Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.103
Filter
1.
Mol Cell ; 77(5): 1066-1079.e9, 2020 03 05.
Article in English | MEDLINE | ID: mdl-31902667

ABSTRACT

Naturally occurring or drug-induced DNA-protein crosslinks (DPCs) interfere with key DNA transactions if not repaired in a timely manner. The unique family of DPC-specific proteases Wss1/SPRTN targets DPC protein moieties for degradation, including stabilized topoisomerase-1 cleavage complexes (Top1ccs). Here, we describe that the efficient DPC disassembly requires Ddi1, another conserved predicted protease in Saccharomyces cerevisiae. We found Ddi1 in a genetic screen of the tdp1 wss1 mutant defective in Top1cc processing. Ddi1 is recruited to a persistent Top1cc-like DPC lesion in an S phase-dependent manner to assist in the eviction of crosslinked protein from DNA. Loss of Ddi1 or its putative protease activity hypersensitizes cells to DPC trapping agents independently from Wss1 and 26S proteasome, implying its broader role in DPC repair. Among the potential Ddi1 targets, we found the core component of Pol II and show that its genotoxin-induced degradation is impaired in ddi1. We propose that the Ddi1 protease contributes to DPC proteolysis.


Subject(s)
DNA Damage , DNA Repair , DNA, Fungal/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Animals , DNA Nucleotidyltransferases/genetics , DNA Nucleotidyltransferases/metabolism , DNA Topoisomerases, Type I/genetics , DNA Topoisomerases, Type I/metabolism , DNA, Fungal/genetics , Gene Expression Regulation, Fungal , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , Proteolysis , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Sf9 Cells , Spodoptera , Transcription, Genetic
2.
J Virol ; 98(4): e0024224, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38446633

ABSTRACT

Viral genomes frequently harbor overlapping genes, complicating the development of virus-vectored vaccines and gene therapies. This study introduces a novel conditional splicing system to precisely control the expression of such overlapping genes through recombinase-mediated conditional splicing. We refined site-specific recombinase (SSR) conditional splicing systems and explored their mechanisms. The systems demonstrated exceptional inducibility (116,700-fold increase) with negligible background expression, facilitating the conditional expression of overlapping genes in adenovirus-associated virus (AAV) and human immunodeficiency virus type 1. Notably, this approach enabled the establishment of stable AAV producer cell lines, encapsulating all necessary packaging genes. Our findings underscore the potential of the SSR-conditional splicing system to significantly advance vector engineering, enhancing the efficacy and scalability of viral-vector-based therapies and vaccines. IMPORTANCE: Regulating overlapping genes is vital for gene therapy and vaccine development using viral vectors. The regulation of overlapping genes presents challenges, including cytotoxicity and impacts on vector capacity and genome stability, which restrict stable packaging cell line development and broad application. To address these challenges, we present a "loxp-splice-loxp"-based conditional splicing system, offering a novel solution for conditional expression of overlapping genes and stable cell line establishment. This system may also regulate other cytotoxic genes, representing a significant advancement in cell engineering and gene therapy as well as biomass production.


Subject(s)
Dependovirus , Genes, Overlapping , Genes, Viral , Genetic Engineering , HIV-1 , RNA Splicing , Humans , Cell Line , Dependovirus/genetics , DNA Nucleotidyltransferases/genetics , DNA Nucleotidyltransferases/metabolism , Gene Expression Regulation, Viral , Genes, Overlapping/genetics , Genes, Viral/genetics , Genetic Engineering/methods , Genetic Therapy/methods , Genetic Vectors/genetics , HIV-1/genetics , RNA Splicing/genetics , Vaccines/biosynthesis , Vaccines/genetics , Viral Genome Packaging/genetics
3.
Nucleic Acids Res ; 51(13): 6857-6869, 2023 07 21.
Article in English | MEDLINE | ID: mdl-37264907

ABSTRACT

Bacterial conjugation is the main mechanism for the dissemination of antibiotic resistance genes. A single DNA strand of the conjugative plasmid is transferred across bacterial membranes covalently bound to a large multi-domain protein, named relaxase, which must be unfolded to traverse the secretion channel. Two tyrosine residues of the relaxase (Y18 and Y26 in relaxase TrwC) play an important role in the processing of conjugative DNA. We have used nanopore technology to uncover the unfolding states that take place during translocation of the relaxase-DNA complex. We observed that the relaxase unfolding pathway depends on the tyrosine residue involved in conjugative DNA binding. Transfer of the nucleoprotein complex is faster when DNA is bound to residue Y18. This is the first time in which a protein-DNA complex that is naturally translocated through bacterial membranes has been analyzed by nanopore sensing, opening new horizons to apply this technology to study protein secretion.


Subject(s)
Conjugation, Genetic , DNA Nucleotidyltransferases , Nanopores , Bacterial Proteins/metabolism , DNA Nucleotidyltransferases/metabolism , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , Plasmids/genetics , Tyrosine/metabolism
4.
Proc Natl Acad Sci U S A ; 119(3)2022 01 18.
Article in English | MEDLINE | ID: mdl-35027456

ABSTRACT

Conditional gene expression is a powerful tool for genetic analysis of biological phenomena. In the widely used "lox-stop-lox" approach, insertion of a stop cassette consisting of a series of stop codons and polyadenylation signals flanked by lox sites into the 5' untranslated region (UTR) of a gene prevents expression until the cassette is excised by tissue-specific expression of Cre recombinase. Although lox-stop-lox and similar approaches using other site-specific recombinases have been successfully used in many experimental systems, this design has certain limitations. Here, we describe the Floxed exon (Flexon) approach, which uses a stop cassette composed of an artificial exon flanked by artificial introns, designed to cause premature termination of translation and nonsense-mediated decay of the mRNA and allowing for flexible placement into a gene. We demonstrate its efficacy in Caenorhabditis elegans by showing that, when promoters that cause weak and/or transient cell-specific expression are used to drive Cre in combination with a gfp(flexon) transgene, strong and sustained expression of green fluorescent protein (GFP) is obtained in specific lineages. We also demonstrate its efficacy in an endogenous gene context: we inserted a flexon into the Argonaute gene rde-1 to abrogate RNA interference (RNAi), and restored RNAi tissue specifically by expression of Cre. Finally, we describe several potential additional applications of the Flexon approach, including more precise control of gene expression using intersectional methods, tissue-specific protein degradation, and generation of genetic mosaics. The Flexon approach should be feasible in any system where a site-specific recombination-based method may be applied.


Subject(s)
Exons , Gene Expression , Recombinases/metabolism , Animals , Caenorhabditis elegans/metabolism , DNA Nucleotidyltransferases , Green Fluorescent Proteins/metabolism , Integrases , Promoter Regions, Genetic , RNA Interference , Recombination, Genetic , Transgenes
5.
Nucleic Acids Res ; 50(2): 1174-1186, 2022 01 25.
Article in English | MEDLINE | ID: mdl-34951450

ABSTRACT

Tyrosine site-specific recombinases (SSRs) represent a versatile genome editing tool with considerable therapeutic potential. Recent developments to engineer and evolve SSRs into heterotetramers to improve target site flexibility signified a critical step towards their broad utility in genome editing. However, SSR monomers can form combinations of different homo- and heterotetramers in cells, increasing their off-target potential. Here, we discover that two paired mutations targeting residues implicated in catalysis lead to simple obligate tyrosine SSR systems, where the presence of all distinct subunits to bind as a heterotetramer is obligatory for catalysis. Therefore, only when the paired mutations are applied as single mutations on each recombinase subunit, the engineered SSRs can efficiently recombine the intended target sequence, while the subunits carrying the point mutations expressed in isolation are inactive. We demonstrate the utility of the obligate SSR system to improve recombination specificity of a designer-recombinase for a therapeutic target in human cells. Furthermore, we show that the mutations render the naturally occurring SSRs, Cre and Vika, obligately heteromeric for catalytic proficiency, providing a straight-forward approach to improve their applied properties. These results facilitate the development of safe and effective therapeutic designer-recombinases and advance our mechanistic understanding of SSR catalysis.


Subject(s)
DNA Nucleotidyltransferases/metabolism , Gene Editing , Genetic Engineering/methods , Recombination, Genetic , HEK293 Cells , Humans
6.
Nucleic Acids Res ; 50(14): 8127-8142, 2022 08 12.
Article in English | MEDLINE | ID: mdl-35849337

ABSTRACT

Integrative and conjugative elements (ICEs) are important drivers of horizontal gene transfer in prokaryotes. They are responsible for antimicrobial resistance spread, a major current health concern. ICEs are initially processed by relaxases that recognize the binding site of oriT sequence and nick at a conserved nic site. The ICESt3/Tn916/ICEBs1 superfamily, which is widespread among Firmicutes, encodes uncanonical relaxases belonging to a recently identified family called MOBT. This family is related to the rolling circle replication initiators of the Rep_trans family. The nic site of these MOBT relaxases is conserved but their DNA binding site is still unknown. Here, we identified the bind site of RelSt3, the MOBT relaxase from ICESt3. Unexpectedly, we found this bind site distantly located from the nic site. We revealed that the binding of the RelSt3 N-terminal HTH domain is required for efficient nicking activity. We also deciphered the role of RelSt3 in the initial and final stages of DNA processing during conjugation. Especially, we demonstrated a strand transfer activity, and the formation of covalent DNA-relaxase intermediate for a MOBT relaxase.


Subject(s)
Bacterial Proteins , Conjugation, Genetic , DNA Nucleotidyltransferases , Gram-Positive Bacteria , Interspersed Repetitive Sequences , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , DNA Nucleotidyltransferases/genetics , DNA Nucleotidyltransferases/metabolism , DNA, Bacterial/genetics , Gene Transfer, Horizontal , Gram-Positive Bacteria/genetics , Plasmids/genetics
7.
PLoS Genet ; 17(3): e1009433, 2021 03.
Article in English | MEDLINE | ID: mdl-33705385

ABSTRACT

Transcriptional rewiring is the regulation of different target genes by orthologous regulators in different organisms. While this phenomenon has been observed, it has not been extensively studied, particularly in core regulatory systems. Several global cell cycle regulators are conserved in the Alphaproteobacteria, providing an excellent model to study this phenomenon. First characterized in Caulobacter crescentus, GcrA and CcrM compose a DNA methylation-based regulatory system that helps coordinate the complex life cycle of this organism. These regulators are well-conserved across Alphaproteobacteria, but the extent to which their regulatory targets are conserved is not known. In this study, the regulatory targets of GcrA and CcrM were analyzed by SMRT-seq, RNA-seq, and ChIP-seq technologies in the Alphaproteobacterium Brevundimonas subvibrioides, and then compared to those of its close relative C. crescentus that inhabits the same environment. Although the regulators themselves are highly conserved, the genes they regulate are vastly different. GcrA directly regulates 204 genes in C. crescentus, and though B. subvibrioides has orthologs to 147 of those genes, only 48 genes retained GcrA binding in their promoter regions. Additionally, only 12 of those 48 genes demonstrated significant transcriptional change in a gcrA mutant, suggesting extensive transcriptional rewiring between these organisms. Similarly, out of hundreds of genes CcrM regulates in each of these organisms, only 2 genes were found in common. When multiple Alphaproteobacterial genomes were analyzed bioinformatically for potential GcrA regulatory targets, the regulation of genes involved in DNA replication and cell division was well conserved across the Caulobacterales but not outside this order. This work suggests that significant transcriptional rewiring can occur in cell cycle regulatory systems even over short evolutionary distances.


Subject(s)
Bacterial Proteins/metabolism , DNA Nucleotidyltransferases/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Bacterial , Transcription, Genetic , Bacterial Proteins/genetics , Caulobacteraceae/genetics , Caulobacteraceae/metabolism , DNA Methylation , DNA Nucleotidyltransferases/genetics , Gene Expression Profiling/methods
8.
J Biol Chem ; 296: 100509, 2021.
Article in English | MEDLINE | ID: mdl-33676891

ABSTRACT

Site-specific recombinases (SSRs) are invaluable genome engineering tools that have enormously boosted our understanding of gene functions and cell lineage relationships in developmental biology, stem cell biology, regenerative medicine, and multiple diseases. However, the ever-increasing complexity of biomedical research requires the development of novel site-specific genetic recombination technologies that can manipulate genomic DNA with high efficiency and fine spatiotemporal control. Here, we review the latest innovative strategies of the commonly used Cre-loxP recombination system and its combinatorial strategies with other site-specific recombinase systems. We also highlight recent progress with a focus on the new generation of chemical- and light-inducible genetic systems and discuss the merits and limitations of each new and established system. Finally, we provide the future perspectives of combining various recombination systems or improving well-established site-specific genetic tools to achieve more efficient and precise spatiotemporal genetic manipulation.


Subject(s)
DNA Nucleotidyltransferases/metabolism , Recombination, Genetic , CRISPR-Cas Systems , Catalysis , Enzyme Activation , Integrases/metabolism , Light , Plants/enzymology , Promoter Regions, Genetic
9.
Mol Microbiol ; 115(6): 1110-1121, 2021 06.
Article in English | MEDLINE | ID: mdl-33244797

ABSTRACT

In Bacillus subtilis, a sporulation-related gene, spsM, is disrupted by SPß prophage, but reconstituted during sporulation through SPß excision. The spsM reconstitution is catalyzed by a site-specific DNA recombinase, SprA, and its cognate recombination directionality factor, SprB. SprB interacts with SprA, directing the SprA-mediated recombination reaction from integration to excision; however, the details of the directionality control remains unclear. Here, we demonstrate the importance of the extreme C-terminal region (ECT) of SprA in the DNA recombination and directionality control. We created a series of SprA C-terminal deletants and examined their DNA-binding and recombination activities. Deletions in the ECT caused a loss of integration and excision activity, the magnitudes of which positively correlated with the deletion size. Gel shift study revealed that the loss of the integration activity was attributable to the failure of synaptic complex formation. The excision deficiency was caused by defective interaction with SprB. Moreover, alanine scanning analysis revealed that Phe532 is essential to interact with SprB. SprAF532A , therefore, showed almost no excision activity, while retaining the integration activity. Collectively, these results suggest that the ECT plays the crucial roles in the interaction of SprA with SprB and possibly in the directional control of the recombination.


Subject(s)
Bacillus Phages/genetics , Bacillus subtilis/genetics , DNA Nucleotidyltransferases/metabolism , Integrases/metabolism , Recombination, Genetic/genetics , Bacillus subtilis/enzymology , Bacillus subtilis/metabolism , DNA, Bacterial/genetics , DNA-Binding Proteins/genetics , Gene Deletion , Integrases/genetics , Lysogeny/genetics
10.
Development ; 146(21)2019 11 04.
Article in English | MEDLINE | ID: mdl-31597657

ABSTRACT

Genetic lineage-tracing techniques are powerful tools for studying specific cell populations in development and pathogenesis. Previous techniques have mainly involved systems for tracing a single gene, which are limited in their ability to facilitate direct comparisons of the contributions of different cell lineages. We have developed a new combinatorial system for tracing all three germ layers using self-cleaving 2A peptides and multiple site-specific recombinases (SSRs). In the resulting TRiCK (TRiple Coloured germ layer Knock-in) mice, the three germ layers are conditionally and simultaneously labelled with distinct fluorescent proteins via embryogenesis. We show that previously reported ectopic expressions of lineage markers are the outcome of secondary gene expression. The results presented here also indicate that the commitment of caudal axial stem cells to neural or mesodermal fate proceeds without lineage fluctuations, contrary to the notion of their bi-potency. Moreover, we developed IMES, an optimized tissue clearing method that is highly compatible with a variety of fluorescent proteins and immunostaining, and the combined use of TRiCK mice and IMES can facilitate comprehensive analyses of dynamic contributions of all three germ layers.


Subject(s)
Cell Lineage , Gene Expression Regulation, Developmental , Gene Knock-In Techniques , Germ Layers/cytology , Animals , Brain/metabolism , Crosses, Genetic , DNA Nucleotidyltransferases/metabolism , Embryonic Stem Cells/cytology , Endoderm/cytology , Endothelium, Vascular/cytology , Female , Genotype , Green Fluorescent Proteins/metabolism , HEK293 Cells , Heart/embryology , Humans , Imaging, Three-Dimensional , Liver/embryology , Male , Mesoderm/cytology , Mice , Mice, Inbred C57BL , Myocardium/cytology , Neural Plate/cytology
11.
Nat Chem Biol ; 16(5): 577-586, 2020 05.
Article in English | MEDLINE | ID: mdl-32094923

ABSTRACT

Receptor tyrosine kinases (RTKs) are transmembrane receptors of great clinical interest due to their role in disease. Historically, therapeutics targeting RTKs have been identified using in vitro kinase assays. Due to frequent development of drug resistance, however, there is a need to identify more diverse compounds that inhibit mutated but not wild-type RTKs. Here, we describe MaMTH-DS (mammalian membrane two-hybrid drug screening), a live-cell platform for high-throughput identification of small molecules targeting functional protein-protein interactions of RTKs. We applied MaMTH-DS to an oncogenic epidermal growth factor receptor (EGFR) mutant resistant to the latest generation of clinically approved tyrosine kinase inhibitors (TKIs). We identified four mutant-specific compounds, including two that would not have been detected by conventional in vitro kinase assays. One of these targets mutant EGFR via a new mechanism of action, distinct from classical TKI inhibition. Our results demonstrate how MaMTH-DS is a powerful complement to traditional drug screening approaches.


Subject(s)
High-Throughput Screening Assays/methods , Protein Kinase Inhibitors/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line , Cell Line, Tumor , DNA Nucleotidyltransferases/genetics , Drug Discovery , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Genes, Reporter , Humans , Luciferases/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mutation , Phosphorylation/drug effects , Reproducibility of Results , Small Molecule Libraries/pharmacology , Staurosporine/analogs & derivatives , Staurosporine/pharmacology
12.
Appl Microbiol Biotechnol ; 106(13-16): 5167-5178, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35851417

ABSTRACT

Glaesserella parasuis is an important bacterial pathogen that affects the swine industry worldwide. Research on the pathogenic mechanism and genetically engineered vaccine remains undeveloped because an effective markerless and multiple-gene knockout system is unavailable for G. parasuis yet. To establish a markerless knockout, deleted allelic genes with kanamycin resistance (KanR) cassettes were introduced into the genome of G. parasuis by using natural transformation with suicide plasmids. Then, the KanR cassette was excised with a thermosensitive plasmid pGF conferring a constitutive Flp expression. To realize the markerless and multiple-gene knockout, plasmid pGAF was constructed by placing the Flp gene under the control of an arabinose-inducible promoter. Firstly, pGAF was introduced into G. parasuis by electroporation, and the marked mutants were produced following natural transformation. Finally, the KanR cassette was excised from the genome by the inducible expression of Flp upon arabinose action. Based on the natural transformation and the inducible expression of Flp, the markerless single-gene knockout mutants of ΔhsdR, ΔneuA2, ΔespP2, Δapd, and ΔnanH were constructed. In addition, a five-gene knockout mutant of ΔhsdRΔneuA2ΔespP2ΔapdΔnanH was generated by successive natural transformation with five suicide plasmids. Taken together, a markerless and multiple-gene deletion system was established for G. parasuis in the present study for the first time. This system is simple, efficient, and easy to manipulate for G. parasuis; thus, our technique will substantially aid the understanding of the etiology, pathogenesis, and genetic engineering of G. parasuis and other bacteria that can be naturally transformed in laboratory conditions. KEY POINTS: • Flp recombinase excised the KanR gene flanked by FRT sites in Glaesserella parasuis. • The regulatory expression of Flp enabled a multiple-gene knockout forG. parasuis. • The technique will promote the understanding of Glässer's disease pathogens.


Subject(s)
Arabinose , Haemophilus parasuis , Animals , DNA Nucleotidyltransferases/genetics , Gene Knockout Techniques , Haemophilus parasuis/genetics , Haemophilus parasuis/metabolism , Humans , Swine
13.
Nucleic Acids Res ; 48(22): 12804-12816, 2020 12 16.
Article in English | MEDLINE | ID: mdl-33270859

ABSTRACT

HK022 coliphage site-specific recombinase Integrase (Int) can catalyze integrative site-specific recombination and recombinase-mediated cassette exchange (RMCE) reactions in mammalian cell cultures. Owing to the promiscuity of the 7 bp overlap sequence in its att sites, active 'attB' sites flanking human deleterious mutations were previously identified that may serve as substrates for RMCE reactions for future potential gene therapy. However, the wild type Int proved inefficient in catalyzing such RMCE reactions. To address this low efficiency, variants of Int were constructed and examined by integrative site-specific recombination and RMCE assays in human cells using native 'attB' sites. As a proof of concept, various Int derivatives have demonstrated successful RMCE reactions using a pair of native 'attB' sites that were inserted as a substrate into the human genome. Moreover, successful RMCE reactions were demonstrated in native locations of the human CTNS and DMD genes whose mutations are responsible for Cystinosis and Duchene Muscular Dystrophy diseases, respectively. This work provides a steppingstone for potential downstream therapeutic applications.


Subject(s)
Bacteriophage HK022/genetics , Genetic Therapy , Integrases/genetics , Recombination, Genetic/genetics , Bacteriophage HK022/enzymology , DNA Nucleotidyltransferases/genetics , Genome, Human/genetics , Humans
14.
Nucleic Acids Res ; 48(1): 472-485, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31745551

ABSTRACT

Site-specific recombinases (SSRs) such as the Cre/loxP system are useful genome engineering tools that can be repurposed by altering their DNA-binding specificity. However, SSRs that delete a natural sequence from the human genome have not been reported thus far. Here, we describe the generation of an SSR system that precisely excises a 1.4 kb fragment from the human genome. Through a streamlined process of substrate-linked directed evolution we generated two separate recombinases that, when expressed together, act as a heterodimer to delete a human genomic sequence from chromosome 7. Our data indicates that designer-recombinases can be generated in a manageable timeframe for precision genome editing. A large-scale bioinformatics analysis suggests that around 13% of all human protein-coding genes could be targetable by dual designer-recombinase induced genomic deletion (dDRiGD). We propose that heterospecific designer-recombinases, which work independently of the host DNA repair machinery, represent an efficient and safe alternative to nuclease-based genome editing technologies.


Subject(s)
Base Sequence , Chromosomes, Human, Pair 7/chemistry , DNA Nucleotidyltransferases/genetics , Gene Editing/methods , Genome, Human , Sequence Deletion , Chromosomes, Human, Pair 7/metabolism , Cloning, Molecular , Computational Biology/methods , DNA Nucleotidyltransferases/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Loci , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
15.
Nucleic Acids Res ; 48(16): 8914-8926, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32789491

ABSTRACT

The Apl protein of bacteriophage 186 functions both as an excisionase and as a transcriptional regulator; binding to the phage attachment site (att), and also between the major early phage promoters (pR-pL). Like other recombination directionality factors (RDFs), Apl binding sites are direct repeats spaced one DNA helix turn apart. Here, we use in vitro binding studies with purified Apl and pR-pL DNA to show that Apl binds to multiple sites with high cooperativity, bends the DNA and spreads from specific binding sites into adjacent non-specific DNA; features that are shared with other RDFs. By analysing Apl's repression of pR and pL, and the effect of operator mutants in vivo with a simple mathematical model, we were able to extract estimates of binding energies for single specific and non-specific sites and for Apl cooperativity, revealing that Apl monomers bind to DNA with low sequence specificity but with strong cooperativity between immediate neighbours. This model fit was then independently validated with in vitro data. The model we employed here is a simple but powerful tool that enabled better understanding of the balance between binding affinity and cooperativity required for RDF function. A modelling approach such as this is broadly applicable to other systems.


Subject(s)
Attachment Sites, Microbiological , DNA Nucleotidyltransferases/metabolism , DNA, Viral/genetics , Lysogeny , Myoviridae/metabolism , Viral Proteins/metabolism , Binding Sites , DNA-Binding Proteins/metabolism , Promoter Regions, Genetic , Protein Binding , Recombination, Genetic
16.
Nucleic Acids Res ; 48(11): 6198-6209, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32379323

ABSTRACT

Group II introns are self-splicing ribozymes and mobile genetic elements. Splicing is required for both expression of the interrupted host gene and intron retromobility. For the pRS01 plasmid-encoded Lactococcus lactis group II intron, Ll.LtrB, splicing enables expression of the intron's host relaxase protein. Relaxase, in turn, initiates horizontal transfer of the conjugative pRS01 plasmid and stimulates retrotransposition of the intron. Little is known about how splicing of bacterial group II introns is influenced by environmental conditions. Here, we show that low temperatures can inhibit Ll.LtrB intron splicing. Whereas autocatalysis is abolished in the cold, splicing is partially restored by the intron-encoded protein (IEP). Structure profiling reveals cold-induced disruptions of key tertiary interactions, suggesting that a kinetic trap prevents the intron RNA from assuming its native state. Interestingly, while reduced levels of transcription and splicing lead to a paucity of excised intron in the cold, levels of relaxase mRNA are maintained, partially due to diminished intron-mediated mRNA targeting, allowing intron spread by conjugal transfer. Taken together, this study demonstrates not only the intrinsic cold sensitivity of group II intron splicing and the role of the IEP for cold-stress adaptation, but also maintenance of horizontal plasmid and intron transfer under cold-shock.


Subject(s)
Cold Temperature , Conjugation, Genetic , Introns/genetics , Lactococcus lactis/genetics , RNA Splicing , Base Sequence , Cold-Shock Response , DNA Nucleotidyltransferases/metabolism , Evolution, Molecular , Gene Transfer, Horizontal , Plasmids/genetics , Plasmids/metabolism , RNA, Catalytic/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retroelements
17.
PLoS Genet ; 15(6): e1008193, 2019 06.
Article in English | MEDLINE | ID: mdl-31242181

ABSTRACT

Mechanisms for highly efficient chromosome-associated equal segregation, and for maintenance of steady state copy number, are at the heart of the evolutionary success of the 2-micron plasmid as a stable multi-copy extra-chromosomal selfish DNA element present in the yeast nucleus. The Flp site-specific recombination system housed by the plasmid, which is central to plasmid copy number maintenance, is regulated at multiple levels. Transcription of the FLP gene is fine-tuned by the repressor function of the plasmid-coded partitioning proteins Rep1 and Rep2 and their antagonist Raf1, which is also plasmid-coded. In addition, the Flp protein is regulated by the host's post-translational modification machinery. Utilizing a Flp-SUMO fusion protein, which functionally mimics naturally sumoylated Flp, we demonstrate that the modification signals ubiquitination of Flp, followed by its proteasome-mediated degradation. Furthermore, reduced binding affinity and cooperativity of the modified Flp decrease its association with the plasmid FRT (Flp recombination target) sites, and/or increase its dissociation from them. The resulting attenuation of strand cleavage and recombination events safeguards against runaway increase in plasmid copy number, which is deleterious to the host-and indirectly-to the plasmid. These results have broader relevance to potential mechanisms by which selfish genomes minimize fitness conflicts with host genomes by holding in check the extra genetic load they pose.


Subject(s)
DNA Nucleotidyltransferases/genetics , Repetitive Sequences, Nucleic Acid/genetics , SUMO-1 Protein/genetics , Transcription, Genetic , Chromosome Segregation/genetics , DNA Copy Number Variations/genetics , DNA Replication/genetics , Genome, Fungal/genetics , Intracellular Signaling Peptides and Proteins/genetics , Protein Binding/genetics , Protein Processing, Post-Translational/genetics , Proto-Oncogene Proteins c-raf/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Sumoylation/genetics , Trans-Activators/genetics
18.
J Neurosci ; 40(37): 7169-7186, 2020 09 09.
Article in English | MEDLINE | ID: mdl-32801153

ABSTRACT

Conditional gene inactivation and restoration are powerful tools for studying gene functions in the nervous system and for modeling neuropsychiatric diseases. The combination of the two is necessary to interrogate specific cell types within defined developmental stages. However, very few methods and animal models have been developed for such purpose. Here we present a versatile method for conditional gene inactivation and in situ restoration through reversibly inverting a critical part of its endogenous genomic sequence by Cre- and Flp-mediated recombinations. Using this method, we generated a mouse model to manipulate Mecp2, an X-linked dosage-sensitive gene whose mutations cause Rett syndrome. Combined with multiple Cre- and Flp-expressing drivers and viral tools, we achieved efficient and reliable Mecp2 inactivation and restoration in the germline and several neuronal cell types, and demonstrated phenotypic reversal and prevention on cellular and behavioral levels in male mice. This study not only provides valuable tools and critical insights for Mecp2 and Rett syndrome, but also offers a generally applicable strategy to decipher other neurologic disorders.SIGNIFICANCE STATEMENT Studying neurodevelopment and modeling neurologic disorders rely on genetic tools, such as conditional gene regulation. We developed a new method to combine conditional gene inactivation and restoration on a single allele without disturbing endogenous expression pattern or dosage. We applied it to manipulate Mecp2, a gene residing on X chromosome whose malfunction leads to neurologic disease, including Rett syndrome. Our results demonstrated the efficiency, specificity, and versatility of this new method, provided valuable tools and critical insights for Mecp2 function and Rett syndrome research, and offered a generally applicable strategy to investigate other genes and genetic disorders.


Subject(s)
Gene Targeting/methods , Methyl-CpG-Binding Protein 2/metabolism , Phenotype , Rett Syndrome/genetics , Animals , DNA Nucleotidyltransferases/genetics , DNA Nucleotidyltransferases/metabolism , Germ-Line Mutation , Integrases/genetics , Integrases/metabolism , Male , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Inbred C57BL , Movement , Neurons/metabolism , Neurons/physiology , Rett Syndrome/pathology
19.
Dev Biol ; 457(1): 9-12, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31550481

ABSTRACT

We describe a new methodology for genetically labeling single cell lineages in Drosophila called DMARCM. The system offers ultra-low frequency labeling, linear induction, consistent labeling among individuals and virtually no background signal. We compare this technique to an existing approach, which has been widely adopted. We demonstrate how application of DMARCM in the gastrointestinal epithelium permits the effects of labeling frequency on tumorigenic stem cell growth to be distinguished in an established tumor model.


Subject(s)
Cell Lineage , Cytological Techniques/methods , Drosophila/cytology , Animals , DNA Nucleotidyltransferases , Drosophila/growth & development , Gastrointestinal Tract/cytology , Heat-Shock Response , Microscopy, Confocal , Staining and Labeling/methods
20.
J Biol Chem ; 295(19): 6413-6424, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32213599

ABSTRACT

Site-specific recombinases, such as Cre, are a widely used tool for genetic lineage tracing in the fields of developmental biology, neural science, stem cell biology, and regenerative medicine. However, nonspecific cell labeling by some genetic Cre tools remains a technical limitation of this recombination system, which has resulted in data misinterpretation and led to many controversies in the scientific community. In the past decade, to enhance the specificity and precision of genetic targeting, researchers have used two or more orthogonal recombinases simultaneously for labeling cell lineages. Here, we review the history of cell-tracing strategies and then elaborate on the working principle and application of a recently developed dual genetic lineage-tracing approach for cell fate studies. We place an emphasis on discussing the technical strengths and caveats of different methods, with the goal to develop more specific and efficient tracing technologies for cell fate mapping. Our review also provides several examples for how to use different types of DNA recombinase-mediated lineage-tracing strategies to improve the resolution of the cell fate mapping in order to probe and explore cell fate-related biological phenomena in the life sciences.


Subject(s)
Cell Tracking , DNA Nucleotidyltransferases , Recombination, Genetic , Stem Cells/metabolism , Animals , DNA Nucleotidyltransferases/genetics , DNA Nucleotidyltransferases/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL