Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 221
Filter
1.
Langmuir ; 40(19): 10195-10207, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38690801

ABSTRACT

With recent advances in DNA-templated dye aggregation for leveraging and engineering molecular excitons, a need exists for minimizing structural heterogeneity. Holliday Junction complexes (HJ) are commonly used to covalently template dye aggregates on their core; however, the global conformation of HJ is detrimentally dynamic. Here, the global conformation of the HJ is selectively tuned by restricting its position and orientation by using a sheet-like DNA origami construct (DOC) physisorbed on glass. The HJ arms are fixed with four different designed interduplex angles (IDAs). Atomic force microscopy confirmed that the HJs are bound to the surface of DOC with tuned IDAs. Dye orientation distributions were determined by combining dipole imaging and super-resolution microscopy. All IDAs led to dye orientations having dispersed distributions along planes perpendicular to the HJ plane, suggesting that stacking occurred between the dye and the neighboring DNA bases. The dye-base stacking interpretation was supported by increasing the size of the core cavity. The narrowest IDA minimizes structural heterogeneity and suggests dye intercalation. A strong correlation is found between the IDA and the orientation of the dye along the HJ plane. These results show that the HJ imposes restrictions on the dye and that the dye-DNA interactions are always present regardless of global conformation. The implications of our results are discussed for the scalability of dye aggregates using DNA self-assembly. Our methodology provides an avenue for the solid-supported single-molecule characterization of molecular assemblies templated on biomolecules─such as DNA and protein templates involved in light-harvesting and catalysis─with tuned conformations and restricted in position and orientation.


Subject(s)
DNA, Cruciform , Nucleic Acid Conformation , DNA, Cruciform/chemistry , DNA/chemistry , Coloring Agents/chemistry , Microscopy, Atomic Force
2.
J Mol Biol ; 434(21): 167822, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36108776

ABSTRACT

RecG, belonging to the category of Superfamily-2 plays a vital role in rescuing different kinds of stalled fork. The elemental mechanism of the helicase activity of RecG with several non-homologous stalled fork structures resembling intermediates formed during the process of DNA repair has been investigated in the present study to capture the dynamic stages of genetic rearrangement. The functional characterization has been exemplified through quantifying the response of the substrate in terms of their molecular heterogeneity and dynamical response by employing single-molecule fluorescence methods. An elevated processivity of RecG is observed for the stalled fork where progression of lagging daughter strand is ahead as compared to that of the leading strand. Through precise alteration of its function in terms of unwinding, depending upon the substrate DNA, RecG catalyzes the formation of Holliday junction from a stalled fork DNA. RecG is found to adopt an asymmetric mode of locomotion to unwind the lagging daughter strand for facilitating formation of Holliday junction that acts as a suitable intermediate for recombinational repair pathway. Our results emphasize the mechanism adopted by RecG during its 'sliding back' mode along the lagging daughter strand to be 'active translocation and passive unwinding'. This also provide clues as to how this helicase decides and controls the mode of translocation along the DNA to unwind.


Subject(s)
Bacterial Proteins , DNA Helicases , DNA Replication , DNA, Cruciform , DNA Helicases/chemistry , DNA, Cruciform/chemistry , Bacterial Proteins/chemistry
3.
Nature ; 609(7927): 630-639, 2022 09.
Article in English | MEDLINE | ID: mdl-36002576

ABSTRACT

The Holliday junction is a key intermediate formed during DNA recombination across all kingdoms of life1. In bacteria, the Holliday junction is processed by two homo-hexameric AAA+ ATPase RuvB motors, which assemble together with the RuvA-Holliday junction complex to energize the strand-exchange reaction2. Despite its importance for chromosome maintenance, the structure and mechanism by which this complex facilitates branch migration are unknown. Here, using time-resolved cryo-electron microscopy, we obtained structures of the ATP-hydrolysing RuvAB complex in seven distinct conformational states, captured during assembly and processing of a Holliday junction. Five structures together resolve the complete nucleotide cycle and reveal the spatiotemporal relationship between ATP hydrolysis, nucleotide exchange and context-specific conformational changes in RuvB. Coordinated motions in a converter formed by DNA-disengaged RuvB subunits stimulate hydrolysis and nucleotide exchange. Immobilization of the converter enables RuvB to convert the ATP-contained energy into a lever motion, which generates the pulling force driving the branch migration. We show that RuvB motors rotate together with the DNA substrate, which, together with a progressing nucleotide cycle, forms the mechanistic basis for DNA recombination by continuous branch migration. Together, our data decipher the molecular principles of homologous recombination by the RuvAB complex, elucidate discrete and sequential transition-state intermediates for chemo-mechanical coupling of hexameric AAA+ motors and provide a blueprint for the design of state-specific compounds targeting AAA+ motors.


Subject(s)
ATPases Associated with Diverse Cellular Activities , Bacterial Proteins , DNA Helicases , DNA, Cruciform , ATPases Associated with Diverse Cellular Activities/chemistry , ATPases Associated with Diverse Cellular Activities/metabolism , ATPases Associated with Diverse Cellular Activities/ultrastructure , Adenosine Triphosphate/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/ultrastructure , Cryoelectron Microscopy , DNA Helicases/chemistry , DNA Helicases/metabolism , DNA Helicases/ultrastructure , DNA, Cruciform/chemistry , DNA, Cruciform/metabolism , DNA, Cruciform/ultrastructure , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/ultrastructure , Homologous Recombination , Hydrolysis , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Multienzyme Complexes/ultrastructure , Nucleotides , Protein Conformation , Rotation
4.
Nat Commun ; 13(1): 654, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35115525

ABSTRACT

Homologous recombination (HR) is a ubiquitous and efficient process that serves the repair of severe forms of DNA damage and the generation of genetic diversity during meiosis. HR can proceed via multiple pathways with different outcomes that may aid or impair genome stability and faithful inheritance, underscoring the importance of HR quality control. Human Bloom's syndrome (BLM, RecQ family) helicase plays central roles in HR pathway selection and quality control via unexplored molecular mechanisms. Here we show that BLM's multi-domain structural architecture supports a balance between stabilization and disruption of displacement loops (D-loops), early HR intermediates that are key targets for HR regulation. We find that this balance is markedly shifted toward efficient D-loop disruption by the presence of BLM's interaction partners Topoisomerase IIIα-RMI1-RMI2, which have been shown to be involved in multiple steps of HR-based DNA repair. Our results point to a mechanism whereby BLM can differentially process D-loops and support HR control depending on cellular regulatory mechanisms.


Subject(s)
DNA Topoisomerases, Type I/metabolism , DNA, Cruciform/metabolism , DNA-Binding Proteins/metabolism , RecQ Helicases/metabolism , DNA Topoisomerases, Type I/genetics , DNA, Cruciform/chemistry , DNA, Cruciform/genetics , DNA-Binding Proteins/genetics , Humans , Kinetics , Models, Genetic , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Protein Binding , RecQ Helicases/genetics , Recombinational DNA Repair/genetics
5.
ACS Appl Mater Interfaces ; 13(27): 32013-32021, 2021 Jul 14.
Article in English | MEDLINE | ID: mdl-34212714

ABSTRACT

The reported donor donor-acceptor ("DD-A") fluorescence resonance energy transfer (FRET) was typically achieved through random collisions and interactions of DNA molecules in the bulk solution, which has inevitable defects, including weak biological stability, slow reaction kinetics, and low hybridization efficiency. In order to overcome these deficiencies, this work developed a quadrivalent cruciform DNA nanostructure (qCDN)-mediated cascaded catalyzed hairpin assembly (CHA) amplifier for the fluorescence detection of amyloid ß oligomer species (AßOs). First, four H1 and four H2 hairpins were assembled on one qCDN to obtain qCDNH1 and qCDNH2, respectively. In the presence of AßOs, strand C was released from the P1-C hybrid hairpin and then alternately opened qCDNH1 and qCDNH2 to trigger the qCDN-mediated CHA. As a result, double donors in H1 and one acceptor in H2 were mutually closed, and the porous DNA nanonet with a high loading of "DD-A" FRET binary probes was formed. The FRET efficiency was approximately 78%, and the initial reaction rate was 25-fold faster than the conventional CHA. The detection limit of AßOs was as low as 0.69 pM. The combination of the "DD-A" FRET binary probes and qCDN-mediated cascaded amplifier exhibited great promise for detecting biomarkers with trace levels.


Subject(s)
Amyloid beta-Peptides/chemistry , DNA, Cruciform/chemistry , Fluorescence Resonance Energy Transfer/methods , Limit of Detection , Nanostructures/chemistry , Protein Multimerization , Protein Structure, Quaternary
6.
Molecules ; 26(4)2021 Feb 18.
Article in English | MEDLINE | ID: mdl-33670583

ABSTRACT

Blocking the PD-1/PD-L1 pathway can diminish immunosuppression and enhance anticancer immunity. PD-1/PD-L1 blockade can be realized by aptamers, which have good biocompatibility and can be synthesized in quantity economically. For in vivo applications, aptamers need to evade renal clearance and nuclease digestion. Here we investigated whether DNA nanostructures could be used to enhance the function of PD-L1 aptamers. Four PD-L1 aptamers (Apt) were built into a Holliday Junction (HJ) to form a tetravalent DNA nanostructure (Apt-HJ). The average size of Apt-HJ was 13.22 nm, which was above the threshold for renal clearance. Apt-HJ also underwent partial phosphorothioate modification and had improved nuclease resistance. Compared with the monovalent PD-L1 aptamer, the tetravalent Apt-HJ had stronger affinity to CT26 colon cancer cells. Moreover, Apt-HJ markedly boosted the antitumor efficacy in vivo vs. free PD-L1 aptamers without raising systemic toxicity. The results indicate that multiple aptamers attached to a DNA nanostructure may significantly improve the function of PD-L1 aptamers in vivo.


Subject(s)
Antineoplastic Agents/pharmacology , Aptamers, Nucleotide/chemistry , B7-H1 Antigen/metabolism , DNA, Cruciform/chemistry , Animals , Cell Line, Tumor , Mice, Inbred BALB C , Nanostructures/chemistry
7.
Nucleic Acids Res ; 49(5): 2803-2815, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33619520

ABSTRACT

Homologous recombination forms and resolves an entangled DNA Holliday Junction (HJ) crucial for achieving genetic reshuffling and genome repair. To maintain genomic integrity, specialized resolvase enzymes cleave the entangled DNA into two discrete DNA molecules. However, it is unclear how two similar stacking isomers are distinguished, and how a cognate sequence is found and recognized to achieve accurate recombination. We here use single-molecule fluorescence observation and cluster analysis to examine how prototypic bacterial resolvase RuvC singles out two of the four HJ strands and achieves sequence-specific cleavage. We find that RuvC first exploits, then constrains the dynamics of intrinsic HJ isomer exchange at a sampled branch position to direct cleavage toward the catalytically competent HJ conformation and sequence, thus controlling recombination output at minimal energetic cost. Our model of rapid DNA scanning followed by 'snap-locking' of a cognate sequence is strikingly consistent with the conformational proofreading of other DNA-modifying enzymes.


Subject(s)
DNA Helicases/metabolism , DNA, Cruciform/chemistry , Escherichia coli Proteins/metabolism , Holliday Junction Resolvases/metabolism , Homologous Recombination , DNA Cleavage , Fluorescence Resonance Energy Transfer , Magnesium
8.
Biochem Biophys Res Commun ; 534: 47-52, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33310187

ABSTRACT

HU, a DNA-binding protein, has a helical N-terminal region (NTR) of ∼44 residues and a beta strand- and IDR-rich C-terminal region (CTR) of ∼46 residues. CTR binds to DNA through (i) a clasp (two arginine/lysine-rich, IDR-rich beta hairpins that bind to phosphate groups in the minor groove), (ii) a flat surface (comprising four antiparallel beta strands that abut the major groove), and (iii) a charge cluster (two lysine residues upon a short C-terminal helix). HU forms a dimer displaying extensive inter-subunit CTR-CTR contacts. A single-chain simulacrum of these contacts (HU-Simul) incorporating all DNA-binding elements was created by fusing together the CTRs of Escherichia coli HU-A and Thermus thermophilus HU. HU-Simul is monomeric, binds to dsDNA and cruciform DNA, but not to ssDNA.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , DNA/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Circular Dichroism , DNA/chemistry , DNA, Cruciform/chemistry , DNA, Cruciform/metabolism , DNA, Single-Stranded/metabolism , Electrophoretic Mobility Shift Assay , Protein Engineering/methods , Protein Stability , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Thermus thermophilus/genetics
9.
Nucleic Acids Res ; 48(21): 12407-12414, 2020 12 02.
Article in English | MEDLINE | ID: mdl-33152066

ABSTRACT

The axial stiffness of DNA origami is determined as a function of key nanostructural characteristics. Different constructs of two-helix nanobeams with specified densities of nicks and Holliday junctions are synthesized and stretched by fluid flow. Implementing single particle tracking to extract force-displacement curves enables the measurement of DNA origami stiffness values at the enthalpic elasticity regime, i.e. for forces larger than 15 pN. Comparisons between ligated and nicked helices show that the latter exhibit nearly a two-fold decrease in axial stiffness. Numerical models that treat the DNA helices as elastic rods are used to evaluate the local loss of stiffness at the locations of nicks and Holliday junctions. It is shown that the models reproduce the experimental data accurately, indicating that both of these design characteristics yield a local stiffness two orders of magnitude smaller than the corresponding value of the intact double-helix. This local degradation in turn leads to a macroscopic loss of stiffness that is evaluated numerically for multi-helix DNA bundles.


Subject(s)
DNA, Cruciform/chemistry , DNA, Single-Stranded/chemistry , DNA, Viral/chemistry , Nanostructures/chemistry , Bacteriophage M13/chemistry , Bacteriophage M13/genetics , Biomechanical Phenomena , DNA, Cruciform/genetics , DNA, Cruciform/metabolism , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , DNA, Viral/genetics , DNA, Viral/metabolism , Elasticity , Polynucleotide 5'-Hydroxyl-Kinase/chemistry , Thermodynamics
10.
Molecules ; 25(21)2020 Nov 03.
Article in English | MEDLINE | ID: mdl-33153073

ABSTRACT

Immobile Holliday junctions represent not only the most fundamental building block of structural DNA nanotechnology but are also of tremendous importance for the in vitro investigation of genetic recombination and epigenetics. Here, we present a detailed study on the room-temperature assembly of immobile Holliday junctions with the help of the single-strand annealing protein Redß. Individual DNA single strands are initially coated with protein monomers and subsequently hybridized to form a rigid blunt-ended four-arm junction. We investigate the efficiency of this approach for different DNA/protein ratios, as well as for different DNA sequence lengths. Furthermore, we also evaluate the potential of Redß to anneal sticky-end modified Holliday junctions into hierarchical assemblies. We demonstrate the Redß-mediated annealing of Holliday junction dimers, multimers, and extended networks several microns in size. While these hybrid DNA-protein nanostructures may find applications in the crystallization of DNA-protein complexes, our work shows the great potential of Redß to aid in the synthesis of functional DNA nanostructures under mild reaction conditions.


Subject(s)
DNA, Cruciform/chemistry , DNA, Single-Stranded/chemistry , DNA-Binding Proteins/chemistry , DNA/chemistry , Temperature
11.
Biochem Biophys Res Commun ; 533(4): 919-924, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33010889

ABSTRACT

The SWI/SNF chromatin remodeling complex plays important roles in gene regulation and it is classified as the SWI/SNF complex in yeast and BAF complex in vertebrates. BAF57, one of the subunits that forms the chromatin remodeling complex core, is well conserved in the BAF complex of vertebrates, which is replaced by bap111 in the Drosophila BAP complex and does not have a counterpart in the yeast SWI/SNF complex. This suggests that BAF57 is a key component of the chromatin remodeling complex in higher eukaryotes. BAF57 contains a HMG domain, which is widely distributed among various proteins and functions as a DNA binding motif. Most proteins with HMG domain bind to four-way junction (4WJ) DNA. Here, we report the crystal structure of the HMG domain of BAF57 (BAF57HMG) at a resolution of 2.55 Å. The structure consists of three α-helices and adopts an L-shaped form. The overall structure is stabilized by a hydrophobic core, which is formed by hydrophobic residues. The binding affinity between BAF57HMG and 4WJ DNA is determined as a 295.83 ± 1.05 nM using a fluorescence quenching assay, and the structure revealed 4WJ DNA binding site of BAF57HMG. Our data will serve structural basis in understanding the roles of BAF57 during chromatin remodeling process.


Subject(s)
Chromosomal Proteins, Non-Histone/chemistry , DNA-Binding Proteins/chemistry , DNA/chemistry , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Crystallography, X-Ray , DNA/genetics , DNA/metabolism , DNA, Cruciform/chemistry , DNA, Cruciform/genetics , DNA, Cruciform/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HMG-Box Domains , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Nucleic Acid Conformation , Protein Binding , Protein Domains , Spectrometry, Fluorescence , Static Electricity
12.
FEBS Lett ; 594(24): 4320-4337, 2020 12.
Article in English | MEDLINE | ID: mdl-32936932

ABSTRACT

The Saccharomyces cerevisiae Mus81-Mms4 complex is a highly conserved DNA structure-specific endonuclease that plays essential roles in the processing of recombination intermediates that arise during the repair of stalled replication forks and double-stranded breaks. To identify novel factors functioning conjointly with Mus81-Mms4, we performed a biochemical screen and found that Crp1, a cruciform DNA-recognizing protein that specifically binds to DNA four-way junction structures, could stimulate the Mus81-Mms4 endonuclease. The specific protein interaction between Mus81-Mms4 and Crp1 was responsible for the stimulation observed. Multicopy expression of Crp1 could partially rescue the sensitivity to DNA-damaging agents of the sgs1∆mus81∆21-24N mutant. Our results provide insight into the functional role and interaction of Crp1 with other proteins involved in DNA repair.


Subject(s)
DNA, Cruciform/metabolism , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Flap Endonucleases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , DNA, Cruciform/chemistry , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Endonucleases/chemistry , Endonucleases/genetics , Enzyme Activation , Gene Expression Regulation, Fungal , Kinetics , Mutation , Nucleic Acid Conformation , Protein Binding , Protein Domains , RecQ Helicases/genetics , RecQ Helicases/metabolism , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
13.
Nature ; 586(7830): 623-627, 2020 10.
Article in English | MEDLINE | ID: mdl-32814343

ABSTRACT

During meiosis, crossover recombination connects homologous chromosomes to direct their accurate segregation1. Defective crossing over causes infertility, miscarriage and congenital disease. Each pair of chromosomes attains at least one crossover via the formation and biased resolution of recombination intermediates known as double Holliday junctions2,3. A central principle of crossover resolution is that the two Holliday junctions are resolved in opposite planes by targeting nuclease incisions to specific DNA strands4. The endonuclease activity of the MutLγ complex has been implicated in the resolution of crossovers5-10, but the mechanisms that activate and direct strand-specific cleavage remain unknown. Here we show that the sliding clamp PCNA is important for crossover-biased resolution. In vitro assays with human enzymes show that PCNA and its loader RFC are sufficient to activate the MutLγ endonuclease. MutLγ is further stimulated by a co-dependent activity of the pro-crossover factors EXO1 and MutSγ, the latter of which binds Holliday junctions11. MutLγ also binds various branched DNAs, including Holliday junctions, but does not show canonical resolvase activity, implying that the endonuclease incises adjacent to junction branch points to achieve resolution. In vivo, RFC facilitates MutLγ-dependent crossing over in budding yeast. Furthermore, PCNA localizes to prospective crossover sites along synapsed chromosomes. These data highlight similarities between crossover resolution and the initiation steps of DNA mismatch repair12,13 and evoke a novel model for crossover-specific resolution of double Holliday junctions during meiosis.


Subject(s)
Crossing Over, Genetic , Endonucleases/metabolism , Meiosis , MutL Proteins/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Adenosine Triphosphate/metabolism , Animals , DNA, Cruciform/chemistry , DNA, Cruciform/genetics , DNA, Cruciform/metabolism , Enzyme Activation , Humans , Hydrolysis , Male , Mice , MutS Proteins/metabolism , Protein Binding , Replication Protein C/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
14.
Nucleic Acids Res ; 48(14): 8090-8098, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32597986

ABSTRACT

The DNA four-way (Holliday) junction is the central intermediate of genetic recombination, yet key aspects of its conformational and thermodynamic properties remain unclear. While multiple experimental approaches have been used to characterize the canonical X-shape conformers under specific ionic conditions, the complete conformational ensemble of this motif, especially at low ionic conditions, remains largely undetermined. In line with previous studies, our single-molecule Förster resonance energy transfer (smFRET) measurements of junction dynamics revealed transitions between two states under high salt conditions, but smFRET could not determine whether there are fast and unresolvable transitions between distinct conformations or a broad ensemble of related states under low and intermediate salt conditions. We therefore used an emerging technique, X-ray scattering interferometry (XSI), to directly probe the conformational ensemble of the Holliday junction across a wide range of ionic conditions. Our results demonstrated that the four-way junction adopts an out-of-plane geometry under low ionic conditions and revealed a conformational state at intermediate ionic conditions previously undetected by other methods. Our results provide critical information to build toward a full description of the conformational landscape of the Holliday junction and underscore the utility of XSI for probing conformational ensembles under a wide range of solution conditions.


Subject(s)
DNA, Cruciform/chemistry , Fluorescence Resonance Energy Transfer , Molecular Dynamics Simulation , Osmolar Concentration , X-Ray Diffraction
15.
ACS Chem Biol ; 15(7): 1942-1948, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32469201

ABSTRACT

In this study, we report experimental (Protein Data Bank (PDB) search) and theoretical (RI-MP2/def2-TZVP level of theory) evidence of the nature, stability, and directionality properties of intramolecular halogen bonding interactions (HaBs) between 5-bromo/5-iodoracil bases and backbone phosphate groups in nucleic acids (NAs). A PDB survey revealed relevant examples where intramolecular HaBs are undertaken and serve as a structural source of stability in RNA and DNA molecules. In order to develop suitable energy predictors, we started this investigation by calculating the interaction energy values and both the potential V(r) and kinetic G(r) energy densities (using Bader's "atoms in molecules" theory) of several halogen bond complexes involving 5-bromo/5-iodoracil molecules and biologically relevant electron donors. Once the energy predictors based on V(r)/G(r) energy densities were developed, we analyzed the HaBs observed in the biological examples retrieved from the PDB search in order to estimate the strength of the interaction. As far as our knowledge extends, intramolecular halogen bonds in NAs have not been previously quantified in the literature using this methodology and may be of great importance in understanding their structural properties as well as in the construction of molecular materials with DNA and other biological macromolecules.


Subject(s)
Bromouracil/chemistry , DNA, Cruciform/chemistry , RNA/chemistry , Static Electricity , Uracil/analogs & derivatives , Bromine/chemistry , Bromouracil/metabolism , DNA, Cruciform/metabolism , Databases, Protein , Escherichia coli/chemistry , Exodeoxyribonuclease V/metabolism , Humans , Iodine/chemistry , Models, Chemical , Protein Binding , RNA/metabolism , Splicing Factor U2AF/metabolism , Thermodynamics , Uracil/chemistry , Uracil/metabolism
16.
Int J Nanomedicine ; 15: 2119-2129, 2020.
Article in English | MEDLINE | ID: mdl-32280210

ABSTRACT

PURPOSE: Chemotherapy is the primary treatment for advanced colon cancer, but its efficacy is often limited by severe toxicities. Targeted therapy in the form of selectively drug delivery system (SDDS) is an important strategy to reduce adverse effects. Here, we aim to design a novel SDDS with potential for practical application using biocompatible components and scalable production process, for targeted delivery of doxorubicin (Dox) to colon cancer cells. METHODS: The SDDS was made of a self-assembled DNA nano-cross (Holliday junction, or HJ) functionalized by four AS1411 aptamers (Apt-HJ) and loaded with Dox. RESULTS: Apt-HJ had an average size of 12.45 nm and a zeta potential of -11.6 mV. Compared with the monovalent AS1411 aptamer, the quadrivalent Apt-HJ showed stronger binding to target cancer cells (CT26). A complex of Apt-HJ and doxorubicin (Apt-HJ-Dox) was formed by intercalating Dox into the DNA structure of Apt-HJ, with each complex carrying approximately 17 Dox molecules. Confocal microscopy revealed that Apt-HJ-Dox selectively delivered Dox into CT26 colon cancer cells but not the control cells. Moreover, Apt-HJ-Dox achieved targeted killing of CT26 cancer cells in vitro and reduced the damage to control cells. Importantly, compared with free Dox, Apt-HJ-Dox significantly enhanced the antitumor efficacy in vivo without boosting the adverse effects. CONCLUSION: These results suggest that Apt-HJ-Dox has application potential in targeted treatment of colon cancer.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Aptamers, Nucleotide/chemistry , Colonic Neoplasms/drug therapy , Doxorubicin/administration & dosage , Drug Delivery Systems/methods , Animals , Aptamers, Nucleotide/administration & dosage , CHO Cells , Cell Line, Tumor , Cricetulus , DNA, Cruciform/chemistry , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Nanostructures/chemistry , Neoplasms, Experimental/drug therapy , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/chemistry
17.
Nat Commun ; 11(1): 1417, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32184398

ABSTRACT

Holliday junctions (HJs) are key DNA intermediates in genetic recombination and are eliminated by nuclease, termed resolvase, to ensure genome stability. HJ resolvases have been identified across all kingdoms of life, members of which exhibit sequence-dependent HJ resolution. However, the molecular basis of sequence selectivity remains largely unknown. Here, we present the chloroplast resolvase MOC1, which cleaves HJ in a cytosine-dependent manner. We determine the crystal structure of MOC1 with and without HJs. MOC1 exhibits an RNase H fold, belonging to the retroviral integrase family. MOC1 functions as a dimer, and the HJ is embedded into the basic cleft of the dimeric enzyme. We characterize a base recognition loop (BR loop) that protrudes into and opens the junction. Residues from the BR loop intercalate into the bases, disrupt the C-G base pairing at the crossover and recognize the cytosine, providing the molecular basis for sequence-dependent HJ resolution by a resolvase.


Subject(s)
Arabidopsis/enzymology , Chloroplasts/enzymology , DNA, Cruciform/metabolism , Glycine max/enzymology , Oryza/enzymology , Plant Proteins/chemistry , Plant Proteins/metabolism , Recombinases/chemistry , Recombinases/metabolism , Arabidopsis/chemistry , Arabidopsis/genetics , Arabidopsis/metabolism , Base Sequence , Chloroplasts/chemistry , Chloroplasts/genetics , DNA, Cruciform/chemistry , DNA, Cruciform/genetics , DNA, Plant/chemistry , DNA, Plant/genetics , DNA, Plant/metabolism , Oryza/chemistry , Oryza/genetics , Oryza/metabolism , Recombinases/genetics , Glycine max/chemistry , Glycine max/genetics , Glycine max/metabolism
18.
Nature ; 577(7792): 701-705, 2020 01.
Article in English | MEDLINE | ID: mdl-31969709

ABSTRACT

Transcription challenges the integrity of replicating chromosomes by generating topological stress and conflicts with forks1,2. The DNA topoisomerases Top1 and Top2 and the HMGB family protein Hmo1 assist DNA replication and transcription3-6. Here we describe the topological architecture of genes in Saccharomyces cerevisiae during the G1 and S phases of the cell cycle. We found under-wound DNA at gene boundaries and over-wound DNA within coding regions. This arrangement does not depend on Pol II or S phase. Top2 and Hmo1 preserve negative supercoil at gene boundaries, while Top1 acts at coding regions. Transcription generates RNA-DNA hybrids within coding regions, independently of fork orientation. During S phase, Hmo1 protects under-wound DNA from Top2, while Top2 confines Pol II and Top1 at coding units, counteracting transcription leakage and aberrant hybrids at gene boundaries. Negative supercoil at gene boundaries prevents supercoil diffusion and nucleosome repositioning at coding regions. DNA looping occurs at Top2 clusters. We propose that Hmo1 locks gene boundaries in a cruciform conformation and, with Top2, modulates the architecture of genes that retain the memory of the topological arrangements even when transcription is repressed.


Subject(s)
DNA, Fungal/chemistry , DNA, Superhelical/chemistry , Genes, Fungal , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Chromatin Assembly and Disassembly , DNA Replication , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , DNA, Cruciform/chemistry , DNA, Cruciform/genetics , DNA, Cruciform/metabolism , DNA, Fungal/genetics , DNA, Fungal/metabolism , DNA, Superhelical/genetics , DNA, Superhelical/metabolism , G1 Phase , Gene Expression Regulation, Fungal , High Mobility Group Proteins/metabolism , Mutation , Nucleic Acid Hybridization , Nucleosomes/chemistry , Nucleosomes/genetics , Nucleosomes/metabolism , Open Reading Frames/genetics , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA, Fungal/chemistry , RNA, Fungal/genetics , RNA, Fungal/metabolism , S Phase , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/metabolism , Transcription, Genetic
19.
Sci Rep ; 10(1): 393, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31941902

ABSTRACT

Fowlpox virus resolvase (Fpr) is an endonuclease that cleaves a broad range of branched DNA structures, including the Holliday junction (HJ), with little sequence-specificity. To better understand the mechanisms underlying its relaxed substrate specificity, we determined the crystal structures of Fpr and that in a novel complex with HJ at 3.1-Å resolution. In the Fpr-HJ complex, two Fpr dimers use several distinct regions to interact with different DNA structural motifs, showing versatility in DNA-binding. Biochemical and solution NMR data support the existence of non-canonical modes of HJ interaction in solution. The binding of Fpr to various DNA motifs are mediated by its flat DNA-binding surface, which is centered on a short loop spanning K61 to I72 and flanked by longer α-helices at the outer edges, and basic side grooves near the dimer interface. Replacing the Fpr loop K61~I72 with a longer loop from Thermus thermophilus RuvC (E71~A87) endows Fpr with an enhanced selectivity toward HJ cleavage but with a target sequence preference distinct from that of RuvC, highlighting a unique role of this loop region in Fpr-HJ interaction. Our work helps explain the broad substrate selectivity of Fpr and suggests a possible mode of its association with poxvirus hairpin telomeres.


Subject(s)
DNA, Cruciform/metabolism , DNA, Viral/metabolism , Fowlpox virus/enzymology , Recombinases/chemistry , Recombinases/metabolism , Crystallography, X-Ray , DNA, Cruciform/chemistry , DNA, Viral/chemistry , Models, Molecular , Protein Conformation , Substrate Specificity
20.
Nucleic Acids Res ; 48(2): 719-735, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31740957

ABSTRACT

Mechanisms that ensure repair of double-strand DNA breaks (DSBs) are instrumental in the integration of foreign DNA into the genome of transgenic organisms. After pronuclear microinjection, exogenous DNA is usually found as a concatemer comprising multiple co-integrated transgene copies. Here, we investigated the contribution of various DSB repair pathways to the concatemer formation. We injected mouse zygotes with a pool of linear DNA molecules carrying unique barcodes at both ends and obtained 10 transgenic embryos with 1-300 transgene copies. Sequencing the barcodes allowed us to assign relative positions to the copies in concatemers and detect recombination events that occurred during integration. Cumulative analysis of approximately 1,000 integrated copies reveals that over 80% of them underwent recombination when their linear ends were processed by synthesis-dependent strand annealing (SDSA) or double-strand break repair (DSBR). We also observed evidence of double Holliday junction (dHJ) formation and crossing over during the concatemer formations. Sequencing indels at the junctions between copies shows that at least 10% of DNA molecules introduced into the zygotes are ligated by non-homologous end joining (NHEJ). Our barcoding approach, verified with Pacific Biosciences Single Molecule Real-Time (SMRT) long-range sequencing, documents high activity of homologous recombination after DNA microinjection.


Subject(s)
DNA Breaks, Double-Stranded , DNA/chemistry , Homologous Recombination/genetics , Transgenes/genetics , Animals , Animals, Genetically Modified , DNA/genetics , DNA Barcoding, Taxonomic , DNA End-Joining Repair/genetics , DNA Repair/genetics , DNA, Cruciform/chemistry , DNA, Cruciform/genetics , Mice , Zygote/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...