Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.835
Filter
Add more filters

Publication year range
1.
Cell ; 179(3): 772-786.e19, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31626774

ABSTRACT

Understanding neural circuits requires deciphering interactions among myriad cell types defined by spatial organization, connectivity, gene expression, and other properties. Resolving these cell types requires both single-neuron resolution and high throughput, a challenging combination with conventional methods. Here, we introduce barcoded anatomy resolved by sequencing (BARseq), a multiplexed method based on RNA barcoding for mapping projections of thousands of spatially resolved neurons in a single brain and relating those projections to other properties such as gene or Cre expression. Mapping the projections to 11 areas of 3,579 neurons in mouse auditory cortex using BARseq confirmed the laminar organization of the three top classes (intratelencephalic [IT], pyramidal tract-like [PT-like], and corticothalamic [CT]) of projection neurons. In depth analysis uncovered a projection type restricted almost exclusively to transcriptionally defined subtypes of IT neurons. By bridging anatomical and transcriptomic approaches at cellular resolution with high throughput, BARseq can potentially uncover the organizing principles underlying the structure and formation of neural circuits.


Subject(s)
Auditory Cortex/metabolism , Nerve Net/metabolism , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Animals , Brain Mapping , Humans , Integrases/genetics , Mice , Neurites/metabolism , Pyramidal Cells/metabolism , Pyramidal Tracts/metabolism
2.
Immunity ; 56(5): 1027-1045.e8, 2023 05 09.
Article in English | MEDLINE | ID: mdl-36791722

ABSTRACT

Genetic tools to target microglia specifically and efficiently from the early stages of embryonic development are lacking. We generated a constitutive Cre line controlled by the microglia signature gene Crybb1 that produced nearly complete recombination in embryonic brain macrophages (microglia and border-associated macrophages [BAMs]) by the perinatal period, with limited recombination in peripheral myeloid cells. Using this tool in combination with Flt3-Cre lineage tracer, single-cell RNA-sequencing analysis, and confocal imaging, we resolved embryonic-derived versus monocyte-derived BAMs in the mouse cortex. Deletion of the transcription factor SMAD4 in microglia and embryonic-derived BAMs using Crybb1-Cre caused a developmental arrest of microglia, which instead acquired a BAM specification signature. By contrast, the development of genuine BAMs remained unaffected. Our results reveal that SMAD4 drives a transcriptional and epigenetic program that is indispensable for the commitment of brain macrophages to the microglia fate and highlight Crybb1-Cre as a tool for targeting embryonic brain macrophages.


Subject(s)
Macrophages , Microglia , Mice , Animals , Microglia/metabolism , Macrophages/metabolism , Integrases/genetics , Integrases/metabolism , Brain/metabolism
3.
Immunity ; 54(10): 2209-2217.e6, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34551314

ABSTRACT

CD4+ T cells share common developmental pathways with CD8+ T cells, and upon maturation, CD4+ T conventional T (Tconv) cells lack phenotypic markers that distinguish these cells from FoxP3+ T regulatory cells. We developed a tamoxifen-inducible ThPOKCreERT2.hCD2 line with Frt sites inserted on either side of the CreERT2-hCD2 cassette, and a Foxp3Ametrine-FlpO strain, expressing Ametrine and FlpO in Foxp3+ cells. Breeding these mice resulted in a CD4conviCreERT2-hCD2 line that allows for the specific manipulation of a gene in CD4+ Tconv cells. As FlpO removes the CreERT2-hCD2 cassette, CD4+ Treg cells are spared from Cre activity, which we refer to as allele conditioning. Comparison with an E8IiCreERT2.GFP mouse that enables inducible targeting of CD8+ T cells, and deletion of two inhibitory receptors, PD-1 and LAG-3, in a melanoma model, support the fidelity of these lines. These engineered mouse strains present a resource for the temporal manipulation of genes in CD4+ T cells and CD4+ Tconv cells.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Cell Lineage/immunology , Gene Editing/methods , Integrases/genetics , Alleles , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Line , Mice
4.
Mol Cell ; 81(4): 756-766.e8, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33472056

ABSTRACT

Bacillus subtilis structural maintenance of chromosomes (SMC) complexes are topologically loaded at centromeric sites adjacent to the replication origin by the partitioning protein ParB. These ring-shaped ATPases then translocate down the left and right chromosome arms while tethering them together. Here, we show that the site-specific recombinase XerD, which resolves chromosome dimers, is required to unload SMC tethers when they reach the terminus. We identify XerD-specific binding sites in the terminus region and show that they dictate the site of unloading in a manner that depends on XerD but not its catalytic residue, its partner protein XerC, or the recombination site dif. Finally, we provide evidence that ParB and XerD homologs perform similar functions in Staphylococcus aureus. Thus, two broadly conserved factors that act at the origin and terminus have second functions in loading and unloading SMC complexes that travel between them.


Subject(s)
Bacillus subtilis/enzymology , Bacterial Proteins/metabolism , Chromosomes, Bacterial/metabolism , Integrases/metabolism , Staphylococcus aureus/enzymology , Bacillus subtilis/genetics , Bacterial Proteins/genetics , Chromosomes, Bacterial/genetics , DNA Primase/genetics , DNA Primase/metabolism , Integrases/genetics , Staphylococcus aureus/genetics
5.
Mol Cell ; 73(4): 727-737.e3, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30709710

ABSTRACT

CRISPR-Cas immunity requires integration of short, foreign DNA fragments into the host genome at the CRISPR locus, a site consisting of alternating repeat sequences and foreign-derived spacers. In most CRISPR systems, the proteins Cas1 and Cas2 form the integration complex and are both essential for DNA acquisition. Most type V-C and V-D systems lack the cas2 gene and have unusually short CRISPR repeats and spacers. Here, we show that a mini-integrase comprising the type V-C Cas1 protein alone catalyzes DNA integration with a preference for short (17- to 19-base-pair) DNA fragments. The mini-integrase has weak specificity for the CRISPR array. We present evidence that the Cas1 proteins form a tetramer for integration. Our findings support a model of a minimal integrase with an internal ruler mechanism that favors shorter repeats and spacers. This minimal integrase may represent the function of the ancestral Cas1 prior to Cas2 adoption.


Subject(s)
CRISPR-Associated Proteins/genetics , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , DNA, Bacterial/genetics , Endodeoxyribonucleases/genetics , Endonucleases/genetics , Escherichia coli Proteins/genetics , Escherichia coli/genetics , Gene Editing/methods , Integrases/genetics , Base Pairing , CRISPR-Associated Proteins/metabolism , DNA, Bacterial/metabolism , Endodeoxyribonucleases/metabolism , Endonucleases/metabolism , Escherichia coli/enzymology , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial , Integrases/metabolism , Nucleotide Motifs , Substrate Specificity
6.
PLoS Genet ; 20(4): e1011231, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38578806

ABSTRACT

Integrons are adaptive devices that capture, stockpile, shuffle and express gene cassettes thereby sampling combinatorial phenotypic diversity. Some integrons called sedentary chromosomal integrons (SCIs) can be massive structures containing hundreds of cassettes. Since most of these cassettes are non-expressed, it is not clear how they remain stable over long evolutionary timescales. Recently, it was found that the experimental inversion of the SCI of Vibrio cholerae led to a dramatic increase of the cassette excision rate associated with a fitness defect. Here, we question the evolutionary sustainability of this apparently counter selected genetic context. Through experimental evolution, we find that the integrase is rapidly inactivated and that the inverted SCI can recover its original orientation by homologous recombination between two insertion sequences (ISs) present in the array. These two outcomes of SCI inversion restore the normal growth and prevent the loss of cassettes, enabling SCIs to retain their roles as reservoirs of functions. These results illustrate a nice interplay between gene orientation, genome rearrangement, bacterial fitness and demonstrate how integrons can benefit from their embedded ISs.


Subject(s)
Bacteria , Integrons , Integrons/genetics , Bacteria/genetics , DNA Transposable Elements , Integrases/genetics
7.
Development ; 150(3)2023 02 15.
Article in English | MEDLINE | ID: mdl-36734327

ABSTRACT

Tracing and manipulating cells in embryos are essential to understand development. Lipophilic dye microinjections, viral transfection and iontophoresis have been key to map the origin of the progenitor cells that form the different organs in the post-implantation mouse embryo. These techniques require advanced manipulation skills and only iontophoresis, a demanding approach of limited efficiency, has been used for single-cell labelling. Here, we perform lineage tracing and local gene ablation using cell-permeant Cre recombinase (TAT-Cre) microinjection. First, we map the fate of undifferentiated progenitors to the different heart chambers. Then, we achieve single-cell recombination by titrating the dose of TAT-Cre, which allows clonal analysis of nascent mesoderm progenitors. Finally, injecting TAT-Cre to Mycnflox/flox embryos in the primitive heart tube revealed that Mycn plays a cell-autonomous role in maintaining cardiomyocyte proliferation. This tool will help researchers identify the cell progenitors and gene networks involved in organ development, helping to understand the origin of congenital defects.


Subject(s)
Integrases , Stem Cells , Mice , Animals , Microinjections , Integrases/genetics , Gene Targeting
8.
Nucleic Acids Res ; 52(8): e43, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38587185

ABSTRACT

Microbiology and synthetic biology depend on reverse genetic approaches to manipulate bacterial genomes; however, existing methods require molecular biology to generate genomic homology, suffer from low efficiency, and are not easily scaled to high throughput. To overcome these limitations, we developed a system for creating kilobase-scale genomic modifications that uses DNA oligonucleotides to direct the integration of a non-replicating plasmid. This method, Oligonucleotide Recombineering followed by Bxb-1 Integrase Targeting (ORBIT) was pioneered in Mycobacteria, and here we adapt and expand it for Escherichia coli. Our redesigned plasmid toolkit for oligonucleotide recombineering achieved significantly higher efficiency than λ Red double-stranded DNA recombineering and enabled precise, stable knockouts (≤134 kb) and integrations (≤11 kb) of various sizes. Additionally, we constructed multi-mutants in a single transformation, using orthogonal attachment sites. At high throughput, we used pools of targeting oligonucleotides to knock out nearly all known transcription factor and small RNA genes, yielding accurate, genome-wide, single mutant libraries. By counting genomic barcodes, we also show ORBIT libraries can scale to thousands of unique members (>30k). This work demonstrates that ORBIT for E. coli is a flexible reverse genetic system that facilitates rapid construction of complex strains and readily scales to create sophisticated mutant libraries.


Subject(s)
Escherichia coli , Oligonucleotides , Plasmids , Escherichia coli/genetics , Oligonucleotides/genetics , Plasmids/genetics , Integrases/genetics , Integrases/metabolism , Genome, Bacterial/genetics , Genetic Engineering/methods , Gene Knockout Techniques , Transcription Factors/genetics , Transcription Factors/metabolism
9.
PLoS Genet ; 19(10): e1010998, 2023 10.
Article in English | MEDLINE | ID: mdl-37831715

ABSTRACT

Archaeal pleomorphic viruses belonging to the Pleolipoviridae family represent an enigmatic group as they exhibit unique genomic features and are thought to have evolved through recombination with different archaeal plasmids. However, most of our understanding of the diversity and evolutionary trajectories of this clade comes from a handful of isolated representatives. Here we present 164 new genomes of pleolipoviruses obtained from metagenomic data of Australian hypersaline lakes and publicly available metagenomic data. We perform a comprehensive analysis on the diversity and evolutionary relationships of the newly discovered viruses and previously described pleolipoviruses. We propose to classify the viruses into five genera within the Pleolipoviridae family, with one new genus represented only by virus genomes retrieved in this study. Our data support the current hypothesis that pleolipoviruses reshaped their genomes through recombining with multiple different groups of plasmids, which is reflected in the diversity of their predicted replication strategies. We show that the proposed genus Epsilonpleolipovirus has evolutionary ties to pRN1-like plasmids from Sulfolobus, suggesting that this group could be infecting other archaeal phyla. Interestingly, we observed that the genome size of pleolipoviruses is correlated to the presence or absence of an integrase. Analyses of the host range revealed that all but one virus exhibit an extremely narrow range, and we show that the predicted tertiary structure of the spike protein is strongly associated with the host family, suggesting a specific adaptation to the host S-layer glycoprotein organization.


Subject(s)
Archaeal Viruses , Viruses , Australia , Viruses/genetics , Archaeal Viruses/genetics , Biological Evolution , Integrases/genetics , Archaea/genetics , Genome, Viral/genetics
10.
Dev Biol ; 514: 78-86, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38880275

ABSTRACT

The second heart field (SHF) plays a pivotal role in heart development, particularly in outflow tract (OFT) morphogenesis and septation, as well as in the expansion of the right ventricle (RV). Two mouse Cre lines, the Mef2c-AHF-Cre (Mef2c-Cre) and Isl1-Cre, have been widely used to study the SHF development. However, Cre activity is triggered not only in the SHF but also in the RV in the Mef2c-Cre mice, and in the Isl1-Cre mice, Cre activation is not SHF-specific. Therefore, a more suitable SHF-Cre line is desirable for better understanding SHF development. Here, we generated and characterized the Prdm1-Cre knock-in mice. In comparison with Mef2c-Cre mice, the Cre activity is similar in the pharyngeal and splanchnic mesoderm, and in the OFT of the Prdm1-Cre mice. Nonetheless, it was noticed that Cre expression is largely reduced in the RV of Prdm1-Cre mice compared to the Mef2c-Cre mice. Furthermore, we deleted Hand2, Nkx2-5, Pdk1 and Tbx20 using both Mef2c-Cre and Prdm1-Cre mice to study OFT morphogenesis and septation, making a comparison between these two Cre lines. New insights were obtained in understanding SHF development including differentiation into cardiomyocytes in the OFT using Prdm1-Cre mice. In conclusion, we found that Prdm1-Cre mouse line is a more appropriate tool to monitor SHF development, while the Mef2c-Cre mice are excellent in studying the role and function of the SHF in OFT morphogenesis and septation.


Subject(s)
Heart , Integrases , Positive Regulatory Domain I-Binding Factor 1 , Animals , Mice , Heart/embryology , Integrases/metabolism , Integrases/genetics , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/metabolism , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Mice, Transgenic , Gene Expression Regulation, Developmental/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Gene Knock-In Techniques
11.
Mol Microbiol ; 121(6): 1200-1216, 2024 06.
Article in English | MEDLINE | ID: mdl-38705589

ABSTRACT

Through their involvement in the integration and excision of a large number of mobile genetic elements, such as phages and integrative and conjugative elements (ICEs), site-specific recombination systems based on heterobivalent tyrosine recombinases play a major role in genome dynamics and evolution. However, despite hundreds of these systems having been identified in genome databases, very few have been described in detail, with none from phages that infect Bacillota (formerly Firmicutes). In this study, we reanalyzed the recombination module of Lactobacillus delbrueckii subsp. bulgaricus phage mv4, previously considered atypical compared with classical systems. Our results reveal that mv4 integrase is a 369 aa protein with all the structural hallmarks of recombinases from the Tn916 family and that it cooperatively interacts with its recombination sites. Using randomized DNA libraries, NGS sequencing, and other molecular approaches, we show that the 21-bp core-attP and attB sites have structural similarities to classical systems only if considering the nucleotide degeneracy, with two 7-bp inverted regions corresponding to mv4Int core-binding sites surrounding a 7-bp strand-exchange region. We also examined the different compositional constraints in the core-binding regions, which define the sequence space of permissible recombination sites.


Subject(s)
Attachment Sites, Microbiological , Bacteriophages , Integrases , Recombination, Genetic , Bacteriophages/genetics , Integrases/metabolism , Integrases/genetics , Attachment Sites, Microbiological/genetics , Lactobacillus delbrueckii/virology , Lactobacillus delbrueckii/genetics , Recombinases/metabolism , Recombinases/genetics , Binding Sites
12.
Annu Rev Genet ; 51: 83-102, 2017 11 27.
Article in English | MEDLINE | ID: mdl-29178817

ABSTRACT

The conditional depletion of a protein of interest (POI) is useful not only for loss-of-function studies, but also for the modulation of biological pathways. Technologies that work at the level of DNA, mRNA, and protein are available for temporal protein depletion. Compared with technologies targeting the pretranslation steps, direct protein depletion (or protein knockdown approaches) is advantageous in terms of specificity, reversibility, and time required for depletion, which can be achieved by fusing a POI with a protein domain called a degron that induces rapid proteolysis of the fusion protein. Conditional degrons can be activated or inhibited by temperature, small molecules, light, or the expression of another protein. The conditional degron-based technologies currently available are described and discussed.


Subject(s)
Gene Expression Regulation , Proteasome Endopeptidase Complex/metabolism , Protein Biosynthesis , Proteomics/methods , Animals , CRISPR-Cas Systems , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Humans , Integrases/genetics , Integrases/metabolism , Light , Morpholinos/genetics , Morpholinos/metabolism , Plants/genetics , Plants/metabolism , Protein Biosynthesis/drug effects , Protein Biosynthesis/radiation effects , Protein Domains , Protein Engineering , Proteolysis/drug effects , Proteolysis/radiation effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Small Molecule Libraries/pharmacology
13.
Arterioscler Thromb Vasc Biol ; 44(6): 1393-1406, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38660804

ABSTRACT

BACKGROUND: Low-dose aspirin is widely used for the secondary prevention of cardiovascular disease. The beneficial effects of low-dose aspirin are attributable to its inhibition of platelet Cox (cyclooxygenase)-1-derived thromboxane A2. Until recently, the use of the Pf4 (platelet factor 4) Cre has been the only genetic approach to generating megakaryocyte/platelet ablation of Cox-1 in mice. However, Pf4-ΔCre displays ectopic expression outside the megakaryocyte/platelet lineage, especially during inflammation. The use of the Gp1ba (glycoprotein 1bα) Cre promises a more specific, targeted approach. METHODS: To evaluate the role of Cox-1 in platelets, we crossed Pf4-ΔCre or Gp1ba-ΔCre mice with Cox-1flox/flox mice to generate platelet Cox-1-/- mice on normolipidemic and hyperlipidemic (Ldlr-/-; low-density lipoprotein receptor) backgrounds. RESULTS: Ex vivo platelet aggregation induced by arachidonic acid or adenosine diphosphate in platelet-rich plasma was inhibited to a similar extent in Pf4-ΔCre Cox-1-/-/Ldlr-/- and Gp1ba-ΔCre Cox-1-/-/Ldlr-/- mice. In a mouse model of tail injury, Pf4-ΔCre-mediated and Gp1ba-ΔCre-mediated deletions of Cox-1 were similarly efficient in suppressing platelet prostanoid biosynthesis. Experimental thrombogenesis and attendant blood loss were similar in both models. However, the impact on atherogenesis was divergent, being accelerated in the Pf4-ΔCre mice while restrained in the Gp1ba-ΔCres. In the former, accelerated atherogenesis was associated with greater suppression of PGI2 biosynthesis, a reduction in the lipopolysaccharide-evoked capacity to produce PGE2 (prostaglandin E) and PGD2 (prostanglandin D), activation of the inflammasome, elevated plasma levels of IL-1ß (interleukin), reduced plasma levels of HDL-C (high-density lipoprotein receptor-cholesterol), and a reduction in the capacity for reverse cholesterol transport. By contrast, in the latter, plasma HDL-C and α-tocopherol were elevated, and MIP-1α (macrophage inflammatory protein-1α) and MCP-1 (monocyte chemoattractant protein 1) were reduced. CONCLUSIONS: Both approaches to Cox-1 deletion similarly restrain thrombogenesis, but a differential impact on Cox-1-dependent prostanoid formation by the vasculature may contribute to an inflammatory phenotype and accelerated atherogenesis in Pf4-ΔCre mice.


Subject(s)
Blood Platelets , Cyclooxygenase 1 , Disease Models, Animal , Integrases , Mice, Inbred C57BL , Mice, Knockout , Platelet Aggregation , Platelet Factor 4 , Receptors, LDL , Animals , Blood Platelets/metabolism , Blood Platelets/drug effects , Blood Platelets/enzymology , Cyclooxygenase 1/metabolism , Cyclooxygenase 1/genetics , Cyclooxygenase 1/deficiency , Platelet Aggregation/drug effects , Platelet Factor 4/genetics , Platelet Factor 4/metabolism , Integrases/genetics , Receptors, LDL/genetics , Receptors, LDL/deficiency , Male , Mice , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/enzymology , Atherosclerosis/prevention & control , Atherosclerosis/blood , Hyperlipidemias/blood , Hyperlipidemias/genetics , Hyperlipidemias/enzymology , Phenotype , Membrane Proteins , Platelet Glycoprotein GPIb-IX Complex
14.
Nature ; 575(7781): 195-202, 2019 11.
Article in English | MEDLINE | ID: mdl-31666704

ABSTRACT

The mammalian cortex is a laminar structure containing many areas and cell types that are densely interconnected in complex ways, and for which generalizable principles of organization remain mostly unknown. Here we describe a major expansion of the Allen Mouse Brain Connectivity Atlas resource1, involving around a thousand new tracer experiments in the cortex and its main satellite structure, the thalamus. We used Cre driver lines (mice expressing Cre recombinase) to comprehensively and selectively label brain-wide connections by layer and class of projection neuron. Through observations of axon termination patterns, we have derived a set of generalized anatomical rules to describe corticocortical, thalamocortical and corticothalamic projections. We have built a model to assign connection patterns between areas as either feedforward or feedback, and generated testable predictions of hierarchical positions for individual cortical and thalamic areas and for cortical network modules. Our results show that cell-class-specific connections are organized in a shallow hierarchy within the mouse corticothalamic network.


Subject(s)
Cerebral Cortex/anatomy & histology , Cerebral Cortex/cytology , Neural Pathways/anatomy & histology , Neural Pathways/cytology , Thalamus/anatomy & histology , Thalamus/cytology , Animals , Axons/physiology , Cerebral Cortex/physiology , Female , Integrases/genetics , Integrases/metabolism , Male , Mice , Mice, Inbred C57BL , Neural Pathways/physiology , Thalamus/physiology
15.
Nature ; 566(7742): 105-109, 2019 02.
Article in English | MEDLINE | ID: mdl-30675057

ABSTRACT

A gene drive biases the transmission of one of the two copies of a gene such that it is inherited more frequently than by random segregation. Highly efficient gene drive systems have recently been developed in insects, which leverage the sequence-targeted DNA cleavage activity of CRISPR-Cas9 and endogenous homology-directed repair mechanisms to convert heterozygous genotypes to homozygosity1-4. If implemented in laboratory rodents, similar systems would enable the rapid assembly of currently impractical genotypes that involve multiple homozygous genes (for example, to model multigenic human diseases). To our knowledge, however, such a system has not yet been demonstrated in mammals. Here we use an active genetic element that encodes a guide RNA, which is embedded in the mouse tyrosinase (Tyr) gene, to evaluate whether targeted gene conversion can occur when CRISPR-Cas9 is active in the early embryo or in the developing germline. Although Cas9 efficiently induces double-stranded DNA breaks in the early embryo and male germline, these breaks are not corrected by homology-directed repair. By contrast, Cas9 expression limited to the female germline induces double-stranded breaks that are corrected by homology-directed repair, which copies the active genetic element from the donor to the receiver chromosome and increases its rate of inheritance in the next generation. These results demonstrate the feasibility of CRISPR-Cas9-mediated systems that bias inheritance of desired alleles in mice and that have the potential to transform the use of rodent models in basic and biomedical research.


Subject(s)
CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , Gene Conversion , Gene Drive Technology/methods , Germ-Line Mutation/genetics , Heterozygote , Homozygote , Alleles , Animals , Breeding , CRISPR-Associated Protein 9/genetics , Chromosomes, Mammalian/genetics , DNA Breaks, Double-Stranded , Disease Models, Animal , Embryo, Mammalian/enzymology , Embryo, Mammalian/metabolism , Female , Integrases/genetics , Integrases/metabolism , Male , Mice , Mice, Transgenic , Monophenol Monooxygenase/genetics , RNA, Guide, Kinetoplastida/genetics , Transgenes/genetics
16.
Nature ; 571(7764): 219-225, 2019 07.
Article in English | MEDLINE | ID: mdl-31189177

ABSTRACT

Conventional CRISPR-Cas systems maintain genomic integrity by leveraging guide RNAs for the nuclease-dependent degradation of mobile genetic elements, including plasmids and viruses. Here we describe a notable inversion of this paradigm, in which bacterial Tn7-like transposons have co-opted nuclease-deficient CRISPR-Cas systems to catalyse RNA-guided integration of mobile genetic elements into the genome. Programmable transposition of Vibrio cholerae Tn6677 in Escherichia coli requires CRISPR- and transposon-associated molecular machineries, including a co-complex between the DNA-targeting complex Cascade and the transposition protein TniQ. Integration of donor DNA occurs in one of two possible orientations at a fixed distance downstream of target DNA sequences, and can accommodate variable length genetic payloads. Deep-sequencing experiments reveal highly specific, genome-wide DNA insertion across dozens of unique target sites. This discovery of a fully programmable, RNA-guided integrase lays the foundation for genomic manipulations that obviate the requirements for double-strand breaks and homology-directed repair.


Subject(s)
CRISPR-Cas Systems/genetics , DNA Transposable Elements/genetics , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , Gene Editing/methods , Mutagenesis, Insertional/methods , RNA, Bacterial/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , CRISPR-Associated Proteins/genetics , CRISPR-Associated Proteins/metabolism , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Escherichia coli/genetics , Genome, Bacterial/genetics , Integrases/genetics , Integrases/metabolism , Mutagenesis, Site-Directed/methods , RNA, Guide, Kinetoplastida/genetics , Substrate Specificity , Vibrio cholerae/genetics
17.
Nucleic Acids Res ; 51(13): 6927-6943, 2023 07 21.
Article in English | MEDLINE | ID: mdl-37254817

ABSTRACT

Casposons are transposable elements containing the CRISPR associated gene Cas1solo. Identified in many archaeal genomes, casposons are discussed as the origin of CRISPR-Cas systems due to their proposed Cas1solo-dependent translocation. However, apart from bioinformatic approaches and the demonstration of Cas1solo integrase and endonuclease activity in vitro, casposon transposition has not yet been shown in vivo. Here, we report on active casposon translocations in Methanosarcina mazei Gö1 using two independent experimental approaches. First, mini-casposons, consisting of a R6Kγ origin and two antibiotic resistance cassettes, flanked by target site duplications (TSDs) and terminal inverted repeats (TIRs), were generated, and shown to actively translocate from a suicide plasmid and integrate into the chromosomal MetMaz-C1 TSD IS1a. Second, casposon excision activity was confirmed in a long-term evolution experiment using a Cas1solo overexpression strain in comparison to an empty vector control under four different treatments (native, high temperature, high salt, mitomycin C) to study stress-induced translocation. Analysis of genomic DNA using a nested qPCR approach provided clear evidence of casposon activity in single cells and revealed significantly different casposon excision frequencies between treatments and strains. Our results, providing the first experimental evidence for in vivo casposon activity are summarized in a modified hypothetical translocation model.


Subject(s)
DNA Transposable Elements , Methanosarcina , Humans , Archaeal Proteins/genetics , Integrases/genetics , Methanosarcina/genetics , Plasmids/genetics , Terminal Repeat Sequences , Translocation, Genetic
18.
Nucleic Acids Res ; 51(11): 5527-5546, 2023 06 23.
Article in English | MEDLINE | ID: mdl-37070184

ABSTRACT

Single-stranded DNA (ssDNA) gapped regions are common intermediates in DNA transactions. Using a new non-denaturing bisulfite treatment combined with ChIP-seq, abbreviated 'ssGap-seq', we explore RecA and SSB binding to ssDNA on a genomic scale in E. coli in a wide range of genetic backgrounds. Some results are expected. During log phase growth, RecA and SSB assembly profiles coincide globally, concentrated on the lagging strand and enhanced after UV irradiation. Unexpected results also abound. Near the terminus, RecA binding is favored over SSB, binding patterns change in the absence of RecG, and the absence of XerD results in massive RecA assembly. RecA may substitute for the absence of XerCD to resolve chromosome dimers. A RecA loading pathway may exist that is independent of RecBCD and RecFOR. Two prominent and focused peaks of RecA binding revealed a pair of 222 bp and GC-rich repeats, equidistant from dif and flanking the Ter domain. The repeats, here named RRS for replication risk sequence, trigger a genomically programmed generation of post-replication gaps that may play a special role in relieving topological stress during replication termination and chromosome segregation. As demonstrated here, ssGap-seq provides a new window on previously inaccessible aspects of ssDNA metabolism.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Rec A Recombinases , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Integrases/genetics , Rec A Recombinases/metabolism
19.
PLoS Genet ; 18(4): e1010177, 2022 04.
Article in English | MEDLINE | ID: mdl-35482826

ABSTRACT

Class 1 integrons are widespread genetic elements playing a major role in the dissemination of antibiotic resistance. They allow bacteria to capture, express and exchange antibiotic resistance genes embedded within gene cassettes. Acquisition of gene cassettes is catalysed by the class 1 integron integrase, a site-specific recombinase playing a key role in the integron system. In in vitro planktonic culture, expression of intI1 is controlled by the SOS response, a regulatory network which mediates the repair of DNA damage caused by a wide range of bacterial stress, including antibiotics. However, in vitro experimental conditions are far from the real lifestyle of bacteria in natural environments such as the intestinal tract which is known to be a reservoir of integrons. In this study, we developed an in vivo model of intestinal colonization in gnotobiotic mice and used a recombination assay and quantitative real-time PCR, to investigate the induction of the SOS response and expression and activity of the class 1 integron integrase, IntI1. We found that the basal activity of IntI1 was higher in vivo than in vitro. In addition, we demonstrated that administration of a subinhibitory concentration of ciprofloxacin rapidly induced both the SOS response and intI1 expression that was correlated with an increase of the activity of IntI1. Our findings show that the gut is an environment in which the class 1 integron integrase is induced and active, and they highlight the potential role of integrons in the acquisition and/or expression of resistance genes in the gut, particularly during antibiotic therapy.


Subject(s)
Integrases , Integrons , Intestines , Animals , Anti-Bacterial Agents/pharmacology , Bacteria/genetics , Drug Resistance, Microbial , Integrases/genetics , Integrases/metabolism , Integrons/genetics , Mice
20.
Genesis ; 62(1): e23582, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38069547

ABSTRACT

Tfap2b, a pivotal transcription factor, plays critical roles within neural crest cells and their derived lineage. To unravel the intricate lineage dynamics and contribution of these Tfap2b+ cells during craniofacial development, we established a Tfap2b-CreERT2 knock-in transgenic mouse line using the CRISPR-Cas9-mediated homologous direct repair. By breeding with tdTomato reporter mice and initiating Cre activity through tamoxifen induction at distinct developmental time points, we show the Tfap2b lineage within the key neural crest-derived domains, such as the facial mesenchyme, midbrain, cerebellum, spinal cord, and limbs. Notably, the migratory neurons stemming from the dorsal root ganglia are visible subsequent to Cre activity initiated at E8.5. Intriguingly, Tfap2b+ cells, serving as the progenitors for limb development, show activity predominantly commencing at E10.5. Across the mouse craniofacial landscape, Tfap2b exhibits a widespread presence throughout the facial organs. Here we validate its role as a marker of progenitors in tooth development and have confirmed that this process initiates from E12.5. Our study not only validates the Tfap2b-CreERT2 transgenic line, but also provides a powerful tool for lineage tracing and genetic targeting of Tfap2b-expressing cells and their progenitor in a temporally and spatially regulated manner during the intricate process of development and organogenesis.


Subject(s)
CRISPR-Cas Systems , Tamoxifen , Mice , Animals , Tamoxifen/pharmacology , Mice, Transgenic , Red Fluorescent Protein , Integrases/genetics , Integrases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL