Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Cell ; 177(4): 1067-1079.e19, 2019 05 02.
Article in English | MEDLINE | ID: mdl-31051099

ABSTRACT

The precise control of CRISPR-Cas9 activity is required for a number of genome engineering technologies. Here, we report a generalizable platform that provided the first synthetic small-molecule inhibitors of Streptococcus pyogenes Cas9 (SpCas9) that weigh <500 Da and are cell permeable, reversible, and stable under physiological conditions. We developed a suite of high-throughput assays for SpCas9 functions, including a primary screening assay for SpCas9 binding to the protospacer adjacent motif, and used these assays to screen a structurally diverse collection of natural-product-like small molecules to ultimately identify compounds that disrupt the SpCas9-DNA interaction. Using these synthetic anti-CRISPR small molecules, we demonstrated dose and temporal control of SpCas9 and catalytically impaired SpCas9 technologies, including transcription activation, and identified a pharmacophore for SpCas9 inhibition using structure-activity relationships. These studies establish a platform for rapidly identifying synthetic, miniature, cell-permeable, and reversible inhibitors against both SpCas9 and next-generation CRISPR-associated nucleases.


Subject(s)
CRISPR-Associated Protein 9/antagonists & inhibitors , CRISPR-Cas Systems/physiology , High-Throughput Screening Assays/methods , CRISPR-Associated Protein 9/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats/physiology , DNA/metabolism , Endonucleases/metabolism , Gene Editing/methods , Genome , Small Molecule Libraries , Streptococcus pyogenes/genetics , Substrate Specificity
2.
Cell ; 174(4): 772-774, 2018 08 09.
Article in English | MEDLINE | ID: mdl-30096306

ABSTRACT

Viruses have evolved inhibitors to counteract the CRISPR immune response, but they are not fully potent and need some time to be expressed after the beginning of infection. In this issue of Cell, Borges et al. and Landsberger et al. show that sequential infection gradually immunosuppresses the host to allow effective CRISPR inhibition.


Subject(s)
Bacteriophages/genetics , Viruses/genetics , Clustered Regularly Interspaced Short Palindromic Repeats
3.
Cell ; 164(4): 710-21, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26853474

ABSTRACT

Type III-A CRISPR-Cas systems defend prokaryotes against viral infection using CRISPR RNA (crRNA)-guided nucleases that perform co-transcriptional cleavage of the viral target DNA and its transcripts. Whereas DNA cleavage is essential for immunity, the function of RNA targeting is unknown. Here, we show that transcription-dependent targeting results in a sharp increase of viral genomes in the host cell when the target is located in a late-expressed phage gene. In this targeting condition, mutations in the active sites of the type III-A RNases Csm3 and Csm6 lead to the accumulation of the target phage mRNA and abrogate immunity. Csm6 is also required to provide defense in the presence of mutated phage targets, when DNA cleavage efficiency is reduced. Our results show that the degradation of phage transcripts by CRISPR-associated RNases ensures robust immunity in situations that lead to a slow clearance of the target DNA.


Subject(s)
CRISPR-Cas Systems , RNA Stability , Staphylococcus Phages/genetics , Staphylococcus epidermidis/immunology , Bacterial Proteins , DNA, Viral/genetics , RNA, Viral/metabolism , Staphylococcus Phages/physiology , Staphylococcus epidermidis/virology , Transcription, Genetic
4.
Cell ; 161(5): 1164-1174, 2015 May 21.
Article in English | MEDLINE | ID: mdl-25959775

ABSTRACT

Immune systems must recognize and destroy different pathogens that threaten the host. CRISPR-Cas immune systems protect prokaryotes from viral and plasmid infection utilizing small CRISPR RNAs that are complementary to the invader's genome and specify the targets of RNA-guided Cas nucleases. Type III CRISPR-Cas immunity requires target transcription, and whereas genetic studies demonstrated DNA targeting, in vitro data have shown crRNA-guided RNA cleavage. The molecular mechanism behind these disparate activities is not known. Here, we show that transcription across the targets of the Staphylococcus epidermidis type III-A CRISPR-Cas system results in the cleavage of the target DNA and its transcripts, mediated by independent active sites within the Cas10-Csm ribonucleoprotein effector complex. Immunity against plasmids and DNA viruses requires DNA, but not RNA, cleavage activity. Our studies reveal a highly versatile mechanism of CRISPR immunity that can defend microorganisms against diverse DNA and RNA invaders.


Subject(s)
CRISPR-Cas Systems , Staphylococcus epidermidis/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , DNA/genetics , DNA/metabolism , RNA/genetics , RNA/metabolism , Ribonucleoproteins/metabolism , Staphylococcus epidermidis/immunology , Staphylococcus epidermidis/virology , Transcription, Genetic
5.
Nature ; 625(7996): 797-804, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38200316

ABSTRACT

Prokaryotic type III CRISPR-Cas systems provide immunity against viruses and plasmids using CRISPR-associated Rossman fold (CARF) protein effectors1-5. Recognition of transcripts of these invaders with sequences that are complementary to CRISPR RNA guides leads to the production of cyclic oligoadenylate second messengers, which bind CARF domains and trigger the activity of an effector domain6,7. Whereas most effectors degrade host and invader nucleic acids, some are predicted to contain transmembrane helices without an enzymatic function. Whether and how these CARF-transmembrane helix fusion proteins facilitate the type III CRISPR-Cas immune response remains unknown. Here we investigate the role of cyclic oligoadenylate-activated membrane protein 1 (Cam1) during type III CRISPR immunity. Structural and biochemical analyses reveal that the CARF domains of a Cam1 dimer bind cyclic tetra-adenylate second messengers. In vivo, Cam1 localizes to the membrane, is predicted to form a tetrameric transmembrane pore, and provides defence against viral infection through the induction of membrane depolarization and growth arrest. These results reveal that CRISPR immunity does not always operate through the degradation of nucleic acids, but is instead mediated via a wider range of cellular responses.


Subject(s)
Bacteriophages , CRISPR-Cas Systems , Membrane Potentials , Staphylococcus aureus , Bacteriophages/immunology , Bacteriophages/metabolism , CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems/genetics , CRISPR-Cas Systems/immunology , Nucleotides, Cyclic/metabolism , RNA, Guide, CRISPR-Cas Systems , Second Messenger Systems , Staphylococcus aureus/cytology , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology , Staphylococcus aureus/virology
6.
Nature ; 629(8011): 410-416, 2024 May.
Article in English | MEDLINE | ID: mdl-38632404

ABSTRACT

Bacteria have adapted to phage predation by evolving a vast assortment of defence systems1. Although anti-phage immunity genes can be identified using bioinformatic tools, the discovery of novel systems is restricted to the available prokaryotic sequence data2. Here, to overcome this limitation, we infected Escherichia coli carrying a soil metagenomic DNA library3 with the lytic coliphage T4 to isolate clones carrying protective genes. Following this approach, we identified Brig1, a DNA glycosylase that excises α-glucosyl-hydroxymethylcytosine nucleobases from the bacteriophage T4 genome to generate abasic sites and inhibit viral replication. Brig1 homologues that provide immunity against T-even phages are present in multiple phage defence loci across distinct clades of bacteria. Our study highlights the benefits of screening unsequenced DNA and reveals prokaryotic DNA glycosylases as important players in the bacteria-phage arms race.


Subject(s)
Bacteria , Bacteriophage T4 , DNA Glycosylases , Bacteria/classification , Bacteria/enzymology , Bacteria/genetics , Bacteria/immunology , Bacteria/virology , Bacteriophage T4/growth & development , Bacteriophage T4/immunology , Bacteriophage T4/metabolism , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , Escherichia coli/genetics , Escherichia coli/virology , Gene Library , Metagenomics/methods , Soil Microbiology , Virus Replication
7.
Mol Cell ; 82(21): 3968-3969, 2022 11 03.
Article in English | MEDLINE | ID: mdl-36332604

ABSTRACT

Hoffmann et al. (2022) demonstrate that RNA-guided transposons are remarkably sequence specific due to the action of a AAA+ ATPase, TnsC, that recruits the transposase to the correct target site.


Subject(s)
Escherichia coli Proteins , Escherichia coli Proteins/genetics , Escherichia coli/genetics , DNA Transposable Elements/genetics , DNA, Bacterial , Clustered Regularly Interspaced Short Palindromic Repeats , DNA-Binding Proteins/genetics , Transposases/genetics , Transposases/metabolism
8.
Mol Cell ; 82(5): 907-919.e7, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35134339

ABSTRACT

Prokaryotic organisms have developed multiple defense systems against phages; however, little is known about whether and how these interact with each other. Here, we studied the connection between two of the most prominent prokaryotic immune systems: restriction-modification and CRISPR. While both systems employ enzymes that cleave a specific DNA sequence of the invader, CRISPR nucleases are programmed with phage-derived spacer sequences, which are integrated into the CRISPR locus upon infection. We found that restriction endonucleases provide a short-term defense, which is rapidly overcome through methylation of the phage genome. In a small fraction of the cells, however, restriction results in the acquisition of spacer sequences from the cleavage site, which mediates a robust type II-A CRISPR-Cas immune response against the methylated phage. This mechanism is reminiscent of eukaryotic immunity in which the innate response offers a first temporary line of defense and also activates a second and more robust adaptive response.


Subject(s)
Bacteriophages , DNA, Viral , Bacteriophages/metabolism , CRISPR-Cas Systems , DNA Restriction Enzymes/genetics , DNA, Viral/genetics , Endonucleases/genetics , Immunity
9.
Nature ; 623(7989): 1001-1008, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37968393

ABSTRACT

Cyclic oligonucleotide-based antiphage signalling systems (CBASS) protect prokaryotes from viral (phage) attack through the production of cyclic oligonucleotides, which activate effector proteins that trigger the death of the infected host1,2. How bacterial cyclases recognize phage infection is not known. Here we show that staphylococcal phages produce a structured RNA transcribed from the terminase subunit genes, termed CBASS-activating bacteriophage RNA (cabRNA), which binds to a positively charged surface of the CdnE03 cyclase and promotes the synthesis of the cyclic dinucleotide cGAMP to activate the CBASS immune response. Phages that escape the CBASS defence harbour mutations that lead to the generation of a longer form of the cabRNA that cannot activate CdnE03. As the mammalian cyclase OAS1 also binds viral double-stranded RNA during the interferon response, our results reveal a conserved mechanism for the activation of innate antiviral defence pathways.


Subject(s)
Bacteria , Nucleotidyltransferases , RNA, Viral , Staphylococcus Phages , Animals , 2',5'-Oligoadenylate Synthetase/metabolism , Bacteria/enzymology , Bacteria/immunology , Evolution, Molecular , Immunity, Innate , Nucleotidyltransferases/metabolism , Oligonucleotides/immunology , Oligonucleotides/metabolism , RNA, Viral/immunology , RNA, Viral/metabolism , Signal Transduction/immunology , Staphylococcus Phages/genetics , Staphylococcus Phages/immunology
10.
Annu Rev Genet ; 54: 93-120, 2020 11 23.
Article in English | MEDLINE | ID: mdl-32857635

ABSTRACT

Prokaryotes have developed numerous defense strategies to combat the constant threat posed by the diverse genetic parasites that endanger them. Clustered regularly interspaced short palindromic repeat (CRISPR)-Cas loci guard their hosts with an adaptive immune system against foreign nucleic acids. Protection starts with an immunization phase, in which short pieces of the invader's genome, known as spacers, are captured and integrated into the CRISPR locus after infection. Next, during the targeting phase, spacers are transcribed into CRISPR RNAs (crRNAs) that guide CRISPR-associated (Cas) nucleases to destroy the invader's DNA or RNA. Here we describe the many different molecular mechanisms of CRISPR targeting and how they are interconnected with the immunization phase through a third phase of the CRISPR-Cas immune response: primed spacer acquisition. In this phase, Cas proteins direct the crRNA-guided acquisition of additional spacers to achieve a more rapid and robust immunization of the population.


Subject(s)
Bacteria/genetics , CRISPR-Cas Systems/genetics , Immunity/genetics , Animals , DNA/genetics , RNA/genetics
11.
Mol Cell ; 78(4): 568-569, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32442502

ABSTRACT

Athukoralage et al. (2020) identify a new anti-CRISPR (Acr) that degrades cA4, a cyclic oligo-adenylate second messenger produced during the type III CRISPR immune response. This provides an effective way by which invaders can bypass downstream CRISPR effectors that rely on this signaling molecule.


Subject(s)
Bacteriophages , Clustered Regularly Interspaced Short Palindromic Repeats , Adenine Nucleotides , CRISPR-Cas Systems , Oligoribonucleotides
12.
Nature ; 592(7855): 611-615, 2021 04.
Article in English | MEDLINE | ID: mdl-33828299

ABSTRACT

Horizontal gene transfer and mutation are the two major drivers of microbial evolution that enable bacteria to adapt to fluctuating environmental stressors1. Clustered, regularly interspaced, short palindromic repeats (CRISPR) systems use RNA-guided nucleases to direct sequence-specific destruction of the genomes of mobile genetic elements that mediate horizontal gene transfer, such as conjugative plasmids2 and bacteriophages3, thus limiting the extent to which bacteria can evolve by this mechanism. A subset of CRISPR systems also exhibit non-specific degradation of DNA4,5; however, whether and how this feature affects the host has not yet been examined. Here we show that the non-specific DNase activity of the staphylococcal type III-A CRISPR-Cas system increases mutations in the host and accelerates the generation of antibiotic resistance in Staphylococcus aureus and Staphylococcus epidermidis. These mutations require the induction of the SOS response to DNA damage and display a distinct pattern. Our results demonstrate that by differentially affecting both mechanisms that generate genetic diversity, type III-A CRISPR systems can modulate the evolution of the bacterial host.


Subject(s)
CRISPR-Cas Systems/genetics , CRISPR-Cas Systems/immunology , Mutagenesis , Mutation , Staphylococcus/genetics , Anti-Bacterial Agents/pharmacology , Bacteriophages/classification , Bacteriophages/physiology , CRISPR-Associated Proteins/metabolism , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Deoxyribonucleases/metabolism , Drug Resistance, Microbial/drug effects , SOS Response, Genetics/drug effects , Staphylococcus/drug effects , Staphylococcus/immunology , Staphylococcus/virology , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Staphylococcus aureus/virology , Staphylococcus epidermidis/drug effects , Staphylococcus epidermidis/genetics , Staphylococcus epidermidis/virology , Time Factors
13.
Nature ; 590(7847): 624-629, 2021 02.
Article in English | MEDLINE | ID: mdl-33461211

ABSTRACT

In the type III CRISPR-Cas immune response of prokaryotes, infection triggers the production of cyclic oligoadenylates that bind and activate proteins that contain a CARF domain1,2. Many type III loci are associated with proteins in which the CRISPR-associated Rossman fold (CARF) domain is fused to a restriction  endonuclease-like domain3,4. However, with the exception of the well-characterized Csm6 and Csx1 ribonucleases5,6, whether and how these inducible effectors provide defence is not known. Here we investigated a type III CRISPR accessory protein, which we name cyclic-oligoadenylate-activated single-stranded ribonuclease and single-stranded deoxyribonuclease 1 (Card1). Card1 forms a symmetrical dimer that has a large central cavity between its CRISPR-associated Rossmann fold and restriction endonuclease domains that binds cyclic tetra-adenylate. The binding of ligand results in a conformational change comprising the rotation of individual monomers relative to each other to form a more compact dimeric scaffold, in which a manganese cation coordinates the catalytic residues and activates the cleavage of single-stranded-but not double-stranded-nucleic acids (both DNA and RNA). In vivo, activation of Card1 induces dormancy of the infected hosts to provide immunity against phage infection and plasmids. Our results highlight the diversity of strategies used in CRISPR systems to provide immunity.


Subject(s)
Adenine Nucleotides/metabolism , CRISPR-Cas Systems/immunology , DNA, Single-Stranded/metabolism , Deoxyribonucleases/metabolism , Endoribonucleases/metabolism , Oligoribonucleotides/metabolism , RNA/metabolism , Staphylococcus/enzymology , Staphylococcus/immunology , Adenine Nucleotides/immunology , Adenosine Triphosphate/metabolism , Bacteriophages/immunology , Bacteriophages/physiology , Biocatalysis , Catalytic Domain , Deoxyribonucleases/chemistry , Deoxyribonucleases/genetics , Endoribonucleases/chemistry , Endoribonucleases/genetics , Enzyme Activation , Ligands , Manganese/chemistry , Manganese/metabolism , Models, Molecular , Oligoribonucleotides/immunology , Plasmids/genetics , Plasmids/metabolism , Protein Multimerization , Rotation , Staphylococcus/growth & development , Staphylococcus/virology , Substrate Specificity
14.
Mol Cell ; 73(2): 264-277.e5, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30503773

ABSTRACT

Type ΙΙΙ CRISPR-Cas systems provide robust immunity against foreign RNA and DNA by sequence-specific RNase and target RNA-activated sequence-nonspecific DNase and RNase activities. We report on cryo-EM structures of Thermococcus onnurineus CsmcrRNA binary, CsmcrRNA-target RNA and CsmcrRNA-target RNAanti-tag ternary complexes in the 3.1 Å range. The topological features of the crRNA 5'-repeat tag explains the 5'-ruler mechanism for defining target cleavage sites, with accessibility of positions -2 to -5 within the 5'-repeat serving as sensors for avoidance of autoimmunity. The Csm3 thumb elements introduce periodic kinks in the crRNA-target RNA duplex, facilitating cleavage of the target RNA with 6-nt periodicity. Key Glu residues within a Csm1 loop segment of CsmcrRNA adopt a proposed autoinhibitory conformation suggestive of DNase activity regulation. These structural findings, complemented by mutational studies of key intermolecular contacts, provide insights into CsmcrRNA complex assembly, mechanisms underlying RNA targeting and site-specific periodic cleavage, regulation of DNase cleavage activity, and autoimmunity suppression.


Subject(s)
Autoimmunity , Bacterial Proteins/metabolism , CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Deoxyribonucleases/metabolism , RNA Stability , RNA, Bacterial/metabolism , RNA-Binding Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Proteins/ultrastructure , CRISPR-Associated Proteins/genetics , CRISPR-Associated Proteins/immunology , CRISPR-Associated Proteins/ultrastructure , CRISPR-Cas Systems/genetics , CRISPR-Cas Systems/immunology , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/immunology , Cryoelectron Microscopy , Deoxyribonucleases/genetics , Deoxyribonucleases/immunology , Deoxyribonucleases/ultrastructure , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli/immunology , Gene Expression Regulation, Bacterial , Models, Molecular , Multiprotein Complexes , Mutation , Nucleic Acid Conformation , Protein Conformation , RNA, Bacterial/genetics , RNA, Bacterial/immunology , RNA, Bacterial/ultrastructure , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , RNA-Binding Proteins/ultrastructure , Structure-Activity Relationship , Thermococcus/enzymology , Thermococcus/genetics , Thermococcus/immunology
15.
Mol Cell ; 73(2): 278-290.e4, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30503774

ABSTRACT

Adaptive immune systems must accurately distinguish between self and non-self in order to defend against invading pathogens while avoiding autoimmunity. Type III CRISPR-Cas systems employ guide RNA to recognize complementary RNA targets, which triggers the degradation of both the invader's transcripts and their template DNA. These systems can broadly eliminate foreign targets with multiple mutations but circumvent damage to the host genome. To explore the molecular basis for these features, we use single-molecule fluorescence microscopy to study the interaction between a type III-A ribonucleoprotein complex and various RNA substrates. We find that Cas10-the DNase effector of the complex-displays rapid conformational fluctuations on foreign RNA targets, but is locked in a static configuration on self RNA. Target mutations differentially modulate Cas10 dynamics and tune the CRISPR interference activity in vivo. These findings highlight the central role of the internal dynamics of CRISPR-Cas complexes in self versus non-self discrimination and target specificity.


Subject(s)
Autoimmunity , Bacterial Proteins/immunology , CRISPR-Associated Proteins/immunology , CRISPR-Cas Systems/immunology , Clustered Regularly Interspaced Short Palindromic Repeats/immunology , RNA, Bacterial/immunology , Self Tolerance , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , CRISPR-Associated Proteins/genetics , CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli/immunology , Kinetics , Microscopy, Fluorescence , Mutation , Nucleic Acid Conformation , Protein Conformation , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , Signal Transduction , Single Molecule Imaging/methods , Staphylococcus aureus/enzymology , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology , Staphylococcus epidermidis/enzymology , Staphylococcus epidermidis/genetics , Staphylococcus epidermidis/immunology , Structure-Activity Relationship
16.
Mol Cell ; 71(5): 791-801.e3, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30122537

ABSTRACT

All immune systems use precise target recognition to interrogate foreign invaders. During CRISPR-Cas immunity, prokaryotes capture short spacer sequences from infecting viruses and insert them into the CRISPR array. Transcription and processing of the CRISPR locus generate small RNAs containing the spacer and repeat sequences that guide Cas nucleases to cleave a complementary protospacer in the invading nucleic acids. In most CRISPR systems, sequences flanking the protospacer drastically affect cleavage. Here, we investigated the target requirements of the recently discovered RNA-targeting type VI-A CRISPR-Cas system in its natural host, Listeria seeligeri. We discovered that target RNAs with extended complementarity between the protospacer flanking sequence and the repeat sequence of the guide RNA are not cleaved by the type VI-A nuclease Cas13, neither in vivo nor in vitro. These findings establish fundamental rules for the design of Cas13-based technologies and provide a mechanism for preventing self-targeting in type VI-A systems.


Subject(s)
CRISPR-Cas Systems/genetics , RNA, Guide, Kinetoplastida/genetics , CRISPR-Associated Proteins/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Escherichia coli/genetics , Listeria/genetics , Nucleic Acids/genetics , Ribonucleases/genetics
17.
Mol Cell ; 72(4): 608-609, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30444997

ABSTRACT

Athukoralage et al. (2018) identify a new class of nuclease that degrades cyclic oligoadenylate (cOA), a second messenger that activates non-specific RNA degradation by the type III CRISPR-Cas accessory RNase Csm6/Csx1. This discovery provides a mechanism for regulating the degradation of foreign transcripts during the type III CRISPR immune response.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Ribonucleases , Adenine Nucleotides , CRISPR-Cas Systems , Oligoribonucleotides
18.
Mol Cell ; 71(1): 42-55.e8, 2018 07 05.
Article in English | MEDLINE | ID: mdl-29979968

ABSTRACT

The ability to target the Cas9 nuclease to DNA sequences via Watson-Crick base pairing with a single guide RNA (sgRNA) has provided a dynamic tool for genome editing and an essential component of adaptive immune systems in bacteria. After generating a double-stranded break (DSB), Cas9 remains stably bound to DNA. Here, we show persistent Cas9 binding blocks access to the DSB by repair enzymes, reducing genome editing efficiency. Cas9 can be dislodged by translocating RNA polymerases, but only if the polymerase approaches from one direction toward the Cas9-DSB complex. By exploiting these RNA-polymerase/Cas9 interactions, Cas9 can be conditionally converted into a multi-turnover nuclease, mediating increased mutagenesis frequencies in mammalian cells and enhancing bacterial immunity to bacteriophages. These consequences of a stable Cas9-DSB complex provide insights into the evolution of protospacer adjacent motif (PAM) sequences and a simple method of improving selection of highly active sgRNAs for genome editing.


Subject(s)
CRISPR-Associated Protein 9 , DNA Breaks, Double-Stranded , DNA Repair , Gene Editing , Mouse Embryonic Stem Cells/metabolism , Animals , Bacteria/genetics , Bacteria/metabolism , Bacteria/virology , Bacteriophages/genetics , Bacteriophages/metabolism , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , Cell Line , Mice
19.
Nucleic Acids Res ; 52(17): 10595-10606, 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-38994567

ABSTRACT

Immune responses need to be regulated to prevent autoimmunity. CRISPR-Cas systems provide adaptive immunity in prokaryotes through the acquisition of short DNA sequences from invading viruses (bacteriophages), known as spacers. Spacers are inserted into the CRISPR locus and serve as templates for the transcription of guides used by RNA-guided nucleases to recognize complementary nucleic acids of the invaders and start the CRISPR immune response. In type II-A CRISPR systems, Cas9 uses the guide RNA to cleave target DNA sequences in the genome of infecting phages, and the tracrRNA to bind the promoter of cas genes and repress their transcription. We previously isolated a Cas9 mutant carrying the I473F substitution that increased the frequency of spacer acquisition by 2-3 orders of magnitude, leading to a fitness cost due to higher levels of autoimmunity. Here, we investigated the molecular basis underlying these findings. We found that the I473F mutation decreases the association of Cas9 to tracrRNA, limiting its repressor function, leading to high levels of expression of cas genes, which in turn increase the strength of the type II-A CRISPR-Cas immune response. We obtained similar results for a related type II-A system, and therefore our findings highlight the importance of the interaction between Cas9 and its tracrRNA cofactor in tuning the immune response to balanced levels that enable phage defense but avoid autoimmunity.


Subject(s)
CRISPR-Associated Protein 9 , CRISPR-Cas Systems , CRISPR-Associated Protein 9/metabolism , CRISPR-Associated Protein 9/genetics , Mutation , Escherichia coli/genetics , Bacteriophages/genetics , RNA, Guide, CRISPR-Cas Systems/genetics , RNA, Guide, CRISPR-Cas Systems/metabolism , Promoter Regions, Genetic/genetics
20.
Nature ; 570(7760): 241-245, 2019 06.
Article in English | MEDLINE | ID: mdl-31142834

ABSTRACT

Clustered, regularly interspaced, short palindromic repeat (CRISPR) loci in prokaryotes are composed of 30-40-base-pair repeats separated by equally short sequences of plasmid and bacteriophage origin known as spacers1-3. These loci are transcribed and processed into short CRISPR RNAs (crRNAs) that are used as guides by CRISPR-associated (Cas) nucleases to recognize and destroy complementary sequences (known as protospacers) in foreign nucleic acids4,5. In contrast to most Cas nucleases, which destroy invader DNA4-7, the type VI effector nuclease Cas13 uses RNA guides to locate complementary transcripts and catalyse both sequence-specific cis- and non-specific trans-RNA cleavage8. Although it has been hypothesized that Cas13 naturally defends against RNA phages8, type VI spacer sequences have exclusively been found to match the genomes of double-stranded DNA phages9,10, suggesting that Cas13 can provide immunity against these invaders. However, whether and how Cas13 uses its cis- and/or trans-RNA cleavage activities to defend against double-stranded DNA phages is not understood. Here we show that trans-cleavage of transcripts halts the growth of the host cell and is sufficient to abort the infectious cycle. This depletes the phage population and provides herd immunity to uninfected bacteria. Phages that harbour target mutations, which easily evade DNA-targeting CRISPR systems11-13, are also neutralized when Cas13 is activated by wild-type phages. Thus, by acting on the host rather than directly targeting the virus, type VI CRISPR systems not only provide robust defence against DNA phages but also prevent outbreaks of CRISPR-resistant phage.


Subject(s)
Bacteriophages/immunology , CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems/immunology , Endodeoxyribonucleases/metabolism , Listeria/immunology , Listeria/virology , Bacteriophages/genetics , Bacteriophages/growth & development , CRISPR-Cas Systems/genetics , DNA Viruses/genetics , DNA Viruses/growth & development , DNA Viruses/immunology , Listeria/genetics , Listeria/growth & development , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL