Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
1.
Trends Microbiol ; 32(10): 934-935, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39277459

ABSTRACT

The gut microbiome contains numerous bacteria tied to our health. However, genetically modifying this community remains a major challenge. Brödel et al. take a critical step by engineering bacteriophages to efficiently deliver gene editors without propagation of the genetic cargo, efficiently introducing edits to bacteria residing in the mouse gut.


Subject(s)
Bacteria , Bacteriophages , Gastrointestinal Microbiome , Gene Editing , Bacteriophages/genetics , Bacteriophages/physiology , Animals , Mice , Gene Editing/methods , Bacteria/genetics , Bacteria/virology , Humans
2.
ACS Synth Biol ; 13(9): 2635-2642, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39300908

ABSTRACT

The Cold Spring Harbor Laboratory (CSHL) Summer Course on Synthetic Biology, established in 2013, has emerged as a premier platform for immersive education and research in this dynamic field. Rooted in CSHL's rich legacy of biological discovery, the course offers a comprehensive exploration of synthetic biology's fundamentals and applications. Led by a consortium of faculty from diverse institutions, the course structure seamlessly integrates practical laboratory sessions, exploratory research rotations, and enriching seminars by leaders in the field. Over the years, the curriculum has evolved to cover essential topics such as cell-free transcription-translation, DNA construction, computational modeling of gene circuits, engineered gene regulation, and CRISPR technologies. In this review, we describe the history, development, and structure of the course, and discuss how elements of the course might inform the development of other short courses in synthetic biology. We also demonstrate the course's impact beyond the lab with a summary of alumni contributions to research, education, and entrepreneurship. Through these efforts, the CSHL Summer Course on Synthetic Biology remains at the forefront of shaping the next generation of synthetic biologists.


Subject(s)
Synthetic Biology , Synthetic Biology/methods , Laboratories , Curriculum , Gene Regulatory Networks/genetics , Humans
3.
PLoS Pathog ; 20(8): e1012462, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39159259

ABSTRACT

Multiple peptide resistance factor (MprF) confers resistance to cationic antimicrobial peptides (AMPs) in several pathogens, thereby enabling evasion of the host immune response. The role of MprF in commensals remains, however, uncharacterized. To close this knowledge gap, we used a common gut commensal of animals, Lactiplantibacillus plantarum, and its natural host, the fruit fly Drosophila melanogaster, as an experimental model to investigate the role of MprF in commensal-host interactions. The L. plantarum ΔmprF mutant that we generated exhibited deficiency in the synthesis of lysyl-phosphatidylglycerol (Lys-PG), resulting in increased negative cell surface charge and increased susceptibility to AMPs. Susceptibility to AMPs had no effect on ΔmprF mutant's ability to colonize guts of uninfected flies. However, we observed significantly reduced abundance of the ΔmprF mutant after infection-induced inflammation in the guts of wild-type flies but not of flies lacking AMPs. Additionally, we found that the ΔmprF mutant compared to wild-type L. plantarum induces a stronger intestinal immune response in flies due to the increased release of immunostimulatory peptidoglycan fragments, indicating an important role of MprF in promoting host tolerance to commensals. Our further analysis suggests that MprF-mediated lipoteichoic acid modifications are involved in host immunomodulation. Overall, our results demonstrate that MprF, besides its well-characterized role in pathogen immune evasion and virulence, is also an important commensal resilience factor.


Subject(s)
Drosophila melanogaster , Immune Evasion , Inflammation , Animals , Drosophila melanogaster/immunology , Drosophila melanogaster/microbiology , Immune Evasion/immunology , Inflammation/immunology , Lactobacillus plantarum/immunology , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Antimicrobial Peptides/immunology , Lactobacillaceae/immunology
4.
Nat Commun ; 15(1): 5909, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39003282

ABSTRACT

Many CRISPR-Cas immune systems generate guide (g)RNAs using trans-activating CRISPR RNAs (tracrRNAs). Recent work revealed that Cas9 tracrRNAs could be reprogrammed to convert any RNA-of-interest into a gRNA, linking the RNA's presence to Cas9-mediated cleavage of double-stranded (ds)DNA. Here, we reprogram tracrRNAs from diverse Cas12 nucleases, linking the presence of an RNA-of-interest to dsDNA cleavage and subsequent collateral single-stranded DNA cleavage-all without the RNA necessarily encoding a protospacer-adjacent motif (PAM). After elucidating nuclease-specific design rules, we demonstrate PAM-independent RNA detection with Cas12b, Cas12e, and Cas12f nucleases. Furthermore, rationally truncating the dsDNA target boosts collateral cleavage activity, while the absence of a gRNA reduces background collateral activity and enhances sensitivity. Finally, we apply this platform to detect 16 S rRNA sequences from five different bacterial pathogens using a universal reprogrammed tracrRNA. These findings extend tracrRNA reprogramming to diverse dsDNA-targeting Cas12 nucleases, expanding the flexibility and versatility of CRISPR-based RNA detection.


Subject(s)
CRISPR-Cas Systems , RNA, Guide, CRISPR-Cas Systems , RNA, Guide, CRISPR-Cas Systems/metabolism , RNA, Guide, CRISPR-Cas Systems/genetics , CRISPR-Associated Proteins/metabolism , CRISPR-Associated Proteins/genetics , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/metabolism , DNA/metabolism , DNA/genetics , RNA/metabolism , RNA/genetics , DNA Cleavage , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Gene Editing/methods , Endodeoxyribonucleases/metabolism , Endodeoxyribonucleases/genetics , Francisella/genetics
5.
Nature ; 631(8021): 670-677, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38987591

ABSTRACT

In all organisms, regulation of gene expression must be adjusted to meet cellular requirements and frequently involves helix-turn-helix (HTH) domain proteins1. For instance, in the arms race between bacteria and bacteriophages, rapid expression of phage anti-CRISPR (acr) genes upon infection enables evasion from CRISPR-Cas defence; transcription is then repressed by an HTH-domain-containing anti-CRISPR-associated (Aca) protein, probably to reduce fitness costs from excessive expression2-5. However, how a single HTH regulator adjusts anti-CRISPR production to cope with increasing phage genome copies and accumulating acr mRNA is unknown. Here we show that the HTH domain of the regulator Aca2, in addition to repressing Acr synthesis transcriptionally through DNA binding, inhibits translation of mRNAs by binding conserved RNA stem-loops and blocking ribosome access. The cryo-electron microscopy structure of the approximately 40 kDa Aca2-RNA complex demonstrates how the versatile HTH domain specifically discriminates RNA from DNA binding sites. These combined regulatory modes are widespread in the Aca2 family and facilitate CRISPR-Cas inhibition in the face of rapid phage DNA replication without toxic acr overexpression. Given the ubiquity of HTH-domain-containing proteins, it is anticipated that many more of them elicit regulatory control by dual DNA and RNA binding.


Subject(s)
Bacteriophages , CRISPR-Cas Systems , DNA-Binding Proteins , Gene Expression Regulation, Viral , Helix-Turn-Helix Motifs , RNA-Binding Proteins , Viral Proteins , Bacteriophages/chemistry , Bacteriophages/genetics , Bacteriophages/metabolism , Bacteriophages/ultrastructure , Binding Sites , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , CRISPR-Associated Proteins/metabolism , Cryoelectron Microscopy , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/ultrastructure , Genes, Viral , Models, Molecular , Nucleic Acid Conformation , Pectobacterium carotovorum/virology , Protein Biosynthesis/genetics , Protein Domains , Ribosomes/metabolism , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Messenger/ultrastructure , RNA, Viral/chemistry , RNA, Viral/genetics , RNA, Viral/metabolism , RNA, Viral/ultrastructure , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/ultrastructure , Substrate Specificity , Transcription, Genetic , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism , Viral Proteins/ultrastructure
6.
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
7.
Cell Host Microbe ; 32(6): 875-886.e9, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38754416

ABSTRACT

Plasmid-encoded type IV-A CRISPR-Cas systems lack an acquisition module, feature a DinG helicase instead of a nuclease, and form ribonucleoprotein complexes of unknown biological functions. Type IV-A3 systems are carried by conjugative plasmids that often harbor antibiotic-resistance genes and their CRISPR array contents suggest a role in mediating inter-plasmid conflicts, but this function remains unexplored. Here, we demonstrate that a plasmid-encoded type IV-A3 system co-opts the type I-E adaptation machinery from its host, Klebsiella pneumoniae (K. pneumoniae), to update its CRISPR array. Furthermore, we reveal that robust interference of conjugative plasmids and phages is elicited through CRISPR RNA-dependent transcriptional repression. By silencing plasmid core functions, type IV-A3 impacts the horizontal transfer and stability of targeted plasmids, supporting its role in plasmid competition. Our findings shed light on the mechanisms and ecological function of type IV-A3 systems and demonstrate their practical efficacy for countering antibiotic resistance in clinically relevant strains.


Subject(s)
CRISPR-Cas Systems , Conjugation, Genetic , Klebsiella pneumoniae , Plasmids , Plasmids/genetics , Klebsiella pneumoniae/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Transfer, Horizontal , Bacteriophages/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
8.
Nucleic Acids Res ; 52(10): 6079-6091, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38661215

ABSTRACT

CRISPR-Cas systems can be utilized as programmable-spectrum antimicrobials to combat bacterial infections. However, how CRISPR nucleases perform as antimicrobials across target sites and strains remains poorly explored. Here, we address this knowledge gap by systematically interrogating the use of CRISPR antimicrobials using multidrug-resistant and hypervirulent strains of Klebsiella pneumoniae as models. Comparing different Cas nucleases, DNA-targeting nucleases outperformed RNA-targeting nucleases based on the tested targets. Focusing on AsCas12a that exhibited robust targeting across different strains, we found that the elucidated modes of escape varied widely, restraining opportunities to enhance killing. We also encountered individual guide RNAs yielding different extents of clearance across strains, which were linked to an interplay between improper gRNA folding and strain-specific DNA repair and survival. To explore features that could improve targeting across strains, we performed a genome-wide screen in different K. pneumoniae strains that yielded guide design rules and trained an algorithm for predicting guide efficiency. Finally, we showed that Cas12a antimicrobials can be exploited to eliminate K. pneumoniae when encoded in phagemids delivered by T7-like phages. Altogether, our results highlight the importance of evaluating antimicrobial activity of CRISPR antimicrobials across relevant strains and define critical parameters for efficient CRISPR-based targeting.


Subject(s)
CRISPR-Cas Systems , Klebsiella pneumoniae , RNA, Guide, CRISPR-Cas Systems , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/drug effects , RNA, Guide, CRISPR-Cas Systems/genetics , RNA, Guide, CRISPR-Cas Systems/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Endodeoxyribonucleases/metabolism , Endodeoxyribonucleases/genetics , Klebsiella Infections/drug therapy , Klebsiella Infections/microbiology , CRISPR-Associated Proteins/metabolism , CRISPR-Associated Proteins/genetics , Anti-Bacterial Agents/pharmacology , Drug Resistance, Multiple, Bacterial/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , Genome, Bacterial/genetics , Gene Editing/methods , Humans
9.
Nucleic Acids Res ; 52(7): 4079-4097, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38499498

ABSTRACT

Genome-wide screens have become powerful tools for elucidating genotype-to-phenotype relationships in bacteria. Of the varying techniques to achieve knockout and knockdown, CRISPR base editors are emerging as promising options. However, the limited number of available, efficient target sites hampers their use for high-throughput screening. Here, we make multiple advances to enable flexible base editing as part of high-throughput genetic screening in bacteria. We first co-opt the Streptococcus canis Cas9 that exhibits more flexible protospacer-adjacent motif recognition than the traditional Streptococcus pyogenes Cas9. We then expand beyond introducing premature stop codons by mutating start codons. Next, we derive guide design rules by applying machine learning to an essentiality screen conducted in Escherichia coli. Finally, we rescue poorly edited sites by combining base editing with Cas9-induced cleavage of unedited cells, thereby enriching for intended edits. The efficiency of this dual system was validated through a conditional essentiality screen based on growth in minimal media. Overall, expanding the scope of genome-wide knockout screens with base editors could further facilitate the investigation of new gene functions and interactions in bacteria.


Subject(s)
CRISPR-Cas Systems , Escherichia coli , Gene Editing , Gene Editing/methods , Escherichia coli/genetics , High-Throughput Screening Assays/methods , Genome, Bacterial/genetics , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , Streptococcus/genetics , Streptococcus pyogenes/genetics , Streptococcus pyogenes/enzymology , Machine Learning , RNA, Guide, CRISPR-Cas Systems/genetics
10.
J Bacteriol ; 206(3): e0031723, 2024 03 21.
Article in English | MEDLINE | ID: mdl-38353529

ABSTRACT

Cas9-based gene editing tools have revolutionized genetics, enabling the fast and precise manipulation of diverse bacterial species. However, widely applicable genetic tools for non-model gut bacteria are unavailable. Here, we present a two-plasmid Cas9-based system designed for gene deletion and knock-in complementation in three members of the Klebsiella oxytoca species complex (KoSC), which we applied to study the genetic factors underlying the role of these bacteria in competition against Klebsiella pneumoniae. Firstly, the system allowed efficient and precise full-length gene deletion via enhanced lambda Red expression. Furthermore, we tested the efficiency of two independent, functionally validated complementation strategies. Ultimately, the insertion of universal "bookmark" targets during gene deletion subsequently allows the most optimal genetic complementation in K. oxytoca, Klebsiella michiganensis, and Klebsiella grimontii. This approach offers a significant advantage by enabling the use of a single high-efficiency "bookmark" for complementing other loci or strains, eliminating the need for site-specific design. We revealed that the carbohydrate permease CasA is critical in ex vivo assays for K. pneumoniae inhibition by K. oxytoca but is neither sufficient nor required for K. michiganensis and K. grimontii. Thus, the adaptation of state-of-the-art genetic tools to KoSC allows the identification of species-specific functions in microbial competition. IMPORTANCE: Cas9-based gene editing tools have revolutionized bacterial genetics, yet, their application to non-model gut bacteria is frequently hampered by various limitations. We utilized a two-plasmid Cas9-based system designed for gene deletion in Klebsiella pneumoniae and demonstrate after optimization its utility for gene editing in three members of the Klebsiella oxytoca species complex (KoSC) namely K. oxytoca, Klebsiella michiganensis, and Klebsiella grimontii. We then adapted a recently developed protocol for functional complementation based on universal "bookmark" targets applicable to all tested species. In summary, species-specific adaptation of state-of-the-art genetic tools allows efficient gene deletion and complementation in type strains as well as natural isolates of KoSC members to study microbial interactions.


Subject(s)
CRISPR-Cas Systems , Klebsiella , Klebsiella/genetics , Klebsiella pneumoniae/genetics
12.
Genome Biol ; 25(1): 13, 2024 01 11.
Article in English | MEDLINE | ID: mdl-38200565

ABSTRACT

CRISPR interference (CRISPRi) is the leading technique to silence gene expression in bacteria; however, design rules remain poorly defined. We develop a best-in-class prediction algorithm for guide silencing efficiency by systematically investigating factors influencing guide depletion in genome-wide essentiality screens, with the surprising discovery that gene-specific features substantially impact prediction. We develop a mixed-effect random forest regression model that provides better estimates of guide efficiency. We further apply methods from explainable AI to extract interpretable design rules from the model. This study provides a blueprint for predictive models for CRISPR technologies where only indirect measurements of guide activity are available.


Subject(s)
Algorithms , Clustered Regularly Interspaced Short Palindromic Repeats , Machine Learning
13.
Proc Natl Acad Sci U S A ; 121(6): e2311323121, 2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38294941

ABSTRACT

Microbiota-centric interventions are limited by our incomplete understanding of the gene functions of many of its constituent species. This applies in particular to small RNAs (sRNAs), which are emerging as important regulators in microbiota species yet tend to be missed by traditional functional genomics approaches. Here, we establish CRISPR interference (CRISPRi) in the abundant microbiota member Bacteroides thetaiotaomicron for genome-wide sRNA screens. By assessing the abundance of different protospacer-adjacent motifs, we identify the Prevotella bryantii B14 Cas12a as a suitable nuclease for CRISPR screens in these bacteria and generate an inducible Cas12a expression system. Using a luciferase reporter strain, we infer guide design rules and use this knowledge to assemble a computational pipeline for automated gRNA design. By subjecting the resulting guide library to a phenotypic screen, we uncover the sRNA BatR to increase susceptibility to bile salts through the regulation of genes involved in Bacteroides cell surface structure. Our study lays the groundwork for unlocking the genetic potential of these major human gut mutualists and, more generally, for identifying hidden functions of bacterial sRNAs.


Subject(s)
Bacteroides thetaiotaomicron , RNA, Small Untranslated , Humans , Bacteroides thetaiotaomicron/genetics , RNA, Guide, CRISPR-Cas Systems , Bile , RNA, Bacterial/genetics , RNA, Small Untranslated/genetics
14.
Nucleic Acids Res ; 52(2): 769-783, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38015466

ABSTRACT

CRISPR-Cas systems store fragments of invader DNA as spacers to recognize and clear those same invaders in the future. Spacers can also be acquired from the host's genomic DNA, leading to lethal self-targeting. While self-targeting can be circumvented through different mechanisms, natural examples remain poorly explored. Here, we investigate extensive self-targeting by two CRISPR-Cas systems encoding 24 self-targeting spacers in the plant pathogen Xanthomonas albilineans. We show that the native I-C and I-F1 systems are actively expressed and that CRISPR RNAs are properly processed. When expressed in Escherichia coli, each Cascade complex binds its PAM-flanked DNA target to block transcription, while the addition of Cas3 paired with genome targeting induces cell killing. While exploring how X. albilineans survives self-targeting, we predicted putative anti-CRISPR proteins (Acrs) encoded within the bacterium's genome. Screening of identified candidates with cell-free transcription-translation systems and in E. coli revealed two Acrs, which we named AcrIC11 and AcrIF12Xal, that inhibit the activity of Cas3 but not Cascade of the respective system. While AcrF12Xal is homologous to AcrIF12, AcrIC11 shares sequence and structural homology with the anti-restriction protein KlcA. These findings help explain tolerance of self-targeting through two CRISPR-Cas systems and expand the known suite of DNA degradation-inhibiting Acrs.


Subject(s)
CRISPR-Associated Proteins , Xanthomonas , CRISPR-Cas Systems , Escherichia coli/genetics , Escherichia coli/metabolism , Xanthomonas/genetics , DNA/genetics , CRISPR-Associated Proteins/metabolism
15.
RNA Biol ; 20(1): 666-680, 2023 01.
Article in English | MEDLINE | ID: mdl-37654098

ABSTRACT

CRISPR technologies comprising a Cas nuclease and a guide RNA (gRNA) can utilize multiple gRNAs to enact multi-site editing or regulation in the same cell. Nature devised a highly compact means of encoding gRNAs in the form of CRISPR arrays composed of conserved repeats separated by targeting spacers. However, the capacity to acquire new spacers keeps the arrays longer than necessary for CRISPR technologies. Here, we show that CRISPR arrays utilized by the Cas9 nuclease can be shortened without compromising and sometimes even enhancing targeting activity. Using multiplexed gene repression in E. coli, we found that each region could be systematically shortened to varying degrees before severely compromising targeting activity. Surprisingly, shortening some spacers yielded enhanced targeting activity, which was linked to folding of the transcribed array prior to processing. Overall, shortened CRISPR-Cas9 arrays can facilitate multiplexed editing and gene regulation from a smaller DNA footprint across many bacterial applications of CRISPR technologies.


Subject(s)
CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , DNA Footprinting , Escherichia coli/genetics , Gene Targeting , Bacteria/genetics , Endonucleases
16.
Mol Cell ; 83(17): 3046-3048, 2023 09 07.
Article in English | MEDLINE | ID: mdl-37683609

ABSTRACT

RNA-guided DNA endonucleases such as those from CRISPR-Cas systems were considered limited to prokaryotes. Saito et al.1 reveal that distant eukaryotic relatives of Cas nucleases, called Fanzors, also function as RNA-guided DNA endonucleases and can be harnessed for genome editing.


Subject(s)
Deoxyribonuclease I , Eukaryota , Eukaryota/genetics , Endonucleases/genetics , RNA , DNA/genetics
17.
Nat Microbiol ; 8(9): 1619-1633, 2023 09.
Article in English | MEDLINE | ID: mdl-37500801

ABSTRACT

CRISPR-Cas systems defend prokaryotic cells from invasive DNA of viruses, plasmids and other mobile genetic elements. Here, we show using metagenomics, metatranscriptomics and single-cell genomics that CRISPR systems of widespread, uncultivated archaea can also target chromosomal DNA of archaeal episymbionts of the DPANN superphylum. Using meta-omics datasets from Crystal Geyser and Horonobe Underground Research Laboratory, we find that CRISPR spacers of the hosts Candidatus Altiarchaeum crystalense and Ca. A. horonobense, respectively, match putative essential genes in their episymbionts' genomes of the genus Ca. Huberiarchaeum and that some of these spacers are expressed in situ. Metabolic interaction modelling also reveals complementation between host-episymbiont systems, on the basis of which we propose that episymbionts are either parasitic or mutualistic depending on the genotype of the host. By expanding our analysis to 7,012 archaeal genomes, we suggest that CRISPR-Cas targeting of genomes associated with symbiotic archaea evolved independently in various archaeal lineages.


Subject(s)
Archaea , Symbiosis , Archaea/genetics , Archaea/metabolism , Symbiosis/genetics , Genomics , Plasmids , DNA/metabolism
18.
Sci Rep ; 13(1): 7405, 2023 05 06.
Article in English | MEDLINE | ID: mdl-37149686

ABSTRACT

CRISPR-based gene perturbation enables unbiased investigations of single and combinatorial genotype-to-phenotype associations. In light of efforts to map combinatorial gene dependencies at scale, choosing an efficient and robust CRISPR-associated (Cas) nuclease is of utmost importance. Even though SpCas9 and AsCas12a are widely used for single, combinatorial, and orthogonal screenings, side-by-side comparisons remain sparse. Here, we systematically compared combinatorial SpCas9, AsCas12a, and CHyMErA in hTERT-immortalized retinal pigment epithelial cells and extracted performance-critical parameters for combinatorial and orthogonal CRISPR screens. Our analyses identified SpCas9 to be superior to enhanced and optimized AsCas12a, with CHyMErA being largely inactive in the tested conditions. Since AsCas12a contains RNA processing activity, we used arrayed dual-gRNAs to improve AsCas12a and CHyMErA applications. While this negatively influenced the effect size range of combinatorial AsCas12a applications, it enhanced the performance of CHyMErA. This improved performance, however, was limited to AsCas12a dual-gRNAs, as SpCas9 gRNAs remained largely inactive. To avoid the use of hybrid gRNAs for orthogonal applications, we engineered the multiplex SpCas9-enAsCas12a approach (multiSPAS) that avoids RNA processing for efficient orthogonal gene editing.


Subject(s)
Benchmarking , CRISPR-Cas Systems , Gene Editing , Endonucleases/genetics
19.
Nat Commun ; 14(1): 680, 2023 02 08.
Article in English | MEDLINE | ID: mdl-36754958

ABSTRACT

Bacterial genome editing commonly relies on chromosomal cleavage with Cas nucleases to counter-select against unedited cells. However, editing normally requires efficient recombination and high transformation efficiencies, which are unavailable in most strains. Here, we show that systematically attenuating DNA targeting activity enables RecA-mediated repair in different bacteria, allowing chromosomal cleavage to drive genome editing. Attenuation can be achieved by altering the format or expression strength of guide (g)RNAs; using nucleases with reduced cleavage activity; or engineering attenuated gRNAs (atgRNAs) with disruptive hairpins, perturbed nuclease-binding scaffolds, non-canonical PAMs, or guide mismatches. These modifications greatly increase cell counts and even improve the efficiency of different types of edits for Cas9 and Cas12a in Escherichia coli and Klebsiella oxytoca. We further apply atgRNAs to restore ampicillin sensitivity in Klebsiella pneumoniae, establishing a resistance marker for genetic studies. Attenuating DNA targeting thus offers a counterintuitive means to achieve CRISPR-driven editing across bacteria.


Subject(s)
CRISPR-Cas Systems , Gene Editing , CRISPR-Cas Systems/genetics , DNA/genetics , Genome, Bacterial/genetics , Bacteria/genetics
20.
Nat Biotechnol ; 41(8): 1107-1116, 2023 08.
Article in English | MEDLINE | ID: mdl-36604543

ABSTRACT

Capturing an individual cell's transcriptional history is a challenge exacerbated by the functional heterogeneity of cellular communities. Here, we leverage reprogrammed tracrRNAs (Rptrs) to record selected cellular transcripts as stored DNA edits in single living bacterial cells. Rptrs are designed to base pair with sensed transcripts, converting them into guide RNAs. The guide RNAs then direct a Cas9 base editor to target an introduced DNA target. The extent of base editing can then be read in the future by sequencing. We use this approach, called TIGER (transcribed RNAs inferred by genetically encoded records), to record heterologous and endogenous transcripts in individual bacterial cells. TIGER can quantify relative expression, distinguish single-nucleotide differences, record multiple transcripts simultaneously and read out single-cell phenomena. We further apply TIGER to record metabolic bet hedging and antibiotic resistance mobilization in Escherichia coli as well as host cell invasion by Salmonella. Through RNA recording, TIGER connects current cellular states with past transcriptional states to decipher complex cellular responses in single cells.


Subject(s)
CRISPR-Cas Systems , RNA , RNA/genetics , RNA, Guide, CRISPR-Cas Systems , Bacteria/genetics , Bacteria/metabolism , DNA/genetics , Gene Editing , RNA, Bacterial/genetics
SELECTION OF CITATIONS
SEARCH DETAIL