Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.847
Filter
1.
Proc Natl Acad Sci U S A ; 121(37): e2406854121, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39231208

ABSTRACT

Alzheimer's disease (AD) is a prevalent neurodegenerative disease characterized by cognitive decline and learning/memory impairment associated with neuronal cell loss. Estrogen-related receptor α (ERRα) and ERRγ, which are highly expressed in the brain, have emerged as potential AD regulators, with unelucidated underlying mechanisms. Here, we identified genome-wide binding sites for ERRα and ERRγ in human neuronal cells. They commonly target a subset of genes associated with neurodegenerative diseases, including AD. Notably, Dickkopf-1 (DKK1), a Wnt signaling pathway antagonist, was transcriptionally repressed by both ERRα and ERRγ in human neuronal cells and brain. ERRα and ERRγ repress RNA polymerase II (RNAP II) accessibility at the DKK1 promoter by modulating a specific active histone modification, histone H3 lysine acetylation (H3K9ac), with the potential contribution of their corepressor. This transcriptional repression maintains Wnt signaling activity, preventing tau phosphorylation and promoting a healthy neuronal state in the context of AD.


Subject(s)
Alzheimer Disease , ERRalpha Estrogen-Related Receptor , Intercellular Signaling Peptides and Proteins , Receptors, Estrogen , tau Proteins , Humans , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , tau Proteins/metabolism , tau Proteins/genetics , Phosphorylation , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Receptors, Estrogen/metabolism , Receptors, Estrogen/genetics , Wnt Signaling Pathway/genetics , Neurons/metabolism , Gene Expression Regulation , Animals , Promoter Regions, Genetic , Mice , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Brain/metabolism
2.
Commun Biol ; 7(1): 1086, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39232115

ABSTRACT

Cell-free DNA (cfDNA) has emerged as a pivotal player in precision medicine, revolutionizing the diagnostic and therapeutic landscape. While its clinical applications have significantly increased in recent years, current cfDNA assays have limited ability to identify the active transcriptional programs that govern complex disease phenotypes and capture the heterogeneity of the disease. To address these limitations, we have developed a non-invasive platform to enrich and examine the active chromatin fragments (cfDNAac) in peripheral blood. The deconvolution of the cfDNAac signal from traditional nucleosomal chromatin fragments (cfDNAnuc) yields a catalog of features linking these circulating chromatin signals in blood to specific regulatory elements across the genome, including enhancers, promoters, and highly transcribed genes, mirroring the epigenetic data from the ENCODE project. Notably, these cfDNAac counts correlate strongly with RNA polymerase II activity and exhibit distinct expression patterns for known circadian genes. Additionally, cfDNAac signals across gene bodies and promoters show strong correlations with whole blood gene expression levels defined by GTEx. This study illustrates the utility of cfDNAac analysis for investigating epigenomics and gene expression, underscoring its potential for a wide range of clinical applications in precision medicine.


Subject(s)
Cell-Free Nucleic Acids , Chromatin , Chromatin/genetics , Chromatin/metabolism , Humans , Cell-Free Nucleic Acids/blood , Cell-Free Nucleic Acids/genetics , Promoter Regions, Genetic , Epigenesis, Genetic , Epigenomics/methods , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Nucleosomes/metabolism , Nucleosomes/genetics
3.
BMC Res Notes ; 17(1): 219, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39103906

ABSTRACT

OBJECTIVE: In past work in budding yeast, we identified a nucleosomal region required for proper interactions between the histone chaperone complex yFACT and transcribed genes. Specific histone mutations within this region cause a shift in yFACT occupancy towards the 3' end of genes, a defect that we have attributed to impaired yFACT dissociation from DNA following transcription. In this work we wished to assess the contributions of DNA sequences at the 3' end of genes in promoting yFACT dissociation upon transcription termination. RESULTS: We generated fourteen different alleles of the constitutively expressed yeast gene PMA1, each lacking a distinct DNA fragment across its 3' end, and assessed their effects on occupancy of the yFACT component Spt16. Whereas most of these alleles conferred no defects on Spt16 occupancy, one did cause a modest increase in Spt16 binding at the gene's 3' end. Interestingly, the same allele also caused minor retention of RNA Polymerase II (Pol II) and altered nucleosome occupancy across the same region of the gene. These results suggest that specific DNA sequences at the 3' ends of genes can play roles in promoting efficient yFACT and Pol II dissociation from genes and can also contribute to proper chromatin architecture.


Subject(s)
Nucleosomes , RNA Polymerase II , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Nucleosomes/metabolism , Nucleosomes/genetics , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Transcriptional Elongation Factors/genetics , Transcriptional Elongation Factors/metabolism , Histone Chaperones/genetics , Histone Chaperones/metabolism , DNA, Fungal/genetics , DNA, Fungal/metabolism , Alleles , Base Sequence , Gene Expression Regulation, Fungal , Transcription, Genetic
4.
Nat Commun ; 15(1): 6751, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39117705

ABSTRACT

Despite the well-established significance of transcription factors (TFs) in pathogenesis, their utilization as pharmacological targets has been limited by the inherent challenges in modulating their protein interactions. The lack of defined small-molecule binding pockets and the nuclear localization of TFs do not favor the use of traditional tools. Aptamers possess large molecular weights, expansive blocking surfaces and efficient cellular internalization, making them compelling tools for modulating TF interactions. Here, we report a structure-guided design strategy called Blocker-SELEX to develop inhibitory aptamers (iAptamers) that selectively block TF interactions. Our approach leads to the discovery of iAptamers that cooperatively disrupt SCAF4/SCAF8-RNAP2 interactions, dysregulating RNAP2-dependent gene expression, which impairs cell proliferation. This approach is further applied to develop iAptamers blocking WDR5-MYC interactions. Overall, our study highlights the potential of iAptamers in disrupting pathogenic TF interactions, implicating their potential utility in studying the biological functions of TF interactions and in nucleic acids drug discovery.


Subject(s)
Aptamers, Nucleotide , SELEX Aptamer Technique , Transcription Factors , Aptamers, Nucleotide/pharmacology , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/metabolism , Humans , Transcription Factors/metabolism , Protein Binding , Cell Proliferation/drug effects , RNA Polymerase II/metabolism , HEK293 Cells , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/antagonists & inhibitors
5.
Antonie Van Leeuwenhoek ; 117(1): 110, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39088091

ABSTRACT

Xenodidymella species have a wide range of hosts and can be found as pathogens and saprobes. In this study, two new species of Xenodidymella were found from leaf diseases of three pasture-medicinal plants in Ilam Province, in the west of Iran, and proposed here as X. ilamica and X. scandicis spp. nov. These species were identified based on morphological features and phylogenetic analyses of the internal transcribed spacer regions 1 & 2 and 5.8S nrDNA (ITS), partial beta-tubulin gene (tub2), and partial RNA polymerase II second largest subunit (rpb2) gene. The four Xenodidymella strains isolated in this study were delimited into two sister clades, with the two isolates of X. ilamica from the leaf spot of Colchicum speciosum and Ficaria kochii and two isolates of X. scandicis from leaf blight of Scandix pecten-veneris. Morphologically, X. scandicis produces larger, ostiolate or poroid pycnidia in vitro, while pycnidia in the cultures of X. ilamica are non-ostiolate and smaller. Some pycnidia in old cultures of X. scandicis produce a neck, but a distinct neck in X. ilamica has not been observed. Moreover, three plants under study are new hosts for the genus Xenodidymella.


Subject(s)
Phylogeny , Tubulin , Iran , Tubulin/genetics , Plants, Medicinal/microbiology , Plant Diseases/microbiology , Plant Leaves/microbiology , DNA, Fungal/genetics , RNA Polymerase II/genetics , Sequence Analysis, DNA , DNA, Ribosomal Spacer/genetics
6.
Nat Commun ; 15(1): 7100, 2024 Aug 18.
Article in English | MEDLINE | ID: mdl-39155303

ABSTRACT

The identification of genes involved in replicative stress is key to understanding cancer evolution and to identify therapeutic targets. Here, we show that CDK12 prevents transcription-replication conflicts (TRCs) and the activation of cytotoxic replicative stress upon deregulation of the MYC oncogene. CDK12 was recruited at damaged genes by PARP-dependent DDR-signaling and elongation-competent RNAPII, to repress transcription. Either loss or chemical inhibition of CDK12 led to DDR-resistant transcription of damaged genes. Loss of CDK12 exacerbated TRCs in MYC-overexpressing cells and led to the accumulation of double-strand DNA breaks, occurring between co-directional early-replicating regions and transcribed genes. Overall, our data demonstrate that CDK12 protects genome integrity by repressing transcription of damaged genes, which is required for proper resolution of DSBs at oncogene-induced TRCs. This provides a rationale that explains both how CDK12 deficiency can promote tandem duplications of early-replicated regions during tumor evolution, and how CDK12 targeting can exacerbate replicative-stress in tumors.


Subject(s)
Cyclin-Dependent Kinases , DNA Replication , Transcription, Genetic , Humans , Cyclin-Dependent Kinases/metabolism , Cyclin-Dependent Kinases/genetics , DNA Breaks, Double-Stranded , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Cell Line, Tumor , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , DNA Damage
7.
Mol Cell ; 84(15): 2799-2801, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39121840

ABSTRACT

In this issue, Versluis et al.1 use a highly sensitive live-cell imaging system to examine transcription dynamics and functions of various key transcription elongation regulators at the Hsp70 loci.


Subject(s)
Polytene Chromosomes , Transcription Elongation, Genetic , Animals , Polytene Chromosomes/genetics , Polytene Chromosomes/metabolism , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Transcription, Genetic , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Transcription Factors/metabolism , Transcription Factors/genetics
8.
Int J Mol Sci ; 25(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39125980

ABSTRACT

RNA polymerase II (Pol II) dysfunction is frequently implied in human disease. Understanding its functional mechanism is essential for designing innovative therapeutic strategies. To visualize its supra-molecular interactions with genes and nascent RNA, we generated a human cell line carrying ~335 consecutive copies of a recombinant ß-globin gene. Confocal microscopy showed that Pol II was not homogeneously concentrated around these identical gene copies. Moreover, Pol II signals partially overlapped with the genes and their nascent RNA, revealing extensive compartmentalization. Using a cell line carrying a single copy of the ß-globin gene, we also tested if the binding of catalytically dead CRISPR-associated system 9 (dCas9) to different gene regions affected Pol II transcriptional activity. We assessed Pol II localization and nascent RNA levels using chromatin immunoprecipitation and droplet digital reverse transcription PCR, respectively. Some enrichment of transcriptionally paused Pol II accumulated in the promoter region was detected in a strand-specific way of gRNA binding, and there was no decrease in nascent RNA levels. Pol II preserved its transcriptional activity in the presence of DNA-bound dCas9. Our findings contribute further insight into the complex mechanism of mRNA transcription in human cells.


Subject(s)
RNA Polymerase II , Transcription, Genetic , beta-Globins , Humans , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , beta-Globins/genetics , beta-Globins/metabolism , DNA/metabolism , DNA/genetics , Promoter Regions, Genetic , CRISPR-Associated Protein 9/metabolism , CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems , RNA/genetics , RNA/metabolism , RNA, Guide, CRISPR-Cas Systems/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Cell Line
9.
Mol Cell ; 84(15): 2856-2869.e9, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39121843

ABSTRACT

RNA polymerase II (RNA Pol II)-mediated transcription is a critical, highly regulated process aided by protein complexes at distinct steps. Here, to investigate RNA Pol II and transcription-factor-binding and dissociation dynamics, we generated endogenous photoactivatable-GFP (PA-GFP) and HaloTag knockins using CRISPR-Cas9, allowing us to track a population of molecules at the induced Hsp70 loci in Drosophila melanogaster polytene chromosomes. We found that early in the heat-shock response, little RNA Pol II and DRB sensitivity-inducing factor (DSIF) are reused for iterative rounds of transcription. Surprisingly, although PAF1 and Spt6 are found throughout the gene body by chromatin immunoprecipitation (ChIP) assays, they show markedly different binding behaviors. Additionally, we found that PAF1 and Spt6 are only recruited after positive transcription elongation factor (P-TEFb)-mediated phosphorylation and RNA Pol II promoter-proximal pause escape. Finally, we observed that PAF1 may be expendable for transcription of highly expressed genes where nucleosome density is low. Thus, our live-cell imaging data provide key constraints to mechanistic models of transcription regulation.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , RNA Polymerase II , Transcription, Genetic , Transcriptional Elongation Factors , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Animals , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Transcriptional Elongation Factors/metabolism , Transcriptional Elongation Factors/genetics , HSP70 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/genetics , Positive Transcriptional Elongation Factor B/metabolism , Positive Transcriptional Elongation Factor B/genetics , Promoter Regions, Genetic , CRISPR-Cas Systems , Transcription Factors/metabolism , Transcription Factors/genetics , Polytene Chromosomes/genetics , Polytene Chromosomes/metabolism , Gene Expression Regulation , Phosphorylation , Protein Binding , Heat-Shock Response/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Nucleosomes/metabolism , Nucleosomes/genetics
10.
Proc Natl Acad Sci U S A ; 121(36): e2318527121, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39190355

ABSTRACT

Catalysis and translocation of multisubunit DNA-directed RNA polymerases underlie all cellular mRNA synthesis. RNA polymerase II (Pol II) synthesizes eukaryotic pre-mRNAs from a DNA template strand buried in its active site. Structural details of catalysis at near-atomic resolution and precise arrangement of key active site components have been elusive. Here, we present the free-electron laser (FEL) structures of a matched ATP-bound Pol II and the hyperactive Rpb1 T834P bridge helix (BH) mutant at the highest resolution to date. The radiation-damage-free FEL structures reveal the full active site interaction network, including the trigger loop (TL) in the closed conformation, bonafide occupancy of both site A and B Mg2+, and, more importantly, a putative third (site C) Mg2+ analogous to that described for some DNA polymerases but not observed previously for cellular RNA polymerases. Molecular dynamics (MD) simulations of the structures indicate that the third Mg2+ is coordinated and stabilized at its observed position. TL residues provide half of the substrate binding pocket while multiple TL/BH interactions induce conformational changes that could allow translocation upon substrate hydrolysis. Consistent with TL/BH communication, a FEL structure and MD simulations of the T834P mutant reveal rearrangement of some active site interactions supporting potential plasticity in active site function and long-distance effects on both the width of the central channel and TL conformation, likely underlying its increased elongation rate at the expense of fidelity.


Subject(s)
Catalytic Domain , Magnesium , Molecular Dynamics Simulation , RNA Polymerase II , Transcription, Genetic , RNA Polymerase II/metabolism , RNA Polymerase II/chemistry , RNA Polymerase II/genetics , Magnesium/metabolism , Magnesium/chemistry , Lasers , Protein Conformation , Electrons , Protein Binding , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/chemistry , Binding Sites
11.
Mol Cell ; 84(16): 3005-3007, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39178836

ABSTRACT

Complementary studies by Zhao et al.1 and Chen et al.2 reveal how an intrinsically disordered region in MED13 controls mutually exclusive binding of RNA Polymerase II and CDK8 kinase module to Mediator, switching Mediator and transcription activation on and off.


Subject(s)
Cyclin-Dependent Kinase 8 , Mediator Complex , RNA Polymerase II , Mediator Complex/metabolism , Mediator Complex/genetics , Mediator Complex/chemistry , Cyclin-Dependent Kinase 8/metabolism , Cyclin-Dependent Kinase 8/genetics , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Humans , Intrinsically Disordered Proteins/metabolism , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/genetics , Protein Binding , Transcriptional Activation
12.
Nat Commun ; 15(1): 6597, 2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39097586

ABSTRACT

Cyclin-dependent kinase 7 (Cdk7) is required in cell-cycle and transcriptional regulation owing to its function as both a CDK-activating kinase (CAK) and part of transcription factor TFIIH. Cdk7 forms active complexes by associating with Cyclin H and Mat1, and is regulated by two phosphorylations in the activation segment (T loop): the canonical activating modification at T170 and another at S164. Here we report the crystal structure of the human Cdk7/Cyclin H/Mat1 complex containing both T-loop phosphorylations. Whereas pT170 coordinates basic residues conserved in other CDKs, pS164 nucleates an arginine network unique to the ternary Cdk7 complex, involving all three subunits. We identify differential dependencies of kinase activity and substrate recognition on the individual phosphorylations. CAK function is unaffected by T-loop phosphorylation, whereas activity towards non-CDK substrates is increased several-fold by T170 phosphorylation. Moreover, dual T-loop phosphorylation stimulates multisite phosphorylation of the RNA polymerase II (RNAPII) carboxy-terminal domain (CTD) and SPT5 carboxy-terminal repeat (CTR) region. In human cells, Cdk7 activation is a two-step process wherein S164 phosphorylation precedes, and may prime, T170 phosphorylation. Thus, dual T-loop phosphorylation can regulate Cdk7 through multiple mechanisms, with pS164 supporting tripartite complex formation and possibly influencing processivity, while pT170 enhances activity towards key transcriptional substrates.


Subject(s)
Cyclin-Dependent Kinase-Activating Kinase , Cyclin-Dependent Kinases , Phosphorylation , Humans , Cyclin-Dependent Kinases/metabolism , Cyclin-Dependent Kinases/chemistry , Cyclin-Dependent Kinases/genetics , Cyclin H/metabolism , Cyclin H/chemistry , Cyclin H/genetics , Crystallography, X-Ray , RNA Polymerase II/metabolism , RNA Polymerase II/chemistry , Transcription Factor TFIIH/metabolism , Transcription Factor TFIIH/chemistry , Transcription Factor TFIIH/genetics , Models, Molecular , Transcription Factors/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Protein Domains , Cell Cycle Proteins
13.
Nat Commun ; 15(1): 7089, 2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39154022

ABSTRACT

Transcription-blocking lesions (TBLs) stall elongating RNA polymerase II (Pol II), which then initiates transcription-coupled repair (TCR) to remove TBLs and allow transcription recovery. In the absence of TCR, eviction of lesion-stalled Pol II is required for alternative pathways to address the damage, but the mechanism is unclear. Using Protein-Associated DNA Damage Sequencing (PADD-seq), this study reveals that the p97-proteasome pathway can evict lesion-stalled Pol II independently of repair. Both TCR and repair-independent eviction require CSA and ubiquitination. However, p97 is dispensable for TCR and Pol II eviction in TCR-proficient cells, highlighting repair's prioritization over repair-independent eviction. Moreover, ubiquitination of RPB1-K1268 is important for both pathways, with USP7's deubiquitinase activity promoting TCR without abolishing repair-independent Pol II release. In summary, this study elucidates the fate of lesion-stalled Pol II, and may shed light on the molecular basis of genetic diseases caused by the defects of TCR genes.


Subject(s)
DNA Damage , DNA Repair , RNA Polymerase II , Transcription, Genetic , Ubiquitination , RNA Polymerase II/metabolism , Humans , Ubiquitin-Specific Peptidase 7/metabolism , Ubiquitin-Specific Peptidase 7/genetics , Valosin Containing Protein/metabolism , Valosin Containing Protein/genetics , Proteasome Endopeptidase Complex/metabolism , Excision Repair
14.
Elife ; 132024 Aug 23.
Article in English | MEDLINE | ID: mdl-39177021

ABSTRACT

MYC family oncoproteins regulate the expression of a large number of genes and broadly stimulate elongation by RNA polymerase II (RNAPII). While the factors that control the chromatin association of MYC proteins are well understood, much less is known about how interacting proteins mediate MYC's effects on transcription. Here, we show that TFIIIC, an architectural protein complex that controls the three-dimensional chromatin organisation at its target sites, binds directly to the amino-terminal transcriptional regulatory domain of MYCN. Surprisingly, TFIIIC has no discernible role in MYCN-dependent gene expression and transcription elongation. Instead, MYCN and TFIIIC preferentially bind to promoters with paused RNAPII and globally limit the accumulation of non-phosphorylated RNAPII at promoters. Consistent with its ubiquitous role in transcription, MYCN broadly participates in hubs of active promoters. Depletion of TFIIIC further increases MYCN localisation to these hubs. This increase correlates with a failure of the nuclear exosome and BRCA1, both of which are involved in nascent RNA degradation, to localise to active promoters. Our data suggest that MYCN and TFIIIC exert an censoring function in early transcription that limits promoter accumulation of inactive RNAPII and facilitates promoter-proximal degradation of nascent RNA.


Subject(s)
Chromatin , N-Myc Proto-Oncogene Protein , Promoter Regions, Genetic , RNA Polymerase II , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , N-Myc Proto-Oncogene Protein/metabolism , N-Myc Proto-Oncogene Protein/genetics , Humans , Chromatin/metabolism , Protein Binding , Transcription Factors, TFII/metabolism , Transcription Factors, TFII/genetics , Transcription, Genetic , Cell Line, Tumor
15.
Sci Adv ; 10(27): eadm9740, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38959309

ABSTRACT

Micrococcal nuclease sequencing is the state-of-the-art method for determining chromatin structure and nucleosome positioning. Data analysis is complex due to the AT-dependent sequence bias of the endonuclease and the requirement for high sequencing depth. Here, we present the nucleosome-based MNase accessibility (nucMACC) pipeline unveiling the regulatory chromatin landscape by measuring nucleosome accessibility and stability. The nucMACC pipeline represents a systematic and genome-wide approach for detecting unstable ("fragile") nucleosomes. We have characterized the regulatory nucleosome landscape in Drosophila melanogaster, Saccharomyces cerevisiae, and mammals. Two functionally distinct sets of promoters were characterized, one associated with an unstable nucleosome and the other being nucleosome depleted. We show that unstable nucleosomes present intermediate states of nucleosome remodeling, preparing inducible genes for transcriptional activation in response to stimuli or stress. The presence of unstable nucleosomes correlates with RNA polymerase II proximal pausing. The nucMACC pipeline offers unparalleled precision and depth in nucleosome research and is a valuable tool for future nucleosome studies.


Subject(s)
Drosophila melanogaster , Micrococcal Nuclease , Nucleosomes , Saccharomyces cerevisiae , Nucleosomes/metabolism , Nucleosomes/genetics , Animals , Micrococcal Nuclease/metabolism , Drosophila melanogaster/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Chromatin Assembly and Disassembly , Genome , Promoter Regions, Genetic , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Chromatin/genetics , Chromatin/metabolism , Sequence Analysis, DNA/methods
16.
Mol Cell ; 84(13): 2405-2406, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38996456

ABSTRACT

In this issue of Molecular Cell, Razew et al.1 and Sabath et al.2 assign function to an unexplored module of the Integrator (INT) complex, expanding the toolbox of this genome-wide attenuator of RNA polymerase II (RNAPII) transcription.


Subject(s)
RNA Polymerase II , Transcription, Genetic , RNA Polymerase II/metabolism , RNA Polymerase II/genetics
17.
Methods Mol Biol ; 2805: 137-151, 2024.
Article in English | MEDLINE | ID: mdl-39008179

ABSTRACT

Transcription in developing metazoans is inherently stochastic, involving transient and dynamic interactions among transcriptional machinery. A fundamental challenge with traditional techniques, including fixed-tissue protein and RNA staining, is the lack of temporal resolution. Quantifying kinetic changes in transcription can elucidate underlying mechanisms of interaction among regulatory modules. In this protocol, we describe the successful implementation of a combination of MS2/MCP and PP7/PCP systems in living Drosophila embryos to further our understanding of transcriptional dynamics during development. Our technique can be extended to visualize transcriptional activities of multiple genes or alleles simultaneously, characterize allele-specific expression of a target gene, and quantitatively analyze RNA polymerase II activity in a single-cell resolution.


Subject(s)
Embryonic Development , Gene Expression Regulation, Developmental , Animals , Embryonic Development/genetics , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Embryo, Nonmammalian/metabolism , Drosophila/embryology , Drosophila/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Transcription, Genetic , Drosophila Proteins/genetics , Drosophila Proteins/metabolism
18.
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
19.
Nat Commun ; 15(1): 5859, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38997286

ABSTRACT

Signal-induced transcriptional programs regulate critical biological processes through the precise spatiotemporal activation of Immediate Early Genes (IEGs); however, the mechanisms of transcription induction remain poorly understood. By combining an acute depletion system with several genomics approaches to interrogate synchronized, temporal transcription, we reveal that KAP1/TRIM28 is a first responder that fulfills the temporal and heightened transcriptional demand of IEGs. Acute KAP1 loss triggers an increase in RNA polymerase II elongation kinetics during early stimulation time points. This elongation defect derails the normal progression through the transcriptional cycle during late stimulation time points, ultimately leading to decreased recruitment of the transcription apparatus for re-initiation thereby dampening IEGs transcriptional output. Collectively, KAP1 plays a counterintuitive role by negatively regulating transcription elongation to support full activation across multiple transcription cycles of genes critical for cell physiology and organismal functions.


Subject(s)
RNA Polymerase II , Tripartite Motif-Containing Protein 28 , Tripartite Motif-Containing Protein 28/metabolism , Tripartite Motif-Containing Protein 28/genetics , RNA Polymerase II/metabolism , Humans , Kinetics , Transcription Elongation, Genetic , Genes, Immediate-Early , Transcription, Genetic , Signal Transduction , Transcriptional Activation , Animals
20.
Antonie Van Leeuwenhoek ; 117(1): 109, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39083124

ABSTRACT

The genetic variety and habitats of Camptophora species, generally known as black yeast, have not been clarified. In this study, we re-evaluated Camptophora based on morphological observations and phylogenetic analyses. Because prior investigations on Camptophora only included a few strains/specimens, 24 Camptophora-related strains were newly obtained from 13 leaf samples of various plant species to redefine the genetic and species concepts of Camptophora. Their molecular phylogenetic relationships were examined using small subunit nuclear ribosomal DNA (nSSU, 18S rDNA), the internal transcribed spacer (ITS) rDNA operon, the large subunit nuclear ribosomal DNA (LSU, 28S rDNA), ß-tubulin, the second largest subunit of RNA polymerase II (rpb2), and mitochondrial small subunit DNA (mtSSU). Single- and multi-locus analyses using nSSU-ITS-LSU-rpb2-mtSSU revealed a robust phylogenetic relationship among Camptophora species within Chaetothyriaceae. Camptophora species could be distinguished from other chaetothyriaceous genera by their snake-shaped conidia with microcyclic conidiation and loosely interwoven mycelial masses. Based on the results of phylogenetic analyses, two undescribed lineages were recognized, and Ca. schimae was excluded from the genus. ITS sequence comparison with environmental DNA sequences revealed that the distribution of the genus is restricted to the Asia-Pacific region. Camptophora has been isolated or detected from abrupt sources, and this was attributed to its microcycle. The mechanisms driving genetic diversity within species are discussed with respect to their phyllosphere habitats.


Subject(s)
DNA, Fungal , Phylogeny , DNA, Fungal/genetics , DNA, Ribosomal/genetics , DNA, Ribosomal Spacer/genetics , Spores, Fungal/genetics , Spores, Fungal/cytology , Spores, Fungal/classification , Sequence Analysis, DNA , Plant Leaves/microbiology , RNA Polymerase II/genetics , Ascomycota/genetics , Ascomycota/classification , Tubulin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL