Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.014
Filter
Add more filters

Publication year range
1.
Cell ; 186(22): 4956-4973.e21, 2023 10 26.
Article in English | MEDLINE | ID: mdl-37852260

ABSTRACT

The complement system is a critical part of our innate immune response, and the terminal products of this cascade, anaphylatoxins C3a and C5a, exert their physiological and pathophysiological responses primarily via two GPCRs, C3aR and C5aR1. However, the molecular mechanism of ligand recognition, activation, and signaling bias of these receptors remains mostly elusive. Here, we present nine cryo-EM structures of C3aR and C5aR1 activated by their natural and synthetic agonists, which reveal distinct binding pocket topologies of complement anaphylatoxins and provide key insights into receptor activation and transducer coupling. We also uncover the structural basis of a naturally occurring mechanism to dampen the inflammatory response of C5a via proteolytic cleavage of the terminal arginine and the G-protein signaling bias elicited by a peptide agonist of C3aR identified here. In summary, our study elucidates the innerworkings of the complement anaphylatoxin receptors and should facilitate structure-guided drug discovery to target these receptors in a spectrum of disorders.


Subject(s)
Anaphylatoxins , Receptors, Complement , Signal Transduction , Anaphylatoxins/metabolism , Complement C3a/metabolism , Immunity, Innate , Receptors, Complement/metabolism , Humans , Animals , Mice
2.
Cell ; 186(19): 4204-4215.e19, 2023 09 14.
Article in English | MEDLINE | ID: mdl-37557170

ABSTRACT

Tn7-like transposons have co-opted CRISPR-Cas systems to facilitate the movement of their own DNA. These CRISPR-associated transposons (CASTs) are promising tools for programmable gene knockin. A key feature of CASTs is their ability to recruit Tn7-like transposons to nuclease-deficient CRISPR effectors. However, how Tn7-like transposons are recruited by diverse CRISPR effectors remains poorly understood. Here, we present the cryo-EM structure of a recruitment complex comprising the Cascade complex, TniQ, TnsC, and the target DNA in the type I-B CAST from Peltigera membranacea cyanobiont 210A. Target DNA recognition by Cascade induces conformational changes in Cas6 and primes TniQ recruitment through its C-terminal domain. The N-terminal domain of TniQ is bound to the seam region of the TnsC spiral heptamer. Our findings provide insights into the diverse mechanisms for the recruitment of Tn7-like transposons to CRISPR effectors and will aid in the development of CASTs as gene knockin tools.


Subject(s)
Ascomycota , CRISPR-Associated Proteins , CRISPR-Cas Systems , DNA Transposable Elements , Gene Knock-In Techniques , CRISPR-Associated Proteins/metabolism , CRISPR-Associated Proteins/ultrastructure , Cryoelectron Microscopy , Ascomycota/chemistry , Ascomycota/metabolism , Ascomycota/ultrastructure
3.
Cell ; 184(9): 2441-2453.e18, 2021 04 29.
Article in English | MEDLINE | ID: mdl-33770501

ABSTRACT

Tn7-like transposons have co-opted CRISPR systems, including class 1 type I-F, I-B, and class 2 type V-K. Intriguingly, although these CRISPR-associated transposases (CASTs) undergo robust CRISPR RNA (crRNA)-guided transposition, they are almost never found in sites targeted by the crRNAs encoded by the cognate CRISPR array. To understand this paradox, we investigated CAST V-K and I-B systems and found two distinct modes of transposition: (1) crRNA-guided transposition and (2) CRISPR array-independent homing. We show distinct CAST systems utilize different molecular mechanisms to target their homing site. Type V-K CAST systems use a short, delocalized crRNA for RNA-guided homing, whereas type I-B CAST systems, which contain two distinct target selector proteins, use TniQ for RNA-guided DNA transposition and TnsD for homing to an attachment site. These observations illuminate a key step in the life cycle of CAST systems and highlight the diversity of molecular mechanisms mediating transposon homing.


Subject(s)
Bacteria/genetics , Bacterial Proteins/metabolism , CRISPR-Associated Proteins/metabolism , DNA Transposable Elements/physiology , DNA, Bacterial/metabolism , RNA, Guide, Kinetoplastida , Transposases/metabolism , Bacteria/metabolism , Bacterial Proteins/genetics , CRISPR-Associated Proteins/genetics , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , DNA, Bacterial/genetics , Gene Editing , Recombination, Genetic , Transposases/genetics
4.
Cell ; 182(4): 976-991.e19, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32702314

ABSTRACT

Although complex inflammatory-like alterations are observed around the amyloid plaques of Alzheimer's disease (AD), little is known about the molecular changes and cellular interactions that characterize this response. We investigate here, in an AD mouse model, the transcriptional changes occurring in tissue domains in a 100-µm diameter around amyloid plaques using spatial transcriptomics. We demonstrate early alterations in a gene co-expression network enriched for myelin and oligodendrocyte genes (OLIGs), whereas a multicellular gene co-expression network of plaque-induced genes (PIGs) involving the complement system, oxidative stress, lysosomes, and inflammation is prominent in the later phase of the disease. We confirm the majority of the observed alterations at the cellular level using in situ sequencing on mouse and human brain sections. Genome-wide spatial transcriptomics analysis provides an unprecedented approach to untangle the dysregulated cellular network in the vicinity of pathogenic hallmarks of AD and other brain diseases.


Subject(s)
Alzheimer Disease/pathology , Sequence Analysis, DNA/methods , Transcriptome , Alzheimer Disease/genetics , Amyloid/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Brain/pathology , Complement System Proteins/genetics , Complement System Proteins/metabolism , Disease Models, Animal , Gene Expression Profiling , Humans , Lysosomes/genetics , Lysosomes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Sheath/genetics , Myelin Sheath/metabolism , Oxidative Stress/genetics
5.
Cell ; 175(4): 934-946.e15, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30343903

ABSTRACT

CRISPR-Cas systems confer an adaptive immunity against viruses. Following viral injection, Cas1-Cas2 integrates segments of the viral genome (spacers) into the CRISPR locus. In type I CRISPR-Cas systems, efficient "primed" spacer acquisition and viral degradation (interference) require both the Cascade complex and the Cas3 helicase/nuclease. Here, we present single-molecule characterization of the Thermobifida fusca (Tfu) primed acquisition complex (PAC). We show that TfuCascade rapidly samples non-specific DNA via facilitated one-dimensional diffusion. Cas3 loads at target-bound Cascade and the Cascade/Cas3 complex translocates via a looped DNA intermediate. Cascade/Cas3 complexes stall at diverse protein roadblocks, resulting in a double strand break at the stall site. In contrast, Cas1-Cas2 samples DNA transiently via 3D collisions. Moreover, Cas1-Cas2 associates with Cascade and translocates with Cascade/Cas3, forming the PAC. PACs can displace different protein roadblocks, suggesting a mechanism for long-range spacer acquisition. This work provides a molecular basis for the coordinated steps in CRISPR-based adaptive immunity.


Subject(s)
Actinomycetales/enzymology , CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , CRISPR-Associated Proteins/chemistry , DNA, Viral/metabolism , Protein Multimerization , Single Molecule Imaging
6.
Cell ; 170(1): 35-47.e13, 2017 Jun 29.
Article in English | MEDLINE | ID: mdl-28666121

ABSTRACT

CRISPR-Cas nucleoproteins target foreign DNA via base pairing with a crRNA. However, a quantitative description of protein binding and nuclease activation at off-target DNA sequences remains elusive. Here, we describe a chip-hybridized association-mapping platform (CHAMP) that repurposes next-generation sequencing chips to simultaneously measure the interactions between proteins and ∼107 unique DNA sequences. Using CHAMP, we provide the first comprehensive survey of DNA recognition by a type I-E CRISPR-Cas (Cascade) complex and Cas3 nuclease. Analysis of mutated target sequences and human genomic DNA reveal that Cascade recognizes an extended protospacer adjacent motif (PAM). Cascade recognizes DNA with a surprising 3-nt periodicity. The identity of the PAM and the PAM-proximal nucleotides control Cas3 recruitment by releasing the Cse1 subunit. These findings are used to develop a model for the biophysical constraints governing off-target DNA binding. CHAMP provides a framework for high-throughput, quantitative analysis of protein-DNA interactions on synthetic and genomic DNA. PAPERCLIP.


Subject(s)
DNA-Binding Proteins/analysis , High-Throughput Nucleotide Sequencing/methods , Protein Binding , Sequence Analysis, DNA/methods , CRISPR-Cas Systems , Electrophoretic Mobility Shift Assay , Microscopy, Fluorescence , Nucleotide Motifs
7.
Cell ; 170(1): 48-60.e11, 2017 Jun 29.
Article in English | MEDLINE | ID: mdl-28666122

ABSTRACT

Type I CRISPR systems feature a sequential dsDNA target searching and degradation process, by crRNA-displaying Cascade and nuclease-helicase fusion enzyme Cas3, respectively. Here we present two cryo-EM snapshots of the Thermobifida fusca type I-E Cascade: (1) unwinding 11 bp of dsDNA at the seed-sequence region to scout for sequence complementarity, and (2) further unwinding of the entire protospacer to form a full R-loop. These structures provide the much-needed temporal and spatial resolution to resolve key mechanistic steps leading to Cas3 recruitment. In the early steps, PAM recognition causes severe DNA bending, leading to spontaneous DNA unwinding to form a seed-bubble. The full R-loop formation triggers conformational changes in Cascade, licensing Cas3 to bind. The same process also generates a bulge in the non-target DNA strand, enabling its handover to Cas3 for cleavage. The combination of both negative and positive checkpoints ensures stringent yet efficient target degradation in type I CRISPR-Cas systems.


Subject(s)
Actinobacteria/genetics , Actinobacteria/ultrastructure , CRISPR-Cas Systems , Nucleic Acid Hybridization , Actinobacteria/chemistry , Actinobacteria/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/ultrastructure , Base Sequence , CRISPR-Associated Proteins/chemistry , CRISPR-Associated Proteins/metabolism , CRISPR-Associated Proteins/ultrastructure , Cryoelectron Microscopy , Models, Molecular , RNA, Bacterial/chemistry , RNA, Bacterial/metabolism , RNA, Small Untranslated/chemistry , RNA, Small Untranslated/metabolism
8.
Mol Cell ; 84(3): 463-475.e5, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38242128

ABSTRACT

Type I CRISPR-Cas systems utilize the RNA-guided Cascade complex to identify matching DNA targets and the nuclease-helicase Cas3 to degrade them. Among the seven subtypes, type I-C is compact in size and highly active in creating large-sized genome deletions in human cells. Here, we use four cryoelectron microscopy snapshots to define its RNA-guided DNA binding and cleavage mechanisms in high resolution. The non-target DNA strand (NTS) is accommodated by I-C Cascade in a continuous binding groove along the juxtaposed Cas11 subunits. Binding of Cas3 further traps a flexible bulge in NTS, enabling NTS nicking. We identified two anti-CRISPR proteins AcrIC8 and AcrIC9 that strongly inhibit Neisseria lactamica I-C function. Structural analysis showed that AcrIC8 inhibits PAM recognition through allosteric inhibition, whereas AcrIC9 achieves so through direct competition. Both Acrs potently inhibit I-C-mediated genome editing and transcriptional modulation in human cells, providing the first off-switches for type I CRISPR eukaryotic genome engineering.


Subject(s)
CRISPR-Associated Proteins , Gene Editing , Humans , CRISPR-Cas Systems , Cryoelectron Microscopy , CRISPR-Associated Proteins/metabolism , DNA/metabolism , RNA
9.
Mol Cell ; 84(16): 3141-3153.e5, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39047725

ABSTRACT

The metagenome-derived type I-E and type I-F variant CRISPR-associated complex for antiviral defense (Cascade) complexes, fused with HNH domains, precisely cleave target DNA, representing recently identified genome editing tools. However, the underlying working mechanisms remain unknown. Here, structures of type I-FHNH and I-EHNH Cascade complexes at different states are reported. In type I-FHNH Cascade, Cas8fHNH loosely attaches to Cascade head and is adjacent to the 5' end of the target single-stranded DNA (ssDNA). Formation of the full R-loop drives the Cascade head to move outward, allowing Cas8fHNH to detach and rotate ∼150° to accommodate target ssDNA for cleavage. In type I-EHNH Cascade, Cas5eHNH domain is adjacent to the 5' end of the target ssDNA. Full crRNA-target pairing drives the lift of the Cascade head, widening the substrate channel for target ssDNA entrance. Altogether, these analyses into both complexes revealed that crRNA-guided positioning of target DNA and target DNA-induced HNH unlocking are two key factors for their site-specific cleavage of target DNA.


Subject(s)
CRISPR-Associated Proteins , CRISPR-Cas Systems , DNA Cleavage , DNA, Single-Stranded , Gene Editing , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/genetics , CRISPR-Associated Proteins/metabolism , CRISPR-Associated Proteins/genetics , Gene Editing/methods , R-Loop Structures/genetics , Cryoelectron Microscopy
10.
Mol Cell ; 84(16): 3154-3162.e5, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39111310

ABSTRACT

Canonical prokaryotic type I CRISPR-Cas adaptive immune systems contain a multicomponent effector complex called Cascade, which degrades large stretches of DNA via Cas3 helicase-nuclease activity. Recently, a highly precise subtype I-F1 CRISPR-Cas system (HNH-Cascade) was found that lacks Cas3, the absence of which is compensated for by the insertion of an HNH endonuclease domain in the Cas8 Cascade component. Here, we describe the cryo-EM structure of Selenomonas sp. HNH-Cascade (SsCascade) in complex with target DNA and characterize its mechanism of action. The Cascade scaffold is complemented by the HNH domain, creating a ring-like structure in which the unwound target DNA is precisely cleaved. This structure visualizes a unique hybrid of two extensible biological systems-Cascade, an evolutionary platform for programmable DNA effectors, and an HNH nuclease, an adaptive domain with a spectrum of enzymatic activity.


Subject(s)
CRISPR-Associated Proteins , CRISPR-Cas Systems , Cryoelectron Microscopy , DNA Cleavage , CRISPR-Associated Proteins/metabolism , CRISPR-Associated Proteins/chemistry , CRISPR-Associated Proteins/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Models, Molecular , DNA/metabolism , DNA/genetics , DNA/chemistry , Protein Domains , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , Structure-Activity Relationship , Clustered Regularly Interspaced Short Palindromic Repeats , Protein Binding
11.
Mol Cell ; 83(7): 1093-1108.e8, 2023 04 06.
Article in English | MEDLINE | ID: mdl-36863348

ABSTRACT

The glucagon-PKA signal is generally believed to control hepatic gluconeogenesis via the CREB transcription factor. Here we uncovered a distinct function of this signal in directly stimulating histone phosphorylation for gluconeogenic gene regulation in mice. In the fasting state, CREB recruited activated PKA to regions near gluconeogenic genes, where PKA phosphorylated histone H3 serine 28 (H3S28ph). H3S28ph, recognized by 14-3-3ζ, promoted recruitment of RNA polymerase II and transcriptional stimulation of gluconeogenic genes. In contrast, in the fed state, more PP2A was found near gluconeogenic genes, which counteracted PKA by dephosphorylating H3S28ph and repressing transcription. Importantly, ectopic expression of phosphomimic H3S28 efficiently restored gluconeogenic gene expression when liver PKA or CREB was depleted. These results together highlight a different functional scheme in regulating gluconeogenesis by the glucagon-PKA-CREB-H3S28ph cascade, in which the hormone signal is transmitted to chromatin for rapid and efficient gluconeogenic gene activation.


Subject(s)
Glucagon , Gluconeogenesis , Animals , Mice , Gluconeogenesis/genetics , Glucagon/metabolism , Histones/metabolism , Phosphorylation , 14-3-3 Proteins/metabolism , Liver/metabolism , Fasting/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism
12.
Mol Cell ; 83(11): 1827-1838.e6, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37267904

ABSTRACT

CRISPR-associated transposons (CASTs) are natural RNA-directed transposition systems. We demonstrate that transposon protein TniQ plays a central role in promoting R-loop formation by RNA-guided DNA-targeting modules. TniQ residues, proximal to CRISPR RNA (crRNA), are required for recognizing different crRNA categories, revealing an unappreciated role of TniQ to direct transposition into different classes of crRNA targets. To investigate adaptations allowing CAST elements to utilize attachment sites inaccessible to CRISPR-Cas surveillance complexes, we compared and contrasted PAM sequence requirements in both I-F3b CAST and I-F1 CRISPR-Cas systems. We identify specific amino acids that enable a wider range of PAM sequences to be accommodated in I-F3b CAST elements compared with I-F1 CRISPR-Cas, enabling CAST elements to access attachment sites as sequences drift and evade host surveillance. Together, this evidence points to the central role of TniQ in facilitating the acquisition of CRISPR effector complexes for RNA-guided DNA transposition.


Subject(s)
CRISPR-Associated Proteins , RNA , DNA/genetics , CRISPR-Cas Systems , CRISPR-Associated Proteins/genetics
13.
Mol Cell ; 82(4): 852-867.e5, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35051351

ABSTRACT

Leading CRISPR-Cas technologies employ Cas9 and Cas12 enzymes that generate RNA-guided dsDNA breaks. Yet, the most abundant microbial adaptive immune systems, Type I CRISPRs, are under-exploited for eukaryotic applications. Here, we report the adoption of a minimal CRISPR-Cas3 from Neisseria lactamica (Nla) type I-C system to create targeted large deletions in the human genome. RNP delivery of its processive Cas3 nuclease and target recognition complex Cascade can confer ∼95% editing efficiency. Unexpectedly, NlaCascade assembly in bacteria requires internal translation of a hidden component Cas11 from within the cas8 gene. Furthermore, expressing a separately encoded NlaCas11 is the key to enable plasmid- and mRNA-based editing in human cells. Finally, we demonstrate that supplying cas11 is a universal strategy to systematically implement divergent I-C, I-D, and I-B CRISPR-Cas3 editors with compact sizes, distinct PAM preferences, and guide orthogonality. These findings greatly expand our ability to engineer long-range genome edits.


Subject(s)
Bacterial Proteins/genetics , CRISPR-Associated Proteins/genetics , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Deletion , Gene Editing , Genome, Human , Neisseria lactamica/genetics , Bacterial Proteins/metabolism , CRISPR-Associated Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Neisseria lactamica/enzymology , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism
14.
Mol Cell ; 82(3): 616-628.e5, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35051352

ABSTRACT

Canonical CRISPR-Cas systems utilize RNA-guided nucleases for targeted cleavage of foreign nucleic acids, whereas some nuclease-deficient CRISPR-Cas complexes have been repurposed to direct the insertion of Tn7-like transposons. Here, we established a bioinformatic and experimental pipeline to comprehensively explore the diversity of Type I-F CRISPR-associated transposons. We report DNA integration for 20 systems and identify a highly active subset that exhibits complete orthogonality in transposon DNA mobilization. We reveal the modular nature of CRISPR-associated transposons by exploring the horizontal acquisition of targeting modules and by characterizing a system that encodes both a programmable, RNA-dependent pathway, and a fixed, RNA-independent pathway. Finally, we analyzed transposon-encoded cargo genes and found the striking presence of anti-phage defense systems, suggesting a role in transmitting innate immunity between bacteria. Collectively, this study substantially advances our biological understanding of CRISPR-associated transposon function and expands the suite of RNA-guided transposases for programmable, large-scale genome engineering.


Subject(s)
Bacterial Proteins/genetics , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , DNA Transposable Elements/genetics , DNA, Bacterial/genetics , Escherichia coli/genetics , Evolution, Molecular , Transposases/genetics , Bacterial Proteins/metabolism , DNA, Bacterial/metabolism , Escherichia coli/immunology , Escherichia coli/metabolism , Gene Editing , Gene Expression Regulation, Bacterial , Genetic Variation , Immunity, Innate , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism , Transposases/metabolism
15.
Mol Cell ; 82(6): 1210-1224.e6, 2022 03 17.
Article in English | MEDLINE | ID: mdl-35216669

ABSTRACT

CRISPR-Cas biology and technologies have been largely shaped to date by the characterization and use of single-effector nucleases. By contrast, multi-subunit effectors dominate natural systems, represent emerging technologies, and were recently associated with RNA-guided DNA transposition. This disconnect stems from the challenge of working with multiple protein subunits in vitro and in vivo. Here, we apply cell-free transcription-translation (TXTL) systems to radically accelerate the characterization of multi-subunit CRISPR effectors and transposons. Numerous DNA constructs can be combined in one TXTL reaction, yielding defined biomolecular readouts in hours. Using TXTL, we mined phylogenetically diverse I-E effectors, interrogated extensively self-targeting I-C and I-F systems, and elucidated targeting rules for I-B and I-F CRISPR transposons using only DNA-binding components. We further recapitulated DNA transposition in TXTL, which helped reveal a distinct branch of I-B CRISPR transposons. These capabilities will facilitate the study and exploitation of the broad yet underexplored diversity of CRISPR-Cas systems and transposons.


Subject(s)
CRISPR-Cas Systems , Endonucleases , Cell-Free System/metabolism , DNA/genetics , Endonucleases/genetics , RNA/metabolism
16.
Mol Cell ; 81(22): 4605-4621.e11, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34582793

ABSTRACT

G-protein-coupled receptors (GPCRs), also known as seven transmembrane receptors (7TMRs), typically interact with two distinct signal-transducers, i.e., G proteins and ß-arrestins (ßarrs). Interestingly, there are some non-canonical 7TMRs that lack G protein coupling but interact with ßarrs, although an understanding of their transducer coupling preference, downstream signaling, and structural mechanism remains elusive. Here, we characterize two such non-canonical 7TMRs, namely, the decoy D6 receptor (D6R) and the complement C5a receptor subtype 2 (C5aR2), in parallel with their canonical GPCR counterparts. We discover that D6R and C5aR2 efficiently couple to ßarrs, exhibit distinct engagement of GPCR kinases (GRKs), and activate non-canonical downstream signaling pathways. We also observe that ßarrs adopt distinct conformations for D6R and C5aR2, compared to their canonical GPCR counterparts, in response to common natural agonists. Our study establishes D6R and C5aR2 as ßarr-coupled 7TMRs and provides key insights into their regulation and signaling with direct implication for biased agonism.


Subject(s)
Cell Membrane/metabolism , Protein Conformation , Signal Transduction , beta-Arrestins/chemistry , Animals , GTP-Binding Proteins/chemistry , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Protein Binding , Protein Domains , Protein Structure, Secondary , Protein Transport , Receptor, Anaphylatoxin C5a/metabolism
17.
Mol Cell ; 80(6): 971-979.e7, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33248026

ABSTRACT

CRISPR-Cas adaptive immune systems provide prokaryotes with defense against viruses by degradation of specific invading nucleic acids. Despite advances in the biotechnological exploitation of select systems, multiple CRISPR-Cas types remain uncharacterized. Here, we investigated the previously uncharacterized type I-D interference complex and revealed that it is a genetic and structural hybrid with similarity to both type I and type III systems. Surprisingly, formation of the functional complex required internal in-frame translation of small subunits from within the large subunit gene. We further show that internal translation to generate small subunits is widespread across diverse type I-D, I-B, and I-C systems, which account for roughly one quarter of CRISPR-Cas systems. Our work reveals the unexpected expansion of protein coding potential from within single cas genes, which has important implications for understanding CRISPR-Cas function and evolution.


Subject(s)
Adaptive Immunity/genetics , CRISPR-Associated Proteins/genetics , CRISPR-Cas Systems/genetics , Evolution, Molecular , CRISPR-Associated Proteins/immunology , Prokaryotic Cells/immunology , Prokaryotic Cells/virology , Protein Biosynthesis , Viruses/immunology
18.
Mol Cell ; 77(1): 39-50.e10, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31735642

ABSTRACT

CRISPR-Cas systems encode RNA-guided surveillance complexes to find and cleave invading DNA elements. While it is thought that invaders are neutralized minutes after cell entry, the mechanism and kinetics of target search and its impact on CRISPR protection levels have remained unknown. Here, we visualize individual Cascade complexes in a native type I CRISPR-Cas system. We uncover an exponential relation between Cascade copy number and CRISPR interference levels, pointing to a time-driven arms race between invader replication and target search, in which 20 Cascade complexes provide 50% protection. Driven by PAM-interacting subunit Cas8e, Cascade spends half its search time rapidly probing DNA (∼30 ms) in the nucleoid. We further demonstrate that target DNA transcription and CRISPR arrays affect the integrity of Cascade and affect CRISPR interference. Our work establishes the mechanism of cellular DNA surveillance by Cascade that allows the timely detection of invading DNA in a crowded, DNA-packed environment.


Subject(s)
Bacteria/genetics , CRISPR-Associated Proteins/genetics , CRISPR-Cas Systems/genetics , DNA/genetics , RNA, Guide, Kinetoplastida/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , DNA Replication/genetics , Gene Dosage/genetics
19.
Mol Cell ; 74(1): 132-142.e5, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30872121

ABSTRACT

Bacteria and archaea have evolved sophisticated adaptive immune systems that rely on CRISPR RNA (crRNA)-guided detection and nuclease-mediated elimination of invading nucleic acids. Here, we present the cryo-electron microscopy (cryo-EM) structure of the type I-F crRNA-guided surveillance complex (Csy complex) from Pseudomonas aeruginosa bound to a double-stranded DNA target. Comparison of this structure to previously determined structures of this complex reveals a ∼180-degree rotation of the C-terminal helical bundle on the "large" Cas8f subunit. We show that the double-stranded DNA (dsDNA)-induced conformational change in Cas8f exposes a Cas2/3 "nuclease recruitment helix" that is structurally homologous to a virally encoded anti-CRISPR protein (AcrIF3). Structural homology between Cas8f and AcrIF3 suggests that AcrIF3 is a mimic of the Cas8f nuclease recruitment helix.


Subject(s)
Bacterial Proteins/metabolism , CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , DNA, Bacterial/metabolism , Molecular Mimicry , Pseudomonas aeruginosa/enzymology , RNA, Bacterial/metabolism , RNA, Guide, Kinetoplastida/metabolism , Viral Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/immunology , CRISPR-Associated Proteins/chemistry , CRISPR-Associated Proteins/genetics , CRISPR-Associated Proteins/immunology , Cryoelectron Microscopy , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Models, Molecular , Nucleic Acid Conformation , Protein Conformation , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/immunology , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , RNA, Guide, Kinetoplastida/chemistry , RNA, Guide, Kinetoplastida/genetics , Structure-Activity Relationship , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/immunology
20.
Mol Cell ; 74(5): 936-950.e5, 2019 06 06.
Article in English | MEDLINE | ID: mdl-30975459

ABSTRACT

CRISPR-Cas systems enable microbial adaptive immunity and provide eukaryotic genome editing tools. These tools employ a single effector enzyme of type II or V CRISPR to generate RNA-guided, precise genome breaks. Here we demonstrate the feasibility of using type I CRISPR-Cas to effectively introduce a spectrum of long-range chromosomal deletions with a single RNA guide in human embryonic stem cells and HAP1 cells. Type I CRISPR systems rely on the multi-subunit ribonucleoprotein (RNP) complex Cascade to identify DNA targets and on the helicase-nuclease enzyme Cas3 to degrade DNA processively. With RNP delivery of T. fusca Cascade and Cas3, we obtained 13%-60% editing efficiency. Long-range PCR-based and high-throughput-sequencing-based lesion analyses reveal that a variety of deletions, ranging from a few hundred base pairs to 100 kilobases, are created upstream of the target site. These results highlight the potential utility of type I CRISPR-Cas for long-range genome manipulations and deletion screens in eukaryotes.


Subject(s)
CRISPR-Cas Systems/genetics , Human Embryonic Stem Cells , RNA, Guide, Kinetoplastida/genetics , Sequence Deletion/genetics , Endonucleases/chemistry , Endonucleases/genetics , Escherichia coli/genetics , Gene Editing/methods , Genome, Human/genetics , Genomics , Humans , Ribonucleoproteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL