Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 726
Filter
1.
Anal Chim Acta ; 1316: 342873, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-38969432

ABSTRACT

BACKGROUND: DNA walker-based strategies have gained significant attention in nucleic acid analysis. However, they face challenges related to balancing design complexity, sequence dependence, and amplification efficiency. Furthermore, most existing DNA walkers rely on walking and lock probes, requiring optimization of various parameters like DNA probe sequence, walking-to-lock probe ratio, lock probe length, etc. to achieve optimal performance. This optimization process is time-consuming and adds complexity to experiments. To enhance the performance and reliability of DNA walker nanomachines, there is a need for a simpler, highly sensitive, and selective alternative strategy. RESULTS: A sensitive and rapid miRNA analysis strategy named hairpin-shaped DNA aligner and nicking endonuclease-fueled DNA walker (HDA-NE DNA walker) was developed. The HDA-NE DNA walker was constructed by modifying hairpin-shaped DNA aligner (HDA) probe and substrate report (SR) probe on the surface of AuNPs. Under normal conditions, HDA and SR remained stable. However, in the presence of miR-373, HDA underwent a conformational transition to an activated structure to continuously cleave the SR probe on the AuNPs with the assistance of Nt.AlwI nicking endonuclease, resulting in sensitive miRNA detection with a detection limit as low as 0.23 pM. Additionally, the proposed HDA-NE DNA walker exhibited high selectivity in distinguishing miRNAs with single base differences and can effectively analyze miR-373 levels in both normal and breast cancer patient serums. SIGNIFICANCE: The proposed HDA-NE DNA walker system was activated by a conformational change of HDA probe only in the presence of the target miRNA, eliminating the need for a lock probe and without sequence dependence for SR probe. This strategy demonstrated a rapid reaction rate of only 30 min, minimal background noise, and a high signal-to-noise ratio (S/B) compared to capture/lock-based DNA walker. The method is expected to become a powerful tool and play an important role in disease diagnosis and precision therapy.


Subject(s)
DNA , MicroRNAs , MicroRNAs/blood , MicroRNAs/analysis , Humans , DNA/chemistry , Limit of Detection , Biosensing Techniques/methods , Gold/chemistry , Metal Nanoparticles/chemistry , DNA Probes/chemistry , DNA Probes/genetics , Endonucleases/metabolism , Endonucleases/chemistry , Inverted Repeat Sequences
2.
Int J Biol Macromol ; 272(Pt 1): 132654, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38810854

ABSTRACT

Mre11 is one of important proteins that are involved in DNA repair and recombination by processing DNA ends to produce 3'-single stranded DNA, thus providing a platform for other DNA repair and recombination proteins. In this work, we characterized the Mre11 protein from the hyperthermophilic euryarchaeon Thermococcus barophilus Ch5 (Tba-Mre11) biochemically and dissected the roles of its four conserved residues, which is the first report on Mre11 proteins from Thermococcus. Tba-Mre11 possesses exonuclease activity for degrading ssDNA and dsDNA in the 5'-3' direction, which contrasts with other reported Mre11 homologs. Maximum degradation efficiency was observed with Mn2+ at 80 °C and at pH 7.5-9.5. In addition to possessing 5'-3' exonuclease activity, Tba-Mre11 has endonuclease activity that nicks plasmid DNA and circular ssDNA. Mutational data show that residues D10, D51 and N86 in Tba-Mre11 are essential for DNA degradation since almost no activity was observed for the D10A, D51A and N86A mutants. By comparison, residue D44 in Tba-Mre11 is not responsible for DNA degradation since the D44A mutant possessed the similar WT protein activity. Notably, the D44A mutant almost completely abolished the ability to bind DNA, suggesting that residue D44 is essential for binding DNA.


Subject(s)
Archaeal Proteins , DNA, Single-Stranded , Thermococcus , Thermococcus/enzymology , Thermococcus/genetics , Archaeal Proteins/metabolism , Archaeal Proteins/chemistry , Archaeal Proteins/genetics , DNA, Single-Stranded/metabolism , Exodeoxyribonucleases/metabolism , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/chemistry , Amino Acid Sequence , Endonucleases/metabolism , Endonucleases/chemistry , Endonucleases/genetics , Mutation , Endodeoxyribonucleases
3.
Chembiochem ; 25(12): e202400204, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38602716

ABSTRACT

Pathogenesis-related class 10 (PR-10) proteins play a crucial role in plant defense by acting as ribonucleases. The specific mechanism of action and substrate specificity of these proteins have remained largely unexplored so far. In this study, we elucidate the enzymatic activity of Pru p 1, a PR-10 protein from peach. We demonstrate that this protein catalyzes the endonucleolytic backbone cleavage of RNA substrates into short oligonucleotides. Initial cleavage products, identified through kinetic analysis, can bind again, priming them for further degradation. NMR binding site mapping reveals that the large internal cavity of Pru p 1, which is characteristic for PR-10 proteins, serves as an anchoring site for single-stranded ribonucleotide chains. We propose a structure-based mechanistic model that accounts for the observed cleavage patterns and the inhibitory effect of zeatin, a nucleoside analog, on the ribonuclease activity of Pru p 1.


Subject(s)
Plant Proteins , Binding Sites , Endonucleases/metabolism , Endonucleases/chemistry , Plant Proteins/metabolism , Plant Proteins/chemistry , Plant Proteins/genetics , RNA/metabolism , RNA/chemistry , Substrate Specificity , Prunus persica
4.
IUCrJ ; 11(Pt 3): 374-383, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38656310

ABSTRACT

The large Bunyavirales order includes several families of viruses with a segmented ambisense (-) RNA genome and a cytoplasmic life cycle that starts by synthesizing viral mRNA. The initiation of transcription, which is common to all members, relies on an endonuclease activity that is responsible for cap-snatching. In La Crosse virus, an orthobunyavirus, it has previously been shown that the cap-snatching endonuclease resides in the N-terminal domain of the L protein. Orthobunyaviruses are transmitted by arthropods and cause diseases in cattle. However, California encephalitis virus, La Crosse virus and Jamestown Canyon virus are North American species that can cause encephalitis in humans. No vaccines or antiviral drugs are available. In this study, three known Influenza virus endonuclease inhibitors (DPBA, L-742,001 and baloxavir) were repurposed on the La Crosse virus endonuclease. Their inhibition was evaluated by fluorescence resonance energy transfer and their mode of binding was then assessed by differential scanning fluorimetry and microscale thermophoresis. Finally, two crystallographic structures were obtained in complex with L-742,001 and baloxavir, providing access to the structural determinants of inhibition and offering key information for the further development of Bunyavirales endonuclease inhibitors.


Subject(s)
Antiviral Agents , Endonucleases , La Crosse virus , Triazines , La Crosse virus/drug effects , La Crosse virus/enzymology , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Endonucleases/antagonists & inhibitors , Endonucleases/metabolism , Endonucleases/chemistry , Dibenzothiepins , Morpholines/pharmacology , Morpholines/chemistry , Pyridones/pharmacology , Pyridones/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Fluorescence Resonance Energy Transfer , Humans , Animals , Viral Proteins/antagonists & inhibitors , Viral Proteins/chemistry , Viral Proteins/metabolism
5.
Talanta ; 273: 125909, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38490020

ABSTRACT

The in vitro detection of circulating tumor cells (CTCs) has been proven as a vital method for early diagnosis and evaluation of cancer metastasis, since the existence and number fluctuation of CTCs have shown close correlation with clinical outcomes. However, it remains difficult and technically challenging to realize accurate CTCs detection, due to the rarity of CTCs in the blood samples with complex components. Herein, we reported a CTCs in vitro detection strategy, utilizing a loop amplification strategy based on DNA tetrahedron and nicking endonuclease reaction, as well as the anti-background interference based on lanthanide metal luminescence strategy. In this work, a detection system (ATDN-MLLPs) composed of an aptamer-functionalized tetrahedral DNA nanostructure (ATDN) and magnetic lanthanide luminescent particles (MLLPs) was developed. ATDN targeted the tumor cells via aptamer-antigen recognition and extended three hybridizable target DNA segments from the apex of a DNA tetrahedron to pair with probe DNA on MLLPs. Then, the nicking endonuclease (Nt.BbvCI) recognized the formed double-strand DNA and nicked the probe DNA to release the target DNA for recycling, and the released TbNps served as a high signal-to-noise ratio fluorescence signal source for CTCs detection. With a detection limit of 5 cells/mL, CTCs were selectively screened throughout a linear response range of low orders of magnitude. In addition, the ATDN-MLLPs system was attempted to detect possible existence of CTCs in biological samples in vitro.


Subject(s)
Aptamers, Nucleotide , Biosensing Techniques , Neoplastic Cells, Circulating , Humans , Endonucleases/chemistry , Luminescence , DNA/genetics , DNA/chemistry , DNA Probes/chemistry , Metals , Aptamers, Nucleotide/chemistry , Biosensing Techniques/methods , Limit of Detection , Nucleic Acid Amplification Techniques/methods
6.
Nat Struct Mol Biol ; 31(5): 767-776, 2024 May.
Article in English | MEDLINE | ID: mdl-38321146

ABSTRACT

The bacterial cyclic oligonucleotide-based antiphage signaling system (CBASS) is similar to the cGAS-STING system in humans, containing an enzyme that synthesizes a cyclic nucleotide on viral infection and an effector that senses the second messenger for the antiviral response. Cap5, containing a SAVED domain coupled to an HNH DNA endonuclease domain, is the most abundant CBASS effector, yet the mechanism by which it becomes activated for cell killing remains unknown. We present here high-resolution structures of full-length Cap5 from Pseudomonas syringae (Ps) with second messengers. The key to PsCap5 activation is a dimer-to-tetramer transition, whereby the binding of second messenger to dimer triggers an open-to-closed transformation of the SAVED domains, furnishing a surface for assembly of the tetramer. This movement propagates to the HNH domains, juxtaposing and converting two HNH domains into states for DNA destruction. These results show how Cap5 effects bacterial cell suicide and we provide proof-in-principle data that the CBASS can be extrinsically activated to limit bacterial infections.


Subject(s)
Bacterial Proteins , Nucleotides, Cyclic , Pseudomonas syringae , Nucleotides, Cyclic/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Models, Molecular , Crystallography, X-Ray , Second Messenger Systems , Protein Multimerization , Endonucleases/metabolism , Endonucleases/chemistry , Signal Transduction , Humans
7.
Res Microbiol ; 175(5-6): 104189, 2024.
Article in English | MEDLINE | ID: mdl-38403006

ABSTRACT

Archaeal NurA protein plays a key role in producing 3'-single stranded DNA used for homologous recombination repair, together with HerA, Mre11, and Rad50. Herein, we describe biochemical characteristics and roles of key amino acid residues of the NurA protein from the hyperthermophilic euryarchaeon Thermococcus barophilus Ch5 (Tba-NurA). Tba-NurA possesses 5'-3' exonuclease activity for degrading DNA, displaying maximum efficiency at 45 °C-65 °C and at pH 8.0 in the presence of Mn2+. The thermostable Tba-NurA also possesses endonuclease activity capable of nicking plasmid DNA and circular ssDNA. Mutational data demonstrate that residue D49 of Tba-NurA is essential for exonuclease activity and is involved in binding ssDNA since the D49A mutant lacked exonuclease activity and reduced ssDNA binding. The R96A and R129A mutants had no detectable dsDNA binding, suggesting that residues R96 and R129 are important for binding dsDNA. The abolished degradation activity and reduced dsDNA binding of the D120A mutant suggest that residue D120 is essential for degradation activity and dsDNA binding. Additionally, residues Y392 and H400 are important for exonuclease activity since these mutations resulted in exonuclease activity loss. To our knowledge, it is the first report on biochemical characterization and mutational analysis of the NurA protein from Thermococcus.


Subject(s)
Archaeal Proteins , DNA, Single-Stranded , Thermococcus , Thermococcus/genetics , Thermococcus/metabolism , Thermococcus/enzymology , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Archaeal Proteins/chemistry , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/genetics , DNA Mutational Analysis , Hydrogen-Ion Concentration , Exonucleases/metabolism , Exonucleases/genetics , Exonucleases/chemistry , Temperature , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/chemistry , Protein Binding , DNA, Archaeal/genetics , DNA, Archaeal/chemistry , Endonucleases/genetics , Endonucleases/metabolism , Endonucleases/chemistry
8.
Science ; 383(6682): eadh4859, 2024 Feb 02.
Article in English | MEDLINE | ID: mdl-38301022

ABSTRACT

Ribozymes are catalytic RNAs with diverse functions including self-splicing and polymerization. This work aims to discover natural ribozymes that behave as hydrolytic and sequence-specific DNA endonucleases, which could be repurposed as DNA manipulation tools. Focused on bacterial group II-C introns, we found that many systems without intron-encoded protein propagate multiple copies in their resident genomes. These introns, named HYdrolytic Endonucleolytic Ribozymes (HYERs), cleaved RNA, single-stranded DNA, bubbled double-stranded DNA (dsDNA), and plasmids in vitro. HYER1 generated dsDNA breaks in the mammalian genome. Cryo-electron microscopy analysis revealed a homodimer structure for HYER1, where each monomer contains a Mg2+-dependent hydrolysis pocket and captures DNA complementary to the target recognition site (TRS). Rational designs including TRS extension, recruiting sequence insertion, and heterodimerization yielded engineered HYERs showing improved specificity and flexibility for DNA manipulation.


Subject(s)
DNA Cleavage , Endonucleases , RNA, Catalytic , Animals , Cryoelectron Microscopy , Endonucleases/chemistry , Endonucleases/genetics , Hydrolysis , Introns , Nucleic Acid Conformation , RNA Splicing , RNA, Catalytic/chemistry , RNA, Catalytic/genetics
9.
Nature ; 626(7997): 186-193, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38096901

ABSTRACT

The long interspersed element-1 (LINE-1, hereafter L1) retrotransposon has generated nearly one-third of the human genome and serves as an active source of genetic diversity and human disease1. L1 spreads through a mechanism termed target-primed reverse transcription, in which the encoded enzyme (ORF2p) nicks the target DNA to prime reverse transcription of its own or non-self RNAs2. Here we purified full-length L1 ORF2p and biochemically reconstituted robust target-primed reverse transcription with template RNA and target-site DNA. We report cryo-electron microscopy structures of the complete human L1 ORF2p bound to structured template RNAs and initiating cDNA synthesis. The template polyadenosine tract is recognized in a sequence-specific manner by five distinct domains. Among them, an RNA-binding domain bends the template backbone to allow engagement of an RNA hairpin stem with the L1 ORF2p C-terminal segment. Moreover, structure and biochemical reconstitutions demonstrate an unexpected target-site requirement: L1 ORF2p relies on upstream single-stranded DNA to position the adjacent duplex in the endonuclease active site for nicking of the longer DNA strand, with a single nick generating a staggered DNA break. Our research provides insights into the mechanism of ongoing transposition in the human genome and informs the engineering of retrotransposon proteins for gene therapy.


Subject(s)
DNA, Complementary , Long Interspersed Nucleotide Elements , RNA , Retroelements , Reverse Transcription , Humans , Cryoelectron Microscopy , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Long Interspersed Nucleotide Elements/genetics , Retroelements/genetics , RNA/chemistry , RNA/genetics , RNA/metabolism , Catalytic Domain , Endonucleases/chemistry , Endonucleases/metabolism , Endonucleases/ultrastructure , Genetic Therapy , RNA-Directed DNA Polymerase/chemistry , RNA-Directed DNA Polymerase/metabolism , RNA-Directed DNA Polymerase/ultrastructure , DNA, Single-Stranded/metabolism , DNA Breaks
10.
Nature ; 626(7997): 194-206, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38096902

ABSTRACT

The LINE-1 (L1) retrotransposon is an ancient genetic parasite that has written around one-third of the human genome through a 'copy and paste' mechanism catalysed by its multifunctional enzyme, open reading frame 2 protein (ORF2p)1. ORF2p reverse transcriptase (RT) and endonuclease activities have been implicated in the pathophysiology of cancer2,3, autoimmunity4,5 and ageing6,7, making ORF2p a potential therapeutic target. However, a lack of structural and mechanistic knowledge has hampered efforts to rationally exploit it. We report structures of the human ORF2p 'core' (residues 238-1061, including the RT domain) by X-ray crystallography and cryo-electron microscopy in several conformational states. Our analyses identified two previously undescribed folded domains, extensive contacts to RNA templates and associated adaptations that contribute to unique aspects of the L1 replication cycle. Computed integrative structural models of full-length ORF2p show a dynamic closed-ring conformation that appears to open during retrotransposition. We characterize ORF2p RT inhibition and reveal its underlying structural basis. Imaging and biochemistry show that non-canonical cytosolic ORF2p RT activity can produce RNA:DNA hybrids, activating innate immune signalling through cGAS/STING and resulting in interferon production6-8. In contrast to retroviral RTs, L1 RT is efficiently primed by short RNAs and hairpins, which probably explains cytosolic priming. Other biochemical activities including processivity, DNA-directed polymerization, non-templated base addition and template switching together allow us to propose a revised L1 insertion model. Finally, our evolutionary analysis demonstrates structural conservation between ORF2p and other RNA- and DNA-dependent polymerases. We therefore provide key mechanistic insights into L1 polymerization and insertion, shed light on the evolutionary history of L1 and enable rational drug development targeting L1.


Subject(s)
Endonucleases , Long Interspersed Nucleotide Elements , RNA-Directed DNA Polymerase , Reverse Transcription , Humans , Cryoelectron Microscopy , Endonucleases/chemistry , Endonucleases/genetics , Endonucleases/metabolism , Long Interspersed Nucleotide Elements/genetics , RNA/genetics , RNA-Directed DNA Polymerase/chemistry , RNA-Directed DNA Polymerase/genetics , RNA-Directed DNA Polymerase/metabolism , Crystallography, X-Ray , DNA/biosynthesis , DNA/genetics , Immunity, Innate , Interferons/biosynthesis
11.
Acta Crystallogr F Struct Biol Commun ; 79(Pt 12): 295-300, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-38051309

ABSTRACT

Replication initiator proteins (Reps) from the HUH endonuclease family process specific single-stranded DNA sequences to initiate rolling-circle replication in viruses. Here, the first crystal structure of the apo state of a Rep domain from the smacovirus family is reported. The structure of the human smacovirus 1 Rep domain was obtained at 1.33 Šresolution and represents an expansion of the HUH endonuclease superfamily, allowing greater diversity in bioconjugation-tag applications.


Subject(s)
DNA, Single-Stranded , Endonucleases , Humans , Crystallography, X-Ray , Endonucleases/chemistry , DNA, Viral/genetics
12.
J Chem Inf Model ; 63(21): 6834-6850, 2023 11 13.
Article in English | MEDLINE | ID: mdl-37877218

ABSTRACT

The clustered regularly interspaced short palindromic repeats (CRISPR) technology is an RNA-guided targeted genome-editing tool using Cas family proteins. Two magnesium-dependent nuclease domains of the Cas9 enzyme, termed HNH and RuvC, are responsible for cleaving the target DNA (t-DNA) and nontarget DNA strands, respectively. The HNH domain is believed to determine the DNA cleavage activity of both endonuclease domains and is sensitive to complementary RNA-DNA base pairing. However, the underlying molecular mechanisms of CRISPR-Cas9, by which it rebukes or accepts mismatches, are poorly understood. Thus, investigation of the structure and dynamics of the catalytic state of Cas9 with either matched or mismatched t-DNA can provide insights into improving its specificity by reducing off-target cleavages. Here, we focus on a recently discovered catalytic-active form of the Streptococcus pyogenes Cas9 (SpCas9) and employ classical molecular dynamics and coupled quantum mechanics/molecular mechanics simulations to study two possible mechanisms of t-DNA cleavage reaction catalyzed by the HNH domain. Moreover, by designing a mismatched t-DNA structure called MM5 (C to G at the fifth position from the protospacer adjacent motif region), the impact of single-guide RNA (sgRNA) and t-DNA complementarity on the catalysis process was investigated. Based on these simulations, our calculated binding affinities, minimum energy paths, and analysis of catalytically important residues provide atomic-level details of the differences between matched and mismatched cleavage reactions. In addition, several residues exhibit significant differences in their catalytic roles for the two studied systems, including K253, K263, R820, K896, and K913.


Subject(s)
CRISPR-Cas Systems , Molecular Dynamics Simulation , RNA, Guide, CRISPR-Cas Systems , CRISPR-Associated Protein 9/chemistry , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , DNA/chemistry , RNA/chemistry , Endonucleases/chemistry , Endonucleases/genetics , Endonucleases/metabolism
13.
Nature ; 622(7984): 826-833, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37853119

ABSTRACT

CRISPR systems are widespread in the prokaryotic world, providing adaptive immunity against mobile genetic elements1,2. Type III CRISPR systems, with the signature gene cas10, use CRISPR RNA to detect non-self RNA, activating the enzymatic Cas10 subunit to defend the cell against mobile genetic elements either directly, via the integral histidine-aspartate (HD) nuclease domain3-5 or indirectly, via synthesis of cyclic oligoadenylate second messengers to activate diverse ancillary effectors6-9. A subset of type III CRISPR systems encode an uncharacterized CorA-family membrane protein and an associated NrN family phosphodiesterase that are predicted to function in antiviral defence. Here we demonstrate that the CorA-associated type III-B (Cmr) CRISPR system from Bacteroides fragilis provides immunity against mobile genetic elements when expressed in Escherichia coli. However, B. fragilis Cmr does not synthesize cyclic oligoadenylate species on activation, instead generating S-adenosyl methionine (SAM)-AMP (SAM is also known as AdoMet) by conjugating ATP to SAM via a phosphodiester bond. Once synthesized, SAM-AMP binds to the CorA effector, presumably leading to cell dormancy or death by disruption of the membrane integrity. SAM-AMP is degraded by CRISPR-associated phosphodiesterases or a SAM-AMP lyase, potentially providing an 'off switch' analogous to cyclic oligoadenylate-specific ring nucleases10. SAM-AMP thus represents a new class of second messenger for antiviral signalling, which may function in different roles in diverse cellular contexts.


Subject(s)
Adenosine Triphosphate , Bacteroides fragilis , CRISPR-Cas Systems , Escherichia coli , S-Adenosylmethionine , Second Messenger Systems , Adenosine Triphosphate/metabolism , Bacteroides fragilis/enzymology , Bacteroides fragilis/genetics , Bacteroides fragilis/immunology , CRISPR-Associated Proteins/genetics , CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems/genetics , CRISPR-Cas Systems/immunology , CRISPR-Cas Systems/physiology , Endonucleases/chemistry , Endonucleases/metabolism , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli/immunology , Escherichia coli/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , RNA/immunology , RNA/metabolism , S-Adenosylmethionine/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
14.
Biochim Biophys Acta Gen Subj ; 1867(10): 130424, 2023 10.
Article in English | MEDLINE | ID: mdl-37463618

ABSTRACT

BACKGROUND: S1-like nucleases are widespread enzymes commonly used in biotechnology and molecular biology. Although it is commonly believed that they are mainly Zn2+-dependent acidic enzymes, we have found that numerous members of this family deviate from this rule. Therefore, in this work, we decided to check how broad is the range of non­zinc-dependent S1-like nucleases and what is the molecular basis of their activities. METHODS: S1-like nucleases chosen for analysis were achieved through heterologous expression in appropriate eukaryotic hosts. To characterize nucleases' active-site properties, point mutations were introduced in selected positions. The enzymatic activities of wild-type and mutant nucleases were tested by in-gel nuclease activity assay. RESULTS: We discovered that S1-like nucleases encoded by non-vascular plants and single-celled protozoa, like their higher plant homologues, exhibit a large variety of catalytic properties. We have shown that these individual properties are determined by specific non-conserved active site residues. CONCLUSIONS: Our findings demonstrate that mutations that occur during evolution can significantly alter the catalytic properties of S1-like nucleases. As a result, different ions can compete for particular S1-type nucleases' active sites. This phenomenon undermines the existing classification of S1-like nucleases. GENERAL SIGNIFICANCE: Our findings have numerous implications for applications and understanding the S1-like nucleases' biological functions. For example, new biotechnological applications should take into account their unexpected catalytic properties. Moreover, these results demonstrate that the trinuclear zinc-based model commonly used to characterize the catalytic activities of S1-like nucleases is insufficient to explain the actions of non­zinc-dependent members of this family.


Subject(s)
Endonucleases , Plants , Catalytic Domain , Endonucleases/chemistry , Plants/genetics , Plants/metabolism , Eukaryotic Cells , Catalysis
15.
Nature ; 620(7974): 660-668, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37380027

ABSTRACT

RNA-guided systems, which use complementarity between a guide RNA and target nucleic acid sequences for recognition of genetic elements, have a central role in biological processes in both prokaryotes and eukaryotes. For example, the prokaryotic CRISPR-Cas systems provide adaptive immunity for bacteria and archaea against foreign genetic elements. Cas effectors such as Cas9 and Cas12 perform guide-RNA-dependent DNA cleavage1. Although a few eukaryotic RNA-guided systems have been studied, including RNA interference2 and ribosomal RNA modification3, it remains unclear whether eukaryotes have RNA-guided endonucleases. Recently, a new class of prokaryotic RNA-guided systems (termed OMEGA) was reported4,5. The OMEGA effector TnpB is the putative ancestor of Cas12 and has RNA-guided endonuclease activity4,6. TnpB may also be the ancestor of the eukaryotic transposon-encoded Fanzor (Fz) proteins4,7, raising the possibility that eukaryotes are also equipped with CRISPR-Cas or OMEGA-like programmable RNA-guided endonucleases. Here we report the biochemical characterization of Fz, showing that it is an RNA-guided DNA endonuclease. We also show that Fz can be reprogrammed for human genome engineering applications. Finally, we resolve the structure of Spizellomyces punctatus Fz at 2.7 Å using cryogenic electron microscopy, showing the conservation of core regions among Fz, TnpB and Cas12, despite diverse cognate RNA structures. Our results show that Fz is a eukaryotic OMEGA system, demonstrating that RNA-guided endonucleases are present in all three domains of life.


Subject(s)
Chytridiomycota , Endonucleases , Eukaryota , Fungal Proteins , Gene Editing , RNA , Humans , Archaea/genetics , Archaea/immunology , Bacteria/genetics , Bacteria/immunology , CRISPR-Associated Protein 9/metabolism , CRISPR-Associated Proteins/chemistry , CRISPR-Associated Proteins/metabolism , CRISPR-Associated Proteins/ultrastructure , CRISPR-Cas Systems , DNA Transposable Elements/genetics , Endonucleases/chemistry , Endonucleases/metabolism , Endonucleases/ultrastructure , Eukaryota/enzymology , Gene Editing/methods , RNA/genetics , RNA/metabolism , RNA, Guide, CRISPR-Cas Systems/genetics , RNA, Guide, CRISPR-Cas Systems/metabolism , Cryoelectron Microscopy , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Fungal Proteins/ultrastructure , Evolution, Molecular , Conserved Sequence , Chytridiomycota/enzymology
16.
Virol Sin ; 38(4): 559-567, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37290559

ABSTRACT

Cap-dependent endonuclease (CEN) in the polymerase acidic protein (PA) of influenza A virus (IAV) represents a promising drug target due to its critical role in viral gene transcription. The CEN inhibitor, baloxavir marboxil (BXM), was approved in Japan and the US in 2018 and several other countries subsequently. Along with the clinical use of BXM, the emergence and spread of IAV variants with reduced susceptibility to BXM have aroused serious concern. Herein, we comprehensively characterized the in vitro and in vivo antiviral activities of ZX-7101A, an analogue of BXM. The active form of prodrug ZX-7101 showed broad-spectrum antiviral potency against various IAV subtypes, including pH1N1, H3N2, H7N9 and H9N2, in MDCK cells, and the 50% effective concentration (EC50) was calculated to nanomole level and comparable to that of baloxavir acid (BXA), the active form of BXM. Furthermore, in vivo assays showed that administration of ZX-7101A conferred significant protection against lethal pH1N1 challenge in mice, with reduced viral RNA loads and alleviated pulmonary damage. Importantly, serial passaging of H1N1 virus in MDCK cells under selection pressure of ZX-7101 led to a resistant variant at the 15th passage. Reverse genetic and sequencing analysis demonstrated that a single E18G substitution in the PA subunit contributed to the reduced susceptibility to both ZX-7101 and BXA. Taken together, our results not only characterized a new CEN inhibitor of IAV but also identified a novel amino acid substitution responsible for CEN inhibitor resistance, which provides critical clues for future drug development and drug resistance surveillance.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza A Virus, H7N9 Subtype , Influenza A Virus, H9N2 Subtype , Influenza, Human , Thiepins , Animals , Mice , Humans , Oxazines/pharmacology , Oxazines/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Endonucleases/genetics , Endonucleases/chemistry , Endonucleases/metabolism , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype , Thiepins/pharmacology , Thiepins/therapeutic use , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Influenza, Human/drug therapy , Drug Resistance, Viral/genetics
17.
Nature ; 616(7956): 384-389, 2023 04.
Article in English | MEDLINE | ID: mdl-37020015

ABSTRACT

The widespread TnpB proteins of IS200/IS605 transposon family have recently emerged as the smallest RNA-guided nucleases capable of targeted genome editing in eukaryotic cells1,2. Bioinformatic analysis identified TnpB proteins as the likely predecessors of Cas12 nucleases3-5, which along with Cas9 are widely used for targeted genome manipulation. Whereas Cas12 family nucleases are well characterized both biochemically and structurally6, the molecular mechanism of TnpB remains unknown. Here we present the cryogenic-electron microscopy structures of the Deinococcus radiodurans TnpB-reRNA (right-end transposon element-derived RNA) complex in DNA-bound and -free forms. The structures reveal the basic architecture of TnpB nuclease and the molecular mechanism for DNA target recognition and cleavage that is supported by biochemical experiments. Collectively, these results demonstrate that TnpB represents the minimal structural and functional core of the Cas12 protein family and provide a framework for developing TnpB-based genome editing tools.


Subject(s)
CRISPR-Associated Proteins , DNA Transposable Elements , Deinococcus , Endonucleases , Gene Editing , CRISPR-Associated Proteins/chemistry , CRISPR-Associated Proteins/classification , CRISPR-Associated Proteins/metabolism , CRISPR-Associated Proteins/ultrastructure , CRISPR-Cas Systems/genetics , Cryoelectron Microscopy , Deinococcus/enzymology , Deinococcus/genetics , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA/ultrastructure , DNA Transposable Elements/genetics , Endonucleases/chemistry , Endonucleases/classification , Endonucleases/metabolism , Endonucleases/ultrastructure , Evolution, Molecular , Gene Editing/methods , RNA, Guide, CRISPR-Cas Systems
18.
J Proteome Res ; 22(6): 1660-1681, 2023 06 02.
Article in English | MEDLINE | ID: mdl-37071664

ABSTRACT

The DNA repair scaffold SLX4 has pivotal roles in cellular processes that maintain genome stability, most notably homologous recombination. Germline mutations in SLX4 are associated with Fanconi anemia, a disease characterized by chromosome instability and cancer susceptibility. The role of mammalian SLX4 in homologous recombination depends critically on binding and activating structure-selective endonucleases, namely SLX1, MUS81-EME1, and XPF-ERCC1. Increasing evidence indicates that cells rely on distinct SLX4-dependent complexes to remove DNA lesions in specific regions of the genome. Despite our understanding of SLX4 as a scaffold for DNA repair proteins, a detailed repertoire of SLX4 interactors has never been reported. Here, we provide a comprehensive map of the human SLX4 interactome using proximity-dependent biotin identification (BioID) and affinity purification coupled to mass spectrometry (AP-MS). We identified 221 unique high-confidence interactors, of which the vast majority represent novel SLX4-binding proteins. Network analysis of these hits revealed pathways with known involvement of SLX4, such as DNA repair, and several emerging pathways of interest, including RNA metabolism and chromatin remodeling. In summary, the comprehensive SLX4 interactome we report here provides a deeper understanding of how SLX4 functions in DNA repair while revealing new cellular processes that may involve SLX4.


Subject(s)
DNA Repair , DNA-Binding Proteins , Animals , Humans , DNA-Binding Proteins/metabolism , Endonucleases/chemistry , Endonucleases/genetics , Endonucleases/metabolism , DNA/genetics , Homologous Recombination , Mammals/genetics , Mammals/metabolism , Recombinases/chemistry , Recombinases/genetics , Recombinases/metabolism
19.
Adv Sci (Weinh) ; 10(12): e2204388, 2023 04.
Article in English | MEDLINE | ID: mdl-36825683

ABSTRACT

Chromatin bridges connecting the two segregating daughter nuclei arise from chromosome fusion or unresolved interchromosomal linkage. Persistent chromatin bridges are trapped in the cleavage plane, triggering cytokinesis delay. The trapped bridges occasionally break during cytokinesis, inducing DNA damage and chromosomal rearrangements. Recently, Caenorhabditis elegans LEM-3 and human TREX1 nucleases have been shown to process chromatin bridges. Here, it is shown that ANKLE1 endonuclease, the human ortholog of LEM-3, accumulates at the bulge-like structure of the midbody via its N-terminal ankyrin repeats. Importantly, ANKLE1-/- knockout cells display an elevated level of G1-specific 53BP1 nuclear bodies, prolonged activation of the DNA damage response, and replication stress. Increased DNA damage observed in ANKLE1-/- cells is rescued by inhibiting actin polymerization or reducing actomyosin contractility. ANKLE1 does not act in conjunction with structure-selective endonucleases, GEN1 and MUS81 in resolving recombination intermediates. Instead, ANKLE1 acts on chromatin bridges by priming TREX1 nucleolytic activity and cleaving bridge DNA to prevent the formation of micronuclei and cytosolic dsDNA that activate the cGAS-STING pathway. It is therefore proposed that ANKLE1 prevents DNA damage and autoimmunity by cleaving chromatin bridges to avoid catastrophic breakage mediated by actomyosin contractile forces.


Subject(s)
Chromatin , Endonucleases , Animals , Humans , Endonucleases/chemistry , Endonucleases/genetics , Endonucleases/metabolism , Actomyosin/genetics , Actomyosin/metabolism , Cell Nucleus/metabolism , DNA/metabolism , DNA Damage , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism
20.
Anal Chem ; 95(9): 4564-4569, 2023 03 07.
Article in English | MEDLINE | ID: mdl-36812460

ABSTRACT

Highly sensitive and selective detection of microRNA (miRNA) is becoming more and more important in the discovery, diagnosis, and prognosis of various diseases. Herein, we develop a three-dimensional DNA nanostructure based electrochemical platform for duplicate detection of miRNA amplified by nicking endonuclease. Target miRNA first helps construction of three-way junction structures on the surfaces of gold nanoparticles. After nicking endonuclease-powered cleavage reactions, single-stranded DNAs labeled with electrochemical species are released. These strands can be facilely immobilized at four edges of the irregular triangular prism DNA (iTPDNA) nanostructure via triplex assembly. By evaluating the electrochemical response, target miRNA levels can be determined. In addition, the triplexes can be disassociated by simply changing pH conditions, and the iTPDNA biointerface can be regenerated for duplicate analyses. The developed electrochemical method not only exhibits an excellent prospect in the detection of miRNA but also may inspire the engineering of recyclable biointerfaces for biosensing platforms.


Subject(s)
Biosensing Techniques , Metal Nanoparticles , MicroRNAs , MicroRNAs/genetics , MicroRNAs/analysis , Endonucleases/chemistry , Gold/chemistry , Metal Nanoparticles/chemistry , Nucleic Acid Amplification Techniques/methods , DNA/genetics , DNA/chemistry , Biosensing Techniques/methods , Electrochemical Techniques/methods , Limit of Detection
SELECTION OF CITATIONS
SEARCH DETAIL
...