Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Mol Cell ; 82(23): 4405-4406, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36459983

ABSTRACT

In this issue, Liu et al. present an in-depth study aiming to unravel the structural, biochemical, and physiological aspects of how type III-E CRISPR-Cas systems trigger abortive infection by activating a protease upon target RNA recognition.1.


Subject(s)
CRISPR-Cas Systems , Endopeptidases , Peptide Hydrolases , RNA , Biology
2.
Nucleic Acids Res ; 52(9): 5241-5256, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38647045

ABSTRACT

CRISPR-Cas systems have widely been adopted as genome editing tools, with two frequently employed Cas nucleases being SpyCas9 and LbCas12a. Although both nucleases use RNA guides to find and cleave target DNA sites, the two enzymes differ in terms of protospacer-adjacent motif (PAM) requirements, guide architecture and cleavage mechanism. In the last years, rational engineering led to the creation of PAM-relaxed variants SpRYCas9 and impLbCas12a to broaden the targetable DNA space. By employing their catalytically inactive variants (dCas9/dCas12a), we quantified how the protein-specific characteristics impact the target search process. To allow quantification, we fused these nucleases to the photoactivatable fluorescent protein PAmCherry2.1 and performed single-particle tracking in cells of Escherichia coli. From our tracking analysis, we derived kinetic parameters for each nuclease with a non-targeting RNA guide, strongly suggesting that interrogation of DNA by LbdCas12a variants proceeds faster than that of SpydCas9. In the presence of a targeting RNA guide, both simulations and imaging of cells confirmed that LbdCas12a variants are faster and more efficient in finding a specific target site. Our work demonstrates the trade-off of relaxing PAM requirements in SpydCas9 and LbdCas12a using a powerful framework, which can be applied to other nucleases to quantify their DNA target search.


Subject(s)
CRISPR-Associated Protein 9 , CRISPR-Associated Proteins , CRISPR-Cas Systems , Gene Editing , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , CRISPR-Associated Protein 9/metabolism , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Proteins/metabolism , CRISPR-Associated Proteins/genetics , DNA/metabolism , DNA/genetics , DNA/chemistry , Endodeoxyribonucleases/metabolism , Endodeoxyribonucleases/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Editing/methods , Kinetics , RNA, Guide, CRISPR-Cas Systems/genetics , RNA, Guide, CRISPR-Cas Systems/metabolism
3.
Mol Cell ; 64(6): 1102-1108, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27867010

ABSTRACT

Bacteria commonly exist in high cell density populations, making them prone to viral predation and horizontal gene transfer (HGT) through transformation and conjugation. To combat these invaders, bacteria possess an arsenal of defenses, such as CRISPR-Cas adaptive immunity. Many bacterial populations coordinate their behavior as cell density increases, using quorum sensing (QS) signaling. In this study, we demonstrate that QS regulation results in increased expression of the type I-E, I-F, and III-A CRISPR-Cas systems in Serratia cells in high-density populations. Strains unable to communicate via QS were less effective at defending against invaders targeted by any of the three CRISPR-Cas systems. Additionally, the acquisition of immunity by the type I-E and I-F systems was impaired in the absence of QS signaling. We propose that bacteria can use chemical communication to modulate the balance between community-level defense requirements in high cell density populations and host fitness costs of basal CRISPR-Cas activity.


Subject(s)
Bacterial Proteins/genetics , CRISPR-Cas Systems/immunology , Endodeoxyribonucleases/genetics , Gene Expression Regulation, Bacterial/immunology , Quorum Sensing/genetics , Serratia/genetics , 4-Butyrolactone/analogs & derivatives , 4-Butyrolactone/pharmacology , Bacterial Proteins/immunology , CRISPR-Associated Proteins/genetics , CRISPR-Associated Proteins/immunology , Clustered Regularly Interspaced Short Palindromic Repeats , Endodeoxyribonucleases/immunology , Quorum Sensing/drug effects , Quorum Sensing/immunology , Repressor Proteins/genetics , Repressor Proteins/immunology , Serratia/drug effects , Serratia/immunology
4.
Nucleic Acids Res ; 49(19): 11392-11404, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34614191

ABSTRACT

CRISPR-Cas is a powerful tool for genome editing in bacteria. However, its efficacy is dependent on host factors (such as DNA repair pathways) and/or exogenous expression of recombinases. In this study, we mitigated these constraints by developing a simple and widely applicable genome engineering tool for bacteria which we termed SIBR-Cas (Self-splicing Intron-Based Riboswitch-Cas). SIBR-Cas was generated from a mutant library of the theophylline-dependent self-splicing T4 td intron that allows for tight and inducible control over CRISPR-Cas counter-selection. This control delays CRISPR-Cas counter-selection, granting more time for the editing event (e.g. by homologous recombination) to occur. Without the use of exogenous recombinases, SIBR-Cas was successfully applied to knock-out several genes in three wild-type bacteria species (Escherichia coli MG1655, Pseudomonas putida KT2440 and Flavobacterium IR1) with poor homologous recombination systems. Compared to other genome engineering tools, SIBR-Cas is simple, tightly regulated and widely applicable for most (non-model) bacteria. Furthermore, we propose that SIBR can have a wider application as a simple gene expression and gene regulation control mechanism for any gene or RNA of interest in bacteria.


Subject(s)
Escherichia coli/genetics , Flavobacterium/genetics , Gene Editing/methods , Genome, Bacterial , Pseudomonas putida/genetics , RNA, Bacterial/genetics , Base Pairing , Base Sequence , CRISPR-Cas Systems , Escherichia coli/metabolism , Flavobacterium/metabolism , Gene Knockout Techniques/methods , Homologous Recombination , Introns , Nucleic Acid Conformation , Pseudomonas putida/metabolism , RNA Splicing , RNA, Bacterial/metabolism , Riboswitch
5.
Biochem Soc Trans ; 50(5): 1353-1364, 2022 10 31.
Article in English | MEDLINE | ID: mdl-36282000

ABSTRACT

Type III CRISPR-Cas systems make use of a multi-subunit effector complex to target foreign (m)RNA transcripts complementary to the guide/CRISPR RNA (crRNA). Base-pairing of the target RNA with specialized regions in the crRNA not only triggers target RNA cleavage, but also activates the characteristic Cas10 subunit and sets in motion a variety of catalytic activities that starts with the production of cyclic oligoadenylate (cOA) second messenger molecules. These messenger molecules can activate an extensive arsenal of ancillary effector proteins carrying the appropriate sensory domain. Notably, the CARF and SAVED effector proteins have been responsible for renewed interest in type III CRISPR-Cas due to the extraordinary diversity of defenses against invading genetic elements. Whereas only a handful of CARF and SAVED proteins have been studied so far, many of them seem to provoke abortive infection, aimed to kill the host and provide population-wide immunity. A defining feature of these effector proteins is the variety of in silico-predicted catalytic domains they are fused to. In this mini-review, we discuss all currently characterized type III-associated CARF and SAVED effector proteins, highlight a few examples of predicted CARF and SAVED proteins with interesting predicted catalytic activities, and speculate how they could contribute to type III immunity.


Subject(s)
CRISPR-Associated Proteins , CRISPR-Associated Proteins/genetics , CRISPR-Cas Systems , RNA, Guide, Kinetoplastida , Second Messenger Systems , Catalytic Domain
6.
Mol Cell ; 56(4): 518-30, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25457165

ABSTRACT

CRISPR-Cas is a prokaryotic adaptive immune system that provides sequence-specific defense against foreign nucleic acids. Here we report the structure and function of the effector complex of the Type III-A CRISPR-Cas system of Thermus thermophilus: the Csm complex (TtCsm). TtCsm is composed of five different protein subunits (Csm1-Csm5) with an uneven stoichiometry and a single crRNA of variable size (35-53 nt). The TtCsm crRNA content is similar to the Type III-B Cmr complex, indicating that crRNAs are shared among different subtypes. A negative stain EM structure of the TtCsm complex exhibits the characteristic architecture of Type I and Type III CRISPR-associated ribonucleoprotein complexes. crRNA-protein crosslinking studies show extensive contacts between the Csm3 backbone and the bound crRNA. We show that, like TtCmr, TtCsm cleaves complementary target RNAs at multiple sites. Unlike Type I complexes, interference by TtCsm does not proceed via initial base pairing by a seed sequence.


Subject(s)
Bacterial Proteins/metabolism , CRISPR-Associated Proteins/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , RNA Cleavage , Thermus thermophilus/genetics , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/ultrastructure , Base Sequence , CRISPR-Associated Proteins/chemistry , CRISPR-Associated Proteins/ultrastructure , Endoribonucleases/chemistry , Endoribonucleases/metabolism , Endoribonucleases/ultrastructure , Microscopy, Electron , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Structure, Quaternary , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , Thermus thermophilus/enzymology
7.
Nucleic Acids Res ; 48(6): 3228-3243, 2020 04 06.
Article in English | MEDLINE | ID: mdl-31989168

ABSTRACT

Genome editing has recently made a revolutionary development with the introduction of the CRISPR-Cas technology. The programmable CRISPR-associated Cas9 and Cas12a nucleases generate specific dsDNA breaks in the genome, after which host DNA-repair mechanisms can be manipulated to implement the desired editing. Despite this spectacular progress, the efficiency of Cas9/Cas12a-based engineering can still be improved. Here, we address the variation in guide-dependent efficiency of Cas12a, and set out to reveal the molecular basis of this phenomenon. We established a sensitive and robust in vivo targeting assay based on loss of a target plasmid encoding the red fluorescent protein (mRFP). Our results suggest that folding of both the precursor guide (pre-crRNA) and the mature guide (crRNA) have a major influence on Cas12a activity. Especially, base pairing of the direct repeat, other than with itself, was found to be detrimental to the activity of Cas12a. Furthermore, we describe different approaches to minimize base-pairing interactions between the direct repeat and the variable part of the guide. We show that design of the 3' end of the guide, which is not involved in target strand base pairing, may result in substantial improvement of the guide's targeting potential and hence of its genome editing efficiency.


Subject(s)
Bacterial Proteins/genetics , CRISPR-Associated Proteins/genetics , CRISPR-Cas Systems/genetics , DNA Repair/genetics , Endodeoxyribonucleases/genetics , Gene Editing , CRISPR-Associated Protein 9/genetics , Escherichia coli/genetics , Luminescent Proteins/genetics , Plasmids/genetics , RNA, Guide, Kinetoplastida/genetics
8.
Mol Cell ; 52(1): 135-145, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24119403

ABSTRACT

The CRISPR-Cas system is a prokaryotic host defense system against genetic elements. The Type III-B CRISPR-Cas system of the bacterium Thermus thermophilus, the TtCmr complex, is composed of six different protein subunits (Cmr1-6) and one crRNA with a stoichiometry of Cmr112131445361:crRNA1. The TtCmr complex copurifies with crRNA species of 40 and 46 nt, originating from a distinct subset of CRISPR loci and spacers. The TtCmr complex cleaves the target RNA at multiple sites with 6 nt intervals via a 5' ruler mechanism. Electron microscopy revealed that the structure of TtCmr resembles a "sea worm" and is composed of a Cmr2-3 heterodimer "tail," a helical backbone of Cmr4 subunits capped by Cmr5 subunits, and a curled "head" containing Cmr1 and Cmr6. Despite having a backbone of only four Cmr4 subunits and being both longer and narrower, the overall architecture of TtCmr resembles that of Type I Cascade complexes.


Subject(s)
Bacterial Proteins/metabolism , CRISPR-Associated Proteins/metabolism , RNA, Bacterial/metabolism , Ribonucleases/metabolism , Thermus thermophilus/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , CRISPR-Associated Proteins/chemistry , CRISPR-Associated Proteins/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , High-Throughput Nucleotide Sequencing , Microscopy, Electron , Models, Molecular , Protein Conformation , Protein Subunits , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , Ribonucleases/chemistry , Ribonucleases/genetics , Sequence Analysis, RNA , Spectrometry, Mass, Electrospray Ionization , Structure-Activity Relationship , Thermus thermophilus/genetics
9.
Annu Rev Genet ; 46: 311-39, 2012.
Article in English | MEDLINE | ID: mdl-23145983

ABSTRACT

All organisms need to continuously adapt to changes in their environment. Through horizontal gene transfer, bacteria and archaea can rapidly acquire new traits that may contribute to their survival. However, because new DNA may also cause damage, removal of imported DNA and protection against selfish invading DNA elements are also important. Hence, there should be a delicate balance between DNA uptake and DNA degradation. Here, we describe prokaryotic antiviral defense systems, such as receptor masking or mutagenesis, blocking of phage DNA injection, restriction/modification, and abortive infection. The main focus of this review is on CRISPR (clustered regularly interspaced short palindromic repeats)/Cas (CRISPR-associated), a prokaryotic adaptive immune system. Since its recent discovery, our biochemical understanding of this defense system has made a major leap forward. Three highly diverse CRISPR/Cas types exist that display major structural and functional differences in their mode of generating resistance against invading nucleic acids. Because several excellent recent reviews cover all CRISPR subtypes, we mainly focus on a detailed description of the type I-E CRISPR/Cas system of the model bacterium Escherichia coli K12.


Subject(s)
DNA Helicases/metabolism , Endodeoxyribonucleases/metabolism , Escherichia coli K12/immunology , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial , Amino Acid Motifs , Bacteriophages/genetics , Bacteriophages/immunology , Bacteriophages/pathogenicity , CRISPR-Associated Proteins , DNA Helicases/genetics , DNA, Viral/genetics , DNA, Viral/metabolism , Deoxyribonucleases, Type I Site-Specific/genetics , Deoxyribonucleases, Type I Site-Specific/metabolism , Endodeoxyribonucleases/genetics , Escherichia coli K12/genetics , Escherichia coli K12/metabolism , Escherichia coli K12/virology , Escherichia coli Proteins/genetics , Genes, Bacterial , Lysogeny , Prophages/genetics , Prophages/metabolism , Species Specificity , Virus Internalization
10.
Biochem Soc Trans ; 48(1): 207-219, 2020 02 28.
Article in English | MEDLINE | ID: mdl-31872209

ABSTRACT

Discovered as an adaptive immune system of prokaryotes, CRISPR-Cas provides many promising applications. DNA-cleaving Cas enzymes like Cas9 and Cas12a, are of great interest for genome editing. The specificity of these DNA nucleases is determined by RNA guides, providing great targeting adaptability. Besides this general method of programmable DNA cleavage, these nucleases have different biochemical characteristics, that can be exploited for different applications. Although Cas nucleases are highly promising, some room for improvement remains. New developments and discoveries like base editing, prime editing, and CRISPR-associated transposons might address some of these challenges.


Subject(s)
Bacterial Proteins/genetics , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Proteins/genetics , CRISPR-Cas Systems/genetics , DNA Cleavage , Deoxyribonucleases/genetics , Endodeoxyribonucleases/genetics , RNA, Guide, Kinetoplastida/genetics , Adaptive Immunity , Bacterial Proteins/chemistry , CRISPR-Associated Protein 9/chemistry , CRISPR-Associated Proteins/chemistry , CRISPR-Cas Systems/immunology , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , DNA-Binding Proteins , Deoxyribonucleases/chemistry , Endodeoxyribonucleases/chemistry , Gene Editing/methods , Humans
11.
Mol Cell ; 46(5): 595-605, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22521689

ABSTRACT

The prokaryotic CRISPR/Cas immune system is based on genomic loci that contain incorporated sequence tags from viruses and plasmids. Using small guide RNA molecules, these sequences act as a memory to reject returning invaders. Both the Cascade ribonucleoprotein complex and the Cas3 nuclease/helicase are required for CRISPR interference in Escherichia coli, but it is unknown how natural target DNA molecules are recognized and neutralized by their combined action. Here we show that Cascade efficiently locates target sequences in negatively supercoiled DNA, but only if these are flanked by a protospacer-adjacent motif (PAM). PAM recognition by Cascade exclusively involves the crRNA-complementary DNA strand. After Cascade-mediated R loop formation, the Cse1 subunit recruits Cas3, which catalyzes nicking of target DNA through its HD-nuclease domain. The target is then progressively unwound and cleaved by the joint ATP-dependent helicase activity and Mg(2+)-dependent HD-nuclease activity of Cas3, leading to complete target DNA degradation and invader neutralization.


Subject(s)
DNA Helicases/physiology , DNA, Superhelical/metabolism , Escherichia coli K12/immunology , Escherichia coli Proteins/physiology , Models, Immunological , CRISPR-Associated Proteins , DNA Helicases/genetics , Escherichia coli K12/genetics , Escherichia coli K12/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Nucleic Acid Conformation
12.
Proc Natl Acad Sci U S A ; 114(26): E5122-E5128, 2017 06 27.
Article in English | MEDLINE | ID: mdl-28611213

ABSTRACT

CRISPR-Cas adaptive immune systems capture DNA fragments from invading bacteriophages and plasmids and integrate them as spacers into bacterial CRISPR arrays. In type I-E and II-A CRISPR-Cas systems, this adaptation process is driven by Cas1-Cas2 complexes. Type I-F systems, however, contain a unique fusion of Cas2, with the type I effector helicase and nuclease for invader destruction, Cas3. By using biochemical, structural, and biophysical methods, we present a structural model of the 400-kDa Cas14-Cas2-32 complex from Pectobacterium atrosepticum with bound protospacer substrate DNA. Two Cas1 dimers assemble on a Cas2 domain dimeric core, which is flanked by two Cas3 domains forming a groove where the protospacer binds to Cas1-Cas2. We developed a sensitive in vitro assay and demonstrated that Cas1-Cas2-3 catalyzed spacer integration into CRISPR arrays. The integrase domain of Cas1 was necessary, whereas integration was independent of the helicase or nuclease activities of Cas3. Integration required at least partially duplex protospacers with free 3'-OH groups, and leader-proximal integration was stimulated by integration host factor. In a coupled capture and integration assay, Cas1-Cas2-3 processed and integrated protospacers independent of Cas3 activity. These results provide insight into the structure of protospacer-bound type I Cas1-Cas2-3 adaptation complexes and their integration mechanism.


Subject(s)
Bacterial Proteins/metabolism , CRISPR-Cas Systems/physiology , Endonucleases/metabolism , Multienzyme Complexes/metabolism , Pectobacterium/enzymology , Bacterial Proteins/genetics , Endonucleases/genetics , Multienzyme Complexes/genetics , Pectobacterium/genetics
13.
RNA Biol ; 16(4): 566-576, 2019 04.
Article in English | MEDLINE | ID: mdl-30157725

ABSTRACT

CRISPR-Cas systems provide bacteria and archaea with adaptive immunity against invading genetic elements, such as plasmids, bacteriophages and archaeal viruses. They consist of cas genes and CRISPR loci, which store genetic memories of previously encountered invaders as short sequences termed spacers. Spacers determine the specificity of CRISPR-Cas defence and immunity can be gained or updated by the addition of new spacers into CRISPR loci. There are two main routes to spacer acquisition, which are known as naïve and primed CRISPR adaptation. Naïve CRISPR adaptation involves the de novo formation of immunity, independent of pre-existing spacers. In contrast, primed CRISPR adaptation (priming) uses existing spacers to enhance the acquisition of new spacers. Priming typically results in spacer acquisition from locations near the site of target recognition by the existing (priming) spacer. Primed CRISPR adaptation has been observed in several type I CRISPR-Cas systems and it is potentially widespread. However, experimental evidence is unavailable for some subtypes, and for most systems, priming has only been shown in a small number of hosts. There is also no current evidence of priming by other CRISPR-Cas types. Here, we used a bioinformatic approach to search for evidence of priming in diverse CRISPR-Cas systems. By analysing the clustering of spacers acquired from phages, prophages and archaeal viruses, including strand and directional biases between subsequently acquired spacers, we demonstrate that two patterns of primed CRISPR adaptation dominate in type I systems. In addition, we find evidence of a priming-like pathway in type II CRISPR-Cas systems.


Subject(s)
Adaptation, Physiological/genetics , CRISPR-Cas Systems/genetics , Computational Biology/methods , Genome, Viral , Prophages/genetics
15.
Biochem J ; 473(8): 1063-72, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26929403

ABSTRACT

CRISPR-Cas systems are adaptive immune systems in prokaryotes that provide protection against viruses and other foreign DNA. In the adaptation stage, foreign DNA is integrated into CRISPR (clustered regularly interspaced short palindromic repeat) arrays as new spacers. These spacers are used in the interference stage to guide effector CRISPR associated (Cas) protein(s) to target complementary foreign invading DNA. Cas1 is the integrase enzyme that is central to the catalysis of spacer integration. There are many diverse types of CRISPR-Cas systems, including type I-F systems, which are typified by a unique Cas1-Cas2-3 adaptation complex. In the present study we characterize the Cas1 protein of the potato phytopathogen Pectobacterium atrosepticum, an important model organism for understanding spacer acquisition in type I-F CRISPR-Cas systems. We demonstrate by mutagenesis that Cas1 is essential for adaptation in vivo and requires a conserved aspartic acid residue. By X-ray crystallography, we show that although P. atrosepticum Cas1 adopts a fold conserved among other Cas1 proteins, it possesses remarkable asymmetry as a result of structural plasticity. In particular, we resolve for the first time a flexible, asymmetric loop that may be unique to type I-F Cas1 proteins, and we discuss the implications of these structural features for DNA binding and enzymatic activity.


Subject(s)
CRISPR-Associated Proteins/chemistry , CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems/physiology , Endodeoxyribonucleases/chemistry , Endodeoxyribonucleases/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Crystallization , Crystallography, X-Ray , Pectobacterium/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary
16.
Proc Natl Acad Sci U S A ; 111(16): E1629-38, 2014 Apr 22.
Article in English | MEDLINE | ID: mdl-24711427

ABSTRACT

Prokaryotes encode adaptive immune systems, called CRISPR-Cas (clustered regularly interspaced short palindromic repeats-CRISPR associated), to provide resistance against mobile invaders, such as viruses and plasmids. Host immunity is based on incorporation of invader DNA sequences in a memory locus (CRISPR), the formation of guide RNAs from this locus, and the degradation of cognate invader DNA (protospacer). Invaders can escape type I-E CRISPR-Cas immunity in Escherichia coli K12 by making point mutations in the seed region of the protospacer or its adjacent motif (PAM), but hosts quickly restore immunity by integrating new spacers in a positive-feedback process termed "priming." Here, by using a randomized protospacer and PAM library and high-throughput plasmid loss assays, we provide a systematic analysis of the constraints of both direct interference and subsequent priming in E. coli. We have defined a high-resolution genetic map of direct interference by Cascade and Cas3, which includes five positions of the protospacer at 6-nt intervals that readily tolerate mutations. Importantly, we show that priming is an extremely robust process capable of using degenerate target regions, with up to 13 mutations throughout the PAM and protospacer region. Priming is influenced by the number of mismatches, their position, and is nucleotide dependent. Our findings imply that even outdated spacers containing many mismatches can induce a rapid primed CRISPR response against diversified or related invaders, giving microbes an advantage in the coevolutionary arms race with their invaders.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Escherichia coli K12/genetics , Escherichia coli K12/immunology , Base Pair Mismatch/genetics , Base Sequence , Databases, Genetic , High-Throughput Screening Assays , Molecular Sequence Data , Mutation/genetics , Nucleotide Motifs/genetics , Plasmids/genetics , RNA, Bacterial/genetics
17.
BMC Genomics ; 17: 356, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27184979

ABSTRACT

BACKGROUND: CRISPR (clustered regularly interspaced short palindromic repeats) RNAs provide the specificity for noncoding RNA-guided adaptive immune defence systems in prokaryotes. CRISPR arrays consist of repeat sequences separated by specific spacer sequences. CRISPR arrays have previously been identified in a large proportion of prokaryotic genomes. However, currently available detection algorithms do not utilise recently discovered features regarding CRISPR loci. RESULTS: We have developed a new approach to automatically detect, predict and interactively refine CRISPR arrays. It is available as a web program and command line from bioanalysis.otago.ac.nz/CRISPRDetect. CRISPRDetect discovers putative arrays, extends the array by detecting additional variant repeats, corrects the direction of arrays, refines the repeat/spacer boundaries, and annotates different types of sequence variations (e.g. insertion/deletion) in near identical repeats. Due to these features, CRISPRDetect has significant advantages when compared to existing identification tools. As well as further support for small medium and large repeats, CRISPRDetect identified a class of arrays with 'extra-large' repeats in bacteria (repeats 44-50 nt). The CRISPRDetect output is integrated with other analysis tools. Notably, the predicted spacers can be directly utilised by CRISPRTarget to predict targets. CONCLUSION: CRISPRDetect enables more accurate detection of arrays and spacers and its gff output is suitable for inclusion in genome annotation pipelines and visualisation. It has been used to analyse all complete bacterial and archaeal reference genomes.


Subject(s)
CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Software , Algorithms , Bacteria/genetics , DNA, Intergenic , Databases, Nucleic Acid , Genome , Genomics/methods , Mutagenesis, Insertional , Mutation , Prokaryotic Cells/metabolism , Sequence Deletion , Tandem Repeat Sequences , User-Computer Interface , Workflow
18.
Methods ; 89: 138-48, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26071038

ABSTRACT

Ribonucleoprotein (RNP) complexes play important roles in the cell by mediating basic cellular processes, including gene expression and its regulation. Understanding the molecular details of these processes requires the identification and characterization of protein-RNA interactions. Over the years various approaches have been used to investigate these interactions, including computational analyses to look for RNA binding domains, gel-shift mobility assays on recombinant and mutant proteins as well as co-crystallization and NMR studies for structure elucidation. Here we report a more specialized and direct approach using UV-induced cross-linking coupled with mass spectrometry. This approach permits the identification of cross-linked peptides and RNA moieties and can also pin-point exact RNA contact sites within the protein. The power of this method is illustrated by the application to different single- and multi-subunit RNP complexes belonging to the prokaryotic adaptive immune system, CRISPR-Cas (CRISPR: clustered regularly interspaced short palindromic repeats; Cas: CRISPR associated). In particular, we identified the RNA-binding sites within three Cas7 protein homologs and mapped the cross-linking results to reveal structurally conserved Cas7 - RNA binding interfaces. These results demonstrate the strong potential of UV-induced cross-linking coupled with mass spectrometry analysis to identify RNA interaction sites on the RNA binding proteins.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats/physiology , RNA-Binding Proteins/analysis , RNA-Binding Proteins/metabolism , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/methods , Ultraviolet Rays , Photic Stimulation/methods , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , RNA/analysis , RNA/chemistry , RNA/metabolism , RNA-Binding Proteins/chemistry
19.
Nucleic Acids Res ; 42(11): 7358-69, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24848017

ABSTRACT

U12-type introns are a rare class of introns in the genomes of diverse eukaryotes. In the human genome, they number over 700. A subset of these introns has been shown to be spliced at a slower rate compared to the major U2-type introns. This suggests a rate-limiting regulatory function for the minor spliceosome in the processing of transcripts containing U12-type introns. However, both the generality of slower splicing and the subsequent fate of partially processed pre-mRNAs remained unknown. Here, we present a global analysis of the nuclear retention of transcripts containing U12-type introns and provide evidence for the nuclear decay of such transcripts in human cells. Using SOLiD RNA sequencing technology, we find that, in normal cells, U12-type introns are on average 2-fold more retained than the surrounding U2-type introns. Furthermore, we find that knockdown of RRP41 and DIS3 subunits of the exosome stabilizes an overlapping set of U12-type introns. RRP41 knockdown leads to slower decay kinetics of U12-type introns and globally upregulates the retention of U12-type, but not U2-type, introns. Our results indicate that U12-type introns are spliced less efficiently and are targeted by the exosome. These characteristics support their role in the regulation of cellular mRNA levels.


Subject(s)
Cell Nucleus/metabolism , Exosome Multienzyme Ribonuclease Complex/metabolism , Introns , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional , RNA Stability , RNA, Messenger/metabolism , Cell Line , Cell Nucleus/enzymology , Exosome Multienzyme Ribonuclease Complex/antagonists & inhibitors , Humans , RNA, Small Nuclear/metabolism , RNA-Binding Proteins/antagonists & inhibitors
20.
Nucleic Acids Res ; 42(13): 8516-26, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24990370

ABSTRACT

Clustered regularly interspaced short palindromic repeats (CRISPR), in combination with CRISPR associated (cas) genes, constitute CRISPR-Cas bacterial adaptive immune systems. To generate immunity, these systems acquire short sequences of nucleic acids from foreign invaders and incorporate these into their CRISPR arrays as spacers. This adaptation process is the least characterized step in CRISPR-Cas immunity. Here, we used Pectobacterium atrosepticum to investigate adaptation in Type I-F CRISPR-Cas systems. Pre-existing spacers that matched plasmids stimulated hyperactive primed acquisition and resulted in the incorporation of up to nine new spacers across all three native CRISPR arrays. Endogenous expression of the cas genes was sufficient, yet required, for priming. The new spacers inhibited conjugation and transformation, and interference was enhanced with increasing numbers of new spacers. We analyzed ∼ 350 new spacers acquired in priming events and identified a 5'-protospacer-GG-3' protospacer adjacent motif. In contrast to priming in Type I-E systems, new spacers matched either plasmid strand and a biased distribution, including clustering near the primed protospacer, suggested a bi-directional translocation model for the Cas1:Cas2-3 adaptation machinery. Taken together these results indicate priming adaptation occurs in different CRISPR-Cas systems, that it can be highly active in wild-type strains and that the underlying mechanisms vary.


Subject(s)
CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , CRISPR-Associated Proteins/genetics , Conjugation, Genetic , Pectobacterium/genetics , Plasmids/genetics , Transformation, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL