DNA Nucleotidyltransferases/metabolism , Gene Targeting/methods , Genetic Vectors , Terminal Repeat Sequences , Transduction, Genetic/methods , DNA Nucleotidyltransferases/genetics , Saccharomyces cerevisiae/genetics
12.
Biomaterials ; 35(14): 4345-56, 2014 May.
Article in English | MEDLINE | ID: mdl-24529624

ABSTRACT

Site specific recombinases are frequently used as gene switches in transgenic animals where recombination is induced by drug treatment or by tissue specific recombinase expression. Alternatively, lentiviral gene transfer can be utilized for the genetic modification of a wide variety of cell types, albeit systems for tight control of transcriptional activity are scarce. Here, we combined lentiviral gene transfer and the development of a tightly drug-controlled FLP recombinase for the construction of "All-in-One" inducible gene expression systems. Tight control of FLP activity was achieved through N-terminal fusion with a FKBP12-derived conditional destruction domain and a C-terminal estrogen receptor binding domain making recombination dependent on the presence of Shield-1 and 4-hydroxytamoxifen. Exploiting the capacity of FLP to mediate excision and inversion, "All-in-One" lentiviral gene switch vector systems were generated where drug-induced recombination resulted in abrogation of FLP expression and subsequent overexpression or knockdown of the prototypical tumor suppressor phosphatase and tensin homolog PTEN. "All-in-One" vectors proved their functionality in a variety of hematopoietic cell lines, and primary murine bone marrow cells. Our new vector system thus combines the ease of lentiviral gene transfer and the power of site specific recombinases for analysis of gene function.


Subject(s)
DNA Nucleotidyltransferases/metabolism , Drug Delivery Systems , Genetic Vectors/metabolism , Lentivirus/genetics , Morpholines/pharmacology , Tamoxifen/analogs & derivatives , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Line , Codon/genetics , Female , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred C57BL , PTEN Phosphohydrolase/metabolism , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Tacrolimus Binding Protein 1A/metabolism , Tamoxifen/pharmacology
13.
Mol Ther ; 22(5): 919-28, 2014 May.
Article in English | MEDLINE | ID: mdl-24434935

ABSTRACT

Methods for generating induced pluripotent stem cells (iPSCs) for disease modeling and cell therapies have progressed from integrating vectors to transient delivery of reprogramming factors, avoiding permanent genomic modification. A major limitation of unmodified iPSCs is the assessment of their distribution and contribution to adverse reactions in autologous cell therapy. Here, we report that polycistronic lentiviral vectors with single Flp recombinase (Flp) recognition target (FRT) sites can be used to generate murine iPSCs that are devoid of the reprogramming cassette but carry an intergenic 300-bp long terminal repeat sequence. Performing quantitative polymerase chain reaction on this marker, we could determine genetic identity and tissue contribution of iPSC-derived teratomas in mice. Moreover, we generated iPSCs carrying heterospecific FRT twin sites, enabling excision and recombinase-mediated cassette exchange (RMCE) of the reprogramming cassette for another expression unit of choice. Following screening of iPSCs for "safe harbor" integration sites, expression cassettes were introduced by RMCE into various previously silenced loci of selected single-copy iPSCs. Analysis of DNA methylation showed that RMCE reverted the local epigenetic signature, which allowed transgene expression in undifferentiated iPSCs and in differentiated progeny. These findings support the concept of creating clonotypically defined exchangeable and traceable pluripotent stem cells for disease research and cell therapy.


Subject(s)
Cell Differentiation/genetics , Cell- and Tissue-Based Therapy , DNA Nucleotidyltransferases/genetics , Induced Pluripotent Stem Cells , Terminal Repeat Sequences/genetics , Animals , Cellular Reprogramming , DNA Methylation , Genetic Vectors , Lentivirus/genetics , Mice
14.
Gene ; 515(1): 1-27, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23201421

ABSTRACT

Starting in 1991, the advance of Tyr-recombinases Flp and Cre enabled superior strategies for the predictable insertion of transgenes into compatible target sites of mammalian cells. Early approaches suffered from the reversibility of integration routes and the fact that co-introduction of prokaryotic vector parts triggered uncontrolled heterochromatization. Shortcomings of this kind were overcome when Flp-Recombinase Mediated Cassette Exchange entered the field in 1994. RMCE enables enhanced tag-and-exchange strategies by precisely replacing a genomic target cassette by a compatible donor construct. After "gene swapping" the donor cassette is safely locked in, but can nevertheless be re-mobilized in case other compatible donor cassettes are provided ("serial RMCE"). These features considerably expand the options for systematic, stepwise genome modifications. The first decade was dominated by the systematic generation of cell lines for biotechnological purposes. Based on the reproducible expression capacity of the resulting strains, a comprehensive toolbox emerged to serve a multitude of purposes, which constitute the first part of this review. The concept per se did not, however, provide access to high-producer strains able to outcompete industrial multiple-copy cell lines. This fact gave rise to systematic improvements, among these certain accumulative site-specific integration pathways. The exceptional value of RMCE emerged after its entry into the stem cell field, where it started to contribute to the generation of induced pluripotent stem (iPS-) cells and their subsequent differentiation yielding a variety of cell types for diagnostic and therapeutic purposes. This topic firmly relies on the strategies developed in the first decade and can be seen as the major ambition of the present article. In this context an unanticipated, potent property of serial Flp-RMCE setups concerns the potential to re-open loci that have served to establish the iPS status before the site underwent the obligatory silencing process. Other relevant options relate to the introduction of composite Flp-recognition target sites ("heterospecific FRT-doublets"), into the LTRs of lentiviral vectors. These "twin sites" enhance the safety of iPS re-programming and -differentiation as they enable the subsequent quantitative excision of a transgene, leaving behind a single "FRT-twin". Such a strategy combines the established expression potential of the common retro- and lentiviral systems with options to terminate the process at will. The remaining genomic tag serves to identify and characterize the insertion site with the goal to identify genomic "safe harbors" (GOIs) for re-use. This is enabled by the capacity of "FRT-twins" to accommodate any incoming RMCE-donor cassette with a compatible design.


Subject(s)
Gene Targeting , Recombinases/metabolism , Recombination, Genetic , Animals , Genomics , Humans
15.
FASEB J ; 25(12): 4088-107, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21891781

ABSTRACT

Site-specific recombinases (SSRs) enable novel tag-and-target as well as tag-and-exchange strategies for tailoring mammalian genomes. If used in combination with homologous recombination, which per se is inefficient but can serve to introduce SSR sites, the tagged locus lends itself to repeated modification at largely increased efficiency and specificity. The more conventional SSR-based genetic modifications enable straightforward integration of a transgene with efficiencies depending on both the target locus and the vector composition. Only the more recent tag-and-exchange strategies in conjunction with advanced selection principles enable the clean replacement of a genomically anchored cassette by a donor cassette with the related architecture. Meanwhile this recombinase-mediated cassette exchange (RMCE) concept could be verified for two classes of SSRs, belonging to either the Tyr or the Ser family. Certain members of these open different fields of application that will be discussed with reference to the molecular properties of the respective enzymes. A major aim of our review is to characterize the RMCE-relevant components and describe their optimal utilization in the fields of gene therapy and molecular genomics. Early contributions to the field of experimental animal models will be mentioned considering in vivo modifications enabled by microinjection into oocytes.


Subject(s)
DNA Nucleotidyltransferases/metabolism , Recombination, Genetic , Animals , DNA/genetics , DNA/metabolism , Female , Genetic Therapy , Genomics , Humans , Male , Models, Animal , Models, Genetic
16.
J Mol Biol ; 407(2): 193-221, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21241707

ABSTRACT

Traditional DNA transduction routes used for the modification of cellular genomes are subject to unpredictable alterations, as the cell-intrinsic repair machinery may affect both the integrity of the transgene and the recipient locus. These problems are overcome by recombinase-mediated cassette exchange (RMCE) approaches enabling predictable expression patterns by the nondisruptive insertion of a gene cassette at a pre-characterized genomic locus. The destination is marked by a "tag" consisting of two heterospecific recombination target sites (RTs) at the flanks of a selection marker. Provided on a circular donor vector, an analogous cassette encoding the gene of interest can cleanly replace the resident cassette under the influence of a site-specific recombinase. RMCE was first based on the yeast integrase Flp but had to give way to the originally more active phage-derived Cre enzyme. To be effective, both Tyr-recombinases have to be applied at a considerable concentration, which, in the case of Cre, triggers endonucleolytic activities and therefore cellular toxicity. This review addresses the particularities of both recombination routes depending on the structure of the synaptic complex and on improved integrase and RT variants. While the performance of Flp-RMCE can now firmly rely on optimized Flp variants and multiple sets of functional target sites (FRTs), the Cre system suffers from the promiscuity of its RT mutants, which is explained in molecular terms. At present, RMCE enters applications in the stem cell field. Remarkable efforts are noted in the framework of various mouse mutagenesis programs, which, in their first phase, have targeted virtually all genes and now start to shift their emphasis from gene trapping to gene modification.


Subject(s)
DNA Nucleotidyltransferases/genetics , Gene Transfer Techniques , Genetic Engineering/methods , Animals , Gene Targeting , Mice , Transgenes
17.
J Mol Biol ; 402(1): 52-69, 2010 Sep 10.
Article in English | MEDLINE | ID: mdl-20650281

ABSTRACT

There are strong indications, but as yet no proof, that extended 48-bp Flp recombinase targets (FRTs) represent unique targets in all eukaryotic genomes investigated, and that recombinase-mediated cassette exchange is not hampered by the occurrence of genomic pseudo sites. This encouraged the present study in which we explore the feasibility of exchanging, in a given cell, two distinct genomically anchored cassettes, each flanked by a unique set of two heterospecific FRT sites. Mutant FRTs have to meet two major prerequisites for successful recombinase-mediated cassette exchange: (i) a self-recognition capacity comparable to a pair of FRT wild-type sites (FRTxFRT), and (ii) a negligible cross-interaction if part of a set of heterospecific sites (F'xF). We apply a two-step strategy to explore various newly created FRT spacer mutants for these properties. As a result of our screening steps, we identify combinations of sites that are successfully applied to parallel Flp-mediated genomic targeting ("multiplexing") reactions (i.e., the simultaneous exchange of two separate target cassettes in a given cell).


Subject(s)
DNA Nucleotidyltransferases/genetics , Gene Targeting , Phosphotransferases (Alcohol Group Acceptor)/genetics , Recombinant Fusion Proteins/genetics , Recombination, Genetic , Thymidine Kinase/genetics , DNA Nucleotidyltransferases/chemistry , DNA Nucleotidyltransferases/metabolism , Humans , Mutation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...