Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77.127
Filter
1.
Elife ; 132024 Jul 12.
Article in English | MEDLINE | ID: mdl-38995818

ABSTRACT

Members of the diverse heterochromatin protein 1 (HP1) family play crucial roles in heterochromatin formation and maintenance. Despite the similar affinities of their chromodomains for di- and tri-methylated histone H3 lysine 9 (H3K9me2/3), different HP1 proteins exhibit distinct chromatin-binding patterns, likely due to interactions with various specificity factors. Previously, we showed that the chromatin-binding pattern of the HP1 protein Rhino, a crucial factor of the Drosophila PIWI-interacting RNA (piRNA) pathway, is largely defined by a DNA sequence-specific C2H2 zinc finger protein named Kipferl (Baumgartner et al., 2022). Here, we elucidate the molecular basis of the interaction between Rhino and its guidance factor Kipferl. Through phylogenetic analyses, structure prediction, and in vivo genetics, we identify a single amino acid change within Rhino's chromodomain, G31D, that does not affect H3K9me2/3 binding but disrupts the interaction between Rhino and Kipferl. Flies carrying the rhinoG31D mutation phenocopy kipferl mutant flies, with Rhino redistributing from piRNA clusters to satellite repeats, causing pronounced changes in the ovarian piRNA profile of rhinoG31D flies. Thus, Rhino's chromodomain functions as a dual-specificity module, facilitating interactions with both a histone mark and a DNA-binding protein.


Subject(s)
Chromatin , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone , Drosophila Proteins , Drosophila melanogaster , Animals , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromatin/metabolism , Chromatin/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Evolution, Molecular , Phylogeny , Protein Binding , RNA, Small Interfering/metabolism , RNA, Small Interfering/genetics , Histones/metabolism , Histones/genetics , DNA/metabolism , DNA/genetics
2.
Sci Adv ; 10(27): eadn9423, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38968349

ABSTRACT

DNA origami nanostructures (DOs) are promising tools for applications including drug delivery, biosensing, detecting biomolecules, and probing chromatin substructures. Targeting these nanodevices to mammalian cell nuclei could provide impactful approaches for probing, visualizing, and controlling biomolecular processes within live cells. We present an approach to deliver DOs into live-cell nuclei. We show that these DOs do not undergo detectable structural degradation in cell culture media or cell extracts for 24 hours. To deliver DOs into the nuclei of human U2OS cells, we conjugated 30-nanometer DO nanorods with an antibody raised against a nuclear factor, specifically the largest subunit of RNA polymerase II (Pol II). We find that DOs remain structurally intact in cells for 24 hours, including inside the nucleus. We demonstrate that electroporated anti-Pol II antibody-conjugated DOs are piggybacked into nuclei and exhibit subdiffusive motion inside the nucleus. Our results establish interfacing DOs with a nuclear factor as an effective method to deliver nanodevices into live-cell nuclei.


Subject(s)
Cell Nucleus , DNA , Nanostructures , Cell Nucleus/metabolism , Humans , DNA/chemistry , DNA/metabolism , Nanostructures/chemistry , RNA Polymerase II/metabolism , Cell Line, Tumor , Nanotubes/chemistry
3.
Sci Rep ; 14(1): 16274, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39009614

ABSTRACT

The α-helical antimicrobial peptide Kn2-7 enhances the activation of mouse macrophage-like RAW264.7 induced by DNA containing unmethylated cytosine-guanine motifs (CpG DNA). This enhancement is related to increased cellular uptake of DNA by Kn2-7, but the relevant properties of Kn2-7 are unknown. Physicochemical property analysis revealed that Kn2-7 has high amphipathicity. In contrast, the α-helical antimicrobial peptide L5, which increases the cellular uptake of CpG DNA but does not enhance CpG DNA-induced activation, has low amphipathicity. Kn2-7 derivatives with decreased amphipathicity but the same amino acid composition as Kn2-7 did not enhance CpG DNA-induced activation. On the other hand, L5 derivatives with high amphipathicity but the same amino acid composition as L5 enhanced CpG DNA-induced activation. Cellular uptake of DNA was not increased by the L5 derivatives, indicating that high amphipathicity does not affect DNA uptake. Furthermore, α-helical peptides with reversed sequences relative to the Kn2-7 and L5 derivatives with high amphipathicity were synthesized. The reversed-sequence peptides, which had the same amphipathicity but different amino acid sequences from their counterparts, enhanced CpG DNA-induced activation. Taken together, these observations indicate that the high amphipathicity of α-helical peptides enhances the CpG DNA-induced activation of RAW264.7.


Subject(s)
CpG Islands , Macrophages , Animals , Mice , RAW 264.7 Cells , Macrophages/drug effects , Macrophages/metabolism , DNA/chemistry , DNA/metabolism , Macrophage Activation/drug effects , Protein Conformation, alpha-Helical , DNA Methylation/drug effects , Peptides/chemistry , Peptides/pharmacology , Antimicrobial Peptides/pharmacology , Antimicrobial Peptides/chemistry
4.
Life Sci Alliance ; 7(10)2024 Oct.
Article in English | MEDLINE | ID: mdl-39013578

ABSTRACT

Cancer development and progression are generally associated with gene dysregulation, often resulting from changes in the transcription factor (TF) sequence or expression. Identifying key TFs involved in cancer gene regulation provides a framework for potential new therapeutics. This study presents a large-scale cancer gene TF-DNA interaction network, as well as an extensive promoter clone resource for future studies. Highly connected TFs bind to promoters of genes associated with either good or poor cancer prognosis, suggesting that strategies aimed at shifting gene expression balance between these two prognostic groups may be inherently complex. However, we identified potential for oncogene-targeted therapeutics, with half of the tested oncogenes being potentially repressed by influencing specific activators or bifunctional TFs. Finally, we investigate the role of intrinsically disordered regions within the key cancer-related TF ESR1 in DNA binding and transcriptional activity, and found that these regions can have complex trade-offs in TF function. Altogether, our study broadens our knowledge of the TFs involved in cancer gene regulation and provides a valuable resource for future studies and therapeutics.


Subject(s)
DNA , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Neoplasms , Protein Binding , Transcription Factors , Humans , Neoplasms/genetics , Neoplasms/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , DNA/metabolism , DNA/genetics , Promoter Regions, Genetic/genetics , Oncogenes/genetics , Prognosis , Estrogen Receptor alpha/metabolism , Estrogen Receptor alpha/genetics , Computational Biology/methods
5.
Proc Natl Acad Sci U S A ; 121(28): e2403130121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38950369

ABSTRACT

DNA polymerase κ (Polκ) is a specialized polymerase that has multiple cellular roles such as translesion DNA synthesis, replication of repetitive sequences, and nucleotide excision repair. We have developed a method for capturing DNA synthesized by Polκ utilizing a Polκ-specific substrate, N2-(4-ethynylbenzyl)-2'-deoxyguanosine (EBndG). After shearing of the DNA into 200 to 500 bp lengths, the EBndG-containing DNA was covalently bound to biotin using the Cu(I)-catalyzed alkyne-azide cycloaddition reaction and isolated with streptavidin beads. Isolated DNA was then ligated to adaptors, followed by PCR amplification and next-generation sequencing to generate genome-wide repair maps. We have termed this method polymerase κ sequencing. Here, we present the human genome maps for Polκ activity in an undamaged cell line. We found that Polκ activity was enhanced in GC-rich regions, euchromatin regions, the promoter of genes, and in DNA that is replicated early in the S phase.


Subject(s)
DNA-Directed DNA Polymerase , Fibroblasts , Genome, Human , Humans , DNA-Directed DNA Polymerase/metabolism , Fibroblasts/metabolism , DNA Repair , DNA/metabolism , DNA/genetics , Cell Line , DNA Replication
6.
Brief Bioinform ; 25(4)2024 May 23.
Article in English | MEDLINE | ID: mdl-38975896

ABSTRACT

Mechanisms of protein-DNA interactions are involved in a wide range of biological activities and processes. Accurately identifying binding sites between proteins and DNA is crucial for analyzing genetic material, exploring protein functions, and designing novel drugs. In recent years, several computational methods have been proposed as alternatives to time-consuming and expensive traditional experiments. However, accurately predicting protein-DNA binding sites still remains a challenge. Existing computational methods often rely on handcrafted features and a single-model architecture, leaving room for improvement. We propose a novel computational method, called EGPDI, based on multi-view graph embedding fusion. This approach involves the integration of Equivariant Graph Neural Networks (EGNN) and Graph Convolutional Networks II (GCNII), independently configured to profoundly mine the global and local node embedding representations. An advanced gated multi-head attention mechanism is subsequently employed to capture the attention weights of the dual embedding representations, thereby facilitating the integration of node features. Besides, extra node features from protein language models are introduced to provide more structural information. To our knowledge, this is the first time that multi-view graph embedding fusion has been applied to the task of protein-DNA binding site prediction. The results of five-fold cross-validation and independent testing demonstrate that EGPDI outperforms state-of-the-art methods. Further comparative experiments and case studies also verify the superiority and generalization ability of EGPDI.


Subject(s)
Computational Biology , DNA-Binding Proteins , DNA , Neural Networks, Computer , Binding Sites , DNA/metabolism , DNA/chemistry , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/chemistry , Computational Biology/methods , Algorithms , Protein Binding
7.
Cell Death Differ ; 31(7): 836-843, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38951700

ABSTRACT

The extent to which transcription factors read and respond to specific information content within short DNA sequences remains an important question that the tumor suppressor p53 is helping us answer. We discuss recent insights into how local information content at p53 binding sites might control modes of p53 target gene activation and cell fate decisions. Significant prior work has yielded data supporting two potential models of how p53 determines cell fate through its target genes: a selective target gene binding and activation model and a p53 level threshold model. Both of these models largely revolve around an analogy of whether p53 is acting in a "smart" or "dumb" manner. Here, we synthesize recent and past studies on p53 decoding of DNA sequence, chromatin context, and cellular signaling cascades to elicit variable cell fates critical in human development, homeostasis, and disease.


Subject(s)
Gene Expression Regulation , Tumor Suppressor Protein p53 , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Humans , DNA/metabolism , Animals , Cell Differentiation , Chromatin/metabolism , Protein Binding , Signal Transduction , Binding Sites
8.
Nat Commun ; 15(1): 5822, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38987289

ABSTRACT

DNA polymerase theta (Polθ)-mediated end-joining (TMEJ) repairs DNA double-strand breaks and confers resistance to genotoxic agents. How Polθ is regulated at the molecular level to exert TMEJ remains poorly characterized. We find that Polθ interacts with and is PARylated by PARP1 in a HPF1-independent manner. PARP1 recruits Polθ to the vicinity of DNA damage via PARylation dependent liquid demixing, however, PARylated Polθ cannot perform TMEJ due to its inability to bind DNA. PARG-mediated de-PARylation of Polθ reactivates its DNA binding and end-joining activities. Consistent with this, PARG is essential for TMEJ and the temporal recruitment of PARG to DNA damage corresponds with TMEJ activation and dissipation of PARP1 and PAR. In conclusion, we show a two-step spatiotemporal mechanism of TMEJ regulation. First, PARP1 PARylates Polθ and facilitates its recruitment to DNA damage sites in an inactivated state. PARG subsequently activates TMEJ by removing repressive PAR marks on Polθ.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA Polymerase theta , DNA-Directed DNA Polymerase , Poly (ADP-Ribose) Polymerase-1 , Humans , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly (ADP-Ribose) Polymerase-1/genetics , DNA-Directed DNA Polymerase/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , DNA Damage , Animals , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , DNA/metabolism , DNA/genetics , HEK293 Cells , Poly ADP Ribosylation , Poly(ADP-ribose) Polymerases/metabolism , Poly(ADP-ribose) Polymerases/genetics , Carrier Proteins , Glycoside Hydrolases , Nuclear Proteins
9.
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
10.
Science ; 385(6705): eadl6173, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38991060

ABSTRACT

Isocitrate dehydrogenase 1 (IDH1) is the most commonly mutated metabolic gene across human cancers. Mutant IDH1 (mIDH1) generates the oncometabolite (R)-2-hydroxyglutarate, disrupting enzymes involved in epigenetics and other processes. A hallmark of IDH1-mutant solid tumors is T cell exclusion, whereas mIDH1 inhibition in preclinical models restores antitumor immunity. Here, we define a cell-autonomous mechanism of mIDH1-driven immune evasion. IDH1-mutant solid tumors show selective hypermethylation and silencing of the cytoplasmic double-stranded DNA (dsDNA) sensor CGAS, compromising innate immune signaling. mIDH1 inhibition restores DNA demethylation, derepressing CGAS and transposable element (TE) subclasses. dsDNA produced by TE-reverse transcriptase (TE-RT) activates cGAS, triggering viral mimicry and stimulating antitumor immunity. In summary, we demonstrate that mIDH1 epigenetically suppresses innate immunity and link endogenous RT activity to the mechanism of action of a US Food and Drug Administration-approved oncology drug.


Subject(s)
Immune Evasion , Immunity, Innate , Isocitrate Dehydrogenase , Neoplasms , Animals , Humans , Mice , Cell Line, Tumor , DNA/metabolism , DNA Demethylation , DNA Methylation , DNA Transposable Elements , Epigenesis, Genetic , Glutarates/metabolism , Immunity, Innate/genetics , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Mutation , Neoplasms/immunology , Neoplasms/genetics , Nucleotidyltransferases/genetics , Tumor Escape , Immune Evasion/genetics
11.
Nat Commun ; 15(1): 5727, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38977669

ABSTRACT

DNA replication and transcription generate DNA supercoiling, which can cause topological stress and intertwining of daughter chromatin fibers, posing challenges to the completion of DNA replication and chromosome segregation. Type II topoisomerases (Top2s) are enzymes that relieve DNA supercoiling and decatenate braided sister chromatids. How Top2 complexes deal with the topological challenges in different chromatin contexts, and whether all chromosomal contexts are subjected equally to torsional stress and require Top2 activity is unknown. Here we show that catalytic inhibition of the Top2 complex in interphase has a profound effect on the stability of heterochromatin and repetitive DNA elements. Mechanistically, we find that catalytically inactive Top2 is trapped around heterochromatin leading to DNA breaks and unresolved catenates, which necessitate the recruitment of the structure specific endonuclease, Ercc1-XPF, in an SLX4- and SUMO-dependent manner. Our data are consistent with a model in which Top2 complex resolves not only catenates between sister chromatids but also inter-chromosomal catenates between clustered repetitive elements.


Subject(s)
DNA Topoisomerases, Type II , Heterochromatin , DNA Topoisomerases, Type II/metabolism , DNA Topoisomerases, Type II/genetics , Heterochromatin/metabolism , Animals , Topoisomerase II Inhibitors/pharmacology , Repetitive Sequences, Nucleic Acid/genetics , Poly-ADP-Ribose Binding Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/genetics , DNA Replication , DNA, Superhelical/metabolism , DNA, Superhelical/chemistry , Humans , Mice , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , DNA/metabolism , DNA/chemistry , Interphase
12.
Postepy Biochem ; 70(1): 88-94, 2024 05 23.
Article in English | MEDLINE | ID: mdl-39016228

ABSTRACT

Information coded in DNA is replicated, modified and transmitted from the origins of protein-based life. Analogies of these processes to information processing, transmission and storage in computer systems is straightforward and can be utilized both in analysis of biological data and in development of biologically based technical systems. Transcription and translation processes are regulated by extremely complex regulatory networks, providing control of cell growth, cell cycle and cellular responses to stress. As such, they constitute engineering control systems exerting their actions at many levels of time scale and spatial organization. This work presents an engineering perspective on DNA-related information processing and biochemical process control  in living cells, followed by a review of two-way crosstalk between engineering and biology.


Subject(s)
Protein Biosynthesis , Transcription, Genetic , Transcription, Genetic/physiology , Humans , DNA/genetics , DNA/metabolism , Animals , Gene Expression Regulation
13.
Proc Natl Acad Sci U S A ; 121(30): e2403739121, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39012822

ABSTRACT

Natural kinesin motors are tethered to their cargoes via short C-terminal or N-terminal linkers, whose docking against the core motor domain generates directional force. It remains unclear whether linker docking is the only process contributing directional force or whether linker docking is coupled to and amplifies an underlying, more fundamental force-generating mechanical cycle of the kinesin motor domain. Here, we show that kinesin motor domains tethered via double-stranded DNAs (dsDNAs) attached to surface loops drive robust microtubule (MT) gliding. Tethering using dsDNA attached to surface loops disconnects the C-terminal neck-linker and the N-terminal cover strand so that their dock-undock cycle cannot exert force. The most effective attachment positions for the dsDNA tether are loop 2 or loop 10, which lie closest to the MT plus and minus ends, respectively. In three cases, we observed minus-end-directed motility. Our findings demonstrate an underlying, potentially ancient, force-generating core mechanical action of the kinesin motor domain, which drives, and is amplified by, linker docking.


Subject(s)
Kinesins , Microtubules , Protein Domains , Kinesins/metabolism , Kinesins/chemistry , Microtubules/metabolism , Animals , DNA/metabolism , DNA/chemistry
14.
Nat Cell Biol ; 26(7): 1212-1224, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38961283

ABSTRACT

Despite the demonstrated importance of DNA G-quadruplexes (G4s) in health and disease, technologies to readily manipulate specific G4 folding for functional analysis and therapeutic purposes are lacking. Here we employ G4-stabilizing protein/ligand in conjunction with CRISPR to selectively facilitate single or multiple targeted G4 folding within specific genomic loci. We demonstrate that fusion of nucleolin with a catalytically inactive Cas9 can specifically stabilize G4s in the promoter of oncogene MYC and muscle-associated gene Itga7 as well as telomere G4s, leading to cell proliferation arrest, inhibition of myoblast differentiation and cell senescence, respectively. Furthermore, CRISPR can confer intra-G4 selectivity to G4-binding compounds pyridodicarboxamide and pyridostatin. Compared with traditional G4 ligands, CRISPR-guided biotin-conjugated pyridodicarboxamide enables a more precise investigation into the biological functionality of de novo G4s. Our study provides insights that will enhance understanding of G4 functions and therapeutic interventions.


Subject(s)
CRISPR-Cas Systems , G-Quadruplexes , Nucleolin , RNA-Binding Proteins , Humans , Ligands , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Phosphoproteins/metabolism , Phosphoproteins/genetics , Picolinic Acids/pharmacology , Picolinic Acids/chemistry , Cell Proliferation/drug effects , Cell Differentiation/drug effects , Animals , Cellular Senescence/drug effects , Cellular Senescence/genetics , CRISPR-Associated Protein 9/metabolism , CRISPR-Associated Protein 9/genetics , Promoter Regions, Genetic , Telomere/metabolism , Telomere/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Pyridines/pharmacology , Pyridines/chemistry , DNA/metabolism , DNA/genetics , Mice , Clustered Regularly Interspaced Short Palindromic Repeats , HEK293 Cells , Myoblasts/metabolism , Myoblasts/cytology , Aminoquinolines
15.
Life Sci Alliance ; 7(9)2024 Sep.
Article in English | MEDLINE | ID: mdl-38969365

ABSTRACT

Zn2+ is an essential metal required by approximately 850 human transcription factors. How these proteins acquire their essential Zn2+ cofactor and whether they are sensitive to changes in the labile Zn2+ pool in cells remain open questions. Using ATAC-seq to profile regions of accessible chromatin coupled with transcription factor enrichment analysis, we examined how increases and decreases in the labile zinc pool affect chromatin accessibility and transcription factor enrichment. We found 685 transcription factor motifs were differentially enriched, corresponding to 507 unique transcription factors. The pattern of perturbation and the types of transcription factors were notably different at promoters versus intergenic regions, with zinc-finger transcription factors strongly enriched in intergenic regions in elevated Zn2+ To test whether ATAC-seq and transcription factor enrichment analysis predictions correlate with changes in transcription factor binding, we used ChIP-qPCR to profile six p53 binding sites. We found that for five of the six targets, p53 binding correlates with the local accessibility determined by ATAC-seq. These results demonstrate that changes in labile zinc alter chromatin accessibility and transcription factor binding to DNA.


Subject(s)
Chromatin , DNA , Protein Binding , Transcription Factors , Tumor Suppressor Protein p53 , Zinc , Humans , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Chromatin/metabolism , Chromatin/genetics , Zinc/metabolism , DNA/metabolism , DNA/genetics , Binding Sites , Transcription Factors/metabolism , Transcription Factors/genetics , Promoter Regions, Genetic/genetics , Chromatin Immunoprecipitation Sequencing/methods
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.
Methods Mol Biol ; 2842: 419-447, 2024.
Article in English | MEDLINE | ID: mdl-39012609

ABSTRACT

Chromatin immunoprecipitation (ChIP) is an invaluable method to characterize interactions between proteins and genomic DNA, such as the genomic localization of transcription factors and post-translational modification of histones. DNA and proteins are reversibly and covalently crosslinked using formaldehyde. Then the cells are lysed to release the chromatin. The chromatin is fragmented into smaller sizes either by micrococcal nuclease (MN) or sonication and then purified from other cellular components. The protein-DNA complexes are enriched by immunoprecipitation (IP) with antibodies that target the epitope of interest. The DNA is released from the proteins by heat and protease treatment, followed by degradation of contaminating RNAs with RNase. The resulting DNA is analyzed using various methods, including polymerase chain reaction (PCR), quantitative PCR (qPCR), or sequencing. This protocol outlines each of these steps for both yeast and human cells. This chapter includes a contextual discussion of the combination of ChIP with DNA analysis methods such as ChIP-on-Chip, ChIP-qPCR, and ChIP-Seq, recent updates on ChIP-Seq data analysis pipelines, complementary methods for identification of binding sites of DNA binding proteins, and additional protocol information about ChIP-qPCR and ChIP-Seq.


Subject(s)
Chromatin Immunoprecipitation Sequencing , Humans , Chromatin Immunoprecipitation Sequencing/methods , Chromatin Immunoprecipitation/methods , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Binding Sites , Chromatin/genetics , Chromatin/metabolism , High-Throughput Nucleotide Sequencing/methods
18.
Int J Mol Sci ; 25(13)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-39000034

ABSTRACT

Non-membrane compartments or biomolecular condensates play an important role in the regulation of cellular processes including DNA repair. Here, an ability of XRCC1, a scaffold protein involved in DNA base excision repair (BER) and single-strand break repair, to form protein-rich microphases in the presence of DNA duplexes was discovered. We also showed that the gap-filling activity of BER-related DNA polymerase λ (Pol λ) is significantly increased by the presence of XRCC1. The stimulation of the Pol λ activity was observed only at micromolar XRCC1 concentrations, which were well above the nanomolar dissociation constant determined for the XRCC1-Pol λ complex and pointed to the presence of an auxiliary stimulatory factor in addition to protein-protein interactions. Indeed, according to dynamic light scattering measurements, the stimulation of the Pol λ activity by XRCC1 was coupled with microphase separation in a protein-DNA mixture. Fluorescence microscopy revealed colocalization of Pol λ, XRCC1, and gapped DNA within the microphases. Thus, stimulation of Pol λ activity is caused both by its interaction with XRCC1 and by specific conditions of microphase separation; this phenomenon is shown for the first time.


Subject(s)
DNA Polymerase beta , DNA Repair , X-ray Repair Cross Complementing Protein 1 , X-ray Repair Cross Complementing Protein 1/metabolism , X-ray Repair Cross Complementing Protein 1/genetics , DNA Polymerase beta/metabolism , Humans , DNA/metabolism , Protein Binding
19.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-39000166

ABSTRACT

Pyridoxal-S-methyl-isothiosemicarbazone (PLITSC) is a member of an important group of ligands characterized by different complexation modes to various transition metals. In this contribution, a new complex containing two differently protonated PLITSC ligands ([Fe(PLITSC-H)(PLITSC)]SO4)∙2.5H2O was obtained. The crystal structure was solved by the X-ray analysis and used further for the optimization at B3LYP/6-311++G(d,p)(H,C,N,O,S)/def2-TZVP(Fe) level of theory. Changes in the interaction strength and bond distance due to protonation were observed upon examination by the Quantum Theory of Atoms in Molecules. The protein binding affinity of [Fe(PLITSC-H)(PLITSC)]SO4 towards transport proteins (Bovine Serum Albumin (BSA) and Human Serum Albumin (HSA)) was investigated by the spectrofluorimetric titration and molecular docking. The interactions with the active pocket containing fluorescent amino acids were examined in detail, which explained the fluorescence quenching. The interactions between complex and DNA were followed by the ethidium-bromide displacement titration and molecular docking. The binding along the minor groove was the dominant process involving complex in the proximity of DNA.


Subject(s)
DNA , Molecular Docking Simulation , Protein Binding , Serum Albumin, Bovine , Ligands , DNA/chemistry , DNA/metabolism , Humans , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/metabolism , Crystallography, X-Ray , Thiosemicarbazones/chemistry , Thiosemicarbazones/metabolism , Cattle , Coordination Complexes/chemistry , Coordination Complexes/metabolism , Animals , Protons , Ferric Compounds/chemistry , Serum Albumin, Human/chemistry , Serum Albumin, Human/metabolism , Binding Sites , Iron/chemistry , Iron/metabolism
20.
Int J Mol Sci ; 25(13)2024 Jul 02.
Article in English | MEDLINE | ID: mdl-39000405

ABSTRACT

Extracellular vesicles (EVs) are garnering attention as a safe and efficient biomolecule delivery system. EVs intrinsically play a crucial role in intercellular communication and pathophysiology by transporting functionally active DNA molecules. The internalized DNA pleiotropically affects the recipient cells. Considering these salient features, an intentional incorporation of specific DNA gene cassettes into EVs and their subsequent delivery to the target cells has potential applications in genetic engineering. Moreover, efficient ways to insert the DNA into EVs during their biogenesis is valuable. Our current research is a step in the development of this technology. As such, cancer cells are known to secrete exosomes containing increased amounts of double-stranded DNA than normal cells. The clonal analysis in our previously published data revealed that exosomes released from various cancer cells contained a significantly larger population of NANOGP8 DNA with a 22-base pair insertion in the 3'-untranslated region (UTR) compared to those secreted by normal cells. This finding led us to hypothesize that the 22-base pair insertion may act as a signal to facilitate the incorporation of NANOGP8 DNA into the exosomes. To test this hypothesis, we compared the EV localization of an Enhanced Green Fluorescent Protein (EGFP) gene fused with the NANOGP8 3'-UTR, with and without the 22-base pair insertion. The quantitative PCR analysis showed a significantly higher EGFP DNA accumulation in exosomes released from cells transfected with the gene cassette containing the 3'-UTR with the 22-base pair insertion. The discovery of a DNA localization signal in exosomal DNA's 3'-UTR could pave the way for the development of an EV-based DNA delivery system. This technology will open new possibilities in genetic engineering and innovative therapies using nucleic acid medicine.


Subject(s)
3' Untranslated Regions , Exosomes , Extracellular Vesicles , Exosomes/metabolism , Exosomes/genetics , Humans , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , DNA/genetics , DNA/metabolism , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/genetics , Cell Line, Tumor
SELECTION OF CITATIONS
SEARCH DETAIL
...