Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.987
Filter
1.
J Am Chem Soc ; 146(37): 25416-25421, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39248674

ABSTRACT

Calicheamicin γ1 and related natural products are renowned for their potency in DNA cleavage, serving as the warheads in commercial ADCs used for treating leukemia. Their mechanism of action involves the formation of aryl radicals, which abstract hydrogen atoms from nucleic acids. However, the complex strained enediyne structure of calicheamicin γ1 presents significant challenges in synthesis, resulting in high production costs and limited structural and activity modularity for tuning the therapeutic window. This report describes the development of simple molecular mimics based on diazonium salts, synthesized in fewer than 3 steps, capable of generating aryl radicals upon green or red light irradiation. SAR studies conducted on over 30 analogues reveal a wide range of potencies in DNA cleavage, with EC50 values ranging from low nanomolar to micromolar. Forming benzenoid diradicals does not appear to be necessary for potent DNA cleavage; instead, DNA cleavage can be achieved with radicals distributed among different arenes when connected with proper linkages. The potency is influenced by electronic effects, stereochemistry, orbital orientations, the distance between multiradicals, and the number of diazonium motifs within the molecule. In addition to providing a more cost-effective, efficient, and modular alternative to calicheamicin γ1, this technology offers the potential for enhanced specificity through spatiotemporal control.


Subject(s)
DNA Cleavage , DNA Cleavage/drug effects , Aminoglycosides/chemistry , Photochemical Processes , Enediynes/chemistry , Enediynes/pharmacology , DNA/chemistry , Structure-Activity Relationship , Light , Diazonium Compounds/chemistry , Humans , Molecular Structure
2.
Dalton Trans ; 53(38): 16005-16017, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39289954

ABSTRACT

In this study, new peripherally substituted symmetric zinc and magnesium phthalocyanines (4 and 5) were successfully prepared by cyclotetramerization of the tetrahydropyrimidone (THPM)-linked phthalonitrile 3. The identity of the compounds were confirmed primarily through spectroscopic analysis including NMR, FT-IR, UV-Vis and MALDI-TOF mass spectroscopy. The photophysical and photochemical properties of the synthesized phthalocyanines (Pcs) were examined using UV-Vis absorption and fluorescence emission spectroscopy techniques. The quantum yields of singlet oxygen were found to be 0.50 and 0.33 for compounds 4 and 5 in DMSO, respectively. In addition to photo-physicochemical properties, the enhanced biological activities of compounds 4 and 5 were investigated using a range of biological assays, namely, antibiofilm, microbial cell viability, antioxidant, DNA cleavage, antimicrobial and photodynamic antimicrobial assays. The maximum DPPH inhibition of 4 and 5 was detected as 40.46% and 25.76% at 100 mg L-1, respectively. Fragmentation of the DNA molecule was observed at concentrations of 25 mg L-1, 50 mg L-1 and 100 mg L-1 for 4 and 5. Additionally, effective inhibition of microbial cell viability was observed with the targeted Pcs. The antibiofilm properties of these compounds were found to be concentration-dependent. The biofilm inhibition activities of 4 and 5 were found to be 96.01% and 92.04% for S. aureus, while they were 95.42% and 91.27%, for P. aeruginosa, respectively. The antimicrobial activities of 4 and 5 on different microorganisms were evaluated using the microdilution assay. In the case of photodynamic antimicrobial treatment, the newly synthesized Pcs showed more effective antimicrobial inhibition compared to the control. These findings suggest that compounds 4 and 5 can be used as promising photodynamic antimicrobial agents for the treatment of many diseases, particularly infectious diseases.


Subject(s)
Biofilms , Indoles , Isoindoles , Microbial Sensitivity Tests , Isoindoles/pharmacology , Indoles/chemistry , Indoles/pharmacology , Indoles/chemical synthesis , Biofilms/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Pyrimidinones/pharmacology , Pyrimidinones/chemistry , Pyrimidinones/chemical synthesis , DNA Cleavage/drug effects , Antioxidants/pharmacology , Antioxidants/chemistry , Antioxidants/chemical synthesis , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/chemical synthesis , Anti-Infective Agents/pharmacology , Anti-Infective Agents/chemistry , Anti-Infective Agents/chemical synthesis , Staphylococcus aureus/drug effects , Molecular Structure , Organometallic Compounds/pharmacology , Organometallic Compounds/chemistry , Organometallic Compounds/chemical synthesis
3.
Sci Rep ; 14(1): 21966, 2024 09 20.
Article in English | MEDLINE | ID: mdl-39304674

ABSTRACT

DNA cleavage by nanomaterials has the potential to be utilized as an innovative tool for gene editing. Numerous nanomaterials exhibiting DNA cleavage properties have been identified and cataloged. Yet, the exploitation of property data through data-driven machine-learning approaches remains unexplored. A database was developed, compiling thirty distinctive characteristics, encompassing physical and chemical properties, as well as experimental conditions of nanomaterials that have demonstrated DNA cleavage capability such as in articles published over the past two decades. The DNA cleavage effect and efficiency of nanomaterials were predicted using machine learning algorithms such as support vector machines, deep neural networks, and random forest, and a classification accuracy of 0.93 for the cleavage effect was achieved. Moreover, the potential of utilizing larger datasets to enhance the predictive capacity of models was discussed. The findings indicate the feasibility of predicting nanomaterial properties based on experimental data. Evaluating the performance and effectiveness of the machine learning models trained using the existing data can furnish valuable insights for future materials research endeavors, especially for the design of DNA cleavage with specific sites.


Subject(s)
DNA Cleavage , Machine Learning , Nanostructures , Nanostructures/chemistry , DNA Cleavage/drug effects , DNA/chemistry , DNA/metabolism , Neural Networks, Computer , Support Vector Machine , Algorithms
4.
Adv Mater ; 36(41): e2406910, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39139006

ABSTRACT

Drug-resistant pathogens significantly threaten human health and life. Simply killing drug-resistant pathogens cannot effectively eliminate their threat since the drug-resistant genes (DRGs) released from dead drug-resistant pathogens are difficult to eliminate and can further spread via horizontal gene transfer, leading to the spread of drug resistance. The development of antibacterial materials with sterilization and DRGs cleavage activities is highly crucial. Herein, a living system, Ce-PEA@Bdello, is fabricated with bacterial killing and DRGs cleavage activities for blocking bacterial drug resistance dissemination by engineered Bdellovibrio bacteriovorus (Bdello). Ce-PEA@Bdello is obtained by engineering Bdello with dopamine and a multinuclear cerium (IV) complex. Ce-PEA@Bdello can penetrate and eliminate kanamycin-resistant P. aeruginosa (KanR) biofilms via the synergistic effect of predatory Bdello and photothermal polydopamine under near-infrared light. Additionally, the DNase-mimicking ability of Ce-PEA@Bdello endows it with genome and plasmid DNA cleavage ability. An in vivo study reveals that Ce-PEA@Bdello can eliminate P. aeruginosa (KanR) and cleave DRGs in scald/burn infected wounds to block the spread of drug resistance and accelerate wound healing. This bioactive system constructed from natural living materials offers a promising means for blocking the spread of drug resistance.


Subject(s)
Anti-Bacterial Agents , Bdellovibrio bacteriovorus , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Pseudomonas aeruginosa/drug effects , Biofilms/drug effects , Cerium/chemistry , Cerium/pharmacology , Drug Resistance, Bacterial/drug effects , Animals , Kanamycin/pharmacology , Dopamine/metabolism , Polymers/chemistry , Polymers/pharmacology , Plasmids/metabolism , Plasmids/genetics , DNA Cleavage/drug effects , Indoles
5.
Int J Mol Sci ; 25(16)2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39201583

ABSTRACT

Hyperthermophilic archaea such as Pyrococcus furiosus survive under very aggressive environmental conditions by occupying niches inaccessible to representatives of other domains of life. The ability to survive such severe living conditions must be ensured by extraordinarily efficient mechanisms of DNA processing, including repair. Therefore, in this study, we compared kinetics of conformational changes of DNA Endonuclease Q from P. furiosus during its interaction with various DNA substrates containing an analog of an apurinic/apyrimidinic site (F-site), hypoxanthine, uracil, 5,6-dihydrouracil, the α-anomer of adenosine, or 1,N6-ethenoadenosine. Our examination of DNA cleavage activity and fluorescence time courses characterizing conformational changes of the dye-labeled DNA substrates during the interaction with EndoQ revealed that the enzyme induces multiple conformational changes of DNA in the course of binding. Moreover, the obtained data suggested that the formation of the enzyme-substrate complex can proceed through dissimilar kinetic pathways, resulting in different types of DNA conformational changes, which probably allow the enzyme to perform its biological function at an extreme temperature.


Subject(s)
DNA Cleavage , Pyrococcus furiosus , Pyrococcus furiosus/enzymology , Kinetics , Archaeal Proteins/metabolism , Archaeal Proteins/chemistry , Substrate Specificity , Nucleic Acid Conformation , DNA/metabolism
6.
Nucleic Acids Res ; 52(16): 9777-9787, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39106172

ABSTRACT

Type II topoisomerases (topos) are a ubiquitous and essential class of enzymes that form transient enzyme-bound double-stranded breaks on DNA called cleavage complexes. The location and frequency of these cleavage complexes on DNA is important for cellular function, genomic stability and a number of clinically important anticancer and antibacterial drugs, e.g. quinolones. We developed a simple high-accuracy end-sequencing (SHAN-seq) method to sensitively map type II topo cleavage complexes on DNA in vitro. Using SHAN-seq, we detected Escherichia coli gyrase and topoisomerase IV cleavage complexes at hundreds of sites on supercoiled pBR322 DNA, approximately one site every ten bp, with frequencies that varied by two-to-three orders of magnitude. These sites included previously identified sites and 20-50-fold more new sites. We show that the location and frequency of cleavage complexes at these sites are enzyme-specific and vary substantially in the presence of the quinolone, ciprofloxacin, but not with DNA supercoil chirality, i.e. negative versus positive supercoiling. SHAN-seq's exquisite sensitivity provides an unprecedented single-nucleotide resolution view of the distribution of gyrase and topoisomerase IV cleavage complexes on DNA. Moreover, the discovery that these enzymes can cleave DNA at orders of magnitude more sites than the relatively few previously known sites resolves the apparent paradox of how these enzymes resolve topological problems throughout the genome.


Subject(s)
DNA Cleavage , DNA Gyrase , DNA Topoisomerase IV , DNA Topoisomerases, Type II , Escherichia coli , Escherichia coli/genetics , Escherichia coli/enzymology , DNA Gyrase/metabolism , DNA Gyrase/genetics , DNA Gyrase/chemistry , DNA Topoisomerase IV/metabolism , DNA Topoisomerase IV/genetics , DNA Topoisomerase IV/chemistry , DNA Topoisomerases, Type II/metabolism , DNA Topoisomerases, Type II/genetics , Sequence Analysis, DNA/methods , DNA, Superhelical/metabolism , DNA, Superhelical/chemistry , Ciprofloxacin/pharmacology , High-Throughput Nucleotide Sequencing , DNA/metabolism , DNA/chemistry
7.
Cell ; 187(19): 5238-5252.e20, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39208796

ABSTRACT

Fanzor (Fz) is an ωRNA-guided endonuclease extensively found throughout the eukaryotic domain with unique gene editing potential. Here, we describe the structures of Fzs from three different organisms. We find that Fzs share a common ωRNA interaction interface, regardless of the length of the ωRNA, which varies considerably across species. The analysis also reveals Fz's mode of DNA recognition and unwinding capabilities as well as the presence of a non-canonical catalytic site. The structures demonstrate how protein conformations of Fz shift to allow the binding of double-stranded DNA to the active site within the R-loop. Mechanistically, examination of structures in different states shows that the conformation of the lid loop on the RuvC domain is controlled by the formation of the guide/DNA heteroduplex, regulating the activation of nuclease and DNA double-stranded displacement at the single cleavage site. Our findings clarify the mechanism of Fz, establishing a foundation for engineering efforts.


Subject(s)
DNA Cleavage , DNA , DNA/metabolism , DNA/chemistry , Catalytic Domain , Models, Molecular , RNA, Guide, CRISPR-Cas Systems/metabolism , RNA, Guide, CRISPR-Cas Systems/chemistry , Humans , Endodeoxyribonucleases/metabolism , Endodeoxyribonucleases/chemistry , Gene Editing , CRISPR-Cas Systems
8.
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
9.
J Biol Chem ; 300(9): 107720, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39214308

ABSTRACT

Site-specific nucleases are crucial for genome engineering applications in medicine and agriculture. The ideal site-specific nucleases are easily reprogrammable, highly specific in target site recognition, and robust in nuclease activities. Prokaryotic Argonaute (pAgo) proteins have received much attention as biotechnological tools due to their ability to recognize specific target sequences without a protospacer adjacent motif, but their lack of intrinsic dsDNA unwinding activity limits their utility in key applications such as gene editing. Recently, we developed a pAgo-based system for site-specific DNA cleavage at physiological temperatures independently of the DNA form, using peptide nucleic acids (PNAs) to facilitate unwinding dsDNA targets. Here, we fused catalytically dead pAgos with the nuclease domain of the restriction endonuclease FokI and named this modified platform PNA-assisted FokI-(d)pAgo (PNFP) editors. In the PNFP system, catalytically inactive pAgo recognizes and binds to a specific target DNA sequence based on a programmable guide DNA sequence; upon binding to the target site, the FokI domains dimerize and introduce precise dsDNA breaks. We explored key parameters of the PNFP system including the requirements of PNA and guide DNAs, the specificity of PNA and guide DNA on target cleavage, the optimal concentration of different components, reaction time for invasion and cleavage, and ideal temperature and reaction buffer, to ensure efficient DNA editing in vitro. The results demonstrated robust site-specific target cleavage by PNFP system at optimal conditions in vitro. We envision that the PNFP system will provide higher editing efficiency and specificity with fewer off-target effects in vivo.


Subject(s)
DNA Cleavage , Deoxyribonucleases, Type II Site-Specific , Deoxyribonucleases, Type II Site-Specific/metabolism , Deoxyribonucleases, Type II Site-Specific/chemistry , Deoxyribonucleases, Type II Site-Specific/genetics , Argonaute Proteins/metabolism , Argonaute Proteins/chemistry , Argonaute Proteins/genetics , Gene Editing/methods , DNA/metabolism , DNA/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Peptide Nucleic Acids/metabolism , Peptide Nucleic Acids/chemistry , Escherichia coli/metabolism , Escherichia coli/genetics
10.
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
11.
Nucleic Acids Res ; 52(16): 9788-9802, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39077950

ABSTRACT

The ability to catalyze reversible DNA cleavage and religation is central to topoisomerases' role in regulating DNA topology. In type IIA topoisomerases (Top2), the formation of its DNA cleavage-religation center is driven by DNA-binding-induced structural rearrangements. These changes optimally position key catalytic modules, such as the active site tyrosine of the WHD domain and metal ion(s) chelated by the TOPRIM domain, around the scissile phosphodiester bond to perform reversible transesterification. To understand this assembly process in detail, we report the catalytic core structures of human Top2α and Top2ß in an on-pathway conformational state. This state features an in trans formation of an interface between the Tower and opposing TOPRIM domain, revealing a groove for accommodating incoming G-segment DNA. Structural superimposition further unveils how subsequent DNA-binding-induced disengagement of the TOPRIM and Tower domains allows a firm grasp of the bound DNA for cleavage/religation. Notably, we identified a previously undocumented protein-DNA interaction, formed between an arginine-capped C-terminus of an α-helix in the TOPRIM domain and the DNA backbone, significantly contributing to Top2 function. This work uncovers a previously unrecognized role of the Tower domain, highlighting its involvement in anchoring and releasing the TOPRIM domain, thus priming Top2 for DNA binding and cleavage.


Subject(s)
DNA Cleavage , DNA Topoisomerases, Type II , DNA Topoisomerases, Type II/metabolism , DNA Topoisomerases, Type II/chemistry , Humans , Poly-ADP-Ribose Binding Proteins/chemistry , Poly-ADP-Ribose Binding Proteins/metabolism , DNA/chemistry , DNA/metabolism , Catalytic Domain , Models, Molecular , Protein Binding , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Crystallography, X-Ray
12.
Nucleic Acids Res ; 52(15): 9014-9027, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-38994558

ABSTRACT

CasX (also known as Cas12e), a Class 2 CRISPR-Cas system, shows promise in genome editing due to its smaller size compared to the widely used Cas9 and Cas12a. Although the structures of CasX-sgRNA-DNA ternary complexes have been resolved and uncover a distinctive NTSB domain, the dynamic behaviors of CasX are not well characterized. In this study, we employed single-molecule and biochemical assays to investigate the conformational dynamics of two CasX homologs, DpbCasX and PlmCasX, from DNA binding to target cleavage and fragment release. Our results indicate that CasX cleaves the non-target strand and the target strand sequentially with relative irreversible dynamics. The two CasX homologs exhibited different cleavage patterns and specificities. The dynamic characterization of CasX also reveals a PAM-proximal seed region, providing guidance for CasX-based effector design. Further studies elucidate the mechanistic basis for why modification of sgRNA and the NTSB domain can affect its activity. Interestingly, CasX has less effective target search efficiency than Cas9 and Cas12a, potentially accounting for its lower genome editing efficiency. This observation opens a new avenue for future protein engineering.


Subject(s)
CRISPR-Associated Proteins , CRISPR-Cas Systems , DNA Cleavage , DNA , Fluorescence Resonance Energy Transfer , CRISPR-Associated Proteins/metabolism , CRISPR-Associated Proteins/chemistry , DNA/chemistry , DNA/metabolism , DNA/genetics , Single Molecule Imaging/methods , RNA, Guide, CRISPR-Cas Systems/chemistry , RNA, Guide, CRISPR-Cas Systems/genetics , RNA, Guide, CRISPR-Cas Systems/metabolism , Gene Editing/methods , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Endodeoxyribonucleases/metabolism , Endodeoxyribonucleases/chemistry , Endodeoxyribonucleases/genetics , Protein Conformation
13.
J Biol Chem ; 300(8): 107576, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39009341

ABSTRACT

Enzymes that form filamentous assemblies with modulated enzymatic activities have gained increasing attention in recent years. SgrAI is a sequence specific type II restriction endonuclease that forms polymeric filaments with accelerated DNA cleavage activity and expanded DNA sequence specificity. Prior studies have suggested a mechanistic model linking the structural changes accompanying SgrAI filamentation to its accelerated DNA cleavage activity. In this model, the conformational changes that are specific to filamentous SgrAI maximize contacts between different copies of the enzyme within the filament and create a second divalent cation binding site in each subunit, which in turn facilitates the DNA cleavage reaction. However, our understanding of the atomic mechanism of catalysis is incomplete. Herein, we present two new structures of filamentous SgrAI solved using cryo-EM. The first structure, resolved to 3.3 Å, is of filamentous SgrAI containing an active site mutation that is designed to stall the DNA cleavage reaction, which reveals the enzymatic configuration prior to DNA cleavage. The second structure, resolved to 3.1 Å, is of WT filamentous SgrAI containing cleaved substrate DNA, which reveals the enzymatic configuration at the end of the enzymatic cleavage reaction. Both structures contain the phosphate moiety at the cleavage site and the biologically relevant divalent cation cofactor Mg2+ and define how the Mg2+ cation reconfigures during enzymatic catalysis. The data support a model for the activation mechanism that involves binding of a second Mg2+ in the SgrAI active site as a direct result of filamentation induced conformational changes.


Subject(s)
DNA Cleavage , Deoxyribonucleases, Type II Site-Specific , Deoxyribonucleases, Type II Site-Specific/metabolism , Deoxyribonucleases, Type II Site-Specific/chemistry , Catalytic Domain , DNA/metabolism , DNA/chemistry , Cryoelectron Microscopy , Magnesium/metabolism , Magnesium/chemistry , Cations, Divalent/metabolism , Models, Molecular
14.
Nat Commun ; 15(1): 5789, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987539

ABSTRACT

The outcome of CRISPR-Cas-mediated genome modifications is dependent on DNA double-strand break (DSB) processing and repair pathway choice. Homology-directed repair (HDR) of protein-blocked DSBs requires DNA end resection that is initiated by the endonuclease activity of the MRE11 complex. Using reconstituted reactions, we show that Cas9 breaks are unexpectedly not directly resectable by the MRE11 complex. In contrast, breaks catalyzed by Cas12a are readily processed. Cas9, unlike Cas12a, bridges the broken ends, preventing DSB detection and processing by MRE11. We demonstrate that Cas9 must be dislocated after DNA cleavage to allow DNA end resection and repair. Using single molecule and bulk biochemical assays, we next find that the HLTF translocase directly removes Cas9 from broken ends, which allows DSB processing by DNA end resection or non-homologous end-joining machineries. Mechanistically, the activity of HLTF requires its HIRAN domain and the release of the 3'-end generated by the cleavage of the non-target DNA strand by the Cas9 RuvC domain. Consequently, HLTF removes the H840A but not the D10A Cas9 nickase. The removal of Cas9 H840A by HLTF explains the different cellular impact of the two Cas9 nickase variants in human cells, with potential implications for gene editing.


Subject(s)
CRISPR-Associated Protein 9 , CRISPR-Cas Systems , DNA Breaks, Double-Stranded , DNA , Humans , CRISPR-Associated Protein 9/metabolism , CRISPR-Associated Protein 9/genetics , DNA/metabolism , DNA/genetics , MRE11 Homologue Protein/metabolism , MRE11 Homologue Protein/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , CRISPR-Associated Proteins/metabolism , CRISPR-Associated Proteins/genetics , Gene Editing , Endonucleases/metabolism , Endonucleases/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Endodeoxyribonucleases/metabolism , Endodeoxyribonucleases/genetics , DNA End-Joining Repair , DNA Cleavage , Transcription Factors/metabolism , Transcription Factors/genetics
15.
Chem Biol Drug Des ; 104(1): e14596, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39054402

ABSTRACT

We have conducted an experimental and computational evaluation of new doxorubicin (4a-c) and ß-lapachone (5a-c) analogs. These novel anticancer analogs were previously synthesized, but had not been tested or characterized until now. We have evaluated their antiproliferative and DNA cleavage inhibition properties using breast (MCF-7 and MDA-MB-231) and prostate (PC3) cancer cell lines. Additionally, cell cycle analysis was performed using flow cytometry. Computational studies, including molecular docking, pharmacokinetic properties, and an analysis of DFT and QTAIM chemical descriptors, were performed to gain insights into the electronic structure and elucidate the molecular binding of the new ß-lapachone and doxorubicin analogs with a DNA sequence and Topoisomerase II (Topo II)α. Our results show that 4a analog displays the highest antiproliferative activity in cancer cell lines by inducing cell death. We observed that stacking interactions and hydrogen bonding are essential to stabilize the molecule-DNA-Topo IIα complex. Moreover, 4a and 5a analogs inhibited Topo's DNA cleavage activity. Pharmacodynamic results indicated that studied molecules have favorable adsorption and permeability properties. The calculated chemical descriptors indicate that electron accumulation in quinone rings is relevant to the reactivity and biological activity. Based on our results, 4a is a strong candidate for becoming an anticancer drug.


Subject(s)
Antineoplastic Agents , Cell Proliferation , DNA Topoisomerases, Type II , Doxorubicin , Molecular Docking Simulation , Naphthoquinones , Naphthoquinones/chemistry , Naphthoquinones/pharmacology , Humans , Doxorubicin/pharmacology , Doxorubicin/chemistry , DNA Topoisomerases, Type II/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , MCF-7 Cells , Drug Screening Assays, Antitumor , Topoisomerase II Inhibitors/pharmacology , Topoisomerase II Inhibitors/chemistry , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/metabolism , DNA Cleavage/drug effects
16.
Nat Commun ; 15(1): 5909, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39003282

ABSTRACT

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


Subject(s)
CRISPR-Cas Systems , RNA, Guide, CRISPR-Cas Systems , RNA, Guide, CRISPR-Cas Systems/metabolism , RNA, Guide, CRISPR-Cas Systems/genetics , CRISPR-Associated Proteins/metabolism , CRISPR-Associated Proteins/genetics , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/metabolism , DNA/metabolism , DNA/genetics , RNA/metabolism , RNA/genetics , DNA Cleavage , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Gene Editing/methods , Endodeoxyribonucleases/metabolism , Endodeoxyribonucleases/genetics , Francisella/genetics
17.
Dalton Trans ; 53(28): 11697-11712, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38912924

ABSTRACT

DNA being the necessary element in cell regeneration, controlled cellular apoptosis via DNA binding/cleaving is considered an approach to combat cancer cells. The widely prescribed metallodrug cisplatin has shown interactions with the guanine-N7 center, and a plethora of complexes are continually developed to enhance crosslinking properties as well as covalent and non-covalent interactions. Two pentadentate ligands, L1 (1-(6-(1H-benzo[d]imidazol-2-yl)pyridin-2-yl)-N,N-bis(pyridin-2-ylmethyl)methanamine) and L2 (1-(6-(1-methyl-1H-benzo[d]imidazol-2-yl)pyridin-2-yl)-N,N-bis(pyridin-2-ylmethyl)methanamine), were synthesized together with their respective copper(II) complexes [1](ClO4)2 and [2](ClO4)2, which crystallized in a trigonal bipyramidal fashion. Different analytical and spectroscopic methods confirmed their formation, and their redox behaviour was also examined. The interactions of salmon sperm DNA (ss-DNA) with these two complexes were explored using absorbance spectroscopy, and they both exhibited a binding affinity (Kb) of ∼104 M-1. Fluorescence quenching experiments with ethidium bromide (EB)-bound DNA (EB-DNA) were also performed, and Stern-Volmer constant (KSV) values of 6.93 × 103 and 2.34 × 104 M-1 for [1](ClO4)2 and [2](ClO4)2, respectively, were obtained. Furthermore, DNA conformational changes due to the interactions of both complexes were validated via circular dichroism. We also assessed the DNA cleavage property of these complexes, which resulted in the linearization of circular plasmid DNA. This finding was supported by studying the growth of MDA-MB-231 breast cancer cells upon treatment with both Cu(II) complexes; IC50 values of 5.34 ± 1.02 µM and 0.83 ± 0.18 µM were obtained for [1](ClO4)2 and [2](ClO4)2, respectively. This validates their affinity towards DNA, and these insights can be further utilized for non-platinum based economical metallodrug development based on first row transition metals.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Copper , DNA , Pyridines , Copper/chemistry , Copper/pharmacology , Humans , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Ligands , DNA/chemistry , DNA/metabolism , Pyridines/chemistry , Pyridines/pharmacology , Cell Line, Tumor , DNA Cleavage/drug effects , Drug Screening Assays, Antitumor , Salmon , Animals , Cell Proliferation/drug effects , Molecular Structure
18.
Inorg Chem ; 63(24): 11450-11458, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38823006

ABSTRACT

Two Ru(II) complexes, [Ru(pydppn)(bim)(py)]2+ [2; pydppn = 3-(pyrid-2'-yl)-4,5,9,16-tetraaza-dibenzo[a,c]naphthacene; bim = 2,2'-bisimidazole; py = pyridine] and [Ru(pydppn)(Me4bim)(py)]2+ [3; Me4bim = 2,2'-bis(4,5-dimethylimidazole)], were synthesized and characterized, and their photophysical properties, DNA binding, and photocleavage were evaluated and compared to [Ru(pydppn)(bpy)(py)]2+ (1; bpy = 2,2'-bipyridine). Complexes 2 and 3 exhibit broad 1MLCT (metal-to-ligand charge transfer) transitions with maxima at ∼470 nm and shoulders at ∼525 and ∼600 nm that extend to ∼800 nm. These bands are red-shifted relative to those of 1, attributed to the π-donating ability of the bim and Me4bim ligands. A strong signal at 550 nm is observed in the transient absorption spectra of 1-3, previously assigned as arising from a pydppn-centered 3ππ* state, with lifetimes of ∼19 µs for 1 and 2 and ∼270 ns for 3. A number of methods were used to characterize the mode of binding of 1-3 to DNA, including absorption titrations, thermal denaturation, relative viscosity changes, and circular dichroism, all of which point to the intercalation of the pydpppn ligand between the nucleobases. The photocleavage of plasmid pUC19 DNA was observed upon the irradiation of 1-3 with visible and red light, attributed to the sensitized generation of 1O2 by the complexes. These findings indicate that the bim ligand, together with pydppn, serves to shift the absorption of Ru(II) complexes to the photodynamic therapy window, 600-900 nm, and also extend the excited state lifetimes for the efficient production of cytotoxic singlet oxygen.


Subject(s)
Coordination Complexes , DNA , Photochemotherapy , Photosensitizing Agents , Plasmids , Ruthenium , Singlet Oxygen , DNA/chemistry , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Coordination Complexes/chemical synthesis , Coordination Complexes/radiation effects , Ruthenium/chemistry , Ruthenium/pharmacology , Plasmids/chemistry , Singlet Oxygen/metabolism , Singlet Oxygen/chemistry , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/radiation effects , Molecular Structure , DNA Cleavage/drug effects , DNA Cleavage/radiation effects
19.
Nature ; 630(8016): 484-492, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38811729

ABSTRACT

The CRISPR system is an adaptive immune system found in prokaryotes that defends host cells against the invasion of foreign DNA1. As part of the ongoing struggle between phages and the bacterial immune system, the CRISPR system has evolved into various types, each with distinct functionalities2. Type II Cas9 is the most extensively studied of these systems and has diverse subtypes. It remains uncertain whether members of this family can evolve additional mechanisms to counter viral invasions3,4. Here we identify 2,062 complete Cas9 loci, predict the structures of their associated proteins and reveal three structural growth trajectories for type II-C Cas9. We found that novel associated genes (NAGs) tended to be present within the loci of larger II-C Cas9s. Further investigation revealed that CbCas9 from Chryseobacterium species contains a novel ß-REC2 domain, and forms a heterotetrameric complex with an NAG-encoded CRISPR-Cas-system-promoting (pro-CRISPR) protein of II-C Cas9 (PcrIIC1). The CbCas9-PcrIIC1 complex exhibits enhanced DNA binding and cleavage activity, broader compatibility for protospacer adjacent motif sequences, increased tolerance for mismatches and improved anti-phage immunity, compared with stand-alone CbCas9. Overall, our work sheds light on the diversity and 'growth evolutionary' trajectories of II-C Cas9 proteins at the structural level, and identifies many NAGs-such as PcrIIC1, which serves as a pro-CRISPR factor to enhance CRISPR-mediated immunity.


Subject(s)
Bacteria , Bacteriophages , CRISPR-Associated Protein 9 , CRISPR-Cas Systems , Bacteria/virology , Bacteria/genetics , Bacteria/immunology , Bacteriophages/genetics , Bacteriophages/immunology , Chryseobacterium/genetics , Chryseobacterium/immunology , Chryseobacterium/virology , CRISPR-Associated Protein 9/chemistry , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , CRISPR-Cas Systems/immunology , DNA Cleavage , Genetic Loci/genetics , Models, Molecular , Protein Domains
20.
Nat Chem Biol ; 20(6): 664-665, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38730193
SELECTION OF CITATIONS
SEARCH DETAIL