Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Cell ; 184(2): 323-333.e9, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33306959

ABSTRACT

The December 2019 outbreak of a novel respiratory virus, SARS-CoV-2, has become an ongoing global pandemic due in part to the challenge of identifying symptomatic, asymptomatic, and pre-symptomatic carriers of the virus. CRISPR diagnostics can augment gold-standard PCR-based testing if they can be made rapid, portable, and accurate. Here, we report the development of an amplification-free CRISPR-Cas13a assay for direct detection of SARS-CoV-2 from nasal swab RNA that can be read with a mobile phone microscope. The assay achieved ∼100 copies/µL sensitivity in under 30 min of measurement time and accurately detected pre-extracted RNA from a set of positive clinical samples in under 5 min. We combined crRNAs targeting SARS-CoV-2 RNA to improve sensitivity and specificity and directly quantified viral load using enzyme kinetics. Integrated with a reader device based on a mobile phone, this assay has the potential to enable rapid, low-cost, point-of-care screening for SARS-CoV-2.


Subject(s)
COVID-19 Nucleic Acid Testing/methods , Cell Phone/instrumentation , Optical Imaging/methods , RNA, Viral/analysis , Viral Load/methods , Animals , COVID-19 Nucleic Acid Testing/economics , COVID-19 Nucleic Acid Testing/instrumentation , CRISPR-Cas Systems , Cell Line , Coronavirus Nucleocapsid Proteins/genetics , Humans , Nasopharynx/virology , Optical Imaging/instrumentation , Phosphoproteins/genetics , Point-of-Care Testing , RNA Interference , RNA, Viral/genetics , Sensitivity and Specificity , Viral Load/economics , Viral Load/instrumentation
2.
Cell ; 175(2): 488-501.e22, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30270045

ABSTRACT

Detection of viruses by innate immune sensors induces protective antiviral immunity. The viral DNA sensor cyclic GMP-AMP synthase (cGAS) is necessary for detection of HIV by human dendritic cells and macrophages. However, synthesis of HIV DNA during infection is not sufficient for immune activation. The capsid protein, which associates with viral DNA, has a pivotal role in enabling cGAS-mediated immune activation. We now find that NONO is an essential sensor of the HIV capsid in the nucleus. NONO protein directly binds capsid with higher affinity for weakly pathogenic HIV-2 than highly pathogenic HIV-1. Upon infection, NONO is essential for cGAS activation by HIV and cGAS association with HIV DNA in the nucleus. NONO recognizes a conserved region in HIV capsid with limited tolerance for escape mutations. Detection of nuclear viral capsid by NONO to promote DNA sensing by cGAS reveals an innate strategy to achieve distinction of viruses from self in the nucleus.


Subject(s)
Capsid Proteins/immunology , Nuclear Matrix-Associated Proteins/immunology , Nuclear Matrix-Associated Proteins/physiology , Octamer Transcription Factors/immunology , Octamer Transcription Factors/physiology , RNA-Binding Proteins/immunology , RNA-Binding Proteins/physiology , Capsid/metabolism , Capsid Proteins/metabolism , Capsid Proteins/physiology , Cell Nucleus/metabolism , DNA, Viral/genetics , DNA, Viral/immunology , DNA-Binding Proteins , Dendritic Cells/immunology , HIV Infections/immunology , HIV-1/genetics , HIV-1/immunology , HIV-2/genetics , HIV-2/immunology , Host-Pathogen Interactions , Humans , Immunity, Innate/immunology , Macrophages/immunology , Membrane Proteins/metabolism , Nuclear Matrix-Associated Proteins/metabolism , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/physiology , RNA-Binding Proteins/metabolism , Signal Transduction/immunology
3.
Cell ; 170(6): 1224-1233.e15, 2017 Sep 07.
Article in English | MEDLINE | ID: mdl-28844692

ABSTRACT

CRISPR-Cas9 proteins function within bacterial immune systems to target and destroy invasive DNA and have been harnessed as a robust technology for genome editing. Small bacteriophage-encoded anti-CRISPR proteins (Acrs) can inactivate Cas9, providing an efficient off switch for Cas9-based applications. Here, we show that two Acrs, AcrIIC1 and AcrIIC3, inhibit Cas9 by distinct strategies. AcrIIC1 is a broad-spectrum Cas9 inhibitor that prevents DNA cutting by multiple divergent Cas9 orthologs through direct binding to the conserved HNH catalytic domain of Cas9. A crystal structure of an AcrIIC1-Cas9 HNH domain complex shows how AcrIIC1 traps Cas9 in a DNA-bound but catalytically inactive state. By contrast, AcrIIC3 blocks activity of a single Cas9 ortholog and induces Cas9 dimerization while preventing binding to the target DNA. These two orthogonal mechanisms allow for separate control of Cas9 target binding and cleavage and suggest applications to allow DNA binding while preventing DNA cutting by Cas9.


Subject(s)
CRISPR-Cas Systems , Endonucleases/antagonists & inhibitors , Viral Proteins/metabolism , Amino Acid Sequence , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacteriophages/genetics , Bacteriophages/metabolism , Endonucleases/chemistry , Endonucleases/genetics , Endonucleases/metabolism , Evolution, Molecular , HEK293 Cells , Humans , Protein Domains , Sequence Alignment
4.
Nucleic Acids Res ; 52(D1): D590-D596, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-37889041

ABSTRACT

CRISPR-Cas enzymes enable RNA-guided bacterial immunity and are widely used for biotechnological applications including genome editing. In particular, the Class 2 CRISPR-associated enzymes (Cas9, Cas12 and Cas13 families), have been deployed for numerous research, clinical and agricultural applications. However, the immense genetic and biochemical diversity of these proteins in the public domain poses a barrier for researchers seeking to leverage their activities. We present CasPEDIA (http://caspedia.org), the Cas Protein Effector Database of Information and Assessment, a curated encyclopedia that integrates enzymatic classification for hundreds of different Cas enzymes across 27 phylogenetic groups spanning the Cas9, Cas12 and Cas13 families, as well as evolutionarily related IscB and TnpB proteins. All enzymes in CasPEDIA were annotated with a standard workflow based on their primary nuclease activity, target requirements and guide-RNA design constraints. Our functional classification scheme, CasID, is described alongside current phylogenetic classification, allowing users to search related orthologs by enzymatic function and sequence similarity. CasPEDIA is a comprehensive data portal that summarizes and contextualizes enzymatic properties of widely used Cas enzymes, equipping users with valuable resources to foster biotechnological development. CasPEDIA complements phylogenetic Cas nomenclature and enables researchers to leverage the multi-faceted nucleic-acid targeting rules of diverse Class 2 Cas enzymes.


Subject(s)
CRISPR-Associated Proteins , CRISPR-Cas Systems , Databases, Genetic , Endodeoxyribonucleases , CRISPR-Cas Systems/genetics , Phylogeny , CRISPR-Associated Proteins/chemistry , CRISPR-Associated Proteins/classification , CRISPR-Associated Proteins/genetics , Endodeoxyribonucleases/chemistry , Endodeoxyribonucleases/classification , Endodeoxyribonucleases/genetics , Encyclopedias as Topic
5.
Nature ; 568(7752): E8-E10, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30944483

ABSTRACT

In this Article, owing to issues with the first 30 nucleotides of the sgRNA, which run in the opposite direction, corrections have been made to the Protein Data Bank (PDB) accessions in the 'Data availability' section, and this also affects Figs. 3, 4, Extended Data Fig. 6, Supplementary Table 1 and Supplementary Video 1. The original Article has been corrected online. See the accompanying Amendment for further details.

6.
Nature ; 566(7743): 218-223, 2019 02.
Article in English | MEDLINE | ID: mdl-30718774

ABSTRACT

The RNA-guided CRISPR-associated (Cas) proteins Cas9 and Cas12a provide adaptive immunity against invading nucleic acids, and function as powerful tools for genome editing in a wide range of organisms. Here we reveal the underlying mechanisms of a third, fundamentally distinct RNA-guided genome-editing platform named CRISPR-CasX, which uses unique structures for programmable double-stranded DNA binding and cleavage. Biochemical and in vivo data demonstrate that CasX is active for Escherichia coli and human genome modification. Eight cryo-electron microscopy structures of CasX in different states of assembly with its guide RNA and double-stranded DNA substrates reveal an extensive RNA scaffold and a domain required for DNA unwinding. These data demonstrate how CasX activity arose through convergent evolution to establish an enzyme family that is functionally separate from both Cas9 and Cas12a.


Subject(s)
CRISPR-Associated Proteins/classification , CRISPR-Associated Proteins/ultrastructure , CRISPR-Cas Systems/genetics , Gene Editing , CRISPR-Associated Proteins/chemistry , CRISPR-Associated Proteins/metabolism , Cryoelectron Microscopy , DNA/chemistry , DNA/metabolism , DNA/ultrastructure , DNA Cleavage , Escherichia coli/genetics , Evolution, Molecular , Gene Silencing , Genome, Bacterial/genetics , Genome, Human/genetics , Humans , Models, Molecular , Nucleic Acid Conformation , Protein Domains , RNA, Guide, Kinetoplastida/metabolism
7.
Mol Cell ; 66(3): 373-383.e3, 2017 May 04.
Article in English | MEDLINE | ID: mdl-28475872

ABSTRACT

CRISPR adaptive immunity pathways protect prokaryotic cells against foreign nucleic acids using CRISPR RNA (crRNA)-guided nucleases. In type VI-A CRISPR-Cas systems, the signature protein Cas13a (formerly C2c2) contains two separate ribonuclease activities that catalyze crRNA maturation and ssRNA degradation. The Cas13a protein family occurs across different bacterial phyla and varies widely in both protein sequence and corresponding crRNA sequence conservation. Although grouped phylogenetically together, we show that the Cas13a enzyme family comprises two distinct functional groups that recognize orthogonal sets of crRNAs and possess different ssRNA cleavage specificities. These functional distinctions could not be bioinformatically predicted, suggesting more subtle co-evolution of Cas13a enzymes. Additionally, we find that Cas13a pre-crRNA processing is not essential for ssRNA cleavage, although it enhances ssRNA targeting for crRNAs encoded internally within the CRISPR array. We define two Cas13a protein subfamilies that can operate in parallel for RNA detection both in bacteria and for diagnostic applications.


Subject(s)
Bacterial Proteins/metabolism , CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Escherichia coli/enzymology , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional , RNA, Bacterial/metabolism , Ribonucleases/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , CRISPR-Associated Proteins/chemistry , CRISPR-Associated Proteins/genetics , Escherichia coli/genetics , Nucleic Acid Conformation , Phylogeny , RNA Precursors/chemistry , RNA Precursors/genetics , RNA Stability , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , Ribonucleases/chemistry , Ribonucleases/genetics , Structure-Activity Relationship , Substrate Specificity
8.
Nucleic Acids Res ; 50(1): 522-535, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34904671

ABSTRACT

The Drosophila behaviour/human splicing (DBHS) proteins are a family of RNA/DNA binding cofactors liable for a range of cellular processes. DBHS proteins include the non-POU domain-containing octamer-binding protein (NONO) and paraspeckle protein component 1 (PSPC1), proteins capable of forming combinatorial dimers. Here, we describe the crystal structures of the human NONO and PSPC1 homodimers, representing uncharacterized DBHS dimerization states. The structures reveal a set of conserved contacts and structural plasticity within the dimerization interface that provide a rationale for dimer selectivity between DBHS paralogues. In addition, solution X-ray scattering and accompanying biochemical experiments describe a mechanism of cooperative RNA recognition by the NONO homodimer. Nucleic acid binding is reliant on RRM1, and appears to be affected by the orientation of RRM1, influenced by a newly identified 'ß-clasp' structure. Our structures shed light on the molecular determinants for DBHS homo- and heterodimerization and provide a basis for understanding how DBHS proteins cooperatively recognize a broad spectrum of RNA targets.


Subject(s)
DNA-Binding Proteins/metabolism , RNA-Binding Proteins/metabolism , RNA/metabolism , Dimerization , Humans , Models, Molecular , Protein Conformation , RNA Splicing
9.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Article in English | MEDLINE | ID: mdl-33758097

ABSTRACT

Most glioblastomas (GBMs) achieve cellular immortality by acquiring a mutation in the telomerase reverse transcriptase (TERT) promoter. TERT promoter mutations create a binding site for a GA binding protein (GABP) transcription factor complex, whose assembly at the promoter is associated with TERT reactivation and telomere maintenance. Here, we demonstrate increased binding of a specific GABPB1L-isoform-containing complex to the mutant TERT promoter. Furthermore, we find that TERT promoter mutant GBM cells, unlike wild-type cells, exhibit a critical near-term dependence on GABPB1L for proliferation, notably also posttumor establishment in vivo. Up-regulation of the protein paralogue GABPB2, which is normally expressed at very low levels, can rescue this dependence. More importantly, when combined with frontline temozolomide (TMZ) chemotherapy, inducible GABPB1L knockdown and the associated TERT reduction led to an impaired DNA damage response that resulted in profoundly reduced growth of intracranial GBM tumors. Together, these findings provide insights into the mechanism of cancer-specific TERT regulation, uncover rapid effects of GABPB1L-mediated TERT suppression in GBM maintenance, and establish GABPB1L inhibition in combination with chemotherapy as a therapeutic strategy for TERT promoter mutant GBM.


Subject(s)
Brain Neoplasms/genetics , GA-Binding Protein Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Telomerase/genetics , Animals , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Astrocytes , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , DNA Damage/drug effects , Drug Resistance, Neoplasm/genetics , Female , GA-Binding Protein Transcription Factor/genetics , Gene Knockdown Techniques , Gene Knockout Techniques , Glioblastoma/drug therapy , Glioblastoma/pathology , HEK293 Cells , Humans , Mice , Mutation , Promoter Regions, Genetic/genetics , Protein Isoforms/metabolism , Temozolomide/pharmacology , Xenograft Model Antitumor Assays
10.
J Biol Chem ; 298(11): 102563, 2022 11.
Article in English | MEDLINE | ID: mdl-36209820

ABSTRACT

RNA-binding proteins of the DBHS (Drosophila Behavior Human Splicing) family, NONO, SFPQ, and PSPC1 have numerous roles in genome stability and transcriptional and posttranscriptional regulation. Critical to DBHS activity is their recruitment to distinct subnuclear locations, for example, paraspeckle condensates, where DBHS proteins bind to the long noncoding RNA NEAT1 in the first essential step in paraspeckle formation. To carry out their diverse roles, DBHS proteins form homodimers and heterodimers, but how this dimerization influences DBHS localization and function is unknown. Here, we present an inducible GFP-NONO stable cell line and use it for live-cell 3D-structured illumination microscopy, revealing paraspeckles with dynamic, twisted elongated structures. Using siRNA knockdowns, we show these labeled paraspeckles consist of GFP-NONO/endogenous SFPQ dimers and that GFP-NONO localization to paraspeckles depends on endogenous SFPQ. Using purified proteins, we confirm that partner swapping between NONO and SFPQ occurs readily in vitro. Crystallographic analysis of the NONO-SFPQ heterodimer reveals conformational differences to the other DBHS dimer structures, which may contribute to partner preference, RNA specificity, and subnuclear localization. Thus overall, our study suggests heterodimer partner availability is crucial for NONO subnuclear distribution and helps explain the complexity of both DBHS protein and paraspeckle dynamics through imaging and structural approaches.


Subject(s)
Paraspeckles , RNA, Long Noncoding , Humans , Dimerization , RNA-Binding Proteins/metabolism , Gene Expression Regulation , RNA, Long Noncoding/genetics
11.
Nat Chem Biol ; 17(1): 10-19, 2021 01.
Article in English | MEDLINE | ID: mdl-33328654

ABSTRACT

Many bacterial and archaeal organisms use clustered regularly interspaced short palindromic repeats-CRISPR associated (CRISPR-Cas) systems to defend themselves from mobile genetic elements. These CRISPR-Cas systems are classified into six types based on their composition and mechanism. CRISPR-Cas enzymes are widely used for genome editing and offer immense therapeutic opportunity to treat genetic diseases. To realize their full potential, it is important to control the timing, duration, efficiency and specificity of CRISPR-Cas enzyme activities. In this Review we discuss the mechanisms of natural CRISPR-Cas regulatory biomolecules and engineering strategies that enhance or inhibit CRISPR-Cas immunity by altering enzyme function. We also discuss the potential applications of these CRISPR regulators and highlight unanswered questions about their evolution and purpose in nature.


Subject(s)
Archaea/genetics , Bacteria/genetics , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Expression Regulation, Archaeal , Gene Expression Regulation, Bacterial , Antibiosis/genetics , Archaea/metabolism , Archaea/virology , Bacteria/metabolism , Bacteria/virology , Bacteriophages/genetics , Bacteriophages/growth & development , Bacteriophages/metabolism , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , Gene Editing/methods , Genetic Engineering/methods , Humans , Interspersed Repetitive Sequences , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism
12.
Nat Chem Biol ; 17(9): 982-988, 2021 09.
Article in English | MEDLINE | ID: mdl-34354262

ABSTRACT

Direct, amplification-free detection of RNA has the potential to transform molecular diagnostics by enabling simple on-site analysis of human or environmental samples. CRISPR-Cas nucleases offer programmable RNA-guided RNA recognition that triggers cleavage and release of a fluorescent reporter molecule, but long reaction times hamper their detection sensitivity and speed. Here, we show that unrelated CRISPR nucleases can be deployed in tandem to provide both direct RNA sensing and rapid signal generation, thus enabling robust detection of ~30 molecules per µl of RNA in 20 min. Combining RNA-guided Cas13 and Csm6 with a chemically stabilized activator creates a one-step assay that can detect severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA extracted from respiratory swab samples with quantitative reverse transcriptase PCR (qRT-PCR)-derived cycle threshold (Ct) values up to 33, using a compact detector. This Fast Integrated Nuclease Detection In Tandem (FIND-IT) approach enables sensitive, direct RNA detection in a format that is amenable to point-of-care infection diagnosis as well as to a wide range of other diagnostic or research applications.


Subject(s)
COVID-19/genetics , CRISPR-Cas Systems/genetics , RNA, Viral/genetics , SARS-CoV-2/genetics , Humans , Reverse Transcriptase Polymerase Chain Reaction
13.
Nucleic Acids Res ; 48(9): 4698-4708, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32286628

ABSTRACT

The increasing use of CRISPR-Cas9 in medicine, agriculture, and synthetic biology has accelerated the drive to discover new CRISPR-Cas inhibitors as potential mechanisms of control for gene editing applications. Many anti-CRISPRs have been found that inhibit the CRISPR-Cas adaptive immune system. However, comparing all currently known anti-CRISPRs does not reveal a shared set of properties for facile bioinformatic identification of new anti-CRISPR families. Here, we describe AcRanker, a machine learning based method to aid direct identification of new potential anti-CRISPRs using only protein sequence information. Using a training set of known anti-CRISPRs, we built a model based on XGBoost ranking. We then applied AcRanker to predict candidate anti-CRISPRs from predicted prophage regions within self-targeting bacterial genomes and discovered two previously unknown anti-CRISPRs: AcrllA20 (ML1) and AcrIIA21 (ML8). We show that AcrIIA20 strongly inhibits Streptococcus iniae Cas9 (SinCas9) and weakly inhibits Streptococcus pyogenes Cas9 (SpyCas9). We also show that AcrIIA21 inhibits SpyCas9, Streptococcus aureus Cas9 (SauCas9) and SinCas9 with low potency. The addition of AcRanker to the anti-CRISPR discovery toolkit allows researchers to directly rank potential anti-CRISPR candidate genes for increased speed in testing and validation of new anti-CRISPRs. A web server implementation for AcRanker is available online at http://acranker.pythonanywhere.com/.


Subject(s)
Bacterial Proteins/genetics , CRISPR-Associated Protein 9/antagonists & inhibitors , Machine Learning , Bacterial Proteins/chemistry , Prophages/genetics , Proteome , Sequence Analysis, Protein , Streptococcus/enzymology , Streptococcus/genetics
14.
Plant Cell ; 29(3): 461-473, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28298518

ABSTRACT

Bowman-Birk Inhibitors (BBIs) are a well-known family of plant protease inhibitors first described 70 years ago. BBIs are known only in the legume (Fabaceae) and cereal (Poaceae) families, but peptides that mimic their trypsin-inhibitory loops exist in sunflowers (Helianthus annuus) and frogs. The disparate biosynthetic origins and distant phylogenetic distribution implies these loops evolved independently, but their structural similarity suggests a common ancestor. Targeted bioinformatic searches for the BBI inhibitory loop discovered highly divergent BBI-like sequences in the seedless, vascular spikemoss Selaginella moellendorffii Using de novo transcriptomics, we confirmed expression of five transcripts in S. moellendorffii whose encoded proteins share homology with BBI inhibitory loops. The most highly expressed, BBI3, encodes a protein that inhibits trypsin. We needed to mutate two lysine residues to abolish trypsin inhibition, suggesting BBI3's mechanism of double-headed inhibition is shared with BBIs from angiosperms. As Selaginella belongs to the lycopod plant lineage, which diverged ∼200 to 230 million years before the common ancestor of angiosperms, its BBI-like proteins imply there was a common ancestor for legume and cereal BBIs. Indeed, we discovered BBI sequences in six angiosperm families outside the Fabaceae and Poaceae. These findings provide the evolutionary missing links between the well-known legume and cereal BBI gene families.


Subject(s)
Selaginellaceae/metabolism , Trypsin Inhibitors/metabolism , Evolution, Molecular , Fabaceae/metabolism , Magnoliopsida/metabolism , Plant Proteins/metabolism , Poaceae/metabolism
15.
Methods ; 212: 10-11, 2023 04.
Article in English | MEDLINE | ID: mdl-36792042

Subject(s)
RNA , RNA/genetics
17.
Nucleic Acids Res ; 44(9): 3989-4004, 2016 05 19.
Article in English | MEDLINE | ID: mdl-27084935

ABSTRACT

Nuclear proteins are often given a concise title that captures their function, such as 'transcription factor,' 'polymerase' or 'nuclear-receptor.' However, for members of the Drosophila behavior/human splicing (DBHS) protein family, no such clean-cut title exists. DBHS proteins are frequently identified engaging in almost every step of gene regulation, including but not limited to, transcriptional regulation, RNA processing and transport, and DNA repair. Herein, we present a coherent picture of DBHS proteins, integrating recent structural insights on dimerization, nucleic acid binding modalities and oligomerization propensity with biological function. The emerging paradigm describes a family of dynamic proteins mediating a wide range of protein-protein and protein-nucleic acid interactions, on the whole acting as a multipurpose molecular scaffold. Overall, significant steps toward appreciating the role of DBHS proteins have been made, but we are only beginning to understand the complexity and broader importance of this family in cellular biology.


Subject(s)
Gene Expression Regulation/genetics , Nuclear Matrix-Associated Proteins/metabolism , Nuclear Proteins/metabolism , Octamer Transcription Factors/metabolism , PTB-Associated Splicing Factor/metabolism , RNA Processing, Post-Transcriptional/genetics , RNA Splicing Factors/metabolism , RNA-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Caenorhabditis elegans , DNA Repair/genetics , DNA-Binding Proteins , Drosophila melanogaster , Humans , Nuclear Matrix-Associated Proteins/ultrastructure , Nuclear Proteins/ultrastructure , Octamer Transcription Factors/ultrastructure , PTB-Associated Splicing Factor/ultrastructure , Protein Interaction Maps , Protein Structure, Secondary , RNA-Binding Proteins/ultrastructure , Transcription, Genetic/genetics , Transcriptional Activation/genetics
18.
mSphere ; 9(3): e0069623, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38376217

ABSTRACT

Fungal infections cause a large health burden but are treated by only a handful of antifungal drug classes. Chromatin factors have emerged as possible targets for new antifungals. These targets include the reader proteins, which interact with posttranslationally modified histones to influence DNA transcription and repair. The YEATS domain is one such reader recognizing both crotonylated and acetylated histones. Here, we performed a detailed structure/function analysis of the Candida albicans YEATS domain reader Yaf9, a subunit of the NuA4 histone acetyltransferase and the SWR1 chromatin remodeling complex. We have previously demonstrated that the homozygous deletion mutant yaf9Δ/Δ displays growth defects and is avirulent in mice. Here we show that a YEATS domain mutant expected to inactivate Yaf9's chromatin binding does not display strong phenotypes in vitro, nor during infection of immune cells or in a mouse systemic infection model, with only a minor virulence reduction in vivo. In contrast to the YEATS domain mutation, deletion of the C-terminal domain of Yaf9, a protein-protein interaction module necessary for its interactions with SWR1 and NuA4, phenocopies the null mutant. This shows that the C-terminal domain is essential for Yaf9 roles in vitro and in vivo, including C. albicans virulence. Our study informs on the strategies for therapeutic targeting of Yaf9, showing that approaches taken for the mammalian YEATS domains by disrupting their chromatin binding might not be effective in C. albicans, and provides a foundation for studying YEATS proteins in human fungal pathogens.IMPORTANCEThe scarcity of available antifungal drugs and rising resistance demand the development of therapies with new modes of action. In this context, chromatin regulation may be a target for novel antifungal therapeutics. To realize this potential, we must better understand the roles of chromatin regulators in fungal pathogens. Toward this goal, here, we studied the YEATS domain chromatin reader Yaf9 in Candida albicans. Yaf9 uses the YEATS domain for chromatin binding and a C-terminal domain to interact with chromatin remodeling complexes. By constructing mutants in these domains and characterizing their phenotypes, our data indicate that the Yaf9 YEATS domain might not be a suitable therapeutic drug target. Instead, the Yaf9 C-terminal domain is critical for C. albicans virulence. Collectively, our study informs how a class of chromatin regulators performs their cellular and pathogenesis roles in C. albicans and reveals strategies to inhibit them.


Subject(s)
Chromatin , Saccharomyces cerevisiae Proteins , Humans , Animals , Mice , Chromatin/genetics , Histones/genetics , Candida albicans/genetics , Candida albicans/metabolism , Saccharomyces cerevisiae Proteins/genetics , Antifungal Agents , Homozygote , Sequence Deletion , Transcription Factors/genetics , Transcription Factors/metabolism , Protein Interaction Domains and Motifs , Mammals
19.
PLoS One ; 17(4): e0263547, 2022.
Article in English | MEDLINE | ID: mdl-35436289

ABSTRACT

Short segments of RNA displace one strand of a DNA duplex during diverse processes including transcription and CRISPR-mediated immunity and genome editing. These strand exchange events involve the intersection of two geometrically distinct helix types-an RNA:DNA hybrid (A-form) and a DNA:DNA homoduplex (B-form). Although previous evidence suggests that these two helices can stack on each other, it is unknown what local geometric adjustments could enable A-on-B stacking. Here we report the X-ray crystal structure of an RNA-5'/DNA-3' strand exchange junction at an anisotropic resolution of 1.6 to 2.2 Å. The structure reveals that the A-to-B helical transition involves a combination of helical axis misalignment, helical axis tilting and compression of the DNA strand within the RNA:DNA helix, where nucleotides exhibit a mixture of A- and B-form geometry. These structural principles explain previous observations of conformational stability in RNA/DNA exchange junctions, enabling a nucleic acid architecture that is repeatedly populated during biological strand exchange events.


Subject(s)
Nucleic Acids , RNA , DNA/chemistry , Nucleic Acid Conformation , Nucleotides , RNA/chemistry
20.
Nat Struct Mol Biol ; 29(4): 395-402, 2022 04.
Article in English | MEDLINE | ID: mdl-35422516

ABSTRACT

In bacterial defense and genome editing applications, the CRISPR-associated protein Cas9 searches millions of DNA base pairs to locate a 20-nucleotide, guide RNA-complementary target sequence that abuts a protospacer-adjacent motif (PAM). Target capture requires Cas9 to unwind DNA at candidate sequences using an unknown ATP-independent mechanism. Here we show that Cas9 sharply bends and undertwists DNA on PAM binding, thereby flipping DNA nucleotides out of the duplex and toward the guide RNA for sequence interrogation. Cryogenic-electron microscopy (cryo-EM) structures of Cas9-RNA-DNA complexes trapped at different states of the interrogation pathway, together with solution conformational probing, reveal that global protein rearrangement accompanies formation of an unstacked DNA hinge. Bend-induced base flipping explains how Cas9 'reads' snippets of DNA to locate target sites within a vast excess of nontarget DNA, a process crucial to both bacterial antiviral immunity and genome editing. This mechanism establishes a physical solution to the problem of complementarity-guided DNA search and shows how interrogation speed and local DNA geometry may influence genome editing efficiency.


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