Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113.324
Filter
Add more filters

Publication year range
1.
Annu Rev Biochem ; 93(1): 139-161, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38598855

ABSTRACT

CRISPR-Cas (clustered regularly interspaced short palindromic repeats-CRISPR-associated nuclease) defense systems have been naturally coopted for guide RNA-directed transposition on multiple occasions. In all cases, cooption occurred with diverse elements related to the bacterial transposon Tn7. Tn7 tightly controls transposition; the transposase is activated only when special targets are recognized by dedicated target-site selection proteins. Tn7 and the Tn7-like elements that coopted CRISPR-Cas systems evolved complementary targeting pathways: one that recognizes a highly conserved site in the chromosome and a second pathway that targets mobile plasmids capable of cell-to-cell transfer. Tn7 and Tn7-like elements deliver a single integration into the site they recognize and also control the orientation of the integration event, providing future potential for use as programmable gene-integration tools. Early work has shown that guide RNA-directed transposition systems can be adapted to diverse hosts, even within microbial communities, suggesting great potential for engineering these systems as powerful gene-editing tools.


Subject(s)
CRISPR-Cas Systems , DNA Transposable Elements , RNA, Guide, CRISPR-Cas Systems , Transposases , DNA Transposable Elements/genetics , RNA, Guide, CRISPR-Cas Systems/genetics , RNA, Guide, CRISPR-Cas Systems/metabolism , Transposases/metabolism , Transposases/genetics , Gene Editing/methods , Bacteria/genetics , Plasmids/metabolism , Plasmids/genetics , Clustered Regularly Interspaced Short Palindromic Repeats
2.
Cell ; 187(5): 1206-1222.e16, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38428395

ABSTRACT

Plasmids are extrachromosomal genetic elements that often encode fitness-enhancing features. However, many bacteria carry "cryptic" plasmids that do not confer clear beneficial functions. We identified one such cryptic plasmid, pBI143, which is ubiquitous across industrialized gut microbiomes and is 14 times as numerous as crAssphage, currently established as the most abundant extrachromosomal genetic element in the human gut. The majority of mutations in pBI143 accumulate in specific positions across thousands of metagenomes, indicating strong purifying selection. pBI143 is monoclonal in most individuals, likely due to the priority effect of the version first acquired, often from one's mother. pBI143 can transfer between Bacteroidales, and although it does not appear to impact bacterial host fitness in vivo, it can transiently acquire additional genetic content. We identified important practical applications of pBI143, including its use in identifying human fecal contamination and its potential as an alternative approach to track human colonic inflammatory states.


Subject(s)
Bacteria , Gastrointestinal Tract , Metagenome , Plasmids , Humans , Bacteria/genetics , Bacteroidetes/genetics , Feces/microbiology , Plasmids/genetics
3.
Cell ; 187(19): 5223-5225, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39303689

ABSTRACT

DdmDE is a novel plasmid defense system that was discovered in the seventh pandemic Vibrio cholerae strain of the biotype O1 EI Tor. In this issue of Cell, Yang and coworkers reveal the mechanisms underlying the assembly and activation of the DdmDE defense system.


Subject(s)
Plasmids , Vibrio cholerae , Plasmids/metabolism , Plasmids/genetics , Vibrio cholerae/genetics , Vibrio cholerae/metabolism , Vibrio cholerae/enzymology , Argonaute Proteins/metabolism , Argonaute Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , DNA Helicases/metabolism , DNA Helicases/genetics
4.
Cell ; 187(19): 5253-5266.e16, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39173632

ABSTRACT

Horizontal gene transfer is a key driver of bacterial evolution, but it also presents severe risks to bacteria by introducing invasive mobile genetic elements. To counter these threats, bacteria have developed various defense systems, including prokaryotic Argonautes (pAgos) and the DNA defense module DdmDE system. Through biochemical analysis, structural determination, and in vivo plasmid clearance assays, we elucidate the assembly and activation mechanisms of DdmDE, which eliminates small, multicopy plasmids. We demonstrate that DdmE, a pAgo-like protein, acts as a catalytically inactive, DNA-guided, DNA-targeting defense module. In the presence of guide DNA, DdmE targets plasmids and recruits a dimeric DdmD, which contains nuclease and helicase domains. Upon binding to DNA substrates, DdmD transitions from an autoinhibited dimer to an active monomer, which then translocates along and cleaves the plasmids. Together, our findings reveal the intricate mechanisms underlying DdmDE-mediated plasmid clearance, offering fundamental insights into bacterial defense systems against plasmid invasions.


Subject(s)
Bacterial Proteins , Gene Transfer, Horizontal , Plasmids , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , DNA/metabolism , DNA Helicases/metabolism , DNA, Bacterial/metabolism , DNA, Bacterial/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Models, Molecular , Plasmids/metabolism , Plasmids/genetics
5.
Cell ; 185(17): 3248-3262.e20, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35985290

ABSTRACT

Bacteria encode sophisticated anti-phage systems that are diverse and versatile and display high genetic mobility. How this variability and mobility occurs remains largely unknown. Here, we demonstrate that a widespread family of pathogenicity islands, the phage-inducible chromosomal islands (PICIs), carry an impressive arsenal of defense mechanisms, which can be disseminated intra- and inter-generically by helper phages. These defense systems provide broad immunity, blocking not only phage reproduction, but also plasmid and non-cognate PICI transfer. Our results demonstrate that phages can mobilize PICI-encoded immunity systems to use them against other mobile genetic elements, which compete with the phages for the same bacterial hosts. Therefore, despite the cost, mobilization of PICIs may be beneficial for phages, PICIs, and bacteria in nature. Our results suggest that PICIs are important players controlling horizontal gene transfer and that PICIs and phages establish mutualistic interactions that drive bacterial ecology and evolution.


Subject(s)
Bacteriophages , Genomic Islands , Bacteria/genetics , Bacteriophages/genetics , Gene Transfer, Horizontal , Immune System , Plasmids
6.
Cell ; 184(3): 643-654.e13, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33482082

ABSTRACT

Epstein-Barr virus (EBV) is an oncogenic human herpesvirus that persists as a multicopy episome in proliferating host cells. Episome maintenance is strictly dependent on EBNA1, a sequence-specific DNA-binding protein with no known enzymatic activities. Here, we show that EBNA1 forms a cell cycle-dependent DNA crosslink with the EBV origin of plasmid replication oriP. EBNA1 tyrosine 518 (Y518) is essential for crosslinking to oriP and functionally required for episome maintenance and generation of EBV-transformed lymphoblastoid cell lines (LCLs). Mechanistically, Y518 is required for replication fork termination at oriP in vivo and for formation of SDS-resistant complexes in vitro. EBNA1-DNA crosslinking corresponds to single-strand endonuclease activity specific to DNA structures enriched at replication-termination sites, such as 4-way junctions. These findings reveal that EBNA1 forms tyrosine-dependent DNA-protein crosslinks and single-strand cleavage at oriP required for replication termination and viral episome maintenance.


Subject(s)
Cell Cycle , Cross-Linking Reagents/chemistry , DNA, Viral/metabolism , Epstein-Barr Virus Nuclear Antigens/metabolism , Plasmids/metabolism , Replication Origin , Virus Replication/physiology , Amino Acid Sequence , B-Lymphocytes/metabolism , Cell Line , DNA Adducts/metabolism , DNA Replication , Endonucleases/metabolism , Epstein-Barr Virus Nuclear Antigens/chemistry , Epstein-Barr Virus Nuclear Antigens/genetics , Humans , Mutation/genetics , Protein Binding , Recombination, Genetic/genetics , Tyrosine/metabolism
7.
Annu Rev Biochem ; 89: 77-101, 2020 06 20.
Article in English | MEDLINE | ID: mdl-32569517

ABSTRACT

DNA synthesis technology has progressed to the point that it is now practical to synthesize entire genomes. Quite a variety of methods have been developed, first to synthesize single genes but ultimately to massively edit or write from scratch entire genomes. Synthetic genomes can essentially be clones of native sequences, but this approach does not teach us much new biology. The ability to endow genomes with novel properties offers special promise for addressing questions not easily approachable with conventional gene-at-a-time methods. These include questions about evolution and about how genomes are fundamentally wired informationally, metabolically, and genetically. The techniques and technologies relating to how to design, build, and deliver big DNA at the genome scale are reviewed here. A fuller understanding of these principles may someday lead to the ability to truly design genomes from scratch.


Subject(s)
DNA/genetics , Gene Editing/methods , Gene Transfer Techniques , Genes, Synthetic , Genetic Engineering/methods , Genome , CRISPR-Cas Systems , DNA/chemistry , DNA/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Oligonucleotides/chemical synthesis , Oligonucleotides/metabolism , Plasmids/chemistry , Plasmids/metabolism , Poliovirus/genetics , Poliovirus/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Spheroplasts/genetics , Spheroplasts/metabolism
8.
Cell ; 178(3): 624-639.e19, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31348889

ABSTRACT

Recent breakthroughs with synthetic budding yeast chromosomes expedite the creation of synthetic mammalian chromosomes and genomes. Mammals, unlike budding yeast, depend on the histone H3 variant, CENP-A, to epigenetically specify the location of the centromere-the locus essential for chromosome segregation. Prior human artificial chromosomes (HACs) required large arrays of centromeric α-satellite repeats harboring binding sites for the DNA sequence-specific binding protein, CENP-B. We report the development of a type of HAC that functions independently of these constraints. Formed by an initial CENP-A nucleosome seeding strategy, a construct lacking repetitive centromeric DNA formed several self-sufficient HACs that showed no uptake of genomic DNA. In contrast to traditional α-satellite HAC formation, the non-repetitive construct can form functional HACs without CENP-B or initial CENP-A nucleosome seeding, revealing distinct paths to centromere formation for different DNA sequence types. Our developments streamline the construction and characterization of HACs to facilitate mammalian synthetic genome efforts.


Subject(s)
Centromere/metabolism , Chromosomes, Artificial, Human/metabolism , DNA, Satellite/metabolism , Binding Sites , Cell Line, Tumor , Centromere/genetics , Centromere Protein A/genetics , Centromere Protein A/metabolism , Centromere Protein B/deficiency , Centromere Protein B/genetics , Centromere Protein B/metabolism , Epigenesis, Genetic , Humans , Nucleosomes/chemistry , Nucleosomes/metabolism , Plasmids/genetics , Plasmids/metabolism
9.
Cell ; 179(1): 205-218.e21, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31522888

ABSTRACT

The molecular chaperone HSP90 facilitates the folding of several client proteins, including innate immune receptors and protein kinases. HSP90 is an essential component of plant and animal immunity, yet pathogenic strategies that directly target the chaperone have not been described. Here, we identify the HopBF1 family of bacterial effectors as eukaryotic-specific HSP90 protein kinases. HopBF1 adopts a minimal protein kinase fold that is recognized by HSP90 as a host client. As a result, HopBF1 phosphorylates HSP90 to completely inhibit the chaperone's ATPase activity. We demonstrate that phosphorylation of HSP90 prevents activation of immune receptors that trigger the hypersensitive response in plants. Consequently, HopBF1-dependent phosphorylation of HSP90 is sufficient to induce severe disease symptoms in plants infected with the bacterial pathogen, Pseudomonas syringae. Collectively, our results uncover a family of bacterial effector kinases with toxin-like properties and reveal a previously unrecognized betrayal mechanism by which bacterial pathogens modulate host immunity.


Subject(s)
Arabidopsis Proteins/metabolism , Bacterial Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Molecular Mimicry/immunology , Plant Immunity/physiology , Adenosine Triphosphatases/metabolism , Arabidopsis/immunology , Arabidopsis/metabolism , Arabidopsis/microbiology , Bacterial Proteins/chemistry , HEK293 Cells , HSP90 Heat-Shock Proteins/chemistry , HeLa Cells , Host Microbial Interactions/immunology , Humans , Phosphorylation , Plasmids/genetics , Protein Binding , Protein Folding , Protein Kinases/metabolism , Pseudomonas syringae/metabolism , Saccharomyces cerevisiae/metabolism
10.
Cell ; 179(1): 106-119.e16, 2019 Sep 19.
Article in English | MEDLINE | ID: mdl-31539491

ABSTRACT

Genes are often transcribed by multiple RNA polymerases (RNAPs) at densities that can vary widely across genes and environmental conditions. Here, we provide in vitro and in vivo evidence for a built-in mechanism by which co-transcribing RNAPs display either collaborative or antagonistic dynamics over long distances (>2 kb) through transcription-induced DNA supercoiling. In Escherichia coli, when the promoter is active, co-transcribing RNAPs translocate faster than a single RNAP, but their average speed is not altered by large variations in promoter strength and thus RNAP density. Environmentally induced promoter repression reduces the elongation efficiency of already-loaded RNAPs, causing premature termination and quick synthesis arrest of no-longer-needed proteins. This negative effect appears independent of RNAP convoy formation and is abrogated by topoisomerase I activity. Antagonistic dynamics can also occur between RNAPs from divergently transcribed gene pairs. Our findings may be broadly applicable given that transcription on topologically constrained DNA is the norm across organisms.


Subject(s)
DNA, Bacterial/genetics , DNA, Superhelical/genetics , DNA-Directed RNA Polymerases/genetics , Escherichia coli/genetics , Transcription, Genetic , DNA-Directed RNA Polymerases/chemistry , Gene Expression Regulation, Bacterial/genetics , Glucose/pharmacology , Glycosides/pharmacology , Isopropyl Thiogalactoside/pharmacology , Kinetics , Lac Operon/drug effects , Lac Operon/genetics , Plasmids/genetics , Promoter Regions, Genetic/genetics , RNA, Bacterial/genetics , Real-Time Polymerase Chain Reaction , Rifampin/pharmacology
11.
Cell ; 179(1): 165-179.e18, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31539494

ABSTRACT

The three-dimensional organization of chromosomes can have a profound impact on their replication and expression. The chromosomes of higher eukaryotes possess discrete compartments that are characterized by differing transcriptional activities. Contrastingly, most bacterial chromosomes have simpler organization with local domains, the boundaries of which are influenced by gene expression. Numerous studies have revealed that the higher-order architectures of bacterial and eukaryotic chromosomes are dependent on the actions of structural maintenance of chromosomes (SMC) superfamily protein complexes, in particular, the near-universal condensin complex. Intriguingly, however, many archaea, including members of the genus Sulfolobus do not encode canonical condensin. We describe chromosome conformation capture experiments on Sulfolobus species. These reveal the presence of distinct domains along Sulfolobus chromosomes that undergo discrete and specific higher-order interactions, thus defining two compartment types. We observe causal linkages between compartment identity, gene expression, and binding of a hitherto uncharacterized SMC superfamily protein that we term "coalescin."


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes, Archaeal/metabolism , Sulfolobus/cytology , Sulfolobus/genetics , Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosomes, Archaeal/genetics , DNA Replication/genetics , DNA, Archaeal/metabolism , DNA-Binding Proteins/metabolism , Gene Expression , Genetic Loci/genetics , Models, Genetic , Multiprotein Complexes/metabolism , Plasmids/genetics , Protein Binding/genetics , Transcription, Genetic
12.
Cell ; 174(2): 338-349.e20, 2018 07 12.
Article in English | MEDLINE | ID: mdl-29937223

ABSTRACT

Macromolecular crowding has a profound impact on reaction rates and the physical properties of the cell interior, but the mechanisms that regulate crowding are poorly understood. We developed genetically encoded multimeric nanoparticles (GEMs) to dissect these mechanisms. GEMs are homomultimeric scaffolds fused to a fluorescent protein that self-assemble into bright, stable particles of defined size and shape. By combining tracking of GEMs with genetic and pharmacological approaches, we discovered that the mTORC1 pathway can modulate the effective diffusion coefficient of particles ≥20 nm in diameter more than 2-fold by tuning ribosome concentration, without any discernable effect on the motion of molecules ≤5 nm. This change in ribosome concentration affected phase separation both in vitro and in vivo. Together, these results establish a role for mTORC1 in controlling both the mesoscale biophysical properties of the cytoplasm and biomolecular condensation.


Subject(s)
Cytoplasm/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Diffusion , HEK293 Cells , Humans , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/genetics , Nanoparticles/chemistry , Nanoparticles/metabolism , Particle Size , Plasmids/genetics , Plasmids/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Rheology , Ribosomes/metabolism , Saccharomyces cerevisiae/metabolism , Tuberous Sclerosis Complex 1 Protein/antagonists & inhibitors , Tuberous Sclerosis Complex 1 Protein/genetics , Tuberous Sclerosis Complex 1 Protein/metabolism
13.
Cell ; 174(2): 259-270.e11, 2018 07 12.
Article in English | MEDLINE | ID: mdl-29937224

ABSTRACT

Many community- and hospital-acquired bacterial infections are caused by antibiotic-resistant pathogens. Methicillin-resistant Staphylococcus aureus (MRSA) predisposes humans to invasive infections that are difficult to eradicate. We designed a closed-loop gene network programming mammalian cells to autonomously detect and eliminate bacterial infections. The genetic circuit contains human Toll-like receptors as the bacterial sensor and a synthetic promoter driving reversible and adjustable expression of lysostaphin, a bacteriolytic enzyme highly lethal to S. aureus. Immunomimetic designer cells harboring this genetic circuit exhibited fast and robust sense-and-destroy kinetics against live staphylococci. When tested in a foreign-body infection model in mice, microencapsulated cell implants prevented planktonic MRSA infection and reduced MRSA biofilm formation by 91%. Notably, this system achieved a 100% cure rate of acute MRSA infections, whereas conventional vancomycin treatment failed. These results suggest that immunomimetic designer cells could offer a therapeutic approach for early detection, prevention, and cure of pathogenic infections in the post-antibiotic era.


Subject(s)
Biomimetics/methods , Methicillin-Resistant Staphylococcus aureus/physiology , Staphylococcal Infections/prevention & control , Alkaline Phosphatase/blood , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Disk Diffusion Antimicrobial Tests , Female , HEK293 Cells , Humans , Lipopolysaccharide Receptors/genetics , Lysostaphin/metabolism , Lysostaphin/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Mice , Mice, Inbred C57BL , Plasmids/genetics , Plasmids/metabolism , Staphylococcal Infections/microbiology , Staphylococcal Infections/veterinary , Toll-Like Receptor 1/genetics , Toll-Like Receptor 2/genetics , Toll-Like Receptor 6/genetics , Transcription Factor AP-1/metabolism
14.
Cell ; 174(2): 271-284.e14, 2018 07 12.
Article in English | MEDLINE | ID: mdl-29887373

ABSTRACT

The small intestinal tuft cell-ILC2 circuit mediates epithelial responses to intestinal helminths and protists by tuft cell chemosensory-like sensing and IL-25-mediated activation of lamina propria ILC2s. Small intestine ILC2s constitutively express the IL-25 receptor, which is negatively regulated by A20 (Tnfaip3). A20 deficiency in ILC2s spontaneously triggers the circuit and, unexpectedly, promotes adaptive small-intestinal lengthening and remodeling. Circuit activation occurs upon weaning and is enabled by dietary polysaccharides that render mice permissive for Tritrichomonas colonization, resulting in luminal accumulation of acetate and succinate, metabolites of the protist hydrogenosome. Tuft cells express GPR91, the succinate receptor, and dietary succinate, but not acetate, activates ILC2s via a tuft-, TRPM5-, and IL-25-dependent pathway. Also induced by parasitic helminths, circuit activation and small intestinal remodeling impairs infestation by new helminths, consistent with the phenomenon of concomitant immunity. We describe a metabolic sensing circuit that may have evolved to facilitate mutualistic responses to luminal pathosymbionts.


Subject(s)
Intestine, Small/physiology , Tritrichomonas/metabolism , Acetates/metabolism , Animals , Dietary Fiber/metabolism , Energy Metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/parasitology , Interleukins/genetics , Interleukins/metabolism , Intestinal Mucosa/cytology , Intestine, Small/microbiology , Intestine, Small/parasitology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microbiota , Plasmids/genetics , Plasmids/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Interleukin/metabolism , Receptors, Interleukin-17/genetics , Receptors, Interleukin-17/metabolism , Succinic Acid/metabolism , TRPM Cation Channels/metabolism , Tritrichomonas/growth & development , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism
15.
Cell ; 170(4): 774-786.e19, 2017 Aug 10.
Article in English | MEDLINE | ID: mdl-28802045

ABSTRACT

Conflicts between transcription and replication are a potent source of DNA damage. Co-transcriptional R-loops could aggravate such conflicts by creating an additional barrier to replication fork progression. Here, we use a defined episomal system to investigate how conflict orientation and R-loop formation influence genome stability in human cells. R-loops, but not normal transcription complexes, induce DNA breaks and orientation-specific DNA damage responses during conflicts with replication forks. Unexpectedly, the replisome acts as an orientation-dependent regulator of R-loop levels, reducing R-loops in the co-directional (CD) orientation but promoting their formation in the head-on (HO) orientation. Replication stress and deregulated origin firing increase the number of HO collisions leading to genome-destabilizing R-loops. Our findings connect DNA replication to R-loop homeostasis and suggest a mechanistic basis for genome instability resulting from deregulated DNA replication, observed in cancer and other disease states.


Subject(s)
DNA Replication , Transcription, Genetic , DNA Damage , DNA Replication Timing , Genomic Instability , HEK293 Cells , Humans , Plasmids
16.
Mol Cell ; 84(5): 883-896.e7, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38309275

ABSTRACT

DNA loop-extruding SMC complexes play crucial roles in chromosome folding and DNA immunity. Prokaryotic SMC Wadjet (JET) complexes limit the spread of plasmids through DNA cleavage, yet the mechanisms for plasmid recognition are unresolved. We show that artificial DNA circularization renders linear DNA susceptible to JET nuclease cleavage. Unlike free DNA, JET cleaves immobilized plasmid DNA at a specific site, the plasmid-anchoring point, showing that the anchor hinders DNA extrusion but not DNA cleavage. Structures of plasmid-bound JetABC reveal two presumably stalled SMC motor units that are drastically rearranged from the resting state, together entrapping a U-shaped DNA segment, which is further converted to kinked V-shaped cleavage substrate by JetD nuclease binding. Our findings uncover mechanical bending of residual unextruded DNA as molecular signature for plasmid recognition and non-self DNA elimination. We moreover elucidate key elements of SMC loop extrusion, including the motor direction and the structure of a DNA-holding state.


Subject(s)
DNA , Endonucleases , DNA/metabolism , Plasmids/genetics , Prokaryotic Cells , Cell Cycle Proteins/metabolism
17.
Mol Cell ; 84(14): 2785-2796.e4, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38936361

ABSTRACT

The bacterial world offers diverse strains for understanding medical and environmental processes and for engineering synthetic biological chassis. However, genetically manipulating these strains has faced a long-standing bottleneck: how to efficiently transform DNA. Here, we report imitating methylation patterns rapidly in TXTL (IMPRINT), a generalized, rapid, and scalable approach based on cell-free transcription-translation (TXTL) to overcome DNA restriction, a prominent barrier to transformation. IMPRINT utilizes TXTL to express DNA methyltransferases from a bacterium's restriction-modification systems. The expressed methyltransferases then methylate DNA in vitro to match the bacterium's DNA methylation pattern, circumventing restriction and enhancing transformation. With IMPRINT, we efficiently multiplex methylation by diverse DNA methyltransferases and enhance plasmid transformation in gram-negative and gram-positive bacteria. We also develop a high-throughput pipeline that identifies the most consequential methyltransferases, and we apply IMPRINT to screen a ribosome-binding site library in a hard-to-transform Bifidobacterium. Overall, IMPRINT can enhance DNA transformation, enabling the use of sophisticated genetic manipulation tools across the bacterial world.


Subject(s)
Cell-Free System , DNA Methylation , Protein Biosynthesis , Transcription, Genetic , Escherichia coli/genetics , Escherichia coli/metabolism , Transformation, Bacterial , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , Plasmids/genetics , Plasmids/metabolism , DNA Modification Methylases/metabolism , DNA Modification Methylases/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
18.
Mol Cell ; 83(12): 2122-2136.e10, 2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37267947

ABSTRACT

To spread, transposons must integrate into target sites without disruption of essential genes while avoiding host defense systems. Tn7-like transposons employ multiple mechanisms for target-site selection, including protein-guided targeting and, in CRISPR-associated transposons (CASTs), RNA-guided targeting. Combining phylogenomic and structural analyses, we conducted a broad survey of target selectors, revealing diverse mechanisms used by Tn7 to recognize target sites, including previously uncharacterized target-selector proteins found in newly discovered transposable elements (TEs). We experimentally characterized a CAST I-D system and a Tn6022-like transposon that uses TnsF, which contains an inactivated tyrosine recombinase domain, to target the comM gene. Additionally, we identified a non-Tn7 transposon, Tsy, encoding a homolog of TnsF with an active tyrosine recombinase domain, which we show also inserts into comM. Our findings show that Tn7 transposons employ modular architecture and co-opt target selectors from various sources to optimize target selection and drive transposon spread.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , DNA Transposable Elements , Plasmids , DNA Transposable Elements/genetics , Recombinases/genetics , Tyrosine/genetics
19.
Nature ; 630(8018): 961-967, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38740055

ABSTRACT

Although eukaryotic Argonautes have a pivotal role in post-transcriptional gene regulation through nucleic acid cleavage, some short prokaryotic Argonaute variants (pAgos) rely on auxiliary nuclease factors for efficient foreign DNA degradation1. Here we reveal the activation pathway of the DNA defence module DdmDE system, which rapidly eliminates small, multicopy plasmids from the Vibrio cholerae seventh pandemic strain (7PET)2. Through a combination of cryo-electron microscopy, biochemistry and in vivo plasmid clearance assays, we demonstrate that DdmE is a catalytically inactive, DNA-guided, DNA-targeting pAgo with a distinctive insertion domain. We observe that the helicase-nuclease DdmD transitions from an autoinhibited, dimeric complex to a monomeric state upon loading of single-stranded DNA targets. Furthermore, the complete structure of the DdmDE-guide-target handover complex provides a comprehensive view into how DNA recognition triggers processive plasmid destruction. Our work establishes a mechanistic foundation for how pAgos utilize ancillary factors to achieve plasmid clearance, and provides insights into anti-plasmid immunity in bacteria.


Subject(s)
Argonaute Proteins , Bacterial Proteins , Plasmids , Vibrio cholerae , Argonaute Proteins/chemistry , Argonaute Proteins/metabolism , Argonaute Proteins/ultrastructure , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/ultrastructure , Cryoelectron Microscopy , Deoxyribonucleases/chemistry , Deoxyribonucleases/metabolism , Deoxyribonucleases/ultrastructure , DNA Helicases/chemistry , DNA Helicases/metabolism , DNA Helicases/ultrastructure , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Models, Molecular , Plasmids/genetics , Plasmids/immunology , Plasmids/metabolism , Protein Domains , Protein Multimerization , Vibrio cholerae/genetics , Vibrio cholerae/immunology , Vibrio cholerae/pathogenicity
20.
Mol Cell ; 82(23): 4487-4502.e7, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36427491

ABSTRACT

CRISPR-Cas are prokaryotic adaptive immune systems. Cas nucleases generally use CRISPR-derived RNA guides to specifically bind and cleave DNA or RNA targets. Here, we describe the experimental characterization of a bacterial CRISPR effector protein Cas12m representing subtype V-M. Despite being less than half the size of Cas12a, Cas12m catalyzes auto-processing of a crRNA guide, recognizes a 5'-TTN' protospacer-adjacent motif (PAM), and stably binds a guide-complementary double-stranded DNA (dsDNA). Cas12m has a RuvC domain with a non-canonical catalytic site and accordingly is incapable of guide-dependent cleavage of target nucleic acids. Despite lacking target cleavage activity, the high binding affinity of Cas12m to dsDNA targets allows for interference as demonstrated by its ability to protect bacteria against invading plasmids through silencing invader transcription and/or replication. Based on these molecular features, we repurposed Cas12m by fusing it to a cytidine deaminase that resulted in base editing within a distinct window.


Subject(s)
CRISPR-Associated Proteins , CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems , DNA/genetics , Plasmids , RNA , RNA, Guide, Kinetoplastida/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL